CN117858707A - CBL-B modulators and uses thereof - Google Patents

CBL-B modulators and uses thereof Download PDF

Info

Publication number
CN117858707A
CN117858707A CN202280040238.XA CN202280040238A CN117858707A CN 117858707 A CN117858707 A CN 117858707A CN 202280040238 A CN202280040238 A CN 202280040238A CN 117858707 A CN117858707 A CN 117858707A
Authority
CN
China
Prior art keywords
cancer
nitrogen
sulfur
oxygen
ring
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280040238.XA
Other languages
Chinese (zh)
Inventor
S·M·利特德莫拉代
A·V·韦斯特
T·贝克
J·C·阿雷吉
D·卡斯塔格纳
J·R·格林伍德
S·拉菲
F·麦克罗伯
Y·张
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Lindbergh Cleo Co
Original Assignee
Lindbergh Cleo Co
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Lindbergh Cleo Co filed Critical Lindbergh Cleo Co
Priority claimed from PCT/US2022/071633 external-priority patent/WO2022217276A1/en
Publication of CN117858707A publication Critical patent/CN117858707A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides compounds, compositions thereof, and methods of using the same to inhibit Cbl-b and to treat Cbl-b mediated disorders.

Description

CBL-B modulators and uses thereof
Cross reference to related applications
U.S. provisional application No. 63/264,940 filed on 3/12/2021; and priority rights of U.S. provisional application No. 63/173,121, filed on 4/9 of 2021, each of which is incorporated herein by reference in its entirety.
Technical Field
The present invention relates to compounds and methods useful for inhibiting E3 ligase Casitas B lineage lymphoma B (Cbl-B). The invention also provides pharmaceutically acceptable compositions comprising the compounds of the invention and methods of using the compositions in the treatment of various disorders.
Background
Ubiquitination is a post-translational modification that regulates the function and fate of proteins involved in physiological processes. The addition of ubiquitin to target proteins occurs through a three-step enzymatic process involving three enzymes. The first enzyme E1 catalyzes ubiquitin activation. The activated ubiquitin is then transferred from E1 to ubiquitin conjugating enzyme E2. The third enzyme or E3 ligase imparts substrate specificity and directly catalyzes the transfer of ubiquitin from E2 into the protein substrate. The addition of polyubiquitin chains to proteins serves as a signal that leads to degradation by proteasome to peptides of ubiquitin conjugated proteins. Additionally, polyubiquitination and monoubiquitination can also alter the cellular localization, function, and interaction of protein substrates with proteins required for downstream activity and signaling events.
Ubiquitination controls a number of biological processes that are often deregulated in disease, including cell cycle, DNA repair, differentiation, and innate and adaptive immunity. Thus, the discovery of molecules that regulate components of the ubiquitin-proteasome system represents an attractive therapeutic opportunity for a wide range of conditions including cancer and autoimmune diseases.
The compounds and compositions described herein are generally useful for inhibiting E3 ligase Casitas B lineage lymphoma B (Cbl-B).
Cbl-b is a RING finger E3 ligase and is a member of the highly conserved Cbl protein family, consisting of three Cbl genes in mammals: cbl, cbl-b and Cbl-c. The Cbl protein interacts with a target protein through its protein-protein interaction domain, allowing modulation of multiple signaling pathways involving Tyrosine Kinase (TK) signaling in a variety of cell types. The structure of the Cbl protein consists of an amino terminal Tyrosine Kinase Binding Domain (TKBD), a Linker Helical Region (LHR) and a truly interesting new gene (RING) domain, followed by the carboxy-terminal region of the binding site containing the Src homology 2 (SH 2) domain and the Src homology 3 (SH 3) domain. Cbl TKBD consists of a four-helix bundle (4H), EF hand and variant SH2 domain that binds to a substrate, such as activated TK, in a phospho-tyrosine dependent manner.
Ubiquitination of activated receptor TK by Cbl-b regulates assembly of endocytic proteins at both membrane and sortilin endosomes to facilitate lysosomal targeting, degradation and signal termination. Cbl-b is also important for down-regulating signaling from antigen and cytokine receptors through ubiquitination of the receptor chain and associated cytoplasmic TK, leading to inactivation and/or proteasome degradation.
Cbl-b is expressed in the immune cell lineage and acts as the primary regulator of immune cell activation and maintenance of peripheral tolerance. Cbl-b down regulates adaptive immune system signaling by establishing a threshold for activation of antigen receptors. In T cells, cbl-b requires that the co-stimulatory signal produce a productive immune response upon T Cell Receptor (TCR) engagement. Mice lacking Cbl-b, and more specifically RING Zn in Cbl-b, refer to the ligase domain, showing tumor rejection mediated by cd8+ T cells.
In addition, cbl-b modulates the activity of multiple cell lineages involved in innate immunity, including NK cells, antigen presenting Dendritic Cells (DCs), and monocytes. Thus, the function of Cbl-b may be cell type dependent due to the complexity and diversity of the protein targets of Cbl-b in a variety of immune cells.
Novel therapeutic approaches aimed at removing inhibitory signals in immune cells to enhance the productive immune system have recently been of interest. Given the central role of Cbl-b in regulating multiple signaling mechanisms in both innate and adaptive immunity, inhibition of Cbl-b provides a therapeutic opportunity for inclusion of cancer immunotherapy.
Cbl-b inhibitors may enhance the activity of cancer vaccines. For example, adoptive transfer of Cbl-b-/-cd8+ T cells in combination with DC vaccine has been reported to delay tumor growth. In addition, cbl-b-/-T cells are resistant to inhibition of PDL-1/PD-1 in vitro and in vivo, which supports the principle combination of Cbl-b inhibitors with anti-PD-1/PD-L1 checkpoint blockade.
The enhanced expression of Cbl-b correlates with a better prognosis in lung adenocarcinoma. In addition, mutations in the RING finger domains of the Cbl protein and Cbl-b linker sequences can be found in a variety of conditions and cancers, including juvenile myelomonocytic leukemia (JMML), pre-leukemia chronic myelomonocytic leukemia (CMML), myeloproliferative neoplasms (MPN), and Acute Myelogenous Leukemia (AML). These observations indicate that impaired degradation of activated TK represents an important cancer mechanism involving Cbl proteins. Consistently, several reports have demonstrated that Epidermal Growth Factor Receptor (EGFR) and platelet-derived growth factor receptor alpha (PDGFRa) are ubiquitinated by Cbl-b. Ubiquitination of these receptors promotes proteasome-dependent degradation in various cancer lineages. Degradation of EGFR by Cbl-b results in proliferation of lung and gastric cancer cells and mediates epithelial-mesenchymal transition (EMT) in metastatic breast and gastric cancers. Additionally, amplification and mutation of both EGFR and PDGFR are major drivers of oncogenic transformation and are common in many types of cancer. Thus, cbl-b inhibition represents an opportunity for both tumor-intrinsic and tumor-extrinsic therapies.
Disclosure of Invention
It has now been found that the compounds of the present invention and pharmaceutically acceptable compositions thereof are effective as inhibitors of Cbl-b. In certain embodiments, the invention provides compounds of formula (la) presented herein.
The compounds of the invention and pharmaceutically acceptable compositions thereof are useful for treating various diseases, disorders, or conditions associated with modulating the immune system involving Cbl-b. Such diseases, disorders, or conditions include the diseases, disorders, or conditions described herein.
The compounds provided by the invention can also be used for researching Cbl-b enzyme in biological and pathological phenomena; study of ubiquitination occurring in body tissues; and comparative evaluation of novel Cbl-b inhibitors or other modulators of cell cycle, DNA repair, differentiation, and innate and adaptive immunity in vitro or in vivo.
Detailed Description
1. General description of certain embodiments of the invention:
in certain aspects, the invention provides a compound of formula I:
or a pharmaceutically acceptable salt thereof, wherein L, X, Y, R 1 、R 2 、R 3 、R 6 、R 8 、R 9 Each of m, n, p, q and t, alone or in combination, is as defined above and as described in the embodiments herein.
In some embodiments, the present invention provides a pharmaceutical composition comprising a compound of formula I and a pharmaceutically acceptable carrier, adjuvant or diluent.
In some embodiments, the invention provides a method of treating a Cbl-b mediated disease, disorder, or condition, the method comprising administering to a patient in need thereof a compound of formula I or a pharmaceutically acceptable salt thereof.
2. Compounds and definitions:
the compounds of the present invention comprise compounds generally described herein and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. For the purposes of the present invention, chemical elements are identified according to the periodic Table of the elements (Periodic Table of the Elements), CAS version, handbook of chemistry and Physics (Handbook of Chemistry and Physics), 75 th edition. Furthermore, general principles of organic chemistry are described in "organic chemistry (Organic Chemistry)", thomas Sorrell, university of Sosaritot, books press (University Science Books, sausalito): 1999 "[ March's Advanced Organic Chemistry)", 5 th edition, editions Smith, M.B., and March, J., john Wiley & Sons, new York, the entire contents of which are hereby incorporated by reference.
As used herein, the term "aliphatic" or "aliphatic group" means a straight (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is fully saturated or contains one or more unsaturated units, or a monocyclic or bicyclic hydrocarbon that is fully saturated or contains one or more unsaturated units but is not aromatic (also referred to herein as "carbocyclic", "alicyclic" or "cycloalkyl") having a single point of attachment to the rest of the molecule. Unless otherwise indicated, aliphatic groups contain 1-6 aliphatic carbon atoms. In some embodiments, the aliphatic group contains 1 to 5 aliphatic carbon atoms. In other embodiments, the aliphatic group contains 1-4 aliphatic carbon atoms. In still other embodiments, the aliphatic group contains 1-3 aliphatic carbon atoms, and in still other embodiments, the aliphatic group contains1-2 aliphatic carbon atoms. In some embodiments, "alicyclic" (or "carbocycle" or "cycloalkyl") refers to an aromatic monocyclic C that is fully saturated or contains one or more unsaturated units, but not has a single point of attachment to the remainder of the molecule 3 -C 6 And (3) hydrocarbons. Suitable aliphatic groups include, but are not limited to, straight or branched substituted or unsubstituted alkyl, alkenyl, alkynyl and hybrids thereof, such as (cycloalkyl) alkyl, (cycloalkenyl) alkyl or (cycloalkyl) alkenyl.
As used herein, the term "bridged bicyclic" refers to any bicyclic ring system having at least one bridge, i.e., carbocyclic or heterocyclic, saturated or partially unsaturated. As defined by IUPAC, a "bridge" is an unbranched chain of atoms or an atom or bond connecting two bridgeheads, wherein a "bridgehead" is any backbone atom of a ring system that is bonded to three or more backbone atoms (excluding hydrogen). In some embodiments, the bridge Lian Shuanghuan group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Such bridge Lian Shuanghuan groups are well known in the art and include those set forth below, wherein each group is attached to the remainder of the molecule at any substitutable carbon or nitrogen atom. Unless otherwise indicated, the bridge Lian Shuanghuan groups are optionally substituted with one or more substituents listed for the aliphatic group. Additionally or alternatively, any substitutable nitrogen of the bridge Lian Shuanghuan group is optionally substituted. Exemplary bridged bicyclic rings include:
the term "lower alkyl" refers to C 1-4 Linear or branched alkyl. Exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl.
The term "lower haloalkyl" refers to C substituted with one or more halogen atoms 1-4 Linear or branched alkyl.
The term "heteroatom" means one of oxygen, sulfur, nitrogen, phosphorus or siliconOne or more (comprising any oxidized form of nitrogen, sulfur, phosphorus or silicon; quaternized form of any basic nitrogen or; substitutable nitrogen of a heterocycle, e.g. N (as in 3, 4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR + (as in N-substituted pyrrolidinyl)).
As used herein, the term "unsaturated" means that a portion has one or more unsaturated units.
As used herein, the term "divalent C 1-8 (or C) 1-6 ) Saturated or unsaturated straight or branched hydrocarbon chain "refers to straight or branched divalent alkylene, alkenylene, and alkynylene chains as defined herein.
The term "alkylene" refers to a divalent alkyl group. "alkylene chain" is polymethylene, i.e., - (CH) 2 ) n -wherein n is a positive integer, preferably 1 to 6, 1 to 4, 1 to 3, 1 to 2 or 2 to 3. A substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced by substituents. Suitable substituents include those described below for substituted aliphatic groups.
The term "alkenylene" refers to a divalent alkenyl group. A substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced by a substituent. Suitable substituents include those described below for substituted aliphatic groups.
The term "halogen" means F, cl, br or I.
The term "aryl" as used in "aralkyl", "aralkoxy" or "aryloxyalkyl" alone or as part of a larger moiety refers to a monocyclic or bicyclic ring system having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic, and wherein each ring in the system contains 3 to 7 ring members. The term "aryl" may be used interchangeably with the term "aryl ring". In certain embodiments of the present invention, "aryl" refers to an aromatic ring system including, but not limited to, phenyl, biphenyl, naphthyl, anthracenyl, and the like, which may carry one or more substituents. As used herein, the term "aryl" also includes within its scope groups in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, phthalimidyl, naphthalimidyl, phenanthridinyl, tetrahydronaphthyl, or the like.
The terms "heteroaryl" and "heteroaryl-" used alone or as part of a larger moiety, such as "heteroarylalkyl" or "heteroarylalkoxy", refer to groups having 5 to 10 ring atoms sharing 6, 10, or 14 pi electrons in a ring array; preferably 5, 6 or 9 ring atoms; and has one to five heteroatoms in addition to carbon atoms. The term "heteroatom" refers to nitrogen, oxygen or sulfur and includes any oxidized form of nitrogen or sulfur and any quaternized form of basic nitrogen. Heteroaryl groups include, but are not limited to: thienyl, furyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl and pteridinyl. As used herein, the terms "heteroaryl" and "heteroaryl-" also include groups in which a heteroaromatic ring is fused to one or more aryl, alicyclic, or heterocyclic rings, wherein the group or point of attachment is located on the heteroaromatic ring or one of the rings to which the heteroaromatic ring is fused, unless otherwise indicated. Non-limiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzothiazolyl, quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, and tetrahydroisoquinolinyl. Heteroaryl groups may be monocyclic or bicyclic. The term "heteroaryl" may be used interchangeably with the terms "heteroaryl ring", "heteroaryl group" or "heteroaromatic", any of which terms comprise an optionally substituted ring. The term "heteroarylalkyl" refers to an alkyl group substituted with a heteroaryl group, wherein the alkyl and heteroaryl moieties are independently optionally substituted.
Such as the bookAs used herein, the terms "heterocycle", "heterocyclyl", "heterocyclic group" and "heterocycle (heterocyclic ring)" are used interchangeably and refer to a stable 5-to 7-membered monocyclic or 7-10-membered bicyclic heterocyclic moiety, saturated or partially unsaturated, having one or more, preferably one to four heteroatoms in addition to carbon atoms, as defined above. The term "nitrogen" when used with respect to a ring atom of a heterocycle includes substituted nitrogen. As examples, in saturated or partially unsaturated rings having 0 to 3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3, 4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or + NR (as in N-substituted pyrrolidinyl).
The heterocycle may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure, and any of the ring atoms may be optionally substituted. Examples of such saturated or partially unsaturated heterocyclyl groups include, but are not limited to, tetrahydrofuranyl, tetrahydrothiophenylpyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazaperyl, oxazepinyl, thiazepinyl, morpholinyl, 2-oxa-6-azaspiro [3.3] heptane and quinuclidinyl. The terms "heterocycle", "heterocyclyl (heterocyclic group)", "heterocyclic moiety" and "heterocyclic group" are used interchangeably herein and also include groups in which the heterocyclyl ring is fused to one or more aryl, heteroaryl or alicyclic rings, such as indolinyl, 3H-indolyl, chromanyl, phenanthridinyl or tetrahydroquinolinyl. The heterocyclyl may be monocyclic or bicyclic. The term "heterocycloalkyl" refers to an alkyl group substituted with a heterocyclyl group, wherein the alkyl and heterocyclyl moieties are independently optionally substituted.
As used herein, the term "partially unsaturated" refers to a ring moiety that contains at least one double or triple bond. The term "partially unsaturated" is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as defined herein.
As described herein, the compounds of the invention may contain an "optionally substituted" moiety. Generally, the term "substituted", whether preceded by the term "optionally" or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an "optionally substituted" group may have the appropriate substituent at each substitutable position of the group, and where more than one position in any given structure may be substituted with more than one substituent selected from the specified group, the substituents may be the same or different at each position. The combinations of substituents contemplated by the present invention are preferably those resulting in the formation of stable or chemically viable compounds. As used herein, the term "stable" refers to a compound that does not substantially change when subjected to conditions that allow it to be produced, detected, and in certain embodiments recovered, purified, and used for one or more of the purposes disclosed herein.
Suitable monovalent substituents on the substitutable carbon atom of an "optionally substituted" group are independently halogen; - (CH) 2 ) 0-4 R o ;-(CH 2 ) 0-4 OR o ;-O(CH 2 ) 0-4 R o 、-O-(CH 2 ) 0-4 C(O)OR;-(CH 2 ) 0-4 CH(OR o ) 2 ;-(CH 2 ) 0- 4 SR o ;-(CH 2 ) 0-4 Ph, which may be substituted with R; - (CH) 2 ) 0-4 O(CH 2 ) 0-1 Ph, which may be substituted with R; -ch=chph, which may be substituted by R; - (CH) 2 ) 0-4 O(CH 2 ) 0-1 -pyridinyl, which may be substituted by R; -NO 2 ;-CN;-N 3 ;-(CH 2 ) 0-4 N(R o ) 2 ;-(CH 2 ) 0-4 N(R o )C(O)R o ;-N(R o )C(S)R o ;-(CH 2 ) 0-4 N(R o )C(O)NR o 2 ;-N(R o )C(S)NR o 2 ;-(CH 2 ) 0-4 N(R o )C(O)OR o ;-N(R o )N(R o )C(O)R o ;-N(R o )N(R o )C(O)NR o 2 ;-N(R o )N(R o )C(O)OR o ;-N(R o )C(NR o )N(R o ) 2 ;-(CH 2 ) 0-4 C(O)R o ;-C(S)R o ;-(CH 2 ) 0-4 C(O)OR o ;-(CH 2 ) 0-4 C(O)SR o ;-(CH 2 ) 0-4 C(O)OSiR o 3 ;-(CH 2 ) 0-4 OC(O)R o ;-OC(O)(CH 2 ) 0-4 SR;-(CH 2 ) 0-4 SC(O)R o ;-(CH 2 ) 0-4 C(O)NR o 2 ;-C(S)NR o 2 ;-C(S)SR;-SC(S)SR、-(CH 2 ) 0-4 OC(O)NR o 2 ;-C(O)N(OR o )R o ;-C(O)C(O)R o ;-C(O)CH 2 C(O)R o ;-C(NOR o )R o ;-(CH 2 ) 0-4 SSR o ;-(CH 2 ) 0-4 S(O) 2 R o ;-(CH 2 ) 0-4 S(O) 2 OR o ;-(CH 2 ) 0-4 OS(O) 2 R o ;-S(O) 2 NR o 2 ;-(CH 2 ) 0-4 S(O)R o ;-N(R o )S(O) 2 NR o 2 ;-N(R o )S(O) 2 R o ;-N(OR o )R o ;-C(NH)NR o 2 ;-(CH 2 ) 0-4 P(O) 2 R o ;-(CH 2 ) 0-4 P(O)R o 2 ;-(CH 2 ) 0-4 OP(O)R o 2 ;-(CH 2 ) 0-4 OP(O)(OR o ) 2 ;-SiR o 3 ;-(C 1-4 Linear or branched alkylene) O-N (R) o ) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or- (C) 1-4 Straight or branched chain alkylene) C (O) O-N (R) o ) 2 Wherein each R is o May be substituted as defined below and independently hydrogen, C 1-6 Aliphatic, -CH 2 Ph、-O(CH 2 ) 0-1 Ph、-CH 2 - (5-6 membered heteroaryl ring) or a 5-6 membered saturated, partially unsaturated or aryl ring having 0 to 4 heteroatoms independently selected from nitrogen, oxygen or sulfur, or two independently occurring R's, although defined above o Together with the intervening atoms thereof form a 3-to 12-membered saturated, partially unsaturated or aryl monocyclic or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur, which may be substituted as defined below.
R o (or by two independently-occurring R' s o Ring formed with an intervening atom) is independently halogen, - (CH) 2 ) 0-2 R · - (halo R) · )、-(CH 2 ) 0-2 OH、-(CH 2 ) 0-2 OR · 、-(CH 2 ) 0-2 CH(OR · ) 2 (halo) R · )、-CN、-N 3 、-(CH 2 ) 0-2 C(O)R · 、-(CH 2 ) 0-2 C(O)OH、-(CH 2 ) 0-2 C(O)OR · 、-(CH 2 ) 0-2 SR · 、-(CH 2 ) 0- 2 SH、-(CH 2 ) 0-2 NH 2 、-(CH 2 ) 0-2 NHR · 、-(CH 2 ) 0-2 NR · 2 、-NO 2 、-SiR · 3 、-OSiR · 3 、-C(O)SR · 、-(C 1-4 Straight-chain OR branched alkylene) C (O) OR · or-SSR · Wherein each R is · Unsubstituted or substituted with one or more halogens only in the case of the preceding "halo" and independently selected from C 1-4 Aliphatic series; -CH 2 Ph;-O(CH 2 ) 0-1 Ph; or a 5-6 membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. R is R o Suitable divalent substituents on saturated carbon atoms of (c) comprise =o and =s.
Synthesis of an "optionally substituted" group on a saturated carbon atomSuitable divalent substituents include the following: =o, =s, =nnr * 2 、=NNHC(O)R * 、=NNHC(O)OR * 、=NNHS(O) 2 R * 、=NR * 、=NOR * 、-O(C(R * 2 )) 2-3 O-or-S (C (R) * 2 )) 2-3 S-, wherein each independently occurs R * Selected from hydrogen; c (C) 1-6 Aliphatic, which may be substituted as defined below; or an unsubstituted 5-6 membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents bonded to the ortho-substitutable carbon of the "optionally substituted" group include: -O (CR) * 2 ) 2- 3 O-, wherein each independently occurs R * Selected from hydrogen; c (C) 1-6 Aliphatic, which may be substituted as defined below; or an unsubstituted 5-6 membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
R * Suitable substituents on the aliphatic radical of (2) comprise halogen; -R · The method comprises the steps of carrying out a first treatment on the surface of the - (halo R) · );-OH;-OR · The method comprises the steps of carrying out a first treatment on the surface of the -O (halo R) · );-CN;-C(O)OH;-C(O)OR · ;-NH 2 ;-NHR · ;-NR · 2 or-NO 2 Wherein each R is · Unsubstituted or substituted with one or more halogens only in the case of the preceding with "halo", and is independently C 1-4 Aliphatic series; -CH 2 Ph;-O(CH 2 ) 0- 1 Ph; or a 5-6 membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
Suitable substituents on the substitutable nitrogen of an "optionally substituted" group include Or->Each of which is->Independently hydrogen; c (C) 1-6 Aliphatic, which may be substituted as defined below; unsubstituted-OPh; or an unsubstituted 5-6 membered saturated, partially unsaturated or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur, or, in spite of the above definition, two independently occurringTogether with the intervening atoms thereof form an unsubstituted 3-12 membered saturated, partially unsaturated or aryl monocyclic or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.
Suitable substituents on the aliphatic radical of (2) are independently halogen; -R · The method comprises the steps of carrying out a first treatment on the surface of the - (halo R) · );-OH;-OR · The method comprises the steps of carrying out a first treatment on the surface of the -O (halo R) · );-CN;-C(O)OH;-C(O)OR · ;-NH 2 ;-NHR · ;-NR · 2 or-NO 2 Wherein each R is · Unsubstituted or substituted with one or more halogens only in the case of the preceding with "halo", and is independently C 1-4 Aliphatic series; -CH 2 Ph;-O(CH 2 ) 0-1 Ph; or a 5-6 membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
As used herein, the term "pharmaceutically acceptable salts" refers to those salts that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S.M. Berge et al, J.pharmaceutical Sciences, incorporated herein by reference, describe in detail pharmaceutically acceptable salts, 1977,66,1-19. Pharmaceutically acceptable salts of the compounds of the invention include salts derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable non-toxic acid addition salts are salts of amino groups with inorganic acids (such as hydrochloric, hydrobromic, phosphoric, sulfuric and perchloric) or organic acids (such as acetic, oxalic, maleic, tartaric, citric, succinic or malonic) or by using other methods used in the art (such as ion exchange). Other pharmaceutically acceptable salts include adipic acid, alginates, ascorbates, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphor, sulfonate, camphorsulfonate, citrate, cyclopentane propionate, digluconate, dodecyl sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoic acid, hexanoate, hydroiodic acid, 2-hydroxyethanesulfonate, lactobionate, lactate, laurate, dodecyl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectate, persulfate, 3-phenylpropionate, phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, sodium p-toluenesulfonate, undecanoate, valerate, and the like.
Salts derived from suitable bases include alkali metal salts, alkaline earth metal salts, ammonium salts and N + (C 1-4 Alkyl group 4 And (3) salt. Representative alkali metal or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Where appropriate, additional pharmaceutically acceptable salts comprise nontoxic ammonium, quaternary ammonium and amine cations formed using counterions such as halides, hydroxides, carboxylates, sulphates, phosphates, nitrates, lower alkyl sulphonates and aryl sulphonates.
Unless otherwiseStated otherwise, structures described herein are also meant to encompass all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structures; for example, the R and S configuration, Z and E double bond isomers, and Z and E conformational isomers for each asymmetric center. Thus, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the compounds of the invention are within the scope of the invention. Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention. In addition, unless otherwise indicated, structures depicted herein are also intended to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, having a composition comprising deuterium or tritium substituted for hydrogen or with 13 C-or 14 Compounds of the structure of the invention in which C-enriched carbon replaces carbon are within the scope of the invention. Such compounds may be used, for example, as analytical tools, probes in bioassays, or as therapeutic agents according to the present invention. In certain embodiments, warhead portion R of the provided compounds 1 Including one or more deuterium atoms. In certain embodiments, ring B of the provided compounds may be substituted with one or more deuterium atoms.
Unless marked as an absolute configuration, structures as drawn represent relative configurations. The present invention contemplates individual enantiomers and racemic mixtures.
As used herein, a "Cbl-b inhibitor" is a molecule that reduces, inhibits, or otherwise decreases one or more of the biological activities of Cbl-b (e.g., ubiquitination, modulation of tyrosine kinase signaling, or modulation of immune cell activation, and maintenance of peripheral tolerance). Inhibition with a Cbl-b inhibitor is not necessarily indicative of total elimination of Cbl-b activity. In contrast, the activity may be reduced by a statistically significant amount, comprising, for example, at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 95% or 100% of the activity of Cbl-b, as compared to an appropriate control. In some embodiments, the Cbl-b inhibitor reduces, inhibits or otherwise reduces the ubiquitination activity of Cbl-b. In some of these embodiments, the Cbl-b inhibitor reduces, inhibits or otherwise reduces Cbl-b mediated ubiquitination of the tyrosine kinase. The disclosed compounds bind directly to CBl-b and inhibit its ubiquitination activity.
By "specific inhibitor" is meant an agent that reduces, inhibits or otherwise reduces the activity of a defined target that is more active than an unrelated target. For example, an amount of a Cbl-b specific inhibitor that reduces at least one biological activity of Cbl-b is statistically greater than the inhibitory effect of an antagonist on any other protein (e.g., other E3 ligase). In some embodiments, the inhibitor is IC to the target 50 IC that is an antagonist to non-target 50 Is about 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5%, 1%, 0.1%, 0.01%, 0.001% or less. The compounds disclosed herein may or may not be specific Cbl-b inhibitors. The amount of specific Cbl-b inhibitor that reduces the biological activity of Cbl-b is statistically greater than the inhibitory effect of the antagonist on any other protein (e.g., other E3 ligase). In certain embodiments, the Cbl-b inhibitor specifically inhibits ubiquitination activity of Cbl-b. In some of these embodiments, the Cbl-b inhibitor pair Cbl-b IC 50 Is an IC of a Cbl-b inhibitor to another RING finger E3 ligase or other type of E3 ligase (e.g., cullin-RING ligase) 50 Is about 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 0.1%, 0.01%, 0.001% or less.
The compounds of the invention may be tethered to a detectable moiety. It will be appreciated that such compounds may be used as imaging agents. One of ordinary skill in the art will recognize that the detectable moiety may be attached to the provided compounds by suitable substituents. As used herein, the term "suitable substituent" refers to a moiety that is capable of covalently linking to a detectable moiety. Such moieties are well known to those of ordinary skill in the art and include groups containing, for example, carboxylate moieties, amino moieties, thiol moieties, or hydroxyl moieties, to name a few. It should be understood that such portions may be direct or throughA tethered group such as a divalent saturated or unsaturated hydrocarbon chain is attached to the provided compounds. In some embodiments, such moieties may be chemically linked by clicking. In some embodiments, such moieties may be attached by 1, 3-cycloaddition of azide to alkyne, optionally in the presence of a copper catalyst. Methods using click chemistry are known in the art and include roscovtsev et al, international edition of applied chemistry (angel. Chem. Int. Ed.) 2002, 412596-99 and Sun et al, "bioconjugation chemistry," 2006,17,52-57。
as used herein, the term "detectable moiety" is used interchangeably with the term "label" and refers to any moiety that can be detected, such as a primary label and a secondary label. Such as radioactive isotopes (e.g., tritium, 32 P、 33 P、 35 S or 14 C) Primary labels, such as mass labels and fluorescent labels, are signal generating reporter groups that can be detected without further modification. The detectable moiety also comprises luminescent and phosphorescent groups.
As used herein, the term "secondary label" refers to moieties such as biotin and various protein antigens that require the presence of a second intermediate to produce a detectable signal. For biotin, the secondary intermediate may comprise a streptavidin-enzyme conjugate. For antigen labeling, the secondary intermediate may comprise an antibody-enzyme conjugate. Some fluorescent groups act as secondary labels because they transfer energy to another group during non-radiative Fluorescence Resonance Energy Transfer (FRET), and the second group produces a detected signal.
As used herein, the terms "fluorescent label," "fluorescent dye," and "fluorophore" refer to a moiety that absorbs light energy at a defined excitation wavelength and emits light energy at a different wavelength. Examples of fluorescent labels include, but are not limited to: alexa Fluor dyes (Alexa Fluor 350, alexa Fluor 488, alexa Fluor 532, alexa Fluor 546, alexa Fluor 568, alexa Fluor 594, alexa Fluor 633, alexa Fluor 660, and Alexa Fluor 680), AMCA-S, BODIPY dyes (BODIPY FL, BODIPY R6G, BODIPY TMR, BODIPY TR, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 589, BODIPY 581/591, IPY 630/650, IPY 650/665), carboxy rhodamine 6G, carboxy-X-Rhodamine (ROX), waterfall Blue (Cascade Blue), cascade Yellow 343, coumarin, cyanine dyes (Cy3, cy5, 3.5, cypo5), cypoxy TMR, BODIPY 58/591, BODIPY 581, BODIPY 581, BOPY 581 (ROX), cascade Blue, cascade 3, cascade Yellow, cas 3.5, cy2, cypo5 '; 5' -dichloro-2 ',7' -dimethoxy-fluorescein, DM-NERF, eosin, erythrosin, fluorescein, FAM, hydroxycoumarin, IRDye (IRD 40, IRD 700, IRD 800), JOE, lissamine rhodamine B (Lissamine rhodamine B), marina Blue (Marina Blue), methoxycoumarin, naphthylfluorescein, oregon green 488, oregon green 500, oregon green 514, pacific Blue, pyMPO, pyrene, rhodamine B, rhodamine 6G, rhodamine green, rhodamine Red, rhodol green, 2',4',5',7' -tetrabromosulfone-fluorescein, tetramethyl-rhodamine (TMR), carboxytetramethyl rhodamine (TAMRA), texas Red (Texas Red), texas Red-X.
As used herein, the term "mass tag" refers to any moiety that can be uniquely detected by means of mass using Mass Spectrometry (MS) detection techniques. Examples of mass labels include electrophoretic release labels such as N- [3- [4'- [ (p-methoxytetrafluorobenzyl) oxy ] phenyl ] -3-methyl glyceryl ] isopiperidinoic acid, 4' - [2,3,5, 6-tetrafluoro-4- (pentafluorophenoxy) ] methylacetophenone, and derivatives thereof. The synthesis and use of these mass tags is described in U.S. Pat. nos. 4,650,750, 4,709,016, 5,360,8191, 5,516,931, 5,602,273, 5,604,104, 5,610,020 and 5,650,270. Other examples of mass labels include, but are not limited to, nucleotides, dideoxynucleotides, oligonucleotides of varying length and base composition, oligopeptides, oligosaccharides and other synthetic polymers of varying length and monomer composition. A large number of neutral and charged organic molecules (biomolecules or synthetic compounds) of suitable mass range (100-2000 daltons) can also be used as mass labels.
As used herein, the terms "measurable affinity" and "measurably inhibit" mean a measurable change in Cbl-b ubiquitination activity between a sample comprising a compound of the present invention or a composition thereof and a Cbl-b E ligase, and an equivalent sample comprising a Cbl-b E3 ligase in the absence of the compound or composition thereof.
3. Description of exemplary embodiments:
as described above, in certain embodiments, the present invention provides a compound of formula I:
or a pharmaceutically acceptable salt thereof, wherein:
ring a is a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; or a 5-6 membered saturated or partially unsaturated heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
each R 1 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
ring B is a divalent phenyl or divalent 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
each R 2 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CH 2 OR、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
ring C is a divalent phenyl or divalent 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
each R 3 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl ring; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; and a 4-8 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur;
Or two R 3 Radicals and each R 3 The atoms to which the groups are attached optionally together form a fused 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring; fused 5-6 membered monocyclic aryl ring; a fused 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; or a fused 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
x is N, N + -O - 、NR 4 、CR 4 Or C-L-R 9
R 4 Is hydrogen, oxo,Halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
y is N, N + -O - 、NR 5 、CR 5 Or C-L-R 9
R 5 Is hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
R 6 is hydrogen or C 1-3 Aliphatic series; or (b)
R 4 And R is 6 Optionally together with the intervening atoms thereof form a 5-to 6-membered partially unsaturated fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, optionally with R 7 U instance substitutions of (2);
each R 7 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
each R 8 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2
-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
l is a covalent bond; or L is C 1-4 A divalent saturated or unsaturated linear or branched hydrocarbon chain, wherein one or two methylene units in the chain are optionally and independently replaced by: -C (R) 2 -、-C(OR)(R)-、-N(R)-、-O-、-S-、-S(O)-、-S(O) 2 -、-S(O)N(R)-、-N(R)S(O)-、-S(O) 2 N(R)-、-N(R)S(O) 2 -C (O) -, -C (O) O-, -OC (O) -, -C (O) N (R) -, -N (R) C (O) -, -C (O) N (R) O-, -ON (R) C (O) -, -OC (O) N (R) -, -N (R) C (O) O-, or-N (R) C (O) N (R) -;
R 9 is halogen, -CN, -NO 2 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-CF 2 R、-CF 3 、-C(R) 2 OR、-C(R) 2 N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-C(S)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-Si(OR)(R) 2 、-Si(R) 3 、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or R is 9 Is an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 4-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spirobicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; and 8-10 membered partially aromatic or heteroaromatic bicyclic heterocycle having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
R 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 、R 8 and R is 9 Each instance independently and optionally by R A V example substitutions of (1), wherein each R A Independently oxo, halogen, -CN, -NO 2 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(R) 2 OR、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 、-P(O)(R)OR、-P(O)(R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or an optionally substituted group selected from: c (C) 1-6 FatA family; a phenyl group; a naphthyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spiro ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; and a 6-11 membered saturated or partially unsaturated bicyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur;
each R is independently hydrogen, -CN, halogen, or an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl group; a naphthyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 7-12 membered saturated or partially unsaturated bicyclic heterocycle having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spiro ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-11 membered saturated or partially unsaturated bicyclic carbocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; or:
Two R groups on the same atom optionally together with the atoms form an optionally substituted 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur;
m is 0, 1, 2, 3, 4 or 5;
n is 0, 1, 2, 3 or 4;
p is 0, 1, 2, 3 or 4;
q is 0, 1, 2 or 3;
t is 0 or 1;
u is 0, 1, 2, 3 or 4; and is also provided with
Each instance of v is independently 0, 1, 2, 3, 4, or 5; and is also provided with
Wherein the method comprises the steps ofRepresents a single bond or a double bond.
As defined generally above, ring a is a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; or a 5-6 membered saturated or partially unsaturated heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In certain embodiments, ring a is a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In certain embodiments, ring a is a 5-6 membered saturated or partially unsaturated heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In certain embodiments, ring a is a 5 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, ring a is furyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, pyrazolyl, pyrrolyl, thiazolyl, thienyl, triazinyl, or triazolyl.
In certain embodiments, ring a is selected from
In certain embodiments, ring A and R thereof 1 Substituents taken together are
In certain embodiments, ring A and R thereof 1 Substituents taken together are
In some embodiments, ring a is selected from those depicted in table 1 below.
In some embodiments, ring A and R thereof 1 Substituents together are selected from those depicted in table 1 below.
As generally defined above, each R 1 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series.
In certain embodiments, R 1 Is halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series.
In certain embodiments, R 1 Is halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)S(O) 2 R or-N (R) S (O) R; or optionally substituted-Me, -Et, -Pr, -i-Pr, -n-Bu, -s-Bu, -t-Bu, straight or branched pentyl or straight or branched hexyl.
In certain embodiments, R 1 Is hydrogen, -Me or-OH.
In certain embodiments, R 1 Is oxo.
In some embodiments, R 1 Selected from those depicted in table 1 below.
Ring B is a divalent phenyl group or a divalent 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, as generally defined above.
In some embodiments, ring B is a divalent phenyl, furyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, pyrimidinyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, thiazolyl, thienyl, triazinyl, or triazolyl group.
In some embodiments, ring B is a divalent phenyl, pyrazolyl, imidazolyl, or pyridinyl group.
In some embodiments, ring B is a divalent group selected from:
in certain embodiments, ring B and R thereof 2 Substituents taken together are
In certain embodiments, ring B and R thereof 2 Substituents taken together are selected from
In certain embodiments, ring B and R thereof 2 Substituents taken together are
In certain embodiments, ring B and R thereof 2 Substituents taken together are
In certain embodiments, ring B and R thereof 2 Substituents taken together are(e.g.)>). In certain embodiments, ring B and R thereof 2 Substituents together are->
In certain embodiments, ring B is selected from
In certain embodiments, ring B and R thereof 2 Substituents taken together are selected from
In some embodiments, ring B is selected from those depicted in table 1 below.
In some embodiments, ring B and R thereof 2 Substituents together are selected from those depicted in table 1 below.
As generally defined above, each R 2 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series.
In certain embodiments, R 2 Is hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series.
In certain embodiments, R 2 Is halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)S(O) 2 R, -N (R) S (O) R, or optionally substituted C 1-6 Aliphatic series.
In certain embodiments, R 2 Is optionally substituted-Me, -Et, -Pr, -i-Pr, -n-Bu, -s-Bu, -t-Bu, straight or branched pentyl or straight or branched hexyl.
In certain embodiments, R 2 Is hydrogen, -Me, -Et, -CF 3 -F, -Cl, -Br, -CN or-C (O) NH 2
In certain embodiments, when ring B and R thereof 2 Substituents taken together are In which t is 1.
In certain embodiments, ring B and R thereof 2 Substituents taken together are Wherein t is 1.
In certain embodiments, when ring B and R thereof 2 Substituents taken together are In which t is 1.
In certain embodiments, ring B and R thereof 2 Substituents taken together are />Wherein t is 1.
In some embodiments, R 2 Selected from those depicted in table 1 below.
Ring C is a divalent phenyl group or a divalent 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, as generally defined above.
In some embodiments, ring C is a divalent phenyl group or a divalent 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
in some embodiments, ring C is a divalent phenyl, pyrimidinyl, pyrazinyl, pyridazinyl, pyridinyl, or pyrimidinyl group.
In certain embodiments, ring C is
In certain embodiments, ring C is
In some embodiments, ring C is selected from those depicted in table 1 below.
In certain embodiments, ring C and R thereof 3 Substituents taken together are
/>
/>
In certain embodiments, ring C and R thereof 3 Substituents taken together are
In certain embodiments, ring C and R thereof 3 Substituents taken together areIn certain embodiments, ring C and R thereof 3 Substituents together are->In certain embodiments, ring C and R thereof 3 Substituents together are->In certain embodiments, ring C and R thereof 3 Substituents together are->
In certain embodiments, ring C and R thereof 3 Substituents taken together are
In some embodiments, ring C and R thereof 3 Substituents together are selected from those depicted in table 1 below.
As generally defined above, each R 3 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl ring; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; and a 4-8 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; or two R 3 Radicals and each R 3 The atoms to which the groups are attached optionally together form a fused 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring; fused 5-6 membered monocyclic aryl ring; a fused 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; or a fused 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In certain embodiments, R 3 Is hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl ring; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; having 1-2A 4-8 membered saturated or partially unsaturated monocyclic heterocycle independently selected from heteroatoms of nitrogen, oxygen and sulfur;
in certain embodiments, R 3 Is an optionally substituted 8-10 membered partially unsaturated or heteroaromatic bicyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In certain embodiments, two R' s 3 Radicals and each R 3 The atoms to which the groups are attached together form a fused 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring; fused 5-6 membered monocyclic aryl ring; a fused 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; or a fused 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In certain embodiments, R 3 Is hydrogen, oxo, halogen, -CN, -NO 2 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 -S (NR) (O) R, -N (R) S (O) R, -N (R) CN; or optionally substituted C 1-6 Aliphatic or optionally substituted 3-7 membered saturated or partially unsaturated monocyclic carbocycles.
In certain embodiments. Each R 3 independently-H, halogen, -OR, -NR 2 、-C(O)R、-C(O)OR、-C(O)NR 2 、-C(O)N(R)OR、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)NR 2 、-N(R)S(O) 2 R or-N (R) S (O) R; or R is 3 Independently of each instance of (C) is optionally substituted 1-6 Aliphatic series; or a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring.
In certain embodiments, R 3 Is hydrogen, oxo, -Me, -Et, -Pr, -i-Pr, straight or branched butyl, straight or branched pentyl or straight or branched hexyl, -CF 3 、-F、-Cl、-Br、-OH、-OMe、-OEt、-NH 2 />
In certain embodiments, R 3 Is that
In certain embodiments, R 3 Is hydrogen, -Me, -Et, -CF 3 、-Cl、-OH、-OMe、-OEt、
In certain embodiments, R 3 Is thatIn certain embodiments, R 3 Is->In certain embodiments, R 3 Is->In certain embodiments, R 3 is-CF 3
In some embodiments, R 3 Selected from those depicted in table 1 below.
X is N, N, as defined generally above + -O - 、NR 4 、CR 4 Or C-L-R 9
In certain embodiments, X is N. In certain embodiments, X is N + -O - . In certain embodiments, X is NR 4 . In certain embodiments, X is NMe. In certain embodiments, X is CR 4 . In certain embodiments, X is CH. In some casesIn embodiments, X is C-L-R 9
In some embodiments, X is selected from those depicted in table 1 below.
R is as defined generally above 4 Is hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series.
In certain embodiments, R 4 Is hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series.
In certain embodiments, R 4 Is hydrogen.
In some embodiments, R 4 is-OH, -CF 3 、-CH 2 OH、-CH 2 NH 2 Or methyl.
In some embodiments, R 4 Is oxo.
In some embodiments, R 4 Selected from those depicted in table 1 below.
Y is N, N as defined generally above + -O - 、NR 5 、CR 5 Or C-L-R 9
In certain embodiments, Y is N. In certain embodiments, Y is N + -O - . In certain embodiments, Y is NR 5 . In certain embodiments, Y is NH. In certain embodiments, Y is NMe. In certain embodiments, Y isIn certain embodiments, Y is CR 5 . In certain embodiments, Y is CH. In certain embodiments, Y is C-L-R 9
In some embodiments, Y is selected from those depicted in table 1 below.
R is as defined generally above 5 Is hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series.
In certain embodiments, R 5 Is hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series.
In certain embodiments, R 5 Is hydrogen.
In some embodiments, R 5 is-OH, -CF 3 、-CH 2 OH、-CH 2 NH 2 Or methyl.
In some embodiments, R 5 Is oxo.
In certain embodiments, R 5 Is that
In some embodiments, R 5 Selected from those depicted in table 1 below.
R is as defined generally above 6 Is hydrogen or C 1-3 Aliphatic series; or R is 4 And R is 6 Optionally together with the intervening atoms thereof form a 5-to 6-membered partially unsaturated fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, optionally with R 7 Are substituted for the u instances of (2).
In certain embodiments, R 6 Is hydrogen or C 1-3 Aliphatic series; or R is 4 And R is 6 Optionally together with the intervening atoms thereof form a 5-to 6-membered partially unsaturated fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, optionally with R 7 Are substituted for the u instances of (2).
In certain embodiments, R 6 Is hydrogen. In certain embodiments, R 6 Is methyl. In certain embodiments, R 6 Is ethyl.
In certain embodiments, R 4 And R is 6 Forms, together with the intervening atoms thereof, a 5-to 6-membered partially unsaturated condensed ring having 0 to 2 heteroatoms independently selected from nitrogen, oxygen and sulfur, except nitrogen, wherein the condensed ring is optionally substituted with R 7 Are substituted for the u instances of (2).
In some casesIn embodiments, R 4 And R is 6 Together with the intervening atoms, form a 5-membered partially unsaturated condensed ring having 0 heteroatoms in addition to nitrogen.
In certain embodiments, R 4 And R is 6 Together with intermediate atoms thereof to form
In some embodiments, R 6 Selected from those depicted in table 1 below.
In some embodiments, R 4 And R is 6 Together with its intervening atoms, are selected from those depicted in table 1 below.
As generally defined above, each R 7 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series.
In some embodiments, R 7 Is hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 、-P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series.
In some embodiments, R 7 Is hydrogen. In some embodiments, R 7 Is methyl.
In some embodiments, R 7 Selected from those depicted in table 1 below.
As generally defined above, each R 8 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series.
In some embodiments, R 8 Is hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series.
In some embodiments, R 8 Is hydrogen.
In some embodiments, R 8 Is halogen, -OH, -OCH 3 、-NHCH 3 、-CF 3 、-CH 2 OH、-CH 2 NH 2 Or methyl.
In some embodiments, R 8 Is halogen, -OCH 3 、-NHCH 3 or-CH 2 NH 2
In some embodiments, R 8 Is halogen. In some embodiments, R 8 is-OH. In some embodiments, R 8 is-OCH 3 . In some embodiments, R 8 is-NHCH 3 . In some embodiments, R 8 is-CF 3 . In some embodiments, R 8 is-CH 2 OH. In some embodiments, R 8 is-CH 2 NH 2 . In some embodiments, R 8 Is methyl.
In some embodiments, R 8 is-C (O) R. In some embodiments, R 8 Is C (O) CH 3
In some embodiments, R 8 Is oxo.
In some embodiments, R 8 Is fluorine. In some embodiments, R 8 Is chlorine. In some embodiments, R 8 Is bromine. In some embodiments, R 8 is-CF 3
In some embodiments, R 8 Selected from those depicted in table 1 below.
L is a covalent bond, as generally defined above; or L is C 1-4 A divalent saturated or unsaturated linear or branched hydrocarbon chain, wherein one or two methylene units in the chain are optionally and independently replaced by: -C (R) 2 -、-N(R)-、-O-、-S-、-S(O)-、-S(O) 2 -、-S(O)N(R)-、-N(R)S(O)-、-S(O) 2 N(R)-、-N(R)S(O) 2 -C (O) -, -C (O) O-, -OC (O) -, -C (O) N (R) -, -N (R) C (O) -, -C (O) N (R) O-, -ON (R) C (O) -, -OC (O) N (R) -, -N (R) C (O) O-, or-N (R) C (O) N (R) -.
In certain embodiments, L is a covalent bond. In certain embodiments, L is C 1-4 A divalent saturated or unsaturated linear or branched hydrocarbon chain, wherein one or two methylene units in the chain are optionally and independently replaced by:-C(R) 2 -、-N(R)-、-O-、-S-、-S(O)-、-S(O) 2 -、-S(O)N(R)-、-N(R)S(O)-、-S(O) 2 N(R)-、-N(R)S(O) 2 -C (O) -, -C (O) O-, -OC (O) -, -C (O) N (R) -, -N (R) C (O) -, -C (O) N (R) O-, -ON (R) C (O) -, -OC (O) N (R) -, -N (R) C (O) O-, or-N (R) C (O) N (R) -.
In certain embodiments, L is-CH 2 -、-CHMe-、-CMe 2 -、-CH 2 CH 2 -。-O-、-CH 2 O-、-OCH 2 -、-CH 2 OCH 2 -、-CH 2 CH 2 OCH 2 -、-CH 2 OCH 2 CH 2 -、-OCH 2 CH 2 -、-CH 2 CH 2 O-、-CH 2 NH-、-CH 2 NHCH 2 -、-CH 2 NHCH 2 CH 2 -、-NHCH 2 -or-C (O) -.
In certain embodiments, L is-CH 2 -。
In some embodiments, L is selected from those depicted in table 1 below.
R is as defined generally above 9 Is halogen, -CN, -NO 2 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-CF 2 R、-CF 3 、-C(R) 2 OR、-C(R) 2 N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-C(S)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-Si(OR)(R) 2 、-Si(R) 3 、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or R is 9 Is an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 4-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; having 1-4 independentlyA 5-6 membered monocyclic heteroaryl ring of heteroatoms selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spirobicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; and 8-10 membered partially aromatic or heteroaromatic bicyclic heterocycle having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In certain embodiments, R 9 Is halogen, -CN, -NO 2 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-CF 2 R、-CF 3 、-C(R) 2 OR、-C(R) 2 N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-C(S)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-Si(OR)(R) 2 、-Si(R) 3 、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or R is 9 Is an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 4-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spirobicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; and 8-10 membered partially aromatic or heteroaromatic bicyclic heterocycle having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In certain embodiments, R 9 Is halogen, -CN, -NO 2 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-CF 2 R、-CF 3 、-C(R) 2 OR、-C(R) 2 N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-C(S)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-Si(OR)(R) 2 、-Si(R) 3 、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2
In certain embodiments, R 9 Is methyl, i-Pr, OCH 3 or-OH.
In certain embodiments, R 9 Is that-NHMe、/> />
/>
/>
/>
In certain embodiments, R 9 Is a fluorophore, e.gSubstituted C 1-6 Aliphatic series.
In certain embodiments, R 9 Is a fluorophore, e.gSubstituted C 1-6 Aliphatic series.
In certain embodiments, R 9 Is 2,2' - ((oxybis (b-2, 1-diyl)) bis (oxy)) bis (b-1-amine) substituted with a fluorophore
In certain embodiments, R 9 Is 2,2' - ((oxybis (b-2, 1-diyl)) bis (oxy)) bis (b-1-amine) substituted with a fluorophore
In certain embodiments, R 9 Is that or-N (R) Me.
In certain embodiments, R 9 Is thator-NHMe.
In some embodiments, R 9 Selected from those depicted in table 1 below.
In some embodiments, -L-R 9 Is that/>/>
In certain embodiments, -L-R 9 Is that/>
In certain embodiments, -L-R 9 Is that />
/>
/>
In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->
In further embodiments, -L-R 9 The R group of (2) is not hydrogen, -CN or halogen.
In certain embodiments, -L-R 9 Is that/>
In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->At a certain positionIn some embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In some embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->
In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is that
In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In some casesIn embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In some embodiments,-L-R 9 Is->In certain embodiments, -L-R 9 Is->
In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In some implementationsIn the examples, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->/>
In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In some embodiments of the present invention, in some embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is thatIn certain embodiments, -L-R 9 Is->In certain embodiments, -L-R 9 Is that
In certain embodiments, -L-R 9 Is that
In certain embodiments, -L-R 9 Is that/>
In a further embodiment, -L-R 9 The R group of (2) is not hydrogen, -CN or halogen.
In certain embodiments, -L-R 9 Is that
In certain embodiments, -L-R 9 Is that
In some embodiments, -L-R 9 Selected from those depicted in table 1 below.
In certain embodiments, when ring B is unsubstituted phenyl, -L-R 9 Selected from the group consisting of
or-L-R 9 Selected from-> />
In further embodiments, -L-R 9 The R group of (2) is not hydrogen, -CN or halogen.
R is as defined generally above 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 、R 8 And R is 9 Each instance independently and optionally by R A V example substitutions of (1), wherein each R A Independently oxo, halogen, -CN, -NO 2 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(R) 2 OR、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 、-P(O)(R)OR、-P(O)(R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl group; a naphthyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spiro ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; and 6-11 membered saturated or partially unsaturated bicyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In certain embodiments, R 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 、R 8 Or R is 9 Independently and optionally by R A V example substitutions of (1), wherein each R A Independently oxo, halogen, -CN, -NO 2 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(R) 2 OR、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 、-P(O)(R)OR、-P(O)(R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl group; a naphthyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spiro ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; and 6-11 membered saturated or partially unsaturated bicyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In some embodiments, R A Is oxo, halogen, -CN, -NO 2 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(R) 2 OR、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 、-P(O)(R)OR、-P(O)(R) 2 Or optionally substitutedFrom the following groups: c (C) 1-6 Aliphatic series; a phenyl group; a naphthyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spiro ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; and 6-11 membered saturated or partially unsaturated bicyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In some embodiments, R A Is methyl, -OH, fluorine, -CO 2 Me or-NH 2
In some embodiments, R A Selected from those depicted in table 1 below.
As generally defined above, each R is independently hydrogen, -CN, halogen, or an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl group; a naphthyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 7-12 membered saturated or partially unsaturated bicyclic heterocycle having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spiro ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-11 membered saturated or partially unsaturated bicyclic carbocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; or two R groups on the same atom optionally together with the atoms form an optionally substituted 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring; having 1-2 groups independently selected from nitrogen, oxygen and sulfur 3-7 membered saturated or partially unsaturated monocyclic heterocycle of heteroatoms of (2).
In some embodiments, R is hydrogen, -CN, halogen, or an optionally substituted group selected from the group consisting of: c (C) 1-6 Aliphatic series; a phenyl group; a naphthyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 7-12 membered saturated or partially unsaturated bicyclic heterocycle having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spiro ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-11 membered saturated or partially unsaturated bicyclic carbocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; or two R groups on the same atom optionally together with the atoms form an optionally substituted 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring; 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In some embodiments, R is hydrogen.
In some embodiments, R is —cn or halogen.
In some embodiments, R is an optionally substituted group selected from the group consisting of: c (C) 1-6 Aliphatic series; a phenyl group; a naphthyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 7-12 membered saturated or partially unsaturated bicyclic heterocycle having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; with 0-3 being independently selected from6-10 membered saturated or partially unsaturated spirocyclic rings of heteroatoms of nitrogen, oxygen and sulfur; a 6-11 membered saturated or partially unsaturated bicyclic carbocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In some embodiments, R is hydrogen. In some embodiments, R is CF 3 . In some embodiments, R is C (O) OC (CH 3 ). In some embodiments, R is optionally substituted C 1-6 Aliphatic series.
In some embodiments, two R groups on the same atom together with the atom form an optionally substituted 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring; 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In some embodiments, R is selected from those depicted in table 1 below.
As defined generally above, m is 0, 1, 2, 3, 4 or 5.
In certain embodiments, m is 0, 1, 2, 3, 4, or 5.
In some embodiments, m is 0. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments, m is 5.
In some embodiments, m is 0 or 1. In some embodiments, m is 0, 1, or 2. In some embodiments, m is 0, 1, 2, or 3. In some embodiments, m is 1 or 2. In some embodiments, m is 1, 2, or 3. In some embodiments, m is 1, 2, 3, or 4. In some embodiments, m is 2 or 3. In some embodiments, m is 2, 3, or 4. In some embodiments, m is 3 or 4. In some embodiments, m is 3, 4, or 5.
In some embodiments, m is selected from the values represented in the compounds depicted in table 1 below.
As generally defined above, n is 0, 1, 2, 3 or 4.
In certain embodiments, n is 0, 1, 2, 3, or 4.
In certain embodiments, n is 0. In certain embodiments, n is 1. In certain embodiments, n is 2. In certain embodiments, n is 3. In certain embodiments, n is 4.
In some embodiments, n is 0 or 1. In some embodiments, n is 0, 1, or 2. In some embodiments, n is 0, 1, 2, or 3. In some embodiments, n is 1 or 2. In some embodiments, n is 1, 2, or 3. In some embodiments, n is 1, 2, 3, or 4. In some embodiments, n is 2 or 3. In some embodiments, n is 2, 3, or 4. In some embodiments, n is 3 or 4.
In some embodiments, n is selected from the values represented in the compounds depicted in table 1 below.
As defined generally above, p is 0, 1, 2, 3 or 4.
In certain embodiments, p is 0, 1, 2, 3, or 4.
In certain embodiments, p is 0. In certain embodiments, p is 1. In certain embodiments, p is 2. In certain embodiments, p is 3. In certain embodiments, p is 4.
In some embodiments, p is 0 or 1. In some embodiments, p is 0, 1, or 2. In some embodiments, p is 0, 1, 2, or 3. In some embodiments, p is 0, 1, 2, 3, or 4. In some embodiments, p is 1 or 2. In some embodiments, p is 1, 2, or 3. In some embodiments, p is 1, 2, 3, or 4. In some embodiments, p is 2 or 3. In some embodiments, p is 2, 3, or 4. In some embodiments, p is 3 or 4.
In some embodiments, p is selected from the values represented in the compounds depicted in table 1 below.
Q is 0, 1, 2 or 3, as generally defined above.
In certain embodiments, q is 0, 1, 2, or 3.
In certain embodiments, q is 0. In certain embodiments, q is 1. In certain embodiments, q is 2. In certain embodiments, q is 3.
In certain embodiments, q is 0, 1, or 2. In certain embodiments, q is 1, 2, or 3. In certain embodiments, q is 1 or 2. In certain embodiments, q is 2 or 3.
In some embodiments, q is selected from the values represented in the compounds depicted in table 1 below.
As defined generally above, t is 0 or 1.
In certain embodiments, t is 0 or 1.
In certain embodiments, t is 0. In certain embodiments, t is 1.
In some embodiments, t is selected from those depicted in table 1 below.
U is 0, 1, 2, 3 or 4, as generally defined above.
In some embodiments, u is 0, 1, 2, 3, or 4.
In some embodiments, u is 0. In some embodiments, u is 1. In some embodiments, u is 2. In some embodiments, u is 3. In some embodiments, u is 4.
In some embodiments, u is selected from those depicted in table 1 below.
Each instance of v is independently 0, 1, 2, 3, 4, or 5, as generally defined above.
In some embodiments, v is 0, 1, 2, 3, 4, or 5.
In some embodiments, v is 0. In some embodiments, v is 1. In some embodiments, v is 2. In some embodiments, v is 3. In some embodiments, v is 4. In some embodiments, v is 5.
In some embodiments, v is 0 or 1. In some embodiments, v is 0, 1, or 2. In some embodiments, v is 0, 1, 2, or 3. In some embodiments, v is 1 or 2. In some embodiments, v is 1, 2, or 3. In some embodiments, v is 1, 2, 3, or 4. In some embodiments, v is 2 or 3. In some embodiments, v is 2, 3, or 4. In some embodiments, v is 3 or 4. In some embodiments, v is 3, 4, or 5.
In some embodiments, v is selected from those depicted in table 1 below.
As generally defined above,represents a single bond or a double bond.
In some embodiments of the present invention, in some embodiments,representing a single bond. In some embodiments, ->Representing a double bond.
In some embodiments of the present invention, in some embodiments,selected from those depicted in table 1 below.
In some embodiments, the present invention provides compounds of formulas II-a through II-zz:
/>
/>
/>
/>
/>
OR a pharmaceutically acceptable salt thereof, wherein-OR, -NHR, -NR-OR-N (R) 2 R groups of (2) are not hydrogen, -CN or halogen, and wherein X, Y, ring A, ring B, ring C, R 1 、R 2 、R 3 、R 6 、R 8 Each of m, n, p, and q, singly or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the present invention provides compounds of formulas II-a through II-zz:
/>
/>
/>
/>
/>
/>
or a pharmaceutically acceptable salt thereof, wherein X, Y, ring A, ring B, ring C, R 1 、R 2 、R 3 、R 6 、R 8 Each of m, n, p, and q, singly or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the invention provides compounds of formula II-k, II-l, II-m, II-n, II-o, II-p, II-q, II-R, II-s, II-t, II-u, II-v, II-y, II-z, II-cc, II-dd, II-ee, II-ff, II-gg, II-hh, II-nn, II-oo, II-pp, II-qq, II-rr, II-ss, II-uu, II-v, II-xx, II-yy, II-zz, II-aaa, II-bbb, II-ccc, II-ddd or II-eee, wherein R is hydrogen, thereby forming the formulae II-k-a, II-l-a, II-m-a, II-n-a, II-o-a, II-p-a, II-q-a, II-R-a, II-s-a, II-t-a, II-u-a, II-v-a, II-y-a, II-z-a, II-cc-a, II-dd-a, II-ee-a, II-ff-a, II-gg-a, II-hh-a, II-nn-a, II-oo-a, II-pp-a, II-qq-a, II-rr-a, II-ss-a, II-uu-a, II-vv-a, II-xx-a, II-yy-a, II-zz-a, II-aaa-a, II-bbb-a, II-ccc-a, II-ddd-a or II-eee-a compounds:
/>
/>
/>
/>
In some embodiments, the present invention provides compounds of formula II-nn, II-qq, or II-xx, wherein R is hydrogen, thereby forming compounds of formula II-nn-a, II-qq-a, or II-xx-a, respectively:
or a pharmaceutically acceptable salt thereof, wherein ring A, ring B, ring C, R 1 、R 2 、R 3 、R 8 Each of m, n, p, and q, singly or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the invention provides compounds of formula II-ee, II-ff or II-yy, wherein R is CH 3 Whereby a compound of the formula II-ee-b, II-ff-b or II-yy-b, respectively, is formed:
or a pharmaceutically acceptable salt thereof, wherein ring A, ring B, ring C, R 1 、R 2 、R 3 、R 8 Each of m, n, p, and q, singly or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the invention provides compounds of formula II-ee, II-ff or II-yy, wherein R is C (O) OC (CH 3 ) Whereby a compound of formula II-ee-c, II-ff-c or II-yy-c, respectively, is formed:
/>
or a pharmaceutically acceptable salt thereof, wherein ring A, ring B, ring C, R 1 、R 2 、R 3 、R 8 Each of m, n, p, and q, singly or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the present invention provides compounds of formula II-gg, II-hh, or II-zz, wherein R is cyclopropyl, thereby forming compounds of formula II-gg-b, II-hh-b, or II-zz-b, respectively:
Or a pharmaceutically acceptable salt thereof, wherein R is cyclopropyl, and wherein ring a, ring B, ring C, R 1 、R 2 、R 3 、R 8 Each of m, n, p, and q, singly or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the invention provides compounds of formulae II '-a to II' -ww:
/>
/>
/>
/>
/>
or a pharmaceutically acceptable salt thereof, wherein X, Y, ring A, ring B, ring C, R 1 、R 2 、R 3 、R 6 、R 8 Each of m, n, p, and q, singly or in combination, is as defined above and described in the embodiments herein. In some embodiments of the embodiments, R in OR is not hydrogen. In some embodiments of the embodiments, R in OR is not-CN ORHalogen.
In some embodiments, the invention provides compounds of formula II '-a, II' -b, II '-c, II' -d, II '-gg, or II' -hh, wherein R is hydrogen, thereby forming compounds of formula II "-a, II" -b, II "-c, II" -d, II "-gg, or II" -hh, respectively:
/>
or a pharmaceutically acceptable salt thereof, wherein X, Y, ring A, ring B, ring C, R 1 、R 2 、R 3 、R 6 、R 8 Each of m, n, p, and q, singly or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the invention provides compounds of formula II ' -g, II ' -h or II ' -jj wherein R is methyl, trifluoromethyl or cyclopropyl, thus, the compounds of the formula II ' -g-a, II ' -h-a, II ' -g-b, II ' -h-b II "-g-c, II" -h-c, II "-jj-a, II" -jj-b or II "-jj-c compounds:
/>
Or a pharmaceutically acceptable salt thereof, wherein X, Y, ring A, ring B, ring C, R 1 、R 2 、R 3 、R 6 、R 8 Each of m, n, p, and q, singly or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the invention provides compounds of formulae II '-ww to II' -zz, II '-aaa, and II' -bbb:
/>
or a pharmaceutically acceptable salt thereof, wherein X, Y, ring A, ring B, ring C, R 1 、R 2 、R 3 、R 6 、R 8 Each of m, n, p, and q, singly or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the present invention provides compounds of formula III-a, III-b, or III-c:
or a pharmaceutically acceptable salt thereof, wherein X, Y, ring A, ring C, L, R 1 、R 3 、R 6 、R 8 、R 9 Each of m, p, q, and t, singly or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the invention provides compounds of formula III-d, III-e, III-f, III-g, III-h, or III-i:
/>
or a pharmaceutically acceptable salt thereof, wherein X, Y, ring A, ring C, L,R 1 、R 3 、R 6 、R 8 、R 9 Each of m, p, q, and t, singly or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the present invention provides compounds of formula III-j, III-k, III-l, III-m, or III-n.
/>
Or a pharmaceutically acceptable salt thereof, wherein X, Y, ring A, ring C, R 1 、R 3 、R 6 、R 8 Each of m, p and q, and t, individually or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the present invention provides compounds of formula IV-a, IV-b, IV-c, IV-d, IV-e, or IV-f:
or a pharmaceutically acceptable salt thereof, wherein X, Y, ring A, L, R 1 、R 3 、R 6 、R 8 、R 9 Each of m, p, q, and t, singly or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the present invention provides compounds of formulas IV-g through IV-r:
/>
or a pharmaceutically acceptable salt thereof, wherein ring A, L, R 1 、R 3 、R 6 、R 8 、R 9 Each of m, p, q, and t, singly or in combination, is as defined above and described in the embodiments herein.
In some embodiments, the present invention provides compounds of formula IV-s, IV-t, IV-u, IV-v or IV-w.
/>
Or a pharmaceutically acceptable salt thereof, wherein X, Y, ring A, R 1 、R 3 、R 6 、R 8 Each of m, p and q, and t, individually or in combination, is as defined above and described in the embodiments herein.
In certain aspects, the invention provides compounds of formula V:
or a pharmaceutically acceptable salt thereof, wherein:
ring a is a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; or a 5-6 membered saturated or partially unsaturated heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
Each R 1 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
R 10 、R 11 、R 12 and R is 13 Each independently is R 3 The method comprises the steps of carrying out a first treatment on the surface of the Or R is 10 、R 11 、R 12 And R is 13 Together with the atoms to which each is attached, form a phenyl ring; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; or a 4-8 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; or R is as valence bond requires 11 And R is 13 Absence of;
ring C is a divalent phenyl or divalent 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
each R 3 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl ring; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; and a 4-8 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur;
or two R 3 Radicals and each R 3 The atoms to which the groups are attached optionally together form a fused 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring; fused 5-6 membered monocyclic aryl ring; a fused 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; or a fused 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
x is N, N + -O - 、NR 4 、CR 4 Or C-L-R 9
R 4 Is hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
y is N, N + -O - 、NR 5 、CR 5 Or C-L-R 9
R 5 Is hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
R 6 is hydrogen or C 1-3 Aliphatic series; or (b)
R 4 And R is 6 Optionally together with the intervening atoms thereof form a 5-to 6-membered partially unsaturated fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, optionally with R 7 U instance substitutions of (2);
each R 7 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
each R 8 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
l is a covalent bond; or L is C 1-4 A divalent saturated or unsaturated linear or branched hydrocarbon chain, wherein one or two methylene units in the chain are optionally and independently replaced by: -C (R) 2 -、-C(OR)(R)-、-N(R)-、-O-、-S-、-S(O)-、-S(O) 2 -、-S(O)N(R)-、-N(R)S(O)-、-S(O) 2 N(R)-、-N(R)S(O) 2 -C (O) -, -C (O) O-, -OC (O) -, -C (O) N (R) -, -N (R) C (O) -, -C (O) N (R) O-, -ON (R) C (O) -, -OC (O) N (R) -, -N (R) C (O) O-, or-N (R) C (O) N (R) -;
R 9 is halogen, -CN, -NO 2 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-CF 2 R、-CF 3 、-C(R) 2 OR、-C(R) 2 N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-C(S)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-Si(OR)(R) 2 、-Si(R) 3 、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or R is 9 Is an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 4-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spirobicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; and 8-10 membered partially aromatic or heteroaromatic bicyclic heterocycle having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
R 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 、R 8 and R is 9 Each instance independently and optionally by R A V example substitutions of (1), wherein each R A Independently oxo, halogen, -CN, -NO 2 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(R) 2 OR、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 、-P(O)(R)OR、-P(O)(R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl group; a naphthyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spiro ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; and a 6-11 membered saturated or partially unsaturated bicyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur;
each R is independently hydrogen, -CN, halogen, or an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl group; a naphthyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 7-12 membered saturated or partially unsaturated bicyclic heterocycle having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spiro ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; 6-11 membered saturated or unsaturated moieties having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur A partially unsaturated bicyclic carbocyclic ring; or:
two R groups on the same atom optionally together with the atoms form an optionally substituted 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur;
m is 0, 1, 2, 3, 4 or 5;
p is 0, 1, 2, 3 or 4;
q is 0, 1, 2 or 3;
t is 0 or 1;
u is 0, 1, 2, 3 or 4;
each instance of v is independently 0, 1, 2, 3, 4, or 5; and is also provided with
Wherein the method comprises the steps ofRepresents a single bond or a double bond.
Exemplary compounds of the present invention are listed in table 1 below.
TABLE 1 selected Compounds
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
In some embodiments, the invention provides a compound listed in table 1 above, or a pharmaceutically acceptable salt thereof. In some embodiments, the invention provides the compounds listed in table 1 above.
In some embodiments, the present invention provides a pharmaceutical composition comprising a compound disclosed herein (described in the embodiments herein, alone or in combination) or a pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable carrier, excipient, or diluent. For example, in some embodiments, the present invention provides a pharmaceutical composition comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, as defined above, together with a pharmaceutically acceptable carrier, excipient or diluent. In some embodiments, the present invention provides a pharmaceutical composition comprising a compound of formula I as defined above together with a pharmaceutically acceptable carrier, excipient or diluent. In some embodiments, the present invention provides a pharmaceutical composition comprising a compound listed in table 1 above, or a pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable carrier, excipient, or diluent. In some embodiments, the present invention provides a pharmaceutical composition comprising a compound listed in table 1 above in combination with a pharmaceutically acceptable carrier, excipient, or diluent.
Exemplary compounds of the present invention are listed in table 2 below.
TABLE 2 selected Compounds
/>
In some embodiments, the invention provides a compound of formula I, or a pharmaceutically acceptable salt thereof, as defined above, or a pharmaceutical composition comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, as defined above, in combination with a pharmaceutically acceptable carrier, adjuvant, or vehicle, for use as a medicament.
In some embodiments, the invention also provides a compound described herein (e.g., a compound of formula I) or a pharmaceutical composition described herein for use in a method for inhibiting Cbl-b as described herein and/or for use in a method of treating a Cbl-b-dependent disorder as described herein. In some embodiments, the invention also provides a compound described herein (e.g., a compound of formula I) or a pharmaceutical composition described herein for use in a method for inhibiting Cbl-b as described herein. In some embodiments, the invention also provides a compound described herein (e.g., a compound of formula I) or a pharmaceutical composition described herein for use in a method for treating a Cbl-b dependent disorder as described herein.
In some embodiments, the invention also provides a compound described herein (e.g., a compound of formula I) or a pharmaceutical composition described herein for use in a method for modulating Cbl-b as described herein and/or for use in a method for treating a Cbl-b dependent disorder as described herein. In some embodiments, the invention also provides a compound described herein (e.g., a compound of formula I) or a pharmaceutical composition described herein for use in a method for modulating Cbl-b as described herein. In some embodiments, the invention also provides a compound described herein (e.g., a compound of formula I) or a pharmaceutical composition described herein for use in a method for treating a Cbl-b dependent disorder as described herein.
4. General methods for providing the Compounds of the invention
The compounds of the present invention may generally be prepared or isolated by synthetic and/or semisynthetic methods known to those of skill in the art for similar compounds and by methods described in detail in the examples herein.
5. Use, formulation and administration
Pharmaceutically acceptable compositions
According to another embodiment, the present invention provides a composition comprising a compound of the present invention or a pharmaceutically acceptable derivative thereof, and a pharmaceutically acceptable carrier, adjuvant or vehicle. In certain embodiments, the amount of the compound in the compositions of the invention is such that Cbl-b, or a mutant thereof, of the biological sample or patient is effectively measurably inhibited. In certain embodiments, the compositions of the present invention are formulated for administration to a patient in need of such compositions. In some embodiments, the compositions of the present invention are formulated for oral administration to a patient.
As used herein, the term "patient" means an animal, preferably a mammal and most preferably a human.
The term "pharmaceutically acceptable carrier, adjuvant or vehicle" refers to a non-toxic carrier, adjuvant or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that can be used in the compositions of the invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (such as human serum albumin), buffer substances (such as phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts), colloidal silica, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and lanolin.
By "pharmaceutically acceptable derivative" is meant any non-toxic salt, ester salt or other derivative of a compound of the invention that is capable of providing, directly or indirectly, a compound of the invention or an active metabolite or residue thereof after administration to a recipient.
As used herein, the term "active metabolite or residue thereof" means that the metabolite or residue thereof is also an inhibitor of Cbl-b or a mutant thereof.
The subject matter disclosed herein includes prodrugs, metabolites, derivatives, and pharmaceutically acceptable salts of the compounds of the invention. Metabolites include compounds produced by a method comprising contacting a compound of the invention with a mammal for a period of time sufficient to produce a metabolite thereof. If the compounds of the invention are bases, the desired pharmaceutically acceptable salts may be prepared by any suitable method available in the art, for example, treating the free base with the following acids: inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, methanesulfonic acid, phosphoric acid, and the like; or organic acids such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, and the like; pyranoid acids such as glucuronic acid and galacturonic acid; alpha-hydroxy acids such as citric acid or tartaric acid; amino acids such as aspartic acid or glutamic acid; aromatic acids such as benzoic acid or cinnamic acid; such as p-toluenesulfonic acid, ethanesulfonic acid and the like. If the compounds of the present invention are acids, the desired pharmaceutically acceptable salts may be prepared by any suitable method, for example, treating the free acid with an inorganic or organic base such as an amine (primary, secondary or tertiary), an alkali metal hydroxide or alkaline earth metal hydroxide, or the like. Illustrative examples of suitable salts include, but are not limited to, amino acids derived from, for example, glycine and arginine; ammonia, primary, secondary and tertiary amines; organic salts of cyclic amines such as piperidine, morpholine or piperazine; and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
The compounds of the invention may be in the form of "prodrugs" which comprise compounds having moieties that can be metabolized in vivo. In general, prodrugs are metabolized in vivo to the active drug by esterases or by other mechanisms. Examples of prodrugs and their uses are well known in the art (see, e.g., berge et al, (1977), "pharmaceutically acceptable salts (Pharmaceutical Salts)", "journal of pharmaceutical science," 66:1-19). Prodrugs can be prepared in situ during the final isolation and purification of the compound, or by separately reacting the purified compound in its free acid form or hydroxy group with a suitable esterifying agent. The hydroxyl groups can be converted to esters by treatment with carboxylic acids. Examples of prodrug moieties include substituted and unsubstituted, branched or unbranched lower alkyl ester moieties (e.g., propionate), lower alkenyl esters, di-lower alkyl-amino lower alkyl esters (e.g., dimethylaminoethyl esters), amido lower alkyl esters (e.g., acetoxymethyl esters), acyloxy lower alkyl esters (e.g., pivaloyloxymethyl esters), aryl esters (phenyl esters), aryl-lower alkyl esters (e.g., benzyl esters), substituted (e.g., with methyl, halo or methoxy substituents) aryl and aryl-lower alkyl esters, amides, lower alkylamides, di-lower alkylamides, and hydroxyamides. Prodrugs that are converted to the active form by other in vivo mechanisms are also included. In various aspects, the compounds of the invention are prodrugs of any formula herein.
The compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, bucally, vaginally, or by an implantable reservoir. As used herein, the term "parenteral" encompasses subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the composition is administered orally, intraperitoneally, or intravenously. The sterile injectable form of the compositions of the invention may be an aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a parenterally-acceptable, nontoxic diluent or solvent, for example, as a solution in 1, 3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents commonly used in the formulation of pharmaceutically acceptable dosage forms, including emulsions and suspensions. Other commonly used surfactants such as Tween (Tween), span (Span) and other emulsifying agents or bioavailability enhancers commonly used in the preparation of pharmaceutically acceptable solid, liquid or other dosage forms may also be used for formulation purposes.
The injectable formulations may be sterilized, for example, by filtration through bacterial-retaining filters or by incorporating sterilizing agents in the form of sterile solid compositions which may be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
In order to prolong the effect of the compounds of the invention, it is generally desirable to slow the absorption of the compounds from subcutaneous or intramuscular injection. This can be achieved by using a liquid suspension of a poorly water-soluble crystalline or amorphous material. Thus, the rate of absorption of a compound depends on its rate of dissolution, which in turn may depend on crystal size and form. Alternatively, delayed absorption of the parenterally administered compound form is achieved by dissolving or suspending the compound in an oily vehicle. Injectable depot forms are prepared by forming a microencapsulated matrix of the compound in a biodegradable polymer such as polylactide-polyglycolide. Depending on the ratio of compound to polymer and the nature of the particular polymer employed, the rate of release of the compound may be controlled. Examples of other biodegradable polymers include poly (orthoesters) and poly (anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with human tissue.
Alternatively, for rectal or vaginal administration, the pharmaceutically acceptable compositions of the invention may be administered in the form of suppositories. The suppositories may be prepared by mixing the agent with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.
The pharmaceutically acceptable compositions of the present invention may also be administered topically, especially where the therapeutic target comprises a region or organ that is readily accessible by topical application, diseases of the eye, skin or lower intestinal tract. Suitable topical formulations for each of these regions or organs are easy to prepare.
Topical application to the lower intestinal tract may be achieved in the form of a rectal suppository formulation (see above) or a suitable enema formulation. Topical transdermal patches may also be used.
For topical application, the provided pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active ingredient suspended or dissolved in one or more carriers. Carriers for topical application of the compounds of the invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compounds, emulsifying wax and water. Alternatively, the provided pharmaceutically acceptable compositions may be formulated in a suitable lotion or cream containing the active component suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetostearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
Dosage forms for topical or transdermal administration of the compounds of the invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active ingredient is admixed under sterile conditions with a pharmaceutically acceptable carrier and any required preservatives or buffers as may be required. Ophthalmic formulations, ear drops and eye drops are also contemplated as falling within the scope of the present invention. In addition, the present invention contemplates the use of transdermal patches that have the additional advantage of allowing the compound to be delivered to the body in a controlled manner. Such dosage forms may be prepared by dissolving or dispersing the compound in an appropriate medium. Absorption enhancers may also be used to increase the flux of the compound across the skin. The rate may be controlled by providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.
For ophthalmic use, the provided pharmaceutically acceptable compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or preferably as solutions in isotonic, pH adjusted sterile saline (with or without preservatives such as benzalkonium chloride). Alternatively, for ophthalmic use, the pharmaceutically acceptable compositions may be formulated in ointments such as petrolatum.
The pharmaceutically acceptable compositions of the present invention may also be administered by nasal aerosol or by inhalation. Such compositions are prepared according to techniques well known in the art of pharmaceutical formulation and may be prepared as solutions in saline using benzyl alcohol or other suitable preservatives, absorption promoters for enhanced bioavailability, fluorocarbons and/or other conventional solubilizing or dispersing agents.
More preferably, the pharmaceutically acceptable compositions of the present invention are formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, the pharmaceutically acceptable compositions of the present invention are not administered with food. In other embodiments, the pharmaceutically acceptable compositions of the invention are administered with food.
The pharmaceutically acceptable compositions of the present invention may be administered orally in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, the usual carriers include lactose and corn starch. Lubricants such as magnesium stearate are also typically added. For oral administration in capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. Certain sweeteners, flavoring agents or coloring agents may also be added if desired.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In such solid dosage forms, the active compound is admixed with: at least one inert pharmaceutically acceptable excipient or carrier, such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and silicic acid; b) Binders, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and acacia; c) Humectants, such as glycerol; d) Disintegrants, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; e) Solution retarders, such as paraffin; f) Absorption promoters, such as quaternary ammonium compounds; g) Humectants, for example cetyl alcohol and glycerol monostearate; h) Adsorbents such as kaolin and bentonite; and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate and mixtures thereof. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid compositions of a similar type may also be used as fillers in soft-filled gelatin capsules using excipients such as lactose or milk sugar, high molecular weight polyethylene glycols and the like, as well as in hard-filled gelatin capsules. Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical compounding arts. The dosage form may optionally contain an opacifying agent and may also be of a composition such that the dosage form releases the active ingredient only or preferentially, optionally in a delayed manner, in a particular portion of the intestinal tract. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be used as fillers in soft-filled gelatin capsules using excipients such as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like, and in hard-filled gelatin capsules.
The active compound may also be in microencapsulated form together with one or more excipients as described above. Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings, release control coatings and other coatings well known in the pharmaceutical compounding arts. In such solid dosage forms, the active compound may be admixed with at least one inert diluent (such as sucrose, lactose or starch). In addition to inert diluents, such dosage forms may normally include additional substances such as tabletting lubricants and other tabletting aids, such as magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. The dosage form may optionally contain an opacifying agent and may also be of a composition such that the dosage form releases the active ingredient only or preferentially, optionally in a delayed manner, in a particular portion of the intestinal tract. Examples of embedding compositions that can be used include polymeric substances and waxes.
Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compound, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. In addition to inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
The amount of a compound of the present invention that can be combined with a carrier material to produce a single dosage form of the composition will vary depending upon the subject being treated, the particular mode of administration. Preferably, the compositions provided should be formulated such that a dose of between 0.01 and 100mg/kg, 0.01 and 50mg/kg or 1 and 25mg/kg, body weight/day of the compound can be administered to a patient receiving these compositions.
It will also be appreciated that the specific dosage and treatment regimen for any particular patient will depend upon a variety of factors including the activity of the particular compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination and the judgment of the treating physician and the severity of the particular disease undergoing therapy. The amount of the compound of the invention in the composition will also depend on the particular compound in the composition.
The compounds of the present invention are preferably formulated in dosage unit form for ease of administration and dose uniformity. As used herein, the expression "dosage unit form" refers to physically discrete units of medicament suitable for the patient to be treated. However, it will be appreciated that the total daily amount of the compounds and compositions of the invention will be at the discretion of the attendant physician within the scope of sound medical judgment.
Use of compounds and pharmaceutically acceptable compositions
The compounds and compositions described herein are generally useful for inhibiting the E3 ligase activity of one or more enzymes. In some embodiments, the E3 ligase inhibited by the compounds and methods of the invention is Cbl-b.
The compounds disclosed herein are useful for inhibiting the enzyme Cbl-b. In one embodiment, the presently disclosed subject matter relates to a method of inhibiting Cbl-b comprising contacting Cbl-b with an effective amount of a compound of the present invention or a pharmaceutical composition described herein.
The disclosed compounds may be used in a method for inhibiting Cbl-b. Such methods comprise contacting Cbl-b with an effective amount of a compound disclosed herein. By "contacting" is meant bringing the compound into sufficient proximity with the isolated Cbl-b enzyme or Cbl-b expressing cell such that the compound is able to bind to Cbl-b and inhibit said Cbl-b. The compound may be contacted with Cbl-b in vitro or in vivo by administering the compound to a subject.
In one aspect, provided herein is a method of inhibiting Cbl-b in a biological sample. The method comprises contacting the sample with a compound disclosed herein (e.g., a compound of formula I), a pharmaceutically acceptable salt thereof, or a pharmaceutical composition disclosed herein (e.g., a composition comprising a compound disclosed herein [ e.g., a compound of formula I ]) and a pharmaceutically acceptable carrier, adjuvant, or vehicle. As used herein, the term "biological sample" includes, but is not limited to: a cell culture or extract thereof; a biopsy material obtained from a mammal or an extract thereof; and blood, saliva, urine, stool, semen, tears, or other bodily fluids or extracts thereof.
The present disclosure provides methods of inhibiting Cbl-b in a patient. The methods comprise administering to a patient a compound disclosed herein (e.g., a compound of formula I), a pharmaceutically acceptable salt thereof, or a pharmaceutical composition disclosed herein (e.g., a composition comprising a compound disclosed herein [ e.g., a compound of formula I ]) and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
The compounds disclosed herein may or may not be selective Cbl-b inhibitors. The amount of selective Cbl-b inhibitor that inhibits the biological activity of Cbl-b is statistically greater than the amount of inhibitor that inhibits any other protein (e.g., other E3 ligase). In some of these embodiments, the Cbl-b inhibitor pair Cbl-b IC 50 IC being a Cbl-b inhibitor against another E3 ligase 50 Is about 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 0.1%, 0.01%, 0.001% or less.
Any method known in the art for measuring the ligase activity of Cbl-b may be used to determine whether Cbl-b is inhibited, including in vitro kinase assays, immunoblots with antibodies specific for ubiquitinated targets of Cbl-b, or measurement of downstream biological effects of Cbl-b ligase activity.
The compounds disclosed herein may be used to treat Cbl-b dependent disorders. As used herein, a "Cbl-b dependent disorder" is a pathological condition in which Cbl-b activity is essential for the occurrence or maintenance of the pathological condition.
Accordingly, in one aspect, provided herein is a method of treating a Cbl-b mediated disorder, disease, or condition in a patient. The method comprises administering to the patient a compound disclosed herein (e.g., a compound of formula I), a pharmaceutically acceptable salt thereof, or a pharmaceutical composition disclosed herein (e.g., a composition comprising a compound disclosed herein [ e.g., a compound of formula I ]) and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
Provided herein are compounds and pharmaceutical compositions that inhibit Cbl-b enzyme, and methods of treatment using such compounds and pharmaceutical compositions. The compounds and compositions may be used in methods for modulating the immune system, for treating diseases, and for treating cells in vivo, in vitro, or ex vivo.
T cell activation and T cell tolerance are strictly controlled processes that regulate the immune response to tumors while preventing autoimmunity. Tolerance prevents the immune system from attacking cells expressing "self" antigens. During peripheral tolerance, T cells that recognize "self" antigens (i.e., autoreactive T cells) become functionally unresponsive or absent upon encountering "self" antigens outside the thymus. Thus, the peripheral tolerability process is important for the prevention of autoimmune diseases. Typically, cancer cells are removed by activated T cells that recognize tumor antigens expressed on the surface of the cancer cells. However, in cancer, the tumor microenvironment may support tolerance of T cells to cancer cells, which allows the cancer cells to avoid recognition and removal by the immune system. The ability of cancer cells to avoid tumor immune surveillance may contribute to uncontrolled tumor growth. Thus, T cell tolerance may be a form of T cell dysfunction. The general principles of T cell dysfunction are well known in the art (see Schietingger et al, trends immunology), 35:51-60,2014. Additional types of T cell dysfunction that may lead to uncontrolled tumor growth include T cell depletion, T cell senescence, and/or T cell failure. Thus, treatment of T cell dysfunction, for example by increasing T cell activation, increasing T cell proliferation, decreasing T cell tolerance, and/or decreasing T cell depletion, is beneficial for preventing or treating cancer. Additional cells of the immune system are important for the recognition and removal of cancer cells during immune surveillance. For example, natural Killer (NK) cells are lymphocytes of the innate immune system that are capable of identifying and killing Cancer cells (see Martinez-Losato et al, clinical Cancer research (Clin Cancer Res.), 21:5048-5056,2015). Recent studies have also shown that B Cell subsets with different phenotypes and functions exhibit different effects in anti-tumor responses (see Sarovaria et al, cell Mol immunol.), 14:662-674,2017. NK cells and B cells can also be therapeutic targets for preventing or treating cancer due to their role in tumor monitoring.
Cbl-b is a RING type E3 ligase that plays an important role in the immune system as it acts as a negative regulator of immune activation. Cbl-b plays an important role in reducing T cell activation, thereby enhancing T cell tolerance. Studies have found that Cbl-b deficient T cells exhibit a lower threshold for activation of antigen recognizing receptors and costimulatory molecules (e.g., CD 28). For example, the loss of Cbl-b in T cells negates the need for CD28 co-stimulation during T cell activation and proliferation (see Bachmailer et al, nature, 403:211-216,2000). Such cbl-b-/-T cells are largely non-resistant to T cells, a tolerating mechanism in which T cells are functionally inactivated and T cell proliferation is severely impaired (see Jeon et al, immunology, 21:167-177,2004; and Schwartz et al, immunology annual review (Annu Rev immunol.), 21:305-34,2003). To support this, loss of Cbl-b in Cbl-b knockout mice resulted in impaired induction of T cell tolerance and increased autoimmunity (see Jeon et al, immunology, 21:167-177,2004). Importantly, the loss of Cbl-b in mice also resulted in a robust anti-tumor response that was largely dependent on cytotoxic T cells. One study showed that cbl-b-/-cd8+ T cells were resistant to T regulatory cell mediated inhibition and exhibited enhanced activation and tumor infiltration. Untreated cbl-b-/-CD8+ T cells are sufficient to mediate rejection of established tumors (see Loeser et al J Exp Med.) (204:879-891,2007). Recent studies have shown that Cbl-b also plays a role in NK cell activation. The genetic deletion of Cbl-b or targeted inactivation of its E3 ligase activity allows NK cells to spontaneously reject metastatic tumors in a mouse model (see Paolino et al Nature 507:508-512,2014).
Provided herein are compounds and compositions that are potent inhibitors of Cbl-b and can be used in novel methods for treating diseases such as cancer. In some embodiments, the compounds and compositions provided herein can be used in methods of modulating the immune system, such as increasing activation of T cells, NK cells, and B cells, as well as for treating such cells in vivo, in vitro, or ex vivo.
Provided herein are methods for modulating the activity of immune cells (e.g., T cells, B cells, or NK cells) as by contacting the immune cells with an effective amount of a Cbl-B inhibitor or a composition thereof described herein. Further provided are methods of modulating a response in vivo in an individual in need thereof (e.g., an individual having cancer), wherein the method comprises administering an effective amount of a Cbl-b inhibitor described herein or a composition thereof.
Additionally, cbl-b inhibitors are provided for use as therapeutically active substances. Cbl-b inhibitors are provided for use in the treatment or prevention of diseases or conditions associated with Cbl-b activity. Likewise, cbl-b inhibitors for use in the treatment of cancer are provided. Further provided is the use of a Cbl-b inhibitor in the manufacture of a medicament for the treatment or prophylaxis of a disease or condition associated with Cbl-b activity. Also provided is the use of a Cbl-b inhibitor in the manufacture of a medicament for the treatment of cancer.
Furthermore, the present disclosure provides therapeutic methods, medicaments and uses comprising Cbl-b inhibitors as part of a combination therapy for treating cancers involving one or more of immune checkpoint inhibitors, anti-tumor agents and radiation therapy.
In some embodiments of the methods of treatment, medicaments and uses of the present disclosure, the cancer is a hematologic cancer, such as lymphoma, leukemia or myeloma. In other embodiments of the methods of treatment, medicaments and uses of the present disclosure, the cancer is a non-hematologic cancer, such as a sarcoma, cancer or melanoma.
Hematological cancers include, but are not limited to, one or more leukemias, such as B-cell acute lymphoblastic leukemia ("BALL"), T-cell acute lymphoblastic leukemia ("tal"), acute Lymphoblastic Leukemia (ALL); one or more chronic leukemias include, but are not limited to, chronic Myelogenous Leukemia (CML) and Chronic Lymphocytic Leukemia (CLL); additional hematologic cancers or hematological conditions include, but are not limited to, B cell prolymphocytic leukemia, blast plasmacytoid dendritic cell neoplasm, burkitt's lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small or large cell follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma, marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-hodgkin's lymphoma, plasmablastoid lymphoma, plasmacytoid dendritic cell neoplasm, waldenstrom macroglobulinemia, and "pre-leukemia," which are a diverse collection of blood conditions that are combined by ineffective production (or dysplasia) of myeloid blood cells.
Non-hematologic cancers include, but are not limited to, neuroblastoma, renal cell carcinoma, colon cancer, colorectal cancer, breast cancer, epithelial squamous cell carcinoma, melanoma, gastric cancer, brain cancer, lung cancer (e.g., NSCLC), pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, prostate cancer, testicular cancer, thyroid cancer, uterine cancer, adrenal cancer, and head and neck cancer.
In some aspects, the effectiveness of administration of a Cbl-b inhibitor in treating a disease or disorder, such as cancer, is measured by assessing clinical outcome, such as reduction in tumor size or tumor number and/or survival. In certain embodiments, "treating Cancer" includes assessing a patient's response to a treatment regimen according to a response assessment criteria for solid tumors as described (RECIST version 1.1) (see, e.g., eisenhauer et al, european journal of Cancer (Eur J Cancer), 45:228-247,2009; and Nishino et al, U.S. journal of radiology (Am J Roentgenol), 195:281-289,2010). Determining that the response criteria for an objective anti-tumor response comprises a Complete Response (CR) according to RECIST 1.1; partial Response (PR); progressive Disease (PD); and Stable Disease (SD).
Thus, in some embodiments, the Cbl-b mediated disorder is hematologic cancer. In one aspect, provided herein are methods of treating hematologic cancer in a patient. The method comprises administering to the patient a compound disclosed herein (e.g., a compound of formula I), a pharmaceutically acceptable salt thereof, or a pharmaceutical composition disclosed herein (e.g., a composition comprising a compound disclosed herein [ e.g., a compound of formula I ]) and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
More generally, in some embodiments, the Cbl-b mediated disorder is a non-hematologic cancer. In one aspect, provided herein are methods of treating non-hematologic cancers in a patient. The method comprises administering to the patient a compound disclosed herein (e.g., a compound of formula I), a pharmaceutically acceptable salt thereof, or a pharmaceutical composition disclosed herein (e.g., a composition comprising a compound disclosed herein [ e.g., a compound of formula I ]) and a pharmaceutically acceptable carrier, adjuvant, or vehicle. In some embodiments, the non-hematologic cancer is neuroblastoma, renal cell carcinoma, colon cancer, colorectal cancer, breast cancer, epithelial squamous cell carcinoma, melanoma, gastric cancer, brain cancer, lung cancer (e.g., NSCLC), pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, prostate cancer, testicular cancer, thyroid cancer, uterine cancer, adrenal cancer, and head and neck cancer. In some embodiments, the non-hematologic cancer is colon cancer. In some embodiments, the non-hematologic cancer is liver cancer. In some embodiments, the non-hematologic cancer is lung cancer. In some embodiments, the non-hematologic cancer is breast cancer. In some embodiments, the non-hematologic cancer is brain cancer.
Cbl-b inhibitors have also been reported to provide benefits to patients suffering from cancer. Thus, in some embodiments, the Cbl-b mediated disorder is cancer. In one aspect, provided herein are methods of treating cancer in a patient. The method comprises administering to the patient a compound disclosed herein (e.g., a compound of formula I), a pharmaceutically acceptable salt thereof, or a pharmaceutical composition disclosed herein (e.g., a composition comprising a compound disclosed herein [ e.g., a compound of formula I ]) and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
Examples of cancers that may be treated using the compounds of the present disclosure include, but are not limited to, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, cutaneous or intraocular malignant melanoma, uterine cancer, ovarian cancer, rectal cancer, anal region cancer, gastric cancer, testicular cancer, uterine cancer, fallopian tube cancer, endometrial cancer (carcinoma of the endometrium), endometrial cancer (endometrial cancer), cervical cancer, vaginal cancer, vulval cancer, hodgkin's disease, non-hodgkin's lymphoma, esophageal cancer, small intestine cancer, endocrine system cancer, thyroid cancer, parathyroid cancer, adrenal gland cancer, soft tissue sarcoma, urinary tract cancer, penile cancer, chronic or acute leukemia including acute myeloid leukemia, chronic myeloid leukemia, acute lymphocytic leukemia and chronic lymphocytic leukemia, childhood solid tumors, lymphocytic lymphoma, bladder cancer, renal cancer or urinary tract cancer, pelvis cancer, central nervous system neoplasm (CNS), primary CNS lymphoma, tumor angiogenesis, spinal axis tumors, brain stem glioma, pituitary adenoma, karposi's epidermis sarcoma (Kaposi's), cancer induced by T cells, cancer of the cell, and combinations thereof.
In some embodiments, cancers that may be treated using the compounds of the present disclosure include, but are not limited to, solid tumors (e.g., prostate, colon, esophageal, endometrial, ovarian, uterine, renal, liver, pancreatic, gastric, breast, lung, head and neck, thyroid, glioblastoma, sarcoma, bladder, etc.), hematological cancers (e.g., lymphomas, leukemias, such as Acute Lymphoblastic Leukemia (ALL), acute Myeloid Leukemia (AML), chronic Lymphoblastic Leukemia (CLL), chronic Myeloid Leukemia (CML), DLBCL, mantle cell lymphoma, non-Hodgkin's lymphoma (including recurrent or refractory NHL and recurrent follicular), hodgkin's lymphoma (Hodgkin's lymphoma), or multiple myeloma), and combinations of such cancers.
In certain embodiments, the cancer is brain cancer, leukemia, skin cancer, prostate cancer, thyroid cancer, colon cancer, lung cancer, or sarcoma. In another embodiment, the cancer is selected from the group consisting of: glioma, glioblastoma multiforme, paraganglioma, supratentorial primitive neuroectodermal tumor, acute myelogenous leukemia, myelodysplastic syndrome, chronic myelogenous leukemia, melanoma, breast, prostate, thyroid, colon, lung, central chondrosarcoma, central and periosteal osteoma, fibrosarcoma and cholangiocarcinoma.
In certain embodiments, the cancer is selected from brain and spinal cancers, head and neck cancers, leukemia and blood cancers, skin cancers, cancers of the reproductive system, cancers of the gastrointestinal system, liver and bile duct cancers, kidney and bladder cancers, bone cancers, lung cancers, malignant mesothelioma, sarcomas, lymphomas, adenocarcinomas, thyroid, heart tumors, germ cell tumors, malignant neuroendocrine (carcinoid) tumors, cancer of the middle-line tract, and unknown primary cancers (cancers where metastatic cancer was found but the initial cancer site is unknown). In certain embodiments, the cancer is present in an adult patient; in further embodiments, the cancer is present in a pediatric patient. In certain embodiments, the cancer is AIDS-related.
In further embodiments, the cancer is selected from brain cancer and spinal cancer. In particular embodiments, the cancer is selected from the group consisting of: anaplastic astrocytoma, glioblastoma, astrocytoma and sensory neuroblastoma (olfactory blastoma). In particular embodiments, the brain cancer is selected from the group consisting of: astrocytomas (e.g., capillary astrocytomas, ependymal giant astrocytomas, diffuse astrocytomas, polymorphic yellow astrocytomas, anaplastic astrocytomas, giant glioblastomas, secondary glioblastomas, primary adult glioblastomas and primary pediatric glioblastomas), oligodendrogliomas (e.g., oligodendrogliomas and anaplastic oligodendrogliomas), oligodendroastrocytomas (e.g., oligodendroastrocytomas and anaplastic oligodendrogliomas), ependymomas (e.g., mucinous papillary ependymomas and anaplastic ependymomas), medulloblastomas, primitive neuroectodermal tumors, schwannomas, meningiomas, atypical meningiomas, anaplastic meningiomas, pituitary adenomas, brain stem gliomas, cerebellar astrocytomas/glioblastomas, optic pathways and hypothalamic gliomas and primary central nervous system lymphomas. In specific examples of these embodiments, the brain cancer is selected from the group consisting of: gliomas, glioblastoma multiforme, paragangliomas, and supratentorial primitive neuroectodermal tumors (sps).
In particular embodiments, the cancer is selected from head and neck cancer, including nasopharyngeal cancer, nasal and paranasal sinus cancer, hypopharyngeal cancer, oral cancer (e.g., squamous cell carcinoma, lymphoma and sarcoma), lip cancer, oropharyngeal cancer, salivary gland tumor, laryngeal cancer (e.g., laryngeal squamous cell carcinoma, rhabdomyosarcoma), and ocular cancer or eye cancer. In particular embodiments, the eye cancer is selected from the group consisting of intraocular melanoma and retinoblastoma.
In specific embodiments, the cancer is selected from leukemia and hematological cancer. In specific embodiments, the cancer is selected from the group consisting of: myeloproliferative neoplasms, myelodysplastic syndrome, myelodysplastic/myeloproliferative neoplasms, acute Myelogenous Leukemia (AML), myelodysplastic syndrome (MDS), chronic Myelogenous Leukemia (CML), myeloproliferative neoplasms (MPN), post-MPN AML, post-MDS AML, del (5 q) -associated high risk MDS or AML, embryonic chronic myelogenous leukemia, angioimmunoblastic lymphoma, acute lymphoblastic leukemia, langerhans' histiocytosis (Langerans cell histiocytosis), hairy cell leukemia, and plasmacytoid neoplasms including plasmacytoma and multiple myeloma. The leukemias referred to herein may be acute or chronic.
In particular embodiments, the cancer is selected from skin cancer. In particular embodiments, the skin cancer is selected from the group consisting of: melanoma, squamous cell carcinoma, and basal cell carcinoma.
In particular embodiments, the cancer is selected from cancers of the reproductive system. In particular embodiments, the cancer is selected from the group consisting of: breast cancer, cervical cancer, vaginal cancer, ovarian cancer, prostate cancer, penile cancer, and testicular cancer. In specific examples of these embodiments, the cancer is a breast cancer selected from the group consisting of ductal cancer and phyllostatic tumor. In specific examples of these embodiments, the breast cancer may be a male breast cancer or a female breast cancer. In specific examples of these embodiments, the cancer is cervical cancer selected from the group consisting of squamous cell carcinoma and adenocarcinoma. In specific examples of these embodiments, the cancer is ovarian cancer selected from the group consisting of epithelial cancer.
In specific embodiments, the cancer is selected from cancers of the gastrointestinal system. In particular embodiments, the cancer is selected from the group consisting of: esophageal cancer, gastric cancer (also known as gastric cancer), gastrointestinal carcinoid tumors, pancreatic cancer, gall bladder cancer, colorectal cancer, and anal region cancer. In examples of these embodiments, the cancer is selected from the group consisting of: esophageal squamous cell carcinoma, esophageal adenocarcinoma, gastric adenocarcinoma, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, gastric lymphoma, gastrointestinal lymphoma, pancreatic solid pseudopapilloma, pancreatic blastoma, islet cell tumor, pancreatic cancer including acinar cell carcinoma and ductal adenocarcinoma, gall bladder adenocarcinoma, colorectal adenocarcinoma, and anal squamous cell carcinoma.
In specific embodiments, the cancer is selected from liver cancer and cholangiocarcinoma. In a particular embodiment, the cancer is liver cancer (hepatocellular carcinoma). In a particular embodiment, the cancer is cholangiocarcinoma (bile duct cancer); in examples of these embodiments, the cholangiocarcinoma is selected from the group consisting of intrahepatic cholangiocarcinoma and extrahepatic cholangiocarcinoma.
In specific embodiments, the cancer is selected from kidney cancer and bladder cancer. In particular embodiments, the cancer is a kidney cancer selected from the group consisting of: renal cell carcinoma, wilms tumor (Wilms tumor), and transitional cell carcinoma. In particular embodiments, the cancer is bladder cancer selected from the group consisting of: urothelial cancer (transitional cell carcinoma), squamous cell carcinoma, and adenocarcinoma.
In particular embodiments, the cancer is selected from bone cancer. In particular embodiments, the bone cancer is selected from the group consisting of: osteosarcoma, osteomalignant fibrous histiocytoma, ewing's sarcoma (Ewing sarcoma) and chordoma.
In specific embodiments, the cancer is selected from lung cancer. In particular embodiments, the lung cancer is selected from the group consisting of: non-small cell lung cancer, bronchogenic tumors, and pleural and lung blastomas.
In particular embodiments, the cancer is selected from malignant mesothelioma. In particular embodiments, the cancer is selected from the group consisting of epithelial mesothelioma and sarcoma.
In particular embodiments, the cancer is selected from sarcomas. In particular embodiments, the sarcoma is selected from the group consisting of: central chondrosarcoma, central and periosteal osteomas, fibrosarcoma, tenascus hyaline cell sarcoma and kaposi's sarcoma.
In particular embodiments, the cancer is selected from lymphomas. In particular embodiments, the cancer is selected from the group consisting of: hodgkin's lymphoma (e.g., reed-stenberg cell), non-hodgkin's lymphoma (e.g., diffuse large B-cell lymphoma, follicular lymphoma, mycosis fungoides, sezary syndrome), primary central nervous system lymphoma, cutaneous T-cell lymphoma, and primary central nervous system lymphoma.
In particular embodiments, the cancer is selected from adenocarcinomas. In particular embodiments, the cancer is selected from the group consisting of: adrenocortical carcinoma, pheochromocytoma, paraganglioma, pituitary tumor, thymoma and thymus carcinoma.
In specific embodiments, the cancer is selected from thyroid cancer. In particular embodiments, the thyroid cancer is selected from the group consisting of: medullary thyroid carcinoma, papillary thyroid carcinoma, and follicular thyroid carcinoma.
In specific embodiments, the cancer is selected from germ cell tumors. In particular embodiments, the cancer is selected from the group consisting of: malignant extracranial germ cell tumors and malignant extragonadal germ cell tumors. In specific examples of these embodiments, the malignant extragonadal germ cell tumor is selected from the group consisting of non-seminomas and seminomas.
In specific embodiments, the cancer is selected from cardiac tumors. In particular embodiments, the cardiac tumor is selected from the group consisting of: malignant teratoma, lymphoma, rhabdomyosarcoma, hemangiosarcoma, chondrosarcoma, infantile fibrosarcoma and synovial sarcoma.
In some embodiments, cancers that can be treated with the compounds of the present disclosure include melanoma (e.g., metastatic malignant melanoma), renal cancer (e.g., clear cell cancer), prostate cancer (e.g., hormone refractory prostate cancer), breast cancer, triple negative breast cancer, colon cancer, and lung cancer (e.g., non-small cell lung cancer and small cell lung cancer). Additionally, the present disclosure includes refractory or recurrent malignancies whose growth can be inhibited using the compounds of the present disclosure.
In some embodiments, the diseases and indications treatable with the compounds of the present disclosure include, but are not limited to, hematological cancer, sarcoma, lung cancer, gastrointestinal cancer, genitourinary tract cancer, liver cancer, bone cancer, nervous system cancer, gynecological cancer, and skin cancer.
Exemplary hematological cancers include lymphomas and leukemias, such as Acute Lymphoblastic Leukemia (ALL), acute Myeloid Leukemia (AML), acute Promyelocytic Leukemia (APL), chronic Lymphoblastic Leukemia (CLL), chronic Myelogenous Leukemia (CML), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma, non-hodgkin's lymphoma (including recurrent or refractory NHL and recurrent follicular), hodgkin's lymphoma, myeloproliferative diseases (e.g., primary Myelofibrosis (PMF), polycythemia Vera (PV), and primary thrombocythemia (ET)), myelodysplastic syndrome (MDS), T-cell acute lymphocytic lymphoma (T-ALL), multiple myeloma, cutaneous T-cell lymphoma, waldenstrom's macroglobulinemia (Waldenstrom's macroglobulinemia), hairy cell lymphoma, chronic lineage lymphoma, and Burkitt's lymphoma (Burkitt's lymphoma).
Exemplary sarcomas include chondrosarcoma, ewing's sarcoma, osteosarcoma, rhabdomyosarcoma, angiosarcoma, fibrosarcoma, liposarcoma, myxoma, rhabdomyoma, rhabdomyosarcoma, fibroma, lipoma, hamartoma, and teratoma.
Exemplary lung cancer includes non-small cell lung cancer (NSCLC), small cell lung cancer, bronchogenic carcinoma (squamous cell carcinoma, undifferentiated small cell carcinoma, undifferentiated large cell carcinoma, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, chondromatous hamartoma, and mesothelioma.
Exemplary gastrointestinal cancers include esophageal cancer (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), gastric cancer (carcinoma, lymphoma, leiomyosarcoma), pancreatic cancer (ductal adenocarcinoma, insulinoma, glucagon tumor, gastrinoma, carcinoid, schwann intestinal peptide tumor), small intestine cancer (adenocarcinoma, lymphoma, carcinoid, kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large intestine cancer (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, smooth muscle cancer), and colorectal cancer.
Exemplary genitourinary tract cancers include renal cancer (adenocarcinoma, wilms' tumor [ nephroblastoma ]), bladder and urinary tract cancers (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate cancer (adenocarcinoma, sarcoma), and testicular cancer (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumor, lipoma).
Exemplary liver cancers include liver cancer (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, and hemangioma.
Exemplary bone cancers include, for example, osteogenic sarcomas (osteosarcoma), fibrosarcomas, malignant fibrous histiocytomas, chondrosarcomas, ewing's sarcoma, malignant lymphomas (reticulosarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochondral tumors (bone exochondral osteowarts), benign chondrias, chondroblastomas, chondromyxoid fibromas, osteoid osteomas, and giant cell tumors.
Exemplary nervous system cancers include head bone cancer (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meningioma (meningioma, glioblastoma), brain cancer (astrocytoma, medulloblastoma, glioma, ependymoma, germ cell tumor (pineal tumor), glioblastoma multiforme, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), and spinal cord cancer (neurofibroma, meningioma, glioma, sarcoma), neuroblastoma, and hermite-Du Duosi disease (lhemitte-Duclos disease).
Exemplary gynaecological cancers include uterine cancer (endometrial cancer), cervical cancer (cervical cancer, pre-tumor cervical atypical hyperplasia), ovarian cancer (serous cyst adenocarcinoma, mucinous cyst adenocarcinoma, unclassified cancer), granulosa cell tumors, sertoli-Leydig cell tumor, asexual cell tumors, malignant teratoma), vulval cancer (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vaginal cancer (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), and fallopian tube (carcinoma).
Exemplary skin cancers include melanoma, basal cell carcinoma, squamous cell carcinoma, kaposi's sarcoma, meckel cell skin carcinoma (Merkel cell skin cancer), nevus dysplasia nevus, lipoma, hemangioma, dermal fibroma, and nevus dysplasia nevus. In some embodiments, diseases and indications treatable using the compounds of the present disclosure include, but are not limited to, sickle cell disease (e.g., sickle cell anemia), triple Negative Breast Cancer (TNBC), myelodysplastic syndrome, testicular cancer, cholangiocarcinoma, esophageal cancer, and urothelial cancer.
Exemplary head and neck cancers include glioblastoma, melanoma, rhabdomyosarcoma, lymphosarcoma, osteosarcoma, squamous cell carcinoma, adenocarcinoma, oral cancer, laryngeal cancer, nasopharyngeal cancer, nasal and paranasal sinus cancer, thyroid cancer, and parathyroid cancer.
The disclosed compounds may be administered in any suitable manner known in the art. In some embodiments, the compounds of the invention, or pharmaceutically acceptable salts, prodrugs, metabolites, or derivatives thereof, are administered intravenously, intramuscularly, subcutaneously, topically, orally, transdermally, intraperitoneally, intraorbitally, implantable, inhaled, intrathecally, intraventricularly, intratumorally, or intranasally.
In some embodiments, the Cbl-b inhibitor is administered continuously. In other embodiments, the Cbl-b inhibitor is administered intermittently. Furthermore, treating a subject with an effective amount of a Cbl-b inhibitor may comprise a single treatment or may comprise a series of treatments.
It will be appreciated that the appropriate dosage of the active compound will depend upon a number of factors within the knowledge of the ordinarily skilled physician or veterinarian. The dosage of the active compound will vary, for example, depending on the age, weight, general health, sex and diet of the subject, the time of administration, the route of administration, the rate of excretion, and any combination of drugs.
It will also be appreciated that the effective dose of a compound of the invention or a pharmaceutically acceptable salt, prodrug, metabolite, or derivative thereof for use in therapy may be increased or decreased over the course of a particular therapy. The variation in dosage may be produced by the results of a diagnostic assay and become apparent.
In some embodiments, the Cbl-b inhibitor is administered to the subject at a dose of between about 0.001 μg/kg and about 1000mg/kg, including but not limited to about 0.001 μg/kg, 0.01 μg/kg, 0.05 μg/kg, 0.1 μg/kg, 0.5 μg/kg, 1 μg/kg, 10 μg/kg, 25 μg/kg, 50 μg/kg, 100 μg/kg, 250 μg/kg, 500 μg/kg, 1mg/kg, 5mg/kg, 10mg/kg, 25mg/kg, 50mg/kg, 100mg/kg, and 200 mg/kg.
As used herein, the term "treating (treatment, treat and treating)" refers to reversing, alleviating, delaying the onset of, or inhibiting the progression of a disease or disorder or one or more symptoms thereof as described herein. In some embodiments, the treatment may be administered after one or more symptoms are produced. In other embodiments, the treatment may be administered in the absence of symptoms. For example, treatment may be administered to a susceptible individual prior to onset of symptoms (e.g., based on symptom history and/or based on genetic or other susceptibility factors). Treatment may also be continued after the symptoms subside, for example, to prevent or delay recurrence thereof.
In some embodiments, the compounds of the invention may be used to prevent or reduce the risk of developing any of the diseases mentioned herein; for example, preventing or reducing the risk of developing a disease, condition or disorder in an individual who may have been previously diagnosed with the disease, condition or disorder but has not undergone or exhibited the pathology or symptomology of the disease.
The term "administration" or "administering" encompasses the route by which a compound is introduced into a subject to perform its intended function. Examples of routes of administration that may be used include injection (subcutaneous, intravenous, parenteral, intraperitoneal, intrathecal), topical, oral, inhalation, rectal and transdermal.
The term "effective amount" encompasses an amount effective to achieve the desired result in a dosimeter and for the desired period of time. The effective amount of the compound may vary depending on factors such as the disease state, age and weight of the subject, and the ability of the compound to elicit a desired response in the subject. The dosage regimen may be adjusted to provide the optimal therapeutic response.
As used herein, the phrases "systemic administration (systemic administration and administered systemically)" and "peripheral administration (peripheral administration and administered peripherally)" mean administration of a compound, drug or other material such that it enters the patient's system and thus undergoes metabolism and other similar processes.
The phrase "therapeutically effective amount" means an amount of a compound of the invention that (i) treats or prevents a particular disease, condition, or disorder, (ii) alleviates, ameliorates, or eliminates one or more symptoms of a particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of a particular disease, condition, or disorder described herein.
The term "subject" refers to an animal, such as a mammal, including, but not limited to, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, and the like. In certain embodiments, the subject is a human.
Combination therapy
Depending on the particular condition or disease to be treated, additional therapeutic agents that are typically administered in order to treat the condition may be administered in combination with the compounds and compositions of the present invention. As used herein, an additional therapeutic agent that is typically administered in order to treat a particular disease or condition is referred to as "suitable for the disease or condition being treated.
In certain embodiments, the provided combination or composition thereof is administered in combination with another therapeutic agent. Examples of agents that may also be combined with the combinations of the present invention include, but are not limited to: for the treatment of Alzheimer's Disease, e.gAnd->For the treatment of HIV, such as ritonavir (ritonavir);for the treatment of Parkinson's Disease, such as L-DOPA/carbidopa (carbidopa), entacapone (entacapone), ropinirole (ropinirole), pramipexole (pramipexole), bromocriptine (bromocriptine), pergolide (pergolide), trihexyphenyl (trihexyphenyl) and amantadine; agents for treating Multiple Sclerosis (MS), such as interferon-beta (e.g., ++>And->)、/>And mitoxantrone (mitoxantrone); for the treatment of asthma, e.g. albuterol and +.>Agents for the treatment of schizophrenia, such as reproprxa, risperidol, sirolimus (seroque) and haloperidol (haloperidol); anti-inflammatory agents such as corticosteroids, TNF blockers, IL-1RA, azathioprine, cyclophosphamide and sulfasalazine; immunomodulatory and immunosuppressant agents such as cyclosporine, tacrolimus (tacrolimus), rapamycin (rapamycin), mycophenolate mofetil (mycophenolate mofetil), interferons, corticosteroids, cyclophosphamide, azathioprine and sulfasalazine; neurotrophic factors, such as acetylcholinesterase inhibitors, MAO inhibitors, interferons, anticonvulsants, ion channel blockers, riluzole (riluzole), and antiparkinsonian agents; agents for the treatment of cardiovascular diseases such as beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers and statins (statins); agents for treating liver diseases, such as corticosteroids, cholestyramine, interferons and antiviral agents; agents for treating hematological disorders, such as corticosteroids, anti-leukemia agents and growth factors; agents that prolong or improve pharmacokinetics, such as cytochrome P450 inhibitors (i.e., inhibitors of metabolic breakdown) and CYP3A4 inhibition Agents (e.g., ketoconazole (ketoconazole) and ritonavir); and agents for treating immunodeficiency disorders, such as gamma globulin.
In certain embodiments, the combination therapies of the invention, or pharmaceutically acceptable compositions thereof, are administered in combination with a monoclonal antibody or siRNA therapeutic.
These additional agents may be administered separately from the provided combination therapies as part of a multi-dose regimen. Alternatively, those agents may be part of a single dosage form, which is mixed in a single composition with the compounds of the present invention. If administered as part of a multi-dose regimen, the two active agents may be delivered simultaneously, sequentially, or within a period of time of each other (typically five hours of each other).
As used herein, the terms "combination," "combined," and related terms refer to the simultaneous or sequential administration of therapeutic agents in accordance with the present invention. For example, the combination of the invention may be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms or together in a single unit dosage form.
The amount of additional therapeutic agent present in the compositions of the present invention will not exceed the amount typically administered in compositions comprising the therapeutic agent as the sole active agent. Preferably, the amount of additional therapeutic agent in the presently disclosed compositions will be in the range of about 50% to 100% of the amount typically present in compositions comprising the agent as the sole therapeutically active agent.
In one embodiment, the present invention provides a composition comprising a compound of formula I and one or more additional therapeutic agents. The therapeutic agent may be administered with the compound of formula I or may be administered before or after administration of the compound of formula I. Suitable therapeutic agents are described in further detail below. In certain embodiments, the compound of formula I may be administered up to 5 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, or 18 hours prior to the therapeutic agent. In other embodiments, the compound of formula I may be administered up to 5 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, or 18 hours after the therapeutic agent.
In some embodiments, the invention provides a method of treating a metabolic disorder, disease, or condition described herein, comprising administering a compound of the invention in combination with one or more agents. Suitable agents that may be used in combination with the compounds of the present invention include antidiabetic agents, antiobesity agents (including appetite suppressants), antihyperglycemic agents, lipid lowering agents and antihypertensive agents.
Suitable antidiabetic agents that may be used in combination with the provided compounds or combinations thereof include, but are not limited to, other acetyl-coa carboxylase (ACC) inhibitors, DGAT-1 inhibitors, AZD7687, LCQ908, DGAT-2 inhibitors, monoacylglycerol O-acyltransferase inhibitors, PDE-10 inhibitors, AMPK activators, sulfonylureas (e.g., acetohexanamide, chlorpropamide, glibenclamide, glipizide, glibenclamide, blimipiride, gliclazide, glimeperide, gliquidone, gliclazide, tolazamide, tolbutamide, meglitide), meglitide, alpha amylase inhibitors (e.g., amylin (tendamistat), treastatin, AL-3688), alpha-glucosidase inhibitors (e.g., acarbose), alpha-glucosidase inhibitors (e.g., lipolytic (adiplosin), carboglibose (camigibose), ethylgliclate (emiglite), miglitol (miglitol), voglibose (voglibose), pradimicin-Q (pradimicin-Q), sarbostatin), PPAR-gamma agonists (e.g., balaglitazone), ciglitazone, dapaglitazone, englitazone), iximazone (isaglitazone), pioglitazone (pioglitazone), roside-Q (rosiglitazone), pioglitazone), troglitazone), PPAR-alpha/gamma agonists (e.g., CLX-0940, GW-1536, GW-1929, GW-2433, KRP-297, L-796449, LR-90, MK-0767, SB-219994), biguanides (e.g., metformin (metformin), buformin (buformin)), GLP-1 modulators (exenatide-3, exenatide-4), liraglutide (liraglutide), abilutide (albiglutide), exenatide (yetida), tasraglutide (lissenatide), dulcin (dulaglide), semaphoride (semaphoride), N, N-9924, TTP-054, PTP-1B inhibitor (Japanese Luo Dusi quinine, hyssopal extract), SIRT-1 inhibitor (e.g. resveratrol, GSK2245840, GSK 184072), DPP-IV inhibitor (e.g. sitagliptin, vildagliptin, alogliptin, duloxetine, lin Gelie-line, saxagliptin), insulin secretagogue, fatty acid oxidation inhibitor, A2 antagonist, JNK inhibitor, glucokinase activator (e.g. TTP-399, TTP-355, TTP-547, AZD1656, ARRY403, MK-0599, TAK-329, AZD5658, GKM-001), insulin mimetic, insulin, glycogen phosphorylase inhibitors (e.g., GSK 1362885), VPAC2 receptor agonists, SGLT2 inhibitors (dapagliflozin), canagliflozin (canagliflozin), BI-10733, tolagliflozin (tofogliflozin), ASP-1941, THR1474, TS-071, ISIS388626, LX 4211), glucagon receptor modulators, GPR119 modulators (e.g., MBX-2982, GSK1292263, APD597, PSN 821), FGF21 derivatives, TGR5 (GPBAR 1) receptor agonists (e.g., INT 777), GPR40 agonists (e.g., TAK-875), GPR120 agonists, nicotinic acid receptor (HM 74A) activators, SGLT1 inhibitors (e.g., GSK 1614235), carnitine palmitoyl transferase inhibitors, fructose 1, 6-bisphosphatase inhibitors, aldose reductase inhibitors, mineralocorticoid receptor inhibitors, TORC2 inhibitors, CCR5 inhibitors, PKC (e.g., PKC- α, PKC- β, PKC- γ) inhibitors, fatty acid synthase inhibitors, serine palmitoyl transferase inhibitors, GPR81 modulators, GPR39 modulators, GPR43 modulators, GPR41 modulators, GPR105 modulators, kv1.3 inhibitors, retinol binding protein 4 inhibitors, glucocorticoid receptor modulators, somatostatin receptors (e.g., SSTR1, SSTR2, SSTR3, SSTR 5) inhibitors, PDHK2 inhibitors, PDHK4 inhibitors, MAP4K4 inhibitors, IL1- β modulators, and RXR- α modulators.
Suitable lipid lowering agents that may be used in combination with the provided compounds or compositions thereof include, but are not limited to: cholic acid chelators, HMG-CoA reductase inhibitors, HMG-CoA synthase inhibitors, cholesterol absorption inhibitors, acyl-CoA-cholesterol acyltransferase (ACAT) inhibitors, CETP inhibitors, squalene synthase inhibitors, PPAR-alpha agonists, FXR receptor modulators, LXR receptor modulators, lipoprotein synthesis inhibitors, inhibitors of the renin-angiotensin system, PPAR-delta partial agonists, bile acid reabsorption inhibitors, PPAR-gamma agonists, triglyceride synthesis inhibitors, microsomal triglyceride transfer inhibitors, transcription regulators, squalene epoxidase inhibitors, low density lipoprotein receptor inducers, platelet aggregation inhibitors, 5-LO or FLAP inhibitors, niacin and niacin-binding chromium.
Suitable anti-obesity agents include, but are not limited to, 11-betA-hydroxysteroid dehydrogenase 1 inhibitors, stearoyl-CoA desaturase (SCD-1) inhibitors, MCR-4 agonists, CCK-A agonists, monoamine reuptake inhibitors, sympathomimetics, betA-3-adrenoreceptor agonists, dopamine receptor agonists (e.g., bromocriptine), melanocyte stimulating hormone and analogs thereof, 5-HT2c agonists (e.g., lorcaserin (bermask)), melanin concentrating hormone antagonists, leptin analogs, leptin agonists, galanin antagonists, lipase inhibitors (e.g., tetrahydrofat suppressant/Orlistat), anorexiA (e.g., bombesin agonists (bombesin agonists)), NPY antagonists (e.g., virucit (velnereipine)), PY 3-36 (and analogs thereof), BRS3 modulators, AGP 3 antagonists, opioid antagonists, oxadoline antagonists, e.g., MTP-35B, MTP-35B, 35H, MTP-related inhibitors (e.g., 35H), e.g., 35B-35, MTP-35, or other inhibitors, ZGN-433), agents having mixed modulating activity at two or more of glucagon, GIP and GLP1 receptors (e.g., MAR-701, ZP 2929), norepinephrine reuptake inhibitors, opioid antagonists (e.g., naltrexone), CB1 receptor antagonists or inverse agonists, ghrelin agonists (ghrelin agonists) or antagonists, gastrin modulators and analogs thereof, monoamine uptake inhibitors (e.g., tesofensine), and combinations (e.g., bupropion) plus zonisamide (emamectin), pramlintide (pramlintide) plus mevaltriptin, bupropion plus naltrexone (Contrave), phentermine plus topiramate (qsymida).
In some embodiments, the anti-obesity agent used in combination with the provided compounds or compositions thereof is selected from the group consisting of gut selective MTP inhibitors (e.g., dirlotapide, imperatorin (impatiapide), R56918), CCK-A agonists, 5-HT2C agonists (e.g., lorcaserin/bellvisk), MCR4 agonists, lipase inhibitors (e.g., cetiristat), PYY3-36 (including analogs and pegylated analogs thereof), opioid antagonists (e.g., naltrexone), oleoyl introns, ornipide (obiepitide), pramlintide, tesrofen, leptin, bromocriptine, orlistat, and AOD-9604.
In another embodiment, the invention provides a method of treating an inflammatory disease, disorder or condition by administering to a patient in need thereof a compound of formula I and one or more additional therapeutic agents. Such additional therapeutic agents may be small molecule or recombinant biological agents and include, for example, acetaminophen, non-steroidal anti-inflammatory drugs (NSAIDS) (e.g., aspirin, ibuprofen, naproxen, etodolac)And celecoxib), colchicine +.>Corticosteroids (e.g. prednisone, prednisolone, methyl)Prednisolone, hydrocortisone, and the like), probenecid, allopurinol, febuxostat Sulfasalazine->Antimalarial agents (e.g. hydroxychloroquine +.>And chloroquine->Methotrexate>) Gold salts (e.g. thioglucogold->Gold thiomalate->And auranofin->) D-penicillamine (>Or->) Azathioprine->Cyclophosphamide->Chlorambucil->CyclosporineLeflunomide->And "anti-TNF" agents (e.g., etanercept->Infliximab->Golimumab->Cetuzumab polyethylene glycol ∈ ->And adalimumab->) "anti-IL-1" agents (e.g., anakinra +)>And Li Naxi p) Carbamab->anti-Jak inhibitors (e.g. tofacitinib), antibodies (e.g. rituximab)>) "anti-T cell" agents (e.g. Abapyrite>) "anti-IL-6" agents (e.g. tolizumab>) Diclofenac, cortisone, hyaluronic acid (++>Or->) Monoclonal antibodies (e.g. tani beads), anticoagulants (e.g. heparin) (-je>Or->) And warfarinAntidiarrheal (e.g. diphenoxylate +.>And loperamide->) Bile acid binders (e.g. cholestyramine), alosetron->Lubiprostone->Laxatives (e.g. magnesium emulsion, polyethylene glycol +.>And->) Anticholinergic or antispasmodic agents (e.g. dicyclo-amine +.>Beta-2 agonists (e.g. salbutamol (/ -A)>HFA、HFA), levosalbutamol +.>Ocinalin->Pibuterol acetate Terbutaline sulfate->Salmeterol xinafoate>And formoterol) Anticholinergic agents (e.g. ipratropium bromide +)>And tiotropium bromide->) Inhaled corticosteroids (such as beclomethasone dipropionate (/ -)>And->) Triamcinolone acetonideMomethasone->Budesonide->And flunisolide Cromolyn sodium->Methylxanthines (e.g. theophylline +)>And aminophylline), igE antibodies (e.g. omalizumab +.>) Nucleoside reverse transcriptase inhibitors (e.g. zidovudine->Abacavir->Abacavir/lamivudine->Abacavir/lamivudine/zidovudineDedanosin->Emtricitabine->Lamivudine->Lamivudine/zidovudine->Stavudine->And zalcitabine->Non-nucleoside reverse transcriptase inhibitors (e.g. delavirdine +.>Efavirenz->NevirapineAnd itravirin>Nucleotide reverse transcriptase inhibitors (e.g. tenofovir) Protease inhibitors (e.g. amprenavir->Atazanavir->Darunavir->Phosphanavir->Indinavir->Lopinavir and ritonavirNefinavir->Ritonavir->Saquinavir (+)>Or (b)) And telanavir->) An entry inhibitor (e.g. Enfuvirtide->And malavir->) An integrase inhibitor (e.g. raltegravir->AdriamycinVincristine->Bortezomib->And lenalidomide- >Combined dexamethasone->) Or any one or more combinations thereof.
In another embodiment, the invention provides a method of treating hematological malignancies comprising administering to a patient in need thereof a compound of formula I and one or more additional therapeutic agents selected from the group consisting of: rituximabCyclophosphamide->Adriamycin->VincristinePrednisone, hedgehog signaling inhibitors, BTK inhibitors, JAK/ubijak inhibitors, PI3K inhibitors, SYK inhibitors, and combinations thereof.
In another embodiment, the invention provides a method of treating a solid tumor comprising administering to a patient in need thereof a compound of formula I and one or more additional therapeutic agents selected from the group consisting of: rituximabCyclophosphamide->Adriamycin->VincristinePrednisone, hedgehog signaling inhibitors, BTK inhibitors, JAK/ubijak inhibitors, PI3K inhibitors, SYK inhibitors, and combinations thereof.
In another embodiment, the invention provides a method of treating hematological malignancies comprising administering to a patient in need thereof a compound of formula I and a hedgehog (Hh) signaling pathway inhibitor. In some embodiments, the hematological malignancy is DLBCL (Ramirez et al, "define causative factors (Defining causative factors contributing in the activation of hedgehog signaling in diffuse large B-cell lymphoma) that promote activation of hedgehog signaling in diffuse large B-cell lymphomas)" (2012) issued online on day 7 and 17 and incorporated herein by reference in its entirety.
In another embodiment, the invention provides a method of treating diffuse large B-cell lymphoma (DLBCL) comprising administering to a patient in need thereof a compound of formula I and one or more additional therapeutic agents selected from the group consisting of: rituximabCyclophosphamide->Doxorubicin->Vincristine->Prednisone, hedgehog signaling inhibitors, and combinations thereof.
In another embodiment, the invention provides a method of treating multiple myeloma comprising administering to a patient in need thereof a compound of formula I and one or more additional therapeutic agents selected from the group consisting of lenalidomideIs a group of (2)And (3) combining: bortezomib (bortezomib)>And dexamethasone (dexamethasone)>Hedgehog signaling inhibitors, BTK inhibitors, JAK/ubijak inhibitors, TYK2 inhibitors, PI3K inhibitors, SYK inhibitors.
In another embodiment, the invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I and a BTK inhibitor, wherein the disease is selected from inflammatory bowel disease, arthritis, cutaneous lupus erythematosus, systemic Lupus Erythematosus (SLE), vasculitis, idiopathic Thrombocytopenic Purpura (ITP), rheumatoid arthritis, psoriatic arthritis, osteoarthritis, still's disease, juvenile arthritis, diabetes, myasthenia gravis, hashimoto's thyroiditis, ord's thyroiditis, graves ' disease, autoimmune thyroiditis, sjogren's syndrome, multiple sclerosis, systemic sclerosis, neurosis, guillain-Barre syndrome, hashimoto-Barre syndrome acute disseminated encephalomyelitis, addison's disease, ocular clonus-myoclonus syndrome, ankylosing spondylitis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, autoimmune gastritis, pernicious anemia, celiac disease, goodpasture's syndrome, idiopathic thrombocytopenic purpura, optic neuritis, scleroderma, primary biliary cirrhosis, rate's syndrome, takayasu's arteritis, temporal arteritis, warm autoimmune hemolytic anemia, wegener's granulomatosis, psoriasis, alopecia universalis, bezier's disease, chronic fatigue, autonomic nervous disorders, membranous glomerulonephropathy, endometriosis, interstitial cystitis, pemphigus vulgaris, bullous pemphigoid, neuromyotonia, scleroderma, vulvodynia, hyperproliferative diseases, rejection of transplanted organs or tissues, acquired immunodeficiency syndrome (AIDS, also known as HIV), type 1 diabetes, graft versus host disease, transplantation, transfusion, anaphylaxis, allergies (e.g., allergic to plant pollen, latex, drugs, foods, insect poisons, animal hair, animal dander, dust mites, or cockroaches), type I hypersensitivity, allergic conjunctivitis, allergic rhinitis and atopic dermatitis, asthma, appendicitis, atopic dermatitis, asthma, allergy, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, cholecystitis, chronic graft rejection, colitis, conjunctivitis, crohn's disease, cystitis, lacrimal gland inflammation, dermatitis, dermatomyositis, encephalitis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, inflammation of the liver, inflammation of the stomach, and inflammation of the liver epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, henoch-Schonlein purpura (Henoch-Schonlein purpura), hepatitis, hidradenitis suppurativa, immunoglobulin A nephropathy, interstitial lung disease, laryngitis, mastitis, meningitis, myelitis, myocarditis, myositis, nephritis, ovaritis, orchitis, osteomyelitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleurisy, phlebitis, pneumonia, pulmonary infection, polymyositis, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendinitis, tonsillitis, ulcerative colitis, uveitis, vaginitis, vasculitis or vulvitis, B-cell proliferative disorders such as diffuse large B-cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia, acute lymphocytic leukemia, B-cell prolymphocytic leukemia, lymphoplasmacytic lymphoma/Waldenstem megalobulinemia, splenic marginal zone lymphoma, multiple myeloma (also known as plasma cell myeloma), non-Hodgkin's lymphoma, plasmacytoma, extranodal marginal zone B-cell lymphoma, nodal marginal zone B-cell lymphoma, mantle cell lymphoma, mediastinal (thymus) large B-cell lymphoma, intravascular large B-cell lymphoma, primary exudative lymphoma, burkitt's lymphoma/leukemia or lymphomatoid granulomatosis, breast cancer, prostate cancer or mast cell carcinoma (e.g., mast cell tumors, mast cell leukemia, mast cell sarcoma, systemic mastocytosis), bone cancers, colorectal cancers, pancreatic cancers, bone and joint diseases including, but not limited to, rheumatoid arthritis, seronegative spondyloarthropathies (including ankylosing spondylitis, psoriatic arthritis and rayleigh's disease), bezier's disease, sjogren's syndrome, systemic sclerosis, osteoporosis, bone cancers, bone metastases, thromboembolic disorders (e.g., myocardial infarction, angina, restenosis after angioplasty, restenosis after aortic coronary artery bypass surgery, stroke, transient ischemia, peripheral arterial occlusive disorders, pulmonary embolism, myocardial infarction, vascular graft, and vascular graft, deep vein thrombosis), inflammatory pelvic disorders, urethritis, skin sunburn, sinusitis, pneumonia, encephalitis, meningitis, myocarditis, nephritis, osteomyelitis, myositis, hepatitis, gastritis, enteritis, dermatitis, gingivitis, appendicitis, pancreatitis, cholecystitis, agaropectinemia, psoriasis, allergy, crohn's disease, irritable bowel syndrome, ulcerative colitis, sjogren's syndrome, tissue graft rejection, hyperacute rejection of transplanted organs, asthma, allergic rhinitis, chronic Obstructive Pulmonary Disease (COPD), autoimmune polyadenylic disease (also known as autoimmune polyadenylic syndrome), autoimmune alopecia, pernicious anemia, glomerulonephritis, dermatomyositis, multiple sclerosis, scleroderma, vasculitis autoimmune hemolytic and thrombocytopenia, pulmonary hemorrhagic nephritis syndrome, atherosclerosis, edison's disease, parkinson's disease, alzheimer's disease, diabetes, septic shock, cutaneous lupus erythematosus, systemic Lupus Erythematosus (SLE), rheumatoid arthritis, psoriatic arthritis, juvenile arthritis, osteoarthritis, chronic idiopathic thrombocytopenic purpura, waldenstrom's macroglobulinemia, myasthenia gravis, hashimoto's thyroiditis, atopic dermatitis, degenerative joint disease, vitiligo, autoimmune hypopituitary disease, green-barre syndrome, bezier's disease, scleroderma, mycosis, acute inflammatory responses (such as acute respiratory distress syndrome and ischemia/reperfusion injury), and grignard disease.
In another embodiment, the invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I and a PI3K inhibitor, wherein said disease is selected from the group consisting of cancer, a neurodegenerative disorder, an angiogenic disorder, a viral disease, an autoimmune disease, an inflammatory disorder, a hormone-related disease, a condition associated with organ transplantation, an immunodeficiency disorder, a destructive bone disorder, a proliferative disorder, an infectious disease, a condition associated with cell death, thrombin-induced platelet aggregation, chronic Myelogenous Leukemia (CML), chronic Lymphocytic Leukemia (CLL), liver disease, a pathological immune condition involving T cell activation, a cardiovascular disorder, and a CNS disorder.
In another embodiment, the invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I and a PI3K inhibitor, wherein said disease is selected from the group consisting of brain, kidney (e.g., renal Cell Carcinoma (RCC)), liver, adrenal gland, bladder, breast, stomach (stomach), gastric (graphic) tumor, ovary, colon, rectum, prostate, pancreas, lung, vagina, endometrium, cervix, testis, genitourinary tract, esophagus, larynx, skin, benign or malignant tumors of bone or thyroid, cancer or solid tumors, sarcomas, glioblastomas, neuroblastomas, multiple myeloma, or gastrointestinal cancer, in particular colon cancer or colorectal adenoma or neck and head tumors, epidermal hyperproliferation, psoriasis, prostatic hyperplasia, neoplasia, epithelial neoplasia, adenoma, adenocarcinoma, keratoacanthoma, epidermoid carcinoma, large cell carcinoma, non-small cell lung carcinoma, lymphomas (including, for example, non-hodgkin's lymphoma (NHL) and hodgkin's lymphoma (also known as hodgkin's disease or hodgkin's disease)), breast cancer, follicular carcinoma, undifferentiated carcinoma, mastoid carcinoma, seminoma, melanoma or leukemia, diseases including Cowden syndrome (Cowden syndrome), hermite-Du Duosi disease (lhemitte-Dudos disease) and Bannayan-zona syndrome), or diseases of abnormal activation of the PI 3K/b pathway, asthma of any type or origin, including endogenous (non-allergic) asthma and exogenous (allergic) asthma, asthma, moderate asthma, severe asthma, bronchitis, exercise-induced asthma, occupational asthma and asthma induced after bacterial infection, acute Lung Injury (ALI), adult/Acute Respiratory Distress Syndrome (ARDS), chronic obstructive pulmonary, airway or lung disease (COPD, COAD or COLD), dyspnea comprising or associated with chronic bronchitis, emphysema and exacerbation of airway Gao Minxing caused by other medications, particularly other medications inhalation treatments, bronchitis of any type or origin, including but not limited to acute, arachnoid inhalation, catarrhal, grippy, chronic or tuberculous bronchitis, pneumoconiosis of any type or origin (inflammatory (usually occupational) lung disease, often accompanied by airway obstruction, whether chronic or acute, and caused by repeated inhalation of dust), including, for example, alumino-pneumoconiosis, coal-pneumoconiosis, asbestosis, chalicosis, lash-drop, siderosis, silicosis, smoke-and-dust, lofoler's syndrome, eosinophilic pneumonia, parasitic (especially metazoan) infections (including tropical eosinophilia), bronchopulmonary aspergillosis, polyarteritis nodosa (including Churg-Strauss syndrome), eosinophilic granuloma and eosinophil-related pathologies affecting the airways caused by drug reactions, psoriasis, contact dermatitis, atopic dermatitis, alopecia areata, erythema multiforme, dermatitis herpetiformis, scleroderma, vitiligo, hypersensitivity vasculitis, urticaria, bullous pemphigoid, lupus erythematosus, herpes, acquired epidermolysis bullosa, conjunctivitis, keratoconjunctivitis sicca and vernal conjunctivitis, diseases affecting the nose including allergic rhinitis, inflammatory diseases involving autoimmune reactions or having an autoimmune component or etiology, including autoimmune blood conditions such as hemolytic anemia, aplastic anemia, pure erythrocyte aplastic anemia and idiopathic thrombocytopenia, cutaneous lupus erythematosus, systemic lupus erythematosus, rheumatoid arthritis, polychondritis, scleroderma, wegener granulomatosis (Wegener granulamatosis), dermatomyositis, chronic active hepatitis, myasthenia gravis, steven-Johnson syndrome (Steven-Johnson syndrome), idiopathic steatorrhea, autoimmune inflammatory bowel disease (e.g., ulcerative colitis and Crohn's disease), endocrinological disorders, griff's disease, sarcoidosis, alveolar inflammation, chronic allergic pneumonia, multiple sclerosis, primary biliary cirrhosis, uveitis (anterior and posterior), keratoconjunctivitis sicca and vernal keratoconjunctivitis, interstitial pulmonary fibrosis, psoriatic arthritis and glomerulonephritis (with and without nephrotic syndrome), e.g. including idiopathic nephrotic syndrome or morbid renal diseases, restenosis, cardiac hypertrophy, atherosclerosis, myocardial infarction, ischemic stroke and congestive heart failure, alzheimer's disease, parkinson's disease, amyotrophic lateral sclerosis, huntington's disease and cerebral ischemia and traumatic injury, glutamate neurotoxicity and hypoxia-induced neurodegenerative diseases.
In some embodiments, the invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I and a Bcl-2 inhibitor, wherein said disease is an inflammatory disorder, an autoimmune disorder, a proliferative disorder, an endocrine disorder, a neurological disorder, or a disorder associated with transplantation. In some embodiments, the disorder is a proliferative disorder, lupus, or lupus nephritis. In some embodiments, the proliferative disorder is chronic lymphocytic leukemia, diffuse large B-cell lymphoma, hodgkin's disease, small cell lung cancer, non-small cell lung cancer, myelodysplastic syndrome, lymphoma, hematological neoplasm, or solid tumor.
In some embodiments, the disease is an autoimmune disorder, an inflammatory disorder, a proliferative disorder, an endocrine disorder, a neurological disorder, or a disorder associated with transplantation. In some embodiments, the JH2 binding compound is a compound of formula I. Other suitable JH2 domain binding compounds include those described in WO 2014074660A1, WO 2014074661A1, WO 2015089143A1, the entire contents of each of which are incorporated herein by reference. Suitable JH1 domain binding compounds include those described in WO 2015131080A1, the entire contents of which are incorporated herein by reference.
The compounds of the present invention may also be used in combination with other therapeutic compounds to advantage. In some embodiments, the other therapeutic compound is an antiproliferative compound. Such antiproliferative compounds include, but are not limited to: an aromatase inhibitor; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule active compounds; an alkylating compound; histone deacetylase inhibitors; compounds that induce cellular differentiation processes; a cyclooxygenase inhibitor; an MMP inhibitor; an mTOR inhibitor; an anti-tumor antimetabolite; a platinum compound; compounds targeting protein or lipid kinases/reducing their activity and further anti-angiogenic compounds; a compound that targets, reduces or inhibits the activity of a protein or lipid phosphatase; gonadotrophin releasing hormone agonists; antiandrogens; methionine aminopeptidase inhibitors; matrix metalloproteinase inhibitors; bisphosphonates; a biological response modifier; an anti-proliferative antibody; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; a proteasome inhibitor; a compound for use in the treatment of hematological malignancies; a compound that targets Flt-3, reduces or inhibits its activity; hsp90 inhibitors such as 17-AAG (17-allylamino geldanamycin, NSC 330507), 17-DMAG (17-dimethylaminoethylamino-17-desmethoxy-geldanamycin, NSC 707545), IPI-504, CNF1010, CNF2024, CNF1010 from Kang Fuma pharmaceutical company (Conforma Therapeutics); temozolomide Kinesin spindle protein inhibitors such as SB715992 or SB743921 from the company glaxoSmithKline, or pentamidine/chlorpromazine from the company CombinatRx, kangbei Torilex; MEK inhibitors such as ARRY142886 from alai biopharmaceutical company (Array BioPharma), AZD6244 from asrilikang company (AstraZeneca), PD181461 from Pfizer, and leucovorin. As used herein, the term "aromatase inhibitor" relates to inhibiting estrogen productionFor example compounds which convert the substrates androstenedione and testosterone to estrone and estradiol, respectively. The term includes, but is not limited to: steroids, in particular atamestane, exemestane and formestane, and in particular non-steroids, in particular aminoglutethimide, roflumilast, pyridodomide, trilostane, testosterone, ketoconazole, voldazole, fadrozole, anastrozole and letrozole. Exemestane is under the trade name aromas TM And (5) selling. Fumesteine is known under the trade name Lentaron TM And (5) selling. Fadrozole is sold under the trademark Afema TM And (5) selling. Anastrozole is known under the trade name Arimidx TM And (5) selling. Letrozole is under the trade name Femara TM Or Femar TM And (5) selling. Ammonia glutethimide is known by the trade name Orimeten TM And (5) selling. The compositions of the invention comprising a chemotherapeutic agent as an aromatase inhibitor are particularly useful for the treatment of hormone receptor positive tumors, such as breast tumors.
As used herein, the term "antiestrogen" refers to a compound that antagonizes the effects of estrogen at estrogen receptor levels. The term includes, but is not limited to tamoxifen (tamoxifen), fulvestrant (fulvestrant), raloxifene (Raloxifene), and Raloxifene hydrochloride (Raloxifene hydrochloride). Tamoxifen is known under the trade name Nolvadex TM And (5) selling. Raloxifene hydrochloride is known by the trade name Evista TM And (5) selling. Fulvestrant may be under the trade name Faslodex TM And (3) application. The combinations of the invention comprising a chemotherapeutic agent as antioestrogen are particularly useful for the treatment of oestrogen receptor positive tumours, such as breast tumours.
As used herein, the term "anti-androgen" refers to any substance capable of inhibiting the biological effects of androgens and includes, but is not limited to bicalutamide (Casodex TM ). As used herein, the term "gonadotropin-releasing hormone agonist" includes, but is not limited to, abarelix (abarelix), goserelin (goserelin), and goserelin acetate (goserelin acetate). Goserelin can be used as a quotientThe designation Zoladex TM And (3) application.
As used herein, the term "topoisomerase I inhibitor" includes, but is not limited to: topotecan (topotecan), gem Ma Tikang (gimatecan), irinotecan (irinotecan), camptothecin (Camptothecian) and analogs thereof, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148. Irinotecan can be prepared, for example, according to the trademark Camptosar TM Applied in a form of sale. Topotecan is known by the trade name Hycamptin TM And (5) selling.
As used herein, the term "topoisomerase II inhibitor" includes, but is not limited to, anthracyclines, such as doxorubicin (including liposome formulations, such as Caelyx TM ) Daunorubicin (daunorubicin), epirubicin (epirubicin), idarubicin (idarubicin) and nemorubicin (nemorubicin), anthraquinones mitoxantrone (mitoxantrone) and loxoxantrone (loxoxantrone), and polifexolone toxin (podophyllotoxine), etoposide (etoposide) and teniposide (teniposide). Etoposide is under the trade name etoposics TM And (5) selling. Teniposide is sold under the trade name VM 26-Bristol. Doxorubicin is known under the trade name Acriblastin TM Or Adriamycin TM And (5) selling. Epirubicin is known under the trade name Farmorubicin TM And (5) selling. Idarubicin is under the trade name Zavedos TM And (5) selling. Mitoxantrone is sold under the trade name Novantron.
The term "microtubule active agent" relates to microtubule stabilizing compounds, microtubule destabilizing compounds, and tubulin polymerization inhibitors, including but not limited to: taxanes (taxane), such as paclitaxel (paclitaxel) and docetaxel (docetaxel); vinca alkaloids, such as vinblastine or vinblastine sulfate, vincristine or vincristine sulfate and vinorelbine (vinorelbine); discodermolide (discodermolide); colchicine and epothilones and derivatives thereof. Paclitaxel is under the trade name Taxol TM And (5) selling. Docetaxel is known by the trade name Taxote TM And (5) selling. Vinblastine sulfate is sold under the trade name Vinblastin R.P TM And (5) selling. Vincristine sulfate is known by the trade name Farmsin TM And (5) selling.
As used herein, the term "alkylating agent" packageIncluding but not limited to cyclophosphamide, ifosfamide, melphalan (melphalan) or nitrosourea (BCNU or glidel). Cyclophosphamide is known by the trade name cyclophosphamide TM And (5) selling. Ifosfamide is known under the trade name Holoxan TM And (5) selling.
The term "histone deacetylase inhibitor" or "HDAC inhibitor" relates to a compound that inhibits histone deacetylase and has antiproliferative activity. This includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).
The term "anti-tumor antimetabolites" includes, but is not limited to, 5-fluorouracil or 5-FU, capecitabine (Capecitabine), gemcitabine (gemcitabine), DNA demethylating compounds such as 5-azacytidine and decitabine (decubine), methotrexate and edatrazine (edetrexate), and folic acid antagonists such as pemetrexed (pemetrexed). Capecitabine is known under the trade name Xeloda TM And (5) selling. Gemcitabine is known under the trade name Gemzar TM And (5) selling.
As used herein, the term "platinum compound" includes, but is not limited to, carboplatin, cisplatin (cis-platin), cisplatin (cisplatinum), and oxaliplatin (oxaliplatin). Carboplatin may be available, for example, under the trademark Carboplat TM Applied in a form of sale. Oxaliplatin can be used, for example, under the trademark Eloxatin TM Applied in a form of sale.
As used herein, the term "compound that targets/reduces the activity of a protein or lipid kinase" or another anti-angiogenic compound includes, but is not limited to: protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, such as a) compounds that target, reduce or inhibit the activity of, e.g. target, PDGFR, especially PDGF receptor-inhibiting compounds, such as N-phenyl-2-pyrimidine-amine derivatives, e.g. imatinib, SU101, SU6668 and GFB-111; b) Compounds that target, reduce or inhibit the activity of Fibroblast Growth Factor Receptor (FGFR); c) Compounds targeting insulin-like growth factor receptor I (IGF-IR), reducing or inhibiting its activity, e.g. compounds targeting IGF-IR, reducing or inhibiting its activity, especially inhibiting kinase activity of IGF-I receptor A compound, or an antibody targeting the extracellular domain of IGF-I receptor or a growth factor thereof; d) Compounds or inhibitors of ephrin B4 that target, reduce or inhibit the activity of the Trk receptor tyrosine kinase family; e) Compounds that target, reduce or inhibit the activity of the AxI receptor tyrosine kinase family; f) Compounds that target Ret receptor tyrosine kinase, reduce or inhibit its activity; g) Compounds that target, reduce or inhibit the activity of, kit/SCFR receptor tyrosine kinase, such as imatinib; h) A compound that targets, reduces or inhibits the activity of, a C-Kit receptor tyrosine kinase as part of the PDGFR family, such as a compound that targets, reduces or inhibits the activity of, in particular a compound that inhibits the C-Kit receptor, such as imatinib; i) Targeting c-Abl family members, gene fusion products (e.g., BCR-Abl kinase) and mutants thereof, compounds that reduce or inhibit their activity, such as targeting c-Abl family members and gene fusion products thereof, such as N-phenyl-2-pyrimidine-amine derivatives, compounds that reduce or inhibit their activity, such as imatinib or nilotinib (AMN 107); PD180970; AG957; NSC 680410; PD173955 from Parkedavis (Parkedavis); or dasatinib (BMS-354825); j) Protein Kinase C (PKC) and Raf families that target serine/threonine kinases, reduce or inhibit members thereof; MEK, SRC, JAK/pan JAK, FAK, PDK, PKB/Akt, ras/MAPK, PI3K, SYK, BTK and members of the TEC family; and/or a compound comprising an activity of a member of the cyclin dependent kinase family (CDK) of staurosporine derivatives, such as midostaurin; examples of additional compounds include UCN-01, sha Fenge (safingol), BAY 43-9006, bryostatin 1, perfosine; illitofosine (Ilmofosine); RO 318220 and RO 320432; GO 6976; isis 3521; LY333531/LY379196; isoquinoline compounds; FTI; PD184352 or QAN697 (P13K inhibitor) or AT7519 (CDK inhibitor); k) Compounds targeting, reducing or inhibiting the activity of a protein tyrosine kinase inhibitor, e.g. compounds targeting, reducing or inhibiting the activity of a protein tyrosine kinase inhibitor, comprise imatinib mesylate (Gleevec TM ) Or tyrosine phosphorylation inhibitors such as tyrosine phosphorylation inhibitor A23/RG-50810; AG 99; tyrosine phosphorusAn acidification inhibitor AG 213; tyrosine phosphorylation inhibitor AG 1748; tyrosine phosphorylation inhibitor AG 490; tyrosine phosphorylation inhibitor B44; a tyrosine phosphorylation inhibitor B44 (+) enantiomer; tyrosine phosphorylation inhibitor AG 555; AG 494; tyrosine phosphorylation inhibitors AG 556, AG957 and adapalene (4- { [ (2, 5-dihydroxyphenyl) methyl]Amino } -benzoic acid adamantyl ester; NSC 680410, adapalene); l) targeting receptor tyrosine kinases (EGFR as homo-or heterodimers) 1 ErbB2, erbB3, erbB 4) and mutants thereof, compounds that reduce or inhibit their activity, such as compounds that target the epidermal growth factor receptor family, reduce or inhibit their activity, in particular compounds that inhibit EGF receptor tyrosine kinase family members, such as EGF receptors, erbB2, erbB3 and ErbB4 or compounds, proteins or antibodies that bind to EGF or EGF-related ligands, CP 358774, ZD 1839, ZM 105180; trastuzumab (Herceptin) TM ) Cetuximab (Erbitux) TM ) Iressa, tarceva, OSI-774, cl-1033, EKB-569, GW-2016, E1.1, E2.4, E2.5, E6.2, E6.4, E2.11, E6.3, or E7.6.3; 7H-pyrrolo- [2,3-d ]Pyrimidine derivatives; m) a compound that targets, reduces or inhibits the activity of the c-Met receptor, such as a compound that targets, reduces or inhibits the activity of c-Met, in particular a compound that inhibits the kinase activity of the c-Met receptor, or an antibody that targets the extracellular domain of c-Met or binds HGF; n) compounds that target the kinase, decrease or inhibit activity of one or more JAK family members (JAK 1/JAK2/JAK3/TYK2 and/or ubiquity), including but not limited to PRT-062070, SB-1578, barytetratinib (baricitinib), pacetinib (pacritinib), mo Meiluo tinib (momellotinib), VX-509, AZD-1480, TG-101348, tofacitinib (tofacitinib), and robustatinib; o) kinases that target PI3 kinases (PI 3 ks), compounds that reduce or inhibit their activity, including but not limited to ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, bupanib (buparlisib), pick anib (pictreliib), PF-4691502, BYL-719, daponib (dactylisib), XL-147, XL-765, and idelalisib; targeting, lowering or q)Compounds that inhibit signaling of hedgehog (Hh) or smooth receptor (SMO) pathways, including but not limited to cyclopamine, vemod gemi, itraconazole (itraconazole), ibrimode gemib (rimodegab) and IPI-926 (saridedigib).
As used herein, the term "PI3K inhibitor" includes, but is not limited to, compounds having inhibitory activity against one or more enzymes of the phosphatidylinositol-3-kinase family, including, but not limited to, PI3kα, PI3kγ, PI3kδ, PI3kβ, PI3K-c2α, PI3K-c2β, PI3K-c2γ, vps34, p110- α, p110- β, p110- γ, p110- δ, p85- α, p85- β, p55- γ, p150, p101, and p87. Examples of PI3K inhibitors that may be used in the present invention include, but are not limited to: ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, bupanib, pickert-nixie, PF-4691502, BYL-719, daponixie, XL-147, XL-765 and Edalanib.
As used herein, the term "BTK inhibitor" includes, but is not limited to, compounds having inhibitory activity against Bruton's Tyrosine Kinase (BTK), including, but not limited to AVL-292 and ibrutinib.
As used herein, the term "SYK inhibitor" includes, but is not limited to, compounds having inhibitory activity against spleen tyrosine kinase (SYK), including, but not limited to, PRT-062070, R-343, R-333, exesell (Excellair), PRT-062607, and Fostamatinib (Fostamatinib).
As used herein, the term "Bcl-2 inhibitor" includes, but is not limited to, compounds having inhibitory activity against B-cell lymphoma 2 protein (Bcl-2), including, but not limited to ABT-199, ABT-731, ABT-737, apogossypol (apogosypol), pan-Bcl-2 inhibitor of Ai Senda (ascnta), curcumin (and analogs thereof), dual Bcl-2/Bcl-xL inhibitor (infinite pharmaceutical company (Infinity Pharmaceuticals)/nova pharmaceutical company (Novartis Pharmaceuticals)), sodium orlistat (Genasense) (G3139), HA14-1 (and analogs thereof; see WO 2008118802), navitos (navitocrax) (and analogs thereof, see US 7390799), NH-1 (apogossypol (Shenayng Pharmaceutical University)), obatos (obaclax) (and analogs thereof), S-001 (Gloria Pharmaceuticals)), compound of the family of sciences of twos (400), and the family of michausex (uniosc). In some embodiments, the Bcl-2 inhibitor is a small molecule therapeutic. In some embodiments, the Bcl-2 inhibitor is a peptidomimetic.
Further examples of BTK inhibiting compounds and conditions treatable by such compounds in combination with the compounds of the invention can be found in WO 2008039218 and WO 2011090760, the entire contents of which are incorporated herein by reference.
Additional examples of SYK inhibiting compounds and conditions treatable by such compounds in combination with the compounds of the invention can be found in WO 2003063794, WO 2005007623 and WO 2006078846, the entire contents of which are incorporated herein by reference.
Additional examples of PI3K inhibiting compounds and conditions treatable by such compounds in combination with the compounds of the invention may be found in WO 2004019973, WO 2004089925, WO 2007016176, US8138347, WO 2002088112, WO 2007084786, WO 2007129161, WO 2006122806, WO 2005113554 and WO 2007044729, the entire contents of which are incorporated herein by reference.
Further examples of JAK inhibiting compounds and conditions treatable by such compounds in combination with the compounds of the present invention can be found in WO 2009114512, WO 200810009943, WO 2007053452, WO 2000142246 and WO 2007070514, the entire contents of which are incorporated herein by reference.
Additional anti-angiogenic compounds include compounds such as thalidomide (thamate) which have another mechanism for their activity, such as being unrelated to protein or lipid kinase inhibition TM ) And TNP-470.
Examples of proteasome inhibitors that may be used in combination with the compounds of the present invention include, but are not limited to, bortezomib (bortezomib), disulfiram (disufiram), epigallocatechin-3-gallate (EGCG), salidroamide A (salinosporamide A), carfilzomib (Carfilzomib), ONX-0912, CEP-18770, and MLN9708.
The compound that targets, reduces or inhibits the activity of a protein or lipid phosphatase is, for example, an inhibitor of phosphatase 1, phosphatase 2A or CDC25, such as okadaic acid or a derivative thereof.
Compounds that induce the cell differentiation process include, but are not limited to, retinoic acid, alpha-gamma-or delta-tocopherol or alpha-gamma-or delta-tocotrienol.
As used herein, the term cyclooxygenase inhibitor includes, but is not limited to, cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acids and derivatives, such as celecoxib (Celebrex) TM ) Rofecoxib (Vioxx) TM ) Etoricoxib (etoricoxib), valdecoxib (valdecoxib) or 5-alkyl-2-arylaminophenylacetic acid, such as 5-methyl-2- (2 '-chloro-6' -fluoroanilino) phenylacetic acid, lomecoxib (lumiracoxib).
As used herein, the term "bisphosphonate" includes, but is not limited to, etidronic acid (etidronic acid), clodronic acid (Clodronic acid), tiludronic acid (tiludronic acid), pamidronic acid (pamidronic acid), alendronic acid (alendronic acid), ibandronic acid (ibandronic acid), risedronic acid (Risedronic acid), and zoledronic acid (zoledronic acid). Etidronic acid is known under the trade name Didronel TM And (5) selling. Chlorophosphonic acid is known under the trade name Bonefos TM And (5) selling. Teluphosphonic acid is known under the trade name Skelid TM And (5) selling. Pamidronate is known under the trade name Aredia TM And (5) selling. Alendronic acid is known under the trade name Fosamax TM And (5) selling. Ibandronic acid is known under the trade name Bondraat TM And (5) selling. Risedronic acid is known under the trade name Actonel TM And (5) selling. Zoledronic acid is known under the trade name Zometa TM And (5) selling. The term "mTOR inhibitor" relates to compounds that inhibit mammalian target of rapamycin (mTOR) and have antiproliferative activity, such as sirolimus (sirolimus)Everolimus (everolimus) (Certifan) TM ) CCI-779 and ABT578.
As used herein, the term "heparanase inhibitor" refers to a compound that targets, reduces or inhibits the degradation of heparin sulfate. The term includes, but is not limited to, PI-88. As used herein, the term "biological response modifier" refers to a lymphokine or an interferon.
As used herein, the term "inhibitors of Ras oncogenic isoforms" such as H-Ras, K-Ras or N-Ras refers to compounds that target Ras, reduce or inhibit the oncogenic activity of Ras; for example, "farnesyl transferase inhibitors", such as L-744832, DK8G557 or R115777 (Zarnestra) TM ). As used herein, the term "telomerase inhibitor" refers to a compound that targets telomerase, reduces or inhibits its activity. Compounds that target, reduce or inhibit the activity of telomerase, especially compounds that inhibit the telomerase receptor, such as telomerase chalone.
As used herein, the term "methionine aminopeptidase inhibitor" refers to a compound that targets methionine aminopeptidase, reduces or inhibits its activity. Compounds that target methionine aminopeptidase, reduce or inhibit its activity include, but are not limited to Ban Ge amide (bengamide) or derivatives thereof.
As used herein, the term "proteasome inhibitor" refers to a compound that targets, reduces, or inhibits the activity of a proteasome. Compounds that target proteasome, reduce or inhibit its activity include, but are not limited to, bortezomib (Velcade) TM ) And MLN 341.
As used herein, the term "matrix metalloproteinase inhibitor" or ("MMP" inhibitor) includes, but is not limited to: collagen peptidomimetics and non-peptidomimetics inhibitors, tetracycline derivatives, such as the hydroxamic acid peptidomimetic inhibitor, batimastat (bat) and its orally bioavailable analogs, marimastat (BB-2516), priomastat (AG 3340), horse betastat (metastat) (NSC 683551), BMS-279251, BAY 12-9566, TAA211, MMI270B or AAJ996.
As used herein, the term "compound for treating hematological malignancies" includes, but is not limited to: FMS-like tyrosine kinase inhibitors, which are compounds that target, reduce or inhibit the activity of FMS-like tyrosine kinase receptor (Flt-3R); interferon, 1-beta-D-arabinofuranosyl cytosine (ara-c) and thionyl chloride (bisulfan); ALK inhibitors, which are compounds that target, reduce or inhibit anaplastic lymphoma kinase and Bcl-2 inhibitors.
Compounds that target, reduce or inhibit the activity of, FMS-like tyrosine kinase receptor (Flt-3R), in particular compounds, proteins or antibodies that inhibit members of the Flt-3R receptor kinase family, such as PKC412, midostaurin, staurosporine derivatives, SU11248 and MLN518.
As used herein, the term "HSP90 inhibitor" includes, but is not limited to, compounds that target, reduce, or inhibit the intrinsic atpase activity of HSP 90; compounds that degrade, target, reduce or inhibit HSP90 client proteins via the ubiquitin proteasome pathway. Compounds that target HSP90, reduce or inhibit its intrinsic atpase activity, in particular compounds, proteins or antibodies that inhibit the atpase activity of HSP90, such as 17-allylamino, 17-desmethoxygeldanamycin (17 AAG), geldanamycin derivatives; other geldanamycin related compounds; radicicol and HDAC inhibitors.
As used herein, the term "anti-proliferative antibody" includes, but is not limited to, trastuzumab (Herceptin TM ) Trastuzumab DM1, erbitux, bevacizumab (Avastin) TM ) RituximabPRO64553 (anti-CD 40) and 2C4 antibodies. Antibodies refer to intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
For the treatment of Acute Myeloid Leukemia (AML), the compounds of the invention can be used in combination with standard leukemia therapies, in particular with therapies for the treatment of AML. In particular, the compounds of the invention may be administered in combination with, for example, farnesyl transferase inhibitors and/or other drugs useful in the treatment of AML such as daunomycin, adriamycin, ara-C, VP-16, teniposide, mitoxantrone, idarubicin, carboplatin (carboplatin) and PKC 412. In some embodiments, the invention provides a method of treating AML associated with ITD and/or D835Y mutations comprising administering a compound of the invention with one or more FLT3 inhibitors. In some embodiments, the FLT3 inhibitor is selected from the group consisting of quinotidinib (AC 220), staurosporine derivatives (e.g., midostaurin or letatinib), sorafenib (sorafenib), tandutinib (tandutinib), LY-2401401, LS-104, EB-10, famitinib (famitinib), NOV-110302, NMS-P948, AST-487, G-749, SB-1317, S-209, SC-110219, AKN-028, non-dastinib (feldtinib), cerzasertib (tozasertib), and sunitinib (sunitinib). In some embodiments, the FLT3 inhibitor is selected from the group consisting of olaquindox, midostaurin, letatinib, sorafenib, and sunitinib.
Other anti-leukemia compounds include, for example, the pyrimidine analog Ara-C, which is a 2' -alpha-hydroxy ribose (arabinoside) derivative of deoxycytidine. Purine analogs of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine phosphate are also included. Targeting Histone Deacetylase (HDAC) inhibitors, compounds that reduce or inhibit their activity, such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA), inhibit the activity of enzymes known as histone deacetylases. Specific HDAC inhibitors include compounds disclosed in MS275, SAHA, FK228 (original name FR 901228), trichostatin a and US 6,552,065, including but not limited to N-hydroxy-3- [4- [ [ [2- (2-methyl-1H-indol-3-yl) -ethyl ] -amino ] methyl ] phenyl ] -2E-2-acrylamide or a pharmaceutically acceptable salt thereof and N-hydroxy-3- [4- [ (2-hydroxyethyl) {2- (1H-indol-3-yl) ethyl ] -amino ] methyl ] phenyl ] -2E-2-acrylamide or a pharmaceutically acceptable salt thereof, especially a lactate salt. As used herein, a somatostatin receptor antagonist refers to a compound that targets, treats, or inhibits the somatostatin receptor, such as octreotide (octreotide) and SOM230. Tumor cell damage methods refer to methods such as ionizing radiation. The term "ionizing radiation" mentioned above and below means ionizing radiation generated as electromagnetic rays (such as X-rays and gamma rays) or particles (such as alpha and beta particles). Ionizing radiation is provided in radiation therapy, but is not limited thereto, and is known in the art. See Hellman, principle of radiation therapy (Principles of Radiation Therapy), cancer: oncology principles and practices (Cancer, in Principles and Practice of Oncology), devita (Devita) et al, editors, 4 th edition, volume 1, pages 248-275 (1993).
Also included are EDG binders and ribonucleotide reductase inhibitors. As used herein, the term "EDG binder" refers to a class of immunosuppressants that regulate lymphocyte recirculation, such as FTY720. The term "ribonucleotide reductase inhibitor" refers to pyrimidine or purine nucleoside analogs, including but not limited to: fludarabine (fludarabine) and/or cytosine arabinoside (ara-C), 6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine (especially for ALL in combination with ara-C) and/or pentastatin. Ribonucleotide reductase inhibitors are in particular hydroxyurea or 2-hydroxy-1H-isoindole-1, 3-dione derivatives.
Also specifically those compounds, proteins or monoclonal antibodies comprising VEGF, such as 1- (4-chloroanilino) -4- (4-pyridylmethyl) phthalazine or a pharmaceutically acceptable salt thereof, 1- (4-chloroanilino) -4- (4-pyridylmethyl) phthalazine succinate; angiostatin TM ;Endostatin TM The method comprises the steps of carrying out a first treatment on the surface of the Anthranilic acid amides; ZD4190; ZD6474; SU5416; SU6668; bevacizumab; or anti-VEGF antibodies or anti-VEGF receptor antibodies, such as rhuMAb and RHIFAB, VEGF aptamers such as Makeup (Macugon); FLT-4 inhibitor, FLT-3 inhibitor, VEGFR-2IgGI antibody, vascular proliferation ribozyme (Angiozyme) (RPI 4610) and bevacizumab (Avastin) TM )。
Photodynamic therapy, as used herein, refers to therapy that uses certain chemicals known as photoactive compounds to treat or prevent cancer. Examples of photodynamic therapy include the use of e.g. Visudyne TM And porphin sodium (porfimer sodium).
As used herein, angiogenesis inhibiting steroids refer to compounds that block or inhibit angiogenesis such as anecortave, triamcinolone, hydrocortisone, 11-alpha-epihydrocortisone, deoxycortisol (Cortexolone), 17 alpha-hydroxyprogesterone, corticosterone, deoxycorticosterone, testosterone, estrone, and dexamethasone.
Implants containing corticosteroids refer to compounds such as fluocinolone acetonide and dexamethasone.
Other chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds, and antagonists; a biological response modifier, preferably a lymphokine or an interferon; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or other compounds or compounds having other or unknown mechanisms of action.
The compounds of the invention may also be used as co-therapeutic compounds for use in combination with other drugs such as anti-inflammatory, bronchodilatory or antihistamine drugs, especially for the treatment of obstructive or inflammatory airways diseases as referred to hereinbefore, e.g. as therapeutic activity potentiators of such drugs or as a method of reducing required administration or potential side effects of such drugs. The compounds of the present invention may be mixed with other drugs in the form of a fixed pharmaceutical composition or they may be administered separately before, simultaneously with or after the other drugs. Thus, the present invention comprises a compound of the present invention as described above in combination with an anti-inflammatory, bronchodilatory, antihistamine or anti-tussive drug substance, said compound of the present invention and said drug substance being in the same or different pharmaceutical compositions.
Suitable anti-inflammatory agents include steroids, particularly glucocorticosteroids such as budesonide, beclomethasone dipropionate, fluticasone propionate (fluticasone propionate), ciclesonide (ciclesonide) or mometasone furoate (mometasone furoate); a non-steroidal glucocorticoid receptor agonist; LTB4 antagonists such as LY293111, CGS025019C, CP-195543, SC-53228, BIIL 284, ONO 4057, SB 209247; LTD4 antagonists such as montelukast (montelukast) and zafirlukast (zafirlukast); PDE4 inhibitors, e.g. cilomilastGelanin SmithKline, roflumilast, byk Gulden, V-11294A, BAY19-8004, bayer, SCH-351591-young company (Schering-Plough)), arofylline (Arofylline) (eimeric medical drug company (Almirall Prodesfarma)), PD189659/PD168787 (parkdavi company (Parke-Davis)), AWD-12-281 (Asta medical), CDC-801 (Celgene)), seiid (TM) CC-10004 (neo-base), VM554/UM565 (Vernalis), T-440 (Tanabe), KW-4490 (co-fermentation industry company (Kyowa Hakko Kogyo)); a2a agonist; a2b antagonist; and beta-2 adrenoceptor agonists such as salbutamol (oxymethylene-t-epinephrine), oxacinal, terbutaline, salmeterol, fenoterol (fenoterol), procaterol (procaterol) and especially formoterol and pharmaceutically acceptable salts thereof. Suitable bronchodilators include anticholinergic or antimuscarinic compounds, in particular ipratropium bromide, oxitropium bromide, tiotropium salts and CHF 4226 (Chiesi) and glycopyrrolate.
Suitable antihistamines include cetirizine hydrochloride (Cetirizine hydrochloride), acetaminophen, chloromattin fumarate (Clemastine fumarate), promethazine, loratadine (loratidine), desloratadine (desloratidine), diphenhydramine (diphenhydramine) and fexofenadine hydrochloride (fexofenadine), atorvastatin (activine), astemizole (astemizole), azelastine (azelastine), ebastine (ebastine), epinastine (epinastine), mizolastine (mizostatin) and tefalatine (tefenadine).
Other useful combinations of the compounds of the invention with anti-inflammatory agents are in combination with, for example, antagonists of the following chemokine receptors: CCR-1, CCR-2, CCR-3, CCR-4, CCR-5, CCR-6, CCR-7, CCR-8, CCR-9 and CCR10, CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, particularly CCR-5 antagonists such as the first-come-young (Schering-Plough) antagonists SC-351125, SCH-55700 and SCH-D and the Wuta-Co (Takeda) antagonists such as N- [ [4- [ [ [6, 7-dihydro-2- (4-methylphenyl) -5H-benzo-cyclohepten-8-yl ] carbonyl ] amino ] phenyl ] -methyl ] tetrahydro-N, N-dimethyl-2H-pyran-4-ammonium chloride (TAK-770).
The structure of The active compound identified by code, common or trade name may be taken from The actual version or database of The standard schema "Merck Index", such as international patent (Patents International) (e.g., IMS world publication (IMS World Publications)).
Exemplary Immunotorhinocerology agents
In some embodiments, the one or more other therapeutic agents are immunooncology agents. As used herein, the term "immunooncology agent" refers to an agent effective to enhance, stimulate, and/or up-regulate an immune response in a subject. In some embodiments, administration of an immunooncology agent with a compound of the present invention has a synergistic effect in treating cancer.
The immunooncology agent may be, for example, a small molecule drug, an antibody or a biological molecule or a small molecule. Examples of biological immunooncology agents include, but are not limited to, cancer vaccines, antibodies, and cytokines. In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the monoclonal antibody is humanized or human.
In some embodiments, the immunooncology agent is (i) an agonist of a stimulatory (including co-stimulatory) receptor or (ii) an antagonist of an inhibitory (including co-inhibitory) signal on a T cell, both of which result in an expanded antigen-specific T cell response.
Certain stimulatory and inhibitory molecules are members of the immunoglobulin superfamily (IgSF). An important family of membrane-bound ligands that bind to co-stimulatory or co-inhibitory receptors is the B7 family, which includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6. Another family of membrane-bound ligands that bind to co-stimulatory or co-inhibitory receptors are TNF family molecules that bind to members of the cognate TNF receptor family, including CD40 and CD40L, OX-40, OX-40L, CD, CD27L, CD, CD30L, 4-1BBL, CD137 (4-1 BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LT beta R, LIGHT, dcR3, HVEM, VEGI/TL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFR1, lymphotoxin alpha/TNF beta, TNFR2, TNFalpha, LT beta R, lymphotoxin alpha 1 beta 2, FAS, FASL, RELT, DR, TROY, NGFR.
In some embodiments, the immunooncology agent is a cytokine that inhibits T cell activation (e.g., IL-6, IL-10, TGF- β, VEGF, and other immunosuppressive cytokines) or a cytokine that stimulates T cell activation for stimulating an immune response.
In some embodiments, the combination of a compound of the invention and an immunooncology agent may stimulate a T cell response. In some embodiments, the immunooncology agent is: (i) Antagonists of proteins that inhibit T cell activation (e.g., immune checkpoint inhibitors), such as CTLA-4, PD-1, PD-L2, LAG-3, TIM-3, galectin 9, CEACAM-1, BTLA, CD69, galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1 and TIM-4; or (ii) agonists of proteins that stimulate T cell activation, such as B7-1, B7-2, CD28, 4-1BB (CD 137), 4-1BBL, ICOS, ICOS-L, OX, OX40L, GITR, GITRL, CD70, CD27, CD40, DR3, and CD28H.
In some embodiments, the immunooncology agent is an antagonist of an inhibitory receptor on NK cells or an agonist of an activating receptor on NK cells. In some embodiments, the immunooncology agent is an antagonist of KIR, such as Li Ruilu mab (lirilumab).
In some embodiments, the immunooncology agent is an agent that inhibits or depletes macrophages or monocytes, including but not limited to CSF-1R antagonists, such as CSF-1R antagonist antibodies, including RG7155 (WO 11/70024, WO11/107553, WO11/131407, WO13/87699, WO13/119716, WO 13/132044) or FPA-008 (WO 11/140249; WO13169264; WO 14/036357).
In some embodiments, the immunooncology agent is selected from agonists that bind to positive co-stimulatory receptors, blockers, antagonists and one or more agents that systematically increase the frequency of anti-tumor T cells by inhibiting receptor signaling, agents that overcome different immunosuppressive pathways within the tumor microenvironment (e.g., blocking inhibition of receptor engagement (e.g., PD-L1/PD-1 interactions), depleting or inhibiting Treg (e.g., using anti-CD 25 monoclonal antibodies (e.g., daclizumab)) or by ex vivo anti-CD 25 bead depletion), inhibit metabolic enzymes such as IDO, or reverse/prevent T cell energy or depletion), and agents that trigger innate immune activation and/or inflammation at the tumor site.
In some embodiments, the immunooncology agent is a CTLA-4 antagonist. In some embodiments, the CTLA-4 antagonist is an antagonistic CTLA-4 antibody. In some embodiments, the antagonistic CTLA-4 antibody is YERVOY (ipilimumab) or tremelimumab (tremelimumab).
In some embodiments, the immunooncology agent is a PD-1 antagonist. In some embodiments, the PD-1 antagonist is administered by infusion. In some embodiments, the immunooncology agent is an antibody or antigen-binding portion thereof that specifically binds to a programmed death-1 (PD-1) receptor and inhibits PD-1 activity. In some embodiments, the PD-1 antagonist is an antagonistic PD-1 antibody. In some embodiments, the antagonistic PD-1 antibody is OPDIVO (nivolumab), keyruda (pembrolizumab), or MEDI-0680 (AMP-514; wo 2012/145493). In some embodiments, the immunooncology agent may be Pidilizumab (CT-011). In some embodiments, the immunooncology agent is a recombinant protein consisting of the extracellular domain of PD-L2 (B7-DC), referred to as AMP-224, fused to the Fc portion of IgG 1.
In some embodiments, the immunooncology agent is a PD-L1 antagonist. In some embodiments, the PD-L1 antagonist is an antagonistic PD-L1 antibody. In some embodiments, the PD-L1 antibody is MPDL3280A (RG 7446; WO 2010/077634), dewaruzumab (durvalumab) (MEDI 4736), BMS-936559 (WO 2007/005874) and MSB0010718C (WO 2013/79174).
In some embodiments, the immunooncology agent is a LAG-3 antagonist. In some embodiments, the LAG-3 antagonist is an antagonistic LAG-3 antibody. In some embodiments, the LAG3 antibody is BMS-986016 (WO 10/19570, WO 14/08218), or IMP-731 or IMP-321 (WO 08/132601, WO 009/44273).
In some embodiments, the immunooncology agent is a CD137 (4-1 BB) agonist. In some embodiments, the CD137 (4-1 BB) agonist is an agonistic CD137 antibody. In some embodiments, the CD137 antibody is Wu Ruilu mab (urelumab) or PF-05082566 (WO 12/32433).
In some embodiments, the immunooncology agent is a GITR agonist. In some embodiments, the GITR agonist is an agonistic GITR antibody. In some embodiments, the GITR antibody is BMS-986153, BMS-986156, TRX-518 (WO 006/105021, WO 009/009116) or MK-4166 (WO 11/028683).
In some embodiments, the immunooncology agent is an indoleamine (2, 3) -dioxygenase (IDO) antagonist. In some embodiments, the IDO antagonist is selected from Ai Kaduo stat (epacoadostat) (INCB 024360, incytte); indoximod (NLG-8189, tourmaline Gene Co., ltd. (NewLink Genetics Corporation)); capetinib (capmannitib) (INC 280, novartis); GDC-0919 (Genntech/Roche); PF-06840003 (pyroxene); BMS F001287 (Bristol-Myers Squibb, bai Zhi Mei Gui Bao Co.); phy906/KD108 (Huider pharmaceutical Co.); an enzyme that breaks down kynurenine (kinase, ikena oncology, previously known as Kyn pharmaceutical corporation); NLG-919 (WO 09/73620, WO009/1156652, WO11/56652, WO 12/142237).
In some embodiments, the immunooncology agent is an OX40 agonist. In some embodiments, the OX40 agonist is an agonistic OX40 antibody. In some embodiments, the OX40 antibody is MEDI-6383 or MEDI-6469.
In some embodiments, the immunooncology agent is an OX40L antagonist. In some embodiments, the OX40L antagonist is an antagonistic OX40 antibody. In some embodiments, the OX40L antagonist is RG-7888 (WO 06/029879).
In some embodiments, the immunooncology agent is a CD40 agonist. In some embodiments, the CD40 agonist is an agonistic CD40 antibody. In some embodiments, the immunooncology agent is a CD40 antagonist. In some embodiments, the CD40 antagonist is an antagonistic CD40 antibody. In some embodiments, the CD40 antibody is Lu Kamu mab (lucatumumab) or dactyluzumab (daceatuzumab).
In some embodiments, the immunooncology agent is a CD27 agonist. In some embodiments, the CD27 agonist is an agonistic CD27 antibody. In some embodiments, the CD27 antibody is varlimumab (varlimumab).
In some embodiments, the immunooncology agent is MGA271 (for B7H 3) (WO 11/109400).
In some embodiments, the immunooncology agent is Ab Fu Shan antibody (abago mab), adelomab (adecatumab), alfutuzumab (afutuzumab), alemtuzumab (alemtuzumab), ting An Moshan antibody (anatumomab mafenatox), apolizumab (apolizumab), ati Li Shan antibody (atezolizumab), abauzumab (avelumab), boluzumab (blinatumomab), BMS-936559, katuzumab (cataxomab), devaluzumab, ai Kaduo stavumab, epatuzumab (epratuzumab), indomadod, oxuzumab (inotuzumab ozogamicin), irituzumab (i-tuzumab), i-Sha Tuo mab (isauximab), lanlizumab (labzolizumab), MED14736, L3280, oxuzumab (oxytuzumab), oxuzumab (oxypuzumab), or oxytuzumab (oxypuzumab), oxytuzumab (37) or oxytuzumab (oxypuzumab).
In some embodiments, the immunooncology agent is an immunostimulatory agent. For example, antibodies blocking the PD-1 and PD-L1 inhibition axes may release activated tumor-reactive T cells and have been shown in clinical trials to induce a sustained anti-tumor response in increasing tumor histology, including some tumor types that are not normally considered immunotherapy-sensitive. See, e.g., okazaki, t.et al, (2013) [ natural immunology (nat. Immunol.) ] 14,1212-1218; zou et al, (2016) science of sci.Transl.Med.) 8. anti-PD-1 antibody NawuzumabBai-Shi Mitsu Guibao, also known as ONO-4538, MDX1106, and BMS-936558) has shown to improve during or after previous anti-angiogenic therapiesPotential for overall survival of RCC patients undergoing disease progression.
In some embodiments, the immunomodulatory therapeutic specifically induces apoptosis of tumor cells. Approved immunomodulatory therapeutic agents that may be used in the present invention include pomalidomideNew base company); lenalidomide (+)>New base company); ingenol mebutate (jol mebutate)>LEO pharmaceutical company (LEO Pharma)).
In some embodiments, the immunooncology agent is a cancer vaccine. In some embodiments, the cancer vaccine is selected from the group consisting of Cepuliucel T (sipuleucel-T)Dandelion (Dendreon)/Wilang pharmaceutical company (Valeant Pharmaceuticals)), which has been approved for the treatment of asymptomatic or minimally symptomatic metastatic castration-resistant (hormone refractory) prostate cancer; hetai Li Moji Lahcapapray (talimogene laherparepvec)Weak BioVex/Advance (Amgen), previously known as T-VEC), is a genetically modified oncolytic virus therapy approved for the treatment of unresectable skin, subcutaneous and lymphadenopathy in melanoma. In some embodiments, the immunooncology agent is selected from oncolytic virus therapies such as pexavew (pexastimogene devacirepvec) (Pexavec/JX-594, silaljen Inc. (Silaljen)/under the name Jennerex biomedical Co., ltd. (Jennerex Biotherapeutics)), a thymidine kinase- (TK-) deficient vaccinia virus engineered to express GM-CSF for hepatocellular carcinoma (NCT 02562755) and melanoma (NCT 004)29312 A) is provided; pealarorep (pelaroep) (-jerusalem)>Oncolytic biotechnology company (Oncolytics Biotech)), a variant of respiratory enteroorphan virus (reovirus) that does not replicate in non-RAS activated cells, for many cancers, including colorectal cancer (NCT 01622543); prostate cancer (NCT 01619813); squamous cell carcinoma of head and neck (NCT 01166542); pancreatic cancer (NCT 00998322); and non-small cell lung cancer (NSCLC) (NCT 00861627); enadienotoucirev (NG-348, pi Si oex, formerly ColoAd 1), an adenovirus engineered to express full-length CD80 and an antibody fragment specific for the T cell receptor CD3 protein, for ovarian cancer (NCT 02028117); metastatic or advanced epithelial tumors, such as colorectal cancer, bladder cancer, head and neck squamous cell carcinoma, and salivary gland carcinoma (NCT 02636036); ONCOS-102 (Targovix/Oncos), an adenovirus engineered to express GM-CSF for melanoma (NCT 03003676); peritoneal disease, colorectal or ovarian cancer (NCT 02963831); GL-ONC1 (GLV-1 h68/GLV-1h153, ginesia Co., ltd. (Genlux GmbH)), a vaccinia virus engineered to express beta-galactosidase (beta-gal)/beta-glucuronidase or beta-gal/human sodium iodide symporter (hNIS), respectively, in peritoneal metastasis (NCT 01443260); fallopian tube cancer, ovarian cancer (NCT 02759588); or CG0070 (Cold Genesys), an adenovirus engineered to express GM-CSF for bladder cancer (NCT 02365818).
In some embodiments, the immunooncology agent is selected from JX-929 (SillaJen/biological pharmaceutical Co., ltd./under the name Jennerex), which is a TK-and vaccinia growth factor-deficient vaccinia virus engineered to express cytosine deaminase, capable of converting the prodrug 5-fluorocytosine to the cytotoxic drug 5-fluorouracil; TG01 and TG02 (tarwox/original name of ondx), which are peptide-based immunotherapeutic agents targeting refractory RAS mutations; and TILT-123 (TILT Bio-pharmaceutical Co., ltd. (TILT Biotherapeutics),it is an engineered adenovirus, named: ad5/3-E2F-delta24-hTNFα -IRES-hIL20; and VSV-GP (Vira therapeutics), a Vesicular Stomatitis Virus (VSV) engineered to express Glycoprotein (GP) of lymphocytic choriomeningitis virus (LCMV), which can be further engineered to express CD8 designed to increase antigen specificity + Antigen of T cell response.
In some embodiments, the immunooncology agent is a T cell engineered to express a chimeric antigen receptor or CAR. T cells engineered to express such chimeric antigen receptors are referred to as CAR-T cells.
CARs have been constructed that consist of a binding domain that can be derived from a natural ligand, a single chain variable fragment (scFv) derived from a monoclonal antibody specific for a cell surface antigen, fused to an internal domain that is the functional end of a T Cell Receptor (TCR), such as the CD 3-zeta signaling domain from a TCR that can generate an activation signal in T lymphocytes. Upon antigen binding, such CARs attach to endogenous signaling pathways in effector cells and produce an activation signal similar to that initiated by the TCR complex.
For example, in some embodiments, the CAR-T cell is one of the cells described in U.S. Pat. No. 8,906,682 (June et al; incorporated herein by reference in its entirety), which discloses a CAR-T cell engineered to include an extracellular domain having an antigen binding domain (e.g., a domain that binds to the domain of CD 19), an intracellular signaling domain fused to the zeta chain of the T cell antigen receptor complex (e.g., CD3 zeta). When expressed in T cells, CARs are able to redirect antigen recognition based on antigen binding specificity. In the case of CD19, the antigen is expressed on malignant B cells. More than 200 clinical trials are currently underway, using CAR-T for various indications.
[https://clinicaltrials.gov/ct2/resultsterm=chimeric+antigen+receptors&pg=1]。
In some embodiments, the immunostimulatory agent is an activator of retinoic acid receptor-related orphan receptor gamma (rorγt). Roryt is a transcription factor that plays a key role in the differentiation and maintenance of type 17 effector subsets of cd4+ (Th 17) and cd8+ (Tc 17) T cells and in the differentiation of IL-17 expressing innate immune cell subsets such as NK cells. In some embodiments, the activator of roryt is LYC-55716 (lycra corporation (lycra)) which is currently in the clinical trial evaluation of the treatment of solid tumors (NCT 02929862).
In some embodiments, the immunostimulatory agent is an agonist or activator of a toll-like receptor (TLR). Suitable activators of TLRs include agonists or activators of TLR9, such as SD-101 (Dynavex). SD-101 is an immunostimulatory CpG that is being studied against B cells, follicles, and other lymphomas (NCT 02254772). Agonists or activators of TLR8 that can be used in the present invention include Mo Tuo mod (VTX-2337, ventirx pharmaceutical company (VentiRx Pharmaceuticals)), which is being studied for head and neck squamous cell carcinoma (NCT 02124850) and ovarian cancer (NCT 02431559).
Other immunological oncology agents that may be used in the present invention include Wu Ruilu mab (BMS-663513, BAIMEISHIGULAO Co.), an anti-CD 137 monoclonal antibody; warfarin (CDX-1127, celldex treatment company (Celldex Therapeutics)), an anti-CD 27 monoclonal antibody; BMS-986178 (Bai Shi Gui Bao Co.), an anti-OX 40 monoclonal antibody; li Ruilu mab (lirilumab) (IPH 2102/BMS-986015, company infnate Pharma, BAIMEISHIGULAR), an anti-KIR monoclonal antibody; mo Nali bead mab (IPH 2201, company as a specialty drug, company as aslicon), an anti-NKG 2A monoclonal antibody; anlixiviamab (GS-5745, gilead Sciences), an anti-MMP 9 antibody; MK-4166 (Merck & Co.)) an anti-GITR monoclonal antibody.
In some embodiments, the immunostimulatory agent is selected from the group consisting of eltuzumab (eltuzumab), mifamurtide (mifamurtide), an agonist or activator of toll-like receptor, or an activator of roryt.
In some embodiments, the immunostimulatory therapeutic agent is recombinant human interleukin 15 (rhIL-15). rhIL-15 has been tested clinically as a therapy for melanoma and renal cell carcinoma (NCT 01021059 and NCT 01369888) and leukemia (NCT 02689453). In some embodiments, the immunostimulatory agent is recombinant human interleukin 12 (rhIL-12). In some embodiments, the IL-15 based immunotherapeutic agent is heterodimeric IL-15 (hetIL-15, nova/oldham (Admune)), a fusion complex consisting of a synthetic form of endogenous IL-15 complexed to the soluble IL-15 binding protein IL-15 receptor alpha chain (IL 15: sIL-15 RA), which has been tested in phase 1 clinical trials for melanoma, renal cell carcinoma, non-small cell lung carcinoma, and head and neck squamous cell carcinoma (NCT 02452268). In some embodiments, recombinant human interleukin 12 (rhIL-12) is NM-IL-12 (neugelicines, inc.), NCT02544724, or NCT02542124.
In some embodiments, the immunooncology agent is selected from the group consisting of the immunooncology agents described in Jerry l.adams et al, "major opportunity for small molecules in immunooncology (Big opportunities for small molecules in immuno-oncology)," Cancer Therapy (2015, volume 14, pages 603-622, the contents of which are incorporated herein by reference in their entirety. In some embodiments, the immunooncology agent is selected from the examples described in table 1 of Jerry l.adams et al. In some embodiments, the immunooncology agent is a small molecule that targets an immunooncology target selected from those listed in table 2 of Jerry l.adams et al. In some embodiments, the immunooncology agent is a small molecule agent selected from those listed in table 2 of Jerry l.adams et al.
In some embodiments, the immunooncology agent is selected from Peter l.touogood, "small molecule immunooncology therapeutic (Small molecule immuno-oncology therapeutic agents)", bioorganic & pharmaceutical chemistry rapid (Bioorganic & Medicinal Chemistry Letters) 2018, volume 28, pages 319-329, the contents of which are incorporated herein by reference in their entirety. In some embodiments, the immunooncology agent is an agent that targets a pathway as described in Peter l.
In some embodiments, the immunooncology agent is selected from the group consisting of Sandra L.Ross et al, "bispecific T cell adaptorsAntibody constructs can mediate bystander tumor cell killing (Bispecific T cell engager)antibody constructs can mediate bystander tumor cell killing) ", public science library: complex (PLoS ONE) 12 (8): e0183390, the contents of which are incorporated herein by reference in their entirety. In some embodiments, the immunooncology agent is the bispecific T cell adapter +.>Antibody constructs. In some embodiments, bispecific T cell adaptors +.>The antibody construct is a CD19/CD3 bispecific antibody construct. In some embodiments, bispecific T cell adaptors +. >The antibody construct is an EGFR/CD3 bispecific antibody construct. In some embodiments, bispecific T cell adaptors +.>The antibody construct activates T cells. In some embodiments, bispecific T cell adaptors +.>The antibody construct activates T cells, which release cytokines that induce up-regulation of intercellular adhesion molecule 1 (ICAM-1) and FAS on bystander cells. In some embodiments, bispecific T cell adaptors +.>The antibody construct activates T cells, causing induced bystander cell lysis. In some embodiments, the bystander cells are in a solid tumor. In some embodiments, the lysed bystander cells are close to + ->Activated T cells. In some embodiments, the bystander cells comprise tumor-associated antigen (TAA) negative cancer cells. In some embodiments, the bystander cells comprise EGFR-negative cancer cells. In some embodiments, the immunooncology agent is an antibody that blocks the PD-L1/PD1 axis and/or CTLA 4. In some embodiments, the immunooncology agent is ex vivo expanded tumor-infiltrating T cells. In some embodiments, the immunooncology agent is a bispecific antibody construct or Chimeric Antigen Receptor (CAR) that directly links T cells to a tumor-associated surface antigen (TAA).
Exemplary immune checkpoint inhibitors
In some embodiments, the immunooncology agent is an immune checkpoint inhibitor as described herein.
As used herein, the term "checkpoint inhibitor" relates to an agent that can be used to prevent cancer cells from avoiding the immune system of a patient. One of the main mechanisms of anti-tumor immune destruction is called "T cell depletion", which is caused by prolonged exposure to antigens that cause upregulation of inhibitory receptors. These inhibitory receptors act as immune checkpoints in order to prevent uncontrolled immune responses.
PD-1 and co-inhibitory receptors such as cytotoxic T lymphocyte antigen 4 (CTLA-4, B and T lymphocyte attenuation factors (BTLA; CD 272), T cell immunoglobulin and mucin domain-3 (Tim-3), lymphocyte activating gene-3 (Lag-3; CD 223) and the like are commonly referred to as checkpoint mediators.
In some embodiments, the immune checkpoint inhibitor is an antibody to PD-1. PD-1 binds to the programmed cell death 1 receptor (PD-1) to prevent binding of the receptor to the inhibitory ligand PDL-1, thereby exceeding the ability of the tumor to suppress the host's anti-tumor immune response.
In some embodiments, the checkpoint inhibitor is a biologic therapeutic or a small molecule. In some embodiments, the checkpoint inhibitor is a monoclonal antibody, a humanized antibody, a fully human antibody, a fusion protein, or a combination thereof. In some embodiments, the checkpoint inhibitor inhibits a checkpoint protein selected from CTLA-4, PDL2, PDL, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligand, or a combination thereof. In some embodiments, the checkpoint inhibitor interacts with a ligand selected from the group consisting of CTLA-4, PDLl, PDL2, PDl, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligand, or a combination thereof. In some embodiments, the checkpoint inhibitor is an immunostimulatory agent, a T cell growth factor, an interleukin, an antibody, a vaccine, or a combination thereof. In some embodiments, the interleukin is IL-7 or IL-15. In some embodiments, the interleukin is glycosylated IL-7. In another aspect, the vaccine is a Dendritic Cell (DC) vaccine.
Checkpoint inhibitors comprise any agent that blocks or inhibits the inhibitory pathway of the immune system in a statistically significant manner. Such inhibitors may comprise small molecule inhibitors or may comprise antibodies or antigen binding fragments thereof that bind to and block or inhibit immune checkpoint receptors or antibodies that bind to and block or inhibit immune checkpoint receptor ligands. Illustrative checkpoint molecules that may be directed to blocking or inhibition include, but are not limited to: CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, GAL, LAG3, TIM3, VISTA, KIR, 2B4 (belonging to the CD2 family of molecules and memorizing CD8 in all NK, γδ and memory) + (αβ) T cells), CD160 (also known as BY 55), CGEN-15049, CHK1 and CHK2 kinases, A2aR and various B-7 family ligands. The B7 family ligands include, but are not limited to, B7-1, B7-2, B7-DC, B7-H1, B7-H2, B7-H3, B7-H4, B7-H5,B7-H6 and B7-H7. Checkpoint inhibitors comprise antibodies or antigen binding fragments thereof, other binding proteins, biotherapeutic agents or small molecules that bind to and block or inhibit the activity of one or more of the following: CTLA-4, PDL1, PDL2, PD1, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160 and CGEN-15049. Illustrative immune checkpoint inhibitors include, but are not limited to, trimeumab (CTLA-4 blocking antibody), anti-OX 40, PD-Ll monoclonal antibody (anti-B7-Hl; MEDI 4736), MK-3475 (PD-1 blocking agent), nivolumab (anti-PDl antibody), CT-011 (anti-PDl antibody), BY55 monoclonal antibody, AMP224 (anti-pdl antibody), BMS-936559 (anti-pdl antibody), MPLDL3280A (anti-pdl antibody), MSB0010718C (anti-pdl antibody), and ipilimumab (anti-CTLA-4 checkpoint inhibitor). Checkpoint protein ligands include, but are not limited to, PD-Ll, PD-L2, B7-H3, B7-H4, CD28, CD86, and TIM-3.
In certain embodiments, the immune checkpoint inhibitor is selected from the group consisting of a PD-1 antagonist, a PD-L1 antagonist, and a CTLA-4 antagonist. In some embodiments, the checkpoint inhibitor is selected from the group consisting of: nawu monoclonal antibody Ipimab->And pembrolizumab->In some embodiments, the checkpoint inhibitor is selected from the group consisting of nivolumab (anti-PD-1 antibody,/-)>Bai Shi Guibao Co., ltd.); pembrolizumab (anti-PD-1 antibody,merck corporation); ipilimumab (anti-CTLA-4 antibody,>bai Shi Mei Gui Bao GongA department); dewaruzumab (anti-PD-L1 antibody,>african company); alemtuzumab (atezolizumab) (anti-PD-L1 antibody,/-for example>Gene tek).
In some embodiments, the checkpoint inhibitor is selected from the group consisting of: langmuir antibody (MK-3475), nafimbrizumab (BMS-936558), pirimizumab (CT-011), AMP-224, MDX-1105, MEDI4736, MPDL3280A, BMS-936559, ipidizumab, li Lishan antibody (lirlumab), IPH2101, pammizumabAnd trimeumab.
In some embodiments, the immune checkpoint inhibitor is REGN2810 (regen), which is an anti-PD-1 antibody tested in patients with: basal cell carcinoma (NCT 03132636); NSCLC (NCT 03088540); squamous cell carcinoma of the skin (NCT 02760498), lymphoma (NCT 02651662) and melanoma (NCT 03002376); pittuzumab (CureTech), also known as CT-011, an antibody that binds to PD-1, in clinical trials against diffuse large B-cell lymphoma and multiple myeloma; avermectin The company dugine/merck company KGaA), also known as MSB 0010718C), which is a fully human IgG1 anti-PD-L1 antibody, in clinical trials against non-small cell lung cancer, merkel (Merkel) cell carcinoma, mesothelioma, solid tumor, renal cancer, ovarian cancer, bladder cancer, head and neck cancer, and gastric cancer; or PDR001 (nowa corporation), which is an inhibitory antibody that binds to PD-1, in clinical trials against non-small cell lung cancer, melanoma, triple negative breast cancer and advanced or metastatic solid tumors. Tramadol (CP-675,206; african Co.) is already being directed against multiple drugsA fully human monoclonal antibody directed against CTLA-4 studied in a clinical trial for an indication comprising: mesothelioma, colorectal cancer, renal cancer, breast cancer, lung cancer and non-small cell lung cancer, pancreatic ductal adenocarcinoma, pancreatic cancer, germ cell cancer, head and neck squamous cell carcinoma, hepatocellular carcinoma, prostate cancer, endometrial cancer, metastatic cancer in the liver, liver cancer, large B-cell lymphoma, ovarian cancer, cervical cancer, metastatic polymorphic thyroid cancer, urothelial cancer, fallopian tube cancer, multiple myeloma, bladder cancer, soft tissue sarcoma and melanoma. AGEN-1884 (An Dijun S company (Agenus)) is an anti-CTLA 4 antibody studied in a phase 1 clinical trial against advanced solid tumor (NCT 02694822).
In some embodiments, the checkpoint inhibitor is an inhibitor of T cell immunoglobulin mucin containing protein-3 (TIM-3). TIM-3 inhibitors that may be used in the present invention include TSR-022, LY3321367, and MBG453.TSR-022 (Tesaro Co., tesaro)) is an anti-TIM-3 antibody studied in solid tumors (NCT 02817633). LY3321367 (Eli Lilly) is an anti-TIM-3 antibody studied in solid tumors (NCT 03099109). MBG453 (North Corp.) is an anti-TIM-3 antibody studied in advanced malignancies (NCT 02608268).
In some embodiments, the checkpoint inhibitor is an inhibitor of a T cell immune receptor or TIGIT (an immune receptor on certain T cells and NK cells) having Ig and ITIM domains. TIGIT inhibitors that may be used in the present invention include BMS-986207 (bai-mei-shi noble) anti-TIGIT monoclonal antibody (NCT 02913313); OMP-313M32 (Oncomeded Co., oncomeded)); and anti-TIGIT monoclonal antibodies (NCT 03119428).
In some embodiments, the checkpoint inhibitor is an inhibitor of lymphocyte activation gene-3 (LAG-3). LAG-3 inhibitors that may be used in the present invention include BMS-986016 and REGN3767 and IMP321.BMS-986016 (BAIMEISHIGULAY Co.) an anti-LAG-3 antibody is being studied in glioblastoma and glioma (NCT 02658981). REGN3767 (Regeneron), also an anti-LAG-3 antibody, is being studied in malignancy (NCT 03005782). IMP321 (Immutep Co., ltd. (Immutep S.A.)) is a LAG-3-Ig fusion protein, which is being studied in melanoma (NCT 02676869), adenocarcinoma (NCT 02614833) and metastatic breast cancer (NCT 00349934).
Checkpoint inhibitors that may be used in the present invention include OX40 agonists. OX40 agonists being studied in clinical trials comprise PF-04518600/PF-8600 (pyro), an agonistic anti-OX 40 antibody in metastatic renal cancer (NCT 03092856) and advanced cancers and neoplasms (NCT 02554812; NCT 05082566); GSK3174998 (merck corporation), an agonistic anti-OX 40 antibody in the phase 1 cancer assay (NCT 02528357); MEDI0562 (medical immune)/aliskir, a kind of anti-OX 40 antibody in advanced solid tumors (NCT 02318394 and NCT 02705482); MEDI6469, an agonistic anti-OX 40 antibody (medical immune/aliskir) in patients with colorectal cancer (NCT 02559024), breast cancer (NCT 01862900), head and neck cancer (NCT 02274155) and metastatic prostate cancer (NCT 01303705); and BMS-986178 (Bai Shi Gui Bao Co.), an agonistic anti-OX 40 antibody in advanced cancer (NCT 02737475).
Checkpoint inhibitors that may be used in the present invention include CD137 (also known as 4-1 BB) agonists. CD137 agonists being studied in clinical trials comprise Wu Tuolu mab (utominiumab) (PF-05082566, pyro), an agonistic anti-CD 137 antibody in diffuse large B cell lymphoma (NCT 02951156) as well as advanced cancers and neoplasms (NCT 02554812 and NCT 05082566); wu Ruilu mab (BMS-663513, BAIMEISHIGUOBO Co.), an agonistic anti-CD 137 antibody in melanoma and skin cancer (NCT 02652455) glioblastoma and glioma (NCT 02658981); and CTX-471 (Compass therapeutics (Compass Therapeutics)), an agonistic anti-CD 137 antibody in metastatic or locally advanced malignant disease (NCT 03881488).
Checkpoint inhibitors that may be used in the present invention include CD27 agonists. CD27 agonists being studied in clinical trials comprise: the agonistic anti-CD 27 antibodies in vallizumab (CDX-1127,Celldex Therapeutics), head and neck squamous cell carcinoma, ovarian carcinoma, colorectal carcinoma, renal cell carcinoma and glioblastoma (NCT 02335918), lymphoma (NCT 01460134), glioma and astrocytoma (NCT 02924038).
Checkpoint inhibitors that may be used in the present invention include glucocorticoid-induced tumor necrosis factor receptor (GITR) agonists. GITR agonists being studied in clinical trials included TRX518 (acuminate biomedical corporation (Leap Therapeutics)), an agonistic anti-GITR antibody in malignant melanoma and other malignant solid tumors (NCT 01239134 and NCT 02628574); GWN323 (nowa corporation), an agonistic anti-GITR antibody in solid tumors and lymphomas (NCT 02740270); INCAGN01876 (because of Corp/An Dijun S), an agonistic anti-GITR antibody in advanced cancers (NCT 02697591 and NCT 03126110); MK-4166 (merck corporation), an agonistic anti-GITR antibody in solid tumors (NCT 02132754); and MEDI1873 (medical immunology/aslicon), an agonistic hexamer GITR-ligand molecule with a human IgG1 Fc domain in advanced solid tumors (NCT 02583165).
Checkpoint inhibitors that may be used in the present invention include inducible T cell costimulatory (ICOS, also known as CD 278) agonists. ICOS agonists being studied in clinical trials comprise: MEDI-570 (medical immune company), an agonistic anti-ICOS antibody in lymphoma (NCT 02520791); GSK3359609 (merck), an agonistic anti-ICOS antibody in stage 1 (NCT 02723955); JTX-2011 (Jiang Si, pharmaceutical Co., ltd. (Jounce Therapeutics)), an agonistic anti-ICOS antibody in phase 1 (NCT 02904226).
Checkpoint inhibitors that may be used in the present invention include killer IgG-like receptor (KIR) inhibitors. KIR inhibitors being studied in clinical trials comprise: li Ruilu mab (IPH 2102/BMS-986015, from Intra-specialty pharmaceutical Co., BAISHEMEISULANSIS Guibao), an anti-KIR antibody in leukemia (NCT 01687387, NCT02399917, NCT02481297, NCT 02599649), multiple myeloma (NCT 02252263) and lymphoma (NCT 01592370); IPH2101 (1-7F 9, genetics specialty chemicals) in myeloma (NCT 01222286 and NCT 01217203); and IPH4102 (from endo-specialty pharmaceutical company), an anti-KIR antibody (KIR 3DL 2) that binds to three domains of long cytoplasmic tails in lymphomas (NCT 02593045).
Checkpoint inhibitors that may be used in the present invention include CD47 inhibitors of the interaction between CD47 and signal-modulating protein α (SIRPa). The CD47/SIRPa inhibitors being studied in clinical trials comprise: ALX-148 (Allisia medicine Co., ltd (Alexo Therapeutics)), an antagonistic variant in phase 1 (NCT 03013218) that binds to CD47 and prevents CD47/SIRPa mediated signaling (SIRPa); TTI-621 (SIRPa-Fc, trillion pharmaceutical company (Trillium Therapeutics)), a soluble recombinant fusion protein produced in phase 1 clinical trials (NCT 02890368 and NCT 02663518) by linking the N-terminus of the CD47 binding domain of SIRPa with the Fc domain of human IgG1, acting by binding to human CD47 and preventing it from delivering its "do not eat" signal to macrophages; CC-90002 (New York Co.) an anti-CD 47 antibody in leukemia (NCT 02641002); and Hu5F9-G4 (Forty Seven corporation (Forty Seven, inc.)) in colorectal neoplasms and solid tumors (NCT 02953782), acute bone leukemia (NCT 02678338), and lymphomas (NCT 02953509).
Checkpoint inhibitors that may be used in the present invention include CD73 inhibitors. CD73 inhibitors being studied in clinical trials comprise MEDI9447 (medical immunology), an anti-CD 73 antibody in solid tumors (NCT 02503774); and BMS-986179 (BASEMENSHIGULAR Co.) an anti-CD 73 antibody in solid tumor (NCT 02754141).
Checkpoint inhibitors that may be used in the present invention comprise agonists of the interferon gene stimulatory protein (STING, also known as transmembrane protein 173 or TMEM 173). Agonists of STING being studied in clinical trials include: MK-1454 (merck), an agonistic synthetic cyclic dinucleotide in lymphoma (NCT 03010176); and ADU-S100 (MIW 815, aduro Biotech/North), an agonistic synthetic cyclic dinucleotide in phase 1 (NCT 02675439 and NCT 03172936).
Checkpoint inhibitors that may be used in the present invention include CSF1R inhibitors. CSF1R inhibitors being studied in clinical trials include: pexidatinib (PLX 3397, plakexicam company (Plexxikon)), a small-molecule inhibitor of CSF1R in colorectal, pancreatic, metastatic and advanced cancers (NCT 02777710), melanoma, non-small cell lung cancer, head and neck squamous cell carcinoma, gastrointestinal stromal tumor (GIST), and ovarian cancer (NCT 02452424); and IMC-CS4 (LY 3022855, gillyx (Lilly)), an anti-CSF-1R antibody in pancreatic cancer (NCT 03153410), melanoma (NCT 03101254) and solid tumors (NCT 02718911); and BLZ945 (4- [2 ((1R, 2R) -2-hydroxycyclohexylamino) -benzothiazol-6-yloxy ] -pyridine-2-carboxylic acid methylamide, nohua corporation), an orally available inhibitor of CSF1R in advanced solid tumors (NCT 02829723).
Checkpoint inhibitors that may be used in the present invention include NKG2A receptor inhibitors. The NKG2A receptor inhibitors being studied in clinical trials comprise Mo Nali bead mab (IPH 2201, in-house specialty drug company), an anti-NKG 2A antibody in head and neck neoplasms (NCT 02643550) and chronic lymphocytic leukemia (NCT 02557516).
In some embodiments, the immune checkpoint inhibitor is selected from nivolumab, pembrolizumab, ipilimumab, avistuzumab, dewaruzumab, alemtuzumab, or Pierizumab.
The compounds of the invention may also be used in combination with known methods of treatment, such as administration of hormones or radiation. In certain embodiments, the provided compounds are useful as radiosensitizers, particularly for treating tumors that exhibit poor sensitivity to radiation therapy.
The compounds of the invention may be administered alone or in combination with one or more other therapeutic compounds, with the possible combination therapies taking the form of a fixed combination or administration of the compounds of the invention with one or more other therapeutic compounds being staggered or administered independently of each other or in combination with one or more other therapeutic compounds. Alternatively or additionally, the compounds of the present invention may be administered in combination with chemotherapy, radiation therapy, immunotherapy, phototherapy, surgical intervention or a combination of these, in particular for oncological therapy. Long-term therapy is also possible, as is adjuvant therapy in the context of other therapeutic strategies as described above. Other possible treatments are therapies that maintain the patient's state after tumor regression, or even chemopreventive therapies, e.g. in patients at risk.
These additional agents may be administered separately from the compositions containing the compounds of the invention as part of a multi-dose regimen. Alternatively, those agents may be part of a single dosage form, which is mixed in a single composition with the compounds of the present invention. If administered as part of a multi-dose regimen, the two active agents may be delivered simultaneously, sequentially, or within a period of time of each other (typically five hours of each other).
As used herein, the terms "combination," "combined," and related terms refer to the simultaneous or sequential administration of therapeutic agents in accordance with the present invention. For example, the compounds of the invention may be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms or together in a single unit dosage form. Accordingly, the present invention provides a single unit dosage form comprising a compound of the invention, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant or vehicle.
The amount of both the compound of the invention and the additional therapeutic agent (in those compositions comprising additional therapeutic agents as described above) that can be combined with the carrier material to produce a single dosage form will vary depending upon the subject being treated and the particular mode of administration. Preferably, the composition of the present invention should be configured such that a dose of 0.01 to 100mg/kg body weight/day of the compound of the present invention can be administered.
In those compositions that include additional therapeutic agents, the additional therapeutic agents and the compounds of the invention may act synergistically. Thus, the amount of additional therapeutic agent in such compositions will be less than would be required in monotherapy utilizing the therapeutic agent alone. In such compositions, an additional therapeutic agent may be administered at 0.01-1,000 μg/kg body weight/day.
The amount of additional therapeutic agent present in the compositions of the present invention will not exceed the amount typically administered in compositions comprising the therapeutic agent as the sole active agent. Preferably, the amount of additional therapeutic agent in the presently disclosed compositions will be in the range of about 50% to 100% of the amount typically present in compositions comprising the agent as the sole therapeutically active agent.
The compounds of the present invention or pharmaceutical compositions thereof may also be incorporated into compositions for coating implantable medical devices such as prostheses, prosthetic valves, vascular grafts, stents and catheters. For example, vascular stents have been used to overcome restenosis (restenosis of the vessel wall after injury). However, patients using stents or other implantable devices are at risk of clot formation or platelet activation. These undesirable effects may be prevented or reduced by pre-coating the device with a pharmaceutically acceptable composition comprising a kinase inhibitor. Implantable devices coated with the compounds of the present invention are another embodiment of the present invention.
Illustration of an example
As depicted in the examples below, in certain exemplary embodiments, compounds were prepared according to the following general procedure. It will be appreciated that although the general method depicts the synthesis of certain compounds of the present invention, the following general methods and other methods known to those of ordinary skill in the art may be applied to all compounds and subclasses and species of each of these compounds as described herein. Additional compounds of the present invention are prepared by methods substantially similar to those described herein in the examples and known to those of skill in the art.
Synthesis of intermediates
Synthesis of methyl 2- (bromomethyl) -3- (trifluoromethyl) benzoate (intermediate A3)
Step 1: synthesis of methyl 2-methyl-3- (trifluoromethyl) benzoate (A-2)
To a solution of 2-methyl-3- (trifluoromethyl) benzoic acid (A-1) (10 g,1 eq, 49 mmol) in MeOH (100 mL) was added sulfuric acid (0.24 g,0.13mL,0.05 eq, 2.4 mmol),and stirred at 75℃for 3 days. Most of the solvent was removed under vacuum. The reaction mixture was diluted with EtOAc (50 mL) and treated with NaHCO 3 Is washed with saturated aqueous solution (3X 10 mL) and brine (10 mL). The combined organic layers were dried (MgSO 4 ) Filtered and concentrated in vacuo to give the sub-title compound (a-2) (9.2 g,39mmol,80%,93% purity) as a pale orange oil. 1 H NMR(400MHz,DMSO-d6)δ7.96(dd,J=7.8,1.4Hz,1H),7.90(dd,J=7.9,1.4Hz,1H),7.52(tt,J=7.9,0.9Hz,1H),3.87(s,3H),2.54(d,J=1.8Hz,3H)。
Step 2: synthesis of methyl 2- (bromomethyl) -3- (trifluoromethyl) benzoate (A-3)
To the product (A-2) (2.00 g,1 equivalent, 9.17 mmol) from step 1 above in CCl 4 To a solution in (45 mL) were added NBS (2.45 g,1.5 eq, 13.8 mmol) and BPO (88 mg,75Wt%,0.3 eq, 2.75 mmol) successively and stirred at reflux overnight. The reaction mixture was diluted with DCM (10 mL) and NaHCO 3 Is washed with saturated aqueous solution (2X 10 mL), saturated aqueous solution (2X 10 mL) of NH4Cl, and brine (10 mL). The combined organic layers were dried (MgSO 4 ) Filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (40 g cartridge, 0-10% etoac/isohexane) to give the title compound (a-3) (2.10 g,7.0mmol,76%,99% purity) as a clear colorless oil. 1 H NMR (400 MHz, chloroform-d) δ8.08-8.02 (m, 1H), 7.82 (dd, j=7.9, 1.4hz, 1H), 7.49 (td, j=7.9, 0.9hz, 1H), 5.11 (s, 2H), 3.99 (s, 3H).
Synthesis of methyl 2- (bromomethyl) -5- (hydroxymethyl) -3- (trifluoromethyl) benzoate (intermediate B-5)
Step 1: synthesis of methyl 5-bromo-2-methyl-3- (trifluoromethyl) benzoate (B-1)
To a solution of methyl 2-methyl-3- (trifluoromethyl) benzoate (a-2) (5.00 g,1 eq, 22.9 mmol) in AcOH (45 mL) and nitric acid (21.2 g,15.1mL,68wt%,10 eq, 229 mmol) at 0 ℃ was added Br2 (4.03 g,1.30mL,1.1 equivalents, 25.2 mmol). To the solution was added dropwise a solution of silver nitrate (5.06 g,1.3 equivalents, 29.8 mmol) in water (12 mL) at 0 ℃. The resulting mixture was stirred at 0℃for 3 hours. The mixture was diluted with water (50 mL) and extracted with DCM (50 mL). The combined organic layers were washed with brine (20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (80 g cartridge, 0-20% EtOAc/isohexane) to give the sub-title compound (B-1) (5.74 g,94% purity) as a colorless oil. 1 H NMR(400MHz,DMSO-d6)δ8.16(d,J=2.2Hz,1H),8.08(d,J=2.2Hz,1H),3.90(s,3H),2.50(q,J=1.7Hz,3H)。
Step 2: synthesis of 3- (methoxycarbonyl) -4-methyl-5- (trifluoromethyl) benzoic acid (B-2)
To a solution of the product (B-1) (5.74 g,94Wt%,1 eq., 18.2 mmol) from step 1 above in DMF (85 mL) was added oxalic acid (2.45 g,1.5 eq., 27.2 mmol), DIPEA (2.35 g,3.16mL,1 eq., 18.2 mmol), ac under N2 at room temperature 2 O (2.78 g,2.57mL,1.5 eq, 27.2 mmol), pd (OAc) 2 (408 mg,0.1 eq, 1.82 mmol) and Xantphos (2.10 g,0.2 eq, 3.63 mmol). The resulting mixture was stirred at 100℃for 16 hours. The mixture was cooled to room temperature, diluted with water (50 mL) and extracted with DCM (3×50 mL). The combined organic layers were washed with brine (50 mL), dried (MgSO 4), filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (220 g cartridge, 10-50% MTBE/isohexane) to give the sub-title compound (B-2) (1.57 g,95% purity) as a colorless solid. M/z 261.0 (M-H) - (ES-). 1 H NMR(400MHz,DMSO-d6)δ13.69(bs,1H),8.47(d,J=1.8Hz,1H),8.29(d,J=1.9Hz,1H),3.90(s,3H),2.66–2.56(m,3H)。
Step 3: synthesis of 4- (bromomethyl) -3- (methoxycarbonyl) -5- (trifluoromethyl) benzoic acid (B-3)
To the product (B-2) (1.51 g,1 equivalent, 5.76 mmol) from step 2 above in CCl 4 To a solution of (55 mL) was added NBS (1.54 g,1.5 eq, 8.64 mmol) and BPO (558 mg,75Wt%,0.3 eq, 1.73 mmol). The resulting mixture was stirred under reflux for 64 hours. Cooling the mixtureTo room temperature and concentrated in vacuo. The crude product was purified by silica gel chromatography (120 g cartridge, 0-50% MTBE/isohexane) (dry loaded) to give the sub-title compound (B-3) (1.65 g,99% purity) as a colorless solid. M/z 339.1 and 341.1 (M-H) - (ES-). 1 H NMR(400MHz,DMSO-d6)δ13.9(s,1H),8.57(d,J=1.8Hz,1H),8.34(d,J=1.8Hz,1H),5.07(s,2H),3.95(s,3H)。
Step 4: synthesis of methyl 2- (bromomethyl) -5- (hydroxymethyl) -3- (trifluoromethyl) benzoate (B-4)
To a solution of the product (B-3) from step 3 above (1.65 g,1 equivalent, 4.84 mmol) in dry THF (45 mL) was added BH 3 THF (1.25 g,14.5mL,1 mol, 3 eq, 14.5 mmol). The mixture obtained is put in N 2 Stirring was carried out at room temperature for 48 hours under an atmosphere. The mixture was slowly quenched with MeOH and concentrated in vacuo. The crude product was purified by silica gel chromatography (80 g cartridge, 10-60 MTBE/hexane) to give the sub-title compound (B-4) (1.54 g,97% purity) as a clear oil. 1 H NMR (400 mhz, dmso-d 6) delta 8.05 (d, j=1.8 hz, 1H), 7.90 (s, 1H), 5.03 (s, 2H), 4.63 (s, 2H), 3.92 (s, 3H). No exchangeable protons were observed.
Step 5: synthesis of methyl 2- (bromomethyl) -5-formyl-3- (trifluoromethyl) benzoate (B-5)
To a solution of product (B-4) (500 mg,1 eq, 1.53 mmol) from step 4 above and in DCM (25 mL) was added DMP (973 mg,1.5 eq, 2.29 mmol). The mixture was stirred at room temperature for 2 hours. The reaction mixture was diluted with DCM (10 mL) and transferred to a separatory funnel. The layer was treated with Na 2 S 2 O 3 Is washed with saturated aqueous solution (3X 10 mL) and then with NaHCO 3 Is washed with brine (3X 10 mL). The combined organic extracts were dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (24 g cartridge, 5-20% mtbe/isohexane) to give the title compound (B-5) (317 mg,99% purity) as a colorless oil. M/z 325.00 and 327.00 (M+H) + (ES+)。 1 H NMR (400 MHz, chloroform-d) δ10.12 (s, 1H), 8.56 (d, j=1.8 hz, 1H), 8.45 (d, j=1.8 hz, 1H), 5.08 (s, 2H), 3.86 (s, 3H)。
Synthesis of 6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (intermediate C-1)
To a solution of intermediate B-4 (200 mg,1 eq., 611. Mu. Mol) in MeOH (2 mL) was added ammonia (125 mg,1.05mL,7 mol, 12 eq., 7.34 mmol). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with MeOH and adsorbed onto silica gel and concentrated in vacuo. The crude product was purified by silica gel chromatography (12 g cartridge, 0-10% MeOH/DCM) to give the title compound (C-1) as a clear white solid (100 mg,0.43mmol,70%,99% purity). M/z 232.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.86(s,1H),7.88(d,J=14.9Hz,2H),5.52(t,J=5.8Hz,1H),4.67(d,J=5.8Hz,2H),4.53(s,2H)。
Synthesis of 3- (2-bromophenyl) -4-methyl-4H-1, 2, 4-triazole (intermediate D-5)
Step 1: synthesis of 2- (2-bromobenzoyl) -N-methylhydrazine-1-thioformamide (D-3)
To a solution of 2-bromobenzoic acid (D-1) (10.2 g,1 eq, 50.6 mmol) and 1-amino-3-methyl-thiourea (D-2) (5.32 g,1.0 eq, 50.6 mmol) in DMF (100 mL) was added HATU (23.1 g,1.2 eq, 60.7 mmol) in portions at 0deg.C. DIPEA (32.7 g,44.1mL,5.0 eq, 253 mmol) was then added under nitrogen at the same temperature. The resulting mixture was stirred at room temperature for 16 hours. Water (200 mL) was added and the aqueous layer extracted with EtOAc (3X 200 mL). The combined organic layers were washed with brine (3×200 mL) and dried (Na 2 SO 4 ) Then concentrated in vacuo to give the crude sub-title compound (D-3) (15.3 g,37mmol,73%,70% purity) as a yellow oil, which was used without purification. M/z 286/288 (M-H) - (ES-)。
Step 2: synthesis of 5- (2-bromophenyl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (D-4)
A solution of the product (D-3) from step 1 above (15.3 g,70wt%,1.0 eq., 37.2 mmol) in NaOH solution (4.31 g,108mL,1.0 mol, 2.90 eq., 108 mmol) was stirred overnight at 50 ℃. Water (300 mL) was added and the mixture was acidified with HCl (1M in water) to pH 5 at 0deg.C. The aqueous layer was extracted with EtOAc (3X 200 mL). The organic extracts were combined, dried (Na 2 SO 4 ) And concentrated in vacuo to give the sub-title compound (D-4) (6.55 g,21mmol,55%,85% purity), M/z 270/272 (M+H) as a yellow oil + (ES+)。
Step 3: synthesis of 3- (2-bromophenyl) -4-methyl-4H-1, 2, 4-triazole (D-5)
To a stirred mixture of product (D-4) (6.55 g,30wt%,1 eq, 7.27 mmol) from step 2 above in DCM (80 mL) was added AcOH (874 mg, 833. Mu.L, 2 eq, 14.5 mmol) dropwise at 0deg.C. Dropwise adding H to the above mixture at 0deg.C 2 O 2 (30 wt%) (4.95 g,4.46mL,30Wt%,6 eq, 43.6 mmol). The resulting mixture was stirred at room temperature for 1.5 hours. The resulting mixture was diluted with water (200 mL). The mixture was treated with saturated NaHCO 3 Alkalizing to pH 8 in the water solution. The aqueous layer was extracted with DCM (3X 200 mL). The organic phase was dried (Na 2 SO 4 ) And then concentrated in vacuo. The crude product was purified by silica gel chromatography (80 g cartridge, 0-10% MeOH/DCM) to give an impure material that was triturated with DCM (10 mL) and the precipitate filtered. The filtrate was concentrated in vacuo and purified by RP flash C18 chromatography (40 g cartridge, 0-100% mecn/10mM ammonium bicarbonate) to give the title compound (D-5) as a pale tan solid (400 mg,1.5mmol,21%,90% purity). M/z 238.1/240.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.63(s,1H),7.86–7.81(m,1H),7.59–7.50(m,3H),3.48(s,3H)。
Synthesis of 2, 6-dichloro-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridine (intermediate E-7)
Step 1: synthesis of methyl 2- (2, 6-dichloropyridin-4-yl) benzoate (E-3)
To a solution of methyl 2-bromobenzoate (E-1) (765 mg,0.500mL,1 equivalent, 3.56 mmol) and 2, 6-dichloro-4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine (E-2) (1.34 g,80Wt%,1.1 equivalent, 3.91 mmol) in 1, 4-dioxane (24 mL) and water (6 mL) was added Pd (dppf) Cl2.DCM (145 mg,0.05 equivalent, 178. Mu. Mol) and potassium carbonate (1.47 g,3 equivalent, 10.7 mmol) degassed three times with nitrogen and heated overnight at 80 ℃. The reaction mixture was passed through a celite plug eluting with EtOAc (30 mL). The filtrate was concentrated in vacuo and the crude product was purified by silica gel chromatography (24 g cartridge, 0-10% MeOH/DCM) to give the sub-title compound (E-3) as a pale yellow solid (873.6 mg,2.9mmol,83%,95% purity). 1 H NMR(400MHz,DMSO-d6)δ8.00–7.95(m,1H),7.77–7.69(m,1H),7.67–7.61(m,1H),7.56(s,2H),7.50–7.46(m,1H),3.68(s,3H)。
Step 2: synthesis of 2- (2, 6-dichloropyridin-4-yl) benzoic acid (E-4)
To a solution of the product (E-3) from step 1 above (8.38 g,93Wt%,1 eq, 27.6 mmol) in THF (10 mL) was added a solution of LiOH (292 mg,10mL,1 eq, 27.6 mmol) in water (10 mL) and the mixture was stirred at 40℃for 16 h. The reaction mixture was concentrated in vacuo. The residue was dissolved in water (30 mL), acidified with 1M HCl, and extracted with EtOAc (3×50 mL). The organic extracts were combined and dried (MgSO 4 ) And concentrated in vacuo to give the sub-title compound (E-4) (7.34 g,27mmol,97%,98% purity) as an off-white solid. M/z 268.1/270.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ13.14(s,1H),7.95(dd,J=7.7,1.4Hz,1H),7.76–7.64(m,1H),7.64–7.57(m,1H),7.56(s,2H),7.48–7.42(m,1H)。
Step 3: synthesis of 2- (2, 6-dichloropyridin-4-yl) benzoyl) -N-methylhydrazine-1-thiocarboxamide (E-5)
The product (E-4) from step 2 above (6.67 g,1 eq, 24.9 mmol) and 1-amino-3-methyl-thiourea (D-2) (2.88 g,1.1 eq, 27.4 mmol) were dissolved in EtOAc (10 mL). DIPEA (7.33 g,9.88mL,2.28 eq, 56.7 mmol) was then added followed by dropwise addition of T3P (23.7 g,22.2mL,50wt%,1.5 eq, 37.3 mmol). The resulting mixture was then stirred at 70℃for 16 hours. The mixture was diluted with water (20 mL) and the precipitate was filtered to give the sub-title compound (E-5) (8.84 g,24.9mmol, 100%) as a brown solid, which was used without further purification or analysis.
Step 4: synthesis of 5- (2, 6-dichloropyridin-4-yl) phenyl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (E-6)
A solution of the product (E-5) (8.85 g,1 eq, 24.9 mmol) from step 3 above in NaOH solution (2.99 g,37.3mL,2 mol, 3 eq, 74.7 mmol) was stirred at 50℃for 16 hours. Water (100 mL) was added and the mixture was acidified to pH 5 with HCl (1M in water). The precipitate was filtered and concentrated in vacuo to give the sub-title compound (E-6) (3.53 g,10mmol,41%,98% purity) as a pale brown solid. 1 H NMR(400MHz,DMSO-d6)δ13.85(s,1H),7.83–7.68(m,4H),7.44(s,2H),3.26–3.23(m,3H)。
Step 5: synthesis of 5- (2, 6-dichloropyridin-4-yl) phenyl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (E-7)
To a solution of the product (E-6) (3.54 g,98Wt%,1 eq., 10.3 mmol) from step 4 above in DCM (90 mL) and AcOH (18.5 g,17.7mL,30 eq., 309 mmol) with stirring at 0deg.C was added H dropwise 2 O 2 (30% in water) (11.7 g,10.5mL,30Wt%,10 equivalents, 103 mmol). The mixture was stirred at this temperature for 1 hour before concentrating in vacuo. The residue was dissolved in water and basified with NaOH (2M) to pH 10, then organics were separated and aqueous phase extracted with EtOAc (2 x100 mL) and organics were washed with brine (50 mL). The organics were combined, dried (MgSO 4 ) Filtered and concentrated in vacuo to give the title compound (E-7) (2.94 g,9.2mmol,89%,95% purity) as a pale yellow solid. M/z305.2/307.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.54 (s, 1H), 7.85-7.67(m,4H),7.26(s,2H),3.42(s,3H)。
Synthesis of 2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -3-oxo-7- (trifluoromethyl) isoindoline-5-carbaldehyde (intermediate Q-1)
/>
Compound P-2 (167 mg,1 eq, 360. Mu. Mol) and Dess-Martin periodate (Dess-Martin periodinane) (229 mg,1.5 eq, 539. Mu. Mol) were stirred in DCM (6 mL) at room temperature for 90 min. The reaction mixture was diluted with DCM (10 mL) and NaHCO 3 Saturated aqueous (10 mL) was washed. The organics were separated, dried (MgSO 4 ) Filtered and concentrated in vacuo to give the title compound (Q-1) (0.21 g,360 μmol,100%,80% purity) as a yellow solid, which was used in the next step without further purification. M/z 463.2 (M+H) + (ES+)。
Synthesis of 3- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) aniline (intermediate AAB-8)
Step 1: synthesis of ethyl 1-methyl-5- (3-nitrophenyl) pyrazole-4-carboxylate (AAB-3)
To a stirred mixture of 5-bromo-1-methylpyrazole-4-carboxylic acid ethyl ester (AAB-1) (1.00 g,1 eq, 4.29 mmol), 3-nitrophenylboronic acid (AAB-2) (1.07 g,1.5 eq, 6.44 mmol) and sodium carbonate (1.36 g,3 eq, 12.9 mmol) in 1, 2-dimethoxy-ethane (15 mL) and water (1.5 mL) at room temperature under nitrogen was added Pd (dppf) Cl 2 DCM (700.7 mg,0.2 eq., 858. Mu. Mol). The resulting mixture was stirred overnight at 90 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by silica gel column chromatography eluting with petroleum ether/EtOAc (1/1) to give the sub-title compound (AAB-3) as a brown solid (900 mg,3.27mmol, 76%). m/z 275.3(M+H) + (ES+)。
Step 2: synthesis of 1-methyl-5- (3-nitrophenyl) pyrazole-4-carboxylic acid (AAB-4)
To a stirred mixture of the product from step 1 above (AAB-3) (500 mg,1 eq, 1.82 mmol) in MeOH (8 mL) and THF (8 mL) was added a solution of NaOH (803 mg,5 eq, 9.08 mmol) in water (4 mL) at room temperature. The resulting mixture was stirred at 60℃for 1.5 hours. The mixture was cooled to room temperature, diluted with water and acidified to pH 5 with HCl (1M aqueous solution) at 0 ℃. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo to give the sub-title compound (AAB-4) (440 mg,1.78mmol, 98%) as a white solid. M/z 248.2 (M+H) + (ES+)。
Step 3: synthesis of 1-methyl-N- [ (methylaminomethylsulfonyl) amino ] -5- (3-nitrophenyl) pyrazole-4-carboxamide (AAB-5)
To a stirred mixture of the product from step 2 above (AAB-4) (360 mg,1 eq, 1.46 mmol) in DMF (5 mL) was added HATU (831 mg,1.5 eq, 2.18 mmol) and DIPEA (560 mg,3 eq, 4.37 mmol) at 0deg.C. The resulting mixture was stirred at room temperature for 0.5 hours. 4-methyl-3-thiosemicarbazide (D-2) (161 mg,1.05 eq, 1.53 mmol) was added to the above mixture at room temperature. The resulting mixture was stirred at room temperature overnight. The reaction was concentrated to give the sub-title compound (AAB-5) which was used directly in the next step. M/z335.4 (M+H) + (ES+)。
Step 4: synthesis of 4-methyl-5- [ 1-methyl-5- (3-nitrophenyl) pyrazol-4-yl ] -1,2, 4-triazole-3-thiol (AAB-6)
A solution of the product from step 3 above (AAB-5) (4816 mg,1 eq., 1.45 mmol) in NaOH (aqueous solution, 1M) (10 mL) was stirred at 60℃for 3 hours. The mixture was cooled to room temperature, diluted with water, acidified to pH 5 with HCl (aqueous, 1M) and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. Passing the crude product through reverse phase flash column chromatographyThe process was purified under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (10% ACN up to 80% within 20 minutes); detector, UV 254/220nm to give the sub-title compound (AAB-6) as a brown solid (160 mg, 5.6. Mu. Mol, 35%). M/z 317.3 (M+H) + (ES+)。
Step 5: synthesis of 4-methyl-3- [ 1-methyl-5- (3-nitrophenyl) pyrazol-4-yl ] -1,2, 4-triazole (AAB-7)
To a stirred solution of the product from step 4 above (AAB-6) (160 mg,1 eq, 506. Mu. Mol) in DCM (20 mL) at 0deg.C was added acetic acid (30 mg,1 eq, 506. Mu. Mol) and hydrogen peroxide (344 mg,30Wt%,6 eq, 3.04 mmol). The resulting mixture was stirred at room temperature for 3 hours, then concentrated in vacuo and purified by silica gel column chromatography eluting with DCM/MeOH (10/1) to give the sub-title compound (AAB-7) as a brown oil (120 mg,422 μmol, 83%). M/z 285.3 (M+H) + (ES+)。
Step 6: synthesis of 3- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] aniline (intermediate AAB-8)
Pd/C39 (44.9 mg,10Wt%,0.1 eq., 42.2. Mu. Mol) was added in portions to a stirred solution of the product (AAB-7) (120 mg,1 eq., 422. Mu. Mol) from step 5 above in MeOH (13 mL) at room temperature under nitrogen. The mixture was hydrogenated under a hydrogen atmosphere using a hydrogen balloon at room temperature for 2 hours. The mixture was filtered through a pad of celite and concentrated in vacuo. The filtrate was concentrated to give the subtitle compound (AAB-8) (60 mg,236 μmol, 56%) as a white solid and used in the next step without further purification. M/z 255.3 (M+H) + (ES+)。
Synthesis of methyl 5- ((5-azaspiro [2.4] heptan-5-yl) methyl) -2- (bromomethyl) -3- (trifluoromethyl) benzoate (intermediate AAC-2)
To intermediate (B-5) (325 mg,1 eq, 1.00 mmol) and 5-azaspiro [2.4]]To a stirred solution of heptane, HCl (AAC-1) (134 mg,1 eq, 1.00 mmol) in DCM (10 mL) was added Et 3 N (101 mg,1 equivalent, 1.00 mmol) and NaBH (OAc) 3 (424 mg,2 equivalents, 2.00 mmol). The resulting mixture was stirred at room temperature for 3 hours, then concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (10% ACN up to 50% within 10 minutes); detector, UV 254/220nm. This gave the title compound (AAC-2) (65 mg, 160. Mu. Mol, 16%) as a yellow oil. M/z 406.2 (M+H) + (ES+)。
Synthesis of 3- (5-bromo-1-methyl-1H-pyrazol-4-yl) -4-methyl-4H-1, 2, 4-triazole (intermediate AAH-4)
Step 1: synthesis of 5-bromo-1-methyl-1H-pyrazole-4-carboxylic acid (AAH-1)
To a solution of 5-bromo-1-methyl-1H-pyrazole-4-carboxylic acid ethyl ester (AAB-1) (1.2 g,1 eq, 5.17 mmol) in THF (20 mL) and water (5 mL) was added LiOH (373 mg,3 eq, 15.5 mmol) at room temperature. The resulting solution was stirred at 50℃for 16 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was diluted with water and the mixture was acidified with HCl (aqueous, 1M) to pH 3. The solid was collected by filtration to give the sub-title compound (AAH-1) (780 mg,3.82mmol, 74%) as an off-white solid. M/z 205.0 (M+H) + (ES+)。
Step 2: synthesis of 2- (5-bromo-1-methyl-1H-pyrazole-4-carbonyl) -N-methylhydrazine-1-thiocarboxamide (AAH-2)
To a stirred solution of the product from step 1 above (AAH-1) (780 mg,1 eq, 3.82 mmol) and 4-methyl-3-thiosemicarbazide (D-2) (481 mg,1.2 eq, 4.58 mmol) in DMF (8 mL) was added DIPEA (1.48 g,3 eq, 11.46 mmol) and HATU (1.6 g,1.1 eq, 4.2 mmol) at room temperature. The resulting mixture was stirred at room temperature for 16 hours, then concentrated in vacuo. Mixing the obtained mixture The compound was diluted with water and extracted with DCM (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ) Filtration and concentration in vacuo gave the sub-title compound (AAH-2) (930 mg,3.19mmol, 84%) as a yellow oil. M/z 292.2 (M+H) + (ES+)。
Step 3: synthesis of 5- (5-bromo-1-methyl-1H-pyrazol-4-yl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (AAH-3)
A mixture of the product from step 2 above (AAH-2) (930 mg,1 eq, 3.19 mmol) in NaOH (aqueous solution, 1M) (10 mL) was stirred overnight at room temperature. The mixture was diluted with water. The mixture was acidified with HCl (aqueous solution) to pH 3. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and ACN (24% ACN up to 41% in 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (AAH-3) (630 mg,2.30mmol, 72%) as a yellow solid. M/z 274.1 (M+H) + (ES+)。
Step 4: synthesis of 3- (5-bromo-1-methyl-1H-pyrazol-4-yl) -4-methyl-4H-1, 2, 4-triazole (AAH-4)
To a solution of the product from step 3 above (AAH-3) (630 mg,1 eq, 2.3 mmol) in DCM (10 mL) and acetic acid (5 mL) at 0deg.C was added hydrogen peroxide (2.63 mL,30Wt%,10 eq, 23 mmol) and the resulting mixture was stirred for 2 hours. The mixture was diluted with water and saturated NaHCO 3 The (aqueous) solution is alkalized to pH 8. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was applied to silica gel column chromatography with DCM/MeOH (20/1) to give the title compound (AAH-4) as a white solid (400 mg,1.66mmol, 72%). M/z 242.1 (M+H) + (ES+)。
Synthesis of 2- (3- (3- (5-hydroxy-4-methyl-4H-1, 2, 4-triazol-3-yl) pyridin-4-yl) phenyl) -4- (trifluoromethyl) isoindolin-1-one (intermediate AAS-6)
Step 1: synthesis of (3- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) phenyl) boronic acid (AAS-2)
To a stirred mixture of intermediate (A-3) (100 mg,1 eq, 330. Mu. Mol) and 3-aminophenylboronic acid (AAS-1) (55 mg,1.2 eq, 400. Mu. Mol) in DMF (3 mL) was added DIPEA (218 mg,5 eq, 1.68 mmol) at room temperature under nitrogen. The resulting mixture was stirred under nitrogen at 60 ℃ for 16 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and ACN (45% ACN up to 55% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AAS-2) (91 mg, 283. Mu. Mol, 84%) as a white solid. M/z 322.1 (M+H) + (ES+)。
Step 2: synthesis of 4- (3- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) phenyl) nicotinic acid (AAS-4)
Pd (dppf) Cl was added to a stirred mixture of the product from step 1 above (AAS-2) (60 mg,1 eq, 180. Mu. Mol) and 4-bromopyridine-3-carboxylic acid (AAS-3) (42 mg,1.1 eq, 200. Mu. Mol) and potassium phosphate (77 mg,3 eq, 560. Mu. Mol) in 1, 4-dioxane (4 mL) and water (1 mL) at room temperature under nitrogen atmosphere 2 DCM (27 mg,0.2 eq, 30 mmol). The resulting mixture was stirred under nitrogen at 80 ℃ for 3 hours. The mixture was cooled to room temperature and the residue was diluted with water. The mixture was acidified to pH 4 with HCl (aqueous solution) and the resulting mixture was extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. This gave the sub-title compound (AAS-4) (65 mg, 163. Mu. Mol, 87%) as a white solid. M/z 399.1 (M+H) + (ES+)。
Step 3: synthesis of N-methyl-2- (4- (3- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) phenyl) nicotinoyl) hydrazine-1-thiocarboxamide (AAS-5)
To a stirred solution of the product (AAS-4) (60 mg,1 eq., 150. Mu. Mol) from step 2 above and 4-methyl-3-thiosemicarbazide (D-2) (19 mg,1.2 eq., 180. Mu. Mol) in EtOAc (5 mL) at room temperature was added T 3 P (191 mg,4 eq, 600. Mu. Mol) and DIPEA (116 mg,6 eq, 910. Mu. Mol). The resulting mixture was stirred at room temperature for 16 hours. The resulting mixture was concentrated in vacuo and the crude product was used directly in the next step without further purification. M/z 486.1 (M+H) + (ES+)。
Step 4: synthesis of 2- (3- (3- (5-hydroxy-4-methyl-4H-1, 2, 4-triazol-3-yl) pyridin-4-yl) phenyl) -4- (trifluoromethyl) isoindolin-1-one (AAS-6)
NaOH (24.6 mg,5 eq, 610. Mu. Mol) was added to the crude product (AAS-5) from step 3 above in DMSO (8 mL) under nitrogen at room temperature. The resulting mixture was stirred under nitrogen at 50 ℃ for 48 hours and then cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (40% ACN up to 60% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 20X 250mm 5um; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 25B to 50B in 10 minutes; a detector, UV 254/210nm; retention time: 9.78. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AAS-6) (22.9 mg,51 μmol, 27%) as a white solid. M/z 452.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ9.03(d,J=0.8Hz,1H),8.77(d,J=5.2Hz,1H),8.13(d,J=7.7Hz,1H),8.04–7.93(m,3H),7.80(t,J=7.7Hz,1H),7.68–7.63(m,1H),7.59(t,J=8.0Hz,1H),7.30–7.23(m,1H),5.21(s,2H),2.83(s,3H)。
Example 1: synthesis of (S) -2- (2-methoxy-5- (1- (4-methyl-4H-1, 2, 4-triazol-3-yl) propan-2-yl) phenyl) -6- (pyrrolidin-1-ylmethyl) -4- (trifluoromethyl) isoindolin-1-one (F-9)
Step 1: synthesis of 1- (4-methoxy-3-nitrophenyl) ethan-1-ol (F-2)
To a stirred solution of 1- (4-methoxy-3-nitrophenyl) ethan-1-one (F-1) (1.0 g,1 eq, 5.1 mmol) in EtOH (20 mL) at room temperature was added NaBH in portions 4 (0.29 g,1.5 eq, 7.6 mmol) and the reaction stirred for 2 hours. The reaction mixture was concentrated in vacuo. The reaction mixture was diluted with EtOAc (25 mL) and transferred to a separatory funnel. Layer with NH 4 Saturated aqueous solution of Cl (3X 5 mL), naHCO 3 Is washed with brine (3X 5 mL). The combined organic layers were collected, dried (MgSO 4 ) Filtered and concentrated in vacuo to give the sub-title compound (F-2) (930 mg,4.6mmol,91%,98% purity) as a pale yellow oil. 1 H NMR(400MHz,DMSO-d 6 )δ7.81(dd,J=2.2,0.7Hz,1H),7.61(ddd,J=8.7,2.3,0.6Hz,1H),7.31(d,J=8.7Hz,1H),5.33(d,J=4.4Hz,1H),4.74(qd,J=6.4,4.4Hz,1H),3.90(s,3H),1.32(d,J=6.5Hz,3H)。
Step 2: synthesis of 4- (1-bromoethyl) -1-methoxy-2-nitrobenzene (F-3)
To a solution of product (F-2) from step 1 above (0.930 g,98wt%,1 eq, 4.62 mmol) in ice AcOH (4 mL) was added dropwise a solution of HBr (33% in ice AcOH) (2.27 g,1.52mL,33wt%,2 eq, 9.24 mmol). The reaction mixture was stirred at room temperature for 16 hours. The mixture was heated to 90 ℃ and stirred for an additional 16 hours. The mixture was diluted with EtOAc (20 mL), water (3X 10 mL), naHCO 3 Is washed with saturated solution (3X 10 mL) and brine (10 mL), dried (MgSO) 4 ) Filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (40 g cartridge, 0-50% EtOAc/isohexane) to give the subtitle as a pale yellow oilCompound (F-3) (383mg, 1.4mmol,30%,94% purity). 1 H NMR (400 MHz, chloroform-d) delta 7.92 (d, j=2.4 hz, 1H), 7.63 (dd, j=8.7, 2.4hz, 1H), 7.07 (d, j=8.8 hz, 1H), 5.18 (q, j=7.0 hz, 1H), 3.97 (s, 3H), 2.04 (d, j=7.0 hz, 3H).
Step 3: synthesis of (1- (4-methoxy-3-nitrophenyl) ethyl) triphenylphosphine bromide (F-4)
Triphenylphosphine (361 mg,1 eq, 1.38 mmol) was added to a solution of product (F-3) (381 mg,94wt%,1 eq, 1.38 mmol) from step 2 above in toluene (5 mL) and stirred at reflux for 72 hours. The mixture was cooled to room temperature. The precipitate was filtered, washed with toluene (2 x5 mL), then hexane (2 x5 mL) and dried under vacuum to give the sub-title compound (F-4) (320 mg,0.55mmol,40%,90% purity) as a pale brown solid. M/z 442.4 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d 6 )δ7.97–7.89(m,3H),7.81–7.70(m,12H),7.35(d,J=1.8Hz,1H),7.32(d,J=1.3Hz,2H),5.77(dq,J=15.2,7.5Hz,1H),3.90(s,3H),1.72(dd,J=18.5,7.3Hz,3H)。
Step 4: (E) Synthesis of-3- (2- (4-methoxy-3-nitrophenyl) prop-1-en-1-yl) -4-methyl-4H-1, 2, 4-triazole (F-6)
Sodium bis (trimethylsilyl) amide (2M in THF) (132 mg, 360. Mu.L, 2 moles, 2 equivalents, 720. Mu. Mol) was added to a solution of the product (F-4) from step 3 above (320 mg,1.7 equivalents, 612. Mu. Mol) in THF (5 mL) and stirred at room temperature for 1 hour. The reaction was cooled to-40 ℃ and THF (1 mL) containing 4-methyl-4H-1, 2, 4-triazole-3-carbaldehyde (F-5) (40.0 mg,1 eq, 360 μmol) was added. The mixture was stirred at-40 ℃ for 1 hour and then warmed to room temperature overnight. Adding saturated NH 4 Cl (10 mL) and the mixture was partitioned between DCM (25 mL) and water (20 mL). Using NH for the organic layer 4 Saturated aqueous solution of Cl (2X 5 mL), naHCO 3 Is washed with saturated aqueous solution (2X 5 mL) and brine (5 mL). The combined organic layers were collected, dried (MgSO 4 ) Filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (12 g cartridge, 0-50% EtOAc/isohexane) to give the sub-title compound (F-6) (40 mg, 0) as a pale yellow oil14mmol,40%,99% purity). M/z 275.2 (M+H) + (ES+). 1 H NMR (400 MHz, chloroform-d) δ9.06 (s, 1H), 7.72 (d, j=2.3 hz, 1H), 7.41 (dd, j=8.7, 2.4hz, 1H), 7.03 (d, j=8.8 hz, 1H), 6.38 (d, j=1.6 hz, 1H), 3.97 (s, 3H), 3.59 (s, 3H), 2.38 (d, j=1.5 hz, 3H).
Step 5: synthesis of 2-methoxy-5- (1- (4-methyl-4H-1, 2, 4-triazol-3-yl) propan-2-yl) aniline (F-7)
To a solution of the product (F-6) from step 4 above (39 mg,1 eq, 0.14 mmol) in EtOH (2 mL) was added Pd/C39 (4 mg,10wt%,0.03 eq, 4. Mu. Mol). The reaction mixture was stirred at room temperature at 5atm H 2 Stirred for 16 hours. The catalyst was filtered and the solvent was removed under vacuum. The process was repeated 3 times, filtered and fresh catalyst was added. The crude product was loaded onto a column containing SCX (150 mg) of MeOH. The column was washed with MeOH (30 mL) and the product eluted with MeOH (30 mL) containing 0.7M ammonia. The ammoniacal MeOH extract was concentrated in vacuo to give the sub-title compound (F-7) (15 mg,55 μmol,39%,90% purity) as a dark orange oil. M/z 247.2 (M+H) + (ES+)。 1 H NMR (400 MHz, chloroform-d) delta 7.91 (s, 1H), 6.66 (d, j=8.8 hz, 1H), 6.48-6.43 (m, 2H), 3.80 (s, 3H), 3.20-3.10 (m, 4H), 3.03 (dd, j=14.4, 6.7hz, 1H), 2.81 (dd, j=14.4, 7.6hz, 1H), 1.35 (d, j=7.0 hz, 3H). The exchangeable protons are not visible.
Step 6: synthesis of (S) -2- (2-methoxy-5- (1- (4-methyl-4H-1, 2, 4-triazol-3-yl) propan-2-yl) phenyl) -6- (pyrrolidin-1-ylmethyl) -4- (trifluoromethyl) isoindolin-1-one (F-9)
To a solution of intermediate B-5 (24 mg,99Wt%,1.2 eq, 73. Mu. Mol) and pyrrolidine (6.5 mg, 7.6. Mu.L, 1.5 eq, 91. Mu. Mol) was added NaBH (OAc) 3 (32 mg,2.5 eq, 0.15 mmol) in DCM (2 mL) and stirred at room temperature for 2 h. The reaction mixture was diluted with DCM (10 mL) and NaHCO 3 Saturated aqueous (10 mL) was washed. The organic layer was dried (phase separator) and concentrated in vacuo. The resulting gum was dissolved in EtOH (1 mL) and EtOH (1 mL) containing the product from step 5 (15 mg,1 eq., 61. Mu. Mol) was added followed by DIPEA (12 mg, 16. Mu.L, 1.5 eq., 91. Mu. Mol) and stirred at 60 ℃Mix overnight. The reaction mixture was diluted with EtOAc (10 mL) and with NH 4 Saturated aqueous solution of Cl (5 mL), naHCO 3 Is washed with saturated aqueous solution (5 mL) and brine (5 mL). The combined organic layers were dried (MgSO 4 ) Filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (4 g cartridge, 0-10% meoh/DCM) to give a racemic mixture of the sub-title compounds (11 mg) as a pale yellow oil. The crude product was purified by chiral SFC (Waters, basic (0.1% ammonia), IH 10X250mm, 5um, 25% MeOH (0.1% ammonia), 75% CO 2 Purification to give the title compound (S) enantiomer (F-9) (3.1 mg, 5.7. Mu. Mol,9%,95% pure). M/z 514.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d) 4 )δ8.29(s,1H),8.10(s,1H),8.01(s,1H),7.28–7.21(m,2H),7.11(d,J=8.3Hz,1H),4.98(d,J=10.0Hz,2H),3.89(s,2H),3.85(s,3H),3.44(s,3H),3.30(d,J=7.6Hz,1H),3.14(dd,J=14.8,6.5Hz,1H),3.06(dd,J=14.8,8.3Hz,1H),2.63(d,J=5.3Hz,4H),1.88(p,J=3.1Hz,4H),1.44(d,J=7.0Hz,3H)。
Example 2: synthesis of 2- (4-methoxy-2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (G-4)
Step 1: synthesis of 3- (4 ' -methoxy-3 ' -nitro- [1,1' -biphenyl ] -2-yl) -4-methyl-4H-1, 2, 4-triazole (G-2)
To a solution of 2- (4-methoxy-3-nitrophenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (G-1) (141 mg,1.2 eq, 504. Mu. Mol) and 3- (2-bromophenyl) -4-methyl-4H-1, 2, 4-triazole (D-5) (100 mg,1.0 eq, 420. Mu. Mol) in degassed 1, 4-dioxane (2 mL) and water (0.5 mL) was added Pd (dppf) Cl in sequence 2 DCM (34.3 mg,0.1 eq, 42.0. Mu. Mol) and K 2 CO 3 (174 mg,3 equivalents, 1.26 mmol) and stirred at 90℃overnight. The reaction mixture was diluted with EtOAc (10 mL) and with NH 4 Cl saturated aqueous solution (1X 5 mL), naHCO 3 Saturated aqueous solution (1X 5)mL) and brine (5 mL). The combined organic layers were dried (MgSO 4 ) Filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (12G cartridge, 0-30% meoh/DCM) to give the sub-title compound (G-2) as a pale yellow oil. M/z 310.9 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.45(s,1H),7.74–7.66(m,1H),7.63(dd,J=8.8,1.8Hz,2H),7.60–7.56(m,2H),7.36(dd,J=8.8,2.3Hz,1H),7.30(d,J=8.8Hz,1H),3.91(s,3H),3.13(s,3H)
Step 2: synthesis of 4-methoxy-2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-amine (G-3)
To a solution of the product (G-2) from step 1 above (65 mg,1 equivalent, 0.21 mmol) in EtOH (2 mL) was added Pd/C39 (6.5 mg,10Wt%,0.029 equivalent, 6.1. Mu. Mol) and at 26℃at 5atm H 2 Stirred for 16 hours. The catalyst was filtered and the solvent removed in vacuo to give the sub-title compound (G-3) (59 mg,0.20mmol,95% purity) as a pale yellow solid. M/z 280.9 (M+H) + (ES+). 1H NMR (400 MHz, chloroform-d) δ8.42 (s, 1H), 7.66-7.35 (m, 5H), 6.74 (d, J=4.3 Hz, 1H), 6.67 (s, 1H), 3.85 (s, 3H), 3.03 (s, 3H). No two exchangeable protons were observed.
Step 3: synthesis of 2- (4-methoxy-2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (G-4)
To a solution of product (G-3) (31 mg,1 eq, 0.11 mmol) and intermediate A-3 (39 mg,1.2 eq, 0.13 mmol) from step 2 above in EtOH (1 mL) was added DIPEA (21 mg, 25. Mu.L, 1.5 eq, 0.17 mmol) and stirred at 60℃for 3 days. The crude product was purified by preparative HPLC (waters, acids (0.1% formic acid), acids, waters X-Select prep-C18, 5 μm, 30X100mm column, 0-100% MeCN/water) to give the title compound (G-4) as a white solid (13 mg,28 μmol,25%,99% purity). M/z 465.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.43 (s, 1H), 8.12 (d, j=7.6 hz, 1H), 8.03-7.98 (m, 1H), 7.84-7.77 (m, 1H), 7.76-7.68 (m, 2H), 7.64-7.55 (m, 2H), 7.28 (dq, j=4.5, 2.4hz, 2H), 7.18 (d, j=9.2 hz, 1H)),4.97(d,J=1.6Hz,2H),3.89(s,3H),3.16(s,3H)。
Example 3: synthesis of methyl 2'- (4-methyl-4H-1, 2, 4-triazol-3-yl) -5- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) - [1,1' -biphenyl ] -3-carboxylate (H-5)
Step 1: synthesis of 2'- (4-methyl-4H-1, 2, 4-triazol-3-yl) -5-nitro- [1,1' -biphenyl ] -3-carboxylic acid (H-2)
To a solution of (3- (methoxycarbonyl) -5-nitrophenyl) boronic acid (H-1) (138 mg,1.2 eq, 615. Mu. Mol) and intermediate D-5 (122 mg,1.0 eq, 512. Mu. Mol) in degassed 1, 4-dioxane (3 mL) and water (0.75 mL) was added Pd (dppf) Cl in sequence 2 DCM (41.8 mg,0.1 eq, 51.2. Mu. Mol) and K 2 CO 3 (212 mg,3 equivalents, 1.54 mmol) and stirred at 90℃overnight. The reaction mixture was diluted with EtOAc (10 mL) and with NH 4 Saturated aqueous solution of Cl (5 mL), naHCO 3 Is washed with saturated aqueous solution (5 mL) and brine (5 mL). The aqueous phase was treated with CHCl 3 IPA (7:3, 5X10 mL) extraction and drying of the combined organic extracts (MgSO 4 ) Filtered and concentrated in vacuo to give the sub-title compound (H-2) (106 mg,0.31mmol,61%,95% purity) as a pale yellow oil. M/z 325.3 (M+H) + (ES+);323.1(M-H) - (ES-)。 1 H NMR (400 MHz, methanol-d) 4 ) δ8.81 (s, 1H), 8.77 (dd, j=2.3, 1.4hz, 1H), 8.33 (t, j=2.0 hz, 1H), 8.20 (t, j=1.6 hz, 1H), 7.89 (ddd, j=7.8, 5.7,3.0hz, 1H), 7.83 (dt, j=7.8, 1.1hz, 1H), 7.78-7.75 (m, 2H), 3.42 (s, 3H). The exchangeable protons are not visible.
Step 2: synthesis of methyl 2'- (4-methyl-4H-1, 2, 4-triazol-3-yl) -5-nitro- [1,1' -biphenyl ] -3-carboxylate (H-3)
To a solution of the product (H-2) from step 1 (100 mg,1 eq, 308. Mu. Mol) in MeOH (25 mL) was added H 2 SO 4 (1.51 mg, 0.826. Mu.L, 0.05 eq, 15.4. Mu. Mol) and stirred at 80℃for 3 days. The reaction mixture was concentrated in vacuo and the crude product was passed throughPurification by silica gel chromatography (12 g cartridge, 0-10% MeOH/DCM) gave the sub-title compound (H-3) (90 mg,0.24mmol,78%,90% purity) as a dark brown oil. M/z 339.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d) 4 )δ8.74(dd,J=2.2,1.5Hz,1H),8.49(s,1H),8.31(t,J=2.0Hz,1H),8.19(t,J=1.6Hz,1H),7.87–7.77(m,2H),7.77–7.68(m,2H),3.98(s,3H),3.34(s,3H)。
Step 3: synthesis of methyl 5-amino-2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-carboxylate (H-4)
To a solution of the product (H-3) from step 2 (90 mg,90Wt%,1 eq, 0.24 mmol) in EtOH (3 mL) was added Pd/C39 (9 mg,10Wt%,0.04 eq, 8. Mu. Mol). The reaction mixture was subjected to H at 5atm at 26 ℃and 2 Stirred for 16 hours. The catalyst was filtered and the solvent removed in vacuo to give the sub-title compound (H-4) (73 mg,0.22mmol,94%,95% purity) as a pale yellow solid. M/z 309.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d) 4 ) Delta 8.42 (s, 1H), 7.77-7.71 (m, 1H), 7.65-7.59 (m, 3H), 7.34-7.27 (m, 1H), 7.12 (t, j=1.6 hz, 1H), 6.76 (dd, j=2.3, 1.7hz, 1H), 3.85 (s, 3H), 3.14 (s, 3H). The exchangeable protons are not visible.
Step 4: synthesis of methyl 2'- (4-methyl-4H-1, 2, 4-triazol-3-yl) -5- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) - [1,1' -biphenyl ] -3-carboxylate (H-5)
To a solution of product (H-4) (35 mg,1 eq, 0.11 mmol) from step 3 and intermediate A-3 (40 mg,1.2 eq, 0.14 mmol) in MeOH (2.5 mL) was added DIPEA (22 mg, 26. Mu.L, 1.5 eq, 0.17 mmol). The mixture was stirred at 60 ℃ for more than 5 days. The solvent was removed in vacuo and the crude product was purified by silica gel chromatography (12 g cartridge, 0-20% meoh/DCM) to give the title compound (H-5) (14 mg,27 μmol,24%,95% purity) as a pale yellow solid. M/z 493.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d) 4 )δ8.54(dd,J=2.2,1.4Hz,1H),8.46(s,1H),8.13(d,J=7.6Hz,1H),8.05–7.99(m,2H),7.85–7.79(m,3H),7.77(t,J=1.5Hz,1H),7.73–7.69(m,2H),5.15(s,2H),3.97(s,3H),3.34(s,3H)。
Example 4: synthesis of 2'- (4-methyl-4H-1, 2, 4-triazol-3-yl) -5- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) - [1,1' -biphenyl ] -3-carboxylic acid (I-1)
To a solution of methyl compound H-5 (10 mg,1 eq, 20. Mu. Mol) in MeOH (2 mL) was added NaOH (0.97 mg, 24. Mu.L, 1 mol, 1.2 eq, 24. Mu. Mol) and stirred at room temperature for 5 days. The reaction mixture was diluted with water (10 mL) and extracted with EtOAc (5 mL). The aqueous layers were combined, acidified with 1M aqueous HCl and extracted with DCM (3×10 mL). The organic extracts were combined, washed with brine (10 mL), dried (MgSO 4), and concentrated in vacuo to give the title compound (I-1) (7 mg,0.01mmol, 70%) as a white solid. M/z479.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ13.15(s,1H),8.59(s,1H),8.40(s,1H),8.08(dd,J=16.7,7.7Hz,2H),7.85(s,1H),7.81(t,J=7.7Hz,1H),7.73(s,2H),7.68–7.59(m,2H),7.49(d,J=1.5Hz,1H),5.16(s,2H),3.14(s,3H)。
Example 5: synthesis of 2- (4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (trifluoromethyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (J-4)
Step 1: synthesis of 4- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -6- (trifluoromethyl) pyridin-2-amine (J-2)
To a solution of 6- (trifluoromethyl) pyridin-2-amine (J-1) (162 mg,1 eq, 999. Mu. Mol) in THF (1 mL) was added 4, 5-tetramethyl-1, 3, 2-dioxaborolan (153 mg, 174. Mu.L, 1.2 eq, 1.20 mmol) and stirred at room temperature for 1 hour. Adding [ Ir (OMe) COD ]]2 (9.94 mg,0.015 eq, 15.0. Mu. Mol), 3,4,7, 8-tetramethyl-1, 10-phenanthroline (7.08 mg,0.03 eq, 30.0. Mu. Mol) and 4,4', 5' -octamethyl-2, 2' -bis (1, 3, 2-dioxaborolan) 127mg,0.5 eq, 500. Mu. Mol) and the mixture was stirred at 80℃for 8 hours. The mixture was cooled to room temperature, diluted with MeOH (5 mL) and concentrated in vacuo. The residue was dissolved in DCM, passed through a silica gel plug and washed with DCM (350 mL). The washings were concentrated in vacuo to give the sub-title compound (J-2) (80 mg,0.27mmol,28%,99% purity) as a white solid. M/z 288.9 (M+H) + (ES+)。 1 H NMR (400 MHz, chloroform-d) delta 7.34 (s, 1H), 7.05 (s, 1H), 1.35 (s, 12H). No exchangeable protons were observed.
Step 2: synthesis of 4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (trifluoromethyl) pyridin-2-amine (J-3)
To a solution of the product (J-2) (70.0 mg,1.0 eq, 243. Mu. Mol) from step 1 and intermediate D-5 (57.9 mg,1.0 eq, 243. Mu. Mol) in degassed 1, 4-dioxane (2 mL) and water (0.5 mL) was added Pd (dppf) Cl in sequence 2 DCM (19.8 mg,0.1 eq, 24.3. Mu. Mol) and K 2 CO 3 (101 mg,3 eq, 729. Mu. Mol) and stirred at 90℃overnight. The reaction mixture was concentrated in vacuo and the crude product was purified by silica gel chromatography (12 g cartridge, 0-30% MeOH/DCM) to give the sub-title compound (J-3) (62 mg,0.16mmol,64%,80% purity) as a pale green solid. M/z 320.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 7.82-7.75 (m, 1H), 7.73-7.63 (m, 3H), 6.69 (d, j=1.3 hz, 1H), 6.58 (d, j=0.7 hz, 1H), 3.31 (s, 3H). No exchangeable protons were observed.
Step 3: synthesis of 2- (4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (trifluoromethyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (J-4)
To a solution of product (J-3) (30 mg,1 eq, 94. Mu. Mol) from step 2 and intermediate A-3 (33 mg,1.2 eq, 0.11 mmol) in MeOH (1 mL) was added DIPEA (18 mg, 21. Mu.L, 1.5 eq, 0.14 mmol) and the mixture was stirred at 60℃for 3 days. The solvent was removed in vacuo and the crude product was purified by silica gel chromatography (12 g cartridge, 0-40% meoh/DCM) to give 90% pure product. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic water XBridge BEH C18 ODB prep columnPurification of 5 μm, 30mm X100 mm, 0-100% MeCN/water) to give the title compound (J-4) (8 mg,0.02mmol,20%,99% purity) as a pale yellow solid. M/z 504.5 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.52(d,J=8.5Hz,1H),8.15(d,J=2.0Hz,1H),7.95(d,J=7.0Hz,1H),7.92–7.84(m,2H),7.77(ddd,J=9.1,7.3,1.5Hz,2H),7.32(dd,J=8.5,7.1Hz,1H),6.84(d,J=1.4Hz,1H),6.62(s,2H),6.42(s,1H),3.86(s,3H)。
Example 6: synthesis of 2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) -5- (trifluoromethyl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (K-3)
Step 1: synthesis of 2'- (4-methyl-4H-1, 2, 4-triazol-3-yl) -5- (trifluoromethyl) - [1,1' -biphenyl ] -3-amine (K-2)
To 3-bromo-5- (trifluoromethyl) aniline (K-1) (100 mg, 58.9. Mu.L, 1 eq, 417. Mu. Mol) and B 2 (pin) 2 (116 mg,1.1 eq, 458. Mu. Mol) in degassed 1, 4-dioxane (1.5 mL) was added Pd (dppf) Cl in sequence 2 DCM (34.0 mg,0.1 eq, 41.7. Mu. Mol) and KOAc (123 mg,3 eq, 1.25 mmol) and stirred at 100℃for 2 hours. The reaction mixture was cooled to room temperature and intermediate D-5 (99.2 mg,1 eq, 417 μmol) was added to 1, 4-dioxane (1.5 mL) and K 2 CO 3 (173 mg,3 eq, 1.25 mmol) in water (0.75 mL). The reaction mixture was stirred at 100 ℃ overnight. The reaction mixture was concentrated in vacuo and the crude product was purified by silica gel chromatography (12 g cartridge, 0-30% MeOH/DCM) to give the sub-title compound (K-2) (81 mg,0.25mmol,60%,98% purity) as a pale green solid. M/z 319.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.41(s,1H),7.70–7.64(m,1H),7.61–7.51(m,3H),6.76(s,1H),6.63(t,J=1.9Hz,1H),6.37(s,1H),5.64(s,2H),3.05(s,3H)。
Step 2: synthesis of 2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) -5- (trifluoromethyl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (K-3)
To a solution of product (K-2) (32 mg,1.5 eq, 0.10 mmol) and intermediate A-3 (20 mg,1 eq, 67. Mu. Mol) from step 1 above in DMF (1 mL) was added DIPEA (13 mg, 15. Mu.L, 1.5 eq, 0.10 mmol) and stirred at 60℃for more than 2 days. The reaction mixture was concentrated in vacuo and the crude product was purified by Jin Guijiao chromatography (12 g cartridge, 0-20% meoh/DCM) followed by preparative HPLC (wok, acid (0.1% formic acid), acid, wok X-Select CSH C18 ODB, 5 μm, 30X100mm column, 0-100% mecn/water) to give the title compound (K-3) (3 mg,4 μmol,6%,65% purity) as a colorless oil. M/z 503.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.45 (s, 1H), 8.40 (s, 1H), 8.15 (d, j=7.5 hz, 1H), 8.03 (d, j=7.8 hz, 1H), 7.95 (s, 1H), 7.82 (p, j=4.2 hz, 3H), 7.73-7.65 (m, 2H), 7.36 (s, 1H), 5.18 (s, 2H), 3.31 (s, 3H).
Example 7: synthesis of 2- (4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -1H-pyrrolo [2,3-b ] pyridin-6-yl) -4- (trifluoromethyl) isoindolin-1-one (L-5)
Step 1: synthesis of 4-bromo-1H-pyrrolo [2,3-b ] pyridine 7-oxide (L-2)
At 0℃to 4-bromo-1H-pyrrolo [2,3-b ]]To a solution of pyridine (L-1) (1.00 g,1 eq, 5.08 mmol) in DCM (25 mL) was added 3-chlorophenylperoxyacid (1.88 g,70Wt%,1.5 eq, 7.61 mmol). The mixture was slowly warmed to room temperature and stirred overnight. The reaction mixture was diluted with DCM (10 mL) and NaHCO 3 Saturated aqueous solution of (a) and Na 2 S 2 O 3 Is washed with a 50/50 mixture of saturated aqueous solutions (3X 10 mL) and brine (10 mL). The combined organic layers were dried (MgSO 4 ) Filtered and concentrated in vacuo to give the sub-title compound (L-2) (520 mg,2.4mmol,48%,99% purity) as a pale brown solid. M/z 213.1/215.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.13 (d, j=6.6 Hz,1H) 7.61 (d, j=3.5 hz,1 h), 7.44 (d, j=6.6 hz,1 h), 6.68 (d, j=3.5 hz,1 h). No exchangeable protons were observed.
Step 2: synthesis of 4-bromo-1H-pyrrolo [2,3-b ] pyridin-6-amine (L-3)
To a solution of the product (L-2) (719 mg,1 eq, 2.44 mmol) from step 1 above in pyridine (35 mL) was added 4-methylbenzenesulfonyl chloride (604 mg,1.3 eq, 3.17 mmol) and stirred at room temperature overnight. Pyridine was removed under vacuum and 2-aminoethan-1-ol (10.4 g,10.3ml,70 equivalents, 171 mmol) was added to the residue and stirred at room temperature for 1 hour. The reaction mixture was diluted with DCM (25 mL) and water (25 mL) and extracted with DCM (5X 10 mL). The combined organic layers were dried (MgSO 4 ) Filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (40 g cartridge, 0-10% MeOH/DCM) to give the sub-title compound (L-3) as a pale brown solid (300 mg,1.4mmol,57%,99% purity). M/z 212.1/214.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ11.13(s,1H),7.01(dd,J=3.5,2.4Hz,1H),6.50(s,1H),6.12(dd,J=3.4,2.1Hz,1H),5.82(s,2H)。
Step 3: synthesis of 2- (4-bromo-1H-pyrrolo [2,3-b ] pyridin-6-yl) -4- (trifluoromethyl) isoindolin-1-one (L-4)
To a solution of product (L-3) (50 mg,1 eq, 0.24 mmol) and intermediate A-3 (84 mg,1.2 eq, 0.28 mmol) from step 2 above in DMF (2 mL) was added DIPEA (61 mg, 72. Mu.L, 2.0 eq, 0.47 mmol) and stirred overnight at 60 ℃. The reaction mixture was cooled to room temperature and concentrated in vacuo. The solid was washed with a small amount of DCM, filtered and dried to give the sub-title compound (L-4) (61 mg,0.15mmol,65%,99% purity) as a light brown solid. M/z 396.1/398.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ12.14(s,1H),8.63(s,1H),8.14(d,J=7.7Hz,1H),8.07(dd,J=8.0,3.1Hz,1H),7.81(t,J=7.7Hz,1H),7.53(d,J=3.7Hz,1H),6.42(d,J=3.5Hz,1H),5.32(s,2H)。
Step 4: synthesis of 2- (4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -1H-pyrrolo [2,3-b ] pyridin-6-yl) -4- (trifluoromethyl) isoindolin-1-one (L-5)
To the product (L-4) (20 mg,1.0 equivalent, 50. Mu. Mol) from the above step 3 and B 2 (pin) 2 (14 mg,1.1 eq, 56. Mu. Mol) to a solution in degassed 1, 4-dioxane (1 mL) was added Pd (dppf) Cl in sequence 2 DCM (4.1 mg,0.1 eq, 5.0. Mu. Mol) and KOAc (15 mg,3 eq, 0.15 mmol). The mixture was stirred at 100℃for 2 hours. The reaction mixture was cooled to room temperature and a solution of intermediate D-5 (14 mg,1.2 eq, 61 μmol) in 1, 4-dioxane (1 mL) was added followed by K 2 CO 3 (21 mg,3 eq, 0.15 mmol) in water (0.5 mL). The reaction mixture was stirred at 100 ℃ overnight. The reaction mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by silica gel chromatography (12 g cartridge, 0-20% MeOH/DCM) followed by recrystallization in EtOH to give the title compound (L-5) (7 mg,0.01mmol,20%,99% purity) as a clear white solid. M/z 475.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ11.84(s,1H),8.33(s,1H),8.13–8.00(m,3H),7.84–7.65(m,5H),7.37(t,J=2.9Hz,1H),6.17(dd,J=3.6,1.7Hz,1H),5.32(s,2H),3.30(s,3H)。
Example 8: synthesis of 2- (1-ethyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -1H-pyrrolo [2,3-b ] pyridin-6-yl) -4- (trifluoromethyl) isoindolin-1-one (M-2)
Step 1: synthesis of 2- (4-bromo-1-ethyl-1H-pyrrolo [2,3-b ] pyridin-6-yl) -4- (trifluoromethyl) isoindolin-1-one (M-1)
To a solution of intermediate L-4 (30 mg,1 eq, 76. Mu. Mol) in DMF (1 mL) was added NaH (3.2 mg,60Wt%,1.05 eq, 80. Mu. Mol) at 0deg.C, and the suspension was stirred at 0deg.C for 1 hour. Iodoethane (12 mg,6.4 μl,1.05 eq, 80 μmol) was added dropwise at 0deg.C. The resulting mixture was slowly warmed to room temperature and stirred at this temperature for 1 hour. The solution was quenched with water and extracted with EtOAc (10 mL). The organic extract is treated with NH 4 Saturated aqueous solution of Cl (5 mL), naHCO 3 Saturated aqueous solution (5 mL)And brine (5 mL). The combined organic extracts were dried (MgSO 4 ) Filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (12 g cartridge, 0-30% EtOAc/isohexane) to give the sub-title compound (M-1) (21 mg,47 μmol,62%,95% purity) as a clear white solid. M/z 424.0and 426.3 (M+H) + (ES+)。 1 H NMR (400 MHz, chloroform-d) δ8.71 (s, 1H), 8.14 (d, j=7.6 hz, 1H), 7.87 (d, j=7.7 hz, 1H), 7.66 (t, j=7.7 hz, 1H), 7.23 (d, j=3.5 hz, 1H), 6.49 (d, j=3.5 hz, 1H), 5.33 (s, 2H), 4.32 (q, j=7.3 hz, 2H), 1.52 (t, j=7.3 hz, 3H).
Step 2: synthesis of 2- (1-ethyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -1H-pyrrolo [2,3-b ] pyridin-6-yl) -4- (trifluoromethyl) isoindolin-1-one (M-2)
To the product (M-1) (21 mg,1.0 equivalent, 50. Mu. Mol) from step 1 above and B 2 (pin) 2 (14 mg,1.1 eq, 54. Mu. Mol) to a solution in degassed 1, 4-dioxane (1 mL) was added Pd (dppf) Cl in sequence 2 DCM (4.0 mg,0.1 eq, 5.0. Mu. Mol) and KOAc (15 mg,3 eq, 0.15 mmol) and stirred at 100℃for 2 hours. The reaction mixture was cooled to room temperature and a solution of intermediate D-5 (14 mg,1.2 eq, 59 μmol) in 1, 4-dioxane (1 mL) was added followed by K 2 CO 3 (21 mg,3 eq, 0.15 mmol) in water (0.5 mL). The reaction mixture was stirred at 100 ℃ overnight. The crude product was purified by RP flash C18 chromatography (12 g cartridge, 15-100% (0.1% formic acid/MeCN)/(0.1% formic acid/water)) to give the title compound (M-2) (1.81 mg,3.4 μmol,5.1%,95% purity) as a pale yellow solid. M/z 503.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.38 (s, 1H), 8.12 (d, j=7.7 hz, 1H), 8.03 (s, 1H), 8.00 (d, j=7.7 hz, 1H), 7.91-7.87 (m, 1H), 7.86-7.78 (m, 2H), 7.77-7.70 (m, 2H), 7.41 (d, j=3.6 hz, 1H), 6.39 (d, j=3.6 hz, 1H), 5.39 (s, 2H), 4.38 (q, j=7.2 hz, 2H), 3.41 (s, 3H), 1.52 (t, j=7.3 hz, 3H).
Example 9: synthesis of 2- (5- (hydroxymethyl) -2'- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (N-5)
Step 1: synthesis of 2'- (4-methyl-4H-1, 2, 4-triazol-3-yl) -5-nitro- [1,1' -biphenyl ] -3-carboxylic acid (N-2)
To a solution of (3- (methoxycarbonyl) -5-nitrophenyl) boronic acid (N-1) (227 mg,1.2 eq, 1.01 mmol) and intermediate D-5 (200 mg,1.0 eq, 840. Mu. Mol) in degassed 1, 4-dioxane (4 mL) and water (1 mL) was added Pd (dppf) Cl in sequence 2 DCM (68.6 mg,0.1 eq, 84.0. Mu. Mol) and K 2 CO 3 (348 mg,3 eq., 2.52 mmol). The mixture was stirred at 90 ℃ overnight. The reaction mixture was diluted with DCM (5 mL) and 1M NaOH (10 mL) and extracted with DCM (3X 5 mL). The aqueous extracts were combined and acidified with concentrated HCl and extracted with DCM/IPA (70:30) (5X 10 mL). The combined organic layers were dried (MgSO 4 ) Filtered and concentrated in vacuo. The crude product was purified by RP flash C18 chromatography (12 g cartridge, 15-65% (0.1% formic acid/MeCN)/(0.1% formic acid/water)) to give the sub-title compound (N-2) as a clear white solid (255 mg,0.72mmol,86%,92% purity). M/z 325.3 (M+H) + (ES+);323.1(M-H) - (ES-). 1 H NMR(400MHz,DMSO-d6)δ13.74(s,1H),8.52(dd,J=2.2,1.4Hz,1H),8.43(s,1H),8.15(t,J=2.0Hz,1H),8.02(t,J=1.6Hz,1H),7.78–7.73(m,2H),7.69–7.63(m,2H),3.19(s,3H)。
Step 2: synthesis of (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) -5-nitro- [1,1' -biphenyl ] -3-yl) methanol (N-3)
To a solution of the product (N-2) (235 mg,1 eq., 725. Mu. Mol) from step 1 above in anhydrous THF (25 mL) at 0deg.C was added BH 3 THF (311 mg,3.62mL,1 mole, 5 equivalents, 3.62 mmol). The resulting mixture was slowly warmed to room temperature and taken up in N 2 Stirring is carried out for 16 hours under an atmosphere at this temperature. The mixture was slowly quenched with MeOH and concentrated in vacuo. The crude product was purified by RP flash C18 chromatography (12 g cartridge, 15-75% (0.1% formic acid/MeCN)/(0.1% formic acid/water)) to give the sub-title compound (N-3) (16 mg,51 μmol, 7.0) as a clear white solid% purity, 98%). M/z 311.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.48-8.39 (m, 1H), 8.21 (s, 1H), 7.96-7.90 (m, 1H), 7.77 (ddt, J=17.5, 7.8,2.9Hz, 2H), 7.71-7.57 (m, 3H), 4.68 (s, 2H), 3.28-3.21 (m, 3H). No exchangeable protons were observed.
Step 3: synthesis of (5-amino-2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) methanol (N-4)
To a solution of the product (N-3) from step 2 above (15 mg,1 eq, 48. Mu. Mol) in EtOH (1 mL) was added Pd/C39 (2 mg,10Wt%,0.04 eq, 2. Mu. Mol). The reaction mixture was subjected to H at 5atm at 45 DEG C 2 Stirred for 16 hours. The catalyst was filtered and the solvent removed in vacuo to give the sub-title compound (N-4) (15 mg,48 μmol,100%,90% purity) as a pale yellow solid. M/z 281.6 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.37 (s, 1H), 7.70 (ddd, j=8.5, 6.7,2.0hz, 1H), 7.65-7.62 (m, 1H), 7.61-7.54 (m, 2H), 6.71 (t, j=1.8 hz, 1H), 6.50 (d, j=1.6 hz, 1H), 6.45 (t, j=2.0 hz, 1H), 4.44 (s, 2H), 3.08 (s, 3H). Three exchangeable protons were not observed.
Step 4: synthesis of 2- (5- (hydroxymethyl) -2'- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (N-5)
To a solution of the product (N-4) (16 mg,1 eq, 57. Mu. Mol) from step 3 above and intermediate A-3 (20 mg,1.2 eq, 68. Mu. Mol) in DMF (1 mL) was added DIPEA (11 mg, 13. Mu.L, 1.5 eq, 86. Mu. Mol) and stirred at 60℃for 3 days. The solvent was removed in vacuo and the crude product was purified by RP flash C18 chromatography (4 g cartridge, 15-65% (0.1% formic acid/MeCN)/(0.1% formic acid/water)) to give the title compound (N-5) (1.96 mg,3.8 μmol,6.7%,90% purity) as a pale yellow solid. M/z 465.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.40 (s, 1H), 8.12 (d, j=7.6 hz, 1H), 8.00 (d, j=7.6 hz, 1H), 7.89 (s, 1H), 7.81 (d, j=7.9 hz, 1H), 7.79-7.74 (m, 2H), 7.66-7.63 (m, 3H), 7.15 (s, 1H), 5.10 (s, 2H), 4.65 (s, 2H), 3.24 (s, 3H). No exchangeable protons were observed.
Example 10: synthesis of N-methyl-2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) -5- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) - [1,1' -biphenyl ] -3-carboxamide (O-3)
Step 1: synthesis of N-methyl-2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) -5-nitro- [1,1' -biphenyl ] -3-carboxamide (O-1)
To a solution of intermediate N-2 (100 mg,1 eq, 308. Mu. Mol) in DMF (5 mL) was added HATU (129 mg,1.1 eq, 339. Mu. Mol) and NEt 3 (93.6 mg, 129. Mu.L, 3 equivalents, 925. Mu. Mol). The mixture was stirred at room temperature for 20 minutes, then methylamine (14.4 mg,231 μl,2 mol, 1.5 eq, 463 μl) was slowly added. The mixture was stirred at room temperature for 16 hours. The solvent was removed in vacuo and the crude product was purified by silica gel chromatography (12 g cartridge, 0-20% meoh/DCM) to give the sub-title compound (O-1) (55.3 mg,0.16mmol,51%,95% purity) as a pale yellow solid. M/z 337.9 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol l-d 4) delta 8.64 (t, j=1.9 hz, 1H), 8.45 (s, 1H), 8.13 (dt, j=4.3, 1.7hz, 2H), 7.86-7.77 (m, 2H), 7.76-7.68 (m, 2H), 3.01 (s, 3H), 2.96 (s, 3H). No exchangeable protons were observed.
Step 2: synthesis of 5-amino-N-methyl-2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-carboxamide (O-2)
To a solution of the product (O-1) from step 1 above (76 mg,90Wt%,1 eq, 0.20 mmol) in EtOH (3 mL) was added Pd/C39 (8 mg,10Wt%,0.04 eq, 8. Mu. Mol). The reaction mixture was subjected to H at 5atm at 45 DEG C 2 Stirred for 16 hours. The catalyst was filtered and the solvent removed in vacuo to give the sub-title compound (O-2) (65 mg,0.19mmol,94%,90% purity) as a pale yellow solid. M/z 308.4 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4). δ8.39 (s, 1H), 7.73 (ddd, j=7.8, 5.2,3.4hz, 1H), 7.66 (dt, j=7.8, 1.0hz, 1H), 7.64-7.56 (m, 2H), 7.05 (t, j=1.9 hz, 1H), 6.96 (t, j=1.6 hz, 1H), 6.58 (t, j=1.9 hz, 1H), 3.13 (s, 3H), 2.89 (s, 3H). Not yetThree exchangeable protons are observed.
Step 3: synthesis of N-methyl-2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) -5- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) - [1,1' -biphenyl ] -3-carboxamide (O-3)
To a solution of product (O-2) (65 mg,1 eq, 0.21 mmol) and intermediate A-3 (75 mg,1.2 eq, 0.25 mmol) from step 2 above in DMF (2.5 mL) was added DIPEA (41 mg, 48. Mu.L, 1.5 eq, 0.32 mmol). The mixture was stirred at 60℃for 3 days. The solvent was removed in vacuo and the crude product purified by RP flash C18 chromatography (12 g cartridge, 15-65% MeCN/10mM ammonium bicarbonate) to give the title compound (O-3) (7 mg,0.01mmol,7%,99% purity) as a white solid. M/z 492.4 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.43 (s, 1H), 8.22 (t, j=1.8 hz, 1H), 8.12 (d, j=7.7 hz, 1H), 8.01 (d, j=7.7 hz, 1H), 7.94 (t, j=1.9 hz, 1H), 7.83-7.77 (m, 3H), 7.67 (dd, j=3.8, 2.8hz, 2H), 7.64 (t, j=1.6 hz, 1H), 5.13 (s, 2H), 3.36 (s, 3H), 2.97 (s, 3H). No exchangeable protons were observed.
Example 11: synthesis of 6- (hydroxymethyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (P-2)
DIPEA (159 mg, 214. Mu.L, 1.5 eq, 1.38 mmol) was added to 2'- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ]]A solution of 3-amine (P-1) (230 mg,1 eq, 917. Mu. Mol) D-5 and intermediate B-4 (300 mg,1 eq, 917. Mu. Mol) in EtOH (16 mL) was stirred at 60℃for 18 hours, then at room temperature for 16 hours. The reaction mixture was cooled down and the precipitate was filtered to give the title compound (P-2) (167 mg,360 μmol, 39.2%) as a white solid. M/z 465.2 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.39(s,1H),8.02–7.97(m,2H),7.95(s,1H),7.75–7.65(m,3H),7.62–7.56(m,2H),7.37(t,J=8.0Hz,1H),6.90(d,J=7.7Hz,1H),5.58(t,J=5.8Hz,1H),5.09(s,2H),4.71(d,J=5.8Hz,2H),3.08(s,3H)。
Example 12: synthesis of (S) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6- ((3-methylpiperidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (R-2)
DCM (1 mL) containing (S) -3-methylpiperidine, HCl (R-1) (7.1 mg,1.1 eq, 52. Mu. Mol) and DIPEA (18 mg, 25. Mu.L, 3.0 eq, 0.14 mmol) was added to DCM (1 mL) containing intermediate Q-1 (22 mg,1 eq, 48. Mu. Mol) followed by NaBH (OAc) 3 (20 mg,2.0 equivalents, 95. Mu. Mol). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with DCM (10 mL) and NaHCO 3 Saturated aqueous (10 mL) was washed. The organic extract was dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column 0-100% MeCN/water) to give the title compound (R-2) as a white solid (3.5 mg,6.4 μmol,13%,99% purity). M/z 546.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.40 (s, 1H), 8.07 (s, 1H), 7.99 (s, 1H), 7.89 (ddd, j=8.2, 2.3,0.9hz, 1H), 7.80-7.73 (m, 3H), 7.67-7.61 (m, 2H), 7.46 (t, j=8.0 hz, 1H), 7.13 (ddd, j=7.8, 1.0hz, 1H), 5.08 (d, j=1.7 hz, 2H), 3.72 (s, 2H), 3.22 (s, 3H), 2.91-2.80 (m, 2H), 2.08-1.97 (m, 1H), 1.82-1.57 (m, 5H), 1.00-0.87 (m, 4H).
Example 13: synthesis of (R) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6- ((3-methylpiperidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (S-2)
DCM (1 mL) containing (R) -3-methylpiperidine, HCl (S-1) (7.1 mg,1.1 eq, 52. Mu. Mol) and DIPEA (18 mg, 25. Mu.L, 3.0 eq, 0.14 mmol) was added to the reaction mixture containing intermediate Q- 1 (22 mg,1 eq, 48. Mu. Mol) in DCM (1 mL) followed by NaBH (OAc) 3 (20 mg,2.0 equivalents, 95. Mu. Mol). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with DCM (10 mL) and NaHCO 3 Saturated aqueous (10 mL) was washed. The organic extract was dried (phase separator) and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column 0-100% MeCN/water) to give the title compound (S-2) as a white solid (4 mg,48 μmol,20%,99% purity). At 2.58 minutes, M/z546.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.40 (s, 1H), 8.07 (s, 1H), 7.99 (s, 1H), 7.89 (ddd, j=8.3, 2.3,0.9hz, 1H), 7.80-7.73 (m, 3H), 7.67-7.62 (m, 2H), 7.46 (t, j=8.0 hz, 1H), 7.13 (dt, j=7.9, 1.1hz, 1H), 5.08 (d, j=1.6 hz, 2H), 3.72 (s, 2H), 3.22 (s, 3H), 2.91-2.79 (m, 2H), 2.08-1.97 (m, 1H), 1.80-1.57 (m, 5H), 1.01-0.87 (m, 4H).
Example 14: synthesis of 6- ((((1S, 2S) -2-hydroxycyclopentyl) amino) methyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (T-2)
DCM (1 mL) containing (1S, 2S) -2-aminocyclopentane-1-ol, HCl (T-1) (10 mg,1.1 eq, 76. Mu. Mol) and DIPEA (27 mg, 36. Mu.L, 3.0 eq, 0.21 mmol) was added to DCM (1 mL) containing intermediate Q-1 (40 mg,80Wt%,1 eq, 69. Mu. Mol), followed by NaBH (OAc) 3 (29 mg,2.0 equivalents, 0.14 mmol). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with DCM (10 mL) and NaHCO 3 Saturated aqueous (10 mL) was washed. The organic phase was dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (woth, basic (0.3% ammonia), basic, woth XBridge BEH C18 ODB, 5 μm, 30x100mm column 0-100% MeCN/water) to give the title compound (T-2) as a white solid (8 mg,0.01mmol,20%99% purity). M/z 548.2 (M+H) + (ES+). 1 H NMR (400 mhz, dmso-d 6) delta 8.39 (s, 1H), 8.04 (s, 1H), 8.00 (d, j=7.9 hz, 2H), 7.75-7.65 (m, 3H), 7.60-7.56 (m, 2H), 7.37 (t, j=8.0 hz, 1H), 6.94-6.88 (m, 1H), 5.07 (s, 2H), 4.52 (d, j=4.2 hz, 1H), 3.91 (s, 2H), 3.80 (dt, j=8.5, 4.3hz, 1H), 3.08 (s, 3H), 2.78-2.69 (m, 1H), 1.87-1.75 (m, 2H), 1.61-1.52 (m, 2H), 1.44-1.35 (m, 1H), 1.35-1.24 (m, 1H). No exchangeable protons were observed.
Example 15: synthesis of ((3-fluoroazetidin-1-yl) methyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (U-2)
To a solution of 3-fluoroazetidine, HCl (U-1) (8.5 mg,1.1 eq, 76. Mu. Mol) and DIPEA (27 mg, 36. Mu.L, 3.0 eq, 0.21 mmol) in DCM (2 mL) was added intermediate Q-1 (40 mg,80wt%,1 eq, 69. Mu. Mol) followed by NaBH (OAc) 3 (29 mg,2.0 equivalents, 0.14 mmol). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with DCM (10 mL) and NaHCO 3 Saturated aqueous (10 mL) was washed. The organic phase was dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (U-2) as a white solid (4.6 mg,8.7 μmol,13%,99% purity). M/z 522.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.39(s,1H),8.00–7.95(m,2H),7.91(s,1H),7.75–7.64(m,3H),7.62–7.56(m,2H),7.37(t,J=8.0Hz,1H),6.91(d,J=7.4Hz,1H),5.21(dq,J=57.8,5.2Hz,1H),5.08(s,2H),3.85(s,2H),3.64–3.53(m,2H),3.26–3.20(m,1H),3.20–3.14(m,1H),3.08(s,3H)。
Example 16: synthesis of (S) -6- ((3-fluoropyrrolidin-1-yl) methyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (V-2)
DCM (1 mL) containing (S) -3-fluoropyrrolidine, HCl (V-1) (9.6 mg,1.1 eq., 76. Mu. Mol) and DIPEA (27 mg, 36. Mu.L, 3.0 eq., 0.21 mmol) was added to DCM (1 mL) containing intermediate Q-1 (40 mg,80Wt%,1 eq., 69. Mu. Mol) followed by NaBH (OAc) 3 (29 mg,2.0 equivalents, 0.14 mmol). The reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with DCM (10 mL) and NaHCO 3 Saturated aqueous (10 mL) was washed. The organic phase was dried (phase separator) and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (V-2) as a white solid (7.2 mg,13 μmol,19%,99% purity). M/z 536.3 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.39(s,1H),8.02–7.97(m,2H),7.95(s,1H),7.76–7.65(m,3H),7.62–7.56(m,2H),7.38(t,J=8.0Hz,1H),6.91(dd,J=7.7,1.5Hz,1H),5.32–5.12(m,1H),5.09(s,2H),3.84(s,2H),3.08(s,3H),2.87–2.76(m,2H),2.74–2.59(m,1H),2.42–2.34(m,1H),2.25–2.07(m,1H),1.99–1.81(m,1H)。
Example 17: synthesis of 6- ((((1R, 2R) -2-hydroxycyclopentyl) amino) methyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (W-2)
DCM (1 mL) containing (1R, 2R) -2-aminocyclopentan-1-ol, HCl (W-1) (10 mg,1.1 eq, 76. Mu. Mol) and DIPEA (27 mg, 36. Mu.L, 3.0 eq, 0.21 mmol) was added to DCM (1 mL) containing intermediate Q-1 (40 mg,80wt%,1 eq, 69. Mu. Mol) followed by NaBH (OAc) 3 (29 mg,2.0 equivalents, 0.14 mmol). The reaction mixture was stirred at room temperature overnight. Additional NaBH (OAc) was added 3 (29 mg,2.0 equivalents, 0.14 mmol) and the reaction was stirred for an additional 2 hours. The reaction mixture was diluted with DCM (10 mL) and NaHCO 3 Is washed with a saturated aqueous solution (10 mL). The organic phase was dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% mecn/water) to give the title compound (W-2) as a white solid (6.36 mg,11 μmol,17%,99% purity). M/z 548.2 (M+H) + (ES+). 1 H NMR (400 mhz, dmso-d 6) delta 8.39 (s, 1H), 8.04 (s, 1H), 8.01-7.95 (m, 2H), 7.76-7.65 (m, 3H), 7.61-7.55 (m, 2H), 7.38 (t, j=8.0 hz, 1H), 6.93-6.88 (m, 1H), 5.08 (s, 2H), 4.51 (d, j=4.1 hz, 1H), 3.91 (s, 2H), 3.79 (q, j=5.2, 4.5hz, 1H), 3.08 (s, 3H), 2.73 (q, j=5.7 hz, 1H), 1.87-1.75 (m, 2H), 1.57 (p, j=7.4 hz, 2H), 1.45-1.35 (m, 1H), 1.36-1.25 (m, 1H). No exchangeable protons were observed.
Example 18: synthesis of (R) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6- ((2-methylmorpholino) methyl) -4- (trifluoromethyl) isoindolin-1-one (X-2)
To a solution of (R) -2-methylmorpholino, HCl (X-1) (14 mg,1.5 eq, 0.10 mmol) and DIPEA (27 mg, 36. Mu.L, 3.0 eq, 0.21 mmol) in DCM (2 mL) was added intermediate Q-1 (40 mg,80Wt%,1 eq, 69. Mu. Mol) and the reaction mixture was stirred for 30 min. Adding NaBH (OAc) 3 (29 mg,2.0 equivalents, 0.14 mmol) and the reaction mixture was stirred at 40℃overnight. The reaction mixture was diluted with DCM (10 mL) and NaHCO 3 Is washed with a saturated aqueous solution (10 mL). The organic phase was dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters X-Bridge BEH C18 ODB, 5 μm, 30X100mm column, 0-100% MeCN/water) to give the title compound (X-2) as a white solid (4.8 mg,8.3 μmol,12%,95% purity). m/z 548.3(M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.39(s,1H),7.99(d,J=7.0Hz,2H),7.94(s,1H),7.75–7.64(m,3H),7.63–7.56(m,2H),7.38(t,J=8.0Hz,1H),6.91(d,J=7.7Hz,1H),5.08(s,2H),3.79–3.70(m,1H),3.68(s,2H),3.57–3.46(m,2H),3.08(s,3H),2.66(dd,J=27.2,11.3Hz,2H),2.14–2.05(m,1H),1.84–1.74(m,1H),1.03(d,J=6.2Hz,3H)。
Example 19: synthesis of (R) -6- ((3-fluoropyrrolidin-1-yl) methyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (Y-2)
To (R) -3-fluoropyrrolidine-HCl (Y-1) (8.1 mg,1.2 eq, 65. Mu. Mol) and DIPEA (21 mg, 28. Mu.L, 3.0 eq, 0.16 mmol) in CHCl 3 To a solution in (4 mL) was added intermediate Q-1 (25 mg,1 eq, 0.054 mmol) and the reaction mixture was stirred for 30 min. Adding NaBH (OAc) 3 (23 mg,2 equivalents, 0.11 mmol) and the reaction mixture was stirred at 40℃overnight. The crude product was taken up with NaHCO 3 (5 mL) quench and add DCM (10 mL). The organic phase was extracted, dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (Y-2) as a tan solid (5 mg,9 μmol,20%,99% purity). M/z 536.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.39(s,1H),8.11–7.88(m,3H),7.77–7.64(m,3H),7.63–7.54(m,2H),7.38(dd,J=9.4,6.7Hz,1H),6.91(d,J=7.8Hz,1H),5.32–5.13(m,1H),5.09(s,2H),3.84(s,2H),3.09(s,3H),2.89–2.74(m,2H),2.74–2.60(m,1H),2.42–2.30(m,1H),2.25–2.11(m,1H),1.98–1.83(m,1H)。
Example 20: synthesis of 6- ((((1R, 2S) -2-hydroxycyclopentyl) amino) methyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (Z-2)
To (1S, 2R) -2-aminocyclopentan-1-ol, HCl (Z-1) (8.9 mg,1.2 eq., 65. Mu. Mol) and DIPEA (21 mg, 28. Mu.L, 3.0 eq., 0.16 mmol) in CHCl 3 To a solution in (4 mL) was added intermediate Q-1 (25 mg,1 eq, 0.054 mmol) and the reaction mixture was stirred for 30 min. Adding NaBH (OAc) 3 (23 mg,2 equivalents, 0.11 mmol) and the reaction mixture was stirred at 40℃overnight. The crude product was taken up with NaHCO 3 Quench and add DCM (10 mL) to a saturated aqueous solution (5 mL). The organics were extracted, dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative reverse phase HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (Z-2) as a white solid (8 mg,0.01mmol,30%,99% purity). M/z 548.2 (M+H) + (ES+). 1 H NMR (400 MHz, DMSO-d 6) delta 8.39 (s, 1H), 8.05 (s, 1H), 8.02-7.97 (m, 2H), 7.75-7.64 (m, 3H), 7.62-7.56 (m, 2H), 7.37 (t, J=8.0 Hz, 1H), 6.91 (dt, J=7.7, 1.3Hz, 1H), 5.08 (s, 2H), 4.45-4.36 (m, 1H), 4.00-3.84 (m, 3H), 3.07 (s, 3H), 2.81-2.71 (m, 1H), 1.75-1.53 (m, 4H), 1.48-1.33 (m, 2H). No exchangeable protons were observed.
Example 21: synthesis of 6- ((((1S, 2R) -2-hydroxycyclopentyl) amino) methyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AA-2)
To (1R, 2S) -2-aminocyclopentan-1-ol, HCl (AA-1) (8.9 mg,1.2 eq., 65. Mu. Mol) and DIPEA (21 mg, 28. Mu.L, 3.0 eq., 0.16 mmol) in CHCl 3 To a solution in (4 mL) was added intermediate Q-1 (25 mg,1 eq, 0.054 mmol) and the reaction mixture was stirred for 30 min. Adding NaBH (OAc) 3 (23 mg,2 equivalents, 0.11 mmol) and the reaction mixture was brought to 40℃Stir overnight. The crude product was taken up with NaHCO 3 Quench and add DCM (10 mL) to a saturated aqueous solution (5 mL). The organics were extracted, dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (woth, basic (0.3% ammonia), woth XBridge BEH C18 ODB 1, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (AA-2) as a white solid (3 mg,5 μmol,10%,95% purity). M/z 548.2 (M+H) + (ES+). 1 H NMR (400 MHz, DMSO-d 6) delta 8.39 (s, 1H), 8.05 (s, 1H), 8.02-7.97 (m, 2H), 7.75-7.65 (m, 3H), 7.62-7.56 (m, 2H), 7.37 (t, J=8.0 Hz, 1H), 6.91 (d, J=7.6 Hz, 1H), 5.08 (s, 2H), 4.45-4.28 (m, 1H), 4.00-3.85 (m, 3H), 3.08 (s, 3H), 2.82-2.71 (m, 1H), 1.75-1.54 (m, 4H), 1.48-1.36 (m, 2H). No exchangeable protons were observed.
Example 22: synthesis of (S) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6- ((2-methylmorpholino) methyl) -4- (trifluoromethyl) isoindolin-1-one (AB-2)
To (S) -2-methylmorpholino (AB-1) (6.6 mg,1.2 eq, 65. Mu. Mol) and DIPEA (21 mg, 28. Mu.L, 3.0 eq, 0.16 mmol) in CHCl 3 To a solution in (4 mL) was added intermediate Q-1 (25 mg,1 eq, 0.054 mmol) and the reaction mixture was stirred for 30 min. Adding NaBH (OAc) 3 (23 mg,2 equivalents, 0.11 mmol) and the reaction mixture was stirred at 40℃overnight. The crude product was taken up with NaHCO 3 Quench and add DCM (10 mL) to a saturated aqueous solution (5 mL). The organics were extracted, dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (woth, basic (0.3% ammonia), woth XBridge BEH C18 ODB 1, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (9 mg,0.02mmol,30%,99% purity) as a white solid. M/z 548.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.39(s,1H),7.99(dd,J=8.1,2.0Hz,2H),7.94(s,1H),7.75–7.65(m,3H),7.63–7.56(m,2H),7.38 (t, j=8.0 hz, 1H), 6.94-6.88 (m, 1H), 5.08 (s, 2H), 3.78-3.70 (m, 1H), 3.68 (s, 2H), 3.58-3.47 (m, 2H), 2.73-2.59 (m, 2H), 2.10 (td, j=11.3, 3.2hz, 1H), 1.84-1.76 (m, 1H), 1.03 (d, j=6.3 hz, 3H). Three protons are masked by the water peak.
Example 23: synthesis of 2-chloro-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (pyrrolidin-1-yl) pyridine (AC-3)
Step 1: synthesis of 2-chloro-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (pyrrolidin-1-yl) pyridine (AC-1)
Intermediate E-7 (60.0 mg,1 eq, 197. Mu. Mol) was dissolved in pyrrolidine (0.43 g,0.50mL,30 eq, 6.0 mmol) and heated to 80℃for 1 hour. The reaction mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by silica gel chromatography (12 g cartridge, 0-10% MeOH/DCM) to give the sub-title compound (AC-1) (54.5 mg, 160. Mu. Mol, 81.6%) as a yellow oil. M/z 340.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.48 (s, 1H), 7.80-7.73 (m, 1H), 7.73-7.60 (m, 3H), 6.51 (d, j=1.2 hz, 1H), 5.99 (d, j=1.2 hz, 1H), 3.31 (s, 4H), 3.27-3.23 (m, 3H), 2.02-1.97 (m, 4H).
Step 2: synthesis of 2-chloro-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (pyrrolidin-1-yl) pyridine (AC-3)
A mixture of RuPhos (2.42 mg,0.05 eq, 5.18. Mu. Mol), naOtBu (4.98 mg,0.5 eq, 51.8. Mu. Mol), the product (AC-1) from step 1 above (39.1 mg,90Wt%,1 eq, 104. Mu. Mol), intermediate AC-2 (31.2 mg,1.5 eq, 155. Mu. Mol) and Pd-171 (RuPhos Pd (crotyl) Cl) (3.44 mg,0.05 eq, 5.18. Mu. Mol) was dissolved in 1, 4-dioxane (1.5 mL). The reaction mixture was taken up in N 2 (x 3) purge and heat to 100 ℃ overnight. The reaction mixture was concentrated in vacuo and the crude product was purified by reverse phase prep HPLC (basic (0.3% ammonia), basic, woterse XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give a white colorThe title compound (AC-3) (3.57 mg, 10. Mu. Mol,9.6%,95% purity) was in solid form. M/z 505.4 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.45 (s, 1H), 8.11 (d, j=7.7 hz, 1H), 7.99 (d, j=7.7 hz, 1H), 7.82-7.75 (m, 3H), 7.71-7.62 (m, 3H), 5.94 (s, 1H), 5.29 (s, 2H), 3.42-3.36 (m, 7H), 2.08-1.99 (m, 4H).
Example 24: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (AD-3) and 2- (6- (dimethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (AD-4)
/>
Step 1: synthesis of 6-chloro-N-ethyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-amine (AD-1) and 6-chloro-N, N-dimethyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-amine (AD-2)
To a solution of intermediate E-7 (33 mg,1 eq, 0.11 mmol) in DMF (2 mL) was added K sequentially 2 CO 3 (23 mg,1.5 eq, 0.17 mmol) and ethylamine (75 mg,0.84ml,2 mol, 15 eq, 1.7 mmol). The reaction mixture was stirred in a sealed tube at 100 ℃ overnight. The crude product was purified by RP flash C18 chromatography (12 g cartridge, 15-75% MeCN/10mM ammonium bicarbonate) to give an inseparable mixture of the sub-title compounds AD-1 (32 mg,31 μmol,27%,30% purity) and AD-2 (32 mg,71 μmol,64%,70% purity) as a light brown gum. M/z 314.3 (M+H) + (es+), 30% and 70% purity (diode array) at 1.51 and 1.59 minutes. 6-chloro-N-ethyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-amine (AD-1) (30%): 1 h NMR (400 MHz, methanol-d 4) delta 8.46 (s, 1H), 7.76-7.56 (m, 4H), 6.33 (d, j=1.2 hz, 1H), 6.08 (d, j=1.2 hz, 1H), 3.25 (s, 3H), 3.17 (q, j=7.2 hz, 2H), 1.13 (t, j=7.2 hz, 3H). No exchangeable protons were observed. 6-chloro-N, N-dimethyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-amine (A)D-2)(70%): 1 H NMR (400 MHz, methanol-d 4) delta 8.44 (s, 1H), 7.77-7.54 (m, 4H), 6.47 (d, j=1.0 hz, 1H), 6.10 (d, j=1.1 hz, 1H), 3.21 (s, 3H), 2.93 (s, 6H).
Step 2: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (AD-3) and 2- (6- (dimethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (AD-4)
At N 2 To a solution of the products (AD-1) and (AD-2) (32 mg,70Wt%,2.3 eq., 71. Mu. Mol) from step 1 above and intermediate C-1 (33 mg,4.6 eq., 0.14 mmol) in 1, 4-dioxane (1 mL) at room temperature was added K 3 PO 4 (42 mg,6.5 eq, 0.20 mmol), xantPhos (12 mg,0.65 eq, 0.020 mmol) and Pd (OAc) 2 (2 mg,0.3 eq, 0.01 mmol). The mixture was stirred at 100℃for 16 hours. Addition of additional Pd (OAc) 2 (2 mg,0.3 eq, 0.01 mmol) and dppf (11 mg,0.6 eq, 0.02 mmol), and the reaction was stirred at 100℃for 16 h. The reaction mixture was diluted with DCM and filtered through a plug of silica gel. The silica gel plug was washed with 10% MeOH/DCM (50 mL) and the solvent was removed in vacuo. Pd-171 (662.62 g/mol,3.3mg,0.15 eq, 0.05 mmol), ruPhos (2.3 mg,0.15 eq, 0.05 mmol), naOtBu (6 mg,13 eq, 0.6 mmol) and 1, 4-dioxane (1 mL) were added to the residue and the mixture was stirred at 100℃for 16 hours. The reaction mixture was diluted with DCM and filtered through a plug of silica eluting with 10% MeOH in DCM (50 mL). The filtrate was concentrated in vacuo and the crude product was purified by reverse phase prep HPLC (wok, acid (0.1% formic acid), acidic wok X-Select CSH C18 ODB, 5 μm, 30X100mm column, 0-100% MeCN/water) to give the title compounds (AD-3) and (AD-4) as white solids: 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (AD-3) (6 mg,0.01mmol,10%,99% purity). M/z 509.5 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.46 (s, 1H), 8.07 (s, 1H), 7.97 (s, 1H), 7.81-7.70%m, 2H), 7.70-7.61 (m, 2H), 7.57 (s, 1H), 6.06 (s, 1H), 5.22 (s, 2H), 4.81 (s, 2H), 3.44 (s, 3H), 3.29 (q, j=7.2 hz, 2H), 1.23 (t, j=7.2 hz, 3H). No two exchangeable protons were observed. 2- (6- (dimethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (AD-4) in the form of a white solid (15 mg,29 μmol,29%,99% purity). M/z 509.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.42 (s, 1H), 8.03 (s, 1H), 7.93 (s, 1H), 7.79-7.70 (m, 2H), 7.70-7.57 (m, 3H), 6.05 (s, 1H), 5.19 (s, 2H), 4.77 (s, 2H), 3.33-3.27 (m, 3H), 2.98 (s, 6H). No exchangeable protons were observed.
Example 25: synthesis of (R) -2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((3-methylpiperidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (AE-2)
Step 1: synthesis of 6-chloro-N-ethyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-amine (AD-1)
Intermediate E-7 (330 mg,1 eq, 1.08 mmol) and ethylamine (2.10 g,23.3mL,2 moles, 43 eq, 46.5 mmol) were added to a microwave vial and irradiated at 100℃for 20 hours. The reaction mixture was concentrated in vacuo. The crude product was purified by silica gel chromatography (24 g cartridge, 0-20% MeOH/DCM) to give the sub-title compound (AD-1) (326 mg,1.0mmol,92%,96% pure) as a pale yellow solid. M/z 313.9 (M+H) + (ES+)。
Step 2: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (AD-3)
To a solution of the product (AD-1) (226 mg,85Wt%,1 equivalent, 612. Mu. Mol) from step 1 above and intermediate C-1 (212 mg,1.5 equivalent, 918. Mu. Mol) in 1, 4-dioxane (6 mL) was added RuPhos (14.3 mg,0.05 equivalent, 30.6. Mu. Mol), naO in this order t Bu (64.7 mg,1.1 eq, 673. Mu.m)mol) and Pd-171 (RuPhos Pd (crotyl) Cl) (20.3 mg,0.05 eq, 30.6. Mu. Mol). The mixture obtained is put in N 2 Stirring is carried out at 100℃for 16 hours. The solvent was removed in vacuo and the crude product was purified by silica gel chromatography (12 g cartridge, 0-20% MeOH/DCM) to give the sub-title compound (AD-3) as a clear white solid (152 mg,0.30mmol,48%,99% purity). M/z 509.2 (M+H) + (ES+)。
Step 3: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -3-oxo-7- (trifluoromethyl) isoindoline-5-carbaldehyde (AE-1)
Manganese dioxide (1.18 g,60 equivalents, 13.6 mmol) was added to chloroform (25 mL) containing the product (AD-3) from step 2 above (116 mg,99Wt%,1 equivalent, 226. Mu. Mol), and stirred at 40℃for 18 hours. The reaction mixture was filtered through a celite plug, washing with 1, 4-dioxane (25 mL) and DCM (25 mL). The filtrate was concentrated in vacuo to give the sub-title compound (AE-1) (79 mg,0.15mmol,65%,94% purity) as a pale yellow solid. M/z 507.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ10.21(s,1H),8.52(s,1H),8.47(d,J=6.7Hz,2H),7.72(td,J=7.4,1.6Hz,1H),7.66–7.54(m,3H),7.50(d,J=1.2Hz,1H),6.73(t,J=5.5Hz,1H),5.95(d,J=1.2Hz,1H),5.28(s,2H),3.33(s,3H),3.17(q,J=6.4Hz,2H),1.13(t,J=7.1Hz,3H)。
Step 4: synthesis of (R) -2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((3-methylpiperidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (AE-2)
NaBH (OAc) 3 (65 mg,6.0 eq, 0.30 mmol) was added to a stirred solution of the product (AE-1) (26 mg,99Wt%,1 eq, 51. Mu. Mol), (R) -3-methylpiperidine-HCl (S-1) (14 mg,2.0 eq, 0.10 mmol) and DIPEA (20 mg, 26. Mu.L, 3.0 eq, 0.15 mmol) from step 3 above in chloroform (2 mL) and stirred at room temperature for 3 days. The reaction mixture was diluted with DCM (5 mL) and NaHCO 3 Is washed with a saturated aqueous solution (6 mL). The organic layer was separated and dried (MgSO 4 ) And concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (03% ammonia), basic Woldie XBIdge BEH C18 ODB, 5 μm, 30X100mm column 0-100% MeCN/water) to give the title compound (AE-2) (8 mg,0.01mmol,30%,100% purity) as a flocculent white solid. M/z 590.6 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.95(s,1H),7.91(s,1H),7.71(td,J=7.5,1.7Hz,1H),7.66–7.53(m,3H),7.48(d,J=1.2Hz,1H),6.69(t,J=5.4Hz,1H),5.93(d,J=1.3Hz,1H),5.16(s,2H),3.63(s,2H),3.23–3.11(m,2H),2.76–2.67(m,2H),1.93(t,J=10.9Hz,1H),1.68–1.56(m,4H),1.51–1.43(m,1H),1.12(t,J=7.2Hz,3H),0.89–0.85(m,1H),0.82(d,J=6.0Hz,3H)。CH 3 Is masked by the water peak.
Example 26: synthesis of (S) -2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) 6- ((3-methylpiperidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (AF-1)
To a solution of intermediate AE-1 (30 mg,99wt%,1 eq, 59 μmol) and DIPEA (23 mg,31 μL,3.0 eq, 0.18 mmol) in DCM (2 mL) was added (S) -3-methylpiperidine-HCl (R-1) (8.7 mg,1.1 eq, 65 μmol). The reaction was stirred for 1 hour and then NaBH (OAc) was added 3 (25 mg,2.0 equivalents, 0.12 mmol) and stirred at room temperature overnight. The reaction mixture was diluted with DCM (5 mL) and NaHCO 3 Is washed with saturated aqueous solution (5 mL) and brine (5 mL). The combined organic layers were dried (MgSO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase prep HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (AF-1) (3 mg,5 μmol,9%,98% purity). M/z 590.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.47 (s, 1H), 8.05 (s, 1H), 7.97 (s, 1H), 7.80-7.70 (m, 2H), 7.69-7.60 (m, 2H), 7.56 (d, j=1.2 hz, 1H), 6.07 (d, j=1.2 hz, 1H), 5.23 (s, 2H), 3.70 (s, 2H), 3.45 (s, 3H), 3.30 (q, j=7.2 hz, 2H), 2.85 (dd, j=15.1, 8.9hz, 2H), 2.02(td, j=11.3, 3.1hz, 1H), 1.83-1.57 (m, 5H), 1.24 (t, j=7.2 hz, 3H), 0.90 (m, 4H). No exchangeable protons were observed.
Example 27: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((3-fluoroazetidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (AG-1)
NaBH (OAc) 3 (52 mg,6.0 eq, 0.25 mmol) was added to a stirred solution of intermediate AE-1 (21 mg,99Wt%,1 eq, 41. Mu. Mol), 3-fluoroazetidine-HCl (U-1) (9.2 mg,2.0 eq, 82. Mu. Mol) and DIPEA (16 mg, 21. Mu.L, 3.0 eq, 0.12 mmol) in chloroform (2 mL) and left at room temperature for 16 hours. The reaction mixture was diluted with DCM (5 mL) and NaHCO 3 Is washed with a saturated aqueous solution (6 mL). The organic layer was extracted, dried (MgSO 4 ) And concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (AG-1) (9 mg,0.02mmol,40%,100% purity) as a flocculent white solid. M/z 566.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.95(s,1H),7.90(s,1H),7.75–7.67(m,1H),7.65–7.54(m,3H),7.48(d,J=1.2Hz,1H),6.69(t,J=5.4Hz,1H),5.93(d,J=1.2Hz,1H),5.33–5.10(m,3H),3.84(s,2H),3.65–3.53(m,2H),3.28–3.13(m,4H),1.12(t,J=7.2Hz,3H)。CH 3 Is masked by the water peak.
Example 28: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -3-oxo-7- (trifluoromethyl) isoindoline-5-carbaldehyde (AH-2)
NaBH (OAc) 3 (52 mg,6.0 eq, 0.25 mmol)To a stirred solution of intermediate AE-1 (21 mg,99Wt%,1 eq, 41. Mu. Mol), (S) - (+) -3-pyrrolidinol-HCl (AH-1) (10 mg,2.0 eq, 82. Mu. Mol) and DIPEA (16 mg, 21. Mu.L, 3.0 eq, 0.12 mmol) in chloroform (2 mL) was added and stirred at room temperature for 16 hours. The reaction mixture was diluted with DCM (5 mL) and NaHCO 3 Is washed with a saturated aqueous solution (6 mL). The organic layer was extracted, dried (MgSO 4 ) And concentrated in vacuo. The crude product was purified by preparative HPLC (woth, basic (0.3% ammonia), basic woth XBridge DEH C18 ODB, 5 μm, 30x100mm, 0-100% MeCN/water) to give the title compound (AH-2) as a flocculent white solid (21 mg,99wt%,1 eq, 41 μmol). M/z 578.1 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.97(s,1H),7.92(s,1H),7.75–7.67(m,1H),7.65–7.53(m,3H),7.48(d,J=1.2Hz,1H),6.69(t,J=5.4Hz,1H),5.94(d,J=1.3Hz,1H),5.16(s,2H),4.72(d,J=4.4Hz,1H),4.21(s,1H),3.82–3.72(m,2H),3.23–3.12(m,2H),2.73–2.58(m,2H),2.48–2.39(m,1H),2.36(dd,J=9.6,3.6Hz,1H),2.08–1.95(m,1H),1.63–1.51(m,1H),1.13(t,J=7.2Hz,3H)。CH 3 Is masked by the water peak.
Example 29: synthesis of 6- ((6-azaspiro [2.5] oct-6-yl) methyl) -2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AI-2)
NaBH (OAc) 3 (65 mg,6.0 eq, 0.30 mmol) was added to intermediate AE-1 (26 mg,99Wt%,1 eq, 51. Mu. Mol), 6-azaspiro [2.5]]A stirred solution of octane-HCl (AI-1) (15 mg,2.0 eq, 0.10 mmol) and DIPEA (20 mg, 26. Mu.L, 3.0 eq, 0.15 mmol) in chloroform (2 mL) was stirred at room temperature for 3 days. The reaction mixture was diluted with DCM (5 mL) and NaHCO 3 Is washed with a saturated aqueous solution (6 mL). The organic layer was extracted, dried (MgSO 4 ) And concentrated in vacuo. The crude product was purified by preparative HPLC (Wo-Tech, basic (0.3% ammonia), basicPurification of Wolth XBridge BEH C18 ODB, 5 μm, 30X100mm column, 0-100% MeCN/water) to give the title compound (AI-2) (10 mg, 17. Mu. Mol,33%,100% purity) as a flocculent white solid. M/z 602.5 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.97(s,1H),7.93(s,1H),7.75–7.67(m,1H),7.66–7.54(m,3H),7.48(d,J=1.2Hz,1H),6.69(t,J=5.4Hz,1H),5.93(d,J=1.2Hz,1H),5.16(s,2H),3.70(s,2H),3.23–3.13(m,2H),2.45–2.41(m,4H),1.37–1.32(m,4H),1.13(t,J=7.2Hz,3H),0.25(s,4H)。CH 3 Is masked by the water peak.
Example 30: synthesis of (S) -2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((3-fluoropyrrolidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (AJ-1)
NaBH (OAc) 3 (52 mg,6.0 eq, 0.25 mmol) was added to a stirred solution of intermediate AE-1 (21 mg,99Wt%,1 eq, 41. Mu. Mol), (S) - (+) -3-fluoropyrrolidine-HCl (V-1) (10 mg,2 eq, 82. Mu. Mol) and DIPEA (16 mg, 21. Mu.L, 3.0 eq, 0.12 mmol) in chloroform (2 mL) and stirred at room temperature overnight. The reaction mixture was diluted with DCM (5 mL) and NaHCO 3 Is washed with a saturated aqueous solution (6 mL). The organic layer was extracted, dried (MgSO 4 ) And concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% mecn/water) to give the title compound (AJ-1) (9 mg,0.02mmol,40%,100% purity) as a flocculent white solid. M/z 580.5 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.98(s,1H),7.94(s,1H),7.75–7.67(m,1H),7.65–7.53(m,3H),7.48(d,J=1.2Hz,1H),6.69(t,J=5.4Hz,1H),5.94(d,J=1.3Hz,1H),5.22(d,J=50.2Hz,3H),3.83(s,2H),3.23–3.12(m,2H),2.88–2.79(m,1H),2.82–2.75(m,1H),2.74–2.57(m,1H),2.43–2.32(m,1H),2.27–2.07(m,1H),1.98–1.82(m,1H),1.13(t,J=7.2Hz,3H)。CH 3 Is masked by the water peak.
Example 31: synthesis of 6- ((4, 4-difluoro-3-methylpiperidin-1-yl) methyl) -2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AK-2)
NaBH (OAc) 3 (65 mg,6.0 eq, 0.30 mmol) was added to a stirred solution of intermediate AE-1 (26 mg,99Wt%,1 eq, 51. Mu. Mol), rac-4, 4-difluoro-3-methylpiperidine-HCl (AK-1) (17 mg,2.0 eq, 0.10 mmol) and DIPEA (20 mg, 26. Mu.L, 3.0 eq, 0.15 mmol) in chloroform (2 mL) and stirred at room temperature for 3 days. The reaction mixture was diluted with DCM (5 mL) and NaHCO 3 Is washed with a saturated aqueous solution (6 mL). The organic layer was extracted, dried (MgSO 4 ) And concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (AK-2) (13 mg,21 μmol,41%,100% purity) M/z 626.6 (m+h) as a white solid + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.99(s,1H),7.94(s,1H),7.75–7.67(m,1H),7.65–7.54(m,3H),7.48(d,J=1.1Hz,1H),6.69(t,J=5.3Hz,1H),5.94(s,1H),5.16(s,2H),3.74(s,2H),3.21–3.13(m,2H),2.77–2.72(m,2H),2.32(s,2H),2.13(s,1H),2.09–2.00(m,2H),1.13(t,J=7.2Hz,3H),0.92(d,J=6.4Hz,3H)。CH 3 Is masked by the water peak.
Example 32: synthesis of (S) -2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((2-methylmorpholino) methyl) -4- (trifluoromethyl) isoindolin-1-one (AL-1)
NaBH (OAc) 3 (68 mg,6.0 equivalents, 0.32 mmol) was addedTo a stirred solution of intermediate AE-1 (33 mg,82Wt%,1 eq, 53. Mu. Mol), (2S) -2-methylmorpholine (AB-1) (11 mg,2.0 eq, 0.11 mmol) and DIPEA (6.9 mg, 9.2. Mu.L, 1.0 eq, 53. Mu. Mol) in chloroform (2 mL) was added and left overnight at room temperature. The reaction mixture was diluted with DCM (5 mL) and NaHCO 3 Is washed with a saturated aqueous solution (6 mL). The organic layer was separated, dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% mecn/water) to give the title compound (AL-1) (11 mg,18 μmol,34%,99% purity) as a flocculent white solid. M/z 592.5 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.98(s,1H),7.93(s,1H),7.75–7.67(m,1H),7.65–7.53(m,3H),7.48(d,J=1.3Hz,1H),6.69(t,J=5.4Hz,1H),5.94(s,1H),5.16(s,2H),3.74(d,J=11.2Hz,1H),3.66(s,2H),3.57–3.46(m,2H),3.23–3.13(m,2H),2.69(d,J=11.1Hz,1H),2.62(d,J=11.3Hz,1H),2.09(td,J=11.4,3.2Hz,1H),1.79(t,J=10.5Hz,1H),1.13(t,J=7.1Hz,3H),1.02(d,J=6.2Hz,3H)。CH 3 The signal is masked by the water peak.
Example 33: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (morpholinomethyl) -4- (trifluoromethyl) isoindolin-1-one (AM-2)
NaBH (OAc) 3 (68 mg,6.0 eq, 0.32 mmol) was added to a stirred solution of intermediate AE-1 (33 mg,82Wt%,1 eq, 53. Mu. Mol), morpholino (AM-1) (9.3 mg,2.0 eq, 0.11 mmol) and DIPEA (6.9 mg, 9.2. Mu.L, 1.0 eq, 53. Mu. Mol) in chloroform (2 mL) and stirred at room temperature overnight. The reaction mixture was diluted with DCM (5 mL) and NaHCO 3 Is washed with a saturated aqueous solution (6 mL). The organic layer was separated, dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (Wolth, basic (0.3% ammonia), basic, wolth XBLID BEH C18 ODB, 5 μm, 30X100mm column, 0-100% MeCN/water) to give the title compound (AM-2) (12 mg, 21. Mu. Mol,39%,100% purity) as a flocculent white solid. M/z 578.5 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.98(s,1H),7.93(s,1H),7.75–7.67(m,1H),7.65–7.54(m,3H),7.48(s,1H),6.69(t,J=5.4Hz,1H),5.94(s,1H),5.16(s,2H),3.68(s,2H),3.59(t,J=4.6Hz,4H),3.18(q,J=6.7Hz,2H),2.40(s,4H),1.13(t,J=7.2Hz,3H)。CH 3 The signal is masked by the water peak.
Example 34: synthesis of (R) -2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((2-methylmorpholino) methyl) -4- (trifluoromethyl) isoindolin-1-one (AN-1)
NaBH (OAc) 3 (63 mg,6.0 eq, 0.29 mmol) was added to a stirred solution of intermediate AE-1 (30 mg,83Wt%,1 eq, 49. Mu. Mol), (R) -2-methyl-morpholino-HCl (X-1) (14 mg,2.0 eq, 98. Mu. Mol) and DIPEA (19 mg, 25. Mu.L, 3.0 eq, 0.15 mmol) in chloroform (2 mL) and stirred at room temperature for 3 days. Additional DIPEA (38 mg, 50. Mu.L, 5.9 eq, 0.29 mmol), chloroform (1.0 mL) and NaBH (OAc) were added 3 (30 mg,2.9 eq, 0.14 mmol) and the reaction mixture was stirred at room temperature overnight. Adding NaBH 4 (10 mg,5.4 eq, 0.26 mmol) and the reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with DCM (4 mL) and NaHCO 3 Is washed with a saturated aqueous solution (10 mL). The organic phase was separated, dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (AN-1) as a flocculent white solid (7 mg,0.01mmol,20%,99% purity). M/z 592.5 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.98(s,1H),7.93(s,1H),7.75–7.67(m,1H),7.66–7.53(m,3H),7.48(d,J=1.2Hz,1H),6.73–6.65(m,1H),5.94(s,1H),5.18–5.12(m,2H),3.77–3.70(m,1H),3.66(s,2H),3.55–3.46(m,2H),3.20–3.15(m,2H),2.69(d,J=11.1Hz,1H),2.62(d,J=11.3Hz,1H),2.15–2.04(m,1H),1.79(dd,J=11.2,9.9Hz,1H),1.13(t,J=7.1Hz,3H),1.02(d,J=6.3Hz,3H)。CH 3 The signal is masked by the water peak.
Example 35: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((((1R, 2R) -2-hydroxycyclopentyl) amino) methyl) -4- (trifluoromethyl) isoindolin-1-one (AO-1)
NaBH (OAc) 3 (63 mg,6.0 eq, 0.29 mmol) was added to a stirred solution of intermediate AE-1 (30 mg,83Wt%,1 eq, 49. Mu. Mol), (1R, 2R) -2-aminocyclopentan-1-ol-HCl (W-1) (14 mg,2.0 eq, 98. Mu. Mol) and DIPEA (19 mg, 25. Mu.L, 3.0 eq, 0.15 mmol) in chloroform (2 mL) and stirred at room temperature overnight. Additional DIPEA (38 mg, 50. Mu.L, 5.9 eq, 0.29 mmol), chloroform (1.0 mL) and NaBH (OAc) 3 (30 mg,2.9 equivalents, 0.14 mmol) and stirred at room temperature overnight. Adding NaBH 4 (10 mg,5.4 eq, 0.26 mmol) and the reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with DCM (4 mL) and NaHCO 3 Is washed with a saturated aqueous solution (10 mL). The organic phase was separated, dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (AO-1) as a flocculent white solid (8 mg,0.01mmol,30%,100% purity). M/z 592.6 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),8.02(s,1H),7.98(s,1H),7.75–7.67(m,1H),7.65–7.53(m,3H),7.48(d,J=1.2Hz,1H),6.68(t,J=5.3Hz,1H),5.94(d,J=1.2Hz,1H),5.15(s,2H),4.51(d,J=4.2Hz,1H),3.92–3.87(m,2H),3.83–3.76(m,1H),3.23–3.14(m,2H),2.75–2.71(m,1H),2.38–2.34(m,1H),1.88–1.74(m,2H),1.63–1.51(m,2H),1.46–1.36(m,1H),1.36–1.23(m,1H),1.13(t,J=7.2Hz,3H)。CH 3 Is masked by the water peak.
Example 36: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((((1S, 2S) -2-hydroxycyclopentyl) amino) methyl) -4- (trifluoromethyl) isoindolin-1-one (AP-1)
NaBH (OAc) 3 (63 mg,6.0 eq, 0.29 mmol) was added to a stirred solution of intermediate AE-1 (30 mg,83Wt%,1 eq, 49. Mu. Mol), trans- (1S, 2S) -2-aminocyclopentanol-HCl (T-1) (14 mg,2.0 eq, 98. Mu. Mol) and DIPEA (19 mg, 25. Mu.L, 3.0 eq, 0.15 mmol) in chloroform (2 mL) and stirred at room temperature for 3 days. Additional DIPEA (38 mg, 50. Mu.L, 5.9 eq, 0.29 mmol), chloroform (1.0 mL) and NaBH (OAc) were added 3 (30 mg,2.9 eq, 0.14 mmol) and the reaction was stirred at room temperature overnight. Adding NaBH 4 (10 mg,5.4 eq, 0.26 mmol) and the reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with DCM (4 mL) and NaHCO 3 Is washed with a saturated aqueous solution (10 mL). The organic phase was separated, dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (AP-1) as a flocculent white solid (8 mg,0.01mmol,30%,100% purity). M/z 592.4 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),8.02(s,1H),7.98(s,1H),7.75–7.67(m,1H),7.65–7.53(m,3H),7.48(d,J=1.2Hz,1H),6.68(t,J=5.4Hz,1H),5.94(d,J=1.2Hz,1H),5.15(s,2H),4.51(d,J=4.2Hz,1H),3.89(s,2H),3.84–3.75(m,1H),3.23–3.12(m,2H),2.76–2.70(m,1H),2.39–2.34(m,1H),1.88–1.75(m,2H),1.63–1.51(m,2H),1.46–1.37(m,1H),1.37–1.23(m,1H),1.13(t,J=7.2Hz,3H)。CH 3 Is masked by the water peak.
Example 37: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((((1R, 2S) -2-hydroxycyclopentyl) amino) methyl) -4- (trifluoromethyl) isoindolin-1-one (AQ-1)
Intermediate AE-1 (30 mg,99Wt%,1 eq, 59. Mu. Mol), (1S, 2R) -2-aminocyclopentan-1-ol-HCl (Z-1) (16 mg,2.0 eq, 0.12 mmol) and DIPEA (23 mg, 30. Mu.L, 3.0 eq, 0.18 mmol) were combined in CHCl 3 The mixture in (2 mL) was stirred at 40℃for 30 min. NaBH (OAc) is then added 3 (75 mg,6.0 equivalents, 0.35 mmol) and the resulting suspension was stirred at room temperature overnight. The reaction mixture was diluted with DCM (4 mL) and NaHCO 3 Saturated aqueous (10 mL) was washed. The organic phase was separated and concentrated in vacuo to give a crude product which was purified by preparative HPLC (woth, basic (0.3% ammonia), basic, woth XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (AQ-1) as a white solid (14 mg,23 μmol,39%,97% purity). M/z 592.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),8.01(d,J=16.6Hz,2H),7.71(td,J=7.4,1.7Hz,1H),7.65–7.53(m,3H),7.48(d,J=1.2Hz,1H),6.68(t,J=5.4Hz,1H),5.93(d,J=1.4Hz,1H),5.15(s,2H),4.40(d,J=3.6Hz,1H),3.99–3.91(m,2H),3.91–3.83(m,1H),3.33(s,3H),3.20–3.14(m,2H),2.78–2.74(m,1H),2.31–2.27(m,1H),1.75–1.54(m,4H),1.48–1.38(m,2H),1.13(t,J=7.1Hz,3H)。
Example 38: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((((1S, 2R) -2-hydroxycyclopentyl) amino) methyl) -4- (trifluoromethyl) isoindolin-1-one (AR-1)
Intermediate AE-1 (30 mg,99Wt%,1 eq., 59. Mu. Mol), (1R, 2S) -2-aminocyclopentan-1-ol-HCl (AA-1) (16 mg,2.0 eq, 0.12 mmol) and DIPEA (23 mg, 30. Mu.L, 3.0 eq, 0.18 mmol) in CHCl 3 The mixture in (2 mL) was stirred at 40℃for 30 min. NaBH (OAc) is then added 3 (75 mg,6.0 equivalents, 0.35 mmol) and the resulting suspension was stirred at room temperature overnight. The reaction mixture was diluted with DCM (4 mL) and NaHCO 3 Saturated aqueous (10 mL) was washed. The organic phase was separated and concentrated in vacuo to give a crude product which was purified by preparative HPLC (woth, basic (0.3% ammonia), basic, woth XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (AR-1) as a white solid (15 mg,25 μmol,42%,98% purity). M/z 591.9 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),8.01(d,J=16.6Hz,2H),7.71(td,J=7.4,1.7Hz,1H),7.65–7.53(m,3H),7.48(d,J=1.2Hz,1H),6.68(t,J=5.4Hz,1H),5.93(d,J=1.3Hz,1H),5.15(s,2H),4.40(d,J=3.6Hz,1H),3.99–3.91(m,2H),3.91–3.83(m,1H),3.33(s,3H),3.20–3.14(m,2H),2.78–2.74(m,1H),2.32–2.27(m,1H),1.73–1.54(m,4H),1.48–1.34(m,2H),1.13(t,J=7.1Hz,3H)。
Example 39: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (methoxymethyl) -4- (trifluoromethyl) isoindolin-1-one (AS-2)
Step 1: synthesis of 6- (chloromethyl) -2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AS-1)
Thionyl chloride (7.1 mg, 4.4. Mu.L, 1.1 eq, 60. Mu. Mol) was added to a suspension of intermediate AD-3 (30 mg,92Wt%,1 eq, 54. Mu. Mol) in DCM (2 mL) and the resulting mixture was stirred at room temperature overnight. More thionyl chloride (65 mg, 40. Mu.L, 10 eq, 0.54 mmol) was added and stirring continued at room temperature overnight to give the sub-title compound for direct use in the next step The product (AS-1). M/z 527.2 (M+H) + (ES+)
Step 2: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (methoxymethyl) -4- (trifluoromethyl) isoindolin-1-one (AS-2)
Sodium methoxide (0.11 g,0.55mL,5.4 moles, 60 equivalents, 3.3 mmol) was added to DCM (0.8 mL) and MeOH (2 mL) containing the product from step 1 above (AS-1) (30 mg,1 equivalent, 57. Mu. Mol). The reaction mixture was stirred at room temperature overnight. The reaction mixture was purified by addition of dioxane containing 4M HCl followed by NaHCO 3 The saturated aqueous solution was adjusted to neutral pH. The bulk solvent was removed in vacuo and the residue partitioned between 1:1DCM-EtOAc (10 mL) and water (10 mL). The organic layer was concentrated in vacuo to give an orange gum. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (AS-2) AS a white solid (10 mg,19 μmol,37%,98% purity). M/z 523.4 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.99(s,1H),7.94(s,1H),7.75–7.67(m,1H),7.66–7.53(m,3H),7.49(d,J=1.2Hz,1H),6.69(t,J=5.5Hz,1H),5.93(d,J=1.3Hz,1H),5.17(s,2H),4.62(s,2H),3.36(s,3H),3.29(s,3H),3.21–3.13(m,2H),1.12(t,J=7.2Hz,3H)。
Example 40: synthesis of 2- [3- [2- (4-methyl-1, 2, 4-triazol-3-yl) imidazol-1-yl ] phenyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AAA-8)
Step 1: synthesis of ethyl 1- (3-bromophenyl) imidazole-2-carboxylate (AAA-3)
To a stirred mixture of ethyl 1H imidazole-2-carboxylate (AAA-1) (1.00 g,1 eq, 7.1 mmol) and 3-bromophenyl boronic acid (AAA-2) (2.87 g,2 eq, 14.3 mmol) in 1, 2-dichloroethane (20 mL) was added copper acetate (1.94 g,1.5 eq, 10.7 mmol) and pyridine (1.13 g,2 eq, 14.3 mmol) at room temperature.The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by silica gel column chromatography eluting with petroleum ether/EtOAc (1/1) to give the sub-title compound (AAA-3) as a white solid (500 mg,1.7mmol, 24%). M/z 295.1/297.1 (M+H) + (ES+)
Step 2: synthesis of 1- (3-bromophenyl) imidazole-2-carbohydrazide (AAA-4)
To a stirred mixture of the product (AAA-3) from step 1 above (400 mg,1 eq, 1.36 mmo) in EtOH (8 mL) was added dropwise hydrazine hydrate (678 mg,80Wt%,10 eq, 13.6 mmo) at room temperature. The resulting mixture was stirred at 80 ℃ overnight. The mixture was cooled to room temperature and concentrated in vacuo to give the sub-title compound (AAA-4) (330 mg,1.17mmol, crude) as a white solid. M/z 281.1/283.1 (M+H) + (ES+)。
Step 3:1- (3-bromophenyl) -N- [ (methylsulfamoyl) amino ] imidazole-2-carboxamide (AAA-5)
To a stirred solution of the product (AAA-4) from step 2 above (320 mg,1 eq, 1.14 mmol) in EtOH (10 mL) was added methyl isothiocyanate (99.8 mg,1.2 eq, 1.37 mmol) at room temperature. The resulting mixture was stirred at 80℃for 6 hours. The mixture was cooled to room temperature and concentrated to give the sub-title compound (AAA-5) (350 mg, 991. Mu. Mol, 87%) as a white solid. M/z 354.2/356.2 (M+H) + (ES+)。
Step 4: synthesis of 5- [1- (3-bromophenyl) imidazol-2-yl ] -4-methyl-1, 2, 4-triazole-3-thiol (AAA-6)
A mixture of the product (AAA-5) (330 mg,1 eq, 0.93 mmol) from step 3 above in NaOH (aqueous solution) (10 mL,1 mol, 10 eq, 10 mmol) was stirred at 60℃for 3 hours. The resulting mixture was diluted with water, acidified to pH 6 with HCl (1M aqueous solution) and extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (2×30 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo to give the sub-title compound (AAA-6) (300 mg, 895. Mu. Mol, 96%) as a white solid. M/z 336.2/338.2 (M+H) + (ES+)。
Step 5: synthesis of 3- [1- (3-bromophenyl) imidazol-2-yl ] -4-methyl-1, 2, 4-triazole (AAA-7)
To a stirred solution of the product from step 4 above (AAA-6) (300 mg,1 eq, 892. Mu. Mol) in DCM (20 mL) was added dropwise acetic acid (54 mg,1 eq, 892. Mu. Mol) and hydrogen peroxide (607 mg,30Wt%,6 eq, 5.35 mmol) at 0deg.C. The resulting mixture was stirred at room temperature for 3 hours. The resulting mixture was concentrated in vacuo. The residue was purified by column chromatography on silica gel eluting with DCM/MeOH (10/1) to give the sub-title compound (AAA-7) (240 mg, 792. Mu. Mol, 88%) M/z 304.2/306.2 (M+H) as a brown oil + (ES+)。
Step 6: synthesis of 2- [3- [2- (4-methyl-1, 2, 4-triazol-3-yl) imidazol-1-yl ] phenyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AAA-8)
To the product (AAA-7) (50 mg,1 equivalent, 0.16 mmol), intermediate (AC-2) (36 mg,1.1 equivalent, 0.18 mmol) and Cs from step 5 above under a nitrogen atmosphere 2 CO 3 To a solution of (161 mg,3 eq, 493. Mu. Mol) in 1, 4-dioxane (3 mL) was added tris (dibenzylideneacetone) dipalladium (30 mg,0.2 eq, 33. Mu. Mol) and Xantphos (19 mg,0.2 eq, 33. Mu. Mol). The resulting mixture was stirred overnight at 120 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature, then diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography under the following conditions: column, C18 silica gel; mobile phase, ACN/water (0.1% nh 4 HCO 3 ) A gradient of 10% to 80% over 20 minutes; detector, UV 254/220nm and purification by preparative HPLC under the following conditions (column: XBridge Shield RP OBD column, 30X 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 -H 2 O), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 26% B to 48% B in 7 minutes, UV detection at 254/210 nm; retention time: 5.93). The product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the product as a white solidThe title compound (AAA-8) (20.9 mg, 49. Mu. Mol, 28%). M/z 425.2 (M+H) + (ES+). 1 H NMR(300MHz,MeOH-d4)δ8.61(s,1H),8.14–8.03(m,2H),7.99(d,J=7.7Hz,1H),7.95–7.85(m,1H),7.84–7.68(m,2H),7.55(t,J=8.2Hz,1H),7.45(d,J=1.3Hz,1H),7.28–7.18(m,1H),5.18(s,2H),3.90(s,3H)。
Example 41: synthesis of 6- ((5-azaspiro [2.4] heptan-5-yl) methyl) -2- (3- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) phenyl) -4- (trifluoromethyl) isoindolin-1-one (AAD-1)
Et was added to a stirred mixture of intermediate (AAB-8) (50 mg,1 eq, 197. Mu. Mol) and intermediate (AAC-2) (160 mg,2 eq, 393. Mu. Mol) in EtOH (6 mL) at room temperature 3 N (100 mg,5 eq, 983. Mu. Mol). The resulting mixture was stirred overnight at 80 ℃ under nitrogen atmosphere. The resulting mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBridge Shield RP18 OBD column, 19×250mm,10 μm; mobile phase a: water (0.1% FA), mobile phase B: meOH; flow rate: 25 ml/min; gradient: 10% B to 65% B in 7 minutes; a detector, UV 210/254nm; retention time: 6.15. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AAD-1) (4.9 mg,9.0 μmol, 4.5%) as a white solid. M/z 548.2 (M+H) + (ES+). 1 H NMR(300MHz,MeOH-d4)δ8.45(s,1H),8.12–7.93(m,4H),7.89(s,1H),7.59(t,J=8.0Hz,1H),7.30–7.20(m,1H),5.17(s,2H),4.00(s,3H),3.87(s,2H),3.43(s,3H),2.83(t,J=7.0Hz,2H),2.58(s,2H),1.89(t,J=6.9Hz,2H),0.59(s,4H)。
Example 42: synthesis of 2-cyclopropyl-1-methyl-N- [3- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] phenyl ] -6-oxopyrimidine-4-carboxamide (AAE-6)
Step 1: synthesis of 2-cyclopropyl-6-oxo-1H-pyrimidine-4-carboxylic acid (AAE-3)
To a stirred mixture of sodium 1, 4-diethoxy-1, 3, 4-trioxybutane-2-carboxylate (AAE-1) (8.70 g,1 eq, 41.4 mmol) in water (50 mL) was added NaOH (2.85 g,1.72 eq, 71.3 mmol) and cyclopropanecarboxamide, HCl (AAE 2) (5.00 g,1 eq, 41.5 mmol) in portions at room temperature. The resulting mixture was stirred at 70 ℃ overnight. The residue was acidified to pH 1 with concentrated HCl (aqueous solution). The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and ACN (10% ACN up to 50% in 10 minutes); detector, UV 254/220nm. The resulting mixture was concentrated in vacuo. This gave the sub-title compound (AAE-3) (4 g,22.2mmol, 54%) as a white solid. M/z 181.1 (M+H) + (ES+)。
Step 2: synthesis of methyl 2-cyclopropyl-1-methyl-6-oxopyrimidine-4-carboxylate (AAE-4)
To a stirred mixture of the product from step 1 above (AAE-3) (1.00 g,1 eq, 5.55 mmol) and DMF (25 mL) was added methyl iodide (1.9 g,2.5 eq, 13.9 mmol) at room temperature. The resulting mixture was stirred at 50℃for 16 hours. The mixture was cooled to room temperature and the resulting mixture was extracted with EtOAc (3×150 mL). The combined organic layers were washed with brine (2×150 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (0% ACN up to 100% within 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (AAE-4) (356 mg,1.71mmol, 31%) as a yellow solid. M/z 209.1 (M+H) + (ES+)。
Step 3: synthesis of 2-cyclopropyl-1-methyl-6-oxopyrimidine-4-carboxylic acid (AAE-5)
To a stirred mixture of the product from step 2 above (AAE-4) (356 mg,1 eq., 1.72 mmol) in MeOH (8 mL) was added water (2 mL) and LiOH (206 mg,5 eq., 8.59 mmol) at room temperature.The resulting mixture was stirred at 60℃for 4 hours. The mixture was cooled to room temperature and the residue was acidified with concentrated HCl (aqueous solution) to pH 5. The resulting mixture was extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (0% ACN up to 100% within 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (AAE-5) (300 mg, 759. Mu. Mol, 90%) as a white solid. M/z 195.1 (M+H) + (ES+)。
Step 4: synthesis of 2-cyclopropyl-1-methyl-N- [3- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] phenyl ] -6-oxopyrimidine-4-carboxamide (AAE-6)
EDCI (75.8 mg,2 equivalents, 0.4 mmol) was added to a stirred mixture of the product (AAE-5) (38.4 mg,1 equivalents, 120. Mu. Mol) from step 3 above and intermediate (AAB-8) (50.3 mg,1 equivalents, 120. Mu. Mol) in pyrazine (1 mL) at room temperature. The resulting mixture was stirred at 80℃for 1 hour. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and ACN (0% ACN up to 100% in 20 minutes); detector, UV 254/220nm. The crude product was purified by preparative HPLC (column: XBridge Shield RP OBD column, 19X 250mm,10 μm; mobile phase A: water (0.1% FA), mobile phase B: meOH, flow rate: 25 ml/min; gradient: 10% B to 50% B in 7 min; detector, UV 210/254nm; retention time: 6.15) to give the title compound (AAE-6) (24.6 mg, 57.1. Mu. Mol, 29%) as a white solid. M/z 431.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.45(s,1H),7.90–7.83(m,2H),7.78(m,1H),7.51(t,J=7.9Hz,1H),7.18(m,1H),7.00(s,1H),3.97(s,3H),3.78(s,3H),3.39(s,3H),2.30(m,1H),1.44–1.36(m,2H),1.25(m,2H)。
Example 43: synthesis of 2- [3- [ 1-ethyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] phenyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AAF-9)
Step 1: synthesis of ethyl 1-ethyl-3-iodo-1H-pyrazole-4-carboxylate (AAF-2)
To a stirred solution of ethyl 3-iodo-1H-pyrazole-4-carboxylate (AAF-1) (500 mg,1 eq, 1.88 mmol) in DMF (10 mL) at room temperature was added ethyl iodide (440 mg,1.5 eq, 2.82 mmol) and Cs 2 CO 3 (1.53 g,2.5 eq, 4.70 mmol). The resulting mixture was diluted with water (50 mL). The resulting mixture was extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (3×30 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography eluting with petroleum ether/EtOAc 10/1 to give the sub-title compound (AAF-2) as a yellow oil (340 mg,1.15mmol, 62%). M/z 295.0 (M+H) + (ES+)。
Step 2: synthesis of ethyl 1-ethyl-3- (3-nitrophenyl) -1H-pyrazole-4-carboxylate (AAF-3)
To a stirred solution of the product (AAF-2) (340 mg,1 eq., 1.16 mmol) from step 1 above and 3-nitrophenylboronic acid (AAB-2) (232 mg,1.2 eq., 1.39 mmol) in 1, 4-dioxane (6 mL) under nitrogen at room temperature was added K 2 CO 3 (479 mg,3 equivalents, 3.47 mmol) and water (1.5 mL). Pd (dppf) Cl was added to the above mixture at room temperature under nitrogen atmosphere 2 DCM (85 mg,0.1 eq., 116. Mu. Mol). The resulting mixture was stirred under nitrogen at 80 ℃ for an additional 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The resulting mixture was diluted with water and extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (2×30 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using petroleum ether in EtOAc 5/1 to give the sub-title compound (AAF-3) as a yellow oil (335 mg,1.16mmol, 100%). M/z 290.1 (M+H) + (ES+)。
Step 3: synthesis of 1-ethyl-3- (3-nitrophenyl) -1H-pyrazole-4-carboxylic acid (AAF-4)
To a stirred solution of the product from step 2 above (AAF-3) (335 mg,1 eq, 1.16 mmol) in THF (5 mL) at room temperature was added LiOH (83 mg,3 eq, 3.47 mmol) and water (1 mL) in portions. The resulting mixture was stirred at 60℃for 16 hours. The mixture was cooled to room temperature and acidified to pH 3 with HCl (aqueous solution). The precipitated solid was collected by filtration and washed with EtOAc (3×5 mL). The resulting solid was dried in an oven in vacuo to give the sub-title compound (AAF-4) (300 mg,1.15mmol, 99%) as a white solid. M/z 262.1 (M+H) + (ES+)。
Step 4: synthesis of 1-ethyl-N- (2-methylhydrazine-1-thiocarbonyl) -3- (3-nitrophenyl) -1H-pyrazole-4-carboxamide (AAF-5)
To a stirred solution of the product from step 3 above (AAF-4) (300 mg,1 eq, 1.15 mmol) and 4-methyl-3-thiosemicarbazide (D-2) (146 mg,1.2 eq, 1.39 mmol) in DMF (6 mL) was added HATU (480 mg,1.1 eq, 1.26 mmol) and DIPEA (1.48 g,10 eq, 11.5 mmol) at room temperature. The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. The resulting mixture was used directly in the next step without further purification. M/z349.1 (M+H) + (ES+)。
Step 5: synthesis of 5- (1-ethyl-3- (3-nitrophenyl) -1H-pyrazol-4-yl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (AAF-6)
NaOH (aqueous solution, 10M) (6 mL) was added to the reaction from step 4 above. The resulting mixture was stirred at room temperature under nitrogen atmosphere for 16 hours. The resulting mixture was extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (3×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was dissolved in DMF (10 mL). The resulting mixture was concentrated in vacuo. This gave the sub-title compound (AAF-6) (350 mg,1.06 mmol) as a yellow oil. M/z 331.1 (M+H) + (ES+).
Step 6: synthesis of 3- (1-ethyl-3- (3-nitrophenyl) -1H-pyrazol-4-yl) -4-methyl-4H-1, 2, 4-triazole (AAF-7)
At 0 ℃, get readyTo a stirred solution of the product of step 5 above (AAF-6) (350 mg,1 eq, 1.06 mmol) in DCM (5 mL) was added acetic acid (7 mL) and water (7 mL). The resulting mixture was stirred at room temperature for 16 hours. The mixture was treated with saturated NaHCO 3 The (aqueous) solution is alkalized to pH 10. The resulting mixture was extracted with DCM (3X 50 mL). The combined organic layers were washed with brine (3×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo to give the sub-title compound (AAF-7) as a green oil (290 mg,973 μmol, 77%). M/z 299.1 (M+H) + (ES+)。
Step 7: synthesis of 3- (1-ethyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-3-yl) aniline (AAF-8)
Pd/C39 (10.35 mg,10wt%,0.1 eq., 97. Mu. Mol) was added to a stirred solution of the product from step 6 above (AAF-7) (290 mg,1 eq., 972. Mu. Mol) in MeOH at room temperature under a nitrogen atmosphere. The resulting mixture was stirred at room temperature under a hydrogen atmosphere for 3 hours. Filtering the resulting mixture; the filter cake was washed with MeOH (3X 5 mL). The filtrate was concentrated in vacuo. The resulting oil was dried in oven under vacuum to give the sub-title compound (AAF-8) as an orange oil (262 mg,977 μmol, 57%). M/z 269.1 (M+H) + (ES+)。
Step 8: synthesis of 2- (3- (1-ethyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-3-yl) phenyl) -4- (trifluoromethyl) isoindolin-1-one (AAF-9)
Et is added to a stirred solution of the product from step 7 above (AAF-8) (262 mg,1 eq., 976. Mu. Mol) and EtOH (5 mL) at room temperature 3 N (290 mg,3 eq, 2.93 mmol) and intermediate (A-3) (290 mg,1 eq, 976. Mu. Mol). The residue was purified by preparative TLC using petroleum ether/EtOAc (15/1). The crude product was purified by preparative HPLC under the following conditions (column: YMC-Triart Diol Hilic,20 x 150mm 5um, mobile phase a: water (0.1% nh) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 30B to 50B within 10 minutes; 254/210 nm) to give the title compound (AAF-9) (40.6 mg, 90. Mu. Mol, 9%) as a white solid. m-z 453.1(M+H) + (ES+). 1 H NMR(300MHz,MeOH-d4)δ8.57(s,1H),8.13–8.05(m,2H),7.97(d,J=7.7Hz,1H),7.94–7.83(m,2H),7.77(t,J=7.7Hz,1H),7.47(t,J=8.0Hz,1H),7.36–7.31(m,1H),5.12(s,2H),4.36(q,J=7.3Hz,2H),3.42(s,3H),1.60(t,J=7.3Hz,3H)。
Example 44: synthesis of 2- (2 ' - (4-methyl-4H-1, 2, 4-triazol-3-yl) -5' - (trifluoromethyl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAG-8)
Step 1: synthesis of methyl 4 '-nitro-5- (trifluoromethyl) - [1,1' -biphenyl ] -2-carboxylate (AAG-2)
To methyl 2-bromo-4- (trifluoromethyl) benzoate (AAG-1) (2.5 g,1 eq., 9.00 mmol), intermediate (AAB-2) (2.3 g,1.5 eq., 13.6 mmol) and K under nitrogen 2 CO 3 (3.7 g,3 eq, 27.0 mmol) in 1, 4-dioxane (50 mL) and Pd (dppf) Cl was added 2 DCM (663 mg,0.1 eq, 910. Mu. Mol). The resulting mixture was stirred under nitrogen at 80 ℃ for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The reaction was concentrated and purified by silica gel column chromatography eluting with petroleum ether/EtOAc (5/1) to give the sub-title compound (AAG-2) (2.3 g,7.08mmol, 78%) as a brown yellow solid. M/z 326.1 (M+H) + (ES+)。
Step 2: synthesis of 3 '-nitro-5- (trifluoromethyl) - [1,1' -biphenyl ] -2-carboxylic acid (AAG-3)
To a solution of the product from step 1 above (AAG-2) (500 mg,1 eq., 1.54 mmol) in THF (10 mL) and water (2 mL) was added LiOH (184 mg,5 eq., 7.70 mmol). The resulting solution was stirred at 60℃for 4 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was diluted with water. The pH of the solution was adjusted to 3 with HCl (aqueous, 1M). The solid was collected by filtrationTo give the sub-title compound (AAG-3) (380 mg,1.22mmol, 79%) as a pale yellow solid. M/z 312.0 (M+H) + (ES+)。
Step 3: synthesis of N-methyl-2- (3 '-nitro-5- (trifluoromethyl) - [1,1' -biphenyl ] -2-carbonyl) hydrazine-1-thiocarboxamide (AAG-4)
To a stirred mixture of the product from step 2 above (AAG-3) (380 mg,1 eq, 1.22 mmol) and 4-methyl-3-thiosemicarbazide (D-2) (193 mg,1.5 eq, 1.83 mmol) and DIPEA (473 mg,3 eq, 3.66 mmol) in DMF (12 mL) was added HATU (557 mg,1.2 eq, 1.47 mmol) at room temperature. The residue was purified by reverse flash chromatography under the following conditions: column, C18 silica gel; mobile phase, ACN/water (0.1% FA), gradient 10% to 100% in 20 min; detector, UV 254/220nm. The product-containing fractions were combined and evaporated to give the sub-title compound (AAG-4) (306 mg,769 μmol, 63%) as a yellow solid. M/z 399.1 (M+H) + (ES+)。
Step 4: synthesis of 4-methyl-5- (3 '-nitro-5- (trifluoromethyl) - [1,1' -biphenyl ] -2-yl) -4H-1,2, 4-triazole-3-thiol (AAG-5)
To a stirred solution of the product from step 3 above (AAG-4) (559 mg,1 eq, 1.40 mmol) in DMF (10 mL) was added NaOH (aqueous solution, 1M) (8 mL) at room temperature. The resulting mixture was stirred at 50 ℃ overnight. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (10% ACN up to 70% within 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (AAG-5) (220 mg, 579. Mu. Mol, 41%) as a yellow solid. M/z 381.1 (M+H) + (ES+)。
Step 5: synthesis of 4-methyl-3- (3 '-nitro-5- (trifluoromethyl) - [1,1' -biphenyl ] -2-yl) -4H-1,2, 4-triazole (AAG-6)
At 0deg.C, the product (AAG-5) (220 mg,1 eq., 580. Mu. Mol) from step 4 above was added to DCMTo a stirred mixture of 3 mL) were added acetic acid (0.93 mL) and hydrogen peroxide (0.72 mL,30 wt%). The resulting mixture was stirred at room temperature for 2 hours. The residue was basified with NaOH (aqueous, 1M) to pH 8. The resulting mixture was diluted with water and extracted with DCM (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The reaction was purified by reverse flash chromatography under the following conditions: column, C18 silica gel; mobile phase, ACN/water (0.1% NH 4 HCO 3 ) Gradient from 10% to 70% over 20 minutes; detector, UV 254/220nm. The product-containing fractions were combined and evaporated to give the sub-title compound (AAG-6) (108 mg,310 μmol, 54%) as a pale yellow oil. M/z 349.1 (M+H) + (ES+)。
Step 6: synthesis of 2' - (4-methyl-4H-1, 2, 4-triazol-3-yl) -5' - (trifluoromethyl) - [1,1' -biphenyl ] -3-amine (AAG-7)
Pd/C39 (1 mg,10Wt%,0.03 eq, 9.4. Mu. Mol) was added to a stirred solution of the product (AAG-6) (108 mg,1 eq, 310. Mu. Mol) from step 5 above in acetic acid (2 ml) at room temperature under an atmosphere of hydrogen. The resulting mixture was stirred at room temperature under a hydrogen atmosphere for 2 hours. Filtering the resulting mixture; the filter cake was washed with acetic acid (2 x2 mL). The residue was purified by reverse flash chromatography under the following conditions: column, C18 silica gel; mobile phase, ACN/water (0.1% nh 4 HCO 3 ) A gradient of 10% to 60% over 20 minutes; detector, UV 254/220nm. This gave the sub-title compound (AAG-7) (65 mg, 200. Mu. Mol, 14.4%) as an off-white solid. M/z 319.1 (M+H) + (ES+)。
Step 7: synthesis of 2- (2 ' - (4-methyl-4H-1, 2, 4-triazol-3-yl) -5' - (trifluoromethyl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAG-8)
Et is added to a stirred solution of the product from step 6 above (AAG-7) (65 mg,1 eq, 200. Mu. Mol) in EtOH (10 ml) at room temperature 3 N (62 mg,3 eq, 620. Mu. Mol) and intermediate (A-3) (61 mg,1 eq, 200. Mu. Mol). The resulting mixture was stirred under nitrogen at 80 ℃ for 2 hours. The mixture was cooled to room temperature and held at true temperature Concentrating in the air. The residue was purified by reverse flash chromatography under the following conditions: column: sun Fire preparation type C18 OBD column, 19 x 150mm,5 μm 10nm; mobile phase a: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 40% B to 68% B in 7 minutes; wavelength: 254/210nm; retention time: 6.32 to give the title compound (AAG-8) (20.9 mg, 42. Mu. Mol, 13%) as a white solid. M/z 503.1 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.44(s,1H),8.11(d,J=7.6Hz,1H),8.05–7.75(m,7H),7.49(t,J=8.0Hz,1H),7.15–7.13(m,1H),5.12(s,2H),3.25(s,3H)。
Example 45: synthesis of 2- (3-ethoxy-5- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) phenyl) -4- (trifluoromethyl) isoindolin-1-one (AAI-7)
Step 1: synthesis of 1-bromo-3-ethoxy-5-nitrobenzene (AAI-2)
To a stirred solution of 3-bromo-5-nitrophenol (AAI-1) (1.09 g,1 eq, 5.00 mmol) in DMF (10 mL) at room temperature was added K 2 CO 3 (1.73 g,2.50 equivalents, 12.5 mmol) and ethyl iodide (1.60 g,2 equivalents, 10.0 mmol). The resulting mixture was stirred at room temperature overnight. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. This gave the sub-title compound (AAI-2) (1.08 g,4.41mmol, 88%) as a yellow solid. M/z246.1/248.1 (M+H) + (ES+)。
Step 2: synthesis of 2- (3-ethoxy-5-nitrophenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (AAI-4)
Into a 250-mL round bottom flask purged with an inert nitrogen atmosphere and maintained was placed dioxane (50 mL) containing the product (AAI-2) (2.46 g,1 equivalent, 10.0 mmol) from step 1 above and bis (pinacolato) diboron (AAI-3) (3.05 g,1.2 equivalent, 12.0 mmol) followed by the addition of potassium acetate (2.45)g,2.5 equivalents, 25.0 mmol) and Pd (dppf) Cl 2 DCM (409 mg,0.05 eq., 501. Mu. Mol). The resulting solution was stirred at 100℃for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (50/1). This gave the sub-title compound (AAI-4) (2 g,6.82mmol, 68%) as a brown solid. M/z 294.1 (M+H) + (ES+)。
Step 3:3- (5- (3-ethoxy-5-nitrophenyl) -1-methyl-1H-pyrazol-4-yl) -4-methyl-4H-1, 2, 4-triazole (AAI-5)
A25-mL round bottom flask purged with an inert nitrogen atmosphere was charged with a solution containing the product (AAI-4) (145 mg,1.2 eq, 496. Mu. Mol), intermediate (AAH-5) (100 mg,1 eq, 413. Mu. Mol) and K from step 2 above under a nitrogen atmosphere at room temperature 2 CO 3 (171 mg,3 eq, 1.24 mmol) dioxane (5 mL) and water (1 mL), then Pd (dppf) Cl was added 2 DCM (30 mg,0.1 eq, 41. Mu. Mol). The resulting solution was stirred under nitrogen at 100 ℃ for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). This gave the sub-title compound (AAI-5) (96.3 mg, 294. Mu. Mol, 71%) as a brown solid. M/z 329.1 (M+H) + (ES+)。
Step 4: synthesis of 3-ethoxy-5- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) aniline (AAI-6)
To a 25-mL round bottom flask under nitrogen was placed MeOH (5 mL) containing the product from step 3 above (AAI-5) (96 mg,1 eq, 293. Mu. Mol) and Pd/C39 (31 mg,10Wt%,0.1 eq, 29.3. Mu. Mol) was added to the above mixture under hydrogen. The resulting solution was stirred at room temperature for 3 hours. The resulting mixture was filtered and the filter cake was washed with EtOAc (3×5 mL). The filtrate was concentrated in vacuo to give a brown/yellow solidThe sub-title compound (AAI-6) (71.7 mg, 240. Mu. Mol, 82%) in the form of a body. M/z 299.2 (M+H) + (ES+)。
Step 5: synthesis of 2- (3-ethoxy-5- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) phenyl) -4- (trifluoromethyl) isoindolin-1-one (AAI-7)
Into a 25-mL round bottom flask at room temperature was placed EtOH (3 mL) containing the product from step 4 above (AAI-6) (71.7 mg,1 eq, 240. Mu. Mol) and intermediate (A-3) (107 mg,1.5 eq, 360. Mu. Mol) followed by Et 3 N (73 mg,3 eq, 720. Mu. Mol). The resulting solution was stirred at 80℃for 3 hours. The resulting mixture was cooled to room temperature and concentrated. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared C18 OBD column, 20×150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 30B to 60B in 7 minutes; a detector, UV 210/254nm; retention time: 6.32. this gave the title compound (AAI-7) (6.0 mg, 12. Mu. Mol, 5.1%) as a white solid. M/z 483.1 (M+H) + (ES+). 1 H NMR(300MHz,MeOH-d4)δ8.45(s,1H),8.10(d,J=7.7Hz,1H),7.98(d,J=7.8Hz,1H),7.86(s,1H),7.78(t,J=7.7Hz,1H),7.64(t,J=2.1Hz,1H),7.49(s,1H),6.82–6.75(m,1H),5.15(s,2H),4.14–3.95(m,5H),3.41(s,3H),1.39(t,J=7.0Hz,3H)。
Example 46: synthesis of 2- (5 ' -bromo-2 ' - (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAJ-9)
Step 1: synthesis of methyl 4-bromo-2-iodobenzoate (AAJ-2)
To a stirred solution of 4-bromo-2-iodobenzoic acid (AAJ-1) (5.00 g,1 eq, 15.3 mmol) in MeOH (30 mL) at room temperature was added sulfuric acid (5 mL). The resulting mixture was stirred at 80 ℃ for 3 hours, then cooled to room temperature and concentrated in vacuo. The resulting mixture was diluted with water. The solution was treated with saturated sodium carbonate solution (water Solution) alkalized to pH 8. The resulting mixture was extracted with DCM (3X 300 mL). The combined organic layers were washed with brine (2×300 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. This gave the sub-title compound (AAJ-2) (4.7 g,13.8mmol, 90%) as a yellow solid. M/z 340.9/342.9 (M+H) + (ES+)。
Step 2: synthesis of 5-bromo-3 '-nitro- [1,1' -biphenyl ] -2-carboxylic acid methyl ester (AAJ-3)
To a solution of the product (AAJ-2) (1.7 g,1 equivalent, 5.00 mmol) from step 1 above and 3-nitrophenylboronic acid (AAB-2) (832 mg,1 equivalent, 4.98 mmol) in 1, 4-dioxane (25 mL) and water (5 mL) under a nitrogen atmosphere was added K 2 CO 3 (2.1 g,3 eq, 15.0 mmol) and Pd (dppf) Cl 2 DCM (365 mg,0.1 eq. 499. Mu. Mol). After stirring at 80 ℃ for 3 hours, the mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography petroleum ether/EtOAc (3/1) to give the sub-title compound (AAJ-3) (1.5 g,4.48mmol, 90%) as a yellow solid. M/z 336.0/338.0 (M+H) + (ES+)。
Step 3: synthesis of 5-bromo-3 '-nitro- [1,1' -biphenyl ] -2-carboxylic acid (AAJ-4)
To a stirred solution of the product from step 2 above (AAJ-3) (830 mg,1 eq, 2.47 mmol) in THF (8 mL) and water (2 mL) at room temperature was added LiOH (177 mg,3 eq, 7.41 mmol). The resulting mixture was stirred at 60 ℃ for 2 hours, then cooled to room temperature and concentrated in vacuo. The residue was diluted with water. The pH of the solution was adjusted to 3 with HCl (aqueous, 1M). The solid was collected by filtration to give the sub-title compound (AAJ-4) (750 mg,2.34mmol, 94%) as a brown solid. M/z 322.0/324.0 (M+H) + (ES+).
Step 4: synthesis of 2- (5-bromo-3 '-nitro- [1,1' -biphenyl ] -2-carbonyl) -N-methylhydrazine-1-thiocarboxamide (AAJ-5)
To the product (AAJ-4) from step 3 above (1.1 g at 0 ℃CTo a stirred solution of 1 equivalent, 3.42 mmol) and pyridine (1.6 g,6 equivalents, 20.5 mmol) in DMF (5 mL) was added T3P (4.4 g,4 equivalents, 13.7 mmol) and 4-methyl-3-thiosemicarbazide (D-2) (1.1 g,3 equivalents, 10.2 mmol). The resulting mixture was stirred at room temperature overnight. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (30% ACN up to 70% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AAJ-5) (1 g,2.44mmol, 72%) as a yellow solid. M/z 409.0/411.0 (M+H) + (ES+)。
Step 5: synthesis of 5- (5-bromo-3 '-nitro- [1,1' -biphenyl ] -2-yl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (AAJ-6)
To a stirred solution of the product from step 4 above (AAJ-5) (1.0 g,1 eq, 2.44 mmol) in DMF (8 mL) was added NaOH (aqueous solution, 1M) (15 mL) at room temperature. The resulting mixture was stirred at 50 ℃ overnight. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (30% ACN up to 50% in 7 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AAJ-6) (600 mg,1.54mmol, 63%) as a white solid. M/z 390.0392.0 (M+H) + (ES+)。
Step 6:3- (5-bromo-3 '-nitro- [1,1' -biphenyl ] -2-yl) -4-methyl-4H-1, 2, 4-triazole (AAJ-7)
To a stirred solution of the product from step 5 above (AAJ-6) (140 mg,1 eq, 358. Mu. Mol) in DCM (5 mL) at 0deg.C was added acetic acid (43 mg,2 eq, 716. Mu. Mol) and hydrogen peroxide solution (61 mg,30Wt%,5 eq, 1.79 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The resulting mixture was diluted with water. The solution was saturated with NaHCO 3 The solution was basified to pH 8. The resulting mixture was extracted with DCM (3X 80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. This results inTo the sub-title compound (AAJ-7) as a yellow solid (120 mg, 335. Mu. Mol, 93%). M/z359.0/361.0 (M+H) + (ES+)。
Step 7: synthesis of 5' -bromo-2 ' - (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-amine (AAJ-8)
To the product (AAJ-7) (130 mg,1 eq., 362. Mu. Mol) from step 6 above and NH at room temperature 4 Zinc powder (95 mg,4 eq, 1.45 mmol) was added to a stirred solution of Cl (97 mg,5 eq, 1.81 mmol) in MeOH (5 mL). The resulting mixture was stirred at 70℃for 2 hours. The resulting mixture was cooled to room temperature and filtered. The filtrate was concentrated in vacuo to give the sub-title compound (AAJ-8) (100 mg,305 μmol, 84%) as a yellow solid. M/z 329.2/331.2 (M+H) + (ES+).
Step 8:2- (5 ' -bromo-2 ' - (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAJ-9)
Et is added to a stirred solution of the product (AAJ-8) (50 mg,1 eq., 152. Mu. Mol) from step 7 above and intermediate (A-3) (50 mg,1.1 eq., 167. Mu. Mol) in EtOH (5 mL) at room temperature 3 N (46 mg,3 eq, 450. Mu. Mol). The resulting mixture was stirred at 80 ℃ for 3 hours and then cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and ACN (30% ACN up to 70% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19X 150mm 5um; mobile phase a: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 40B to 60B in 10 minutes; a detector, UV 254/210nm; retention time: 10.38. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AAJ-9) (20.8 mg,41 μmol, 27%) as a white solid. M/z 513.2 (M+H) + (ES+). 1 H NMR(300MHz,MeOH-d4)δ8.39(s,1H),8.10(d,J=7.6Hz,1H),8.05–7.82(m,3H),7.86–7.75(m,3H),7.55(d,J=8.21Hz,1H),7.45(t,J=7.95Hz,1H),7.10–7.07(m,1H),5.10(s,2H),3.21(s,3H)。
Example 47: synthesis of 2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAK-2)
Step 1: synthesis of 2'- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-amine (P-1)
Pd/C39 (30 mg,10Wt%,0.5 eq., 283. Mu. Mol) was added to a solution of intermediate (AAJ-8) (200 mg,1 eq., 559. Mu. Mol) in MeOH (10 mL) under a hydrogen atmosphere. The resulting mixture was stirred at room temperature under a hydrogen atmosphere for 3 hours. After the reaction was completed, the reaction mixture was filtered. The filtrate was evaporated in vacuo to give the sub-title compound (P-1) (135 mg,540 μmol, 96%) as a yellow solid. M/z 251.1 (M+H) + (ES+)。
Step 2:2- (-2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAK-2)
Et is added to a stirred solution of the product (P-1) (80 mg, 319. Mu. Mol,1 eq.) from step 1 above and intermediate (A-3) (104 mg,1.1 eq., 351. Mu. Mol) in EtOH (5 mL) at room temperature 3 N (97.0 mg,3 eq., 962. Mu. Mol). The resulting mixture was stirred at 80 ℃ for 3 hours, and the resulting mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (30% ACN up to 70% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: YMC-Triart Diol Hilic, 20X 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 . H 2 O), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 34B to 64B in 7 minutes; a detector, UV 254/210nm; retention time: 6.32. combining the product-containing fractions and partially in trueEvaporated in air and lyophilized overnight to give the sub-title compound (AAK-2) (16.9 mg,39 μmol, 12%) as a white solid. M/z 435.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.40(s,1H),8.16-7.97(m,3H),7.87-7.77(m,1H),7.73-7.62(m,3H),7.62-7.52(m,2H),7.38(t,J=8.0Hz,1H),6.97-6.81(m,1H),5.12(s,2H),3.08(s,3H)。
Example 48: synthesis of 2- [3- [ 1-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] phenyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AAL-8)
Step 1: synthesis of ethyl 1-methyl-3- (3-nitrophenyl) -1H-pyrazole-4-carboxylate (AAL-2)
To a stirred solution of 3-bromo-1-methylpyrazole-4-carboxylic acid ethyl ester (AAL-1) (1.00 g,1 eq, 4.29 mmol) and intermediate (AAB-2) (860 mg,1.2 eq, 4.07 mmol) in 1, 4-dioxane (20 mL) and water (5 mL) at room temperature under nitrogen atmosphere was added K 2 CO 3 (1.78 g,3 eq, 12.9 mmol) and Pd (dppf) Cl 2 DCM (350 mg,0.1 eq, 429. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature, and the resulting mixture was diluted with water (40 mL). The resulting mixture was extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography eluting with hexane/EtOAc (5/1) to give the sub-title compound (AAL-2) (1 g,3.63mmol, 85%) as a white solid. M/z 276.1 (M+H) + (ES+).
Step 2: synthesis of 1-methyl-3- (3-nitrophenyl) -1H-pyrazole-4-carboxylic acid (AAL-3)
To a stirred solution of the product from step 1 above (AAL-2) (1.00 g,1 eq, 3.63 mmol) in THF (25 mL) and water (5 mL) at room temperature was added LiOH (261 mg,3 eq, 10.9 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature and the resulting mixture was treated with water @50 mL) was diluted. The mixture was acidified with HCl (aqueous, 1M) to pH 3. The precipitated solid was collected by filtration and washed with water (3×5 mL). This gave the sub-title compound (AAL-3) (800 mg,3.24mmol, 89%) as a white solid. M/z 248.1 (M+H) + (ES+)。
Step 3: synthesis of N-methyl-2- (1-methyl-3- (3-nitrophenyl) -1H-pyrazole-4-carbonyl) hydrazine-1-thiocarboxamide (AAL-4)
To a stirred solution of the product from step 1 above (AAL-3) (500 mg,1 eq, 2.02 mmol) and 4-methyl-3-thiosemicarbazide (D-2) (277 mg,1.3 eq, 2.63 mmol) in DMF (25 mL) was added DIPEA (784 mg,3 eq, 6.07 mmol) and HATU (923 mg,1.2 eq, 2.43 mmol) at room temperature. The resulting mixture was stirred at room temperature for 3 hours. The resulting mixture was used directly in the next step without further purification. M/z 335.1 (M+H) + (ES+)。
Step 4: synthesis of 4-methyl-5- (1-methyl-3- (3-nitrophenyl) -1H-pyrazol-4-yl) -4H-1,2, 4-triazole-3-thiol (AAL-5)
NaOH (aqueous solution, 1M) (30 mL) was added to the above product (AAL-4) from step 3 above at room temperature, and the resulting mixture was stirred for 2 days. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (10% ACN up to 50% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AAL-5) (500 mg,1.58mmol, 88%) as a yellow oil. M/z 317.0 (M+H) + (ES+)。
Step 5: synthesis of 4-methyl-3- (1-methyl-3- (3-nitrophenyl) -1H-pyrazol-4-yl) -4H-1,2, 4-triazole (AAL-6)
To a stirred solution of the product from step 4 above (AAL-5) (560 mg,1 eq, 1.77 mmol) in DCM (20 mL) at 0deg.C was added acetic acid (2.78 mL,27.4 eq, 48.5 mmol) and hydrogen peroxide (1.85 mL,44.9 eq, 79.4 mmol), and the resulting mixture was stirred at 0deg.C for 2 hours. The mixture was treated with saturated NaHCO 3 The (aqueous) solution was basified to pH8. The resulting mixture was partitioned with EtOAc (3x80 mL) of the extract. The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. This gave the sub-title compound (AAL-6) (500 mg,1.76mmol, 99%) as a yellow oil. M/z 285.1 (M+H) + (ES+)。
Step 6: synthesis of 3- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-3-yl) aniline (AAL-7)
Pd/C39 (30 mg,10Wt%,0.2 eq, 281. Mu. Mol) was added to a solution of the product from step 5 above (AAL-6) (400 mg,1 eq, 1.41 mmol) in MeOH (20 mL) in a 50mL round bottom flask under nitrogen. The mixture was hydrogenated at room temperature under a hydrogen atmosphere overnight using a hydrogen balloon, then filtered through a celite pad and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1) to give the sub-title compound (AAL-7) as an off-white solid (300 mg,1.18mmol, 84%). M/z 255.1 (M+H) + (ES+)。
Step 7: synthesis of 2- (3- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-3-yl) phenyl) -4- (trifluoromethyl) isoindolin-1-one (AAL-8)
Et is added to a stirred solution of the product (AAL-7) (100 mg,1 eq., 393. Mu. Mol) from step 6 above and intermediate (A-3) (117 mg,1 eq., 338. Mu. Mol) in EtOH (4 mL) at room temperature 3 N (119 mg,3 eq, 1.18 mmol). The resulting mixture was stirred at 80℃for 4 hours. The mixture was cooled to room temperature and the crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (10% ACN up to 50% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge Shield RP OBD column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 22% B to 38% B in 10 minutes; a detector, UV 254nm; retention time: 9.72 To give the sub-title compound as a white solid(AAL-8)(31.4mg,72μmol,18%)。m/z 439.0(M+H) + (ES+). 1 H NMR(300MHz,CD3OD)δ8.69(s,1H),8.13–8.05(m,2H),8.00–7.93(m,2H),7.88–7.84(m,1H),7.77(t,J=7.7Hz,1H),7.48(t,J=8.0Hz,1H),7.38–7.33(m,1H),5.12(s,2H),4.08(s,3H),3.46(s,3H)。
Example 49: synthesis of 2- (4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AAM-3)
Step 1: synthesis of 2-chloro-4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridine (AAM-2)
Intermediate (AAH-4) (200 mg,1 eq, 826. Mu. Mol), 2-chloropyridin-4-ylboronic acid (AAM-1) (65 mg,0.5 eq, 413. Mu. Mol) and NaHCO were added to the mixture under a nitrogen atmosphere at room temperature 3 (aqueous solution, 1M) (2 mL) Pd (dppf) Cl was added to a stirred solution of 1, 4-dioxane (2 mL) 2 DCM (68 mg,0.1 eq, 83. Mu. Mol). The final reaction mixture was irradiated with microwave radiation at 100 ℃ under nitrogen atmosphere for 30 minutes. The mixture was cooled to room temperature, and the resulting mixture was diluted with water (30 mL). The resulting mixture was extracted with EtOAc (3×30 mL) and the combined organic layers were washed with brine (2×30 mL), dried (Na 2 SO 4 ) And concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (30/1) to give the sub-title compound (AAM-2) as a yellow solid (34 mg, 124. Mu. Mol, 12%). M/z 275.1/277.1 (M+H) + (ES+)。
Step 2:2- (4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AAM-3)
Pd (OAc) was added to a stirred solution of the product from step 1 above (AAM-2) (34 mg,1 eq, 124. Mu. Mol), intermediate (AC-2) (50 mg,2 eq, 248. Mu. Mol) and potassium phosphate (53 mg,2 eq, 248. Mu. Mol) in 1, 4-dioxane (4 mL) at room temperature under nitrogen atmosphere 2 (3mg,0.1Equivalent weight, 12. Mu. Mol) and Xantphos (14 mg,0.2 equivalent, 25. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 12 hours. The mixture was cooled to room temperature, and the resulting mixture was diluted with water (30 mL). The resulting mixture was extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (30 mL), dried (Na 2 SO 4 ) And concentrated in vacuo. The residue was purified by preparative TLC with DCM/MeOH (15/1) and the crude product was purified by preparative HPLC under the following conditions (column: YMC-Triart Diol Hilic,20 x 150mm5um; mobile phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 23B to 43B in 7 minutes; a detector, UV 210/254nm; retention time: 8.97 To give the sub-title compound (AAM-3) (6.8 mg,15 μmol, 12%) as a white solid. M/z 440.0 (M+H) + (ES+). 1 H NMR(400MHz,CD3OD)δ8.57–8.51(m,3H),8.13(d,J=7.7Hz,1H),8.02(d,J=7.7Hz,1H),7.94(s,1H),7.79(t,J=7.7Hz,1H),7.24(dd,J=5.1,1.6Hz,1H),5.34(s,2H),4.06(s,3H),3.60(s,3H)。
Example 50: synthesis of 2- (6-chloro-4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AAN-7)
Step 1: synthesis of 5- (2, 6-dichloropyridin-4-yl) -1-methyl-1H-pyrazole-4-carboxylic acid ethyl ester (AAN-2)
To intermediate (AAB-1) (1.00 g,1 equivalent, 4.29 mmol) and 2, 6-dichloropyridin-4-ylboronic acid (AAN-1) (823 mg,1 equivalent, 4.29 mmol) were added under a nitrogen atmosphere at room temperature 2 CO 3 (1.77 g,3 eq., 12.9 mmol) Pd (dtbpf) Cl was added to a stirred solution of 1, 4-dioxane (37.5 mL) and water (7.5 mL) 2 DCM (280 mg,0.1 eq, 420. Mu. Mol). The resulting mixture was stirred overnight at 60 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by column chromatography on silica gel,elution with petroleum ether/EtOAc (5/1) afforded the sub-title compound (AAN-2) as a yellow solid (550 mg,18.3mmol, 55%). M/z 300.0/302.0 (M+H) + (ES+)。
Step 2: synthesis of 5- (2, 6-dichloropyridin-4-yl) -1-methyl-1H-pyrazole-4-carboxylic acid (AAN-3)
To a solution of the product from step 1 above (AAN-2) (550 mg,1 eq, 1.83 mmol) in THF (20 mL) and water (5 mL) was added LiOH (439 mg,10 eq, 18.3 mmol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight and then cooled to room temperature. The mixture was acidified with HCl (aqueous, 1M) to pH 3. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. This gave the sub-title compound (AAN-3) (850 mg,3.12mmol, crude product) as a yellow solid. M/z 272.1/274.1 (M+H) + (ES+)。
Step 3: synthesis of 2- (5- (2, 6-dichloropyridin-4-yl) -1-methyl-1H-pyrazole-4-carbonyl) -N-methylhydrazine-1-thiocarboxamide (AAN-4)
To a stirred solution of the product from step 2 above (AAN-3) (850 mg,1 eq, 3.12 mmol) and DIPEA (1.21 g,3 eq, 9.37 mmol) in DMF (10 mL) was added 4-methyl-3-thiosemicarbazide (D-2) (399 mg,1 eq, 3.12 mmol) and HATU (1.19 g,1 eq, 3.12 mmol) at 0deg.C. The resulting mixture was stirred at room temperature for 2 hours and the crude product was used directly in the next step without any further purification. M/z 359.2/361.2 (M+H) + (ES+)。
Step 4: synthesis of 5- (5- (2, 6-dichloropyridin-4-yl) -1-methyl-1H-pyrazol-4-yl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (AAN-5)
NaOH (aqueous, 1M) (12.5 mL) was added to the reaction mixture (AAN-4) from step 3 above at 0deg.C. The resulting mixture was stirred at room temperature overnight. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (36% ACN up to 48% in 10 minutes); detector, UV 254/220nm. Will contain the productFractions were combined and concentrated in vacuo. This gave the sub-title compound (AAN-5) (620 mg,18.1mmol, 92%) as a white solid. M/z 341.2/343.2 (M+H) + (ES+)。
Step 5: synthesis of 2, 6-dichloro-4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridine (AAN-6)
To a solution of the product from step 4 above (AAN-5) (620 mg,1 eq, 1.81 mmol) in DCM (10 mL) was added acetic acid (218.24 mg,2 eq, 3.63 mmol) and hydrogen peroxide (309 mg,30Wt%,5 eq, 9.08 mmol) at 0deg.C. The resulting mixture was stirred at room temperature for 1 hour, then the mixture was taken up in saturated NaHCO 3 The (aqueous, 1M) solution was basified to pH 8. The resulting mixture was diluted with water and extracted with DCM (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. This gave the sub-title compound (AAN-6) (360 mg,1.17mmol, 58%) as a white solid. M/z 309.1/311.1 (M+H) + (ES+)。
Step 6: synthesis of 2- (6-chloro-4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AAN-7)
Pd (OAc) was added to a stirred solution of the product (AAN-6) (31 mg,1 eq, 100. Mu. Mol) and intermediate (AC-2) (20 mg,1 eq, 100. Mu. Mol) from step 5 above in 1, 4-dioxane (5 mL) under nitrogen at room temperature 2 (2.24 mg,0.1 eq, 10. Mu. Mol) and Xantphos (12 mg,0.2 eq, 20. Mu. Mol). The resulting mixture was stirred under nitrogen at 100 ℃ for 3 hours, then cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19 x 150mm,5 μm 10nm; mobile phase a: water (0.1% fa), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 30% B to 60% B,60% B in 7 minutes; wavelength: 254/210nm; retention time: 6.32. this gave the title compound (AAN-7) (8.3 mg,18mmol, 27%) as a white solid. M/z 474.0 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.53(s,1H),8.49(d,J=1.2Hz,1H),8.12(d,J=7.7Hz,1H),8.03(d,J=7.7Hz,1H),7.96(s,1H),7.79(t,J=7.7Hz,1H),7.35(d,J=1.2Hz,1H),5.29(s,2H),4.06(s,3H),3.68(s,3H)。
Example 51: synthesis of 2- (6-hydroxy-4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AAO-1)
Intermediate (AAN-7) (75 mg,1 eq, 150. Mu. Mol) and ethylamine (36 mg,5 eq, 10. Mu. Mol) and Cs were added to the mixture under a nitrogen atmosphere at room temperature 2 CO 3 (206 mg,4 eq, 630. Mu. Mol) Pd (OAc) was added to the stirred mixture of 1, 4-dioxane 2 (3.6 mg,0.1 eq, 20. Mu. Mol) and XantPhos (18 mg,0.2 eq, 30 mmol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere, then cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19X 150mm,5um; mobile phase a: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 30B to 60B in 7 minutes; a detector, UV 254/210nm; retention time: 6.08. this gave the title compound (AAO-1) (9.1 mg, 20. Mu. Mol, 12.49%) as a white solid. M/z 456.0 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.51(s,1H),8.07(d,J=7.7Hz,1H),7.99(d,J=7.7Hz,1H),7.78–7.75(s,2H),7.75(t,J=7.7Hz,1H),6.46(d,J=1.2Hz,1H),5.24(s,2H),4.05(s,3H),3.58(s,3H)。
Example 52: synthesis of 2-cyclopropyl-N- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6-oxo-1, 6-dihydropyrimidine-4-carboxamide (AAP-1)
Intermediate (P-1) (50 mg,1 eq, 200. Mu. Mol) was added to an 8mL sealed tube at 0deg.C) Intermediate (AAE-3) (43 mg,1.2 eq, 240. Mu. Mol) and DIPEA (77 mg,3 eq, 599. Mu. Mol) in DMF (3 mL) was added HATU (114 mg,1.5 eq, 300. Mu. Mol). The resulting mixture was stirred at room temperature overnight. The crude product was purified by preparative HPLC (column: sunFire preparative C18 OBD column, 19.150 mm,5 μm 10nm; mobile phase A: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 15% B to 35% B,35% B; wavelength: 254/210nm; retention time: 9.77) to give the title compound (AAP-1) (22 mg, 70. Mu. Mol, 25%) as a white solid. M/z 413.3 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.39(s,1H),7.79–7.65(m,4H),7.63–7.60(m,2H),7.34(t,J=7.9Hz,1H),7.01–6.92(m,2H),3.18(s,3H),2.04–1.95(m,1H),1.43–1.36(m,2H),1.25–1.17(m,2H)。
Example 53: synthesis of 6- ((5-azaspiro [2.4] heptan-5-yl) methyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAQ-1)
To a 20mL sealed tube at room temperature was added intermediate (AAC-2) (100 mg,1 eq, 246. Mu. Mol), intermediate (P-1) (74 mg,1.2 eq, 295. Mu. Mol) and Et 3 N (75 mg,3 eq. 738. Mu. Mol) EtOH (3 mL). The resulting mixture was stirred at 80℃for 16 hours. The resulting mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: YMC-act three C18 ExRS,30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 43% B to 63% B,63% B in 9 minutes; wavelength: 254/220nm; retention time: 8.33 To give the title compound (AAQ-1) (23 mg,42 μmol, 18%) as a white solid. M/z 544.2 (M+H) + (ES+). 1 H NMR(300MHz,MeOH-d4)δ8.39(s,1H),8.08(s,1H),8.01(s,1H),7.89–7.83(m,1H),7.78–7.70(m,3H),7.67–7.60(m,2H),7.44(t,J=8.0Hz,1H),7.15–7.08(m,1H),5.07(s,2H),3.87(s,2H),3.21(s,3H),2.84(t,J=7.0Hz,2H),2.58(s,2H),1.89(t,J=6.9Hz,2H),0.66–0.53(m,4H)。
Example 54: synthesis of 2- (5-ethoxy-2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAR-5)
Step 1: synthesis of 2- (3-ethoxy-5-nitrophenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (AAR-2).
Pd (dppf) Cl was added to a solution of 1-bromo-3-ethoxy-5-nitrobenzene (AAI-1) (500 mg,1 eq, 2.03 mmol), intermediate (AAI-3) (1.55 mg,3 eq, 6.10 mmol) and potassium acetate (598 mg,3 eq, 6.10 mmol) in 1, 4-dioxane (10 mL) under nitrogen 2 DCM (294 mg,0.2 eq., 410. Mu. Mol). After stirring overnight at 100 ℃ under nitrogen atmosphere, the resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (10% ACN up to 50% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and evaporated to give the sub-title compound (AAR-2) as a pale reddish brown solid (445 mg,1.52mmol, 75%). M/z 294.1 (M+H) + (ES+)。
Step 2: synthesis of 3- (3 ' -ethoxy-5 ' -nitro- [1,1' -biphenyl ] -2-yl) -4-methyl-4H-1, 2, 4-triazole (AAR-3)
To the product (AAR-2) (231 mg,1 equivalent, 790. Mu. Mol), intermediate (D-5) (225 mg,1.2 equivalent, 950. Mu. Mol) and K from step 1 above were added under a nitrogen atmosphere 2 CO 3 (327 mg,3 eq, 2.36 mmol) in 1, 4-dioxane (5 mL) Pd (dppf) Cl was added 2 DCM (115 mg,0.2 eq., 160. Mu. Mol). After stirring for 2 hours at 100 ℃ under nitrogen atmosphere, the resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phaseWater (0.1% NH) 4 HCO 3 ) And ACN (10% ACN up to 50% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and evaporated to give the sub-title compound (AAR-3) (137 mg,423 μmol, 62%) as a pale reddish brown solid. M/z 325.1 (M+H) + (ES+)。
Step 3: synthesis of 5-ethoxy-2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-amine (AAR-4)
Pd/C39 (9 mg,10Wt%,0.1 eq, 85. Mu. Mol) was added to a solution of the product from step 2 above (AAR-3) (281mg, 1 eq, 870. Mu. Mol) in MeOH (10 mL) in a 50mL round bottom flask under nitrogen. The mixture was hydrogenated under a hydrogen atmosphere at room temperature for 4 hours. Filtering the resulting mixture; the filter cake was washed with MeOH (3X 5 mL). The filtrate was concentrated in vacuo. This gave the sub-title compound (AAR-4) (230 mg, 782. Mu. Mol, 90%) as a pale green solid. M/z 295.1 (M+H) + (ES+)。
Step 4: synthesis of 2- (5-ethoxy-2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAR-5).
Et is added to a stirred solution of the product (AAR-4) (50 mg,1 eq., 170. Mu. Mol) from step 3 above and intermediate (A-3) (61 mg,1.2 eq., 200. Mu. Mol) in EtOH (2 mL) at room temperature 3 N (26 mg,1.5 eq, 260 mmol). The resulting mixture was stirred under nitrogen at 80 ℃ for 12 hours. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: YMC-act Triart C18 ExRS,30 x 250,5um; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 35B to 65B in 7 minutes; a detector, UV 254/210nm; retention time: 6.32 To give the title compound (AAR-5) (20.6 mg,43 μmol, 25%) as an off-white solid. M/z 479.1 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.29(s,1H),7.97(d,J=7.6Hz,1H),7.87–7.86(m,1H),7.82–7.71(m,3H),7.51–7.50(m,2H),7.58(t,J=2.2Hz,1H),7.27(t,J=1.7Hz,1H),6.61(dd,J=2.3,1.4Hz,1H),5.08(d,J=1.7Hz,2H),3.97(q,J=7.0Hz,2H),3.20(s,3H),1.38(t,J=7.0Hz,3H)。
Example 55: synthesis of 2- (6-chloro-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AAT-1)
Pd (OAc) was added to a stirred solution of intermediate (E-7) (140 mg,1 eq, 450. Mu. Mol) and intermediate (AC-2) (92 mg,1 eq, 450 mmol) in 1, 4-dioxane at room temperature under a nitrogen atmosphere (195 mg,2 eq, 910. Mu. Mol) 2 (10 mg,0.1 eq, 40. Mu. Mol) and Xantphos (53 mg,0.2 eq, 90. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19 x 150mm,5 μm 10nm; mobile phase a: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 30% B to 60% B in 7 minutes, wavelength: 254/210nm; retention time: 6.32. this gave the title compound (AAT-1) (11 mg, 23. Mu. Mol, 5%) as a white solid. M/z 470.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.52(s,1H),8.21(d,J=1.3Hz,1H),8.11(d,J=7.7Hz,1H),8.01(d,J=7.8Hz,1H),7.85–7.67(m,5H),7.18(d,J=1.3Hz,1H),5.23(s,2H),3.59(s,3H)。
Example 56: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AAU-1)
Intermediate (AAT-1) (60 mg,1 equivalent, 120. Mu. Mol) and ethylamine (52 mg,5 equivalent, 640. Mu. Mol) and Cs were added to the mixture under a nitrogen atmosphere at room temperature 2 CO 3 (333 mg,8 eq, 1.02 mmol) in 1, 4-dioxane (2 mL)) Pd (OAc) was added to the stirred mixture of (C) 2 (3 mg,0.1 eq, 10. Mu. Mol) and XantPhos (15 mg,0.2 eq, 20. Mu. Mol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBridge Shield RP18 OBD column, 19×250mm,10 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 25 ml/min; gradient: 45% B to 59% B in 10 minutes; wavelength: 254/210nm; retention time: 8.25. this gave the title compound (AAU-1) (2.1 mg, 4.4. Mu. Mol, 3.4%) as a white solid. M/z 479.1 (M+H) + (ES+) 1 H NMR(400MHz,MeOH-d4)δ8.46(s,1H),8.09(d,J=7.7Hz,1H),7.98(d,J=7.7Hz,1H),7.78–7.71(m,3H),7.69–7.59(m,2H),7.56(d,J=1.3Hz,1H),6.06(d,J=1.3Hz,1H),5.25(s,2H),3.44(s,3H),3.32–3.26(m,2H),1.23(t,J=7.2Hz,3H)。
Example 57: synthesis of 2- (2 '- (4-methyl-1H-imidazol-5-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAV-6)
Step 1: synthesis of 2- (2 '-bromo- [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAV-2)
Pd (dppf) Cl was added to a stirred solution of intermediate (AAS-2) (1.00 g,1 equivalent, 3.11 mmol), 1-bromo-2-iodo-benzene (AAV-1) (1.7 g,2 equivalent, 6.23 mmol) and potassium acetate (610 mg,2 equivalent, 6.23 mmol) in 1, 4-dioxane (10 mL) and water (1 mL) at room temperature under nitrogen atmosphere 2 DCM (254 mg, 311. Mu. Mol). The resulting mixture was stirred under nitrogen at 80 ℃ for 1 hour, then cooled to room temperature, concentrated in vacuo and purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (10% ACN up to 50% within 10 minutes); a detector, UV 254/220nm to give the subtitle compound (AAV-2) as a pale yellow solid (800 mg,1.86mmol, 59%)。m/z 432.2/434.2(M+H) + (ES+)。
Step 2: synthesis of 2- (2 '- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAV-3)
Pd (dppf) Cl was added to a stirred solution of the product (AAV-2) (700 mg,1 eq, 1.62 mmol), intermediate (AAI-3) (777 mg,2 eq, 3.24 mmol) and potassium acetate (317 mg,2 eq, 3.24 mmol) from step 1 above in 1, 4-dioxane (10 mL) under nitrogen 2 DCM (132 mg,0.1 eq, 162. Mu. Mol). The resulting mixture was stirred under nitrogen at 90 ℃ for 48 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/1) to give the sub-title compound (AAV-3) as a pale yellow solid (300 mg,626 μmol, 39%). M/z 380.2 (M+H) + (ES+)。
Step 3: 4-methyl-5- (3 '- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) - [1,1' -biphenyl ] -2-yl) -1H-imidazole-1-carboxylic acid tert-butyl ester (AAV-5)
To a stirred solution of the product from step 2 above (AAV-3) (300 mg,1 eq, 626 μmol), 5-bromo-4-methyl-imidazole-1-carboxylic acid tert-butyl ester (AAV-4) (490 mg,3 eq, 1.88 mmol) and potassium phosphate (265 mg,2 eq, 1.25 mmol) in 1, 4-dioxane (3 mL) and water (0.3 mL) was added Ephos Pd G4 (60 mg,0.1 eq, 0.06 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (10% ACN up to 50% within 10 minutes); the detector, UV 254/220nm, to give the sub-title compound (AAV-5) (40 mg, 75. Mu. Mol, 12%) as an off-white solid. M/z 534.2 (M+H) + (ES+)。
Step 4: synthesis of 2- [3- [2- (4-methyl-1H-imidazol-5-yl) phenyl ] -4- (trifluoromethyl) isoindolin-1-one (AAV-6)
To the product (AA) from step 3 above under a nitrogen atmosphereV-5) (40 mg,1 eq, 75. Mu. Mol) to a stirred solution of EtOAc (2 mL) was added HCl (aqueous solution, 2M) (2 mL). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 1 hour. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBridge-prepared C18 OBD column, 19 x 150mm,5 μm; mobile phase a: water (0.05% TFA), mobile phase B: ACN; flow rate: 20 ml/min; gradient: 50% B to 65% B,65% B in 4.5 minutes; wavelength: 254/210nm; retention time: 4.35. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AAV-6) (1.1 mg,2.5 μmol, 3.4%) as a white solid. M/z 434.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.10(d,J=7.64Hz,1H),7.98(d,J=7.76Hz,1H),7.91–7.86(m,1H),7.78(t,J=7.71Hz,1H),7.59(s,1H),7.58–7.46(m,5H),7.40(t,J=7.95Hz,1H),7.15(d,J=7.80Hz,1H),4.98(s,2H),2.05(s,1H),1.74(s,3H)。
Example 58: synthesis of N- (4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) -6- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) pyridin-2-yl) methanesulfonamide (AAW-2)
Pd was added to a stirred mixture of the product from the intermediate (AAN-7) (112 mg,1 eq, 237. Mu. Mol), methanesulfonamide (AAW-1) (45 mg,2 eq, 470. Mu. Mol) and potassium phosphate (125.4 mg,2.5 eq, 590. Mu. Mol) in 1, 4-dioxane (8 mL) at room temperature under nitrogen atmosphere 2 (dba) 3 (22 mg,0.1 eq, 20. Mu. Mol) and Xantphos (27 mg,0.2 eq, 40. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The resulting mixture was cooled to room temperature and the crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (25% ACN up to 35% in 8 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC on a solid phasePurification was carried out under the following conditions: column: sunFire preparation type C18 OBD column, 30X 250mm 5um; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 10B to 30B in 10 minutes; a detector, UV 254/210nm; retention time: 7.53. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AAW-2) (32.8 mg,62 μmol, 25%) as a white solid. M/z 533.1 (M+H) + (ES+). 1 H NMR(300MHz,MeOH-d4)δ8.52(s,1H),8.18–8.07(m,2H),8.01(d,J=7.7Hz,1H),7.92(s,1H),7.78(t,J=7.7Hz,1H),6.74(d,J=1.2Hz,1H),5.34(s,2H),4.06(s,3H),3.60(s,3H),3.30(s,3H)。
Example 59: synthesis of 2- (2 '- (1-methyl-1H-1, 2, 4-triazol-5-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAX-2)
In a 40-mL vial purged and maintained with an inert nitrogen atmosphere at room temperature under nitrogen atmosphere was placed water (3.5 mL) and 1, 4-dioxane (16.5 mL) containing intermediate (AAX-1) (300 mg,1 equivalent, 1.26 mmol), intermediate (AAS-2) (445 mg,1.1 equivalent, 1.39 mmol) and potassium phosphate (8011 mg,3 equivalent, 3.78 mmol). Pd (DtBPF) Cl was added at room temperature under a nitrogen atmosphere 2 (164 mg,0.2 eq, 252. Mu. Mol) and the resulting solution was stirred at 100℃overnight. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (2×30 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: (column: XBridge-prepared C18 OBD column, 30X 100mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 39% B to 59% B,59% B in 10 minutes; wavelength: 254/220nm; retention time: 9.67). This gave the title compound (AAX-2) as a white solid(45.5mg,104μmol,8%)。m/z 435.0(M+H) + (ES+)。 1 H NMR (400 MHz, chloroform-d) δ8.11 (d, j=6.6 hz, 2H), 7.99 (s, 1H), 7.87 (d, j=7.7 hz, 1H), 7.70-7.62 (m, 4H), 7.56 (d, j=7.9 hz, 1H), 7.45 (t, j=8.0 hz, 1H), 7.40 (s, 1H), 7.14 (d, j=7.7 hz, 1H), 4.80 (s, 2H), 3.21 (s, 3H).
Example 60: synthesis of 2- [3- [3- (4-methyl-1, 2, 4-triazol-3-yl) -pyridin-4-yl ] -phenyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AAY-6)
Step 1: synthesis of 4- (3-nitrophenyl) nicotinic acid (AAY-2)
To 4-bromonicotinic acid (AAY-1) (200 mg,1 equivalent, 990. Mu. Mol), intermediate (AAB-2) (198 mg,1.2 equivalent, 1.18 mmol) and K under a nitrogen atmosphere at room temperature 2 CO 3 (411 mg,3 eq, 2.97 mmol) in 1, 4-dioxane (8 mL) was added Pd (dppf) Cl 2 DCM (145 mg,0.2 eq., 190. Mu. Mol). The resulting mixture was stirred under nitrogen at 85 ℃ for another 4 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and ACN (20% ACN up to 40% in 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (AAY-2) (178 mg, 729. Mu. Mol, 73%) M/z245.1 (M+H) + (ES+), as a white solid.
Step 2: synthesis of N-methyl-2- (4- (3-nitrophenyl) nicotinoyl) hydrazine-1-thiocarboxamide (AAY-3)
To a stirred solution of the product from step 1 above (AAY-2) (60 mg,1 eq, 240. Mu. Mol) and 1-amino-3-methyl thiourea (D-2) (31 mg,1.2 eq, 29. Mu. Mol) in EtOAc (2 mL) at room temperature was added T 3 P (313 mg,4 eq, 980. Mu. Mol) and DIPEA (191 mg,6 eq, 1.47 mmol). The resulting mixture was stirred at room temperature for 3 hours. The crude product was used directly in the next step without further purification. M/z 332.1 (M+H) + (ES+).
Step 3: synthesis of 4-methyl-5- (4- (3-nitrophenyl) pyridin-3-yl) -4H-1,2, 4-triazole-3-thiol (AAY-4)
NaOH (aqueous solution, 1M) (10 mL) was added to the reaction mixture (AAY-3) from step 2 above at 0deg.C. The resulting mixture was stirred at 80 ℃ for 2 hours and then cooled to room temperature. The residue was diluted with water and acidified to pH 4 with HCl (aqueous, 1M). The resulting mixture was extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. This gave the sub-title compound (AAY-4) (55 mg, 176. Mu. Mol, 71%) as a white solid. M/z 314.1 (M+H) + (ES+)。
Step 4: synthesis of 3- (4-methyl-4H-1, 2, 4-triazol-3-yl) -4- (3-nitrophenyl) pyridine (AAY-5)
To a stirred mixture of the product from step 3 above (AAY-4) (50 mg,1 eq, 160. Mu. Mol) and acetic acid (19 mg,2 eq, 320. Mu. Mol) in DCM (3 mL) at 0deg.C was added hydrogen peroxide (27 mg,30Wt%,5 eq, 800. Mu. Mol), and the resulting mixture was stirred at room temperature for 2 hours. The residue was diluted with water and saturated NaHCO 3 (aqueous solution, 1M) alkalization to pH 8. The resulting mixture was extracted with DCM (3×50 mL) and the combined organic layers were washed with brine (30 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. This gave the sub-title compound (AAY-5) (40 mg, 142. Mu. Mol, 89%) as a white solid. M/z 282.1 (M+H) + (ES+)。
Step 5: synthesis of 3- (3- (4-methyl-4H-1, 2, 4-triazol-3-yl) pyridin-4-yl) aniline (AAY-6)
Palladium on carbon (1.5 mg,10wt%,0.1 eq, 14. Mu. Mol) was added to a solution of the product from step 4 above (AAY-5) (40 mg,1 eq, 140. Mu. Mol) in MeOH (8 mL) in a 10mL pressure tank reactor under nitrogen. The mixture was hydrogenated under a hydrogen atmosphere at room temperature using a hydrogen balloon for 2 hours, then filtered through a celite pad and concentrated in vacuo. This gave the sub-title compound (AAY-6) (31 mg, 123. Mu. Mol, 57%) as a white solid. M/z 252.1 (M+H) + (ES+)。
Step 6:2- (3- (3- (4-methyl-4H-1, 2, 4-triazol-3-yl) pyridin-4-yl) phenyl) -4- (trifluoromethyl) isoindolin-1-one (AAY-7)
DIPEA (60 mg,5 eq, 590. Mu. Mol) was added to a stirred mixture of the product (AAY-6) (30 mg,1 eq, 110. Mu. Mol) from step 5 above and intermediate (A-3) (71 mg,2 eq, 230. Mu. Mol) in DMF (6 mL) under nitrogen at room temperature. The resulting mixture was stirred under nitrogen at 80 ℃ for 16 hours. The resulting mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (30% ACN up to 45% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 30 x 100mm,5um; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 21B to 41B in 10 minutes; a detector, UV 254/210nm; retention time: 10. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AAY-7) (1.9 mg,4.3 μmol, 3%) as a white solid. M/z 436.0 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.88(d,J=5.2Hz,1H),8.80(d,J=0.8Hz,1H),8.49(s,1H),8.12(d,J=7.6Hz,1H),8.03–7.94(m,2H),7.88–7.75(m,3H),7.53(t,J=8.0Hz,1H),7.20–7.13(m,1H),5.12(s,2H),3.23(s,3H)。
Example 61: synthesis of 2- (2 '- (4-methylisoxazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAZ-7)
Step 1: (E) Synthesis of 2-bromobenzaldehyde oxime (AAZ-2)
Into a 100-mL round bottom flask was placed E containing 2-bromo-benzaldehyde (AAZ-1) (1.00 g,1 equivalent, 5.40 mmol), hydroxylamine, HCl (751 mg,2 equivalent, 10.8 mmol) and sodium carbonate (2.86 g,5 equivalent, 27.0 mmol)tOH (24.00 mL) and water (6.00 mL). The resulting solution was stirred at room temperature for 2 hours. The resulting solution was diluted with water, extracted with EtOAc (3×50 mL), and the organic layers were combined and concentrated. This gave the sub-title compound (AAZ-2) (1.06 g, crude product) as a white solid. M/z 200.0/202.0 (M+H) + (ES+)。
Step 2: synthesis of (Z) -2-bromo-N-hydroxyphenyliminochloride (AAZ-3)
A50-mL round bottom flask was charged with DMF (20.00 mL) containing the product from step 1 above (AAZ-2) (1.06 g,1 eq, 5.29 mmol) followed by 1-chloropyrrolidine-2, 5-dione (707 mg,1 eq, 5.29 mmol). The resulting solution was stirred at room temperature for 2 hours, then diluted with water, extracted with EtOAc (3×50 mL), and the organic layers were combined and concentrated. This gave the sub-title compound (AAZ-3) (1.25 g, crude product) as a yellow oil. M/z 233.9/235.9 (M+H) + (ES+)。
Step 3: synthesis of 3- (2-bromophenyl) -5-ethoxy-4-methyl-4, 5-dihydroisoxazole (AAZ-5)
Into a 50-mL round bottom flask at room temperature was placed DMF (20 mL) containing the product from step 2 above (AAZ-3) (1.25 g,1 eq, 5.33 mmol), added ether, ethyl propenyl (AAZ-4) (918 mg,2 eq, 10.7 mmol) and NaHCO 3 (896 mg,2 equivalents, 10.7 mmol) and the resulting solution was stirred at room temperature overnight. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was applied to a silica gel column with EtOAc/petroleum ether (1/6). This gave the sub-title compound (AAZ-5) as a yellow oil (800 mg,2.83mmol, 53%). M/z 284.0/286.0 (M+H) + (ES+).
Step 4: synthesis of 3- (2-bromophenyl) -4-methylisoxazole (AAZ-6)
A25-mL round-bottomed flask was charged with TFA (3 mL) containing the product from step 3 above (AAZ-5) (800 mg,1 eq, 2.82 mmol) at room temperature. The resulting solution was stirred at 80 ℃ for 3 hours, then the resulting mixture was cooled to room temperature and concentrated. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). Will be combined withThe organic layer was washed with brine (2×80 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was applied to a silica gel column with EtOAc/petroleum ether (1/4). This gave the sub-title compound (AAZ-6) as a yellow oil (620 mg,2.62mmol, 93%). M/z 238.0/240.0 (M+H) + (ES+)。
Step 5:2- (2 '- (4-methylisoxazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AAZ-7)
A25-mL round bottom flask was charged with the product from step 1 (AAZ-6) (50 mg,1 eq, 210. Mu. Mol), intermediate (AAS-2) (67 mg,1 eq, 210. Mu. Mol) and K under nitrogen at room temperature 2 CO 3 (87 mg,3 eq, 630. Mu. Mol) of 1, 4-dioxane (5 mL) and water (1 mL) followed by Pd (dppf) Cl 2 DCM (15 mg,0.1 eq, 21. Mu. Mol). The resulting solution was stirred overnight at 80 ℃ under nitrogen atmosphere, then cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared C18 OBD column, 30×150mm,5um; mobile phase a: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 57B to 82B in 9 minutes; a detector, UV 220/254nm; retention time: 8.85. this gave the title compound (AAZ-7) (41.3 mg, 95. Mu. Mol, 45%) as a white solid. M/z 435.3 (M+H) + (ES+). 1 H NMR(300MHz,DMSO-d6)δ8.60(d,J=1.2Hz,1H),8.11–8.04(m,2H),8.01–7.93(m,1H),7.85–7.73(m,2H),7.72–7.62(m,2H),7.60–7.47(m,2H),7.42(t,J=8.0Hz,1H),7.09–7.00(m,1H),5.12(s,2H),1.46(d,J=1.1Hz,3H)。
Example 62: synthesis of 2- (2 '- (1, 3, 4-oxadiazol-2-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (ABA-4)
Step 1: synthesis of 2-bromobenzoyl hydrazine (ABA-2)
At room temperature under nitrogen atmosphere, to a 100-mL circle purged and maintained with an inert nitrogen atmosphereInto the bottom flask was placed EtOH (40 mL) containing ethyl 2-bromobenzoate (ABA-1) (1.00 g,1 eq., 4.37 mmol) and hydrazine hydrate (2.20 g,80Wt%,10 eq., 43.7 mmol). The resulting mixture was stirred overnight at 60 ℃ under nitrogen atmosphere and then cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was used directly in the next step without any further purification. This gave the sub-title compound (ABA-2) (800 mg,3.72mmol, 85%) as a yellow solid. M/z 215.0/217.0 (M+H) + (ES+)。
Step 2: synthesis of 2- (2-bromophenyl) -1,3, 4-oxadiazole (ABA-3)
A50 mL round bottom flask purged with an inert nitrogen atmosphere and maintained at room temperature was charged with (diethoxymethoxy) ethane (15 mL) containing the product (ABA-2) from step 1 above (800 mg,1 eq, 3.72 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 3 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And ACN (10% ACN up to 50% in 8 minutes); detector, UV 254/220nm to give the sub-title compound (ABA-3) as a yellow solid (300 mg,1.34mmol, 36%). M/z 225.0/227.0 (M+H) + (ES+)。
Step 3:2- (2 '- (1, 3, 4-oxadiazol-2-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (ABA-4)
Into a 40-mL vial purged and maintained with an inert nitrogen atmosphere were placed 1, 4-dioxane (6 mL) and water (1.5 mL) containing the product (ABA-3) (95 mg,1 equivalent, 424. Mu. Mol) from step 2 above, intermediate (AAS-2) (136 mg,1 equivalent, 424. Mu. Mol), and potassium phosphate (225 mg,2.5 equivalent, 1.06 mmol). Pd (DtBPF) Cl was added at room temperature under a nitrogen atmosphere 2 (55 mg,0.2 eq, 85. Mu. Mol) and subjecting the resulting mixture to nitrogenStirring is carried out for 6 hours at 100℃under an atmosphere. The mixture was cooled to room temperature and the resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XBridge preparative C18 OBD column, 30X 100mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 40% B to 60% B,60% B within 10 minutes; wavelength: 254/220nm; retention time: 10.38 To give the title compound (ABA-4) (64.1 mg,152 μmol, 36%) as a white solid. M/z 422.0 (M+H) + (ES+)。 1 H NMR (300 MHz, chloroform-d) delta 8.28 (s, 1H), 8.14 (d, j=7.6 hz, 1H), 8.05-8.02 (m, 1H), 7.94-87.86 (m, 2H), 7.83-7.82 (m, 1H), 7.71-7.64 (m, 2H), 7.60-7.54 (m, 2H), 7.45 (t, j=7.9 hz, 1H), 7.08 (d, j=7.7 hz, 1H), 5.03 (s, 2H).
Example 63: synthesis of 2- [3- [4- (4-methyl-1, 2, 4-triazol-3-yl) pyridin-3-yl ] phenyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (ABB-5)
Step 1: synthesis of 2- (3-bromoisonicotinyl) -N-methylhydrazine-1-thioformamide (ABB-2)
To a 40-mL vial purged and maintained with an inert nitrogen atmosphere under nitrogen atmosphere at 0deg.C was placed a solution containing 3-bromoisonicotinic acid (ABB-1) (202 mg,1 eq, 1.00 mmol) and 4-methyl-3-thiosemicarbazide (D-2) (116 mg,1.1 eq, 1.10 mmol) in DMF (10 mL) followed by T3P (1.57 g,4 eq, 4.40 mmol) and DIPEA (1.09 mL,98Wt%,6 eq, 6.60 mmol). The resulting solution was stirred at room temperature overnight. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo to give the sub-title compound (ABB-2) (220 mg,761 μmol, 76%) as a yellow solid. M/z 289.2/291.2 (M+H) + (ES+)。
Step 2: synthesis of 5- (3-bromopyridin-4-yl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (ABB-3)
A40-mL vial purged with an inert nitrogen atmosphere and maintained at room temperature was placed in a flask containing the product (ABB-2) from step 1 above (220 mg,1 eq, 761. Mu. Mol) and NaOH (aqueous, 1M) (3.8 mL,5 eq, 3.81 mmol) in DMF (10 mL). The resulting mixture was stirred overnight at 50 ℃ under nitrogen atmosphere and then cooled to. The resulting mixture was diluted with water and extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (2×30 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (ABB-3) as a white solid (190 mg, 704. Mu. Mol, 92%). M/z 271.0/273.0 (M+H) + (ES+)。
Step 3: synthesis of 3-bromo-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) pyridine (ABB-4)
DCM (4 mL) containing the product from step 2 above (ABB-3) (123 mg,1 eq, 452. Mu. Mol) and acetic acid (54 mg,2 eq, 904. Mu. Mol) was placed in a 20-mL vial purged and maintained with an inert nitrogen atmosphere at room temperature. Hydrogen peroxide (92 mg,30wt%,6 eq, 2.71 mmol) was then added at 0 ℃ under nitrogen atmosphere. The resulting solution was stirred at room temperature under nitrogen atmosphere for 2 hours. The mixture was diluted with water and extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (2×30 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (50% ACN up to 60% within 20 minutes); detector, UV 254/220nm to give the sub-title compound (ABB-4) as a white solid (80 mg, 336. Mu. Mol, 52%). M/z 239.1/241.1 (M+H) + (ES+)。
Step 4: synthesis of 2- (3- (4- (4-methyl-4H-1, 2, 4-triazol-3-yl) pyridin-3-yl) phenyl) -4- (trifluoromethyl) isoindolin-1-one (ABB-5).
Put into a 20-mL vial purged and maintained with an inert nitrogen atmosphere at room temperatureComprises the product (ABB-4) (80 mg,1 equivalent, 335. Mu. Mol), intermediate (AAS-2) (118 mg,1.1 equivalent, 369. Mu. Mol) and K from step 3 above 2 CO 3 (139 mg,3 equivalents, 1.00 mmol) of 1, 4-dioxane (5 mL) and water (1 mL). Pd (dppf) Cl was added at room temperature under nitrogen atmosphere 2 DCM (49 mg,0.2 eq, 67. Mu. Mol) and then the mixture was stirred overnight at 80 ℃. The mixture was cooled to room temperature and the mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative HPLC under the following conditions (column: XBridge preparative C18 OBD column, 30X 100mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 23% B to 43% B in 10 minutes; wavelength: 254/220nm; retention time: 9.67 To give the title compound (ABB-5) (23.4 mg, 16%) as a white solid. M/z 436.0 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.94(d,J=0.8Hz,1H),8.82(d,J=5.1Hz,1H),8.47(s,1H),8.11(d,J=7.6Hz,1H),8.03–7.91(m,2H),7.84(t,J=2.0Hz,1H),7.83–7.75(m,1H),7.72–7.71(m,1H),7.52(t,J=8.0Hz,1H),7.16–7.14(m,1H),5.12(s,2H),3.26(s,3H)。
Example 64: synthesis of 6- ((5-azaspiro [2.4] heptan-5-yl) methyl) -2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ABC-7)
Step 1: synthesis of methyl 5-bromo-2- (bromomethyl) -3- (trifluoromethyl) benzoate (ABC-1).
To a stirred solution of intermediate (B-1) (2.70 g,1 eq, 9.09 mmol) in carbon tetrachloride (80 mL) was added NBS (2.43 g,1.5 eq, 13.6 mmol) and BPO (700 mg,0.3 eq, 2.73 mmol) at room temperature under nitrogen. The resulting mixture was subjected to nitrogen atmosphere at 80℃Stirred for 16 hours, then cooled to room temperature and concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (50/1) to give the sub-title compound (ABC-1) (2.7 g,7.18mmol, 79%) as a yellow solid. M/z 377.0/379.0 (M+H) + (ES+)。
Step 2: synthesis of 6-bromo-4- (trifluoromethyl) isoindolin-1-one (ABC-2)
A solution of the product from step 1 above (ABC-1) (2.70 g,1 eq., 7.18 mmol) in ammonia MeOH solution (80 ml, 7M) was stirred at room temperature for 16 hours. The resulting mixture was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (2/1) to give the sub-title compound (ABC-2) (1.2 g,4.32mmol, 60%) as a yellow solid. M/z 279.0/281.0 (M+H) + (ES+)。
Step 3: synthesis of 4- (trifluoromethyl) -6-vinylisoindolin-1-one (ABC-3).
Pd (PPh) was added to a stirred solution of the product (ABC-2) (1.00 g,1 equivalent, 3.57 mmol), tri-tert-butyl (vinyl) stannane (1.36 g,1.2 equivalent, 4.29 mmol) and cesium fluoride (1.08 g,2 equivalent, 7.14 mmol) from step 2 above in dioxane (30 mL) at room temperature under nitrogen atmosphere 3 ) 2 Cl 2 (501 mg, 714. Mu. Mol,0.2 eq). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The resulting mixture was diluted with water. The resulting mixture was extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (2/1) to give the sub-title compound (ABC-3) as a yellow solid (620 mg,2.73mmol, 76%). M/z 228.1 (M+H) + (ES+)。
Step 4: synthesis of 3-oxo-7- (trifluoromethyl) isoindoline-5-carbaldehyde (ABC-4)
To a solution of the product (ABC-3) (620 mg,1 eq, 2.73 mmol), citric acid (682 mg,1.3 eq, 3.55 mmol) and NMO (416 mg,1.3 eq, 3.55 mmol) from step 3 above in water (10 mL) was added potassium osmium dihydrate (VI) (101 mg,0.1 whenThe amount of 273. Mu. Mol) of tBuOH (10 mL) and the mixture was stirred at room temperature for 2 hours. Sodium periodate (1.17 mg,2 equivalents, 5.46 mmol) was added to the above mixture at room temperature, and the resulting mixture was stirred at room temperature for another 3 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (0% ACN up to 40% in 12 minutes); detector, UV 254/220nm. This gave the sub-title compound (ABC-4) (500 mg,2.17mmol, 80%) as a yellow solid. M/z 230.0 (M+H) + (ES+)。
Step 5: synthesis of 6- ((5-azaspiro [2.4] heptan-5-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (ABC-5)
To the product (ABC-4) (200 mg,1 eq., 873. Mu. Mol) from step 4 above and 5-azaspiro [2.4 ] under nitrogen at room temperature]To a stirred solution of heptane, HCl (AAC-1) (117 mg,1 eq., 873. Mu. Mol) in MeOH (5 mL) was added NaBH 3 CN (274 mg,5 eq., 4.37 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 6 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and ACN (10% ACN up to 50% in 15 minutes); detector, UV 254/220nm. This gave the sub-title compound (ABC-5) as a yellow oil (93 mg, 300. Mu. Mol, 34%). M/z 311.1 (M+H) + (ES+)。
Step 6: synthesis of 6-chloro-N-ethyl-4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-amine (ABC-6).
K was added to a stirred mixture of intermediate (AAN-6) (1.00 g,1 eq., 3.24 mmol) and ethylamine, HCl (2.64 g,10 eq., 32.4 mmol) in NMP (25 mL) at room temperature under nitrogen 2 CO 3 (4.47 g,10 equivalents, 32.4 mmol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere and then cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and ACN (10% ACN up to 30% in 20 minutes); detector, UV 254/220nm to give the sub-title compound (ABC-6) as a brown solid (800 mg,2.52mmol, 78%). M/z 318.1/320.1 (M+H) + (ES+)。
Step 7:6- ((5-azaspiro [2.4] heptan-5-yl) methyl) -2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) above isoindolin-1-one (ABC-7)
Into a 25-mL round bottom flask was placed 1, 4-dioxane (3 mL) containing the product from step 6 (ABC-6) (50.00 mg,1 eq, 157. Mu. Mol), the product from step 5 above (ABC-5) (49 mg,1 eq, 157. Mu. Mol) and potassium phosphate (100 mg,3 eq, 471. Mu. Mol) at room temperature, followed by palladium acetate (7 mg,0.2 eq, 31. Mu. Mol) and XantPhos (36 mg,0.4 eq, 63. Mu. Mol). The resulting solution was stirred at 80 ℃ overnight, then cooled to room temperature and concentrated. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared C18OBD column, 30×100mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 40B to 60B in 10 minutes; a detector, UV 220/254nm; retention time: 9.67. this gave the title compound (ABC-7) (10.2 mg, 11%) as a white solid. M/z 592.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),8.08(s,1H),8.02(s,1H),7.87(s,1H),7.65(d,J=1.2Hz,1H),6.20(d,J=1.2Hz,1H),5.26(d,J=1.8Hz,2H),4.04(s,3H),3.91(s,2H),3.50(s,3H),3.42–3.34(m,2H),2.88(t,J=7.0Hz,2H),2.62(s,2H),1.90(t,J=7.0Hz,2H),1.25(t,J=7.2Hz,3H),0.65–0.55(m,4H)。
Example 65: synthesis of 6- ((5-azaspiro [2.4] heptan-5-yl) methyl) -2- (6-chloro-4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ABD-1)
1, 4-dioxane (3 mL) containing intermediate (ABC-5) (20 mg,1 eq, 64. Mu. Mol), intermediate (AAN-6) (20 mg,1 eq, 64. Mu. Mol) and potassium phosphate (41 mg,3 eq, 193. Mu. Mol) was placed in a 25-mL round bottom flask under nitrogen at room temperature, followed by palladium acetate (3 mg,0.2 eq, 13. Mu. Mol) and XantPhos (15 mg,0.4 eq, 26. Mu. Mol). The resulting solution was stirred overnight at 80 ℃ under nitrogen atmosphere. The resulting mixture was cooled to room temperature and concentrated. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared C18OBD column, 30×150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 45B to 75B in 7 minutes; a detector, UV 210/254nm; retention time: 4.85. this gave the title compound (ABD-1) (1.2 mg, 2.1. Mu. Mol, 3.2%) as a white solid. M/z 583.1 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.52(s,1H),8.48(d,J=1.17Hz,1H),8.10(s,1H),8.04(s,1H),7.96(s,1H),7.34(d,J=1.2Hz,1H),5.29–5.24(m,2H),4.05(s,3H),3.87(s,2H),3.68(s,3H),2.83(t,J=7.0Hz,2H),2.57(s,2H),1.89(t,J=7.0Hz,2H),0.61–0.54(m,4H)。
Example 66: synthesis of 6- ((5-azaspiro [2.4] heptan-5-yl) methyl) -2- (6-hydroxy-4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ABE-1)
To a 25-mL round bottom flask was placed 1, 4-dioxane (3 mL) containing intermediate (ABC-5) (60 mg,1 eq, 193. Mu. Mol), intermediate (AAN-6) (72 mg,1.2 eq, 232. Mu. Mol) and potassium phosphate (123 mg,3 eq, 579. Mu. Mol) under nitrogen atmosphere at room temperature, followed by palladium acetate (9 mg,0.2 eq, 39. Mu. Mol) and XantPhos (45 mg,0.4 eq, 77. Mu. Mol). The obtained solution was subjected to nitrogen gas Stirred overnight at 80 ℃ under an atmosphere, then cooled to room temperature and concentrated. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared C18 OBD column, 30×150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 45B to 75B in 7 minutes; a detector, UV 210/254nm; retention time: 4.85. this gave the title compound (ABE-1) (11.0 mg, 9.9%) as a white solid. M/z 565.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.50(s,1H),8.07(s,1H),8.02(s,1H),7.89(s,1H),7.84(s,1H),6.43(s,1H),5.22(s,2H),4.04(s,3H),3.88(s,2H),3.55(s,3H),2.85(t,J=7.0Hz,2H),2.59(s,2H),1.89(t,J=6.9Hz,2H),0.64–0.54(m,4H)。
Example 67: synthesis of 4- ((5-azaspiro [2.4] heptan-5-yl) methyl) -6-cyclopropyl-N- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) pyridine amide (ABF-8)
Step 1: synthesis of methyl 6-cyclopropyl-4-methylpyridine carboxylate (ABF-3)
Pd (dppf) Cl was added to a stirred solution of methyl 6-chloro-4-methylpyridine-2-carboxylate (ABF-1) (2.00 g,1 equivalent, 10.8 mmol), cyclopropylboronic acid (ABF-2) (1.39 g,1.5 equivalent, 16.2 mmol) and potassium phosphate (4.58 g,2 equivalent, 21.6 mmol) in dioxane (40 mL) at room temperature under nitrogen atmosphere 2 DCM (788 mg,0.1 eq, 1.08 mmol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen, then cooled to room temperature, diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (10/1) to give the sub-title compound (ABF-3) as a yellow oil (750 mg,3.92mmol, 36%). M/z 192.1 (M+H) + (ES+)。
Step 2: synthesis of methyl 6-cyclopropyl-4-formylpyridinium formate (ABF-4)
A solution of the product from step 1 above (ABF-3) (931 mg,1 eq., 4.87 mmol), selenium dioxide (810 mg,1.5 eq., 7.30 mmol) and acetic anhydride (1.99 g,4 eq., 19.5 mmol) in acetic acid (18.6 mL) was stirred at 105℃for 8 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/5) to give the sub-title compound (ABF-4) as a pale yellow oil (460 mg,2.24mmol, 86%). M/z 206.1 (M+H) + (ES+)。
Step 3: synthesis of isopropyl 4- ((5-azaspiro [2.4] heptan-5-yl) methyl) -6-cyclopropylpicolinate (ABF-5)
To the product (ABF-4) from step 2 above (600 mg,1 eq, 2.92 mmol) and 5-azaspiro [2.4] under nitrogen at 0 ℃ ]To a stirred mixture of heptane, HCl (AAC-1) (781 mg,2 eq, 5.85. Mu. Mol) in DCM (30 mL) was added NaBH 3 CN (735 mg,4 equivalents, 11.7 mmol) and Ti (OiPr) 4 (3.32 g,4 equivalents, 11.7 mmol) and the resulting mixture was stirred. The resulting mixture was concentrated in vacuo and purified by preparative TLC using EtOAc/petroleum ether (1/1) to give the sub-title compound (ABF-5) as a light brown oil (274 mg,873 μmol, 26%). M/z 315.2 (M+H) + (ES+)。
Step 4: synthesis of 4- ((5-azaspiro [2.4] heptan-5-yl) methyl) -6-cyclopropylpicolinic acid (ABF-6)
To a stirred solution of the product from step 3 above (ABF-5) (300 mg,1 eq, 1.05 mmol) in a mixture of THF (10 mL) and water (2 mL) was added LiOH (125 mg,5 eq, 5.24 mmol) at room temperature. The resulting mixture was stirred at 70 ℃ for 2 hours, then the resulting mixture was cooled to room temperature and concentrated in vacuo. The mixture was acidified with HCl (aqueous, 1M) to pH 6. The resulting mixture was extracted with (chloroform: isopropanol=3:1) (5×30 mL). The combined organic layers were washed with brine (2×30 mL), dried (Na 2 SO 4 ) And concentrated in vacuo. This gave the sub-title compound (ABF-6) (170 mg, 625. Mu. Mol, 60%) as an off-white solid. M/z 273.2 (M+H) + (ES+)。
Step 5: synthesis of 4- ((5-azaspiro [2.4] heptan-5-yl) methyl) -6-cyclopropylpyridinamide (ABF-7)
To a stirred solution of the product (ABF-6) (50 mg,1 eq, 184. Mu. Mol) from step 4 above and DIPEA (71 mg,3 eq, 552. Mu. Mol) in DMF (1.5 mL) at room temperature was added HATU (105 mg,1.5 eq, 276. Mu. Mol) and NH 4 Cl (98 mg,10 eq, 1.84 mmol). The resulting mixture was stirred at room temperature for 2 hours, then concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (ABF-7) as a pale yellow oil (39 mg, 144. Mu. Mol, 78%). M/z 272.2 (M+H) + (ES+)。
Step 6: synthesis of 4- ((5-azaspiro [2.4] heptan-5-yl) methyl) -6-cyclopropyl-N- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) pyridine amide (ABF-8)
Pd (OAc) was added to a solution of intermediate (ABC-6) (40 mg,1 eq, 126. Mu. Mol), the product from step 5 above (ABF-7) (34 mg,1 eq, 126. Mu. Mol) and potassium phosphate (53 mg, 252. Mu. Mol,2 eq) in 1, 4-dioxane (1.5 mL) at room temperature under a nitrogen atmosphere 2 (3 mg,0.1 eq, 13. Mu. Mol) and Xantphos (15 mg,0.2 eq, 25. Mu. Mol). After stirring overnight at 100 ℃ under nitrogen, the resulting mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XBridge prep. C18 OBD column, 30 x 100mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 40% B to 60% B in 9 minutes; wavelength: 254/220nm; retention time: 8.85 To give the title compound (ABF-8) (9.1 mg,16 μmol, 13%) as a white solid. M/z 553.5 (M+H) + (ES+). 1 H NMR(300MHz,DMSO-d6)δ10.01(s,1H),8.47(s,1H),7.87(d,J=2.9Hz,2H),7.51(d,J=1.4Hz,1H),7.31(d,J=1.2Hz,1H),6.86(t,J=5.4Hz,1H),6.24(d,J=1.2Hz,1H),3.89(s,3H),3.68(s,2H),3.44(s,3H),3.22(dd,J=7.3,5.5Hz,2H),2.69(t,J=6.8Hz,2H),2.45(s,2H),2.29–2.24(m,1H),1.77(t,J=6.8Hz,2H),1.19–0.95(m,7H),0.55–0.47(m,4H)。
Example 68: synthesis of 2- (6- (aminomethyl) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ABG-3)
Step 1: synthesis of tert-butyl ((4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) -6- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) pyridin-2-yl) methyl) carbamate (ABG-2)
To intermediate (AAN-7) (150 mg,1 eq, 317. Mu. Mol), N- [ (trifluoro-. Lamda.4-borane) methyl, under nitrogen at room temperature]To a stirred mixture of tert-butyl carbamate (ABG-1) (125 mg,2 eq, 633. Mu. Mol), potassium (25 mg,2 eq, 633. Mu. Mol) and potassium phosphate (134 mg,2 eq, 633. Mu. Mol) in dioxane (10 mL) and water (2 mL) were added SPhos Pd gen.3 (49 mg,0.2 eq, 63. Mu. Mol) and SPhos (52 mg,0.4 eq, 127. Mu. Mol). The mixture was stirred under nitrogen at 90 ℃ for 15 hours, then cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (10% ACN up to 60% within 20 minutes); detector, UV 254/220nm, to give the sub-title compound (ABG-2) as a white solid (155 mg, 273. Mu. Mol, 86%). M/z 569.1 (M+H) + (ES+)。
Step 2: synthesis of 2- (6- (aminomethyl) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ABG-3)
To the product (ABG-2) from step 1 above (30 mg,1 whenTo a stirred mixture of 50. Mu. Mol in DCM (2 mL) was added TFA (0.4 mL). The mixture was stirred at room temperature for 2 hours, then concentrated and purified by preparative HPLC under the following conditions: column: XBridge-prepared C18 OBD column, 30 x 100mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 25B to 50B in 9 minutes; a detector, UV 254/220nm; retention time: 8.85 to give the title compound (ABG-3) (10.4 mg, 22. Mu. Mol, 40%) as a white solid. M/z 469.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.30(d,J=1.3Hz,1H),8.10(dd,J=15.6,7.7Hz,2H),7.97(s,1H),7.81(t,J=7.7Hz,1H),7.38(d,J=1.3Hz,1H),5.30(s,2H),3.92–3.90(m,5H),3.54(s,3H)。
Example 69: synthesis of 2- (6-ethoxy-4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ABH-2)
Step 1: synthesis of 2-chloro-6-ethoxy-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) -pyrazol-3-yl ] -pyridine (ABH-1)
Intermediate (AAN-6) (100 mg,1 eq, 320. Mu. Mol) and NaOEt (44 mg,2 eq, 640. Mu. Mol) were added to a stirred solution of EtOH (8 mL) at room temperature. The resulting mixture was stirred at 60℃for 16 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. This gave the sub-title compound (ABH-1) (80 mg, 251. Mu. Mol, 77%) as a white solid. M/z 319.1/321.1 (M+H) + (ES+)。
Step 2: synthesis of 2- (6-ethoxy-4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ABH-2)
To the product (A) from step 1 above under a nitrogen atmosphere at room temperatureBH-1) (80 mg,1 eq, 251. Mu. Mol), intermediate (AC-2) (60 mg,1.2 eq, 300. Mu. Mol) and potassium phosphate (106.5 mg,0.50mmol,2.0 eq) in dioxane (8 mL) was added Xantphos (29 mg,0.2 eq, 50. Mu. Mol) and Pd (OAc) 2 (6 mg,0.1 eq, 20. Mu. Mol). The resulting mixture was stirred under nitrogen at 100 ℃ for 16 hours. The resulting mixture was cooled to room temperature and the crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (30% ACN up to 40% in 8 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19X 150mm 5um; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 31B to 51B within 10 minutes; a detector, UV 254/210nm; retention time: 9.67. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ABH-2) (54.7 mg,113 μmol, 44%) as a white solid. M/z484.1 (M+H) + (ES+). 1 H NMR(300MHz,MeOH-d4)δ8.50(s,1H),8.10–7.96(m,3H),7.91(s,1H),7.78(t,J=7.7Hz,1H),6.62(d,J=1.2Hz,1H),5.31(s,2H),4.53–4.39(m,2H),4.04(s,3H),3.58(s,3H),1.45(t,J=7.1Hz,3H)。
Example 70: synthesis of 2- (6- (isopropylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ABI-1)
To intermediate (AAN-7) (95 mg,1 equivalent, 200. Mu. Mol) and isopropylamine (59 mg,5 equivalent, 1.00 mmol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 (523 mg,8 eq, 1.60 mmol) to a stirred mixture of dioxane (8 mL) was added Pd (OAc) 2 (5 mg,0.1 eq, 20. Mu. Mol) and XantPhos (23 mg,0.2 eq, 40. Mu. Mol). The mixture was subjected to nitrogenThe mixture was stirred at 100℃for 16 hours under an atmosphere, and then cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (40% ACN up to 70% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 30X 150mm 5um; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 33B to 58B in 9 minutes; a detector, UV 254/210nm; retention time: 8.85. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ABI-1) (10.6 mg,21 μmol, 10%) as a white solid. M/z 497.1 (M+H) + (ES+) 1 H NMR(300MHz,MeOH-d4)δ8.49(s,1H),8.09(d,J=7.7Hz,1H),7.99(d,J=7.7Hz,1H),7.86(s,1H),7.77(t,J=7.7Hz,1H),7.64(d,J=1.2Hz,1H),6.20(d,J=1.2Hz,1H),5.27(s,2H),4.12–3.97(m,4H),3.51(s,3H),1.25(d,J=6.4Hz,6H)。
Example 71: synthesis of 2-cyclopropyl-N- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6-methylpyrimidine-4-carboxamide (ABJ-4)
Step 1: synthesis of 2-cyclopropyl-6-methylpyrimidine-4-carboxylic acid (ABJ-2)
Toluene (15 mL) containing methyl 2-chloro-6-methylpyrimidine-4-carboxylate (ABJ-1) (1.00 g,1 eq, 5.36 mmol), cyclopropylboronic acid (ABF-2) (921 mg,2 eq, 10.7 mmol) and potassium phosphate (4.55 g,4 eq, 21.4 mmol) was added to a 40mL sealed tube at room temperature under nitrogen atmosphere. Pd (OAc) was added to the above mixture over 3 minutes at room temperature under nitrogen atmosphere 2 (60 mg,0.05 eq, 268. Mu. Mol) and PCy3 (150 mg,0.1 eq, 536. Mu. Mol). The resulting mixture was stirred overnight at 110 ℃ under nitrogen, then cooled to room temperature, diluted with water and EtOAc (3X 80 mL) extraction. The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (36% ACN up to 48% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ABJ-2) as a yellow oil (660 mg,3.71mmol, 69%). M/z 179.1 (M+H) + (ES+)。
Step 2: synthesis of 2-cyclopropyl-6-methylpyrimidine-4-carboxamide (ABJ-3)
A40 mL sealed tube was charged with the product (ABJ-2) (284 mg,1 equivalent, 3.00 mmol) from step 1 above, NH, at room temperature 4 Cl (803 mg,5 eq, 15.0 mmol) and DIPEA (3.87 g,10 eq, 30.0 mmol) in THF (10 mL). HATU (2.28 g,2 equivalents, 5.99 mmol) was added to the above mixture at 0 ℃ over 3 minutes, and the resulting mixture was stirred at room temperature for 3 hours. The resulting mixture was concentrated in vacuo to give the sub-title compound (ABJ-3) (310 mg,1.75mmol, 58%) as a white solid. M/z 178.1 (M+H) + (ES+)。
Step 3: synthesis of 2-cyclopropyl-N- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6-methylpyrimidine-4-carboxamide (ABJ-4)
1, 4-dioxane (4 mL) containing intermediate (ABC-6) (55 mg,1 eq, 173. Mu. Mol), potassium phosphate (110 mg,3 eq, 519. Mu. Mol) and product (ABJ-3) (37 mg,1.2 eq, 208. Mu. Mol) from step 2 above was added to an 8mL sealed tube under nitrogen at room temperature. Pd was added to the above mixture over 3 minutes at room temperature under nitrogen atmosphere 2 (dba) 3 (32 mg,0.2 eq, 35. Mu. Mol) and XantPhos (40 mg,0.4 eq, 69. Mu. Mol) and the resulting mixture was stirred at 100℃for 3 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC under the following conditions (column: XBridge preparative C18 OBD column, 30X 100mm,5 μm)The method comprises the steps of carrying out a first treatment on the surface of the Mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 25% B to 50% B in 9 minutes; wavelength: 254/220nm; retention time: 7.53 To give the title compound (ABJ-4) (31.8 mg,69 μmol, 40%) as a white solid. M/z 459.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ9.97(s,1H),8.47(s,1H),7.88(s,1H),7.75(s,1H),7.29(d,J=1.3Hz,1H),6.90(t,J=5.4Hz,1H),6.28(d,J=1.3Hz,1H),3.89(s,3H),3.44(s,3H),3.30–3.19(m,2H),2.53(s,3H),2.37–2.26(m,1H),1.18–1.05(m,7H)。
Example 72: synthesis of 2-cyclopropyl-N- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6- (2- (pyrrolidin-1-yl) ethoxy) pyrimidine-4-carboxamide (ABK-7)
Step 1: synthesis of methyl 2-cyclopropyl-6-oxo-1H-pyrimidine-4-carboxylate (ABK-1)
Sulfuric acid (5 mL) was added dropwise to a stirred mixture of intermediate (AAE-3) (5.00 g,1 eq, 27.8 mmol) in MeOH (150 mL) at 0 ℃. The mixture was stirred at 60 ℃ for 2 hours and then cooled to room temperature. The resulting mixture was concentrated in vacuo and the crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (0% ACN up to 30% within 10 minutes); detector, UV 254/220nm. The reaction was concentrated in vacuo to give the sub-title compound (ABK-1) (3.78 g,19.5mmol, 70%) as a brown solid. M/z 195.1 (M+H) + (ES+)。
Step 2: synthesis of methyl 6- (2-chloroethoxy) -2-cyclopropylpyrimidine-4-carboxylate (ABK-3)
To the product (ABK-1) (140 mg,1 equivalent, 721. Mu. Mol) from step 1 above and K under a nitrogen atmosphere at room temperature 2 CO 3 (498 mg,5 eq, 3.61 mmol) to a stirred mixture of NMP (8 mL) was added 1-bromo-2-chloroethane (ABK-2) 517mg,5 eq, 3.61 mmol). The mixture was stirred at 60 ℃ for 1 hour and then cooled to room temperature. The mixture was concentrated and purified by preparative TLC using petroleum ether/EtOAc (10/1) to give the sub-title compound (ABK-3) as a pale yellow oil (115 mg,448 μmol, 62%). M/z 257.1/259.1 (M+H) + (ES+)。
Step 3: synthesis of methyl 2-cyclopropyl-6- (2- (pyrrolidin-1-yl) ethoxy) pyrimidine-4-carboxylate (ABK-5)
To a stirred mixture of the product (ABK-3) (115 mg,1 eq, 448 mmol) from step 2 above and DIPEA (173 mg,3 eq, 1.34 mmol) in NMP (5 mL) was added pyrrolidine (ABK-4) (127 mg,4 eq, 1.79 mmol) at room temperature under nitrogen. The mixture was stirred at 80 ℃ for 3 hours, then the mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (0% ACN up to 45% in 25 minutes); detector, UV 254/220nm. The reaction was concentrated in vacuo to give the sub-title compound (ABK-5) (37 mg,127 μmol, 28%) as a white solid. M/z 292.2 (M+H) + (ES+)。
Step 4: synthesis of 2-cyclopropyl-6- (2- (pyrrolidin-1-yl) ethoxy) pyrimidine-4-carboxylic acid (ABK-6)
To a stirred mixture of product (ABK-5) (37 mg,1 eq, 127. Mu. Mol) from step 3 above and LiOH (15 mg,5 eq, 636. Mu. Mol) in MeOH (2 mL), THF (2 mL) and water (1 mL) under a nitrogen atmosphere at room temperature. The mixture was stirred at room temperature for 2 hours, and then concentrated to give the sub-title compound (ABK-6) (30 mg, 108. Mu. Mol, 86%) as a white solid. M/z 278.1 (M+H) + (ES+)。
Step 5: synthesis of 2-cyclopropyl-N- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6- (2- (pyrrolidin-1-yl) ethoxy) pyrimidine-4-carboxamide (ABK-7)
Under nitrogen atmosphereEDCI (41 mg,2 eq, 216. Mu. Mol) was added to a stirred mixture of the product (ABK-6) (30 mg,1 eq, 108. Mu. Mol) from step 4 above and intermediate (P-1) (27 mg,1 eq, 108. Mu. Mol) in pyridine (2 mL) at room temperature. The mixture was stirred under nitrogen at 80 ℃ for 2 hours. The mixture was cooled to room temperature. The mixture was concentrated and purified by preparative TLC using DCM/MeOH (15/1). And the mixture was then purified by preparative HPLC under the following conditions: (column: sunfire preparation type C18 column, 30. Times.150 mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 10% B to 35% B in 7 min; wavelength: 254/220nm; retention time: 6.45) to give the title compound (ABK-7) (4.7 mg, 9.2. Mu. Mol, 8.5%) as a white solid. M/z 510.4 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.38(s,1H),7.81–7.67(m,4H),7.66–7.57(m,2H),7.36(t,J=7.9Hz,1H),7.33(s,1H),7.02–6.70(m,1H),4.67–4.70(m,2H),3.36(s,2H),3.19(s,3H),3.13(s,4H),2.38–2.32(m,1H),2.05–1.96(m,4H),1.26–1.23(m,1H),1.18–1.13(m,2H)。
Example 73: synthesis of 2-cyclopropyl-N- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6-oxo-1- (2- (pyrrolidin-1-yl) ethyl) -1, 6-dihydropyrimidine-4-carboxamide (ABL-4)
Step 1: synthesis of methyl 2-cyclopropyl-6-oxo-1- (2- (pyrrolidin-1-yl) ethyl) -1, 6-dihydropyrimidine-4-carboxylate (ABL-1)
To a stirred mixture of intermediate (ABK-3) (800 mg,1 eq, 3.12 mmol) and pyrrolidine (ABK-4) (887 mg,4 eq, 12.5 mmol) in NMP (20 mL) was added DIPEA (1.21 mg,3 eq, 9.35 mmol) at room temperature under nitrogen atmosphere, and the mixture was stirred at 80℃for 3 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (10% ACN up to 50% within 10 minutes); detector, UV 254/220nm to give the sub-title compound (ABL-1) as a brown/yellow oil (370 mg,1.27mmol, 41%). M/z 292.2 (M+H) + (ES+)。
Step 2: synthesis of 2-cyclopropyl-6-oxo-1- (2- (pyrrolidin-1-yl) ethyl) -1, 6-dihydropyrimidine-4-carboxylic acid (ABL-2)
A solution of the product from step 1 above (ABL-1) (120 mg,1 eq., 410. Mu. Mol) and LiOH (49 mg,5 eq., 2.06 mmol) in MeOH (3 mL), THF (3 mL) and water (1.5 mL) was stirred at room temperature for 2 hours. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and ACN (0% ACN up to 10% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ABL-2) (40 mg, 144. Mu. Mol, 35%) as a white solid. M/z 278.1 (M+H) + (ES+)。
Step 3: 2-cyclopropyl-6-oxo-1- (2- (pyrrolidin-1-yl) ethyl) -1, 6-dihydropyrimidine-4-carboxamide (ABL-3)
To the product (ABL-2) (40 mg,1 equivalent, 144. Mu. Mol) from step 2 above and NH at room temperature 4 To a stirred solution of Cl (39 mg,5 eq, 720. Mu. Mol) in DMF (10 mL) was added HATU (110 mg,2 eq, 288 mmol) and DIPEA (186 mg,10 eq, 1.44 mmol). The resulting mixture was stirred at room temperature for 2 hours. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (30% ACN up to 50% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ABL-3) (35 mg, 126. Mu. Mol, 87%) as a white solid. M/z 277.2 (M+H) + (ES+)。
Step 4: 2-cyclopropyl-N- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6-oxo-1- (2- (pyrrolidin-1-yl) ethyl) -1, 6-dihydropyrimidine-4-carboxamide (ABK-4)
Pd was added to a stirred mixture of the product (ABL-3) (40 mg,1 eq, 144 mmol) and intermediate (ABC-6) (69 mg,1.5 eq, 216. Mu. Mol) from step 3 above and potassium phosphate (61 mg,2 eq, 288. Mu. Mol) in 1, 4-dioxane (8 mL) at room temperature under nitrogen atmosphere 2 (dba) 3 (26 mg,0.2 eq, 28. Mu. Mol) and Xantphos (34 mg,0.4 eq, 57. Mu. Mol). The resulting mixture was stirred under nitrogen at 120 ℃ for 3 hours and then cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (20% ACN up to 30% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 30X 100mm 5um; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 16B to 36B within 10 minutes; a detector, UV 254/210nm; retention time: 10.58. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ABL-4) (8.1 mg,14 μmol, 9%) as a yellow solid. M/z 558.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),7.86(s,1H),7.39(d,J=1.2Hz,1H),6.99(s,1H),6.22(d,J=1.2Hz,1H),4.55–4.41(m,2H),4.01(s,3H),3.49(s,3H),3.32–3.28(m,2H),2.94–2.82(m,2H),2.75–2.68(m,4H),2.40–2.29(m,1H),1.90–1.82(m,4H),1.43–1.33(m,2H),1.36–1.25(m,2H),1.22(t,J=7.2Hz,3H)。
Example 74: synthesis of 2-cyclopropyl-N- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6-oxo-1- (2- (pyrrolidin-1-yl) ethyl) -1, 6-dihydropyrimidine-4-carboxamide (ABM-1)
To intermediate (ABL-2) (40 mg,1 eq, 144 μm) under nitrogen at room temperatureTo a stirred mixture of ol) and intermediate (P-1) (36 mg,1 eq, 144 mmol) in pyrazine (6 mL) was added EDCI (55 mg,2 eq, 288. Mu. Mol). The resulting mixture was stirred under nitrogen at 80 ℃ for 16 hours and then cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (50% ACN up to 65% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 30X 100mm 5um; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 20B to 45B within 10 minutes; a detector, UV 254/210nm; retention time: 7.53. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ABM-1) (9.2 mg,18 μmol, 12%) as a white solid. M/z 510.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.39(s,1H),7.79–7.57(m,6H),7.35(t,J=7.9Hz,1H),7.03–6.96(m,2H),4.56–4.47(m,2H),3.19(s,3H),2.94–2.86(m,2H),2.76–2.68(m,4H),2.38–2.28(m,1H),1.92–1.81(m,4H),1.48–1.39(m,2H),1.32–1.23(m,2H)。
Example 75: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((3-fluoroazetidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (ABN-2)
Step 1: synthesis of 6- ((3-fluoroazetidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (ABN-1)
To a stirred solution of intermediate (ABC-4) (100 mg,1 eq., 436. Mu. Mol) and 3-fluoroazetidine, HCl (U-1) (131 mg,4 eq., 1.74 mmol) in MeOH (5 mL) at room temperature was added NaBH 3 CN (137 mg,5 eq., 2.18 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphereAnd (5) at night. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (35% ACN up to 65% in 8 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ABN-1) (70 mg, 243. Mu. Mol, 56%) as a white solid. M/z 289.1 (M+H) + (ES+)。
Step 2: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((3-fluoroazetidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (ABN-2)
To a stirred solution of the product (ABN-1) (30 mg,1 eq, 104. Mu. Mol), intermediate (ABC-6) (33 mg,1 eq, 104. Mu. Mol) and potassium phosphate (44 mg,2 eq, 208. Mu. Mol) from step 1 above in 1, 4-dioxane (2 mL) under nitrogen at room temperature was added Xanthos (12 mg,0.2 eq, 21. Mu. Mol) and Pd (OAc) 2 (2.3 mg,0.1 eq, 10. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The resulting mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using petroleum ether/EtOAc (2/1). The crude product was purified by preparative HPLC under the following conditions: column: sunfire preparation type C18 column, 30×150mm 5um; mobile phase a: water (0.1% fa), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 8% B to 32% B in 10 minutes; a detector, UV 254/210nm; retention time: 9.67. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ABN-2) (16.5 mg,29 μmol, 28%) as a white solid. M/z 570.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),8.04(s,1H),7.95(s,1H),7.87(s,1H),7.65(d,J=1.2Hz,1H),6.20(d,J=1.2Hz,1H),5.31–5.19(m,3H),4.04(s,3H),3.96(s,2H),3.82–3.71(m,2H),3.50(s,3H),3.41–3.34(m,4H),1.25(t,J=7.2Hz,3H)。
Example 76: synthesis of 2- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) - [1, 1-biphenyl ] -3-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (ABO-5)
Step 1: synthesis of 2- (2-bromo-5-fluorobenzoyl) -N-methylhydrazine-1-thiocarboxamide (ABO-2)
To a stirred mixture of 2-bromo-5-fluorobenzoic acid (ABO-1) (2.19 g,1 eq, 10.0 mmol) and 4-methyl-3-thiosemicarbazide (D-2) (1.05 g,1 eq, 10.0 mmol) in DMF (40 mL) was added DIPEA (7.75 g,6 eq, 60.0 mmol) and T at room temperature 3 P (12.7 g,4 equivalents, 40.0 mmol). The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo. The crude mixture was used directly in the next step without further purification. M/z 306.0/308.0 (M+H) + (ES+)。
Step 2: synthesis of 5- (2-bromo-5-fluorophenyl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (ABO-3)
NaOH (aqueous solution, 1M) was added to the stirred mixture of the product (ABO-2) from step 1 above at room temperature, and the resulting mixture was stirred at 100℃overnight. The residue was neutralized to pH 5 with HCl (aqueous, 1M). The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (35% ACN up to 50% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ABO-3) (1 g,3.48mmol, 45%) as a white solid. M/z 288.0/290.0 (M+H) + (ES+)。
Step 3: synthesis of 3- (2-bromo-5-fluorophenyl) -4-methyl-4H-1, 2, 4-triazole (ABO-4)
To the product (ABO-3) (230 mg,1 eq., 798. Mu. Mol) from step 2 above was stirred and mixed in DCM (4 mL) at 0deg.CAcetic acid (96 mg,2 equivalents, 1.60 mmol) and hydrogen peroxide (136 mg,5 equivalents, 4.00 mmol) were added to the compound. The resulting mixture was stirred at room temperature for 2 hours, then saturated NaHCO 3 The solution (aqueous solution) was alkalized to pH 9. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (36% ACN up to 48% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ABO-4) as a white solid (145 mg, 568. Mu. Mol, 71%). M/z 256.0/258.0 (M+H) + (ES+)。
Step 4: synthesis of 2- (4 ' -fluoro-2 ' - (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (ABO-5)
Pd (DtBPF) Cl was added to a stirred mixture of the product (ABO-4) (232 mg,1 eq, 906. Mu. Mol), intermediate (AAS-2) (29 mg,1 eq, 906. Mu. Mol) and potassium phosphate (577 mg,3 eq, 2.72 mmol) from step 3 above in dioxane (5 ml) and water (1 ml) at room temperature under nitrogen atmosphere 2 (59 mg,0.1 eq, 91. Mu. Mol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere, then cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And ACN (10% ACN up to 50% within 10 minutes); detector, UV 254/220nm. The crude product was purified by preparative HPLC under the following conditions (column: XBridge prep. C18 OBD column, 30 x 100mm,5um; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 35% B to 55% B in 10 minutes; wavelength: 254/220nm; retention time: 7.13 To give the title compound (ABO-5) (10.8 mg,24 μmol, 2.6%) as a white solid. M/z 453.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.43(s,1H),8.12–8.02(m,2H),8.02–7.96(m,1H),7.80(t,J=7.7Hz,1H),7.77–7.69(m,1H),7.67(t,J=2.0Hz,1H),7.64–7.54(m,1H),7.54–7.46(m,1H),7.38(t,J=8.0Hz,1H),6.92–6.85(m,1H),5.12(d,J=1.8Hz,2H),3.14(s,3H)。
Example 77: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((3-fluoropyrrolidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (ABP-3)
Step 1: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -3-oxo-7- (trifluoromethyl) isoindoline-5-carbaldehyde (ABP-1)
To a stirred solution of intermediate (ABC-4) (150 mg,1 eq, 650. Mu. Mol), intermediate (ABC-6) (146 mg,0.7 eq, 450. Mu. Mol) and potassium phosphate (278 mg,2 eq, 1.31 mmol) in dioxane (10 mL) under nitrogen at room temperature was added XantPhos (76 mg,0.2 eq, 130. Mu. Mol) and Pd (OAc) 2 (15 mg,0.1 eq, 60. Mu. Mol). The resulting mixture was stirred under nitrogen at 100 ℃ for 2 hours, then cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (60% ACN up to 70% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ABP-1) (180 mg, 353. Mu. Mol, 53%) as a yellow solid. M/z 511.2 (M+H) + (ES+)。
Step 2: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((3-fluoropyrrolidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (ABP-3)
To the product (ABP-1) (20 mg,1 eq, 30. Mu. Mol) from step 1 above and 3-fluoropyrrolidine, HCl (ABP-2) (20 mg,4 eq, 150 mmol) in DCM (2 m) under nitrogen at room temperatureAdding NaBH (OAc) to the stirred mixture in L) 3 (17 mg,2 eq, 70. Mu. Mol) and Et 3 N (12 mg,3 eq, 110. Mu. Mol). The resulting mixture was stirred at room temperature under nitrogen for 1 hour, and the resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and ACN (20% ACN up to 30% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 30X 150mm 5um; mobile phase a: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 40B to 60B in 10 minutes; a detector, UV 254/210nm; retention time: 6.97. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ABP-3) (2.3 mg,4 μmol, 10%) as a white solid. M/z 584.1 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),8.07(s,1H),8.00(s,1H),7.87(s,1H),7.66(d,J=1.2Hz,1H),6.20(d,J=1.2Hz,1H),5.26–5.14(m,3H),4.04(s,3H),3.95–3.82(m,2H),3.50(s,3H),3.40–3.36(m,2H),2.99–2.85(m,2H),2.85–2.68(m,1H),2.57–2.46(m,1H),2.35–2.15(m,1H),2.14–1.95(m,1H),1.25(t,J=7.2Hz,3H)。
Example 78: synthesis of 6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) picolinic acid (ABQ-1)
DMF (2 mL) containing intermediate (ABC-6) (30 mg,1 eq, 94. Mu. Mol), oxalic acid (13 mg,1.5 eq, 141. Mu. Mol), DIPEA (18.3 mg,1.5 eq, 140. Mu. Mol) and acetic anhydride (14 mg,1.5 eq, 141. Mu. Mol) was added to an 8mL sealed tube under nitrogen at room temperature. Pd (OAc) was added to the above mixture at room temperature under nitrogen atmosphere 2 (2 mg,0.1 eq, 10. Mu. Mol) and XantPhos (11 mg,0.2 eq, 20. Mu. Mol). The resulting mixture was stirred overnight at 100℃under a nitrogen atmosphere, howeverThe mixture was then cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 2% B to 20% B over 9 min; wavelength: 254/220nm; retention time: 6.32) to give the title compound (ABQ-1) (5.2 mg, 16. Mu. Mol, 17%) as a white solid. M/z 328.1 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.50(s,1H),7.92(s,1H),7.26(d,J=1.4Hz,1H),6.78(s,1H),4.00(s,3H),3.55(s,3H),3.34(s,1H),3.32(s,1H),1.23(t,J=7.2Hz,3H)。
Example 79: synthesis of 6- ((5-azaspiro [2.4] heptan-5-yl) methyl) -2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ABR-1)
To a stirred mixture of intermediate (AD-1) (30 mg,1 equivalent, 90. Mu. Mol), intermediate (ABC-5) (30 mg, 90. Mu. Mol,1 equivalent) and potassium phosphate (41 mg,2 equivalent, 190. Mu. Mol) in 1, 4-dioxane (1.5 mL) was added XantPhos (11 mg,0.2 equivalent, 20. Mu. Mol) and Pd (OAc) at room temperature under nitrogen atmosphere 2 (2 mg,0.1 eq, 10 mmol). The resulting mixture was stirred under nitrogen at 100 ℃ for 5 hours, then cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBridge-prepared C18 OBD column, 30 x 100mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 48% B to 72% B in 10 minutes; wavelength: 254/220nm; retention time: 9.67. this gave the title compound (ABR-1) (2.7 mg, 4.6. Mu. Mol, 3.6%) as a white solid. M/z 588.4 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.45(s,1H),8.07(s,1H),8.00(s,1H),7.80–7.69(m,2H),7.69–7.58(m,2H),7.56(d,J=1.3Hz,1H),6.05(d,J=1.2Hz,1H),5.22(s,2H),3.86(s,2H),3.44(s,3H),3.32–3.26(m,2H),2.83(t,J=6.9Hz,2H),2.57(s,2H),1.89(t,J=7.0Hz,2H),1.22(t,J=7.2Hz,3H),0.59(d,J=4.6Hz,4H)。
Example 80: synthesis of 2- (6-cyclopropyl-5- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ABS-1)
PhMe (8 mL) containing intermediate (AAT-1) (100 mg,1 eq, 213. Mu. Mol), intermediate (ABF-2) (73 mg,4 eq, 851. Mu. Mol) and potassium phosphate (136 mg,3 eq, 639. Mu. Mol) was placed in a 40-mL vial purged and maintained with an inert nitrogen atmosphere at room temperature, and tricyclohexylphosphine (6 mg,0.1 eq, 21. Mu. Mol) and Pd (OAc) were added 2 (5 mg,0.1 eq, 21. Mu. Mol) and the resulting mixture was stirred overnight at 100℃under a nitrogen atmosphere. The mixture was cooled to room temperature and the resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by preparative HPLC (column: sunfire preparative C18 column, 30X 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 45% B to 75% B in 7 min; wavelength: 254/220nm; retention time: 6.32) to give the title compound (ABS-1) (37.8 mg, 79. Mu. Mol, 36%) as a white solid. M/z 476.0 (M+H) + (ES+). 1 H NMR(300MHz,MeOH-d4)δ8.47(s,1H),8.12–8.02(m,2H),7.97(d,J=7.7Hz,1H),7.85–7.60(m,5H),6.91(d,J=1.4Hz,1H),5.23–5.16(m,2H),3.46(s,3H),2.09–2.00(m,1H),1.02–1.00(m,4H)。
Example 81: synthesis of 2- (6- ((2-hydroxyethyl) amino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ABT-2)
Step 1: synthesis of 2- ((6-chloro-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) amino) ethan-1-ol (ABT-1).
Intermediate (E-7) (100 mg,1 eq, 328. Mu. Mol), K, was placed in a 40-mL vial purged and maintained with an inert nitrogen atmosphere at room temperature under a nitrogen atmosphere 2 CO 3 (457 mg,10 eq, 3.28 mmol) and ethanolamine (200 mg,10 eq, 3.28 mmol) NMP (1.5 mL). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and the resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And ACN (30% ACN up to 40% in 25 minutes); detector, UV 254/220nm to give the sub-title compound (ABT-1) as a white solid (70 mg,212 μmol, 65%). M/z 330.1/332.1 (M+H) + (ES+)。
Step 2: synthesis of 2- (6- ((2-hydroxyethyl) amino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ABT-2)
1, 4-dioxane (8 mL) containing the product (ABT-1) (69 mg,1 equivalent, 209. Mu. Mol), intermediate (AC-2) (63 mg,1.5 equivalent, 314. Mu. Mol) and potassium phosphate (89 mg,2 equivalent, 418. Mu. Mol) from step 1 above, was placed in a 40-mL vial purged with an inert nitrogen atmosphere and maintained at room temperature, and XantPhos (24 mg,0.2 equivalent, 42. Mu. Mol) and Pd were added 2 (dba) 3 -CHCl 3 (22 mg,0.1 eq, 21. Mu. Mol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and the resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XBridge preparative C18 OBD column, 30X 100mm,5 μm; flow)Phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 23% B to 43% B in 10 minutes; wavelength: 254/220nm; retention time: 9.67 To give the title compound (ABT-2) (30.7 mg,62 μmol, 29%) as a white solid. M/z 495.2 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.46(s,1H),8.08(d,J=7.6Hz,1H),7.97(d,J=7.7Hz,1H),7.79–7.60(m,5H),7.55(d,J=1.2Hz,1H),6.14(d,J=1.3Hz,1H),5.24(s,2H),3.73(t,J=5.8Hz,2H),3.46–3.43(m,5H)。
Example 82: synthesis of 2-cyclopropyl-N- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- (2- (pyrrolidin-1-yl) ethoxy) pyrimidine-4-carboxamide (ABU-2)
Step 1: synthesis of 2-cyclopropyl-6- (2- (pyrrolidin-1-yl) ethoxy) pyrimidine-4-carboxamide (ABU-1)
To intermediate (ABK-6) (100 mg,1 eq, 361. Mu. Mol), HATU (274 mg,2 eq, 721. Mu. Mol) and NH at RT 4 To a stirred mixture of Cl (96 mg,5 eq, 1.80 mmol) in THF (20 mL) was added DIPEA (463 mg,10 eq, 3.61 mmol). The mixture was stirred at room temperature for 2 hours. The mixture was concentrated and purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (ABU-1) as a brown/yellow solid (75 mg, 253. Mu. Mol, 75%). M/z 277.2 (M+H) + (ES+)。
Step 2: synthesis of 2-cyclopropyl-N- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- (2- (pyrrolidin-1-yl) ethoxy) pyrimidine-4-carboxamide (ABU-2)
To a stirred mixture of the product (ABU-1) (70 mg,1 eq, 253. Mu. Mol) and the intermediate (ABC-6) (97 mg,1.2 eq, 304. Mu. Mol) from step 1 above and potassium phosphate (161 mg, 760. Mu. Mol,3 eq) in dioxane (5 mL) under nitrogen at room temperaturePd addition 2 (dba) 3 (46 mg,0.2 eq, 51. Mu. Mol) and Xantphos (59 mg,0.4 eq, 101. Mu. Mol). The mixture was stirred under nitrogen at 100 ℃ for 15 hours. The mixture was cooled to room temperature and the resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (0% ACN up to 70% in 40 minutes); detector, UV 254/220nm. The reaction was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: (column: XBridge preparation type OBD C18 column, 30 x 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 33% B to 42% B in 9 minutes; wavelength: 254/220nm; retention time: 8.2 To give the title compound (ABU-2) (21.2 mg, 38. Mu. Mol, 15%) M/z 558.2 (M+H) as a pale yellow solid + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),7.86(s,1H),7.44(d,J=1.2Hz,1H),7.26(s,1H),6.22(d,J=1.2Hz,1H),4.59(t,J=5.7Hz,2H),4.02(s,3H),3.49(s,3H),3.36(s,1H),3.31(s,1H),2.95(t,J=5.8Hz,2H),2.69–2.68(m,4H),2.35–2.28(m,1H),1.89–1.84(m,4H),1.28–1.10(m,7H)。
Example 83: synthesis of tert-butyl ((6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) methyl) carbamate (intermediate ABV-1)
1, 4-dioxane (3.33 mL) containing intermediate (ABC-6) (30 mg,1 equivalent, 94. Mu. Mol), intermediate (ABG-1) (90 mg,4 equivalent, 378. Mu. Mol) and potassium phosphate (40 mg,2 equivalent, 189. Mu. Mol) and water (0.66 mL) were placed in a 40-mL vial purged and maintained with an inert nitrogen atmosphere. Under nitrogen atmosphere at room temperature, S-PHOSPDG was added 3 (8 mg,0.1 eq, 9. Mu. Mol) and SPhOs (8 mg,0.2 eq, 19. Mu. Mol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen, then cooled to room temperature, diluted with water and quenched with EtOAc (3 x20 mL) extraction. The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XBridge preparative OBD C18 column, 30X 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 25% B to 50% B in 8 minutes; wavelength: 254/220nm; retention time: 6.27 To give the title compound (ABV-1) (5.1 mg,12 μmol, 12%) as a white solid. M/z 413.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.46(s,1H),7.84(s,1H),6.43(s,1H),6.22(s,1H),4.17(s,2H),3.94(s,3H),3.37(s,3H),3.30–3.27(m,2H),1.46(s,9H),1.18(t,J=7.2Hz,3H)。
Example 84: synthesis of 6-cyclopropyl-N- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4-methylpyridine amide (ABW-3)
Step 1: synthesis of 6-cyclopropyl-4-methylpyridine carboxylic acid (ABW-1)
To a stirred solution of intermediate (ABF-3) (200 mg,1 eq, 1.04 mmol) in THF (2 mL), meOH (2 mL) and water (1 mL) at room temperature was added LiOH (125 mg,5 eq, 5.20 mmol). The resulting mixture was stirred at room temperature for 2 hours. The mixture was concentrated in vacuo. The residue was diluted with water and the solution was acidified with HCl (aqueous, 1M) to pH 3. The solid was collected by filtration to give the sub-title compound (ABW-1) (180 mg,1.02mmol, 97%) as a yellow solid. M/z 178.1 (M+H) + (ES+)。
Step 2: synthesis of 6-cyclopropyl-4-methylpyridine amide (ABW-2)
To the product (ABW-1) (110 mg,1 equivalent, 620. Mu. Mol) from step 1 above and NH at room temperature 4 To a stirred solution of Cl (166 mg,5 eq, 3.10 mmol) in THF (5 mL) were added HATU (706 mg,3 eq, 1.86 mmol) and DIPEA (802 mg,10 eq, 6.20 mmol).The resulting mixture was stirred at room temperature for 3 hours, then concentrated in vacuo. The residue was purified by preparative TLC using petroleum ether/EtOAc (1/1). This gave the sub-title compound (ABW-2) (80 mg, 454. Mu. Mol, 73%) as a white solid. M/z 177.1 (M+H) + (ES+)。
Step 3: synthesis of 6-cyclopropyl-N- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) -pyrazol-3-yl ] -pyridin-2-yl ] -4-methylpyridine-2-carboxamide (ABW-3)
To a stirred solution of the product (ABW-2) (32 mg,1 eq, 180. Mu. Mol), intermediate (ABC-6) (58 mg,1 eq, 180. Mu. Mol) and potassium phosphate (77 mg,2 eq, 360. Mu. Mol) from step 2 above in 1, 4-dioxane (8 mL) under nitrogen at room temperature was added Xanthos (21 mg,0.2 eq, 36. Mu. Mol) and Pd 2 (dba) 3 (17 mg,0.1 eq, 18. Mu. Mol). The resulting mixture was stirred under nitrogen at 80 ℃ for 3 hours, then cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC with DCM/MeOH (10/1) and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30×150mm 5um; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 42% B to 67% B in 8 minutes; a detector, UV 254/210nm; retention time: 6.92. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ABW-3) (19.9 mg,43 μmol, 23%) as a white solid. M/z 458.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),7.86(s,1H),7.79(s,1H),7.45(d,J=1.3Hz,1H),7.34(s,1H),6.18(d,J=1.3Hz,1H),4.03(s,3H),3.48(s,3H),3.32–3.29(m,2H),2.44(s,3H),2.21–2.15(m,1H),1.22(t,J=7.2Hz,3H),1.17–1.04(m,4H)。
Example 85: synthesis of 2- [3- (ethylamino) -5- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] phenyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (ABX-6)
Step 1: synthesis of 2- (3-bromo-5-nitrophenyl) -4- (trifluoromethyl) isoindolin-1-one (ABX-2).
Into a 250-mL round bottom flask purged with an inert nitrogen atmosphere and maintained at room temperature was placed DMF (50 mL) containing 3-bromo-5-nitroaniline (ABX-1) (2.17 g,1 eq, 10.0 mmol) followed by addition of intermediate (A-3) (5.94 g,2 eq, 20.0 mmol) and DIPEA (12.9 g,10 eq, 100.0 mmol). The resulting solution was stirred at 80 ℃ overnight. The resulting mixture was diluted with water and extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (65% ACN up to 80% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ABX-2) as a yellow solid (2 g,4.99mmol, 50%). M/z 401.1/403.1 (M+H) + (ES+)。
Step 2: synthesis of 2- (3-amino-5-bromophenyl) -4- (trifluoromethyl) isoindolin-1-one (ABX-3)
Into a 250-mL round bottom flask purged with an inert nitrogen atmosphere and maintained at room temperature was placed EtOH (80 mL) and water (20 mL) from the product from step 1 above (ABX-2) (2.00 g,1 eq., 4.99 mmol) and then iron powder (1.39 g,5 eq., 24.9 mmol) and NH were added at room temperature under a nitrogen atmosphere 4 Cl (2.67 g,10 equivalents, 49.9 mmol). The resulting solution was stirred under nitrogen at 80 ℃ for 16 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (5/1). This gave (1.5 g,4.05mmol, 81%) of the sub-title compound (ABX-3) as a yellow solid. M/z 371.2/373.2 (M+H) + (ES+)。
Step 3:2- (3-bromo-5- (ethylamino) phenyl) -4- (trifluoromethyl) isoindolin-1-one (ABX-4)
Into a 100-mL round bottom flask was placed a solution containing the product from step 2 above (ABX-3) (1.30 g,1 eq., 3.50 mmol) and acetaldehyde (123 mg,0.8 eq., 2.80 mmol) in DCM (30 mL). The mixture was stirred at room temperature for 30 minutes, then NaBH (OAc) was added at 0deg.C 3 (123 mg,0.8 eq, 2.80 mmol). The resulting solution was stirred at room temperature for 16 hours, and the resulting mixture was diluted with water and extracted with DCM (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by preparative TLC using DCM/MeOH (10/1). This gave the sub-title compound (ABX-4) as a yellow solid (800 mg,2.01mmol, 57%). M/z 399.2/401.2 (M+H) + (ES+)。
Step 4: synthesis of 2- (3- (ethylamino) -5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -4- (trifluoromethyl) isoindolin-1-one (ABX-5)
A100-mL round bottom flask purged with an inert nitrogen atmosphere and maintained at room temperature was charged with 1, 4-dioxane (20 mL) containing the product (ABX-4) (3.00 g,1 eq, 8.39 mmol), bis (pinacolato) diboron (AAI-3) (1.02 g,2 eq, 4.00 mmol) and potassium acetate (560 mg,3 eq, 6.01 mmol) from step 3 above. Pd (dppf) Cl was added at room temperature under nitrogen atmosphere 2 DCM (147 mg,0.1 eq, 200. Mu. Mol) and the resulting solution was stirred under nitrogen at 80℃for 4 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (22% ACN up to 45% in 8 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ABX-5) as a brown/yellow solid (550 mg,1.23mmol, 16%). M/z 447.2 (M+H) + (ES+)。
Step 5:2- (3- (ethylamino) -5- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) phenyl) -4- (trifluoromethyl) isoindolin-1-one (ABX-6)
Under nitrogen gasA25-mL round bottom flask was charged with the product from step 4 (ABX-5) (100 mg,1 eq, 224. Mu. Mol), intermediate (AAH-4) (54 mg,1 eq, 224. Mu. Mol) and K at room temperature under an atmosphere 2 CO 3 (93 mg,3 eq, 672. Mu. Mol) 1, 4-dioxane (4 mL) and water (1 mL) followed by Pd (dppf) Cl 2 DCM (18 mg,0.1 eq, 22. Mu. Mol). The resulting solution was stirred overnight at 80 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared C18OBD column, 19×150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 24B to 49B in 9 minutes; a detector, UV 210/254nm; retention time: 8.9. this gave the title compound (ABX-6) (34.6 mg, 72. Mu. Mol, 32%) as a white solid. M/z 482.1 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.45(s,1H),8.10(d,J=7.6Hz,1H),8.01–7.95(m,1H),7.84(s,1H),7.82–7.74(m,1H),7.23(t,J=2.1Hz,1H),7.14(t,J=1.7Hz,1H),6.39(dd,J=2.1,1.4Hz,1H),5.12(s,2H),4.01(s,3H),3.36(s,3H),3.11(q,J=7.2Hz,2H),1.22(t,J=7.2Hz,3H)。
Example 86: synthesis of 2- [5- (ethylamino) -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1, 1-biphenyl ] -3-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (ABY-1)
A25-mL round bottom flask was charged with intermediate (ABX-5) (80 mg, 179. Mu. Mol,1 eq.) intermediate (D-5) (43 mg,1 eq., 179. Mu. Mol) and K under nitrogen at room temperature 2 CO 3 (74 mg,3 eq, 538. Mu. Mol) of 1, 4-dioxane (4 mL) and water (1 mL) followed by Pd (dppf) Cl 2 DCM (15 mg,0.1 eq, 18. Mu. Mol). The resulting solution was stirred overnight at 80 ℃ under nitrogen atmosphere, then cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared C18 OBD column, 30×150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 48B to 66B in 8 minutes; a detector, UV 210/254nm; retention time: 6.05. this gave 1.1 (20.3 mg, 42. Mu. Mol, 24%) as a white solid. M/z 478.1 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.39(s,1H),8.09(d,J=7.6Hz,1H),7.97(d,J=7.7Hz,1H),7.82–7.68(m,3H),7.64–7.54(m,2H),7.20(t,J=2.1Hz,1H),6.92(t,J=1.8Hz,1H),6.30(t,J=1.8Hz,1H),5.04(s,2H),3.19(s,3H),3.04(q,J=7.2Hz,2H),1.21(t,J=7.2Hz,3H)。
Example 87: synthesis of 6-cyclopropyl-N- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (hydroxymethyl) picolinamide (ABZ-7)
Step 1: synthesis of (2, 6-dichloropyridin-4-yl) methanol (ABZ-2)
To a stirred solution of methyl 2, 6-dichloropyridine-4-carboxylate (ABZ-1) (8.20 g,1 eq, 39.8 mmol) in a mixture of MeOH (100 mL) and THF (20 mL) at 0deg.C was added NaBH 4 (7.53 g,199mmol,5 eq.). The resulting mixture was stirred at room temperature for 1 hour, then at 0℃by addition of saturated NH 4 Cl solution (aq) (60 mL) was quenched. The resulting mixture was diluted with water and extracted with EtOAc (3×500 mL). The combined organic layers were washed with brine (2×500 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. This gave the sub-title compound (ABZ-2) (7 g,39.5mmol, 99%) as a white solid. M/z 178.0/180.0 (M+H) + (ES+)。
Step 2: synthesis of (2-chloro-6-cyclopropylpyridin-4-yl) methanol (ABZ-3)
To the product (ABZ-2) (3.56 g,1 equivalent, 20.0 mmol), cyclopropylboronic acid (ABF-2) (1.89 g,1.1 equivalent, 22.0 mmol) from step 1 above, and K under a nitrogen atmosphere at room temperature 2 CO 3 (8.29 g,3 eq, 60.0 mmol) in a mixture of dioxane (20 mL) and water (1 mL) Pd (dppf) Cl was added to the solution 2 DCM (1.46 g,0.1 eq, 2.00 mmol). The resulting mixture was stirred overnight at 90 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (5/1) to give the sub-title compound (ABZ-3) (2.1 g,11.5mmol, 57%) as a yellow solid. M/z 184.0/186.0 (M+H) + (ES+)。
Step 3: synthesis of methyl 6-cyclopropyl-4- (hydroxymethyl) picolinate (ABZ-4)
To the product (ABZ-3) (2.10 g,1 equivalent, 11.4 mmol) from step 2 above and Et at room temperature under a nitrogen atmosphere 3 To a stirred solution of N (3.47 g,3 equivalents, 34.3 mmol) in MeOH (40 mL) was added Pd (dppf) Cl 2 DCM (418 mg,0.05 eq., 572. Mu. Mol). The resulting mixture was stirred overnight at 70 ℃ under carbon monoxide atmosphere. The resulting mixture was cooled to room temperature and concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (2/1) to give the sub-title compound (ABZ-4) (1.82 g,8.79mmol, 77%) as a pale yellow oil. M/z 208.0 (M+H) + (ES+)。
Step 4: synthesis of 6-cyclopropyl-4- (hydroxymethyl) picolinic acid (ABZ-5)
To a stirred solution of the product from step 3 above (ABZ-4) (200 mg,1 eq., 965 μmol) in THF (2 mL), meOH (2 mL) and water (1 mL) at room temperature was added LiOH (116 mg,5 eq., 4.83 mmol). The resulting mixture was stirred at room temperature for 2 hours, then concentrated in vacuo. The residue was diluted with water and acidified to pH 3 with HCl (aqueous, 1M). The solid was collected by filtration to give the sub-title compound (ABZ-5) (180 mg,932 μmol, 97%) as a white solid. M/z 194.1 (M+H) + (ES+)。
Step 5: synthesis of 6-cyclopropyl-4- (hydroxymethyl) pyridine amide (ABZ-6)
At 0deg.C, to the product (ABZ-5) (160 mg,1 eq., 820. Mu. Mol) from step 4 above and NH 4 To a stirred solution of Cl (222 mg,5 eq, 4.14 mmol) in THF (5 mL) were added HATU (945 mg,3 eq, 2.48 mmol) and DIPEA (1.00 g, 10)Equivalent, 8.28 mmol) and the resulting mixture was stirred at room temperature overnight. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (40% ACN up to 80% in 8 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ABZ-6) (120 mg, 625. Mu. Mol, 75%) as a white solid. M/z 193.1 (M+H) + (ES+)。
Step 6: synthesis of 6-cyclopropyl-N- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (hydroxymethyl) picolinamide (ABZ-7)
To a stirred mixture of the product (ABZ-6) (48 mg,1 eq, 250. Mu. Mol), intermediate (ABC-6) (79 mg,1 eq, 250. Mu. Mol) and potassium phosphate (1599 mg,3 eq, 749. Mu. Mol) from step 5 above in 1, 4-dioxane (4 mL) under nitrogen at room temperature was added Xanthos (29 mg,0.2 eq, 50. Mu. Mol) and Pd 2 (dba) 3 (23 mg,0.1 eq, 25. Mu. Mol) and the resulting mixture was stirred under nitrogen at 100℃for 2 hours. The resulting mixture was cooled to room temperature and diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using petroleum ether/EtOAc (1/1). The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30×150mm 5um; mobile phase a: water (10% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 29% B to 54% B in 8 minutes; a detector, UV 254/210nm; retention time: 6.6. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ABZ-7) (17.6 mg,37 μmol, 14.88%) as a white solid. M/z 474.4 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),7.96(d,J=1.4Hz,1H),7.86(s,1H),7.47(m,2H),6.19(d,J=1.2Hz,1H),4.72(s,2H),4.03(s,3H),3.48(s,3H),3.31-3.19(m,2H),2.26-2.20(m,1H),1.22(t,J=7.2Hz,3H),1.16-1.06(m,4H)。
Example 88: synthesis of (R) -2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((3-fluoropyrrolidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (ACA-1)
Et is added to a stirred solution of intermediate (ABP-1) (30 mg,1 eq, 50. Mu. Mol) and (3R) -3-fluoropyrrolidine, HCl (Y-1) (21 mg,4 eq, 230. Mu. Mol) in DCM (5 mL) at room temperature 3 N (18 mg,3 eq, 170. Mu. Mol) and NaBH (OAc) 3 (25 mg,2 equivalents, 110. Mu. Mol). The resulting mixture was stirred at room temperature for 1 hour, then concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1). The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 30X 150mm 5um; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 43B to 68B in 8 minutes; a detector, UV 254/210nm; retention time: 6.53. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ACA-1) (5.2 mg,8.9 μmol, 15%) as a white solid. M/z 584.3 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),8.07(s,1H),8.00(s,1H),7.86(s,1H),7.66(s,1H),6.20(s,1H),5.31–5.09(m,3H),4.04(s,3H),3.94–3.82(m,2H),3.50(s,3H),3.50–3.36(t,J=7.2Hz,2H),2.99–2.85(m,2H),2.84–2.68(m,1H),2.56–2.46(m,1H),2.33–2.16(m,1H),2.14–1.96(m,1H),1.25(t,J=7.2Hz,3H)。
Example 89: synthesis of (S) -2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((3-fluoropyrrolidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (ACB-1)
Et is added to a stirred solution of intermediate (ABP-1) (30 mg,1 eq, 50. Mu. Mol) and (S) -3-fluoropyrrolidine, HCl (V-1) (21 mg,4 eq, 230. Mu. Mol) in DCM (5 mL) at room temperature 3 N (18 mg,3 eq, 170. Mu. Mol) and NaBH (OAc) 3 (25 mg,2 eq, 110. Mu. Mol) and the resulting mixture was stirred at room temperature for 1 hour. The mixture was concentrated in vacuo and the residue was purified by preparative TLC using DCM/MeOH (15/1). The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 30X 150mm 5um; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 37B to 54B in 10 minutes; a detector, UV 254/210nm; retention time: 8.2. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ACB-1) (8.2 mg,14 μmol, 23%) as a white solid. M/z 584.3 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),8.07(s,1H),8.00(s,1H),7.87(s,1H),7.66(d,J=1.2Hz,1H),6.20(d,J=1.2Hz,1H),5.31–5.24(m,3H),4.04(s,3H),3.94–3.82(m,2H),3.50(s,3H),3.40–3.34(m,2H),2.96–2.87(m,2H),2.92–2.76(m,1H),2.56–2.45(m,1H),2.35–2.15(m,1H),2.14–1.95(m,1H),1.25(t,J=7.2Hz,3H)。
Example 90: synthesis of 2- (4-methyl-4H-1, 2, 4-triazol-3-yl) -3'- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) - [1,1' -biphenyl ] -4-carbonitrile (ACC-8)
Step 1: synthesis of methyl 4-cyano-3 '-nitro- [1,1' -biphenyl ] -2-carboxylate (ACC-2)
To a 250mL round bottom flask was added 1, 4-dioxane (40 mL) containing methyl 2-bromo-5-cyanobenzoate (ACC-1) (3.00 g,10.5mmol,1 eq.) intermediate (AAB-2) (2.09 g,12.5mmol,1.2 eq.) and potassium phosphate (2.89 g,20.9mmol,2 eq.) under nitrogen at room temperature. At nitrogen Pd (dppf) Cl was added to the above mixture at room temperature under a gas atmosphere 2 DCM (760 mg,0.1 eq, 1.05 mmol) and the resulting mixture was stirred under nitrogen at 80℃for 3 hours. The mixture was cooled to room temperature and diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography petroleum ether/EtOAc (4/1) to give the sub-title compound (ACC-2) (2.2 g,7.79mmol, 75%) as a yellow oil. M/z 283.1 (M+H) + (ES+)。
Step 2: synthesis of 4-cyano-3 '-nitro- [1,1' -biphenyl ] -2-carboxylic acid (ACC-3)
To a 20mL sealed tube were added THF (8 mL) and water (2 mL) containing the product (ACC-2) (846 mg,1 eq, 3.00 mmol) from step 1 above at room temperature. LiOH (215 mg,3 equivalents, 8.99 mmol) was added to the above mixture at room temperature over 3 minutes, and the resulting mixture was stirred at 60 ℃ for 3 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was diluted with water and the solution was acidified with HCl (aqueous, 1M) to pH 3. The solid was collected by filtration to give the sub-title compound (ACC-3) (650 mg,2.42mmol, 81%) as a yellow oil. M/z 269.1 (M+H) + (ES+)。
Step 3:2- (4-cyano-3 '-nitro- [1,1' -biphenyl ] -2-carbonyl) -N-methylhydrazine-1-thiocarboxamide (ACC-4)
DMF (10 mL) containing the product (ACC-3) (600 mg,1 eq, 2.24 mmol) from step 2 above, 1-amino-3-methyl thiourea (D-2) (704 mg,3 eq, 6.71 mmol) and DIPEA (867 mg,6.71mmol,3 eq) was added to a 20mL sealed tube at room temperature. HATU (851 mg,1 eq, 2.24 mmol) was added to the above mixture at 0 ℃ over 3 min, and the resulting mixture was stirred at room temperature overnight. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (30% ACN up to 70% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gives a small as a white solidThe title compound (ACC-4) (450 mg,1.27mmol, 57%). M/z 356.1 (M+H) + (ES+)。
Step 4:2- (5-mercapto-4-methyl-4H-1, 2, 4-triazol-3-yl) -3 '-nitro- [1,1' -biphenyl ] -4-carbonitrile (ACC-5)
To a stirred solution of the product (ACC-4) from step 3 above (430 mg,1 eq, 1.21 mmol) in DMF (8 mL) was added NaOH (aqueous solution, 1M) (15 mL) at room temperature. The resulting mixture was stirred at 50 ℃ overnight. The mixture was cooled to room temperature and the crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (30% ACN up to 50% in 7 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ACC-5) (120 mg, 356. Mu. Mol, 29%) as a yellow solid. M/z 338.1 (M+H) + (ES+)。
Step 5:2- (4-methyl-4H-1, 2, 4-triazol-3-yl) -3 '-nitro- [1,1' -biphenyl ] -4-carbonitrile (ACC-6)
To a stirred solution of the product from step 4 above (ACC-5) (100 mg,1 eq, 296 μmol) in DCM (5 mL) was added acetic acid (53 mg,3 eq, 888 μmol) and hydrogen peroxide solution (168 mg,30wt%,5 eq, 1.48 mmol) at 0 ℃. The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was diluted with water. The solution was saturated with NaHCO 3 The solution (aqueous solution) was alkalized to pH 8. The resulting mixture was extracted with DCM (3×50 mL) and the combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. This gave the sub-title compound (ACC-6) (85 mg, 278. Mu. Mol, 94%) as a yellow solid. M/z 306.1 (M+H) + (ES+)。
Step 6:3 '-amino-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (ACC-7)
MeOH (5 mL) containing the product (ACC-6) from step 5 above (80 mg,1 eq, 262. Mu. Mol) was added to a 20mL sealed tube at room temperature under a nitrogen atmosphere. Pd/C39 (3 mg,10wt%,0.1 eq, 26. Mu. Mol) was added to the above mixture at room temperature under a nitrogen atmosphere . The resulting mixture was stirred at room temperature under a hydrogen atmosphere for 3 hours. Filtering the resulting mixture; the filter cake was washed with MeOH (3X 2 mL). The filtrate was concentrated in vacuo to give the subtitle compound (ACC-7) (50 mg,182 μmol, 69%) as a colorless oil. M/z 276.1 (M+H) + (ES+)。
Step 7:2- (4-methyl-4H-1, 2, 4-triazol-3-yl) -3'- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) - [1,1' -biphenyl ] -4-carbonitrile (ACC-8)
EtOH (2 mL) containing the product (ACC-7) from step 6 above (20 mg,1 eq, 73. Mu. Mol) and intermediate (A-3) (24 mg,1.1 eq, 80. Mu. Mol) was added to an 8mL sealed tube at room temperature. Et is added to the above mixture at room temperature over 5 minutes 3 N (30 uL,3 eq, 219. Mu. Mol). The resulting mixture was stirred at 80 ℃ overnight. The mixture was cooled to room temperature and the resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (10% ACN up to 50% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 34% B to 48% B in 10 minutes; wavelength: 254/220nm; retention time: 9.8 To give the title compound (ACC-8) (2.0 mg,4.3 μmol, 6%) as a white solid. M/z 460.1 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.44(s,1H),8.14–7.91(m,6H),7.85–7.76(m,2H),7.50(t,J=8.0Hz,1H),7.13(d,J=8.0Hz,1H),5.11(s,2H),3.24(s,3H)。
Example 91: synthesis of 2- (4-methyl-4H-1, 2, 4-triazol-3-yl) -3'- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) - [1,1' -biphenyl ] -4-carboxamide (ACD-1)
To compound (ACC-8) (10 mg,1 equivalent, 22. Mu. Mol) and K at 0 ℃ 2 CO 3 To a stirred mixture of (9 mg,3 eq, 66. Mu. Mol) in DMSO (1 mL) was added hydrogen peroxide (0.3 mL,30 Wt%). The resulting mixture was stirred at room temperature for 20 minutes, then quenched with aqueous sodium thiosulfate (2 mL) at 0 ℃. The mixture was extracted with EtOAc (3×10 mL) and the combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 25% B to 45% B in 9 minutes; wavelength: 254/220nm; retention time: 8.5 To give the title compound (ACD-1) (6 mg,13 μmol, 58%) as a white solid. M/z 478.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.43(s,1H),8.23–8.16(m,2H),8.11–8.01(m,4H),7.84–7.70(m,3H),7.55(s,1H),7.41(t,J=8.0Hz,1H),6.95–6.93(m,1H),5.13(s,2H),3.10(s,3H)。
Example 92: synthesis of (R) -2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((3-methylpiperidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (ACE-1)
Intermediate-containing (ABP-1) (50 mg,1 eq, 98. Mu. Mol), (R) -3-methylpiperidine, HCl (53 mg,4 eq, 392. Mu. Mol) and Et are placed in a 40-mL vial purged and maintained with an inert nitrogen atmosphere at room temperature 3 N (30 mg,3 eq, 294. Mu. Mol) DCM (4 mL). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 1 hour. NaBH (OAc) was added to the above mixture at room temperature 3 (42 mg,2 eq, 196. Mu. Mol) and the resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2X 00 mL) and dried(Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XBridge preparative OBD C18 column, 30X 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 58% B to 83% B in 8 minutes; wavelength: 254/210nm; retention time: 7.08 To give the title compound (ACE-1) (18.8 mg,31 μmol, 32%) as a white liquid. M/z 594.4 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.47(s,1H),7.97(s,1H),7.92(s,1H),7.88(s,1H),7.57(d,J=1.2Hz,1H),6.92(t,J=5.4Hz,1H),6.25(d,J=1.2Hz,1H),5.20(s,2H),3.90(s,3H),3.64(s,2H),3.47(s,3H),3.29–3.24(m,2H),2.74–2.70(m,2H),1.96–1.91(m,1H),1.71–1.40(m,5H),1.17(t,J=7.1Hz,3H),0.90–0.81(m,4H)。
Example 93: synthesis of (S) -2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((3-methylpiperidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (ACF-1)
Intermediate (ABP-1) (50 mg, 98. Mu. Mol,1.0 eq), (S) -3-methylpiperidine, HCl (R-1) (53 mg,4 eq, 392. Mu. Mol) and Et are placed in a 40-mL vial purged and maintained with an inert nitrogen atmosphere at room temperature 3 N (30 mg,3 eq, 294. Mu. Mol) DCM (4 mL). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 1 hour. NaBH (OAc) was added to the above mixture at room temperature 3 (42 mg,2 eq, 196. Mu. Mol) and the resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XBridge preparative OBD C18 column, 30X 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60Milliliters per minute; gradient: 50% B to 70% B within 9 minutes; wavelength: 254/220nm; retention time: 8.83 To give the title compound (ACF-1) (18.8 mg,32 μmol, 32%) as a white liquid. M/z 594.4 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),8.04(s,1H),7.97(s,1H),7.86(s,1H),7.66(d,J=1.2Hz,1H),6.20(d,J=1.2Hz,1H),5.26(s,2H),4.04(s,3H),3.70(s,2H),3.51(s,3H),3.41–3.34(m,2H),2.87–2.81(m,2H),2.05–1.98(m,1H),1.82–1.55(m,5H),1.25(t,J=7.2Hz,3H),0.99–0.77(m,4H)。
Example 94: synthesis of 6- (aminomethyl) -2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ACG-3)
Step 1: synthesis of N- ((2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -3-oxo-7- (trifluoromethyl) isoindolin-5-yl) methyl) -2-methylpropan-2-sulfinamide (ACG-2)
To a stirred mixture of intermediate (ABP-1) (50 mg,1 eq, 98. Mu. Mol) and t-butylsulfinamide (ACG-1) (36 mg,3 eq, 294. Mu. Mol) in THF (2 mL) under nitrogen at room temperature was added Ti (OiPr) 4 (139 mg,5 eq, 490. Mu. Mol) and NaBH 4 (11 mg,3 equivalents, 294. Mu. Mol). The resulting mixture was stirred under nitrogen at 60 ℃ for 1 hour. The mixture was cooled to room temperature, and the resulting mixture was diluted with water. The resulting mixture was extracted with EtOAc (3×20 mL) and the combined organic layers were washed with water (3×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (ACG-2) (50 mg, 81. Mu. Mol, 83%) as a yellow solid. M/z 616.2 (M+H) + (ES+)。
Step 2:6- (aminomethyl) -2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ACG-3)
A mixture of the product (ACG-2) (50 mg,1 equivalent, 81. Mu. Mol) from step 1 above in 1, 4-dioxane and a mixture of HCl in 1, 4-dioxane (1 mL, 4M) were stirred at room temperature for 1 hour. The resulting mixture was diluted with water. The resulting mixture was extracted with EtOAc (3×20 mL) and the combined organic layers were washed with water (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 30% B to 43% B in 9 minutes; wavelength: 254/220nm; retention time: 8.72 To give the title compound (ACG-3) (21 mg,41 μmol, 51%) as a white solid. M/z 512.3 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.47(s,1H),8.03(d,J=15.6Hz,2H),7.89(s,1H),7.58(d,J=1.2Hz,1H),6.91(t,J=5.4Hz,1H),6.25(d,J=1.2Hz,1H),5.19(s,2H),3.94–3.90(m,5H),3.48(s,3H),3.29–3.21(m,2H),1.17(t,J=7.1Hz,3H)。
Example 95: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (ACH-1)
To a stirred mixture of intermediate (ABP-1) (40 mg,1 eq, 78. Mu. Mol) in THF (5 ml) at room temperature was added NaBH 4 (6 mg,2 equivalents, 156. Mu. Mol). The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was diluted with water. The resulting mixture was extracted with EtOAc (3×30 mL) and the combined organic layers were washed with brine (3×30 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1). Will be thickThe product was purified by preparative HPLC under the following conditions: (column: XBridge preparation type OBD C18 column, 30 x 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 40% B to 60% B,60% B in 8 minutes; wavelength: 254/220nm; retention time: 7.83 To give the title compound (ACH-1) (20 mg,39 μmol, 5%) as a white solid. M/z 513.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.47(s,1H),7.98(d,J=18.3Hz,2H),7.89(s,1H),7.58(d,J=1.2Hz,1H),6.92(t,J=5.4Hz,1H),6.25(d,J=1.2Hz,1H),5.57(t,J=5.8Hz,1H),5.20(s,2H),4.70(d,J=5.8Hz,2H),3.90(s,3H),3.47(s,3H),3.30–3.23(m,2H),1.17(t,J=7.1Hz,3H)。
Example 96: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- (((2-hydroxyethyl) amino) methyl) -4- (trifluoromethyl) isoindolin-1-one (ACI-1)
A solution of intermediate (ABP-1) (20 mg,1 eq, 39. Mu. Mol) and ethanolamine (12 mg,5 eq, 195. Mu. Mol) in DCM (5 mL) was stirred at room temperature for 1 hour. NaBH was added to the above mixture at 0deg.C over 0.5 hours 4 (3 mg,2 eq, 78. Mu. Mol) and the resulting mixture was stirred at room temperature for 1 hour. The reaction was quenched with MeOH (2 mL) at 0 ℃, and the resulting mixture was diluted with DCM (30 mL) and concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (5/1). The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 17% B to 42% B in 10 minutes; wavelength: 254/220nm to give the title compound (ACI-1) (19.7 mg, 35. Mu. Mol, 87%) as a white solid. M/z 556.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.47(s,1H),8.01(d,J=15.6Hz,2H),7.89(s,1H),7.58(d,J=1.2Hz,1H),6.91(t,J=5.4Hz,1H),6.25(d,J=1.2Hz,1H),5.19(s,2H),4.49(t,J=5.4Hz,1H),3.95-3.85(m,5H),3.52–3.47(m,5H),3.31-3.22(m,2H),2.57(t,J=5.8Hz,2H),1.17(t,J=7.1Hz,3H)。
Example 97: synthesis of methyl 1- (4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) pyridin-2-yl) cyclopropane-1-carboxylate (ACJ-6)
Step 1: synthesis of tert-butyl 2- (4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) pyridin-2-yl) acetate (ACJ-2)
XPhos (10 mg,0.2 eq, 21. Mu. Mol) and Pd were added dropwise to a stirred solution of intermediate (AAT-1) (50 mg, 106. Mu. Mol,1 eq) in THF (5 mL) at room temperature under nitrogen atmosphere 2 (dba) 3 (9.74 mg,0.1 eq, 11. Mu. Mol). The resulting mixture was stirred under nitrogen at 60 ℃ for 10 minutes. Tert-butyl 2- (bromozincate) acetate (ACJ-1) (139 mg,5 eq., 530. Mu. Mol) was added to the system at 60℃under a nitrogen atmosphere. The resulting mixture was stirred under nitrogen at 60 ℃ for 5 hours and then cooled to room temperature. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (ACJ-2) as an off-white solid (52 mg, 94. Mu. Mol, 80%). M/z 550.2 (M+H) + (ES+)。
Step 2: synthesis of tert-butyl 1- (4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) pyridin-2-yl) cyclopropane-1-carboxylate (ACJ-4)
DMF (12 mL) containing the product from step 1 above (ACJ-2) (60 mg,1 eq, 109. Mu. Mol) and (2-bromoethyl) diphenyltriflic sulfonium (ACJ-3) (145 mg,3 eq, 327. Mu. Mol) was stirred at room temperature for 10 min. Et is then added to the system at room temperature 3 N (55 mg,5 eq, 545. Mu. Mol). The resulting mixture was stirred at room temperature for 48 hours. The reaction was then quenched by the addition of ice water (3 mL) at 0 ℃. And is combined withThe resulting mixture was extracted with EtOH (3X 20 mL). The combined organic layers were washed with water (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (ACJ-4) as a white solid (40 mg, 69. Mu. Mol, 57%). M/z 576.2 (M+H) + (ES+)。
Step 3:1- (4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) pyridin-2-yl) cyclopropane-1-carboxylic acid (ACJ-5)
The product from step 2 above (ACJ-4) (30 mg,1 eq, 52. Mu. Mol) in DCM (3 mL) and TFA (1 mL) were stirred at room temperature for 2 hours. The crude product was used directly in the next step without further purification. M/z 520.2 (M+H) + (ES+)。
Step 4: synthesis of methyl 1- (4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) pyridin-2-yl) cyclopropane-1-carboxylate (ACJ-6)
To a 10mL vial was added the product from step 3 above (ACJ-5) (10 mg,1 eq, 19. Mu. Mol) and MeOH (3 mL) at room temperature. Sulfuric acid (0.2 mL) was added to the above mixture at 0deg.C. The resulting mixture was stirred at 60 ℃ for an additional 2 hours. The mixture was cooled to room temperature. The reaction was then quenched by the addition of ice water (2 mL) at 0 ℃. The resulting mixture was extracted with EtOAc (3×10 mL). The combined organic layers were washed with water (2×10 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC (column: xselect CSH OBD column 30X 150mm 5um; mobile phase A: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 2% B to 23% B in 7 min; wavelength: 254/220 nm) to give the title compound (ACJ-6) (0.8 mg, 1.50. Mu. Mol, 7.4%) as a white solid. M/z534.1 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.49(s,1H),8.26(d,J=1.5Hz,1H),8.10(dd,J=17.0,7.6Hz,2H),7.85–7.74(m,2H),7.72–7.57(m,3H),7.02(d,J=1.4Hz,1H),5.23(s,2H),3.63(s,3H),3.32(s,3H),1.53(q,J=4.0,3.5Hz,2H),1.40(q,J=4.6,4.1Hz,2H)。
Example 98: synthesis of 2- (6-cyclopropyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (ACK-3)
Step 1: synthesis of 2- (6-chloro-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -3-oxo-7- (trifluoromethyl) isoindoline-5-carbaldehyde (ACK-1)
To 2, 6-dichloro-4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl under nitrogen at room temperature]Pyridine (E-7) (100 mg,1 eq, 328. Mu. Mol), intermediate (ABC-4) (68 mg,0.9 eq, 295. Mu. Mol) and potassium phosphate (209 mg,3 eq, 984. Mu. Mol) were added XantPhos (19 mg,0.1 eq, 33. Mu. Mol) and Pd (OAc) to a stirred mixture of dioxane (4 mL) 2 (7 mg,0.1 eq, 33. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The resulting mixture was concentrated in vacuo and the residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (ACK-1) as a yellow oil (60 mg,120 μmol, 37%). M/z 498.1 (M+H) + (ES+)。
Step 2:2- (6-cyclopropyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -3-oxo-7- (trifluoromethyl) isoindoline-5-carbaldehyde (ACK-2)
To a stirred mixture of the product (ACK-1) (150 mg,1 eq, 301. Mu. Mol), intermediate (ABF-2) (52 mg,2 eq, 602. Mu. Mol) and potassium phosphate (192 mg,3 eq, 904. Mu. Mol) in toluene (3 mL) from step 1 above was added tricyclohexylphosphine (8 mg,0.1 eq, 30. Mu. Mol) and Pd (AcO) at room temperature under a nitrogen atmosphere 2 (7 mg,0.1 eq, 30 mmol). The resulting mixture was stirred under nitrogen at 110 ℃3 hours. The mixture was cooled to room temperature and concentrated in vacuo. The precipitated solid was collected by filtration and washed with EtOAc (3×5 mL). The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (ACK-2) as a yellow oil (80 mg, 159. Mu. Mol, 53%). M/z 504.2 (M+H) + (ES+)。
Step 3:2- (6-cyclopropyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (ACK-3)
To a stirred solution of the product (ACK-2) (30 mg,1 eq, 60. Mu. Mol) from step 2 above in THF (5 mL) at room temperature was added NaBH 4 (4.51 mg,2 equivalents, 119. Mu. Mol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of ice water (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 40% B to 60% B in 8 minutes; wavelength: 254/220nm; retention time: 7.83 To give the title compound (ACK-3) (2.9 mg,5.74 μmol, 10%) as a white solid. M/z 506.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.49(s,1H),8.06–7.93(m,3H),7.79–7.73(m,1H),7.71–7.61(m,3H),6.81(d,J=1.4Hz,1H),5.59(t,J=5.8Hz,1H),5.18(s,2H),4.71(d,J=5.8Hz,2H),3.38(s,3H),2.09–2.00(m,1H),1.02–0.95(m,2H),0.95–0.88(m,2H)。
Example 99: synthesis of 6- ((5-azaspiro [2.4] heptan-5-yl) methyl) -2- (6-cyclopropyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ACL-1)
To intermediate (ACK-2) (30 mg,1 equivalent, 60. Mu. Mol) and 5-azaspiro [2.4] under a nitrogen atmosphere at room temperature ]To a stirred mixture of heptane (AAC-1) (9 mg,1.5 eq, 90. Mu. Mol) in DCM (5 mL) was added Et 3 N (18 mg,3 eq, 180. Mu. Mol) and NaBH (OAc) 3 (63 mg,5 eq, 300. Mu. Mol). The resulting mixture was stirred under nitrogen at 40 ℃ for 2 hours. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC (column: XBridge preparative OBD C18 column, 30.150 mm,5 μm; mobile phase A: water (0.1% NH4HCO 3), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 62% B to 82% B in 8 min; wavelength: 254/220nm; retention time: 7.66) to give the title compound (ACL-1) (2.9 mg, 4.96. Mu. Mol, 8%) as a white solid. M/z 585.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.49(s,1H),8.04–7.99(m,2H),7.96(s,1H),7.78–7.73(m,1H),7.70–7.61(m,3H),6.82(d,J=1.4Hz,1H),5.17(s,2H),3.80(s,2H),3.38(s,3H),2.70(t,J=6.8Hz,2H),2.46(s,2H),2.10–2.02(m,1H),1.77(t,J=6.8Hz,2H),1.01–0.95(m,2H),0.95–0.89(m,2H),0.55–0.47(m,4H)。
Example 100: synthesis of 2- (6- (ethylamino) -4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ACM-8)
Step 1: synthesis of ethyl 2- (2, 6-dichloropyridin-4-yl) -5-fluorobenzoate (ACM-2)
To ethyl 2-bromo-5-fluorobenzoate (ACM-1) (4.94 g,1 eq, 20.0 mmol), intermediate (AAN-1) (4.60 g,1.2 eq, 24.0 mmol) and K under a nitrogen atmosphere at room temperature 2 CO 3 (8.29 g,3 eq, 60.0 mmol) Pd (DtBPF) Cl was added to a stirred mixture of 1, 4-dioxane (80 mL) 2 (1.30 g,0.1 eq, 2.00 mmol). The resulting mixture was stirred under nitrogen at 60 ℃ for 5 hours. The resulting mixture was cooled to room temperature and diluted with water. The resulting mixture was extracted with EtOAc (3×300 mL). The combined organic layers were washed with brine (2×300 mL), dried (Na 2 SO 4 ) Filter and in trueConcentrating in the air. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (36% ACN up to 48% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ACM-2) (4.6 g,14.7mmol, 73%) as a brown solid. M/z 314.0/316.0 (M+H) + (ES+)。
Step 2: synthesis of 2- (2, 6-dichloropyridin-4-yl) -5-fluorobenzoic acid (ACM-3)
To a stirred mixture of the product (ACM-2) from step 1 above (2.3 g,1 eq, 7.32 mmol) in THF (30 mL) was added LiOH (700 mg,4 eq, 29.3 mmol) over 3 minutes at room temperature. The resulting mixture was stirred at 60 ℃ for an additional 3 hours. The resulting mixture was cooled to room temperature. The residue was neutralized to pH 5 with HCl (aqueous, 1M). The resulting mixture was diluted with water and extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude mixture was used directly in the next step without further purification. M/z 286.0/288.0 (M+H) + (ES+)。
Step 3: synthesis of 2- (2, 6-dichloropyridin-4-yl) -5-fluorobenzoyl) -N-methylhydrazine-1-thiocarboxamide (ACM-4)
To a stirred mixture of the product (ACM-3) (2.00 g,1 eq, 6.99 mmol) from step 2 above and 4-methyl-3-thiosemicarbazide (D-2) (740 mg,1 eq, 6.99 mmol) in THF (30 mL) was added DIPEA (2.71 g,3 eq, 21.0 mmol) and HATU (2.66 g,1 eq, 6.99 mmol) at room temperature. The resulting mixture was stirred at room temperature for 2 hours. The crude mixture was used directly in the next step without further purification. M/z373.0/375.0 (M+H) + (ES+)。
Step 4: synthesis of 5- (2, 6-dichloropyridin-4-yl) -5-fluorophenyl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (ACM-5)
NaOH (aqueous solution, 10M) was added to the stirred mixture of the product (ACM-4) from step 3 above at room temperature. The resulting mixture was subjected to a temperature of 100 ℃Stir overnight. The residue was cooled to room temperature and acidified to pH 5 with HCl (aqueous, 1M). The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude mixture was used directly in the next step without further purification. M/z 355.0/357.0 (M+H) + (ES+)。
Step 5: synthesis of 2, 6-dichloro-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridine (ACM-6)
To a stirred mixture of the product from step 4 above (ACM-5) (1 g,1 eq, 2.81 mmol) in DCM (10 mL) was added acetic acid (340 mg,2 eq, 5.63 mmol) and hydrogen peroxide (480 g,5 eq, 14.1 mmol) at room temperature. The resulting mixture was stirred at room temperature for 2 hours. The mixture was treated with saturated NaHCO 3 (aqueous solution) neutralization to pH 9. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (50% ACN up to 70% within 15 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ACM-6) (800 mg,2.48mmol, 88%) as a white solid. M/z 323.0/325.0 (M+H) + (ES+)。
Step 6: synthesis of 6-chloro-N-ethyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-amine (ACM-7)
To a stirred mixture of the product (ACM-6) (400 mg,1 eq, 1.24 mmol) from step 5 above and ethylamine, HCl (578 mg,10 eq, 12.4 mmol) in NMP (5 mL) at room temperature was added K 2 CO 3 (1.71 g,10 equivalents, 12.3 mmol). The resulting mixture was stirred at 100 ℃ overnight. The mixture was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (36% ACN up to 48% in 10 minutes); the light emitted by the light source is detected by the detector,UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo to give the sub-title compound (ACM-7) (300 mg,906 μmol, 73%) as a white solid. M/z332.1/334.1 (M+H) + (ES+)。
Step 7: synthesis of 2- (6- (ethylamino) -4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ACM-8)
To a stirred mixture of the product (ACM-7) (150 mg,1 eq, 452. Mu. Mol), intermediate (AC-2) (91 mg,1 eq, 452 mmol) and potassium phosphate (287 mg,3 eq, 1.36 mmol) from step 6 above in 1, 4-dioxane (2 mL) was added XantPhos (52 mg,0.2 eq, 90. Mu. Mol) and Pd under nitrogen at room temperature 2 (dba) 3 (41 mg,0.1 eq, 45. Mu. Mol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The resulting mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (20% ACN up to 60% in 25 minutes); detector, UV 254/220nm. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 41% B to 59% B in 10 minutes; wavelength: 254/220nm; retention time: 9.5 To give the title compound (ACM-8) (12 mg,24 μmol, 5.3%) as a white solid. M/z 497.2 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.47(s,1H),8.08(d,J=7.7Hz,1H),7.97(d,J=7.7Hz,1H),7.80–7.71(m,2H),7.56–7.47(m,2H),7.45–7.42(m,1H),6.03(d,J=1.3Hz,1H),5.23(s,2H),3.45(s,3H),3.32–3.26(m,2H),1.22(t,J=7.2Hz,3H)。
Example 101: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- (2- (pyrrolidin-1-yl) ethoxy) -4- (trifluoromethyl) isoindolin-1-one (ACN-5)
Step 1: synthesis of 6- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ACN-1)
Pd (dppf) Cl was added to a stirred solution of intermediate (ABC-2) (200 mg,1 eq, 714. Mu. Mol), intermediate (AAI-3) (5.44 g,30 eq, 21.4 mmol) and potassium acetate (210 mg,3 eq, 2.14 mmol) in 1, 4-dioxane (20 mL) at room temperature under nitrogen atmosphere 2 DCM (116 mg,0.2 eq., 143. Mu. Mol). The resulting mixture was stirred under nitrogen at 80 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by silica gel column chromatography eluting with DCM/MeOH (15:1) to give the sub-title compound (ACN-1) as a brown/yellow solid (180 mg,550 μmol, 69%). M/z 328.1 (M+H) + (ES+)。
Step 2: synthesis of 6-hydroxy-4- (trifluoromethyl) isoindolin-1-one (ACN-2)
To a solution of the product from step 1 above (ACN-1) (400 mg,1 eq, 1.22 mmol) in THF (3 mL) and hydrogen peroxide (1 mL,30 wt%) at room temperature was added acetic acid (1 mL). The resulting mixture was stirred at room temperature for 2 hours. The mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography eluting with DCM/MeOH (15:1) to give the sub-title compound (ACN-2) as an off-white solid (100 mg,460 μmol, 34%). M/z 218.0 (M+H) + (ES+)。
Step 3: synthesis of 6- (2- (pyrrolidin-1-yl) ethoxy) -4- (trifluoromethyl) isoindolin-1-one (ACN-4)
To a stirred solution of the product (ACN-2) (100 mg,1 eq, 461 μmol), epothilone (ACN-3) (265 mg,5 eq, 2.31 mmol) and triphenylphosphine (362 mg,1.38mmol,3 eq) from step 2 above in THF (15 mL) under a nitrogen atmosphere at room temperature was added DIAD (186 mg,2 eq, 922 μmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. The mixture was acidified with HCl (aqueous, 1M) to pH 5. The resulting mixture was extracted with DCM (2X 40 mL).The combined aqueous layers were spin dried. The crude product was purified by preparative HPLC (column: xselect CSH OBD column 30 x 150mm 5um, n; mobile phase A: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 2% B to 23% B over 7 min; wavelength: 254/220 nm) to give the sub-title compound (ACN-4) as an off-white solid (70 mg, 223. Mu. Mol, 44%). M/z 315.1 (M+H) + (ES+)。
Step 4: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- (2- (pyrrolidin-1-yl) ethoxy) -4- (trifluoromethyl) isoindolin-1-one (ACN-5)
To a stirred solution of the product (ACN-4) (40 mg, 127. Mu. Mol,1 equivalent), potassium phosphate (81 mg,3 equivalent, 381. Mu. Mol) and intermediate (ABC-6) (40 mg,1 equivalent, 127. Mu. Mol) from step 3 above in 1, 4-dioxane (5 ml) under nitrogen at room temperature was added XantPhos (15 mg,0.2 equivalent, 25. Mu. Mol) and Pd 2 (dba) 3 (12 mg,0.1 eq, 13. Mu. Mol). The final reaction mixture was irradiated with microwave radiation at 100 ℃ overnight under nitrogen atmosphere. The mixture was cooled to room temperature and acidified to pH 5 with HCl (aqueous, 1M). The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC (column: xselect CSH OBD column 30X 150mm 5um, n; mobile phase A: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 2% B to 23% B over 7 min; wavelength: 254/220 nm) to give the title compound (ACN-5) (13.2 mg, 22. Mu. Mol, 17%) as a pale yellow solid. M/z 596.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.47(s,1H),7.89(s,1H),7.56(s,3H),6.92(t,J=5.4Hz,1H),6.25(d,J=1.2Hz,1H),5.13(d,J=1.8Hz,2H),4.27(t,J=5.7Hz,2H),3.90(s,3H),3.46(s,3H),3.32–3.19(m,2H),2.85(t,J=5.7Hz,2H),2.62–2.53(m,4H),1.83–1.54(m,4H),1.17(t,J=7.1Hz,3H)。
Example 102: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ACO-1)
To intermediate (AAN-7) (80 mg,1 eq, 169. Mu. Mol), THF (422. Mu.L, 2M,5 eq, 845. Mu. Mol) in ethylamine-containing solution and Cs under nitrogen at room temperature 2 CO 3 (165 mg,3 eq, 500. Mu. Mol) Pd was added to a solution in 1, 4-dioxane (5 mL) 2 (dba) 3 (31 mg, 34. Mu. Mol,0.2 eq.) and XantPhos (39 mg,0.4 eq., 68. Mu. Mol). After stirring overnight at 80 ℃ under nitrogen atmosphere, the resulting mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by column chromatography on silica gel eluting with DCM/MeOH (10/1) and preparative HPLC under the following conditions: column: sunfire preparation type C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: ACN; flow rate: 60 ml/min; gradient: 30% B to 50% B in 9 minutes; a detector, 254/220nm; retention time: 8.77. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ACO-1) (5.5 mg,11 μmol, 6.5%) as a white solid. M/z 483.1 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.50(s,1H),8.09(d,J=7.6Hz,1H),7.99(d,J=7.6Hz,1H),7.87(s,1H),7.79–7.75(m,1H),7.66(d,J=1.2Hz,1H),6.20(d,J=1.3Hz,1H),5.28(s,2H),4.04(s,3H),3.50(s,3H),3.40–3.36(m,2H),1.25(t,J=7.2Hz,3H)。
Example 103: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((2-hydroxyethoxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (ACP-3)
Step 1: synthesis of 6- (chloromethyl) -2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ACP-1)
Under nitrogen atmosphere at 0 deg.CTo a stirred solution of intermediate (ACH-1) (50 mg,1 eq, 98. Mu. Mol) in DCM (10 mL) was added thionyl chloride (35 mg,3 eq, 294. Mu. Mol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 1 hour. The reaction was quenched with MeOH (3 mL) at 0 ℃ and concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (8/1) to give the sub-title compound (ACP-1) (31 mg, 58. Mu. Mol, 60%) as an off-white solid. M/z 531.2/533.2 (M+H) + (ES+)。
Step 2: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((2-hydroxyethoxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (ACP-3)
A solution of ethylene glycol (ACP-2) (18 mg,5 eq, 290. Mu. Mol) and sodium hydride (0.5 mg,60Wt%, 20. Mu. Mol,0.2 eq) in THF (10 mL) was stirred under nitrogen at 0deg.C for 30 min. The product (ACP-1) (31 mg,1 equivalent, 58. Mu. Mol) from step 1a was added to the above mixture at 0℃over 30 minutes, and the resulting mixture was stirred for another 1 hour. The reaction was then quenched by the addition of ice water (2 mL) at 0 ℃. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (6/1). The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (10% ACN up to 35% within 20 minutes); detector, UV 254/220nm. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 30% B to 38% B in 10 minutes; wavelength: 254/220nm to give the title compound (ACP-3) (4.5 mg, 7.8. Mu. Mol, 13%) as a white solid. M/z 577.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.02(d,J=16.4Hz,2H),7.89(s,1H),7.58(d,J=1.2Hz,1H),6.93(t,J=5.5Hz,1H),6.25(d,J=1.2Hz,1H),5.21(s,2H),4.73-4.71(m,3H),3.90(s,3H),3.63-3.49(m,4H),3.47(s,3H),3.32-3.22(m,2H),1.17(t,J=7.1Hz,3H)。
Example 104: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6-hydroxy-4- (trifluoromethyl) isoindolin-1-one (ACQ-7)
Step 1: synthesis of methyl 2-methyl-5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3- (trifluoromethyl) benzoate (ACQ-1)
Pd (dppf) Cl was added to a stirred solution of intermediate (B-1) (800 mg,1 eq, 2.69 mmol), intermediate (AAI-3) (752 mg,1.1 eq, 2.96 mmol) and potassium acetate (793 mg,3 eq, 8.08 mmol) in dioxane (15 mL) at room temperature under nitrogen atmosphere 2 DCM (219 mg,0.1 eq., 269. Mu. Mol). The resulting mixture was stirred under nitrogen at 100 ℃ for 16 hours. The mixture was cooled to room temperature and the resulting mixture was filtered; the filter cake was washed with petroleum ether (3×5 mL). The filtrate was concentrated in vacuo to give the crude sub-title compound (ACQ-1) (900 mg,2.61mmol, 96%) which was used directly in the next step without further purification. M/z 345.1 (M+H) + (ES+)。
Step 2: synthesis of methyl 5-hydroxy-2-methyl-3- (trifluoromethyl) benzoate (ACQ-2)
To a 25mL round bottom flask was added the product from step 1 above (ACQ-1) (890 mg,1 eq, 2.59 mmol) and acetic acid (6 mL) at room temperature. Hydrogen peroxide (3 mL,30 wt%) and THF (6 mL) were added to the above mixture at 0 ℃. The resulting mixture was stirred at room temperature for an additional 2 hours. The resulting mixture was extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using petroleum ether/EtOAc (10/1) to give the sub-title compound (ACQ-2) as an off-white solid (480 mg,2.05mmol, 79%). M/z 235.1 (M+H) + (ES+)。
Step 3: synthesis of methyl 5- ((tert-butyldimethylsilyl) oxy) -2-methyl-3- (trifluoromethyl) benzoate (ACQ-3)
A solution of the product from step 2 above (ACQ-2) (200 mg,1 eq, 854. Mu. Mol), TBSCl (154 mg,1.2 eq, 1.03 mmol) and imidazole (140 mg,2.4 eq, 2.05 mmol) in DMF (10 mL) was stirred at room temperature overnight. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The residue was purified by preparative TLC using petroleum ether/EtOAc (20/1) to give the sub-title compound (ACQ-3) as a colorless oil (210 mg,603 μmol, 71%). M/z 349.1 (M+H) + (ES+)。
Step 4: synthesis of methyl 2- (bromomethyl) -5- ((tert-butyldimethylsilyl) oxy) -3- (trifluoromethyl) benzoate (ACQ-4)
A solution of the product from step 3 above (ACQ-3) (100 mg, 287. Mu. Mol,1 eq.), NBS (77 mg,1.5 eq., 430. Mu. Mol) and benzoyl peroxide (22 mg,0.3 eq., 86. Mu. Mol) in carbon tetrachloride (5 mL) was stirred overnight at 80 ℃. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using petroleum ether/EtOAc (20/1) to give the sub-title compound (ACQ-4) as a brown/yellow solid (100 mg,234 μmol, 82%). M/z 427.0/429.0 (M+H) + (ES+)。
Step 5: synthesis of 6-hydroxy-4- (trifluoromethyl) isoindolin-1-one (ACQ-5)
A solution of the product from step 4 above (ACQ-4) (100 mg,1 eq, 234. Mu. Mol) in MeOH containing ammonia (5 mL, 7M) was stirred at room temperature for 2 hours. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1) to give the sub-title compound (ACQ-5) as an off-white solid (50 mg, 230. Mu. Mol, 98%). M/z 218.0 (M+H) + (ES+)。
Step 6: synthesis of 6- ((tert-butyldimethylsilyl) oxy) -4- (trifluoromethyl) isoindolin-1-one (ACQ-6)
A solution of the product from step 5 above (ACQ-5) (50 mg,1 eq, 230. Mu. Mol), TBSCl (42 mg,1.2 eq, 276. Mu. Mol) and imidazole (38 mg,2.4 eq, 552. Mu. Mol) in DMF (5 mL) was stirred at room temperature overnight. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (0% ACN up to 100% within 24 minutes); detector, UV 254/220nm. This gave the sub-title compound (ACQ-6) (60 mg, 181. Mu. Mol, 79%) as a colorless oil. M/z 332.1 (M+H) + (ES+)。
Step 7: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6-hydroxy-4- (trifluoromethyl) isoindolin-1-one (ACQ-7)
Pd was added to a solution of the product (ACQ-6) (50 mg,0.151mmol,1.0 eq.) from step 6 above and intermediate (ABC-6) (43 mg,0.9 eq., 136. Mu. Mol) and potassium phosphate (19 mg,3 eq., 90. Mu. Mol) in dioxane (10 mL) at room temperature under nitrogen atmosphere 2 (dba) 3 (3 mg,0.1 eq, 3. Mu. Mol) and Xantphos (17 mg,0.2 eq, 30. Mu. Mol). The resulting mixture was stirred under nitrogen at 100 ℃ for 4 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (0% ACN up to 50% within 24 minutes); detector, UV 254/220nm. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 35% B to 60% B in 8 minutes; wavelength: 254/220nm to give the title compound (ACQ-7) (5.4 mg, 10.8. Mu. Mol, 7.1%) as a white solid. M/z 499.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ10.64(s,1H),8.48(s,1H),7.88(s,1H),7.56(s,1H),7.35(s,2H),6.91(t,J=5.5Hz,1H),6.23(s,1H),5.09(s,2H),3.89(s,3H),3.46(s,3H),3.31–3.25(m,2H),1.16(t,J=7.1Hz,3H)。
Example 105: synthesis of 2- (4- (4-chloro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (ethylamino) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ACR-7)
Step 1: synthesis of 5-chloro-2- (2, 6-dichloropyridin-4-yl) benzoic acid (ACR-2)
Into a 250-mL round bottom flask purged with an inert nitrogen atmosphere and maintained was placed dioxane (50 mL) and water (10 mL) containing 2-bromo-5-chlorobenzoic acid (ACR-1) (5.00 g,1 equivalent, 21.2 mmol), intermediate (AAN-1) (4.07 g,1 equivalent, 21.2 mmol) and potassium phosphate (13.52 g,3 equivalent, 63.7 mmol), then Pd (DtBPF) Cl was added at room temperature under a nitrogen atmosphere 2 (1.38 g,0.1 eq, 2.12 mmol). The resulting solution was stirred overnight at 60 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And ACN (36% ACN up to 48% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ACR-2) (1.8 g,5.95mmol, 28%) as a brown solid. M/z 302.9/304.9 (M+H) + (ES+)。
Step 2: synthesis of 2- (5-chloro-2- (2, 6-dichloropyridin-4-yl) benzoyl) -N-methylhydrazine-1-thiocarboxamide (ACR-3).
Into a 100-mL round bottom flask purged with an inert nitrogen atmosphere and maintained at room temperature was placed DMF (40 mL) containing the product (ACR-2) (1.80 g,1 eq, 5.95 mmol) from step 1 above and DIPEA (2.31 g,3 eq, 17.9 mmol) and then 4-methyl-3-thiosemicarbazide (D-2) (630 mg,1 eq, 5) was added at room temperature95 mmol) and T3P (4.73 g,2.5 eq, 14.9 mmol). The resulting solution was stirred at room temperature for 6 hours, and the resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (60% ACN up to 85% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ACR-3) as a brown/yellow solid (720 mg,1.85mmol, 30%). M/z 389.0/391.0 (M+H) + (ES+)。
Step 3: synthesis of 5- (5-chloro-2- (2, 6-dichloropyridin-4-yl) phenyl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (ACR-4)
A50-mL round-bottomed flask was charged with a NaOH solution (aqueous solution, 1M) (20 mL) containing the product (ACR-3) (650 mg,1.67mmol,1 eq.) from step 2 above at room temperature. The resulting solution was stirred at room temperature for 4 hours. The resulting solution was concentrated in vacuo to give the sub-title compound (ACR-4) (620 mg,1.61mmol, 96%) as a brown/yellow solid, which was used directly in the next step without any further purification. M/z 371.0/373.0 (M+H) + (ES+)。
Step 4: synthesis of 2, 6-dichloro-4- (4-chloro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridine (ACR-5).
A50 mL round bottom flask purged with an inert nitrogen atmosphere and maintained at room temperature was charged with DCM (20 mL) containing the product (ACR-4) from step 3 above (600 mg,1 eq, 1.61 mmol). Acetic acid (194 mg,2 equivalents, 3.23 mmol) and hydrogen peroxide solution (290 mg,30wt%,5 equivalents, 8.05 mmol) were added at room temperature, and the resulting solution was stirred for 4 hours. The resulting mixture was diluted with water and extracted with DCM (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. This gave the sub-title compound (ACR-5) as a brown/yellow solid (500 mg,1.47mmol, 91%). M/z 339.0/341.0 (M+H) + (ES+)。
Step 5: synthesis of 6-chloro-4- (4-chloro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -N-ethylpyridin-2-amine (ACR-6)
A100-mL round bottom flask was charged with NMP (10 mL) containing the product from step 4 above (ACR-5) (500 mg,1 eq, 1.47 mmol) and ethylamine, HCl (664 mg,10 eq, 14.7 mmol) at room temperature, then K was added at room temperature under a nitrogen atmosphere 2 CO 3 (2.03 g,10 equivalents, 14.7 mmol). The resulting solution was stirred under nitrogen at 100 ℃ for 16 hours. The mixture was cooled to room temperature and the crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (20% ACN up to 43% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ACR-6) (480 mg,1.38mmol, 94%) as a white solid. M/z348.1/350.1 (M+H) + (ES+)。
Step 6:2- (4- (4-chloro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (ethylamino) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ACR-7)
1, 4-dioxane (3 mL) containing the product (ACR-6) (70 mg,1 eq, 201. Mu. Mol), intermediate (AC-2) (40 mg,1 eq, 201. Mu. Mol) and potassium phosphate (128 mg,3 eq, 603. Mu. Mol) from step 5 above was placed in a 25-mL round bottom flask under nitrogen at room temperature, then Pd was added 2 (dba) 3 -CHCl 3 (42 mg,0.2 eq, 40. Mu. Mol) and Xantphos (47 mg,0.4 eq, 80. Mu. Mol). The resulting solution was stirred overnight at 100 ℃ under nitrogen atmosphere. The resulting mixture was cooled to room temperature and concentrated. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared C18 OBD column, 19×150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: ACN; flow rate: 25 ml/min; gradient: 50B to 65B in 9 minutes; a detector, UV 210/254nm; retention time: 6.67. this gave the title compound (ACR-7) (10.6 mg, 21. Mu. Mol, 10%) as a white solid. M/z 513.1 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.47(s,1H),8.08(d,J=7.7Hz,1H),7.97(d,J=7.8Hz,1H),7.81–7.66(m,4H),7.52(d,J=1.3Hz,1H),6.03(d,J=1.3Hz,1H),5.23(s,2H),3.44(s,3H),3.29–3.26(m,2H),1.22(t,J=7.2Hz,3H)。
Example 106: synthesis of (S) -2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- (3-hydroxypyrrolidine-1-carbonyl) -4- (trifluoromethyl) isoindolin-1-one (ACS-4)
Step 1: synthesis of 3-oxo-7- (trifluoromethyl) isoindoline-5-carboxylic acid (ACS-1)
DMF (10 mL) containing intermediate (ABC-2) (300 mg,1 equivalent, 1.07 mmol), oxalic acid (145 mg,1.5 equivalent, 1.60 mmol), DIPEA (208 mg,1.5 equivalent, 1.61 mmol) and acetic anhydride (164 mg,1.5 equivalent, 1.60 mmol) was added to a 40mL sealed tube at room temperature. Pd (OAc) was added to the above mixture over 3 minutes at room temperature under nitrogen atmosphere 2 (24 mg,0.1 eq, 107. Mu. Mol) and XantPhos (124 mg,0.2 eq, 210. Mu. Mol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere before cooling to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% fa) and ACN (36% ACN up to 48% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ACS-1) (120 mg, 489. Mu. Mol, 46%) as a yellow oil. M/z 246.0 (M+H) + (ES+)。
Step 2: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -3-oxo-7- (trifluoromethyl) isoindoline-5-carboxylic acid (ACS-2)
1, 4-dioxane (5 mL) containing the product from step 1 above (ACS-1) (100 mg,1 eq, 408. Mu. Mol), intermediate (ABC-6) (143 mg,1.1 eq, 449. Mu. Mol) and potassium phosphate (260 mg,3 eq, 1.22 mmol) was added to a 20mL sealed tube at room temperature. At room temperature under nitrogen atmosphereNext, pd (OAc) was added to the above mixture over 3 minutes 2 (9 mg,0.1 eq, 41. Mu. Mol) and XantPhos (47 mg, 82. Mu. Mol,0.2 eq) and the resulting mixture was stirred under nitrogen at 100℃for 3 hours. The mixture was cooled to room temperature and the crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (36% ACN up to 48% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ACS-2) (60 mg, 114. Mu. Mol, 28%) as a yellow oil. M/z 527.2 (M+H) + (ES+)。
Step 3:2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- [ (3S) -3-hydroxypyrrolidine-1-carbonyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (ACS-4)
DMF (2 mL) containing the product from step 2 above (ACS-2) (50 mg,1 eq, 95. Mu. Mol), (3S) -pyrrolidin-3-ol (ACS-3) (10 mg,1.2 eq, 114. Mu. Mol) and DIPEA (37 mg,3 eq, 285. Mu. Mol) was added to an 8mL sealed tube at room temperature. HATU (54 mg,1.5 eq, 143 μmol) was added to the above mixture at 0 ℃ over 5 min, and the resulting mixture was stirred at room temperature for 3 hours. The mixture was concentrated in vacuo and the crude product was purified by prep HPLC under the following conditions (column: XBridge prep OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 25% B to 36% B in 9 minutes; wavelength: 254/220nm; retention time: 8.9 To give the title compound (ACS-4) (19.2 mg,32 μmol, 34%) as a white solid. M/z 596.4 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),8.23(d,J=7.7Hz,1H),8.15(d,J=4.5Hz,1H),7.87(s,1H),7.65(d,J=1.2Hz,1H),6.22(d,J=1.3Hz,1H),5.33(s,2H),4.54–4.42(m,1H),4.04(s,3H),3.88–3.52(m,4H),3.51(s,3H),3.42–3.35(m,3H),2.21–1.94(m,2H),1.25(t,J=7.2Hz,3H)。
Example 107: synthesis of (R) -2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- (3-hydroxypyrrolidine-1-carbonyl) -4- (trifluoromethyl) isoindolin-1-one (ACT-2)
DMF (2 mL) containing intermediate (ACS-2) (50 mg,1 eq, 95. Mu. Mol), (3R) -pyrrolidin-3-ol (ACT-1) (10 mg,1.2 eq, 114. Mu. Mol) and DIPEA (37 mg,3 eq, 285. Mu. Mol) was added to an 8mL sealed tube at room temperature. HATU (54 mg,143 μmol,1.5 eq.) was added to the above mixture at 0 ℃ over 5 minutes, and the resulting mixture was stirred at room temperature for 3 hours. The mixture was concentrated in vacuo and the crude product was purified by prep HPLC under the following conditions (column: XBridge prep OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 25% B to 36% B in 9 minutes; wavelength: 254/220nm; retention time: 8.8 To give the title compound (ACT-2) (18.4 mg, 31. Mu. Mol, 33%) as a white solid. M/z 596.4 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),8.23(d,J=7.6Hz,1H),8.16(d,J=4.5Hz,1H),7.87(s,1H),7.66(d,J=1.3Hz,1H),6.22(d,J=1.2Hz,1H),5.34(d,J=1.7Hz,2H),4.54–4.43(m,1H),4.04(s,3H),3.86–3.52(m,4H),3.51(s,3H),3.43–3.35(m,3H),2.22–1.97(m,2H),1.25(t,J=7.2Hz,3H)。
Example 108: synthesis of 2- (6- (1- (hydroxymethyl) cyclopropyl) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ACU-2)
Step 1: synthesis of 1- (4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (1-oxo-4- (trifluoromethyl) isoindolin-2-yl) pyridin-2-yl) cyclopropane-1-carboxylic acid (ACU-1)
Intermediate (ACJ-4) (60 mg,1 eq, 104. Mu. Mol) in DCM (3 mL) and TFA (1 mL)Is stirred at room temperature for 2 hours. The resulting mixture was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 520.2 (M+H) + (ES+)。
Step 2:2- (6- (1- (hydroxymethyl) cyclopropyl) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (ACU-2)
CDI (7 mg,1.3 eq, 46. Mu. Mol) was added to a stirred solution of the product from step 1 above (ACU-1) (20 mg,1 eq, 35. Mu. Mol) in THF (1 mL) at room temperature. After 30 minutes, the mixture was added to NaBH at 0℃under a nitrogen atmosphere 4 (7 mg, 175. Mu. Mol,5 eq.) in THF (1 mL) and water (0.5 mL). The resulting mixture was stirred at room temperature for 2 hours, then concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 42% B to 67% B in 8 minutes; wavelength: 254/220nm; retention time: 6.62 To give the title compound (ACU-2) (3 mg,5.9 μmol, 17%) as a white solid. M/z 506.0 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.47(s,1H),8.18(d,J=1.3Hz,1H),8.11(d,J=7.7Hz,1H),8.00(d,J=7.7Hz,1H),7.85–7.76(m,3H),7.73–7.64(m,2H),7.08(d,J=1.4Hz,1H),5.30–5.25(m,2H),3.87(s,2H),3.45(s,3H),1.22(m,2H),1.01(m,2H)。
Example 109: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((1-methylazetidin-3-yl) oxy) -4- (trifluoromethyl) isoindolin-1-one (ACV-2)
To a stirred solution of intermediate (ACQ-7) (40 mg,1 eq, 80. Mu. Mol) and 1-methylazetidin-3-ol (ACV-1) (28 mg,4 eq, 320. Mu. Mol) in THF (10 mL) at room temperature was added triphenylAlkylphosphines (105 mg,5 eq, 400. Mu. Mol) and DIAD (81 mg,5 eq, 400. Mu. Mol). The resulting mixture was stirred at room temperature overnight and then acidified to pH 3 with HCl (aqueous solution, 4M). The resulting mixture was diluted with water and extracted with DCM (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ) Filtered and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 37% B to 44% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (ACV-2) (3.3 mg, 5.8. Mu. Mol, 7%) as an off-white solid. M/z 568.4 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.87(s,1H),7.52(d,J=1.2Hz,1H),7.48(d,J=2.3Hz,1H),7.35(d,J=2.3Hz,1H),6.24(d,J=1.2Hz,1H),5.16–5.07(m,2H),5.06–4.94(m,1H),3.89(s,3H),3.81–3.72(m,2H),3.43(s,3H),3.25(q,J=7.2Hz,2H),3.19–3.07(m,2H),2.33(s,3H),1.15(t,J=7.2Hz,3H)。
Example 110: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- (isopropoxymethyl) -4- (trifluoromethyl) isoindolin-1-one (ACW-1)
NaOiPr (32 mg, 395. Mu. Mol,5 eq.) was added to a stirred solution of intermediate (ACP-1) (42 mg, 79. Mu. Mol,1 eq.) in iPrOH (5 mL) at room temperature. The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of ice water (3 mL) at 0 ℃. The mixture was concentrated in vacuo and the residue was purified by TLC with DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 52% B to 62% B in 8 minutes; wavelength: 254/220nm) to give the title compound (ACW-1) (16.8 mg,30 μmol, 38%) as a white solid. M/z 555.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.47(s,1H),8.00-7.95(m,2H),7.89(s,1H),7.57(d,J=1.2Hz,1H),6.93(t,J=5.3Hz,1H),6.25(d,J=1.3Hz,1H),5.21(s,2H),4.67(s,2H),3.90(s,3H),3.75-3.66(m,1H),3.47(s,3H),3.31-3.23(m,2H),1.22-1.13(m,9H)。
Example 111: synthesis of (R) -2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((3-hydroxypyrrolidin-1-yl) methyl) -4- (trifluoromethyl) isoindolin-1-one (AT-1)
NaBH (OAc) 3 (75 mg,6.0 eq, 0.35 mmol) was added to intermediate AE-1 (30 mg,99Wt%,1 eq, 59. Mu. Mol) and (R) -pyrrolidin-3-ol (ACU-1) (20 mg,4.0 eq, 0.23 mmol) in CHCl 3 (2 mL) and stirred at room temperature overnight. The reaction mixture was diluted with DCM (4 mL) and NaHCO 3 Is washed with a saturated aqueous solution (10 mL). The organic extract was dried (phase separator) and concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18ODB, 5 μm, 30x100mm column 0-100% MeCN/water) to give the title compound (AT-1) as a pale yellow solid (16 mg,26 μmol,44%,94% purity). M/z 576.3 (M-H) - (ES-). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.97(s,1H),7.92(s,1H),7.71(td,J=7.4,1.6Hz,1H),7.65–7.53(m,3H),7.48(d,J=1.2Hz,1H),6.69(t,J=5.5Hz,1H),5.94(d,J=1.2Hz,1H),5.16(s,2H),4.72(d,J=4.3Hz,1H),4.23–4.19(m,1H),3.82–3.72(m,2H),3.33(s,3H),3.21–3.12(m,2H),2.73–2.58(m,2H),2.48–2.39(m,1H),2.39–2.32(m,1H),2.08–1.95(m,1H),1.61–1.53(m,1H),1.13(t,J=7.1Hz,3H)。
Example 112: synthesis of (R) -6- ((4, 4-difluoro-3-methylpiperidin-1-yl) methyl) -2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AU-2)
NaBH (OAc) 3 (94 mg,6.0 eq, 0.44 mmol) was added to a stirred solution of intermediate AE-1 (30 mg,99Wt%,0.79 eq, 59. Mu. Mol), (R) -4, 4-difluoro-3-methylpiperidine (AU-1) (10 mg,1 eq, 74. Mu. Mol) and DIPEA (29 mg, 38. Mu.L, 3.0 eq, 0.22 mmol) in DCM (2 mL) and stirred at room temperature overnight. The reaction mixture was diluted with DCM (5 mL) and NaHCO 3 Is washed with a saturated aqueous solution (6 mL). The organic layer was separated and concentrated in vacuo. The crude product was purified by RP flash C18 chromatography (12 g cartridge, 15-80% MeCN/10mM ammonium bicarbonate) to give the title compound (AU-2) (4.5 mg,7.1 μmol,9.6%,99% purity) as a white solid. M/z 626.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.99(s,1H),7.94(s,1H),7.71(td,J=7.5,1.7Hz,1H),7.64–7.54(m,3H),7.48(d,J=1.2Hz,1H),6.69(t,J=5.5Hz,1H),5.94(d,J=1.2Hz,1H),5.16(s,2H),3.74(d,J=2.5Hz,2H),3.29(s,3H),3.22–3.12(m,2H),2.80–2.68(m,2H),2.36–1.84(m,5H),1.13(t,J=7.2Hz,3H),0.92(d,J=6.4Hz,3H)。
Example 113: synthesis of 6- ((((1R, 2S) -2-hydroxycyclopentyl) oxy) methyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AV-3)
Step 1: synthesis of 6- (chloromethyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] 3-yl) -4- (trifluoromethyl) isoindolin-1-one (AV-1)
A stirred solution of intermediate (P-2) (35 mg,1 eq, 75. Mu. Mol) in DCM (2 mL) was treated with SOCl 2 (22 mg, 14. Mu.L, 2.5 eq, 0.19 mmol) was treated dropwise. The resulting suspension was stirred at room temperature for 30 minutes. The volatiles were removed in vacuoAnd the resulting crude subtitle compound (AV-1) was used as a solution without further purification. 1 H NMR(400MHz,DMSO-d6)δ9.05(s,1H),8.15(d,J=4.9Hz,2H),7.96–7.89(m,1H),7.85–7.78(m,1H),7.76–7.64(m,4H),7.44(t,J=8.0Hz,1H),7.01(d,J=7.7Hz,1H),5.17(s,2H),5.02(s,2H),3.29(s,3H)。
Step 2: synthesis of 6- ((((1R, 2S) -2-hydroxycyclopentyl) oxy) methyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AV-3)
To a vial containing the product from step 1 (AV-1) (20 mg,1 eq, 41 μmol), TBAB (6.7 mg,0.5 eq, 21 μmol) and (1 r,2 s) -cyclopentane-1, 2-diol (AV-2) (21 mg,5 eq, 0.21 mmol) was added DCM (2 mL). A solution of KOH (aqueous solution) (12 mg,1mL,20wt%,1 eq, 41. Mu. Mol) was added and the reaction was kept at room temperature for 2 hours without stirring. The reaction was diluted with water (10 mL) and extracted with EtOAc (3×10 mL). The organics were combined, dried (MgSO 4 ) Filtered and evaporated. The crude product was purified by RP flash C18 chromatography (12 g cartridge, 15-65% MeCN/10mM ammonium bicarbonate) to give the title compound (AV-3) (3 mg,5 μmol,10%,99% purity) as a white solid. M/z 549.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.39(s,1H),8.05(d,J=17.9Hz,2H),8.00(dd,J=8.1,2.3Hz,1H),7.75–7.65(m,3H),7.60–7.56(m,2H),7.38(t,J=8.0Hz,1H),6.93–6.88(m,1H),5.10(s,2H),4.80(d,J=12.7Hz,1H),4.70(d,J=12.8Hz,1H),4.49(d,J=4.8Hz,1H),4.05–3.98(m,1H),3.72(td,J=6.2,3.8Hz,1H),3.08(s,3H),1.82–1.65(m,4H),1.62–1.53(m,1H),1.48–1.40(m,1H)。
Example 114: synthesis of 6- ((((1-hydroxycyclobutyl) methyl) amino) methyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AW-2)
NaBH (OAc) 3 (82 mg,6.0 eq, 0.39 mmol)) To a stirred solution of intermediate (Q-1) (30 mg,99Wt%,1 eq, 64. Mu. Mol), 1- (aminomethyl) cyclobutan-1-ol (AW-1) (7.8 mg,1.2 eq, 77. Mu. Mol) and DIPEA (25 mg, 34. Mu.L, 3 eq, 0.19 mmol) in DCM (5 mL) was added and stirred at room temperature overnight. Adding NaBH 4 (7.3 mg,3 equivalents, 0.19 mmol) and the reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with DCM (5 mL) and NaHCO 3 Is washed with a saturated aqueous solution (6 mL). The organic layer was separated, dried (phase separator) and concentrated in vacuo. The crude product was purified by RP flash C18 chromatography (12 g cartridge, 25-55% MeCN/10mM ammonium bicarbonate) to give the title compound (AW-2) (5 mg,9 μmol,10%,95% purity) as a white solid. M/z 548.2 (M+H) + (ES+). 1 H NMR (400 MHz, DMSO-d 6) delta 8.39 (s, 1H), 8.05 (s, 1H), 8.03-7.97 (m, 2H), 7.75-7.65 (m, 3H), 7.61-7.55 (m, 2H), 7.37 (t, J=8.0 Hz, 1H), 6.90 (dt, J=7.7, 1.3Hz, 1H), 5.08 (s, 2H), 4.93 (s, 1H), 3.96 (s, 2H), 3.08 (s, 3H), 2.04-1.96 (m, 2H), 1.96-1.85 (m, 2H), 1.67-1.55 (m, 1H), 1.44-1.32 (m, 1H). Three protons are masked by DMSO peaks.
Example 115: synthesis of 6- ((((1-hydroxycyclobutyl) methyl) amino) methyl) -2-phenyl-4- (trifluoromethyl) isoindolin-1-one (AX-3)
Step 1: synthesis of 6- (hydroxymethyl) -2-phenyl-4- (trifluoromethyl) isoindolin-1-one (AX-1)
A solution of intermediate (B-4) (135 mg,1 eq, 413. Mu. Mol) in aniline (500 mg, 489. Mu.L, 13 eq, 5.37 mmol) was stirred at 100℃for 30 minutes (microwave). The reaction mixture was then allowed to stand at room temperature overnight. The reaction mixture was diluted with EtOAc (10 mL) and washed with 1M HCl (aq) (2×10 mL). The organic layer was diluted with DCM (10 mL), passed through a phase separator and concentrated in vacuo to give the sub-title compound (AX-1) (84 mg,0.26mmol,63%,95% purity) as a flocculent off-white solid. m/z 305.7 &307.2(M-H) - (ES-). 1 H NMR(400MHz,DMSO-d6)δ8.02(s,1H),7.98–7.88(m,3H),7.50–7.41(m,2H),7.26–7.17(m,1H),5.59(t,J=5.8Hz,1H),5.20(d,J=1.8Hz,2H),4.71(d,J=5.8Hz,2H)。
Step 2: synthesis of 3-oxo-2-phenyl-7- (trifluoromethyl) isoindoline-5-carbaldehyde (AX-2)
Manganese dioxide (0.68 g,30 equivalents, 7.8 mmol) was added to CHCl containing product (AX-1) from step 1 above (84 mg,95Wt%,1 equivalent, 0.26 mmol) 3 (10 mL) and stirred at room temperature throughout the weekend. The reaction mixture was filtered through a celite plug (substituted with 10mL MeOH). The filtrate was evaporated to give the sub-title compound (AX-2) (74 mg,0.22mmol,86%,92% purity) as a pale yellow solid. 1 H NMR(400MHz,DMSO-d6)δ10.22(s,1H),8.56(s,1H),8.49(s,1H),8.00–7.92(m,2H),7.52–7.43(m,2H),7.29–7.19(m,1H),5.35(s,2H)。
Step 3: synthesis of 6- ((((1-hydroxycyclobutyl) methyl) amino) methyl) -2-phenyl-4- (trifluoromethyl) isoindolin-1-one (AX-3)
The product (AX-2) (20 mg,99Wt%,1 equivalent, 65. Mu. Mol), 1-aminomethyl cyclobutanol (AW-1) (6.6 mg,1 equivalent, 65. Mu. Mol) and DIPEA (25 mg, 33. Mu.L, 3.0 equivalent, 0.19 mmol) from step 2 above were reacted with CHCl 3 The mixture in (2 mL) was stirred at room temperature for 5 min. NaBH (OAc) is then added 3 (82 mg,6.0 eq, 0.39 mmol) and the resulting suspension was stirred at room temperature overnight. The reaction temperature was raised to 40 ℃ and stirring was continued overnight. Adding NaBH 4 (10 mg,4.1 eq, 0.26 mmol) and stirring was continued at room temperature overnight. The reaction mixture was diluted with DCM (4 mL) and NaHCO 3 Saturated aqueous (10 mL) was washed. The organic phase was separated and evaporated to give the crude product. The crude product was purified by preparative HPLC (Woltzian, basic (0.3% ammonia), basic, woltzian XBridge BEH C18 ODB column,Purification on a 5 μm, 30x100mm column, 30-100% MeCN/water) afforded the title compound (AX-3) (6 mg,0.01mmol,20%,94% purity) as a white solid. M/z 391.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.06(s,1H),8.01(s,1H),7.97–7.90(m,2H),7.51–7.42(m,2H),7.26–7.17(m,1H),5.20(s,2H),4.93(s,1H),3.96(s,2H),3.29(s,1H),2.53(s,2H),2.05–1.84(m,4H),1.65–1.55(m,1H),1.44–1.33(m,1H)。
Example 116: synthesis of 6-bromo-2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AY-1)
DIPEA (258 mg, 347. Mu.L, 1.5 eq, 1.99 mmol) was added to a solution of intermediate P-1 (365 mg,1.1 eq, 1.46 mmol) and intermediate ABC-1 (500 mg,1 eq, 1.33 mmol) in benzotrifluoride (3 mL) and the reaction stirred at 60℃for 16 h. The volatiles were evaporated and the crude product was dissolved in EtOH (20 mL) and stirred at 60 ℃ for 16 hours. The reaction mixture was concentrated in vacuo and the crude product was purified by RP flash C18 chromatography (40 g cartridge, 15-80% MeCN/10mM ammonium hydroxide) to give the title compound (AY-1) as a yellow solid (161 mg,0.30mmol,23%,96% purity). M/z 513/515 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.39(s,1H),8.27(s,1H),8.23(d,J=1.7Hz,1H),7.98(dd,J=8.0,2.2Hz,1H),7.74–7.70(m,1H),7.69–7.66(m,2H),7.62–7.57(m,2H),7.38(t,J=7.9Hz,1H),6.91(d,J=7.8Hz,1H),5.08(s,2H),3.08(s,3H)。
Example 117: synthesis of 6- (((2-hydroxy-2-methylpropyl) amino) methyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (AZ-2)
NaBH (OAc) 3 (82 mg,6.0 eq, 0.39 mmol) was added to intermediate Q-1 (30 mg,99Wt%,1 eq, 64. Mu. Mol), 1-amino-2-methylpropan-2-ol (AZ-1) (6.9 mg,1.2 eq, 77. Mu. Mol) and DIPEA (25 mg, 34. Mu.L, 3 eq, 0.19 mmol) at DIn a stirred solution in CM (5 mL) and stirred at room temperature overnight. Adding NaBH 4 (7.3 mg,3 equivalents, 0.19 mmol) and the reaction mixture was stirred at room temperature overnight. The reaction mixture was diluted with DCM (5 mL) and NaHCO 3 Is washed with a saturated aqueous solution (6 mL). The organic layer was separated, dried (phase separator) and concentrated in vacuo. The crude product was purified by RP flash C18 chromatography (12 g cartridge, 25-55% MeCN/10mM ammonium bicarbonate) to give the title compound (AZ-2) (10 mg,18 μmol,28%,97% purity) as a white solid. M/z 536.1 (M+H) + (ES+). 1 H NMR (400 MHz, DMSO-d 6) delta 8.39 (s, 1H), 8.04 (s, 1H), 8.02-7.96 (m, 2H), 7.75-7.65 (m, 3H), 7.62-7.55 (m, 2H), 7.37 (t, J=8.0 Hz, 1H), 6.91 (dt, J=7.7, 1.2Hz, 1H), 5.08 (s, 2H), 4.23 (s, 1H), 3.93 (s, 2H), 3.08 (s, 3H), 2.36 (s, 2H), 1.11 (s, 6H). No exchangeable protons were observed.
Example 118: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((piperidin-4-yloxy) methyl) -4- (trifluoromethyl) isoindolin-1-one, HCl (BA-4)
Step 1: synthesis of tert-butyl 4- ((3-oxo-7- (trifluoromethyl) isoindolin-5-yl) methoxy) piperidine-1-carboxylate (BA-2)
Will contain Cs 2 CO 3 (0.26G, 4.5 eq, 0.82 mmol) of water (0.4 mL) was added to toluene (3 mL) containing intermediate ABC-2 (54 mg,95Wt%,1 eq, 184. Mu. Mol), ((1- (tert-butoxycarbonyl) piperidin-4-yl) oxy) methyl) potassium trifluoroborate (BA-1) (88 mg,1.5 eq, 0.28 mmol) and RuPhos Pd G3 (15.4 mg,0.1 eq, 18.4. Mu. Mol). The reaction mixture was sparged with nitrogen for 1 minute and stirred at 100 ℃ for 30 minutes (microwaves). The reaction mixture was filtered through a celite plug, washing with MeOH (10 mL). The filtrate was evaporated under reduced pressure and the crude product was purified by RP flash C18 chromatography (24 g cartridge, 0-100% MeCN/10mM ammonium bicarbonate) to give the sub-title compound (BA-2) as a colourless gum (14 mg,33 μmol,18%,99% pure)Degree). M/z 359.2 (M-tBu+H) + (ES+)。
Step 2: synthesis of tert-butyl 4- ((2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -3-oxo-7- (trifluoromethyl) isoindolin-5-yl) methoxy) piperidine-1-carboxylate (BA-3)
Pd-171 (RuPhos Pd (crotyl) Cl) (2.1 mg,0.10 eq., 3.2. Mu. Mol) and RuPhos (1.5 mg,0.10 eq., 3.2. Mu. Mol) were added to a mixture of NaOtBu (3.0 mg,1 eq., 32. Mu. Mol), intermediate AD-1 (10 mg,99Wt%,1 eq., 32. Mu. Mol) and the product (BA-2) (14 mg,99Wt%,1.05 eq., 33. Mu. Mol) from step 1 above in 1, 4-dioxane (1.2 mL) and NMP (0.3 mL). The reaction mixture was sparged with nitrogen for 2 minutes and then stirred at 120 ℃ for 3 hours (microwave). Additional RuPhos (1.5 mg,0.10 eq, 3.2 μmol) and Pd-171 (RuPhos Pd (crotyl) Cl) (2.1 mg,0.10 eq, 3.2 μmol) were added to the reaction mixture, which was sparged with nitrogen for 2 minutes and then stirred at 120 ℃ for an additional 1 hour (microwave). The reaction mixture was filtered and concentrated in vacuo. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the sub-title compound (BA-3) as a colorless solid (10 mg,14 μmol,44%,97% purity). M/z 692.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),8.00(s,1H),7.95(s,1H),7.75–7.67(m,1H),7.66–7.53(m,3H),7.49(d,J=1.2Hz,1H),6.69(t,J=5.4Hz,1H),5.93(d,J=1.3Hz,1H),5.17(s,2H),4.73(s,2H),3.71–3.58(m,2H),3.33(s,3H),3.23–3.13(m,2H),3.07–3.03(m,3H),1.90–1.83(m,2H),1.45(ddd,J=13.0,8.7,4.1Hz,2H),1.39(s,9H),1.13(t,J=7.2Hz,3H)。
Step 3: synthesis of 2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((piperidin-4-yloxy) methyl) -4- (trifluoromethyl) isoindolin-1-one, HCl (BA-4)
HCl (4M in 1, 4-dioxane) (13 mg, 88. Mu.L, 4 moles, 25 equivalents, 0.35 mmol) was added to 1 containing the product (BA-3) from step 2 above (10 mg,97Wt%,1 equivalent, 14. Mu. Mol)4-dioxane (1 mL) and the mixture was stirred at room temperature overnight. The reaction mixture was concentrated in vacuo to give the title compound (BA-4) (9.0 mg,13 μmol,92%,90% purity) as a pale yellow glass. M/z 592.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 9.37 (s, 1H), 8.16 (s, 1H), 8.05 (s, 1H), 7.99-7.92 (m, 1H), 7.91-7.87 (m, 1H), 7.84-7.81 (m, 1H), 7.05 (s, 1H), 6.43 (s, 1H), 5.26 (s, 2H), 4.83 (s, 2H), 3.96-3.88 (m, 1H), 3.80-3.75 (m, 1H), 3.73 (s, 3H), 3.71-3.67 (m, 1H), 3.65-3.58 (m, 1H), 3.49-3.36 (m, 4H), 3.26-3.15 (m, 2H), 2.22-2.11 (m, 2H), 2.09-1.96 (m, 2H), 1.80-3.75 (m, 1H), 3.65-3.58 (m, 1H), 3.65 (j=3.7.2 hz.
Example 119: synthesis of tert-butyl 4- ((2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -3-oxo-7- (trifluoromethyl) isoindolin-5-yl) methoxy) piperidine-1-carboxylate (BB-1)
Will contain Cs 2 CO 3 (85 mg,4.5 eq, 0.26 mmol) of water (0.2 mL) was added to toluene (1.5 mL) containing compound (AY-1) (30 mg,99Wt%,1 eq, 58. Mu. Mol), ((1- (tert-butoxycarbonyl) piperidin-4-yl) oxy) methyl) potassium trifluoroborate (BA-1) (28 mg,1.5 eq, 87. Mu. Mol) and RuPhos Pd G3 (4.8 mg,0.1 eq, 5.8. Mu. Mol). The reaction mixture was sparged with nitrogen for 1 minute and then stirred at 100 ℃ for 2 hours (microwaves). Additional RuPhos Pd G3 (4.8 mg,0.1 eq, 5.8 μmol) was added and stirred at 120 ℃ for 2 hours (microwave). The reaction mixture was dissolved with EtOAc (10 mL) and NaHCO was added 3 Saturated aqueous solutions (10 mL). The organic layer was separated and the aqueous extracted with EtOAc (2×10 mL). The organic layers were combined, dried (MgSO 4 ) Filtered and evaporated. The crude product was purified by RP flash C18 chromatography (12 g cartridge, 10-60% MeCN/10mM ammonium bicarbonate) followed by preparative HPLC (woth, basic (0.3% ammonia), basic, woth XBridge BEH C18 ODB, 5 μm, 30x100mM column, 0-100% MeCN/water) to give the title compound (BB-1) (5 mg,8 μmol,10%,99% pure). M/z 648.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.39(s,1H),8.02(s,1H),8.02–7.98(m,1H),7.96(s,1H),7.72(ddd,J=8.4,5.6,3.0Hz,1H),7.68(dd,J=4.8,2.6Hz,2H),7.61–7.57(m,2H),7.38(t,J=8.0Hz,1H),6.91(dt,J=7.7,1.2Hz,1H),5.10(s,2H),4.74(s,2H),3.64(tt,J=8.4,4.6Hz,3H),3.08(s,3H),3.05(s,2H),1.90–1.82(m,2H),1.45(ddd,J=12.9,8.6,4.0Hz,2H),1.40(s,9H)。
Example 120: synthesis of 2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6- (((1-methylpiperidin-4-yl) oxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (BC-4)
Step 1: synthesis of Potassium trifluoro (((1-methylpiperidin-4-yl) oxy) methyl) borate (BC-3)
To a stirred solution of NaH (174 mg,60wt%,3 eq, 4.34 mmol) in THF (3 mL) at 0 ℃ was added 1-methylpiperidin-4-ol (BC-1) (500 mg,3 eq, 4.34 mmol). The mixture was stirred at 0 ℃ for 15 minutes and at room temperature for 30 minutes. Potassium (bromomethyl) trifluoroborate (BC-2) (29 mg,1 eq, 1.45 mmol) was then added at 0deg.C, and the mixture was stirred at room temperature for 20 hours. The reaction mixture was then quenched with 4.5M potassium bifluoride (1.02 g,2.89ml,4.5 moles, 9 equivalents, 13.0 mmol) and the solution stirred for 30 minutes. The mixture was evaporated under reduced pressure and dried in vacuo. The crude product was suspended in Et 2 O (10 mL) and filtered to remove organic impurities. The resulting solid was suspended in acetone (10 mL) and filtered to remove additional inorganic impurities. The organic phase was then evaporated to give the sub-title compound (BC-3) (140 mg,0.12mmol,8.2%,20% purity) as a yellow gum, the crude product was used in the next step without further purification or analysis.
Step 2: synthesis of 2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6- (((1-methylpiperidin-4-yl) oxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (BC-4)
Will contain Cs 2 CO 3 (0.14G, 4.5 eq, 0.43 mmol) of water (0.2 mL) was added to toluene (1.5 mL) containing compound (AY-1) (50 mg,99Wt%,1 eq, 96. Mu. Mol), product (BC-3) from step 1 above (34 mg,1.5 eq, 0.14 mmol) and RuPhos Pd G3 (8.1 mg,0.1 eq, 9.6. Mu. Mol). The reaction mixture was sparged with nitrogen for 1 minute and stirred at 100 ℃ for 2 hours (microwave). The reaction mixture was dissolved with EtOAc (10 mL) and treated with NaHCO 3 Is washed with a saturated aqueous solution (10 mL). The organic layer was separated and the aqueous yellow solution extracted with EtOAc (2X 10 mL). The organics were combined, dried (MgSO 4 ) Filtered and evaporated. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), basic, waters X-Select CSH C18 ODB, 5 μm, 30X100mm column, 0-100% MeCN/water) to give the title compound (BC-4) (4 mg,7 μmol,7%,99% purity). M/z 562.3 (M+H) + (ES+)。 1 H NMR (400 mhz, dmso-d 6) delta 8.39 (s, 1H), 8.03-7.97 (m, 2H), 7.95 (s, 1H), 7.72 (ddd, j=8.3, 5.5,2.9hz, 1H), 7.68 (dd, j=4.5, 2.5hz, 2H), 7.61-7.57 (m, 2H), 7.38 (t, j=8.0 hz, 1H), 6.91 (d, j=8.0 hz, 1H), 5.10 (s, 2H), 4.71 (s, 2H), 3.08 (s, 3H), 2.64-2.56 (m, 2H), 2.15 (s, 3H), 2.04 (t, j=10.7 hz, 2H), 1.89 (d, j=12.7 hz, 2H), 1.55 (d, j=9.8 hz, 1H). One proton is masked by the water peak.
Example 121: synthesis of 2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6- (((tetrahydro-2H-pyran-4-yl) oxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (BD-3)
Step 1: synthesis of Potassium trifluoro (((tetrahydro-2H-pyran-4-yl) oxy) methyl) borate (BD-2)
To a stirred solution of NaH (196 mg,60wt%,3 eq, 4.90 mmol) in THF (3 mL) was added tetrahydro-2H-pyran-4-ol (BD-1) (500 mg,3 eq, 4.90 mmol) at 0 ℃. The mixture was stirred at 0 ℃ for 15 minutes and at room temperature for 30 minutes. Then potassium (bromomethyl) trifluoroborate (BC-2) (328 mg) was added at 0deg.C1 equivalent, 1.63 mmol) and the mixture was stirred at room temperature for 20 hours. The reaction mixture was quenched with 4.5M potassium bifluoride (1.15 g,3.26ml,4.5 moles, 9 equivalents, 14.7 mmol) and the solution stirred for 30 minutes. The mixture was evaporated under reduced pressure and dried in vacuo. The crude product was suspended in Et 2 O (10 mL) and filtered to remove organic impurities. The resulting solid was suspended in acetone (10 mL) and filtered to remove additional inorganic impurities. The organic phase was then evaporated to give the sub-title compound (BD-2) (45 mg,0.20mmol, 12%).
Step 2: synthesis of 2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6- (((tetrahydro-2H-pyran-4-yl) oxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (BD-3)
Will contain Cs 2 CO 3 (0.14G, 4.5 eq, 0.43 mmol) of water (0.2 mL) was added to toluene (1.5 mL) containing compound (AY-1) (50 mg,99Wt%,1 eq, 96. Mu. Mol), product (BD-2) from step 1 above (28 mg,1.3 eq, 0.13 mmol) and RuPhos Pd G3 (8.1 mg,0.1 eq, 9.6. Mu. Mol). The reaction mixture was sparged with nitrogen for 1 minute and then stirred at 100 ℃ for 2 hours (microwaves). The reaction mixture was extracted with EtOAc (10 mL) and NaHCO 3 Is dissolved in saturated aqueous solution (10 mL). The organic layer was separated and the aqueous extracted with EtOAc (2×10 mL). The organic extracts were combined and dried (MgSO 4 ) Filtered and evaporated. The crude product was purified by preparative HPLC (waters, basic (0.1% ammonia), basic, waters XBridge BEH C18 ODB, 5 μm, 30x100mm column, 0-100% MeCN/water) to give the title compound (BD-3) as a white solid (5 mg,9 μmol,9%,99% purity). M/z 549.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.39(s,1H),8.03(s,1H),7.99(dd,J=7.9,2.2Hz,1H),7.97(s,1H),7.74–7.69(m,1H),7.69–7.66(m,2H),7.61–7.56(m,2H),7.38(t,J=8.0Hz,1H),6.93–6.88(m,1H),5.10(s,2H),4.75(s,2H),3.83(dt,J=11.7,4.2Hz,2H),3.65(tt,J=8.8,4.0Hz,1H),3.36(ddd,J=11.9,9.9,2.7Hz,2H),3.08(s,3H),1.94–1.87(m,2H),1.50(dtd,J=13.3,9.5,4.1Hz,2H)。
Example 122; synthesis of 3'- (6- ((((1R, 2S) -2-hydroxycyclopentyl) amino) methyl) -1-oxo-4- (trifluoromethyl) isoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (BE-3)
Step 1: synthesis of 3'- (6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) isoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (BE-1)
DIPEA (105 mg, 141. Mu.L, 1.5 eq, 908. Mu. Mol) was added to a solution of intermediate ACC-8 (200 mg,1.2 eq, 726. Mu. Mol) and intermediate B-4 (198 mg,1 eq, 605. Mu. Mol) in EtOH (20 mL) and stirred at 60℃for 16 hours. Additional intermediate B-4 (99.0 mg,0.5 eq, 303 μmol) was added and the reaction stirred at 60 ℃ for 16 hours. The volatiles were evaporated and the crude product was purified by RP flash C18 chromatography (24 g cartridge, 25-55% MeCN/10mM ammonium bicarbonate) to give the sub-title compound (BE-1) as a white solid (255 mg,0.25mmol,42%,48% purity). M/z 490.2 (M+H) + (ES+)。
Step 2: synthesis of 3'- (6-formyl-1-oxo-4- (trifluoromethyl) isoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (BE-2)
DCM (25 mL) containing the product from step 1 above (BE-1) (257 mg,1 eq, 525. Mu. Mol) and dess-Martin periodate (334 mg,1.5 eq, 788. Mu. Mol) was stirred at 40℃for 16 h. The reaction mixture was diluted with DCM (10 mL) and NaHCO 3 Saturated aqueous (10 mL) was washed. The organics were separated, dried (MgSO 4 ) Filtered and evaporated to give the crude product. The crude product was purified by preparative HPLC (waters, basic (0.3% ammonia), XBridge BEH C18 ODB 1 from waters, 5 μm, 30x100mm column, 20-100% MeCN/water) to give the sub-title compound (BE-2) as a white solid (11 mg,22 μmol,4.3%,99% purity). M/z 488.1 (M+H) + (ES+)。
Step 3: synthesis of 3'- (6- ((((1R, 2S) -2-hydroxycyclopentyl) amino) methyl) -1-oxo-4- (trifluoromethyl) isoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (BE-3)
NaBH (OAc) 3 (34 mg,6.0 eq, 0.16 mmol) was added to a stirred solution of the product (BE-2) (13 mg,99Wt%,1 eq, 26. Mu. Mol), (1S, 2R) -2-aminocyclopentan-1-ol, HCl (Z-1) (11 mg,3 eq, 79. Mu. Mol) and DIPEA (17 mg, 23. Mu.L, 5 eq, 0.13 mmol) from step 2 above in DCM (2 mL) and left overnight at room temperature. The reaction mixture was diluted with DCM (5 mL) and NaHCO 3 Saturated aqueous (6 mL) was washed. The organic layer was separated and evaporated to dryness to give the crude product. The crude product was purified by preparative HPLC (waters, basic (0.1% ammonia), XBridge BEH C18 ODB 1 from waters, 5 μm, 30x100mm column, 25-100% MeCN/water) to give the title compound (BE-3) as a white solid (4 mg,7 μmol,30%,99% purity). M/z 573.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),8.19(dd,J=8.1,1.8Hz,1H),8.12(d,J=1.8Hz,1H),8.07–7.99(m,3H),7.89(d,J=8.0Hz,1H),7.72(t,J=2.0Hz,1H),7.41(t,J=8.0Hz,1H),6.93(d,J=7.7Hz,1H),5.09(s,2H),4.40(d,J=3.5Hz,1H),4.01–3.82(m,3H),3.16(s,3H),2.80–2.70(m,1H),2.34–2.24(m,1H),1.75–1.54(m,4H),1.47–1.34(m,2H)。
Example 123: synthesis of 6- (hydroxymethyl) -2- {6- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (ACX-2)
Step 1: synthesis of 2- {6- [ 6-formyl-1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -3-oxo-7- (trifluoromethyl) -1H-isoindole-5-carbaldehyde (ACX-1)
To intermediate (E-7) (100 mg,1.0 eq, 328. Mu. Mol) and intermediate (ABC-4) (113 mg,1.5 eq, 492. Mu. Mol) in 1, 4-dioxane at room temperature under a nitrogen atmospherePotassium phosphate and Pd (OAc) were added to the stirred mixture in alkane (5 mL) 2 (7.4 mg,0.1 eq, 33. Mu. Mol) and XantPhos (19 mg,0.1 eq, 33. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The resulting mixture was extracted with EtOAc (3×30 mL). The combined organic layers were washed with water (2×30 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (ACX-1) (50 mg, 65. Mu. Mol,22%,90% purity) as a yellow solid. M/z 691.1 (M+H) + (ES+)。
Step 2: synthesis of 6- (hydroxymethyl) -2- {6- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (ACX-2)
To a stirred solution of the product (ACX-1) (30 mg,1 eq, 43. Mu. Mol) from step 1 above in THF (5 mL) at room temperature under nitrogen was added NaBH 4 (3.3 mg,2 equivalents, 86. Mu. Mol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 1 hour. Will be quenched with MeOH (1 mL) at room temperature. The resulting mixture was concentrated in vacuo. The crude product (25 mg) was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 55% B in 10 minutes; wavelength: 254/220nm; retention time: 9.07 To give the title compound (ACX-2) (3.1 mg,4.4 μmol,9.3%,91 purity) as a white solid. M/z 695.0 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO–d6)δ8.50(s,1H),8.05(d,J=1.7Hz,4H),7.99(s,2H),7.81–7.76(m,1H),7.75–7.66(m,3H),5.61(s,2H),5.29(s,4H),4.72(s,4H),3.58(s,3H)。
Example 124: synthesis of 2- [6- (ethylamino) -4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -6- { [ (3S) -3-methylpiperidin-1-yl ] methyl } -3H-isoindol-1-one (ACY-4)
Step 1: synthesis of 3-oxo-isoindoline-5-carbaldehyde (ACY-2)
To a 50-mL round-bottomed flask purged and maintained with an inert nitrogen atmosphere at room temperature under a nitrogen atmosphere was placed DMSO (20 mL) containing 6-bromo-2, 3-isoindolin-1-one (ACY-1) (424 mg,1 eq, 2.00 mmol), t-butyl isocyanate (199mg, 1.2 eq, 2.40 mmol) and sodium formate (272 mg,2 eq, 4.00 mmol), followed by Pd (OAc) 2 (45 mg,0.1 eq, 200. Mu. Mol) and DPPE (159 mg,0.2 eq, 400. Mu. Mol). The resulting solution was stirred overnight at 120 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 48% in 15 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ACY-2) (150 mg,0.87mmol,47%,93% purity) as a yellow solid. M/z 162.0 (M+H) + (ES+)。
Step 2: synthesis of (S) -6- ((3-methylpiperidin-1-yl) methyl) isoindolin-1-one (ACY-3)
Into a 250-mL round bottom flask purged with an inert nitrogen atmosphere and maintained at room temperature was placed a solution containing the product (ACY-2) (150 mg,1 eq, 931. Mu. Mol) from step 1 above and (3S) -3-methylpiperidine (R-1) (185 mg,2 eq, 1.86 mmol) in DCM (15 mL) and then Et was added at room temperature 3 N (188 mg,2 eq, 1.86 mmol). NaBH (OAc) was added to the above mixture over 4 hours at 0deg.C 3 (399mg, 2 equivalents, 1.86 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The resulting mixture was diluted with water and extracted with DCM (3×50 mL). The combined organic layers were washed with brine (2×50 mL) and dried (Na 2SO 4). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (ACY-3) (90 mg, 295. Mu. Mol,39%,80% purity) as a yellow solid. M/z 245.2 (M+H) + (ES+)。
Step 3: synthesis of (S) -2- (6- (ethylamino) -4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((3-methylpiperidin-1-yl) methyl) isoindolin-1-one (ACY-4)
A25-mL round bottom flask purged with an inert nitrogen atmosphere was charged at room temperature with a flask containing the product (ACY-3) (45 mg,1 eq, 184. Mu. Mol), intermediate (AD-1) (58 mg, 184. Mu. Mol,1 eq) and K from step 2 above 3 PO 4 (117 mg,3 eq, 552. Mu. Mol) 1, 4-dioxane (3 mL) and then Pd (OAc) was added at room temperature under a nitrogen atmosphere 2 (8.3 mg,0.2 eq, 37. Mu. Mol) and XantPhos (43 mg,0.4 eq, 74. Mu. Mol). The resulting mixture was stirred under nitrogen at 100 ℃ for an additional overnight. The resulting mixture was cooled to room temperature, diluted with water and extracted with DCM (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ) And (5) drying. After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 column, 30 x 150mm,5 μm, mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 11% B to 25% B in 10 minutes; wavelength: 254/210nm; retention time: 6.32. this gave the title compound (ACY-4) (15.3 mg, 28. Mu. Mol,15%,96% purity) as a white solid. M/z 522.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH–d4)δ8.45(s,1H),7.89(s,1H),7.82–7.63(m,4H),7.68–7.60(m,2H),7.54(d,J=1.3Hz,1H),6.06(d,J=1.2Hz,1H),5.11(s,2H),4.10(s,2H),3.44(s,3H),3.34(s,1H),3.32(s,1H),3.26–3.11(m,2H),2.53(t,J=11.9Hz,1H),2.25(t,J=12.1Hz,1H),1.83(s,3H),1.71(d,J=12.9Hz,1H),1.23(t,J=7.2Hz,3H),1.09(d,J=11.2Hz,1H),0.95(d,J=6.4Hz,3H)。
Example 125: synthesis of 4- {2- [ (1- {2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl } -5,8, 11-trioxa-2-azatride-din-13-yl) carbamoyl ] ethyl } -2, 2-difluoro-12- (1H-pyrrol-2-yl) -1λ5, 3-diaza-2-boronic acid tricyclic [7.3.0.0 {3,7} ] dodecane-1 (12), 4,6,8, 10-pentaen-1-yl cation-2-anion) (ACZ-5)
Step 1: synthesis of tert-butyl N- (1- {2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl } -5,8, 11-trioxa-2-azatridecan-13-yl) carbamate (ACZ-2)
To intermediate (ABP-1) (70 mg,1 eq, 137. Mu. Mol) and N- (2- {2- [2- (2-aminoethoxy) ethoxy) at room temperature]To a stirred solution of tert-butyl ethoxy } ethyl) carbamate (ACZ-1) (52 mg,1.3 eq, 178. Mu. Mol) in DCM (2 mL) was added NaBH (OAc) 3 (87 mg,3 equivalents, 411. Mu. Mol). The resulting mixture was stirred at 40℃for 3 hours. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1) to give the sub-title compound (ACZ-2) as a yellow solid (93 mg,123 μmol,85%,92% purity). M/z 699.3 (M+H) + (ES+)。
Step 2: synthesis of 6- (13-amino-5, 8, 11-trioxa-2-azatridecan-1-yl) -2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one) (ACZ-3)
To a stirred solution of the product from step 1 above (ACZ-2) (88 mg,1 eq, 112 μmol) in DCM (1 mL) at room temperature was added TFA (0.3 mL) dropwise. The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 17% B over 10 min; wavelength: 254/220nm; retention time: 9.67) to give the sub-title compound (ACZ-3) as a pale yellow solid (65 mg,103 μmol,86%,95% purity). M/z 599.3 (M+H) + (ES+)
Step 3: synthesis of 4- {2- [ (1- {2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl } -5,8, 11-trioxa-2-azatride-din-13-yl) carbamoyl ] ethyl } -2, 2-difluoro-12- (1H-pyrrol-2-yl) -1λ5, 3-diaza-2-boronic acid tricyclic [7.3.0.0 {3,7} ] dodecane-1 (12), 4,6,8, 10-pentaen-1-yl cation-2-anion) (ACZ-5)
To the product (ACZ-3) (13 mg,1 equivalent, 22. Mu. Mol) from step 2 above and Et at room temperature 3 N (6 mg,3 eq, 57. Mu. Mol) to a stirred solution in DMF (1 mL) was added 4- {3- [ (2, 5-dioxopyrrolidin-1-yl) oxy)]-3-oxopropyl } -2, 2-difluoro-12- (1H-pyrrol-2-yl) -1λ5, 3-diaza-2-boronic acid tricyclic [7.3.0.0 {3,7}]Dodecane-1 (12), 4,6,8, 10-penten-1-yl positive ion-2-negative ion (ACZ-4) (6.5 mg,0.8 eq, 15. Mu. Mol). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was concentrated in vacuo. The residue was purified by preparative HPLC on a column (XBridge prep OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 50% B in 9 minutes; wavelength: 254/220nm; retention time: 8.77 To give the title compound (ACZ-5) (7.2 mg,7.2 μmol,38%,98% purity) as a violet solid. M/z 998.4 (M+H) + (ES+). 1 H NMR(400MHz,MeOH–d4)δ8.49(d,J=1.0Hz,1H),8.00–7.84(m,3H),7.62(d,J=1.3Hz,1H),7.19–7.07(m,4H),6.94–6.79(m,2H),6.34–6.15(m,3H),5.14(s,2H),4.02(d,J=1.1Hz,3H),3.90(s,2H),3.68–3.57(m,10H),3.54(t,J=5.3Hz,2H),3.50(d,J=1.0Hz,3H),3.37(d,J=5.2Hz,2H),3.35(d,J=6.0Hz,2H),3.22(t,J=7.8Hz,2H),2.76(t,J=5.1Hz,2H),2.60(t,J=7.8Hz,2H),1.25–1.19(m,3H)。
Example 126: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- { [ (1-methylpiperidin-4-yl) oxy ] methyl } -4- (trifluoromethyl) -3H-isoindol-1-one (ADA-2)
Step 1: synthesis of 6- (chloromethyl) -2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (ADA-1)
SOCl was added dropwise to a stirred solution of intermediate (ACH-1) (197mg, 1 eq., 384. Mu. Mol) in DCM (39 mL) under nitrogen at 0deg.C 2 (137 mg,3 equivalents, 1.15 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 1 hour. The reaction was quenched with MeOH (5 mL) at 0 ℃ and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1) to give the sub-title compound (ADA-1) (171 mg,271 μmol,84%,84% pure) as a yellow solid. M/z 531.2/533.2 (M+H) + (ES+)。
Step 2: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- { [ (1-methylpiperidin-4-yl) oxy ] methyl } -4- (trifluoromethyl) -3H-isoindol-1-one (ADA-2)
To a stirred solution of the product from step 1 above (ADA-1) (23 mg,43 μmol,1 eq) and 1-methylpiperidin-4-ol (BC-1) (25 mg,5 eq, 215 μmol) in DCM (2 mL) at room temperature was added TBAB (7 mg,0.5 eq, 21 μmol) and 1mL of 20% KOH aqueous solution. The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water (20 mL), extracted with DCM (3×20 mL), and extracted with (Na 2 SO 4 ) And (5) drying. After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (6/1) and by preparative HPLC under the following conditions (column: XBridge prep OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 32% B to 42% B in 9 minutes; wavelength: 254/220nm; retention time: 8.58 To give the title compound (ADA-2) (2.3 mg,4.4 μmol,8.5%,97% purity) as a white solid. M/z 522.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH–d4)δ8.50(s,1H),8.07(s,1H),7.95(s,1H),7.87(s,1H),7.65(d,J=1.2Hz,1H),6.20(d,J=1.2Hz,1H),5.26(s,2H),4.75(s,2H),4.04(s,3H),3.59(s,1H),3.51(s,3H),3.41–3.36(m,2H),2.76(s,2H),2.30(s,5H),2.01(s,2H),1.76(s,2H),1.25(t,J=7.2Hz,3H)。
Example 127: synthesis of (rac) -2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] oxy } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (ADB-1)
To a stirred solution of intermediate (ADA-1) (88 mg,1 eq, 166 μmol) and (1 r,2 s) -cyclopentane-1, 2-diol (AV-2) (85 mg,5 eq, 830 μmol) in DCM (2 mL) at room temperature was added TBAB (27 mg,0.5 eq, 83 μmol) and 1mL of 20% KOH aqueous solution. The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with DCM (3×30 mL). The combined organic layers were washed with brine (3×30 mL), dried (Na 2 SO 4 ) And concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 41% B to 51% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (ADB-1) (3.9 mg, 6.5. Mu. Mol,3.9%,98% purity) as a white solid. M/z 597.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.48(s,1H),8.06(d,J=15.2Hz,2H),7.89(s,1H),7.58(d,J=1.2Hz,1H),6.94(t,J=5.4Hz,1H),6.25(d,J=1.2Hz,1H),5.21(s,2H),4.80(d,J=12.8Hz,1H),4.69(d,J=12.8Hz,1H),4.50(d,J=4.8Hz,1H),4.05–3.89(m,1H),3.90(s,3H),3.75–3.68(td,J=6.1,3.7Hz,1H),3.47(s,3H),3.32–3.23(m,2H),1.82–1.64(m,4H),1.62–1.52(m,1H),1.52–1.40(m,1H),1.17(t,J=7.1Hz,3H)。
Example 128: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- [ (oxetan-3-yloxy) methyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (ADC-2)
To a stirred solution of intermediate (ADA-1) (35 mg,1 eq, 66 μmol) and oxetan-3-ol (ADC-1) (50 mg,10 eq, 660 μmol) in DCM (1 mL) was added TBAB (11 mg,0.5 eq, 33 μmol) and 1mL of 20% KOH in water at room temperature. The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was diluted with water. The aqueous layer was extracted with DCM (3X 20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35% B to 45% B in 9 minutes; wavelength: 254/220nm; retention time: 8.63 To give the title compound (ADC-2) (4.2 mg,7.4 μmol,11%,98% purity) as a white solid. M/z569.0 (M+H) + (ES+). 1 H NMR(400MHz,MeOH–d4)δ8.50(s,1H),8.08(s,1H),7.98(s,1H),7.87(s,1H),7.66(d,J=1.2Hz,1H),6.21(d,J=1.2Hz,1H),5.27(s,2H),4.87–4.82(m,2H),4.81–4.75(m,1H),4.72–4.62(m,4H),4.04(s,3H),3.51(s,3H),3.37(d,J=7.2Hz,2H),1.25(t,J=7.2Hz,3H)。
Example 129: synthesis of 4- {2- [ (1- {2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl } -5,8, 11-trioxa-2-azatride-din-13-yl) carbamoyl ] ethyl } -2, 2-difluoro-10, 12-dimethyl-1λ5, 3-diaza-2-boronic acid tricyclic [7.3.0.0 {3,7} ] dodecane-1 (12), 4,6,8, 10-penten-1-yl positive ion-2-negative ion (ADD-2)
Intermediate (ACZ-3) (7.8 mg,0.8 eq, 26. Mu. Mol) and Et at room temperature 3 N(To a stirred solution of 10mg,3 eq, 99. Mu. Mol) in DMF (1.5 mL) was added HATU (17 mg,1.3 eq, 43 mmol) and 6- ({ [2- (2-aminoethoxy) ethyl group]Amino } methyl) -2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl]Pyridin-2-yl]-4- (trifluoromethyl) -3H-isoindol-1-one (ADD-1) (20 mg,1 eq, 33. Mu. Mol). The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1) and by preparative HPLC under the following conditions (column: XB ridge preparative OBD C18 column, 30X 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 50% B in 9 minutes; wavelength: 254/220nm; retention time: 8.73 To give the title compound (ADD-2) (13.4 mg,14 μmol,41%,98% purity) as an orange solid. M/z 961.0 (M+H) + (ES+). 1 H NMR(400MHz,MeOH–d4)δ8.49(s,1H),8.01(s,1H),7.95(s,1H),7.87(s,1H),7.65(d,J=1.2Hz,1H),7.32(s,1H),6.92(d,J=4.0Hz,1H),6.26(d,J=4.0Hz,1H),6.20(d,J=1.2Hz,1H),6.15(s,1H),5.18(s,2H),4.03(s,3H),3.93(s,2H),3.70–3.58(m,10H),3.52(d,J=9.4Hz,5H),3.38–3.35(m,4H),3.16(t,J=7.7Hz,2H),2.78(t,J=5.2Hz,2H),2.58(t,J=7.7Hz,2H),2.45(s,3H),2.23(s,3H),1.24(t,J=7.2Hz,3H)。
Example 130: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- [ (1, 2-oxazol-4-yloxy) methyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (ADE-2)
To a stirred solution of intermediate (ADA-1) (25 mg,1 eq, 47. Mu. Mol) and 1, 2-oxazol-4-ol (ADE-1) (0 (20 mg,5 eq, 235. Mu. Mol) in DCM (2 mL) at room temperature was added dropwise TABA (8 mg,0.5 eq, 24. Mu. Mol) and 1mL of 20% aqueous KOH solutionWashed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1). The crude product was purified by preparative HPLC on a column (XBridge prep C18 OBD column, 30 x 100mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 24% B to 54% B in 7 minutes; wavelength: 254/220 nm) to give the title compound (ADE-2) (2.6 mg,8.9%,93% purity) as a white solid. M/z 580.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ9.26(s,1H),8.47(s,1H),8.37(s,1H),8.00–7.82(m,3H),7.55(d,J=1.2Hz,1H),6.93(t,J=5.3Hz,1H),6.25(d,J=1.2Hz,1H),5.19(s,2H),4.33(s,2H),3.89(s,3H),3.47(s,3H),3.30–3.21(m,2H),1.17(t,J=7.1Hz,3H)。
Example 131: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (ADF-2)
/>
DIPEA (38 mg,3 equiv., 294. Mu. Mol) was added dropwise to a stirred solution of intermediate (ABP-1) (50 mg,1 equiv., 98. Mu. Mol) and (1S, 2R) -2-aminocyclopentan-1-ol (Z-1) (20 mg,2 equiv., 196. Mu. Mol) in MeOH (2 mL) at room temperature. The resulting mixture was stirred at room temperature for 3 hours. NaBH was added to the above mixture at room temperature 4 (15 mg,4 eq, 392. Mu. Mol). The resulting mixture was stirred at room temperature for an additional 2 hours. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 7% B to 31% B in 7 min; wavelength: 254/220nm; retention time: 6.81) to give the title compound (ADF-2) (15.1 mg, 25. Mu. Mol,25%,98% purity) as a yellow solid. m/z 596.4 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.45(s,1H),8.10(d,J=15.4Hz,2H),7.86(s,1H),7.49(d,J=1.2Hz,1H),6.24(d,J=1.2Hz,1H),5.20(s,2H),4.14–4.07(m,3H),3.87(s,3H),3.45(s,3H),3.25(q,J=7.2Hz,2H),3.14–3.04(m,1H),1.90–1.79(m,1H),1.78–1.65(m,2H),1.63–1.40(m,3H),1.15(t,J=7.2Hz,3H)。
Example 132: synthesis of 3'- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (ADG-3)
Step 1: synthesis of 3 '-amino-2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (ADG-2)
To an 8mL sealed tube at room temperature was added 4-bromo-3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADG-1) (60 mg,1 eq, 228. Mu. Mol), 3-aminophenylboronic acid (AAS-1) (31 mg,1 eq, 228. Mu. Mol) and K 2 CO 3 (95 mg,3 eq, 684. Mu. Mol) 1, 4-dioxane (2 mL) water (0.2 mL). Pd (dppf) Cl was added to the above mixture 2 . DCM (17 mg,0.1 eq, 23. Mu. Mol) at room temperature under nitrogen. The resulting mixture was stirred under nitrogen at 100 ℃ for an additional 3 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (36% MeCN up to 48% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ADG-2) (35 mg, 114. Mu. Mol,56%,90% purity) as a white solid. M/z 276.1 (M+H) + (ES+)。
Step 2: synthesis of 3'- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (ADG-3)
To an 8mL sealed tube at room temperature was added a solution containing the product (ADG-2) from step 1 above (20 mg,1 equivalent, 73. Mu. Mol) and intermediate (B-4) (36 mg,1.5 equivalent, 109. Mu. Mol) EtOH (1 mL). Et was added dropwise to the above mixture at room temperature over 3 minutes 3 N (22 mg,3 eq, 219. Mu. Mol). The resulting mixture was stirred at 80 ℃ for an additional overnight. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19X 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30B to 60B in 7 minutes; a detector, UV 254/210nm; retention time: 6.08 to give the title compound (ADG-3) (1.7 mg, 3.5. Mu. Mol,4.7%,98% purity) as a white solid. M/z 490.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.47(s,1H),8.25–8.10(m,1H),8.13(d,J=1.8Hz,1H),8.08–7.87(m,4H),7.74(s,1H),7.42(t,J=8.0Hz,1H),6.93(d,J=7.7Hz,1H),5.60(t,J=5.8Hz,1H),5.11(s,2H),4.71(d,J=5.7Hz,2H),3.17(s,3H)。
Example 133: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] oxy } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (ADH-1)
The racemic compound (ADB-1) (25 mg,1 eq., 42. Mu. Mol) was separated by chiral preparative HPLC under the following conditions (column: CHIRAL ART cellulose-SB, 2X 25cm, 5. Mu.m; mobile phase A: MTBE: MEOH=2:1, mobile phase B: hex (0.5% 2M NH3-MeOH) -HPLC; flow rate: 45 ml/min; gradient: 55% B to 55% B over 30 min; wavelength: 220/254nm; retention time 1 (min): 23.4; retention time 2 (min): 26.9) to give the title compound (ADH-1) (10.8 mg, 18. Mu. Mol,43%,99% purity) as a white solid. M/z 597.0 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO–d6)δ8.48(s,1H),8.05(d,J=15.2Hz,2H),7.89(s,1H),7.58(d,J=1.2Hz,1H),6.94(tJ=5.4 hz, 1H), 6.25 (d, j=1.2 hz, 1H), 5.21 (s, 2H), 4.80 (d, j=12.8 hz, 1H), 4.69 (d, j=12.8 hz, 1H), 4.50 (d, j=4.8 hz, 1H), 4.07-3.96 (m, 1H), 3.90 (s, 3H), 3.78-3.64 (m, 1H), 3.47 (s, 3H), 3.31-3.20 (m, 2H), 1.82-1.64 (m, 4H), 1.64-1.50 (m, 1H), 1.51-1.37 (m, 1H), 1.17 (t, j=7.1 hz, 3H). Column: CHIRAL ART cellulose-SB, 4.6 x 100mm,3 μm; mobile phase a: MTBE (0.1% DEA): hex: meoh=30:55:15; flow rate: 1 ml/min; retention time: 8.490.
example 134: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- ({ [ (1S, 2R) -2-hydroxycyclopentyl ] oxy } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (ADI-1)
The racemic compound (ADB-1) (25 mg,1 eq., 42. Mu. Mol) was separated by chiral preparative HPLC under the following conditions (column: CHIRAL ART cellulose-SB, 2X 25cm, 5. Mu.m; mobile phase A: MTBE: MEOH=2:1, mobile phase B: hex (0.5% 2M NH3-MeOH) -HPLC; flow rate: 45 ml/min; gradient: 55% B to 55% B over 30 min; wavelength: 220/254nm; retention time 1 (min): 23.4; retention time 2 (min): 26.9) to give the title compound (ADI-1) (10.4 mg, 17. Mu. Mol,41%,98% purity) as a white solid. M/z597.0 (M+H) + (ES+)。 1 H NMR (400 mhz, dmso-d 6) delta 8.48 (s, 1H), 8.05 (d, j=15.2 hz, 2H), 7.89 (s, 1H), 7.57 (d, j=1.2 hz, 1H), 6.94 (t, j=5.4 hz, 1H), 6.25 (d, j=1.2 hz, 1H), 5.21 (s, 2H), 4.80 (d, j=12.8 hz, 1H), 4.69 (d, j=12.8 hz, 1H), 4.50 (d, j=4.8 hz, 1H), 4.06-3.95 (m, 1H), 3.90 (s, 3H), 3.77-3.63 (m, 1H), 3.47 (s, 3H), 3.30-3.20 (m, 2H), 1.86-1.62 (m, 4H), 1.62-1.51 (m, 1.51), 1.38 (m, 1.38-1H). Column: CHIRAL ART cellulose-SB, 4.6 x 100mm,3 μm; mobile phase a: MTBE (0.1% DEA): hex: meoh=30:55:15; flow rate: 1 ml/min; retention time: 9.545.
example 135: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (ADJ-1)
DIPEA (51 mg,2 eq, 392. Mu. Mol) was added dropwise to a stirred solution of intermediate (ABP-1) (100 mg,1 eq, 196. Mu. Mol) and 1- (aminomethyl) cyclobutan-1-ol, HCl (AW-1) (40 mg,2 eq, 392. Mu. Mol) in MeOH (15 mL) at room temperature. The resulting mixture was stirred at room temperature for 2 hours. Drop wise adding NaBH to the above mixture at 0deg.C 4 (30 mg,4 eq, 784. Mu. Mol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by addition of 5mL of ice water at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% fa), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 8% B to 32% B over 7 min; wavelength: 254/220nm; retention time: 6.12) to give the title compound (ADJ-1) as an off-white solid (50.1 mg,84 μmol,43%,99% purity). M/z 596.4 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),8.14(d,J=4.6Hz,1H),8.09(s,1H),7.87(s,1H),7.65(t,J=1.2Hz,1H),6.21(d,J=1.2Hz,1H),5.28(d,J=2.5Hz,2H),4.25–4.19(m,2H),4.04(s,3H),3.51(s,3H),3.42–3.34(m,2H),2.94(d,J=1.5Hz,2H),2.18–2.05(m,4H),1.84–1.71(m,1H),1.60–1.48(m,1H),1.25(t,J=7.2Hz,3H)。
Example 136: synthesis of 3'- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carboxamide (ADK-1)
At 0℃to compound (ADG-3) (30 mg,1 equivalent, 60. Mu. Mol) and K 2 CO 3 (25 mg,3 equivalents,180. Mu. Mol) to a stirred solution of DMSO (1 mL) was added hydrogen peroxide solution (0.5 mL,30 Wt%). The resulting mixture was stirred at room temperature for an additional 3 hours. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% fa) and MeCN (0% MeCN up to 50% in 15 min); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunfire preparation type C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% fa), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 15% B to 35% B in 10 minutes, detector, UV 254/220nm; retention time: 9.4. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ADK-1) (1.2 mg,2.3 μmol,3.9%,95% purity) as a white solid. M/z 508.1 (M+H) + (ES+). 1 H NMR(400MHz,MeOH–d4)δ8.44(s,1H),8.28–8.20(m,1H),8.16–8.07(m,2H),7.98(s,1H),7.95–7.79(m,1H),7.70(s,2H),7.48(t,J=8.0Hz,1H),7.14(d,J=7.7Hz,1H),5.09(s,2H),4.82(s,2H),3.24(s,3H)。
Example 137: synthesis of 6- ({ [ (1R, 2R) -2-hydroxycyclopentyl ] oxy } methyl) -2- [2'- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (ADL-4)
Step 1: synthesis of 6- (hydroxymethyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (ADL-1)
Et is added to a stirred mixture of intermediate (P-1) (104 mg,0.42mmol,1 eq.) and intermediate (B-4) (136 mg,1 eq., 0.42 mmol) in EtOH (5 mL) under a nitrogen atmosphere at room temperature 3 N (126 mg,3 eq, 1.25 mmol). The resulting mixture was stirred overnight at 60 ℃ under nitrogen atmosphere. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 30% in 20 minutes); detector, UV 254/220nm to give the sub-title compound (ADL-1) (70 mg,0.14mmol,36%,91% purity) as a yellow solid. M/z 465.1 (M+H) + (ES+)。
Step 2: synthesis of 6- (chloromethyl) -4- (1, 1-difluoroethyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) isoindolin-1-one (ADL-2)
To a 25mL round bottom flask at 0deg.C was added a solution containing the product (ADL-1) (40 mg,1 eq, 86. Mu. Mol) from step 1 above and SOCl 2 (31 mg,3 eq, 0.26 mmol) DCM (3 mL). The resulting mixture was stirred at room temperature overnight. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 30% in 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (ADL-2) (25 mg, 47. Mu. Mol,60%,90% purity) as a yellow solid. M/z479.1/481.1 (M+H) + (ES+)。
Step 3: synthesis of 6- ((((1R, 2R) -2-hydroxycyclopentyl) oxy) methyl) -2- (2 '- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -4- (trifluoromethyl) isoindolin-1-one (ADL-4)
To the product (ADL-2) (20 mg,1 eq, 41. Mu. Mol) from step 2 above and (1R, 2R) -cyclopentane-1, 2-diol (ADL-3) (8.5 mg,2 eq, 82. Mu. Mol) in DCM (1.5 mL) and H under nitrogen at room temperature 2 To a stirred mixture in O (1.5 mL) were added NaOH (5 mg,3 eq, 0.12 mmol) and TBAHS (14 mg,1 eq, 41. Mu. Mol). The resulting mixture was stirred overnight at 60 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XBridge prep. C18OBD column, 30 x 100mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 28% B to 53% B in 9 minutes; wavelength: 254/220nm; retention time: 8.87 To give the title compound (ADL-4) (2.1 mg,3.8 μmol,9.2%,99% purity) as a white solid. M/z 549.3 (M+H) + (ES+). 1 H NMR(400MHz,MeOH–d4)δ8.40(s,1H),8.09(s,1H),7.96(s,1H),7.92–7.85(m,1H),7.81–7.71(m,3H),7.67–7.59(m,2H),7.45(t,J=8.0Hz,1H),7.15–7.08(m,1H),5.08(s,2H),4.77(s,2H),4.23–4.15(m,1H),3.89–3.82(m,1H),3.21(s,3H),2.10–1.94(m,2H),1.83–1.66(m,3H),1.66–1.54(m,1H)。
Example 138: synthesis of 3'- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -6- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-carbonitrile (ADM-6)
Step 1: synthesis of 2-bromo-4-cyano-N- [ (methylsulfamoyl) -amino ] -benzamide (ADM-2)
To a stirred solution of 2-bromo-4-cyanobenzoic acid (ADM-1) (500 mg,1 eq, 2.21 mmol) and 4-methyl-3-thiosemicarbazide (D-2) (698 mg,3 eq, 6.63 mmol) in DMF (15 mL) at 0deg.C was added T 3 P (2.82 g,4 equivalents, 8.84 mmol) and pyridine (1.05 g,6 equivalents, 13.3 mmol). The resulting mixture was stirred at room temperature for 16 hours. The crude mixture was used directly in the next step without further purification. M/z 313.0/315.0 (M+H) + (ES+)。
Step 2: synthesis of 3-bromo-4- (4-methyl-5-sulfonyl-1, 2, 4-triazol-3-yl) -benzonitrile (ADM-3)
The resulting mixture from step 1 above (ADM-2) and aqueous NaOH (22 mL,1M,10 equivalents, 22.1 mmol) were stirred at 80℃for 2 hours. The mixture was cooled to room temperature. The residue was diluted with water. The pH of the solution was adjusted to 4 with 1N HCl. The resulting mixture was extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (ADM-3) (140 mg,0.43mmol,21%,90% purity) as a white solid. M/z 295.0/297.0 (M+H) + (ES+)。
Step 3: synthesis of 3-bromo-4- (4-methyl-1, 2, 4-triazol-3-yl) -benzonitrile (ADM-4)
To a stirred solution of the product from step 2 above (ADM-3) (50 mg,1 eq, 160. Mu. Mol) and hydrogen peroxide (12 mg,2 eq, 330. Mu. Mol) in DCM (10 mL) at 0deg.C was added AcOH (51 mg,5 eq, 840. Mu. Mol). The resulting mixture was stirred at room temperature for 16 hours. The mixture was treated with saturated NaHCO 3 (aqueous solution) neutralization to pH 9. The resulting mixture was extracted with DCM (3X 50 mL). The combined organic layers were washed with brine (2×30 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (ADM-4) (40 mg, 146. Mu. Mol,89%,96% purity) as a white solid. M/z 263.0/265.0 (M+H) + (ES+)。
Step 4: synthesis of 3 '-amino-6- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-carbonitrile (ADM-5)
To a stirred solution of the product (ADM-4) (40 mg,1 eq, 152. Mu. Mol) from step 3 above and 3-aminophenylboronic acid (AAS-1) (23 mg,1.1 eq, 160. Mu. Mol) in 1, 4-dioxane (4 mL) and water (1 mL) under nitrogen at room temperature was added K 2 CO 3 (63 mg,3 eq, 450. Mu. Mol) and Pd (dppf) Cl 2 DCM (22 mg,0.2 eq, 30. Mu. Mol). The resulting mixture was stirred under nitrogen at 80 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1) to give the sub-title compound (ADM-5) (30 mg,101 μmol,71%,93% purity) as a white solid. M/z 276.1 (M+H) + (ES+)。
Step 5: synthesis of 3'- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -6- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-carbonitrile (ADM-6)
To a stirred solution of the product (ADM-5) (30 mg,1 eq., 100. Mu. Mol) from step 4 above and intermediate (B-4) (89 mg,2.5 eq., 270. Mu. Mol) in EtOH (2 mL) at room temperature was added Et 3 N (55 mg,5 eq, 540. Mu. Mol). The resulting mixture was stirred at 80℃for 16 hours. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water(0.1% FA) and MeCN (30% MeCN up to 45% in 10 min); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 30 x 100mm,5 μm; mobile phase a: water (0.05% TFA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 24B to 46B in 7 minutes; a detector, UV 254/210nm; retention time: 6.75. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ADM-6) (4.6 mg,8.9 μmol,8%,95% purity) as a white solid. M/z 490.0 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.45(s,1H),8.17–8.07(m,2H),8.03–7.92(m,3H),7.87–7.78(m,2H),7.53–7.44(m,1H),7.15–7.07(m,1H),5.10(s,2H),4.82(s,2H),3.23(s,3H)。
Example 139: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (ADN-2)
Step 1: synthesis of 2-chloro-6-cyclopropyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridine (ADN-1).
To a 50-mL round-bottomed flask purged and maintained with an inert nitrogen atmosphere under nitrogen atmosphere was placed toluene (15 mL) containing intermediate (ACM-6) (650 mg,1 eq, 2.01 mmol), cyclopropylboronic acid (ABF-2) (207 mg,1.2 eq, 2.41 mmol) and potassium phosphate (1.28 g,3 eq, 6.03 mmol) at room temperature. Pd (OAc) was then added at room temperature under a nitrogen atmosphere 2 (59 mg,0.13 eq, 261. Mu. Mol) and tricyclohexylphosphine (73 mg,0.13 eq, 261. Mu. Mol). The resulting solution was stirred under nitrogen at 110 ℃ for 3 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). This gave the sub-title compound (AND-1) (310 mg, 945. Mu. Mol, 47%) as a brown-yellow solid. M/z329.1/331.1 (M+H) + (ES+)。
Step 2: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (ADN-2)
To the product (ADN-1) (16 mg,1 equivalent, 49. Mu. Mol), intermediate (AC-2) (10 mg,1 equivalent, 49. Mu. Mol) and K from step 1 above were added under nitrogen at room temperature 3 PO 4 (31 mg,3 eq, 147. Mu. Mol) to a stirred mixture of 1, 4-dioxane (2 mL) was added Pd (OAc) 2 (2.2 mg,0.2 eq, 10. Mu. Mol) and XantPhos (11 mg,0.4 eq, 20. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 60% B in 10 minutes; wavelength: 254/220nm; retention time: 9.9 To give the title compound (ADN-2) (6.2 mg,12 μmol,25%,97% purity) as a white solid. M/z 494.0 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.50(s,1H),8.10(d,J=7.7Hz,1H),8.04(d,J=1.4Hz,1H),7.99(d,J=7.7Hz,1H),7.84–7.73(m,2H),7.61–7.33(m,2H),6.91(d,J=1.4Hz,1H),5.21(s,2H),3.49(s,3H),2.10–2.00(m,1H),1.05–0.98(m,4H)。
Example 140: synthesis of 6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -2- {3- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] phenyl } -4- (trifluoromethyl) -3H-isoindol-1-one (ADO-2)
Step 1: synthesis of 2- {3- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] phenyl } -3-oxo-7- (trifluoromethyl) -1H-isoindole-5-carbaldehyde (ADO-1)
To intermediate (B-5) (150 mg,1 equivalent, 461. Mu. Mol), intermediate (AAB-8) (129 mg, 507. Mu. Mol,1.1 equivalent) and silver nitrate (235 mg,3 equivalent, 1.38 mmol) at room temperatureIn MeCN (6 mL) and H 2 O (3 mL) in a stirred solution. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 50% within 24 minutes); detector, UV 254/220nm. This gave the sub-title compound (ADO-1) (90 mg, 177. Mu. Mol,42%,92% purity) as an off-white solid. M/z 467.1 (M+H) + (ES+)。
Step 2: synthesis of 6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -2- {3- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] phenyl } -4- (trifluoromethyl) -3H-isoindol-1-one (ADO-2)
To the product (ADO-1) (80 mg,1 eq, 172. Mu. Mol) from step 1 above and (1S, 2R) -2-aminocyclopentan-1-ol, HCl (Z-1) (35 mg,1.5 eq, 258. Mu. Mol) and Et at room temperature 3 N (52 mg,3 eq, 516. Mu. Mol) in MeOH (10 mL). The resulting mixture was stirred at room temperature overnight. Adding NaBH to the mixture at 0deg.C 4 (97 mg,15 eq, 2.58 mmol). The resulting mixture was stirred at 30 ℃ for an additional 2 days. The mixture was cooled to room temperature. The reaction was then quenched by the addition of 2mL of ice water at 0 ℃ and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 50% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (ADO-2) (19.4 mg, 35. Mu. Mol,21%,98% purity) as a white solid. M/z 552.0 (M+H) + (ES+).1H NMR(400MHz,MeOH-d4)δ8.46(s,1H),8.12(s,1H),8.07–8.02(m,2H),7.98–7.94(m,1H),7.89(s,1H),7.59(t,J=8.0Hz,1H),7.28–7.22(m,1H),5.17(s,2H),4.20–4.14(m,1H),4.10–3.95(m,5H),3.43(s,3H),3.01–2.93(m,1H),1.98–1.68(m,4H),1.63–1.49(m,2H)。
Example 141: synthesis of 4- [2- (ethylamino) -6- [ 1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADP-8)
Step 1: synthesis of methyl 5-cyano-2- (2, 6-dichloropyridin-4-yl) benzoate (ADP-2)
To methyl 2-bromo-5-cyanobenzoate (ADP-1) (2.40 g,1 equivalent, 10.0 mmol), 2, 6-dichloropyridin-4-ylboronic acid (AAN-1) (1.92 g,1 equivalent, 10.0 mmol) and K under a nitrogen atmosphere at room temperature 2 CO 3 (4.15 g,3 equivalents, 30 mmol) in 1, 4-dioxane (30 mL) and H 2 Pd (dtBPF) Cl was added to a stirred solution in O (3 mL) 2 (652 mg,0.1 eq, 1.00 mmol). The resulting mixture was stirred under nitrogen at 60 ℃ for 3 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (4/1) to give the sub-title compound (ADP-2) (2.2 g,5.96mmol,72%,83% purity) as an off-white solid. M/z 307.0/309.0 (M+H) + (ES+)。
Step 2: synthesis of 5-cyano-2- (2, 6-dichloropyridin-4-yl) benzoic acid (ADP-3)
To a stirred solution of the product from step 1 above (ADP-2) (2.20 g,1 eq, 7.16 mmol) and LiOH (860 mg,5 eq, 35.8 mmol) in THF (30 mL) and H2O (10 mL) at room temperature. The resulting mixture was stirred at room temperature for 3 hours. The mixture was acidified to pH3 with aqueous HCl (4M) at 0 ℃. The resulting mixture was diluted with water and extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (2×200 mL) and dried (Na 2SO 4). After filtration, the filtrate was concentrated in vacuo to give the sub-title compound (ADP-3) (2.1 g,6.11mmol,95%,85% purity) as an off-white solid. M/z 293.0/295.0 (M+H) + (ES+).
Step 3: synthesis of 5-cyano-2- (2, 6-dichloropyridin-4-yl) -N- [ (methylaminomethylsulfonyl) amino ] benzamide (ADP-4)
To the product (ADP-3) (2.10 g,1 equivalent, 7.17 mmol), 4-methyl-3-thiosemicarbazide (D-2) (753 mg,1 equivalent, 7.17 mmol), T from step 2 above at room temperature 3 P (50% in DMF) (9.12 g,50wt%,4 equivalents, 28.7 mmol) and DIPEA (5.56 g,6 equivalents, 43.0 mmol) in DMF (20 mL). The resulting mixture was stirred at room temperature for 3 hours. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 40% in 24 minutes); detector, UV 254/220nm. This gave the sub-title compound (ADP-4) (1.9 g,4.51mmol,70%,90% purity) as an off-white solid. M/z 380.0/382.0 (M+H) + (ES+)。
Step 4: synthesis of 4- (2, 6-dichloropyridin-4-yl) -3- (4-methyl-5-sulfonyl-1, 2, 4-triazol-3-yl) benzonitrile (ADP-5)
The product (ADP-4) (1.90 g,1 equivalent, 5.00 mmol) from step 3 above was taken up in NaHCO 3 The solution in water (20 mL, 1M) was stirred at 80℃for 1 hour. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 40% in 25 minutes); detector, UV 254/220nm. This gave the sub-title compound (ADP-5) (1.3 g,3.42mmol,61%,95% purity) as an off-white solid. M/z 362.0/364.0 (M+H) + (ES+)。
Step 5: synthesis of 4- (2, 6-dichloropyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADP-6)
To the product (ADP-5) (1.30 g,1 equivalent, 3.59 mmol) from step 4 above, H at room temperature 2 O 2 (610 mg,30Wt%,5 equivalents, 17.9 mmol), acOH (431 mg,2 equivalents, 7.18 mmol) in DCM (20 mL). The resulting mixture was stirred at room temperature for 2 hours. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 45% in 25 minutes); detector, UV 254/220nm. This gave the sub-title compound (ADP-6) (750 mg,2.05mmol,63% purity) as an off-white solid. M/z 330.0/332.0 (M+H) + (ES+)。
Step 6: synthesis of 4- [ 2-chloro-6- (ethylamino) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADP-7)
To the product (ADP-6) (350 mg,1 equivalent, 1.06 mmol) from step 5 above, etOH at room temperature 2 -HCl (864 mg,10 equivalents, 10.6 mmol) and K 2 CO 3 (1.46 g,10 equivalents, 10.6 mmol) in NMP (20 mL). The resulting mixture was stirred at 120℃for 6 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 50% in 27 minutes); detector, UV 254/220nm. This gave the sub-title compound (ADP-7) (260 mg,0.71mmol,72%,92% purity) as an off-white solid. M/z339.1/341.1 (M+H) + (ES+)。
Step 7: synthesis of 4- [2- (ethylamino) -6- [ 1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADP-8)
To the product (ADP-7) (40 mg,1 equivalent, 118. Mu. Mol), 4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (AC-2) (29 mg,1.2 equivalent, 142. Mu. Mol) and K from step 6 above under a nitrogen atmosphere at room temperature 3 PO 4 (75 mg,3 eq., 354. Mu. Mol) to a stirred solution of 1, 4-dioxane (5 mL) was added Pd (OAc) 2 (2.65 mg,0.1 eq, 12. Mu. Mol) and XantPhos (14 mg,0.2 eq, 24. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (40% MeCN up to 80% in 20 minutes); detector, UV 254/220nm. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN;flow rate: 60 ml/min; gradient: 46% B to 62% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (ADP-8) (7.7 mg, 15. Mu. Mol,13%,98% purity) as an off-white solid. M/z 504.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.51(s,1H),8.17-8.04(m,3H),7.98(d,J=7.8Hz,1H),7.90(d,J=8.1Hz,1H),7.76(t,J=7.7Hz,1H),7.53(s,1H),6.08(s,1H),5.24(s,2H),3.48(s,3H),3.31-3.25(m,2H),1.23(t,J=7.2Hz,3H)。
Example 142: synthesis of 2- (4-methyl-1, 2, 4-triazol-3-yl) -3'- (6- { [ (3S) -3-methylpiperidin-1-yl ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) - [1,1' -biphenyl ] -4-carbonitrile (ADQ-2)
Step 1:3'- (6-formyl-1-oxo-4- (trifluoromethyl) isoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile
The reaction was carried out in a similar manner to step 2 for the synthesis of BE-3 (example 122).
Step 2: synthesis of 2- (4-methyl-1, 2, 4-triazol-3-yl) -3'- (6- { [ (3S) -3-methylpiperidin-1-yl ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) - [1,1' -biphenyl ] -4-carbonitrile (ADQ-2)
To a stirred solution of intermediate (ADQ-1) (50 mg,1 eq, 103. Mu. Mol) and (S) -3-methylpiperidine, HCl (R-1) (31 mg,3 eq, 309. Mu. Mol) in DCM (10 mL) at room temperature was added DIPEA (16 mg, 124. Mu. Mol,1.2 eq) and NaBH (OAc) 3 (44 mg,2 equivalents, 206. Mu. Mol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was quenched with MeOH (1 mL) at 0 ℃ and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 65% B in 9 minutes; wavelength: 254/220 nm) to obtain a displayThe title compound (ADQ-2) (23.5 mg, 41. Mu. Mol,40%,99% purity) was obtained as a white solid. M/z 571.4 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.47(s,1H),8.24–8.17(m,1H),8.13(d,J=1.7Hz,1H),8.08–8.01(m,1H),7.98(s,1H),7.96–7.86(m,2H),7.72(t,J=2.0Hz,1H),7.42(t,J=8.0Hz,1H),6.97–6.90(m,1H),5.11(s,2H),3.65(s,2H),3.17(s,3H),2.77–2.68(m,2H),1.99–1.89(m,1H),1.70–1.56(m,4H),1.52–1.44(m,1H),0.92–0.80(m,4H)。
Example 143: synthesis of 2- { 6-cyclopropyl-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one) (ADR-3)
Step 1: synthesis of 2-chloro-6-cyclopropyl-4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridine (ADR-1)
Intermediate (AAN-6) (30 mg,1 eq, 97. Mu. Mol), cyclopropylboronic acid (ABF-2) (14 mg,1.7 eq, 165. Mu. Mol) and K were added to the mixture under a nitrogen atmosphere at room temperature 3 PO 4 (41 mg,2 eq, 194. Mu. Mol) Pd (dppf) Cl was added to a stirred mixture of 1, 4-dioxane (1 mL) 2 DCM (7.1 mg,0.1 eq, 10. Mu. Mol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 50% within 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (ADR-1) (20 mg, 57. Mu. Mol,65%,90% purity) as a white solid. M/z 315.1/317.1 (M+H) + (ES+)。
Step 2: synthesis of 6- ((((1-hydroxycyclobutyl) methyl) amino) methyl) -4- (trifluoromethyl) isoindolin-1-one (ADR-2)
1- (aminomethyl) cyclobutan-1-ol (AW-1) (33 mg,1.5 asAmounts of 327. Mu. Mol) and DIPEA (85 mg,3 eq., 654. Mu. Mol) in MeOH (6 mL) were stirred at room temperature for 30 min. Intermediate (ABC-4) (50 mg,1 eq, 218. Mu. Mol) was added to the above mixture at room temperature. NaBH was added to the above mixture at room temperature over 2 hours 4 (41 mg,5 equivalents, 1.09 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by addition of 2mL of ice water at 0 ℃. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). The residue was purified by preparative TLC using DCM/petroleum ether (1/1) to give the sub-title compound (ADR-2) (36 mg,105 μmol,51%,92% purity) as an off-white solid. M/z 315.1 (M+H) + (ES+)。
Step 3: synthesis of 2- { 6-cyclopropyl-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one) (ADR-3)
To the product (ADR-2) (100 mg,1 equivalent, 318. Mu. Mol) from the above step 2, the product (ADR-1) (100 mg,1 equivalent, 318. Mu. Mol) from the above step 1, and Cs under a nitrogen atmosphere at room temperature 2 CO 3 (207 mg,2 eq. 636. Mu. Mol) to a stirred solution of RuPhos ring palladium complex Gen.3 (53 mg,0.2 eq., 64. Mu. Mol) and RuPhos (59 mg,0.4 eq., 127. Mu. Mol) in 1, 4-dioxane (15 mL) were added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 12% B to 23% B in 11 min; wavelength: 254/220nm; retention time: 13.1) to give the title compound (ADR-3) (64.1 mg, 106. Mu. Mol) as a white solid 34%,98% purity). M/z 593.4 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.49(s,1H),8.26–8.15(m,2H),8.07(d,J=10.7Hz,2H),7.97(s,1H),7.23(d,J=1.4Hz,1H),5.21(s,2H),3.98(s,2H),3.91(s,3H),3.55(s,3H),2.58–2.53(m,2H),2.15(p,J=6.4Hz,1H),2.04–1.97(m,2H),1.96–1.85(m,2H),1.69–1.57(m,1H),1.44–1.32(m,1H),1.03(d,J=6.3Hz,4H)。
Example 144: synthesis of 3- [2- (ethylamino) -6- [ 1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -4- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADS-6)
Step 1: synthesis of 4-cyano-2- (2, 6-dichloropyridin-4-yl) benzoic acid (ADS-1)
To 2-bromo-4-cyanobenzoic acid (ADM-1) (2.00 g,1 equivalent, 8.85 mmol), 2, 6-dichloropyridin-4-ylboronic acid (AAN-1) (1.19 g,0.7 equivalent, 6.19 mmol) and K under a nitrogen atmosphere at room temperature 3 PO 4 (3.67 g,3 eq, 26.5 mmol) Pd (dtBPF) Cl was added to a stirred solution of 1, 4-dioxane (20 mL) and water (1 mL) 2 (577 mg,0.1 eq, 0.89 mmol). The resulting mixture was stirred under nitrogen at 60 ℃ for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and MeCN (50% MeCN up to 70% in 10 min); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ADS-1) (350 mg,1.08mmol,14%,90% purity) as a yellow solid. M/z 293.0/295.0 (M+H) + (ES+)。
Step 2: 4-cyano-2- (2, 6-dichloropyridin-4-yl) -N- [ (methylamino methylsulfonyl) amino ] benzamide (ADS-2)
At room temperature, to the reaction mixture from step 1To a stirred solution of product (ADS-1) (350 mg,1 eq, 1.19 mmol) and 4-methyl-3-thiosemicarbazide (D-2) (151 mg,1.2 eq, 1.43 mmol) in DMF (5 mL) was added T 3 P (50% in EA) (2.28 g,50wt%,6 eq., 7.16 mmol). The resulting mixture was stirred at room temperature for 2 hours. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and MeCN (52% MeCN up to 73% in 10 min); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ADS-2) (330 mg,0.80mmol,73%,92% purity) as a yellow solid. M/z 380.0/382.0 (M+H) + (ES+)。
Step 3: synthesis of 3- (2, 6-dichloropyridin-4-yl) -4- (4-methyl-5-sulfonyl-1, 2, 4-triazol-3-yl) benzonitrile (ADS-3)
To an 8mL vial at room temperature was added the product (ADS-2) (330 mg,1 eq, 0.87 mmol) from step 2 above and NaHCO 3 (6 mL, 1M). The resulting mixture was stirred at 80℃for 1 hour. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (44% MeCN up to 65% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ADS-3) (300 mg,0.79mmol,95% purity) as a yellow solid. M/z 362.0/364.0 (M+H) + (ES+)。
Step 4: synthesis of 3- (2, 6-dichloropyridin-4-yl) -4- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADS-4)
To a stirred solution of the product from step 3 above (ADS-3) (300 mg,1 eq, 0.83 mmol) and AcOH (100 mg,2 eq, 1.66 mmol) in DCM (10 mL) at 0deg.C was added H 2 O 2 (469.7 mg,30Wt%,5 equivalents, 4.15 mmol). The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and MeCN (56% MeCN up to 83% in 10 min); detector, UV 254220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ADS-4) (150 mg,0.41mmol,50%,91% purity) as a yellow solid. M/z 330.0/332.0 (M+H) + (ES+)。
Step 5: synthesis of 3- [ 2-chloro-6- (ethylamino) pyridin-4-yl ] -4- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADS-5)
To the product (ADS-4) from step 4 above (100 mg,1 eq, 0.30 mmol) and EtOH under nitrogen at room temperature 2 To a stirred solution of HCl (741mg, 30 equivalents, 9.09 mmol) in NMP (10 mL) was added K 2 CO 3 (1.26 g,30 equivalents, 9.09 mmol). The resulting mixture was stirred overnight at 110 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and MeCN (60% MeCN up to 80% in 10 min); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ADS-5) (80 mg,0.21mmol,78%,90% purity) as a yellow oil. M/z 339.1/341.1 (M+H) + (ES+)
Step 6: synthesis of 3- [2- (ethylamino) -6- [ 1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -4- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADS-6)
To the product (ADS-5) (80 mg,1 eq, 0.24 mmol), intermediate (AC-2) (57 mg,1.2 eq, 0.28 mmol) and K from step 5 above, under nitrogen at room temperature 3 PO 4 (150 mg,3 eq, 0.71 mmol) to a stirred solution of 1, 4-dioxane (3 mL) was added Pd (OAc) 2 (5.3 mg,0.1 eq, 24. Mu. Mol) and XantPhos (27 mg,0.2 eq, 47. Mu. Mol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10 minInner 36% MeCN up to 70%); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 65% B in 9 minutes; wavelength: 254/220nm; retention time: 8.48 To give the title compound (ADS-6) (19.8 mg,39 μmol,17%,99% purity) as a white solid. M/z 504.0 (M+H) + (ES+) 1 H NMR(400MHz,MeOH-d4)δ8.51(s,1H),8.11–8.07(m,2H),8.04–7.96(m,2H),7.86–7.73(m,2H),7.51(d,J=1.3Hz,1H),6.09(d,J=1.3Hz,1H),5.24(s,2H),3.48(s,3H),3.29(d,J=7.2Hz,2H),1.24(t,J=7.2Hz,3H)。
Example 145: synthesis of 4- [2- (ethylamino) -6- (6- { [ (3S) -3-methylpiperidin-1-yl ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADT-2)
Step 1: synthesis of 6- { [ (3S) -3-methylpiperidin-1-yl ] methyl } -4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (ADT-1)
To an 8mL sealed tube was added intermediate (ABC-4) (100 mg,1 eq, 0.44 mmol), et at room temperature 3 N (265 mg,6 eq, 2.62 mmol) and (S) -3-methylpiperidine, HCl (R-1) (43 mg,1 eq, 0.44 mmol) DCM (3 mL). NaBH (OAc) was added to the above mixture at 0deg.C over 10 minutes 3 (555 mg,6 equivalents, 2.62 mmol). The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 50% in 15 minutes); detector, UV 254/220nm, to give the sub-title compound (ADT-1) (41 mg,0.12mmol,30%,90% purity) as a yellow oil. M/z 313.1 (M+H)) + (ES+)。
Step 2: synthesis of 4- [2- (ethylamino) -6- (6- { [ (3S) -3-methylpiperidin-1-yl ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADT-2)
To the product (ADT-1) (70 mg,1 equivalent, 0.22 mmol), intermediate (ADP-7) (75.93 mg,1 equivalent, 0.22 mmol) and Cs from step 1 above, under a nitrogen atmosphere at room temperature 2 CO 3 (146 mg,2 eq, 0.45 mmol) to a stirred solution of RuPhos (42 mg,0.4 eq, 90. Mu. Mol) and RuPhos cyclopalladium complex Gen.3 (37 mg,0.2 eq, 45. Mu. Mol) in 1, 4-dioxane (8 mL) were added. The final reaction mixture was irradiated with microwave radiation at 100 ℃ under nitrogen atmosphere for 1 hour. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/1) to give the crude product. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 10% B in 7 minutes; wavelength: 254/220nm; retention time: 6.2 To give the title compound (ADT-2) (9.9 mg,16 μmol,7.1%,99% purity) as a yellow solid. M/z 615.4 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.51 (s, 1H), 8.11-8.02 (m, 3H), 7.96 (s, 1H), 7.90 (d, j=8.1 hz, 1H), 7.52 (d, j=1.2 hz, 1H), 6.08 (d, j=1.3 hz, 1H), 5.23-5.19 (m, 2H), 3.69 (s, 2H), 3.48 (s, 3H), 3.29 (t, j=7.2 hz, 2H), 2.90-2.78 (m, 2H), 2.07-1.98 (m, 1H), 1.81-1.56 (m, 5H), 1.23 (t, j=7.2 hz, 3H), 0.95-0.89 (m, 4H).
Example 146: synthesis of 4- [2- (ethylamino) -6- (6- { [ (3R) -3-methylpiperidin-1-yl ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADU-2)
Step 1: synthesis of 6- { [ (3R) -3-methylpiperidin-1-yl ] methyl } -4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (ADU-1)
To a stirred solution of (3R) -3-methylpiperidine, HCl (S-1) (57 mg,1.2 eq, 0.42 mmol) and DIPEA (68 mg,1.5 eq, 0.52 mmol) in DCM (10 mL) at room temperature. The resulting mixture was stirred at room temperature for 20 minutes. Intermediate (ABC-4) (80 mg,1 eq, 0.35 mmol) was added to the above mixture at room temperature. The resulting mixture was stirred at room temperature for an additional 1 hour. NaBH (OAc) was added to the above mixture at room temperature 3 (370 mg,5 eq, 1.75 mmol). The resulting mixture was stirred at room temperature overnight. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 60% in 24 minutes); detector, UV 254/220nm. This gave the sub-title compound (ADU-1) (60 mg,0.18mmol,55%,92% purity) as an off-white solid. M/z 313.1 (M+H) + (ES+)。
Step 2: synthesis of 4- [2- (ethylamino) -6- (6- { [ (3R) -3-methylpiperidin-1-yl ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADU-2)
To the product (ADU-1) (60 mg,1 equivalent, 0.19 mmol), intermediate (ADP-7) (72 mg,1.1 equivalent, 0.21 mmol) and Cs from step 1 above were added under nitrogen at room temperature 2 CO 3 (125 mg,2 eq, 0.38 mmol) to a stirred solution of RuPhos ring palladium complex gen.3 (32 mg,0.2 eq, 38. Mu. Mol) and RuPhos (36 mg,0.4 eq, 77. Mu. Mol) in 1, 4-dioxane (10 mL). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (20% MeCN up to 100% in 25 minutes); detector, UV 254/220nm. The crude product was purified by preparative HPLC under the following conditions (column: XBIdge-prepared OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 65% B to 78% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (ADU-2) (27.3 mg, 44. Mu. Mol,23%,99% purity) as a pale yellow solid. M/z 615.0 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.50(s,1H),8.13–7.87(m,5H),7.52(d,J=1.3Hz,1H),6.08(d,J=1.3Hz,1H),5.21(s,2H),3.70(s,2H),3.48(s,3H),3.29(t,J=7.2Hz,2H),2.84(t,J=12.1Hz,2H),2.06–1.92(m,1H),1.86–1.51(m,5H),1.23(t,J=7.2Hz,3H),0.89(d,J=5.4Hz,4H)。
Example 147: synthesis of 3'- [6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (ADV-1)
A solution of intermediate (ADQ-1) (mg, 1 equivalent, 0.10 mmol) and 1- (aminomethyl) cyclobutan-1-ol (AW-1) (31 mg,3 equivalent, 0.31 mmol) in MeOH (10 mL) was stirred at 60℃for 1 hour. Adding NaBH to the mixture at 0deg.C 4 (8 mg,2 equivalents, 0.21 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was carried out at 0℃with H 2 O (1 mL) was quenched and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1). The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 5% B to 25% B over 10 min; wavelength: 254/220 nm) to give the title compound (ADV-1) as a white solid (17.5 mg,30 μmol,30%,99% purity). M/z 573.0 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO–d6)δ8.46(s,1H),8.21(d,J=1.8Hz,1H),8.19(d,J=1.9Hz,1H),8.13(d,J=1.8Hz,1H),8.05(d,J=12.6Hz,3H),7.90(d,J=8.1Hz,1H),7.73(d,J=2.4Hz,1H),7.42(t,J=8.0Hz,1H),6.93(d,J=7.7Hz,1H),5.10(s,2H),4.97(s,1H),3.98(s,2H),3.17(s,3H),2.55(s,2H),2.06–1.86(m,4H),1.66–1.59(m,1H),1.48–1.34(m,1H)。
Example 148: synthesis of N- [ 2-amino-5- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -3- (trifluoromethyl) phenyl ] -4' -cyano-2 ' - (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-carboxamide (ADW-5)
Step 1: synthesis of tert-butyl 4' -cyano-2 ' - (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-carboxylate (ADW-2)
K was added to a solution of intermediate (ADG-1) (160 mg,1 eq, 608. Mu. Mol) and (3- (tert-butoxycarbonyl) phenyl) boronic acid (ADW-1) (135 mg,1 eq, 608. Mu. Mol) in dioxane (5 mL) and water (1 mL) at room temperature under a nitrogen atmosphere 2 CO 3 (252 mg,3 eq., 1.82 mmol) and Pd (dppf) Cl 2 DCM (45 mg,0.1 eq, 61. Mu. Mol). After stirring at 80 ℃ for 3 hours under nitrogen atmosphere, the mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (12/1) to give the sub-title compound (ADW-2) as a yellow solid (150 mg, 417. Mu. Mol, 68%). M/z 361.2 (M+H) + (ES+)。
Step 2: synthesis of 4' -cyano-2 ' - (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-carboxylic acid (ADW-3)
To a stirred solution of the product from step 1 above (ADW-2) (145 mg,1 eq, 402 μmol) in DCM (3 mL) was added TFA (1 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 305.1 (M+H) + (ES+)。
Step 3: synthesis of N- [ 2-amino-5- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -3- (trifluoromethyl) phenyl ] -4' -cyano-2 ' - (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-carboxamide (ADW-5)
To the product (ADW-3) (20 mg,1 equivalent, 66. Mu. Mol) from step 2 above and (1S, 2R) -2- ({ [3, 4-diamino-5- (trifluoromethyl) phenyl) at room temperature]To a stirred solution of methyl } amino) cyclopentan-1-ol (ADW-4) (19 mg,1 eq, 66 μmol) in pyridine (1.5 mL) was added EDCI (26 mg,2 eq, 0.13 mmol). The resulting mixture was stirred at 60℃for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: MECN; flow rate: 60 ml/min; gradient: 28% B to 38% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (ADW-5) (2.9 mg, 5.0. Mu. Mol,7.7%,99% purity) as a white solid. M/z 576.0 (M+H) + (ES+) 1 H NMR(400MHz,DMSO-d6)δ9.84(s,1H),8.49(s,1H),8.24–8.17(m,1H),8.14(d,J=1.7Hz,1H),8.03–7.91(m,3H),7.48–7.30(m,3H),7.16(d,J=7.7Hz,1H),5.19(s,2H),4.39(s,1H),3.99–3.91(m,1H),3.70–3.54(m,2H),3.16(s,3H),2.77(d,J=5.8Hz,1H),1.74–1.48(m,4H),1.46–1.31(m,2H)。
Example 149: synthesis of 4- [2- (ethylamino) -6- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADX-2)
Step 1: synthesis of 6- (hydroxymethyl) -4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (ADX-1)
Intermediate (ABC-4) (90 mg,1 eq, 0.39 mmol) and NaBH at room temperature 4 (45 mg,3 eq, 1.18 mmol) in MeOH (10 mL). The resulting mixture was stirred at room temperature for 2 hours. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0).1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 40% in 24 minutes); detector, UV 254/220nm. This gave the sub-title compound (ADX-1) (70 mg,0.29mmol,77% purity) as an off-white solid. M/z 232.1 (M+H) + (ES+)。
Step 2: synthesis of 4- [2- (ethylamino) -6- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADX-2)
To the product (ADX-1) (50 mg,1 equivalent, 0.22 mmol), intermediate (ADP-7) (88 mg,1.2 equivalent, 0.26 mmol) and K from step 1 above were added under nitrogen at room temperature 3 PO 4 (138 mg,3 eq, 0.65 mmol) in 1, 4-dioxane (5 mL) was added Xantphos (25 mg,0.2 eq, 43. Mu. Mol) and Pd (OAc) 2 (5 mg,0.1 eq, 22. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 6 hours. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (0% MeCN up to 50% in 25 minutes); detector, UV 254/220nm. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 33% B to 44% B in 11 minutes; wavelength: 254/220 nm) to give the title compound (ADX-2) (7.2 mg, 13. Mu. Mol,6.2%,99% purity) as a pale yellow solid. M/z 534.2 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.50(s,1H),8.16–8.04(m,3H),7.98–7.87(m,2H),7.53(d,J=1.3Hz,1H),6.08(d,J=1.3Hz,1H),5.22(s,2H),4.81(s,2H),3.48(s,3H),3.29(t,J=7.2Hz,2H),1.23(t,J=7.2Hz,3H)。
Example 150: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- [ (2-hydroxy-2-methylpropyloxy) methyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (ADY-2)
To a stirred solution of intermediate (ADA-1) (40 mg,1 eq, 75 μmol) and 2-methyl-propane-1, 2-diol (ADY-1) (68 mg,10 eq, 0.75 mmol) in DCM (5 mL) was added TBAB (13 mg,0.5 eq, 37 μmol) and aqueous KOH (5 mL,20 wt%) at room temperature. The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (8/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35% B to 46% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (ADY-2) (3.6 mg, 6.1. Mu. Mol,8.2%,99% purity) as a white solid. M/z 585.0 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH–d4)δ8.49(s,1H),8.09(s,1H),8.00(s,1H),7.87(s,1H),7.66(d,J=1.2Hz,1H),6.20(d,J=1.2Hz,1H),5.27(s,2H),4.77(s,2H),4.04(s,3H),3.50(s,3H),3.43–3.32(m,4H),1.33–1.20(m,9H)。
Example 151: synthesis of 2- { 6-cyclopropyl-4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (ADZ-2)
Step 1: synthesis of 2-chloro-6-cyclopropyl-4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -pyridine (ADZ-1)
To a stirred solution of intermediate (E-7) (450 mg,1 eq, 1.47 mmol) and cyclopropylboronic acid (ABF-2) (177 mg,1.4 eq, 2.06 mmol) in toluene (15 mL) under nitrogen at room temperature was added potassium phosphate (939 mg,3 eq, 4.45 mmol), PCy 3 (62 mg,0.1 eq, 220)Mu mol) and Pd (AcO) 2 (50 mg,0.1 eq, 220. Mu. Mol). The resulting mixture was stirred under nitrogen at 110 ℃ for 16 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: c18 silica gel; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (20% MeCN up to 40% in 10 minutes); detector, UV 254nm. This gave the sub-title compound (ADZ-1) (120 mg, 387. Mu. Mol, 26%) as a white solid. M/z 311.1/313.1 (M+H) + (ES+)。
Step 2: synthesis of 2- (6-cyclopropyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((((1-hydroxycyclobutyl) methyl) amino) methyl) -4- (trifluoromethyl) isoindolin-1-one (ADZ-2)
To intermediate (ADR-2) (40 mg,1 equivalent, 0.13 mmol), the product from step 1 above (ADZ-1) (40 mg,1 equivalent, 0.13 mmol) and Cs under nitrogen at room temperature 2 CO 3 (83 mg,0.25mmol,2 eq.) to a stirred solution of RuPhos (24 mg,0.4 eq., 51. Mu. Mol) and RuPhos cyclopalladium complex Gen.3 (21 mg,0.2 eq., 25. Mu. Mol) in 1, 4-dioxane (1 mL) were added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19X 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30B to 60B in 7 minutes; a detector, UV 254/210nm; retention time: 6.08. this gave the title compound (ADZ-2) (17.3 mg, 29. Mu. Mol,23%,98% purity) as a white solid. M/z 589.4 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.20(s,1H),8.14(s,1H),8.08(d,J=1.4Hz,1H),7.84–7.76(m,1H),7.80–7.70(m,2H),7.73–7.63(m,2H),6.94(d,J=1.4Hz,1H),5.24(s,2H),4.33(s,2H),3.45(s,3H),3.06(s,2H),2.22–2.12(m,4H),2.12–2.01(m,1H),1.85–1.73(m,1H),1.65–1.53(m,1H),1.02(d,J=6.4Hz,4H)。
Example 152: synthesis of 3'- (6- { [ (2-hydroxyethyl) amino ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AEA-2)
A solution of intermediate (ADQ-1) (20 mg,1 equivalent, 41. Mu. Mol) and ethanolamine (AEA-1) (26 mg,10 equivalent, 0.41 mmol) in MeOH (5 mL) was stirred at 60℃for 0.5 h. Adding NaBH to the mixture at 0deg.C 4 (4 mg,2 equivalents, 82. Mu. Mol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was carried out at 0℃with H 2 O (1 mL) was quenched and concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (8/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 28% B to 37% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (AEA-2) (10.0 mg, 18. Mu. Mol,45%,97% purity) as a white solid. M/z 533.0 (M+H) + (ES+). 1 H NMR(400MHz,MeOH–d4)δ8.44(s,1H),8.15–8.08(m,2H),8.08–8.00(m,2H),7.97–7.90(m,2H),7.85–7.79(m,1H),7.49(t,J=8.0Hz,1H),7.16–7.08(m,1H),5.08(s,2H),4.01(s,2H),3.71(t,J=5.6Hz,2H),3.24(s,3H),2.77(t,J=5.6Hz,2H)。
Example 153: synthesis of 3'- (6- (((2-methoxyethyl) amino) methyl) -1-oxo-4- (trifluoromethyl) isoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AEB-2)
/>
A solution of intermediate (ADQ-1) (20 mg,1 eq, 41. Mu. Mol) and 2-methoxyethylamine (AEB-1) (31 mg,10 eq, 0.41 mmol) in MeOH (5 mL) was stirred at 60℃for 0.5 h. Adding NaBH to the mixture at 0deg.C 4 (4 mg,2 equivalents, 82. Mu. Mol). The resulting mixture was stirred at room temperature for 1 hour. Will beReaction with H at 0deg.C 2 O was quenched and concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (8/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 34% B to 44% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AEB-2) (5.4 mg, 9.4. Mu. Mol,23%,95% purity) as a white solid. M/z 547.0 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH–d4)δ8.44(s,1H),8.14–8.08(m,2H),8.07–8.00(m,2H)7.97–7.90(m,2H),7.83(t,J=2.0Hz,1H),7.49(t,J=8.0Hz,1H),7.12(d,J=7.6Hz,1H),5.09(s,2H),4.00(s,2H),3.55(t,J=5.3Hz,2H),3.38(s,3H),3.24(s,3H),2.80(t,J=5.3Hz,2H)。
Example 154: synthesis of 4- [2- (ethylamino) -6- (6- { [ (2-methoxyethyl) amino ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEC-2)
Step 1: synthesis of 6- { [ (2-methoxyethyl) amino ] methyl } -4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (AEC-1)
2-methoxyethyl-1-amine (AEB-1) (52 mg, 0.52. Mu. Mol,1.2 eq.) and Et 3 A stirred solution of N (66 mg,1.5 eq, 0.65 mmol) in DCM (5 mL) was stirred at room temperature for 10 min. Intermediate (ABC-4) (100 mg,1 eq, 0.44 mmol) was added to the above mixture at room temperature. The resulting mixture was stirred at room temperature for 2 hours. NaBH (OAc) was added to the above mixture at room temperature 3 (463mg, 5 eq, 2.18 mmol). The resulting mixture was stirred at room temperature overnight. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 50% in 20 minutes); detector, UV 254/220nm. This gives the subtitle as an off-white solidCompound (AEC-1) (80 mg,0.26mmol,64%,94% purity). M/z 289.1 (M+H) + (ES+)。
Step 2:4- [2- (ethylamino) -6- (6- { [ (2-methoxyethyl) amino ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEC-2)
To intermediate (ADP-7) (40 mg,1 equivalent, 0.12 mmol), the product from step 1 above (AEC-1) (41 mg,1.2 equivalent, 0.14. Mu. Mol) and Cs under nitrogen at room temperature 2 CO 3 (77 mg,2 eq, 0.24 mmol) to a stirred solution of RuPhos ring palladium complex gen.3 (20 mg,0.2 eq, 24. Mu. Mol) and RuPhos (22 mg,0.4 eq, 47. Mu. Mol) in 1, 4-dioxane (5 mL) was added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 50% in 20 minutes); detector, UV 254/220nm. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 50% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (AEC-2) (10 mg, 16. Mu. Mol,14%,96% purity) as a pale yellow solid. M/z 591.5 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),8.13–8.01(m,4H),7.90(d,J=8.1Hz,1H),7.53(d,J=1.3Hz,1H),6.08(d,J=1.3Hz,1H),5.22(s,2H),4.07(s,2H),3.58(t,J=5.3Hz,2H),3.47(s,3H),3.39(s,3H),3.29(t,J=7.2Hz,2H),2.89(t,J=5.3Hz,2H),1.23(t,J=7.2Hz,3H)。
Example 155: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- { [ (1-hydroxycyclobutyl) methoxy ] methyl } -4- (trifluoromethyl) -3H-isoindol-1-one (AED-2)
To a stirred solution of intermediate (ADA-1) (46 mg,1 eq, 87 μmol) and 1- (hydroxymethyl) cyclobutan-1-ol (AED-1) (89 mg,10 eq, 0.87 mmol) in DCM (5 mL) was added TBAB (14 mg,0.5 eq, 43 μmol) and aqueous KOH (5 mL,20 wt%) at room temperature. The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (8/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 41% B to 51% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AED-2) (4.8 mg, 8.0. Mu. Mol,9.3%,99% purity) as a white solid. M/z 597.1 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH–d4)δ8.50–8.44(m,1H),8.08(s,1H),8.00(s,1H),7.84(s,1H),7.64(d,J=1.2Hz,1H),6.18(d,J=1.2Hz,1H),5.25(s,2H),4.78(s,2H),4.01(s,3H),3.57(s,2H),3.48(s,3H),3.40–3.32(m,2H),2.21–2.11(m,2H),2.11–1.99(m,2H),1.84–1.70(m,1H),1.63–1.51(m,1H),1.23(t,J=7.2Hz,3H)。
Example 156: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AEE-2)
Step 1: synthesis of 6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -2, 3-dihydro-isoindol-1-one (AEE-1)
To a stirred solution of intermediate (ACY-2) (170 mg,1 eq, 1.06 mmol) and 1- (aminomethyl) cyclobutan-1-ol (AW-1) (213 mg,2 eq, 2.11 mmol) in MeOH (10 mL) at room temperature under nitrogen was added NaBH 3 CN (265 mg,4 equivalents, 4.22 mmol) and AcOH (63 mg,1 equivalents, 1.06 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 40% in 15 minutes); detector, UV 254/220nm. This gave the sub-title compound (AEE-1) (120 mg,0.46mmol,46%,95% purity) as a white solid. M/z 247.1 (M+H) + (ES+)。
Step 2: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AEE-2)
Intermediate (ABC-6) (50 mg,1 eq., 0.16 mmol), product (AEE-1) from step 1 above (39 mg,1 eq., 0.16 mmol) and K were added to the mixture under nitrogen at room temperature 3 PO 4 (67 mg,0.31mmol,2 eq.) in 1, 4-dioxane (5 mL) was added XantPhos (18 mg,0.2 eq., 31. Mu. Mol) and Pd (OAc) 2 (3.5 mg,0.1 eq, 16. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and MeCN (5% MeCN up to 50% in 15 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: 30 x 150mm,5 μm for Sunfire preparative C18 column; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 10% B to 35% B in 10 minutes; a detector, UV 254/220nm; retention time: 6.7. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AEE-2) (10.3 mg,19 μmol,12%,98 purity) as a white solid. M/z 528.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.47(s,1H),8.24(s,2H),7.88(s,1H),7.76(s,1H),7.66(d,J=1.2Hz,2H),7.57(d,J=1.2Hz,1H),6.86(t,J=5.4Hz,1H),6.20(d,J=1.2Hz,1H),5.05(s,2H),3.89(s,5H),3.53–3.42(m,4H),2.55(s,3H),2.45(s,2H),2.05–1.83(m,2H),1.64–1.57(m,1H),1.44–1.32(m,1H),1.26–1.10(m,3H)。
Example 157: synthesis of 2- (6- (ethylamino) -4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (AEF-1)
To intermediate (ACM-7) (50 mg,1 equivalent, 0.15 mmol), intermediate (ADX-1) (35 mg,1 equivalent, 0.15 mmol) and Cs 2 CO 3 (98 mg,2 eq, 0.30 mmol) to a stirred mixture of 1, 4-dioxane (3 mL) was added Ruphos (28 mg,0.4 eq, 60. Mu. Mol) and Ruphos ring palladium complex Gen.3 (25 mg,0.2 eq, 30. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water. The resulting mixture was extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 42% B to 52% B, UV 254/220nm in 8 minutes; retention time: 7.07. this gave the title compound (AEF-1) (29.6 mg, 56. Mu. Mol,37%,99% purity) as an off-white solid. M/z 527.0 (M+H) + (ES+) 1 H NMR(400MHz,DMSO-d6)δ8.50(s,1H),7.97(d,J=20.4Hz,2H),7.65(d,J=8.6,5.8Hz,1H),7.57(td,J=8.5,2.7Hz,1H),7.53–7.50(m,1H),7.46(d,J=1.2Hz,1H),6.71(t,J=5.4Hz,1H),5.92(d,J=1.2Hz,1H),5.59(t,J=5.8Hz,1H),5.16(s,2H),4.70(d,J=5.4Hz,2H),3.38(s,3H),3.21–3.14(m,2H),1.13(t,J=7.2Hz,3H)。
Example 158: synthesis of 4- { 2-cyclopropyl-6- [ 1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEG-3)
Step 1: synthesis of 4- (2-chloro-6-cyclopropylpyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEG-2)
Pd (PPh) was added to a stirred mixture of intermediate (ADP-6) (80 mg,1 eq, 0.24 mmol) and bromine (cyclopropyl) zinc (AEG-1) (226 mg,5 eq, 1.21 mmol) in THF (3 mL) at room temperature under nitrogen atmosphere 3 ) 4 (56 mg,0.2 eq, 48. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The reaction was then quenched by addition of 2mL of ice water at 0 ℃. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 60% in 25 minutes); detector, UV 254/220nm to give the sub-title compound (AEG-2) (25 mg,69 μmol,31%,93% purity) as a white solid. M/z336.1/338.1 (M+H) + (ES+)。
Step 2: synthesis of 4- { 2-cyclopropyl-6- [ 1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEG-3)
To the product (AEG-2) (13 mg,1 equivalent, 39. Mu. Mol), intermediate (AC-2) (9.3 mg,1.2 equivalent, 47. Mu. Mol) and Cs from step 1 above, under a nitrogen atmosphere at room temperature 2 CO 3 (25 mg,2 eq, 78. Mu. Mol) to a stirred solution of RuPhos (7.2 mg,0.4 eq, 16. Mu. Mol) and RuPhos cyclopalladium complex Gen.3 (6.5 mg,0.2 eq, 8. Mu. Mol) in 1, 4-dioxane (2 mL) were added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 1 hour. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 ) Mobile phase B: MECN; flow rate: 60 ml/min; gradient: 50% B to 60% B within 9 minutes; wavelength: 254/220nm; retention time: 825) to give the title compound (AEG-3) (3.1 mg, 6. Mu. Mol,16%,98% purity) as a white solid. M/z 501.0 (M+H) + (ES+). 1 H NMR(300MHz,DMSO–d6)δ8.54(s,1H),8.26–8.17(m,2H),8.15–7.96(m,3H),7.91–7.75(m,2H),6.89(s,1H),5.19(s,2H),3.46(s,3H),2.06(d,J=8.6Hz,1H),1.04–1.88(m,4H)。
Example 159: synthesis of 2- { 6-cyclopropyl-4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AEH-1)
To intermediate (AEE-1) (40 mg,1 equivalent, 0.16 mmol), intermediate (ADZ-1) (50 mg,1 equivalent, 0.16 mmol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 To a stirred solution of (106 mg,0.32mmol,2 eq.) in 1, 4-dioxane (5 mL) were added RuPhos (30 mg,0.4 eq., 0.06 mmol) and RuPhos ring palladium complex gen.3 (27 mg,0.2 eq., 0.03 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (50% MeCN up to 80% in 15 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30×150mm,5um; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 72B to 82B in 8 minutes; a detector, UV 254/210nm; retention time: 5.68. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AEH-1) (20.8 mg,39 μmol,25%,99% purity) as a white solid. M/z 521.4 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.48(s,1H),8.21(s,1H),8.03(d,J=1.4Hz,1H),7.81–7.73(m,2H),7.74–7.59(m,5H),6.79(d,J=1.4Hz,1H),5.01(s,2H),3.89(s,2H),3.38(s,3H),2.57(s,2H),2.08–1.97(m,3H),1.93–1.84(m,2H),1.63–1.51(m,1H),1.42–1.28(m,1H),0.95(d,J=6.3Hz,4H)。
Example 160: synthesis of 2- { 6-cyclopropyl-4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -5-fluoro-6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AEI-5)
Step 1: synthesis of 6-vinyl-5-fluoro-2, 3-dihydro-isoindol-1-one (AEI-2)
Pd (PPh) was added to a stirred solution of 6-bromo-5-fluoro-2, 3-isoindol-1-one (AEI-1) (200 mg,1 eq, 0.87 mmol) tributyl (vinyl) stannane (331 mg,1.2 eq, 1.04 mmol) and CsF (265 mg,2 eq, 1.74 mmol) in 1, 4-dioxane (3 mL) at room temperature under nitrogen atmosphere 3 ) 2 Cl 2 (61 mg,0.1 eq, 87. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (30% MeCN up to 50% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AEI-2) (120 mg,0.62mmol,78%,92% purity) as a white solid. M/z 178.1 (M+H) + (ES+)。
Step 2: synthesis of 6-fluoro-3-oxo-1, 2-dihydro-isoindole-5-carbaldehyde (AEI-3)
To a stirred solution of the product (AEI-2) (120 mg,1 eq, 0.67 mmol) from step 1 above and NMO (106 mg,1.3 eq, 0.90 mmol) in tBuOH (3 mL) and water (3 mL) at room temperature was added K 2 OsO 4.2 H 2 O (13 mg,0.06 eq, 0.04 mmol) and 2-hydroxy propane-1, 2, 3-tricarboxylic acid (173 mg,1.3 eq, 0.90 mmol). After 2 hours, sodium periodate (289 mg,2 equivalents, 1.35 mmol) was added to the above mixture at room temperature.The resulting mixture was stirred at room temperature for an additional 1 hour. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (20% MeCN up to 30% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AEI-3) (100 mg,0.50mmol,83%,90% purity) as a white solid. M/z 180.0 (M+H) + (ES+)。
Step 3: synthesis of 5-fluoro-6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -2, 3-dihydro-isoindol-1-one (AEI-4)
To a stirred solution of the product from step 2 above (AEI-3) (100 mg,1 eq, 0.56 mmol) and 1- (aminomethyl) cyclobutan-1-ol (AW-1) (153 mg,2 eq, 1.12 mmol) in MeOH (5 mL) at room temperature was added NaBH 3 CN (105 mg,3 equivalents, 1.67 mmol) and AcOH (3.4 mg,0.1 equivalents, 56. Mu. Mol). The resulting mixture was stirred at room temperature overnight. The residue was purified by preparative TLC using DCM/MeOH (12/1) to give the sub-title compound (AEI-4) (100 mg,0.34mmol,68%,91% pure) as a yellow solid. M/z265.1 (M+H) + (ES+)。
Step 4:2- { 6-cyclopropyl-4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -5-fluoro-6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AEI-5)
To the product (AEI-4) (50 mg,1 eq, 0.19 mmol), intermediate (ADZ-1) (65 mg,1.1 eq, 0.21 mmol) and Cs from step 3 above, under a nitrogen atmosphere at room temperature 2 CO 3 (185 mg,0.57mmol,3 eq.) Pd (OAc) was added to a stirred solution of DMF (3 mL) 2 (8.5 mg,0.2 eq, 38. Mu. Mol) and XantPhos (44 mg,0.4 eq, 76. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (70% MeCN up to 90% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. Passing the crude product throughPreparative HPLC was purified (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 10% B to 40% B in 7 min; wavelength: 254/220nm; retention time: 6.3) to give the title compound (AEI-5) (5.0 mg, 9.1. Mu. Mol,4.9%,99% purity) as a white solid. M/z 539.4 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO–d6)δ8.48(s,1H),8.31(s,1H),8.00(d,J=1.4Hz,1H),7.90(d,J=6.7Hz,1H),7.76–7.72(m,1H),7.67–7.61(m,3H),7.54(d,J=9.5Hz,1H),6.80(d,J=1.5Hz,1H),5.01(s,2H),3.87(s,2H),3.37(s,3H),2.57(s,2H),2.05–1.97(m,3H),1.94–1.86(m,2H),1.62(d,J=10.7Hz,1H),1.47–1.35(m,1H),0.96–0.83(m,4H)。
Example 161: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((((1-hydroxycyclobutyl) methyl) (methyl) amino) methyl) -4- (trifluoromethyl) isoindolin-1-one (AEJ-3)
Step 1:6- ((((1-hydroxycyclobutyl) methyl) (methyl) amino) methyl) -4- (trifluoromethyl) isoindolin-1-one (AEJ-2)
To intermediate (ABC-4) (75 mg,0.327mmol,1.00 eq.) and 1- [ (methylamino) methyl under nitrogen at room temperature]To a stirred mixture of cyclobutan-1-ol (AEJ-1) (75 mg,2 eq, 0.65 mmol) in MeOH (5 mL) was added Et 3 N (66 mg,2 eq, 0.65 mmol). The resulting mixture was stirred under nitrogen at 60 ℃ for 2 hours. Adding NaBH to the mixture at 0deg.C 3 CN (82 mg,4 equivalents, 1.31 mmol). The resulting mixture was stirred at room temperature overnight. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 60% in 10 minutes); detector, UV 254/220nm to give the sub-title compound (AEJ-2) as a yellow solid (86 mg,0.23mmol,80%89% purity). M/z 329.1 (M+H) + (ES+)。
Step 2: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((((1-hydroxycyclobutyl) methyl) (methyl) amino) methyl) -4- (trifluoromethyl) isoindolin-1-one (AEJ-3)
To the product (AEJ-2) (50 mg,1 eq, 0.15 mmol), intermediate (ABC-6) (48 mg,1 eq, 0.15 mmol) and Cs from step 1 above, under a nitrogen atmosphere at room temperature 2 CO 3 (99 mg,2 eq, 0.30 mmol) to a stirred mixture of dioxane (5 mL) was added RuPhos ring palladium complex Gen.3 (25 mg,0.2 eq, 30. Mu. Mol) and RuPhos (28 mg,0.4 eq, 61. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 34% B to 42% B in 9 minutes; wavelength: 254/220nm; retention time: 8.35 To give the title compound (AEJ-3) (36.1 mg, 58. Mu. Mol,35%,98% purity) as a white solid. M/z 610.0 (M+H) + (ES+). 1 H NMR(400MHz,MeOH–d4)δ8.46(s,1H),8.08(d,J=21.3Hz,2H),7.85(s,1H),7.62(d,J=1.2Hz,1H),6.19(d,J=1.2Hz,1H),5.25(d,J=1.8Hz,2H),4.03(d,J=17.0Hz,5H),3.48(s,3H),3.36(t,J=7.2Hz,2H),2.85(s,2H),2.51(s,3H),2.20–2.01(m,4H),1.85–1.73(m,1H),1.59–1.43(m,1H),1.23(t,J=7.2Hz,3H)。
Example 162: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- ({ [ (1-hydroxycyclobutyl) methyl ] (methyl) amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AEK-3)
Step 1: synthesis of 6- ((((1-methoxycyclobutyl) methyl) (methyl) amino) methyl) -4- (trifluoromethyl) isoindolin-1-one (AEK-2)
Intermediate (ABC-4) (50 mg,1 eq, 0.22 mmol) and [ (1-methoxycyclobutyl) methyl are introduced into an intermediate (ABC-4) (N.sub.1)]To a stirred mixture of (meth) amine (AEK-1) (56 mg,2 eq, 0.45 mmol) in MeOH (5 mL) was added Et 3 N (44 mg,2 eq, 0.44 mmol). The resulting mixture was stirred under nitrogen at 60 ℃ for 2 hours. Adding NaBH to the mixture at 0deg.C 3 CN (55 mg,4 equivalents, 0.87 mmol). The resulting mixture was stirred at room temperature overnight. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 40% in 15 minutes); detector, UV 254/220nm to give the sub-title compound (AEK-2) (65 mg,0.17mmol,87%,90% purity) as a yellow solid. M/z 343.2 (M+H) + (ES+)。
Step 2: synthesis of 2- (6- (ethylamino) -4- (1-methyl-4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1H-pyrazol-5-yl) pyridin-2-yl) -6- ((((1-methoxycyclobutyl) methyl) (methyl) amino) methyl) -4- (trifluoromethyl) isoindolin-1-one (AEK-3)
To the product (AEK-2) (60 mg,1 equivalent, 0.18 mmol), intermediate (ABC-6) (56 mg,1 equivalent, 0.18 mmol) and Cs from step 1 above, under a nitrogen atmosphere at room temperature 2 CO 3 To a stirred mixture of (99 mg,0.30mmol,2 eq.) in 1, 4-dioxane (5 mL) were added RuPhos ring palladium complex gen.3 (29 mg,0.2 eq., 35 μmol) and RuPhos (33 mg,0.4 eq., 70 μmol). The resulting mixture was stirred under nitrogen at 80 ℃ for 2 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and MeCN (0% MeCN up to 30% in 10 min); detector, UV 254/220nm. The crude product was purified by preparative HPLC (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 11% B to 2 in 10 min)6% B; wavelength: 254/220nm; retention time: 11.5 To give the title compound (AEK-3) (11.0 mg, 17. Mu. Mol,10%,99% purity) as a white solid. M/z 624.4 (M+H) + (ES+). 1 H NMR(400MHz,MeOH–d4)δ8.47(s,1H),8.02(d,J=19.3Hz,2H),7.84(s,1H),7.63(d,J=1.2Hz,1H),6.17(d,J=1.2Hz,1H),5.23(d,J=1.8Hz,2H),4.01(s,3H),3.78(s,2H),3.48(s,3H),3.35(t,J=7.2Hz,2H),3.20(s,3H),2.66(s,2H),2.32(s,3H),2.17–2.06(m,2H),2.02–1.92(m,2H),1.79–1.68(m,1H),1.59–1.43(m,1H),1.22(t,J=7.2Hz,3H)。
Example 163: synthesis of 4- [2- (ethylamino) -6- [6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEL-2)
Step 1: synthesis of 6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (AEL-1)
To (1S, 2R) -2-aminocyclopentan-1-ol, HCl (Z-1) (144 mg,1.2 eq, 1.05 mmol) and Et at room temperature 3 N (399 mg,3 eq, 2.62 mmol) in DCM (3 mL). The resulting mixture was stirred at room temperature for 30 minutes. Intermediate (ABC-4) (200 mg,1 eq, 0.87 mmol) was added to the above mixture at room temperature. The resulting mixture was stirred at room temperature for an additional 1 hour. NaBH (OAc) was added to the above mixture at room temperature 2 (925 mg,5 equivalents, 4.37 mmol). The resulting mixture was stirred at room temperature overnight. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 50% within 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (AEL-1) (200 mg,0.60mmol,73%,94% purity) as an off-white solid. M/z 315.1 (M+H) + (ES+)。
Step 2: synthesis of 4- [2- (ethylamino) -6- [6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEL-2)
To intermediate (ADP-7) (40 mg,1 equivalent, 0.12 mmol), the product from step 1 above (AEL-1) (41 mg,1.1 equivalent, 0.13 mmol) and Cs under nitrogen at room temperature 2 CO 3 (115 mg,3 eq, 0.35 mmol) to a stirred solution of RuPhos ring palladium complex gen.3 (20 mg,0.2 eq, 24. Mu. Mol) and RuPhos (22 mg,0.4 eq, 47. Mu. Mol) in 1, 4-dioxane (10 mL) was added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The mixture was cooled to room temperature. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 50% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (AEL-2) (19.6 mg, 31. Mu. Mol,27%,98% purity) as a pale yellow solid. M/z 617.4 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.47(s,1H),8.12–7.97(m,4H),7.87(d,J=8.1Hz,1H),7.51(d,J=1.3Hz,1H),6.05(d,J=1.2Hz,1H),5.18(s,2H),4.17–4.12(m,1H),4.04–3.88(m,2H),3.45(s,3H),3.27(t,J=7.2Hz,2H),2.99–2.90(m,1H),1.93–1.66(m,4H),1.65–1.46(m,2H),1.20(t,J=7.2Hz,3H)。
Example 164: synthesis of 2- (6-cyclopropyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((((1-hydroxycyclobutyl) methyl) amino) methyl) -5-methoxyisoindolin-1-one (AEM-9)
Step 1: synthesis of 5-bromo-4-methoxy-2-methylbenzoic acid (AEM-2)
To 4-methoxy-2-methylbenzoic acid (AEM-1) (3.00 g,1 eq., 18.1 mmol) and Fe (710 mg,0.7 eq., 12.6 mmol) in CHCl under nitrogen at room temperature 3 Br was added to the stirred mixture in (40 mL) 2 (2.89 g,1 eq, 18.1 mmol). The resulting mixture was stirred at room temperature under nitrogen overnight. The resulting mixture was diluted with water. The resulting mixture was extracted with DCM (3X 200 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 30% in 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (AEM-2) (2.85 g,10.4mmol,64%,89% purity) as a white solid. M/z 245.0/247.0 (M+H) + (ES+)。
Step 2: synthesis of 5-bromo-4-methoxy-2-methylbenzoic acid methyl ester (AEM-3)
Thionyl chloride (962mg, 0.7 eq., 8.08 mmol) was added dropwise to a stirred mixture of the product from step 1 above (AEM-2) (2.83 g,1 eq., 11.5 mmol) in MeOH (30 mL) at room temperature. The resulting mixture was stirred at 70℃for 3 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water. The resulting mixture was extracted with DCM (3X 150 mL). The combined organic layers were washed with brine (2×150 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (3/1) to give the sub-title compound (AEM-3) (2.46 g,8.58mmol,82%,90% purity) as a white solid. M/z 259.0/261.0 (M+H) + (ES+)。
Step 3: synthesis of methyl 5-bromo-2- (bromomethyl) -4-methoxybenzoate (AEM-4)
To the product (AEM-3) from step 2 above (2.44 g,1 eq., 9.42 mmol) and AIBN (1.55 g,1 eq., 9.42 mmol) in CHCl under a nitrogen atmosphere at 0deg.C 3 NBS (1.76 g,1.05 eq, 10.0 mmol) was added to the stirred mixture in (30 mL). The resulting mixture was stirred under nitrogen at 90 ℃ for 4 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water. The resulting mixture was extracted with DCM (3X 100 mL). Will be combined The organic layer was washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (1/1) to give the sub-title compound (AEM-4) (1.64 g,4.27mmol,52%,88% purity) as an off-white solid. M/z 338.9/340.9 (M+H) + (ES+)。
Step 4: synthesis of 6-bromo-5-methoxyisoindolin-1-one (AEM-5)
To the product (AEM-4) (1.44 g,1 eq., 4.26 mmol) from step 3 above in an NH-containing atmosphere at room temperature under nitrogen 3 The mixture was stirred in MeOH (30 mL, 7M). The resulting mixture was stirred under nitrogen at 120 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water. The resulting mixture was extracted with DCM (3X 100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with DCM/MeOH (20/1) to give the sub-title compound (AEM-5) (760 mg,2.9mmol,74%,92% purity) as a white solid. M/z 242.0/244.0 (M+H) + (ES+)。
Step 5: synthesis of 5-methoxy-6-vinyl isoindolin-1-one (AEM-6)
Pd (PPh) was added to a stirred mixture of the product (AEM-5) (750 mg,1 eq, 3.10 mmol), tributyl (vinyl) stannane (1.18 g,1.2 eq, 3.72 mmol) and CsF (941 mg,2 eq, 6.20 mmol) from step 4 above in 1, 4-dioxane (10 mL) at room temperature under nitrogen atmosphere 3 ) 2 Cl 2 (435 mg,0.2 eq, 0.62 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water. The resulting mixture was extracted with DCM (3X 80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (20/1) to give the sub-title compound (AEM-6) (550 mg,2.62mmol,94%,90% purity) as a white solid. m-z 190.1(M+H) + (ES+)。
Step 6: synthesis of 6-methoxy-3-oxo-isoindoline-5-carbaldehyde (AEM-7)
To a stirred mixture of the product (AEM-6) (550 mg,1 eq, 2.91 mmol) from step 5 above and NMO (443 mg,1.3 eq, 3.78 mmol) in tBuOH (3 mL) and water (3 mL) at room temperature was added citric acid (726 mg,1.3 eq, 3.78 mmol) and K 2 OsO 4 (97 mg,0.1 eq, 0.29 mmol) and sodium periodate (1.24 g,2 eq, 5.81 mmol). The resulting mixture was stirred at room temperature for 3 hours. The resulting mixture was diluted with water. The resulting mixture was extracted with DCM (3X 80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (25% MeCN up to 35% in 9 minutes); detector, UV 254/220nm. This gave the sub-title compound (AEM-7) (130 mg,0.65mmol,23%,95% purity) as a white solid. M/z 192.1 (M+H) + (ES+)。
Step 7: synthesis of 2- (6-cyclopropyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6-methoxy-3-oxoisoindoline-5-carbaldehyde (AEM-8)
To the product (AEM-7) (100 mg,1 eq., 0.52 mmol), intermediate (ADZ-1) (163 mg,1 eq., 0.52 mmol) and Cs from step 6 above, under a nitrogen atmosphere at room temperature 2 CO 3 (349mg, 2 eq, 1.05 mmol) to a stirred mixture of 1, 4-dioxane (3 mL) was added Ruphos (98 mg,0.4 eq, 0.21 mmol) and Ruphos ring palladium complex gen.3 (87 mg,0.2 eq, 0.11 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water. The resulting mixture was extracted with DCM (3X 50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1) to give the sub-title compound (AEM-8) as a white solid(70 mg,0.14mmol,29%,95% purity). M/z 466.2 (M+H) + (ES+)。
Step 8: synthesis of 2- (6-cyclopropyl-4- (2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((((1-hydroxycyclobutyl) methyl) amino) methyl) -5-methoxyisoindolin-1-one (AEM-9)
To a stirred mixture of the product (AEM-8) (40 mg,1 eq, 86. Mu. Mol) from step 7 above and 1- (aminomethyl) cyclobutan-1-ol (AW-1) (26 mg,3 eq, 0.26 mmol) in DCM (3 mL) under nitrogen at room temperature was added Et 3 N (17 mg,2 eq, 0.17 mmol) and NaBH (OAc) 3 (73 mg,4 equivalents, 0.34 mmol). The resulting mixture was stirred at room temperature under nitrogen overnight. The resulting mixture was diluted with water. The resulting mixture was extracted with DCM (3X 10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO- 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 33% B to 45% B in 9 minutes; wavelength: 254/220nm; retention time: 8.2. this gave the title compound (AEM-9) (13.4 mg, 24. Mu. Mol,28%,98% purity) as a white solid. M/z 551.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.00(d,J=1.4Hz,1H),7.79–7.58(m,5H),7.32(s,1H),6.76(d,J=1.4Hz,1H),4.97(s,2H),4.91(s,1H),3.84(d,J=51.3Hz,5H),3.38(s,3H),2.56(s,2H),2.03–1.98(m,3H),1.98–1.86(m,2H),1.68–1.58(m,1H),1.50–1.37(m,1H),0.94(d,J=6.4Hz,4H)。
The by-product was isolated from the purification and this yielded compound (AER-1) (6.3 mg,14 μmol,22%,98% purity) as a white solid. M/z 438.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.01(d,J=1.4Hz,1H),7.79–7.57(m,5H),7.28(d,J=2.2Hz,1H),7.07(d,J=8.5,2.3Hz,1H),6.76(d,J=1.4Hz,1H),4.98(s,2H),3.87(s,3H),3.36(s,3H),2.10–1.94(m,1H),0.98–0.91(m,4H)。
Example 165: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (2-methoxyethyl) amino ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEN-1)
To intermediate (AEG-2) (30 mg,1 equivalent, 89. Mu. Mol), intermediate (AEC-1) (31 mg,1.2 equivalent, 0.11 mmol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 (58 mg,2 eq, 0.18 mmol) to a stirred solution of RuPhos (17 mg,0.4 eq, 36. Mu. Mol) and RuPhos ring palladium complex Gen.3 (15 mg,0.2 eq, 18. Mu. Mol) in 1, 4-dioxane (4 mL) were added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 60% B in 9 minutes; wavelength: 254/220nm; retention time: 8.83 To give the title compound (AEN-1) (7.8 mg,13 μmol,15%,98% purity) as a white solid. M/z 588.4 (M+H) + (ES+). 1 H NMR(300MHz,DMSO–d6)δ8.54(s,1H),8.25–8.16(m,2H),8.05–7.96(m,3H),7.87(d,J=8.5Hz,1H),6.89(d,J=1.4Hz,1H),5.15(s,2H),3.90(s,2H),3.47(s,3H),3.40(t,J=5.6Hz,2H),3.25–3.22(m,3H),2.64(t,J=5.7Hz,2H),2.11–2.00(m,1H),1.04–0.87(m,4H)。
Example 166: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (2-hydroxyethyl) amino ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEO-2)
Step 1: synthesis of 6- (((2-hydroxyethyl) amino) methyl) -4- (trifluoromethyl) isoindolin-1-one (AEO-1)
To a stirred solution of intermediate (ABC-4) (150 mg,1 eq., 0.66 mmol) and ethanolamine (AEA-1) (48 mg,1.2 eq., 0.79 mmol) in DCM (20 mL) at RT was added NaBH (OAc) 3 (694 mg,5 eq, 3.28 mmol) and Et 3 N (99 mg,1.5 eq, 0.98 mmol). The resulting mixture was stirred at room temperature under nitrogen overnight. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (5/1). This gave the sub-title compound (AEO-1) (180 mg,0.56mmol,99%,85% purity) as a white solid. M/z 275.1 (M+H) + (ES+)。
Step 2: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (2-hydroxyethyl) amino ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEO-2)
To intermediate (AEG-2) (40 mg,1 equivalent, 0.12 mmol), the product from step 1 above (AEO-1) (39 mg,1.2 equivalent, 0.14 mmol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 (78 mg,2 eq, 0.24 mmol) to a stirred solution of RuPhos ring palladium complex gen.3 (20 mg,0.2 eq, 24. Mu. Mol) and RuPhos (22 mg,0.4 eq, 48. Mu. Mol) in 1, 4-dioxane (5 mL) was added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (5/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 52% B in 8 minutes; wavelength: 254/220nm; retention time: 7.33 To give the title compound (AEO-2) (12.9 mg, 22. Mu. Mol,19%,99% purity) as a white solid. M/z 574.3 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO–d6)δ8.54(s,1H),8.24–7.95(m,5H),7.87(d,J=8.5Hz,1H),6.90(d,J=1.4Hz,1H),5.16(s,2H),4.55(s,1H),3.94(s,2H),3.48(d,J=9.7Hz,5H),2.60(t,J=5.8Hz,2H),2.11–2.01(m,1H),1.02–0.89(m,4H)。
Example 167: synthesis of 4- [2- (ethylamino) -6- [ 1-oxo-6- (pyrrolidin-1-yl) -4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEP-2)
/>
Step 1: synthesis of 6- (pyrrolidin-1-yl) -4- (trifluoromethyl) isoindolin-1-one (AEP-1)
Cs was added to a stirred mixture of intermediate (ABC-2) (140 mg,1 eq, 0.50 mmol) and pyrrolidine (ABK-4) (356 mg,10 eq, 5.00 mmol) in 1, 4-dioxane (5 mL) at room temperature under nitrogen atmosphere 2 CO 3 (407 mg,2.5 eq, 1.25 mmol) and SPhos Pd Gen.3 (41 mg,0.2 eq, 0.10 mmol) and SPhos (82 mg,0.4 eq, 0.20 mmol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (36% MeCN up to 48% in 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (AEP-1) (40 mg,0.13mmol,30%,90% purity) as a white solid. M/z 271.1 (M+H) + (ES+)。
Step 2: synthesis of 4- (2- (ethylamino) -6- (1-oxo-6- (pyrrolidin-1-yl) -4- (trifluoromethyl) isoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AEP-2)
To intermediate (ADP-7) (35 mg,1 equivalent, 0.13 mmol), the product from step 1 above (AEP-1) (44 mg,1.0 equivalent, 0.13 mmol) and Cs under nitrogen at room temperature 2 CO 3 (84 mg,2 eq, 0.26 mmol) to a stirred solution in 1, 4-dioxane (5 mL) was added RuPhos ring palladium complex Gen.3 (22 mg,0.2 eq, 26. Mu. Mol) and RuPhos (24 mg,0.4 eq, 52. Mu. Mol). The resulting mixture was stirred overnight at 120 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19X 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30B to 60B in 7 minutes; a detector, UV 254/210nm; retention time: 6.08. this gave the title compound (AEP-2) (15.7 mg, 26. Mu. Mol,20%,95% purity) as a white solid. M/z 573.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.52(s,1H),8.20–8.11(m,2H),7.78(d,J=8.0Hz,1H),7.41(d,J=1.2Hz,1H),7.04–6.97(m,2H),6.77(t,J=5.4Hz,1H),5.97(d,J=1.4Hz,1H),5.00(s,2H),3.40(s,3H),3.38–3.32(m,4H),3.23–3.15(m,2H),2.05–1.96(m,4H),1.13(t,J=7.2Hz,3H)。
Example 168: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- ({ [ (1-methoxycyclobutyl) methyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AEQ-3)
Step 1: synthesis of 6- ({ [ (1-methoxycyclobutyl) methyl ] amino } methyl) -4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (AEQ-2)
Et is added to a stirred mixture of intermediate (ABC-4) (60 mg,1 eq, 0.26 mmol) and 1- (1-methoxycyclobutyl) methylamine (AEQ-1) (60 mg,2 eq, 0.52 mmol) in MeOH (1 mL) at room temperature under nitrogen atmosphere 3 N (53 mg,2 eq, 0.52 mmol) and AcOH (16 mg,1 eq, 0.26 mmol). NaBH was added to the above mixture at 0deg.C over 30 minutes 3 CN (66 mg,4 equivalents, 1.05 mmol). The resulting mixture was stirred at 60 ℃ overnight. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (0% MeCN up to 30% in 15 minutes); detector, UV 254/220nm, to give the sub-title compound (AEQ-2) (40 mg,0.14mmol,47%,95% purity) as a yellow solid. M/z 271.1 (M+H) + (ES+)。
Step 2: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- ({ [ (1-methoxycyclobutyl) methyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AEQ-3)
To the product (AEQ-2) (40 mg,1 equivalent, 0.12 mmol), 6-intermediate (ABC-6) (39 mg,1 equivalent, 0.12 mmol) and Cs from step 1 above, under a nitrogen atmosphere at room temperature 2 CO 3 To a stirred mixture of (79 mg,0.24mmol,2 eq.) in 1, 4-dioxane (5 mL) were added RuPhos ring palladium complex gen.3 (11 mg,0.2 eq., 24 μmol) and RuPhos (23 mg,0.4 eq., 49 μmol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 10% B to 30% B over 10 min; wavelength: 254/220nm; retention time: 9.3) to give the title compound (AEQ-3) (3.2 mg,5.1 μmol,4.2%,98% purity) as a white solid. M/z 610.3 (M+H) + (ES+). 1 HNMR(400MHz,MeOH–d4)δ8.46(s,1H),8.07(d,J=18.9Hz,2H),7.84(s,1H),7.64(d,J=1.2Hz,1H),6.18(d,J=1.2Hz,1H),5.25(s,2H),4.07(s,2H),4.01(s,3H),3.48(s,3H),3.36(t,J=7.2Hz,2H),3.15(s,3H),2.84(s,2H),2.25–2.12(m,2H),1.98–1.88(m,2H),1.78–1.71(m,1H),1.62–1.48(m,1H),1.23(t,J=7.2Hz,3H)。
Example 170: synthesis of 4- (2-cyclopropyl-6- (6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) isoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AES-1)
To intermediate (AEG-2) (25 mg, 74. Mu. Mol,1 eq), intermediate (ADX-1) (17 mg,1 eq, 74. Mu. Mol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 To a stirred mixture of (49 mg,0.15mmol,2 eq.) in 1, 4-dioxane (2 mL) was added Ruphos (14 mg,0.4 eq., 30. Mu. Mol) and Ruphos ring palladium complex gen.3 (12 mg,0.2 eq., 15. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water. The resulting mixture was extracted with DCM (3X 5 mL). The combined organic layers were washed with brine (2×5 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 50% B in 10 minutes, UV 254/220nm; retention time: 10.17. this gave the title compound (AES-1) (4.7 mg, 8.8. Mu. Mol,12%,99% purity) as a white solid. M/z 531.3 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.25–8.18(m,2H),8.04–7.94(m,3H),7.88(d,J=8.5Hz,1H),6.89(d,J=1.4Hz,1H),5.58(t,J=5.8Hz,1H),5.17(s,2H),4.70(d,J=5.8Hz,2H),3.47(s,3H),2.12–2.01(m,1H),1.00–0.91(m,4H)。
Example 171: synthesis of 4- [2- (ethylamino) -6- [6- ({ [ (1R, 2S) -2-fluorocyclopentyl ] amino } methyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AET-6)
Step 1: synthesis of (1R, 2R) -2- [ (diphenylmethylene) amino ] cyclopentan-1-ol (AET-2)
(1R, 2R) -2-aminocyclopentan-1-ol, HCl (W-1) (500 mg,1 eq., 3.63 mmol), benzophenone (AET-1) (659 mg,1 eq., 3.63 mmol) and Et 3 A stirred solution of N (730 mg,2 eq, 7.27 mmol) in DCM (20 mL) was stirred at room temperature overnight. The resulting mixture was extracted with EtOAc (3×20 mL). The combined organic layers were washed with water (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/5) to give the sub-title compound (AET-2) as a brown oil (400 mg,1.39mmol,41%,92% purity). M/z 266.2 (M+H) + (ES+)。
Step 2: synthesis of N- [ (1R, 2S) -2-fluorocyclopentyl ] -1, 1-diphenylazomethine (AET-3)
DAST (607 mg,2 eq., 3.78 mmol) was added dropwise to a stirred solution of the product from step 1 above (AET-2) (500 mg,1 eq., 1.88 mmol) in DCM (10 mL) at-20 ℃. The reaction liquid was concentrated in vacuo. This gave the sub-title compound (AET-3) (500 mg,1.78mmol,99%,95% purity) as a brown oil. M/z 268.1 (M+H) + (ES+)。
Step 3: synthesis of (1R, 2S) -2-fluorocyclopentane-1-amine (AET-4)
To a stirred solution of the product from step 2 above (AET-3) (500 mg,1 eq, 1.87 mmol) in dioxane (10 mL) was added HCl (gas) at room temperature. The resulting mixture was stirred at 80℃for 3 hours. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. This gave the sub-title compound (AET-4) (180 mg,1.55mmol,93% purity) as a brown oil. M/z 104.1 (M+H) + (ES+)。
Step 4: synthesis of 6- ({ [ (1R, 2S) -2-fluorocyclopentyl ] amino } methyl) -4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (AET-5)
The product (AET-4) (81 mg,1.2 eq, 0.79 mmol) from step 3 above, et 3 A stirred solution of N (338 mg,4 eq, 2.62 mmol) in DCM (10 mL) was stirred at room temperature for 30 min. Intermediate (ABC-4) (150 mg,1 equivalent, 0.66 mmol). The resulting mixture was stirred at room temperature overnight. NaBH (OAc) was added to the above mixture at room temperature 3 (124 mg,5 eq, 3.28 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was then quenched by addition of 2mL of ice water at 0 ℃. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (0% MeCN up to 40% in 18 minutes); detector, UV 254/220nm. This gave the sub-title compound (AET-5) (70 mg,0.21mmol,34%,93% purity) as a brown oil. M/z 317.1 (M+H) + (ES+)。
Step 5: synthesis of 4- [2- (ethylamino) -6- [6- ({ [ (1R, 2S) -2-fluorocyclopentyl ] amino } methyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AET-6)
To the product (AET-5) (50 mg,1 eq., 0.16 mmol), intermediate (ADP-7) (59 mg,0.17mmol,1.1 eq.) and Cs from step 5 above, under a nitrogen atmosphere at room temperature 2 CO 3 (103 mg,2 eq, 0.32 mmol) to a stirred solution of RuPhos ring palladium complex gen.3 (26 mg,0.2 eq, 32. Mu. Mol) and RuPhos (30 mg,0.4 eq, 63. Mu. Mol) in 1, 4-dioxane (10 mL) was added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (30% MeCN up to 60% in 20 minutes); detector, UV 254/220nm. The product was purified by preparative HPLC on a column (XBridge prep OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 55% B in 10 minutes; wavelength: 254/220 nm) to give the title compound (AET-6) (8.3 mg, 13. Mu. Mol,8.5%,98% purity) as a pale yellow solid. M/z 619.3 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.47(s,1H),8.16–8.03(m,3H),7.98(s,1H),7.87(d,J=8.1Hz,1H),7.51(d,J=1.3Hz,1H),6.05(d,J=1.3Hz,1H),5.18(d,J=1.9Hz,2H),4.99–4.78(m,1H),3.99(s,2H),3.45(s,3H),3.27(t,J=7.2Hz,2H),3.22–3.10(m,1H),2.11–1.84(m,3H),1.81–1.69(m,2H),1.49–1.39(m,1H),1.20(t,J=7.2Hz,3H)。
Example 172: synthesis of 6- { [ (1-aminocyclobutyl) methoxy ] methyl } -2- {4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6- (methylamino) pyridin-2-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (AEU-8)
/>
Step 1: synthesis of methyl 5-bromo-2-methyl-3- (trifluoromethyl) benzoate (AEU-2)
To a stirred mixture of methyl 2-methyl-3- (trifluoromethyl) benzoate (AEU-1) (1.00 g,1 eq., 4.58 mmol) in AcOH (20 mL) at room temperature was added HNO 3 (2.89 g,10 equivalents, 45.8 mmol) and Br 2 (952 mg,1.3 eq, 5.96 mmol). AgNO-containing solution was added to the above mixture at 0deg.C for 10 minutes 3 (1.01 mg,1.3 eq, 5.96 mmol) H 2 O (1 mg). The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/5) to give the sub-title compound (AEU-2) (1.25 g,3.80mmol,92%,90% purity) as a yellow solid. M/z 297.0/299.0 (M+H) + (ES+)。
Step 2: synthesis of methyl 2-methyl-5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3- (trifluoromethyl) benzoate (AEU-3)
Pd (dppf) Cl was added to a stirred mixture of the product (AEU-2) (2.40 g,1 eq., 6.06 mmol), bis (pinacolato) diboron (AAI-3) (1.69 g,1.1 eq., 6.67 mmol) and KOAc (1.78 g,18.2mmol,3 eq.) from step 1 above in 1, 4-dioxane (20 mL) at room temperature under nitrogen atmosphere 2 DCM (490 mg,0.1 eq, 0.61 mmol). Mixing the obtained mixtureThe mixture was stirred under nitrogen at 100 ℃ for an additional 12 hours. The mixture was cooled to room temperature. Filtering the resulting mixture; the filter cake was washed with EtOAc (15 mL). The filtrate was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 343.1 (M+H) + (ES+)。
Step 3: synthesis of methyl 5-hydroxy-2-methyl-3- (trifluoromethyl) benzoate (AEU-4)
To a stirred mixture of the product from step 2 above (AEU-3) (2.40 g,1 eq, 6.97 mmol) in AcOH (20 mL) at room temperature was added H 2 O 2 (5 mL,30 wt%): THF (5 mL) =1:1. The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (5/1) to give the sub-title compound (AEU-4) (1.3 g,5.27mmol,76%,95% purity) as a yellow solid. M/z 235.1 (M+H) + (ES+)。
Step 4: synthesis of methyl 5-hydroxy-2-methyl-3- (trifluoromethyl) benzoate (AEU-5)
To the product (AEU-4) from step 3 above (1.30 g,1 eq., 5.27 mmol) and CH at room temperature 3 I (898 mg,1.2 eq, 6.33 mmol) in DMF (10 mL) was added K 2 CO 3 (2.19 g,3 equivalents, 15.8 mmol). The resulting mixture was stirred at room temperature for 12 hours. The resulting mixture was concentrated in vacuo. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (1/1) to give the sub-title compound (AEU-5) (1.1 g,3.99mmol,76%,90% purity) as a yellow solid. M/z 249.1 (M+H) + (ES+)。
Step 5: synthesis of methyl 2- (bromomethyl) -5-methoxy-3- (trifluoromethyl) benzoate (AEU-6)
To the product (AEU-5) from step 4 above (1.10 g,1 eq., 4.43 mmol) and NBS (1.18 g,1.5 eq., 6.65 mmol) in CHCl at room temperature 3 BPO (3411 mg,0.3 eq, 1.33 mmol) was added to the stirred mixture in (20 mL). The resulting mixture was stirred under nitrogen at 80 ℃ for 12 hours. The mixture was cooled to room temperature. The resulting mixture was used directly in the next step without further purification. M/z 327.0/329.0 (M+H) + (ES+)。
Step 6: synthesis of 6-methoxy-4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (AEU-7)
At room temperature, to NH 3 To a stirred mixture in MeOH (20 mL) was added the product from step 5 above (AEU-6) (1.1 g,1 eq, 3.36 mmol). The resulting mixture was stirred at room temperature for 30 minutes. The resulting mixture was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (1/1) to give the sub-title compound (AEU-7) (1.0 g,3.98mmol,90%,92% purity) as a white solid. M/z 232.1 (M+H) + (ES+)。
Step 7: synthesis of 4- [2- (ethylamino) -6- [ 6-methoxy-1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEU-8)
To the product (AEU-7) (500 mg,14.7 eq., 2.17 mmol), intermediate (ADP-7) (50 mg,1 eq., 0.15 mmol) and Cs from step 6 above under nitrogen at 100℃were added 2 CO 3 (144 mg,3 eq, 0.44 mmol) to a stirred mixture of 1, 4-dioxane (20 mL) was added RuPhos ring palladium complex Gen.3 (23 mg,0.2 eq, 30. Mu. Mol) and RuPhos (28 mg,0.4 eq, 59. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1). The crude product was purified by preparative HPLC under the following conditions (column: XBIdge preparative OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 65% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (AEU-8) (25.7 mg, 48. Mu. Mol,33%,99% purity) as a green solid. M/z 534.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.51(s,1H),8.23–8.09(m,2H),7.78(d,J=8.0Hz,1H),7.60–7.51(m,2H),7.41(d,J=1.2Hz,1H),6.79(t,J=5.4Hz,1H),5.99(d,J=1.3Hz,1H),5.14–5.05(m,2H),3.93(s,3H),3.41(s,3H),3.26–3.14(m,2H),1.13(t,J=7.1Hz,3H)。
Example 173: synthesis of 4- {2- [6- (cyclopropoxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -6- (ethylamino) pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEV-3)
Step 1: synthesis of 4- {2- [6- (chloromethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -6- (ethylamino) pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEV-1)
To a stirred solution of intermediate (ADX-2) (89 mg,1 eq, 0.17 mmol) in DCM (8 mL) at 0deg.C was added SOCl 2 (60 mg,3 equivalents, 0.50 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was quenched with MeOH (2 mL) at 0 ℃ and concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (10/1) to give the sub-title compound (AEV-1) (50 mg, 86. Mu. Mol,54%,95% purity) as a yellow solid. M/z 552.1/554.1 (M+H) + (ES+)。
Step 2: synthesis of 4- {2- [6- (cyclopropoxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -6- (ethylamino) pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEV-3)
To a stirred solution of the product (AEV-1) (43 mg,1 eq, 78. Mu. Mol) from step 1 above and cyclopropyl alcohol (AEV-2) (45 mg,10 eq, 0.78 mmol) in DCM (5 mL) at room temperature was added TBAB (13 mg,0.5 eq)39. Mu. Mol) and aqueous KOH (5 mL,20 wt%). The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (8/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 65% B in 10 minutes; wavelength: 254/220 nm) to give the title compound (AEV-3) (6.9 mg, 12. Mu. Mol,15%,99% purity) as a yellow solid. M/z 574.0 (M+H) + (ES+) 1 H NMR(400MHz,DMSO–d6)δ8.52(s,1H),8.20–8.11(m,2H),7.96(d,J=15.3Hz,2H),7.79(d,J=8.0Hz,1H),7.43(d,J=1.3Hz,1H),6.80(t,J=5.4Hz,1H),5.98(d,J=1.3Hz,1H),5.16(s,2H),4.70(s,2H),3.47–3.38(m,4H),3.25–3.15(m,2H),1.13(t,J=7.1Hz,3H),0.66–0.57(m,2H),0.54–0.46(m,2H)。
Example 174: synthesis of 4- { 2-ethoxy-6- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEW-2)
Step 1: synthesis of 4- (2-chloro-6-ethoxypyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEW-1)
To a stirred solution of intermediate (ADP-6) (68 mg,1 eq, 0.21 mmol) in EtOH (4 mL) was added EtONa (17 mg,1.2 eq, 0.25 mmol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature and purified by preparative TLC using EtOAc/petroleum ether (1/1). This gave the sub-title compound (AEW-1) (60 mg,0.15mmol,86% pure) as a white oil. M/z 340.1/342.1 (M+H) + (ES+)。
Step 2: synthesis of 4- { 2-ethoxy-6- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEW-2)
To the product (AEW-1) (50 mg,1 eq., 0.14 mmol), intermediate (ADX-1) (68 mg,2 eq., 0.29 mmol) and Cs from step 1 above, under nitrogen at room temperature 2 CO 3 To a stirred solution of (96 mg,0.29mmol,2.0 eq.) in 1, 4-dioxane (3 mL) was added RuPhos (27 mg,0.4 eq., 59. Mu. Mol) and RuPhos cyclopalladium complex Gen.3 (25 mg,0.2 eq., 29. Mu. Mol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (2×30 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/1). The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30×150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 72B to 82B in 8 minutes; a detector, UV 254/210nm; retention time: 5.68. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AEW-2) (7.1 mg,13 μmol,9%,98% purity) as a white solid. M/z535.0 (M+H) + (ES+). 1 H NMR(400MHz,MeOH–d4)δ8.49(s,1H),8.15–8.04(m,3H),7.96(s,1H),7.90(d,J=8.0Hz,1H),7.84(d,J=1.2Hz,1H),6.48(d,J=1.2Hz,1H),5.22(s,2H),4.79(s,2H),4.45–4.35(m,2H),3.53(s,3H),1.40(t,J=7.1Hz,3H)。
Example 175: synthesis of 4- {2- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEX-2)
Step 1: synthesis of 4- (2-chloropyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEX-1)
Intermediate (ADP-6) (200 mg,1 eq, 0.61 mmol) and Zn (390 mg,10 eq, 6.06 mmol), aqueous NaCl (1.2 mL, 12M) containing NH 3 H 2 O (0.2 mL,25 wt%) in DCM (2 mL) was stirred overnight at 50 ℃. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (10% MeCN up to 60% in 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (AEX-1) (60 mg,0.18mmol,33%,90% purity) as a white solid. M/z 296.1/298.1 (M+H) + (ES+)。
Step 2:4- {2- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEX-2)
To the product (AEX-1) (60 mg,1 equivalent, 0.20 mmol), intermediate (ADX-1) (52 mg,1.1 equivalent, 0.22 mmol) and Cs from step 1 above under a nitrogen atmosphere at room temperature 2 CO 3 (132 mg,2 eq, 0.41 mmol) to a stirred solution of RuPhos ring palladium complex gen.3 (34 mg,0.2 eq, 41. Mu. Mol) and RuPhos (38 mg,0.4 eq, 81. Mu. Mol) in 1.4-dioxane (10 mL). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (30% MeCN up to 50% in 15 minutes); detector, UV 254/220nm. The product was purified by preparative HPLC on a column (XBridge prep OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 33% B to 44% B in 11 minutes; wavelength: 254/220 nm) to give the title compound (AEX-2) (25.3 mg, 51. Mu. Mol,25%,98% purity) as a white solid. M/z 491.2 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.56–8.40(m,2H),8.34–8.30(m,1H),8.18–8.03(m,3H),8.00–7.90(m,2H),7.11–7.05(m,1H),5.23(s,2H),4.79(s,2H),3.53(s,3H)。
Example 176: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AEY-1)
To intermediate (ADN-1) (50 mg,0.15mmol,1 eq.), intermediate (ADR-2) (48 mg,1 eq., 0.15 mmol) and Cs under nitrogen at room temperature 2 CO 3 To a stirred mixture of (99 mg,0.304mmol,2.0 eq.) in 1, 4-dioxane (3 mL) was added RuPhos (28 mg,0.4 eq., 61. Mu. Mol) and RuPhos ring palladium complex gen.3 (25 mg,0.2 eq., 30. Mu. Mol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19X 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30B to 60B in 7 minutes; a detector, UV 254/210nm; retention time: 6.08. this gave the title compound (AEY-1) (30.8 mg, 49. Mu. Mol,92%,97% purity) as a white solid. M/z 607.3 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.45(s,1H),8.10(s,1H),8.06–8.00(m,2H),7.82–7.73(m,1H),7.58–7.49(m,1H),7.49–7.43(m,1H),6.88(d,J=1.4Hz,1H),5.18(s,2H),4.09(s,2H),3.45(s,3H),2.80(s,2H),2.17–1.98(m,5H),1.81–1.68((m,1H),1.59–1.45(m,1H),0.99(d,J=6.5Hz,4H)。
Example 177: synthesis of 4- [2- (ethylamino) -6- (6- { [ (1-hydroxycyclobutyl) methoxy ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AEZ-1)
At room temperature, toward the middleTo a stirred solution of bulk (AEV-1) (40 mg,1 eq, 74. Mu. Mol) and 1- (hydroxymethyl) cyclobutan-1-ol (AED-1) (76 mg,10 eq, 0.74 mmol) in DCM (5 mL) was added TBAB (12 mg,0.5 eq, 37. Mu. Mol) and aqueous KOH (5 m,20 Wt%). The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (12/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 60% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (AEZ-1) (9.6 mg, 15. Mu. Mol,21%,99% purity) as a white solid. M/z 618.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.52(s,1H),8.23–8.08(m,2H),8.01(d,J=14.1Hz,2H),7.79(d,J=8.0Hz,1H),7.44(d,J=1.2Hz,1H),6.81(t,J=5.4Hz,1H),5.98(d,J=1.3Hz,1H),5.22–5.08(m,3H),4.76(s,2H),3.45(s,2H),3.41(s,3H),3.25–3.15(m,2H),2.09–2.00(m,2H),1.99–1.88(m,2H),1.73–1.58(m,1H),1.55–1.39(m,1H),1.13(t,J=7.1Hz,3H)。
Example 178: synthesis of 4- [2- (ethylamino) -6- [6- (methylamino) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AFA-2)
Step 1: synthesis of 6- (methylamino) -4- (trifluoromethyl) isoindolin-1-one (AFA-1)
Intermediate (ABC-2) (280 mg,1 eq, 1.00 mmol) and methylamino, HCl (155 mg,5 eq, 5.00 mmol) and Cs are added to the mixture under nitrogen at room temperature 2 CO 3 (2.28 g,7.00mmol,7 eq.) Pd was added to a stirred mixture of 1, 4-dioxane (20 mL) 2 (dba) 3 (92mg,0.1 equivalent, 0.10 mmol) and t-BuBrettPhos (48 mg,0.1 equivalent, 0.10 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 60% in 20 minutes); detector, UV 254/220nm, to give the sub-title compound (AFA-1) (39 mg,0.16mmol,17%,93% purity) as a yellow solid. M/z 231.1 (M+H) + (ES+)。
Step 2: synthesis of 4- [2- (ethylamino) -6- [6- (methylamino) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AFA-2)
To a stirred mixture of the product (AFA-1) (35 mg,1 eq, 0.15 mmol) from step 1 above and intermediate (ADP-7) (52 mg,1 eq, 0.15 mmol) in 1, 4-dioxane (5 mL) was added RuPhos ring palladium complex Gen.3 (13 mg,0.1 eq, 15. Mu. Mol) and RuPhos (14 mg,0.2 eq, 30. Mu. Mol) under nitrogen at room temperature. The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge Shield RP, 18 OBD column, 19 x 250mm,10 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 25 ml/min; gradient: 48% B to 60% B in 9 minutes; wavelength: 254/220nm; retention time: 8.03 To give the title compound (AFA-2) (11.7 mg,22 μmol,14%,99% purity) as a white solid. M/z 533.0 (M+H) + (ES+). 1 H NMR(300MHz,DMSO–d6)δ8.52(s,1H),8.22–8.10(m,2H),7.80–7.74(m,1H),7.42(d,J=1.2Hz,1H),7.12(d,J=2.1Hz,1H),7.02(d,J=2.2Hz,1H),6.77(t,J=5.4Hz,1H),6.51(d,J=5.3Hz,1H),5.96(d,J=1.3Hz,1H),4.98(s,2H),3.40(s,3H),3.23–3.13(m,2H),2.78(d,J=4.9Hz,3H),1.12(t,J=7.1Hz,3H)。
Example 179: synthesis of 2- [6- (ethylamino) -4- [ 4-methoxy-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AFB-8)
Step 1: synthesis of 2- (2, 6-dichloropyridin-4-yl) -5-methoxybenzoate (AFB-2)
To methyl 2-bromo-5-methoxybenzoate (AFB-1) (1.00 g,1 eq, 4.08 mmol), 2, 6-dichloropyridin-4-ylboronic acid (AAN-1) (860 mg,1.1 eq, 4.49 mmol) and K under a nitrogen atmosphere at room temperature 2 CO 3 (1.69 g,12.2mmol,3 eq.) in 1, 4-dioxane (15 ml) and H 2 Pd (dtBPF) Cl was added to the stirred mixture in O (1.5 mL) 2 (530 mg,0.2 eq, 0.82 mmol). The resulting mixture was stirred under nitrogen at 60 ℃ for 6 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (20% MeCN up to 80% in 20 minutes); detector, UV 254/220nm to give the sub-title compound (AFB-2) (871 mg,2.58mmol,66%,92% purity) as a yellow solid. M/z 312.0/314.0 (M+H) + (ES+)。
Step 2: synthesis of 2- (2, 6-dichloropyridin-4-yl) -5-methoxybenzoic acid (AFB-3)
The product from step 1 above (AFB-2) (823mg, 1 eq, 2.63 mmol) was taken up in THF (12 mL) and H 2 A solution in O (4 mL) was treated with LiOH (310 g,5 eq, 13.2 mmol). The resulting mixture was stirred at 70℃for 3 hours. The mixture was cooled to room temperature. Concentrating the mixtureHCl is acidified to pH 2. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 298.0/230.0 (M+H) + (ES+)。
Step 3: synthesis of 2- (2, 6-dichloropyridin-4-yl) -5-methoxy-N- [ (methylamino methylsulfonyl) amino ] benzamide (AFB-4)
To a stirred mixture of the product (AFB-3) (583 mg,1 eq, 2.63 mmol) from step 2 above in DMF (15 ml) was added DIPEA (2.03 g,6 eq, 15.8 mmol) and T at 0deg.C 3 P (50% in EA) (5.01 g,50wt%,6 eq., 15.8 mmol). 4-methyl-3-thiosemicarbazide (D-2) (206 mg,1 equivalent, 1.96 mmol) was added to the above mixture at 0 ℃. The resulting mixture was stirred at room temperature for 1.5 hours. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (10% MeCN up to 50% within 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (AFB-4) (500 mg,1.24mmol,49%,90% purity) as a white solid. M/z 365.0/367.0 (M+H) + (ES+)。
Step 4: synthesis of 5- [2- (2, 6-dichloropyridin-4-yl) -5-methoxyphenyl ] -4-methyl-1, 2, 4-triazole-3-thiol (AFB-5)
To a stirred mixture of the product from step 3 above (AFB-4) (500 mg,1 eq, 1.30 mmol) in DMF (10 ml) was added aqueous NaOH (5.2 mL,10M,4 eq, 5.20 mmol). The resulting mixture was stirred at 80℃for 1.5 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water. The mixture was acidified to pH 4 with concentrated HCl. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 70% in 15 minutes); detector, UV 254/220nm to give the sub-title compound (AFB-5) (225 mg,0.57mmol,77%,93% purity) as a white solid. M/z 367.0/369.0 (M+H) + (ES+)。
Step 5: synthesis of 2, 6-dichloro-4- [ 4-methoxy-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridine (AFB-6)
To a stirred mixture of the product from step 4 above (AFB-5) (225 mg,1 eq., 0.62 mmol) in DCM (10 ml) was added dropwise AcOH (184 mg,5 eq., 3.07 mmol) and H at 0deg.C 2 O 2 (347 mg,30Wt%,15 equivalents, 10.2 mmol). The resulting mixture was stirred at room temperature for 45 minutes. The resulting mixture was diluted with water. The mixture was treated with NaHCO 3 Is alkalized to pH 8. Filtering the resulting mixture; the filter cake was washed with brine (3×3 mL). The filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 40% in 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (AFB-6) (92 mg,0.27mmol,70%,97% purity) as a white solid. M/z 335.0/337.0 (M+H) + (ES+)。
Step 6: synthesis of 6-chloro-N-ethyl-4- [ 4-methoxy-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-amine (AFB-7)
To a stirred mixture of the product (AFB-6) from step 5 above (168 mg,1 equivalent, 0.50 mmol) in NMP (15 ml) at room temperature was added K 2 CO 3 (2.08 g,15.0mmol,30 eq.) and EtOH 2 -HCl (123 mg,3 eq, 1.50 mmol). The resulting mixture was stirred at 120℃for 7 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 50% within 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (AFB-7) (126 mg,0.33mmol,67%,90% purity) as a white solid. M/z344.1/346.1 (M+H) + (ES+)。
Step 7: synthesis of 2- [6- (ethylamino) -4- [ 4-methoxy-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AFB-8)
To a stirred mixture of the product (AFB-7) from step 6 above (120 mg,1 eq, 0.35 mmol) and intermediate (AC-2) (78 mg,1.1 eq, 0.39 mmol) in 1, 4-dioxane (10 mL) was added Cs 2 CO 3 (227 mg,2 equivalents, 0.70 mmol). RuPhos (67 mg,0.4 eq, 0.14 mmol) and RuPhos ring palladium complex Gen.3 (58 mg,0.2 eq, 0.07 mmol) were added to the above mixture under nitrogen at room temperature. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 1 hour. The mixture was cooled to room temperature. The resulting mixture was diluted with water. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 65% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (AFB-8) (58.8 mg,0.11mmol,33%,99% purity) as a green solid. M/z 509.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.46(s,1H),8.05(m,2H),7.78(t,J=7.7Hz,1H),7.56–7.43(m,2H),7.27(d,J=8.6,2.7Hz,1H),7.11(d,J=2.7Hz,1H),6.64(t,J=5.4Hz,1H),5.87(s,1H),5.18(s,2H),3.87(s,3H),3.32(s,3H),3.23–3.09(m,2H),1.12(t,J=7.1Hz,3H)。
Example 180: synthesis of methyl 4- [2- (ethylamino) -6- [ 1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzoate (AFC-1)
Under nitrogen atmosphereTo a stirred solution of compound (ADP-8) (10 mg,1 eq, 20. Mu. Mol) in MeOH (1 mL) at room temperature under an atmosphere was added 1, 4-dioxane (1 mL, 4M) containing HCl (gas). The resulting mixture was stirred under nitrogen at 60 ℃ for 2 days. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 51% B in 9 minutes; wavelength: 254/220nm; retention time: 8.05 To give the title compound (AFC-1) (2.7 mg,4.8 μmol,24%,96% purity) as a pale yellow solid. M/z 537.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.49(s,1H),8.28–8.23(m,1H),8.13–8.00(m,3H),7.81–7.74(m,2H),7.53(s,1H),6.79(t,J=5.4Hz,1H),5.96(s,1H),5.19(s,2H),3.92(s,3H),3.31(s,3H)3.21–3.12(m,2H),1.13(t,J=7.2Hz,3H)。
Example 181: synthesis of 2- [6- (ethylamino) -4- [4- (hydroxymethyl) -2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AFD-1)
To a stirred solution of compound (AFC-1) (40 mg,1 eq, 75. Mu. Mol) in MeOH (5 mL) under nitrogen at 0deg.C was added NaBH 4 (28 mg,10 equivalents, 0.75 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 days. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 51% B in 9 minutes; wavelength: 254/220nm; retention time: 8.05 To give the title compound (AFD-1) (5.4 mg,11 μmol,14%,99% purity) as a white solid. M/z 509.0 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO–d6)δ8.46(s,1H),8.09–8.01(m,2H),7.78(t,J=7.7Hz,1H),7.66–7.61(m,1H),7.56(d,J=7.9Hz,1H),7.53–7.47(m,2H),6.69(t,J=5.4Hz,1H),5.91(d,J=1.3Hz,1H),5.40(t,J=5.7Hz,1H),5.19(s,2H),4.63(d,J=5.2Hz,2H),3.29(s,3H),3.22–3.07(m,2H),1.12(t,J=7.1Hz,3H)。
Example 182: synthesis of 4- [2- (ethylamino) -6- {6- [ (2-hydroxy-2-methylpropyloxy) methyl ] -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl } pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AFE-1)
To a stirred solution of intermediate (AEV-1) (40 mg,1 eq, 72. Mu. Mol) and 2-methyl-propane-1, 2-diol (ADY-1) (65 mg,10 eq, 0.72 mmol) in DCM (5 mL) was added TBAB (12 mg,0.5 eq, 36. Mu. Mol) and aqueous KOH (5 mL,20 Wt%) at room temperature. The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (12/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 42% B to 62% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AFE-1) (3.4 mg, 5.6. Mu. Mol,7.7%,99% purity) as a yellow solid. M/z 606.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.52(s,1H),8.22–8.11(m,2H),8.00(d,J=15.0Hz,2H),7.79(d,J=8.0Hz,1H),7.48–7.39(m,1H),6.81(t,J=5.4Hz,1H),6.01–5.93(m,1H),5.17(s,2H),4.73(s,2H),4.47(s,1H),3.41(s,3H),3.26(s,2H),3.22–3.15(m,2H),1.19–1.05(m,9H)。
Example 183: synthesis of 2- [6- (ethylamino) -4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -6- { [ (1-hydroxycyclobutyl) methoxy ] methyl } -4- (trifluoromethyl) -3H-isoindol-1-one (AFF-2)
Step 1: synthesis of 6- (chloromethyl) -2- [6- (ethylamino) -4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AFF-1)
To a stirred solution of compound (AEF-1) (417 mg,1 eq, 0.79 mmol) in DCM (15 mL) at 0deg.C was added SOCl 2 (283 mg,3 eq, 2.38 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was quenched with MeOH (1 mL) at 0 ℃ and concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (10/1) to give the sub-title compound (AFF-1) (351 mg,0.59mmol,81%,92% purity) as a yellow solid. M/z 545.1/547.1 (M+H) + (ES+)。
Step 2: synthesis of 2- [6- (ethylamino) -4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -6- { [ (1-hydroxycyclobutyl) methoxy ] methyl } -4- (trifluoromethyl) -3H-isoindol-1-one (AFF-2)
To a stirred solution of the product (AFF-1) (40 mg,1 eq, 73. Mu. Mol) from step 1 above and 1- (hydroxymethyl) cyclobutan-1-ol (AED-1) (75 mg,10 eq, 0.73 mmol) in DCM (5 mL) was added TBAB (12 mg,0.5 eq, 36. Mu. Mol) and aqueous KOH (5 mL,20 Wt%) at room temperature. The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 52% B to 64% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (AFF-2) (5.8 m) as a white solidg, 9.4. Mu. Mol,13%,99% purity). M/z 611.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.49(s,1H),8.01(d,J=14.7Hz,2H),7.68–7.50(m,3H),7.47–7.43(m,1H),6.72(t,J=5.4Hz,1H),5.97–5.88(m,1H),5.16(s,2H),5.10(s,1H),4.83–4.74(m,2H),3.45(s,2H),3.38(d,J=2.9Hz,3H),3.24–3.14(m,2H),2.08–2.00(m,2H),1.99–1.89(m,2H),1.72–1.58(m,1H),1.55–1.40(m,1H),1.13(t,J=7.1Hz,3H)。
Example 184: synthesis of 2- [6- (ethylamino) -4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -6- [ (2-hydroxy-2-methylpropyloxy) methyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AFG-1)
To a stirred solution of intermediate (AFF-1) (35 mg,1 eq, 64. Mu. Mol) and 2-methyl-propane-1, 2-diol (ADY-1) (58 mg,10 eq, 0.64. Mu. Mol) in DCM (5 mL) at room temperature was added TBAB (11 mg,0.5 eq, 32. Mu. Mol) and aqueous KOH (5 mL,20 Wt%). The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 60% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (AFG-1) (3.3 mg, 5.4. Mu. Mol,8.5%,98% purity) as an off-white solid. M/z 599.1 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO–d6)δ8.49(s,1H),7.99(d,J=16.2Hz,2H),7.68–7.41(m,4H),6.71(t,J=5.4Hz,1H),5.92(s,1H),5.17(s,2H),4.73(s,2H),4.46(s,1H),3.37(s,3H),3.26(s,2H),3.23–3.10(m,2H),1.21–1.06(m,9H)。
Example 185: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AFH-1)
To intermediate (ADN-1) (60 mg,1 equivalent, 0.18 mmol), intermediate (AEE-1) (54 mg,1.2 equivalent, 0.22 mmol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 (119 mg,2 eq, 0.36 mmol) to a stirred solution of RuPhos (34 mg,0.4 eq, 73. Mu. Mol) and RuPhos ring palladium complex Gen.3 (31 mg,0.2 eq, 36. Mu. Mol) in 1, 4-dioxane (8 mL). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1). The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 10% B to 40% B over 7 min; wavelength: 254/220nm; retention time: 6.6) to give the title compound (AFH-1) (22.8 mg,42 μmol,23%,99% purity) as a white solid. M/z 539.6 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.51(s,1H),7.98(d,J=1.4Hz,1H),7.77–7.68(m,2H),7.65(d,J=1.8Hz,2H),7.60(t,J=8.6Hz,2H),6.80(d,J=1.5Hz,1H),4.96(d,J=35.4Hz,3H),3.86(s,2H),3.43(s,3H),2.53(s,2H),1.99(t,J=8.8Hz,3H),1.94–1.84(m,2H),1.60(d,J=10.6Hz,1H),1.46–1.32(m,1H),0.95(d,J=6.4Hz,4H)。
Example 186: synthesis of 2- { 6-cyclopropyl-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AFI-1)
/>
To the intermediate at room temperature under nitrogen atmosphere(ADR-1) (50 mg,1 equivalent, 0.16 mmol), intermediate (AEE-1) (39 mg,1 equivalent, 0.16 mmol) and Cs 2 CO 3 (104 mg,0.32mmol,2 eq.) to a stirred mixture of 1, 4-dioxane (5 mL) was added RuPhos (30 mg,0.4 eq., 64. Mu. Mol) and RuPhos ring palladium complex Gen.3 (27 mg,0.2 eq., 32. Mu. Mol). The resulting mixture was stirred overnight at 80 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 27% B to 37% B in 9 minutes; wavelength: 254/220nm; retention time: 8.9 to give the title compound (AFI-1) (19 mg, 35. Mu. Mol,22%,96% purity) as a white solid. M/z 525.5 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.48(s,1H),8.23(d,J=1.3Hz,1H),7.90(s,1H),7.83(s,1H),7.70(d,J=7.4Hz,1H),7.64(d,J=7.8Hz,1H),7.07(d,J=1.4Hz,1H),5.07(s,2H),4.02(s,3H),3.95(s,2H),3.55(s,3H),2.72(s,2H),2.14–2.00(m,5H),1.73(s,1H),1.56–1.44(m,1H),1.15–1.06(m,2H),1.06–0.97(m,2H)。
Example 187: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AFJ-1)
To intermediate (AEG-2) (60 mg,1 equivalent, 0.18 mmol), intermediate (AEE-1) (57 mg,1.3 equivalent, 0.23 mmol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 (116 mg,2 eq, 0.36 mmol) to a stirred solution of 1, 4-dioxane (8 mL) was added RuPhos (33 mg,0.4 eq, 72. Mu. Mol) and RuPhos ring palladium complex Gen.3 (30 mg,0.2 eq, 36. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH(15/1) purification. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 48% B in 8 minutes; wavelength: 254/220nm; retention time: 7.3 To give the title compound (AFJ-1) (3.7 mg,6.6 μmol,3.8%,97% purity) as a white solid. M/z 546.5 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.54(s,1H),8.25–8.18(m,2H),7.99(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.75(s,1H),7.65(t,J=1.3Hz,2H),6.88(d,J=1.4Hz,1H),5.00(s,2H),4.91(s,1H),3.86(s,2H),3.48(s,3H),2.53(s,2H),2.09–1.95(m,3H),1.89(d,J=12.1,9.4Hz,2H),1.64–1.57(m,1H),1.40(d,J=10.9,8.9Hz,1H),1.00–0.93(m,4H)。
Example 188: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- (hydroxymethyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AFK-1)
To intermediate (ADN-1) (45 mg,1 equivalent, 0.14 mmol), intermediate (ADX-1) (38 mg,1.2 equivalent, 0.16 mmol) and Cs under nitrogen at room temperature 2 CO 3 (134 mg,3 eq, 0.41 mmol) to a stirred solution of RuPhos ring palladium complex Gen.3 (23 mg,0.2 eq, 27. Mu. Mol) and Ruphos (26 mg,0.4 eq, 55. Mu. Mol) in 1, 4-dioxane (3 mL) were added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1). The crude product was purified by preparative HPLC (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 34% B to 64% B in 7 min;wavelength: 254/220nm; retention time: 6.3 To give the title compound (AFK-1) (19.4 mg,37 μmol,27%,99% purity) as a white solid. M/z 524.0 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO–d6)δ8.51(s,1H),8.01–7.96(m,3H),7.74–7.71(m,1H),7.64–7.53(m,2H),6.81(d,J=1.5Hz,1H),5.59(t,J=5.8Hz,1H),5.16(s,2H),4.70(d,J=5.8Hz,2H),3.43(s,3H),2.07–2.01(m,1H),0.99–0.93(m,4H)。
Example 189: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- { [ (1-hydroxycyclobutyl) methoxy ] methyl } -4- (trifluoromethyl) -3H-isoindol-1-one (AFL-2)
Step 1: synthesis of 6- (chloromethyl) -2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (AFL-1)
To a stirred solution of compound (AFK-1) (40 mg,1 eq, 76. Mu. Mol) in DCM (3 mL) at 0deg.C was added SOCl 2 (14 mg,1.5 eq, 0.11 mmol). The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was diluted with water. The resulting mixture was extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC (DCM/MeOH 15/1) to give the sub-title compound (AFL-1) (25 mg, 44. Mu. Mol,60%,95% pure) as a yellow solid. M/z 542.1/544.1 (M+H) + (ES+)。
Step 2: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- { [ (1-hydroxycyclobutyl) methoxy ] methyl } -4- (trifluoromethyl) -3H-isoindol-1-one (AFL-2)
To a stirred solution of the product from step 1 above (AFL-1) (25 mg,1 eq, 46. Mu. Mol) and 1- (hydroxymethyl) cyclobutan-1-ol (AED-1) (47 mg,10 eq, 0.46 mmol) in DCM (5 mL) at room temperature was addedTBAB (7.4 mg,0.5 eq, 23. Mu. Mol). To the above mixture was added an aqueous KOH solution (5 mL,20 Wt%). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water. The resulting mixture was extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1). The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 20% B to 50% B over 7 min; wavelength: 254/220nm; retention time: 6.3) to give the title compound (AFL-2) as a white solid (4.8 mg,7.5 μmol,16%,95% purity). M/z 608.1 (M+H) + (ES+). 1 H NMR(400MHz,MeOH–d4)δ8.47(s,1H),8.08(s,1H),8.01–8.00(m,2H),7.79–7.77(m,1H),7.57–7.45(m,2H),6.88(d,J=1.5Hz,1H),5.17(s,2H),4.77(s,2H),3.57(s,2H),3.46(s,3H),2.19–2.13(m,2H),2.09–2.01(m,3H),1.79–1.76(m,1H),1.61–1.54(m,1H),1.00–0.98(m,4H)。
Example 190: synthesis of 2- [6- (ethylamino) -4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -6- [ (methoxymethoxy) methyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AFM-2)
To a stirred solution of intermediate (AFF-1) (35 mg,1 eq, 64. Mu. Mol) and 2-methoxyethanol (AFM-1) (49 mg,10 eq, 0.64 mmol) in DCM (5 mL) was added TBAB (11 mg,0.5 eq, 32. Mu. Mol) and aqueous KOH (5 mL,20 Wt%) at room temperature. The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (10/1). The crude product was purified by preparative HPLC under the following conditionsPurification under (column: XBridge-prepared OBD C18 column, 30X 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 55% B to 65% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (AFM-2) (3.5 mg, 5.9. Mu. Mol,9.5%,99% purity) as a white solid. M/z 585.5 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.52(s,1H),7.98(d,J=14.8Hz,2H),7.67–7.50(m,3H),7.43(s,1H),6.71(s,1H),5.93(s,1H),5.16(s,2H),4.70(s,2H),3.65–3.62(m,2H),3.53–3.51(m,2H),3.38(s,3H),3.27(s,3H),3.22–3.15(m,2H),1.13(t,J=7.2Hz,3H)。
Example 191: synthesis of 2- [4' -fluoro-2 ' - (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl ] -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AFN-3)
Step 1: synthesis of 4' -fluoro-2 ' - (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-amine) (AFN-1)
To intermediate (ABO-4) (400 mg,1 equivalent, 1.56 mmol), 3-aminophenylboronic acid (AAS-1) (235 mg,1.1 equivalent, 1.72 mmol) and K under a nitrogen atmosphere at room temperature 2 CO 3 (649 mg,3 eq, 4.68 mmol) in 1, 4-dioxane (15 mL) and H 2 Pd (dppf) Cl was added to the stirred mixture in O (1.5 mL) 2 DCM (127 mg,0.1 eq, 0.16 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 6 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1) to give the sub-title compound (AFN-1) (283 mg,0.97mmol,67%,92% purity) as a white solid. M/z 269.1 (M+H) + (ES+)。
Step 2: synthesis of 3- {3 '-bromo-4-fluoro- [1,1' -biphenyl ] -2-yl } -4-methyl-1, 2, 4-triazole (AFN-2)
Under nitrogen atmosphere at 0 ℃, to t-BuNO 2 To a stirred mixture of (231 mg,6 eq, 2.24 mmol) in MeCN (8 mL) was added CuBr (80 mg,1.5 eq, 0.56 mmol). The resulting mixture was stirred under nitrogen atmosphere at 0 ℃ for 1 hour. To the above mixture was added MeCN (1 mL) containing the product (AFN-1) from step 1 above (100 mg,1 eq, 0.37 mmol) at room temperature. The resulting mixture was stirred at room temperature for 10 minutes. The resulting mixture was then stirred at 50℃for 1.5 hours. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1) to give the sub-title compound (AFN-2) (64 mg,0.18mmol,52%,95% pure) as a white solid. M/z332.0/334.0 (M+H) + (ES+)。
Step 3: synthesis of 2- [4' -fluoro-2 ' - (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl ] -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AFN-3)
To a stirred mixture of the product (AFN-2) (46 mg,1 eq, 0.14 mmol) from step 2 above and intermediate (ADR-2) (48 mg,1.1 eq, 0.15 mmol) in 1, 4-dioxane (15 mL) at room temperature was added Cs 2 CO 3 (90 mg,2 equivalents, 0.28 mmol). RuPhos (26 mg,0.4 eq, 55. Mu. Mol) and RuPhos ring palladium complex Gen.3 (24 mg,0.2 eq, 29. Mu. Mol) were added to the above mixture under nitrogen atmosphere at room temperature. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 1 hour. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; gradient: 38% MeCN up to 48% in 9 minutes; wavelength: 254/220 nm) to give the title compound (AFN-3) (6.2 mg, 11. Mu. Mol,7.7%,96% purity). M/z 566.1 (M+H) + (ES+)。 1 H NMR(300MHz,DMSO–d6)δ8.42(s,1H),8.01(m,3H),7.76–7.33(m,5H),6.88(d,J=7.7Hz,1H),5.08(s,2H),4.94(s,1H),3.96(s,2H),3.13(s,3H),2.53(s,2H),2.05–1.86(m,4H),1.61(m,1H),1.44–1.34(m,1H)。
Example 192: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -3H-isoindol-1-one (AFO-2)
Step 1: synthesis of 6- ((((1R, 2S) -2-hydroxycyclopentyl) amino) methyl) isoindolin-1-one (AFO-1)
To MeOH (10 mL) containing stirred solution (1S, 2R) -2-aminocyclopentan-1-ol (Z-1) (151 mg,1.2 eq, 1.49 mmol) was added DIPEA (640 mg,4 eq, 5.00 mmol) at room temperature. The resulting mixture was stirred at 60℃for 10 minutes. Intermediate (ACY-2) (200 mg,1 eq, 1.24 mmol) was then added at room temperature. The resulting mixture was stirred at 60℃for 2 hours. Adding NaBH to the above mixture at 0deg.C 4 (235 mg,5 eq, 6.21 mmol). The resulting mixture was stirred at room temperature for 1 hour. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 20% in 20 minutes); detector, UV 254/220nm to give the sub-title compound (AFO-1) (144 mg,0.51mmol,47%,88% purity) as a white solid. M/z 247.1 (M+H) + (ES+)。
Step 2: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -3H-isoindol-1-one (AFO-2)
To intermediate (ADN-1) (50 mg,1 equivalent, 0.15 mmol), the product from step 1 above (AFO-1) (37 mg,1 equivalent, 0.15 mmol) and Cs under nitrogen at room temperature 2 CO 3 (99 mg,2 eq, 0.30 mmol) in 1, 4-dioxaneTo a stirred solution of RuPhos (28 mg,0.4 eq, 61. Mu. Mol) and RuPhos ring palladium complex Gen.3 (25 mg,0.2 eq, 30. Mu. Mol) were added in alkane (6 mL). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35% B to 52% B in 9 minutes; wavelength: 254/220nm; retention time: 8.03 To give the title compound (AFO-2) (23.0 mg,42 μmol,28%,99% purity) as a white solid. M/z 539.2 (M+H) + (ES+). 1 H NMR(300MHz,DMSO-d6)δ8.51(s,1H),7.97(d,J=1.4Hz,1H),7.80–7.68(m,2H),7.68–7.53(m,4H),6.81(d,J=1.4Hz,1H),5.00(s,2H),4.39(s,1H),4.00–3.90(m,1H),3.91–3.73(m,2H),3.44(s,3H),2.83–2.69(m,1H),2.26(s,1H),2.08–1.95(m,1H),1.77–1.47(m,4H),1.48–1.31(m,2H),0.95(d,J=6.4Hz,4H)。
Example 193: synthesis of 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -6- { [ (1-fluorocyclobutyl) methoxy ] methyl } -4- (trifluoromethyl) -3H-isoindol-1-one (AFP-2)
To a stirred solution of intermediate (ADA-1) (40 mg,1 eq, 75 μmol) and (1-fluorocyclobutyl) MeOH (78 mg,10 eq, 0.75 mmol) in DCM (3 mL) was added TBAB (49 mg,2 eq, 0.15 mmol) and aqueous KOH (3 mL,20 wt%) at room temperature. The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC under the following conditions (column: sunfirePreparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 43% B to 67% B in 7 minutes; wavelength: 254/220nm; retention time: 6.8 To give the title compound (AFP-2) (14.9 mg,25 μmol,33%,99% purity) as a white solid. M/z 599.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.47(s,1H),8.00(d,J=16.4Hz,2H),7.89(s,1H),7.57(d,J=1.2Hz,1H),6.93(t,J=5.4Hz,1H),6.25(d,J=1.3Hz,1H),5.21(s,2H),4.77(s,2H),3.89(s,3H),3.69(d,J=24.6Hz,2H),3.47(s,3H),3.29–3.20(m,2H),2.28–2.15(m,4H),1.82–1.72(m,1H),1.56–1.45(m,1H),1.17(t,J=7.2Hz,3H)。
Example 194: synthesis of 2- { 6-cyclopropyl-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclopentyl) methyl ] amino } methyl) -3H-isoindol-1-one (AFQ-3)
Step 1: synthesis of 6- ((((1-hydroxycyclopentyl) methyl) amino) methyl) isoindolin-1-one (AFQ-2)
To a stirred solution of 1- (aminomethyl) cyclopentan-1-ol (AFQ-1) (172 mg,1.2 eq, 1.49 mmol) in MeOH (10 mL) was added DIPEA (640 mg,4 eq, 4.96 mmol) and intermediate (ACY-2) (200 mg,1 eq, 1.24 mmol) at room temperature. The resulting mixture was stirred at 60℃for 2 hours. Then NaBH is added at 0 DEG C 4 (235 mg,5 eq, 6.21 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by addition of 2mL of ice water at 0 ℃. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 30% in 20 minutes); detector, UV 254/220nm to give the sub-title compound (AFQ-2) (131 mg,0.45mmol,40%,90% purity) as a white solid. M/z 261.2 (M+H) + (ES+)。
Step 2: synthesis of 2- { 6-cyclopropyl-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclopentyl) methyl ] amino } methyl) -3H-isoindol-1-one (AFQ-3)
To intermediate (ADR-1) (60 mg,1 equivalent, 0.19 mmol), the product from step 1 above (AFQ-2) (50 mg,1 equivalent, 0.19 mmol) and Cs under nitrogen at room temperature 2 CO 3 (124 mg,2 eq, 0.38 mmol) to a stirred mixture of 1, 4-dioxane (5 mL) was added RuPhos (36 mg,0.4 eq, 76. Mu. Mol) and RuPhos ring palladium complex Gen.3 (32 mg,0.2 eq, 38. Mu. Mol). The resulting mixture was stirred overnight at 80 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19X 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30B to 60B in 7 minutes; a detector, UV 254/210nm; retention time: 6.08. this gave the title compound (AFQ-3) (24.5 mg, 45. Mu. Mol,24%,99% purity) as a white solid. M/z 539.5 (M+H) + (ES+). 1 H NMR(300MHz,MeOH-d4)δ8.49(d,J=4.5Hz,1H),8.22(d,J=1.3Hz,1H),7.92(d,J=7.7Hz,2H),7.83–7.69(m,2H),7.10(d,J=1.3Hz,1H),5.11(s,2H),4.29(s,2H),4.02(s,3H),3.56(s,3H),2.99(s,2H),2.17–2.03(m,1H),1.70(t,J=7.8Hz,8H),1.64(s,2H),1.16–1.07(m,2H)。
Example 195: synthesis of 4- (2- {6- [2- (dimethylamino) -1-hydroxyethyl ] -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl } -6- (ethylamino) pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AFR-7)
Step 1: synthesis of 6- (1-ethoxyvinyl) -4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (AFR-2)
Intermediate (ABC-2) (1.5 g,1 eq, 5.36 mmol), tributyl (1-ethoxyvinyl) stannane (AFR-1) (2.32 g,1.2 eq, 6.43 mmol) was added to 1, 4-dioxane (15 m) at room temperature under nitrogen atmospherePd (PPh) was added to the stirred solution in L) 3 ) 2 Cl 2 (376 mg,0.1 eq, 0.54 mmol). The resulting mixture was stirred overnight at 80 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (1/3) to give the sub-title compound (AFR-2) (1.2 g,3.76mmol,83%,85% purity) as a brown yellow solid. M/z 272.1 (M+H) + (ES+)。
Step 2: synthesis of 6-acetyl-4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (AFR-3)
To a stirred solution of the product from step 1 above (AFR-2) (600 mg,1 eq, 2.21 mmol) and TsOH (1.90 g,5 eq, 11.1 mmol) in DCM (20 mL) at room temperature. The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (3/1) to give the sub-title compound (AFR-3) as an off-white solid (420 mg,1.59mmol,78%,92% purity). M/z 244.1 (M+H) + (ES+)。
Step 3: synthesis of 6- (2-bromoacetyl) -4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (AFR-4)
A stirred solution of the product from step 2 above (AFR-3) (300 mg,1 eq, 1.23 mmol) and bromine (197mg, 1 eq, 1.23 mmol) in HBr (10 mL) was stirred at 70℃for 1 hour at room temperature. The mixture was cooled to room temperature. The residue was purified by preparative TLC using DCM/MeOH (15/1) to give the sub-title compound (AFR-4) as an off-white solid (170 mg,0.48mmol,43%,90% purity). M/z 322.0/324.0 (M+H) + (ES+)。
Step 4: synthesis of 6- [2- (dimethylamino) acetyl ] -4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (AFR-5)
The product from step 3 above (AFR-4) (230 mg,1 eq, 0.71 mmol) and dimethylamine (2M in THF) (0.02 mL,10 eq, 0.31 mmol) were combined in THF (3mL) was stirred at room temperature for 2 hours. The crude product was used directly in the next step without further purification. M/z 287.1 (M+H) + (ES+)。
Step 5: synthesis of 6- [2- (dimethylamino) -1-hydroxyethyl ] -4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (AFR-6)
Adding NaBH to the mixture at 0deg.C 4 (152 mg,5 eq, 4.02 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of 2mL of MeOH at 0 ℃. The residue was purified by preparative TLC using DCM/MeOH (15/1) to give the sub-title compound (AFR-6) (130 mg,0.40mmol,56% pure) as an off-white solid. M/z 289.1 (M+H) + (ES+)。
Step 6: synthesis of 4- (2- {6- [2- (dimethylamino) -1-hydroxyethyl ] -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl } -6- (ethylamino) pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AFR-7)
To intermediate (ADP-7) (130 mg,1 equivalent, 0.38 mmol), the product from step 5 above (AFR-6) (122 mg,1.1 equivalent, 0.42 mmol) and Cs under nitrogen at room temperature 2 CO 3 (250 mg,2 eq, 0.77 mmol) to a stirred solution of RuPhos (72 mg,0.4 eq, 0.15 mmol) and RuPhos cyclopalladium complex Gen.3 (64 mg,0.2 eq, 77. Mu. Mol) in 1, 4-dioxane (10 mL). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The residue was purified by preparative TLC using DCM/MeOH (15/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 20% B to 30% B in 7 minutes; wavelength: 254/220 nm) to give the title compound (AFR-7) (55 mg, 92. Mu. Mol,24%,99% purity) as a yellow solid. M/z 591.6 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.48(s,1H),8.12–7.96(m,4H),7.87(d,J=8.1Hz,1H),7.50(d,J=1.4Hz,1H),6.06(d,J=1.2Hz,1H),5.20–5.15(m,2H),5.02–4.98(m,1H),3.46(s,3H),3.27(t,J=7.2Hz,2H),2.71–2.63(m,1H),2.55–2.49(m,1H),2.38(s,6H),1.20(t,J=7.2Hz,3H)。
Example 196: synthesis of 6-acetyl-2- { 6-cyclopropyl-4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (AFS-1)
To an 8mL sealed tube was added under nitrogen at room temperature intermediate (ADZ-1) (30 mg,1 eq, 97. Mu. Mol), intermediate (AFR-3) (35 mg,1.5 eq, 0.15 mmol) and Cs 2 CO 3 (94 mg,3 eq, 0.29 mmol) of 1, 4-dioxane (1 mL). Pd (OAc) was added to the above mixture over 2 minutes at room temperature under nitrogen atmosphere 2 (2.2 mg,0.1 eq, 10. Mu. Mol) and XantPhos (11 mg,0.2 eq, 19. Mu. Mol). The resulting mixture was stirred under nitrogen at 100 ℃ for an additional 3 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18OBD column, 19X 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30B to 60B in 7 minutes; a detector, UV 254/210nm; retention time: 6.08 to give the title compound (AFS-1) (5.0 mg, 9.5. Mu. Mol,9.8%,98% purity) as a white solid. M/z 518.1 (M+H) + (ES+). 1 H NMR(400MHz,MeOH–d4)δ8.61(d,J=1.3Hz,1H),8.47(d,J=9.2Hz,2H),8.03(d,J=1.4Hz,1H),7.85–7.73(m,2H),7.73–7.61(m,2H),6.93(d,J=1.4Hz,1H),5.25(d,J=1.6Hz,2H),3.46(s,3H),2.73(s,3H),2.10–2.00(m,1H),1.07–0.92(m,4H)。
Example 197: synthesis of 2- { 6-cyclopropyl-4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- [ (1S) -1- { [ (1-hydroxycyclobutyl) methyl ] amino } ethyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AFT-2). Stereochemistry was arbitrarily specified.
Step 1: synthesis of 2- { 6-cyclopropyl-4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- (1- { [ (1-hydroxycyclobutyl) methyl ] amino } ethyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AFT-1)
To an 8mL sealed tube was added MeOH (1 mL) containing compound (AFS-1) (40 mg,1 eq, 77. Mu. Mol), 1- (aminomethyl) cyclobutan-1-ol, HCl (AW-1) (16 mg,2 eq, 0.15 mmol) and AcOH (14 mg,3 eq, 0.23 mmol) at room temperature. NaBH was added to the above mixture at room temperature over 2 minutes 3 CN (24 mg,5 eq, 0.39 mmol). The resulting mixture was stirred at room temperature overnight. Filtering the resulting mixture; the filter cake was washed with MeOH (2X 2 mL). The filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 48% in 20 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AFT-1) (11 mg, 17. Mu. Mol,24%,95% purity) as a white solid. M/z 603.4 (M+H) + (ES+)。
Step 2: synthesis of 2- { 6-cyclopropyl-4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- [ (1S) -1- { [ (1-hydroxycyclobutyl) methyl ] amino } ethyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AFT-2)
The product (AFT-1) from step 1 above (40 mg,1 eq, 77. Mu. Mol) was isolated by chiral preparative HPLC under the following conditions: column: CHIRALPAK IG,2×25cm,5 μm; mobile phase a: hex: dcm=3:1 (0.5% 2m NH 3 MeOH) -HPLC, mobile phase B: etOH-HPLC; flow rate: 20 ml/min; gradient: 30% B to 30% B in 30 minutes; wavelength: 220/254nm; retention time 1 (min): 14.24; retention time 2 (minutes): 21.388; sample solvent: etOH-HPLC; injection volume: 4mL. This gave the title compound (AFT-2) (2.0 mg, 3.2. Mu. Mol,18%,98% purity) as a white solid. M/z 603.4 (M+H) + (ES+)。 1 H NMR (400 MHz, meOH-d 4) delta 8.45 (s, 1H), 8.14-7.98 (m, 3H), 7.83-7.73 (m, 2H), 7.73-7.60 (m, 2H), 6.89 (d, J=1.4 Hz, 1H), 5.18 (d, J=1.8 Hz, 2H), 4.10-3.95 (m, J=6.6 Hz, 1H), 3.44 (s, 3H), 2.65 (d, J=11.8 Hz, 1H), 2.45 (d, J=11.8 Hz, 1H), 2.15-1.92 (m, 5H), 1.82-1.63 (m, 1H), 1.53-1.35 (m, 4H), 1.05-0.83 (m, 4H). Column: CHIRALPAK IG-3,4.6 x 50mm 3um; mobile phase a: hex: dcm=3:1) (0.1% DEA): etoh=70:30; flow rate: 1 ml/min; retention time: 2.712.
Example 198: synthesis of 2- { 6-cyclopropyl-4- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- [ (1R) -1- { [ (1-hydroxycyclobutyl) methyl ] amino } ethyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AFU-1). Stereochemistry was arbitrarily specified.
The racemic product (AFT-1) (40 mg,1 eq., 77. Mu. Mol) was separated by chiral preparative HPLC under the following conditions: column: CHIRALPAK IG,2×25cm,5 μm; mobile phase a: hex: dcm=3:1 (0.5% 2m NH 3 MeOH) -HPLC, mobile phase B: etOH-HPLC; flow rate: 20 ml/min; gradient: 30% B to 30% B in 30 minutes; wavelength: 220/254nm; retention time 1 (min): 14.24; retention time 2 (minutes): 21.388; sample solvent: etOH-HPLC; injection volume: 4mL. This gave the title compound (AFU-1) (2.5 mg, 4.0. Mu. Mol,23%,97% purity) as a white solid. M/z 603.4 (M+H) + (ES+)。 1 H NMR (400 MHz, meOH-d 4) delta 8.45 (s, 1H), 8.14-7.99 (m, 3H), 7.84-7.72 (m, 2H), 7.72-7.60 (m, 2H), 6.90 (d, J=1.4 Hz, 1H), 5.18 (d, J=1.6 Hz, 2H), 4.10-3.97 (m, 1H), 3.44 (s, 3H), 2.65 (d, J=11.8 Hz, 1H), 2.45 (d, J=11.9 Hz, 1H), 2.15-1.96 (m, 5H), 1.79-1.63 (m, 1H), 1.52-1.35 (m, 4H), 1.06-0.94 (m, 4H). Column: CHIRALPAK IG-3,4.6 x 50mm,3 μm; mobile phase a: hex: dcm=3:1) (0.1% DEA): etoh=70:30; flow rate: 1 ml/min; retention time: 4.162.
Example 199: synthesis of 2- { 6-cyclopropyl-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl } -6- { [ (1-hydroxycyclobutyl) methoxy ] methyl } -4- (trifluoromethyl) -3H-isoindol-1-one (AFV-3)
Step 1: synthesis of 2- { 6-cyclopropyl-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl } -6- (hydroxymethyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AFV-1)
To intermediate (ADX-1) (100 mg,1 equivalent, 0.43 mmol), intermediate (ADR-1) (150 mg,1.1 equivalent, 0.48 mmol) and Cs under nitrogen at room temperature 2 CO 3 (423 mg,3 eq, 1.30 mmol) to a stirred solution of RuPhos ring palladium complex gen.3 (72 mg,0.2 eq, 87. Mu. Mol) and RuPhos (81 mg,0.4 eq, 0.17 mmol) in 1, 4-dioxane (4 mL) was added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AFV-1) (150 mg,0.26mmol,68%,90% purity) as a yellow solid. M/z510.2 (M+H) + (ES+)。
Step 2: synthesis of 6- (chloromethyl) -2- { 6-cyclopropyl-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (AFV-2)
SOCl was added dropwise to a stirred solution of the product (AFV-1) (100 mg,1 eq, 0.20 mmol) from step 1 above in DCM (5 mL) under nitrogen at 0deg.C 2 (35 mg,1.5 eq, 0.29 mmol). The resulting mixture was stirred under nitrogen atmosphere at 0 ℃ for 2 hours. The reaction was quenched with MeOH (2 mL) at 0 ℃ and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AFV-2) (80 mg,0.14mmol,77%,92% purity) as an off-white solid. M/z 528.1/530.1 (M+H) + (ES+)。
Step 3: synthesis of 2- { 6-cyclopropyl-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl } -6- { [ (1-hydroxycyclobutyl) methoxy ] methyl } -4- (trifluoromethyl) -3H-isoindol-1-one (AFV-3)
To a stirred solution of the product from step 2 above (AFV-2) (40 mg,1 eq, 76. Mu. Mol) and 1- (hydroxymethyl) cyclobutan-1-ol (AED-1) (77 mg,10 eq, 0.76 mmol) in DCM (3 mL) was added TBAB (12 mg,0.5 eq, 38. Mu. Mol) and aqueous KOH (5 mL,20 Wt%) at room temperature. The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 15% B to 75% B in 8 minutes; wavelength: 254/220nm; retention time: 6.48 To give the title compound (AFV-3) (9.3 mg,15 μmol,21%,99% purity) as a white solid. M/z 594.1 (M+H) + (ES+) 1 H NMR(400MHz,CD3OD)δ8.48(s,1H),8.23(d,J=1.3Hz,1H),8.09(s,1H),8.02(s,1H),7.90(s,1H),7.10(d,J=1.3Hz,1H),5.22(s,2H),4.78(s,2H),4.02(s,3H),3.56(d,J=4.8Hz,5H),2.19–2.01(m,5H),1.78–1.72(m,1H),1.61–1.53(m,1H),1.11–1.01(m,4H)。
Example 200: synthesis of 2- {4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6- (prop-1-en-2-yl) pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AFW-3)
Step 1: synthesis of 2-chloro-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6- (prop-1-en-2-yl) pyridine (AFW-2)
In nitrogen atmosphereIntermediate (ACM-6) (200 mg,1 eq, 0.62 mmol) and 4, 5-tetramethyl-2- (prop-1-en-2-yl) -1,3, 2-dioxaborolan (AFW-1) (62 mg,0.6 eq, 0.37 mmol) were added at room temperature 2 CO 3 (257 mg,3 equivalents, 1.86 mmol) in 1, 4-dioxane (10 mL) and H 2 Pd (dppf) Cl was added to the stirred solution in O (1 mL) 2 DCM (101 mg,0.2 eq, 0.12 mmol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 50% in 27 minutes); detector, UV 254/220nm. This gave the sub-title compound (AFW-2) (50 mg,0.14mmol,25%,95% purity) as a brown solid. M/z 329.1/331.1 (M+H) + (ES+)。
Step 2: synthesis of 2- {4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6- (prop-1-en-2-yl) pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AFW-3)
To the product (AFW-2) (40 mg,1 equivalent, 0.12 mmol), intermediate (AEE-1) (33 mg,1.1 equivalent, 0.13 mmol) and Cs from step 1 above, under a nitrogen atmosphere at room temperature 2 CO 3 (79 mg,2 eq, 0.24 mmol) to a stirred solution of RuPhos (23 mg,0.4 eq, 49. Mu. Mol) and RuPhos cyclopalladium complex Gen.3 (20 mg,0.2 eq, 24. Mu. Mol) in 1, 4-dioxane (5 mL) were added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 50% in 15 minutes); detector, UV 254/220nm. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 48% B in 8 minutes; wavelength: 254/220 nm) to give the title compound as a white solidAFW-3 (9.6 mg, 17. Mu. Mol,15%,98% purity). M/z 539.4 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.51(s,1H),8.25(d,J=1.3Hz,1H),7.87–7.76(m,2H),7.74–7.56(m,4H),7.00(d,J=1.3Hz,1H),5.94(t,J=1.4Hz,1H),5.32(t,J=1.8Hz,1H),5.11(s,2H),4.92(s,1H),3.87(s,2H),3.40(s,3H),2.54(s,2H),2.07(s,3H),2.04–1.95(m,2H),1.95–1.84(m,2H),1.66–1.55(m,1H),1.45–1.32(m,1H)。
Example 201: synthesis of 2- {4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-isopropylpyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AFX-1)
A stirred solution of compound (AFW-3) (65 mg,1 eq, 0.12 mmol) and Pd/C form 87L (10 mg,0.78 eq, 94. Mu. Mol) in THF (10 mL) was stirred at room temperature under nitrogen. The resulting mixture was stirred at room temperature under a hydrogen atmosphere for 2 hours. Filtering the resulting mixture; the filter cake was washed with MeOH (3X 5 mL). The filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 31% B to 48% B in 10 minutes; wavelength: 254/220 nm) to give the title compound (AFX-1) (2.2 mg, 3.5. Mu. Mol,3.4%,95% purity) as a white solid. M/z 541.5 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (d, j=15.2 hz, 1H), 8.22 (d, j=1.5 hz, 1H), 7.86 (d, j=1.6 hz, 1H), 7.82-7.76 (m, 1H), 7.72-7.68 (m, 1H), 7.65 (d, j=7.9 hz, 1H), 7.57-7.50 (m, 1H), 7.50-7.44 (m, 1H), 6.77 (d, j=1.5 hz, 1H), 5.12 (s, 2H), 3.98 (s, 2H), 3.40 (s, 3H), 2.99-2.88 (m, 1H), 2.75 (s, 2H), 2.15-1.97 (m, 4H), 1.78-1.68 (m, 1H), 1.58-1.44 (m, 1H), 6.24 (d, 1.5hz, 1H).
Example 202: synthesis of 2- (4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6-isopropylpyridin-2-yl) -6-methylisoindolin-1-one (AFY-1)
The by-product of example 201 was obtained as a compound (AFY-1) (2 mg, 4.5. Mu. Mol,3.8%,99% purity) in the form of a white solid. M/z 442.4 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.47(s,1H),8.20(d,J=1.4Hz,1H),7.83–7.75(m,1H),7.64(s,1H),7.58–7.42(m,4H),6.75(d,J=1.4Hz,1H),5.07(s,2H),3.39(s,3H),3.00–2.90(m,1H),2.46(s,3H),1.23(d,J=6.9Hz,6H)。
Example 203: synthesis of 2- [6- (ethylamino) -4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] oxy } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AFZ-1)
To a stirred solution of intermediate (AFF-1) (40 mg,1 eq, 75. Mu. Mol) and (1R, 2S) -cyclopentane-1, 2-diol (AV-2) (38 mg,5 eq, 0.38 mmol) in DCM (2 mL) was added TBAB (12 mg,0.5 eq, 37. Mu. Mol) and aqueous KOH (2 mL,20 Wt%) at room temperature. The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with DCM (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (20/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35% B to 68% B in 9 minutes; wavelength: 254/220nm; retention time: 8.73 To give the title compound (AFZ-1) (10.2 mg, 16. Mu. Mol,22%,98% purity) as a white solid. M/z 611.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.49(s,1H),8.04(d,J=14.0Hz,2H),7.66–7.61(m,1H),7.60–7.56(m,1H),7.56–7.49(m,1H),7.44(d,J=1.2Hz,1H),6.72(t,J=5.4Hz,1H),5.93(d,J=1.3Hz,1H),5.16(s,2H),4.79(d,J=12.8Hz,1H),4.69(d,J=12.8Hz,1H),4.49(d,J=4.8Hz,1H),4.05–3.98(m,1H),3.39–3.34(s,3H),3.74–3.68(m,1H),3.22–3.11(m,2H),1.84–1.64(m,4H),1.62–1.53(m,1H),1.48–1.38(m,1H),1.13(t,J=7.2Hz,3H)。
Example 204: synthesis of 2- { 6-cyclopropyl-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl } -6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] oxy } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AGA-1)
To 6- (chloromethyl) -2- { 6-cyclopropyl-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl at room temperature]To a stirred solution of pyridin-2-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (AFV-2) (40 mg,1 eq, 76 μmol) and (1 r,2 s) -cyclopentane-1, 2-diol (AV-2) (77 mg,10 eq, 0.76 mmol) in DCM (3 mL) was added TBAB (12 mg,0.5 eq, 38 μmol), aqueous KOH (3 mL,20 wt%). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 5% B to 95% B in 8 minutes; wavelength: 254/220nm; retention time: 5.75 To give the title compound (AGA-1) (13.1 mg,22 μmol,29%,98% purity) as a white solid. M/z 594.3 (M+H) + (ES+). 1 H NMR(400MHz,CD3OD)δ8.49(s,1H),8.22(d,J=1.3Hz,1H),8.10(s,1H),8.03(s,1H),7.90(s,1H),7.10(d,J=1.3Hz,1H),5.22(s,2H),4.84–4.72(m,2H),4.19–4.10(m,1H),4.02(s,3H),3.86–3.82(m,1H),3.56(s,3H),2.16–2.05(m,1H),1.90–1.79(m,4H),1.73–1.70(m,1H),1.59–1.55(m,1H),1.10–1.01(m,4H)。
Example 205: synthesis of 2- [6- (ethylamino) -4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -6- { [ (1-fluorocyclobutyl) methoxy ] methyl } -4- (trifluoromethyl) -3H-isoindol-1-one (AGB-1)
To a stirred solution of intermediate (AFF-1) (40 mg,1 eq, 75. Mu. Mol) and (1-fluorocyclobutyl) MeOH (39 mg,5 eq, 0.38 mmol) in DCM (2 mL) was added TBAB (12 mg,0.5 eq, 37. Mu. Mol) and aqueous KOH (2 mL,20 Wt%) at room temperature. The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with DCM (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (20/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 49% B to 69% B in 10 minutes; wavelength: 254/220nm; retention time: 9.22 To give the title compound (AGB-1) (9.0 mg,15 μmol,19%,99% purity) as a white solid. M/z 614.2 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO–d6)δ8.49(s,1H),7.99(d,J=16.7Hz,2H),7.67–7.42(m,4H),6.72(t,J=5.4Hz,1H),5.92(d,J=1.3Hz,1H),5.21–5.11(m,2H),4.77(s,2H),3.73(s,1H),3.67(s,1H),3.38(s,3H),3.21–3.12(m,2H),2.29–2.11(m,4H),1.82–1.69(m,1H),1.57–1.43(m,1H),1.13(t,J=7.2Hz,3H)。
Example 206: synthesis of 4- [2- (ethylamino) -6- (1-oxo-6- { [ (2-oxocyclopentyl) amino ] methyl } -4- (trifluoromethyl) -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGC-5)
Step 1: synthesis of 6- ({ [ (1R, 2R) -2-hydroxycyclopentyl ] amino } methyl) -4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (AGC-1)
A solution of intermediate (ABC-4) (200 mg,1 eq, 0.87 mmol) and (1R, 2R) -2-aminocyclopentan-1-ol, HCl (W-1) (132 mg,1.5 eq, 1.31 mmol) in MeOH (20 mL) was stirred at 60℃for 2 hours. Adding NaBH to the mixture at 0deg.C 4 (132 mg,4 eq, 3.49 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by addition of 5mL of ice water at 0 ℃. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/1) to give the sub-title compound (AGC-1) (210 mg,0.60mmol,69% purity) as a white solid. M/z 315.1 (M+H) + (ES+)。
Step 2: synthesis of tert-butyl N- [ (1R, 2R) -2-hydroxycyclopentyl ] -N- { [ 3-oxo-7- (trifluoromethyl) -1, 2-dihydro-isoindol-5-yl ] methyl } carbamate (AGC-2)
To the product from step 1 above (AGC-1) (200 mg,1 eq., 0.64 mmol) and (Boc) at room temperature 2 O (153 mg,1.1 eq, 0.70 mmol) in THF (15 mL) was added with NaHCO 3 (160 mg,3 equivalents, 1.91 mmol). The resulting mixture was stirred at 60℃for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/5) to give the sub-title compound (AGC-2) (100 mg,0.21mmol,34%,88% purity) as a yellow oil. M/z 415.2 (M+H) + (ES+)。
Step 3: synthesis of tert-butyl N- { [ 3-oxo-7- (trifluoromethyl) -1, 2-dihydro-isoindol-5-yl ] methyl } -N- (2-oxocyclopentyl) carbamate (AGC-3)
A solution of the product from step 2 above (AGC-2) (100 mg,1 eq, 0.24 mmol) and PCC (156 mg,3 eq, 0.72 mmol) in DCM (15 mL) was stirred at room temperature overnight. Filtering the resulting mixture; the filter cake was washed with DCM (3X 5 mL). The filtrate was concentrated in vacuo. This gave the sub-title compound (AGC-3) (70 mg,0.15mmol,63%,90% purity) as a brown yellow oil. M/z 413.2 (M+H) + (ES+)。
Step 4: synthesis of tert-butyl N- [ (2- {4- [ 4-cyano-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6- (ethylamino) pyridin-2-yl } -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl) methyl ] -N- (2-oxocyclopentyl) carbamate (AGC-4)
To intermediate (ADP-7) (40 mg,1 equivalent, 0.12 mmol), the product from step 3 above (AGC-3) (54 mg,1.1 equivalent, 0.13 mmol) and Cs under nitrogen at room temperature 2 CO 3 (18 mg,2 eq, 0.24 mmol) to a stirred solution of RuPhos (22 mg,0.4 eq, 47. Mu. Mol) and RuPhos ring palladium complex Gen.3 (20 mg,0.2 eq, 24. Mu. Mol) in 1, 4-dioxane (10 mL) were added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. Filtering the resulting mixture; the filter cake was washed with MeOH (2X 2 mL). The filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AGC-4) (35 mg, 47. Mu. Mol,37%,96% purity) as a yellow solid. M/z 715.2 (M+H) + (ES+)。
Step 5: synthesis of 4- [2- (ethylamino) -6- (1-oxo-6- { [ (2-oxocyclopentyl) amino ] methyl } -4- (trifluoromethyl) -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGC-5)
To a 25mL round bottom flask was added 1, 4-dioxane (5 mL) containing the product from step 4 above (AGC-4) (30 mg,1 eq, 42. Mu. Mol) and HCl (gas) at room temperature. The resulting mixture was stirred at room temperature for 2 hours. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 55% B in 8 minutes; 55% B; wavelength: 254/220nm; retention time: 7.2 To give the title compound (AGC-5) (2.4 mg,3.8 μmol,9.0%,96% purity) as a white solid. M/z 615.2 (M+H) + (ES+). 1 H NMR(300MHz,CHCl3-d)δ8.43(d,J=4.1Hz,1H),8.10–7.97(m,4H),7.83(d,J=8.1Hz,1H),7.48–7.39(m,1H),6.09–6.00(m,1H),5.23–5.09(m,2H),4.44–4.11(m,2H),3.50(t,J=9.8Hz,1H),3.41(d,J=3.0Hz,3H),3.22(d,J=7.2Hz,2H),2.49–2.37(m,2H),2.30–1.96(m,2H),1.89–1.55(m,2H),1.17(t,J=7.1Hz,3H)。
Example 207: synthesis of 4- [2- (6- { [ (2-cyclopropoxyethyl) amino ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) -6- (ethylamino) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGD-2)
To a stirred solution of intermediate (AEV-1) (50 mg,1 eq, 91. Mu. Mol) and 2-cyclopropyloxyethyl-1-amine, HCl (AGD-1) (92 mg,10 eq, 0.91 mmol) in DCM (5 mL) was added TBAB (12 mg,0.5 eq, 36. Mu. Mol) and aqueous KOH (5 mL,20 Wt%) at room temperature. The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was extracted with EtOAc (3×10 mL). The combined organic layers were washed with water (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC under the following conditions: (column: XBridge preparation type OBD C18 column, 30X 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 44% B to 54% B in 9 minutes; wavelength: 254/220nm; retention time: 8.9 To give the title compound (AGD-2) (6.5 mg,10 μmol,12%,99% purity) as an off-white solid. M/z 617.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.47(s,1H),8.17–8.06(m,1H),8.05–8.00(m,2H),7.96(s,1H),7.87(d,J=8.0Hz,1H),7.50(d,J=1.2Hz,1H),6.04(d,J=1.2Hz,1H),5.17(s,2H),3.95(s,2H),3.64(t,J=5.4Hz,2H),3.45(s,3H),3.34(s,1H),3.26(t,J=7.2Hz,2H),2.75(t,J=5.3Hz,2H),1.20(t,J=7.2Hz,3H),0.58–0.48(m,2H),0.47–0.42(m,2H)。
Example 208: synthesis of 2- [6- (ethylamino) -4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -6- [ (2-hydroxyethoxy) methyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AGE-3)
Step 1: synthesis of 6- ({ 2- [ (tert-butyldimethylsilyl) oxy ] ethoxy } methyl) -2- [6- (ethylamino) -4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AGE-2)
To intermediate (AFF-1) (40 mg,1 eq, 73. Mu. Mol) and 2- [ (tert-butyldimethylsilyl) oxy at room temperature]To a stirred solution of ethanol (AGE-1) (129 mg,10 eq, 0.73 mmol) in DCM (5 mL) was added TBAB (12 mg,0.5 eq, 36. Mu. Mol) and aqueous KOH (5 mL,20 wt%). The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was extracted with EtOAc (3×10 mL). The combined organic layers were washed with water (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AGE-2) as a yellow oil (26 mg, 36. Mu. Mol,47%,95% purity). M/z 685.3 (M+H) + (ES+)。
Step 2: synthesis of 2- [6- (ethylamino) -4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -6- [ (2-hydroxyethoxy) methyl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AGE-3)
THF (5 mL) containing the product from step 1 above (AGE-2) (30 mg,1 eq, 44. Mu. Mol) and TEA.3HF (0.5 mL) was added to a 25mL round bottom flask at room temperature. The resulting mixture was stirred at room temperature overnight. The mixture was concentrated in vacuo. Will beThe crude product was purified by preparative HPLC under the following conditions (column: xselect CSH F-phenyl OBD column, 19X 250mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meOH-HPLC; flow rate: 25 ml/min; gradient: 65% B to 70% B in 9 minutes; wavelength: 254/220nm; retention time: 8.67 To give the title compound (AGE-3) (10.5 mg,18 μmol,42%,98% purity) as a white solid. M/z 571.1 (M+H) + (ES+). 1 H NMR(300MHz,DMSO-d6)δ8.49(s,1H),8.00(d,J=11.1Hz,2H),7.70–7.45(m,3H),7.42(d,J=1.2Hz,1H),5.94(d,J=1.3Hz,1H),5.17(s,2H),4.70(s,2H),3.60–3.51(m,4H),3.39(s,3H),3.28–3.01(m,2H),1.17–1.08(m,3H)。
Example 209: synthesis of 6- { [ (1-aminocyclobutyl) methoxy ] methyl } -2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AGF-3)
Step 1: synthesis of tert-butyl N- {1- [ ({ 2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl } methoxy) methyl ] cyclobutyl } carbamate (AGF-2)
To intermediate (ADA-1) (30 mg,1 eq, 57. Mu. Mol) and N- [1- (hydroxymethyl) cyclobutyl at RT]To a stirred solution of tert-butyl carbamate (AGF-1) (114 mg,10 eq, 0.57 mmol) in DCM (3 mL) was added TBAB (9 mg,0.5 eq, 29. Mu. Mol) and aqueous KOH (3 mL,20 Wt%). The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AGF-2) (25 mg, 34. Mu. Mol,64%,95% purity) as a yellow solid. M/z 696.2 (M+H) + (ES+)。
Step 2: synthesis of 6- { [ (1-aminocyclobutyl) methoxy ] methyl } -2- [6- (ethylamino) -4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AGF-3)
To a stirred solution of the product from step 1 above (AGF-2) (25 mg,1 eq, 36 μmol) in DCM (4 mL) was added HCl (gas) -containing 1, 4-dioxane (2 mL,4 m) at room temperature. The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 40% B in 10 minutes; wavelength: 254/220nm; retention time: 9.09 To give the title compound (AGF-3) (12.6 mg,20 μmol,59%,96% purity) as a white solid. M/z 596.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.47(s,1H),8.02(d,J=16.3Hz,2H),7.88(s,1H),7.57(d,J=1.1Hz,1H),6.93(t,J=5.4Hz,1H),6.25(d,J=1.2Hz,1H),5.21(s,2H),4.74(s,2H),3.89(s,3H),3.46(s,3H),3.40(s,2H),3.29–3.05(m,2H),2.00–1.94(m,2H),1.81–1.75(m,2H),1.73–1.63(m,1H),1.62–1.52(m,1H),1.17(t,J=7.2Hz,3H)。
Example 210: synthesis of 6- { [ (1-aminocyclobutyl) methoxy ] methyl } -2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (AGG-2)
Step 1: synthesis of tert-butyl N- (1- { [ (2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl) methoxy ] methyl } cyclobutyl) carbamate (AGG-1)
To intermediate (AFL-1) (50 mg,1 eq, 92. Mu. Mol) and N- [1- (hydroxymethyl) cyclobutyl at RT]To a stirred solution of tert-butyl carbamate (AGF-1) (279 mg,15 eq, 1.38 mmol) in DCM (8 mL) was added TBAB (15 mg,0.5 eq, 46. Mu. Mol). To the above mixture was added aqueous KOH solution (8 ml,20 wt%) at room temperature. The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water. The resulting mixture was extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC (DCM/MeOH 12/1) to give the sub-title compound (AGG-1) (35 mg, 45. Mu. Mol,54%,90% pure). M/z 707.1 (M+H) + (ES+)。
Step 2: synthesis of 6- { [ (1-aminocyclobutyl) methoxy ] methyl } -2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (AGG-2)
To a stirred solution of the product from step 1 above (AGG-1) (35 mg,1 eq, 50 μmol) in DCM (2 mL) was added dropwise 1, 4-dioxane (1 mL,4 m) containing HCl (gas) at 0 ℃. The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was diluted with water. The mixture was neutralized to pH 7 with saturated ammonium bicarbonate. The resulting mixture was extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 44% B to 54% B in 8 minutes; wavelength: 254/220nm; retention time: 7.93 To give the title compound (AGG-2) (3.2 mg,5.2 μmol,11%,99% purity) as a white solid. M/z 607.1 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.46(s,1H),8.11–7.96(m,3H),7.79–7.73(m,1H),7.58–7.43(m,2H),6.90(d,J=1.4Hz,1H),5.18(s,2H),4.78(s,2H),3.60(d,J=2.0Hz,2H),3.46(s,3H),2.14–1.99(m,5H),1.92–1.70(m,2H),1.00(d,J=6.6Hz,4H)。
Example 211: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- (hydroxymethyl) -3H-isoindol-1-one (AGH-2)
Step 1: synthesis of 6- (hydroxymethyl) -2, 3-dihydro-isoindol-1-one (AGH-1)
To a stirred mixture of intermediate (ACY-2) (150 mg,1 eq, 0.93 mmol) in THF (20 mL) at 0deg.C was added NaBH 4 (70 mg,2 equivalents, 1.86 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was quenched with MeOH (2 mL) at 0deg.C. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 50% in 12 minutes); detector, UV 254/220nm, to give the sub-title compound (AGH-1) (95 mg,0.54mmol,63%,92% purity) as a white solid. M/z 164.1 (M+H) + (ES+)。
Step 2: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- (hydroxymethyl) (AGH-2)
To the product (AGH-1) (89 mg,1.5 eq., 0.55 mmol), intermediate (ADN-1) (120 mg,1 eq., 0.37 mmol) and Cs from step 1 above, under nitrogen at room temperature 2 CO 3 To a stirred mixture of (317 mg,1.10mmol,3 eq.) in 1, 4-dioxane (2 mL) was added RuPhos ring palladium complex gen.3 (57 mg,0.2 eq., 73 μmol) and RuPhos (68 mg,0.4 eq., 0.15 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 33% B to 47% B in 9 minutes; wavelength:254/220 nm) to give the title compound (AGH-2) (7 mg, 15. Mu. Mol,4.1%,96% purity) as a white solid. M/z 456.1 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO–d6)δ8.51(s,1H),7.99(d,J=1.3Hz,1H),7.82–7.46(m,6H),6.79(d,J=1.3Hz,1H),5.37(t,J=5.8Hz,1H),5.01(s,2H),4.61(d,J=5.7Hz,2H),3.43(s,3H),2.01(m,1H),0.95(d,J=6.4Hz,4H)。
Example 212: synthesis of 6- { [ (1-aminocyclobutyl) methoxy ] methyl } -2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -3H-isoindol-1-one (AGI-3)
Step 1: synthesis of 6- (chloromethyl) -2- (6-cyclopropyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) isoindolin-1-one (AGI-1)
To a stirred mixture of intermediate (AGH-2) (115 mg,1 eq, 0.25 mmol) in DCM (10 mL) at 0deg.C was added SOCl 2 (0.15 mL). The resulting mixture was stirred at room temperature for 0.5 hours. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AGI-1) (80 mg,0.16mmol,53%,92% pure) as a white solid. M/z 474.1/476.1 (M+H) + (ES+)。
Step 2: synthesis of tert-butyl (1- (((2- (6-cyclopropyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -3-oxoisoindolin-5-yl) methoxy) methyl) cyclobutyl) carbamate (AGI-2)
To the product (AGI-1) (30 mg,1 equivalent, 32. Mu. Mol) from step 1 above and N- [1- (hydroxymethyl) cyclobutyl at room temperature]To a stirred mixture of tert-butyl carbamate (AGF-1) (127 mg,10 eq, 0.32 mmol) in DCM (5 mL) was added TBAB (10 mg,0.5 eq, 16. Mu. Mol). Aqueous KOH solution (5 mL,20 wt%) was added to the above mixture over 1 minute at room temperature. The resulting mixture was stirred at room temperature for an additional 0.5 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2x10 mL), and drying (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AGI-2) (30 mg, 43. Mu. Mol,74%,92% purity) as a white solid. M/z 639.1 (M+H) + (ES+)。
Step 3: synthesis of 6- (((1-aminocyclobutyl) methoxy) methyl) -2- (6-cyclopropyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) isoindolin-1-one (AGI-3)
To a stirred solution of the product from step 2 above (AGI-2) (30 mg,1 eq, 47 μmol) in DCM (3 mL) at 0 ℃ was added HCl (gas) -containing 1, 4-dioxane (1 mL,4 m). The resulting mixture was stirred at room temperature for 2 hours. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge Shield RP OBD column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 26% B to 42% B in 10 minutes; wavelength: 254/220 nm) to give the title compound (AGI-3) (3.1 mg, 5.6. Mu. Mol,12%,98% purity) as a white solid. M/z 539.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.51(s,1H),7.98(d,J=1.3Hz,1H),7.79–7.50(m,6H),6.80(d,J=1.4Hz,1H),5.03(s,2H),4.65(s,2H),3.43(s,3H),3.36–3.35(m,2H),2.05–1.92(m,3H),1.83–1.63(m,3H),1.61–1.51(m,1H),0.96(d,J=6.4Hz,4H)。
Example 213: synthesis of 4- [2- (6- { [ (1-aminocyclobutyl) methoxy ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) -6- (ethylamino) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGJ-2)
Step 1: synthesis of tert-butyl N- (1- { [ (2- {4- [ 4-cyano-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6- (ethylamino) pyridin-2-yl } -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl) methoxy ] methyl } cyclobutyl) carbamate (AGJ-1)
To intermediate (AEV-1) (60 mg,1 eq, 0.11 mmol) and N- [1- (hydroxymethyl) cyclobutyl at RT ]To a stirred mixture of tert-butyl carbamate (AGF-1) (219 mg,10 equivalents, 1.09 mmol) in DCM (10 mL) was added TBAB (18 mg,0.5 equivalents, 54. Mu. Mol). Aqueous KOH solution (8 ml,20 wt%) was added to the above mixture at room temperature over 2 minutes. The resulting mixture was stirred at room temperature for an additional 0.5 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AGJ-1) (40 mg,52 μmol,51%,94% purity) as a white solid. M/z 717.2 (M+H) + (ES+)。
Step 2: synthesis of 4- [2- (6- { [ (1-aminocyclobutyl) methoxy ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) -6- (ethylamino) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGJ-2)
To a stirred mixture of the product (AGJ-1) from step 1 above (50 mg,1 eq, 70 μmol) in DCM (6 mL) was added HCl (gas) -containing 1, 4-dioxane (2 mL,4 m) at room temperature. The resulting mixture was stirred at room temperature for 0.5 hours. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 70% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (AGJ-2) (3.4 mg, 5.3. Mu. Mol,7.7%,97% purity) as a yellow solid. M/z 617.2 (M+H) + (ES+). 1 H NMR(300MHz,DMSO–d6)δ8.52(s,1H),8.22–8.12(m,2H),8.01(d,J=11.3Hz,2H),7.79(d,J=8.0Hz,1H),7.45(d,J=1.2Hz,1H),6.82(t,J=5.5Hz,1H),5.97(d,J=1.3Hz,1H),5.17(s,2H),4.75(s,2H),3.43–3.39(m,5H),3.25–3.14(m,2H),2.05–1.92(m,2H),1.80(m,2H),1.72–1.53(m,2H),1.13(t,J=7.1Hz,3H)。
Example 214: synthesis of 4- (2- {6- [ (1R) -2- (dimethylamino) -1-hydroxyethyl ] -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl } -6- (ethylamino) pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGK-1). Stereochemistry was arbitrarily specified.
Compound (AFR-7) (50 mg,1 eq, 85 μmol) was purified by chiral HPLC under the following conditions (column: CHIRALPAK IG,2×25cm,5 μm; mobile phase a: hex: dcm=3:1 (0.5% 2m NH) 3 MeOH) -HPLC, mobile phase B: etOH-HPLC; flow rate: 20 ml/min; gradient: 30% B to 30% B within 20 minutes; wavelength: 220/254nm; retention time 1 (min): 11.51; retention time 2 (minutes): 17.34 To give the title compound (AGK-1) (14.9 mg,24 μmol,30%,97% purity) as a yellow solid. M/z 591.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.48(s,1H),8.11–8.07(m,2H),8.03(d,J=1.7Hz,1H),7.99(s,1H),7.87(d,J=8.1Hz,1H),7.51–7.46(m,1H),6.08–6.03(m,1H),5.19(s,2H),5.08–5.00(m,1H),3.46(s,3H),3.27(t,J=7.2Hz,2H),2.77–2.68(m,1H),2.62–2.54(m,1H),2.44(s,6H),1.20(t,J=7.2Hz,3H)。
Column: CHIRALPAK IG-3,4.6 x 50mm,3um; mobile phase a: (Hex: dcm=3:1) (0.1% DEA): etoh=70:30; flow rate: 1 ml/min; retention time: 2.745.
Example 215: synthesis of 4- (2- {6- [ (1S) -2- (dimethylamino) -1-hydroxyethyl ] -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl } -6- (ethylamino) pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGL-1). Stereochemistry was arbitrarily specified.
Compound (AFR-7) (50 mg,1 eq, 85 μmol) was purified by chiral HPLC under the following conditions (column: CHIRALPAK IG,2×25cm,5 μm; mobile phase a: hex: dcm=3:1 (0.5% 2m NH) 3 -MeOH) -HPLC, mobile phase B: etOH-HPLC; flow rate: 20 ml/min; gradient: 30% B to 30% B within 20 minutes; wavelength: 220/254nm; retention time 1 (min): 11.51; retention time 2 (minutes): 17.34 To give the title compound (AGL-1) (4.1 mg,6.7 μmol,8.2%,97% purity) as a yellow solid. M/z 591.2 (M+H) + (ES+)。 1 H NMR (400 MHz, meOH-d 4) delta 8.48 (s, 1H), 8.15-7.98 (m, 4H), 7.87 (d, J=8.1 Hz, 1H), 7.53-7.46 (m, 1H), 6.11-6.00 (m, 1H), 5.19 (s, 2H), 5.09-5.00 (m, 1H), 3.46 (s, 3H), 3.29-3.23 (m, 2H), 2.82-2.69 (m, 1H), 2.69-2.59 (m, 1H), 2.47 (s, 6H), 1.21 (t, J=7.2 Hz, 3H). Column: CHIRALPAK IG-3,4.6 x 50mm,3 μm; mobile phase a: (Hex: dcm=3:1) (0.1% DEA): etoh=70:30; flow rate: 1 ml/min; retention time: 4.004.
Example 216: synthesis of 4- { 2-cyclopropyl-6- [6- (1-hydroxycyclobutyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGM-5)
Step 1: synthesis of methyl 5- (1-hydroxycyclobutyl) -2-methyl-3- (trifluoromethyl) benzoate (AGM-2)
To a 40-mL vial purged and maintained with an inert nitrogen atmosphere was placed THF (10 mL) containing methyl 5-bromo-2-methyl-3- (trifluoromethyl) benzoate (B-1) (400 mg,1 eq, 1.35 mmol) followed by i-PrMgCl (554 mg,4 eq, 5.38 mmol) at 0deg.C. The resulting mixture was stirred under nitrogen at 0℃for 2 hours, then cyclobutanone (AGM-1) (566 mg,6 eq., 8.08 mmol) was added at 0 ℃. The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. The resulting mixture was diluted with water and extracted with DCM (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (10% MeCN up to 50% in 15 minutes); detection ofUV 254/220nm to give the sub-title compound (AGM-2) (120 mg,0.37mmol,31%,90% purity) as a white solid. M/z 289.1 (M+H) + (ES+)。
Step 2: synthesis of methyl 2- (bromomethyl) -5- (1-hydroxycyclobutyl) -3- (trifluoromethyl) benzoate (AGM-3)
A250-mL round bottom flask maintained under an inert nitrogen atmosphere was charged with CCl containing the product (AGM-2) from step 1 above (270 mg,1 eq, 0.94 mmol) at room temperature 4 (15 mL) then NBS (250 mg,1.5 eq, 1.41 mmol) and BPO (72 mg,0.3 eq, 0.28 mmol) were added at 0deg.C. The resulting mixture was stirred at 80 ℃ overnight. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product mixture was used directly in the next step without further purification. M/z 267.0/269.0 (M+H) + (ES+)。
Step 3: synthesis of 6- (1-hydroxycyclobutyl) -4- (trifluoromethyl) isoindolin-1-one (AGM-4).
NH containing the product (AGM-3) from step 2 above (150 mg,1 equivalent, 0.41 mmol) was placed in a 40-mL vial purged and maintained with an inert nitrogen atmosphere 3 MeOH solution (10 mL, 25%). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 3 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with DCM (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 70% in 15 minutes); detector, UV 254/220nm. This gave the sub-title compound (AGM-4) (100 mg,0.34mmol,90%,92% purity) as a white solid. M/z 272.1 (M+H) + (ES+)。
Step 4: synthesis of 4- (2-cyclopropyl-6- (6- (1-hydroxycyclobutyl) -1-oxo-4- (trifluoromethyl) isoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AGM-5)
Under nitrogen atmosphere at room temperature, the reaction was conductedA20-mL vial purged and maintained under an inert nitrogen atmosphere was placed in the vial containing the product from step 3 above (AGM-4) (30 mg,1 equivalent, 0.11 mmol), intermediate (AEG-2) (37 mg,1 equivalent, 0.11 mmol) and Cs 2 CO 3 (72 mg,2 eq, 0.22 mmol) of 1, 4-dioxane (6 mL) followed by addition of RuPhos Ring Palladium Complex Gen.3 (19 mg,0.2 eq, 22. Mu. Mol) and RuPhos (21 mg,0.4 eq, 44. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The aqueous layer was extracted with DCM (3X 30 mL). The combined organic layers were washed with brine (2×30 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 48% B to 58% B in 9 minutes; wavelength: 571nm; retention time: 8.63 To give the title compound (AGM-5) (13.2 mg, 23. Mu. Mol,21%,99% purity) as a white solid.
m/z 571.3(M+H) + (ES+). 1 H NMR(300MHz,DMSO-d6)δ8.55(s,1H),8.22(h,J=1.8Hz,2H),8.08(d,J=12.4Hz,2H),7.98(d,J=1.3Hz,1H),7.88(d,J=8.5Hz,1H),6.91(d,J=1.4Hz,1H),5.98(s,1H),5.17(s,2H),3.48(s,3H),2.49–2.42(m,2H),2.41–2.26(m,2H),2.14–1.95(m,2H),1.83–1.67(m,1H),1.03–0.87(m,4H)。
Example 217: synthesis of 4- { 2-cyclopropyl-6- [ 1-oxo-6- ({ [2- (trifluoromethoxy) ethyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGN-3)
Step 1: synthesis of 4- { 2-cyclopropyl-6- [ 6-formyl-1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGN-1)
Intermediate (AES-1) (60 mg,1 eq, 0.11 mmol) and dess-Martin peroxideA solution of iodoalkane (72 mg,1.5 eq, 0.17 mmol) in DCM (15 mL) was stirred at room temperature for 1 hour. Filtering the resulting mixture; the filter cake was washed with DCM (3X 3 mL). The filtrate was concentrated in vacuo to give the sub-title compound (AGN-1) (40 mg,72 μmol,60%,95% purity) as a yellow solid. M/z 529.2 (M+H) + (ES+)。
Step 2: synthesis of 4- { 2-cyclopropyl-6- [ 1-oxo-6- ({ [2- (trifluoromethoxy) ethyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGN-3)
To a stirred solution of the product from step 1 above (AGN-1) (20 mg,1 eq, 36. Mu. Mol) and 2- (trifluoromethoxy) ethan-1-amine, HCl (AGN-2) (9.4 mg,2 eq, 72. Mu. Mol) in MeOH (15 mL) was added DIPEA (19 mg,4 eq, 0.14 mmol) at room temperature. The resulting mixture was stirred at room temperature for 2 hours. Adding NaBH to the mixture at 0deg.C 4 (7 mg,5 equivalents, 0.18 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by addition of 2mL of ice water at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 8% B to 32% B over 7 min; wavelength: 254/220nm; retention time: 6.12) to give the title compound (AGN-3) (1.7 mg,2.6 μmol,7.2%,98 purity) as an off-white solid. M/z 642.1 (M+H) + (ES+)。
6.95(d,J=1.5Hz,1H),5.17(s,2H),4.15(t,J=5.4Hz,2H),4.01(s,2H),3.49(s,3H),2.93(t,J=5.4Hz,2H),2.12–1.98(m,1H),1.04–0.97(m,4H)。
Example 218: synthesis of 4- (2-cyclopropyl-6- (6- (((2-methoxyethyl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AGO-2)
Step 1: synthesis of 6- (((2-methoxyethyl) amino) methyl) isoindolin-1-one (AGO-1)
To a stirred solution of intermediate (ACY-2) (60 mg,1 eq, 0.37 mmol) in DCM (8 mL) was added DIPEA (192 mg,4 eq, 1.49 mmol) at room temperature. The resulting mixture was stirred at 60℃for 10 minutes. 2-methoxyethyl-1-amine (39 mg,1.4 eq, 0.52 mmol) was then added at room temperature. The resulting mixture was stirred at 60℃for 2 hours. Finally, naBH is added at 0 DEG C 4 (399mg, 5 equivalents, 1.86 mmol). The resulting mixture was stirred at room temperature for 1 hour. The mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 20% in 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (AGO-1) (45 mg,0.19mmol,55%,95% purity) as a yellow crude solid. M/z 221.1 (M+H) + (ES+)。
Step 2: synthesis of 4- (2-cyclopropyl-6- (6- (((2-methoxyethyl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AGO-2)
To intermediate (AEG-2) (60 mg,0.18mmol,1 eq.) the product from step 1 above (AGO-1) (43 mg,1.1 eq., 0.20 mmol) and Cs under nitrogen at room temperature 2 CO 3 (175 mg,3 eq, 0.54 mmol) to a stirred solution of RuPhos (33 mg,0.4 eq, 72. Mu. Mol) and RuPhos ring palladium complex Gen.3 (30 mg,0.2 eq, 36. Mu. Mol) in 1, 4-dioxane (8 mL) were added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1). The crude product was purified by preparative HPLC under the following conditions (column: XBridge prep. OBD C18 column, 19 x 250mm,5 μm; mobile phase a: water (0.1% nh) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 25 ml/min; gradient: 25% B to 70% B in 8 minutes; wavelength: 254/220nm; retention time: 7.67 To give the title compound (AGO-2) (34.1 mg, 64. Mu. Mol,36%, 98%) as a white solidPurity). M/z 520.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.54(s,1H),8.25–8.18(m,2H),7.98(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.74(s,1H),7.63(d,J=1.2Hz,2H),6.89(d,J=1.4Hz,1H),5.00(s,2H),3.82(s,2H),3.48(s,3H),3.40(t,J=5.7Hz,2H),3.24(s,3H),2.64(t,J=5.7Hz,2H),2.08–2.02(m,1H),1.00–0.93(m,4H)。
Example 219: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclopentyl) methyl ] amino } methyl) -3H-isoindol-1-one (AGP-1)
To intermediate (ADN-1) (70 mg,0.21mmol,1 eq.), intermediate (AFQ-2) (67 mg,1.2 eq., 0.26 mmol) and Cs under nitrogen at room temperature 2 CO 3 (208 mg,0.64mmol,3 eq.) to a stirred solution in 1, 4-dioxane was added RuPhos (10 mg,0.1 eq., 21. Mu. Mol) and RuPhos ring palladium complex Gen.3 (18 mg,0.1 eq., 21. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 50% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (AGP-1) (51.9 mg, 93. Mu. Mol,44%,99% purity) as a white solid. M/z 553.4 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO–d6)δ8.50(s,1H),7.94(d,J=1.4Hz,1H),7.80–7.70(m,2H),7.67–7.51(m,4H),6.81(d,J=1.4Hz,1H),5.00(s,2H),3.83(s,2H),3.43(s,3H),2.50(s,2H),2.03(m,1H),1.70–1.45(m,8H),0.96(m,4H)。
Example 220: synthesis of 2- { 6-ethoxy-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AGQ-2)
Step 1: synthesis of 2-chloro-6-ethoxy-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridine (AGQ-1)
To a 20mL sealed tube was added EtOH (10 mL) containing intermediate (ACM-6) (300 mg,1 eq, 0.93 mmol) at room temperature. To the above mixture was added EtONa (94.76 mg,1.5 eq, 1.39 mmol) over 1 minute at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 70% in 15 minutes); detector, UV 254/220nm, to give the sub-title compound (AGQ-1) (135 mg,0.39mmol,44%,95% purity) as a yellow solid. M/z 333.1/335.1 (M+H) + (ES+)。
Step 2: synthesis of 2- { 6-ethoxy-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AGQ-2)
To the product (AGQ-1) (70 mg,1 equivalent, 0.21 mmol), intermediate (ADR-2) (79 mg,1.2 equivalent, 0.25 mmol) and Cs from step 1 above, at room temperature under nitrogen atmosphere 2 CO 3 (137 mg,2 eq, 0.42 mmol) to a stirred solution of RuPhos (39 mg,0.4 eq, 84. Mu. Mol) and RuPhos ring palladium complex Gen.3 (35 mg,0.2 eq, 42. Mu. Mol) in 1, 4-dioxane (10 mL) were added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 1 hour. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) and by preparative HPLC under the following conditions (column: XBridge prep OBD C18 column, 30 x 150mm,5 μm; mobile phase a: Water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 70% B in 8 minutes; wavelength: 254/220nm; retention time: 6.62 To give the title compound (AGQ-2) (37.5 mg,60 μmol,29%,99% purity) as a white solid. M/z 611.4 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.51(s,1H),8.04(d,J=10.2Hz,2H),7.81(d,J=1.2Hz,1H),7.74–7.68(m,1H),7.63–7.55(m,2H),6.33(d,J=1.2Hz,1H),5.19(s,2H),4.94(s,1H),4.34–4.26(m,2H),3.95(s,2H),3.43(s,3H),2.53(s,2H),2.05–1.94(m,2H),1.94–1.84(m,2H),1.66–1.57(m,1H),1.44–1.32(m,4H)。
Example 221: synthesis of 2- { 6-ethoxy-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AGR-1)
To intermediate (AGQ-1) (60 mg,1 equivalent, 0.18 mmol), intermediate (AEE-1) (49 mg,1.1 equivalent, 0.20 mmol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 (118 mg,2 eq, 0.36 mmol) to a stirred mixture of 1, 4-dioxane (8 mL) was added RuPhos (34 mg,0.4 eq, 72. Mu. Mol) and RuPhos ring palladium complex Gen.3 (30 mg,0.2 eq, 36. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 1 hour. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 27% B to 42% B in 10 minutes; wavelength: 254/220nm; retention time: 9.2 To give the title compound (AGR-1) (25.2 mg,0.45mmol,25%,96% purity) as a white solid. M/z 543.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.51(s,1H),7.83–7.55(m,7H),6.28(d,J=1.2Hz,1H),5.05(s,2H),4.91(s,1H),4.33(q,J=7.0Hz,2H),3.86(s,2H),3.44(s,3H),2.53(s,2H),2.00(t,J=8.8,3.1Hz,2H),1.89(d,J=12.2,9.5Hz,2H),1.64–1.54(m,1H),1.47–1.30(m,4H)。
Example 222: synthesis of 6- { [ (1-aminocyclobutyl) methoxy ] methyl } -2- {4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6- (methylamino) pyridin-2-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (AGS-2)
Step 1: synthesis of tert-butyl N- {1- [ ({ 2- [6- (ethylamino) -4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -3-oxo-1- (trifluoromethyl) -1H-isoindol-5-yl } methoxy) methyl ] cyclobutyl } carbamate (AGS-1)
To intermediate (AFF-1) (40 mg,1 eq, 73. Mu. Mol) and N- [1- (hydroxymethyl) cyclobutyl]To a stirred mixture of tert-butyl carbamate (AGF-1) (148 mg,10 equivalents, 0.73 mmol) in DCM (2 mL) was added aqueous KOH (2 mL,20 wt%). TBAB (12 mg,0.5 eq, 36. Mu. Mol) was added to the above mixture over 2 minutes at room temperature. The resulting mixture was stirred at room temperature for 30 minutes. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AGS-1) (20 mg, 27. Mu. Mol,38%,96% purity) as a white solid. M/z 710.4 (M+H) + (ES+)。
Step 2: synthesis of 6- { [ (1-aminocyclobutyl) methoxy ] methyl } -2- {4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6- (methylamino) pyridin-2-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (AGS-2)
To a stirred mixture of the product from step 1 above (AGS-1) (18 mg,1 eq, 25 μmol) in DCM (5 mL) was added HCl (gas) -containing 1, 4-dioxane (1 mL,4 m) at room temperature. The resulting mixture was stirred at room temperature for 0.5 hours. The mixture is put inConcentrating in vacuum. The crude product was purified by preparative HPLC on a column (XBridge Shield RP, 18 OBD column, 19 x 250mm,10 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 25 ml/min; gradient: 40% B to 70% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AGS-2) (3.3 mg, 5.1. Mu. Mol,21%,95% purity) as a white solid. M/z 610.4 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO–d6)δ8.48(s,1H),8.02(d,J=18.6Hz,2H),7.70–7.47(m,3H),7.45(s,1H),6.71(t,J=5.4Hz,1H),5.92(s,1H),5.17(s,2H),4.75(s,2H),3.47(s,3H),3.17(m,4H),2.05–1.84(m,4H),1.77–1.62(m,2H),1.13(t,J=7.1Hz,3H)。
Example 223: synthesis of 4- { 2-cyclopropyl-6- [ 1-oxo-6- ({ [2- (trifluoromethoxy) ethyl ] amino } methyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGT-3)
Step 1: synthesis of 4- (2-cyclopropyl-6- (6- (hydroxymethyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AGT-1)
To intermediate (AEG-2) (60 mg,1 equivalent, 0.18 mmol), intermediate (AGH-1) (38 mg,1.3 equivalent, 0.23 mmol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 To a stirred mixture of (116 mg,0.36mmol,2 eq.) in 1, 4-dioxane (10 mL) were added RuPhos ring palladium complex gen.3 (30 mg,0.2 eq., 36 μmol) and RuPhos (33 mg,0.4 eq., 72 μmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1) to give the sub-title compound (AGT-1) (55 mg,0.12mmol,66%,90% purity) as a white solid. M/z 463.2 (M+H) + (ES+)。
Step 2: synthesis of 4- (2-cyclopropyl-6- (6-formyl-1-oxo-isoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AGT-2)
A solution of the product from step 1 above (AGT-1) (20 mg,1 eq, 43. Mu. Mol) and DMP (24 mg,1.3 eq, 56. Mu. Mol) in DCM (6 mL) was stirred at room temperature for 1 hour. Filtering the resulting mixture; the filter cake was washed with MeOH (3X 2 mL). The filtrate was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z461.2 (M+H) + (ES+)。
Step 3: synthesis of 4- { 2-cyclopropyl-6- [ 1-oxo-6- ({ [2- (trifluoromethoxy) ethyl ] amino } methyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGT-3)
To a stirred solution of the product from step 2 above (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and 2- (trifluoromethoxy) ethan-1-amine, HCl (13 mg,1.5 eq, 98. Mu. Mol) in MeOH (15 mL) was added DIPEA (51 mg,2 eq, 0.39 mmol) at room temperature. The resulting mixture was stirred at room temperature for 2 hours. Adding NaBH to the mixture at 0deg.C 4 (30 mg,4 equivalents, 0.78 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of 2mL of ice water at 0 ℃ and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 8% B to 32% B over 7 min; wavelength: 254/220nm; retention time: 6.12) to give the title compound (AGT-3) (10.4 mg,17 μmol,27%,95% purity) as a white solid. M/z 574.1 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.48(s,1H),8.15–8.03(m,3H),7.96–7.77(m,2H),7.73–7.56(m,2H),6.92(s,1H),5.03(s,2H),4.17(t,J=5.3Hz,2H),3.97(s,2H),3.49(s,3H),2.96(t,J=5.4Hz,2H),2.08–1.97(m,1H),1.08–0.94(m,4H)。
Example 224: synthesis of 2- [4' -chloro-2 ' - (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl ] -6- ({ [ (1-hydroxycyclopentyl) methyl ] amino } methyl) -3H-isoindol-1-one (AGU-10)
Step 1: synthesis of 2-bromo-5-chloro-N- [ (methylaminomethylsulfonyl) -amino ] -benzamide (AGU-1)
To a stirred mixture of 2-bromo-5-chlorobenzoic acid (ACR-1) (4.90 g,1 eq, 20.8 mmol) and DIPEA (8.07 g,3 eq, 62.4 mmol) in DMF (50 mL) at 0 ℃ was added 4-methyl-3-thiosemicarbazide (D-2) (2.63 g,1.2 eq, 25.0 mmol) and a T-containing solution 3 EtOAc (26.5 g,50Wt%,4 equivalents, 83.2 mmol) of P. The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and MeCN (0% MeCN up to 60% in 30 min); UV 254/220nm to give the sub-title compound (AGU-1) (4.5 g,11.5mmol,67%,82% purity) as a yellow solid. M/z 321.9/323.9 (M+H) + (ES+)。
Step 2: synthesis of 5- (2-bromo-5-chlorophenyl) -4-methyl-1, 2, 4-triazole-3-thiol (AGU-2)
At 0deg.C, naOH (2.79 g,69.745mmol,5 eq.) was added to H 2 To a stirred mixture of O (200 g) was added the product from step 1 above (AGU-1) (4.50 g,1 eq., 14.0 mmol). The resulting mixture was stirred at 60℃for 2 hours. The mixture was acidified to pH 4 with HCl (aqueous, 1M) at 0 ℃. The resulting mixture was diluted with water and extracted with DCM (3×300 mL). The combined organic layers were washed with brine (2×300 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (AGU-2) (4.3 g,10.6mmol, crude product, 75% purity) as a yellow solid. M/z 303.9/305.9 (M+H) + (ES+)。
Step 3: synthesis of 3- (2-bromo-5-chlorophenyl) -4-methyl-1, 2, 4-triazole (AGU-3)
To a stirred mixture of the product from step 2 above (AGU-2) (4.3 g,1 eq, 14.1 mmol) in DCM (50 mL) was added AcOH (1.70 g,2 eq, 28.2 mmol) dropwise at 0 ℃. Dropwise adding H to the above mixture at 0deg.C 2 O 2 (2.40 g,30Wt%,5 equivalents, 70.6 mmol). The resulting mixture was stirred at room temperature for 2 hours. The mixture was treated with saturated NaHCO 3 (aqueous, 1M) acidified to pH 7. The resulting mixture was diluted with water and extracted with DCM (3×300 mL). The combined organic layers were washed with brine (2×300 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (AGU-3) (2.8 g,8.30mmol,73%,80% purity) as a yellow solid. M/z 271.0/273.0 (M+H) + (ES+)。
Step 4: synthesis of 4' -chloro-2 ' - (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-amine (AGU-4)
To the product (AGU-3) (600 mg,1 eq, 2.20 mmol), 3-aminophenylboronic acid (AAS-1) (332 mg,1.1 eq, 2.42 mmol) and K from step 3 above under a nitrogen atmosphere at room temperature 2 CO 3 (913 mg,3 eq, 6.61 mmol) to a stirred solution of 1, 4-dioxane (10 mL) and water (1 mL) was added Pd (dppf) Cl 2 DCM (178 mg,0.1 eq, 0.22 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1) to give the sub-title compound (AGU-4) as a brown yellow solid (500 mg,1.58mmol,80%,90% purity). M/z 285.1/287.1 (M+H) + (ES+)。
Step 5: synthesis of 4- (bromomethyl) -3- (methoxycarbonyl) benzoic acid (AGU-6)
To 3- (methoxycarbonyl) -4-methylbenzoic acid (AGU-5) (2.00 g,1 eq., 10.3 mmol) and NBS (2.20 g,1.2 eq., 12.4 mmol) in CHCl at room temperature 3 BPO (792 mg,0.3 eq, 3.09 mmol) was added to the stirred solution in (60 mL). The resulting mixture was stirred at 80 ℃ overnight. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (1/6) to give the sub-title compound (AGU-6) (2.4 g,7.68mmol,85%,87% purity) as an off-white solid. M/z 273.0/275.0 (M+H) + (ES+)。
Step 6: synthesis of methyl 2- (bromomethyl) -5- (hydroxymethyl) benzoate (AGU-7)
To a stirred solution of the product from step 1 above (AGU-6) (2.00 g,1 eq., 7.32 mmol) in THF (40 mL) at 0deg.C under nitrogen was added drop wise BH 3 THF (18 mL,1M,2.5 eq, 18.3 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. The reaction was quenched with MeOH (10 mL) at 0deg.C. The resulting mixture was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (3/1) to give the sub-title compound (AGU-7) (1.1 g,3.75mmol,58%,88% purity) as an off-white solid. M/z 259.0/261.0 (M+H) + (ES+)。
Step 7: synthesis of methyl 2- (bromomethyl) -5-formylbenzoate (AGU-8)
To a stirred solution of the product from step 6 above (AGU-7) (1.20 g,1 eq., 4.63 mmol) and IBX (1.95 g,1.5 eq., 6.95 mmol) in EtOAc (20 mL) at room temperature. The resulting mixture was stirred at 70℃for 3 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/3) to give the sub-title compound (AGU-8) (700 mg,2.46mmol,59%,90% purity) as a brown yellow oil. M/z 257.0/259.0 (M+H) + (ES+)。
Step 8: synthesis of 2- [4' -chloro-2 ' - (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl ] -3-oxo-1H-isoindole-5-carbaldehyde (AGU-9)
To a stirred solution of the product from step 4 above (AGU-4) (400 mg,1 eq., 1.41 mmol) and the product from step 7 above (AGU-8) (433 mg,1.2 eq., 1.69 mmol) in MeCN (10 mL) and water (5 mL) at 0deg.C was added AgNO 3 (358 mg,1.5 eq, 2.11 mmol). The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 50% in 25 minutes); detector, UV 254/220nm. This gave the sub-title compound (AGU-9) (260 mg,0.57mmol,43%,94% purity) as a brown yellow solid. M/z 429.1/431.1 (M+H) + (ES+)。
Step 9: synthesis of 2- [4' -chloro-2 ' - (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl ] -6- ({ [ (1-hydroxycyclopentyl) methyl ] amino } methyl) -3H-isoindol-1-one (AGU-10)
To a stirred solution of the product from step 8 above (AGU-9) (70 mg,1 eq, 0.16 mmol), 1- (aminomethyl) cyclopentan-1-ol (AFQ-1) (23 mg,1.2 eq, 0.20 mmol) and DIPEA (63 mg,3 eq, 0.49 mmol) in MeOH (10 mL) at room temperature. The resulting mixture was stirred at 60℃for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (31 mg,5 equivalents, 0.82 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of 2mL of ice water at 0 ℃ and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 31% B to 48% B in 10 minutes; wavelength: 254/220 nm) to give the title compound (AGU-10) (24.4 mg, 45. Mu. Mol,28%,98% purity) as a white solid. M/z 528.4/530.4 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.43(s,1H),8.01–7.92(m,1H),7.85–7.55(m,7H),7.39(t,J=8.0Hz,1H),6.94–6.86(m,1H),4.86(s,2H),3.84(s,2H),3.11(s,3H),2.49(s,2H),1.75–1.62(m,2H),1.60–1.36(m,6H)。
Example 225: synthesis of 2- [4' -chloro-2 ' - (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl ] -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AGV-1)
At room temperature, to the intermediate(AGU-9) (70 mg,1 eq, 0.16 mmol), 1- (aminomethyl) cyclobutan-1-ol, HCl (AW-1) (20 mg,1.2 eq, 0.20 mmol) and DIPEA (63 mg,3 eq, 0.49 mmol) in MeOH (10 mL). The resulting mixture was stirred at 60℃for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (31 mg,5 equivalents, 0.82 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of 2mL of ice water at 0 ℃ and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 31% B to 48% B in 10 minutes; wavelength: 254/220 nm) to give the title compound (AGV-1) (29.3 mg, 56. Mu. Mol,35%,98% purity) as a white solid. M/z 514.4/516.4 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.43(s,1H),8.03–7.96(m,1H),7.84–7.73(m,2H),7.72–7.63(m,4H),7.59(d,J=7.8Hz,1H),7.37(t,J=8.0Hz,1H),6.92–6.82(m,1H),4.89(d,J=14.0Hz,3H),3.87(s,2H),3.10(s,3H),2.53(s,2H),2.06–1.95(m,2H),1.95–1.81(m,2H),1.70–1.53(m,1H),1.45–1.31(m,1H)。
Example 226: synthesis of 2- {4- [ 4-chloro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-cyclopropylpyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AGW-6)
Step 1: synthesis of 5-chloro-2- (2, 6-dichloropyridin-4-yl) benzoic acid (AGW-1).
Into a 250-mL round bottom flask purged and maintained with an inert nitrogen atmosphere at room temperature under a nitrogen atmosphere was placed a flask containing 2-bromo-5-chlorobenzoic acid (ACR-1) (5.00 g,1 eq, 21.2 mmol), 2, 6-dichloropyridin-4-ylboronic acid (AAN-1) (4.07 g,1 eq, 21.2 mmol) and K 3 PO 4 (13.5 g,3 eq, 63.7 mmol) 1, 4-dioxane (50 mL) and water (10 mL) followed by Pd (DtBPF))Cl 2 (1.38 g,0.1 eq, 2.12 mmol). The resulting solution was stirred overnight at 60 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×300 mL). The combined organic layers were washed with brine (2×300 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 70% in 20 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AGW-1) (1.8 g,5.32mmol,28%,89% purity) as a brown solid. M/z 301.9/303.9 (M+H) + (ES+)。
Step 2: synthesis of 2- (5-chloro-2- (2, 6-dichloropyridin-4-yl) benzoyl) -N-methylhydrazine-1-thiocarboxamide (AGW-2)
To a 100-mL round bottom flask purged with an inert nitrogen atmosphere and maintained at room temperature was placed a solution containing the product from step 1 above (AGW-1) (1.80 g,1 eq, 5.95 mmol) and DIPEA (2.31 g,3 eq, 17.9 mmol) in DMF (40 mL), followed by the addition of 4-methyl-3-thiosemicarbazide (D-2) (630 mg,1 eq, 5.95 mmol) and HATU (4.73 g,2.5 eq, 14.9 mmol) at room temperature. The resulting solution was stirred at room temperature for 6 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (60% MeCN up to 85% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AGW-2) (720 mg,1.70mmol,30%,92% purity) as a brown yellow solid. M/z 389.0/391.0 (M+H) + (ES+)。
Step 3: synthesis of 5- (5-chloro-2- (2, 6-dichloropyridin-4-yl) phenyl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (AGW-3).
Into a 50-mL round bottom flask was placed a flask containing the contents from the above stepsA solution of the product from step 2 (AGW-2) (650 mg,1 eq., 1.67 mmol) in NaOH in water (20 mL, 1M). The resulting solution was stirred at room temperature for 4 hours. The resulting solution was concentrated in vacuo. The crude product was used directly in the next step without any further purification. This gave the sub-title compound (AGW-3) (620 mg,1.37mmol, crude product, 82% purity) as a brown yellow solid. M/z 371.0/373.0 (M+H) + (ES+)。
Step 4: synthesis of 2, 6-dichloro-4- (4-chloro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridine (AGW-4).
A50-mL round-bottom flask, purged with an inert nitrogen atmosphere and maintained at room temperature, was charged with DCM (20 mL) containing the product from step 3 above (AGW-3) (600 mg,1 eq, 1.61 mmol). AcOH (194 mg,2 eq, 3.23 mmol) and H were then added at room temperature 2 O 2 (912 mg,30wt%,5 equivalents, 8.05 mmol). The resulting solution was stirred at room temperature for 4 hours. The resulting mixture was diluted with water and extracted with DCM (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (AGW-4) as a brown-yellow solid (500 mg,1.18mmol, crude, 80%). M/z 339.0/341.0 (M+H) + (ES+)。
Step 5: synthesis of 2-chloro-4- [ 4-chloro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-cyclopropylpyridine (AGW-5)
To the product (AGW-4) (200 mg,1 equivalent, 0.58 mmol), cyclopropylboronic acid (ABF-2) (75.8 mg,1.5 equivalent, 0.88 mmol) and K from step 4 above under a nitrogen atmosphere at room temperature 3 PO 4 (250 mg,2 eq, 1.17 mmol) in 1, 4-dioxane (5 mL) was added Pd (dppf) Cl 2 DCM (43 mg,0.05mmol,0.1 eq). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18;mobile phase, water (0.1% FA) and MeCN (20% MeCN up to 70% in 10 min); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AGW-5) (110 mg,0.29mmol,54%,90% purity) as a white solid. M/z 345.1/347.1 (M+H) + (ES+)。
Step 6: synthesis of 2- {4- [ 4-chloro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-cyclopropylpyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AGW-6)
To the product (AGW-5) (80 mg,1 equivalent, 0.23 mmol), intermediate (AEE-1) (57 mg,1 equivalent, 0.23 mmol) and Cs from step 5 above under a nitrogen atmosphere at room temperature 2 CO 3 (151 mg,2 eq, 0.46 mmol) to a stirred solution of RuPhos ring palladium complex gen.3 (39 mg,0.2 eq, 0.04 mmol) and RuPhos (43 mg,0.4 eq, 0.09 mmol) in 1, 4-dioxane (5 mL) was added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (2×30 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (20% MeCN up to 60% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30×150,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 43B to 49B in 8 minutes; a detector, UV 254/210nm; retention time: 7.70. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AGW-6) (5.3 mg,9.2 μmol,4.1%,97% purity) as a white solid. M/z 555.3/557.3 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.47(s,1H),8.05(d,J=1.4Hz,1H),7.85–7.77(m,2H),7.77–7.66(m,3H),7.65–7.59(m,1H),6.85(d,J=1.5Hz,1H),5.03(s,2H),3.94(s,2H),3.46(s,3H),2.71(s,2H),2.15–2.05(m,2H),1.99–2.00(m,3H),1.75–1.72(m,1H),1.54–1.47(m,1H),1.05–0.91(m,4H)。
Example 227: synthesis of 2- [6- (ethylamino) -4- [4- (4-methyl-1, 2, 4-triazol-3-yl) -1- (2-methylpropyl) pyrazol-3-yl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AGX-8)
Step 1: synthesis of ethyl 3-iodo-1- (2-methylpropyl) pyrazole-4-carboxylate (AGX-1)
To a stirred mixture of ethyl 3-iodo-1H-pyrazole-4-carboxylate (AAF-1) (2.66 g,1 eq, 10.0 mmol) in DMF (50 mL) under nitrogen at room temperature was added 1-iodo-2-methylpropane (2.76 g,1.5 eq, 15.0 mmol) and K 2 CO 3 (4.15 g,3 equivalents, 30.0 mmol). The resulting mixture was stirred at room temperature under nitrogen overnight. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (20% MeCN up to 80% in 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (AGX-1) (1.5 g,4.19mmol,47%,90% purity) as a white solid. M/z 323.0 (M+H) + (ES+)。
Step 2: synthesis of ethyl 3- (2, 6-dichloropyridin-4-yl) -1- (2-methylpropyl) pyrazole-4-carboxylate (AGX-2)
To the product (AGX-1) (640 mg,1 equivalent, 1.99 mmol), 2, 6-dichloropyridin-4-ylboronic acid (AAN-1) (383mg, 1 equivalent, 1.99 mmol) from step 1 above and K under a nitrogen atmosphere at room temperature 2 CO 3 (823 mg,3 eq, 5.96 mmol) in 1, 4-dioxane (25 mL) and H 2 Pd (DtBPF) Cl was added to the stirred mixture in O (5 mL) 2 (129 mg,0.1 eq, 0.20 mmol). The resulting mixture was stirred overnight at 60 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; flow ofPhase, water (0.1% NH) 4 HCO 3 ) And MeCN (10% MeCN up to 50% within 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (AGX-2) (540 mg,1.46mmol,79%,92% purity) as a white solid. M/z 342.1/344.1 (M+H) + (ES+)。
Step 3: synthesis of 3- (2, 6-dichloropyridin-4-yl) -1-isobutyl-1H-pyrazole-4-carboxylic acid (AGX-3)
To the product (AGX-2) from step 2 above (530 mg,1 eq, 1.55 mmol) in THF (8 mL) and H at room temperature 2 To the stirred mixture in O (2 mL) was added LiOH (185 mg,5 eq, 7.75 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (AGX-3) (470 mg,1.34mmol,96% pure) as a white solid. M/z 314.0/316.0 (M+H) + (ES+)。
Step 4: synthesis of 2- (3- (2, 6-dichloropyridin-4-yl) -1-isobutyl-1H-pyrazole-4-carbonyl) -N-methylhydrazine-1-thiocarboxamide (AGX-4)
To a stirred mixture of the product from step 3 above (AGX-3) (460 mg,1 eq, 1.46 mmol) and 1-amino-3-methyl thiourea (D-2) (185 mg,1.2 eq, 1.76 mmol) in THF (5 mL) was added HATU (557 mg,1 eq, 1.46 mmol) and DIPEA (618 mg,3 eq, 4.39 mmol) at 0deg.C. The resulting mixture was stirred overnight at room temperature under an air atmosphere. The resulting mixture was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 401.1/403.1 (M+H) + (ES+)。
Step 5: synthesis of 5- (3- (2, 6-dichloropyridin-4-yl) -1-isobutyl-1H-pyrazol-4-yl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (AGX-5)
NaOH (12.5 mL, 1M) was added dropwise to the reaction mixture of the product (AGX-4) from step 4 above at 0deg.C. The resulting mixture was stirred at room temperature overnight. Mixing the obtained mixtureThe mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 48% in 20 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AGX-5) (400 mg,0.97mmol,70%,93% purity) as a white solid. M/z 383.1/385.1 (M+H) + (ES+)。
Step 6: synthesis of 2, 6-dichloro-4- [4- (4-methyl-1, 2, 4-triazol-3-yl) -1- (2-methylpropyl) pyrazol-3-yl ] pyridine (AGX-6)
To a stirred mixture of the product (AGX-5) from step 5 above (400 mg,1 eq, 1.04 mmol) in DCM (5 mL) at 0deg.C was added AcOH (125 mg,2 eq, 2.09 mmol) and H 2 O 2 (177 mg,30Wt%,5 equivalents, 5.22 mmol). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (20% MeCN up to 60% in 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (AGX-6) (360 mg,0.97mmol,98%,94% purity) as a white solid. M/z 351.1/353.1 (M+H) + (ES+)。
Step 7: synthesis of 6-chloro-N-ethyl-4- [4- (4-methyl-1, 2, 4-triazol-3-yl) -1- (2-methylpropyl) pyrazol-3-yl ] pyridin-2-amine (AGX-7).
To a stirred mixture of the product from step 6 above (AGX-6) (100 mg,1 eq, 0.29 mmol) and ethylamino (128 mg,10 eq, 2.85 mmol) in NMP (2 mL) under nitrogen at room temperature was added K 2 CO 3 (393 mg,10 equivalents, 2.85 mmol). The resulting mixture was stirred overnight at 120 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (10% MeCN up to 50% in 20 minutes); detector, UV 254/220nm. This gives the sub-title compound (AGX-7) (90 m) as a white solidg,0.23mmol,88%,90% purity). M/z 360.2/362.2 (M+H) + (ES+)。
Step 8: synthesis of 2- [6- (ethylamino) -4- [4- (4-methyl-1, 2, 4-triazol-3-yl) -1- (2-methylpropyl) pyrazol-3-yl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AGX-8)
To the product (AGX-7) (40 mg,1 equivalent, 0.11 mmol), intermediate (AC-2) (22 mg,1 equivalent, 0.11 mmol) and Cs from step 7 above, under a nitrogen atmosphere at room temperature 2 CO 3 (72 mg,2 eq, 0.22 mmol) to a stirred mixture of 1, 4-dioxane (2 mL) was added Ruphos (21 mg,0.4 eq, 44. Mu. Mol) and Ruphos ring palladium complex Gen.3 (19 mg,0.2 eq, 22. Mu. Mol). The resulting mixture was stirred overnight at 80 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19X 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30B to 60B in 7 minutes; a detector, UV 254/210nm; retention time: 6.08. this gave the title compound (AGX-8) (11.9 mg, 22. Mu. Mol,20%,99% purity) as a white solid. M/z 525.2 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.59(s,1H),8.06(d,J=7.6Hz,2H),7.98–7.91(m,1H),7.78–7.69(m,1H),7.61(d,J=1.2Hz,1H),6.41(d,J=1.2Hz,1H),5.23(d,J=1.7Hz,2H),4.11(d,J=7.3Hz,2H),3.50(s,3H),3.37–3.27(m,2H),2.40–2.25(m,1H),1.23(t,J=7.2Hz,3H),1.00(d,J=6.7Hz,6H)。
Example 228: synthesis of tert-butyl N- [ (2- {4- [ 4-cyano-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-cyclopropylpyridin-2-yl } -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl) methyl ] -N-methylcarbamate (AGY-3)
Step 1: synthesis of 6- [ (methylamino) methyl ] -4- (trifluoromethyl) -2, 3-dihydro-isoindol-1-one (AGY-1)
MeOH (5 mL) containing intermediate (ABC-4) (200 mg,1 eq, 0.87 mmol), methylamine (136 mg,5 eq, 4.37 mmol) and AcOH (524 mg,10 eq, 8.73 mmol) was added to a 20mL sealed tube at room temperature. NaBH was added to the above mixture over 2 days at room temperature 3 CN (274 mg,5 eq., 4.37 mmol). The resulting mixture was stirred at room temperature overnight. Filtering the resulting mixture; the filter cake was washed with MeOH (3 mL). The filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (30% MeCN up to 60% in 15 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AGY-1) (112 mg,0.42mmol,53%,92% purity) as a yellow oil. M/z 245.1 (M+H) + (ES+)。
Step 2: synthesis of tert-butyl N-methyl-N- { [ 3-oxo-7- (trifluoromethyl) -1, 2-dihydro-isoindol-5-yl ] methyl } carbamate (AGY-2)
To an 8mL sealed tube at room temperature was added a solution containing the product (AGY-1) (80 mg,1 equivalent, 0.33 mmol) from step 1 above and Boc 2 O (143 mg,2 eq, 0.66 mmol) methyl (2 mL). DMAP (4 mg,0.1 eq, 33. Mu. Mol) was added to the above mixture over 3 minutes at room temperature. The resulting mixture was stirred at room temperature for 3 hours. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (36% MeCN up to 48% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AGY-2) (68 mg,0.19mmol,60%,95% purity) as a yellow oil. M/z 345.1 (M+H) + (ES+)。
Step 3: synthesis of tert-butyl N- [ (2- {4- [ 4-cyano-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-cyclopropylpyridin-2-yl } -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl) methyl ] -N-methylcarbamate (AGY-3)
To an 8mL sealed tube at room temperature was added a solution containing the product (AGY-2) (50 mg,1 equivalent, 0.15 mmol), intermediate (AEG-2) (54 mg,0.16mmol,1.1 equivalent) and Cs from step 2 above 2 CO 3 (95 mg,2 eq, 0.29 mmol) of 1, 4-dioxane (1 mL). Pd (OAc) was added to the above mixture over 3 minutes at room temperature under nitrogen atmosphere 2 (3.3 mg,0.1 eq, 14. Mu. Mol), xantPhos (17 mg,0.2 eq, 29. Mu. Mol). The resulting mixture was stirred under nitrogen at 100 ℃ for an additional 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19X 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30B to 60B in 7 minutes; a detector, UV 254/210nm; retention time: 6.08 to give the title compound (AGY-3) as a white solid (4.8 mg, 7.2. Mu. Mol,5.1%,97% purity). M/z 644.2 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),8.18–8.01(m,3H),7.99–7.90(m,2H),7.85(s,1H),6.95(d,J=1.4Hz,1H),5.19(s,2H),4.59(d,J=15.8Hz,2H),3.49(s,3H),2.92(s,3H),2.12–2.02(m1H),1.48(d,J=17.9Hz,9H),1.08–0.96(m,4H)。
Example 229: synthesis of 4- (2-cyclopropyl-6- {6- [ (methylamino) methyl ] -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AGZ-1)
DCM (1 mL) containing compound (AGY-3) (30 mg,1 eq, 47. Mu. Mol) was added to an 8mL sealed tube at room temperature. TFA (53 mg,10 equivalents, 0.47 mmol) was added dropwise to the above mixture over 1 minute at room temperature. The resulting mixture was stirred at room temperature for an additional 2 hours. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19X 150mm,5 μm; mobile phase a: water0.1%NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30B to 60B in 7 minutes; a detector, UV 254/210nm; retention time: 6.08 to give the title compound (AGZ-1) (9.7 mg, 17. Mu. Mol,38%,97% purity) as a white solid. M/z 544.2 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.48(s,1H),8.18–7.98(m,5H),7.92(d,J=8.1Hz,1H),6.95(d,J=1.5Hz,1H),5.19(s,2H),4.01(s,2H),3.48(s,3H),2.49(s,3H),2.13–2.02(m,1H),1.09–0.96(m,4H)。
Example 230: synthesis of 2- (4 ' -fluoro-2 ' - (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6- ((((1-hydroxycyclobutyl) methyl) amino) methyl) isoindolin-1-one (AHA-2)
Step 1: synthesis of 2- (4 ' -fluoro-2 ' - (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -3-oxoisoindoline-5-carbaldehyde (AHA-1)
Intermediate (AFN-1) (300 mg,1 eq, 1.12 mmol) and intermediate (AGU-8) (431 mg,1.5 eq, 1.68 mmol) were added to MeCN (6 mL) and H 2 A solution in O (3 mL) was treated with AgNO at room temperature 3 (284 mg,1.5 eq, 1.68 mmol) was treated overnight. The precipitated solid was collected by filtration and washed with MeCN (2 x5 mL). The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (20% MeCN up to 60% in 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (AHA-1) (120 mg,0.27mmol,26%,92% purity) as a yellow solid. M/z 413.1 (M+H) + (ES+)。
Step 2: synthesis of 2- (4 ' -fluoro-2 ' - (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6- ((((1-hydroxycyclobutyl) methyl) amino) methyl) isoindolin-1-one (AHA-2)
To the product (AHA-1) (50 mg,1 equivalent, 0.12 mmol) from step 1 above and 1- (amino) at 60℃CTo a stirred mixture of methyl) cyclobutan-1-ol, HCl (15 mg,1.2 eq, 0.15 mmol) in MeOH (5 mL) was added DIPEA (31 mg,2 eq, 0.24 mmol) for 1 hour. NaBH was added to the above mixture over 0.5 minutes at room temperature 4 (9 mg,2 equivalents, 0.24 mmol). The resulting mixture was stirred at room temperature for 1 hour. At 0℃with NH 4 The reaction was quenched with saturated aqueous Cl (1 mL). The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 5% B to 30% B over 7 min; wavelength: 254/220nm; retention time: 6.4) to give the title compound (AHA-2) as an off-white solid (18.8 mg,37 μmol,31%,98% purity). M/z 498.4 (M+H) + (ES+). 1 H NMR(300MHz,DMSO-d6)δ8.43(s,1H),8.22(s,1H),8.01–7.91(m,1H),7.78(s,1H),7.73–7.54(m,4H),7.49(m,1H),7.39–7.30(t,J=8.0Hz,1H),6.90–6.81(m,1H),4.88(s,2H),3.91(s,2H),3.12(s,3H),2.57(s,2H),2.11–1.80(m,4H),1.71–1.56(m,1H),1.49–1.29(m,1H)。
Example 231: synthesis of 2- (4 ' -fluoro-2 ' - (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -3-yl) -6- ((((1-hydroxycyclopentyl) methyl) amino) methyl) isoindolin-1-one (AHB-1)
DIPEA (47 mg,3 equiv., 0.36 mmol) was added dropwise to a stirred mixture of intermediate (AHA-1) (50 mg,1 equiv., 0.12 mmol) and 1- (aminomethyl) cyclopentan-1-ol (AFQ-1) (14 mg,1 equiv., 0.12 mmol) in MeOH (3 mL) at 60℃for 1 hour. NaBH was added to the above mixture over 0.5 minutes at room temperature 4 (14 mg,3 equivalents, 0.36 mmol). The resulting mixture was stirred at room temperature for 1 hour. At 0℃with NH 4 The reaction was quenched with saturated aqueous Cl (1 mL). The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: sunfire preparation Type C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 8% B to 23% B in 10 minutes; wavelength: 254/220nm; retention time: 10.5 To give the title compound (AHB-1) (17.8 mg, 33. Mu. Mol,26%,95% purity) as an off-white solid. M/z 512.5 (M+H) + (ES+). 1 H NMR(300MHz,DMSO-d6)δ8.43(s,1H),8.20-8.16(d,J=3.0Hz,1H),8.00–7.93(m,1H),7.80(s,1H),7.74–7.54(m,5H),7.52–7.43(m,1H),7.37(t,J=8.0Hz,1H),6.91–6.84(m,1H),4.88(s,2H),3.93(s,2H),3.12(s,3H),2.57(d,J=2.2Hz,2H),1.75–1.62(m,2H),1.58–1.44(m,6H)。
Example 232: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AHC-1)
To intermediate (AFO-1) (70 mg,1 equivalent, 0.28 mmol), intermediate (AEG-2) (115 mg,1.2 equivalent, 0.34 mmol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 (185 mg,2 eq, 0.57 mmol) to a stirred solution of RuPhos (53 mg,0.4 eq, 0.11 mmol) and RuPhos cyclopalladium complex Gen.3 (48 mg,0.2 eq, 57. Mu. Mol) in 1, 4-dioxane (5 mL). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 1 hour. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (20/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 60% B in 8 minutes; wavelength: 254/220nm; retention time: 7.8 To give the title compound (AHC-1) (18.2 mg, 33. Mu. Mol,12%,99% purity) as a white solid. M/z 546.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.54(s,1H),8.24–8.19(m,2H),7.98(d,J=1.4Hz,1H),7.90–7.74(m,2H),7.69–7.61(m,2H),6.89(d,J=1.4Hz,1H),5.00(s,2H),4.39(s,1H),3.96–3.76(m,3H),3.48(s,3H),2.80–2.72(m,1H),2.09–1.99(m,1H),1.70–1.50(m,4H),1.44–1.33(m,2H),1.01–0.92(m,4H)。
Example 233: synthesis of 2- [6- (ethylamino) -4- [4- (4-methyl-1, 2, 4-triazol-3-yl) -1-propylpyrazol-3-yl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AHD-9)
Step 1: synthesis of 3-iodo-1-propyl-1H-pyrazole-4-carboxylic acid ethyl ester (AHD-2)
To a 500-mL round bottom flask at room temperature was added K to a stirred mixture of ethyl 3-iodo-1H-pyrazole-4-carboxylate (AAF-1) (2.60 g,1 eq, 9.77 mmol) and iodopropane (AHD-1) (2.49 g,1.5 eq, 14.6 mmol) in DMF (20 mL) 2 CO 3 (4.05 g,3 equivalents, 29.3 mmol). The resulting mixture was stirred at room temperature overnight. The mixture was acidified with HCl (aqueous, 1M) to pH 4. The resulting mixture was diluted with water and extracted with EtOAc (3×300 mL). The combined organic layers were washed with brine (2×300 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 80% in 20 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AHD-2) (1.3 g,3.80mmol,43%,90% purity) as a colorless oil. M/z 309.0 (M+H) + (ES+)。
Step 2: synthesis of 3- (2, 6-dichloropyridin-4-yl) -1-propyl-1H-pyrazole-4-carboxylic acid ethyl ester (AHD-3).
To a stirred mixture of the product (AHD-2) (600 mg,1 eq, 1.95 mmol) from step 1 above and 2, 6-dichloropyridin-4-ylboronic acid (AAN-1) (373 mg,1 eq, 1.95 mmol) in 1, 4-dioxane (5 mL) and water (1 mL) at room temperature under a nitrogen atmosphere was addedAdding K 2 CO 3 (803 mg,3 equivalents, 5.84 mmol). Pd (DtBPF) Cl was added to the above mixture at room temperature under a nitrogen atmosphere 2 (127 mg,0.1 eq, 0.20 mmol). The resulting mixture was stirred overnight at 60 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (10% MeCN up to 60% in 15 minutes); detector, UV 254/220nm, to give the sub-title compound (AHD-3) (400 mg,1.09mmol,63%,89% purity) as a white solid. M/z 328.1/330.1 (M+H) + (ES+)。
Step 3: synthesis of 3- (2, 6-dichloropyridin-4-yl) -1-propyl-1H-pyrazole-4-carboxylic acid (AHD-4)
To a stirred mixture of product (AHD-3) (400 mg,1 eq, 1.22 mmol) from step 2 above and LiOH (88 mg,3 eq, 3.65 mmol) in THF (16 mL) at room temperature was added water (4 mL). The resulting mixture was stirred at 60℃for 2 hours. The mixture was acidified with HCl (aqueous, 1M) to pH 4. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (AHD-4) (340 mg,0.91mmol,93% pure) as a white solid. M/z 300.0/302.0 (M+H) + (ES+)。
Step 4: synthesis of 2- (3- (2, 6-dichloropyridin-4-yl) -1-propyl-1H-pyrazole-4-carbonyl) -N-methylhydrazine-1-thiocarboxamide (AHD-5)
To a stirred mixture of the product (AHD-4) (284 mg,1 eq, 0.98 mmol) from step 3 above in THF (20 mL) at 0deg.C was added dropwise the T-containing 3 EtOAc (1.25 g,50Wt%,4 equivalents, 3.92 mmol) of P. DIPEA (380 mg,3 eq, 2.94 mmol) and 1-amino-3-methyl thiourea (D-2) (124 mg,1.2 eq, 1.18 mmol) were added to the above mixture at 0deg.C. The resulting mixture was stirred at room temperature for 2 hours. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 50% in 30 minutes); detector, UV 254/220nm, to give the sub-title compound (AHD-5) (300 mg,0.72mmol,79%,92% purity) as a yellow solid. M/z 387.0/389.0 (M+H) + (ES+)。
Step 5: synthesis of 5- (3- (2, 6-dichloropyridin-4-yl) -1-propyl-1H-pyrazol-4-yl) -4-methyl-4H-1, 2, 4-triazole-3-thiol (AHD-6)
To a 50mL vial was added an aqueous NaOH solution (20 mL) containing the product from step 4 above (AHD-5) (300 mg,1 eq., 0.78 mmol) at room temperature. The resulting mixture was stirred at 60℃for 2 hours. The mixture was acidified with HCl (aqueous, 1M) to pH 4. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (AHD-6) (300 mg,0.61mmol, crude product, 75% purity) as a yellow solid. M/z 369.0/371.0 (M+H) + (ES+)。
Step 6: synthesis of 2, 6-dichloro-4- (4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1-propyl-1H-pyrazol-3-yl) pyridine (AHD-7)
To the product (AHD-6) (760 mg,1 eq., 2.06 mmol) from step 5 above and H at 0deg.C 2 O 2 To a stirred solution of (350 mg,30wt%,5 eq, 10.3 mmol) in DCM (20 mL) was added AcOH (247 mg,2 eq, 4.12 mmol). The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 40% in 15 minutes); detector, UV 254/220nm, to give the sub-title compound (AHD-7) (560 mg,1.50mmol,81%,90% purity) as a yellow solid. M/z 337.1/339.1 (M+H) + (ES+)。
Step 7: synthesis of 6-chloro-N-ethyl-4- (4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1-propyl-1H-pyrazol-3-yl) pyridin-2-amine (AHD-8)
The reaction mixture was subjected to the above-mentioned steps at room temperature under a nitrogen atmosphere6 (AHD-7) (150 mg,1 eq., 0.30 mmol) and ethylamine, HCl (242 mg,10 eq., 2.97 mmol) in NMP (3 mL) were added K 2 CO 3 (410 mg,10 equivalents, 2.97 mmol). The resulting mixture was stirred overnight at 120 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (0% MeCN up to 38% in 20 minutes); detector, UV 254/220nm, to give the sub-title compound (AHD-8) (110 mg,0.29mmol,80%,92% purity) as a colourless solid. M/z 346.1/348.1 (M+H) + (ES+)。
Step 8: synthesis of 2- (6- (ethylamino) -4- (4- (4-methyl-4H-1, 2, 4-triazol-3-yl) -1-propyl-1H-pyrazol-3-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AHD-9)
Cs was added to a stirred solution of the product (AHD-8) (50 mg,1 eq, 0.15 mmol) from step 7 above and intermediate (AC-2) (29 mg,1 eq, 0.15 mmol) in 1, 4-dioxane (5 mL) at room temperature under nitrogen atmosphere 2 CO 3 (94 mg,2 equivalents, 0.29 mmol). RuPhos (27 mg,0.4 eq, 58. Mu. Mol) and RuPhos ring palladium complex Gen.3 (24 mg,0.2 eq, 29. Mu. Mol) were added to the above mixture under nitrogen atmosphere at room temperature. The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 60% B in 9 minutes; wavelength: 254/220nm; retention time: 8.88 To give the title compound (AHD-9) (13.8 mg, 27. Mu. Mol,19%,99% purity) as a white solid. M/z 511.2 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.59(s,1H),8.09–8.03(m,2H),7.98–7.91(m,1H),7.78–7.69(m,1H),7.61(d,J=1.2Hz,1H),6.41(d,J=1.2Hz,1H),5.25–5.21(m,2H),4.27(t,J=7.0Hz,2H),3.49(s,3H),3.34(d,J=7.2Hz,2H),2.07–1.94(m,2H),1.23(t,J=7.2Hz,3H),1.00(t,J=7.4Hz,3H)。
Example 234: synthesis of 4- { 2-ethoxy-6- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AHE-4)
Step 1: synthesis of 4- {2- [6- (chloromethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -6-ethoxypyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AHE-1)
To a stirred solution of intermediate (AEW-2) (80 mg,1 eq, 0.15 mmol) in DCM (10 mL) at 0deg.C was added SOCl dropwise 2 (0.4 mL,22.5 eq, 3.36 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was quenched with MeOH (1 mL) at 0deg.C. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1) to give the sub-title compound (AHE-1) (61.5 mg,0.11mmol,67% pure) as a yellow solid. M/z 553.1/555.1 (M+H) + (ES+)。
Step 2: synthesis of tert-butyl N- {2- [ (2- {4- [ 4-cyano-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-ethoxypyridin-2-yl } -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl) methoxy ] ethyl } -N-methylcarbamate (AHE-3)
To a stirred solution of the product from step 1 above (AHE-1) (60 mg,1 eq, 54. Mu. Mol) and tert-butyl N- (2-hydroxyethyl) -N-methylcarbamate (AHE-2) (190 mg,10 eq, 0.54 mmol) in DCM (2 mL) at room temperature was added TBAB (18 mg,0.5 eq, 27. Mu. Mol) and aqueous KOH (2 mL,20 Wt%). The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue is taken upPurification by preparative TLC using DCM/MeOH (10/1) gave the sub-title compound (AHE-3) (40 mg, 52. Mu. Mol,96%,90% purity) as a yellow solid. M/z 692.2 (M+H) + (ES+)。
Step 3: synthesis of 4- { 2-ethoxy-6- [6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AHE-4)
To a stirred solution of the product from step 2 above (AHE-3) (30 mg,1 eq, 43. Mu. Mol) in DCM (9 mL) at room temperature was added TFA (3 mL). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.05% TFA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 10% B to 20% B over 7 min; wavelength: 254/220nm; retention time: 5.2) to give the title compound (AHE-4) as a yellow solid (8.3 mg,14 μmol,35%,98% purity). M/z592.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.24–8.15(m,2H),8.03(d,J=16.4Hz,2H),7.89–7.83(m,1H),7.81(d,J=1.2Hz,1H),6.43(d,J=1.2Hz,1H),5.22(s,2H),4.70(s,2H),4.33(q,J=7.0Hz,2H),3.59(t,J=5.5Hz,2H),3.48(s,3H),2.76(t,J=5.5Hz,2H),2.34(s,3H),1.36(t,J=7.0Hz,3H)。
Example 235: synthesis of 4- (2-cyclopropyl-6- {6- [ (2-hydroxyethoxy) methyl ] -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AHF-4)
Step 1: synthesis of 4- {2- [6- (chloromethyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] -6-cyclopropylpyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AHF-1)
To a stirred solution of intermediate (AES-1) (60 mg,1 eq, 0.11 mmol) in DCM (10 mL) at 0deg.C was added SOCl dropwise 2 (0.4 mL,29.7 eq)3.36 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was quenched by the addition of MeOH (4 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (20/1) to give the sub-title compound (AHF-1) (46 mg, 76. Mu. Mol,67%,91% purity) as a yellow solid. M/z 549.1/551.1 (M+H) + (ES+)。
Step 2: synthesis of 4- [2- (6- {2- [ (tert-butyldimethylsilyl) oxy ] ethoxy } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AHF-3)
To the product (AHF-1) (40 mg,1 eq, 36. Mu. Mol) from step 1 above and 2- [ (tert-butyldimethylsilyl) oxy group at room temperature]To a stirred solution of ethanol (AHF-2) (128 mg,10 eq, 0.36 mmol) in DCM (2 mL) was added TBAB (12 mg,0.5 eq, 18. Mu. Mol) and aqueous KOH (2 mL,20 wt%). The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1) to give the sub-title compound (AHF-3) (11 mg, 15. Mu. Mol,40%,96% purity) as a yellow solid. M/z 689.3 (M+H) + (ES+)。
Step 3: synthesis of 4- (2-cyclopropyl-6- {6- [ (2-hydroxyethoxy) methyl ] -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AHF-4)
To a stirred solution of the product from step 2 above (AHF-3) (10 mg,1 eq, 15. Mu. Mol) in THF (7 mL) at room temperature was added TEA.3HF (0.5 mL) dropwise. The resulting mixture was stirred at room temperature for 5 hours. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35% B to 60% B in 7 min; wavelength: 254/220nm; retention time: 6.8) to giveThe title compound (AHF-4) (0.9 mg, 1.5. Mu. Mol,11%,98% purity) was in the form of a white solid. M/z 575.0 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.49(s,1H),8.14–7.90(m,6H),6.95(s,1H),5.16(s,2H),4.74(s,2H),3.76(t,J=4.7Hz,2H),3.66(t,J=4.8Hz,2H),3.49(s,3H),2.09–2.02(m,1H),1.31(s,2H),1.01(d,J=6.4Hz,2H)。
Example 236: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- { [ (2-hydroxy-2-methylpropyl) amino ] methyl } -3H-isoindol-1-one (AHG-2)
Step 1: synthesis of 6- { [ (2-hydroxy-2-methylpropyl) amino ] methyl } -2, 3-dihydro-isoindol-1-one (AHG-1).
DIPEA (209 mg,2 eq, 1.61 mmol) was added to a stirred mixture of intermediate (ACY-2) (130 mg,1 eq. 0.81 mmol) and 1-amino-2-methylpropan-2-ol (AZ-1) (144 mg,2 eq., 1.61 mmol) in MeOH (13 mL) at room temperature under nitrogen. NaBH was added to the above mixture at 60℃over 2 hours 4 (61 mg,2 equivalents, 1.61 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was then quenched by addition of 5mL of ice water at 0 ℃. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo to give the sub-title compound (AHG-1) (120 mg,0.37mmol, crude, 73% purity) as a yellow solid. M/z 235.1 (M+H) + (ES+)。
Step 2: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- { [ (2-hydroxy-2-methylpropyl) amino ] methyl } -3H-isoindol-1-one (AHG-2)
To the product (AHG-1) (60 mg,1 eq, 0.26 mmol), intermediate (ADN-1) (84 mg,1 eq, 0.26 mmol) and Cs from step 1 above, under nitrogen at room temperature 2 CO 3 (167 mg,2 eq, 0.51 mmol) to a stirred mixture of 1, 4-dioxane (6 mL) was added RuPhos ring palladium complex Gen.3 (43 mg,0.2 eq, 51. Mu. Mol) and RuPhos (48 mg,0.4 eq, 102. Mu. Mol). The resulting mixture was stirred under nitrogen at 80 ℃ for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 13% B to 28% B over 10 min; wavelength: 254/220nm; retention time: 9.7) to give the title compound (AHG-2) as a white solid (19.8 mg,37 μmol,15%,99% purity). M/z 527.3 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.46(s,1H),8.03(d,J=1.4Hz,1H),7.85(d,J=1.4Hz,1H),7.82–7.74(m,1H),7.74–7.67(m,1H),7.64(d,J=7.8Hz,1H),7.58–7.49(m,1H),7.49–7.43(m,1H),6.86(d,J=1.4Hz,1H),5.04(s,2H),3.99(s,2H),3.46(s,3H),2.61(s,2H),2.06–1.95(m,1H),1.22(s,6H),1.06–0.92(m,4H)。
Example 237: synthesis of 6- { [ (cyclopentylmethyl) amino ] methyl } -2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -3H-isoindol-1-one (AHH-3)
Step 1: synthesis of 6- { [ (cyclopentylmethyl) amino ] methyl } -2, 3-dihydro-isoindol-1-one (AHH-2).
To a stirred mixture of intermediate (ACY-2) (120 mg,1 eq, 0.75 mmol), 1-cyclopentylmethylamine (AHH-1) (111 mg,1.5 eq, 1.12 mmol) and AcOH (22 mg,0.5 eq, 0.37 mmol) in MeOH (24 mL) was added DIPEA (115 mg,1.2 eq, 0.89 mmol) under a nitrogen atmosphere at room temperature. NaBH4 (56 mg,2 equivalents, 1.49 mmol) was added to the above mixture over 2 hours at 60 ℃. The resulting mixture was stirred at room temperatureMix for another 2 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo to give the sub-title compound (AHH-2) (100 mg,0.37mmol,55%,90% purity) as a yellow solid. M/z 245.2 (M+H) + (ES+)。
Step 2: synthesis of 6- { [ (cyclopentylmethyl) amino ] methyl } -2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -3H-isoindol-1-one (AHH-3)
To the product (AHH-2) (50 mg,1 eq., 0.21 mmol), intermediate (ADN-1) (67 mg,1 eq., 0.21 mmol) and Cs from step 1 above under nitrogen at room temperature 2 CO 3 (31 mg,2 eq, 0.41 mmol) to a stirred mixture of 1, 4-dioxane (6 mL) was added RuPhos ring palladium complex Gen.3 (34 mg,0.2 eq, 41. Mu. Mol) and RuPhos (38 mg,0.4 eq, 82. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.05% fa), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 15% B to 31% B over 10 min; wavelength: 254/220nm; retention time: 10.4) to give the title compound (AHH-3) (21.1 mg,37 μmol,18%,95% purity) as a white solid. M/z 537.6 (M+H) + (ES+). 1 H NMR(400MHz,MeOH-d4)δ8.46(s,1H),8.04(d,J=1.4Hz,1H),7.83(s,1H),7.81–7.73(m,1H),7.72–7.61(m,2H),7.58–7.49(m,1H),7.49–7.43(m,1H),6.86(d,J=1.4Hz,1H),5.04(s,2H),3.96(s,2H),3.46(s,3H),2.62(d,J=7.2Hz,2H),2.14–1.95(m,2H),1.89–1.80(m,2H),1.70–1.50(m,4H),1.19(s,2H),1.06–0.92(m,4H)。
Example 238: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (3R) -1-methylpyrrolidin-3-yl ] methoxy } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AHI-4)
Step 1: synthesis of benzyl (3R) -3- { [ (2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl) methoxy ] methyl } pyrrolidine-1-carboxylate (AHI-2)
To a stirred solution of intermediate (AFL-1) (120 mg,1 eq, 0.22 mmol) and benzyl (3R) -3- (hydroxymethyl) pyrrolidine-1-carboxylate (AHI-1) (130 mg,10 eq, 0.55 mmol) in DCM (5 mL) at room temperature was added TBAB (36 mg,0.5 eq, 0.11 mmol) and aqueous KOH (5 mL,20 wt%). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (60% MeCN up to 75% in 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (AHI-2) (77 mg, 96. Mu. Mol,47%,92% purity) as a white solid. M/z 741.3 (M+H) + (ES+)。
Step 2: synthesis of (R) -2- (6-cyclopropyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((pyrrolidin-3-ylmethoxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (AHI-3)
A solution of the product from step 1 above (AHI-2) (77 mg,1 eq, 104. Mu. Mol) and TFA (5 mL) in DCM (5 mL) was stirred at 60℃for 2 hours. The mixture was cooled to room temperature and then concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 607.2 (M+H) + (ES+)。
Step 3:2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (3R) -1-methylpyrrolidin-3-yl ] methoxy } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AHI-4)
To a stirred solution of the product from step 2 above (AHI-3) (63 mg,1 eq, 104. Mu. Mol) and formaldehyde (9.4 mg,1.2 eq, 125. Mu. Mol) in MeOH (5 mL) was added DIPEA (41 mg,3 eq, 0.31 mmol) at room temperature. The resulting mixture was stirred at 60℃for 1 hour. The mixture was cooled to room temperature. NaBH was added to the above mixture at room temperature 3 CN (13 mg,2 equivalents, 0.21 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of 2mL of ice water at 0 ℃ and concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% fa), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 12% B to 42% B over 7 min; wavelength: 254/220 nm) to give the title compound (AHI-4) (11.0 mg,18 μmol,17%,99% purity) as a white solid. M/z 621.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO–d6)δ8.50(s,1H),8.03–7.92(m,3H),7.76–7.68(m,1H),7.66–7.56(m,2H),6.80(s,1H),5.17(s,2H),4.67(s,2H),3.42–3.37(m,5H),2.48–2.32(m,4H),2.32–2.19(m,4H),2.09–2.01(m,1H),1.94–1.80(m,1H),1.47–1.33(m,1H),1.04–0.94(m,2H),0.94–0.84(m,2H)。
Example 239: synthesis of (R) -2- (6-chloro-4- (1- (4-methyl-4H-1, 2, 4-triazol-3-yl) propan-2-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AHJ-11)
Step 1: (E) Synthesis of ethyl-3- (2, 6-dichloropyridin-4-yl) acrylate (AHJ-3)
To a stirred solution of 2, 6-dichloropyridine-4-carbaldehyde (AHJ-1) (8.00 g,1 eq, 45.5 mmol) in DCM (50 mL) was added ethyl 2- (triphenylλ5-phosphanylidene) acetate (AHJ-2) (1.58 g,1 eq, 45.5 mmol) at 0deg.C. The resulting mixture was stirred at room temperature for 1 hour. The mixture was placed in vacuo Concentrating. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (5/1) to give the sub-title compound (AHJ-3) (7.5 g,26.3mmol,72%,86% purity) as a white solid. M/z 246.0/248.0 (M+H) + (ES+)。
Step 2: (2E) Synthesis of (E) -3- (2, 6-dichloropyridin-4-yl) prop-2-enoic acid (AHJ-4)
To a stirred solution of the product from step 1 above (AHJ-3) (9.20 g,1 eq, 37.4 mmol) in THF (160 mL) and water (160 mL) at 0deg.C was added LiOH (4.50 g,5 eq, 187 mmol). The resulting mixture was stirred at room temperature overnight. The mixture was concentrated in vacuo and diluted with water. The pH of the solution was adjusted to 3 with aqueous HCl (1M). The solid was collected by filtration to give the sub-title compound (AHJ-4) (7.3 g,27.2mmol,90%,82% purity) as a yellow solid. M/z 218.0/220.0 (M+H) + (ES+)。
Step 3: synthesis of (4R) -3- [ (2E) -3- (2, 6-dichloropyridin-4-yl) prop-2-enoyl ] -4-phenyl-1, 3-oxazolidin-2-one (AHJ-5)
To the product (AHJ-4) (4.40 g,1 equivalent, 20.2 mmol) from step 2 above and Et at 0deg.C 3 To a solution of N (4.50 g,2.2 eq, 44.4 mmol) in THF (80 mL) was added 2, 2-dimethylpropionyl chloride (2.40 g,1 eq, 20.2 mmol). After stirring for 1 hour, THF (20 mL) containing LiCl (800 g,1 equivalent, 20.2 mmol) and (4R) -4-phenyl-1, 3-oxazolidin-2-one (3.30 g,1 equivalent, 20.2 mmol) was added dropwise to the above mixture at 0 ℃ over 10 minutes. The resulting mixture was stirred at room temperature overnight. The reaction was carried out at 0deg.C with NH 4 Aqueous Cl (5 mL, 1M) was quenched. The resulting mixture was diluted with water and extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (5/1). This gave the sub-title compound (AHJ-5) (5.8 g,14.0mmol,79%,88% purity) as a white solid. M/z 363.0/365.0 (M+H) + (ES+)。
Step 4: synthesis of (4R) -3- [ (3R) -3- (2, 6-dichloropyridin-4-yl) butanoyl ] -4-phenyl-1, 3-oxazolidin-2-one (AHJ-6)
Dropwise adding copper (I) bromide into a 500mL 3-neck round bottom flask at-40 ℃ under nitrogen atmosphere; to THF (100 mL) containing dimethyl sulfide (4.90 g,1.5 eq, 24.0 mmol) was added MeMgBr (16 mL,3m,8.68 eq, 134 mmol) dropwise. After stirring for 40 minutes, BF was added dropwise to the above mixture at-40℃over 5 minutes 3 .Et 2 O (3.40 g,1.5 eq, 24.0 mmol). The resulting mixture was stirred at-40℃for 40 minutes. THF (20 mL) containing the product from step 3 above (AHJ-5) (5.80 g,1 eq., 16.0 mmol) was added dropwise to the above mixture at-40℃over 10 min. The resulting mixture was stirred at-40℃for 30 minutes. The reaction was carried out at 0deg.C with NH 4 Aqueous Cl (5 mL, 1M) was quenched. The resulting mixture was diluted with water and extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (6/1). This gave the sub-title compound (AHJ-6) (1.6 g,3.81mmol,26% pure) as a colourless solid. M/z 379.1/381.1 (M+H) + (ES+)。
Step 5: synthesis of (3R) -3- (2, 6-dichloropyridin-4-yl) butanoylhydrazine (AHJ-7)
To a solution of the product from step 4 above (AHJ-6) (1.50 g,1 eq., 4.00 mmol) in THF (30 mL) was added hydrazine hydrate (64% hydrazine) (246 mg,64Wt%,2 eq., 7.91 mmol) at room temperature. The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo. The crude product was used directly in the next step without any further purification. This gave the sub-title compound (AHJ-7) (950 mg,3.07mmol, crude product, 80% purity) as a white solid. M/z 248.0/250.0 (M+H) + (ES+)。
Step 6: synthesis of (3R) -3- (2, 6-dichloropyridin-4-yl) -N- [ (methylaminomethylsulfonyl) amino ] butanamide (AHJ-8)
To a stirred solution of the product from step 5 above (AHJ-7) (1.50 g,1 eq, 6.05 mmol) in THF (15 mL) at room temperatureMethyl isothiocyanate (530 mg,1.2 equivalents, 7.26 mmol) was added thereto. The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo. The crude product was used directly in the next step without any further purification. This gave the sub-title compound (AHJ-8) (1.5 g,4.08mmol,77%,87% purity) as a white solid. M/z 321.0/323.0 (M+H) + (ES+)。
Step 7: synthesis of 5- [ (2R) -2- (2, 6-dichloropyridin-4-yl) propyl ] -4-methyl-1, 2, 4-triazole-3-thiol (AHJ-9)
To a stirred solution of the product from step 6 above (AHJ-8) (1.50 g,1 eq., 4.67 mmol) in THF (15 mL) at 0deg.C was added aqueous NaOH (16 mL, 1M). The resulting mixture was stirred at room temperature overnight. The mixture was concentrated in vacuo and diluted with water. The pH of the solution was adjusted to 3 with aqueous HCl (1M) at 0deg.C. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (1×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (AHJ-9) (1.0 g,2.81mmol,71% pure) as a white solid. M/z 303.0/305.0 (M+H) + (ES+)。
Step 8: synthesis of 2, 6-dichloro-4- [ (2R) -1- (4-methyl-1, 2, 4-triazol-3-yl) propan-2-yl ] pyridine (AHJ-10)
To a stirred solution of the product from step 7 above (AHJ-9) (1.00 g,1 eq, 3.30 mmol) in DCM (10 mL) at 0deg.C was added H 2 O 2 (224 mg,30Wt%,2 equivalents, 6.60 mmol) and AcOH (390 mg,2 equivalents, 6.60 mmol). The resulting mixture was stirred at room temperature for 3 hours. The mixture was treated with Na 2 CO 3 Is alkalized to pH 8 with saturated aqueous solution (1M). The resulting mixture was diluted with water and extracted with DCM (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (AHJ-10) (800 g,2.73mmol,89%,92% purity) as a yellow solid. M/z 271.0/273.0 (M+H) + (ES+)。
Step 9: synthesis of (R) -2- (6-chloro-4- (1- (4-methyl-4H-1, 2, 4-triazol-3-yl) propan-2-yl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AHJ-11)
To the product (AHJ-10) (150 mg,1 eq, 0.55 mmol), intermediate (AC-2) (45 mg,0.4 eq, 0.22 mmol) and K from step 8 above under nitrogen at room temperature 3 PO 4 (235 mg,2 eq, 1.10 mmol) in 1, 4-dioxane (5 mL) was added XantPhos (64 mg,0.2 eq, 0.11 mmol) and Pd (OAc) 2 (12 mg,0.1 eq, 0.05 mmol). The resulting mixture was stirred under nitrogen at 100 ℃ for 16 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by reverse phase flash under the following conditions: a column, C18 column; mobile phase, water (0.1% FA) and MeCN (20% MeCN up to 70% in 20 minutes); UV detection at 254/220 nm. The product-containing fractions were combined and concentrated in vacuo. The residue was purified by preparative HPLC under the following conditions: column: 5 μm,19×150mm of SunFire preparative C18 OBD column; mobile phase, water (0.1% FA) and MeCN (35% MeCN up to 55% in 10 min); UV detection at 254/220 nm. The product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AHJ-11) (6.5 mg,15 μmol,2.6%,99% purity) as a white solid. M/z 436.3/438.3 (M+H) + (ES+)。 1 H NMR(400MHz,MeOH-d4)δ8.44(d,J=1.2Hz,1H),8.35(s,1H),8.13(d,J=7.7Hz,1H),8.04–7.98(m,1H),7.83–7.74(m,1H),7.19(d,J=1.2Hz,1H),5.24(d,J=2.0Hz,2H),3.69(s,3H),3.52–3.41(m,1H),3.19(d,J=7.6Hz,2H),1.45(d,J=6.9Hz,3H)。
Example 240: synthesis of N- [4- [ (2R) -1- (4-methyl-1, 2, 4-triazol-3-yl) propan-2-yl ] -6- [ 1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-2-yl ] methanesulfonamide (AHK-1)
To a compound (AHJ-11) (60 mg,1 equivalent, 0.14 mmol), methanesulfonamide (AAW-1) (26 mg,2 equivalent, 0.28 mmol) and under a nitrogen atmosphere at room temperature K 3 PO 4 (58 mg,2 eq, 0.28 mmol) in 1, 4-dioxane (4 mL) was added XantPhos (16 mg,0.2 eq, 28. Mu. Mol) and Pd (OAc) 2 (3 mg,0.1 eq, 14. Mu. Mol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 50% in 15 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: YMC-Actus Triart C18, 30X 250mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 H2O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 5B to 35B in 7 minutes; UV 254/210nm; retention time: 6.32). The product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AHK-1) (21.7 mg,43 μmol,32%,99% purity) as a white solid. M/z 495.0 (M+H) + (ES+)。 1 H NMR(300MHz,MeOH–d4)δ8.31(s,1H),8.19–8.08(m,2H),7.99(d,J=7.7Hz,1H),7.77(t,J=7.7Hz,1H),6.66(d,J=1.2Hz,1H),5.29(s,2H),3.64(s,3H),3.44–3.39(m,1H),3.34(s,3H),3.16(d,J=7.6Hz,2H),1.44(d,J=6.9Hz,3H)。
Example 241: synthesis of N- [4- [ (2S) -1- (4-methyl-1, 2, 4-triazol-3-yl) propan-2-yl ] -6- [ 1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-2-yl ] methanesulfonamide (AHL-8)
Step 1: synthesis of (S, E) -3- (3- (2, 6-dichloropyridin-4-yl) acryloyl) -4-phenyloxazolidin-2-one (AHL-1)
At 0deg.C, to intermediate (AHJ-4) (4.40 g,1 eq, 20.2 mmol) and Et 3 To a stirred solution of N (4.49 g,2.2 eq, 44.4 mmol) in THF (80 mL) was added 2, 2-dimethylpropionyl chloride (2.43 g,1 eq, 20.2 mmol). After 1 hour, THF (20 mL) containing LiCl (860 mg,1 eq, 20.2 mmol) and (4S) -4-phenyl-1, 3-oxazolidin-2-one (3.29 g,1 eq, 20.2 mmol) was added dropwise to the above mixture at 0 ℃ over 10 minutes. The resulting mixture was stirred at room temperature overnight. At room temperature, with NH 4 Aqueous Cl (10 mL) quenched the reaction. The resulting mixture was diluted with water and extracted with EtOAc (3×500 mL). The combined organic layers were washed with brine (2×500 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (5/1) to give the sub-title compound (AHL-1) (5.8 g,13.6mmol,79%,85% purity) as a white solid. M/z 363.0/365.0 (M+H) + (ES+)。
Step 2: synthesis of (4S) -3- [ (3S) -3- (2, 6-dichloropyridin-4-yl) butanoyl ] -4-phenyl-1, 3-oxazolidin-2-one (AHL-2)
To a stirred solution of copper (I) bromide, dimethyl sulfate (4.92 g,1.5 eq., 24.0 mmol) in THF (100 mL) was added MeMgBr (15.97 mL,3M,8.68 eq., 134 mmol) dropwise under nitrogen at-40 ℃. After 40 minutes, BF was added dropwise to the above mixture at-40℃over 5 minutes 3 .Et 2 O (3.40 g,1.5 eq, 24.0 mmol). The resulting mixture was stirred at-40℃for 40 minutes. THF (20 mL) containing the product (AHL-1) from step 1 above (5.80 g,1 eq., 16.0 mmol) was added dropwise to the above mixture over 10 minutes at-40 ℃. The resulting mixture was stirred at-40℃for 30 minutes. By adding NH at-40 DEG C 4 The reaction was quenched with saturated aqueous Cl (20 mL). The resulting mixture was diluted with water and extracted with EtOAc (3×400 mL). The combined organic layers were washed with brine (2×400 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (6/1) to give the sub-title compound (AHL-2) (1.6 g,3.68mmol,26%,87% purity) as a colorless solid. M/z 379.1/381.1 (M+H) + (ES+)
Step 3: synthesis of (3S) -3- (2, 6-dichloropyridin-4-yl) butanoylhydrazine (AHL-3)
To a stirred solution of the product from step 2 above (AHL-2) (770 mg,1 eq., 2.03 mmol) in THF (15 mL) was added hydrazine hydrate (64% hydrazine) (126 mg,64Wt%,4.06mmol,2 eq.) at room temperature. The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo. This gave the sub-title compound (AHL-3) (500 mg,1.14mmol,99% purity) as a yellow oil. M/z 348.0/350.0 (M+H) + (ES+)。
Step 4: synthesis of (3S) -3- (2, 6-dichloropyridin-4-yl) -N- [ (methylaminomethylsulfonyl) amino ] butanamide (AHL-4)
To a stirred solution of the product from step 3 above (AHL-3) (1.00 g,1 eq, 4.03 mmol) in THF (30 mL) at room temperature was added methyl isothiocyanate (354 mg,1.2 eq, 4.84 mmol). The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo. This gave the sub-title compound (AHL-4) (1.2 g,2.73mmol,92%,78% purity) as a yellow oil. M/z 321.0/323.0 (M+H) + (ES+)。
Step 5: synthesis of 5- [ (2S) -2- (2, 6-dichloropyridin-4-yl) propyl ] -4-methyl-1, 2, 4-triazole-3-thiol (AHL-5)
To a stirred solution of the product from step 4 above (AHL-4) (1.20 g,1 eq, 3.74 mmol) in THF (30 mL) at room temperature was added dropwise aqueous NaOH (11 mL, 1M). The mixture was acidified to pH 3 with aqueous HCl (1M) at 0 ℃. The resulting mixture was diluted with water (40 mL). The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (AHL-5) (1.0 g,2.58mmol,88%,78% purity) as a yellow oil. M/z 303.0/305.0 (M+H) + (ES+)。
Step 6: synthesis of 2, 6-dichloro-4- [ (2S) -1- (4-methyl-1, 2, 4-triazol-3-yl) propan-2-yl ] pyridine (AHL-6)
To a stirred solution of the product from step 5 above (AHL-5) (1.96 g,1 eq., 6.46 mmol) in DCM (30 mL) at 0deg.C was added AcOH (10 mL,0.03 eq., 0.17 mmol) and H dropwise 2 O 2 (6.80 mL,30Wt%,0.03 eq, 0.20 mmol). The resulting mixture was stirred at 0 ℃ for 2 hours. The mixture was treated with NaHCO 3 Is alkalized to pH8 with saturated aqueous solution (1M). The resulting mixture was diluted with water and extracted with DCM (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 70% in 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (AHL-6) (1.4 g,4.67mmol,80%,90% purity) as a yellow oil. M/z 271.0/273.0 (M+H) + (ES+)。
Step 7: synthesis of 2- [ 6-chloro-4- [ (2S) -1- (4-methyl-1, 2, 4-triazol-3-yl) propan-2-yl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AHL-7)
To the product (AHL-6) (150 mg,1 eq, 0.55 mmol), intermediate (AC-2) (45 mg,0.4 eq, 0.22 mmol) and K from step 6 above under nitrogen at room temperature 3 PO 4 (235 mg,2 eq, 1.11 mmol) in 1, 4-dioxane (5 mL) was added Pd (OAc) 2 (12 mg,0.1 eq, 55. Mu. Mol) and XantPhos (64 mg,0.2 eq, 0.11 mmol). The resulting mixture was stirred under nitrogen at 80 ℃ for 1 hour. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (30% MeCN up to 70% in 15 minutes); detector, UV 254/220nm. This gave the sub-title compound (AHL-7) (120 mg,0.25mmol,50%,92% purity) as a yellow oil. M/z 436.1/438.1 (M+H) + (ES+)。
Step 8: synthesis of N- [4- [ (2S) -1- (4-methyl-1, 2, 4-triazol-3-yl) propan-2-yl ] -6- [ 1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-2-yl ] methanesulfonamide (AHL-8)
To the product (AHL-7) (40 mg,1 eq, 92. Mu. Mol), methanesulfonamide (AAW-1) (17 mg,2 eq, 0.18 mmol) and K from step 7 above, under nitrogen at room temperature 3 PO 4 (39 mg,2 eq, 0.18 mmol) to a stirred solution of 1, 4-dioxane (3 mL) was added Pd (OAc) 2 (2.1 mg,0.1 eq, 9. Mu. Mol) and XantPhos (11 mg,0.2 eq, 18. Mu. Mol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (50% MeCN up to 70% within 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: YMC-Actus Triart C18, 30X 250mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 15B to 45B in 7 minutes; 254/210nm, retention time: 6.03 To give the title compound (AHL-8) (31.5 mg, 63. Mu. Mol,69%,99% purity) as a white solid. M/z 495.1 (M+H) + (ES+). 1 H NMR(400MHz,CD3OD)δ8.33(s,1H),8.21–8.11(m,2H),8.00(d,J=7.7Hz,1H),7.79(t,J=7.7Hz,1H),6.67(d,J=1.2Hz,1H),5.30(s,2H),3.66(s,3H),3.47–3.38(m,1H),3.35(s,3H),3.17(d,J=7.7Hz,2H),1.45(d,J=6.9Hz,3H)。
Example 242: synthesis of 2- (3-ethoxy-5- [3- [ (4-methyl-1, 2, 4-triazol-3-yl) methyl ] oxetan-3-yl ] phenyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AHM-12)
Step 1: synthesis of 1-bromo-3-ethoxy-5-nitrobenzene (AHM-2)
To 3-bromo-5-nitrophenol (AHM-1) (6.00 g,1 eq, 27.5 mmol) and K at 0deg.C 2 CO 3 To a stirred solution of (9.51 g,2.5 eq, 68.7 mmol) in DMF (40 mL) was added ethyl iodide (8.59 g,2 eq, 55 mmol) dropwise. The resulting mixture was stirred at room temperature overnight. The resulting mixture was diluted with water and extracted with EtOAc (3×300 mL). The combined organic layers were washed with brine (2×300 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified rapidly by reverse phase to give the sub-title compound (AHM-2) (6.5 g,23.9mmol,96%,90% purity) as a yellow solid. M/z 246.0/248.0 (M+H) + (ES+)。
Step 2: synthesis of 2- (3-ethoxy-5-nitrophenyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (AHM-3)
Pd (dppf) Cl was added to a stirred mixture of the product (AHM-2) (6.00 g,1 eq., 24.4 mmol), bis (pinacolato) diboron (AAI-3) (7.43 g,1.2 eq., 29.3 mmol) and AcOK (5.98 g,2.5 eq., 61.0 mmol) from step 1 above in 1, 4-dioxane (20 mL) at room temperature under nitrogen atmosphere 2 DCM (89mg, 0.05 eq, 1.22 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (10/1) to give the sub-title compound (AHM-3) (8.49 g,25.0mmol, crude product, 73% purity) as a white solid. M/z 294.1 (M+H) + (ES+)。
Step 3: 3-ethoxy-5-nitrophenylboronic acid (AHM-4)
To a stirred solution of the product from step 2 above (AHM-3) (5.80 g,1 eq., 20.0 mmol) in MeCN (60 mL) was added dropwise, at 80℃methyl boronic acid (10.66 g,9 eq., 178 mmol) and TFA (4.06 mL,2.76 eq., 54.7 mmol). The resulting mixture was stirred at 80℃for 4 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×500 mL) of the extract. The combined organic layers were washed with brine (2×500 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (AHM-4) (7.1 g,24.2mmol, crude product, 72% purity) as a pink solid. M/z 212.1 (M+H) + (ES+)。
Step 4: synthesis of ethyl 2- [3- (3-ethoxy-5-nitrophenyl) oxetan-3-yl ] acetate (AHM-6)
To the product (AHM-4) (2.84 g,1 eq, 13.5 mmol) from step 3 above and Pd (COD) Cl under nitrogen at room temperature 2 (380 mg,0.1 eq, 1.35 mmol) in 1, 4-dioxane (100 mL) was added H containing KOH (2.27 g,3 eq, 40.4 mmol) 2 O (20 mL) and ethyl 2- (oxetan-3-ylidene) acetate (AHM-5) (2.30 g,1.2 eq, 16.2 mmol). The resulting mixture was stirred overnight at 40 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (15/1) to give the sub-title compound (AHM-6) (2.28 g,6.42mmol,55%,87% purity) as a brown solid. M/z 310.1 (M+H) + (ES+)。
Step 5: synthesis of [3- (3-ethoxy-5-nitrophenyl) oxetan-3-yl ] acetic acid (AHM-7)
To the product (AHM-6) (830 mg,1 eq, 2.68 mmol) from step 4 above in THF (15 mL) and H under nitrogen at 0deg.C 2 To a stirred solution of O (5 mL) was added LiOH (321 mg,5 eq, 13.4 mmol). The resulting mixture was stirred at room temperature under nitrogen overnight. The resulting mixture was diluted with water (50 mL). The residue was acidified to pH 4 with aqueous HCl (1M). The precipitated solid was collected by filtration and washed with DCM (3×10 mL). This gave the sub-title compound (AHM-7) (704 mg,2.13mmol,93% pure) as a yellow solid. M/z 282.1 (M+H) + (ES+)。
Step 6: synthesis of 2- [3- (3-ethoxy-5-nitrophenyl) oxetan-3-yl ] -N- [ (methylaminomethylsulfonyl) amino ] acetamide (AHM-8)
To a stirred solution of the product from step 6 above (AHM-7) (704 g,1 eq, 2.51 mmol) and 4-methyl-3-thiosemicarbazide (D-2) (316 mg,1.2 eq, 5.02 mmol) in DMF (15 mL) was added HATU (1.05 g,1.1 eq, 2.8 mmol) and DIPEA (1.62 g,5 eq, 12.5 mmol) at room temperature. The resulting mixture was stirred at room temperature for 3 hours. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (20% MeCN up to 60% in 20 minutes); detector, UV 254/220nm to give the sub-title compound (AHM-8) (700 mg,1.75mmol,76%,92% purity) as a yellow solid. M/z 369.1 (M+H) + (ES+)。
Step 7: synthesis of 5- [ [3- (3-ethoxy-5-nitrophenyl) oxetan-3-yl ] methyl ] -4-methyl-1, 2, 4-triazole-3-thiol (AHM-9)
To a stirred mixture of the product from step 6 above (AHM-8) (700 mg,1 eq., 1.90 mmol) in DMF (15 mL) was added NaOH (152 mg,2 eq., 3.80 mmol) at 0deg.C. The resulting mixture was stirred at room temperature overnight. The mixture was acidified to pH 4 with concentrated HCl at 0 ℃. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (AHM-9) (319 mg,1.55mmol,91%,89% purity) as a white solid. M/z 351.1 (M+H) + (ES+)。
Step 8: synthesis of 3- [ [3- (3-ethoxy-5-nitrophenyl) oxetan-3-yl ] methyl ] -4-methyl-1, 2, 4-triazole (AHM-10).
To the product (AHM-9) (550 mg,1 equivalent, 1.57 mmol) from step 7 above and H at 0deg.C 2 O 2 (0.22 mL,30Wt%,6 equivalents, 9.42 mmol) in DCM (10 mL) was added dropwise AcOH (189 mg,2 equivalents, 3.14 mmol). The resulting mixture was stirred at room temperature for 2 hours. The mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH30/1) to give the sub-title compound (AHM-10) (460 mg,1.30mmol,92%,90% purity) as a brown yellow solid. M/z 319.1 (M+H) + (ES+)。
Step 9: synthesis of 3- [ [3- (3-ethoxy-5-nitrophenyl) oxetan-3-yl ] methyl ] -4-methyl-1, 2, 4-triazole (AHM-11).
A mixture of the product from step 8 above (AHM-10) (400 mg,1 eq, 1.26 mmol) and Pd/C form 87L (148 mg,1 eq, 1.26 mmol) in MeOH (30 mL) was stirred overnight at room temperature under a hydrogen atmosphere. Filtering the resulting mixture; the filter cake was washed with MeOH (3X 3 mL). The filtrate was concentrated in vacuo. This gave the sub-title compound (AHM-11) (290 mg,0.86mmol,80%,85% purity) as a brown yellow solid. M/z 289.2 (M+H) + (ES+)。
Step 10: synthesis of 2- (3-ethoxy-5- [3- [ (4-methyl-1, 2, 4-triazol-3-yl) methyl ] oxetan-3-yl ] phenyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AHM-12)
Into a 25-mL round bottom flask at room temperature was placed EtOH (10 mL) containing the product from step 9 above (AHM-11) (50 mg,1 eq, 0.17 mmol) and intermediate (A-3) (52 mg,1 eq, 0.17 mmol) followed by Et 3 N (88 mg,5 eq, 0.87 mmol). The resulting solution was stirred at 80℃for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 60% B in 7 minutes; wavelength: 254/210nm; retention time: 6.32. this gave the title compound (AHM-12) (24.7 mg, 51. Mu. Mol,30%,98% purity) as a white solid. M/z 473.1 (M+H) + (ES+). 1 H NMR(300MHz,MeOH–d4)δ8.22(s,1H),8.08(d,J=7.7Hz,1H),8.01–7.92(m,1H),7.82–7.71(m,1H),7.48(t,J=2.1Hz,1H),6.85(t,J=1.7Hz,1H),6.26–6.19(m,1H),5.11–4.99(m,6H),4.04–3.91(m,2H),3.64(s,2H),2.95(s,3H),1.36(t,J=7.0Hz,3H)。
Example 243: synthesis of 2- (5- (ethylamino) -4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (AHN-9)
Step 1: (6-chloro-4-iodopyridin-3-yl) (ethyl) carbamic acid tert-butyl ester (AHN-2)
To a stirred solution of tert-butyl N- (6-chloro-4-iodopyridin-3-yl) carbamate (AHN-1) (3.55 g,1 eq, 10.0 mmol) in DMF (50 mL) was added NaH (601 mg,60Wt%,1.5 eq, 15.0 mmol) in portions under nitrogen at 0deg.C. The resulting mixture was stirred at room temperature under nitrogen atmosphere for 0.5 hours. Ethyl iodide (2.34 g,1.5 eq, 15.0 mmol) was added dropwise to the above mixture at 0 ℃. The resulting mixture was stirred at room temperature overnight. The reaction was then quenched by addition of 5mL of ice water at 0 ℃. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 80% in 20 minutes); detector, UV 254/220nm. The product-containing fractions were combined and evaporated to give the sub-title compound (AHN-2) (3.0 g,7.07mmol,78%,90% purity) as a brown solid. M/z 383.0/385.0 (M+H) + (ES+)。
Step 2:2- (5- ((tert-Butoxycarbonyl) (ethyl) amino) -2-chloropyridin-4-yl) -5-fluorobenzoic acid (AHN-4)
The product from step 1 above (AHN-2) (1.52 g,1 eq., 3.97 mmol), 4-fluoro-2- (methoxycarbonyl) phenylboronic acid (AHN-3) (1.18 g,1.5 eq., 5.96 mmol) and K were reacted 2 CO 3 A solution of (220 mg,0.4 eq, 1.59 mmol) in DME (32 mL) and water (8 mL) was placed under nitrogen at room temperature. Pd (PPh) was then added at room temperature under a nitrogen atmosphere 3 ) 4 (918 mg,0.2 eq, 0.80 mmol). The resulting mixture was subjected to nitrogen atmosphere at 100℃CStir overnight. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 80% in 20 minutes); detector, UV 254/220nm. The product-containing fractions were combined and evaporated to give the sub-title compound (AHN-4) (520 mg,1.05mmol,32%,92% purity) as a brown solid. M/z 495.1/497.1 (M+H) + (ES+)。
Step 3: (6-chloro-4- (4-fluoro-2- (2- (methylamino-methylsulfonyl) hydrazine-1-carbonyl) phenyl) pyridin-3-yl) (ethyl) carbamic acid tert-butyl ester (AHN-5)
To a stirred solution of the product from step 2 above (AHN-4) (265 mg,1 eq, 0.67 mmol) and 1-amino-3-methyl thiourea (D-2) (106 mg,1.5 eq, 1.01 mmol) in MeCN (10 mL) was added TCFH (377 mg,2 eq, 1.34 mmol) and NMI (4471 mg,8 eq, 5.37 mmol) at 0deg.C. The resulting mixture was stirred at room temperature for 3 hours. The mixture was concentrated in vacuo. The resulting mixture was used directly in the next step without further purification. M/z 482.1/484.1 (M+H) + (ES+)
Step 4: (6-chloro-4- (4-fluoro-2- (5-mercapto-4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-3-yl) (ethyl) carbamic acid tert-butyl ester (AHN-6)
To a stirred solution of the product from step 3 above (AHN-5) (324 mg,1 eq, 0.67 mmol) in MeCN (10 mL) was added water (10 mL) containing NaOH (403 mg,15 eq, 10.1 mmol) at room temperature. The resulting mixture was stirred at 80 ℃ overnight. The mixture was cooled to room temperature and concentrated in vacuo. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo to give the sub-title compound (AHN-6) (320 mg, crude product) as a brown solid. M/z 464.1/466.1 (M+H) + (ES+)。
Step 5: (6-chloro-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-3-yl) (ethyl) carbamic acid tert-butyl ester (AHN-7)
At the temperature of 0 deg.c,to a stirred solution of the product from step 4 above (AHN-6) (290 mg,1 eq, 0.63 mmol) in DCM (20 mL) was added AcOH (75 mg,2 eq, 1.25 mmol) and hydrogen peroxide (284 mg,30Wt%,4 eq, 2.50 mmol). The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And ACN (10% ACN up to 80% within 20 minutes); detector, UV 254/220nm. The product-containing fractions were combined and evaporated to give the sub-title compound (AHN-7) (80 mg,0.19mmol,30%,95% purity) as a brown solid. M/z 432.2/434.2 (M+H) + (ES+)。
Step 6: (4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6- (6- (hydroxymethyl) -1-oxo-4- (trifluoromethyl) isoindolin-2-yl) pyridin-3-yl) carbamic acid tert-butyl ester (AHN-8)
To the product (AHN-7) (40 mg,1 eq, 0.09 mmol), intermediate (C-1) (26 mg,1.2 eq, 0.11 mmol) and Cs from step 5 above under nitrogen at room temperature 2 CO 3 (91 mg,3 eq, 0.28 mmol) to a solution of RuPhos ring palladium complex Gen.3 (16 mg,0.2 eq, 19. Mu. Mol) and RuPhos (17 mg,0.4 eq, 37. Mu. Mol) in 1, 4-dioxane (4 mL) were added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and MeCN (10% MeCN up to 80% in 20 min); detector, UV 254/220nm. The product-containing fractions were combined and evaporated to give the sub-title compound (AHN-8) (35 mg, 56. Mu. Mol,60%,96% purity) as a brown solid. M/z 627.2 (M+H) + (ES+)。
Step 7: synthesis of 2- (5- (ethylamino) -4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (AHN-9)
To a stirred solution of the product from step 6 above (AHN-8) (35 mg,1 eq, 56. Mu. Mol) in DCM (2 mL) at 0deg.C was added TFA (0.4 mL). The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 36% B to 46% B in 9 minutes; wavelength: 254/220nm; retention time: 8.05). The product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AHN-9) (10.9 mg,21 μmol,36%,97.2% purity) as a white solid. M/z 527.2 (M+H) + (ES+). 1 H NMR(300MHz,Methanol-d4)δ8.39(s,1H),7.97(t,J=14.5Hz,3H),7.79(s,1H),7.71–7.63(m,1H),7.55–7.45(m,2H),5.16(s,2H),4.77(s,2H),3.57(s,3H),3.20–3.01(m,2H),1.16(t,J=7.1Hz,3H)。
Example 244: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (2R) -1-methylpyrrolidin-2-yl ] methoxy } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AHO-4)
/>
Step 1: (2R) -2- { [ (2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl) methoxy ] methyl } pyrrolidine-1-carboxylic acid tert-butyl ester (AHO-2)
To a stirred solution of intermediate (AFL-1) (100 mg,1 eq, 0.18 mmol) and tert-butyl (2R) -2- (hydroxymethyl) pyrrolidine-1-carboxylate (AHO-1) (371 mg,10 eq, 1.85 mmol) in DCM (5 mL) was added TBAB (30 mg,0.5 eq, 92 μmol) and aqueous KOH (5 mL,20 wt%) at room temperature. The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The combined organic layers were washed with brine, dried (Na 2 SO 4 ) And concentrated in vacuo. The crude product was taken upPurification by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (50% MeCN up to 75% in 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (AHO-2) (64 mg, 91. Mu. Mol,49%,92% purity) as a white solid. M/z 707.3 (M+H) + (ES+)。
Step 2:2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- { [ (2R) -pyrrolidin-2-ylmethoxy ] methyl } -4- (trifluoromethyl) -3H-isoindol-1-one (AHO-3)
A solution of the product from step 1 above (AHO-2) (60 mg,1 eq, 85. Mu. Mol) and TFA (4 mL) in DCM (4 mL) was stirred at 60℃for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 607.2 (M+H) + (ES+)。
Step 3:2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (2R) -1-methylpyrrolidin-2-yl ] methoxy } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AHO-4)
To a stirred solution of the product from step 2 above (AHO-3) (58 mg,1 eq, 96. Mu. Mol) and formaldehyde (9 mg,1.2 eq, 0.12 mmol) in MeOH (5 mL) at room temperature was added DIPEA (37 mg,3 eq, 0.29 mmol) and NaBH 4 (12 mg,2 equivalents, 0.19 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of 2mL of MeOH at 0 ℃ and then concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1). The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 14% B to 34% B over 10 min; wavelength: 254/220 nm) to give the title compound (AHO-4) as a white solid (2.5 mg,4.0 μmol,4.2%,99.3% purity). M/z 621.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.50(s,1H),8.06–7.92(m,3H),7.75–7.68(m,1H),7.65–7.51(m,2H),6.79(s,1H),5.17(s,2H),4.70(s,2H),3.57–3.49(m,1H),3.41(s,4H),2.96–2.88(m,1H),2.40(t,J=7.0Hz,1H),2.31(s,3H),2.18–2.09(m,1H),2.08–1.99(m,1H),1.95–1.82(m,1H),1.69–1.58(m,2H),1.56–1.45(m,1H),1.01–0.86(m,4H)。
Example 245: synthesis of 4- (2-cyclopropyl-6- (6- (((1-hydroxycyclobutyl) methoxy) methyl) -1-oxo-4- (trifluoromethyl) isoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AHP-1)
To a stirred solution of intermediate (AHF-1) (60 mg,1 eq, 0.11 mmol) and 1- (hydroxymethyl) cyclobutan-1-ol (AED-1) (112 mg,10 eq, 1.09 mmol) in DCM (5 mL) was added TBAB (18 mg,0.5 eq, 54. Mu. Mol) and aqueous KOH (5 mL,20 Wt%) at room temperature. The resulting mixture was stirred slowly at room temperature for 1 hour. The mixture was diluted with water and extracted with EtOAc. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (60% MeCN up to 75% in 10 minutes); detector, UV 254/220nm. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 52% B to 57% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AHP-1) (11.3 mg, 18. Mu. Mol,16.7%, 99.3%) as a white solid. M/z 615.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.25–8.20(m,2H),8.09–7.96(m,3H),7.87(d,J=8.4Hz,1H),6.89(s,1H),5.18–5.08(m,3H),4.76(s,2H),3.53–3.42(m,5H),2.11–2.01(m,3H),1.99–1.88(m,2H),1.73–1.61(m,1H),1.56–1.42(m,1H),1.06–0.90(m,4H)。
Example 246: synthesis of 2- (6-ethoxy-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((2-hydroxyethoxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (AHQ-4)
Step 1: synthesis of 2- (6-ethoxy-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4- (trifluoromethyl) isoindolin-1-one (AHQ-1)
Intermediate (AGQ-1) (100 mg,1 equivalent, 0.30 mmol), intermediate (C-1) (76 mg,1.1 equivalent, 0.33 mmol) and Cs were reacted under nitrogen at room temperature 2 CO 3 (196 mg,2 eq, 0.60 mmol) in dioxane (10 mL) was placed under a nitrogen atmosphere. RuPhos (56 mg,0.4 eq, 0.12 mmol) and RuPhos ring palladium complex Gen.3 (50 mg,0.2 eq, 0.06 mmol) were added to the above mixture under nitrogen at room temperature. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 1 hour. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AHQ-1) (80 mg,0.15mmol,45%,92% purity) as an off-white solid. M/z 528.2 (M+H) + (ES+)。
Step 2:6- (chloromethyl) -2- (6-ethoxy-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AHQ-2)
To a stirred solution of the product from step 1 above (AHQ-1) (30 mg,1 eq., 57. Mu. Mol) in DCM (6 mL) at 0deg.C was added SOCl dropwise 2 (10 mg,1.5 eq, 86. Mu. Mol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of 2mL of MeOH at 0 ℃. The mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AHQ-2) (27 mg,5.0 μmol,87%,95% purity) as a yellow solid. M/z 546.1/548.1 (M+H) + (ES+)。
Step 3:6- ((2- ((tert-Butyldimethylsilyl) oxy) ethoxy) methyl) -2- (6-ethoxy-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AHQ-3)
To the product (AHQ-2) (70 mg,1 eq, 0.13 mmol) from step 2 above and 2- [ (tert-butyldimethylsilyl) oxy]To a stirred solution of ethanol (AGE-1) (27 mg,1.2 eq, 0.15 mmol) in DCM (6 mL) was added TBAB (21 mg,0.5 eq, 64 μmol) and aqueous KOH (6 mL,20 wt%) and the resulting mixture was stirred under nitrogen at room temperature for 2 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo and the crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 50% within 10 minutes); detector, UV 254/220nm to give the sub-title compound (AHQ-3) as a white solid (22 mg,32 μmol,25%,96% purity). M/z 686.3 (M+H) + (ES+)。
Step 4:2- (6-ethoxy-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((2-hydroxyethoxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (AHQ-4)
Et is added dropwise to a stirred solution of the product from step 3 (AHQ-3) (22 mg,1 eq, 32. Mu. Mol) in THF (3 mL) at room temperature 3 N.3HF (6 mg,1.2 eq, 38. Mu. Mol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative HPLC on a column (XBridge prep OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 60% B in 8 minutes; wavelength: 254/220nm; retention time: 7.8 To obtain the target as a white solidThe title compound (AHQ-4) (3.7 mg, 6.5. Mu. Mol,6.3%,98.5% purity). M/z 572.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.51(s,1H),8.05(s,1H),8.00(s,1H),7.80(s,1H),7.71(s,1H),7.64–7.55(m,2H),6.34(s,1H),5.22(s,2H),4.77–4.69(m,3H),4.32–4.29(m,2H),3.63–3.51(m,4H),3.44(s,3H),1.35–1.41(m,3H)。
Example 247: synthesis of (S) -2- (6-cyclopropyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (((1-methylpyrrolidin-2-yl) methoxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (AHR-4)
Step 1: (S) -2- (((2- (6-cyclopropyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -3-oxo-7- (trifluoromethyl) isoindolin-5-yl) methoxy) methyl) pyrrolidine-1-carboxylic acid benzyl ester (AHR-2)
To a stirred solution of intermediate (AFL-1) (60 mg,1 eq, 0.11 mmol) and benzyl (2S) -2- (hydroxymethyl) pyrrolidine-1-carboxylate (AHR-1) (261 mg,10 eq, 1.11 mmol) in DCM (5 mL) was added TBAB (18 mg,0.5 eq, 56 μmol) and aqueous KOH (5 mL,20 wt%) and the resulting mixture stirred at room temperature for 1 hour. The mixture was then diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (55% MeCN up to 60% in 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (AHR-2) (45 mg, 61. Mu. Mol,55%,95% purity) as a yellow solid. M/z 741.3 (M+H) + (ES+)。
Step 2: (S) -2- (6-cyclopropyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((pyrrolidin-2-ylmethoxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (AHR-3)
Will come fromA solution of the product of step 1 (AHR-2) (40 mg,1 eq., 54. Mu. Mol) and TFA (5 mL) in DCM (5 mL) was stirred at 60℃for 4 hours. The mixture was cooled to room temperature and then concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 607.2 (M+H) + (ES+)。
Step 3: (S) -2- (6-cyclopropyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (((1-methylpyrrolidin-2-yl) methoxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (AHR-4)
To a stirred solution of the product from step 2 above (AHR-3) (40 mg,1 eq, 66. Mu. Mol) and formaldehyde (5.9 mg,1.2 eq, 79. Mu. Mol) in MeOH (5 mL) at room temperature was added DIPEA (26 mg,3 eq, 0.20 mmol) and NaBH 4 (8.3 mg,2 equivalents, 0.13 mmol) and the resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of 2mL of MeOH at 0 ℃ and then concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1). The crude product was purified by preparative HPLC on a column (XBridge Shield RP OBD column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35% B to 53% B in 10 minutes; 53% B; wavelength: 254/220 nm) to give the title compound (AHR-4) (7.1 mg, 11. Mu. Mol,17%,99.4% purity) as a white solid. M/z 621.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.50(s,1H),8.05–7.93(m,3H),7.76–7.69(m,1H),7.66–7.55(m,2H),6.79(d,J=1.4Hz,1H),5.17(s,2H),4.70(s,2H),3.57–3.52(m,1H),3.41(s,3H),3.40–3.36(m,1H),2.98–2.88(m,1H),2.46–2.37m,1H),2.31(s,3H),2.21–2.10(m,1H),2.10–1.99(m,1H),1.95–1.85(m,1H),1.72–1.60(m,2H),1.57–1.47(m,1H),1.02–0.83(m,4H)。
Example 248: synthesis of 4- (2-ethoxy-6- (6- (((2-methoxyethyl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AHS-2)
Step 1:4- (2-ethoxy-6- (6-formyl-1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AHS-1)
A solution of intermediate (AKY-1) (20 mg,1 eq, 43. Mu. Mol) and DMP (27 mg,1.5 eq, 65. Mu. Mol) in DCM (8 mL) was stirred at room temperature for 1 hour. The crude product was used directly in the next step without any further purification. M/z 465.2 (M+H) + (ES+)。
Step 2:4- (2-ethoxy-6- (6- (((2-methoxyethyl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AHS-2)
A solution of the product from step 1 above (AHS-1) (20 mg,1 eq, 43. Mu. Mol), 2-methoxyethyl-1-amine (AEB-1) (5 mg,1.5 eq, 65. Mu. Mol) and DIPEA (17 mg,3 eq, 0.13 mmol) in DCM (8 mL) was stirred at room temperature for 2 hours. Adding NaBH to the mixture at 0deg.C 4 (8 mg,5 equivalents, 0.22 mmol). The resulting mixture was stirred at room temperature overnight. The reaction was then quenched by the addition of 2mL of MeOH at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 33% B to 43% B in 9 minutes; wavelength: 254/220nm; retention time: 8.28 To give the title compound (AHS-2) (4.0 mg, 7.6. Mu. Mol,17%,97.2% purity) as a white solid. M/z 524.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.23–8.14(m,2H),7.85(d,J=8.5Hz,1H),7.81(d,J=1.2Hz,1H),7.75(s,1H),7.64(d,J=1.2Hz,2H),6.37(d,J=1.2Hz,1H),5.05(s,2H),4.39–4.31(m,2H),3.82(s,2H),3.48(s,3H),3.40(t,J=5.7Hz,2H),3.24(s,3H),2.63(t,J=5.7Hz,2H),1.35(t,J=7.0Hz,3H)。
Example 249: synthesis of 2- (4- (4-chloro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6-ethoxypyridin-2-yl) -6- ((((1-hydroxycyclobutyl) methyl) amino) methyl) isoindolin-1-one (AHT-2)
Step 1: 2-chloro-4- [ 4-chloro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-ethoxypyridine (AHT-1)
To a 25mL round bottom flask was added EtOH (10 mL) containing intermediate (AGW-4) (200 mg,1 eq, 0.59 mmol) at room temperature. To the above stirred solution was added EtONa (60 mg,1.5 eq, 0.88 mmol) at room temperature. The resulting mixture was stirred at room temperature for an additional 2 hours. The precipitated solid was collected by filtration and washed with toluene (3×10 mL) to give the sub-title compound (AHT-1) (120 mg,0.13mmol,58%,95% purity) as a pale yellow solid. M/z 349.1/351.1 (M+H) + (ES+)
Step 2:2- (4- (4-chloro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6-ethoxypyridin-2-yl) -6- ((((1-hydroxycyclobutyl) methyl) amino) methyl) isoindolin-1-one (AHT-2)
To the product (AHT-1) (100 mg,1 eq, 0.29 mmol), intermediate (AEE-1) (85 mg,1.2 eq, 0.34 mmol) and K from step 1 above under nitrogen at room temperature 2 CO 3 To a stirred mixture of (119 mg,3 eq, 0.86 mmol) in dioxane (10 mL) was added XantPhos (331 mg,2 eq, 0.57 mmol). The resulting mixture was stirred under nitrogen at 60 ℃ for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (5/1). The crude product was purified by preparative HPLC (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 20% B to 50% B in 7 min; wavelength: 254/220nm; retention time: 6.63) to give the title compound (A) as a white solidHT-2) (24 mg, 4.3. Mu. Mol,14%,98.7% purity). M/z 559.1/561.1 (M+H) + (ES+). 1 H NMR(400MHz,Methanol-d4)δ8.53(s,0.858H),8.45(s,1H),7.98–7.65(m,7H),6.40(d,J=1.3Hz,1H),5.10(s,2H),4.43–4.33(m,2H),4.23(s,2H),3.47(s,3H),3.00(s,2H),2.16–2.08(m,4H),1.81–1.69(m,1H),1.61–1.50(m,1H),1.38(t,J=7.0Hz,3H)。
Example 250: synthesis of 2- [6- (ethylamino) -4- [2- (4-methyl-1, 2, 4-triazol-3-yl) -4- (prop-2-yn-1-yloxy) phenyl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AHU-12)
Step 1:2- (2, 6-dichloropyridin-4-yl) -5-hydroxybenzoic acid methyl ester (AHU-2)
To methyl 2-bromo-5-hydroxybenzoate (AHU-1) (1.4 g,1 equivalent, 6.06 mmol), 2, 6-dichloropyridin-4-ylboronic acid (AAN-1) (1.74 g,1.5 equivalent, 9.09 mmol) and K under a nitrogen atmosphere at room temperature 2 CO 3 (2.51 g,3 eq, 18.2 mmol) to a stirred solution of dioxane (20 mL) and water (2 mL) Pd (DtBPF) Cl was added 2 (399mg, 0.1 eq, 0.61 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (3/1) to give the sub-title compound (AHU-2) (1.2 g,4.04mmol,66%,92% purity) as a brown yellow solid. M/z 298.0/300.0 (M+H) + (ES+)
Step 2: methyl 2- (2, 6-dichloropyridin-4-yl) -5- [ (4-methoxyphenyl) methoxy ] benzoate (AHU-3)
To the product (AHU-2) (1.2 g,1 equivalent, 4.03 mmol) from step 1 above and K 2 CO 3 To a stirred solution of (1.67 g,3 eq, 12.1 mmol) in DMF (20 mL) was added PMB-Cl (0.95 g,1.5 eq, 6.04 mmol) and the resulting mixture was stirred at room temperature overnight. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2 x100 mL), and dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (4/1) to give the sub-title compound (AHU-3) (1.3 g,3.12mmol,77%,92% purity) as a yellow oil. M/z 418.1/420.1 (M+H) + (ES+)
Step 3:2- (2, 6-dichloropyridin-4-yl) -5- [ (4-methoxyphenyl) methoxy ] benzoic acid (AHU-4)
To a stirred solution of the product from step 2 above (AHU-3) (1.3 g,1 eq, 3.11 mmol) and LiOH (372 mg,5 eq, 15.5 mmol) in THF (15 mL) and water (5 mL) and the resulting mixture was stirred overnight at 60 ℃. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 50% within 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (AHU-4) (1.1 g,2.72mmol,88%,91% purity) as an off-white solid. M/z 404.0/406.0 (M+H) + (ES+)
Step 4:2- (2, 6-dichloropyridin-4-yl) -5- [ (4-methoxyphenyl) methoxy ] -N- [ (methylaminomethylsulfonyl) amino ] benzamide (AHU-5)
To a stirred solution of the product from step 3 above (AHU-4) (1.1 g,1 eq, 2.72 mmol) and 4-methyl-3-thiosemicarbazide (D-2) (315 mg,1.1 eq, 2.99 mmol) in DMF (15 mL) was added T 3 P (6.93 g,8 eq, 21.8 mmol) and DIPEA (2.11 g,6 eq, 16.3 mmol) and the resulting mixture was stirred at room temperature for 2 hours. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 50% in 15 minutes); detector, UV 254/220nm. This gave the sub-title compound (AHU-5) (480 mg,0.98mmol,36%,89% purity) as an off-white solid. M/z 491.1/493.1 (M+H) + (ES+)
Step 5:5- [2- (2, 6-dichloropyridin-4-yl) -5- [ (4-methoxyphenyl) methoxy ] phenyl ] -4-methyl-1, 2, 4-triazole-3-thiol (AHU-6)
A solution of the product from step 4 above (AHU-5) (480 mg,1 eq., 0.98 mmol) in aqueous NaOH (10 mL, 1M) was stirred at 80℃for 2 hours at room temperature. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 50% in 30 minutes); detector, UV 254/220nm. This gave the sub-title compound (AHU-6) (360 mg,0.76mmol,78% pure) as an off-white solid. M/z473.1/475.1 (M+H) + (ES+)
Step 6:2, 6-dichloro-4- {4- [ (4-methoxyphenyl) methoxy ] -2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl } pyridine (AHU-7)
The product from step 5 above (AHU-6) (350 mg,1 eq., 0.74 mmol) and AcOH (89 mg,2 eq., 1.48 mmol), H 2 O 2 A solution of (126 mg,30Wt%,5 eq, 3.70 mmol) in DCM (10 mL) was stirred at room temperature for 2 hours. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 50% in 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (AHU-7) (230 mg,0.52mmol,70%,95% purity) as an off-white solid. M/z 441.1/443.1 (M+H) + (ES+)
Step 7: 6-chloro-N-ethyl-4- {4- [ (4-methoxyphenyl) methoxy ] -2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl } pyridin-2-amine (AHU-8)
The product from step 6 above (AHU-7) (220 mg,1 eq., 0.50 mmol) and ethylamine, HCl (31 mg,30 eq., 0.69 mmol), K 2 CO 3 (2.07 g,30 equivalents, 150 mmol) in NMP (15 mL) was stirred at 120℃for 3 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 50% in 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (AHU-8) (170 mg, 0) as an off-white solid.38mmol,76%,95% purity). M/z 450.2/452.2 (M+H) + (ES+)
Step 8:2- [6- (ethylamino) -4- {4- [ (4-methoxyphenyl) methoxy ] -2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl } pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AHU-9)
To the product (AHU-8) (120 mg,0.27mmol,1 eq.) from step 7 above, intermediate (AC-2) (59 mg,1.1 eq., 0.29 mmol) and Cs under nitrogen at room temperature 2 CO 3 (261 mg,3 eq, 0.80 mmol) in DMF (10 mL) was added Pd 2 (dba) 3 (24 mg,0.1 eq, 27. Mu. Mol) and RuPhos (12 mg,0.1 eq, 27. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (30/1) to give the sub-title compound (AHU-9) (90 mg,0.15mmol,55%,92% purity) as an off-white solid. M/z 615.2 (M+H) + (ES+)
Step 9:2- [6- (ethylamino) -4- [ 4-hydroxy-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AHU-10)
A solution of the product from step 8 above (AHU-9) (90 mg,1 eq, 0.15 mmol) and TFA (1 mL) in DCM (3 mL) was stirred at room temperature for 2 hours and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1) to give the sub-title compound (AHU-10) (50 mg,0.10mmol,69%,90% purity) as an off-white solid. M/z 495.2 (M+H) + (ES+)
Step 10:2- [6- (ethylamino) -4- [2- (4-methyl-1, 2, 4-triazol-3-yl) -4- (prop-2-yn-1-yloxy) phenyl ] pyridin-2-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AHU-12)
To the product (AHU-10) (50 mg,1 eq., 0.10 mmol) from step 9 above and K at room temperature 2 CO 3 To a stirred solution of (42 mg,3 eq, 0.30 mmol) in DMF (5 mL) was added propargyl bromide (AHU-11) (18 mg,1.5 eq, 0.15 mmol). The resulting mixture was stirred at room temperature overnight. The resulting mixture was diluted with water and treated with DCM/MeOH (10/1)3x20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1). The product was purified by preparative HPLC on a column (XBridge prep OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 54% B to 58% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AHU-12) (7.0 mg, 13. Mu. Mol,13%,99.5% purity) as an off-white solid. M/z 533.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.47(s,1H),8.10–8.00(m,2H),7.79(d,J=7.7Hz,1H),7.57–7.44(m,2H),7.34–7.28(m,1H),7.17(d,J=2.7Hz,1H),6.65(t,J=5.4Hz,1H),5.89(d,J=1.3Hz,1H),5.18(s,2H),4.94(d,J=2.4Hz,2H),3.65(t,J=2.3Hz,1H),3.35(s,3H),3.20–3.12(m,2H),1.12(t,J=7.2Hz,3H)。
Example 251: synthesis of 4- { 2-cyclopropyl-6- [6- (hydroxymethyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AHV-1)
To intermediate (AEG-2) (60 mg,1 equivalent, 0.18 mmol), intermediate (AGH-1) (38 mg,1.3 equivalent, 0.23 mmol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 (116 mg,2 eq, 0.36 mmol) to a stirred mixture of 1, 4-dioxane was added RuPhos ring palladium complex gen.3 (30 mg,0.2 eq, 36. Mu. Mol) and RuPhos (33 mg,0.4 eq, 72. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 40% B in 10 minutes; wave-guide And (3) length: 254/220nm; retention time: 9.67 To give the title compound (AHV-1) (2.4 mg,2.84%,99.0% purity) as a white solid. M/z 463.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.02 (m, 3H), 7.92 (d, j=8.1 hz, 1H), 7.83 (s, 1H), 7.70-7.57 (m, 2H), 6.92 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 4.71 (s, 2H), 3.50 (s, 3H), 2.07-1.99 (m, 1H), 1.07-0.89 (m, 4H).
Example 252: synthesis of 2- {4- [ 4-chloro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-ethoxypyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AHW-1)
To intermediate (AHT-1) (50 mg,1 eq, 0.14 mmol), intermediate (ADR-2) (54 mg,1.2 eq, 0.17 mmol) and K under nitrogen at room temperature 3 PO 4 (61 mg,2 eq, 0.29 mmol) in dioxane (3 mL) was added in portions Pd (OAc) 2 (3 mg,0.1 eq, 14. Mu. Mol) and XantPhos (17 mg,0.2 eq, 29. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 11% B to 31% B; wavelength: 254/220nm; retention time: 11.3) to give the title compound (AHW-1) as a white solid (17.4 mg,28 μmol,19%,99.8% purity). M/z 627.2/629.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.45 (s, 1H), 8.13 (d, j=26.3 hz, 2H), 7.85-7.68 (m, 4H), 6.47-6.41 (m, 1H), 5.25 (d, j=3.2 hz, 2H), 4.43-4.33 (m, 2H), 4.24 (d, j=6.4 hz, 2H), 3.47 (s, 3H), 2.96 (d, j=5.8 hz, 2H), 2.12 (t, j=9.6 hz, 4H), 1.81-1.71 (m, 1H), 1.62-1.50 (m, 1H), 1.40 (t, j=7.0 hz, 3H).
Example 253: synthesis of 2- (6-cyclopropyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((2-hydroxyethoxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (AHX-2)
Step 1:6- ((2- ((tert-Butyldimethylsilyl) oxy) ethoxy) methyl) -2- (6-cyclopropyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -4- (trifluoromethyl) isoindolin-1-one (AHX-1)
To a solution of intermediate (AFL-1) (20 mg,1 eq, 37 μmol), 2- ((tert-butyldimethylsilyl) oxy) ethan-1-ol (AGE-1) (8 mg,1.2 eq, 44 μmol) in DCM (2 mL) was added TBAB (6 mg,0.5 eq, 18 μmol) and aqueous KOH (2 mL,20 wt%) at room temperature. The resulting mixture was stirred at room temperature under nitrogen atmosphere for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1) to give the sub-title compound (AHX-1) (12 mg, 18. Mu. Mol,48%,95% purity) as a white solid. M/z 682.3 (M+H) + (ES+)。
Example 254: synthesis of 2- (6-cyclopropyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((2-hydroxyethoxy) methyl) -4- (trifluoromethyl) isoindolin-1-one (AHX-2).
Et is added dropwise to a stirred solution of the product (AHX-1) (23 mg,1 eq, 34. Mu. Mol) from step 1 above in THF (3 mL) at room temperature 3 N.3HF (7 mg,1.2 eq, 41. Mu. Mol). The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: sunfire preparative C18 column,30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35% B to 62% B in 10 minutes; wavelength: 254/220nm; retention time: 10.58 To give the title compound (AHX-2) (2.8 mg, 4.9. Mu. Mol,15%,97.9% purity) as a white solid. M/z 568.2 (M+H) + (ES+)。 1 H NMR (300 MHz, methanol-d 4) delta 8.48 (s, 1H), 8.12-7.97 (m, 3H), 7.81-7.74 (m, 1H), 7.58-7.41 (m, 2H), 6.89 (d, J=1.4 Hz, 1H), 5.18 (s, 2H), 4.75 (s, 2H), 3.78-3.72 (m, 2H), 3.69-3.62 (m, 2H), 3.47 (s, 3H), 2.09-1.97 (m, 1H), 1.00 (d, J=6.4 Hz, 4H).
Example 255: synthesis of 2- {7- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -1H-indol-5-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (AHY-5)
Step 1: 7-bromo-1H-indol-5-amine (AHY-2)
To a 40mL sealed tube was added THF (8 mL) containing 7-bromo-5-nitro-1H-indole (AHY-1) (1.00 g,1 eq., 4.15 mmol) and AcOH (2.49 g,10 eq., 41.5 mmol) at room temperature, and iron powder (1.16 g,5 eq., 20.7 mmol) at 0deg.C. The resulting mixture was stirred at room temperature overnight. The resulting mixture was filtered and the filter cake was washed with EtOAc (3×15 mL). The filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 50% within 10 minutes); detector, UV 254/220nm, to give the sub-title compound (AHY-2) (640 mg,3.19mmol,77%,92% purity) as a yellow oil. M/z 211.0/213.0 (M+H) + (ES+)
Step 2:2- (7-bromo-1H-indol-5-yl) -4- (trifluoromethyl) -3H-isoindol-1-one (AHY-3)
To an 8mL sealed tube at room temperature was added a solution containing the product (AHY-2) (100 mg,1 equivalent, 0.47 mmol) from step 1 above and Et 3 N (144 mg,3 eq, 1.42 mmol) EtOH (6 mL). At room temperature, adding into the above mixtureIntermediate (A-3) (211 mg,1.5 eq, 0.71 mmol) was added. The resulting mixture was stirred at 80 ℃ overnight. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 60% in 15 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AHY-3) (86 mg,0.22mmol,46%,95% purity) as a yellow solid. M/z 395.0/397.0 (M+H) + (ES+)
Step 3:2- [7- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-indol-5-yl ] -4- (trifluoromethyl) -3H-isoindol-1-one (AHY-4)
1, 4-dioxane (2 mL) containing the product from step 2 above (AHY-3) (50 mg,1 eq, 0.13 mmol), bis (pinacolato) diboron (AAI-3) (64 mg,2 eq, 0.25 mmol) and KOAc (25 mg,2 eq, 0.25 mmol) was added to an 8mL sealed tube at room temperature under nitrogen atmosphere. Pd (dppf) Cl was added to the above mixture at room temperature under nitrogen atmosphere 2 DCM (19 mg,0.2 eq, 25. Mu. Mol). The resulting mixture was stirred under nitrogen at 110 ℃ for an additional 2 hours. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 50% in 12 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (AHY-4) (35 mg, 79. Mu. Mol,63%,95% purity) as a white solid. M/z 443.2 (M+H) + (ES+)
Step 4:2- {7- [2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -1H-indol-5-yl } -4- (trifluoromethyl) -3H-isoindol-1-one (AHY-5)
To an 8mL sealed tube under nitrogen at room temperature was added a solution containing the product (AHY-4) (30 mg,1 equivalent, 68. Mu. Mol), intermediate (D-5) (24 mg,1.5 equivalent, 0.10 mmol) and K from step 3 above 2 CO 3 (13 mg,2 eq, 0.14 mmol) of 1,4-Dioxane (5 mL). Pd (dppf) Cl was added to the above mixture at room temperature under nitrogen atmosphere 2 DCM (10 mg,0.2 eq., 14. Mu. Mol). The resulting mixture was stirred under nitrogen at 80 ℃ for an additional 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: sunFire preparation type C18 OBD column, 19X 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30B to 60B in 7 minutes; a detector, UV 254/210nm; retention time: 6.08 to give the title compound (AHY-5) (3.0 mg, 6.3. Mu. Mol,9.3%,99.4% purity) as a white solid. M/z 474.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.24 (s, 1H), 8.07 (d, j=7.6 hz, 1H), 7.97-7.90 (m, 2H), 7.86-7.63 (m, 5H), 7.31 (d, j=3.2 hz, 1H), 7.03 (d, j=2.0 hz, 1H), 6.57 (d, j=3.2 hz, 1H), 5.00 (s, 2H), 3.01 (s, 3H).
Example 256: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (oxetan-3-ylmethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AHZ-2)
A solution of intermediate (AGT-2) (20 mg,1 eq, 43. Mu. Mol) and 1- (oxetan-3-yl) methylamine (AHZ-1) (6 mg,1.5 eq, 65. Mu. Mol) in MeOH (6 mL) was stirred at room temperature for 1 hour. Adding NaBH to the mixture at 0deg.C 4 (4 mg,2 equivalents, 86. Mu. Mol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of 2mL of MeOH at 0 ℃. The mixture was acidified with FA to pH 6 and then concentrated in vacuo. The crude product was purified by preparative HPLC (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 11% B to 25% B in 10 min; wavelength: 254/220 nm) to give the title as a white solid Compound (AHZ-2) (3.2 mg, 6.0. Mu. Mol,14%,98.7% purity). M/z 532.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.31–8.19(m,2H),8.06–7.99(m,1H),7.95–7.85(m,1H),7.74–7.63(m,3H),6.87(d,J=1.4Hz,1H),5.00(s,2H),4.66–4.56(m,2H),4.24(t,J=5.9Hz,2H),3.79(s,2H),3.47(s,3H),3.09–3.00(m,1H),2.76(d,J=7.4Hz,2H),2.11–2.01(m,1H),1.03–0.92(m,4H)。
Example 257: synthesis of 4- [2- (6- { [ (cyclobutylmethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIA-2)
A solution of 1-cyclobutylmethylamine, HCl (AIA-1) (7 mg,1.5 eq, 58. Mu. Mol) and DIPEA (6 mg,1.2 eq, 47. Mu. Mol) in MeOH (5 mL) was stirred at room temperature for 10 min. To the above mixture was added intermediate (AGT-2) (18 mg,1 eq, 39. Mu. Mol) at room temperature. The resulting mixture was stirred at room temperature overnight. Adding NaBH to the mixture at 0deg.C 4 (3 mg,2 equivalents, 78. Mu. Mol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of 2mL of MeOH at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 14% B to 34% B over 7 min; wavelength: 254/220 nm) to give the title compound (AIA-2) as a white solid (6.3 mg,12 μmol,30%,99.6% purity). M/z 530.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.23–8.19(m,2H),7.99(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.75(s,1H),7.64(d,J=1.2Hz,2H),6.87(d,J=1.4Hz,1H),5.00(s,2H),3.81(s,2H),3.47(s,3H),2.54(d,J=7.1Hz,2H),2.45–2.40(m,1H),2.06–1.96(m,3H),1.86–1.76(m,2H),1.69–1.60(m,2H),1.01–0.93(m,4H)。
Example 258: synthesis of 4- (2-cyclopropyl-6- (6- (((cyclopropylmethyl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AIB-2)
A solution of intermediate (AGT-2) (20 mg,1 eq, 43. Mu. Mol) and 1-cyclopropylmethylamine (AIB-1) (4.6 mg,1.5 eq, 65. Mu. Mol) in MeOH (8 mL) was stirred at 60℃for 1 hour. Adding NaBH to the mixture at 0deg.C 4 (3.3 mg,2 equivalents, 86. Mu. Mol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of 2mL of MeOH at 0 ℃. The mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (9/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 47% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AIB-2) (0.9 mg, 1.7. Mu. Mol,3.9%,97.8% purity) as a white solid. M/z 516.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.48 (s, 1H), 8.14-8.05 (m, 3H), 7.92 (d, j=8.1 hz, 1H), 7.80 (s, 1H), 7.69-7.60 (m, 2H), 6.90 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 3.90 (s, 2H), 3.49 (s, 3H), 2.46 (d, j=6.9 hz, 2H), 2.05-1.99 (m, 1H), 1.06-0.96 (m, 5H), 0.53-0.49 (m, 2H), 0.16-0.12 (m, 2H).
Example 259: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- { [ (1-hydroxycyclobutyl) methoxy ] methyl } -4-methoxy-3H-isoindol-1-one (AIC-11)
Step 1: 5-bromo-3-hydroxy-2-methylbenzoic acid methyl ester (AIC-2)
At 0℃to 3-ammoniaMethyl-5-bromo-2-methylbenzoate (AIC-1) (10.0 g,1 eq, 41.0 mmol) in concentrated H 2 SO 4 NaNO was added in portions to the stirred solution in (30 mL) 2 (3.39 g,1.2 eq, 49.2 mmol). The resulting mixture was stirred at room temperature overnight. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (8/1) to give the sub-title compound (AIC-2) (6.0 g,23.7mmol,54%,90% purity) as a brown yellow oil. M/z 245.0/247.0 (M+H) + (ES+)。
Step 2: 5-bromo-3-methoxy-2-methylbenzoic acid methyl ester (AIC-3)
To the product (AIC-2) (6.00 g,1 equivalent, 24.5 mmol) from step 1 above and K under a nitrogen atmosphere at room temperature 2 CO 3 (10.2 g,3 eq, 73.4 mmol) in DMF (20 mL) was added dropwise CH 3 I (5.21 g,1.5 eq, 36.7 mmol). The resulting mixture was stirred overnight at 60 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (5/1) to give the sub-title compound (AIC-3) (5.0 g,19.4mmol,73%,92% purity) as a light brown oil. M/z 259.0/261.0 (M+H) + (ES+)
Step 3: 5-bromo-2- (bromomethyl) -3-methoxybenzoic acid methyl ester (AIC-4)
To the product (AIC-3) (5.00 g,1 eq., 19.3 mmol) from step 2 above and NBS (3.78 g,1.1 eq., 21.2 mmol) in CCl under nitrogen at room temperature 4 BPO (495mg, 0.1 eq, 1.93 mmol) was added to the stirred solution in (20 mL). The resulting mixture was stirred overnight at 80 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layersWashed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 338.9/340.9 (M+H) + (ES+)
Step 4: 6-bromo-4-methoxy-2, 3-dihydro-isoindol-1-one (AIC-5)
The product (AIC-4) (5.00 g,1 eq., 14.8 mmol) from step 3 above was reacted with NH 3 A solution in MeOH (20 mL, 7M) was stirred at room temperature for 3 hours. The resulting mixture was concentrated in vacuo. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z242.0/244.0 (M+H) + (ES+)
Step 5: 6-vinyl-4-methoxy-2, 3-dihydro-isoindol-1-one (AIC-6)
To a stirred solution of the product (AIC-5) (6.50 g,1 equivalent, 26.9 mmol) from step 4 above and tributyl (vinyl) stannane (10.2 g,1.2 equivalent, 32.2 mmol) in dioxane (30 mL) was added CsF (12.2 g,3 equivalent, 80.6 mmol) at room temperature under nitrogen atmosphere. Pd (PPh) was added to the above mixture at room temperature under a nitrogen atmosphere 3 ) 2 Cl 2 (1.88 g,0.1 eq, 2.69 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (1/1) to give the sub-title compound (AIC-6) (3.1 g,16.4mmol,56%,91% purity) as an off-white solid. M/z 190.1 (M+H) + (ES+)
Step 6: 7-methoxy-3-oxo-1, 2-dihydro-isoindole-5-carbaldehyde (AIC-7)
To a stirred solution of the product (AIC-6) (3.00 g,1 equivalent, 15.9 mmol) from step 5 above and citric acid (3.96 g,1.3 equivalent, 20.6 mmol) in t-BuOH (20 mL) at room temperature was added NMO (2.41 g,1.3 equivalent, 20.6 mmol) and K 2 OsO 4 .2H 2 O (284 mg,0.1 eq, 1.59 mmol). The resulting mixture was stirred at room temperature for 2 hours. Adding NaIO to the mixture at 0deg.C 4 (4.75 g,2 equivalents, 31.7 mmol). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (1/1) to give the sub-title compound (AIC-7) (1.8 g,9.42mmol,53%,95% purity) as an off-white solid. M/z 192.1 (M+H) + (ES+)
Step 7:6- (hydroxymethyl) -4-methoxy-2, 3-dihydro-isoindol-1-one (AIC-8)
To a stirred solution of the product from step 6 above (AIC-7) (1.8 g,9.42mmol,1 eq.) in MeOH (20 mL) at 0deg.C under nitrogen was added NaBH in portions 4 (1.07 g,3 equivalents, 28.2 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. The reaction was then quenched by the addition of 10mL of MeOH at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 30% in 15 minutes); detector, UV 254/220nm. This gave the sub-title compound (AIC-8) (480 mg,5.08mmol,51%,95% purity) as an off-white solid. M/z 194.1 (M+H) + (ES+)
Step 8:2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- (hydroxymethyl) -4-methoxy-3H-isoindol-1-one (AIC-9)
To the product (AIC-8) (107 mg,1.1 eq., 0.55 mmol), intermediate (ADN-1) (165 mg,1 eq., 0.50 mmol) and Cs from step 7 above, at room temperature under a nitrogen atmosphere 2 CO 3 (327 mg,2 eq, 1.00 mmol) to a stirred solution of dioxane (10 mL) was added Ruphos (94 mg,0.4 eq, 0.20 mmol) and Ruphos ring palladium complex Gen.3 (84 mg,0.2 eq, 0.10 mmol). The mixture was subjected to nitrogenStirring is carried out for 2 hours at 100℃under an atmosphere. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1) to give the sub-title compound (AIC-9) (101 mg,0.21mmol,41% pure) as a yellow solid. M/z 486.2 (M+H) + (ES+)。
Step 9:6- (chloromethyl) -2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -4-methoxy-3H-isoindol-1-one (AIC-10)
To a stirred solution of the product from step 8 above (AIC-9) (98 mg,1 eq, 0.20 mmol) in DCM (10 mL) at 0deg.C was added SOCl dropwise 2 (72 mg,3 equivalents, 0.61 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of 2mL of MeOH at 0 ℃ and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1) to give the sub-title compound (AIC-10) (76 mg,0.15mmol,75%,95% pure) as a yellow solid. M/z 504.2/506.2 (M+H) + (ES+)。
Step 10:2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- { [ (1-hydroxycyclobutyl) methoxy ] methyl } -4-methoxy-3H-isoindol-1-one (AIC-11)
To a stirred solution of the product (AIC-10) (50 mg,1 eq, 99. Mu. Mol) from step 8 above and 1- (hydroxymethyl) cyclobutan-1-ol (AED-1) (101 mg,10 eq, 0.99 mmol) in DCM (5 mL) was added TBAB (16 mg,0.5 eq, 0.05 mmol) and aqueous KOH (5 mL,20 wt%) at room temperature. The resulting mixture was stirred slowly at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1). The crude product was purified by preparative HPLC under the following conditions (column: XBridge Shield RP18 OBD column, 19 x 250mm,10 μm; mobile phase a:water (0.1% NH) 4 HCO 3 ) Mobile phase B: meOH-HPLC; flow rate: 25 ml/min; gradient: 63% B to 65% B in 17 minutes; wavelength: 254/220 nm) to give the title compound (AIC-11) (3.1 mg, 5.4. Mu. Mol,5.4%,98.2% purity) as a white solid. M/z 570.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.50(s,1H),7.96(d,J=1.4Hz,1H),7.73–7.68(m,1H),7.64–7.56(m,2H),7.38–7.27(m,2H),6.80(d,J=1.4Hz,1H),5.02(s,1H),4.90(s,2H),4.66(s,2H),3.93(s,3H),3.42(d,J=5.6Hz,5H),2.09–2.00(m,3H),1.98–1.87(m,2H),1.72–1.62(m,1H),1.53–1.43(m,1H),0.95(d,J=6.4Hz,4H)。
Example 260: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (2R) -2-methoxypropyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AID-2)
A solution of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and (2R) -2-methoxypropan-1-amine (AID-1) (8.7 mg,1.5 eq, 98. Mu. Mol) in MeOH (5 mL) was stirred at 60℃for 1 hour. Adding NaBH to the mixture at 0deg.C 4 (4.9 mg,2 equivalents, 0.13 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃ and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1). The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 5% B to 35% B over 7 min; wavelength: 254/220 nm) to give the title compound (AID-2) as a white solid (14.7 mg,28 μmol,42%,99.8% purity). M/z 534.3 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.54(d,J=2.0Hz,1H),8.24–8.19(m,2H),7.99(d,J=2.0Hz,1H),7.92–7.84(m,1H),7.73(s,1H),7.63(s,2H),6.88(d,J=2.0Hz,1H),5.00(s,2H),3.81(s,2H),3.48(d,J=1.9Hz,3H),3.42–3.38(m,1H),3.23(d,J=2.0Hz,3H)2.49–2.42(m,2H),2.08–2.00(m,1H),1.13–1.06(m,3H),1.00–0.95(m,4H)。
Example 261: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (2-hydroxy-2-methylpropyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIE-1)
To a stirred solution of intermediate (AGT-2) (30 mg,1 eq, 0.06 mmol) and 1-amino-2-methylpropan-2-ol (AZ-1) (5.81 mg,1 eq, 0.06 mmol) in MeOH (5 mL) at 60℃under nitrogen was added Et 3 N (37 mg,5 eq, 0.33 mmol) for 2 hours. Adding NaBH to the mixture at 0deg.C 4 (25 mg,10 equivalents, 0.65 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was diluted with water and extracted with DCM (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 19×250mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 25 ml/min; gradient: 32B to 42B within 10 minutes; a detector, UV 254/210nm; retention time: 9.12. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AIE-1) (13.6 mg,26 μmol,39%,98.6% purity) as a white solid. M/z 534.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.07 (m, 2H), 8.06 (d, j=1.4 hz, 1H), 7.94-7.82 (m, 2H), 7.72-7.66 (m, 1H), 7.61 (d, j=7.8 hz, 1H), 6.92 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 3.91 (s, 2H), 3.50 (s, 3H), 2.54 (s, 2H), 2.06-2.01 (m, 1H), 1.21 (s, 6H), 1.07-1.01 (m, 2H), 1.01-0.95 (m, 2H).
Example 262: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (1-hydroxycyclopentyl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIF-1)
A solution of intermediate (AIG-1) (30 mg,1 eq, 65. Mu. Mol) and 1- (aminomethyl) cyclopentan-1-ol (AFQ-1) (11 mg,1.5 eq, 98. Mu. Mol) in MeOH (6 mL) was stirred overnight at room temperature under a nitrogen atmosphere. NaBH was added to the above mixture at 0deg.C under nitrogen atmosphere 4 (12.32 mg,0.325mmol,5 eq.). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 16% B to 30% B over 10 min; wavelength: 254/220nm; retention time: 9.4) to give the title compound (AIF-1) (9.1 mg,16 μmol,24%,98.0% purity) as a white solid. M/z 560.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.28–8.19(m,2H),7.99(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.75(s,1H),7.65(d,J=1.8Hz,2H),6.88(d,J=1.4Hz,1H),5.00(s,2H),3.85(s,3H),3.48(s,4H),2.09–2.00(m,1H),1.71–1.61(m,2H),1.61–1.29(m,6H),1.03–0.79(m,4H)。
Example 263: synthesis of 4- [2- (6- { [ (cyclopentylmethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIG-1)
Et is added to a stirred solution of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and 1-cyclopentylmethylamine (AHH-1) (6.5 mg,1 eq, 65. Mu. Mol) in MeOH (5 mL) under a nitrogen atmosphere at 60 ℃ 3 N(33mg,5 equivalents, 0.33 mmol) for 2 hours. Adding NaBH to the mixture at 0deg.C 4 (25 mg,10 equivalents, 0.65 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was diluted with water and extracted with DCM (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 19×250mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 48B to 58B in 9 minutes; a detector, UV 254/210nm; retention time: 8.6. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AIG-1) (13.5 mg,25 μmol,38%,99.7% purity) as a white solid. M/z 544.4 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.15-8.03 (m, 3H), 7.92-7.81 (m, 2H), 7.70-7.60 (m, 2H), 6.92 (d, J=1.5 Hz, 1H), 5.03 (s, 2H), 3.89 (s, 2H), 3.50 (s, 3H), 2.54 (d, J=7.3 Hz, 2H), 2.12-2.00 (m, 2H), 1.87-1.77 (m, 2H), 1.67-1.49 (m, 4H), 1.22-1.13 (m, 2H), 1.09-0.92 (m, 4H).
Example 264: synthesis of 4- (2-cyclopropyl-6- (5-fluoro-6- (((2-methoxyethyl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AIH-2)
Step 1: 5-fluoro-6- { [ (2-methoxyethyl) amino ] methyl } -2, 3-dihydro-isoindol-1-one (AIH-1)
To a solution of intermediate (AEI-3) (200 mg,1.12mmol,1 eq.) and 2-methoxyethyl-1-amine (AEB-1) (101 mg,1.2 eq., 1.34 mmol) in MeOH (10 mL) was added AcOH (134 mg,2 eq., 2.23 mmol) at room temperature. The resulting mixture was stirred at room temperature for 1 hour. Then NaBH was added at room temperature 3 CN (140 mg,2 equivalents, 2.23 mmol). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was concentrated in vacuo. The resulting mixture was extracted with DCM (3X 50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AIH-1) (200 mg,0.84mmol,63%,95% pure) as a yellow oil. M/z 239.1 (M+H) + (ES+)。
Step 2:4- (2-cyclopropyl-6- (5-fluoro-6- (((2-methoxyethyl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AIH-2)
Intermediate (AEG-2) (59 mg,1 eq., 0.18 mmol), product (AIH-1) from step 1 above (50 mg,1.2 eq., 0.21 mmol) and Cs were taken up 2 CO 3 A solution of (171 mg,3 eq, 0.53 mmol) in 1, 4-dioxane (10 mL) was stirred at room temperature under nitrogen. Ephos (9.4 mg,0.1 eq, 18. Mu. Mol) and Ephos Pd G4 (16 mg,0.1 eq, 18. Mu. Mol) were added to the above mixture under a nitrogen atmosphere at room temperature. The resulting mixture was stirred under nitrogen at 60 ℃ for an additional 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC (column: XBridge preparative OBD C18 column, 30 x 150mm,5 μm; mobile phase A: water (0.1% NH4HCO3+0.1% NH3.H2O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 37% B to 47% B;47% B; wavelength: 254/220 nm) under the following conditions to give the title compound (AIH-2) (3.1 mg, 5.8. Mu. Mol,3.2%,96.1% purity) as a white solid. M/z 538.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.22–8.19(m,2H),7.95(d,J=1.4Hz,1H),7.88–7.85(m,2H),7.53(d,J=9.5Hz,1H),6.89(d,J=1.4Hz,1H),5.00(s,2H),3.82(s,2H),3.48(s,3H),3.40(t,J=5.6Hz,2H),3.24(s,3H),2.67(t,J=5.6Hz,2H),2.07–2.01(m,1H),0.97(d,J=6.4Hz,4H)。
Example 265: synthesis of 2- { 6-cyclopropyl-4- [2, 4-difluoro-6- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AII-9)
Step 1: 2-bromo-3, 5-difluorobenzoic acid methyl ester (AII-2)
To a stirred mixture of 2-bromo-3, 5-difluorobenzoic acid (AII-1) (2.00 g,1 eq., 8.44 mmol) in MeOH (30 mL) at 0deg.C was added concentrated H 2 SO 4 (10 mL). The resulting mixture was stirred at 70℃for 12 hours. The mixture was cooled to room temperature. The reaction was then quenched by addition of 50mL of ice water at 0 ℃. The residue was taken up with NaHCO 3 Is alkalized to pH 7. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (5/1) to give the sub-title compound (AII-2) (1.1 g,4.4mmol,52%,90% purity) as a yellow solid. M/z 250.9/252.9 (M+H) + (ES+)
Step 2:2- (2, 6-dichloropyridin-4-yl) -3, 5-difluorobenzoic acid methyl ester (AII-3)
To the product (AII-2) (600 mg,1 equivalent, 2.39 mmol), 2, 6-dichloropyridin-4-ylboronic acid (AAN-1) (550 mg,1.2 equivalent, 2.87 mmol) and K from step 1 above were added under a nitrogen atmosphere at room temperature 2 CO 3 (991 mg,7.17mmol,3 eq.) Pd (DtBPF) Cl was added to a stirred mixture in dioxane (20 mL) 2 (156 mg,0.1 eq, 0.24 mmol). The resulting mixture was stirred under nitrogen at 80 ℃ for 4 hours. The mixture was cooled to room temperature and concentrated in vacuo. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phaseWater (0.1% NH) 4 HCO 3 ) And MeCN (10% ACN up to 80% in 10 minutes); detector, UV 254/220nm. This gave the sub-title compound (AII-3) (300 mg,0.95mmol,39%,92% purity) as a yellow solid. M/z 318.0/320.0 (M+H) + (ES+)
Step 3:2- (2, 6-dichloropyridin-4-yl) -3, 5-difluorobenzoic acid (AII-4)
To the product (AII-3) (990 mg,1 eq, 3.11 mmol) from step 2 above in THF (10 mL) and H at room temperature 2 To a stirred mixture in O (2 mL) was added LiOH (2.98 g,40 eq, 124 mmol). The resulting mixture was stirred at 80℃for 12 hours. The mixture was cooled to room temperature. The residue was acidified to pH 4 with concentrated HCl. The mixture was concentrated in vacuo. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The resulting mixture was used directly in the next step without further purification. M/z 304.0/306.0 (M+H) + (ES+)
Step 4:2- (2, 6-dichloropyridin-4-yl) -3, 5-difluoro-N- [ (methylaminomethylsulfonyl) amino ] benzamide (AII-5)
To a stirred mixture of the product (AII-4) (573 mg,1 eq, 1.88 mmol) from step 3 above and 1-amino-3-methyl-thiourea (D-2) (238 mg,2.26mmol,1.2 eq) in DMF (20 mL) was added DIPEA (731 mg,3 eq, 5.65 mmol) and T at room temperature 3 P (899 mg,1.5 eq, 2.83 mmol). The resulting mixture was stirred at room temperature under an air atmosphere for 2 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (10% MeCN up to 60% in 15 minutes); detector, UV 254/220nm. This gave the sub-title compound (AII-5) (500 mg,1.28mmol,68%,90% purity) as a yellow solid. M/z 391.0/393.0 (M+H) + (ES+)
Step 5:5- [2- (2, 6-dichloropyridin-4-yl) -3, 5-difluorophenyl ] -4-methyl-1, 2, 4-triazole-3-thiol (AII-6)
A mixture of the product from step 4 above (AII-5) (490 mg,1 eq, 1.25 mmol) in aqueous NaOH (10 mL, 1M) was stirred at 60℃for 12 hours. The mixture was cooled to room temperature. The residue was acidified to pH 4 with concentrated HCl. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 65% in 15 minutes); detector, UV 254/220nm. This gave the sub-title compound (AII-6) (230 mg,0.62mmol,49%,92% purity) as a yellow solid. M/z 373.0/375.0 (M+H) + (ES+)
Step 6:2, 6-dichloro-4- [2, 4-difluoro-6- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridine (AII-7)
To a stirred mixture of the product from step 5 above (AII-6) (275 mg,1 eq., 0.74 mmol) and AcOH (89 mg,2 eq., 1.47 mmol) in DCM (20 mL) at 0deg.C was added H 2 O 2 (66 mg,30Wt%,5 equivalents, 3.69 mmol). The resulting mixture was stirred at room temperature for 2 hours. The residue was taken up with NaHCO 3 Is alkalized to pH 8. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% ACN up to 50% in 15 minutes); detector, UV 254/220nm. This gave the sub-title compound (AII-7) (173 mg,0.51mmol,69%,95% purity) as a yellow solid. M/z 341.0/343.0 (M+H) + (ES+)
Step 7: 2-chloro-6-cyclopropyl-4- [2, 4-difluoro-6- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridine (AII-8)
To a stirred mixture of the product (AII-7) (168 mg,1 equivalent, 0.49 mmol), cyclopropylboronic acid (ABF-2) (63 mg,1.5 equivalent, 0.74 mmol) from step 6 above in dioxane (10 mL) was added KOAc (97 mg,2 equivalent, 0.98 mmol) and PCy3 (14 mg,0.1 equivalent, 49. Mu. Mol) at room temperature under a nitrogen atmosphere. Pd (OAc) was added to the above mixture at room temperature under nitrogen atmosphere 2 (11 mg,0.1 eq, 49. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AII-8) (130 mg,0.38mmol,76%,91% pure) as a yellow solid. M/z 347.1/349.1 (M+H) + (ES+)
Step 8:2- { 6-cyclopropyl-4- [2, 4-difluoro-6- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AII-9)
To the product (AII-8) (107 mg,1.2 eq., 0.31 mmol), intermediate (AEE-1) (60 mg,1 eq., 0.26 mmol) and Cs from step 7 above, at room temperature under a nitrogen atmosphere 2 CO 3 (252 mg,3 eq, 0.77 mmol) to a stirred mixture of dioxane (10 mL) was added RuPhos ring palladium complex gen.3 (43 mg,0.2 eq, 52 μmol) and RuPhos (48 mg,0.4 eq, 0.10 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (6/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; ladderDegree: 30% B to 55% B in 10 minutes; wavelength: 254/220nm; retention time: 9) To give the title compound (AII-9) (19.6 mg, 35. Mu. Mol,14%,99.1% purity) as a white solid. M/z 557.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.00(d,J=1.1Hz,1H),7.74(d,J=4.9Hz,1H),7.72-7.66(m,1H),7.66-7.61(m,2H),7.51(d,J=8.8,2.7,1.2Hz,1H),6.87(t,J=1.3Hz,1H),5.00(s,2H),3.85(s,2H),3.50(s,3H),2.53(s,2H),2.04-1.87(m,5H),1.60(t,J=13.1,6.6,3.4Hz,1H),1.39(d,J=11.1,8.9Hz,1H),1.00-0.93(m,4H)。
Example 266: synthesis of 2- { 6-ethoxy-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -4-methoxy-3H-isoindol-1-one (AIJ-2)
Step 1:6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -4-methoxy-2, 3-dihydro-isoindol-1-one (AIJ-1)
A mixture of 7-methoxy-3-oxo-1, 2-dihydro-isoindole-5-carbaldehyde (AIC-7) (180 mg,1 eq, 0.94 mmol) and 1- (aminomethyl) cyclobutan-1-ol (AW-1) (114 mg,1.2 eq, 1.13 mmol) in MeOH (10 mL) was stirred at room temperature for 2 hours. Adding NaBH to the mixture at 0deg.C 4 (71 mg,2 equivalents, 1.88 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AIJ-1) (150 mg,0.54mmol,58%,92% purity) as a white solid. M/z 277.2 (M+H) + (ES+)
Step 2:2- { 6-ethoxy-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -4-methoxy-3H-isoindol-1-one (AIJ-2)
The product (AIJ-1) (50 mg,1 equivalent,0.18 mmol), intermediate (AGQ-1) (72 mg,1.2 eq, 0.22 mmol) and Cs 2 CO 3 (177 mg,3 eq, 0.54 mmol) to a stirred mixture of dioxane (10 mL) was added RuPhos ring palladium complex gen.3 (30 mg,0.2 eq, 36 μmol) and RuPhos (34 mg,0.4 eq, 72 μmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 42% B to 52% B in 9 minutes; wavelength: 254/220nm; retention time: 8.2 To give the title compound (AIJ-2) (20.6 mg, 36. Mu. Mol,20%,99.7% purity) as a white solid. M/z 573.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.49(s,1H),7.80(d,J=1.2Hz,1H),7.69(d,J=8.4,5.6Hz,1H),7.62-7.52(m,2H),7.40(d,J=25.0Hz,2H),6.28(d,J=1.3Hz,1H),5.21(s,1H),4.96(s,2H),4.33(m,2H),4.00(s,2H),3.93(s,3H),3.43(s,3H),2.69(s,2H),2.07-1.90(m,4H),1.62(d,J=10.8Hz,1H),1.46-1.39(m,1H),1.34(t,J=7.0Hz,3H)。
Example 267: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (2R) -1-methoxyprop-2-yl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) (AIK-2)
A solution of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) in MeOH (10 mL) was treated with (2R) -1-methoxypropan-2-amine (AIK-1) (7 mg,1.2 eq, 78. Mu. Mol) under a nitrogen atmosphere at 60℃for 1 hour. Adding NaBH to the mixture at 0deg.C 4 (7 mg,3 equivalents, 0.20 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 37% B to 47% B in 9 minutes; wavelength: 254/220nm; retention time: 8.25 To give the title compound (AIK-2) (3.4 mg,6.4 μmol,9.6%,98.3% purity) as a white solid. M/z 534.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.21(d,J=4.2,1.8Hz,2H),7.99(d,J=1.4Hz,1H),7.91-7.84(m,1H),7.75(s,1H),7.63(d,J=1.5Hz,2H),6.88(d,J=1.4Hz,1H),5.00(s,2H),3.84(m,2H),3.48(s,3H),3.35(s,2H),3.25(s,2H),3.21-3.17(m,1H),2.76(m,1H),2.07-2.01(m,1H),0.97(d,J=6.9,1.8Hz,7H)。
Example 268: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (2-isopropoxyethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIL-3)
Step 1:6- { [ (2-isopropoxyethyl) amino ] methyl } -2, 3-dihydro-isoindol-1-one (AIL-2)
A solution of intermediate (ACY-2) (60 mg,1 eq, 0.37 mmol) and 2-isopropoxyethylamine (AIL-1) (46 mg,1.2 eq, 0.45 mmol) in MeOH (4 mL) was stirred at 60℃for 2 h. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (28 mg,2 eq, 0.74 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AIL-2) (40 mg,0.16mmol,43%,95% pure) as a white solid. m/z 248.1(M+H) + (ES+)
Step 2:4- [ 2-cyclopropyl-6- (6- { [ (2-isopropoxyethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIL-3)
To intermediate (AEG-2) (50 mg,1 equivalent, 0.15 mmol), product (AIL-2) from step 1 above (44 mg,1.2 equivalent, 0.18 mmol) and Cs under nitrogen at room temperature 2 CO 3 (34 mg,3 eq, 0.45 mmol) to a stirred mixture of dioxane (10 mL) was added RuPhos ring palladium complex Gen.3 (23 mg,0.2 eq, 30. Mu. Mol) and RuPhos (28 mg,0.4 eq, 60. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 37% B to 44% B in 12 minutes; wavelength: 254/220nm; retention time: 10.75 To give the title compound (AIL-3) (15.8 mg, 29. Mu. Mol,19%,99.4% purity) as a white solid. M/z 548.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.21(d,J=3.5,1.8Hz,2H),7.99(s,1H),7.87(d,J=8.5Hz,1H),7.73(s,1H),7.63(s,2H),6.88(d,J=1.5Hz,1H),5.00(s,2H),3.82(s,2H),3.56-3.41(m,6H),2.61(t,J=5.8Hz,2H),2.05(m,1H),1.08(d,J=6.0Hz,6H),0.97(d,J=6.4Hz,4H)。
Example 269: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (2S) -2-methoxypropyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIN-2)
A solution of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and (2S) -2-methoxypropan-1-amine (AIM-1) (9 mg,1.5 eq, 98. Mu. Mol) in MeOH (5 mL) was stirred at 60℃for 1 hour. Adding NaBH to the mixture at 0deg.C 4 (5 mg,2 equivalents, 0.13 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃ and then concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 10% B to 32% B over 7 min; wavelength: 254/220 nm) to give the title compound (AIM-2) as a white solid (14.2 mg,26 μmol,41%,99.5% purity). M/z 534.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(d,J=1.7Hz,1H),8.28–8.17(m,2H),8.02–7.96(m,1H),7.93–7.81(m,1H),7.75(s,1H),7.65(d,J=6.3Hz,2H),6.93–6.82(m,1H),5.09–4.93(m,2H),3.84(d,J=5.5Hz,2H),3.48(t,J=3.4Hz,3H),3.43–3.37(m,1H),3.25(d,J=4.6Hz,3H),2.55–2.52(m,2H),2.09–1.99(m,1H),1.12–1.04(m,3H),1.05–0.84(m,4H)。
Example 270: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (2-ethoxyethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIN-2)
A solution of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol), DIPEA (25 mg,3 eq, 0.20 mmol) and 2-ethoxyethyl-1-amine (AIN-1) (7 mg,1.2 eq, 78. Mu. Mol) in MeOH (8 mL) was stirred at 60℃for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃ and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: sunfire prep. C18 column, 30 x 150mm,5μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 18% B to 30% B in 10 minutes; wavelength: 254/220nm; retention time: 10.07 To give the title compound (AIN-2) (9.4 mg,18 μmol,26%,97.6% purity) as a white solid. M/z 534.3 (M+H) + (ES+). 1 H NMR(400MHz,Methanol-d4)δ8.50(s,1H),8.14–8.10(m,1H),8.09(d,J=1.5Hz,1H),8.05(d,J=1.4Hz,1H),7.94–7.90(m,1H),7.80(t,J=1.1Hz,1H),7.69–7.64(m,1H),7.63–7.59(m,1H),6.91(d,J=1.4Hz,1H),5.02(s,2H),3.89(s,2H),3.56(t,J=5.4Hz,2H),3.51(d,J=7.0Hz,5H),2.76(t,J=5.4Hz,2H),2.07–1.98(m,1H),1.19(t,J=7.0Hz,3H),1.06–1.01(m,2H),1.01–0.95(m,2H)。
Example 271: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (2-methoxyethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIO-9)
Step 1: 5-bromo-2- (bromomethyl) -3-fluorobenzoic acid methyl ester (AIO-2)
To a stirred solution of methyl 5-bromo-3-fluoro-2-methylbenzoate (AIO-1) (1.50 g,1 equivalent, 6.07 mmol) and NBS (1.30 g,1.2 equivalent, 7.29 mmol) in benzotrifluoride (20 mL) at room temperature was added AIBN (299 mg,0.3 equivalent, 1.82 mmol). The resulting mixture was stirred at 110 ℃ overnight. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo to give the sub-title compound (AIO-2) (1.7 g,5.21mmol,86%,83% purity. M/z 326.9/328.9 (M+H)) as a brown yellow oil + (ES+)
Step 2: 6-bromo-4-fluoro-2, 3-dihydro-isoindol-1-one (AIO-3)
The product (AIO-2) (1.70 g,1 equivalent, 5.22 mmol) from step 1 above was reacted with NH 3 The stirred solution in MeOH (30 mL, 7M) was stirred at room temperature for 2 hours. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (3/1) to give a yellow solidThe sub-title compound (AIO-3) (1.1 g,3.34mmol,92% purity) was in the form of a body. M/z 330.0/332.0 (M+H) + (ES+)
Step 3: 6-vinyl-4-fluoro-2, 3-dihydro-isoindol-1-one (AIO-4)
Pd (PPh) was added to a stirred solution of the product (AIO-3) (1.10 g,1 equivalent, 4.78 mmol), tributyl (vinyl) stannane (1.82 g,1.2 equivalent, 5.74 mmol) and CsF (2.18 g,3 equivalent, 14.3 mmol) from step 2 above in dioxane (35 mL) at room temperature under nitrogen atmosphere 3 ) 2 Cl 2 (336 mg,0.1 eq, 0.48 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (3/1) to give the sub-title compound (AIO-4) as an off-white solid (629 mg,3.55mmol,74%,90% purity). M/z 178.1 (M+H) + (ES+)
Step 4: 7-fluoro-3-oxo-1, 2-dihydro-isoindole-5-carbaldehyde (AIO-5)
The product from step 3 above (AIO-4) (629 mg,1 equivalent, 3.53 mmol) and CA (887 mg,1.3 equivalent, 4.61 mmol), NMO (541 mg,1.3 equivalent, 4.61 mmol), K were combined 2 OsO 4 .2H 2 A solution of O (131 mg,0.1 eq, 0.36 mmol) in t-BuOH (20 mL) and water (20 mL) was stirred at room temperature for 2 hours. Adding NaIO to the mixture at room temperature 4 (1.52 g,2 equivalents, 7.10 mmol). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (3/1) to give the sub-title compound (AIO-5) (314 mg,1.75mmol,49%,92% purity) as an off-white solid. M/z 180.0 (M+H) + (ES+)
Step 5: 4-fluoro-6- (hydroxymethyl) -2, 3-dihydro-isoindol-1-one (AIO-6)
To a stirred solution of the product (AIO-5) (314 mg,1 eq., 1.75 mmol) from step 4 above in MeOH (20 mL) at 0deg.CAdding NaBH in batches into the liquid 4 (333 mg,5 equivalents, 8.78 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (20/1) to give the sub-title compound (AIO-6) (173 mg,0.96mmol,55%,95% purity) as an off-white solid. M/z 182.1 (M+H) + (ES+)
Step 6:4- { 2-cyclopropyl-6- [ 4-fluoro-6- (hydroxymethyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIO-7)
To intermediate (AEG-2) (200 mg,1 equivalent, 0.60 mmol), the product from step 5 above (AIO-6) (108 mg,1 equivalent, 0.60 mmol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 To a stirred solution of (552 mg,3 eq, 1.79 mmol) in dioxane (10 mL) were added X-Phos (114 mg,0.4 eq, 0.24 mmol) and XPhos Pd G3 (101 mg,0.2 eq, 0.12 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AIO-7) (200 mg,0.42mmol,70%,95% pure) as a yellow solid. M/z 481.2 (M+H) + (ES+)
Step 7:4- [ 2-cyclopropyl-6- (4-fluoro-6-formyl-1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIO-8)
A solution of the product from step 6 above (AIO-7) (35 mg,1 eq, 73. Mu. Mol) and DMP (46 mg,1.5 eq, 0.11 mmol) in DCM (10 mL) was stirred at room temperature for 1 hour. The resulting mixture was filtered and the filter cake was washed with DCM (3X 5 mL). The filtrate was concentrated in vacuo. The crude product was used directly in the next step without any further purification. M/z479.2 (M+H) + (ES+)
Step 8:4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (2-methoxyethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIO-9)
To the product from step 7 aboveTo a stirred solution of (AIO-8) (35 mg,1 eq, 73. Mu. Mol) and 2-methoxyethyl-1-amine (AEB-1) (10 mg,1.8 eq, 0.13 mmol) in MeOH (5 mL) was added DIPEA (38 mg,4 eq, 0.29 mmol). The resulting mixture was stirred at room temperature under nitrogen overnight. Adding NaBH to the mixture at 0deg.C 4 (14 mg,5 eq, 0.37 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash under the following conditions (column: XBridge-prepared C18OBD column, 30X 100mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 32% B to 62% B in 25 minutes; wavelength: 254/220 nm) to give the title compound (AIO-9) (14.8 mg, 28. Mu. Mol,37%,99.7% purity) as a white solid. M/z 538.1 (M+H) + (ES+). 1 H NMR(400MHz,Methanol-d4)δ8.49(s,1H),8.17–7.89(m,4H),7.65(s,1H),7.47–7.40(m,1H),6.94(d,J=1.4Hz,1H),5.06(s,2H),3.91(s,2H),3.59–3.44(m,5H),3.35(s,3H),2.77(t,J=5.3Hz,2H),2.09–2.00(m,1H),1.10–0.96(m,4H)。
Example 272: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (2R) -2-hydroxybutyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIP-2)
A solution of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol), (2R) -1-aminobutan-2-ol (AIP-1) (7 mg,1.2 eq, 78. Mu. Mol) and DIPEA (25 mg,3 eq, 0.20 mmol) in MeOH (8 mL) was stirred at 60℃for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on the followingPurification under conditions (column: XBridge-prepared OBD C18 column, 30X 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35% B to 52% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (AIP-2) (11.6 mg, 22. Mu. Mol,33%,98.4% purity) as a white solid. M/z 534.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.16-8.06 (m, 2H), 8.05 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.82 (s, 1H), 7.83-7.80 (m, 1H), 7.63 (d, j=7.9 hz, 1H), 6.92 (d, j=1.5 hz, 1H), 5.02 (s, 2H), 4.03-3.88 (m, 2H), 3.68-3.61 (m, 1H), 3.49 (s, 3H), 2.74-2.68 (m, 1H), 2.61-2.54 (m, 1H), 2.06-1.99 (m, 1H), 1.59-1.37 (m, 2H), 1.10-0.88 (m, 7H).
Example 273: synthesis of 4- (2-cyclopropyl-6- (6- ((2-methoxyethoxy) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AIQ-2)
Step 1:4- (2- (6- (chloromethyl) -1-oxoisoindolin-2-yl) -6-cyclopropylpyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AIQ-1)
To a stirred solution of compound (AHV-1) (60 mg,1 eq, 0.13 mmol) in DCM (5 mL) at 0deg.C was added SOCl 2 (46 mg,3 eq, 0.39 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1) to give the sub-title compound (AIQ-1) (58 mg,0.12mmol,93%,92% pure) as a yellow solid. M/z 481.1/483.1 (M+H) + (ES+)
Step 2:4- (2-cyclopropyl-6- (6- ((2-methoxyethoxy) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AIQ-2)
At room temperature, to the reaction mixture from the aboveTo a stirred solution of the product of step 1 (AIQ-1) (50 mg,1 eq, 0.10 mmol), 2-methoxyethanol (AFM-1) (40 mg,5 eq, 0.52 mmol) and TBAB (16.76 mg, 52. Mu. Mol,0.5 eq) in DCM (2 mL) was added aqueous KOH (2 mL,20 Wt%). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 54% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AIQ-2) (9.2 mg, 18. Mu. Mol,17%,99.4% purity) as a white solid. M/z 521.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.07 (m, 2H), 8.04 (d, j=1.4 hz, 1H), 7.95-7.89 (m, 1H), 7.83-7.79 (m, 1H), 7.70-7.59 (m, 2H), 6.92 (d, j=1.4 hz, 1H), 5.02 (s, 2H), 4.65 (s, 2H), 3.73-3.65 (m, 2H), 3.63-3.59 (m, 2H), 3.51 (s, 3H), 3.38 (s, 3H), 2.13-2.01 (m, 1H), 1.07-1.01 (m, 2H), 1.01-0.94 (m, 2H).
Example 274: synthesis of (S) -4- (2-cyclopropyl-6- (6- (((1-methoxyprop-2-yl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AIR-2)
To a stirred mixture of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) in MeOH (10 mL) was added (2S) -1-methoxypropan-2-amine (AIR-1) (9 mg,1.5 eq, 98. Mu. Mol) at 60 ℃. The resulting mixture was stirred at 60℃for 2 hours. Adding NaBH to the mixture at 0deg.C 4 (5 mg,2 equivalents, 0.13. Mu. Mol). The resulting mixture was stirred at room temperature for 1 hour. Then at 0 ℃ through adding MeOH2 mL) quench the reaction. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 58% B in 8 minutes; wavelength: 254/220nm; retention time: 7.17 To give the title compound (AIR-2) (11.9 mg,22 μmol,34%,99.7% purity) as a white solid. M/z 534.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.27–8.18(m,2H),7.99(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.75(s,1H),7.63(s,2H),6.88(d,J=1.4Hz,1H),5.00(s,2H),3.93–3.77(m,2H),3.48(s,3H),3.23(s,4H),3.22–3.16(m,1H),2.81–2.72(m,1H),2.08–1.98(m,1H),1.06–0.89(m,7H)。
Example 275: synthesis of 4- (2- {6- [ (cyclopentylmethoxy) methyl ] -1-oxo-3H-isoindol-2-yl } -6-cyclopropylpyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIS-2)
To a stirred mixture of intermediate (AIQ-1) (40 mg,1 eq, 83. Mu. Mol) and cyclopentanemethanol (AIT-1) (83 mg,10 eq, 0.83 mmol) in DCM (4 mL) was added TBAB (2.68 mg, 8. Mu. Mol,0.1 eq) at room temperature. To the above mixture was added aqueous KOH solution (4 ml,20 wt%) at room temperature. The resulting mixture was stirred at room temperature for an additional 0.5 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 65% B to 7 in 8 minutes5% B; wavelength: 254/220nm; retention time: 7.98 To give the title compound (AIS-2) (2.1 mg,3.9 μmol,4.6%,99.4% purity) as a white solid. M/z 545.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.23–8.19(m,2H),8.00(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.73–7.60(m,3H),6.87(d,J=1.4Hz,1H),5.02(s,2H),4.58(s,2H),3.47(s,3H),3.33(d,J=7.0Hz,2H),2.20–2.10(m,1H),2.08–1.99(m,1H),1.76–1.61(m,2H),1.59–1.44(m,4H),1.26–1.17(m,2H),1.03–0.91(m,4H)。
Example 276: synthesis of 4- (2-cyclopropyl-6- {6- [ (methylamino) methyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIT-1)
A solution of methylamine (3.1 mg,1.5 eq, 98. Mu. Mol) and DIPEA (10 mg,1.2 eq, 78. Mu. Mol) in MeOH (3 mL) was stirred at room temperature for 10 min. To the above mixture was added intermediate (AGT-2) (30 mg,1 equivalent, 65. Mu. Mol) at room temperature. The resulting mixture was stirred at 60℃for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (5 mg,2 equivalents, 0.13 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 50% B in 7 minutes; wavelength: 254/220 nm) to give the title compound (AIT-1) (3.7 mg, 7.8. Mu. Mol,12%,99.6% purity) as a white solid. M/z 476.2 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.22–8.19(m,2H),7.99(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.73(s,1H),7.63(s,2H),6.87(d,J=1.4Hz,1H),5.00(s,2H),3.75(s,2H),3.47(s,3H),2.27(s,3H),2.07–2.01(m,1H),0.97(d,J=5.9Hz,4H)。
Example 277: synthesis of 2- (4- (4-chloro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6-cyclopropylpyridin-2-yl) -6- (hydroxymethyl) isoindolin-1-one (AIU-1)
To a 25mL round bottom flask was added intermediate (AGW-5) (100 mg,1 eq, 0.29 mmol), intermediate (AGH-1) (57 mg,1.2 eq, 0.35 mmol) and Cs at room temperature under nitrogen atmosphere 2 CO 3 (283 mg,3 eq, 0.87 mmol) of 1, 4-dioxane (15 ml). To the above stirred solution were added CuI (11 mg,0.2 eq, 58 μmol) and (1 s,2 s) -1-N, 2-N-dimethylcyclohexane-1, 2-diamine (16 mg,0.4 eq, 0.12 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred under nitrogen at 100 ℃ for an additional 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 28% B to 38% B in 9 minutes; wavelength: 254 nm) to give the title compound (AIU-1) (7.4 mg, 16. Mu. Mol,5.4%,99.6% purity) as a white solid. M/z 472.0/474.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.51(s,1H),8.00(d,J=1.4Hz,1H),7.85–7.77(m,2H),7.75–7.60(m,4H),6.80(d,J=1.5Hz,1H),5.40–5.35(m,1H),5.01(s,2H),4.61(d,J=5.8Hz,2H),3.42(s,3H)2.06–1.99(m,1H),0.95(d,J=6.4Hz,4H)。
Example 278: synthesis of 2- (4- (4-chloro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6-cyclopropylpyridin-2-yl) -6- (((cyclobutylmethyl) amino) methyl) isoindolin-1-one (AIV-2)
Step 1:2- (4- (4-chloro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6-cyclopropylpyridin-2-yl) -3-oxoisoindoline-5-carbaldehyde (AIV-1)
To a stirred solution of compound (AIU-1) (100 mg,1 eq, 0.21 mmol) in DCM (5 mL) under nitrogen at room temperature was added DMP (178 mg,2 eq, 0.42 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. Filtering the resulting mixture; the filter cake was washed with DCM (3X 5 mL). The filtrate was concentrated in vacuo. The crude product was used directly in the next step without any further purification. M/z 470.1/472.1 (M+H) + (ES+)
Step 2:2- (4- (4-chloro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6-cyclopropylpyridin-2-yl) -6- (((cyclobutylmethyl) amino) methyl) isoindolin-1-one (AIV-2)
To a stirred mixture of the product from step 1 above (AIV-1) (50 mg,1 eq, 0.11 mmol) and 1-cyclobutylmethylamine (AIA-1) (14 mg,1.5 eq, 0.16 mmol) in DCM (5 mL) was added DIPEA (17 mg,1.2 eq, 0.13 mmol) at room temperature. The resulting mixture was stirred at 60℃for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture 4 (8 mg,2 equivalents, 0.21 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (Xselect CSH F-phenyl OBD column, 19 x 250mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 25 ml/min; gradient: 70% B to 85% B in 9 minutes; wavelength: 254/220nm; retention time: 8.3 To give the title compound (AIV-2) (6.8 mg,13 μmol,12%,98.0% purity) as a white solid. M/z 539.2/541.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) δ8.47 (s, 1H), 8.05 (d, j=1.4 hz, 1H), 7.79 (d, j=1 hz1.5Hz,1H),7.78(d,J=2.2Hz,1H),7.74(s,1H),7.73–7.69(m,1H),7.67–7.58(m,2H),6.85(d,J=1.5Hz,1H),5.02(s,2H),3.85(s,2H),3.45(s,3H),2.62(d,J=7.3Hz,2H),2.60–2.47(m,1H),2.15–2.08(m,2H),2.09–1.95(m,1H),1.94–1.78(m,2H),1.76–1.62(m,2H),1.05–0.99(m,2H),0.99–0.93(m,2H)。
Example 279: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (2-methoxy-2-methylpropyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIW-2)
A solution of 2-methoxy-2-methylpropan-1-amine (AIW-1) (8 mg,1.2 eq, 78. Mu. Mol) and DIPEA (34 mg,4 eq, 0.26 mmol) in MeOH (8 mL) was stirred at room temperature for 10 min. To the above mixture was added intermediate (AGT-2) (30 mg,1 equivalent, 65. Mu. Mol). The resulting mixture was stirred at 60 ℃ for another 4 hours. Adding NaBH to the above mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 42% B to 52% B in 8 minutes; wavelength: 254/220nm; retention time: 7.58 To give the title compound (AIW-2) (13.7 mg,25 μmol,38%,99.1% purity) as a white solid. M/z 549.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (d, j=1.5 hz, 1H), 8.14-8.03 (m, 3H), 7.95-7.88 (m, 1H), 7.81 (s, 1H), 7.71-7.64 (m, 1H), 7.61 (d, j=7.7 hz, 1H), 6.91 (d, j=1.6 hz, 1H), 5.02 (s, 2H), 3.89 (s, 2H), 3.50 (d, j=1.6 hz, 3H), 3.15 (d, j=1.5 hz, 3H), 2.56 (d, j=1.6 hz, 2H), 2.07-1.97 (m, 1H), 1.18 (d, j=1.6 hz, 6H), 1.04-0.97 (m, 4H).
Example 280: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (cyclopropylmethyl) (methyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIX-2)
A solution of 1-cyclopropyl-N-methyl methylamine, HCl (AIX-1) (7 mg,1.2 eq., 78. Mu. Mol) and DIPEA (34 mg,4 eq., 0.26. Mu. Mol) in MeOH (8 mL) was stirred at room temperature for 10 min. To the above mixture was added intermediate (AGT-2) (30 mg,1 equivalent, 65. Mu. Mol) at room temperature. The resulting mixture was stirred at room temperature overnight. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 32% B to 62% B in 7 minutes; wavelength: 254nm; retention time: 8.65 To give the title compound (AIX-2) (3.4 mg, 6.4. Mu. Mol,9.4%,95.6% purity) as a white solid. M/z 530.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.07 (m, 2H), 8.05 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.79 (d, j=1.5 hz, 1H), 7.68-7.59 (m, 2H), 6.92 (d, j=1.5 hz, 1H), 5.03 (s, 2H), 3.70 (s, 2H), 3.50 (s, 4H), 2.37-2.26 (m, 5H), 2.07-1.99 (m, 1H), 1.06-1.02 (m, 2H), 1.01-0.96 (m, 2H), 0.58-0.51 (m, 2H), 0.15-0.09 (m, 2H).
Example 281: synthesis of 4- [ 2-cyclopropyl-6- (6- { [2- (methylamino) ethoxy ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIY-2)
Step 1: n- {2- [ (2- {4- [ 4-cyano-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-cyclopropylpyridin-2-yl } -3-oxo-1H-isoindol-5-yl) methoxy ] ethyl } -N-methylcarbamic acid tert-butyl ester (AIY-1)
To a stirred solution of intermediate (AIQ-1) (50 mg,1 eq, 0.10 mmol), tert-butyl N- (2-hydroxyethyl) -N-methylcarbamate (AHE-2) (18 mg,1 eq, 0.10 mmol) and TBAB (17 mg,0.5 eq, 52. Mu. Mol) in DCM (3 mL) was added aqueous KOH (3 mL,20 Wt%) at room temperature. The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1) to give the title compound (AIY-1) (40 mg, 64. Mu. Mol,62%,95% pure) as a brown yellow solid. M/z 620.3 (M+H) + (ES+)
Step 2:4- [ 2-cyclopropyl-6- (6- { [2- (methylamino) ethoxy ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIY-2)
To a stirred solution of the product from step 1 above (AIY-1) (40 mg,1 eq, 65 μmol) in DCM (6 mL) at room temperature was added TFA (2 mL). The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 54% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AIY-2) (6.9 mg, 13. Mu. Mol,20%,98.3% purity) as a white solid. M/z 520.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.08 (m, 2H), 8.04 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.85-7.79 (m, 1H), 7.71-7.62 (m, 2H), 6.93 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 4.65 (s, 2H), 3.66 (d, j=5.0 hz, 2H), 3.50 (s, 3H), 2.87-2.80 (m, 2H), 2.44 (s, 3H), 2.09-1.99 (m, 1H), 1.08-0.95 (m, 4H).
Example 282: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (3R) -1-methyl-2-oxopyrrolidin-3-yl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AIZ-2)
To a stirred solution of intermediate (AGT-2) (30 mg,1 eq, 65 μmol) and (3R) -3-amino-1-methylpyrrolidin-2-one (AIZ-1) (10 mg,1.2 eq, 78 μmol) in MeOH (8 mL) at room temperature was added DIPEA (34 mg,4 eq, 0.26 mmol). The resulting mixture was stirred at 60℃for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 34% B to 53% B in 8 minutes; wavelength: 254/220nm; retention time: 7.89 To give the title compound (AIZ-2) (10.7 mg,19 μmol,29%,99.8% purity) as a white solid. M/z 559.3 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.55(s,1H),8.24–8.19(m,2H),7.97(d,J=1.4Hz,1H),7.89–7.75(m,2H),7.64(d,J=2.2Hz,2H),6.90(d,J=1.4Hz,1H),5.00(s,2H),3.91(s,2H),3.49(s,3H),3.29–3.18(m,3H),2.73(s,3H),2.22–2.13(m,1H),2.08–2.00(m,1H),1.72–1.62(m,1H),1.00–0.94(m,4H)。
Example 283: synthesis of (S) -4- (2-cyclopropyl-6- (6- (((2-hydroxybutyl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AJA-2)
To a stirred solution of intermediate (AGT-2) (30 mg,1 eq, 65 μmol) and (2S) -1-aminobutan-2-ol (AJA-1) (7 mg,1.2 eq, 78 μmol) in MeOH (8 mL) was added DIPEA (25 mg,3 eq, 0.20 mmol) at room temperature. The resulting mixture was stirred at 60℃for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35% B to 45% B in 9 minutes; wavelength: 254/220nm; retention time: 8.2 To give the title compound (AJA-2) (6.7 mg,13 μmol,19%,98.6% purity) as a white solid. M/z 534.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.55-8.47 (m, 1H), 8.15-8.10 (m, 1H), 8.10-8.03 (m, 2H), 7.94-7.90 (m, 1H), 7.83 (d, j=1.8 hz, 1H), 7.72-7.62 (m, 2H), 6.92 (d, j=1.5 hz, 1H), 5.03 (d, j=2.8 hz, 2H), 4.04-3.94 (m, 2H), 3.70-3.62 (m, 1H), 3.49 (d, j=1.7 hz, 3H), 2.78-2.71 (m, 1H), 2.64-2.56 (m, 1H), 2.06-1.99 (m, 1H), 1.54-1.38 (m, 2H), 1.06-0.92 (m, 7H).
Example 284: synthesis of N- (2- (((2- (4- (4-cyano-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6-cyclopropylpyridin-2-yl) -3-oxoisoindolin-5-yl) methyl) amino) ethyl) acetamide (AJB-2)
A solution of intermediate (AGN-1) (40 mg,1 eq, 76. Mu. Mol), DIPEA (39 mg,4 eq, 0.30 mmol) and N- (2-aminoethyl) acetamide (AJB-1) (9 mg,1.2 eq, 91. Mu. Mol) in MeOH (8 mL) was stirred at room temperature overnight. Adding NaBH to the mixture at 0deg.C 4 (14 mg,5 eq, 0.38 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35% B to 45% B in 9 minutes; wavelength: 254/220nm; retention time: 8.47 To give the title compound (AJB-2) (10.4 mg,17 μmol,22%,99.6% purity) as a white solid. M/z 615.2 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.56–8.51(m,1H),8.25–8.18(m,2H),8.07–7.97(m,3H),7.91–7.84(m,1H),7.80(s,1H),6.90(d,J=2.0Hz,1H),5.16(s,2H),3.88(s,2H),3.50–3.45(m,3H),3.14(d,J=6.3Hz,3H),2.55(d,J=5.6Hz,1H),2.11–2.03(m,1H),1.82–1.77(m,3H),1.02–0.90(m,4H)。
Example 285: synthesis of 2- (6-ethoxy-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (hydroxymethyl) -4-methoxyisoindolin-1-one (AJC-1)
Cs was added to a stirred solution of intermediate (AGQ-1) (50 mg,1 eq, 0.15 mmol) and intermediate (AIC-8) (44 mg,1.5 eq, 0.23 mmol) in dioxane (5 mL) at room temperature under nitrogen atmosphere 2 CO 3 (98 mg,2 equivalents, 0.30 mmol). RuPhos (28 mg,0.4 eq, 0.06 mmol) and RuPhos ring palladium complex Gen.3 (25 mg,0.2 eq, 0.03 mmol) were added to the above mixture under nitrogen at room temperature. The resulting mixture was stirred under nitrogen at 100 ℃ for an additional 2 hours. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 36% B to 46% B in 9 minutes; wavelength: 254/220nm; retention time: 8.18 To give the title compound (AJC-1) (17.5 mg,36 μmol,24%,99.5% purity) as a white solid. M/z 490.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.51(s,1H),7.81(d,J=1.3Hz,1H),7.73–7.66(m,1H),7.64–7.54(m,2H),7.33(d,J=1.1Hz,1H),7.24(d,J=1.2Hz,1H),6.27(d,J=1.2Hz,1H),5.40(t,J=5.8Hz,1H),4.94(s,2H),4.61(d,J=5.8Hz,2H),4.37–4.27(m,2H),3.92(s,3H),3.43(s,3H),1.34(t,J=7.1Hz,3H)。
Example 286: synthesis of 3'- (6- (hydroxymethyl) -1-oxoisoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJD-2)
Step 1:3 '-bromo-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJD-1)
At 0℃to t-BuNO 2 (603 mg,6 eq., 5.89 mmol) to a stirred mixture of ACN (10 mL) was added CuBr (211 mg,1.5 eq., 1.47 mmol). The resulting mixture was stirred at 0 ℃ for 1 hour. Adding 3 '-amino-2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] to the above mixture at 0deg.C]4-formonitrile (ADG-2) (270 mg,1 eq, 0.98 mmol). The resulting mixture was stirred at 50 ℃ for an additional 1 hour. The mixture was cooled to room temperature and concentrated in vacuo. The residue was applied to silica gel column chromatography with DCM/methyl (12/1) to give the sub-title compound (AJE-1) (140 mg,0.41mmol,42%,95% purity) as a yellow solid. M/z 339.0/341.0 (M+H) + (ES+)
Step 2:3'- (6- (hydroxymethyl) -1-oxoisoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJD-2)
To the product (AJD-1) from step 1 above (140 mg,1 eq, 0.41 mmol) under nitrogen at room temperatureIntermediate (AGH-1) (81 mg,1.2 eq, 0.50 mmol) and Cs 2 CO 3 (403 mg,3 eq, 1.24 mmol) to a stirred mixture of dioxane (10 mL) was added RuPhos ring palladium complex Gen.3 (69 mg,0.2 eq, 83. Mu. Mol) and RuPhos (77 mg,0.4 eq, 0.17 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 20% B to 40% B in 8 minutes; wavelength: 254/220nm; retention time: 7.53 To give the title compound (AJD-2) (1.9 mg,4.5 μmol,1.1%,99.6% purity) as a white solid. M/z 422.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.47(d,J=1.9Hz,1H),8.19(d,J=8.1,1.8Hz,1H),8.12(d,J=1.9Hz,1H),8.01(d,J=8.3Hz,1H),7.88(d,J=8.0,1.8Hz,1H),7.75-7.69(m,2H),7.62(s,2H),7.41(t,J=8.0Hz,1H),6.91(d,J=7.7Hz,1H),5.37(d,J=6.6,4.8Hz,1H),4.90(s,2H),4.62(d,J=5.7Hz,2H),3.16(d,J=1.8Hz,3H)。
Example 287: synthesis of 3'- (6- (((cyclopropylmethyl) amino) methyl) -1-oxoisoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJE-2)
Step 1:3'- (6-formyl-1-oxo-3H-isoindol-2-yl) -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJE-1)
To a stirred mixture of compound (AJD-2) (30 mg,1 eq, 71. Mu. Mol) in DCM (10 mL) at room temperature was added DMP (36 mg,1.2 eq, 8)5. Mu. Mol). The resulting mixture was stirred at room temperature for 1 hour. Filtering the resulting mixture; the filter cake was washed with DCM (3X 3 mL). The filtrate was concentrated in vacuo. The crude mixture was used directly in the next step without further purification. M/z 420.1 (M+H) + (ES+)
Step 2:3'- (6- (((cyclopropylmethyl) amino) methyl) -1-oxoisoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJE-2)
A mixture of the product from step 1 above (AJE-1) (30 mg,1 eq, 72. Mu. Mol) and 1-cyclopropylmethylamine (AIB-1) (8 mg,1.5 eq, 0.11 mmol) in MeOH (10 mL) was stirred at room temperature for 12 hours. Adding NaBH to the mixture at 0deg.C 4 (5 mg,2 equivalents, 0.14 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 27% B over 10 min; wavelength: 254nm; retention time: 9.47) to give the title compound (AJE-2) (6.0 mg,13 μmol,18%,99.2% purity) as a yellow solid. M/z 475.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),8.25(s,1H),8.19(d,J=8.1,1.8Hz,1H),8.12(d,J=1.8Hz,1H),8.01(d,J=8.3,2.3Hz,1H),7.87(d,J=8.1Hz,1H),7.78(s,1H),7.72–7.58(m,3H),7.41(t,J=8.0Hz,1H),6.92(d,J=7.7Hz,1H),4.89(s,2H),3.90(d,J=4.6Hz,2H),3.16(s,3H),2.45(t,J=5.6Hz,2H),1.00–0.87(m,1H),0.48–0.39(m,2H),0.13(t,J=4.3Hz,2H)。
Example 288: synthesis of 3'- (6- (((2-methoxyethyl) amino) methyl) -1-oxoisoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJF-1)
Intermediate (AJE-1)) A mixture of (30 mg,1 eq, 72. Mu. Mol) and 2-methoxyethyl-1-amine (AEB-1) (6.5 mg,1.2 eq, 86. Mu. Mol) in MeOH (10 mL) was stirred at room temperature for 12 hours. Adding NaBH to the mixture at 0deg.C 4 (5.4 mg,2 equivalents, 0.14 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 27% B over 10 min; wavelength: 254nm; retention time: 9) to give the title compound (AJF-1) (4.3 mg,9.0 μmol,12%,95.9% purity) as a white solid. M/z 479.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.46(s,1H),8.19(d,J=8.1,1.8Hz,1H),8.12(d,J=1.8Hz,1H),8.01(d,J=8.3,2.3Hz,1H),7.87(d,J=8.1Hz,1H),7.76-7.67(m,2H),7.66–7.55(m,2H),7.41(t,J=8.0Hz,1H),6.92(d,J=7.6Hz,1H),4.88(s,2H),3.82(s,2H),3.40(t,J=5.7Hz,2H),3.24(m,3H),3.15(m,3H),2.64(t,J=5.7Hz,2H)。
Example 289: synthesis of 4- (2-cyclopropyl-6- (6- (((1- (methoxymethyl) cyclobutyl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AJG-2)
Intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol), 1- (methoxymethyl) cyclobutan-1-amine (AJG-1) (9 mg,1.2 eq, 78. Mu. Mol) and Ti (Oi-Pr) 4 A solution of (74 mg,4 eq, 0.26 mmol) in THF (8 mL) was stirred overnight at 60 ℃. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XBIdge preparative OBDC18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 46% B to 63% B in 8 minutes; wavelength: 254/220nm; retention time: 7.8 To give the title compound (AJG-2) (3.2 mg,5.7 μmol,8.8%,99.8% purity) as a white solid. M/z 560.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.14-8.10 (m, 1H), 8.10-8.04 (m, 2H), 7.93 (d, j=8.1 hz, 1H), 7.83 (s, 1H), 7.70-7.66 (m, 1H), 7.61 (d, j=7.8 hz, 1H), 6.92 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 3.82 (s, 2H), 3.55 (s, 2H), 3.50 (s, 3H), 3.42 (s, 3H), 2.13-1.95 (m, 5H), 1.89-1.77 (m, 2H), 1.07-1.01m, 2H), 1.01-0.96 (m, 2H).
Example 290: synthesis of 4- (2- (6-acetyl-1-oxoisoindolin-2-yl) -6-cyclopropylpyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AJH-3)
Step 1:6- (1-ethoxyvinyl) -2, 3-dihydro-isoindol-1-one (AJH-1)
Pd (PPh) was added to a stirred solution of intermediate (ACY-1) (2.00 g,1 equivalent, 9.43 mmol) and dibutyl (1-ethoxyvinyl) propylstannane (AFR-1) (3.93 g,1.2 equivalent, 11.3 mmol) in dioxane (15 mL) at room temperature under nitrogen 3 ) 2 Cl 2 (662 mg,0.1 eq, 0.94 mmol). The resulting mixture was stirred overnight at 80 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AJH-1) (1.7 g,8.33mmol,89%,92% pure) as a yellow solid. M/z 204.1 (M+H) + (ES+)
Step 2: 6-acetyl-2, 3-dihydro-isoindol-1-one (AJH-2)
To a stirred solution of the product (AJH-1) (1.7 g,1 eq, 8.33 mmol) from step 1 above in DCM (20 mL) at room temperature was added TsOH #2.88g,2 equivalents, 16.7 mmol). The resulting mixture was stirred at room temperature for 1 hour. The mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AJH-2) (1.2 g,6.85mmol,82%,93% pure) as a yellow solid. M/z 176.1 (M+H) + (ES+)
Step 3:4- (2- (6-acetyl-1-oxoisoindolin-2-yl) -6-cyclopropylpyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AJH-3)
To the product (AJH-2) (25 mg,1.2 eq, 0.14 mmol), intermediate (AEG-2) (40 mg,1 eq, 0.12 mmol) and Cs from step 2 above under nitrogen at room temperature 2 CO 3 (77 mg,2 eq, 0.24 mmol) to a stirred solution in dioxane (8 mL) was added RuPhos (22 mg,0.4 eq, 48. Mu. Mol) and RuPhos ring palladium complex Gen.3 (20 mg,0.2 eq, 24. Mu. Mol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The product was purified by preparative HPLC on a column (XBridge prep OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 50% B in 8 minutes; wavelength: 254/220nm; retention time: 7.67 To give the title compound (AJH-3) (3.2 mg, 6.7. Mu. Mol,5.7%,99.7% purity) as a white solid. M/z 475.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.29–8.24(m,2H),8.24–8.20(m,2H),7.98(d,J=1.5Hz,1H),7.90–7.84(m,2H),6.93(d,J=1.4Hz,1H),5.12(s,2H),3.49(s,3H),2.68(s,3H),2.09–2.03(m,1H),1.02–0.95(m,4H)。
Example 291: synthesis of 4- (2-cyclopropyl-6- (6- (1- ((cyclopropylmethyl) amino) ethyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AJI-1)
To a stirred solution of intermediate (AJH-3) (100 mg,1 eq, 0.21 mmol) and 1-cyclopropylmethylamine (AIB-1) (18 mg,1.2 eq, 0.25 mmol) in DCM (10 mL) at RT was added Ti (Oi-Pr) 4 (180 mg,3 eq, 0.63 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (40 mg,5 equivalents, 1.06 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The resulting mixture was diluted with water and extracted with DCM/MeOH (10/1) (3X 20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 58% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AJI-1) (53 mg,0.10mml,47%,98.3% purity) as a white solid. M/z 530.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.16-8.03 (m, 3H), 7.92 (d, j=8.1 hz, 1H), 7.86-7.77 (m, 1H), 7.70-7.59 (m, 2H), 6.93 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 4.03-3.93 (m, 1H), 3.50 (s, 3H), 2.43-2.32 (m, 1H), 2.27-2.15 (m, 1H), 2.08-1.97 (m, 1H), 1.43 (d, j=6.7 hz, 3H), 1.08-0.86 (m, 5H), 0.54-0.41 (m, 2H), 0.13-0.03 (m, 2H).
Example 292: synthesis of (R) -4- (2-cyclopropyl-6- (6- (1- ((cyclopropylmethyl) amino) ethyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AJJ-1)
The crude product (AJI-1) (50 mg,1 eq,94 μmol) was purified by preparative chiral HPLC under the following conditions (column: CHIRAL ART Amylose-SA, 2X 25cm,5 μm; mobile phase a: mtBE (0.5% 2M NH) 3 MeOH) -HPLC, mobile phase B: etOH-HPLC; flow rate: 20 ml/min; gradient: 50% B to 50% B within 25 minutes; wavelength: 220/254nm, retention time 1 (min): 14.486 To give the title compound (AJJ-1) (19.6 mg, 37. Mu. Mol,39%,99.6% purity) as a white solid. M/z 530.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.10 (m, 1H), 8.10-8.07 (m, 1H), 8.06 (d, j=1.4 hz, 1H), 7.95-7.91 (m, 1H), 7.82-7.77 (m, 1H), 7.70-7.64 (m, 1H), 7.62 (d, j=7.9 hz, 1H), 6.92 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 4.02-3.93 (m, 1H), 3.50 (s, 3H), 2.42-2.32 (m, 1H), 2.25-2.15 (m, 1H), 2.09-1.98 (m, 1H), 1.43 (d, j=6.7 hz, 3H), 1.07-1.02 (m, 2.01-0.95 (m, 0.0.44-0H), 0.0.0.0 (m, 3H). Column: CHIRALPAK IA-3,4.6 x 50mm,3 μm; mobile phase a: mtBE (0.1% DEA): etoh=50:50; flow rate: 1 ml/min; retention time: 2.408.
Example 293: synthesis of (S) -4- (2-cyclopropyl-6- (6- (1- ((cyclopropylmethyl) amino) ethyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AJK-1)
The crude product (AJI-1) (50 mg,1 eq., 94. Mu. Mol) was purified by preparative chiral HPLC under the following conditions (column: CHIRAL ART Amylose-SA, 2X 25cm, 5. Mu.m; mobile phase A: mtBE (0.5% 2M NH) 3 MeOH) -HPLC, mobile phase B: etOH-HPLC; flow rate: 20 ml/min; gradient: 50% B to 50% B within 25 minutes; wavelength: 220/254nm, retention time 2 (min): 22.279 To give the title compound (AJK-1) (17 mg,32 μmol,33%,98.5% purity) as a white solid. M/z 530.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.10 (m, 1H), 8.10-8.07 (m, 1H), 8.06 (d, J=1.4 Hz, 1H), 7.95-7.91 (m, 1H), 7.82-7.77 (m, 1H), 7.70-7.64 (m1H), 7.62 (d, j=7.9 hz, 1H), 6.92 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 4.02-3.93 (m, 1H), 3.50 (s, 3H), 2.42-2.32 (m, 1H), 2.25-2.15 (m, 1H), 2.09-1.98 (m, 1H), 1.43 (d, j=6.7 hz, 3H), 1.07-1.02 (m, 2H), 1.01-0.95 (m, 2H), 0.95-0.87 (m, 1H), 0.52-0.44 (m, 2H), 0.13-0.03 (m, 2H). Column: CHIRALPAK IA-3,4.6 x 50mm,3um; mobile phase a: mtBE (0.1% DEA): etoh=50:50; flow rate: 1 ml/min; retention time: 3.250.
Example 294: synthesis of 4- (2-cyclopropyl-6- (6-hydroxy-1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AJL-2)
To intermediate (AEG-2) (150 mg,1 eq, 0.45 mmol), 6-hydroxy-2, 3-dihydro-isoindol-1-one (AJL-1) (74 mg,1.1 eq, 0.49 mmol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 (29 mg,2 eq, 0.89 mmol) to a stirred solution of dioxane (10 mL) was added RuPhos (84 mg,0.4 eq, 0.18 mmol) and RuPhos ring palladium complex gen.3 (75 mg,0.2 eq, 89 μmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 55% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AJL-2) (9.7 mg,22 μmol,4.8%,99.8% purity) as a white solid. M/z 449.1 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ9.88(s,1H),8.54(s,1H),8.23–8.16(m,2H),7.98(d,J=1.4Hz,1H),7.86(d,J=8.6Hz,1H),7.50(d,J=8.1Hz,1H),7.12–7.04(m,2H),6.85(d,J=1.4Hz,1H),4.90(s,2H),3.45(s,3H),2.07–1.99(m,1H),1.01–0.89(m,4H)。
Example 295: synthesis of (S) -4- (2-cyclopropyl-6- (6- (2, 3-dihydroxypropoxy) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AJM-3)
Step 1:4- [ 2-cyclopropyl-6- (6- { [ (4S) -2, 2-dimethyl-1, 3-dioxolan-4-yl ] methoxy } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AJM-2)
To a stirred solution of compound (AJL-2) (40 mg,1 eq, 89. Mu. Mol) and (4S) -4- (chloromethyl) -2, 2-dimethyl-1, 3-dioxolane (AJM-1) (20 mg,1.5 eq, 0.13 mmol) in DMSO (5 mL) at room temperature was added K 2 CO 3 (37 mg,3 equivalents, 0.27 mmol). The resulting mixture was stirred at 120℃for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1) to give the sub-title compound (AJM-2) (28 mg, 50. Mu. Mol,56%,95% purity) as a colorless oil. M/z 563.2 (M+H) + (ES+)。
Step 2: (S) -4- (2-cyclopropyl-6- (6- (2, 3-dihydroxypropoxy) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AJM-3)
To a stirred solution of the product from step 1 above (AJM-2) (25 mg,1 eq, 44. Mu. Mol) in MeOH (4 mL) at room temperature was added dropwise aqueous HCl (4 mL, 2M). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30X 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate):60 ml/min; gradient: 13% B to 43% B in 10 minutes; wavelength: 254 nm) to give the title compound (AJM-3) (6.7 mg, 13. Mu. Mol,29%,99.8% purity) as a white solid. M/z 523.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.25–8.18(m,2H),7.97(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.60(d,J=8.4Hz,1H),7.30–7.21(m,2H),6.88(d,J=1.5Hz,1H),4.96(d,J=13.3Hz,3H),4.67(d,J=6.1Hz,1H),4.14–4.07(m,1H),4.00–3.92(m,1H),3.87–3.78(m,1H),3.47(s,5H),2.11–2.00(m,1H),0.97(d,J=6.4Hz,4H)。
Example 296: synthesis of (R) -4- (2-cyclopropyl-6- (6- (2, 3-dihydroxypropoxy) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AJN-3)
Step 1:4- [ 2-cyclopropyl-6- (6- { [ (4R) -2, 2-dimethyl-1, 3-dioxolan-4-yl ] methoxy } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AJN-2)
To a stirred solution of compound (AJL-2) (40 mg,1 eq, 89. Mu. Mol) and (4R) -4- (chloromethyl) -2, 2-dimethyl-1, 3-dioxolane (AJN-1) (21 mg,1.5 eq, 0.13 mmol) in DMSO (5 mL) at room temperature was added K 2 CO 3 (25 mg,2 equivalents, 0.18 mmol). The resulting mixture was stirred at 120℃for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1) to give the sub-title compound (AJN-2) (32 mg,57 μmol,64%,95% purity) as a yellow solid. M/z 563.2 (M+H) + (ES+)。
Step 2: (R) -4- (2-cyclopropyl-6- (6- (2, 3-dihydroxypropoxy) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AJN-3)
To a stirred solution of the product from step 1 above (AJN-2) (27 mg,1 eq, 48 μmol) in MeOH (4 mL) at room temperature was added aqueous HCl (4 mL,2 m) dropwise. The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (column: sunfire preparative C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 13% B to 43% B over 10 min; wavelength: 254 nm) to give the title compound (AJN-3) (8.2 mg,16 μmol,33%,99.8% purity) as a white solid. M/z 523.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(d,J=2.8Hz,1H),8.26–8.11(m,2H),7.98(d,J=2.5Hz,1H),7.91–7.82(m,1H),7.64–7.56(m,1H),7.30–7.14(m,2H),6.89(d,J=2.6Hz,1H),5.01–4.84(m,3H),4.73–4.63(m,1H),4.17–4.06(m,1H),3.99–3.91(m,1H),3.82(d,J=7.1Hz,1H),3.47(d,J=3.0Hz,5H),2.11–2.00(m,1H),1.01–0.84(m,4H)。
Example 297: synthesis of 2- (6-ethoxy-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (((((1-hydroxycyclobutyl) methyl) (methyl) amino) methyl) -4-methoxyisoindolin-1-one (AJO-2)
Step 1:2- (6-ethoxy-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -7-methoxy-3-oxoisoindoline-5-carbaldehyde (AJO-1)
To a stirred solution of compound (AJC-1) (50 mg,1 eq, 0.10 mmol) in DCM (5 mL) was added DMP (65 mg,1.5 eq, 0.15 mmol) at room temperature. The resulting mixture was stirred at room temperature for 1 hour. Filtering the resulting mixture; the filter cake was washed with DCM (2X 3 mL). The filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (20/1) to give the sub-title compound (AJO-1) (50 mg,0.10mmol,95%,98% pure) as an off-white solid. M/z 488.2 (M+H) + (ES+)
Step 2:2- (6-ethoxy-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- ((((1-hydroxycyclobutyl) methyl) (methyl) amino) methyl) -4-methoxyisoindolin-1-one (AJO-2)
To the product (AJO-1) (40 mg,1 equivalent, 82. Mu. Mol) from step 1 above and 1- [ (methylamino) methyl group under nitrogen at room temperature ]To a stirred solution of cyclobutan-1-ol, HCl (AEJ-1) (15 mg,1.2 eq, 98. Mu. Mol) in DCM (5 mL) was added Et 3 N (25 mg,3 eq, 0.25 mmol). The resulting mixture was stirred at room temperature under nitrogen overnight. Adding NaBH to the mixture at 0deg.C 4 (16 mg,5 eq, 0.41 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 48% B to 60% B in 8 minutes; wavelength: 254/220nm; retention time: 7.7 To give the title compound (AJO-2) (3.2 mg,5.5 μmol,6.6%,99.7% purity) as a white solid. M/z 587.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.51(s,1H),7.78(d,J=1.2Hz,1H),7.73–7.64(m,1H),7.59(t,J=8.4Hz,2H),7.37–7.24(m,2H),6.28(d,J=1.2Hz,1H),4.92(d,J=17.0Hz,3H),4.38–4.28(m,2H),3.91(s,3H),3.68(s,2H),3.43(s,3H),2.46(s,2H),2.26(s,3H),2.08–2.00(m,2H),1.96–1.85(m,2H),1.68–1.54(m,2H),1.34(t,J=7.1Hz,3H)。
Example 298: synthesis of 3' -cyclopropyl-5 ' - (6- (hydroxymethyl) -1-oxoisoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJP-7)
Step 1: 1-bromo-3-cyclopropyl-5-nitrobenzene (AJP-2)
To a stirred solution of 1-bromo-3-iodo-5-nitrobenzene (AJP-1) (500 mg,1 eq, 1.53 mmol) and cyclopropylboronic acid (ABF-2) (196 mg,1.5 eq, 2.29 mmol) in dioxane (30 mL) at room temperature under nitrogen atmosphere was added K 3 PO 4 (647 mg,2 equivalents, 3.05 mmol). Pd (dppf) Cl was added to the above mixture at room temperature under nitrogen atmosphere 2 DCM (124 mg,0.1 eq, 0.15 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/10) to give the sub-title compound (AJP-2) (400 mg,1.66mmol,94%,90% purity) as a white solid. M/z 242.0/244.0 (M+H) + (ES+)
Step 2: 3-bromo-5-cyclopropylaniline (AJP-3)
To the product (AJP-2) (900 mg,1 eq, 3.72 mmol) from step 1 above and NH under nitrogen at room temperature 4 To a stirred solution of Cl (795 mg,4 eq, 14.9 mmol) in EtOH (40 mL) was added iron powder (1.25 g,6 eq, 22.3 mmol). The resulting mixture was stirred under nitrogen at 80 ℃ for 3 hours. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/8) to give the sub-title compound (AJP-3) as an off-white solid (500 mg,2.37mmol,54%,92% purity). M/z 212.0/214.0 (M+H) + (ES+)
Step 3: 3-cyclopropyl-5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) aniline (AJP-4)
To the product (AJP-3) (500 mg,1 eq, 2.36 mmol) from step 2 above and bis (pinacolato) diboron (AAI-3) (898 mg,1.5 eq, 3.54 mm) under nitrogen at room temperatureol) to a stirred solution of dioxane (25 mL) was added KOAc (694 mg,3 eq, 7.07 mmol). Pd (dppf) Cl was added to the above mixture at room temperature under nitrogen atmosphere 2 DCM (192 mg,0.1 eq, 0.24 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/3) to give the sub-title compound (AJP-4) as an off-white solid (480 mg,1.85mmol,72%,92% purity). M/z 260.2 (M+H) + (ES+)
Step 4:3' -amino-5 ' -cyclopropyl-2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJP-5)
To a stirred solution of 4-bromo-3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ADG-1) (360 mg,1 eq, 1.37 mmol) and the product from step 3 above (AJQ-4) (390 mg,1.1 eq, 1.51 mmol) in dioxane (15 mL) at room temperature under nitrogen atmosphere was added K 2 CO 3 (567 mg,3 equivalents, 4.10 mmol). Pd (dppf) Cl was added to the above mixture at room temperature under nitrogen atmosphere 2 DCM (111 mg,0.1 eq, 0.14 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 3 hours. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (20/1) to give the sub-title compound (AJQ-5) (220 mg,0.70mmol,46%,91% purity) as a pale brown solid. M/z 316.1 (M+H) + (ES+)
Step 5:3' -bromo-5 ' -cyclopropyl-2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJP-6)
A solution of CuBr (150 mg,1.5 eq., 1.05 mmol) and t-BuONO (432 mg,6 eq., 4.19 mmol) in MeCN (12 mL) was stirred under nitrogen at 0deg.C for 1. To the above mixture was added MeCN containing the product (AJP-5) from step 4 above (220 mg,1 eq, 0.70 mmol) under nitrogen at 0 ℃. The resulting mixture was stirred under nitrogen at 50 ℃ for 1 hour. Filtering the resulting mixture; the filter cake was washed with MeOH (3X 5 mL). The filtrate was concentrated in vacuo. Applying the residue to a substrate having DC M/MeOH (5/1) to give the sub-title compound (AJP-6) (165 mg,0.44mmol,53%,94% purity) as a brown yellow solid. M/z 379.0/381.0 (M+H) + (ES+)
Step 6:3' -cyclopropyl-5 ' - (6- (hydroxymethyl) -1-oxoisoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJP-7)
Cs was added to a stirred solution of the product (AJP-6) (40 mg,1 eq, 0.11 mmol) from step 5 above and intermediate (AGH-1) (21 mg,1.2 eq, 0.13 mmol) in dioxane (5 mL) at room temperature under nitrogen atmosphere 2 CO 3 (69 mg,2 equivalents, 0.21 mmol). RuPhos (20 mg,0.4 eq, 42. Mu. Mol) and RuPhos ring palladium complex Gen.3 (18 mg,0.2 eq, 21. Mu. Mol) were added to the above mixture under nitrogen atmosphere at room temperature. The resulting mixture was stirred under nitrogen at 100 ℃ for an additional 2 hours. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 40% B in 8 minutes; wavelength: 254/220nm; retention time: 7.67 To give the title compound (AJP-7) (4.1 mg, 8.9. Mu. Mol,8.4%,99.8% purity) as a white solid. M/z 462.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.20–8.15(m,1H),8.10(d,J=1.7Hz,1H),7.87(d,J=8.1Hz,1H),7.77–7.70(m,2H),7.63–7.56(m,3H),6.44(t,J=1.6Hz,1H),5.39(t,J=5.8Hz,1H),4.92(s,2H),4.62(d,J=5.7Hz,2H),3.12(s,3H),1.92–1.83(m,1H),0.97–0.84(m,2H),0.56–0.46(m,2H)。
Example 299: synthesis of 3' -cyclopropyl-5 ' - (6- (((2-methoxyethyl) amino) methyl) -1-oxoisoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJQ-2)
Step 1:3' -cyclopropyl-5 ' - (6-formyl-1-oxo-3H-isoindol-2-yl) -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJQ-1)
To a stirred solution of compound (AJP-7) (30 mg,1 eq, 65. Mu. Mol) in DCM (5 mL) under nitrogen at room temperature was added DMP (41 mg,1.5 eq, 98. Mu. Mol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 1 hour. The resulting mixture was concentrated in vacuo. Filtering the resulting mixture; the filter cake was washed with MeOH (3X 2 mL). The filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AJQ-1) (28 mg,61 μmol,89%,95% purity) as a pale yellow solid. M/z 460.2 (M+H) + (ES+)
Step 2:3' -cyclopropyl-5 ' - (6- (((2-methoxyethyl) amino) methyl) -1-oxoisoindolin-2-yl) -2- (4-methyl-4H-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJQ-2)
To a stirred solution of the product (AJQ-1) (30 mg,1 eq, 65. Mu. Mol) from step 1 above and 2-methoxyethyl-1-amine (AEB-1) (7 mg,1.5 eq, 98. Mu. Mol) in DCM (4 mL) at room temperature under nitrogen was added Et 3 N (20 mg,3 eq, 0.20 mmol). The resulting mixture was stirred at room temperature under nitrogen overnight. NaBH was added to the above mixture at room temperature 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 25% B to 45% B in 9 minutes; wavelength: 254/220nm; retention time: 8.33 To give the title compound (AJQ-2) (12.0 mg, 23. Mu. Mol,35%,99.1% purity) as a white solid. M/z 519.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.19–8.14(m,1H),8.10(d,J=1.8Hz,1H),7.87(d,J=8.1Hz,1H),7.77–7.67(m,2H),7.65–7.60(m,1H),7.60–7.54(m,2H),6.45(d,J=1.6Hz,1H),4.91(s,2H),3.82(s,2H),3.41(t,J=5.7Hz,2H),3.20(s,3H),3.12(s,3H),2.64(t,J=5.7Hz,2H),1.93–1.82(m,1H),0.98–0.87(m,2H),0.58–0.47(m,2H)。
Example 300: synthesis of 3' -cyclopropyl-5 ' - (6- { [ (cyclopropylmethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (AJR-1)
Et is added to a stirred solution of intermediate (AJQ-1) (30 mg,1 eq, 65. Mu. Mol) and 1-cyclopropylmethylamine (AIB-1) (7 mg,1.5 eq, 98. Mu. Mol) in DCM (4 mL) at room temperature under nitrogen 3 N (20 mg,3 eq, 0.20 mmol). The resulting mixture was stirred at room temperature under nitrogen overnight. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 33% B to 48% B in 8 minutes; wavelength: 254/220nm; retention time: 7.77 To give the title compound (AJR-1) (11.4 mg, 22. Mu. Mol,34%,98.9% purity) as a white solid. M/z 515.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.20–8.15(m,1H),8.10(d,J=1.8Hz,1H),7.87(d,J=8.1Hz,1H),7.77–7.73(m,2H),7.66–7.62(m,1H),7.62–7.47(m,2H),6.45(t,J=1.6Hz,1H),4.91(s,2H),3.85(s,2H),3.12(s,3H),2.39(d,J=6.7Hz,2H),1.93–1.82(m,1H),1.06–0.79(m,3H),0.57–0.46(m,2H),0.46–0.32(m,2H),0.11–0.02(m,2H)。
Example 301: synthesis of 2- (6-cyclopropyl-4- (4-fluoro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) pyridin-2-yl) -6- (((((1-hydroxycyclobutyl) methyl) (methyl) amino) methyl) isoindolin-1-one (AJS-1)
A solution of compound (AFH-1) (20 mg,1 eq, 37. Mu. Mol) and paraformaldehyde (10 mg,3 eq, 0.11 mmol) in MeOH (2 mL) was stirred at 50deg.C, then cooled, and NaBH was added at room temperature 3 CN (7 mg,3 equivalents, 0.11 mmol). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 8% B to 38% B over 10 min; wavelength: 254nm; retention time: 8.38) to give the title compound (AJS-1) (6.2 mg, 11. Mu. Mol,28%,95.7% purity) as a white solid. M/z 553.3 (M+H) + (ES+). 1 H NMR(300MHz,DMSO-d6)δ8.50(s,1H),8.13(s,0.324H),7.97(d,J=1.4Hz,1H),7.81–7.44(m,6H),6.81(d,J=1.4Hz,1H),5.02(s,2H),3.77(s,2H),3.43(s,3H),2.49(s,2H),2.28(s,3H),2.08–1.96(m,3H),1.92(d,J=9.8Hz,2H),1.62(d,J=10.4Hz,1H),1.43–1.26(m,1H),0.95(d,J=6.4Hz,4H)。
Example 302: synthesis of 2- { 6-ethoxy-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] (methyl) amino } methyl) -3H-isoindol-1-one (AJT-1)
Compound (AGR-1) (30 mg,to a stirred mixture of 1 eq, 55. Mu. Mol) and formaldehyde (5 mg,3 eq, 0.17 mmol) in MeOH (10 mL) was added NaBH 3 CN (7 mg,2 equivalents, 0.11 mmol). The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 70% B in 8 minutes; wavelength: 254/220nm; retention time: 6.25 To give the title compound (AJT-1) (3.9 mg, 7.0. Mu. Mol,13%,99.5% purity) as a white solid. M/z 557.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.51(s,1H),7.80(d,J=1.2Hz,1H),7.76–7.67(m,2H),7.65(d,J=1.7Hz,2H),7.61–7.53(m,2H),6.28(d,J=1.2Hz,1H),5.06(s,2H),4.86(s,1H),4.37–4.28(m,2H),3.69(s,2H),3.43(s,3H),2.47(s,2H),2.22(s,3H),2.03(d,J=12.1,8.8,3.2Hz,2H),1.90(d,J=12.0,9.3Hz,2H),1.61(m,1H),1.34(t,J=7.1Hz,4H)。
Example 303: synthesis of 4- (2-cyclopropyl-6- {6- [ (2S) -2-hydroxy-3- (methylamino) propoxy ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AJU-3)
Step 1:4- (2-cyclopropyl-6- {6- [ (2S) -oxaprop-2-ylmethoxy ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AJU-2)
To a stirred solution of intermediate (AJL-2) (60 mg,1 eq, 0.13 mmol) and (S) -epichlorohydrin (AJU-1) (19 mg,1.5 eq, 0.20 mmol) in DMSO (5 mL) at room temperature was added K 2 CO 3 (37 mg,2 equivalents, 0.27 mmol). The resulting mixture was stirred at 60℃for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). Will be combinedThe organic layer was washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1) to give the sub-title compound (AJU-2) (45 mg,89 μmol,67%,95% purity) as a yellow solid. M/z 505.2 (M+H) + (ES+)。
Step 2:4- (2-cyclopropyl-6- {6- [ (2S) -2-hydroxy-3- (methylamino) propoxy ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AJU-3)
To a stirred solution of the product from step 1 above (AJU-2) (42 mg,1 eq, 83. Mu. Mol) and methylamine (4 mg,1.5 eq, 0.12 mmol) in MeOH (5 mL) was added DIPEA (32 mg,3 eq, 0.25 mmol) at room temperature. The resulting mixture was stirred at 60℃for 12 hours. The mixture was cooled to room temperature and then concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 30% B over 10 min; wavelength: 254nm; retention time: 10.6) to give the title compound (AJU-3) (18.9 mg, 35. Mu. Mol,42%,9.6% purity) as a white solid. M/z 536.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.39(s,0.506H),8.20(d,J=8.2Hz,2H),7.96(d,J=1.5Hz,1H),7.87(d,J=7.9Hz,1H),7.62(d,J=8.1Hz,1H),7.28(d,J=8.7Hz,2H),6.90(d,J=1.5Hz,1H),4.95(s,2H),4.13–3.98(m,3H),3.46(s,3H),2.97–2.90(m,1H),2.86–2.79(m,1H),2.47(s,3H),2.11–1.09(m,1H),0.97(d,J=5.3Hz,4H)。
Example 304: synthesis of 4- (2-cyclopropyl-6- {6- [ (2R) -2-hydroxy-3- (methylamino) propoxy ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AJV-3)
Step 1:4- (2-cyclopropyl-6- {6- [ (2R) -oxaprop-2-ylmethoxy ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AJV-2)
To a stirred solution of intermediate (AJL-2) (100 mg,1 eq, 0.22 mmol) and (R) -epichlorohydrin (AJV-1) (31 mg,1.5 eq, 0.34 mmol) in DMSO (5 mL) at room temperature was added K 2 CO 3 (62 mg,2 equivalents, 0.45 mmol). The resulting mixture was stirred at 60℃for 1 hour. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1) to give the sub-title compound (AJV-2) (63 mg,0.12mmol,56% pure) as a yellow solid. M/z 505.2 (M+H) + (ES+)。
Step 2:4- (2-cyclopropyl-6- {6- [ (2R) -2-hydroxy-3- (methylamino) propoxy ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AJV-3)
To a stirred solution of the product from step 1 above (AJV-2) (39 mg,1 eq, 77. Mu. Mol) and methylamine (3.6 mg,1.5 eq, 0.12 mmol) in MeOH (5 mL) was added DIPEA (30 mg,3 eq, 0.23 mmol) at room temperature. The resulting mixture was stirred at 60℃for 12 hours. The mixture was cooled to room temperature and then concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 32% B over 10 min; wavelength: 254 nm) to give the title compound (AJV-3) (20.1 mg, 37. Mu. Mol,48%,99.6% purity) as a white solid. M/z 536.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.39(s,0.549H),8.20(d,J=7.7Hz,2H),7.96–7.87(m,2H),7.62(d,J=8.1Hz,1H),7.31–7.24(m,2H),6.89(d,J=1.4Hz,1H),4.95(s,2H),4.11–3.98(m,3H),3.46(s,3H),2.97–2.90(m,1H),2.84–2.80(m,1H),2.47(s,3H),2.10–2.00(m,1H),1.01–0.90(m,4H)。
Example 305: synthesis of 2- { 6-cyclopropyl-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] (methyl) amino } methyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AJW-1)
To a stirred mixture of compound (ADR-3) (30 mg,1 eq, 51. Mu. Mol) and formaldehyde (5 mg,3 eq, 0.15 mmol) in MeOH (10 mL) at room temperature was added NaBH 3 CN (6.4 mg,2 equivalents, 0.10 mmol). The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 52% B in 10 minutes; wavelength: 254/220nm; retention time: 9.12 To give the title compound (AJW-1) (3.1 mg, 5.1. Mu. Mol,10%,99.4% purity) as a white solid. M/z 607.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.18(d,J=1.3Hz,1H),8.02(d,J=10.0Hz,2H),7.96(s,1H),7.22(d,J=1.3Hz,1H),5.20(s,2H),4.94(s,1H),3.91(s,3H),3.80(s,2H),3.54(s,3H),2.49(s,2H),2.25(s,3H),2.19–2.08(m,1H),2.02(t,J=8.9,3.2Hz,2H),1.90(d,J=12.2,9.3Hz,2H),1.61(m,1H),1.33(d,J=10.8,8.6Hz,1H),1.03(d,J=6.4Hz,4H)。
Example 306: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (1-fluorocyclobutyl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AJX-1)
Intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and 1- (1-fluorocyclobutyl) methylamine (AFP-1) (8 mg,1.2 eq,to a stirred solution of 78. Mu. Mol in MeOH (8 mL) was added DIPEA (25 mg,3 eq., 0.20 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 46% B to 60% B in 8 minutes; wavelength: 254/220nm; retention time: 7.73 To give the title compound (AJX-1) (4.3 mg,7.8 μmol,12%,99.3% purity) as a white solid. M/z 548.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.55(s,1H),δ8.16–8.03(m,3H),7.89–7.86(m,1H),7.77(s,1H),7.64(d,J=2.2Hz,2H),6.90(d,J=1.4Hz,1H),5.00(s,2H),3.91(s,2H),3.49(s,3H),3.28–3.24(m,1H),3.23–3.18(m,1H),2.73(s,4H),2.21–2.13(m,1H),2.08–2.00(m,1H),1.72–1.63(m,1H),1.01–0.94(m,4H)。
Example 307: synthesis of 4- [2- (6- { [ (cyclopentylmethyl) (methyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AJY-1)
To a stirred mixture of compound (AIG-2) (30 mg,1 eq, 55. Mu. Mol) and formaldehyde (2.5 mg,1.5 eq, 83. Mu. Mol) in MeOH (10 mL) at room temperature was added NaBH 3 CN (7 mg,2 equivalents, 0.11 mmol). The resulting mixture was stirred at room temperature for 2 hours. The mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 70% B to 82% B in 8 minutes; wavelength: 254/220nm; retention time: 7.15 To give the title compound (AJY-1) (5.3 mg,9.5 μmol,17%,99.8% purity) as a white solid. M/z 558.2 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.21(d,J=3.5,1.7Hz,2H),7.99(d,J=1.3Hz,1H),7.87(d,J=8.5Hz,1H),7.70-7.58(m,3H),6.87(d,J=1.3Hz,1H),5.01(s,2H),3.51(d,J=33.7Hz,5H),2.27-2.00(m,7H),1.76-1.65(m,2H),1.49(q,J=8.1,6.5Hz,4H),1.18(d,J=13.7,4.1Hz,2H),0.97(d,J=6.4Hz,4H)。
Example 308: synthesis of 4- (2- (6- (1- ((cyclobutylmethyl) amino) ethyl) -1-oxoisoindolin-2-yl) -6-cyclopropylpyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AJZ-1)
Intermediate (AJH-3) (90 mg,1 eq, 0.19 mmol), cyclobutylmethylamine, HCl (AIA-1) (19 mg,1.2 eq, 0.23 mmol) and Ti (Oi-Pr) 4 A solution of (216 mg,4 eq, 0.76 mmol) in THF (12 mL) was stirred overnight at 60 ℃. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (36 mg,5 equivalents, 0.95 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 46% B to 60% B in 8 minutes; wavelength: 254/220nm; retention time: 7.73 To give the title compound (AJZ-1) (2.0 mg, 3.7. Mu. Mol,1.9%,99.8% purity) as a white solid. M/z 544.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4))δ8.50(s,1H),8.14–8.04(m,3H),7.92(d,J=8.0Hz,1H),7.79(d,J=1.6Hz,1H),7.68–7.59(m,2H),6.92(d,J=1.5Hz,1H),5.03(s,2H),3.93–3.84(m,1H),3.50(s,3H),2.54–2.34(m,3H),2.11–1.99(m,3H),1.94–1.74(m,2H),1.68–1.54(m,2H),1.40(d,J=6.6Hz,3H),1.28(s,1H),1.05–0.99(m,3H)。
Example 309: synthesis of (R) -4- (2- (6- (1- ((cyclobutylmethyl) amino) ethyl) -1-oxoisoindolin-2-yl) -6-cyclopropylpyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AKA-1)
The crude product (AJZ-1) (30 mg,1 eq., 55. Mu. Mol) was purified by preparative HPLC under the following conditions (column: CHIRAL ART Amylose-SA, 2X 25cm,5 μm; mobile phase A: mtBE (0.5% 2M NH) 3 MeOH) -HPLC, mobile phase B: etOH-HPLC; flow rate: 20 ml/min; gradient: 50% B to 50% B within 25 minutes; wavelength: 220/254nm; retention time 1 (min): 15.086, sample solvent: etOH-HPLC; injection volume: 1.5 mL) to give the title compound (AKA-1) (9.5 mg, 17. Mu. Mol,32%,99.6% purity) as a white solid. M/z 544.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.05 (m, 3H), 7.92 (d, j=8.0 hz, 1H), 7.80-7.76 (m, 1H), 7.68-7.60 (m, 2H), 6.92 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 3.91-3.84 (m, 1H), 3.50 (s, 3H), 2.52-2.33 (m, 3H), 2.10-1.99 (m, 3H), 1.95-1.84 (m, 1H), 1.84-1.72 (m, 1H), 1.68-1.54 (m, 2H), 1.40 (d, j=6.6 hz, 3H), 1.07-1.01 (m, 2H), 1.01-0.96 (m, 2H). Column: CHIRALPAK IA-3,4.6 x 50mm,3 μm; mobile phase a: mtBE (0.1% DEA): etoh=50:50; flow rate: 1 ml/min; retention time: 2.565.
example 310: synthesis of (S) -4- (2- (6- (1- ((cyclobutylmethyl) amino) ethyl) -1-oxoisoindolin-2-yl) -6-cyclopropylpyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AKB-1)
The crude product (AJZ-1) (30 mg,1 eq., 55. Mu. Mol) was purified by preparative HPLC under the following conditions (column: CHIRAL ART Amylose-SA, 2X 25cm,5 μm; mobile phase A: mtBE (0.5% 2M NH) 3 MeOH) -HPLC, mobile phase B: etOH-HPLC; flow rate: 20 ml/min; gradient: 50% B to 50% B within 25 minutes; wavelength: 220/254nm; retention time 2 (minutes): 24.157; sample solvent: etOH-HPLC; injection volume: 1.5 mL) to give the title compound (AKB-1) (9.8 mg, 18. Mu. Mol,33%,98.5% purity) as a white solid. M/z 544.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.04 (m, 3H), 7.93 (d, j=8.0 hz, 1H), 7.81 (d, j=1.3 hz, 1H), 7.70-7.62 (m, 2H), 6.93 (d, j=1.5 hz, 1H), 5.04 (s, 2H), 3.97 (d, j=6.8 hz, 1H), 3.50 (s, 3H), 2.61-2.54 (m, 1H), 2.53-2.40 (m, 2H), 2.11-2.00 (m, 3H), 1.95-1.86 (m, 1H), 1.85-1.75 (m, 1H), 1.70-1.58 (m, 2H), 1.45 (d, j=6.7 hz, 1H), 1.29 (s, 3H), 1.61-2.54 (m, 1H), 2.53-2.40 (m, 1.02 (m, 2H), 1.02-1.96 (m, 2H). Column: CHIRALPAK IA-3,4.6 x 50mm,3 μm; mobile phase a: mtBE (0.1% DEA): etoh=50:50; flow rate: 1 ml/min; retention time: 3.845.
example 311: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (3, 3-difluorocyclobutyl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKC-2)
A solution of 1- (3, 3-difluorocyclobutyl) methylamine (AKC-1) (11.8 mg,1.5 eq, 98. Mu. Mol) and DIPEA (10 mg,1.2 eq, 78. Mu. Mol) in MeOH (3 mL) was stirred at room temperature for 10 min. To the above mixture was added intermediate (AGT-2) (30 mg,1 equivalent, 65. Mu. Mol) at room temperature. The resulting mixture was stirred at 60 ℃ for an additional 1 hour. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (5 mg,2 equivalents, 0.13 mmol). The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. Will beThe crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 55% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AKC-2) (7.4 mg, 13. Mu. Mol,20%,99.5% purity) as a white solid. M/z 566.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.22–8.19(m,2H),7.99(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.74–7.64(m,3H),6.88(d,J=1.5Hz,1H),5.00(s,2H),3.80(s,2H),3.47(s,3H),2.65–2.57(m,4H),2.31–2.20(m,3H),2.07–2.01(m,1H),0.99–0.94(m,4H)。
Example 312: synthesis of 4- (2- {6- [ ({ bicyclo [1.1.1] pentan-1-ylmethyl } amino) methyl ] -1-oxo-3H-isoindol-2-yl } -6-cyclopropylpyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKD-2)
To intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and 1- { bicyclo [1.1.1 at room temperature]To a stirred solution of pentane-1-yl } methylamine (AKD-1) (13 mg,2 eq, 0.13 mmol) in MeOH (2 mL) was added DIPEA (51 mg,6 eq, 0.39 mmol). The resulting mixture was stirred at room temperature under nitrogen overnight. Adding NaBH to the mixture at 0deg.C 4 (7 mg,3 equivalents, 0.20 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 30% B in 10 min; wavelength: 254nm; retention time: 11.28) to give the title compound (AKD-2) (2.3 mg, 4.2. Mu. Mol,6.0%,99.7% purity) as a white solid. M/z 542.1 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.43(s,0.909H),8.25–8.18(m,2H),7.99(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.73(s,1H),7.62(d,J=1.2Hz,2H),6.88(d,J=1.4Hz,1H),5.00(s,2H),3.79(s,2H),3.48(s,3H),2.47(d,J=3.3Hz,3H),2.09–2.00(m,1H),1.68(s,6H),1.00–0.93(m,4H)。
Example 313: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (1-methoxycyclopentyl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKE-2)
DIPEA (34 mg,4 eq, 0.26 mmol) was added to a stirred solution of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and 1- (1-methoxycyclopentyl) methylamine (AKE-1) (13 mg,1.5 eq, 98. Mu. Mol) in MeOH (6 mL) at room temperature. The resulting mixture was stirred at room temperature under nitrogen overnight. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 30% B in 10 min; wavelength: 254nm; retention time: 9.8) to give the title compound (AKE-2) (11.6 mg, 20. Mu. Mol,31%,99.7% purity) as a white solid. M/z 574.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.16-8.10 (m, 2H), 8.09-8.03 (m, 1H), 7.98-7.92 (m, 1H), 7.90-7.85 (m, 1H), 7.77-7.70 (m, 1H), 7.68-7.64 (m, 1H), 6.94 (d, J=1.4 Hz, 1H), 5.05 (s, 2H), 4.06 (s, 2H), 3.49 (s, 3H), 3.08 (s, 3H), 2.81 (s, 2H), 2.09-2.00 (m, 1H), 1.90-1.80 (m, 2H), 1.74-1.64 (m, 2H), 1.66-1.56 (m, 2H), 1.55-1.44 (m, 2H), 1.09-0.94 (m, 4H).
Example 314: synthesis of 4- {2- [6- ({ [ (1-cyanocyclobutyl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] -6-cyclopropylpyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKF-2)
To a 25mL round bottom flask was added MeOH (5 mL) containing intermediate (AGT-2) (60 mg,1 eq, 0.13 mmol), 1- (aminomethyl) cyclobutane-1-carbonitrile (AKF-1) (22 mg,1.5 eq, 0.20 mmol) at room temperature. DIPEA (51 mg,3 eq, 0.39 mmol) was added to the above mixture at room temperature. The resulting mixture was stirred at 60 ℃ for an additional overnight. Adding NaBH to the mixture at 0deg.C 4 (25 mg,5 eq, 0.65 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 48% B to 52% B in 9 minutes; wavelength: 254 nm) to give the title compound (AKF-2) as a white solid (4.1 mg, 7.3. Mu. Mol,5.8%,99.6% purity). M/z 555.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.14-8.08 (m, 2H), 8.05 (d, j=1.4 hz, 1H), 7.95-7.91 (m, 1H), 7.84 (s, 1H), 7.74-7.69 (m, 1H), 7.62 (s, 1H), 6.93 (d, j=1.5 hz, 1H), 5.03 (s, 2H), 3.96 (s, 2H), 3.51 (s, 3H), 2.88 (s, 2H), 2.47-2.41 (m, 2H), 2.23-2.15 (m, 3H), 2.06-2.01 (m, 2H), 1.06-0.97 (m, 4H).
Example 315: synthesis of 4- { 2-cyclopropyl-6- [ 4-fluoro-6- ({ [ (2S) -1-methoxyprop-2-yl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKG-1)
Intermediate (AIO-8) (30 mg,1 eq, 63. Mu. Mol) and (2S) -1-methoxypropan-2-amine (AIR-1) (7 mg,1.2To a stirred solution of 76. Mu. Mol in MeOH (8 mL) was added DIPEA (24 mg,3 eq., 0.19 mmol). The resulting mixture was stirred at 60℃for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 equivalents, 0.32 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 42% B to 52% B in 8 minutes; wavelength: 254/220nm; retention time: 7.94 To give the title compound (AKG-1) (1.6 mg,2.9 μmol,4.5%,97.8% purity) as a white solid. M/z 552.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.08 (m, 2H), 8.03 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.66 (s, 1H), 7.47-7.43 (m, 1H), 6.95 (d, j=1.5 hz, 1H), 5.06 (s, 2H), 3.96 (d, j=13.8 hz, 1H), 3.86 (d, j=13.8 hz, 1H), 3.50 (s, 3H), 3.36-3.33 (m, 5H), 2.91-2.85 (m, 1H), 2.08-2.01 (m, 1H), 1.08-1.03 (m, 5H), 1.02-0.97 (m, 2H).
Example 316: synthesis of 4- { 2-cyclopropyl-6- [ 4-fluoro-6- ({ [ (2R) -1-methoxyprop-2-yl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKH-1)
To a 25mL round bottom flask was added MeOH (5 mL) containing intermediate (AIO-8) (40 mg,1 eq, 84. Mu. Mol), (2R) -1-methoxypropan-2-amine (AIK-1) (11 mg,1.5 eq, 0.13 mmol) at room temperature. Et is added to the above mixture at room temperature 3 N (25 mg,3 eq, 0.25 mmol). The resulting mixture was stirred at 60 ℃ for an additional overnight. Adding NaBH to the mixture at 0deg.C 4 (16 mg,5 eq, 0.42 mmol). The resulting mixture was stirred at room temperature for an additional 1 hourWhen (1). The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XBridge prep. OBD C18 column, 19 x 250mm,5 μm; mobile phase a: water (0.1% nh) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 25 ml/min; gradient: 45% B to 55% B in 8 minutes; wavelength: 254 nm) to give the title compound (AKH-1) (10.7 mg, 19. Mu. Mol,23%,99.9% purity) as a white solid. M/z 552.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.08 (m, 2H), 8.02 (d, j=1.4 hz, 1H), 7.92 (d, j=8.1 hz, 1H), 7.66 (d, j=1.2 hz, 1H), 7.49-7.44 (m, 1H), 6.95 (d, j=1.4 hz, 1H), 5.07 (s, 2H), 3.99 (d, j=13.8 hz, 1H), 3.90 (d, j=13.8 hz, 1H), 3.49 (s, 3H), 3.40-3.32 (m, 5H), 2.96-2.87 (m, 1H), 2.10-2.01 (m, 1H), 1.10 (d, j=6.5 hz, 3H), 1.06-0.98 (m, 4H).
Example 317: synthesis of 2- { 6-cyclopropyl-4- [ 2-methyl-4- (4-methyl-1, 2, 4-triazol-3-yl) pyrazol-3-yl ] pyridin-2-yl } -6- (hydroxymethyl) -4- (trifluoromethyl) -3H-isoindol-1-one (AKI-1)
To intermediate (ADR-1) (100 mg,1 equivalent, 0.32 mmol), intermediate (ADX-1) (88 mg,1.2 equivalent, 0.38 mmol) and Cs under nitrogen at room temperature 2 CO 3 To a stirred mixture of (311 mg,3 eq, 0.95 mmol) in dioxane (15 mL) was added XPhos Pd G3 (54 mg,0.2 eq, 64. Mu. Mol) and XPhos (61 mg,0.4 eq, 0.13 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC under the following conditions (column: XBridge preparative OBD C18 column, 30X 150mm,5 μm; stream)Mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 33% B to 43% B in 9 minutes; wavelength: 254/220nm; retention time: 8.47 To give the title compound (AKI-1) as a white solid (6.7 mg,13 μmol,4.1%,99.6% purity). M/z 510.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.19(d,J=1.3Hz,1H),8.04–7.93(m,3H),7.23(d,J=1.4Hz,1H),5.58(t,J=5.8Hz,1H),5.20(s,2H),4.70(d,J=5.6Hz,2H),3.90(s,3H),3.55(s,3H),2.14(p,J=6.5Hz,1H),1.03(d,J=5.6Hz,4H)。
Example 318: synthesis of 4- { 2-ethoxy-6- [6- ({ [ (1-fluorocyclobutyl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKJ-1)
A solution of 1- (1-fluorocyclobutyl) methylamine (AFP-1) (11 mg,1.2 eq, 0.10 mmol) and DIPEA (45 mg,4 eq, 0.34 mmol) in MeOH (8 mL) was stirred at room temperature for 5 min. Intermediate (AHS-1) (40 mg,1 eq, 86. Mu. Mol) was added to the above mixture at room temperature. The resulting mixture was stirred at 60 ℃ for an additional overnight. Adding NaBH to the above mixture at 0deg.C 4 (16 mg,5 equivalents, 0.43 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 54% B in 9 minutes; wavelength: 254/220nm; retention time: 8.13 To give the title compound (AKJ-1) (6.0 mg,11 μmol,12%,98.7% purity) as a white solid. M/z 552.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.14-8.05 (m, 2H),7.92–7.80(m,3H),7.71–7.66(m,1H),7.61(d,J=7.8Hz,1H),6.42(d,J=1.3Hz,1H),5.06(s,2H),4.44–4.34(m,2H),3.93(s,2H),3.53(s,3H),2.85(s,1H),2.79(s,1H),2.35–2.12(m,4H),1.88–1.74(m,1H),1.55–1.41(m,1H),1.39(t,J=7.1Hz,3H)。
example 319: synthesis of 4- { 2-cyclopropyl-6- [ 4-fluoro-6- ({ [ (1-fluorocyclobutyl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKK-1)
To a stirred solution of intermediate (AIO-8) (30 mg,1 eq, 63. Mu. Mol) and 1- (1-fluorocyclobutyl) methylamine (AFP-1) (10 mg,1.5 eq, 95. Mu. Mol) in MeOH (6 mL) at room temperature was added DIPEA (49 mg,6 eq, 0.38 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 equivalents, 0.32 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 15% B to 35% B in 10 min; wavelength: 254nm; retention time: 9.43) to give the title compound (AKK-1) (9.0 mg, 16. Mu. Mol,25%,99.9% purity) as a white solid. M/z 566.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.25–8.18(m,2H),7.95(d,J=1.4Hz,1H),7.91–7.84(m,1H),7.64(s,1H),7.53(d,J=10.0Hz,1H),6.92(d,J=1.4Hz,1H),5.07(s,2H),3.87(s,2H),3.51–3.47(m,3H),2.74–2.68(m,2H),2.20–2.10(m,4H),2.01(s,1H),1.74(s,1H),1.51–1.32(m,1H),0.98(d,J=8.2Hz,4H)。
Example 320: synthesis of 4- { 2-cyclopropyl-6- [6- (hydroxymethyl) -5-methoxy-1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKL-2)
Step 1:6- (hydroxymethyl) -5-methoxy-2, 3-dihydro-isoindol-1-one (AKL-1)
Intermediate (AEM-7) (40 mg,1 eq, 0.21 mmol) and NaBH 4 A solution of (16 mg,2 eq, 0.42 mmol) in MeOH (10 mL) was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1) to give the sub-title compound (AKL-1) (20 mg,0.10mmol,49% pure) as a white solid. M/z 194.1 (M+H) + (ES+)
Step 2:4- { 2-cyclopropyl-6- [6- (hydroxymethyl) -5-methoxy-1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKL-2)
To intermediate (AEG-2) (20 mg,1 equivalent, 60. Mu. Mol), the product from step 1 above (AKL-1) (12.7 mg,1.1 equivalent, 66. Mu. Mol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 (58 mg,3 eq, 0.18 mmol) to a solution of RuPhos (11 mg,0.4 eq, 24. Mu. Mol) and RuPhos cyclopalladium complex Gen.3 (10 mg,0.2 eq, 12. Mu. Mol) were added to a solution of 1, 4-dioxane (4 mL). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 48% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AKL-2) (9.2 mg, 18. Mu. Mol,31%,99.8% purity) as a white solid. M/z 493.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.22–8.19(m,2H),7.99(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.68(d,J=8.4Hz,1H),7.17(d,J=8.5Hz,1H),6.80(d,J=1.4Hz,1H),5.14–5.09(m,3H),4.67(d,J=5.5Hz,2H),3.89(s,3H),3.45(s,3H),2.07–2.01(m,1H),0.99–0.90(m,4H)。
Example 321: synthesis of 4- [2- (6- { [ (cyclobutylmethyl) amino ] methyl } -5-methoxy-1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKM-2)
Step 1:4- [ 2-cyclopropyl-6- (6-formyl-5-methoxy-1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKM-1)
A solution of compound (AKL-2) (40 mg,1 eq, 81. Mu. Mol) and DMP (69 mg,2 eq, 0.16 mmol) in DCM (6 mL) was stirred at room temperature for 1 hour. The resulting mixture was filtered and the filter cake was washed with DCM (3X 5 mL). The filtrate was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 491.2 (M+H) + (ES+)
Step 2:4- [2- (6- { [ (cyclobutylmethyl) amino ] methyl } -5-methoxy-1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKM-2)
A solution of 1-cyclobutylmethylamine, HCl (AIA-1) (12 mg,1.2 eq., 98. Mu. Mol) and DIPEA (16 mg,1.5 eq., 0.12 mmol) in MeOH (9 mL) was stirred at room temperature for 10 min. To the above mixture was added the product from step 1 (AKM-1) (40 mg,1 eq., 82. Mu. Mol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (6 mg,2 equivalents, 0.16 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), flowMobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 55% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AKM-2) (14.9 mg, 27. Mu. Mol,32%,98.9% purity) as a white solid. M/z 560.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(t,J=6.6Hz,1H),8.22–8.18(m,2H),7.97(d,J=1.4Hz,1H),7.86(d,J=8.6Hz,1H),7.65(d,J=8.4Hz,1H),7.17(d,J=8.4Hz,1H),6.84(d,J=1.4Hz,1H),5.06(s,2H),3.89(s,3H),3.79(s,2H),3.45(s,3H),2.54(d,J=7.0Hz,2H),2.47–2.40(m,1H),2.06–1.97(m,3H),1.88–1.77(m,2H),1.70–1.61(m,2H),1.04–0.89(m,4H)。
Example 322: synthesis of 4- [2- (5-chloro-6-formyl-1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKN-12)
Step 1: 4-chloro-2-methyl-5-nitrobenzonitrile (AKN-2)
4-chloro-2-methylbenzonitrile (AKN-1) (20.0 g,1 eq., 132 mmol) and KNO 3 (13.3 g,1 eq, 132 mmol) in concentrated H 2 SO 4 The mixture in (140 mL) was stirred at 0deg.C for 2 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (10/1) to give the crude product which was recrystallized to give the sub-title compound (AKN-2) as a white solid (20.0 g,102mmol,77%,90% purity). M/z 197.0/199.0 (M+H) + (ES+)
Step 2: 5-amino-4-chloro-2-methylbenzonitrile (AKN-3)
A mixture of the product (AKN-2) from step 1 above (20.0 g,1 eq, 102 mmol) and Fe (28.4 g,5 eq, 509 mmol) in AcOH (48 mL,8.17 eq, 831 mmol), etOH (200 mL) and water (100 mL) was stirred at room temperature for 3 hours. The resulting mixture was diluted with water and treated with E tOAc (3X 200 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo to give the sub-title compound (AKN-3) (8.0 g,48.2mmol,47%,92% purity) as a yellow solid. M/z167.0/169.0 (M+H) + (ES+)
Step 3: 5-bromo-4-chloro-2-methylbenzonitrile (AKN-4)
The product (AKN-3) (8.0 g,1 eq., 48.0 mmol) from step 2 above, cuBr 2 A mixture of (16.1 g,1.5 eq, 72.0 mmol) and tBu-nitrite (9.90 g,2 eq, 96.0 mmol) in MeCN (133 mL) was stirred at room temperature for 3 hours. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (10/1) to give the sub-title compound (AKN-4) (6.0 g,26.2mmol,54%,90% purity) as a white solid. M/z 229.9/231.9 (M+H) + (ES+)。
Step 4: 5-bromo-4-chloro-2-methylbenzoic acid (AKN-5)
A mixture of the product from step 3 above (AKN-4) (6.0 g,1 eq, 26.0 mmol) and aqueous KOH (280 mL, 2M) was stirred overnight at 110 ℃. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×200 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (AKN-5) (5.0 g,20.2mmol,77% purity) as a white solid. M/z 248.9/250.9 (M+H) + (ES+)
Step 5: 5-bromo-4-chloro-2-methylbenzoic acid methyl ester (AKN-6)
A solution of the product from step 4 above (AKN-5) (5.0 g,1 eq., 20.0 mmol) in DCM (50 mL) and MeOH (50 mL) was treated with TMSCHN at room temperature 2 (11.5 mL,10 equivalents, 200.4 mmol) for 1 hour. The resulting mixture was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (10/1)The above was to give the sub-title compound (AKN-6) (5.0 g,19.1mmol,95%,90% purity) as a white solid. M/z262.9/264.9 (M+H) + (ES+)。
Step 6: 5-bromo-2- (bromomethyl) -4-chlorobenzoic acid methyl ester (AKN-7)
A solution of the product (AKN-6) (5.0 g,1 eq, 190 mmol) from step 5 above in DCE (100 mL) was treated with AIBN (1.25 g,0.4 eq, 7.61 mmol) and NBS (5.07 g,1.5 eq, 28.5 mmol) at room temperature under nitrogen. The resulting mixture was stirred overnight at 80 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The resulting mixture was used directly in the next step without further purification. M/z 342.8/344.8 (M+H) + (ES+)
Step 7: 6-bromo-5-chloro-2, 3-dihydro-isoindol-1-one (AKN-8)
The product (AKN-7) (5.0 g,1 eq., 14.6 mmol) from step 6 above was reacted with an NH-containing catalyst 3 The solution in MeOH (120 mL, 7M) was stirred at room temperature for 16 hours. The precipitated solid was collected by filtration and washed with EtOAc (3×30 mL). The resulting mixture was concentrated in vacuo. This gave the sub-title compound (AKN-8) (3.0 g,12.2mmol,83%,92% purity) as a white solid. M/z 245.9/247.9 (M+H) + (ES+)。
Step 8: 5-chloro-6-vinyl-2, 3-dihydro-isoindol-1-one (AKN-9)
Pd (PPh) was added to a stirred mixture of the product (AKN-8) (500 mg,1 eq, 2.03 mmol), tributyl (vinyl) stannane (643 mg,1 eq, 2.03 mmol) and CsF (616 mg,2 eq, 4.06 mmol) from step 7 above in dioxane (12 mL) at room temperature under nitrogen atmosphere 3 ) 2 Cl 2 (284 mg,0.2 eq, 0.41 mmol). The resulting mixture was stirred under nitrogen at 120 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (20% ACN up to 80% in 15 minutes); detector, UV 254/220nm. This gives the sub-title compound (AKN-9) as a white solid 120mg,0.62mmol,31%,90% purity). M/z 194.0/196.0 (M+H) + (ES+)。
Step 9: 6-chloro-3-oxo-1, 2-dihydro-isoindole-5-carbaldehyde (AKN-10)
To the compound, the product (AKN-9) from step 8 above (312 mg,1 eq, 1.61 mmol), NMO (378 mg,2 eq, 3.22 mmol) and CA (612 mg,2 eq, 3.22 mmol) in t-BuOH (10 mL) and H 2 K was added to the solution in O (10 mL) 2 OsO 4 .2H 2 O (59 mg,0.1 eq, 0.16 mmol) was stirred at room temperature for 2 hours. Adding NaIO to the mixture at room temperature 4 (66 mg,2 equivalents, 0.31 mmol). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The resulting mixture was used directly in the next step without further purification. M/z196.0/198.0 (M+H) + (ES+)
Step 10: 5-chloro-6- (hydroxymethyl) -2, 3-dihydro-isoindol-1-one (AKN-11)
To a stirred mixture of the product (AKN-10) (250 mg,1 eq, 1.28 mmol) from step 9 above in THF (20 mL) at 0deg.C was added NaBH 4 (97 mg,2 equivalents, 2.56 mmol). The resulting mixture was stirred at room temperature for 1 hour. At 0℃by addition of saturated NH 4 Cl (aq) (10 mL) quenched the reaction. The resulting mixture was diluted with water and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 50% in 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (AKN-11) (160 mg,0.81mmol,63%,95% purity) as a white solid. M/z 198.0/200.0 (M+H) + (ES+)。
Step 11:4- {2- [ 5-chloro-6- (hydroxymethyl) -1-oxo-3H-isoindol-2-yl ] -6-cyclopropylpyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKN-12)
To intermediate (AEG-2) (20 mg,1 equivalent, 60. Mu. Mol), product (AKN-11) from step 10 above (13 mg,1.1 equivalent, 66. Mu. Mol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 To a stirred mixture of (58 mg,3 eq, 0.18 mmol) in dioxane (3 mL) was added XPhos Pd G3 (10 mg,0.2 eq, 12 μmol) and XPhos (6 mg,0.2 eq, 12 μmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 1 hour. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 60% B over 10 min; wavelength: 254nm; retention time: 8.73) to give the title compound (AKN-12) (1.0 mg, 2.0. Mu. Mol,3.4%,99.0% purity) as a white solid. M/z 497.3/499.3 (M+H) + (ES+)。 1 H NMR (300 MHz, methanol-d 4) delta 8.51 (s, 1H), 8.17-8.00 (m, 4H), 7.92 (d, j=8.0 hz, 1H), 7.69 (s, 1H), 6.94 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 4.77 (s, 2H), 3.51 (s, 3H), 2.10-1.96 (m, 1H), 1.09-0.90 (m, 4H).
Example 323: synthesis of 4- (2- (5-chloro-6- (((cyclopentylmethyl) amino) methyl) -1-oxoisoindolin-2-yl) -6-cyclopropylpyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AKO-2)
Step 1:4- [2- (5-chloro-6-formyl-1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKO-1)
A solution of compound (AKN-12) (20 mg,1 eq, 40. Mu. Mol) in DCM (1 mL) was treated with DMP (26 mg,1.5 eq, 60. Mu. Mol) at room temperature for 1 hour. The resulting mixture was filtered and the filter cake was washed with DCM (3X 3 mL). The filtrate was concentrated in vacuo. The crude product was purified without further purificationIn the case of the chemical reaction, the reaction mixture was used directly in the next step. M/z 495.1/497.1 (M+H) + (ES+)。
Step 2:4- (2- (5-chloro-6- (((cyclopentylmethyl) amino) methyl) -1-oxoisoindolin-2-yl) -6-cyclopropylpyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AKO-2)
A mixture of the product (AKO-1) from step 1 above (10 mg,1 eq, 20. Mu. Mol) and 1-cyclopentylmethylamine (AHH-1) (2.2 mg,1.1 eq, 22. Mu. Mol) in MeOH (2 mL) was stirred at 60℃for 1 hour. Then NaBH was added at room temperature 4 (2.5 mg,2 equivalents, 40. Mu. Mol) for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 6% B to 36% B over 10 min; wavelength: 254nm; retention time: 10.63) to give the title compound (AKO-2) (3.0 mg, 5.2. Mu. Mol,24%,93.4% purity) as a white solid. M/z 578.3/580.3 (M+H) + (ES+)。 1 H NMR (300 MHz, methanol-d 4) delta 8.47 (s, 1H), 8.18-8.07 (m, 2H), 8.05-7.99 (m, 2H), 7.94-7.82 (m, 2H), 6.97 (d, J=1.4 Hz, 1H), 5.08 (s, 2H), 4.37 (s, 2H), 3.47 (s, 3H), 3.01 (d, J=7.4 Hz, 2H), 2.29-2.17 (m, 1H), 2.10-1.99 (m, 1H), 2.00-1.87 (m, 2H), 1.75-1.60 (m, 4H), 1.34-1.24 (m, 2H), 1.07-0.97 (m, 4H).
Example 324: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (1-hydroxycyclobutyl) methyl ] (methyl) amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKP-1)
A solution of compound (AFJ-1) (40 mg,1 eq, 73. Mu. Mol) and formaldehyde (1 mL) in MeOH (8 mL) was stirred at room temperature overnight. Adding NaBH to the mixture 3 CN (23 mg,0.37mmol,5 eq.). The resulting mixture was stirred at room temperature for an additional 2 days. The obtained mixture is put in trueConcentrating in the air. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 46% B to 60% B in 8 minutes; wavelength: 254/220nm; retention time: 7.95 To give the title compound (AKP-1) (2.6 mg,4.6 μmol,6.3%,99.6% purity) as a white solid. M/z 560.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.14-8.04 (m, 3H), 7.92 (d, j=8.0 hz, 1H), 7.81 (s, 1H), 7.72-7.65 (m, 1H), 7.60 (d, j=7.8 hz, 1H), 6.92 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 3.72 (s, 2H), 3.51 (s, 3H), 2.58 (s, 2H), 2.30 (s, 3H), 2.15-1.96 (m, 5H), 1.82-1.67 (m, 1H), 1.52-1.35 (m, 1H), 1.09-0.94 (m, 4H).
Example 325: synthesis of 4- [2- (6- { 3-azabicyclo [3.1.0] hexane-3-ylmethyl } -4-fluoro-1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKQ-2)
To intermediate (AIO-8) (40 mg,1 eq, 84. Mu. Mol) and 3-azabicyclo [3.1.0] at room temperature under a nitrogen atmosphere ]To a stirred mixture of hexane (AKQ-1) (10 mg,1.5 eq, 0.13 mmol) in DCM (5 mL) was added DIPEA (13 mg,1.2 eq, 0.10 mmol). The resulting mixture was stirred at room temperature overnight. NaBH (OAc) was added to the above mixture at 0deg.C 3 (35 mg,2 equivalents, 0.17 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The resulting mixture was diluted with water and extracted with DCM (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min;gradient: 44% B to 56% B in 8 minutes; wavelength: 254/220nm; retention time: 6.32 To give the title compound (AKQ-2) (2.6 mg,4.8 μmol,5.7%,99.7% purity) as a white solid. M/z 546.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.51 (s, 1H), 8.16-8.07 (m, 2H), 8.01 (d, j=1.4 hz, 1H), 7.95-7.89 (m, 1H), 7.59 (s, 1H), 7.35 (d, j=9.6 hz, 1H), 6.97 (d, j=1.4 hz, 1H), 5.06 (s, 2H), 3.70 (s, 2H), 3.51 (s, 3H), 2.94 (d, j=8.6 hz, 2H), 2.42 (d, j=8.5 hz, 2H), 2.10-2.00 (m, 1H), 1.51-1.26 (m, 2H), 1.22-0.82 (m, 4H), 0.81-0.74 (m, 1H), 0.39 (td, j=7.7, 4.1 hz).
Example 326: synthesis of 4- (2-cyclopropyl-6- { 4-fluoro-6- [ (1S, 4S) -2-oxa-5-azabicyclo [2.2.1] heptan-5-ylmethyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKR-3)
Step 1:4- {2- [6- (chloromethyl) -4-fluoro-1-oxo-3H-isoindol-2-yl ] -6-cyclopropylpyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKR-1)
To a stirred mixture of intermediate (AIO-7) (60 mg,1 eq, 0.13 mmol) in DCM (10 mL) at 0deg.C was added SOCl 2 (0.6 mL). The resulting mixture was stirred at room temperature for 30 minutes. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (9/1) to give the sub-title compound (AKR-1) as a white solid (58 mg,0.12mmol,93% purity). M/z 499.1/501.1 (M+H) + (ES+)
Step 2:4- (2-cyclopropyl-6- { 4-fluoro-6- [ (1S, 4S) -2-oxa-5-azabicyclo [2.2.1] heptan-5-ylmethyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) (AKR-3)
To the product (AKR-1) (58 mg,1 equivalent, 0.12 mmol) from step 1 above and (1S, 4S) -2-oxa-5-azabicyclo [2.2.1] at room temperature ]Heptane (AKR-2) (17 mg,1.5 eq, 0.17 mmol) inEt was added to a stirred mixture in DCM (10 mL) 3 N (35 mg,3 eq, 0.35 mmol). The resulting mixture was stirred at 60℃for 12 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 48% B in 9 minutes; wavelength: 254/210nm; retention time: 8.35 To give the title compound (AKR-3) (13.6 mg,24 μmol,21%,99.9% purity) as a white solid. M/z 562.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.21(h,J=1.7Hz,2H),7.95(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.70(d,J=8.5Hz,1H),7.50(d,J=9.9Hz,1H),6.92(d,J=1.4Hz,1H),5.07(s,2H),4.36(s,1H),3.93(d,J=7.5Hz,1H),3.88-3.78(m,2H),3.54(d,J=7.5,1.9Hz,1H),3.47(s,4H),2.75(d,J=9.9,1.8Hz,1H),2.43(d,J=9.9Hz,1H),2.09-2.01(m,1H),1.87-1.79(m,1H),1.61(d,J=10.0Hz,1H),1.04-0.91(m,4H)。
Example 327: synthesis of 4- [2- (6- { [ (cyclobutylmethyl) amino ] methyl } -4-fluoro-1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKS-1)
Et is added to a stirred solution of intermediate (AIO-8) (30 mg,1 eq, 63. Mu. Mol) and 1-cyclobutylmethylamine (AIA-1) (5 mg,1 eq, 63. Mu. Mol) in MeOH (5 mL) under nitrogen at 60 ℃ 3 N (32 mg,5 eq, 0.32 mmol) for 2 hours. Adding NaBH to the mixture at 0deg.C 4 (24 mg,10 equivalents, 0.63 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge preparationModel OBD C18 column, 19X 250mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 25 ml/min; gradient: 56B to 62B in 8 minutes; a detector, UV 254/210nm; retention time: 7.03. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AKS-1) (12.7 mg,23 μmol,37%,99.5% purity) as a white solid. M/z 548.3 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.24–8.20(m,2H),7.95(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.61(s,1H),7.50(d,J=10.0Hz,1H),6.92(d,J=1.4Hz,1H),5.07(s,2H),3.78(s,2H),3.48(s,3H),2.48(s,2H),2.42–2.38(m,1H),1.87–1.75(m,3H),1.71–1.55(m,4H),1.23(s,1H),1.01–0.96(m,4H)。
Example 328: synthesis of 4- [2- (6- { [ (cyclobutylmethyl) (methyl) amino ] methyl } -4-fluoro-1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKT-1)
To a stirred solution of compound (AKS-1) (30 mg,1 eq., 55. Mu. Mol) and formaldehyde (5 mg,0.165mmol,3 eq.) in MeOH (5 mL) at room temperature was added NaBH 3 CN (7 mg,2 equivalents, 0.11 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was diluted with water and extracted with DCM (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 19×250mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 65B to 76B in 8 minutes; a detector, UV 254/210nm; retention time: 7.85. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the product as a white solid(AKT-1) (2.7 mg,4.8 μmol,8.8%,99.8% purity). M/z 562.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.25–8.18(m,2H),7.95(d,J=1.4Hz,1H),7.87(d,J=8.4Hz,1H),7.57(s,1H),7.45(d,J=9.9Hz,1H),6.92(d,J=1.4Hz,1H),5.08(s,2H),3.55(s,2H),3.47(s,3H),3.17(d,J=5.2Hz,1H),2.39(s,2H),2.11(s,3H),2.03(t,J=10.6Hz,3H),1.84(t,J=9.0Hz,1H),1.76(d,J=9.6Hz,1H),1.63(d,J=9.2Hz,2H),0.98(d,J=6.1Hz,4H)。
Example 329: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (1-ethylcyclopropyl) amino ] methyl } -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKU-2)
To a stirred mixture of 1-ethylcyclopropan-1-amine (AKU-1) (92 mg,10 equivalents, 0.76 mmol) in MeOH (3 ml) was added DIPEA (49 mg,0.38mmol,5 equivalents) at room temperature. The resulting mixture was stirred at room temperature for 5 minutes. Intermediate (AIO-8) (40 mg,1 eq, 76. Mu. Mol) was added to the above mixture at room temperature. The resulting mixture was stirred at 60 ℃ for an additional 1 hour. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (10 mg,3.5 eq, 0.26 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 62% B to 82% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AKU-2) (8.1 mg, 14. Mu. Mol,18%,99.6% purity) as a white solid. M/z 598.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(d,J=2.0Hz,1H),8.23–8.16(m,2H),8.04–7.92(m,3H),7.90–7.85(m,1H),6.90(d,J=1.5Hz,1H),5.14(s,2H),3.88(s,2H),3.47(s,3H),2.11–2.02(m,1H),1.47–1.39(m,2H),1.00–0.86(m,7H),0.52–0.46(m,2H),0.32(t,J=2.9Hz,2H)。
Example 330: synthesis of 4- { 2-cyclopropyl-6- [ 4-fluoro-6- ({ [ (2R) -2-methoxypropyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKV-1)
To a stirred mixture of intermediate (AIO-8) (40 mg,1 eq, 84. Mu. Mol) in MeOH (6 mL) was added (2R) -2-methoxypropan-1-amine (AID-1) (80 mg,11 eq, 0.90 mmol) at room temperature. The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was stirred at 60 ℃ for an additional 1 hour. Adding NaBH to the above mixture at 0deg.C 4 (25 mg,7.9 eq, 0.66 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1). The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 32% B over 10 min; wavelength: 254 nm) to give the title compound (AKV-1) (14.2 mg, 26. Mu. Mol,31%,99.2% purity) as a white solid. M/z 552.1 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.26–8.15(m,2H),7.95(d,J=1.4Hz,1H),7.91–7.80(m,1H),7.62(s,1H),7.51(d,J=9.9Hz,1H),6.95–6.85(m,1H),5.07(s,2H),3.82(s,2H),3.48(s,3H),3.40–3.36(m,1H),3.23(s,3H),2.49–2.39(m,2H),2.13–1.98(m,1H),1.07(d,J=6.1Hz,3H),1.00–0.86(m,4H)。
Example 331: synthesis of 4- { 2-cyclopropyl-6- [ 4-fluoro-6- ({ [ (2S) -2-methoxypropyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKW-1)
To a stirred solution of intermediate (AIO-8) (30 mg,1 eq, 63 μmol) and (2S) -2-methoxypropan-1-amine (AIM-1) (7 mg,1.2 eq, 76 μmol) in MeOH (8 mL) was added DIPEA (24 mg,3 eq, 0.19 mmol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 equivalents, 0.32 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XSelect CSH preparative C18 OBD column, 19 x 250mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 25 ml/min; gradient: 17% B to 47% B over 8 min; wavelength: 254/220nm; retention time: 7.18) to give the title compound (AKW-1) (3.4 mg,6.2 μmol,9.6%,97.8% purity) as a white solid. M/z 552.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.14-8.08 (m, 2H), 8.02 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.67 (s, 1H), 7.46 (d, j=9.6 hz, 1H), 6.96 (d, j=1.4 hz, 1H), 5.07 (s, 2H), 3.95 (s, 2H), 3.57-3.51 (m, 1H), 3.49 (s, 3H), 3.35 (s, 3H), 2.66-2.62 (m, 2H), 2.08-2.00 (m, 1H), 1.13 (d, j=6.1 hz, 3H), 1.07-0.97 (m, 4H).
Example 332: synthesis of 4- { 2-ethoxy-6- [6- (hydroxymethyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKX-1)
To intermediate (AEW-1) (20 mg,1 eq, 59. Mu. Mol), intermediate (AGH-1) (12 mg,1.3 eq, 77. Mu. Mol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 (38 mg,2 eq, 0.12 mmol) in 1, 4-dioxane (8 mL)To the solution of RuPhos (11 mg,0.4 eq, 24. Mu. Mol) and RuPhos ring palladium complex Gen.3 (10 mg,0.2 eq, 12. Mu. Mol) were added. The resulting mixture was stirred at 100℃for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 44% B in 9 minutes; wavelength: 254/220nm; retention time: 8.02 To give the title compound (AKX-1) (3.0 mg, 6.4. Mu. Mol,11%,99.5% purity) as a white solid. M/z 467.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.14-8.05 (m, 2H), 7.90 (d, j=8.0 hz, 1H), 7.86-7.80 (m, 2H), 7.69-7.58 (m, 2H), 6.42 (d, j=1.3 hz, 1H), 5.06 (s, 2H), 4.71 (s, 2H), 4.44-4.34 (m, 2H), 3.53 (s, 3H), 1.39 (t, j=7.0 hz, 3H).
Example 333: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (3S) -3-methylpiperidin-1-yl ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKY-1)
To a stirred solution of intermediate (AKR-1) (30 mg,1 eq, 60. Mu. Mol) and (3S) -3-methylpiperidine (R-1) (9 mg,1.5 eq, 90. Mu. Mol) in DCM (8 mL) at room temperature was added Et3N (30 mg,5 eq, 0.30 mmol). The resulting mixture was stirred at room temperature under nitrogen overnight. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 32% B in 10 min; wavelength: 254nm; retention time: 11.48) to give the title compound (AKY-1) (8.6 mg, 15. Mu. Mol,25%,99.5% purity) as a white solid. M/z 562.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.25–8.18(m,2H),7.94(d,J=1.3Hz,1H),7.87(d,J=8.5Hz,1H),7.55(s,1H),7.46(d,J=9.9Hz,1H),6.93(d,J=1.4Hz,1H),5.07(s,2H),3.67–3.58(m,2H),3.57–3.50(m,3H),2.71(s,2H),2.09–2.00(m,1H),1.94–1.84(m,1H),1.60(s,4H),1.54–1.43(m,1H),0.98(d,J=7.4Hz,4H),0.83–0.79(m,4H)。
Example 334: synthesis of 4- { 2-cyclopropyl-6- [ 4-fluoro-6- ({ [ (1-fluorocyclopropyl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AKZ-2)
A solution of 1- (1-fluorocyclopropyl) methylamine (AKZ-1) (6.7 mg,1.2 eq, 76. Mu. Mol) in MeOH (8 mL) was treated with DIPEA (24 mg,3 eq, 0.19 mmol) at room temperature for 5 min, followed by the addition of intermediate (AIO-8) (30 mg,1 eq, 63. Mu. Mol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 equivalents, 0.32 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 25 ml/min; gradient: 50% B to 66% B in 8 minutes; wavelength: 254/220nm; retention time: 7.62 To give the title compound (AKZ-2) (11 mg, 20. Mu. Mol,32%,99.6% purity) as a white solid. M/z 552.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.14-8.07 (m, 2H), 8.02 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.66 (d, j=1.2 hz, 1H), 7.48-7.43 (m, 1H), 6.95 (d, j=1.4 hz, 1H), 5.06 (s, 2H), 3.97 (s, 2H), 3.49 (s, 3H), 2.92 (d, j=21.5 hz, 2H), 2.09-2.01 (m, 1H), 1.07-0.96 (m, 6H), 0.70-0.62 (m, 2H).
Example 335: synthesis of 4- {2- [ 4-bromo-6- (hydroxymethyl) -1-oxo-3H-isoindol-2-yl ] -6-cyclopropylpyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALA-5)
Step 1: 3-oxo-1, 2-dihydro-isoindole-5-carboxylic acid (ALA-1)
To a stirred solution of 6-bromo-2, 3-isoindolin-1-one (ACY-1) (500 mg,1 eq, 2.36 mmol) and oxalic acid (317 mg,1.2 eq, 2.83 mmol) in DMF (10 mL) at room temperature under nitrogen was added Ac 2 O (361 mg,1.5 eq, 3.54 mmol) and DIPEA (4.57 g,1.5 eq, 3.54 mmol). Xantphos (2793 mg,0.2 eq, 0.47 mmol) and Pd (OAc) were added to the above mixture under nitrogen at room temperature 2 (53 mg,0.1 eq, 0.24 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% MeCN up to 20% in 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (ALB-1) (226 mg,1.28mmol,54% pure) as a white solid. M/z 178.0 (M+H) + (ES+)。
Step 2: 7-bromo-3-oxo-1, 2-dihydro-isoindole-5-carboxylic acid (ALA-2)
To the product (ALA-1) from step 1 above (226 mg,1 equivalent, 1.27 mmol) at room temperature in concentrated H 2 SO 4 Ag was added to the stirred solution in (5 mL) 2 SO 4 (398 mg,1 equivalent, 1.28 mmol) and Br 2 (248 mg,1.2 eq, 1.53 mmol). The resulting mixture was stirred at 80℃for 12 hours. The mixture was cooled to room temperature. The reaction was then quenched by addition of 10mL of ice water at 0 ℃. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (20% MeCN up to 30% in 10 minutes); detector, UV 254/220nm. This gives a yellow solidThe sub-title compound (ALA-2) (150 mg,0.59mmol,46%,92% purity). M/z 256.0/258.0 (M+H) + (ES+)。
Step 3: 7-bromo-3-oxo-1, 2-dihydro-isoindole-5-carboxylic acid methyl ester (ALA-3)
To a stirred solution of the product (ALA-2) from step 2 above (150 mg,1 eq, 0.59 mmol) in MeOH (10 mL) under nitrogen at 0deg.C was added SOCl in portions 2 (209 mg,3 eq, 1.76 mmol). The resulting mixture was stirred under nitrogen at 80 ℃ for 2 hours. The mixture was cooled to room temperature and then concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (12/1) to give the sub-title compound (ALA-3) as a yellow solid (150 mg,0.56mmol,95%,92% purity). M/z 270.0/272.0 (M+H) + (ES+)
Step 4: 4-bromo-6- (hydroxymethyl) -2, 3-dihydro-isoindol-1-one (ALA-4)
To a stirred solution of the product (ALA-3) from step 3 above (150 mg,1 eq, 0.56 mmol) in THF (10 mL) under nitrogen at 0deg.C was added DIBAL-H (g) in portions in THF (5.6 mL,1.0M,10 eq, 5.60 mmol). The resulting mixture was stirred under nitrogen at 0 ℃ for 15 minutes. The reaction was then quenched by the addition of 3mL of ice water at 0 ℃ and then concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (10/1) to give the sub-title compound (ALA-4) as a white solid (90 mg,0.37mmol,67%,90% purity). M/z 242.0/244.0 (M+H) + (ES+)
Step 5:4- {2- [ 4-bromo-6- (hydroxymethyl) -1-oxo-3H-isoindol-2-yl ] -6-cyclopropylpyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALA-5)
To the product (ALA-4) (90 mg,1 equivalent, 0.37 mmol), intermediate (AEG-2) (250 mg,2 equivalent, 0.74 mmol) and Cs from step 4 above, under a nitrogen atmosphere at room temperature 2 CO 3 (243 mg,2 eq, 0.74 mmol) to a stirred solution of RuPhos (69 mg,0.4 eq, 0.15 mmol) and RuPhos ring palladium complex gen.3 (62 mg,0.2 eq, 74. Mu. Mol) in dioxane (10 mL) were added. The resulting mixture was stirred under nitrogen at 60 ℃ for 2 hours. Cooling the mixture toDilute with water and extract with EtOAc (3×20 mL) at room temperature. The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (12/1). The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 25% B to 55% B in 10 min; wavelength: 254 nm) to give the title compound (ALA-5) (6.1 mg, 11. Mu. Mol,30%,98.3% purity) as a white solid. M/z 541.1/543.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.25–8.18(m,2H),7.97(d,J=1.4Hz,1H),7.90–7.82(m,2H),7.74(s,1H),6.88(d,J=1.5Hz,1H),5.50(t,J=5.8Hz,1H),4.91(s,2H),4.62(d,J=5.8Hz,2H),3.47(s,3H),2.13–2.04(m,1H),1.07–0.92(m,4H)。
Example 336: synthesis of 4- [2- (4-bromo-6- { [ (2-methoxyethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALB-2)
Step 1:4- [2- (4-bromo-6-formyl-1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALB-1)
A solution of compound (ALA-5) (60 mg,1 eq, 0.11 mmol) and DMP (62 mg,1.3 eq, 0.14 mmol) in DCM (5 mL) was stirred at room temperature for 1 h. Filtering the resulting mixture; the filter cake was washed with DCM (3X 3 mL). The filtrate was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 539.1/541.1 (M+H) + (ES+)。
Step 2:4- [2- (4-bromo-6- { [ (2-methoxyethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALB-2)
The product (ALB-1) from step 1 above (60 mg,a solution of 1 eq, 0.11 mmol) and 2-methoxyethyl-1-amine (AEB-1) (13 mg,1.5 eq, 0.17 mmol) in MeOH (5 mL) was stirred at 60℃for 1 hour. Adding NaBH to the mixture at 0deg.C 4 (8 mg,2 equivalents, 0.22 mmol). The resulting mixture was stirred at room temperature for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃ and then concentrated in vacuo. The residue was purified by TLC with DCM/MeOH (10/1). The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 32% B over 10 min; wavelength: 254 nm) to give the title compound (ALB-2) (9.7 mg, 16. Mu. Mol,15%,99.5% purity) as a white solid. M/z 598.0/600.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.25–8.16(m,2H),7.96(d,J=1.3Hz,1H),7.90–7.85(m,2H),7.76(s,1H),6.89(d,J=1.4Hz,1H),4.90(s,2H),3.84(s,2H),3.48(s,3H),3.40(t,J=5.6Hz,2H),3.24(s,3H),2.64(t,J=5.6Hz,2H),2.11–2.04(m,1H),1.02–0.94(m,4H)。
Example 337: synthesis of 4- { 2-cyclopropyl-6- [ 4-fluoro-6- ({ [ (2R, 3R) -3-methoxybutan-2-yl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALC-2)
A solution of intermediate (AIO-8) (30 mg,1 eq, 63. Mu. Mol), DIPEA (32 mg,4 eq, 0.25 mmol) and (2R, 3R) -3-methoxybutan-2-amine (ALC-1) (8.4 mg,1.3 eq, 82. Mu. Mol) in MeOH (8 mL) was stirred overnight at 60 ℃. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 equivalents, 0.32 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 43% B to 53% B in 9 minutes; wavelength: 254/220nm; retention time: 8.45 To give the title compound (ALC-2) (8.0 mg, 14. Mu. Mol,23%,99.8% purity) as a white solid. M/z 566.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.16-8.06 (m, 2H), 8.03 (d, j=1.4 hz, 1H), 7.92 (d, j=8.1 hz, 1H), 7.65 (d, j=1.2 hz, 1H), 7.47-7.41 (m, 1H), 6.95 (d, j=1.4 hz, 1H), 5.06 (s, 2H), 3.98 (d, j=13.8 hz, 1H), 3.81 (d, j=13.8 hz, 1H), 3.50 (s, 3H), 3.34 (s, 3H), 3.21-3.10 (m, 1H), 2.63-2.51 (m, 1H), 2.09-1.98 (m, 1H), 1.11 (d, j=6.1 hz, 3H), 1.07-0.97 (m, 7H).
Example 338: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (2-fluoro-2-methylpropyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALD-2)
A solution of 2-fluoro-2-methylpropan-1-amine (ALD-1) (7 mg,1.2 eq, 78. Mu. Mol) in MeOH (20 mL) was treated with DIPEA (34 mg,4 eq, 0.26 mmol) at room temperature for 5 min, followed by the addition of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 52% B in 9 minutes; wavelength: 254/220nm; retention time: 8.83 To give the title compound (ALD-2) (6.1 mg, 11. Mu. Mol,17%,99.8% purity) as a white solid )。m/z 536.1(M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.14-8.07 (m, 2H), 8.05 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.81 (d, j=1.5 hz, 1H), 7.69-7.66 (m, 1H), 7.60 (d, j=7.8 hz, 1H), 6.92 (d, j=1.4 hz, 1H), 5.02 (s, 2H), 3.91 (s, 2H), 3.50 (s, 3H), 2.68 (d, j=19.6 hz, 2H), 2.07-1.99 (m, 1H), 1.39 (s, 3H), 1.33 (s, 3H), 1.06-0.95 (m, 4H).
Example 339: synthesis of 4- { 2-cyclopropyl-6- [6- (hydroxymethyl) -1-oxo-3H-pyrrolo [3,4-c ] pyridin-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALE-7)
Step 1:5- (bromomethyl) -2-chloropyridine-4-carboxylic acid methyl ester (ALE-2)
AIBN (239 mg,0.3 eq, 1.45 mmol) is added to a stirred solution of methyl 2-chloro-5-methylpyridine-4-carboxylate (ALE-1) (900 mg,1 eq, 4.85 mmol) and NBS (949 mg,1.1 eq, 5.33 mmol) in (trifluoromethyl) benzene (25 mL) at room temperature under nitrogen. The resulting mixture was stirred overnight at 80 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/5) to give the sub-title compound (ALE-2) (920 g,3.5mmol,40%,95% purity) as a pale yellow oil. M/z 263.9/265.9 (M+H) + (ES+)。
Step 2: 6-chloro-2H, 3H-pyrrolo [3,4-c ] pyridin-1-one (ALE-3)
The product (ALE-2) from step 1 above (920 mg,1 eq, 3.48 mmol) was taken up in NH 3 A solution in MeOH (10 mL, 7M) was stirred at room temperature under nitrogen for 2 hours. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (0% MeCN up to 20% in 20 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ALE-3) as an off-white solid (400 mg,2.38mmol,58%,90% purity). M/z 169.0/171.0 (M+H) + (ES+)。
Step 3: 6-vinyl-2H, 3H-pyrrolo [3,4-c ] pyridin-1-one (ALE-4)
To a stirred solution of the product (ALE-3) (240 mg,1 eq, 1.42 mmol) from step 2 above and tributyl (vinyl) stannane (552 mg,1.2 eq, 1.71 mmol) in dioxane (12 mL) under nitrogen at room temperature was added CsF (433 mg,2 eq, 2.85 mmol). Pd (PPh) was added to the above mixture at room temperature under a nitrogen atmosphere 3 ) 4 (399 mg,0.2 eq, 0.29 mmol). The resulting mixture was stirred under nitrogen at 100 ℃ for an additional overnight. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and MeCN (0% MeCN up to 20% in 30 min); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ALE-4) (180 mg,1.12mmol,73%,92% purity) as a white solid. M/z 161.1 (M+H) + (ES+)。
Step 4: 1-oxo-2H, 3H-pyrrolo [3,4-c ] pyridine-6-carbaldehyde (ALF-5)
To the product (ALE-4) (180 mg,1 eq, 1.12 mmol) from step 3 above and 2-hydroxypropane-1, 2, 3-tricarboxylic acid (281mg, 1.3 eq, 1.46 mmol) in t-BuOH (5 mL) and H under nitrogen at room temperature 2 To a stirred solution of O (5 mL) was added NMO (171 mg,1.3 eq, 1.46 mmol) and K 2 OsO 4 (41 mg,0.1 eq, 0.11 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. Adding NaIO to the mixture at 0deg.C 4 (481 mg,2 equivalents, 2.25 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 1 hour. The mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (5/1) to give the sub-title compound (ALE-5) (80 mg,0.49mmol,42%,90% purity) as a white solid. M/z 163.0 (M+H) + (ES+)。
Step 5:6- (hydroxymethyl) -2H, 3H-pyrrolo [3,4-c ] pyridin-1-one (ALE-6)
To a stirred solution of the product from step 4 above (ALE-5) (80 mg,1 eq., 0.49 mmol) in MeOH (8 mL) at 0deg.C under nitrogen was added NaBH 4 (93 mg,5 eq, 2.47 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1) to give the sub-title compound (ALE-6) as an off-white solid (40 mg,0.24mmol,44%,93% purity). M/z 165.1 (M+H) + (ES+)。
Step 6:4- { 2-cyclopropyl-6- [6- (hydroxymethyl) -1-oxo-3H-pyrrolo [3,4-c ] pyridin-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALE-7)
Cs was added to a stirred solution of intermediate (AEG-2) (30 mg,1 equivalent, 89 μmol) and product (ALE-6) from step 5 above (16 mg,1.1 equivalent, 98 μmol) in dioxane (5 mL) at room temperature under a nitrogen atmosphere 2 CO 3 (58 mg,2 equivalents, 0.18 mmol). RuPhos (17 mg,0.4 eq, 36. Mu. Mol) and RuPhos ring palladium complex Gen.3 (15 mg,0.2 eq, 18. Mu. Mol) were added to the above mixture under nitrogen atmosphere at room temperature. The resulting mixture was stirred under nitrogen at 100 ℃ for an additional 1 hour. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 24% B to 34% B in 9 minutes; wavelength: 254/220nm; retention time: 8.92 To give the title compound (ALE-7) as a white solid (3.5 mg, 7.6. Mu. Mol,8.4%,99.8% purity). M/z 464.0 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.92(d,J=1.2Hz,1H),8.55(s,1H),8.22(d,J=7.2Hz,2H),7.98(d,J=1.4Hz,1H),7.95–7.86(m,1H),7.77(d,J=1.2Hz,1H),6.95(d,J=1.4Hz,1H),5.62(t,J=5.9Hz,1H),5.12(s,2H),4.68(d,J=5.9Hz,2H),3.49(s,3H),2.10–1.97(m,1H),1.02–0.90(m,4H)。
Example 340: synthesis of 4- [2- (6- { [ (cyclobutylmethyl) amino ] methyl } -1-oxo-3H-pyrrolo [3,4-c ] pyridin-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALF-2)
Step 1:4- (2-ethyl-6- { 6-formyl-1-oxo-3H-pyrrolo [3,4-c ] pyridin-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALF-1)
To a stirred solution of compound (ALE-7) (50 mg,1 eq, 0.11 mmol) in DCM (5 mL) under nitrogen at room temperature was added DMP (23 mg,0.5 eq, 54. Mu. Mol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 1 hour. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (ALF-1) as an off-white solid (50 mg,0.11mmol,98%,94% purity). M/z 462.2 (M+H) + (ES+)。
Step 2:4- [2- (6- { [ (cyclobutylmethyl) amino ] methyl } -1-oxo-3H-pyrrolo [3,4-c ] pyridin-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALF-2)
To a stirred solution of the product from step 1 above (ALF-1) (30 mg,1 eq, 65. Mu. Mol) and DIPEA (25 mg,3 eq, 0.20 mmol) in MeOH (5 mL) was added 1-cyclobutylmethylamine, HCl (AIA-1) (12 mg,1.5 eq, 98. Mu. Mol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature under nitrogen overnight. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 60% B in 8 minutes; wavelength: 254/220nm; retention time: 7.4 To give the title compound (ALF-2) (7.5 mg,14 μmol,21%, 97.1%) as a white solid. M/z 531.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.92(d,J=1.1Hz,1H),8.55(s,1H),8.22(d,J=7.1Hz,2H),7.97(d,J=1.4Hz,1H),7.92–7.82(m,1H),7.78(d,J=1.1Hz,1H),6.95(d,J=1.4Hz,1H),5.11(s,2H),3.89(s,2H),3.49(s,3H),2.55(d,J=7.2Hz,2H),2.47–2.37(m,1H),2.10–1.95(m,3H),1.90–1.74(m,2H),1.70–1.58(m,2H),1.07–0.89(m,4H)。
Example 341: synthesis of 3' -cyclopropyl-5 ' - [ 4-fluoro-6- ({ [ (2R) -1-methoxypropan-2-yl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (ALG-3)
Step 1:3' -cyclopropyl-5 ' - [ 4-fluoro-6- (hydroxymethyl) -1-oxo-3H-isoindol-2-yl ] -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (ALG-1)
To intermediate (AJP-6) (160 mg,1 equivalent, 0.42 mmol), intermediate (AIO-6) (92 mg,1.2 equivalent, 0.51 mmol) and Cs under a nitrogen atmosphere at room temperature 2 CO 3 (412 mg,3 eq, 1.27 mmol) to a stirred mixture of dioxane (10 mL) was added RuPhos (79 mg,0.4 eq, 0.17 mmol) and RuPhos ring palladium complex gen.3 (71 mg,0.2 eq, 0.08 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (ALG-1) (100 mg,0.21mmol,49%,95% pure) as a yellow solid. M/z 480.2 (M+H) + (ES+)
Step 2:3' -cyclopropyl-5 ' - (4-fluoro-6-formyl-1-oxo-3H-isoindol-2-yl) -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (ALG-2)
To a stirred mixture of the product from step 1 above (ALG-1) (40 mg,1 eq, 83. Mu. Mol) in DCM (10 mL) was added DMP (42 mg,1.2 eq, 0.10 mmol) at room temperature. The resulting mixture was stirred at room temperature for 1 hour. Filtering the resulting mixture; the filter cake was washed with DCM (3X 3 mL). The filtrate was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 478.2 (M+H) + (ES+)
Step 3:3' -cyclopropyl-5 ' - [ 4-fluoro-6- ({ [ (2R) -1-methoxyprop-2-yl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (ALG-3)
A solution of the product from step 2 above (ALG-2) (35 mg,1 eq, 73. Mu. Mol) in MeOH (12 mL) was treated with (2R) -1-methoxypropan-2-amine (AIK-1) (9.8 mg,1.5 eq, 0.11 mmol) at 60℃under a nitrogen atmosphere for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the above mixture in portions at 0deg.C 4 (6 mg,2 equivalents, 0.15 mmol). The resulting mixture was stirred at room temperature under an air atmosphere for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 48% B in 8 minutes; wavelength: 254/220nm; retention time: 7.67 To give the title compound (ALG-3) (9.6 mg,13 μmol,24%,99.6% purity) as a white solid. M/z 551.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.48(s,1H),8.17(d,J=8.1,1.8Hz,1H),8.09(d,J=1.8Hz,1H),7.87(d,J=8.1Hz,1H),7.73(d,J=1.8Hz,1H),7.62(s,2H),7.51(d,J=10.0Hz,1H),6.45(s,1H),5.01(s,2H),3.96-3.75(m,2H),3.27(d,J=9.3,6.3Hz,4H),3.18(d,J=9.3,5.5Hz,1H),3.11(s,3H),2.74(p,J=6.2Hz,1H),1.88(t,J=8.6,5.0Hz,1H),1.03-0.87(m,5H),0.58-0.46(m,2H)。
Example 342: synthesis of 3' -cyclopropyl-5 ' - [ 4-fluoro-6- ({ [ (2S) -1-methoxypropan-2-yl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] -2- (4-methyl-1, 2, 4-triazol-3-yl) - [1,1' -biphenyl ] -4-carbonitrile (ALH-1)
A solution of intermediate (ALG-2) (35 mg,1 eq, 73. Mu. Mol) in MeOH (12 mL) was treated with (2S) -1-methoxypropan-2-amine (AIR-1) (10 mg,1.5 eq, 0.11 mmol) at 60℃under a nitrogen atmosphere for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the above mixture in portions at 0deg.C 4 (6 mg,2 equivalents, 0.15 mmol). The resulting mixture was stirred at room temperature under an air atmosphere for 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 48% B in 8 minutes; wavelength: 254/220nm; retention time: 7.67 To give the 3 title compound (ALH-1) (8.8 mg,16 μmol,22%,99.8% purity) as a white solid. M/z 551.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.48(d,J=2.5Hz,1H),8.18(d,J=8.1,1.9Hz,1H),8.10(d,J=1.9Hz,1H),7.87(d,J=8.1Hz,1H),7.77-7.71(m,1H),7.62(t,J=1.8Hz,2H),7.51(d,J=10.0Hz,1H),6.45(t,J=1.6Hz,1H),5.02(s,2H),3.92–3.81(m,2H),3.30-3.26(m,1H),3.24(d,J=2.3Hz,3H),3.18(d,J=9.4,5.5Hz,1H),3.12(d,J=2.5Hz,3H),2.76-2.71(m,1H),1.87(d,J=8.3,4.8Hz,1H),1.01-0.88(m,5H),0.56-0.48(m,2H)。
Example 343: synthesis of 4- { 2-cyclopropyl-6- [ 4-fluoro-6- ({ [ (3R, 4R) -4-hydroxyoxolan-3-yl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALI-2)
A solution of intermediate (AIO-8) (30 mg, 63. Mu. Mol,1 eq.), DIPEA (32 mg,4 eq., 0.25 mmol) and (3R, 4R) -4-aminovalance-3-ol (ALI-1) (8 mg,1.3 eq., 82. Mu. Mol) in MeOH (8 mL) was stirred overnight at 60 ℃. The mixture was cooled to room temperature. NaBH was added to the above mixture at room temperature 3 CN (79 mg,20 equivalents, 1.26 mmol). The resulting mixture was stirred at 60 ℃ for an additional 2 days. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm 5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 19% B to 26% B over 10 min; wavelength: 254nm; retention time: 9.5) to give the title compound (ALI-2) (4.2 mg, 7.4. Mu. Mol,12%,98.1% purity) as a white solid. M/z 566.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.16-8.06 (m, 2H), 8.02 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.69 (s, 1H), 7.48 (d, j=9.7 hz, 1H), 6.95 (d, j=1.4 hz, 1H), 5.07 (s, 2H), 4.31-4.24 (m, 1H), 4.01 (d, j=13.6 hz, 1H), 3.97-3.85 (m, 3H), 3.82-3.76 (m, 1H), 3.50 (d, j=9.7 hz, 4H), 3.38-3.32 (m, 1H), 2.09-1.99 (m, 1H), 1.09-0.95 (m, 4H).
Example 344: synthesis of 4- (2-cyclopropyl-6- {6- [ (3-fluoroazetidin-1-yl) methyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALJ-1)
A solution of 3-fluoroazetidine (U-1) (8 mg,1.2 eq, 0.11 mmol) in DCM (12 mL) was treated with DIPEA (48 mg,4 eq, 0.38 mmol) at room temperature for 5 min, followed by the addition of intermediate (AIQ-1) (45 mg,1 eq, 94. Mu. Mol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 50% B in 8 minutes; wavelength: 254/220nm; retention time: 7.43 To give the title compound (ALJ-1) (26.7 mg,51 μmol,55%,99.6% purity) as a white solid. M/z 520.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.14-8.08 (m, 2H), 8.04 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.75 (s, 1H), 7.61 (d, j=1.2 hz, 2H), 6.92 (d, j=1.4 hz, 1H), 5.25-5.18 (m, 1H), 5.01 (s, 2H), 3.79 (s, 2H), 3.68-3.59 (m, 2H), 3.50 (s, 3H), 3.36-3.33 (m, 1H), 3.29-3.25 (m, 1H), 2.06-1.99m, 1H), 1.05-0.95 (m, 4H).
Example 345: synthesis of 4- {2- [6- (2-aminoprop-2-yl) -1-oxo-3H-isoindol-2-yl ] -6-cyclopropylpyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALK-9)
Step 1: 6-bromo-2- [ (4-methoxyphenyl) methyl ] -3H-isoindol-1-one (ALK-2)
Methyl 5-bromo-2- (bromomethyl) benzoate (ALK-1) (3.00 g,1 eq, 9.74 mmol) and Et 3 N (1.97 g,2 equivalents, 19.5 mmol) and PMB-NH 2 (2.00 g,1.5 eq, 14.6 mmol) in MeOH (50 mL) was stirred overnight at 80 ℃. The mixture was cooled to room temperature and concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (3/1) to give the sub-title compound (ALK-2) (2.5 g,7.55mmol,77%,90% purity) as a brown yellow solid. M/z 332.0/334.0 (M+H) + (ES+)
Step 2:2- {2- [ (4-methoxyphenyl) methyl ] -3-oxo-1H-isoindol-5-yl } acetic acid tert-butyl ester (ALK-4)
The product (ALK-2) (500 mg,1 equivalent, 1.51 mmol) from step 1 above was combined with Pd 2 (dba) 3 A solution of (69 mg,0.05 eq, 75. Mu. Mol) XPhos (72 mg,0.1 eq, 0.15 mmol) in THF (15 mL) was stirred at room temperature under nitrogen for 30 minutes. To the above mixture was added tert-butyl 2- (zinc-. Lamda.2-bromoalkyl) acetate (ALK-3) (1.96 g,5 equivalents, 7.53 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred under nitrogen at 80 ℃ for an additional overnight. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/2) to give the sub-title compound (ALK-4) as an off-white solid (400 mg,1.09mmol,72%,94% purity). M/z 368.2 (M+H) + (ES+)
Step 3:2- {2- [ (4-methoxyphenyl) methyl ] -3-oxo-1H-isoindol-5-yl } -2-methylpropanoic acid tert-butyl ester (ALK-5)
To a stirred solution of the product (ALK-4) from step 2 above (400 mg,1 eq, 1.09 mmol) and t-BuOK (366 mg,3 eq, 3.27 mmol) in THF (10 mL) at 0deg.C was added CH 3 I (773 mg,5 eq, 5.45 mmol). The resulting mixture was stirred at 0 ℃ for 1 hour. The reaction was then quenched by the addition of 5mL of MeOH at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/3) to give the sub-title compound (ALK-5) (230 mg,0.58mmol,53%,92% purity) as a yellow oil. M/z 396.2 (M+H) + (ES+)
Step 4:2- {2- [ (4-methoxyphenyl) methyl ] -3-oxo-1H-isoindol-5-yl } -2-methylpropanoic acid (ALK-6)
A solution of the product from step 3 above (ALK-5) (230 mg,1 eq, 0.58 mmol) and TFA (2 mL) in DCM (6 mL) was stirred at room temperature for 2 hours. The resulting mixture was diluted with water and extracted with DCM (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (ALK-6) (140 mg,0.32mmol,71%,94% purity) as a brown yellow oil. M/z 340.2 (M+H) + (ES+)
Step 5:6- (2-Isocyanopropan-2-yl) -2- [ (4-methoxyphenyl) methyl ] -3H-isoindol-1-one (ALK-7)
To a stirred solution of the product (ALK-6) (130 mg,1 eq, 0.38 mmol) from step 5 above and DPPA (264 mg,2.5 eq, 0.96 mmol) in DCM (6 mL) at room temperature was added Et 3 N (97 mg,2.5 eq, 0.96 mmol). The resulting mixture was stirred at room temperature for 2 hours. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/3) to give the sub-title compound (ALK-7) (80 mg,0.24mmol,62%,90% purity) as a brown yellow solid. M/z 337.2 (M+H) + (ES+)
Step 6:6- (2-aminopropan-2-yl) -2, 3-dihydro-isoindol-1-one (ALK-8)
A solution of the product from step 5 above (ALK-7) (80 mg,1 eq, 0.24 mmol) in TFA (10 mL) was stirred overnight at 100deg.C. The mixture was cooled to room temperature. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (0% MeCN up to 70% in 20 minutes); detector, UV 254/220nm. This gave the sub-title compound (ALK-8) (30 mg,0.16mmol,66%,92% purity) as a yellow solid. M/z 191.1 (M+H) + (ES+)
Step 7:4- {2- [6- (2-aminoprop-2-yl) -1-oxo-3H-isoindol-2-yl ] -6-cyclopropylpyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALK-9)
To the product (ALK-8) (30 mg,1 equivalent, 0.16 mmol), intermediate (AEG-2) (58 mg,1.1 equivalent, 0.17 mmol) and Cs from step 6 above, under a nitrogen atmosphere at room temperature 2 CO 3 (103 mg,2 eq, 0.32 mmol) to a stirred solution of RuPhos ring palladium complex gen.3 (26 mg,0.2 eq, 32. Mu. Mol) and RuPhos (29 mg,0.4 eq, 63. Mu. Mol) in dioxane (5 mL) were added. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XSelect CSH preparative C18 OBD column, 19 x 250mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient:38% B to 48% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (ALK-9) (5.3 mg, 11. Mu. Mol,6.8%,99.5% purity) as a white solid. M/z 490.1 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.21(d,J=6.8Hz,2H),7.97–7.84(m,4H),7.63(d,J=8.0Hz,1H),6.92(d,J=1.4Hz,1H),4.99(s,2H),3.50(s,3H),2.10–2.02(m,1H),1.44(s,6H),1.04–0.94(m,4H)。
Example 346: synthesis of 4- [ 2-cyclopropyl-6- (6- {2- [ (cyclopropylmethyl) amino ] propan-2-yl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALL-2)
/>
To a stirred solution of compound (ALK-9) (30 mg,1 eq, 61. Mu. Mol) and cyclopropanecarbaldehyde (ALL-1) (13 mg,3 eq, 0.18 mmol) in MeOH (5 mL) at room temperature was added DIPEA (48 mg,6 eq, 0.37 mmol). The resulting mixture was stirred at 60℃for 4 hours. The mixture was cooled to room temperature. NaBH (OAc) was added to the above mixture at room temperature 3 (65 mg,5 equivalents, 0.31 mmol). The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column, 30X 150mm,5 μm, n; mobile phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 31% B to 61% B in 10 minutes; wavelength: 254 nm) to give the title compound (ALL-2) (6.6 mg, 12. Mu. Mol,20%,98.8% purity) as a white solid. M/z 544.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.08 (m, 2H), 8.05 (d, j=1.4 hz, 1H), 7.96-7.91 (m, 1H), 7.90-7.88 (m, 1H), 7.84-7.80 (m, 1H), 7.62 (d, j=8.1 hz, 1H), 6.93 (d, j=1.5 hz, 1H), 5.02 (s, 2H), 3.51 (s, 3H), 2.13-1.99 (m, 3H), 1.53 (s, 6H), 1.08-0.94 (m, 4H), 0.92-0.81 (m, 1H), 0.48-0.40 (m, 2H), 0.05-0.02 (m, 2H).
Example 347: synthesis of 4- {2- [ 4-fluoro-6- (hydroxymethyl) -1-oxo-3H-isoindol-2-yl ] -6- (trifluoromethyl) pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALM-3)
Step 1:4- [ 2-chloro-6- (trifluoromethyl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALM-2)
To intermediate (ADG-1) (100 mg,1 eq, 0.38 mmol) and 2-chloro-6- (trifluoromethyl) pyridin-4-ylboronic acid (ALM-1) (94 mg,1.1 eq, 0.42 mmol) in THF (10 mL) and H under a nitrogen atmosphere at room temperature 2 Na was added to the stirred solution in O (1 mL) 2 CO 3 (121 mg,3 eq, 1.14 mmol). Pd (PPh) was added to the above mixture at room temperature under a nitrogen atmosphere 3 ) 4 (44 mg,0.1 eq, 38. Mu. Mol). The resulting mixture was stirred under nitrogen at 80 ℃ for an additional 2 hours. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1) to give the sub-title compound (ALM-2) as an off-white solid (68 mg,0.19mmol,47%,95% purity). M/z 364.0/366.0 (M+H) + (ES+)
Step 2:4- {2- [ 4-fluoro-6- (hydroxymethyl) -1-oxo-3H-isoindol-2-yl ] -6- (trifluoromethyl) pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALM-3)
Cs was added to a stirred solution of the product (ALM-2) (30 mg,1 equivalent, 0.08 mmol) from step 1 above and intermediate (AIO-6) (22 mg,1.5 equivalent, 0.12 mmol) in dioxane (5 mL) at room temperature under nitrogen atmosphere 2 CO 3 (54 mg,2 equivalents, 0.16 mmol). RuPhos (16 mg,0.4 eq, 33. Mu. Mol) and RuPhos ring palladium complex Gen.3 (14 mg,0.2 eq, 16. Mu. Mol) were added to the above mixture under nitrogen atmosphere at room temperature. The resulting mixture was stirred under nitrogen at 100 ℃ for an additional 1 hour. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (15/1). Crude product is producedThe material was purified by preparative HPLC under the following conditions (column: XBridge preparative OBD C18 column, 30 x 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 48% B in 9 minutes; wavelength: 254/220nm; retention time: 8.3 To give the title compound (ALM-3) (3.9 mg,7.7 μmol,9.3%,99.8% purity) as a white solid. M/z 509.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.58(s,1H),8.49(s,1H),8.37–8.24(m,2H),7.99(d,J=8.1Hz,1H),7.65(s,1H),7.50(d,J=10.3Hz,2H),5.53(t,J=5.8Hz,1H),5.14(s,2H),4.63(d,J=5.7Hz,2H),3.59(s,3H)。
Example 348: synthesis of 4- [2- (4-fluoro-6- { [ (2-methoxyethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) -6- (trifluoromethyl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALN-2)
Step 1:4- [2- (4-fluoro-6-formyl-1-oxo-3H-isoindol-2-yl) -6- (trifluoromethyl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALN-1)
To a stirred solution of compound (ALM-3) (55 mg,1 eq, 0.11 mmol) in DCM (5 mL) under nitrogen at room temperature was added DMP (69 mg,1.5 eq, 0.16 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 1 hour. Filtering the resulting mixture; the filter cake was washed with DCM (3X 2 mL). The filtrate was concentrated in vacuo. This gave the sub-title compound (ALN-1) (50 mg, 99. Mu. Mol,86%,96% purity) as an off-white solid. M/z 507.1 (M+H) + (ES+)。
Step 2:4- [2- (4-fluoro-6- { [ (2-methoxyethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) -6- (trifluoromethyl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALN-2)
To the product (ALN-1) (35 mg,1 equivalent, 0.07 mmol) from step 1 above and 2-methoxyethyl at room temperature under a nitrogen atmosphereTo a stirred solution of 1-amine (AEB-1) (8 mg,1.5 eq, 0.10 mmol) in MeOH (5 mL) was added DIPEA (27 mg,3 eq, 0.21 mmol). The resulting mixture was stirred at room temperature under nitrogen overnight. Adding NaBH to the mixture at 0deg.C 4 (13 mg,5 eq, 0.35 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 30% B over 10 min; wavelength: 254nm; retention time: 7.7) to give the title compound (ALN-2) (13.1 mg, 23. Mu. Mol,33%,99.8% purity) as a white solid. M/z 566.1 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.58(s,1H),8.47(d,J=1.3Hz,1H),8.32(d,J=1.7Hz,1H),8.28–8.22(m,1H),7.99(d,J=8.1Hz,1H),7.67(s,1H),7.55(d,J=9.9Hz,1H),7.50(d,J=1.3Hz,1H),5.12(s,2H),3.84(s,2H),3.59(s,3H),3.40(t,J=5.7Hz,2H),3.24(s,3H),2.63(t,J=5.7Hz,2H)。
Example 349: synthesis of 4- { 2-cyclopropyl-6- [ 4-fluoro-6- ({ [ (1-methylcyclobutyl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALO-2)
A solution of 1- (1-methylcyclobutyl) methylamine (ALO-1) (13 mg,1.3 eq., 0.14 mmol) and DIPEA (54 mg,4 eq., 0.42 mmol) in MeOH (8 mL) was stirred at room temperature for 5 min. Intermediate (AIO-8) (50 mg,1 eq, 0.10 mmol) was added to the above mixture at room temperature. The resulting mixture was stirred at 60 ℃ for an additional overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (20 mg,5 eq, 0.52 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The obtained mixture is treated in the following conditionConcentrating in vacuum. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 60% B to 73% B in 8 minutes; wavelength: 254/220nm; retention time: 7.57 To give the title compound (ALO-2) (7.2 mg, 13. Mu. Mol,12%,99.5% purity) as a white solid. M/z 562.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.16-8.06 (m, 2H), 8.03 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.66 (s, 1H), 7.46 (d, j=9.8 hz, 1H), 6.94 (d, j=1.5 hz, 1H), 5.06 (s, 2H), 3.89 (s, 2H), 3.49 (s, 3H), 2.52 (s, 2H), 2.10-1.99 (m, 1H), 1.98-1.75 (m, 4H), 1.74-1.60 (m, 2H), 1.17 (s, 3H), 1.07-0.92 (m, 4H).
Example 350: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (1-methylcyclobutyl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALP-1)
A solution of 1- (1-methylcyclobutyl) methylamine (ALO-1) (8.0 mg,1.2 eq, 78. Mu. Mol) in MeOH (12 mL) was treated with DIPEA (34 mg,4 eq, 0.26 mmol) at room temperature for 5 min. To the above mixture was added intermediate (AGT-2) (30 mg,1 equivalent, 65. Mu. Mol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 65% B in 9 minutesThe method comprises the steps of carrying out a first treatment on the surface of the Wavelength: 254/220nm; retention time: 8.9 To give the title compound (ALP-1) (8.5 mg, 16. Mu. Mol,24%,99.6% purity) as a white solid. M/z 544.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.14-8.05 (m, 3H), 7.92 (d, j=8.1 hz, 1H), 7.81 (s, 1H), 7.71-7.59 (m, 2H), 6.91 (d, j=1.5 hz, 1H), 5.02 (s, 2H), 3.88 (s, 2H), 3.50 (s, 3H), 2.52 (s, 2H), 2.07-1.98 (m, 1H), 1.94-1.75 (m, 4H), 1.72-1.62 (m, 2H), 1.16 (s, 3H), 1.06-0.93 (m, 4H).
Example 351: synthesis of 4- (2-cyclopropyl-6- {6- [ (cyclopropylamino) methyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALQ-2)
To a stirred solution of intermediate (AGT-2) (25 mg,1 eq, 54. Mu. Mol) and aminocyclopropane (ALQ-1) (4 mg,1.2 eq, 65. Mu. Mol) in MeOH (8 mL) was added DIPEA (28 mg,4 eq, 0.22 mmol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (10 mg,5 eq, 0.27 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 41% B to 51% B in 8 minutes; wavelength: 254/220nm; retention time: 7.82 To give the title compound (ALQ-2) (5.3 mg,11 μmol,18%,92.9% purity) as a white solid. M/z 502.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.14-8.04 (m, 3H), 7.92 (d, J=8.0 Hz, 1H), 7.80 (s, 1H), 7.68-7.65 (m, 1H), 7.61 (d, J=7.7 Hz, 1H), 6.91 (d, J=1.4 Hz, 1H), 5.02 (s, 2H), 3.90 (s, 2H), 3.50 (s, 3H), 2.15-1.99 (m, 2H), 1.04-0.96 (m, 4H), 0.50–0.44(m,2H),0.42–0.37(m,2H)。
Example 352: synthesis of 4- (2-cyclopropyl-6- {6- [ (cyclopropylamino) methyl ] -4-fluoro-1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALR-1)
A solution of intermediate (AIO-8) (35 mg,1 eq, 73. Mu. Mol), DIPEA (38 mg,4 eq, 0.29 mmol) and aminocyclopropane (ALQ-1) (6.3 mg,1.5 eq, 0.11 mmol) in MeOH (8 mL) was stirred overnight at 60 ℃. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (14 mg,5 eq, 0.37 mmol). The resulting mixture was stirred at room temperature for another 1 day. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 46% B to 55% B in 8 minutes; wavelength: 254/220nm; retention time: 7.55 To give the title compound (ALR-1) (2.7 mg,5.2 μmol,6.6%,92.3% purity) as a white solid. M/z 520.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.16-8.06 (m, 2H), 8.03 (d, j=1.4 hz, 1H), 7.92 (d, j=8.1 hz, 1H), 7.65 (s, 1H), 7.43 (d, j=9.8 hz, 1H), 6.94 (d, j=1.5 hz, 1H), 5.06 (s, 2H), 3.91 (s, 2H), 3.49 (s, 3H), 2.15-2.09 (m, 1H), 2.08-2.00 (m, 1H), 1.09-0.95 (m, 4H), 0.52-0.43 (m, 2H), 0.41-0.34 (m, 2H).
Example 353: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (3S) -3-methylpiperidin-1-yl ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALS-1)
A solution of (3S) -3-methylpiperidine (R-1) (9 mg, 93. Mu. Mol,1.5 eq.) and DIPEA (32 mg,4 eq., 0.25 mmol) in DCM (8 mL) was stirred at room temperature for 5 min. Intermediate (AIQ-1) (30 mg,1 equivalent, 62. Mu. Mol) was added to the above mixture at room temperature. The resulting mixture was stirred at 60 ℃ for an additional overnight. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 55% B to 65% B in 9 minutes; wavelength: 254/220nm; retention time: 8.18 To give the title compound (ALS-1) (9.5 mg,17 μmol,28%,99.1% purity) as a white solid. M/z 544.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.16-8.07 (m, 2H), 8.05 (d, j=1.3 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.78 (s, 1H), 7.68-7.58 (m, 2H), 6.93 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 3.61 (s, 2H), 3.51 (s, 3H), 2.88-2.77 (m, 2H), 2.07-1.91 (m, 2H), 1.76-1.52 (m, 5H), 1.06-0.93 (m, 4H), 0.85 (d, j=5.6 hz, 4H).
Example 354: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [1- (methoxymethyl) cyclopropyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALT-2)
A solution of 1- (methoxymethyl) cyclopropyl-1-amine (ALT-1) (8.0 mg,1.2 eq, 78. Mu. Mol) in MeOH (8 mL) was treated with DIPEA (34 mg,4 eq, 0.26 mmol) at room temperature for 5 min, followed by the addition of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃.The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 43% B to 53% B in 8 minutes; 53% B; wavelength: 254/220nm; retention time: 7.83 To give the title compound (ALT-2) (9.6 mg, 18. Mu. Mol,27%,99.6% purity) as a white solid. M/z 546.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.14-8.03 (m, 3H), 7.93-7.90 (m, 1H), 7.80-7.78 (m, 1H), 7.67-7.55 (m, 2H), 6.91 (d, J=1.5 Hz, 1H), 5.00 (s, 2H), 3.93 (s, 2H), 3.50 (s, 3H), 3.41 (d, J=13.5 Hz, 5H), 2.06-1.96 (m, 1H), 1.06-0.95 (m, 4H), 0.71-0.65 (m, 2H), 0.57-0.51 (m, 2H).
Example 355: synthesis of 4- (2-cyclopropyl-6- {6- [ (4, 4-difluoropiperidin-1-yl) methyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALU-2)
A solution of 4, 4-difluoropiperidine (ALU-1) (15 mg,1.5 eq, 0.12 mmol) and DIPEA (43 mg,4 eq, 0.33 mmol) in DCM (8 mL) was stirred at room temperature for 5 min. Intermediate (AIQ-1) (40 mg,1 eq, 83. Mu. Mol) was added to the above mixture at room temperature. The resulting mixture was stirred at 60 ℃ for another 4 days. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 47% B to 57% B in 10 minutes; wavelength: 254/220nm; retention time: 9.58 To give the title compound (AIU-2) (9.0 mg,16 μmol,19%,99.4% purity) as a white solid. M/z 566.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H),8.15–8.02(m,3H),7.92(d,J=8.0Hz,1H),7.81(s,1H),7.69–7.58(m,2H),6.93(d,J=1.4Hz,1H),5.02(s,2H),3.69(s,2H),3.51(s,3H),2.59(t,J=5.6Hz,4H),2.09–1.92(m,5H),1.08–0.94(m,4H)。
example 356: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (2, 2-difluoro-2-methoxyethyl) amino ] methyl } -4-fluoro-1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALV-4)
Step 1: 4-fluoro-6- { [ (2, 2-trifluoroethyl) amino ] methyl } -2, 3-dihydro-isoindol-1-one (ALV-2)
To a stirred solution of intermediate (AIO-5) (300 mg,1 eq, 1.68 mmol) and 2, 2-trifluoroethylamine (ALV-1) (332 mg,2 eq, 3.35 mmol) in MeOH (20 mL) was added AcOH (6 mL) at room temperature under nitrogen. NaBH was added to the above mixture at room temperature under nitrogen atmosphere 3 CN (316 mg,3 equivalents, 5.03 mmol). The resulting mixture was stirred under nitrogen at room temperature overnight. Adding NaBH to the mixture at 0deg.C 4 (317 mg,5 equivalents, 8.38 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (ALV-2) as an off-white solid (260 mg,0.99mmol,54%,93% purity). M/z 263.1 (M+H) + (ES+)
Step 2:6- { [ (2, 2-difluoro-2-methoxyethyl) amino ] methyl } -4-fluoro-2, 3-dihydro-isoindol-1-one (ALV-3)
To a stirred solution of the product from step 1 above (ALV-2) (100 mg,1 eq, 0.38 mmol) in THF (5 mL) and MeOH (5 mL) at room temperature under nitrogen atmosphere was added KOH (2.14 g,100 eq, 38.1 mmol). The resulting mixture was stirred overnight at 100 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) was purified to give the sub-title compound (ALV-3) (53 mg,0.19mmol,48%,96% purity) as a white solid. M/z 275.1 (M+H) + (ES+)
Step 3:4- [ 2-cyclopropyl-6- (6- { [ (2, 2-difluoro-2-methoxyethyl) amino ] methyl } -4-fluoro-1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALV-4)
Cs was added to a stirred solution of intermediate (AEG-2) (30 mg,1 equivalent, 0.09 mmol) and product (ALV-3) from step 2 above (27 mg,1.1 equivalent, 0.10 mmol) in dioxane (5 mL) under nitrogen at room temperature 2 CO 3 (58 mg,2 equivalents, 0.18 mmol). RuPhos (17 mg,0.4 eq, 0.04 mmol) and RuPhos ring palladium complex Gen.3 (15 mg,0.2 eq, 0.02 mmol) were added to the above mixture under nitrogen at room temperature. The resulting mixture was stirred under nitrogen at 100 ℃ for an additional 1 hour. The mixture was cooled to room temperature. Filtering the resulting mixture; the filter cake was washed with MeOH (2X 3 mL). The filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (20/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 47% B to 55% B in 9 minutes; wavelength: 254/220nm; retention time: 8.6 To give the title compound (ALV-4) (18.1 mg, 32. Mu. Mol,34%,96.6% purity) as an off-white solid. M/z 574.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.21(d,J=6.8Hz,2H),7.96(d,J=1.3Hz,1H),7.91–7.80(m,1H),7.35(d,J=1.1Hz,1H),7.28(d,J=1.2Hz,1H),6.90(d,J=1.4Hz,1H),4.88(s,2H),3.90(d,J=17.4Hz,5H),3.49(s,3H),3.25–3.04(m,3H),2.04(d,J=6.4Hz,1H),1.00–0.86(m,4H)。
Example 357: synthesis of- [ 2-cyclopropyl-6- (4-fluoro-1-oxo-6- { [ (2, 2-trifluoroethyl) amino ] methyl } -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALW-1)
Cs was added to a stirred solution of intermediate (AEG-2) (30 mg,1 equivalent, 0.09 mmol) and intermediate (ALV-2) (27 mg,1.1 equivalent, 0.10 mmol) in dioxane (5 mL) at room temperature under nitrogen atmosphere 2 CO 3 (58 mg,2 equivalents, 0.18 mmol) and RuPhos ring palladium complex Gen.3 (15 mg,0.2 equivalents, 0.02 mmol) were added. The resulting mixture was stirred under nitrogen at 100 ℃ for an additional 1 hour. The mixture was cooled to room temperature. Filtering the resulting mixture; the filter cake was washed with MeOH (2X 3 mL). The filtrate was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (20/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 47% B to 55% B in 9 minutes; wavelength: 254/220nm; retention time: 8.6 To give the title compound (ALW-1) (17.3 mg,31 μmol,34%,97.8% purity) as a white solid. M/z 562.0 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.26–8.14(m,2H),7.95(d,J=1.4Hz,1H),7.91–7.82(m,1H),7.64(d,J=1.2Hz,1H),7.54–7.48(m,1H),6.93(d,J=1.4Hz,1H),5.08(s,2H),3.91(d,J=4.9Hz,2H),3.48(s,3H),3.26–3.13(m,2H),2.12–1.98(m,1H),1.02–0.93(m,4H)。
Example 358: synthesis of 4- { 2-cyclopropyl-6- [4, 5-difluoro-6- (hydroxymethyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALX-9)
Step 1:3, 4-difluoro-2-methylbenzoic acid methyl ester (ALX-2)
3, 4-difluoro-2-methylbenzoic acid (ALX-1) (5.0 g,1 equivalent, 29.1 mmol) and SOCl 2 (1A mixture of 0.4g,3 equivalents, 87.2 mmol) in MeOH (100 mL) was stirred under nitrogen at 80deg.C for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (3/1). This gave the sub-title compound (ALX-2) (5.0 g,26.9mmol,92% purity) as a yellow solid. M/z 187.1 (M+H) + (ES+)
Step 2: 5-bromo-3, 4-difluoro-2-methylbenzoic acid methyl ester (ALX-3).
To the product (ALX-2) (2.00 g,1 equivalent, 10.7 mmol), br from step 1 above at 0deg.C 2 (6.87 g,4 equivalents, 43.0 mmol) and concentrated HNO 3 (6.77 g,10 eq., 107 mmol) AgNO was added to a stirred mixture in AcOH (40 mL) 3 (2.37 g,1.3 eq, 14.0 mmol). The resulting mixture was stirred at room temperature under nitrogen overnight. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (5/1) to give the sub-title compound (ALX-3) (2.0 g,7.58mmol,70%,90% purity) as a yellow oil. M/z 265.0/267.0 (M+H) + (ES+)
Step 3: methyl 5-bromo-2- (bromomethyl) -3, 4-difluorobenzoate (ALX-4).
To the product (ALX-3) (1.9 g,1 equivalent, 7.17 mmol) from step 2 above and NBS (1.91 g,1.5 equivalent, 10.8 mmol) in CHCl under nitrogen at room temperature 3 BPO (920 mg,0.5 eq, 3.58 mmol) was added to the stirred mixture in (40 mL). The resulting mixture was stirred overnight at 80 ℃ under nitrogen atmosphere. The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was used directly in the next step without any further purification. M/z 344.9/346.9 (M+H) + (ES+)
Step 4: 6-bromo-4, 5-difluoro-2, 3-dihydro-isoindol-1-one (ALX-5).
The product (ALX-4) (3 g, 8.720 mmol,1 eq.) from step 3 above was combined with NH 3 The mixture in MeOH (20 mL) was stirred at room temperature under an air atmosphere for 2 hours. The residue was purified by silica gel column chromatography, diluted with PE/EA (1:1) to give the sub-title compound (ALX-5) (900 mg,3.65mmol,42%,92% purity) as a yellow solid. M/z247.9/249.9 (M+H) + (ES+)。
Step 5: 6-vinyl-4, 5-difluoro-2, 3-dihydro-isoindol-1-one (ALX-6).
Pd (PPh) was added to a stirred mixture of the product (ALZ-5) (700 mg,1 eq, 2.82 mmol), tributyl (vinyl) stannane (1.04 g,1.2 eq, 3.29 mmol) and CsF (857 mg,2 eq, 5.64 mmol) from step 4 above in dioxane (20 mL) at room temperature under nitrogen atmosphere 3 ) 2 Cl 2 (198 mg,0.1 eq, 0.28 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (20/1) to give the sub-title compound (ALX-6) (550 mg,2.82mmol,99% pure) as a yellow solid. M/z 196.1 (M+H) + (ES+)。
Step 6:6, 7-difluoro-3-oxo-1, 2-dihydro-isoindole-5-carbaldehyde (ALX-7).
To the product (ALX-6) (500 mg,1 equivalent, 2.56 mmol) from step 5 above and K under a nitrogen atmosphere at room temperature 2 OSO 4 .2H 2 O (94 mg,0.1 eq, 0.26 mmol) in t-BuOH (20 mL) and H 2 To a stirred mixture of O (20 mL) was added NMO (390 mg,1.3 eq, 3.33 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. Adding NaIO to the mixture at 0deg.C 4 (1.10 g,2 equivalents, 5.12 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (20% MeCN up to 48% in 15 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo.This gave the sub-title compound (ALX-7) (200 mg,1.01mmol,40%,95% purity) as a yellow solid. M/z 198.0 (M+H) + (ES+)。
Step 7:4, 5-difluoro-6- (hydroxymethyl) -2, 3-dihydro-isoindol-1-one (ALX-8).
A solution of the product from step 6 above (ALX-7) (170 mg,1 eq, 0.86 mmol) in MeOH (10 mL) was treated with NaBH at 0deg.C 4 (65 mg,2 eq, 1.72 mmol) of the treated material. The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (20% MeCN up to 45% in 10 minutes); detector, UV 254/220nm. The product-containing fractions were combined and concentrated in vacuo. This gave the sub-title compound (ALX-8) (70 mg,0.35mmol,41%,91% purity) as a yellow solid. M/z 200.0 (M+H) + (ES+)。
Step 8:4- { 2-cyclopropyl-6- [4, 5-difluoro-6- (hydroxymethyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALX-9).
To the product (ALX-8) (30 mg,1 equivalent, 0.15 mmol), intermediate (AEG-2) (40 mg,0.8 equivalent, 0.12 mmol) and Cs from step 7 above under a nitrogen atmosphere at room temperature 2 CO 3 To a stirred mixture of (98 mg,2 eq, 0.30 mmol) in dioxane (5 mL) was added XPhos (29 mg,0.4 eq, 0.06 mmol) and XPhos Pd G3 (26 mg,0.2 eq, 0.03 mmol). The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 36% B to 46% B in 9 minutes; wavelength: 254/220nm; retention time: 8.87 To give a white solid The title compound (ALX-9) (7.2 mg, 14. Mu. Mol,9.6%,99.6% purity) was obtained in the form of a solid. M/z 499.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.25–8.18(m,2H),7.96–7.83(m,2H),7.71(d,J=5.2Hz,1H),6.92(d,J=1.4Hz,1H),5.59(t,J=5.9Hz,1H),5.13(s,2H),4.66(d,J=5.8Hz,2H),3.48(s,3H),2.11–2.00(m,1H),1.01–0.94(m,4H)。
Example 359: synthesis of 4- { 2-cyclopropyl-6- [4, 5-difluoro-1-oxo-6- ({ [2- (trifluoromethoxy) ethyl ] amino } methyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALY-2)
Step 1:4- [ 2-cyclopropyl-6- (4, 5-difluoro-6-formyl-1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALY-1).
A solution of compound (ALX-9) (30 mg,1 eq, 0.06 mmol) in DCM (3 mL) was treated with DMP (33 mg,1.3 eq, 78. Mu. Mol) at room temperature. The resulting mixture was stirred at room temperature under nitrogen atmosphere for 3 hours. The resulting mixture was filtered and the filter cake was washed with DCM (3X 3 mL). The crude product was used directly in the next step without any further purification. M/z 497.1 (M+H) + (ES+)
Step 2:4- { 2-cyclopropyl-6- [4, 5-difluoro-1-oxo-6- ({ [2- (trifluoromethoxy) ethyl ] amino } methyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALY-2).
To a stirred mixture of product (ALY-1) (20 mg,1 eq, 0.04 mmol) from step 1 above and 2- (trifluoromethoxy) ethylamine (AGN-2) (8 mg,1.5 eq, 0.06 mmol) in MeOH (2 mL) was added DIPEA (10 mg,2 eq, 0.08 mmol) at room temperature under nitrogen. The resulting mixture was stirred at room temperature overnight. Adding NaBH to the mixture at 0deg.C 4 (3 mg,2 equivalents, 0.08 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. Mixing the obtained mixtureThe compound was concentrated in vacuo. The residue was purified by preparative HPLC on a column (XBridge prep OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 36% B to 46% B in 9 minutes; wavelength: 254/220nm; retention time: 8.87 To give the title compound (ALY-2) (3.6 mg,5.9 μmol,14%,94.4% purity) as a white solid. M/z 610.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(d,J=5.4Hz,1H),8.22(d,J=6.9Hz,2H),7.94–7.84(m,2H),7.77(d,J=5.2Hz,1H),7.00–6.92(m,1H),5.14(d,J=8.4Hz,2H),4.12(t,J=5.4Hz,2H),3.89(s,2H),3.49(d,J=5.0Hz,3H),2.82(t,J=5.3Hz,2H),2.09–2.04(m,1H),0.98(d,J=8.0Hz,4H)。
Example 360: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [2- (methylamino) ethyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALZ-3)
Step 1: n- (2- { [ (2- {4- [ 4-cyano-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-cyclopropylpyridin-2-yl } -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl) methyl ] amino } ethyl) -N-methylcarbamic acid tert-butyl ester (ALZ-2)
To a stirred mixture of intermediate (AGT-2) (60 mg,1 eq, 0.11 mmol) and tert-butyl N- (2-aminoethyl) -N-methylcarbamate (ALZ-1) (24 mg,1.2 eq, 0.14 mmol) in MeOH (5 mL) was added DIPEA (44 mg,3 eq, 0.34 mmol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. NaBH was added to the above mixture at room temperature 4 (13 mg,3 eq, 0.34 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the residue as a yellow solidThe sub-title compound (ALZ-2) (40 mg, 64. Mu. Mol,51%,95% purity). M/z 619.3 (M+H) + (ES+)
Step 2:4- { 2-cyclopropyl-6- [6- ({ [2- (methylamino) ethyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ALZ-3)
A solution of the product from step 1 above (ALZ-2) (60 mg,1 eq, 87. Mu. Mol) and TFA (2 mL) in DCM (10 mL) was stirred at room temperature for 1 hour. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 25% B over 10 min; wavelength: 254nm; retention time: 11.02) to give the title compound (ALZ-3) (15.5 mg, 30. Mu. Mol,27%,95.0% purity) as a white solid. M/z 519.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.52(d,J=2.5Hz,1H),8.36(s,0.761H),8.28–8.14(m,2H),7.96(d,J=3.8Hz,1H),7.91–7.84(m,1H),7.80(s,1H),7.66(d,J=1.4Hz,2H),6.91(d,J=1.5Hz,1H),5.01(s,2H),3.82(s,2H),3.62(s,3H),2.94(s,2H),2.71(t,J=6.4Hz,2H),2.54(s,3H),2.10–2.01(m,1H),1.05–0.91(m,4H)。
Example 361: synthesis of 4- (2-cyclopropyl-6- {6- [ ({ [1- (hydroxymethyl) cyclobutyl ] methyl } amino) methyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMA-2)
To intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and [1- (aminomethyl) cyclobutyl at room temperature]To a stirred mixture of methanol (AMA-1) (8 mg,1 eq, 65. Mu. Mol) in MeOH (10 mL) was added DIPEA (25 mg,3 eq, 0.20 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. NaBH was added to the above mixture at room temperature 4 (7 mg,3 equivalents, 0.20 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. Then atThe reaction was quenched by the addition of MeOH (2 mL) at 0deg.C. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 50% B in 8 minutes; wavelength: 254/220nm; retention time: 7.83 To give the title compound (AMA-2) (2.3 mg,4.1 μmol,5.7%,90.0% purity) as a white solid. M/z 561.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.16-8.03 (m, 3H), 7.92 (d, J=8.1 Hz, 1H), 7.81 (s, 1H), 7.70-7.60 (m, 2H), 6.92 (d, J=1.3 Hz, 1H), 5.03 (s, 2H), 3.92 (s, 2H), 3.63 (s, 2H), 3.50 (s, 3H), 2.77 (s, 2H), 2.07-1.99 (m, 1H), 1.98-1.75 (m, 6H), 1.10-0.92 (m, 4H).
Example 362: synthesis of N- (2- { [ (2- {4- [ 4-cyano-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-cyclopropylpyridin-2-yl } -3-oxo-1H-isoindol-5-yl) methyl ] amino } ethyl) acetamide (AMB-1)
A solution of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol), N- (2-aminoethyl) acetamide (AJB-1) (8 mg,1.2 eq, 78. Mu. Mol) and DIPEA (34 mg,4 eq, 0.26 mmol) in MeOH (8 mL) was stirred at room temperature overnight. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was acidified with FA to pH 5 at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge Shield RP OBD column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 20% B to 35% B in 10 minutes; wavelength: 254/220nm; retention time: 10.7 To give the title compound (AMB) as a white solid -1) (8.6 mg,16 μmol,23%,96.1% purity). M/z 547.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.48 (s, 1H), 8.15-8.09 (m, 1H), 8.09-8.04 (m, 2H), 7.92 (d, j=8.1 hz, 1H), 7.83-7.78 (m, 1H), 7.68-7.59 (m, 2H), 6.90 (d, j=1.4 hz, 1H), 5.02 (s, 2H), 3.87 (s, 2H), 3.49 (s, 3H), 3.32 (s, 2H), 2.71 (t, j=6.4 hz, 2H), 2.08-1.97 (m, 1H), 1.94 (s, 3H), 1.06-0.93 (m, 4H).
Example 363: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (cyclopropylmethyl) amino ] methyl } -4-fluoro-1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMC-2)
Step 1:6- { [ (cyclopropylmethyl) amino ] methyl } -4-fluoro-2, 3-dihydro-isoindol-1-one (AMC-1)
To a stirred mixture of intermediate (AIO-5) (150 mg,1 eq, 0.84 mmol) and 1-cyclopropylmethylamine (AIB-1) (71 mg,1.2 eq, 1.00 mmol) in MeOH (20 mL) was added DIPEA (325 mg,3 eq, 2.51 mmol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (95 mg,3 equivalents, 2.51 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of 5mL of MeOH at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1) to give the sub-title compound (AMC-1) (130 mg,0.56mmol,66%,94% pure) as a yellow solid. M/z 235.1 (M+H) + (ES+)
Step 2:4- [ 2-cyclopropyl-6- (6- { [ (cyclopropylmethyl) amino ] methyl } -4-fluoro-1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMC-2)
To the product (AMC-1) (130 mg,1 equivalent, 0.56 mmol), intermediate (AEG-2) (186 mg,1 equivalent, 0.56 mmol) and Cs from step 1 above, under a nitrogen atmosphere at room temperature 2 CO 3 (362 mg,2 equivalents, 1.11 mmol) in dioxane (20 mTo the stirred mixture in L) were added RuPhos (104 mg,0.4 eq, 0.22 mmol) and RuPhos ring palladium complex Gen.3 (93 mg,0.2 eq, 0.11 mmol). The resulting mixture was stirred under nitrogen at 60 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (20% MeCN up to 60% in 20 minutes); detector, UV 254/220nm. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 39% B to 59% B in 9 minutes; wavelength: 254/220nm; retention time: 8.05 To give the title compound (AMC-2) as a white solid (29.4 mg,55 purity, 9.9%,99.2% purity). M/z 534.1 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.31–8.06(m,2H),7.95(d,J=1.4Hz,1H),7.92–7.83(m,1H),7.62(s,1H),7.51(d,J=10.0Hz,1H),6.92(d,J=1.4Hz,1H),5.06(s,2H),3.83(s,2H),3.48(s,3H),2.36(d,J=6.6Hz,2H),2.12–1.98(m,1H),1.11–0.94(m,4H),0.93–0.78(m,1H),0.44–0.32(m,2H),0.14–0.04(m,2H)。
Example 364: synthesis of 4- { 2-cyclopropyl-6- [ 4-fluoro-6- ({ [ (2S, 3S) -3-methoxybutan-2-yl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMD-2)
To a stirred mixture of intermediate (AIO-8) (30 mg,1 eq, 63. Mu. Mol) and (2S, 3S) -3-methoxybutane-2-amine (AMD-1) (6 mg,1 eq, 63. Mu. Mol) in DCM (2 mL) at room temperature was added Et 3 N (24 mg,3 eq, 0.19 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. NaBH (OAc) was added to the above mixture at room temperature 3 (7 mg,3 equivalents, 0.19 mmol). Will be spentThe resulting mixture was stirred at room temperature for another 1 hour. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 67% B in 8 minutes; wavelength: 254/220nm; retention time: 6.65 To give the title compound (AMD-2) as a white solid (15.5 mg,27 μmol,44%,99.6% purity). M/z 566.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.25–8.18(m,2H),7.95(d,J=1.4Hz,1H),7.90–7.84(m,1H),7.65(s,1H),7.53(d,J=10.0Hz,1H),6.93(d,J=1.4Hz,1H),5.08(s,2H),3.86(d,J=26.5Hz,2H),3.48(s,3H),3.31(s,1H),3.23(s,4H),2.10–2.02(m,1H),1.11–0.87(m,10H)。
Example 365: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (3, 3-difluoropropyl) amino ] methyl } -4-fluoro-1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AME-2)
DIPEA (24 mg,3 eq, 0.19 mmol) was added to a stirred mixture of intermediate (AIO-8) (30 mg,1 eq, 63. Mu. Mol) and 3, 3-difluoropropan-1-amine (AME-1) (6 mg,1 eq, 63. Mu. Mol) in MeOH (5 mL) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. NaBH was added to the above mixture at room temperature 4 (7 mg,3 equivalents, 0.19 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 42% B to 52% B in 10 minutes; wavelength: 254/220nm;retention time: 8.92 To give the title compound (AME-2) (4.8 mg,8.6 μmol,14%,98.4% purity) as a white solid. M/z 558.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.38–8.08(m,2H),7.95(d,J=1.4Hz,1H),7.91–7.79(m,1H),7.63(s,1H),7.56–7.47(m,1H),6.93(d,J=1.4Hz,1H),6.32–5.99(m,1H),5.07(s,2H),3.81(s,2H),3.48(s,3H),2.59(t,J=7.0Hz,2H),2.17–1.80(m,3H),1.22–0.74(m,4H)。
Example 366: synthesis of 4- [2- (6- { [ (cyclobutylmethyl) (methyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMF-1)
To a stirred mixture of compound (AIA-2) (30 mg,1 eq, 57. Mu. Mol) and HCHO (1.7 mg,1 eq, 57. Mu. Mol) in MeOH (5 mL) was added DIPEA (22 mg,3 eq, 0.17 mmol) at room temperature. The resulting mixture was stirred overnight at 60 ℃ under an air atmosphere. The mixture was cooled to room temperature. AcOH (0.50 m) and NaBH were added to the above mixture at room temperature 3 CN (11 mg,3 equivalents, 0.17 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 15% B to 45% B over 10 min; wavelength: 254nm; retention time: 7.87) to give the title compound (AMF-1) (7.4 mg, 14. Mu. Mol,24%,98.9% purity) as a white solid. M/z 544.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.16-8.00 (m, 3H), 7.92 (d, j=8.1 hz, 1H), 7.78 (s, 1H), 7.68-7.58 (m, 2H), 6.93 (d, j=1.5 hz, 1H), 5.03 (s, 2H), 3.65 (s, 2H), 3.50 (s, 3H), 2.70-2.58 (m, 1H), 2.50 (s, 2H), 2.22 (d, j=3.7 hz, 3H), 2.15-1.98 (m, 3H), 1.98-1.86 (m, 1H), 1.85-1.64 (m, 3H), 1.08-0.94 (m, 4H).
Example 367: synthesis of 2- (4- (4-chloro-2- (4-methyl-4H-1, 2, 4-triazol-3-yl) phenyl) -6-cyclopropylpyridin-2-yl) -6- ((((1-hydroxycyclobutyl) methyl) (methyl) amino) methyl) isoindolin-1-one (AMI-1)
To a 25mL round bottom flask was added compound (AGW-6) (60 mg,1 eq, 0.11 mmol), meOH (5 mL) containing HCHO (1 mL) at room temperature. NaBH was added to the above-mentioned stirred solution at room temperature 3 CN (34 mg,5 eq, 0.54 mmol). The resulting mixture was stirred at 60 ℃ for an additional 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.05% TFA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 32% B over 10 min; wavelength: 254 nm) to give the title compound (AMG-1) (7.2 mg, 13. Mu. Mol,12%,99.8% purity) as a white solid. M/z569.2/571.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.50(s,1H),8.00(s,1H),7.84–7.79(m,1H),7.79–7.74(m,2H),7.69(d,J=8.3Hz,1H),7.64(s,2H),6.80(s,1H),5.00(s,2H),4.89(s,1H),3.86(s,2H),3.41(S,3H)2.53(s,3H),2.09–1.83(m,7H),1.61(d,J=10.7Hz,1H),1.44–1.35(m,1H),0.95(d,J=6.4Hz,4H)。
Example 368: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ (2-hydroxy-2-methylpropyl) (methyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMH-1)
To a stirred mixture of compound (AIE-1) (60 mg,1 eq, 0.11 mmol) and HCHO (17 mg,5 eq, 0.56 mmol) in MeOH (5 mL) was added DIPEA (44 mg,3 eq, 0.34 mmol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. At room temperature Adding NaBH to the mixture 3 CN (21 mg,3 equivalents, 0.34 mmol) and AcOH (0.2 mL). The resulting mixture was stirred at room temperature for an additional 1 hour. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 6% B to 36% B over 10 min; wavelength: 254nm; retention time: 9.4) to give the title compound (AMH-1) (3.6 mg, 6.6. Mu. Mol,5.8%,99.5% purity) as a white solid. M/z 548.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.51 (s, 1H), 8.16-8.07 (m, 2H), 8.04 (d, j=1.4 hz, 1H), 7.96-7.89 (m, 1H), 7.83 (t, j=1.1 hz, 1H), 7.73-7.66 (m, 1H), 7.59 (d, j=7.8 hz, 1H), 6.93 (d, j=1.4 hz, 1H), 5.02 (s, 2H), 3.73 (s, 2H), 3.51 (s, 3H), 2.45 (s, 2H), 2.30 (s, 3H), 2.09-1.98 (m, 1H), 1.21 (s, 6H), 1.08-0.95 (m, 4H).
Example 369: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -4-fluoro-6- (hydroxymethyl) -3H-isoindol-1-one (AMI-1)
To a 25mL round bottom flask was added under nitrogen at room temperature intermediate (ADN-1) (60 mg,1 eq, 0.18 mmol), intermediate (AIO-6) (40 mg,1.2 eq, 0.22 mmol) and Cs 2 CO 3 (178 mg,3 eq, 0.55 mmol) of 1, 4-dioxane (5 mL). RuPhos (34 mg,0.4 eq, 73. Mu. Mol) and RuPhos ring palladium complex Gen.3 (31 mg,0.2 eq, 36. Mu. Mol) were added to the above stirred solution at room temperature under nitrogen atmosphere. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 2 hours. The mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XBridge preparative OBD C18 column, 30X 150mm,5 μm; mobile phase A): water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 65% B in 8 minutes; wavelength: 254 nm) to give the title compound (AMI-1) (7.8 mg,16 μmol,9.0%,99.8% purity) as a white solid. M/z 474.1 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.51(s,1H),7.96(d,J=1.4Hz,1H),7.75–7.69(m,1H),7.64–7.57(m,3H),7.45(d,J=9.9Hz,1H),6.83(d,J=1.4Hz,1H),5.53–5.47(m,1H),5.07(s,2H),4.62(d,J=5.7Hz,2H),3.43(s,3H),2.06–1.98(m,1H),0.99–0.93(m,4H)。
Example 370: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -4-fluoro-6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -3H-isoindol-1-one (AMJ-1)
To a 25mL round bottom flask was added compound (AMI-1) (60 mg,1 eq, 0.13 mmol), DMP (70 mg,1.3 eq, 0.17 mmol) and DCM (5 mL) at room temperature. The resulting mixture was stirred at room temperature for 2 hours. DIPEA (49 mg,3 eq, 0.38 mmol) and 1- (aminomethyl) cyclobutan-1-ol (AW-1) (17 mg,1.3 eq, 0.17 mmol) were added to the above stirred solution at room temperature. The resulting mixture was stirred at 60 ℃ for an additional overnight. The mixture was cooled to room temperature. Adding NaBH to the above stirred solution at 0deg.C 4 (60 mg,5 equivalents, 1.59 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 65% B in 8 minutes; wavelength: 254/220nm; retention time: 7.38). This gave the title compound (AMJ-1) (4.7 mg, 8.5. Mu. Mol,6.6%,99.4% purity) as a white solid. M/z 557.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.47 (s, 1H), 8.01 (d, J=1.4 Hz, 1H), 7.80-7.74 (m, 1H), 7.67 (s, 1H), 7.56-7.45 (m, 3H), 6.89 (d, J=1.4 Hz, 1H), 5.07 (s, 2H), 3.95 (s, 2H), 3.47 (s, 3H), 2.71 (s, 2H), 2.13-1.98 (m, 5H), 1.78-1.70 (m, 1H), 1.56-1.46 (m, 1H), 1.06-0.95 (m, 4H).
Example 371: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [2- (methylamino) ethyl ] amino } methyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMK-3)
Step 1:4- { 2-cyclopropyl-6- [ 6-formyl-1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMK-1)
A solution of compound (AFK-1) (30 mg,1 eq, 57. Mu. Mol) and DMP (31 mg,1.3 eq, 74. Mu. Mol) in DCM (5 mL) was stirred at room temperature under an air atmosphere for 1 hour. Filtering the resulting mixture; the filter cake was washed with MeOH (2X 3 mL). The filtrate was concentrated in vacuo. The crude product mixture was used directly in the next step without further purification. M/z 529.2 (M+H) + (ES+)
Step 2: n- (2- { [ (2- {4- [ 4-cyano-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-cyclopropylpyridin-2-yl } -3-oxo-7- (trifluoromethyl) -1H-isoindol-5-yl) methyl ] amino } ethyl) -N-methylcarbamic acid tert-butyl ester (AMK-2)
To a stirred mixture of the product from step 1 above (AMK-1) (60 mg,1 eq, 0.11 mmol) and tert-butyl N- (2-aminoethyl) -N-methylcarbamate (ALZ-1) (24 mg,1.2 eq, 0.14 mmol) in MeOH (5 mL) was added DIPEA (44 mg,3 eq, 0.34 mmol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. NaBH was added to the above mixture at room temperature 4 (13 mg,3 eq, 0.34 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue is taken up Purification by preparative TLC using DCM/MeOH (10/1) gave the sub-title compound (AMK-2) (40 mg, 58. Mu. Mol,51%,95% pure) as a yellow solid. M/z 687.2 (M+H) + (ES+)
Step 3:4- { 2-cyclopropyl-6- [6- ({ [2- (methylamino) ethyl)]Amino } methyl) -1-oxo-4- (trifluoromethyl) -3H-isoindol-2-yl]Pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMK-3). A solution of the product from step 2 above (AMK-2) (60 mg,1 eq, 87. Mu. Mol) and TFA (2 mL) in DCM (10 mL) was stirred at room temperature for 1 hour. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 25% B over 10 min; wavelength: 254nm; retention time: 11.02) to give the title compound (AMK-3) (15.5 mg, 26. Mu. Mol,27%,86.5% purity) as a white solid. M/z 587.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.37(d,J=5.3Hz,0.814H),8.27–8.17(m,2H),8.17–7.93(m,3H),7.92–7.83(m,1H),6.97–6.84(m,1H),5.16(s,2H),3.89(s,2H),3.49(s,5H),2.85–2.58(m,4H),2.41(s,3H),2.13–2.02(m,1H),1.10–0.85(m,4H)。
Example 372: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -4-fluoro-6- ({ [ (1-hydroxycyclobutyl) methyl ] (methyl) amino } methyl) -3H-isoindol-1-one (AML-1)
To a 100mL round bottom flask at room temperature was added compound (AMJ-1) (50 mg,1 eq, 0.09 mmol) and MeOH (5 mL). Formaldehyde (1 mL) and NaBH were added to the above stirred solution at room temperature 3 CN (17 mg,3 equivalents, 0.27 mmol). The resulting mixture was stirred at 60 ℃ for an additional 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 65% B in 8 minutes; wavelength: 254 nm) to give the title compound (AML-1) (4.4 mg, 8.53%) as a white solid. M/z 571.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.47 (s, 1H), 8.00 (d, J=1.4 Hz, 1H), 7.81-7.76 (m, 1H), 7.67-7.64 (m, 1H), 7.56-7.50 (m, 1H), 7.49-7.45 (m, 2H), 6.89 (d, J=1.4 Hz, 1H), 5.06 (s, 2H), 3.73 (s, 2H), 3.47 (s, 3H), 2.59 (s, 2H), 2.31 (s, 3H), 2.14-1.98 (m, 5H), 1.83-1.74 (m, 1H), 1.51-1.43 (m, 1H), 1.06-0.96 (m, 4H).
Example 373: synthesis of 2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- { [ (2-hydroxy-2-methylpropyl) (methyl) amino ] methyl } -3H-isoindol-1-one (AMM-1)
To a stirred solution of compound (AHG-2) (50 mg,1 eq, 95. Mu. Mol) and HCHO (29 mg,10 eq, 0.95 mmol) in MeOH (10 mL) at room temperature was added DIPEA (49 mg,4 eq, 0.38 mmol) and NaBH 3 CN (24 mg,4 equivalents, 0.38 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 65% B in 8 minutes; wavelength: 254/220nm; retention time: 7.52 To give the title compound (AMM-1) (5.3 mg,9.8 μmol,10%,98.5% purity) as a white solid. M/z 541.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.48 (s, 1H), 8.02 (d, J=1.4 Hz, 1H), 7.83 (t, J=1.0 Hz, 1H), 7.80-7.75 (m, 1H), 7.71-7.67 (m, 1H), 7.61-7.57 (m, 1H), 7.56-7.51 (m, 1H), 7.49-7.45 (m, 1H), 6.86 (d, J=1.4 Hz, 1H), 5.02 (s, 2H), 3.73 (s, 2H), 3.48%s,3H),2.45(s,2H),2.30(s,3H),2.05–1.95(m,1H),1.21(s,6H),1.05–0.99(m,2H),0.99–0.94(m,2H)。
Example 374: synthesis of 4- (2-cyclopropyl-6- {6- [ (1S, 4S) -2-oxa-5-azabicyclo [2.2.1] heptane-5-ylmethyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMN-1)
To intermediate (AIQ-1) (40 mg,1 eq, 83. Mu. Mol) and (1S, 4S) -2-oxa-5-azabicyclo [2.2.1 ] at room temperature]To a stirred solution of heptane (AKR-2) (12 mg,1.5 eq, 0.12 mmol) in DCM (5 mL) was added Et 3 N (2 mg,5 eq, 0.42 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature and concentrated in vacuo. The resulting mixture was diluted with water and extracted with DCM (3×20 mL). The combined organic layers were washed with brine (2×20 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30×150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35B to 45B in 9 minutes; a detector, UV 254/210nm; retention time: 8.05. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (amb-1) (10.2 mg,19 μmol,23%,99.8% purity) as a white solid. M/z 544.2 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.25–8.15(m,2H),7.98(d,J=1.4Hz,1H),7.87(d,J=8.6Hz,1H),7.72(s,1H),7.68–7.61(m,2H),6.89(d,J=1.4Hz,1H),5.00(s,2H),4.36(d,J=2.2Hz,1H),3.94(d,J=7.5Hz,1H),3.90–3.76(m,2H),3.57–3.42(m,5H),2.76–2.70(m,1H),2.42(d,J=10.0Hz,1H),2.09–2.00(m,1H),1.87–1.79(m,1H),1.63–1.54(m,1H),0.97(d,J=6.3Hz,4H)。
Example 378: synthesis of 4- (2-cyclopropyl-6- {6- [ (oxa-4-ylamino) methyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMO-2)
Et is added to a stirred solution of intermediate (AGT-2) (40 mg,1 eq, 87. Mu. Mol) and tetrahydro-2H-pyran-4-amine (AMO-1) (18 mg,2 eq, 0.17 mmol) in MeOH (5 mL) at 60 ℃ 3 N (44 mg,5 eq, 0.44 mmol) for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (33 mg,10 equivalents, 0.87 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30×150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38B to 48B in 8 minutes; a detector, UV 254/210nm; retention time: 7.52. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (AMO-2) (11.9 mg,22 μmol,25%,99.2% purity) as a white solid. M/z 546.2 (M+H) + (ES+). 1 H NMR(300MHz,DMSO-d6)δ8.54(s,1H),8.27-8.13(m,2H),7.98(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.78(s,1H),7.70-7.60(m,2H),6.89(d,J=1.4Hz,1H),5.01(s,2H),3.83(d,J=15.3Hz,4H),3.48(s,3H),3.31-3.20(m,3H),2.07-2.02(m,1H),1.81-1.77(m,2H),1.31-1.24(m,2H),0.98-0.96(m,4H)。
Example 379: synthesis of 4- [ 2-cyclopropyl-6- (4, 5-difluoro-6- { [ (2-methoxyethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMP-1)
In the roomTo a 25mL round bottom flask was added MeOH (5 mL) containing intermediate (ALY-1) (50 mg,1 eq, 0.10 mmol) at temperature. DIPEA (39 mg,3 eq, 0.30 mmol) and 2-methoxyethyl-1-amine (AEB-1) (9 mg,1.2 eq, 0.12 mmol) were added to the above mixture at room temperature. The resulting mixture was stirred at 60 ℃ for an additional 12 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (15 mg,4 eq, 0.40 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 50% B in 9 minutes; wavelength: 254 nm) to give the title compound (AMP-1) (1.5 mg, 2.7. Mu. Mol,2.5%,93.3% purity) as a white solid. M/z 556.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.15-8.08 (m, 2H), 8.00 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.74 (d, j=5.4 hz, 1H), 6.95 (d, j=1.5 hz, 1H), 5.11 (s, 2H), 3.96 (s, 2H), 3.53 (d, j=5.2 hz, 2H), 3.49 (s, 3H), 3.35 (s, 3H), 2.82-2.77 (m, 2H), 2.07-2.02 (m, 1H), 1.08-1.04 (m, 2H), 1.02-0.99 (m, 2H).
Example 380: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ cyclopropyl (methyl) amino ] methyl } -4-fluoro-1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMQ-2)
To a stirred mixture of intermediate (AKR-1) (30 mg,1 eq, 0.06 mmol) and N-methylcyclopropylamine (AMQ-1) (6.4 mg,1.5 eq, 0.09 mmol) in DCM (5 mL) was added DIPEA (23 mg,3 eq, 0.18 mmol) at 60 ℃. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate):60 ml/min; gradient: 3% B to 33% B in 10 minutes; wavelength: 254nm; retention time: 10.72 To give the title compound (AMQ-2) (5.8 mg,11 μmol,18%,99.3% purity) as a white solid. M/z 534.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.16-8.07 (m, 2H), 8.01 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.63 (s, 1H), 7.39 (d, j=9.6 hz, 1H), 6.97 (d, j=1.4 hz, 1H), 5.07 (s, 2H), 3.89 (s, 2H), 3.50 (s, 3H), 2.37 (s, 3H), 2.11-2.00 (m, 1H), 1.92 (s, 1H), 1.09-0.95 (m, 4H), 0.62-0.43 (m, 4H).
Example 381: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (3S) -3-hydroxy-3-methylpiperidin-1-yl ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMR-2)
To a stirred solution of intermediate (AKR-1) (30 mg,1 eq, 0.06 mmol) and (3S) -3-methylpiperidin-3-ol (AMR-1) (8.3 mg,1.2 eq, 72 μmol) in DCM (8 mL) at room temperature was added DIPEA (31 mg,4 eq, 0.24 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1%% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 52% B to 62% B in 9 minutes; wavelength: 254/220nm; retention time: 7.32 To give the title compound (AMR-2) as a white solid (8.6 mg,15 μmol,25%,99.3% purity). M/z 578.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.07 (m, 2H), 8.02 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.66-7.63 (m, 1H), 7.49 (d, j=9.8 hz, 1H), 6.96 (d, j=1.5 hz, 1H), 5.06 (s, 2H), 3.61 (s, 2H), 3.50 (s, 3H), 2.52 (s, 1H), 2.39-2.25 (m, 2H), 2.18 (d, j=10.9 hz, 1H), 2.09-2.01 (m, 1H), 1.85-1.76 (m, 1H), 1.58-1.43 (m, 3H), 1.19 (s, 3H), 1.07-0.97 (m, 4H).
Example 382: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { 2-oxa-6-azaspiro [3.3] heptan-6-ylmethyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMS-2)
To intermediate (AKR-1) (30 mg,1 eq, 0.06 mmol) and 2-oxa-6-azaspiro [3.3] at room temperature]To a stirred solution of heptane (AMS-1) (8.9 mg,1.5 eq, 0.09 mmol) in DCM (8 mL) was added DIPEA (31 mg,4 eq, 0.24 mmol). The resulting mixture was stirred at 60℃for 1 day. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 9% B to 39% B in 10 min; wavelength: 254nm; retention time: 8.45) to give the title compound (AMS-2) (7.6 mg, 14. Mu. Mol,22%,99.3% purity) as a white solid. M/z 562.0 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.29–8.18(m,2H),7.95(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.50(s,1H),7.41(d,J=9.8Hz,1H),6.92(d,J=1.4Hz,1H),5.07(s,2H),4.61(s,4H),3.61(s,2H),3.49(s,3H),3.47(s,4H),2.11–2.00(m,1H),1.01–0.94(m,4H)。
Example 383: synthesis of 4- [ 2-cyclopropyl-6- (6- {2- [ (oxetan-3-ylmethyl) amino ] propan-2-yl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMT-2)
To a stirred solution of intermediate (ALK-9) (30 mg,1 eq, 61 μmol) and oxetane-3-carbaldehyde (AMT-1) (16 mg,3 eq, 0.18 mmol) in MeOH (3 mL) at room temperature was added DIPEA (40 mg,5 eq, 0.31 mmol). The resulting mixture was stirred at 60 ℃2 hours. The mixture was cooled to room temperature. NaBH was added to the above mixture at room temperature 4 (12 mg,5 equivalents, 0.31 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (5/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35% B to 48% B in 9 minutes; wavelength: 254/220 nm) to give the title compound (AMT-2) (4.0 mg, 7.2. Mu. Mol,11%,98.1% purity) as a white solid. M/z 560.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.51 (s, 1H), 8.17-8.09 (m, 2H), 8.05 (d, j=1.4 hz, 1H), 7.97-7.90 (m, 2H), 7.87-7.81 (m, 1H), 7.63 (d, j=8.1 hz, 1H), 6.94 (d, j=1.5 hz, 1H), 5.03 (s, 2H), 4.78-4.72 (m, 2H), 4.26 (t, j=6.0 hz, 2H), 3.51 (s, 3H), 3.06-2.96 (m, 1H), 2.57 (d, j=7.5 hz, 2H), 2.09-1.98 (m, 1H), 1.53 (s, 6H), 1.08-0.96 (m, 4H).
Example 384: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (3-methyloxetan-3-yl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMU-2)
To a stirred mixture of intermediate (AGT-2) (30 mg,1 eq, 64. Mu. Mol), 1- (3-methyloxetan-3-yl) methylamine (AMU-1) (12 mg,1.8 eq, 0.12 mmol) and DIPEA (33.11 mg,0.256mmol,4 eq) in MeOH (8 mL) at room temperature. The resulting mixture was stirred at 60℃for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (10 mg,4 equivalents, 0.26 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. Passing the crude product through a preparative processHPLC was performed under the following conditions (column: XBridge-prepared OBD C18 column, 30X 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 63% B in 9 minutes; wavelength: 254/220nm; retention time: 8.98 To give the title compound (AMU-2) (6.0 mg,11 μmol,17%,98.9% purity) as a white solid. M/z 546.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.17-8.04 (m, 3H), 7.96-7.84 (m, 2H), 7.74-7.70 (m, 1H), 7.65 (d, j=7.9 hz, 1H), 6.93 (d, j=1.4 hz, 1H), 5.04 (s, 2H), 4.46 (d, j=5.9 hz, 2H), 4.35 (d, j=5.9 hz, 2H), 4.01 (s, 2H), 3.49 (s, 3H), 2.89 (s, 2H), 2.07-1.97 (m, 1H), 1.34 (s, 3H), 1.09-0.92 (m, 4H).
Example 385: synthesis of 4- (2-cyclopropyl-6- {6- [ (oxetan-3-ylamino) methyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMV-2)
To a stirred solution of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and oxetan-3-amine (AMV-1) (5.7 mg,1.2 eq, 78. Mu. Mol) in MeOH (12 mL) was added DIPEA (25 mg,3 eq, 0.20 mmol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35% B to 45% B in 8 minutes; wavelength: 254/220nm; retention time: 7.22 To give the title compound (AMV-2) (11.5 mg,22 μm) as a white solidol,34%,99.3% purity). M/z 518.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.14-8.04 (m, 3H), 7.92 (d, j=8.1 hz, 1H), 7.79 (d, j=1.6 hz, 1H), 7.67-7.59 (m, 2H), 6.92 (d, j=1.5 hz, 1H), 5.02 (s, 2H), 4.71 (t, j=6.8 hz, 2H), 4.44 (t, j=6.4 hz, 2H), 4.05-3.98 (m, 1H), 3.80 (s, 2H), 3.49 (s, 3H), 2.06-1.99 (m, 1H), 1.06-0.95 (m, 4H).
Example 386: synthesis of 4- { 2-cyclopropyl-6- [ 4-fluoro-6- ({ [ (1-hydroxycyclobutyl) methyl ] (methyl) amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMW-2)
To intermediate (AKR-1) (30 mg,1 eq, 0.06 mmol) and 1- [ (methylamino) methyl at room temperature]To a stirred solution of cyclobutan-1-ol (AMW-1) (14 mg,2 eq, 0.12 mmol) in DCM (5 mL) was added DIPEA (23 mg,3 eq, 0.18 mmol). The resulting mixture was stirred at 60℃for 4 days. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 4% B to 34% B in 10 min; wavelength: 254 nm) to give the title compound (AMW-2) (7.4 mg, 13. Mu. Mol,21%,98.1% purity) as a white solid. M/z 578.0 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.55(s,1H),8.25–8.20(m,2H),7.94(d,J=1.4Hz,1H),7.89–7.84(m,1H),7.61(s,1H),7.52(d,J=10.1Hz,1H),6.93(d,J=1.4Hz,1H),5.08(s,2H),4.93(s,1H),3.72(s,2H),3.48(s,3H),2.48(s,2H),2.23(s,3H),2.10–1.98(m,3H),1.95–1.84(m,2H),1.67–1.57(m,1H),1.42–1.32(m,1H),1.03–0.93(m,4H)。
Example 387: synthesis of 4- (2- {6- [ (1R, 5S) -2-azabicyclo [3.1.0] hexane-2-ylmethyl ] -4-fluoro-1-oxo-3H-isoindol-2-yl } -6-cyclopropylpyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMX-3)
Step 1:4- [2- (6- { 2-azabicyclo [3.1.0] hex-2-ylmethyl } -4-fluoro-1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMX-2)
To a stirred solution of intermediate (AKR-1) (100 mg,1 eq, 0.20 mmol) and DIPEA (78 mg,3 eq, 0.60 mmol) in DCM (15 mL) under nitrogen at room temperature was added 2-azabicyclo [3.1.0]Hexane, HCl (AMX-1) (29 mg,1.2 eq, 0.24 mmol). The resulting mixture was stirred overnight at 60 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18OBD column 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 18% B to 24% B over 15 min; wavelength: 254nm; retention time: 14.03) to give the sub-title compound (AMX-2) as a white solid (43 mg, 79. Mu. Mol,39%,99% purity). M/z 546.1 (M+H) + (ES+)。
Step 2:4- (2- {6- [ (1R, 5S) -2-azabicyclo [3.1.0] hex-2-ylmethyl ] -4-fluoro-1-oxo-3H-isoindol-2-yl } -6-cyclopropylpyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMX-3)
The product from step 1 (AMX-2) (40 mg,1 eq., 73. Mu. Mol) was purified by chiral preparative HPLC under the following conditions (column: CHIRALPAK IF, 2X 25cm,5 μm; mobile phase A: mtBE (0.5% 2M NH) 3 MeOH) -HPLC, mobile phase B: meOH dcm=1:1— HPLC; flow rate: 17 ml/min; gradient: 20% B to 20% in 55 min; wavelength: 220/254nm; retention time 1 (min): 44.016, sample solvent: etOH-HPLC; injection volume: 1 mL) to give the title compound (AMX-3) (10.5 mg,19 μmol,26%,99.1% purity) as a white solid. M/z 546.1 (M+H) + (ES+)。 1 H NMR (300 MHz, meOH-d 4) δ8.49 (s, 1H), 8.16-8.07 (m, 2H), 8.02 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.69 (d, j=1.2 hz, 1H), 7.51-7.44 (m, 1H), 6.95 (d, j=1.4 hz, 1H), 5.07 (s, 2H), 3.84 (s, 2H), 3.50 (s, 3H), 2.98-2.85 (m, 1H), 2.74-2.65 (m, 1H), 2.22-2.10 (m, 1H), 2.11-1.87 (m, 3H), 1.56-1.46 (m, 1H), 1.09-0.94 (m, 4H), 0.91-0.82 (m, 1H), 0.32-0.21 (m, 1H). Chiral HPLC-PH-NIMB-0898-0: column: CHIRALPAK IF-3,4.6 x 50mm,3 μm; mobile phase a: mtBE (0.1% DEA): (MeOH: dcm=1:1) =80:20; flow rate: 1 ml/min; retention time: 5.437.
Example 388: synthesis of 4- (2- {6- [ (1S, 5R) -2-azabicyclo [3.1.0] hexan-2-ylmethyl ] -4-fluoro-1-oxo-3H-isoindol-2-yl } -6-cyclopropylpyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMY-1)
Compound (AMX-2) (40 mg,1 eq, 73. Mu. Mol) was purified by chiral preparative HPLC under the following conditions (column: CHIRALPAK IF, 2X 25cm, 5. Mu.m; mobile phase A: mtBE (0.5% 2M NH) 3 MeOH) -HPLC, mobile phase B: meOH dcm=1:1— HPLC; flow rate: 17 ml/min; gradient: 20% B to 20% in 55 min; wavelength: 220/254nm; retention time 2 (minutes): 19.329; sample solvent: etOH-HPLC; injection volume: 1 mL) to give the title compound (AMY-1) (7.5 mg,14 μmol,19%,99.1% purity) as a white solid. M/z 546.1 (M+H) + (ES+)。 1 H NMR (300 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.16-8.07 (m, 2H), 8.02 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.69 (d, j=1.2 hz, 1H), 7.51-7.44 (m, 1H), 6.95 (d, j=1.4 hz, 1H), 5.07 (s, 2H), 3.84 (s, 2H), 3.50 (s, 3H), 2.98-2.85 (m, 1H), 2.74-2.65 (m, 1H), 2.22-2.10 (m, 1H), 2.11-1.87 (m, 3H), 1.56-1.46 (m, 1H), 1.09-0.94 (m, 4H), 0.91-0.82 (m, 1H), 0.32-0.21 (m, 1H). Chiral HPLC-PH-NIMB-0898-0: column: CHIRALPAK IF-3,4.6 x 50mm,3 μm; mobile phase a: mtBE (0.1% DEA): (MeOH: dcm=1:1) =80:20; flow rate: 1 ml/min; retention time: 5.995.
Example 389: synthesis of 4- [ 2-cyclopropyl-6- (4, 5-difluoro-6- { [ (2-methoxyethyl) (methyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMZ-3)
Step 1:4- {2- [6- (chloromethyl) -4, 5-difluoro-1-oxo-3H-isoindol-2-yl ] -6-cyclopropylpyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMZ-1)
To a 50mL round bottom flask was added DCM (10 mL) containing intermediate (ALX-9) (100 mg,1 eq, 0.20 mmol) at room temperature. Adding SOCl to the above stirred solution at 0deg.C 2 (72 mg,3 equivalents, 0.60 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of 5mL of MeOH at 0 ℃. The resulting mixture was diluted with water and extracted with DCM (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. This gave the sub-title compound (AMZ-1) (74 mg,0.14mmol,62%,92% purity) as a white solid. M/z 517.1/519.1 (M+H) + (ES+)
Step 2:4- [ 2-cyclopropyl-6- (4, 5-difluoro-6- { [ (2-methoxyethyl) (methyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AMZ-3)
To a 25mL round bottom flask at room temperature was added MeOH (5 mL) containing the product from step 1 above (AMZ-1) (30 mg,1 eq, 58. Mu. Mol) and 2-methoxy-N-methylethyl-1-amine (AMZ-2) (11 mg,2 eq, 0.12 mmol). DIPEA (23 mg,3 eq, 0.17 mmol) was added to the above mixture at room temperature. The resulting mixture was stirred at 60 ℃ for an additional 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/1). The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 4% B to 34% B in 10 min;wavelength: 254 nm) to give the title compound (AMZ-3) (18.0 mg,54%,99.5% purity) as a white solid. M/z 570.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.14-8.08 (m, 2H), 7.98 (d, j=1.4 hz, 1H), 7.92 (d, j=8.1 hz, 1H), 7.75 (d, j=5.2 hz, 1H), 6.96 (d, j=1.4 hz, 1H), 5.10 (s, 2H), 3.79-3.75 (m, 2H), 3.60-3.55 (m, 2H), 3.51 (s, 3H), 3.35 (s, 3H), 2.71-2.65 (m, 2H), 2.29 (s, 3H), 2.09-2.01 (m, 1H), 1.08-0.97 (m, 4H).
Example 390: synthesis of 4- [2- (6- { 3-azabicyclo [3.1.0] hexane-3-ylmethyl } -1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANA-1)
Intermediate (AIQ-1) (20 mg,1 eq, 42. Mu. Mol) and 3-azabicyclo [3.1.0] at room temperature]To a stirred mixture of hexane (AKQ-1) (3.8 mg,1.1 eq, 46. Mu. Mol) in DCM (2 mL) was added Et 3 N (25 mg,6 eq, 0.25 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH) 4 HCO 3 ) And MeCN (20% MeCN up to 80% in 20 minutes); detector, UV 254/220nm. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 63% B in 9 minutes; wavelength: 254/220nm; retention time: 8.98 To give the title compound (ANA-1) (3.2 mg,6.0 μmol,15%,99.9% purity) as a white solid. M/z 528.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.51 (s, 1H), 8.16-8.07 (m, 2H), 8.03 (d, j=1.5 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.74 (s, 1H), 7.64-7.54 (m, 2H), 6.93 (d, j=1.5 hz, 1H), 5.01 (s, 2H), 3.69 (s, 2H), 3.51 (s, 3H), 2.92(d,J=8.7Hz,2H),2.42(d,J=8.8Hz,2H),2.09–1.98(m,1H),1.43–1.35(m,2H),1.08–0.94(m,4H),0.81–0.73(m,1H),0.42–0.32(m,1H)。
Example 391: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (oxetan-3-ylmethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANB-1)
To a stirred solution of intermediate (AIO-8) (30 mg,1 eq, 63. Mu. Mol) and 1- (oxetan-3-yl) methylamine (AHZ-1) (7 mg,1.2 eq, 76. Mu. Mol) in MeOH (8 mL) was added DIPEA (24 mg,3 eq, 0.19 mmol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 equivalents, 0.32 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 34% B to 44% B in 9 minutes; wavelength: 254/220nm; retention time: 8.48 To give the title compound (ANB-1) (7.9 mg,14 μmol,23%,99.8% purity) as a white solid. M/z 550.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.15-8.07 (m, 2H), 8.03 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.65 (d, j=1.1 hz, 1H), 7.46-7.41 (m, 1H), 6.95 (d, j=1.4 hz, 1H), 5.06 (s, 2H), 4.82-4.78 (m, 2H), 4.40 (t, j=6.1 hz, 2H), 3.87 (s, 2H), 3.49 (s, 3H), 3.20-3.11 (m, 1H), 2.90 (d, j=7.4 hz, 2H), 2.08-2.00 (m, 1H), 1.08-0.95 (m, 4H).
Example 392: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (2R, 3S) -3-hydroxy-2-methylpyrrolidin-1-yl ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANC-2)
To a stirred mixture of intermediate (AKR-1) (30 mg,1 eq, 0.06 mmol) and (2 r,3 s) -2-methylpyrrolidin-3-ol (ANC-1) (9 mg,1.5 eq, 0.09 mmol) in DCM (5 mL) was added DIPEA (23 mg,3 eq, 0.18 mmol) at 60 ℃. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm 5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 7% B to 37% B over 10 min; wavelength: 254nm; retention time: 9.03) to give the title compound (ANC-2) (6.1 mg, 11. Mu. Mol,18%,99.8% purity) as a white solid. M/z 564.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.25–8.18(m,2H),7.92(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.55(s,1H),7.44(d,J=9.8Hz,1H),6.94(d,J=1.4Hz,1H),5.07(s,2H),4.01(d,J=13.7Hz,1H),3.70–3.61(m,1H),3.48(s,3H),3.35(d,J=13.8Hz,1H),2.74–2.67(m,1H),2.31–2.21(m,2H),2.11–2.03(m,1H),1.98–1.87(m,1H),1.51–1.41(m,1H),1.12(d,J=6.2Hz,3H),1.03–0.92(m,4H)。
Example 393: synthesis of 4- [2- (6- { 5-azaspiro [2.4] heptan-5-ylmethyl } -1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AND-1)
Intermediate (AIQ-1) (15 mg,1 eq, 31. Mu. Mol), DIPEA (16 mg,4 eq, 0.12 mmol) and 5-azaspiro [2.4]]A solution of heptane (AAC-1) (4 mg,1.3 eq, 0.04 mmol) in DCM (3 mL) was stirred at 60℃for 3 days. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: XBridge prep. OBD C18 column, 30 x 150mm,5 μm;mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 51% B to 61% B in 8 minutes; wavelength: 254/220nm; retention time: 7.35 To give the title compound (AND-1) (6.7 mg, 12. Mu. Mol,39%,99.0% purity) as a white solid. M/z 542.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.13-8.07 (m, 2H), 8.05 (d, j=1.4 hz, 1H), 7.92 (d, j=8.1 hz, 1H), 7.80 (s, 1H), 7.70-7.66 (m, 1H), 7.61 (d, j=7.8 hz, 1H), 6.92 (d, j=1.5 hz, 1H), 5.02 (s, 2H), 3.75 (s, 2H), 3.51 (s, 3H), 2.79 (t, j=7.0 hz, 2H), 2.53 (s, 2H), 2.07-1.99 (m, 1H), 1.85 (t, j=6.9 hz, 2H), 1.06-0.96 (m, 4H), 0.56 (d, j=2.2 hz, 4H).
Example 394: synthesis of 4- [ 2-cyclopropyl-6- (4, 5-difluoro-6- { [ (oxetan-3-ylmethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANE-1)
To a 25mL round bottom flask was added MeOH (5 mL) containing intermediate (ALY-1) (30 mg,1 eq, 61. Mu. Mol) at room temperature. DIPEA (24 mg,3 eq, 0.18 mmol) and 1- (oxetan-3-yl) methylamine (AHZ-1) (6 mg,1.2 eq, 73. Mu. Mol) were added to the above mixture at room temperature. The resulting mixture was stirred at 60 ℃ for an additional 12 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 equivalents, 0.31 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 48% B in 8 minutes; wavelength: 254 nm) to give the title compound (ANE-1) (10.9 mg, 19. Mu. Mol,31%,97.4% purity) as a white solid. M/z 568.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.14-8.08 (m, 2H), 8.00 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.74 (d, j=5.3 hz, 1H), 6.96 (d, j=1.4 hz, 1H), 5.11 (s, 2H), 4.85-4.79 (m, 2H), 4.44-4.38 (m, 2H), 3.93 (s, 2H), 3.49 (s, 3H), 3.20-3.13 (m, 1H), 2.94 (d, j=7.4 hz, 2H), 2.08-2.02 (m, 1H), 1.07-0.98 (m, 4H).
Example 395: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { hexahydrofuro [3,2-b ] pyrrol-4-ylmethyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANF-11)
Step 1: (2Z) -2-azido-3- (furan-2-yl) prop-2-enoic acid ethyl ester (ANF-3)
To a stirred solution of EtONa (4.25 g,1.5 eq, 62.4 mmol) in EtOH (50 mL) was added dropwise furan-2-carbaldehyde (ANF-1) (4.00 g,1 eq, 41.6 mmol) and ethyl 2-azidoacetate (ANF-2) (16.1 g,3 eq, 125 mmol) under a nitrogen atmosphere at 0deg.C. The resulting mixture was stirred under nitrogen atmosphere at 0 ℃ for 2 hours. The resulting mixture was extracted with EtOAc (3×20 mL). The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (10/1) to give the sub-title compound (ANF-3) (2.8 g,13.5mmol,32%,90% purity) as a yellow oil. M/z 208.1 (M+H) + (ES+)
Step 2: 4H-Furano [3,2-b ] pyrrole-5-carboxylic acid ethyl ester (ANF-4)
A solution of the product from step 1 above (ANF-3) (2.8 g,1 eq., 13.5 mmol) in xylene (40 mL) was stirred at 110℃for 2 hours. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (3/1) to give the sub-title compound (ANF-4) (2) as a brown oil.0g,11.1mmol,83%,92% purity). M/z 180.1 (M+H) + (ES+)
Step 3: 4-5-ethylfuro [3,2-b ] pyrrole-4, 5-dicarboxylic acid tert-butyl ester (ANF-5)
To a stirred solution of the product (ANF-4) from step 2 above (2.00 g,1 eq., 11.2 mmol) and DMAP (68 mg,0.05 eq., 0.56 mmol) in MeCN (30 mL) at room temperature was added Boc 2 O (2.92 g,1.2 eq, 13.4 mmol). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (6/1) to give the sub-title compound (ANF-5) (2.5 g,8.96mmol,80%,91% purity) as a brown oil. M/z 280.1 (M+H) + (ES+)
Step 4: 4-5-Ethylhexahydrofuro [3,2-b ] pyrrole-4, 5-dicarboxylic acid tert-butyl ester (ANF-6)
To a stirred solution of the product from step 3 above (ANF-5) (2.5 g,1 eq, 8.95 mmol) in EtOH (50 mL) was added Pd/C39 (495mg, 10wt%,0.52 eq, 4.66 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred overnight at 60 ℃ under a hydrogen atmosphere. The mixture was cooled to room temperature. Filtering the resulting mixture; the filter cake was washed with EtOH (3X 10 mL). The filtrate was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 286.2 (M+H) + (ES+)
Step 5:4- (tert-Butoxycarbonyl) -hexahydrofuro [3,2-b ] pyrrole-5-carboxylic acid (ANF-7)
To a stirred solution of the product from step 4 above (ANF-6) (2.4 g,1 eq, 8.41 mmol) and LiOH (1.01 g,5 eq, 42.1 mmol) in THF (30 mL) and water (10 mL) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. The mixture was acidified to pH 3 with aqueous HCl (1M). The resulting mixture was diluted with water and extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (2×100 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was used directly in the next step without further purification. m/z 2 58.1(M+H) + (ES+)
Step 6: hexahydro-2H-furo [3,2-b ] pyrrole-5-carboxylic acid (ANF-8)
A solution of the product from step 5 above (ANF-7) (2.00 g,1 eq., 7.77 mmol) in TFA (10 mL) in DCM (30 mL) was stirred at room temperature for 2 hours. The resulting mixture was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 158.1 (M+H) + (ES+)
Step 7: hexahydro-2H-furo [3,2-b ] pyrrole (ANF-10)
A stirred solution of the product from step 6 above (ANF-8) (1.00 g,1 eq., 6.36 mmol) and p-methylacetophenone (ANF-9) (85.4 mg,0.1 eq., 0.64 mmol) in cyclohexanol (30 mL) was stirred at 160℃for 4 hours. The mixture was cooled to room temperature. The resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with aqueous HCl (10%) (2×200 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was used directly in the next step without further purification. M/z 114.1 (M+H) + (ES+)
Step 8:4- [ 2-cyclopropyl-6- (4-fluoro-6- { hexahydrofuro [3,2-b ] pyrrol-4-ylmethyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANF-11)
To a stirred solution of intermediate (AKR-1) (100 mg,1 eq., 0.2 mmol) and product (ANF-10) from step 7 above (45 mg,2 eq., 0.40 mmol) in DCM (10 mL) at room temperature was added DIPEA (104 mg,4 eq., 0.8 mmol). The resulting mixture was stirred at 60℃for 4 days. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 4% B to 34% B over 10 min; wavelength: 254 nm) to give the title compound (ANF-11) (28 mg, 49. Mu. Mol,24%,99.3% purity) as a white solid. M/z 576.0 (M+H) + (ES+)。 1 H NMR (400 MHz, meOH-d 4) delta 8.50(s,1H),8.14–8.07(m,2H),8.03–7.90(m,2H),7.65(s,1H),7.45–7.40(m,1H),6.96(d,J=1.4Hz,1H),5.08(s,2H),4.62–4.52(m,1H),4.04–3.87(m,2H),4.84–3.76(m,1H),3.61–3.49(m,4H),3.28–3.23(m,1H),2.94–2.84(m,1H),2.32–2.23(m,1H),2.15–2.01(m,2H),1.87–1.65(m,3H),1.12–0.98(m,4H)。
Example 396: synthesis of rel-4- (2-cyclopropyl-6- (4-fluoro-6- (((3 ar,6 ar) -hexahydro-4H-furo [3,2-b ] pyrrol-4-yl) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (ANG-1)
The crude product (ANF-11) (28 mg,1 eq., 49. Mu. Mol) was purified by preparative chiral HPLC under the following conditions (column: CHIRALPAK ID, 2X 25cm, 5. Mu.m; mobile phase A: mtBE (0.5% 2M NH) 3 MeOH) -HPLC, mobile phase B: meOH dcm=1:1— HPLC; flow rate: 20 ml/min; gradient: 25% B to 25% B in 33 minutes; wavelength: 220/254 nm) to give the title compound (ANG-1) (11.4 mg, 20. Mu. Mol,41%,99.7% purity) as a white solid. M/z 576.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.16-8.06 (m, 2H), 8.01 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.65 (s, 1H), 7.42 (d, j=9.8 hz, 1H), 6.97 (d, j=1.4 hz, 1H), 5.06 (s, 2H), 4.60-4.53 (m, 1H), 4.04-3.96 (m, 1H), 3.96-3.88 (m, 1H), 3.84-3.76 (m, 1H), 3.54 (d, j=26.8 hz, 4H), 3.28-3.22 (m, 1H), 2.92-2.84 (m, 1H), 2.31-2.23 (m, 1H), 2.15-2.00 (m, 2.89 (m, 1H), 3.96-3.88 (m, 1H), 3.80-3.80 (m, 1H). Column: CHIRALPAK ID-3,4.6 x 50mm 3um; mobile phase a: mtBE (0.1% DEA): (MeOH: dcm=1:1) =75:25; flow rate: 1 ml/min; retention time: 6.869.
example 397: synthesis of rel-4- (2-cyclopropyl-6- (4-fluoro-6- (((3 ar,6 ar) -hexahydro-4H-furo [3,2-b ] pyrrol-4-yl) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (ANH-1)
The crude product (ANF-11) (28 mg,1 eq., 49. Mu. Mol) was purified by preparative chiral HPLC under the following conditions (column: CHIRALPAK ID, 2X 25cm, 5. Mu.m; mobile phase A: mtBE (0.5% 2M NH) 3 MeOH) -HPLC, mobile phase B: meOH dcm=1:1— HPLC; flow rate: 20 ml/min; gradient: 25% B to 25% B in 33 minutes; wavelength: 220/254 nm) to give the title compound (ANH-1) (11.4 mg, 20. Mu. Mol,41%,99.9% purity) as a white solid. M/z 576.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.00 (m, 3H), 7.92 (d, j=8.0 hz, 1H), 7.65 (s, 1H), 7.42 (d, j=9.8 hz, 1H), 6.97 (d, j=1.4 hz, 1H), 5.06 (s, 2H), 4.62-4.53 (m, 1H), 4.03-3.89 (m, 2H), 3.85-3.77 (m, 1H), 3.61-3.47 (m, 4H), 3.28-3.21 (m, 1H), 2.94-2.85 (m, 1H), 2.34-2.23 (m, 1H), 2.16-2.01 (m, 2H), 1.83-1.67 (m, 3H), 1.12-0.95 (m, 4H). Column: CHIRALPAK ID-3,4.6 x 50mm 3um; mobile phase a: mtBE (0.1% DEA): (MeOH: dcm=1:1) =75:25; flow rate: 1 ml/min; retention time: 5.844.
example 398: synthesis of 4- (2- {6- [ (cyclobutylamino) methyl ] -1-oxo-3H-isoindol-2-yl } -6-cyclopropylpyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANI-2)
To a 25mL round bottom flask was added intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and MeOH (3 mL) at room temperature. DIPEA (25 mg,3 eq, 0.20 mmol) and cyclobutylamine (ANI-1) (5.6 mg,1.2 eq, 78. Mu. Mol) were added to the above mixture at room temperature. The resulting mixture was stirred at 60 ℃ for an additional 12 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditionsPurification under (column: XBridge-prepared OBD C18 column, 30X 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 41% B to 51% B in 9 minutes; wavelength: 254 nm) to give the title compound (ANI-2) (5.1 mg, 10. Mu. Mol,15%,99.7% purity) as a white solid. M/z 516.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.14-8.04 (m, 3H), 7.92 (d, j=8.0 hz, 1H), 7.79 (s, 1H), 7.68-7.59 (m, 2H), 6.91 (d, j=1.3 hz, 1H), 5.05-4.99 (m, 2H), 3.78 (d, j=2.6 hz, 2H), 3.49 (s, 3H), 3.27 (s, 1H), 2.23-2.14 (m, 2H), 2.05-1.99 (m, 1H), 1.88-1.79 (m, 2H), 1.75-1.65 (m, 2H), 1.06-0.96 (m, 4H).
Example 399: synthesis of (R) -4- (2-cyclopropyl-6- (6- (((1- (oxetan-3-yl) ethyl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (ANJ-2)
To a stirred solution of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol), (1R) -1- (oxetan-3-yl) ethylamine (ANJ-1) (6.6 mg,1 eq, 65. Mu. Mol) in MeOH (3 mL) at 60℃was added Et 3 N (33 mg,5 eq, 0.325 mmol) for 2 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (25 mg,10 equivalents, 0.65 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was diluted with water and extracted with DCM (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 40% B to 50% in 8 minutesB, a step of preparing a composite material; wavelength: 254/220nm; retention time: 7.82. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ANJ-2) (6.4 mg,12 μmol,18%,99.9% purity) as a white solid. M/z 546.7 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.38–8.11(m,2H),7.99(d,J=1.4Hz,1H),7.95–7.83(m,1H),7.76(s,1H),7.72–7.38(m,2H),6.89(d,J=1.4Hz,1H),5.00(s,2H),4.71–4.43(m,2H),4.42–4.16(m,2H),3.86(d,J=14.0Hz,1H),3.73(d,J=13.9Hz,1H),3.48(s,3H),2.84(s,2H),2.09–1.94(m,1H),1.41–0.54(m,7H)。
Example 400: synthesis of (S) -4- (2-cyclopropyl-6- (6- (((1- (oxetan-3-yl) ethyl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (ANK-2)
DIPEA (25 mg,3 eq, 0.20 mmol) was added to a stirred mixture of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and (1S) -1- (oxetan-3-yl) ethylamine (ANK-1) (6.6 mg,1 eq, 65. Mu. Mol) in MeOH (10 mL) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. NaBH was added to the above mixture at room temperature 4 (7.4 mg,3 equivalents, 0.40 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 6% B to 36% B over 10 min; wavelength: 254nm; retention time: 8.8) to give the title compound (ANK-2) (16.6 mg, 30. Mu. Mol,45%,99.2% purity) as a white solid. M/z 546.2 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.25–8.18(m,2H),8.01–7.92(m,1H),7.91–7.84(m,1H),7.76(s,1H),7.69–7.60(m,2H),6.89(d,J=1.5Hz,1H),5.00(s,2H),4.61–4.50(m,2H),4.38–4.22(m,2H),3.90–3.68(m,2H),3.48(s,3H),2.91–2.76(m,2H),2.10–1.99(m,1H),1.02–0.87(m,7H)。
Example 401: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [ (3-fluorooxetan-3-yl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANL-2)
DIPEA (17 mg,2 eq, 0.13 mmol) was added to a stirred mixture of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and 1- (3-fluorooxetan-3-yl) methylamine (ANL-1) (10 mg,1.5 eq, 98. Mu. Mol) in MeOH (1 mL) at room temperature under nitrogen. The resulting mixture was stirred under nitrogen at 60 ℃ for hours. The mixture was cooled to room temperature. Adding NaBH to the mixture 4 (4.9 mg,2 equivalents, 0.13 mmol). The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative HPLC on a column (XBridge prep OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 48% B in 8 minutes; wavelength: 254/220nm; retention time: 7.28 To give the title compound (ANL-2) (2.1 mg,38 μmol,5.8%,98.1% purity) as a white solid. M/z 550.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.25-7.99 (m, 3H), 7.92 (d, j=8.0 hz, 1H), 7.83 (s, 1H), 7.68 (d, j=7.8 hz, 1H), 7.62 (d, j=7.8 hz, 1H), 6.92 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 4.72 (d, j=7.8 hz, 1H), 4.68 (d, j=7.8 hz, 1H), 4.62 (d, j=7.7 hz, 1H), 4.57 (d, j=7.7 hz, 1H), 3.93 (s, 2H), 3.50 (s, 3H), 3.07 (s, 1H), 3.02 (s, 1H), 2.07-1.97 (m, 1H), 1.08 (m, 1.88-0.4H).
Example 402: synthesis of 4- { 2-cyclopropyl-6- [6- ({ [2- (oxetan-3-yl) prop-2-yl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANM-2)
Et is added to a stirred solution of intermediate (AGT-2) (25 mg,1 eq, 54. Mu. Mol) and 2- (oxetan-3-yl) propan-2-amine (ANM-1) (6 mg,1 eq, 54. Mu. Mol) in MeOH (1.5 mL) at room temperature under nitrogen atmosphere 3 N (27 mg,5 eq, 0.27 mmol). The resulting mixture was stirred under nitrogen at 60 ℃ for hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (21 mg,10 equivalents, 0.54 mmol). The resulting mixture was stirred at room temperature for 2 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30×150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 ) Mobile phase B: meCN; flow rate: 60 ml/min; gradient: 44B to 56B in 8 minutes; a detector, UV 254/220nm; retention time: 5.7. the product-containing fractions were combined and partially evaporated in vacuo and lyophilized overnight to give the title compound (ANM-2) (11.1 mg,20 μmol,36%,96.4% purity) as a white solid. M/z 560.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.07 (m, 2H), 8.05 (d, j=1.4 hz, 1H), 7.93 (d, j=8.2 hz, 1H), 7.82 (s, 1H), 7.67-7.63 (m, 1H), 7.59 (d, j=7.9 hz, 1H), 6.92 (d, j=1.4 hz, 1H), 5.02 (s, 2H), 4.74-4.65 (m, 2H), 4.63 (d, j=6.5 hz, 2H), 3.76 (s, 2H), 3.50 (s, 3H), 3.14-3.10 (m, 1H), 2.07-1.98 (m, 1H), 1.23 (s, 6H), 1.07-0.92 (m, 4H).
Example 403: synthesis of (R) -4- (2-cyclopropyl-6- (6- (1- ((oxetan-3-ylmethyl) amino) ethyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (ANN-2)
Step 1:4- (2-cyclopropyl-6- (6- (1- ((oxetan-3-ylmethyl) amino) ethyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (ANN-1)
To a stirred mixture of intermediate (AJH-3) (50 mg,1 eq, 0.11 mmol) and 1- (oxetan-3-yl) methylamine (AHZ-1) (11 mg,1.2 eq, 0.13 mmol) in THF (5 mL) under nitrogen at room temperature was added Ti (Oi-Pr) 4 (60 mg,2 equivalents, 0.21 mmol). The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. Adding NaBH to the mixture at 0deg.C 4 (13 mg,5 eq, 0.55 mmol). The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative HPLC on a column (XBridge prep OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 33% B to 43% B in 8 minutes; wavelength: 254/220nm; retention time: 7.85 To give the sub-title compound (ANN-1) (25 mg,46 μmol,43%,98% purity) as a white solid. M/z 546.3 (M+H) + (ES+)
Step 2: (R) -4- (2-cyclopropyl-6- (6- (1- ((oxetan-3-ylmethyl) amino) ethyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (ANN-2).
The crude product (ANN-1) (25 mg,1 eq., 46. Mu. Mol) from the product of step 1 above was purified by preparative chiral HPLC under the following conditions: (column: CHIRAL ART Cellulose-SA, 2X 25cm,5 μm; mobile phase A: mtBE (0.5% 2M NH) 3 MeOH) -HPLC, mobile phase B: meOH dcm=1:1— HPLC; flow rate: 20 ml/min; gradient: 35% B to 35% B in 25 minutes; wavelength: 220/254nm; retention time 1 (min): 16.877, sample solvent: etOH-HPLC; injection volume: 0.5 mL) to give the title compound (ANN-2) (8.2 mg, 15. Mu. Mol,33%,99.3% purity) as a white solid. M/z 546.3 (M+H) + (ES+)。 1 H NMR (400 mhz, dmso-d 6) delta 8.54 (s, 1H), 8.25-8.18 (m, 2H), 7.98 (d, j=1.4 hz, 1H), 7.91-7.84 (m, 1H), 7.76 (s, 1H), 7.66 (s, 2H), 6.90 (d, j=1.4 hz, 1H), 5.00 (s, 2H), 4.62-4.54 (m, 2H), 4.20-4.15 (m, 2H), 3.82 (s, 1H), 3.48 (s, 3H), 2.97 (s, 1H), 2.70-2.61 (m, 1H), 2.40-2.27 (m, 1H), 2.10-2.00 (m, 1H), 1.25 (d, j=14.hz, 3H), 0.97 (d, j=6.4 hz, 4H). Column: CHIRALPAK IA-3,4.6 x 50mm,3 μm; mobile phase a: mtBE (0.1% DEA): (MeOH: dcm=1:1) =65:35; flow rate: 1 ml/min; retention time: 3.801
Example 404: synthesis of (S) -4- (2-cyclopropyl-6- (6- (1- ((oxetan-3-ylmethyl) amino) ethyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (ANO-1)
The crude product (ANN-1) (25 mg,1 eq, 46. Mu. Mol) was purified by preparative chiral HPLC under the following conditions: (column: CHIRAL ART Cellulose-SA, 2X 25cm,5 μm; mobile phase A: mtBE (0.5% 2M NH) 3 MeOH) -HPLC, mobile phase B: meOH dcm=1:1— HPLC; flow rate: 20 ml/min; gradient: 35% B to 35% B in 25 minutes; wavelength: 220/254nm; retention time 2 (minutes): 21.967; sample solvent: etOH-HPLC; injection volume: 0.5 ml) to give the title compound (ANO-1) (7.7 mg, 14. Mu. Mol,31%,99.8% purity) as a white solid. M/z 546.3 (M+H) + (ES+)。 1 H NMR (400 mhz, dmso-d 6) delta 8.54 (s, 1H), 8.25-8.18 (m, 2H), 7.98 (d, j=1.4 hz, 1H), 7.87 (d, j=8.6 hz, 1H), 7.77 (s, 1H), 7.67 (s, 2H), 6.90 (d, j=1.4 hz, 1H), 5.01 (s, 2H), 4.62-4.54 (m, 2H), 4.21-4.13 (m, 2H), 3.82 (s, 1H), 3.48 (s, 3H), 2.98 (s, 1H), 2.80-2.58 (m, 2H), 2.11-2.00 (m, 1H), 1.28 (s, 3H), 0.97 (d, j=6.4 hz, 4H). Column: CHIRALPAK IA-3,4.6 x 50mm,3 μm; mobile phase a: mtBE (0.1% DEA): (MeOH: dcm=1:1) =65:35; flow rate: 1 ml/min; retention time: 3.023.
Example 405: synthesis of 4- [ 2-cyclopropyl-6- (1-oxo-6- { [ (3S) -oxolan-3-ylamino ] methyl } -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANP-2)
To a stirred solution of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and (3R) -oxolane-3-amine (ANP-1) (7 mg,1.2 eq, 78. Mu. Mol) in MeOH (8 mL) was added DIPEA (25 mg,3 eq, 0.20 mmol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 40% B in 10 minutes; wavelength: 254/220nm; retention time: 9.5 To give the title compound (ANP-2) (11.3 mg,21 μmol,32%,99.0% purity) as a white solid. M/z 532.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.04 (m, 3H), 7.92 (d, j=8.0 hz, 1H), 7.81 (d, j=1.6 hz, 1H), 7.69-7.65 (m, 1H), 7.61 (d, j=7.8 hz, 1H), 6.92 (d, j=1.4 hz, 1H), 5.02 (s, 2H), 3.97-3.88 (m, 1H), 3.88-3.79 (m, 3H), 3.79-3.72 (m, 1H), 3.62-3.58 (m, 1H), 3.50 (s, 3H), 3.43-3.37 (m, 1H), 2.18-1.99 (m, 2H), 1.86-1.77 (m, 1H), 1.06-0.95 (m, 4H).
Example 406: synthesis of 4- [ 2-cyclopropyl-6- (1-oxo-6- { [ (3S) -oxolan-3-ylamino ] methyl } -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANQ-2)
At room temperature under nitrogen atmosphereTo a stirred mixture of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and (3S) -oxolane-3-amine (ANQ-1) (8.5 mg,1.5 eq, 98. Mu. Mol) in MeOH (2 mL) was added DIPEA (17 mg,2 eq, 0.13 mmol). The resulting mixture was stirred under nitrogen at 60 ℃ for 2 hours. The mixture was cooled to room temperature. NaBH (OAc) was added to the above mixture at room temperature under a nitrogen atmosphere 3 (28 mg,2 equivalents, 0.13 mmol). The resulting mixture was stirred at room temperature overnight. Filtering the resulting mixture; the filter cake was washed with MeOH (2X 3 mL). The filtrate was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 6% B to 36% B over 10 min; wavelength: 254nm; retention time: 8.7) to give the title compound (ANQ-2) (11.4 mg, 21. Mu. Mol,33%,99.6% purity) as a white solid. M/z 532.3 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.55(s,1H),8.22(d,J=1.7Hz,1H),8.22–8.18(m,1H),7.97(d,J=1.4Hz,1H),7.91–7.84(m,1H),7.76(s,1H),7.64(d,J=1.3Hz,2H),6.90(d,J=1.4Hz,1H),5.00(s,2H),3.85–3.73(m,3H),3.72–3.60(m,2H),3.49(s,3H),3.48–3.42(m,1H),3.31–3.21(m,1H),2.12–1.99(m,1H),1.99–1.86(m,1H),1.76–1.64(m,1H),1.00–0.93(m,4H)。
Example 407: synthesis of 5-chloro-2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -3H-isoindol-1-one (ANR-2)
Step 1: 5-chloro-6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -2, 3-dihydro-isoindol-1-one (ANR-1)
To a solution of intermediate (AKN-10) (200 mg,1 eq, 1.02 mmol) and (1S, 2R) -2-aminocyclopentan-1-ol (Z-1) (114 mg,1.1 eq, 1.12 mmol) in DCM (20 mL) at room temperature was added Et 3 N (310 mg,3 eq, 3.07 mmol). Will be spentThe resulting mixture was stirred at room temperature for 1 hour. Then NaBH was added at room temperature 3 CN (129 mg,2 equivalents, 2.04 mmol). The resulting mixture was stirred at room temperature for 1 hour. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1) to give the sub-title compound (ANR-1) (200 mg,0.71mmol,70%,95% purity) as a yellow oil. M/z 281.1/283.1 (M+H) + (ES+)
Step 2: 5-chloro-2- { 6-cyclopropyl-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1R, 2S) -2-hydroxycyclopentyl ] amino } methyl) -3H-isoindol-1-one (ANR-2)
To the product (ANR-1) (40 mg,1 equivalent, 0.14 mmol), intermediate (ADN-1) (47 mg,1 equivalent, 0.14 mmol) and Cs from step 1 above, under a nitrogen atmosphere at room temperature 2 CO 3 To a stirred mixture of (139 mg,3 eq, 0.43 mmol) in dioxane (10 mL) were added EPhos (30 mg,0.4 eq, 57 μmol) and EPhos Pd G4 (26 mg,0.2 eq, 28 mmol). The resulting mixture was stirred under nitrogen at 90 ℃ for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 ) Mobile phase B: ACN; flow rate: 60 ml/min; gradient: 50% B to 72% B in 8 minutes; wavelength: 254/220nm; retention time: 7.12 To give the title compound (ANR-2) (1.0 mg,1.7 μmol,1.2%,98.1% purity) as a white solid. M/z 573.1/575.1 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.50(s,1H),7.96(d,J=10.5Hz,2H),7.82(s,1H),7.75-7.69(m,1H),7.60(t,J=8.9Hz,2H),6.83(s,1H),5.02(s,2H),4.46(s,1H),3.96(d,J=39.3Hz,2H),3.44(s,3H),2.90(s,1H),2.02(d,J=6.0Hz,1H),1.64(d,J=39.2Hz,7H),1.00-0.90(m,4H)。
Example 408: synthesis of 2- { 6-ethoxy-4- [ 4-fluoro-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] pyridin-2-yl } -6- ({ [ (1-hydroxycyclobutyl) methyl ] (methyl) amino } methyl) -3H-isoindol-1-one (ANS-1)
To a 20mL sealed tube under nitrogen at room temperature were added intermediate (AGQ-1) (36 mg,1 equivalent, 0.11 mmol), intermediate (AGH-1) (26 mg,1.5 equivalent, 0.16 mmol) and Cs 2 CO 3 (70 mg,2 eq, 0.22 mmol) of 1, 4-dioxane (5 mL). RuPhos ring palladium complex Gen.3 (18 mg,0.2 eq, 22. Mu. Mol) and RuPhos (20 mg,0.4 eq, 43. Mu. Mol) were added to the above mixture under nitrogen atmosphere at room temperature. The resulting mixture was stirred under nitrogen at 100 ℃ for an additional 3 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% FA) and MeCN (10% MeCN up to 50% in 10 minutes); detector, UV 254/220nm. The crude product was purified by preparative HPLC on a column (Xselect CSH C18OBD column 30 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 17% B to 47% B over 10 min; wavelength: 254nm; retention time: 11.03) to give the title compound (ANS-1) (1.5 mg,3.3 μmol,3.0%,99.0% purity). M/z 460.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.47 (s, 1H), 7.83 (t, j=1.6 hz, 2H), 7.78-7.73 (m, 1H), 7.71-7.59 (m, 2H), 7.55-7.49 (m, 1H), 7.47-7.43 (m, 1H), 6.37 (d, j=1.2 hz, 1H), 5.07 (s, 2H), 4.72 (s, 2H), 4.37 (t, j=7.1 hz, 2H), 3.51 (s, 3H), 1.38 (t, j=7.1 hz, 3H).
Example 409: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (2-methoxyethyl) (methyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANT-1)
To a stirred mixture of compound (AIO-9) (38 mg,1 eq, 71. Mu. Mol) and HCHO (6.37 mg,3 eq, 0.21 mmol) in MeOH (10 mL) at room temperature was added NaBH 3 CN(9mg,2 equivalents, 0.14 mmol). The resulting mixture was stirred at room temperature for 2 hours. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC under the following conditions (column: XBridge prep. OBD C18 column, 19 x 250mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 25 ml/min; gradient: 50% B to 68% B in 8 minutes; wavelength: 254/220nm; retention time: 7.55 To give the title compound (ANT-1) (7.4 mg,13 μmol,19%,99.3% purity) as a white solid. M/z 552.1 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.27–8.17(m,2H),7.96–7.85(m,2H),7.58(s,1H),7.47(d,J=10.1Hz,1H),6.93(d,J=1.4Hz,1H),5.08(s,2H),3.63(s,2H),3.47(d,J=4.7Hz,5H),3.24(s,3H),2.55(d,J=5.9Hz,2H),2.19(s,3H),2.06(d,J=7.9,4.8Hz,1H),1.02-0.93(m,4H)。
Example 410: synthesis of 4- (2-cyclopropyl-6- (4-fluoro-6- (((1S, 4S) -1-methyl-2-oxa-5-azabicyclo [2.2.1] heptan-5-yl) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (ANU-3)
Step 1:4- (2-cyclopropyl-6- (4-fluoro-6- ((1-methyl-2-oxa-5-azabicyclo [2.2.1] heptan-5-yl) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (ANU-2)
To intermediate (AKR-1) (60 mg,1 eq, 0.12 mmol) and 1-methyl-2-oxa-5-azabicyclo [2.2.1] at room temperature]To a stirred mixture of heptane (ANU-1) (14 mg,1 eq, 0.12 mmol) in DCM (10 mL) was added DIPEA (47 mg,3 eq, 0.36 mmol). The resulting mixture was stirred at 60 ℃ overnight. The resulting mixture was concentrated in vacuo. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30X 150mm,5 μm, n; stream)Mobile phase a: water (0.1% fa), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 5% B to 35% B in 10 minutes; wavelength: 254nm; retention time: 9.6 To give the title compound (ANU-2) (40 mg, 69. Mu. Mol,58%,99% purity) as a white solid. M/z 576.1 (M+H) + (ES+)
Step 2:4- (2-cyclopropyl-6- (4-fluoro-6- (((1S, 4S) -1-methyl-2-oxa-5-azabicyclo [2.2.1] heptan-5-yl) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (ANU-3)
The crude product (ANU-2) (40 mg, 69. Mu. Mol,1 eq.) from the product of step 1 above was purified by preparative chiral HPLC under the following conditions (column: CHIRALPAK ID, 2X 25cm,5 μm; mobile phase A: hex: DCM=1:1 (0.5% 2M NH) 3 MeOH) -HPLC, mobile phase B: etOH-HPLC; flow rate: 20 ml/min; gradient: 50% B to 50% B within 19 minutes; wavelength: 220/254nm; retention time 1 (min): 10.824 To give the title compound (ANU-3) (10.2 mg, 18. Mu. Mol,15%,99.0% purity) as a white solid. M/z 576.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.16-8.06 (m, 2H), 8.01 (t, j=1.3 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.67 (s, 1H), 7.45 (d, j=9.8 hz, 1H), 6.95 (t, j=1.3 hz, 1H), 5.05 (s, 2H), 4.15 (d, j=7.8 hz, 1H), 3.90 (d, j=2.6 hz, 2H), 3.78-3.71 (m, 1H), 3.50 (d, j=1.1 hz, 4H), 2.82 (d, j=9.9 hz, 1H), 2.56-2.48 (m, 1H), 2.10-2.00 (m, 1H), 2.00-1.92 (m, 1H), 1.9.64 (d, 9hz, 1H), 3.64 (d, 1.6 hz, 2H). Column: CHIRALPAK ID-3,4.6 x 50mm 3um; mobile phase a: (Hex: dcm=1:1) (0.1% DEA): etoh=50:50; flow rate: 1 ml/min; retention time: 2.676.
Example 411: synthesis of 4- (2-cyclopropyl-6- (4-fluoro-6- (((1R, 4R) -1-methyl-2-oxa-5-azabicyclo [2.2.1] heptan-5-yl) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (ANV-1)
Will be thickThe product (ANU-2) (40 mg, 69. Mu. Mol,1 eq.) was purified by preparative chiral HPLC (column: CHIRALPAK ID, 2X 25cm, 5. Mu.m; mobile phase A: hex: DCM=1:1 (0.5% 2M NH3-MeOH) -HPLC; mobile phase B: etOH- -HPLC; flow rate: 20 ml/min; gradient: 50% B to 50% B in 19 min; wavelength: 220/254nm; retention time: 2 min): 17.038) to give the title compound (ANX-1) (4.3 mg, 7.4. Mu. Mol,6.2%,99.4% purity) as a white solid. M/z 576.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.18-8.05 (m, 2H), 8.02 (d, j=1.3 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.68 (s, 1H), 7.45 (d, j=9.8 hz, 1H), 6.96 (d, j=1.4 hz, 1H), 5.06 (s, 2H), 4.15 (d, j=7.8 hz, 1H), 3.90 (d, j=3.0 hz, 2H), 3.78-3.70 (m, 1H), 3.51 (s, 4H), 2.83 (d, j=9.9 hz, 1H), 2.08-1.95 (m, 2H), 1.64 (d, j=9.9 hz, 1.39H), 3.0H, 3.09 (m, 1H). Column: CHIRALPAK ID-3,4.6 x 50mm,3 μm; mobile phase a: (Hex: dcm=1:1) (0.1% dea): etoh=50:50; flow rate: 1 ml/min; retention time: 4.676.
Example 412: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-1-oxo-6- { [ (1, 1-trifluoro-3-methoxyprop-2-yl) amino ] methyl } -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANW-3)
Step 1:4- [ 2-cyclopropyl-6- (4-fluoro-1-oxo-6- { [ (1, 1-trifluoro-3-methoxyprop-2-yl) amino ] methyl } -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANW-2)
A solution of intermediate (AIO-8) (50 mg,1 eq, 0.10 mmol) and 1, 1-trifluoro-3-methoxypropan-2-amine (ANW-1) (22.43 mg,1.5 eq, 0.16 mmol) in DMF (2 mL) was stirred at 60℃for 2 hours. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), flowPhase B: meCN; flow rate: 60 ml/min; gradient: 52% B to 62% B in 9 minutes; wavelength: 254/220nm; retention time: 8.83 To give the sub-title compound (ANW-2) (16 mg,26 μmol,24%,98% purity) as a white solid. M/z 604.0 (M+H) + (ES+)
Step 2:4- [ 2-cyclopropyl-6- (4-fluoro-1-oxo-6- { [ (1, 1-trifluoro-3-methoxyprop-2-yl) amino ] methyl } -3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANW-3)
The product (ANW-2) (15 mg,1 equivalent, 31. Mu. Mol) from step 1 above was reacted with NaBH 4 A solution of (142 mg,120 eq, 3.72 mmol) in MeOH (8 mL) was stirred at room temperature for 3 days. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 52% B to 62% B in 9 minutes; 62% B; wavelength: 254/220nm; retention time: 8.3 To give the title compound (ANW-3) (2.0 mg,3.3 μmol,11%,99.7% purity) as a white solid. M/z 606.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.16-8.07 (m, 2H), 8.02 (d, j=1.5 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.68 (s, 1H), 7.47 (d, j=9.9 hz, 1H), 6.96 (d, j=1.5 hz, 1H), 5.06 (s, 2H), 4.04 (s, 2H), 3.66-3.53 (m, 1H), 3.44 (d, j=57.4 hz, 5H), 3.31 (s, 3H), 2.09-1.97 (m, 1H), 1.08-0.83 (m, 4H).
Example 413: synthesis of 4- [ 2-cyclopropyl-6- (6- { [ methyl (oxetan-3-ylmethyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANX-2)
Stirring of intermediate (AIQ-1) (30 mg,1 eq, 62. Mu. Mol) and methyl (oxetan-3-ylmethyl) amine (ANX-1) (11 mg,1.8 eq, 0.11 mmol) in DCM (10 mL) at RTDIPEA (40 mg,5 eq, 0.31 mmol) was added to the stirred solution. The resulting mixture was stirred at 60℃for 7 days. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 2% B to 32% B in 10 minutes; wavelength: 254nm; retention time: 9.88 To give the title compound (ANX-2) (13.2 mg,24 μmol,39%,99.7% purity) as a white solid. M/z 546.0 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.54(s,1H),8.21(t,2H),7.98(d,J=1.4Hz,1H),7.87(d,J=8.5Hz,1H),7.69–7.63(m,2H),7.60(d,J=7.8,1.5Hz,1H),6.88(d,J=1.4Hz,1H),5.01(s,2H),4.63(d,J=7.8,5.8Hz,2H),4.21(t,J=6.0Hz,2H),3.55–3.47(m,5H),3.22–3.18(m,1H),2.66(d,J=7.5Hz,2H),2.08–2.03(m,4H),0.97(d,J=6.4Hz,4H)。
Example 414: synthesis of 4- { 2-cyclopropyl-6- [ 1-oxo-6- ({ [ (3S) -oxolan-3-ylmethyl ] amino } methyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANY-2)
Intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol), DIPEA (34 mg,4 eq, 0.26 mmol) and 1- [ (3S) -oxolan-3-yl]A solution of methylamine (ANY-1) (9 mg,1.3 eq, 85. Mu. Mol) in MeOH (8 mL) was stirred overnight at 60 ℃. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate:60 ml/min; gradient: 35% B to 45% B in 9 minutes; wavelength: 254/220nm; retention time: 8.3 To give the title compound (ANY-2) (12 mg,22 μmol,34%,99.6% purity) as a white solid. M/z 546.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.16-8.02 (m, 3H), 7.92 (d, J=8.1 Hz, 1H), 7.81 (s, 1H), 7.70-7.55 (m, 2H), 6.91 (d, J=1.4 Hz, 1H), 5.02 (s, 2H), 3.93-3.77 (m, 4H), 3.75-3.66 (m, 1H), 3.52-3.41 (m, 4H), 2.64-2.50 (m, 2H), 2.48-2.38 (m, 1H), 2.12-1.98 (m, 2H), 1.64-1.54 (m, 1H), 1.06-0.90 (m, 4H).
Example 415: synthesis of 4- { 2-cyclopropyl-6- [ 1-oxo-6- ({ [ (3R) -oxolan-3-ylmethyl ] amino } methyl) -3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (ANZ-2)
To intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and 1- [ (3R) -oxolan-3-yl at room temperature ]To a stirred solution of methylamine (ANZ-1) (8 mg,1.2 eq, 78. Mu. Mol) in MeOH (8 mL) was added DIPEA (25 mg,3 eq, 0.20 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 35% B to 45% B in 9 minutes; wavelength: 254/220nm; retention time: 8.33 To give the title compound (ANZ-2) (11.7 mg,21 μmol,33%,99.8% purity) as a white solid. M/z 546.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) δ8.50 (s, 1H), 8.15-8.03 (m, 3H), 7.92 (d, J=8.0 Hz, 1H), 7.80 (t, J=1.1 Hz, 1H), 7.69-7.59(m,2H),6.92(d,J=1.5Hz,1H),5.02(s,2H),3.91–3.77(m,4H),3.75–3.68(m,1H),3.52–3.43(m,4H),2.65–2.53(m,2H),2.47–2.39(m,1H),2.12–1.99(m,2H),1.64–1.54(m,1H),1.07–0.94(m,4H)。
Example 416: synthesis of 4- [2- (6- { [ cyclobutyl (methyl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOA-2)
Et is added to a stirred mixture of intermediate (AIQ-1) (39 mg,1 eq, 81. Mu. Mol) and N-methylcyclobutylamine (AOA-1) (14 mg,2 eq, 0.16 mmol) in EtOH (4 mL) under a nitrogen atmosphere at room temperature 3 N (24 mg,3 eq, 0.24 mmol). The resulting mixture was stirred under nitrogen at 60 ℃ for 6 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 10% B to 80% B in 10 minutes; wavelength: 254nm; retention time: 7.5 To give the title compound (AOA-2) (14.2 mg,27 μmol,33%,99.7% purity) as a white solid. M/z 530.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.16-8.03 (m, 3H), 7.92 (d, j=8.0 hz, 1H), 7.77 (s, 1H), 7.67-7.55 (m, 2H), 6.93 (d, j=1.5 hz, 1H), 5.03 (s, 2H), 3.52 (d, j=8.7 hz, 5H), 2.99-2.89 (m, 1H), 2.15-2.06 (m, 2H), 2.04 (s, 4H), 2.00-1.90 (m, 2H), 1.78-1.66 (m, 2H), 1.28 (s, 1H), 1.07-0.94 (m, 4H).
Example 417: synthesis of (S) -4- (2-cyclopropyl-6- (6- (1- (oxetan-3-ylamino) ethyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AOB-2)
Step 1:4- (2-cyclopropyl-6- (6- (1- (oxetan-3-ylamino) ethyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AOB-1)
To a 25mL round bottom flask was added THF (5 mL) containing intermediate (AJH-3) (100 mg,1 eq, 0.21 mmol) at room temperature. To the above stirred solution were added oxetan-3-amine (AMV-1) (23 mg,1.5 eq, 0.32 mmol) and Ti (Oi-Pr) at room temperature 4 (299 mg,5 eq, 1.06 mmol). The resulting mixture was stirred under nitrogen at 60 ℃ for another 16 hours. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (40 mg,5 equivalents, 1.06 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 55% B in 8 minutes; wavelength: 254/220nm; retention time: 7.8 To give the sub-title compound (AOB-1) (24 mg,45 μmol,22%,98% purity) as a white solid. M/z 532.2 (M+H) + (ES+)
Step 2: (S) -4- (2-cyclopropyl-6- (6- (1- (oxetan-3-ylamino) ethyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AOB-2)
The crude product (AOB-1) (24 mg,0.045mmol,1 eq.) from step 1 above was isolated by chiral preparative HPLC under the following conditions: column: CHIRALPAK ID,2×25cm,5 μm; mobile phase a: hex: dcm=1:1 (0.5% 2m NH3-MeOH) -HPLC, mobile phase B: etOH-HPLC; flow rate: 20 ml/min; gradient: 20% B to 20% B in 26 minutes; wavelength: 220/254nm; retention time 1 (min): 15.85. this gave the title compound (AOB-2) (5.8 mg, 11. Mu. Mol, 10) as a white solid% 99.8% purity). M/z 532.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.07 (m, 2H), 8.06 (d, j=1.4 hz, 1H), 7.92 (d, j=8.1 hz, 1H), 7.79 (s, 1H), 7.67-7.58 (m, 2H), 6.92 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 4.71 (t, j=6.7 hz, 1H), 4.53 (t, j=6.4 hz, 1H), 4.39 (t, j=6.7 hz, 1H), 4.23 (t, j=6.4 hz, 1H), 3.92-3.82 (m, 2H), 3.50 (s, 3H), 2.07-1.98 (m, 1H), 1.41 (d, j=6.6 hz, 1.07-1.95 (m, 1H). Column: CHIRALPAK ID-3,4.6 x 50mm 3 μm; mobile phase a: hex: dcm=1:1) (0.1% dea): etoh=80:20; flow rate: 1 ml/min; retention time: 2.505.
Example 418: synthesis of (R) -4- (2-cyclopropyl-6- (6- (1- (oxetan-3-ylamino) ethyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AOC-1)
The crude product (AOB-1) (24 mg,0.045mmol,1 eq.) was isolated by chiral preparative HPLC under the following conditions: column: CHIRALPAK ID,2×25cm,5 μm; mobile phase a: hex: dcm=1:1 (0.5% 2m NH3-MeOH) -HPLC, mobile phase B: etOH-HPLC; flow rate: 20 ml/min; gradient: 20% B to 20% B in 26 minutes; wavelength: 220/254nm; retention time 2 (minutes): 22.09. this gave the title compound (AOC-1) (7.2 mg, 14. Mu. Mol,14%,99.7% purity) as a white solid. M/z 532.2 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.01 (m, 3H), 7.92 (d, j=8.0 hz, 1H), 7.79 (d, j=1.6 hz, 1H), 7.68-7.58 (m, 2H), 6.92 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 4.71 (t, j=6.7 hz, 1H), 4.53 (t, j=6.4 hz, 1H), 4.39 (t, j=6.8 hz, 1H), 4.23 (t, j=6.4 hz, 1H), 3.93-3.18 (m, 2H), 3.50 (s, 3H), 2.07-1.98 (m, 1H), 1.41 (d, j=6.7 hz, 3H), 1.09-0.94 (m, 4H). Column: CHIRALPAK ID-3,4.6 x 50mm,3um; mobile phase a: hex: dcm=1:1) (0.1% DEA): etoh=80:20; flow rate: 1 ml/min; retention time: 3.255.
Example 419: synthesis of 4- (2-cyclopropyl-6- {6- [ (1S) -1- { 2-oxa-6-azaspiro [3.3] heptan-6-yl } ethyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOD-2)
Step 1:4- (2-cyclopropyl-6- {6- [1- { 2-oxa-6-azaspiro [3.3] heptan-6-yl } ethyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOD-1).
To intermediate (AJH-3) (20 mg,1 eq, 42. Mu. Mol), 2-oxa-6-azaspiro [3.3] under nitrogen at room temperature]Heptane (AMS-1) (5 mg,1.2 eq, 50. Mu. Mol) and Ti (Oi-Pr) 4 (24 mg,2 eq, 84. Mu. Mol) in THF (1 mL) in a stirred mixture 4 (3 mg,2 equivalents, 84. Mu. Mol). The resulting mixture was stirred at room temperature under nitrogen overnight. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (10/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 48% B in 8 minutes; wavelength: 254/220nm; retention time: 7.44 To give the sub-title compound (AOD-1) (15 mg,27 μmol,63%,98% purity) as a white solid. M/z 558.2 (M+H) + (ES+)
Step 2:4- (2-cyclopropyl-6- {6- [ (1S) -1- { 2-oxa-6-azaspiro [3.3] heptan-6-yl } ethyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOD-2).
The crude product (AOD-1) (15 mg,1 eq., 27. Mu. Mol) from the product of step 1 above was purified by preparative chiral HPLC under the following conditions: (column CHIRALPAK IF, 2X 25cm,5 μm; mobile phase A: mtBE (0.5% 2M NH3-MeOH) - - -HPLC; mobile phase B: meOH: DCM=1:1- -HPLC; flow rate: 20 ml/min; gradient: 50% B to 50% B in 15 min; wavelength: 2)20/254nm; retention time 1 (min): 9.004 To give the title compound (AOD-2) (3.2 mg,5.7 μmol,21%,98.3% purity) as a white solid. M/z 558.2 (M+H) + (ES+)。 1 H NMR (300 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.17-8.02 (m, 3H), 7.92 (d, j=8.0 hz, 1H), 7.76 (s, 1H), 7.61 (d, j=1.4 hz, 2H), 6.92 (d, j=1.5 hz, 1H), 5.02 (s, 2H), 4.72 (s, 4H), 3.50 (s, 3H), 3.48-3.38 (m, 3H), 3.28 (s, 2H), 2.06-1.97 (m, 1H), 1.25 (d, j=6.4 hz, 3H), 1.06-0.95 (m, 4H). Column: CHIRALPAK IF-3,4.6 x 50mm,3 μm; mobile phase a: mtBE (0.1% DEA) (MeOH: dcm=1:1) =50:50; flow rate: 1 ml/min; retention time: 1.654.
Example 420: synthesis of 4- (2-cyclopropyl-6- {6- [ (1R) -1- { 2-oxa-6-azaspiro [3.3] heptan-6-yl } ethyl ] -1-oxo-3H-isoindol-2-yl } pyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOE-1)
The crude product (AOD-1) (15 mg,1 eq, 27. Mu. Mol) was purified by preparative chiral HPLC under the following conditions: (column CHIRALPAK IF, 2X 25cm,5 μm; mobile phase A: mtBE (0.5% 2M NH3-MeOH) - - -HPLC; mobile phase B: meOH: DCM=1:1- -HPLC; flow rate: 20 ml/min; gradient: 50% B to 50% B in 15 min; wavelength: 220/254nm; retention time 2 (min): 13.774) to give the title compound (AOE-1) (3.3 mg, 5.9. Mu. Mol,22%,98.8% purity) as a white solid. M/z 558.2 (M+H) + (ES+)。 1 H NMR (300 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.17-8.01 (m, 3H), 7.92 (d, j=8.0 hz, 1H), 7.76 (s, 1H), 7.61 (s, 2H), 6.92 (d, j=1.8 hz, 1H), 5.01 (d, j=2.1 hz, 2H), 4.72 (s, 4H), 3.50 (s, 3H), 3.51-3.38 (m, 3H), 3.28 (d, j=7.0 hz, 2H), 2.15-1.95 (m, 1H), 1.25 (d, j=6.4 hz, 3H), 1.08-0.94 (m, 4H). Column: CHIRALPAK IF-3,4.6 x 50mm,3 μm; mobile phase a: mtBE (0.1% DEA) (MeOH: dcm=1:1) =50:50; flow rate: 1 ml/min; retention time: 3.796.
Example 421: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (4-hydroxy-3-methyl-oxolan-3-yl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOF-9)
Step 1: oxapentalene-3, 4-diol (AOF-2)
At 0 ℃, 3, 6-dioxabicyclo [3.1.0]To a stirred solution of hexane (AOF-1) (20.0 g,1 equivalent, 232 mmol) in water (40 mL) was added concentrated H 2 SO 4 (6 mL). The resulting mixture was stirred at room temperature overnight. The mixture was cooled to room temperature. The mixture was treated with Na 2 CO 3 Is alkalized to pH 8. Filtering the resulting mixture; the filter cake was washed with THF (3×20 mL). The resulting mixture was concentrated in vacuo to give the sub-title compound (AOF-1) (31 g,298mmol, crude product) as a colorless crude oil. M/z 105.0 (M+H) + (ES+)
Step 2:4- [ (tert-Butyldiphenylsilyl) oxy ] oxapentan-3-ol (AOF-3)
To a stirred solution of the product from step 1 above (AOF-2) (30.0 g,1 eq, 288 mmol) and imidazole (19.6 g,1 eq, 288 mmol) in MeCN (50 mL) under nitrogen at room temperature was added TBDPS-Cl (63.4 g,0.8 eq, 231 mmol). The resulting mixture was stirred under nitrogen at 80 ℃ for 4 hours. The mixture was cooled to room temperature and concentrated in vacuo. The residue was applied to silica gel column chromatography with petroleum ether/EtOAc (7/1) to give the sub-title compound (AOF-3) (28 g,81.9mmol,28%,85% purity) as a white solid. M/z 343.2 (M+H) + (ES+)
Step 3:4- [ (tert-Butyldiphenylsilyl) oxy ] oxapent-3-one (AOF-4)
To a stirred solution of the product from step 2 above (AOF-3) (600 mg,1 eq., 1.75 mmol) in DCM (8 mL) was added DMP (2.23 g,3 eq., 5.26 mmol) at room temperature. The resulting mixture was stirred at room temperature overnight. Filtering the resulting mixture; the filter cake was washed with petroleum ether (3×5 mL). The filtrate was concentrated in vacuo. Will be spentThe resulting mixture was diluted with water and extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×80 mL), dried (Na 2 SO 4 ). After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/20) to give the sub-title compound (AOF-4) (480 mg,1.41mmol,80%,92% purity) as a yellow oil. M/z 341.1 (M+H) + (ES+)
Step 4: n- [ (3E) -4- [ (tert-butyldiphenylsilyl) oxy ] oxa-pent-3-ylene ] -2-methylpropane-2-sulfinamide (AOF-5)
To a stirred solution of the product from step 3 above (AOF-4) (1.00 g,1 eq, 2.94 mmol) and t-butylsulfonamide (710 mg,2 eq, 5.87 mmol) in DCM (20 mL) at room temperature under nitrogen was added Ti (i-PrO) 4 (2.50 g,3 equivalents, 8.81 mmol). The resulting mixture was stirred overnight at 60 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The residue was purified by preparative TLC using EtOAc/petroleum ether (1/4) to give the sub-title compound (AOF-5) (1.00 g,77%,90% purity) as a white solid. M/z 444.2 (M+H) + (ES+)
Step 5: n- {4- [ (tert-Butyldiphenylsilyl) oxy ] -3-methyloxypent-3-yl } -2-methylpropan-2-sulfinamide (AOF-6)
To a stirred solution of the product from step 4 above (AOF-5) (1.40 g,1 eq., 3.16 mmol) in THF (50 mL) under nitrogen at-78deg.C was added MeMgBr (753 mg,2 eq., 6.31 mmol) dropwise. The resulting mixture was stirred under nitrogen at-78 ℃ for an additional 1 hour. The reaction was then quenched by addition of 10mL of ice water at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by reverse phase flash column chromatography under the following conditions: column, C18; mobile phase, water (0.1% NH 4 HCO 3 ) And MeCN (10% ACN up to 80% in 30 minutes); detector, UV 254/220nm. This gave the sub-title compound (AOF-6) (195 mg,0.42mmol,13%,95% purity) as a white solid. M/z 460.2 (M+H) + (ES+)。
Step 6:4- [ (tert-Butyldiphenylsilyl) oxy ] -3-methyloxypentan-3-amine (AOF-7)
To a stirred mixture of the product from step 5 above (AOF-6) (195 mg,1 eq, 0.42 mmol) in MeOH (3 mL) was added 1, 4-dioxane (1 mL,4 m) containing HCl (g) in portions under a nitrogen atmosphere at 0 ℃. The resulting mixture was stirred at room temperature under nitrogen for an additional 30 minutes. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (8/1) to give the sub-title compound (AOF-7) (130 mg,0.37mmol,86%,92% purity) as a white solid. M/z 356.2 (M+H) + (ES+)
Step 7:4- (2- {6- [ ({ 4- [ (tert-butyldiphenylsilyl) oxy ] -3-methyloxypent-3-yl } amino) methyl ] -4-fluoro-1-oxo-3H-isoindol-2-yl } -6-cyclopropylpyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOF-8)
To a stirred solution of the product (AOF-7) from step 6 above (130 mg,1 eq, 0.37 mmol) and intermediate (AIO-8) (210 mg,1.2 eq, 0.44 mmol) in DCM (10 mL) under nitrogen at room temperature was added Ti (i-PrO) 4 (312 mg,3 equivalents, 1.10 mmol). The resulting mixture was stirred overnight at 60 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature. NaBH was added to the above mixture at room temperature 3 CN (69 mg,1.10mmol,3 eq.). The resulting mixture was stirred at room temperature for an additional 2 hours. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (12/1) to give the sub-title compound (AOF-8) (237 mg,0.29mmol,79%,95% purity) as a white solid. M/z 818.4 (M+H) + (ES+)
Step 8:4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (4-hydroxy-3-methyl-oxolan-3-yl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOF-9)
Et is added dropwise to a stirred mixture of the product from step 7 above (AOF-8) (170 mg,1 eq, 0.21 mmol) in THF (5 mL) at room temperature under nitrogen atmosphere 3 N.3HF (0.5 mL). The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo. Will beThe residue was purified by preparative TLC using DCM/MeOH (12/1). The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 46% B in 10 minutes; wavelength: 254nm; retention time: 9.07 To give the title compound (AOF-9) (20 mg,34 μmol,17%,99.6% purity) as a white solid. M/z 580.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.53(s,1H),8.22–8.20(m,2H),7.94(d,J=1.4Hz,1H),7.88-7.86(d,J=8.5Hz,1H),7.65(s,1H),7.53-7.51(d,J=10.0Hz,1H),6.93(d,J=1.4Hz,1H),5.20-5.19(d,J=4.4Hz,1H),5.07(s,2H),4.01–3.97(m,1H),3.84–3.80(m,3H),3.63–3.60(m,1H),3.48(d,J=3.8Hz,4H),3.42-3.40(d,J=7.8Hz,1H),2.07–2.04(m,1H),1.13(s,3H),0.98(m,4H)。
Example 422: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (4-hydroxy-3-methyl-oxolan-3-yl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOG-1)
The crude product (AOF-9) (20 mg,1 eq, 34. Mu. Mol) was purified by preparative chiral HPLC under the following conditions (column: CHIRALPAK IE,2X25cm, 5. Mu.m; mobile phase A: mtBE (0.5% 2M NH) 3 MeOH) -HPLC, mobile phase B: etOH-HPLC; flow rate: 20 ml/min; gradient: 50% B to 50% B in 29 minutes; wavelength: 220/254nm; retention time 1 (min): 18.63 To give the title compound (AOG-1) (8.4 mg,15 μmol,42%,99.5% purity) as a white solid. M/z 580.0 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO–d6)δ8.53(s,1H),8.21–8.20(m,2H),7.94(m,1H),7.88–7.86(d,J=8.5Hz,1H),7.65(s,1H),7.53–7.51(d,J=10.0Hz,1H),6.93(m,1H),5.20(s,1H),5.06(s,2H),4.01–3.97(m,1H),3.87–3.76(m,3H),3.63–3.60(m,1H),3.48(d,J=3.5Hz,4H),3.42–3.41(d,J=7.8Hz, 1H), 2.09-2.02 (m, 1H), 1.14 (s, 3H), 1.02-0.95 (m, 4H). Column: CHIRALPAK IE-3,4.6 x 50mm,3 μm; mobile phase a: mtBE (0.1% DEA): etoh=50:50; flow rate: 1 ml/min; retention time: 3.209.
example 423: synthesis of rel-4- { 2-cyclopropyl-6- [ 4-fluoro-6- ({ [ (3R, 4R) -4-hydroxy-3-methyl-oxolan-3-yl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOH-1)
The crude product (AOF-9) (20 mg,1 eq, 34. Mu. Mol) was purified by preparative chiral HPLC under the following conditions (column: CHIRALPAK IE,2X25cm, 5. Mu.m; mobile phase A: mtBE (0.5% 2M NH) 3 MeOH) -HPLC, mobile phase B: etOH-HPLC; flow rate: 20 ml/min; gradient: 50% B to 50% B in 29 minutes; wavelength: 220/254nm; retention time 2 (minutes): 26.30 To give the title compound (AOH-1) (8.6 mg,15 μmol,43%,99.8% purity) as a white solid. M/z 580.0 (M+H) + (ES+)。 1 H NMR (400 MHz, DMSO-d 6) delta 8.53 (s, 1H), 8.21-8.20 (m, 2H), 7.94 (s, 1H), 7.88-7.86 (m, 1H), 7.65 (s, 1H), 7.53-7.51 (d, J=10.0 Hz, 1H), 6.93 (s, 1H), 5.20 (s, 1H), 5.07 (s, 2H), 4.01-3.97 (m, 1H), 3.87-3.76 (m, 3H), 3.63-3.60 (m, 1H), 3.48 (d, J=3.1 Hz, 4H), 3.42-3.41 (d, J=7.8 Hz, 1H), 2.09-2.02 (m, 1H), 1.23 (s, 3H), 1.13 (m, 4H). Column: CHIRALPAK IE-3,4.6 x 50mm,3 μm; mobile phase a: mtBE (0.1% DEA): etoh=50:50; flow rate: 1 ml/min; retention time: 4.632.
example 424: synthesis of 4- (2- {6- [2- (cyclobutylamino) propan-2-yl ] -1-oxo-3H-isoindol-2-yl } -6-cyclopropylpyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOI-1)
To compound (ALK-9) (30 mg,1 equivalent, 61. Mu. Mo) at room temperaturel) and cyclobutanone (AGM-1) (13 mg,3 eq, 0.18 mmol) in DCM (5 mL) to which Ti (Oi-Pr) is added 4 (26 mg,1.5 eq, 91. Mu. Mol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. NaBH (OAc) was added to the above mixture at room temperature 3 (65 mg,5 equivalents, 0.31 mmol). The resulting mixture was stirred at room temperature for another 5 hours. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 51% B to 61% B in 8 minutes; wavelength: 254/220 nm) to give the title compound (AOI-1) (4.1 mg, 7.5. Mu. Mol,12%,97.2% purity) as a white solid. M/z 544.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.51 (s, 1H), 8.15-8.09 (m, 2H), 8.05 (d, j=1.4 hz, 1H), 7.96-7.90 (m, 2H), 7.86-7.81 (m, 1H), 7.61 (d, j=8.1 hz, 1H), 6.94 (d, j=1.4 hz, 1H), 5.02 (s, 2H), 3.51 (s, 3H), 3.09-2.99 (m, 1H), 2.07-2.01 (m, 1H), 1.93-1.84 (m, 2H), 1.79-1.65 (m, 2H), 1.61-1.53 (m, 1H), 1.50 (s, 6H), 1.49-1.38 (m, 1H), 1.06-0.96 (m, 4H).
Example 425: synthesis of 4- {2- [6- ({ [ (1R) -1-cyclobutylethyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] -6-cyclopropylpyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOJ-2)
A solution of (1R) -1-cyclobutylethylamine (AOJ-1) (13 mg,2 eq, 0.13 mmol) and DIPEA (34 mg,4 eq, 0.26 mmol) in MeOH (8 mL) was stirred at room temperature for 5 min. To the above mixture was added intermediate (AGT-2) (30 mg,1 equivalent, 65. Mu. Mol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). Will beThe resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 46% B to 58% B in 8 minutes; wavelength: 254/220nm; retention time: 7.8 To give the title compound (AOJ-2) (15.3 mg, 28. Mu. Mol,43%,99.3% purity) as a white solid. M/z 544.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.49 (s, 1H), 8.14-8.03 (m, 3H), 7.92 (d, j=8.1 hz, 1H), 7.79 (d, j=1.5 hz, 1H), 7.68-7.58 (m, 2H), 6.91 (d, j=1.5 hz, 1H), 5.02 (s, 2H), 3.92 (d, j=13.5 hz, 1H), 3.80 (d, j=13.5 hz, 1H), 3.49 (s, 3H), 2.61-1.51 (m, 1H), 2.33-2.22 (m, 1H), 2.12-1.92 (m, 3H), 1.91-1.80 (m, 1H), 1.78-1.62 (m, 3H), 1.08-0.93 (m, 7H).
Example 426: synthesis of 4- {2- [6- ({ [ (1S) -1-cyclobutylethyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] -6-cyclopropylpyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOK-2)
DIPEA (25 mg,3 eq, 0.20 mmol) was added to a stirred mixture of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) and (1S) -1-cyclobutylethylamine (AOK-1) (12 mg,1.8 eq, 0.12 mmol) in MeOH (8 mL) at room temperature. The resulting mixture was stirred at 60 ℃ for an additional overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (7 mg,3 equivalents, 0.20 mmol). The resulting mixture was stirred at room temperature overnight. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 61% B in 8 minutes; wavelength: 254/220nm; retention time: 7.5 To give the title compound (AOK-2) (15.0 mg,28 μmol,42%,99.4% purity) as a white solid. M/z 544.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.02 (m, 3H), 7.92 (d, j=8.1 hz, 1H), 7.79 (d, j=1.6 hz, 1H), 7.70-7.57 (m, 2H), 6.92 (d, j=1.4 hz, 1H), 5.02 (s, 2H), 3.91 (d, j=13.5 hz, 1H), 3.80 (d, j=13.5 hz, 1H), 3.50 (s, 3H), 2.58-2.46 (m, 1H), 2.33-2.21 (m, 1H), 2.15-1.93 (m, 3H), 1.93-1.82 (m, 1H), 1.78-1.59 (m, 3H), 1.06-0.90 (m, 7H).
Example 427: synthesis of 4- [ 2-cyclopropyl-6- (6- { 2-oxa-6-azaspiro [3.3] heptan-6-ylmethyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOL-1)
To intermediate (AIQ-1) (30 mg,1 eq, 62. Mu. Mol) and 2-oxa-6-azaspiro [3.3] at room temperature]To a stirred solution of heptane (AMS-1) (11 mg,1.8 eq, 0.11 mmol) in DCM (8 mL) was added DIPEA (40 mg,5 eq, 0.31 mmol). The resulting mixture was stirred under an air atmosphere at 60 ℃ for 2 days. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: column: XBIdge-prepared OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 30% B to 40% B in 9 minutes; wavelength: 254/220nm; retention time: 8.9 To give the title compound (AOL-1) (11 mg,20 μmol,32%,99.9% purity) as a white solid. M/z 544.1 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.55(s,1H),8.26–8.18(m,2H),7.98(d,J=1.4Hz,1H),7.91–7.81(m,1H),7.65–7.59(m,2H),7.57–7.53(m,1H),6.89(d,J=1.4Hz,1H),5.00(s,2H),4.61(s,4H),3.59(s,2H),3.48(s,3H),3.30(s,4H),2.08–2.00(m,1H),0.97(d,J=6.4Hz,4H)。
Example 428: synthesis of 4- [2- (6- { [ (2-cyclobutylpropan-2-yl) amino ] methyl } -1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOM-2)
A solution of 2-cyclobutylpropan-2-amine (AOM-1) (9 mg,1.2 eq, 78. Mu. Mol) in MeOH (8 mL) was treated with DIPEA (34 mg,4 eq, 0.26. Mu. Mol) at room temperature for 5 min, followed by the addition of intermediate (AGT-2) (30 mg,1 eq, 65. Mu. Mol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (12 mg,5 eq, 0.33 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of MeOH (2 mL) at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm; mobile phase A: water (0.1% NHHCO- +0.1% NH. HO), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 68% B over 10 min; wavelength: 254nm; retention time: 7.87) to give the title compound (AOM-2) (14.3 mg, 26. Mu. Mol,39%,99.7% purity) as a white solid. M/z 558.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.14-8.07 (m, 2H), 8.03 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.79 (d, j=1.5 hz, 1H), 7.66-7.63 (m, 1H), 7.58 (d, j=7.9 hz, 1H), 6.91 (d, j=1.4 hz, 1H), 4.99 (s, 2H), 3.77 (s, 2H), 3.50 (s, 3H), 2.67-2.57 (m, 1H), 2.05-1.98 (m, 1H), 1.97-1.85 (m, 5H), 1.75-1.66 (m, 1H), 1.11 (s, 6H), 1.05-0.94 (m, 4H).
Example 429: synthesis of 2- { [ (2- {4- [ 4-cyano-2- (4-methyl-1, 2, 4-triazol-3-yl) phenyl ] -6-cyclopropylpyridin-2-yl } -3-oxo-1H-isoindol-5-yl) methyl ] (oxetan-3-ylmethyl) amino } -N, N-dimethylacetamide (AON-2)
To a stirred solution of compound (AHZ-2) (30 mg,1 eq, 56. Mu. Mol) and DIPEA (36 mg,5 eq, 0.28 mmol) in EtOH (5 mL) was added 2-chloro-N, N-dimethylacetamide (AON-1) (34 mg,5 eq, 0.28 mmol) at room temperature under nitrogen. The resulting mixture was stirred overnight at 60 ℃ under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18OBD column 30 x 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 6% B to 36% B over 10 min; wavelength: 254nm; retention time: 9.67) to give the title compound (AON-2) (9.7 mg, 16. Mu. Mol,28%,99.6% purity) as a white solid. M/z 617.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.16-8.07 (m, 2H), 8.04 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.77 (s, 1H), 7.68-7.57 (m, 2H), 6.92 (d, j=1.4 hz, 1H), 5.02 (s, 2H), 4.77-4.69 (m, 2H), 4.29 (t, j=6.0 hz, 2H), 3.77 (s, 2H), 3.50 (s, 3H), 3.35 (s, 2H), 3.28-3.20 (m, 1H), 2.99 (s, 3H), 2.92 (d, j=7.7 hz, 2H), 2.88 (s, 3H), 2.08-1.97 (m, 1H), 1.08-1.94 (m, 4H).
Example 430: synthesis of 4- [ 2-cyclopropyl-6- (6- {2, 5-dioxa-8-azaspiro [3.5] nonan-8-ylmethyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOO-2)
To a stirred solution of intermediate (AIQ-1) (30 mg,1 eq, 62. Mu. Mol) and DIPEA (40 mg,5 eq, 0.31 mmol) in DCM (5 mL) under nitrogen at room temperature was added 2, 5-dioxa-8-azaspiro [3.5]Nonane, HCl (AOO-1) (52 mg,5 eq., 0.31 mmol). The resulting mixture was stirred under nitrogen atmosphere at 60 ℃ for 5 days. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditionsLower purification (column: xselect CSH C18 OBD column 30X 150mm,5 μm; mobile phase A: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 5% B to 35% B over 10 min; wavelength: 254nm; retention time: 10.92) to give the title compound (AOO-2) (17.7 mg, 31. Mu. Mol,49%,99.6% purity) as a white solid. M/z 574.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.51 (s, 1H), 8.16-8.07 (m, 2H), 8.04 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.81 (s, 1H), 7.71-7.64 (m, 1H), 7.62 (d, j=7.9 hz, 1H), 6.94 (d, j=1.4 hz, 1H), 5.03 (s, 2H), 4.54 (d, j=6.7 hz, 2H), 4.41 (d, j=6.7 hz, 2H), 3.70-3.63 (m, 4H), 3.51 (s, 3H), 2.66 (s, 2H), 2.49-2.39 (m, 2H), 2.09-1.98 (m, 1H), 1.09-0.94 (m, 4H).
Example 431: synthesis of rac-4- (2- {6- [ (3R, 5S) -5-cyclobutylmorpholin-3-yl ] -1-oxo-3H-isoindol-2-yl } -6-cyclopropylpyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOP-4)
Step 1: [ (2-amino-2-cyclobutylethoxy) methyl ] tributylstannane (AOP-2)
To a stirred solution of 2-amino-2-cyclobutylethanol, HCl (AOP-1) (500 mg,1 eq, 3.30 mmol) in DMF (25 mL) was added NaH (3996 mg,5 eq, 16.5 mmol) at 0deg.C under nitrogen. The resulting mixture was stirred under nitrogen atmosphere at 0 ℃ for 1 hour. Tributyl (iodomethyl) stannane (2.13 g,1.5 eq, 4.95 mmol) was added to the above mixture under nitrogen at 0 ℃. The resulting mixture was stirred at room temperature for an additional 3 hours. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% nh 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 41% B to 51% B in 9 minutes; wavelength: 254/220nm; retention time: 8.3 To give the sub-title compound (AOP-2) as a brown yellow oil(350 mg,0.84mmol,24%,97% pure). M/z 419.2 (M+H) + (ES+)
Step 2:6- (5-Cyclobutylmorpholin-3-yl) -2, 3-dihydro-isoindol-1-one (AOP-3)
A solution of the product from step 1 above (AOP-2) (280 mg,1 eq, 0.67 mmol) and intermediate (ACY-2) (129 mg,1.2 eq, 0.80 mmol) in DCM (8 mL) was stirred overnight at room temperature under a nitrogen atmosphere. Cu (OTf) was added to the above mixture at room temperature over 1 hour 2 (242 mg,1 eq, 0.67 mmol) and HFIP (4 mL) of 2,6-lutidine (72 mg,1 eq, 0.67 mmol). The resulting mixture was stirred under nitrogen at 60 ℃ for an additional 2 days. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions: (column: XBridge preparation type OBD C18 column, 30 x 150mm,5 μm; mobile phase A: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 20% B to 35% B in 8 minutes; wavelength: 254/220nm; retention time: 7.57 To give the sub-title compound (AOP-3) (70 mg,0.26mmol,35%,98% purity) as an off-white solid. M/z 273.2 (M+H) + (ES+)
Step 3: rac-4- (2- {6- [ (3R, 5S) -5-Cyclobutylmorpholin-3-yl ] -1-oxo-3H-isoindol-2-yl } -6-cyclopropylpyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOP-4)
Cs was added to a stirred solution of intermediate (AEG-2) (20 mg,1 equivalent, 0.06 mmol) and the product from step 2 above (AOP-3) (18 mg,1.1 equivalent, 0.07 mmol) in dioxane (5 mL) at room temperature under nitrogen atmosphere 2 CO 3 (39 mg,2 equivalents, 0.12 mmol). RuPhos (11 mg,0.4 eq, 24. Mu. Mol) and RuPhos ring palladium complex Gen.3 (10 mg,0.2 eq, 12. Mu. Mol) were added to the above mixture under nitrogen atmosphere at room temperature. The resulting mixture was stirred under nitrogen atmosphere at 100 ℃ for 1 hour. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The residue was purified by preparative TLC using DCM/MeOH (20/1). The crude product was purified by preparative HPLC under the following conditions (column: xsel)30 x 150mm,5 μm, n of an ect CSH C18 OBD column; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 8% B to 38% B in 10 minutes; wavelength: 254nm; retention time: 9.08 To give the title compound (AOP-4) (3.5 mg,6.1 μmol,10%,99.7% purity) as a white solid. M/z 572.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.04 (m, 3H), 7.95-7.89 (m, 2H), 7.73-7.69 (m, 1H), 7.63-7.57 (m, 1H), 6.92 (d, j=1.4 hz, 1H), 5.01 (s, 2H), 4.05-3.99 (m, 1H), 3.82-3.76 (m, 2H), 3.51 (s, 3H), 3.25 (d, j=10.7 hz, 1H), 3.11 (t, j=10.7 hz, 1H), 2.97-2.89 (m, 1H), 2.36-2.25 (m, 1H), 2.10-1.80 (m, 7H), 1.05-0.94 (m, 4H).
Example 432: synthesis of rel-4- (2- {6- [ (3R, 5S) -5-cyclobutylmorpholin-3-yl ] -1-oxo-3H-isoindol-2-yl } -6-cyclopropylpyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOQ-1)
The crude product (AOP-4) (20 mg,1 eq, 35. Mu. Mol) was purified by preparative chiral HPLC under the following conditions (column: CHIRALPAK ID, 2X 25cm, 5. Mu.m; mobile phase A: etOH- -HPLC; mobile phase B: hex: DCM=1:1 (0.5% 2M NH) 3 -MeOH) -HPLC; flow rate: 16 ml/min; gradient: 50% B to 50% B in 35 minutes; wavelength: 220/254nm; retention time 2 (minutes): 25.34 To give the title compound (AOQ-1) (10.4 mg,18 μmol,52%,99.8% purity) as an off-white solid. M/z 572.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.07 (m, 2H), 8.04 (d, j=1.4 hz, 1H), 7.95-7.89 (m, 2H), 7.73-7.69 (m, 1H), 7.63-7.57 (m, 1H), 6.92 (d, j=1.4 hz, 1H), 5.01 (s, 2H), 4.05-3.99 (m, 1H), 3.82-3.76 (m, 2H), 3.51 (s, 3H), 3.25 (d, j=10.7 hz, 1H), 3.11 (t, j=10.7 hz, 1H), 2.97-2.89 (m, 1H), 2.36-2.25 (m, 1H), 2.10-1.80 (m, 7H), 1.05-0.94 (m, 4H). Column: CHIRALPAK ID-3,4.6 x 50mm,3 μm; mobile phase a: hex: dcm=1:1) (0.1% DEA): etoh=50:50; flow rate: 1 ml/min; retention time: 3.932.
Example 433: synthesis of rel-4- (2- {6- [ (3R, 5S) -5-cyclobutylmorpholin-3-yl ] -1-oxo-3H-isoindol-2-yl } -6-cyclopropylpyridin-4-yl) -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOR-1)
The crude product (AOP-4) (20 mg,1 eq, 35. Mu. Mol) was purified by preparative chiral HPLC under the following conditions (column: CHIRALPAK ID, 2X 25cm, 5. Mu.m; mobile phase A: etOH- -HPLC; mobile phase B: hex: DCM=1:1 (0.5% 2M NH) 3 -MeOH) -HPLC; flow rate: 16 ml/min; gradient: 50% B to 50% B in 35 minutes; wavelength: 220/254nm; retention time 1 (min): 12.75 To give the title compound (AOR-1) (5.1 mg,8.9 μmol,25%,99.9% purity) as a white solid. M/z 572.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.07 (m, 2H), 8.04 (d, j=1.4 hz, 1H), 7.95-7.89 (m, 2H), 7.73-7.69 (m, 1H), 7.63-7.57 (m, 1H), 6.92 (d, j=1.4 hz, 1H), 5.01 (s, 2H), 4.05-3.99 (m, 1H), 3.82-3.76 (m, 2H), 3.51 (s, 3H), 3.25 (d, j=10.7 hz, 1H), 3.11 (t, j=10.7 hz, 1H), 2.97-2.89 (m, 1H), 2.36-2.25 (m, 1H), 2.10-1.80 (m, 7H), 1.05-0.94 (m, 4H). Column: CHIRALPAK ID-3,4.6 x 50mm,3 μm; mobile phase a: hex: dcm=1:1) (0.1% DEA): etoh=50:50; flow rate: 1 ml/min; retention time: 2.171.
Example 434: synthesis of 4- [2- (6- {2- [ (cyclobutylmethyl) amino ] propan-2-yl } -1-oxo-3H-isoindol-2-yl) -6-cyclopropylpyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOS-2)
To a stirred mixture of compound (ALK-9) (40 mg,1 eq, 82. Mu. Mol) and cyclobutal (AOS-1) (69 mg,10 eq, 0.82 mmol) in MeOH (2.5 mL) was added DIPEA (32 mg,3 eq, 0.25 mmol) at room temperature. NaBH (OAc) was added to the above mixture at room temperature 3 (52 mg,3 eq, 0.25 mmol). The resulting mixture was stirred at 60 ℃ for another 4 days. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 45% B to 60% B in 8 minutes; wavelength: 254/220nm; retention time: 7.72 To give the title compound (AOS-2) (4.5 mg,8.1 μmol,9.9%,99.8% purity) as a white solid. M/z 558.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.48 (s, 1H), 8.16-8.07 (m, 2H), 8.07-7.97 (m, 2H), 7.93 (d, j=8.0 hz, 1H), 7.88-7.75 (m, 2H), 6.98 (d, j=1.5 hz, 1H), 5.09 (s, 2H), 3.48 (s, 3H), 2.68 (d, j=7.4 hz, 2H), 2.54-2.45 (m, 1H), 2.16-2.02 (m, 3H), 1.99-1.76 (m, 8H), 1.71-1.60 (m, 2H), 1.10-0.96 (m, 4H).
Example 435: synthesis of 4- (2-cyclopropyl-6- (4-fluoro-6- ((hexahydro-1H-furo [3,4-b ] pyrrol-1-yl) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AOT-2)
To intermediate (AKR-1) (60 mg,1 eq, 0.12 mmol) and hexahydro-1H-cyclopenta [ c ] at room temperature]To a stirred mixture of furan (AOT-1) (15 mg,1.1 eq, 0.13 mmol) in DCM (5 mL) was added DIPEA (47 mg,3 eq, 0.36 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect peptide CSH C18 19 x 150mm,5 μm; mobile phase a: water (0.1% FA), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 4% B to 34% B over 10 min; wavelength: 254nm; retention time: 9.93) to give the title compound (AOT-2) (5.0 mg,8.7 μmol,7.2%,99.4% purity) as a white solid. M/z 576.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.16-8.07 (m, 2H), 8.02 (d, J=1.4 Hz, 1H), 7.92 (d, J=8.0 Hz, 1H), 7.65 (d, J=1.1 Hz, 1H), 7.46-7.39 (m, 1H), 6.95 (d, J=1.4 Hz, 1H), 5.07 (s, 2H), 3.90 (d, J=12.9 Hz, 1H), 3.86-3.71 (m, 2H), 3.66 (d, J=12.9 Hz, 1H), 3.49 (s, 3H), 3.48-3.39 (m, 1H), 3.24 (t, J=6.1 Hz, 1H), 3.19-3.08 (m, 1H), 3.04-2.98 (m, 1H), 2.98 (m, 2H), 3.86-3.71 (m, 2H), 3.66 (d, J=12.9 Hz, 1H), 3.49 (s, 3H), 3.48-3.39 (m, 1H), 3.24 (m, 1H), 3.98 (1.80-1H), 1.80 (m, 1H), 1.85.1.1.80 (1H).
Example 436: synthesis of 4- (2-cyclopropyl-6- (4-fluoro-6- (((3 aS,6 aS) -hexahydro-1H-furo [3,4-b ] pyrrol-1-yl) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AOU-1)
The crude product (AOT-2) (20 mg,1 eq, 35 μmol) was purified by preparative chiral HPLC under the following conditions (column: CHIRALPAK IG,2×25cm,5 μm; mobile phase a: hex: dcm=1:1 (0.5% 2m NH) 3 MeOH) -HPLC, mobile phase B: IPA-HPLC; flow rate: 20 ml/min; gradient: 50% B to 50% B within 16 minutes; wavelength: 220/254nm; retention time 1 (min): 9.534 To give the title compound (AOU-1) (9.8 mg,17 μmol,49%,99.8% purity) as a white solid. M/z 576.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.16-8.06 (m, 2H), 8.01 (d, j=1.4 hz, 1H), 7.92 (d, j=8.1 hz, 1H), 7.64 (s, 1H), 7.45-7.38 (m, 1H), 6.95 (d, j=1.4 hz, 1H), 5.05 (s, 2H), 3.89 (d, j=12.9 hz, 1H), 3.85-3.71 (m, 2H), 3.65 (d, j=12.9 hz, 1H), 3.51-3.39 (m, 4H), 3.24 (t, j=6.1 hz, 1H), 3.18-3.07 (m, 1H), 3.04-2.87 (m, 2H), 2.71-2.55 (m, 1H), 2.09-1.98 (m, 1H), 3.85-3.71 (m, 2H), 3.65 (m, 1H), 3.51-3.39 (m, 1H), 1.95 (m, 1H). Column: CHIRALPAK IG-3,4.6 x 50mm,3 μm; mobile phase a: hex: dcm=1:1) (0.1% DEA): ipa=50:50; flow rate: 1 ml/min; retention time: 2.236.
Example 437: synthesis of 4- (2-cyclopropyl-6- (4-fluoro-6- (((3 aR,6 aR) -hexahydro-1H-furo [3,4-b ] pyrrol-1-yl) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AOV-1)
The crude product (AOT-2) (20 mg,1 eq, 35 μmol) was purified by preparative chiral HPLC under the following conditions (column: CHIRALPAK IG,2×25cm,5 μm; mobile phase a: hex: dcm=1:1 (0.5% 2m NH) 3 MeOH) -HPLC, mobile phase B: IPA-HPLC; flow rate: 20 ml/min; gradient: 50% B to 50% B within 16 minutes; wavelength: 220/254nm; retention time 2 (minutes): 13.427 To give the title compound (AOV-1) (10.0 mg,17 μmol,50%,99.8% purity) as a white solid. M/z 576.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.16-8.06 (m, 2H), 8.01 (d, j=1.4 hz, 1H), 7.92 (d, j=8.0 hz, 1H), 7.64 (d, j=1.2 hz, 1H), 7.45-7.38 (m, 1H), 6.95 (d, j=1.4 hz, 1H), 5.06 (s, 2H), 3.89 (d, j=12.9 hz, 1H), 3.85-3.70 (m, 2H), 3.65 (d, j=12.9 hz, 1H), 3.52-3.38 (m, 4H), 3.24 (t, j=6.2 hz, 1H), 3.17-3.07 (m, 1H), 3.04-2.96 (m, 1H), 2.96-2.85 (m, 1.71.8-1H), 3.52-3.70 (m, 2H), 3.65 (d, j=12.9 hz, 1H), 3.52-3.80 (m, 1H), 1.96 (m, 1H). Column: CHIRALPAK IG-3,4.6 x 50mm3um; mobile phase a: hex: dcm=1:1) (0.1% DEA): ipa=50:50; flow rate: 1 ml/min; retention time: 3.345.
Example 438: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (2R) -2- (methoxymethyl) azetidin-1-yl ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOW-2)
To a stirred solution of intermediate (AKR-1) (30 mg,1 eq, 60 μmol) and (R) -2- (methoxymethyl) azetidine (AOW-1) (7 mg,1.2 eq, 72 μmol) in DCM (6 mL) at room temperature was added DIPEA (31 mg,4 eq, 0.24 mmol). The resulting mixture was stirred at 60 ℃ overnight. Will beThe mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC under the following conditions (column: xselect CSH C18 OBD column 30 x 150mm,5 μm, n; mobile phase a: water (0.1% NH) 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 38% B to 68% B in 10 minutes; wavelength: 254nm; retention time: 7.87 To give the title compound (AOW-2) (10.5 mg,19 μmol,31%,99.3% purity) as a white solid. M/z 564.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.15-8.07 (m, 2H), 8.02 (d, j=1.5 hz, 1H), 7.94-7.90 (m, 1H), 7.61 (d, j=1.2 hz, 1H), 7.41-7.37 (m, 1H), 6.95 (d, j=1.4 hz, 1H), 5.06 (s, 2H), 3.91 (d, j=13.2 hz, 1H), 3.65 (d, j=13.3 hz, 1H), 3.50 (s, 3H), 3.41-3.36 (m, 2H), 3.33 (s, 1H), 3.31 (s, 3H), 3.29-3.26 (m, 1H), 3.02-2.93 (m, 1H), 2.10-1.96 (m, 3H), 1.08 (m, 4H).
Example 439: synthesis of 4- { 2-cyclopropyl-6- [4, 5-difluoro-6- ({ [ (1-hydroxycyclobutyl) methyl ] amino } methyl) -1-oxo-3H-isoindol-2-yl ] pyridin-4-yl } -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (AOX-1)
A solution of intermediate (ALY-1) (30 mg,1 eq, 0.06 mmol), DIPEA (31 mg,4 eq, 0.24 mmol) and 1- (aminomethyl) cyclobutan-1-ol (AW-1) (12 mg,2 eq, 0.12 mmol) in MeOH (8 mL) was stirred overnight at 40 ℃. The mixture was cooled to room temperature. Adding NaBH to the mixture at 0deg.C 4 (11 mg,5 equivalents, 0.30 mmol). The resulting mixture was stirred at room temperature for an additional 1 hour. The reaction was then quenched by the addition of 2mL of MeOH at 0 ℃. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 44% B to 54% B in 8 minutes; wave-guideAnd (3) length: 254/220nm; retention time: 7.12 To give the title compound (AOX-1) (8.6 mg,15 μmol,24%,98.8% purity) as a white solid. M/z 582.1 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.48 (s, 1H), 8.15-8.06 (m, 2H), 8.00 (d, j=1.4 hz, 1H), 7.91 (d, j=8.0 hz, 1H), 7.77 (d, j=5.2 hz, 1H), 6.95 (d, j=1.5 hz, 1H), 5.10 (s, 2H), 3.99 (s, 2H), 3.49 (s, 3H), 2.72 (s, 2H), 2.16-1.97 (m, 5H), 1.81-1.68 (m, 1H), 1.61-1.48 (m, 1H), 1.09-0.95 (m, 4H).
Example 440: synthesis of (S) -4- (2-cyclopropyl-6- (4-fluoro-6- ((2- (methoxymethyl) pyrrolidin-1-yl) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (AOY-2)
To a stirred mixture of intermediate (AKR-1) (30 mg,1 eq, 0.06 mmol) and (2S) -2- (methoxymethyl) pyrrolidine (AOY-1) (14 mg,2 eq, 0.12 mmol) in DCM (5 mL) was added DIPEA (23 mg,3 eq, 0.18 mmol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 60% B within 9 minutes; wavelength: 254/220nm; retention time: 8.08 To give the title compound (AOY-2) (4.5 mg, 7.8. Mu. Mol,11%,99.9% purity) as a white solid. M/z 578.0 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.51 (s, 1H), 8.16-8.07 (m, 2H), 8.01 (d, J=1.4 Hz, 1H), 7.96-7.89 (m, 1H), 7.65 (s, 1H), 7.46-7.39 (m, 1H), 6.96 (d, J=1.4 Hz, 1H), 5.06 (s, 2H), 4.25 (d, J=13.5 Hz, 1H), 3.51 (s, 4H), 3.50-3.44 (m, 1H), 3.42-3.37 (m, 1H), 3.35 (s, 3H), 2.94-2.85 (m, 1H), 2.85-2.74 (m, 1H), 2.33-2.22 (m, 1H), 2.08-1.91 (m, 2H), 1.79-1.68 (m, 2H), 3.50-3.44 (m, 1H), 3.55-1.95 (m, 1H).
Example 441: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (2R) -2- (fluoromethyl) pyrrolidin-1-yl ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4H-1, 2, 4-triazol-3-yl) benzonitrile (AOZ-2)
To a stirred mixture of intermediate (AKR-1) (30 mg,1 eq, 0.06 mmol) and (2R) -2- (fluoromethyl) pyrrolidine (AOZ-1) (12 mg,2 eq, 0.12 mmol) in DCM (5 mL) was added DIPEA (23 mg,3 eq, 0.18 mmol) at room temperature. The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 60% B within 9 minutes; wavelength: 254/220nm; retention time: 8.98 To give the title compound (AOZ-2) (4.1 mg,7.3 μmol,12%,93.1% purity) as a white solid. M/z 566.3 (M+H) + (ES+)。 1 H NMR (400 MHz, methanol-d 4) delta 8.50 (s, 1H), 8.16-8.07 (m, 2H), 8.00 (d, j=1.4 hz, 1H), 7.95-7.89 (m, 1H), 7.65 (d, j=1.1 hz, 1H), 7.46-7.38 (m, 1H), 6.96 (d, j=1.4 hz, 1H), 5.06 (s, 2H), 4.44 (d, j=5.2 hz, 1H), 4.32 (d, j=5.2 hz, 1H), 4.19 (d, j=13.6 hz, 1H), 3.59 (d, j=13.6 hz, 1H), 3.51 (s, 3H), 3.02-2.87 (m, 2H), 2.39-2.28 (m, 1H), 2.08-1.92 (m, 2H), 1.80-1.80 (m, 1.95H), and 4.95 (m, 1H).
Example 442: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (2S) -2- (fluoromethyl) pyrrolidin-1-yl ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (APA-2)
To intermediate (AKR-1) (30 mg,1 eq, 0.06 mmol) at room temperatureTo a stirred mixture of (2S) -2- (fluoromethyl) pyrrolidine, HCl (APA-1) (11 mg,1.8 eq, 0.11 mmol) in DCM (8 mL) was added DIPEA (31 mg,4 eq, 0.24 mmol). The resulting mixture was stirred at 60 ℃ overnight. The mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; gradient: 50% B to 60% B within 9 minutes; wavelength: 254/220nm; retention time: 8.83 To give the title compound (APA-2) (10.1 mg,18 μmol,29%,97.5% purity) as a white solid. M/z 566.9 (M+H) + (ES+)。 1 H NMR(400MHz,DMSO-d6)δ8.55(s,1H),8.25–8.18(m,2H),7.92–7.84(m,2H),7.61–7.56(m,1H),7.51–7.44(m,1H),6.95(d,J=1.4Hz,1H),5.07(s,2H),4.43–4.31(m,2H),4.12(d,J=13.8Hz,1H),3.58(d,J=13.8Hz,1H),3.49(s,3H),2.98–2.83(m,1H),2.83–2.76(m,1H),2.29–2.23(m,1H),2.09–2.01(m,1H),1.94–1.87(m,1H),1.73–1.52(m,3H),1.02–0.94(m,4H)。
Example 443: synthesis of 4- [ 2-cyclopropyl-6- (4-fluoro-6- { [ (2R) -2- (methoxymethyl) pyrrolidin-1-yl ] methyl } -1-oxo-3H-isoindol-2-yl) pyridin-4-yl ] -3- (4-methyl-1, 2, 4-triazol-3-yl) benzonitrile (APB-2)
To a stirred mixture of intermediate (AKS-1) (30 mg,1 eq, 0.06 mmol) and (2R) -2- (methoxymethyl) pyrrolidine (APB-1) (12 mg,1.8 eq, 0.11 mmol) in MeOH (8 mL) was added DIPEA (39 mg,5 eq, 0.30 mmol) at room temperature. The resulting mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo. The crude product was purified by preparative HPLC on a column (XBridge prep. OBD C18 column, 30 x 150mm,5 μm; mobile phase a: water (0.1% NH 4 HCO 3 +0.1%NH 3 .H 2 O), mobile phase B: meCN; flow rate: 60 ml/min; ladderDegree: 50% B to 60% B in 8 minutes; wavelength: 254/220nm; retention time: 7.9 To give the title compound (APB-2) (17.4 mg,30 μmol,50%,99.8% purity) as a white solid. M/z 578.0 (M+H) + (ES+). 1 H NMR(400MHz,DMSO-d6)δ8.55(s,1H),8.25–8.18(m,2H),7.93(d,J=1.3Hz,1H),7.90–7.84(m,1H),7.57(s,1H),7.46(d,J=9.9Hz,1H),6.95(d,J=1.4Hz,1H),5.07(s,2H),4.17(d,J=13.9Hz,1H),3.49(s,4H),3.42(d,J=9.5,5.4Hz,1H),3.30–3.20(m,4H),2.83–2.68(m,2H),2.23–2.12(m,1H),2.08–2.01(m,1H),1.90–1.84(m,1H),1.70–1.58(m,2H),1.58–1.45(m,1H),1.01–0.94(m,4H)。
Example 444:4- (2-cyclopropyl-6- (6- (2- (((3-methyloxetan-3-yl) methyl) amino) propan-2-yl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (compound # 433), 4- (2-cyclopropyl-6- (6- ((ethyl (oxetan-3-ylmethyl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (compound # 434), synthesis of 4- (2-cyclopropyl-6- (5-fluoro-6- (hydroxymethyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (Compound # 435) and 4- (2-cyclopropyl-6- (5-fluoro-6- ((((1-hydroxycyclobutyl) methyl) amino) methyl) -1-oxoisoindolin-2-yl) pyridin-4-yl) -3- (4-methyl-4H-1, 2, 4-triazol-3-yl) benzonitrile (Compound # 436).
The compounds in table 3 were prepared according to a similar method as described in the examples above.
TABLE 3 characterization data for additional Compounds
Example 445: cbl-b biochemical assay (TR-FRET)
Recombinant human Cbl-b (aa 36-427) was expressed in E.coli (E.coli) and purified and biotinylated in vitro. Proteins were diluted to 12nM in freshly prepared assay buffer consisting of: 50mM HEPES, pH 7.0, 100mM NaCl, 5mM MgCl2, 0.01% Triton-X100, 0.01% BSA and 1mM DTT.
Recombinant human Src (aa 254-536) -GSSGSS-Zap-70 (aa 281-297) fusion protein was expressed in E.coli and purified. Proteins were diluted to 2-5nM in assay buffer and supplemented with ATP to 1mM.
fluorescein-BODIPY-labeled UBED2D (C85K) -Ub was prepared by combining ubiquitin (Ub) labeled with fluorescein-BODIPY maleimide at its N-terminus (Cemerfeeil technologies Co., thermo Fisher catalog number B10250) with E.coli expressed and purified UBED2D (C85K) [ see Dou et al, "Nature Structure and molecular biology (Nature Structural and Molecular Biology) [ 8:982-987,2013 ]]. Recombinant human UBE2D2 (C85K) was expressed in e.coli, purified and ubiquitinated and labelled Bodipy in vitro. In the absence of MgCl 2 The protein was diluted to 200nM in assay buffer (or Cisbio PPI buffer). Streptavidin-terbium was added to 2nM and EDTA to 10mM to provide a binding assay mixture.
Compounds were dissolved in DMSO and diluted to prepare ten-point dilution series. 100nl of each concentration of compound was dispensed in duplicate into 384 well black assay plates using acoustic dispensing. Wells for maximum signal control received only 100nl of DMSO and wells for minimum signal control received 100nl of reference inhibitor compound at a final assay concentration of 100mM to produce 100% inhibition.
Mu.l of diluted Cbl-b enzyme was added to all wells of the assay plate and incubated for 30-60 min at room temperature. The enzyme assay was started by adding 5 μl of Src-Zap/ATP mix to all wells and the plates were incubated for 60 minutes at room temperature. The enzyme reaction was stopped and the binding reaction was initiated by adding 10 μl of binding assay mixture to all wells and incubating the plates at room temperature for 60 minutes prior to the assay reading.
The final assay conditions consisted of: 6nM Cbl-b, 1-2.5nM Src-Zap70, 0.5mM ATP, 1% (v/v) DMSO (enzyme reaction) and 100nM UBE2D2 (C85K) -Ub-FL-BODIPY, 5mM EDTA, 1nM streptavidin-Tb (binding reaction).
HTRF assay signals were measured at 520nm on an Envision microplate reader and reference signals were measured at 485 or 620 nm. Data were normalized using maximum and minimum assay controls: inhibition% = 100- (100 x ((max control) -unknown)/(max control-min control)). Normalized dose response data were fitted using a 4 parameter dose response equation and IC50 of the test compound was deduced.
Table 4 shows the activity of selected compounds of the invention in a TR-FRET biochemical assay. Compounds having an activity designated "A" provide IC 50 Less than or equal to 0.2 mu M; compounds having an activity designated "B" provide IC 50 >0.20 mu M but less than or equal to 1 mu M; compounds designated as "C" provide IC 50 >1 mu M but less than or equal to 10 mu M; compounds designated as "D" provide IC 50 >10 mu M but<50μM。
TABLE 4 results of cbl-b biochemical assay (TR-FRET)
/>
/>
/>
/>
/>
Example 446: C-Cbl biochemical assay (TR-FRET)
Recombinant human c-Cbl (aa 47-435) was expressed in E.coli (E.coli) and purified and biotinylated in vitro. Proteins were diluted to 12nM in freshly prepared assay buffer consisting of: 50mM HEPES, pH 7.0, 100mM NaCl, 5mM MgCl 2 0.01% Triton-X100, 0.01% BSA and 1mM DTT.
Recombinant human Src (aa 254-536) -GSSGSS-Zap-70 (aa 281-297) fusion protein was expressed in E.coli and purified. The protein was diluted to 5-20nM in assay buffer and ATP was added to 1mM.
Recombinant human UBE2D2 (C85K) was expressed in e.coli, purified and ubiquitinated and labelled Bodipy in vitro. In the absence of MgCl 2 The protein was diluted to 200nM in assay buffer (or Cisbio PPI buffer). Streptavidin-terbium was added to 2nM and EDTA to 10mM to provide a binding assay mixture.
Compounds were dissolved in DMSO and diluted to prepare ten-point semilog dilution series. 100nl of each compound concentration was dispensed in duplicate into 384 well black assay plates using acoustic dispensing. Wells for maximum signal control received only 100nl of DMSO and wells for minimum control received 100nl of reference inhibitor compound at a final assay concentration of 100mM to produce 100% inhibition.
Mu.l of diluted c-cbl enzyme was added to all wells of the assay plate and incubated for 30 minutes at room temperature. The enzyme assay was started by adding 5 μl of Src-Zap/ATP mix to all wells and the plates were incubated for 60-90 minutes at room temperature. The enzyme reaction was stopped and the binding reaction was initiated by adding 10 μl of binding assay mixture to all wells and incubating the plates at room temperature for 60 minutes prior to the assay reading.
The final assay conditions consisted of: 6nM C-cbl, 2.5-10nM Src-Zap70, 0.5mM ATP, 1% (v/v) DMSO (enzyme reaction) and 100nM UBE2D2 (C85K) -Ub-FL-BODIPY, 5mM EDTA, 1nM streptavidin-Tb (binding reaction).
At EnvHTRF assay signals were measured at 520nm on an ision microplate reader and reference signals were measured at 485 or 620 nm. Data were normalized using maximum and minimum assay controls: inhibition% = 100- (100 x ((max control) -unknown)/(max control-min control)). Fitting normalized dose response data using a 4 parameter dose response equation and deriving IC for test compounds 50
Table 5 lists the compounds of the invention tested in the TR-FRET c-Cbl biochemical assay. IC for all compounds tested 50 ≤1μM。
TABLE 5 results of c-Cbl biochemical assay (TR-FRET)
Compound ID Compound ID Compound ID
ABI-1 ABY-1 ABX-6
AG-1 AI-2 AE-2
AF-1 AM-2 AL-1
AN-1 AH-2 AAU-1
ACR-7 ACM-8 AK-2
ABR-1 AJ-1 AQ-1
AO-1 AP-1 AR-1
ACT-2 ACS-4 ACG-3
ABN-2 ABP-3 ABC-7
ACE-1 ACF-1 ACA-1
ACB-1 ACI-1 ACH-1
ACW-1 ACP-3 ACV-2
CAN-5 ACO-1 AD-3
AS-2 ABF-8 AD-4
ACC-8 ABO-5 ACL-1
ABS-1 ACU-2 AAQ-1
Z-1 W-2 T-2
AA-2 ABD-1 ACK-3
X-2 R-2 AB-2
Table 5A shows the activity of selected compounds of the invention in a TR-FRET c-Cbl biochemical assay. Compounds having an activity designated "A" provide IC 50 Less than or equal to 0.2 mu M; compounds having an activity designated "B" provide IC 50 >0.20 mu M but less than or equal to 1 mu M; compounds designated as "C" provide IC 50 >1 mu M but less than or equal to 10 mu M; compounds designated as "D" provide IC 50 >10 mu M but<50μM。
TABLE 5A. Cbl-b Biochemical assay (TR-FRET) results
/>
/>
Example 447: jurkat reporter assay (NFAT luciferase)
Commercially available NFAT luciferase constructs were purchased and transfected into Jurkat cells to generate stable cell lines. Jurkat (clone E6-1) cells were engineered to stably express the NFAT luciferase reporter. Cells were maintained in RPMI 1640 medium with 10% FBS, 200 μg/ml hygromycin B (Hygromycin B) and 1% Penicillin (Penicillin)/streptomycin and assayed in the same medium omitting hygromycin B.
Test compounds were dissolved in DMSO (typically at 20 mM) and ten-point semilog dilution series were prepared using acoustic partitioning. The 125nl concentration of each compound was dispensed in duplicate into the wells of 384-well white assay plates, typically providing a highest final assay compound concentration of 100 mM. The assay low control wells received DMSO alone and the high control received standard compound (NDI-996179), providing a final assay concentration of 100 mM.
Jurkat cells were harvested and grown at 4.445X10 5 Individual cells/ml were resuspended in assay medium. 22.5 μl (10,000 cells) was added to the wells of a white 384 well assay plate and incubated at 37℃with 5% CO 2 Incubate for 15 minutes. anti-CD 3 antibody (Siemens technologies # 16-0037-85) was diluted to 10. Mu.g/ml in assay medium. 2.5 μl was added to the wells of the assay plate and incubated at 37℃with 5% CO 2 The medium was incubated for 6 hours. Additional control wells receiving assay medium instead of anti-CD 3 were included.
The assay plate was equilibrated to room temperature and 25 μl was usedReagent (Propranolol)Meger company (Promega) # E2520) was added to all wells. Plates were centrifuged at 100Xg for 1 min and incubated for 10 min. Luminescence was read on an Envision microplate reader.
Data were normalized using high and low assay controls: activation% = 100- (100 x ((high control) -unknown)/(high control-low control)). Normalized data were fitted to a nonlinear regression equation (log inhibitor vs. response variable slope 4 parameter) to derive compound EC50 values. Fold activation relative to baseline was expressed as mean luminescence from high control wells/mean luminescence from low control wells (anti-CD 3 only). Maximum activation of the compounds is also expressed as normalized value for the reference standard compound (maximum activation% of test compound/100)
Table 6 shows selected compounds of the invention tested in the Jurkat reporter assay. EC of all tested compounds 50 ≤1μM。
TABLE 6 Jurkat reporter assay (NFAT luciferase) results
/>
Table 6A shows selected compounds of the invention tested in the Jurkat reporter assay. Compounds having an activity designated "A" provide EC 50 Less than or equal to 0.2 mu M; compounds having an activity designated "B" provide EC 50 >0.20 mu M but less than or equal to 1 mu M; compounds designated as "C" provide EC 50 >1 mu M but less than or equal to 10 mu M; compounds designated "D" provide EC 50 >10 mu M but<50μM。
TABLE 6A Jurkat reporter assay (NFAT luciferase) results
/>
/>
Example 448: t cell alpha LISA assay
Test compounds were dissolved in DMSO (typically at 20 mM) and ten-point semilog dilution series were prepared using acoustic partitioning. The 300nl concentration of each compound was dispensed in duplicate into the wells of 384-well white assay plates, typically providing a maximum final assay compound concentration of 50 mM. The assay low control wells received DMSO alone and the high control received standard compound (NDI-996238), providing a final assay concentration of 50 mM.
Cryopreserved human T cells were thawed and incubated for 24 hours prior to use in RPMI 1640 medium containing 10% HI FBS and 1% penicillin-streptomycin. Cells were harvested and expressed at 1.25x10 6 The individual cells/ml concentration was resuspended in assay medium. Mu.l (50,000 cells) were added to wells of a white 384-well assay plate, and 5. Mu.l (0.5. Mu.g/ml) of anti-CD 28 antibody (Life technologies Co. (Life Technologies) # 16-0289-85) and 5. Mu.l (0.75. Mu.g/ml) of anti-CD 3 antibody (Siemens Feisha technologies Co. # 16-0037-85) were added to the appropriate wells. The plates were then exposed to 5% CO at 37℃ 2 The medium was incubated for 48 hours.
At the end of the incubation period, the assay plates were centrifuged at 300Xg for 5 minutes and 50. Mu.L of supernatant was transferred to the plates for IL-2 analysis. IL2 levels were measured according to the manufacturer's protocol (Perkin Elmer) #AL 221C). Briefly, human IL2 (0.3. Mu.g) was dissolved in 100. Mu.L of 1 XAlphaLISA immunoassay buffer. The semi-logarithmic IL2 standard curve was generated at the highest concentration of 30. Mu.g/mL in different assay media according to the manufacturer's protocol. mu.L of sample or IL-2 standard was added to 384 well Proxiplate along with 8. Mu.L of 2.5 XMIX alpha LISA anti-analyte receptor beads (final 10. Mu.g/mL) and biotinylated antibody anti-analyte (final 1 nM). Plates were incubated at 25℃for 60 minutes in the dark and at 25℃for a further 30 minutes in the dark before the addition of 10. Mu.L of 2 XSA donor beads (final 40. Mu.g/mL). Absorbance was read at 570nm using Envision (perkin elmer)
Data were normalized using high and low assay controls: activation% = 100- (100 x ((high control) -unknown)/(high control-low control)). Normalized data were fitted to a nonlinear regression equation (log inhibitor vs. response variable slope 4 parameter) to derive compound EC50 values. Fold activation relative to baseline was expressed as mean luminescence from high control wells/mean luminescence from low control wells (anti-CD 3/anti-CD 28 alone). Maximum activation of the compounds is also expressed as normalized value (maximum activation% of test compound/100) of the reference standard compound.
Table 7 shows selected compounds of the invention tested in a T cell assay. EC of all tested compounds 50 ≤1μM。
TABLE 7.T cell AlphaLISA assay results
Table 7A shows selected compounds of the invention tested in a T cell assay. EC of all tested compounds 50 ≤0.2μM。
TABLE 7A T cell AlphaLISA assay results
Example 449: TABLE 8 results of hcbl-b Displacement measurement
Table 7 shows selected compounds of the invention tested in the hCbl-b shift assay. EC of all tested compounds 50 ≤10μM。
TABLE 8 results of hcbl-b Displacement measurement
Compound ID Compound ID Compound ID Compound ID
ADR-3 AFK-1 ADV-1 AFM-2
AFO-2 AEF-1 AEI-5 AGU-10
ADZ-2 AEN-1 BE-3 ADX-2
AFT-2 ADJ-1 AGS-2 AFI-1
AFU-1 ADQ-2 AHI-4 AED-2
AEY-1 AEH-1 AEO-2 AGV-1
AGW-6 ADT-2 AFL-2 ADH-1
AGR-1 AGG-2 AFJ-1 AEM-9
AGP-1 AGZ-1 AEQ-3 ADB-1
AFH-1 AES-1 AGA-1 AND-2
AFN-3 AFW-3 AW-2 AT-1
AHG-2 AFQ-3 ACY-4 AEE-2
ADF-2 AFV-3 AFG-1 ADI-1
AGQ-2 AEJ-3 AHC-1 AGY-3
AHH-3 ADO-2 AZ-2 AEA-2
Table 8A shows selected compounds of the invention tested in hCbl-b shift assay. EC of all tested compounds 50 <50nM。
Table 8A. Results of hcbl-b Displacement measurement
Example 450: jurkat pZAP70 HTRF assay
Jurkat wild-type (clone E6-1) cells were maintained in RPMI 1640 medium and assayed in the medium using 10% FBS, 1% L-glutamine and 1% penicillin/streptomycin. Test compounds were dissolved in DMSO (typically at 20 mM) and ten-point semilog dilution series were prepared using acoustic partitioning. 62.5nl of each compound concentration was dispensed in duplicate into the wells of 384-well white assay plates, typically providing a maximum final assay compound concentration of 10 mM. The assay low control wells received DMSO alone and the high control received standard compound (NDI-205422/NDI-206096) to provide a final assay concentration of 10 mM. Harvesting Jurkat cells and at 1X10 7 Individual cells/ml were resuspended in assay medium. Mu.l (100,000 cells) were added to the wells of a white 384-well assay plate containing acoustically assigned compound and incubated at 37℃with 5% CO 2 Incubate for 15 minutes. anti-CD 3 antibody (Siemens technologies # 16-0037-85) was diluted to 30. Mu.g/ml in assay medium and 2. Mu.l was added to the wells of the assay plate at a final concentration of 5. Mu.g/ml. The assay plate was incubated at 37℃with 5% CO 2 Incubate for 3 hours. Phospho-Zap70 (Y-319) was quantified using the Cisbio HTRF system (catalog number 64 ZAPPEH). Cells were lysed by adding 4 μl of lysis buffer for 30 min at room temperature. Mu.l of diluted donor and acceptor antibody mixture was added and the plate was sealed and incubated overnight at room temperature. The assay was read on an Envision microplate reader using HTRF read mode, with laser excitation and emission reading at 665 nm.
Data were normalized using high and low assay controls:
activation% = 100- (100 x ((high control) -unknown)/(high control-low control)).
Normalized data were fitted to a nonlinear regression equation (log inhibitor vs. response variable slope 4 parameter) to derive compound EC50 values. Maximum activation of the compounds is also expressed as normalized value (maximum activation% of test compound/100) of the reference standard compound.
Table 9 shows selected compounds of the invention tested in the Jurkat pZAP70 HTRF assay. Compounds having an activity designated "A" provide EC 50 Less than or equal to 0.2 mu M; compounds having an activity designated "B" provide EC 50 >0.20 mu M but less than or equal to 1 mu M; compounds designated as "C" provide EC 50 >1 mu M but less than or equal to 10 mu M; compounds designated "D" provide EC 50 >10 mu M but<50μM。
TABLE 9 ZAP70 Jurkat assay
/>
Table 9A shows selected compounds of the invention tested in the Jurkat pZAP70 HTRF assay. EC of all tested compounds 50 <1μM。
TABLE 9A.ZAP70 Jurkat assay
/>
While various embodiments of the invention have been described, it will be apparent that the examples can be modified to provide additional embodiments utilizing the compounds and methods of the invention. It is, therefore, to be understood that the scope of the invention is to be defined by the appended claims rather than by the specific embodiments which have been represented by way of example.

Claims (21)

1. A compound of formula I:
or a pharmaceutically acceptable salt thereof, wherein:
ring a is a 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; or a 5-6 membered saturated or partially unsaturated heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
each R 1 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
ring B is a divalent phenyl or divalent 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
each R 2 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CH 2 OR、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
ring C is a divalent phenyl or divalent 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
each R 3 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or an optionally substituted group selected from:C 1-6 aliphatic series; a phenyl ring; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; and a 4-8 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; an optionally substituted 8-10 membered partially unsaturated or heteroaromatic bicyclic heterocycle having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
or two R 3 Radicals and each R 3 The atoms to which the groups are attached optionally together form a fused 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring; fused 5-6 membered monocyclic aryl ring; a fused 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; or a fused 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
X is N, N + -O - 、NR 4 、CR 4 Or C-L-R 9
R 4 Is hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
y is N, N + -O - 、NR 5 、CR 5 Or C-L-R 9
R 5 Is hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
R 6 is hydrogen or C 1-3 Aliphatic series; or (b)
R 4 And R is 6 Optionally together with the intervening atoms thereof form a 5-to 6-membered partially unsaturated fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, optionally with R 7 U instance substitutions of (2);
each R 7 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
each R 8 Independently hydrogen, oxo, halogen, -CN, -NO 2 、-CHF 2 、-CF 3 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or optionally substituted C 1-6 Aliphatic series;
l is a covalent bond; or L is C 1-4 A divalent saturated or unsaturated linear or branched hydrocarbon chain, wherein one or two methylene units in the chain are optionally and independently replaced by: -C (R) 2 -、-C(OR)(R)-、-N(R)-、-O-、-S-、-S(O)-、-S(O) 2 -、-S(O)N(R)-、-N(R)S(O)-、-S(O) 2 N(R)-、-N(R)S(O) 2 -C (O) -, -C (O) O-, -OC (O) -, -C (O) N (R) -, -N (R) C (O) -, -C (O) N (R) O-, -ON (R) C (O) -, -OC (O) N (R) -, -N (R) C (O) O-, or-N (R) C (O) N (R) -;
R 9 Is halogen, -CN, -NO 2 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-CF 2 R、-CF 3 、-C(R) 2 OR、-C(R) 2 N(R) 2 、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-C(S)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R, -N three S (O) (R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-Si(OR)(R) 2 、-Si(R) 3 、-P(O)(R)N(R) 2 -P (O) (R) OR OR-P (O) (R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or R is 9 Is an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 4-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spirobicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; and 8-10 membered partially aromatic having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfurA bicyclic heterocyclic ring of either the family or the heteroaromatic group;
R 1 、R 2 、R 3 、R 4 、R 5 、R 6 、R 7 、R 8 and R is 9 Each instance independently and optionally by R A Wherein each RA is independently oxo, halogen, -CN, -NO 2 、-OR、-SR、-N(R) 2 、-S(O) 2 R、-S(O) 2 N(R) 2 、-S(O)R、-S(O)N(R) 2 、-C(R) 2 OR、-C(O)R、-C(O)OR、-C(O)N(R) 2 、-C(O)N(R)OR、-OC(O)R、-OC(O)N(R) 2 、-N(R)C(O)OR、-N(R)C(O)R、-N(R)C(O)N(R) 2 、-N(R)C(NR)N(R) 2 、-N(R)N(R) 2 、-N(R)S(O) 2 N(R) 2 、-N(R)S(O) 2 R、-N=S(O)(R) 2 、-S(NR)(O)R、-N(R)S(O)R、-N(R)CN、-P(O)(R)N(R) 2 、-P(O)(R)OR、-P(O)(R) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl group; a naphthyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorus, silicon and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spiro ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; and a 6-11 membered saturated or partially unsaturated bicyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur;
Each R is independently hydrogen, -CN, halogen, or an optionally substituted group selected from: c (C) 1-6 Aliphatic series; a phenyl group; a naphthyl group; 3-7 membered saturated or partially unsaturated monocyclic carbocycle; a 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; having 1 to 4 groups independently selected from nitrogen,An 8-10 membered bicyclic heteroaryl ring of heteroatoms of oxygen and sulfur; a 7-12 membered saturated or partially unsaturated bicyclic heterocycle having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 5-8 membered saturated or partially unsaturated bridged bicyclic ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-10 membered saturated or partially unsaturated spiro ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur; a 6-11 membered saturated or partially unsaturated bicyclic carbocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur; or:
two R groups on the same atom optionally together with the atoms form an optionally substituted 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring; a 3-7 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur;
m is 0, 1, 2, 3, 4 or 5;
n is 0, 1, 2, 3 or 4;
p is 0, 1, 2, 3 or 4;
q is 0, 1, 2 or 3;
t is 0 or 1;
u is 0, 1, 2, 3 or 4; and is also provided with
Each instance of v is independently 0, 1, 2, 3, 4, or 5; and is also provided with
Wherein the method comprises the steps ofRepresents a single bond or a double bond.
2. The compound of claim 1, wherein ring B and R thereof 2 Substituents taken together are selected from
3. The compound according to claim 1 or 2, wherein ring B and R thereof 2 Substituents taken together are selected from
4. The compound of claim 1, wherein the compound has the formula II-a to II-eee:
OR a pharmaceutically acceptable salt thereof, wherein-OR, -NR-OR-N (R) 2 The R group of (2) is not hydrogen, -CN or halogen.
5. The compound of claim 1, wherein the compound has the formula II-k-a to II-eee-a:
/>
/>
/>
or a pharmaceutically acceptable salt thereof.
6. The compound of claim 1, wherein the compound has the formula II-a to II' -VV:
/>
/>
/>
/>
/>
or a pharmaceutically acceptable salt thereof.
7. The compound of claim 1, wherein the compound has the formula II '-ww, II' -xx, II '-yy, II' -zz, II '-aaa, or II' -bbb:
/>
/>
or a pharmaceutically acceptable salt thereof.
8. The compound of claim 1, wherein the compound has the formula III-a to III-n:
/>
/>
Or a pharmaceutically acceptable salt thereof.
9. The compound of claim 1, wherein the compound has the formula IV-a to IV-W:
/>
/>
/>
or a pharmaceutically acceptable salt thereof.
10. A compound selected from the compounds depicted in table 1, or a pharmaceutically acceptable salt thereof.
11. A pharmaceutical composition comprising a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant or vehicle.
12. A compound according to any one of claims 1 to 10 or a pharmaceutical composition according to claim 11 for use as a medicament.
13. A method of inhibiting Cbl-b in a biological sample, the method comprising contacting the sample with a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 11.
14. A method of treating a Cbl-b mediated disorder, disease or condition in a patient, the method comprising administering to the patient a compound according to any one of claims 1 to 10 or a pharmaceutically acceptable salt thereof or a pharmaceutical composition according to claim 11.
15. The method of claim 14, wherein the Cbl-b mediated disorder is hematological cancer.
16. The method of claim 15, wherein the hematologic cancer is B-cell acute lymphoblastic leukemia (BALL), T-cell acute lymphoblastic leukemia (tal), acute Lymphoblastic Leukemia (ALL), chronic leukemia, hematologic cancer, or a hematologic condition selected from the group consisting of: b cell prolymphocytic leukemia, blast plasmacytoid dendritic cell neoplasm, burkitt's lymphoma (Burkitfs lymphoma), diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small or large cell follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma, marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin's lymphoma, plasmablasts lymphoma, plasmacytoid dendritic cell neoplasm, waldenstrom macroglobulinemia (Waldenstrom's macroglobulinemia), and pre-leukemia.
17. The method of claim 14, wherein the Cbl-b mediated disorder is bone cancer, pancreatic cancer, skin cancer, head and neck cancer, skin or intraocular malignant melanoma, uterine cancer, ovarian cancer, rectal cancer, anal region cancer, gastric cancer, testicular cancer, uterine cancer, fallopian tube cancer, endometrial cancer (carcinoma ofthe endometrium), endometrial cancer (endometrial cancer), cervical cancer, vaginal cancer, vulval cancer, hodgkin's Disease, non-Hodgkin's lymphoma, esophageal cancer, small intestine cancer, endocrine system cancer, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, urinary tract cancer, penile cancer, a chronic or acute leukemia selected from acute myeloid leukemia, chronic myeloid leukemia, acute lymphocytic leukemia and chronic lymphocytic leukemia, pediatric solid tumor, lymphocytic lymphoma, bladder cancer, renal cancer or urinary tract cancer, renal pelvis cancer, neoplasms of the Central Nervous System (CNS), primary CNS lymphoma, tumor angiogenesis, spinal axis tumor, brain stem glioma, kaposi's sarcoma, cancer of the human face, cancer of the epidermis cell, cancer of the human skin, cancer of the human cell system, cancer of the ear cell, or a combination of cancer.
18. Use of a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 11, in the manufacture of a medicament for treating a Cbl-b mediated disorder, disease or condition in a patient.
19. The use of claim 18, wherein the Cbl-b mediated disorder is hematological cancer.
20. The use of claim 19, wherein the hematologic cancer is B-cell acute lymphoblastic leukemia (BALL), T-cell acute lymphoblastic leukemia (tal), acute Lymphoblastic Leukemia (ALL), chronic leukemia, hematologic cancer, or a hematologic condition selected from the group consisting of: b cell prolymphocytic leukemia, blast plasmacytoid dendritic cell neoplasm, burkitt's lymphoma, diffuse large B cell lymphoma, follicular lymphoma, hairy cell leukemia, small or large cell follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma, marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-hodgkin's lymphoma, plasmablasts lymphoma, plasmacytoid dendritic cell neoplasm, waldenstrom macroglobulinemia and pre-leukemia.
21. The use of claim 18, wherein the Cbl-b mediated disorder is bone cancer, pancreatic cancer, skin cancer, head and neck cancer, cutaneous or intraocular malignant melanoma, uterine cancer, ovarian cancer, rectal cancer, anal region cancer, gastric cancer, testicular cancer, uterine cancer, fallopian tube cancer, endometrial cancer (carcinoma ofthe endometrium), endometrial cancer (endometrial cancer), cervical cancer, vaginal cancer, vulvar cancer, hodgkin's disease, non-hodgkin's lymphoma, esophageal cancer, small intestine cancer, cancer of the endocrine system, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, urinary tract cancer, penile cancer, chronic myeloid leukemia, chronic or acute leukemia selected from acute myeloid leukemia and chronic lymphocytic leukemia, pediatric solid tumors, lymphocytic lymphomas, bladder cancer, kidney or renal cancer, renal pelvis cancer, neoplasm of the Central Nervous System (CNS), primary CNS lymphoma, tumor angiogenesis, spinal cord shaft tumors, stem glioma, pituitary adenoma, kaposi's lymphoma, epidermosarcoma, T cell sarcoma, squamous cell carcinoma, cancer of the environment-induced cancer or a combination of said cancers.
CN202280040238.XA 2021-04-09 2022-04-08 CBL-B modulators and uses thereof Pending CN117858707A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/173,121 2021-04-09
US202163264940P 2021-12-03 2021-12-03
US63/264,940 2021-12-03
PCT/US2022/071633 WO2022217276A1 (en) 2021-04-09 2022-04-08 Cbl-b modulators and uses thereof

Publications (1)

Publication Number Publication Date
CN117858707A true CN117858707A (en) 2024-04-09

Family

ID=90543820

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280040238.XA Pending CN117858707A (en) 2021-04-09 2022-04-08 CBL-B modulators and uses thereof

Country Status (1)

Country Link
CN (1) CN117858707A (en)

Similar Documents

Publication Publication Date Title
US11779578B2 (en) IRAK degraders and uses thereof
US20230089916A1 (en) Irak degraders and uses thereof
AU2020302118A1 (en) IRAK degraders and uses thereof
WO2020010210A1 (en) Mertk degraders and uses thereof
WO2020251971A1 (en) Smarca degraders and uses thereof
WO2020251972A1 (en) Smarca degraders and uses thereof
WO2020206424A1 (en) Stat degraders and uses thereof
WO2020251969A1 (en) Smarca degraders and uses thereof
EP4076524A2 (en) Irak degraders and uses thereof
JP2024514847A (en) CBL-B Modulators and Uses Thereof
JP2024522827A (en) CBL-B regulators and uses thereof
WO2023044046A1 (en) Bcl-xl degraders and uses thereof
AU2020412780A1 (en) SMARCA degraders and uses thereof
EP4259144A1 (en) Smarca degraders and uses thereof
WO2021119159A1 (en) Irak degraders and uses thereof
CN117295737A (en) HPK1 antagonists and uses thereof
CN117120090A (en) HPK1 antagonists and uses thereof
CN117858865A (en) CBL-B modulators and uses thereof
US20230416242A1 (en) Double degraders and uses thereof
CN117858707A (en) CBL-B modulators and uses thereof
WO2023192586A1 (en) Irak degraders and uses thereof
WO2023220425A1 (en) Bcl-xl/bcl-2 degraders and uses thereof
CN117377673A (en) HPK1 antagonists and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination