NZ755543A - Histone deacetylase inhibitors - Google Patents

Histone deacetylase inhibitors

Info

Publication number
NZ755543A
NZ755543A NZ755543A NZ75554317A NZ755543A NZ 755543 A NZ755543 A NZ 755543A NZ 755543 A NZ755543 A NZ 755543A NZ 75554317 A NZ75554317 A NZ 75554317A NZ 755543 A NZ755543 A NZ 755543A
Authority
NZ
New Zealand
Prior art keywords
compound
reaction
reaction mixture
stirred
added
Prior art date
Application number
NZ755543A
Inventor
Shripad Bhagwat
Gregory Luedtke
Original Assignee
Biomarin Pharmaceuticals Inc
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 of NZ755543A publication Critical patent/NZ755543A/en
Application filed by Biomarin Pharmaceuticals Inc filed Critical Biomarin Pharmaceuticals Inc

Links

Abstract

Provided herein are compounds and methods for inhibiting histone deacetylase ("HDAC") enzymes (e.g., HDAC1, HDAC2, and HDAC3).

Description

HISTONE DEACETYLASE INHIBITORS TECHNICAL FIELD Provided herein are compounds and methods of inhibiting histone ylase (“HDAC”) enzymes (e.g., HDACt, HDACZ, and HDACB).
BACKGROUND To date, 18 HDAC s have been identified in humans and there is increasing evidence that the 18 HDAC enzymes in humans are not redundant in function.
HDAC enzymes are classified into three main groups based on their homology to yeast proteins. Class | includes HDAC1, HDACZ, HDACB, and HDACB and have homology to yeast RPD3. HDAC4, HDACE, HDAC7, and HDACQ belong to class Na and have gy to yeast HDACt. HDACS and HDACt 0 contain two catalytic sites and are classified as class llb, whereas HDACt 1 has conserved residues in its catalytic center that are shared by both class I and class II deacetylases and is placed in class IV. These HDAC enzymes contain zinc in their catalytic site and are inhibited by compounds like trichostatin A (TSA) and stat [suberoylanilide hydroxamic acid (SAHA)]. Class III HDAC enzymes are known as sirtuins. They have homology to yeast Sir2, require NAD+ as cofactor, and do not contain zinc in the catalytic site. In general, HDAC inhibitors of zinc-dependent HDAC enzymes include a Zn-binding group, as well as a surface recognition domain.
HDAC enzymes are involved in the tion of a number of cellular ses.
Histone acetyltransferases (HATs) and HDAC s acetylate and deacetylate lysine residues on the N termini of histone proteins thereby affecting transcriptional activity. They have also been shown to regulate post—translational acetylation of at least 50 non—histone proteins such as oc-tubulin (see for example Kahn, N et a/ Biochem J 409 (2008) 581, Dokmanovic, M., eta/Moi Cancer Res 5 (2007) 981).
Altering gene expression through chromatin modification can be accomplished by inhibiting HDAC s. There is evidence that histone acetylation and deacetylation are mechanisms by which transcriptional regulation in a cell — a major event in cell entiation, proliferation, and apoptosis — is achieved. It has been hypothesized that these effects occur h changes in the structure of chromatin by altering the affinity of histone ns for coiled DNA in the nucleosome. Hypoacetylation of histone proteins is believed to increase the interaction of the histone with the DNA phosphate backbone.
Tighter binding between the histone n and DNA can render the DNA inaccessible to transcriptional regulatory ts and machinery. HDAC enzymes have been shown to catalyze the removal of acetyl groups from the s-amino groups of lysine residues present within the N-terminal extension of core histones, thereby g to hypoacetylation of the histones and blocking of the riptional machinery and regulatory elements.
W0 2018/1 19362 Inhibition of HDAC, therefore can lead to histone deacetylase—mediated transcriptional derepression of tumor suppressor genes. For example, cells treated in culture with HDAC tors have shown a consistent induction of the kinase inhibitor p21, which plays an important role in cell cycle arrest. HDAC inhibitors are thought to increase the rate of transcription of p21 by propagating the hyperacetylated state of histones in the region of the p21 gene, thereby making the gene accessible to transcriptional machinery.
Further, non-histone proteins involved in the regulation of cell death and cell-cycle also undergo lysine acetylation and deacetylation by HDAC enzymes and histone acetyl transferase (HATS).
This ce supports the use of HDAC inhibitors in ng various types of cancers. For e, vorinostat (suberoylanilide hydroxamic acid (SAHA)) has been approved by the FDA to treat cutaneous T-cell lymphoma and is being investigated for the treatment of solid and logical tumors. Further, other HDAC inhibitors are in pment for the ent of acute myelogenous leukemia, Hodgkin’s disease, myelodysplastic syndromes and solid tumor cancers.
HDAC inhibitors have also been shown to inhibit pro—inflammatory cytokines, such as those involved in autoimmune and matory disorders (e.g. TNF—oc). For example, the HDAC tor M8275 was shown to slow e progression and joint destruction in collagen-induced arthritis in rat and mouse models. Other HDAC inhibitors have been shown to have efficacy in treating or ameliorating inflammatory disorders or ions in in vivo models or tests for disorders such as Crohn’s disease, colitis, and airway inflammation and hyper-responsiveness. HDAC inhibitors have also been shown to ameliorate spinal cord inflammation, demyelination, and al and axonal loss in experimental autoimmune alomyelitis (see for example Wanf, L., etal, Nat Rev Drug Disc 8 (2009) 969).
Triplet repeat expansion in genomic DNA is associated with many neurological conditions (e.g., neurodegenerative and neuromuscular es) ing myotonic dystrophy, spinal muscular atrophy, fragile X syndrome, Huntington’s disease, spinocerebellar ataxias, amyotrophic lateral sclerosis, Kennedy’s disease, spinal and bulbar muscular atrophy, Friedreich’s ataxia and Alzheimer’s disease. Triplet repeat ion may cause disease by altering gene expression. For example, in Huntington‘s disease, spinocerebellar ataxias, fragile X syndrome, and myotonic dystrophy, expanded repeats lead to gene silencing. ln Friedreich’s ataxia, the DNA abnormality found in 98% of FRDA patients is an unstable hyper-expansion of a GAA triplet repeat in the first intron of the frataxin gene (see Campuzano, et al., Science 271 :1423 (1996)), which leads to frataxin insufficiency resulting in a progressive erebellar neurodegeneration. Since they can affect transcription and potentially correct transcriptional dysregulation, HDAC inhibitors have been tested and have been shown to positively affect neurodegenerative diseases (see Herman, D., et al, Nat Chem Bio 2 551 (2006) for Friedreich’s ataxia, Thomas, E.A., et al, Proc Natl Acad Sci USA 105 15564 (2008) for Huntington’s disease).
HDAC inhibitors may also play a role in ion-related conditions and diseases.
It has indeed become increasingly evident that transcription is likely a key element for long- term memory processes (Alberini, C.M., Physiol Rev 89 121 (2009)) thus highlighting r role for netrant HDAC inhibitors. Although studies have shown that treatment with non—specific HDAC inhibitors such as sodium butyrate can lead to long—term memory formation (Stefanko, D.P., et al, Proc Natl Acad Sci USA 106 9447 (2009)), little is known about the role of specific isoforms. A limited number of studies have shown that, within class I HDAC enzymes, main target of sodium butyrate, the prototypical inhibitor used in cognition studies, HDAC2 (Guan, J-S., etal, Nature 459 55 (2009)) and HDAC3 wn, S.C., etal, J Neurosci 31 764 (2011)) have been shown to regulate memory processes and as such are interesting targets for memory enhancement or extinction in -affecting conditions such as, but not limited to, Alzheimer’s disease, post-traumatic stress disorder or drug addiction.
HDAC inhibitors may also be useful to treat ious e such as viral infections. For example, treatment of HIV infected cells with HDAC inhibitors and anti- iral drugs can eradicate virus from treated cells (Blazkova, J., et a/J Infect Dis. 2012 Sep 1 ;206(5):765-9; Archin, N. M, eta/Nature 2012 Jul 25, 487(7408) .4825) ”QED Some prior disclosed HDAC inhibitors include a moiety of NH2 ,which can metabolize under physiological ions to provide a metabolite OPD — phenylenediamine) ;H2 .OPD is a toxic material. Thus, the need exists for HDAC «,gOLNHQ inhibitors sing a moiety of NH2 w,hich, under physiological conditions, produce lower amounts, or substantially no amounts, of CPD.
SUMMARY Provided herein are compounds of formula (I), or a pharmaceutically acceptable salt thereof, and methods of using compounds of formula (I), e.g., for inhibiting HDAC (e.g., W0 20182’119362 18 one or more of HDAC1, HDAC2, and HDAC3): 1 4 R2 (I), wherein ring A is a 4-7 membered monocyclic heterocycloalkyl ring or a 7-12 membered spiro heterocycloalkyl ring, wherein ring A contains one nitrogen ring atom and optionally contains one additional ring atom independently selected from O, N, and S; R1 is H, C1. 6alkyl, C2_6alkenyl, C1_6hydroxyalky|, C(O)C1_6alkyl, C0.3alkylene-C3.1ocycloalkyl, or C0. ene-C2_5heterocycloa|ky| having 1 or 2 heteroatoms selected from O, S, N, and MG. 4alkyl); R2 is H, F, Cl, or CH3; R3 is C1_3alkyl; R4 is H, F, or Cl; and n is 0, 1, or 2, with the proviso that (a) ring A is not morpholino or thiomorpholino; and (b) when ring A is piperazinyl, R1 is C2_6alkenyl, C1_6hydroxyalkyl, C(O)C1_6alkyl, C0_3alkylene—C3_1ocycloalkyl, or C0_3a|kylene-C2_5cycloheteroalkyl having 1 or 2 heteroatoms selected from O, S, N, and MG. 4a|kyl).
Also provided herein are pharmaceutical compositions comprising a compound as disclosed herein, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. r provided are s of using the compounds as disclosed herein to inhibit HDAC (e.g., one or more of HDAC1, HDAC2, and HDAC3) and methods of treating conditions associated with aberrant HDAC ty by administering a compound sed herein to a subject suffering from such a condition.
DETAILED DESCRIPTION Provided herein are compounds of formula (I), pharmaceutical compositions thereof, and methods of using compounds of formula (I), e.g., for inhibiting HDAC (e.g., one or more of HDAC1, HDAC2, and HDAC3): R1 (R3)n a O ”QNH2 R2 (I), wherein ring A, R‘, R2, R3, and R4 are defined herein.
The compounds provided herein are capable of forming low amounts of CPD under physiological ions (e.g., a pH of about 7.2 and 37 °C). Physiological conditions as disclosed herein are intended to include a ature of about 35 to 40 °C, and a pH of about 7.0 to about 7.4 and more lly include a pH of 7.2 to 7.4 and a temperature of 36 to 38° C in an aqueous environment. By “low amounts” of CPD, as used herein, it is W0 20182’119362 18 ed to mean that the nds disclosed herein generate OPD under physiological conditions for 24 hours at an amount of 30% or less. In some embodiments, the amount of CPD generated at physiological conditions for 24 hours is 25% or less, or 20% or less, or % or less, or 10% or less, or 5% or less, or 1% or less. The amount of CPD generated can be measured indirectly by measuring the amount of resulting acid from the amide hydrolysis of the compound. In some embodiments, the measurement of CPD generated can be performed by administration of the compound as sed herein to a subject, collection of plasma samples over 24 hours, and determining the amount of GDP and/or the relevant acid over that 24 hours.
Definitions The following definitions are used, unless ise described. Specific and l values listed below for radicals, substituents, and ranges, are for illustration only; they do not exclude other defined values or other values within defined ranges for the radicals and substituents.
As used herein, the term “about” preceeding a numerical value refers to a range of values i10% of the vlaue specified.
As used herein, the term ”acceptable” with respect to a ation, composition, or ingredient, means no persistent detrimental effect on the general health of the subject being treated.
As used herein, the term “alkyl,” employed alone or in combination with other terms, refers to a saturated hydrocarbon group that may be straight-chain or branched. In some embodiments, the alkyl group contains 1 to 12, 1 to 8, or 1 to 6 carbon atoms. In certain ments, alkyl includes 1-6 carbon atoms (“C15 alkyl”). In certain embodiments, alkyl includes 1-4 carbon atoms (“C14 alkyl”). In certain ments alkyl includes 1-3 carbon atoms (“C1.3 alkyl”).
As used herein, the term ”alkylene” employed alone or in ation with other terms, refers to a divalent radical formed by removal of a hydrogen atom from alkyl. In some embodiments, the ne group contains 1-3 carbon atoms.
In some ments, alkyl includes methyl, ethyl, yl, isopropyl, n-butyl, tert-butyl, isobutyl, sec-butyl; higher homologs such as 2-methyI-1—butyl, n—pentyl, 3-pentyl, n-hexyl, 1,2,2-trimethylpropyl, n-heptyl, n-octyl, and the like. In some embodiments, the alkyl moiety is methyl, ethyl, n-propyl, isopropyl, n—butyl, isobutyl, utyl, n-pentyl, isopentyl, neopentyl, n-hexyl, or 2,4,4-trimethylpentyl.
As used herein, the term “alkenyl,” employed alone or in combination with other terms, refers to a saturated hydrocarbon group with at least one double bond that may be straight-chain or ed. In some embodiments, the l group contains 2 to 12, 2 to 8, or 2 to 6 carbon atoms. In certain ments, alkenyl e ethenyl, propenyl, 2- methylprop-I-enyl, 1-butenyl, 1-pentenyl, or 1-hexenyl. In certain embodiments alkyl includes 2-6 carbon atoms (“02.6 alkenyl”).
As used herein, the term “cycloalkyl,” employed alone or in combination with other terms, refers to a saturated, cyclic arbon moiety of 3 to 10 carbon atoms. Cycloalkyl includes saturated or partially unsaturated rings, but does not contain an aromatic ring. In certain embodiments, cycloalkyl include a saturated, monocyclic or bicyclic hydrocarbon moiety of 3 to 10 carbon atoms. When a cycloalkyl group ns from 3—10 carbon atoms, it may be referred to herein as ycloalkyl. In some embodiments, the lkyl group contains 3 to 7, or 3 to 6 carbon ring atoms. In some embodiments, cycloalkyl groups include cyclopropyl, cyclobutyl, entyl, cyclohexyl, and cycloheptyl. In some embodiments, cycloalkyl includes cyclopropyl, cyclopentyl, and cyclohexyl. In some embodiments, cycloalkyl es cyclopropyl; or it includes cyclopentyl; or it includes cyclohexyl; or it includes adamantyl.
As used herein, the term “heterocycloalkyl” employed alone or in combination with other terms, refers to a saturated ring system, which has carbon ring atoms and at least one heteroatom ring atom selected from nitrogen, sulfur, and oxygen (independently selected when more than one is present), unless specified otherwise. Heterocycloalkyl includes saturated or partially unsaturated rings, but does not contain an aromatic ring.
Heterocycloalkyl can include fused, bridged and spiro rings. When the heterocycloalkyl group contains more than one heteroatom, the heteroatoms may be the same or ent.
Heterocycloalkyl groups can include mono- or bicyclic (e.g., having 2 fused rings) ring systems. For example, a fused heterocycloalkyl group may comprise two rings that share adjacent atoms (e.g., one covalent bond). Heterocycloalkyl groups can also include bridgehead heterocycloalkyl groups. As used herein, “bridgehead heterocycloalkyl group” refers to a heterocycloalkyl moiety containing at least one head heteroatom (e.g., nitrogen or carbon). The moiety “02.5heterocycloalkyl” and the like refer to heterocycloalkyl rings having at least 2 to 5 ring carbon atoms in addition to at least 1 heteroatom. For example, a CZ heterocycloalkyl can be a three-membered ring with 1 heteroatom in the ring and 2 carbon ring atoms, or a four-membered ring, where there are 2 carbon ring atoms and 2 heteroatoms in the ring, or a five-membered ring, where there are 2 carbon ring atoms and 3 atoms in the ring.
In certain embodiments, heterocycloalkyl includes a monocyclic ring of 4 to 7 ring atoms. In certain embodiments, cycloalkyl includes a spiro ring system of 7 to 12 ring atoms. In certain embodiments, heterocycloalkyl includes 1, 2, or 3 nitrogen ring atoms; or 1 or 2 nitrogen ring atoms; 2 en ring atoms; or 1 nitrogen ring atom. In certain embodiments, heterocycloalkyl es 1 nitrogen ring atom and 1 oxygen or sulfur ring atom.
In certain embodiments, heterocycloalkyl includes azetidinyl, pyrrolidinyl, 2,5- dihydro-1H- pyrrolinyl, 2,5-dihydro-1H—pyrrolyl, piperidinyl, zinyl, pyranyl, ydropyranyl, tetrahydrothiopyranyl, 1,3-dioxinyl, 1,3-dioxanyl, 1,4-dioxinyl, 1,4-dioxanyl, roazepinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, dihydropyridinyl, tetrahydropyridinyl, oxazolinyl, oxazolidinyl, isoxazolidinyl, thiazolinyl, thiazolidinyl, quinuclidinyl, isothiazolidinyl, octahydroindolyl, droisoindolyl, decahydroisoquinolyl, ydrofuryl, 2—azaspiro[3.3]heptanyl, 7-azabicyclo[2.2.1]heptanyl, and 8-azabicyclo[3.2.1]octanyl. In some embodiments, the heterocycloalkyl comprises dinyl, piperazinyl, azetidinyl, azepanyl, or diazepanyl, e.g., piperidinyl or zinyl. In some embodiments, the heterocycloalkyl comprises piperidinyl, piperazinyl, azetidinyl, azepanyl, pyrrolidinyl or diazepanyl. Specifically contemplated spiro heterocycloalkyl groups e azetidinyl ring spiro fused to another azetidinyl ring or a piperidinyl ring, or a piperazinyl ring, and an oxetanyl ring spiro fused to an inyl ring or a piperidinyl ring or a zinyl ring, or a cyclohexyl ring spiro fused to an azetidinyl ring or a piperidinyl ring or a piperazinyl ring.
As used herein, the term “hydroxyalkyl” and the like employed alone or in combination with other terms, refers to an alkyl group having at least one hydroxy group. . In certain embodiments, hydroxyalkyl refers to an alkyl group having 1 hydroxy group. In n embodiments, yalkyl refers to an alkyl group having 1, 2, or 3 y group.
The term “subject” refers to a mammal, such as a mouse, guinea pig, rat, dog, or human. In certain embodiments, mammal e sheep, goat, horse, cat, rabbit, monkey, or cow. The terms “subject” and “patient” are used interchangeably. In certain ments, the subject is a human; or the subject is a human adult; or the subject is a human child.
“Treat,” “treating,” and “treatment,” in the context of treating a disease or disorder, are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or to slowing the progression, spread or worsening of a disease, disorder or condition or of one or more symptoms thereof. Often, the beneficial effects that a subject derives from a therapeutic agent do not result in a complete cure of the disease, disorder or condition.
Although methods and materials similar or equivalent to those described herein can be used in practice or testing, suitable methods and materials are described below. All publications, patent applications, patents, and other references ned herein are incorporated by reference in their entirety. In embodiment of conflict, the present specification, including definitions, will control.
Com ounds of formula I nds of a (I) are provided herein: 1 (R3) R2 (I), ring A is a 4—7 membered clic heterocycloalkyl ring or a 7-12 membered spiro heterocycloalkyl ring, wherein ring A contains one nitrogen ring atom and optionally contains one additional ring atom independently selected from O, N, and S; R1 is H, C1_6alkyl, C2. salkenyl, C1_6hydroxyalkyl, C(O)C1_6alkyl, C0_3alkylene-C3_10cycloalkyl, or C0_3alkylene-C2_ 5heterocycloalky| having 1 or 2 heteroatoms selected from O, S, N, and N(C1_4alkyl); R2 is H, F, CI, or CH3; R3 is C1_3alky|; R4 is H, F, or Cl; and n is 0, 1, or 2, with the proviso that (a) ring A is not morpholino or thiomorpholino; and (b) when ring A is piperazinyl, Ft1 is C2_6alkenyl, C1_6hydroxyalkyl, C(O)C1_6alkyl, C0_3alkylene-C3_1ocycloalkyl, or C0_3alkylene-C2_ 5cycloheteroalkyl having 1 or 2 heteroatoms selected from O, S, N, and N(C1_4alkyl). In some embodiments, R1 is H, C1_6alkyl, C3_6hydroxyalkyl, C3_6alkenyl, or C1_2alkylene-C3_ 10cycloalkyl; R2 is H; R3, if present, is CH3, and R4 is H. In certain embodiments, R1 is C1- , C3_6hydroxyalkyl, or C1.2alkylene-C3_10cycloalkyl; R2 is H; R3, if present, is CH3, and R4 is F.
In some some cases, the compound of formula (I) has the following characteristics: ring A is dinyl, azetidinyl, azepanyl, diazepanyl, pyrrolidinyl, 7;, N); N , >'Il'; R1 is H, C1_6alkyl, C2_6alkenyl, C1_6hydroxyalkyl, C(O)C1_ salkyl, C0_3alkylene—C3_1ocycloalkyl, or C0_3alkylene—C2_5heterocycloalkyl having 1 or 2 heteroatoms selected from O, S, N, and N(C1_4alkyl); R2 is H, F, Cl, or CH3; R3 is kyl; R4 is H, F, or Cl; and n is 0, 1, or 2.
In some some cases, the compound of a (I) has the following characteristics: ring A is piperidinyl, azetidinyl, azepanyl, diazepanyl, pyrrolidinyl, 7!, 0% NED N N Y or >"I; R1 is H, C1_6alkyl, kenyl, C1_6hydroxyalkyl, or C0. , 3alkylene-C3_1ocycloalkyl; R2 is H; R3 is C1_3alkyl; R4 is H or F; and n is 0, 1, or 2.
In some cases, the compound of a (I) has the following characteristics: ring A is piperazinyl; R1 is Czealkenyl, C1_6hydroxyalkyl, or C0.3alkylene-C3_1ocycloalkyl; R2 is H, F, Cl, or CH3; R3 is C1_3alkyl; R4 is H or F; and n is 0, 1, or 2. In some cases, the compound of formula (I) has the following characteristics: ring A is piperazinyl; R1 is C1_6hydroxyalkyl or C0. 3alkylene-C3_1ocycloalkyl; R2 is H; R3 is C1_3alkyl; R4 is H; and n is 0, 1, or 2.
In various embodiments, ring A is a 4-7 ed monocyclic heterocycloalkyl ring or a 7-12 ed spiro heterocycloalkyl ring, wherein ring A contains one nitrogen ring atom and optionally contains one additional ring atom independently selected from O, N, and S. In various embodiments, ring A is a 4-7 membered monocyclic heterocycloalkyl ring or a 7-12 membered spiro heterocycloalkyl ring, wherein ring A contains one nitrogen ring atom and ally contains one additional nitrogen ring atom. In various embodiments, ring A is a 4-7 membered monocyclic heterocycloalkyl ring or a 7-12 membered spiro heterocycloalkyl ring, wherein ring A contains one nitrogen ring atom and optionally contains one oxygen ring atom. In various embodiments, ring A is a 4-7 membered monocyclic cycloalkyl ring or a 7-12 membered spiro cycloalkyl ring, wherein ring A contains one nitrogen ring atom and optionally contains one sulfur ring atom. In various embodiments, ring A is a 7—1 2 membered spiro heterocycloalkyl ring containing one or two nitrogen ring atoms or one nitrogen ring atom and one oxygen ring atom. In various embodiments, ring A is a 7-12 membered spiro heterocycloalkyl ring containing one or two nitrogen ring atoms. In various embodiments, ring A is a 7-12 ed spiro heterocycloalkyl ring containing one nitrogen ring atom. In various embodiments, ring A is a 7-12 membered spiro heterocycloalkyl ring containing two nitrogen ring atoms. In various embodiments, ring A is a 7—1 2 membered spiro heterocycloalkyl ring containing one nitrogen ring atom and one oxygen ring atom. In various cases, ring A is a 4-7 membered monocyclic heterocycloalkyl ring ning one or two nitrogen ring atoms. In various cases, ring A is a 4-7 membered monocyclic heterocycloalkyl ring containing one nitrogen ring atom. In various cases, ring A is a 4-7 ed monocyclic heterocycloalkyl ring ning two nitrogen ring atoms. Some specific ring A moieties contemplated include piperidinyl, piperazinyl, azetidinyl, yl, and diazepanyl. Some specific ring A moieties contemplated e piperidinyl, piperazinyl, azetidinyl, yl, diazepanyl, and pyrrolidinyl. In certain embodiments, ring A is piperidinyl, piperazinyl, or azetidinyl. In certain embodiments, ring A is piperidinyl, piperazinyl, azetidinyl, or pyrrolidinyl. In certain embodiments, ring A is piperidinyl, pyrrolidinyl, or azetidinyl. In certain embodiments, ring A is azetidinyl, azepanyl, or diazepanyl. In n embodiments, ring A is azepanyl or diazepanyl. In certain embodiments, ring A is piperidinyl. In n ments, ring A is piperazinyl. In certain embodiments, ring A is azetidinyl. In certain embodiments, ring A is azepanyl. In certain embodiments, ring A is diazepanyl. In certain embodiments, ring A is pyrrolidinyl. Some ic spiro ring A moieties contemplated include azetidinyl ring spiro fused to another azetidinyl ring or a piperidinyl ring, or a piperazinyl ring, and an oxetanyl ring spiro fused to an inyl ring or a piperidinyl ring or a piperazinyl ring, or a cyclohexyl ring spiro fused to an azetidinyl ring or a piperidinyl ring or a piperazinyl ring. In some cases, ring A can be piperidinyl or piperazinyl. In various cases, ring A is selected from the group consisting of: R‘ can be H, C1_ealkyl, Czealkenyl, C1_6hydroxyalkyl, C(O)C1_6alkyl, CO_3alkylene—Cg_ 1Ocycloalkyl, or CO_3aIkylene-Cz_5heterocycloalkyl having 1 or 2 heteroatoms selected from O, S, N, and N(C1_4alkyl). In some cases, R1 is H. In some cases, R‘ is C1_5alkyl (e.g., methyl, isopropyl, sec-butyl, or CH20(CH3)3). In some cases, R1 is methyl, ethyl, , isopropyl, butyl, sec-butyl, or CHZC(CH3)3). In some cases, R1 is methyl or neopentyl. In some cases R1 is methyl. In some cases R1 is neopentyl. In some cases, R1 is droxyalkyl (e.g., HO><\/). In some cases R1 is HOW. In some cases, R1 is Cs.1ocycl03'ky| or C1. salkylene-Cg_1ocycloalkyl, e.g., the cycloalkyl group is cyclopropyl or chycloalkyl, Le, 2017/0681 18 adamantyl. In some cases, R1 is kylene-C3.1ocycloalkyl, e.g.W or In some cases R1 is3 . or g . In some cases R1 is W. In some cases R1 is . In certain embodiments, R1 is Czealkenyl. In certain embodiments, R1 is M.
In some cases, the compound of formula (I) has the following characteristics: R1 R10y Rag 3 is selected from the group consisting of R R\ R1 O NI U I RE RIN N N N N k 7; f \a’; Y Y and R1 \JN is selected from the group consisting of H, CH3, W, ><\/ HOW, mg and M; R2 is H, F, CI, or CH3; R3 is CH3, R4 is H or F; andnisO,1,or2.
In some cases, the compound of formula (I) has the following characteristics: is selected from the group consisting of (>ny UROI R1 1{DO HE b N q/ , y, ,and R1 \JN is selected from the group consisting of H, CH3, W, XV HOW, Hg ,andM;R2isH;R3isCH3;R4isHorF;andniso,1, or 2. In certain embodiments, n is 0. In certain embodiments n is 1. In certain embodiments, n is 2.
In some cases, the compound of formula (I) has the following characteristics: R1 <:>\ ; RsoN is 7!, R1 is selected from the group consisting of W, HOW, g R2 is H, F, CI, or CH3; R3 is CH3, R4 is H or F; , and N; andnisO,1,or2.
In some cases, the compound of formula (I) has the following characteristics: R1 ®\/ R\[\(/\|N is >11? is Wor HOXV ;R2 is H; R3 is CH3; R4 is H; and nis0,1,or2.
In some cases, the compound of a (I) has the following teristics: ring A is piperidinyl; R1 is H, C1_6alkyl, C2_6alkenyl, C1_6hydroxyalkyl, or C0_3alkylene-C3_ oalkyl; R2 is H, F, Cl, or CH3; R3 is C1.3alkyl; R4 is H, F, or Cl; and n is 0, 1, or 2. In some cases, the compound of formula (I) has the following teristics: ring A is piperidinyl; R‘ is H, C1_6alkyl, Czealkenyl, C1_6hydroxyalkyl, or C0.3alkylene-C3_1ocycloalkyl; R2 is H; R3 is C1_3alkyl; R4 is H or F; and n is 0, 1, or 2.
In some cases, the compound of formula (I) has the following characteristics: ring A is azetidinyl; R1 is H, C1.5alkyl, C2_6alkenyl, C1_6hydroxyalkyl, or C0_3alkylene-C3_1ocycloalkyl; R2 is H, F, CI, or CH3; R3 is kyl; R4 is H, F, or Cl; and n is 0, 1, or 2. In some cases, the compound of formula (I) has the following teristics: ring A is azetidinyl; R1 is H or C0. 3alkylene-C3_1ocycloalkyl; R2 is H; R3 is kyl; R4 is H; and n is 0, 1, or 2. In some cases, the compound of formula (I) has the following characteristics: ring A is azetidinyl; R1 is H or kylene-C3_1ocycloalkyl; R2 is H; R4 is H; and n is 0.
In various cases, R2 is H. In some cases, R2 is F. In some cases, R2 is CI. In some cases, R2 is CH3.
For compounds of formula (I), n can be 0. In certain embodiments, when n is 1 or 2, R3 is C1_3alkyl, and can be, e.g., CH3. In certain embodiments, when n is 2, each R3 can be substituted at the same atom of ring A, or at different atoms of ring A. In certain embodiments, when n is 2, each R3 is CH3 and each R3 is substituted at the same atom of ring A. In certain embodiments, when n is 2, each R3 is CH3 and each R3 is substituted at different atoms of ring A.
R4 can be H, or can be F, or can be CI. In various cases, R4 is H or F. In some cases, R4 is H. In some cases, R4 is F.
Some specific nds contemplated herein are listed in Table 1.
Table 1 Ex Structure 238 CF3€§€3C3§I WO 19362 3_/9 4oo 1 4009 1 oo 1 4_/ 2 4oo5 49 1 4oo6 4|_7 00 4_/ _/ 4|_ 7| oo 4_/ oo 4oo0 4_/9 4oo3 4oo 2 Some specific compounds plated include those listed in Table 2.
Table 2 556 ./‘/\\, ,..»~’\ xxx ,\ l 1 H NHLfl In certain embodiments, the compound or salt thereof is selected from Table 1. In certain ments, the compound or salt thereof is ed from Table 2. In certain embodiments the compound or salt thereof is selected from Table 1 and Table 2.
In certain embodiments, the compound or salt f is selected from the group consisting of compounds 485, 486, 479, 480, 483, 484, 482, 481, 489, 490, 491, 492, 487, 488, 477, 477-I, 477-Il, 478, 4781, 47811, 356, 359, 357, 379, 181, 472, 238, 241, 176, 171, 172, 174, 175, 354, 169, 161, 162, 163, 146, 147, 555, and 556, or a single stereoisomer or mixture of stereoisomers thereof.
Compounds of formula (I) bed herein may contain one or more asymmetric centers and thus occur as racemates and c mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. While shown without respect to the stereochemistry in formula (I), the present disclosure includes such optical isomers (enantiomers) and diastereomers; as well as the racemic and resolved, enantiomerically pure R and S stereoisomers; as well as other es of the R and S stereoisomers and pharmaceutically acceptable salts thereof. The use of these compounds is intended to cover the racemic mixture or either of the chiral enantiomers.
W0 20182’119362 18 One skilled in the art will also recognize that it is possible for tautomers to exist for the compounds described . The disclosure includes all such tautomers even though not shown in the formulas herein. All such isomeric forms of such compounds are expressly included in the t disclosure.
Optical s can be obtained in pure form by standard procedures known to those skilled in the art, and include, but are not limited to, diastereomeric salt formation, kinetic resolution, and tric synthesis. See, for example, Jacques, et aI., Enantiomers, Racemates and tions (Wiley Interscience, New York, 1981); Wilen, S.H., et aI., Tetrahedron 3322725 (1977); Eliel, E.L. Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); Wilen, S.H. Tables of Resolving Agents and Optical Resolutions p. 268 (EL. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN 1972), each of which is incorporated herein by reference in their entireties. It is also understood that this disclosure encompasses all possible regioisomers, and mixtures thereof, which can be obtained in pure form by rd separation procedures known to those skilled in the art, and include, but are not limited to, column chromatography, thin-layer chromatography, and high- performance liquid chromatography.
Compounds described herein can also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium, preferably deuterium.
The compounds described herein also include pharmaceutically able salts of the compounds disclosed herein. As used herein, the term aceutically acceptable salt” refers to a salt formed by the addition of a pharmaceutically acceptable acid or base to a compound sed herein. As used herein, the phrase “pharmaceutically acceptable” refers to a substance that is acceptable for use in pharmaceutical applications from a toxicological perspective and does not adversely interact with the active ingredient.
Pharmaceutically able salts, including mono— and bi— salts, include, but are not d to, those derived from organic and inorganic acids such as, but not limited to, acetic, lactic, citric, cinnamic, ic, succinic, fumaric, maleic, c, ic, malic, oxalic, propionic, hydrochloric, hydrobromic, phosphoric, nitric, sulfuric, glycolic, pyruvic, methanesulfonic, ethanesulfonic, toluenesulfonic, salicylic, benzoic, and similarly known acceptable acids.
Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418; Journal of Pharmaceutical Science, 66, 2 (1977); and "Pharmaceutical Salts: Properties, Selection, and Use A Handbook; Wermuth, C. G. and Stahl, P. H. (eds.) Verlag ica Chimica Acta, Zurich, 2002 [ISBN 90— 26-8] each of which is orated herein by reference in their entireties.
Methods of Use All the compounds and ceutical compositions ed herein can be used in any of the methods provided herein.
Provided herein are methods of inhibiting one or more HDAC enzymes (e.g., HDAC1 or HDACZ; e.g., HDAC3) or more than one HDAC (e.g., HDAC1 and HDAC2; e.g., HDAC1 and HDAC3; e.g., HDAC2 or HDAC3; e.g., HDAC1, HDAC2, and HDAC3) using a compound or a salt thereof as disclosed herein. In some embodiments, the methods can include contacting one or more HDAC enzymes (e.g., HDAC1 or HDAC2; e.g., HDAC3) in a sample with a compound or a salt thereof as disclosed herein. In other embodiments, the methods can include administering a compound or a salt thereof as disclosed herein to a subject (e.g., a mammal, such as a human).
A histone deacetylase , as described herein, can be any polypeptide having features characteristic of polypeptides that catalyze the l of the acetyl group (deacetylation) from acetylated target proteins. Features characteristic of HDAC enzymes are known in the art (see, for example, Finnin et al., 1999, Nature, 401 :188). Thus, an HDAC enzyme can be a polypeptide that ses gene ription by deacetylating the s-amino groups of conserved lysine residues located at the N-termini of histones, e.g., H3, H4, H2A, and H28, which form the nucleosome. HDAC enzymes also deacetylate other proteins such as p53, E2F, d-tubulin, and MyoD (see, for example, Annemieke et al., 2003, Biochem. J., 370:737). HDAC enzymes can also be localized to the s and certain HDAC s can be found in both the nucleus and also the cytoplasm.
Compounds described herein can interact with any HDAC enzyme. In some embodiments, the compounds described herein will have at least about 2—fold (e.g., at least about 5-fold, 10-fold, d, or 20—fold) greater activity to inhibit one or more class I HDAC enzymes (e.g., HDAC1, HDAC2, or HDAC3) as compared to one or more other HDAC enzymes (e.g., one or more HDAC enzymes of class Ila, llb, or IV).
In some embodiments, a compound or a salt thereof as disclosed herein selectively inhibits HDAC3, e.g., ively ts HDAC3 over HDAC1 and HDAC2 (e.g. exhibiting 5-fold or greater selectivity, e.g. exhibiting 25-fold or greater selectivity). While not wishing to be bound by theory, it is ed that HDAC3-selective inhibitors can increase expression of frataxin, and can therefore be useful in the treatment of neurological conditions (e.g., neurological conditions associated with reduced frataxin expression, such as Friedreich’s ataxia). It is also believed that HDACS inhibition plays an important role in memory consolidation (McQuown SC etal, J Neurosci 31 764 (2011)). Selective inhibitors of HDACS provide advantages for treatment of ogical conditions over the use of broad- spectrum HDAC inhibitors by ng ties associated with inhibition of other HDAC W0 119362 18 enzymes. Such ic HDAC3 inhibitors can provide a higher therapeutic index, resulting in better tolerance by patients during c or long-term treatment.
In some further embodiments, compounds selectively inhibit HDACt and/or HDAC2 (e.g. exhibiting 5-fold or greater selectivity, e.g. exhibiting 25-fold or greater selectivity). Inhibition of HDACt and/or 2 can be useful in treating cancer, or another disease as disclosed herein.
In some ments, a compound or a salt thereof as disclosed herein exhibits enhanced brain penetration. For example, brain/plasma ratios of greater than about 0.25 (e.g., greater than about 0.50, greater than about 1.0, greater than about 1.5, or greater than about 2.0) are observed when rats, mice, dogs, or monkeys are dosed with some of the compounds disclosed herein. In some embodiments, a compound or a salt thereof as disclosed herein selectively inhibits HDAC3, e.g., selectively inhibits HDAC3 over HDACt and HDAC2 (e.g., exhibiting 5-fold or greater selectivity, e.g. exhibiting 25-fold or greater selectivity) and exhibits enhanced brain penetration. In some embodiments, a compound described herein selectively inhibits HDACt and/or HDAC2, e.g., selectively inhibit HDACt and/or HDAC2 over HDAC3 (e.g., exhibiting 5-fold or greater selectivity, e.g. exhibiting 25— fold or greater selectivity) and ts enhanced brain penetration.
Compounds with ed brain penetration are suitable for therapies targeting the brain (e.g., neurological conditions such as Friedreich’s ataxia, myotonic dystrophy, spinal muscular atrophy, fragile X syndrome, Huntington’s e, spinocerebellar ataxia, Kennedy’s disease, amyotrophic lateral sclerosis, spinal and bulbar muscular y, and Alzheimer’s disease; a memory impairment condition, frontotemportal dementia; post- traumatic stress disorder; a drug ion).
Provided herein are methods of treating a disease or disorder mediated by HDAC in a subject (e.g., a , such as a human) in need thereof, which include administering a compound or a salt thereof as disclosed herein to the subject.
Further provided herein are methods of preventing a e or disorder mediated by HDAC in a subject (e.g., a mammal, such as a human) in need f. Prevention can include delaying the onset of or reducing the risk of developing, a disease, disorder, or condition or symptoms f.
The disclosure further provides a method of treating a cancer in patient in need thereof, comprising stering a therapeutically effective amount of an HDAC inhibitor as described herein, or salt thereof. In some embodiments, the cancer is a solid tumor, neoplasm, carcinoma, sarcoma, leukemia, or lymphoma. In some embodiments, leukemias include acute leukemias and c leukemias such as acute cytic leukemia (ALL), W0 20182’119362 18 acute myeloid leukemia, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML) and Hairy Cell Leukemia; lymphomas such as cutaneous T-cell lymphomas (CTCL), aneous peripheral T—cell lymphomas, lymphomas associated with human T- cell lymphotrophic virus (flTLV) such as adult T-cell leukemia/lymphoma (ATLL), Hodgkin's e and non-Hodgkin's mas, large-cell lymphomas, diffuse large B-cell lymphoma (DLBCL); Burkitt's lymphoma; primary central s system (CNS) lymphoma; multiple a; childhood solid tumors such as brain , neuroblastoma, retinoblastoma, Wilm's tumor, bone tumors, and soft-tissue sarcomas, common solid tumors of adults such as head and neck cancers (e.g., oral, eal and esophageal), genitor- urinary cancers (e.g., prostate, bladder, renal, uterine, ovarian, testicular, rectal and colon), lung cancer, breast cancer.
In some embodiments, the cancer is (a) Cardiac: sarcoma sarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; (b) Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; (c) Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, ma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, noma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular a, villous adenoma, hamartoma, leiomyoma); (d) urinary tract: kidney (adenocarcinoma, Wilm's tumor (nephroblastoma), lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell oma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); (e) Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, cellular adenoma, hemangioma; (f) Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, ant fibrous histiocytoma, chondrosarcoma, Ewing's a, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochrondroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, omyxofibroma, osteoid osteoma and giant cell tumors; (g) Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, oblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiform, endroglioma, noma, retinoblastoma, congenital tumors), spinal cord (neurofibroma, meningioma, glioma, sarcoma); (h) Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma, serous cystadenocarcinoma, us enocarcinoma), sified oma (granulosa-thecal cell tumors, Sertoli-Leydig cell , dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma), embryonal rhabdomyosarcoma, ian tubes noma); (i) Hematologic: blood (myeloid ia [acute and c], acute lymphoblastic leukemia, chronic cytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, .non-Hodgkin’s lymphoma (malignant lymphoma); (j) Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi’s sarcoma, moles dysplastic nevi, lipoma, angioma, ofibroma, keloids, psoriasis; and (k) Adrenal glands: neuroblastoma conditions.
In another aspect, provided is a method of treating an inflammatory er in patient in need f, comprising administering a therapeutically effective amount of a compound as described herein, or salt thereof. In some embodiments, the inflammatory disorder is an acute and chronic matory disease, autoimmune disease, allergic disease, disease associated with oxidative stress, and diseases characterized by ar hyperproliferation. Non-limiting examples are inflammatory conditions of a joint including rheumatoid arthritis (RA) and psoriatic arthritis; inflammatory bowel diseases such as Crohn's disease and ulcerative colitis; spondyloarthropathies; scleroderma; psoriasis ding T-cell mediated psoriasis) and inflammatory dermatoses such an dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria; vasculitis (e.g., necrotizing, cutaneous, and hypersensitivity vasculitis); eosinophilic myositis, eosinophilic fasciitis; cancers with leukocyte infiltration of the skin or organs, ischemic injury, including cerebral ischemia (e.g., brain injury as a result of trauma, epilepsy, hemorrhage or stroke, each of which may lead to neurodegeneration); HIV, heart failure, chronic, acute or malignant liver disease, autoimmune thyroiditis; systemic lupus erythematosus, Sjorgren's syndrome, lung diseases (e.g., ARDS); acute atitis; amyotrophic lateral sclerosis (ALS); Alzheimer's disease; cachexia/anorexia; asthma; atherosclerosis; chronic fatigue syndrome, fever; diabetes (e.g., insulin diabetes or le onset diabetes); glomerulonephritis; graft versus host ion (e.g., in transplantation); hemorrhagic shock; hyperalgesia: inflammatory bowel disease; le sclerosis; myopathies (e.g., muscle protein metabolism, esp. in sepsis); rthritis; osteoporosis; Parkinson's disease; pain; preterm labor; psoriasis; reperfusion injury; ne-induced toxicity (e.g., septic shock, endotoxic shock); side effects from radiation therapy, temporal mandibular joint disease, tumor metastasis; or an inflammatory condition resulting from strain, sprain, cartilage , trauma such as burn, orthopedic surgery, infection or other disease processes.
W0 20182’119362 18 Allergic diseases and conditions, include but are not limited to respiratory allergic diseases such as asthma, allergic rhinitis, hypersensitivity lung diseases, hypersensitivity pneumonitis, eosinophilic pneumonias (e.g., Loeffler's syndrome, chronic eosinophilic pneumonia), delayed-type hypersensitivity, interstitial lung diseases (ILD) (e.g., idiopathic pulmonary fibrosis, or ILD associated with toid arthritis, systemic lupus erythematosus, ankylosing spondylitis, systemic sclerosis, Sjogren's syndrome, polymyositis or dermatomyositis); ic anaphylaxis or hypersensitivity responses, drug ies (e.g., to penicillin, cephalosporins), insect sting allergies, and the like.
In another , provided is a method of preventing or treating a memory-related disorder in patient in need thereof, comprising administering a eutically ive amount of a compound as described herein. Compounds can be used to treat patients with memory impairments ated with direct cognitive disorders such as amnesia, dementia and delirium, temportal dementia; anxiety disorders such as phobias, panic disorders, psychosocial stress (e.g. as seen in disaster, catastrophe or violence s), obsessive— sive disorder, generalized anxiety er and post-traumatic stress disorder; mood disorders such as depression and bipolar disorder; and psychotic disorders such as schizophrenia and delusional disorder. Memory impairment, a hallmark of neurodegenerative diseases such as, but not limited to, Parkinson’s, mer’s, gton’s, amyotrophic l sclerosis (ALS), spinocerebellar ataxia, as well as aging, can also be treated by using a compound disclosed herein. In addition, compounds sed can be used to treat drug addiction through extinction of drug-seeking behavior.
HDAC inhibitors, e.g., HDACt and/or HDAC2 selective inhibitors, may also be useful to treat sickle cell disease (SCD) and B-thalassemia (bT). They may also be useful in treating mood disorders or brain disorders with altered in—mediated lasticity (Schoreder, et al., PLoS ONE 8(8): e71323 (2013)).
In another aspect, provided is a method of preventing or treating a hemoglobin disorder in patient in need thereof, comprising administering a therapeutically effective amount of a compound as described herein, or salt thereof. nds can be used to treat patients with sickle cell anemia or B—thalassemia. In various embodiments, the compound is a selective HDAC1 and/or HDAC2 inhibitor and is used to prevent or treat the hemoglobin disorder (e.g., sickle cell anemia or B-thalassemia).
Further provided is a method of preventing or treating a mood disorder or brain disorders with altered chromatin—mediated neuroplasticity in patient in need thereof, comprising administering a therapeutically effective amount of a compound as described herein, or salt thereof. Compounds as described herein can be used to treat patients with a mood disorder.
W0 20182’119362 18 In a further , this application features methods of treating a neurological condition (e.g., Friedreich’s ataxia (FRDA), myotonic dystrophy, spinal muscular atrophy, fragile X me, Huntington’s disease, a erebellar ataxia, Kennedy’s disease, amyotrophic l sclerosis, Niemann Pick, Pitt Hopkins, spinal and bulbar muscular atrophy, Alzheimer’s disease or schizophrenia, bipolar disorder, and related diseases) that include administering a compound described herein or salt thereof to a patient having a neurological condition.
In another aspect, provided herein is the use of a compound described herein or salt f in the preparation of a medicament for the treatment or prevention of a neurological condition (e.g., Friedreich’s ataxia, myotonic dystrophy, spinal muscular atrophy, fragile X syndrome, Huntington’s disease, a spinocerebellar ataxia, Kennedy’s disease, amyotrophic lateral sclerosis, Niemann Pick, Pitt Hopkins, spinal and bulbar muscular y, or Alzheimer’s disease); a -affecting condition or disease, a cancer; or an inflammatory disorder, or a dium falciparum infection (e.g., a).
Further provided herein is a method of using a compound or a salt thereof as disclosed herein to inhibit class I histone deacetylases, wherein this inhibition results in an in vitro increased frataxin mRNA expression in eich’s ataxia patient peripheral blood mononuclear cells (PBMCs). In other embodiments, compounds or a salt thereof disclosed herein inhibit in vitro proliferation of colorectal cancer cells in a dose-dependent fashion. In further embodiments, compounds or a salt thereof disclosed herein se long term memory in vivo using the novel object recognition gm.
In a further aspect, provide herein is a kit for the treatment or prevention of a disorder ed from a neurological disorder (e.g., eich’s ataxia, ic dystrophy, spinal muscular atrophy, fragile X syndrome, Huntington’s disease, a spinocerebellar ataxia, y’s disease, amyotrophic lateral sclerosis, spinal and bulbar muscular atrophy, or Alzheimer’s disease), a memory-affecting condition or disease, a cancer, an inflammatory disorder, or a Plasmodium falciparum infection (e.g., malaria) in a patient in need thereof, comprising (i) a compound described herein or a salt thereof; and (ii) instructions comprising a direction to administer said compound to said patient.
In another aspect, provided are methods of treating a neurological condition (e.g., Friedreich’s ataxia, ic dystrophy, spinal muscular atrophy, fragile X syndrome, Huntington’s disease, spinocerebellar ataxias, Kennedy’s disease, amyotrophic lateral sclerosis, spinal and bulbar muscular atrophy, or Alzheimer’s e) that include performing any of the above methods, ating the candidate nd or a salt thereof in a pharmaceutical composition, and administering the pharmaceutical composition to a patient having a neurological condition.
W0 20182’119362 18 HDAC inhibitors have been shown to have antimalarial activity (Andrews, et al., 2000, Int. J. Parasitol., 30:761-768; Andrews, et al., Antimicrob. Agents Chemother., 52:1454-61). The present disclosure provides methods of treating a Plasmodium falciparum infection (e.g., malaria) in a patient in need thereof.
HDAC inhibitors may also be useful to treat infectious disease such as viral infections. For example, treatment of HIV infected cells with HDAC inhibitors and anti- retroviral drugs can eradicate virus from treated cells (Blazkova, J., et al J Infect Dis. 2012 Sep 1 ):765-9; Archin, N.M., et al Nature 2012 Jul 25, 487(7408):482-5). The present disclosure provides methods of treating a HIV infection in need thereof.
In n embodiments, provided is a method of treating any of the diseases or disorders described herein comprising stering to a t in need of treatment f a compound or salt thereof according to any of the various embodiments sed Pharmaceutical itions HDAC inhibitors as disclosed herein can be administered neat or formulated as ceutical itions. Pharmaceutical compositions e an appropriate amount of the HDAC inhibitor in combination with an appropriate carrier and optionally other useful ingredients. For example, the other useful ingredients include, but not limited to, encapsulating materials or additives such as absorption accelerators, antioxidants, binders, buffers, coating , coloring agents, diluents, disintegrating agents, emulsifiers, extenders, fillers, flavoring agents, ants, lubricants, perfumes, preservatives, propellants, releasing agents, izing agents, sweeteners, solubilizers, wetting agents and mixtures thereof.
In certain embodiments, optionally in combination with any or all of the above various embodiments, provided herein is pharmaceutical composition of a compound disclosed herein, for example a compound of formula (I), a compound of Table 1, or a compound of Table 2, or stereoisomers thereof, or a pharmaceutically acceptable salt f, and one or more pharmaceutically acceptable carriers.
In n embodiments, the pharmaceutical composition comprises a compound of formula (I), or stereoisomers thereof, or a pharmaceutically acceptable salt thereof, and one or more ceutically acceptable carriers. In certain embodiments, pharmaceutical composition comprises a compound of Table 1, or a compound of Table 2, or stereoisomers thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers. In certain embodiments, pharmaceutical composition comprises a compound of Table 1, or stereoisomers thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers. In certain embodiments, W0 20182’119362 18 pharmaceutical composition comprises a compound of Table 2, or stereoisomers thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers.
Thus, provided herein are pharmaceutical compositions comprising a compound described herein and one or more pharmaceutically acceptable carriers. The pharmaceutical compositions are administered to a t in need thereof by any route which makes the compound bioavailable. In one embodiment, the composition is a solid formulation adapted for oral stration. In another embodiment, the composition is a tablet, , or capsule; or the composition is a tablet. In one embodiment, the composition is a liquid formulation d for oral administration. In one embodiment, the composition is a liquid ation adapted for eral stration. In another embodiment, the composition is a on, suspension, or emulsion; or the composition is a solution. In another embodiment, solid form compositions can be ted, shortly before use, to liquid form compositions for either oral or parenteral administration. These particular solid form compositions are provided in unit dose form and as such are used to provide a single liquid dosage unit.
These and other pharmaceutical compositions and processes for ing the same are well known in the art. (See, for example, Remington: The e and Practice of Pharmacy (D. B. Troy, Editor, 21st Edition, Lippincott, Williams & Wilkins, 2006).
The dosages may be varied depending on the requirement of the patient, the severity of the ion being treating and the particular compound being employed.
Determination of the proper dosage for a particular situation can be determined by one skilled in the l arts. The total daily dosage may be divided and administered in portions throughout the day or by means ing continuous delivery.
The compounds and compositions described herein may be administered initially in a suitable dosage that may be adjusted as required, depending on the desired clinical response. In certain embodiments, the compounds are administered to a subject at a daily dosage of between 0.01 to about 50 mg/kg of body weight. In other embodiments, the dose is from 1 to 1000 mg/day. In certain embodiments, the daily dose is from 1 to 750 mg/day; or from 10 to 500 mg/day.
In another embodiment, the pharmaceutical composition is in unit dosage form.
The composition can be subdivided into unit doses containing appropriate quantities of the active component(s). The unit dosage form can be a tablet, capsule, or powder in a vial or ampule, or it may be the appropriate number of any of these in a packaged form. The unit dosage form can be a packaged form, the package containing discrete quantities of ition such as packeted tablets, capsules, or powders in vials or ampules. The quantity of active compound(s) in a unit dose of the composition may be varied or adjusted W0 20182’119362 18 from about 1 mg to about 100 mg, or from about 1 mg to about 50 mg, or from about 1 mg to about 25 mg, according to the particular application.
General Synthesis of nds of formula (l[ Compounds of the present disclosure can be conveniently prepared in accordance with the procedures outlined in the Examples section, from commercially available starting materials, compounds known in the ture, or readily prepared intermediates, by ing conventional synthetic methods and procedures known to those skilled in the art.
Conventional synthetic methods and procedures for the preparation of c molecules and functional group transformations and manipulations can be readily obtained from the relevant scientific literature or from standard textbooks in the field. It will be appreciated that, where typical or preferred process ions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures. Those skilled in the art of organic synthesis will ize that the nature and order of the tic steps presented may be varied for the purpose of zing the formation of the nds described .
EXAMPLES Synthesis of specific compounds was performed as follows.
Abbreviations Abbreviation Meaning ACN acetonitrile ACOH acetic acid Boc tert—butyloxycarbonyl 80020 di-tert-butyl dicarbonate 082003 cesium carbonate DCE 1,2—dichloroethane DCM romethane DIPEA diisoproylethylamine DMAP 4-dimethylaminopyridine DMF dimethylformamide DMSO dimethyl sulfoxide EtOH ethanol EDCI 1-ethyl(3-dimethylaminopropyl)carbodiimide Fmoc fluorenylmethyloxycarbonyl HATU O—(7-azabenzotriazol-1 -y|)-N,N,N’,N’-tetramethyluronium hexafluorophosphate HCI hydrochloric acid W0 2018l119362 Abbreviation Meaning HPLC high performance liquid chromatography Hz Hertz (s' ) HOBt hydroxybenzotriazole K2003 ium carbonate T3P Propylphosphonic acid anhydride LC-MS liquid chromatography-mass spectrometry mg milligram MeOH methanol mL milliliter mm millimeter pm micrometer mmol millimole pL microliter mM olar uM micromolar NMR Nuclear Magnetic Resonance NaH sodium hydride STAB sodiumtriacetoxyborohyride TEA triethylamine TFA trifluoroacetic acid THF tetrahydrofuran TLC thin layer chromatography Synthetic Scheme for Compound 485 and Compound 486: BrfiO/ O 0/ Red. ammni / 4"” HC' '" ”Wane Boc—N —> Boc—WmK/Ns\/<jjiO/—. yQJiO A'ky'a—>"°” R——Nm \\/NH K2003, MeCN, \\/N RT 1 h Step'3 1 2HC| RT 16 h Step-2 5a-b Step-1 We 0 NaOH 4M HCI/Dioxane N —’ R‘“ N Ste 6 R—NO “Hz M60“ H20 (1 1) HATU Pyridine K/N H 9' NHBDC Step-4 'b Reflux, 12h 8a-b Compound- 485 Step—5 Compound-486 Compound- 485 Compound-486 Step 1: Synthesis of utyl 4—(4-(methoxycarbonyl)benzyl)-1 ,4-diazepane carboxylate (3): To a stirred solution of compound 1 (1.92 g, 1.1 eq.) and compound 2 (2 g, 1 eq.) in ACN (20 mL), potassium carbonate (1.8 g, 1.5 eq.) was added. The reaction mixture was stirred at room temperature for 16 h. The tion of reaction was monitored by TLC. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over anhydrous NaZSO4, filtered and concentrated under reduced pressure to afford the title compound 3 which was used for next step without further purification.
Step 2: Synthesis of methyl 4-((1,4-diazepanyl)methyl)benzoate hydrochloride (4): To a stirred on of compound 3 (2.8 g, 1 eq.) in 1,4-dioxane (10 mL), 4M HCI in dioxane (20 mL) was added at 0 °C. The resulting on mass was stirred at room temperature for 1 h. After completion of on, the reaction mixture was concentrated under reduced pressure, the resulting e was triturated with diethyl ether and dried under vacuum to afford the title nd 4 as HCI salt.
Step 3: Synthesis of compound 5a for Compound 485: To a stirred solution of compound 4 (1 eq.) and cyclopropyl carboxaldehyde (1.2 eq.) in DCM (10 vol.), acetic acid (6 eq.) was added and stirred at room temperature for 30 min. To this, sodium triacetoxyborohydride (STAB) (3 eq.) was added at room temperature. The resulting reaction mixture was stirred at room temperature for overnight. The reaction mixture was then quenched with sat. NaHCOa solution and extracted with DCM. The combined organic extracts were washed with water and brine, dried over anhydrous NaZSO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the d compound 5a.
Step 3: Synthesis of nd 5b for Compound 486: To a solution of compound 4 (1 eq) in ethanol (10 vol.), TEA (2.5 eq) and 2,2-dimethyloxirane (1.5 eq) were added. The reaction mixture was heated at 80 °C for 12 h. The completion of reaction was monitored by TLC. The reaction mixture was allowed to cool, concentrated to give the crude product which was purified by silica gel column chromatography to afford the desired compound 5b.
Step 4: l procedure for synthesis of compound 6a-b: To a stirred solution of nd 5 (1 eq.) in methanol: water (1:1), NaOH (1.5 eq.) was added at room temperature. The above mixture was heated to 80 0C for 5 to 6 h. The progress of the on was monitored by TLC. After tion of reaction, the reaction mixture was concentrated and the resulting residue was dissolved in water and washed with diethyl ether.
The aqueous layer was neutralized to pH = 7 using 1N HCI at 0 0C. The solid obtained was filtered, washed with water and dried under vacuum to afford the desired compound 6.
Step 5: General procedure for synthesis of compound 8a-b: To a stirred on of compound 6 (1 eq.) and tert—butyl (2—aminophenyl)carbamate (1.2 eq.) in ACN, pyridine (6 eq.) and HATU (1.5 eq.) were added at room temperature. The on mixture was stirred at 90 °C for overnight and the reaction progress was monitored by TLC and LCMS. After completion of on, the reaction mixture was concentrated and resulting residue was portioned between water and ethyl e. The organic layer was separated, washed with water and 1% HCI to remove traces of pyridine, dried over anhydrous NaZSO4 and concentrated. The crude residue was purified by silica gel column chromatography to afford the desired compound 8.
No Structure 1 mm J \\/N HN\BOC 2 Nqfi”\\/ HN ‘Boc HO 8b Step 6: Synthesis of N-(2-aminophenyl)((4-(cyclopropylmethyl)-1,4-diazepan yI)methyI) ide trihydrochloride (Compound 485): To a stirred solution of compound 8 (1 eq.) in 1,4-dioxane (5 vol.), 4M HCI in e (5 vol.) was added. The reaction e was stirred at room temperature for 1 h. The completion of reaction was monitored TLC. The reaction mixture was concentrated, resulting residue was triturated with diethyl ether and dried under vacuum to afford the title compound 485 as a HCI salt. 1H NMR (400 MHz, 6): 6 10.12 (s, 1H), 8.12 (d, J: 7.2 Hz, 2H), 7.82 (d, J: 8.0 Hz, 2H), 7.33 (d, J: 7.2 Hz, 1H), 7.16 — 7.08 (m, 2H), 6.95 — 6.93 (m, 1H), 4.46 (s, 2H), 3.67 — 3.34 (m, 8H), 3.05 — 3.04 (m, 2H), 2.33 — 2.26 (m, 2H), 1.12 — 1.10 (m, 1H), 0.65 - 0.63 (m, 2H), 0.42 — 0.40 (m, 2H); LCMS: Calculated for C23H30N4O for free base : 378.24; Observed: 379.15 (M+1)*.
Step 6: Synthesis of N-(2-aminophenyl)—4-((4-(2-hydroxymethylpropyl)-1,4- diazepan-1—yl)methyl) benzamide (Compound 486): To a stirred solution of nd 8 (1 eq.) in 1,4-dioxane (5 vol.), 4M HCI in dioxane (5 vol.), was added. The resulting reaction mass was stirred at room temperature for 1 h. The completion of on was red TLC. The reaction mixture was concentrated under reduced pressure. The residue was basified with sat. NaHC03 on and extracted with ethyl acetate. The organic layer was separated, washed with water and brine, dried over anhydrous NaZSO4 and concentrated.
The crude t was purified by silica gel column chromatography and preparative HPLC to afford the title compound 486. ] 1H NMR (400 MHz, DMSO-d6) 6 9.61 (s, 1H), 7.93 (d, J: 7.8 Hz, 2H), 7.44 (d, J = 7.8 Hz, 2H), 7.16 (d, J: 7.8 Hz, 1H), 6.97 (t, J: 7.7 Hz, 1H), 6.78 (d, J: 7.9 Hz, 1H), 6.59 (t, J: 7.5 Hz, 1H), 4.88 (s, 2H), 3.96 (s, 1H), 3.67 (s, 2H), 2.83 — 2.79 (m, 4H), 2.67 — 2.56 (m, 4H), 2.38 (s, 2H), 1.70 — 1.68 (m, 2H), 1.07 (s, 6H); LCMS: Calculated for C23H32N402: 396.25; Observed: 396.95 (M+1)+.
Synthetic Scheme for Compound 479 and Compound 480: BrfiO/ :Cl/fi Red. ammn/ 2 Alkylation HCI in Dioxane Mfio/ —> K2C03. MeCN N Step-3 RT, 16 h RT 1 h Step-1 Step-_2 o OBDCW/Q 0 Dioxane. HCI RN/W H RN NaOH N —’ A/ H—*7NH2 R‘N/\l H SteP-3 (DH/Q MeOH:H20( 1: 1) N N HATU, Py, ACN, A/ HNt Step-.4 reflux 8a-b Compound-479 Step-5 Compound-480 HO>—A Compound-479 Compound-480 Step 1: Synthesis of tert—butyl -(4—(methoxycarbonyl)benzyI) methylpiperazinecarboxylate (3): To a stirred solution of compound 1 (1.92 g, 1.1 eq.) and compound 2 (2 g, 1 eq.) in ACN (20 mL), potassium carbonate (1.81 g, 1.5 eq.) was added.
The reaction mixture was stirred at room temperature for 16 h. The completion of reaction was monitored by TLC. The on mixture was diluted with water and extracted with ethyl acetate. The combined organic ts were washed with water, brine, dried over anhydrous NaZSO4, filtered and concentrated under reduced pressure to get the crude product which was purified by silica gel column chromatography to afford the title compound Step 2: sis of methyl (R)—4-((3-methylpiperazinyl)methyl)benzoate hydrochloride (4): To a stirred solution of compound 3 (2.9 g, 1 eq.) in 1,4—dioxane (5 mL), 4M HCI in dioxane (15 mL) was added. The resulting reaction mass was d at room temperature for 1 h. The tion of reaction was monitored by TLC. The reaction mixture was concentrated under reduced pressure, resulting residue was triturated with diethyl ether and dried under vacuum to afford the title compound 4 as HCI salt.
Step 3: Synthesis of compound 5a for compound-479: To a stirred solution of compound 4 (1 eq.) and cyclopropyl carboxaldehyde (1.2 eq.) in DCM (10 vol.) was added acetic acid (6 eq.) and sodium triacetoxyborohydride (STAB) (3 eq.) at room temperature.
Reaction e was stirred at room temperature for overnight. The reaction mixture was then ed with sat. NaH003 solution and extracted with DCM. The combined organic extracts were washed with water and brine, dried over ous NaZSO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the desired compound 5a.
Step 3: Synthesis of compound 5b for nd 480: To a solution of compound 4 (1 eq.) in l (10 vol.), TEA (2.5 eq.) and 2,2—dimethyloxirane (1.5 eq.) were added and the reaction mixture was heated at 80 °C for 12 h. The progress of reaction was monitored by TLC. After completion of reaction, the reaction mixture was allowed to cool, concentrated to give a crude compound which was purified by silica gel column chromatography to afford the desired compound 5b.
Step 4: General procedure for synthesis of compound 6a-b: To stirred solution of compound 5 (1 eq.) in methanol:water (1:1) was added NaOH (1.5 eq.) at room temperature.
The reaction mixture was heated to 60 °C for 5 to 6 h. The progress of reaction was monitored by TLC. After completion of reaction, the reaction e was concentrated and the resulting residue was dissolved in water and washed with diethyl ether. The aqueous layer was neutralized to pH = 7 using 1N HCI at 0 °C. The solid obtained was filtered, washed with water and dried under vacuum to provide the desired compound 6.
] Step 5: General procedure for synthesis of compound 8a-b: To a stirred solution of compound 6 (1 eq.) and tert—butyl (2-aminophenyl)carbamate (1.1 eq.) in ACN, pyridine (5 eq.) and HATU (1.5 eq.) were added at room temperature. After stirring the reaction mixture at 80 °C for 12 to 16 h, the reaction progress was red by TLC and LCMS. After completion of reaction, the reaction mixture was concentrated and the residue was partitioned n water and ethyl acetate. The c layer was separated, washed with water and 1% HCI to remove traces of pyridine, dried over anhydrous NaZSO4 and concentrated. The crude residue was purified by silica gel column chromatography to afford the desired compound 8.
Structure Step 6: Synthesis of (R)—N-(2-aminophenyl)((4-(cyc|opropy|methyl) methylpiperazinyl)methyl)benzamide (Compound 479): To a stirred solution of compound 8 (1 eq) in 1,4-dioxane (5 vol.), 4M HCI in dioxane (5 vol.) was added. The resulting reaction mass was stirred at room temperature for 1 h. The tion of reaction was monitored TLC. The reaction mixture was concentrated under reduced pressure. The residue was basified with sat. NaHC03 solution and extracted with ethyl e. The combined organic layers were washed with water and brine, dried over anhydrous Na2804 and concentrated.
The crude residue was purified by silica gel column tography and preparative HPLC to afford the desired compound 479. 1H NMR (400 MHz, DMSO-d6) 6 9.62 (s, 1H), 7.93 (d, J: 7.9 Hz, 2H), 7.42 (d, J = 7.9 Hz, 2H), 7.19 — 7.12 (m, 1H), 7.01 —6.88 (m, 1H), 6.78 — 6.76 (m, 1H), 6.64— 6.55 (m, 1H), 4.88 (s, 2H), 3.50 (s, 2H), 2.93 — 2.91 (m, 1H), 2.65 — 2.53 (m, 3H), 2.17 — 2.09 (m, 2H), 1.88— 1.86 (m, 1H), 0.93 (d, J: 6.1 Hz, 3H), 0.81 — 0.79 (m, 1H), 0.46 — 0.42 (m, 2H), 0.06 — 0.05 (m, 2H), 2H merged in solvent peak; LCMS: Calculated for C23H30N40: 378.24; Observed: 379.05 (M+1 )1.
Step 6: Synthesis of (R)—N-(2-aminophenyl)((4-(2-hydroxy-2—methylpropyl) methylpiperazinyl)methyl)benzamide (Compound 480): To a stirred solution of compound 8 (0.3 g, 1 eq.) in 1,4-dioxane (5 vol.), 4M HCI in dioxane (5 vol.) was added. The resulting reaction mass was stirred at room temperature for 1 h. The completion of reaction was monitored TLC. The reaction e was concentrated under reduced pressure. The residue was basified with sat. NaHC03 solution and extracted with ethyl acetate. The combined organic layers were washed with water and brine, dried over anhydrous NaZSO4 and concentrated. The crude residue was ed by silica gel column chromatography and preparative HPLC to afford title compound 480. ] 1H NMR (400 MHz, DMSO-d6) 6 9.62 (s, 1H), 7.93 (d, J: 7.9 Hz, 2H), 7.42 (d, J = 7.9 Hz, 2H), 7.16 (d, J: 7.9 Hz, 1H), 6.97 (t, J: 7.6 Hz, 1H), 6.78 (d, J: 7.9 Hz, 1H), 6.60 (t, J: 7.6 Hz, 1H), 4.88 (s, 2H), 3.96 (s, 1H), 3.49 (s, 2H), 3.10 — 2.98 (m, 1H), 2.46 — 2.42 (m, 4H), 2.38 — 2.33 (m, 2H), 2.26 — 2.24 (m, 1H), 2.09 — 1.92 (m, 1H), 1.06 (d, J: 4.8 Hz, 6H), 0.94 (d, J: 6.0 Hz, 3H). LCMS: Calculated for 023H32N402: 396.25; Observed: 397.20 (M+1)*.
Synthetic Scheme for Compound 483 and Compound 484: Ofio O Boc \ N —> B00N/fi 0/ 0’ RA/Alkylation RN 0/ 4M HCIIn e Ejfio fl #VNs N N Step--3 STAB DCM Step2 1 5a-b RT 16 h Step-1 BDC\E/Q O R R oc 4M HCI/Dioxane N/fi N/fi N NaOH N/W —> H N NHZ MeOH: H20 (1: 1) N N Step--6 DIPEA T3P DCM RT Step--4 ab (For Comp483) Compound-483 HATU Pyridine ACN, Compound-4B4 RT (For Comp-484) Step-5 nd-483 compound-484 Step 1: Synthesis of tert—butyl 4—(4-(methoxycarbonyl)benzyl)-2,2- dimethylpiperazine—1-carboxylate (3): To a stirred solution of compound 1 (0.4 g, 1 eq.) and aldehyde 2 (0.367 g, 1.2 eq.) in DCM (15 mL), sodium triacetoxyborohydride (STAB) (0.553 g, 1.4 eq.) was added at room ature. The resulting reaction mixture was stirred at room temperature for 16. The completion of reaction was monitored by TLC and LCMS. The reaction mixture was portioned between DCM and water. The organic layer was washed with water and brine, dried over NaZSO4 and ated to get the crude product which was purified by silica gel column chromatography to afford the title compound 3.
Step 2: Synthesis of methyl 4-((3,3-dimethylpiperazin-1—yl)methyl)benzoate hydrochloride (4): To a stirred solution of compound 3 (0.5 g, 1 eq.) in 1,4—dioxane (5 mL), 4M HCl in dioxane (15 mL) was added. The resulting on was d at room temperature for 1 h. The reaction completion was red by TLC. The reaction mixture was concentrated and the resulting residue was triturated with n-pentane and dried under vacuum to afford the title compound 4 as HCI salt.
Step 3: Synthesis of compound 5a for compound 483: To a stirred solution of nd 4 (1 eq.) and ropyl carboxaldehyde (1.2 eq.) in DCM (10 mL), acetic acid (6 eq.) was added and d at room temperature for 30 min. To this, sodium triacetoxyborohydride (STAB) (3 eq.) was added at room temperature. The resulting reaction mixture was stirred at room temperature for overnight. The reaction completion was monitored by TLC and LCMS. The reaction mixture was quenched with sat. NaHC03 solution and extracted with DCM. The combined organic layers were washed with water and brine, dried over NaZSO4 and evaporated to get the crude t which was purified by silica gel column chromatography to afford the desired compound 5a.
Step 3: Synthesis of compound 5b for compound 484: To a solution of nd 4 (1 eq.) in ethanol (10 vol.), TEA (3 eq.) and 2,2—dimethyloxirane (2.6 eq.) were added and the reaction mixture was heated at 80 0C for 12 h. The reaction completion was monitored by TLC. The reaction mixture was d to cool, concentrated to give the crude compound which was purified by silica gel column chromatography to afford the desired compound 5.
No ure 1 mfio Homfio5b Step 4: General procedure for synthesis of compound 6a-b: To stirred solution of compound (1.0 eq.) in methanol:water (1:1) was added NaOH (1.5 eq.) at room temperature.
The above mixture was heated to 60 °C for 12 to 18 h. The progress of the reaction was monitored by TLC. After completion of reaction, the reaction mixture was concentrated and the resulting residue was dissolved in water and washed with diethyl ether. The aqueous layer was lized to pH = 7 using 1N HCI at 0 °C. The solid obtained was filtered, washed with water and dried under vacuum to provide the desired compound 6.
Step 5: Synthesis of compound 8a for compound 483: To a stirred on of nd 6a (1 eq.) and compound 7 (1.2 eq.) in DCM (10 vol.), DIPEA (2 eq.) and T3P (1.5 eq.) were added at room temperature. The reaction mixture was stirred at room temperature for 12 h. The reaction completion was monitored by TLC and LCMS. The reaction mixture was portioned between DCM and water. The c layer was washed with water and brine, dried over NaZSO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the desired compound 8a.
Step 5: Synthesis of compound 8b for compound 484: To a stirred solution of compound 6b (1 eq.) and compound 7 (1.1 eq.) in ACN (10 vol., pyridine (5 eq.) and HATU (1.5 eq.) were added at room temperature. After stirring the reaction mixture at 80 °C for 12 h, the reaction completion was monitored by TLC and LCMS. The reaction mixture was concentrated and resulting residue was partitioned between water and ethyl acetate. The organic layer was washed with water and 1% HCI to remove traces of pyridine, dried over NazSO4 and concentrated. The crude residue was purified by silica gel column chromatography to afford the desired nd 8b.
‘Boc 8a Step 6: Synthesis of N-(2-aminophenyl)((4-(cyclopropylmethyl)-3,3- dimethylpiperazin-1—yl)methyl)benzamide und 483): To a stirred solution of compound 8a (1 eq.) in 1,4-dioxane (5vol.), 4M HCI in dioxane (5 vol.) was added. The resulting reaction mass was stirred at room temperature for 1 h. The reaction tion was monitored TLC. The reaction mixture was concentrated under reduced pressure. The residue was ed with sat. NaHCOs on and extracted with ethyl acetate. The combined organic layers were washed with water and brine, dried over NaZSO4 and concentrated. The crude residue was purified by silica gel column tography and preparative HPLC to afford the desired compound 483. 1H NMR (400 MHz, DMSO-d6) 6 9.60 (s, 1H), 7.92 (d, J: 7.9 Hz, 2H), 7.41 (d, J = 7.9 Hz, 2H), 7.18— 7.11 (m, 1H), 7.00 —6.91 (m, 1H), 6.76 (d, J: 7.9 Hz, 1H), 6.58 (t, J: 7.5 Hz, 1H), 4.87 (s, 2H), 3.47 (s, 2H), 2.63 — 2.61 (m, 2H), 2.41 — 2.31 (m, 2H), 2.20 — 2.10 (m, 4H), 0.92 (s, 6H), 0.74— 0.72 (m, 1H), 0.41 —0.38 (m, 2H), 0.31 — 0.21 (m, 2H); LCMS: Calculated for C24H32N4O: 392.26; Observed: 393.20 (M+1)+.
Step 6: Synthesis of N-(2-aminophenyl)((4-(2-hydroxymethylpropyl)-3,3- dimethylpiperaziny|)methy|)benzamide (Compound 484): To a stirred solution of compound 8b (0.15 g, 1 eq.) in 1,4—dioxane (5vol.), 4M HCI in dioxane (5 vol.) was added.
The resulting reaction mass was stirred at room temperature for 1 h. The reaction completion was monitored TLC. The reaction mixture was concentrated under reduced pressure. The residue was basified with sat. NaH003 solution and extracted with ethyl acetate. The ed organic layers were washed with water and brine, dried over NaZSO4 and concentrated. The crude residue was purified by silica gel column chromatography and preparative HPLC to afford the desired compound 484. 1H NMR (400 MHz, DMSO-d6) 6 9.62 (s, 1H), 7.93 (d, J: 8.0Hz, 2H), 7.42 (d, J = 7.6 Hz, 2H), 7.16 (d, J: 7.6 Hz, 1H), 6.99 — 6.96 (m, 1H), 6.78 — 6.76 (m, 1H), 6.61 — 6.59 (m, 1 H), 4.88 (s, 2H), 3.93 (s, 1 H), 3.32 (s, 2H), 2.74 — 2.72 (m, 2H), 2.46 — 2.32 (m, 2H), 2.10 — 2.00 (m, 4H), 1.05 (s, 6H), 0.93 (s, 6H); LCIVIS: Calculated for C24H34N402: 410.27; Observed: 411.25 (M+1)+.
Synthetic Scheme for Compound 481 and Compound 482: Bfi“ o BOG Boc\ Red. ammn/ / _.u\ N/mH Dfio / 4M HCI in Dioxane Alkylation O/ N —> N K200a MeCN S—>tep-3 Ste -2 1 P 4 RT 15 h Step-1 BocxM/Q NaOH R /fil\k/©)\.n“ NH: —>N OH 7 RN ONQ MeOH:H20(1;1) DIPEA TSP, DCM, RT NBOWRON.» Step-4 Ga-b (For Comp-481) nd-482 HATU Pyridine ACN, nd-481 RT (For 82) Step-5 HO>—A:.
Compound-481 Compound-482 Step 1: Synthesis of utyl (S)(4-(methoxycarbonyl)benzyl) methylpiperazine—1—carboxylate (3): To a d solution of compound 1 (2 g, 1 eq.) and compound 2 (2.29 g, 1 eq.) in ACN (20 mL), potassium carbonate (4.2 g, 3 eq.) was added.
The reaction mixture was stirred at room ature for 16 h. The reaction completion was monitored by TLC. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over anhydrous NaZSO4, filtered and concentrated under d pressure to provide a crude e which was purified by silica gel column chromatography to afford title compound 3.
] Step 2: Synthesis of methyl (S)((2-methylpiperaziny|)methyl)benzoate hydrochloride (3): To a stirred solution of compound 3 (2.8 g, 1 eq.) in 1,4-dioxane (15 mL), 4M HCI in dioxane (5 mL) was added. The reaction mixture was stirred at room temperature for 1 h. The reaction completion was monitored by TLC. The reaction mixture was concentrated and the resulting residue was ated with n—pentane, l ether and dried under vacuum to afford title compound 4 as HCI salt.
Step 3: Synthesis of nd 5a for Compound 481: To a stirred solution of compound 4 (, 1 eq.) and cyclopropyl carboxaldehyde (1.2 eq.) in DCM (10 vol.), acetic acid (6 eq.) was added and stirred at room temperature for 30 min. To this, sodium triacetoxyborohydride (STAB) (3 eq.) was added at room temperature. The resulting reaction mixture was stirred at room temperature for overnight. The reaction tion was monitored by TLC and LCMS. The reaction mixture was quenched with sat. NaHC03 solution and extracted with DCM. The combined organic layers were washed with water and brine, dried over NaZSO4 and evaporated to afford the desired compound 5a.
Step 3: Synthesis of compound 5b for nd 482: To a solution of compound 4( 1 eq.) in l (10 vol.), TEA (3 eq.) and 2,2-dimethyloxirane ( 1.5 eq.) were added and the reaction mixture was heated at 80 °C for 12 h. The reaction completion was monitored by TLC. The reaction mixture was allowed to cool, concentrated to afford the desired compound 5b.
Step 4: General procedure for synthesis of compound 6a-b: To a stirred solution of compound 5 (1.0 eq.) in ol:water (1 :1) was added NaOH (1.5 eq.) at room temperature. The above mixture was heated to 90 0C for 5 h. The reaction completion was monitored by TLC. The reaction e was concentrated and the resulting residue was dissolved in water and washed with diethyl ether. The aqueous layer was neutralized to pH = 7 using 1N HCI at 0 0C. The solid obtained was filtered, washed with water and dried under vacuum to afford the desired nd 6.
Structure Step 5: Synthesis of compound 8a for Compound 481: To a stirred solution of compound 6a (1 eq.) and compound 7 (1 eq.) in DCM (10 vol.), DIPEA (2 eq.) T3P (1.5 eq.) was added at room temperature. After stirring the reaction mixture at ambient temperature for overnight, the reaction mixture was portioned n DCM and water. The combined organic extracts were washed with water and brine, dried over Na2SO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the desired compound 8a.
Step 5: sis of compound 8b for Compound 482: To a d solution of compound 6b (1 eq) and compound 7 (1.1 eq.) in ACN (10 vol.), pyridine (5 eq.) and HATU (1.5 eq.) were added at room temperature. After stirring the reaction mixture at 80 °C for overnight, the reaction mixture was , concentrated and resulting residue was partitioned between water and ethyl acetate. The combined organic extracts were washed with water and 1% HCI to remove traces of pyridine, dried over Na2SO4 and concentrated.
The crude residue was purified by silica gel column chromatography to afford d compound 8b.
Structure Step 6: Synthesis of (S)-N-(2-aminophenyl)((4-(2-hydroxymethylpropyl) methylpiperaziny|)methy|)benzamide (Compound 482): To a stirred solution of compound 8b (1 eq.) in 1,4-dioxane (5vo|.), 4M HCI in dioxane ) was added. The resulting on mass was stirred at room temperature for 1 h. The reaction completion was monitored TLC.
The reaction mixture was concentrated under reduced pressure. The crude residue was purified by preparative HPLC to afford the desired compound 482. 1H NMR (400 MHz, DMSO-dB) 6 9.60 (s, 1H), 7.91 (d, J: 7.6 Hz, 2H), 7.40 (d, J = 8.0 Hz, 2H), 7.18— 7.11 (m, 1H), 6.97 —6.93 (m, 1H), 6.76 (d, J: 8.0 Hz, 1H), 6.60 —6.56 (m, 1H), 4.87 (s, 2H), 4.03 — 3.92 (m, 2H), 3.24 — 3.22 (m, 1 H), 2.75 — 2.73 (m, 2H), 2.66 — 2.63 (m, 1 H), 2.44 — 2.42 (m, 1 H), 2.29 — 2.19 (m, 1H), 2.19 — 2.04 (m, 4H), 1.08 — 1.02 (m, 9H); LCMS: ated for C23H32N402: 396.25; Observed: 397 (M+1)+.
Step 6: Synthesis of (S)—N-(2-aminophenyl)((4-(cyclopropylmethyl) methylpiperaziny|)methyl)benzamide (Compound-481): To a stirred solution of compound 8a (0.15 g, 1 eq) in 1,4-dioxane (5 vol.), 4M HCI in dioxane (5 vol.) was added. The resulting reaction mass was d at room temperature for 1 h. The reaction completion was monitored TLC. The reaction mixture was concentrated under reduced pressure. The residue was basified with sat. NaH003 solution and extracted with ethyl acetate. The combined organic extracts were washed with water and brine, dried over NaZSO4 and concentrated. The crude residue was purified by silica gel column chromatography and preparative HPLC to afford the desired compound 481. 1H NMR (400 MHz, DMSO-d6) 6 9.61 (s, 1H), 7.93 (d, J: 8.0 Hz, 2H), 7.42 (d, J = 7.6 Hz, 2H), 7.19 — 7.12 (m, 1H), 7.01 —6.92 (m, 1H), 6.78 (d, J: 6.8 Hz, 1H), 6.60 (t, J: 7.6 Hz, 1H), 4.88 (s, 2H), 4.04 — 4.00 (m, 1H), 3.22 — 3.18 (m, 1H), 2.79 — 2.64 (m, 2H), 2.61 — 2.53 (m, 1H), 2.43 — 2.41 (m, 1H), 2.19 —2.05 (m, 4H), 2.00 - 1.92 (m, 1H), 1.08 (d, J: 6.4 Hz, 3H), 0.80 — 0.78 (m, 1H), 0.44 — 0.41 (m, 2H), 0.06 — 0.02 (m, 2H); LCMS: Calculated for N4O: 378.24; Observed: 379.20 (M+1)+. tic Scheme for Compound 489 and nd 490: Br\/©)\O/ O HCI Red ammn./ Boo\N/fiy BDC ..\\ / ‘N 0/ HN ‘ O fl>RAlkl t K:,NH 4M HCIIn Dioxane K,N N K2C03 MeCN Step--3 1 Step--2 Step-_1 4 aiive nemisli/ 0 mmMg NaOH ‘ -“‘\ 7 NH2 \QAOCEMD HECEIIn Me0H1H20(1:1) HATU DIPEA tep--6 RN/j‘fiu 2 Step-4 é DMF rt ' Step--5 Compound-489 Compound-490 Compound-489 Compound-490 ] Step 1: Synthesis of tert—butyl (3R,5S)-4—(4-(methoxycarbony|)benzy|)—3,5- dimethylpiperazine—1-carboxy|ate (3): To a stirred solution of compound 1 (2.1 g, 1 eq.) and compound 2 (2.3 g, 1 eq.) in ACN (20 mL), potassium carbonate (4.1 g, 3 eq.) was added.
The reaction mixture was d at room temperature for 16 h. The on completion was monitored by TLC. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with water and brine, dried over anhydrous NaZSO4, filtered and concentrated under reduced pressure to give a crude residue which was purified by silica gel column chromatography to afford compound 3.
] Step 2: Synthesis of methyl 4—(((2H,6S)-2,6—dimethylpiperazin—1 - yl)methyl)benzoate hydrochloride (4): To a stirred solution of compound 3 (2.5 g, 1 eq.) in 1,4-dioxane (5 mL), 4M HCI in e (3 mL) was added. The reaction mixture was d at room temperature for 1 h. The reaction completion was monitored by TLC. The reaction mixture was trated and the resulting residue was triturated with n—pentane and dried under vacuum to afford the desired compound 4 as HCI salt.
Step 3: Synthesis of compound 5a for Compound-489: To a stirred solution of compound 4 (1 eq.) and cyclopropyl carboxaldehyde (1.2 eq.) in DCM (10 vol.), acetic acid (6 eq.) was added and stirred at room temperature for 30 min. To this, sodium triacetoxyborohydride (STAB) (3 eq.) was added at room temperature. The ing reaction mixture was stirred at room temperature for overnight. The reaction completion was red by TLC and LCMS. The reaction mixture was quenched with sat. NaH003 on and extracted with DCM. The combined organic extracts were washed with water and brine, dried over NazSO4 and evaporated to afford the desired compound 5a.
Step 3: Synthesis of compound 5b for Compound 490: To a solution of compound 4 (1 eq.) in ethanol (10 vol.), TEA (3 eq.) and 2,2-dimethyloxirane (1.5 eq.) were added and the on mixture was heated at 80 0C for 12 h. The reaction completion was monitored by TLC. The reaction mixture was allowed to cool and concentrated to afford the desired compound 5b.
Step 4: General procedure for synthesis of compound 6a-b: To a stirred solution of compound 5 (1.0 eq.) in methano|:water (1 :1) was added NaOH (1.5 eq.) at room temperature. The reaction mixture was heated to 90 0C for 5 h. The reaction completion was monitored by TLC. The reaction mixture was concentrated and the resulting e was dissolved in water and washed with diethyl ether. The aqueous layer was neutralized to pH = 7 using 1N HCI at 0 0C. The solid obtained was filtered, washed with water and dried under vacuum to afford the desired compound 6.
Step 5: General procedure for synthesis of compound 8a-b: To a stirred solution of compound 6 (1 eq.) and compound 7 (1.2 eq.) in DMF (5 mL), DIPEA (3 eq.) was added and stirred for 10 min. To this, HATU (1.5 eq.) was added and the reaction mixture was stirred at room ature for overnight. The reaction progress was monitored by TLC and LClVIS. After completion of reaction, the reaction mixture was partitioned between ethyl acetate and water. The organic layer was washed with water and brine, dried over ous NaZSO4, filtered and evaporated to get the crude t which was purified by silica gel column tography to afford the d compound 8.
Kz/N HN‘Boc I\:/N HN\BOC ] Step 6: Synthesis of N-(2—aminophenyl)(((2132,6S)(cyclopropylmethyl)-2,6- dimethylpiperazin-1—yl)methyl)benzamide (Compound 489): To a stirred solution of compound 8a (1 eq.) in oxane (5 vol.), 4M HCI in dioxane (5vol.) was added. The resulting reaction mass was stirred at room temperature for 1 h. The reaction completion was monitored TLC. The on mixture was concentrated under reduced pressure. The residue was basified with sat. NaHC03 solution and extracted with ethyl acetate. The W0 20182’119362 18 combined organic extracts were washed with water and brine, dried over NaZSO4, filtered and concentrated. The crude e was purified by silica gel column chromatography and preparative HPLC to afford the title compound 489. 1H NMR (400 MHz, DMSO-d6) 6 9.58 (s, 1H), 7.90 (d, J: 7.9 Hz, 2H), 7.48 (d, J = 7.9 Hz, 2H), 7.16 (d, J: 7.2 Hz, 1H), 7.01 — 6.92 (m, 1H), 6.77 (d, J: 6.8 Hz, 1H), 6.61 — 6.59 (m, 1 H), 4.88 (s, 2H), 3.78 (s, 2H), 2.84 — 2.81 (m, 2H), 2.60 — 2.56 (m, 2H), 2.14 — 2.05 (m, 2H), 1.77 (t, J: 10.6 Hz, 2H), 0.92 (d, J: 6.0 Hz, 6H), 0.87 — 0.73 (m, 1 H), 0.49 — 0.38 (m, 2H), 0.10 — 0.03 (m, 2H); LCMS: ated for C24H32N40: ; Observed: 393.20 Step 6: Synthesis of N-(2-aminophenyI)(((2R,6S)(2-hydroxy methylpropyI)-2,6-dimethylpiperazinyl)methyl)benzamide und 490): To a stirred solution of compound 8b (1 eq.) in 1,4-dioxane (5vol.), 4M HCI in dioxane (5 vol.) was added. The resulting reaction mass was stirred at room temperature for 1 h. The reaction completion was monitored TLC. The reaction mixture was concentrated under reduced pressure. The residue was basified with sat. NaH003 solution and extracted with ethyl acetate. The combined organic extracts were washed with water and brine, dried over NaZSO4, filtered and concentrated. The crude residue was purified by silica gel column chromatography and preparative HPLC to afford the title compound 490. 1H NMR (400 MHz, DMSO-d6) 6 9.58 (s, 1H), 7.90 (d, J: 7.9 Hz, 2H), 7.48 (d, J = 7.9 Hz, 2H), 7.15 (d, J: 8.0 Hz, 1H), 6.98 — 6.94 (m, 1H), 6.77 (d, J: 7.2 Hz, 1H), 6.64 — 6.55 (m, 1H), 4.88 (s, 2H), 4.03 (s, 1H), 3.77 (s, 2H), 2.86 — 2.78 (m, 2H), 2.60 — 2.57 (m, 2H), 2.13 (s, 2H), 1.96 (t, J: 10.6 Hz, 2H), 1.07 (s, 6H), 0.89 (d, J: 6.0 Hz, 6H); LCMS: Calculated for C24H34N402: 410.27; Observed: 411.25 (M+1)+.
Synthetic Scheme for Compound 491 and Compound 492: Boc~N/fi 0/ O O \7<“H HCI fio2 Boc\N/Wfio/ / 4M HCI in e HN/mfiko 0\ N STAB, DCM —>s R 1 tep-2 3 4 Step-1 O HRH/q] . R‘N/fifio/ R\N/Wfig}, 7 Boc NH2 RA/Alkylation NaOH steps RN MeOH-Hzo (1.1). % Pyridine, HATU', 90 C, 5h AcN, Reflux 6a-b Step-4 Step-5 O O RN/W 4N Dioxane.HCl N RN/W H RN H HN\ Step-6 RN NH2 8a-b Compound-491 HO > Compound-492 nd-491 nd-492 Step 1: Synthesis of tert—butyl 4—(4-(methoxycarbonyl)benzyI)-3,3- dimethylpiperazine—1-carboxy|ate (3): To a stirred solution of amine compound 2 (0.5 g, 1 eq) and aldehyde 1 (0.421 g, 1.1 eq) in DCM (10 mL), sodium triacetoxyborohydride (STAB) (0.693 g, 1.4 eq) was added. The reaction mixture was stirred at room temperature for overnight; the reaction progress was monitored by TLC and LCMS. After completion of reaction, the reaction mixture was partitioned between DCM and water. The organic layers were separated, washed with water and brine, dried over NaZSO4 and ated to get the crude product which was purified by silica gel column chromatography to afford the desired compound 3.
Step 2: Synthesis of methyl 4-((2,2-dimethylpiperazin-1—yl)methyl)benzoate hloride (4): To a stirred solution of Boc nd 3 (0.6 g, 1 eq) in 1,4-dioxane (5 mL), 4M HCI in dioxane (5 mL) was added. The reaction mixture was stirred at room temperature for 1 h. The reaction progress was monitored by TLC. After completion, the reaction mixture was concentrated and the resulting residue was triturated with ane and dried under vacuum to give the d compound 4 as HCI salt.
Step 3: Synthesis of compound 5a: To a stirred solution of amine compound 4 (1 eq) and corresponding aldehyde (1.2 eq) in DCM (10 vol.), acetic acid (6 eq) was added and 2017/0681 18 stirred at room temperature for 30 min. To this, sodium triacetoxyborohydride (STAB) (3 eq) was added at room temperature. The resulting reaction mixture was stirred at room temperature for overnight; the on progress was monitored by TLC and LCMS. After completion, the reaction mixture was quenched with sat. NaH003 solution and extracted with DCM. The organic layers were separated, washed with water and brine, dried over NaZSO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the desired compound 5a.
Step 3: Synthesis of compound 5b: To a solution of compound 4 (0.4 g, 1 eq) in ethanol (5 vol.), TEA (2.5 eq) and 2,2-dimethyloxirane (1.5 eq) were added and the reaction mixture was heated at 80 °C for 12 h. The progress of reaction was monitored by TLC. After completion, the reaction mixture was allowed to cool, concentrated to give a crude compound which was purified by silica gel column chromatography to afford the desired nd 5b.
No Structure ‘ Wood“5a Step 4: Synthesis of compound 6a—b: To stirred solution of ester compound in Methanol: Water (1:1) was added NaOH (1.5 eq) at room temperature. The above mixture was heated to 90 °C for 5h. The progress of the reaction was monitored by TLC. After completion of reaction, the reaction mixture was concentrated and the ing e was portioned between l ether and water. The aqueous layer was neutralized to pH = 7 using 1N HCI at 0 °C. The solid obtained was ed, washed with water and dried under vacuum to provide the desired nd.
No Structure W0 20182’119362 18 Step 5: Synthesis of compound 8 a—b: To a stirred solution of acid compound 6 (1 eq) and amine (1.1 eq) in ACN, pyridine (6 eq) and HATU (1.5 eq) were added at room temperature. After stirring the reaction mixture at 80 0C for 12 h, the reaction progress was monitored by TLC and LCMS. After completion, the reaction mixture was concentrated and ing residue was partitioned between water and ethyl acetate. The organic layers were ted, washed with water and 1% HCI to remove traces of pyridine, dried over NaZSO4 and concentrated. The crude residue was ed by silica gel column chromatography to provide the desired compound.
Step 6: Synthesis of N—(2-aminophenyl)((4—(2-hydroxymethylpropyl)-2,2— dimethylpiperazin-1—yl)methyl)benzamide (Compound 492): To a stirred solution of Boc compound 8b (1 eq) in 1,4—dioxane ), 4M HCI in dioxane (5 vol.) was added. The resulting reaction mass was d at room temperature for 1 h. The progress of the reaction was monitored TLC. After completion, the reaction mixture was concentrated under reduced pressure. The residue was basified with sat. NaHC03 solution and extracted with ethyl acetate. The organic layers were separated, washed with water and brine, dried over NagSO4 and trated. The crude residue was ed by silica gel column tography /prep. HPLC to provide the desired compound. 1H NMR (400 MHz, DMSO-dB) 6 9.59 (s, 1H), 7.91 (d, J: 8.4 Hz, 2H), 7.43 (d, J = 8.4 Hz, 2H), 7.16 (d, J: 7.6 Hz, 1H), 6.99 — 6.94 (m, 1H), 6.79 — 6.77 (m, 1H), 6.62 — 6.57 (m, 1H), 4.88 (s, 2H), 4.02 (s, 1H), 3.60 — 3.51 (m, 2H), 2.42 — 2.33 (m, 6H), 2.12 (s, 2H), 1.11 — 1.08 (m, 12H); LCMS Calculated for C24H34N402: 410.27; Observed: 411.30 (M + 1)+.
Step 6: Synthesis of N-(2-aminophenyl)—4-((4-(cyc|opropy|methyl)-2,2- dimethylpiperazinyl)methyl)benzamide (Compound 491): To a stirred solution of Boc compound 8a (0.14 g, 1 eq) in 1,4-dioxane (5 vol.), 4M HCI in dioxane (5 vol.) was added.
The resulting reaction mass was stirred at room ature for 1 h. The progress of the reaction was monitored TLC. After completion, the reaction mixture was concentrated under reduced pressure. The residue was basified with sat. NaHC03 solution and extracted with ethyl acetate. The organic layers were separated, washed with water and brine, dried over NazSO4 and concentrated. The crude residue was purified by silica gel column chromatography /prep. HPLC to provide the desired compound. 1H NMR (400 MHz, DMSO-dB) 6 9.59 (s, 1H), 7.91 (d, J: 8.4 Hz, 2H), 7.43 (d, J = 8.4 Hz, 2H), 7.16 (d, J: 7.6 Hz, 1H), 6.98 — 6.94 (m, 1H), 6.79 — 6.77 (m, 1H), 6.61 — 6.59 (m, 1H), 4.87 (s, 2H), 3.60 — 3.52 (m, 2H), 2.34 - 2.25 (m, 6H), 2.12 — 2.10 (m, 2H), 1.12 (s, 6H), 0.88 — 0.75 (m, 1H), 0.49 — 0.39 (m, 2H), 0.09 — 0.03 (m, 2H); LCMS Calculated for C24H32N4O: 392.26; ed: 393.30 (M + 1)+.
Synthetic Scheme for Compound 487 and Compound 488: N’B°° RNAIK N’B°° “H 4M HCI/Dioxane NaOH (S) _U;lam, (S) Mrs?) Step-1 fl) —, HN{flR) ,N R) R’ (R)NNW MeOH1120(1: 1) R Step-2 MeCN,r1, 16h Step4 1 3a-b Za-b . . Step-3 Relative Stereochemlstry Boc\N o o ”7 I: R\ J} RNi'SN y” N (s) OH (R) T3P DIPEA (R) 8a:H/©oc—>HCI.DioxaneStep5 NH2 DCM, [1 63") nd-487 Compound-488 Compound-487 Compound-488 Step 1: Synthesis of compound 2a: To a stirred solution of amine compound 1 (1 eq) and cyclopropyl carboxaldehyde (1.2 eq) in DCM (10 vol.), acetic acid (6 eq) was added and stirred at room temperature for 30 min. To this, sodium triacetoxyborohydride (STAB) (3 eq) was added at room ature. The resulting reaction mixture was stirred at room temperature for overnight; the reaction progress was red by TLC and LCMS. After completion, the reaction mixture was ed with sat. NaHCO3 solution and extracted with DCM. The organic layers were separated, washed with water and brine, dried over NaZSO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the desired compound 2a.
Step 1: Synthesis of compound 2b: To a solution of compound 1 (1 eq) in ethanol (10 mL), TEA (3 eq) and 2,2-dimethyloxirane ( 1.5 eq) were added and the reaction mixture was heated at 80 °C for 12 h. The progress of reaction was monitored by TLC. After completion, the reaction mixture was allowed to cool, concentrated to give a crude compound which was purified by silica gel column chromatography to afford the desired compound 2b.
Step 2: Synthesis of nds 3 a—b: To a stirred solution of Boc compound 3 (1 eq) in 1,4-dioxane (5 vol.), 4M HCI in dioxane(5vo|.) was added. The reaction mixture was stirred at room temperature for 1 h. The reaction progress was red by TLC. After completion, the reaction mixture was concentrated and the resulting residue was triturated with n-pentane and dried under vacuum to give the desired compound 3 as HCI salt.
Step 3: sis of compound 5a—b: To a stirred solution of nd 3 (1 eq) and compound 4 (1 eq) in ACN, potassium carbonate (3 eq) was added. The reaction mixture was stirred at room temperature for 16 h. The progress of reaction was monitored by TLC. After completion, the on mixture was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over anhydrous NazSO4, filtered and concentrated under reduced pressure to provide a crude residue which was purified by silica gel column chromatography to afford compound 5.
No ure 1 N/(lfl o/ 5a 2 HO%/\N/('SN o/ )Q/N Step 4: Synthesis of nd 6a—b: To stirred solution of ester compound in Methanol: Water (1 :1) was added NaOH (1.5 eq) at room temperature. The above mixture was heated to 90 0C for 5h. The progress of the reaction was monitored by TLC. After completion of on, the reaction mixture was concentrated and the resulting residue was partitioned between diethyl ether and water. The aqueous layer was neutralized to pH = 7 using 1N HCI at 0 °C. The solid obtained was filtered, washed with water and dried under vacuum to provide the desired nd.
No Structure 1 Nflfl OH )Q/N 2 HO§/\N78% OH Step 5: Synthesis of compound 8a—b: To a d solution of compound 6 (1 eq) and compound 7 (1.2 eq) in DCM, DIPEA (2 eq) and T3P (1.5 eq) were added at room temperature. The reaction mixture was stirred at room temperature for 12 h. The reaction progress was monitored by TLC and LCMS. After completion of reaction, the reaction e was portioned between DCM and water. The organic layers were ted, washed with water and brine, dried over Na2804 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the desired compound 8.
No Structure 1 V/\Ngsfl N (R) H N HN\ 8a 2 HoyN/(lh N (R) H N HN‘Boc 8b Step 6: Synthesis of N—(2-aminophenyI)(((3R,5S)(cyclopropylmethyI)-3,5- dimethylpiperazin-1—yl)methyl)benzamide (Compound 487): To a stirred solution of Boc compound 8a (0.18 g, 1 eq) in 1,4—dioxane (5 vol.), 4M HCI in dioxane (5 vol.) was added.
The resulting on mass was stirred at room temperature for 1 h. The progress of the reaction was monitored TLC. After completion, the reaction mixture was trated under reduced pressure. The residue was ed with sat. NaHC03 solution and extracted with W0 20182’119362 2017/0681 18 ethyl acetate. The organic layers were separated, washed with water and brine, dried over NaZSO4 and trated. The crude residue was purified by silica gel column chromatography /prep. HPLC to provide the desired compound. ] 1H NMR (400 MHz, DMSO-d6) 6 9.62 (s, 1H), 7.94 (d, J: 8.0 Hz, 2H), 7.42 (d, J = 8.4 Hz, 2H), 7.16 (d, J: 8.0 Hz, 1H), 7.01 — 6.92 (m, 1H), 6.78 (d, J: 8.0 Hz, 1H), 6.64 — 6.55 (m, 1 H), 4.89 (s, 2H), 3.46 (s, 2H), 2.80 — 2.78 (m, 2H), 2.66 — 2.64 (m, 2H), 2.59 — 2.57 (m, 2H), 1.74 (t, J: 10.4 Hz, 2H), 0.95 (d, J: 6.0 Hz, 6H), 0.85 — 0.83 (m, 1 H), 0.43 — 0.39 (m, 2H), 0.07 — 0.06 (m, 2H); LCMS Calculated for C24H32N40: 392.26; Observed: 393.20 (M + 1)+.
Step 6: sis of minophenyl)—4-(((3R,5S)(2-hydroxy methylpropyl)-3,5-dimethylpiperazinyl)methyl)benzamide (Compound 488): To a stirred solution of Boc compound 8b (1 eq) in 1,4—dioxane (5 vol.), 4M HCI in dioxane (5 vol.) was added. The resulting reaction mass was stirred at room temperature for 1 h. The progress of the reaction was monitored TLC. After completion, the reaction mixture was concentrated under reduced pressure. The residue was basified with sat. NaH003 solution and extracted with ethyl acetate. The organic layers were separated, washed with water and brine, dried over NaZSO4 and concentrated. The crude residue was purified by prep. HPLC to provide the desired compound. 1H NMR (400 MHz, DMSO-d6) 6 9.61 (s, 1H), 7.92 (d, J: 7.2 Hz, 2H), 7.42 (d, J = 7.6 Hz, 2H), 7.15 (d, J: 8.0 Hz, 1H), 6.96 (t, J: 7.6 Hz, 1H), 6.77 (d, J: 8.0 Hz, 1H), 6.58 (t, J: 7.2 Hz, 1H), 4.88 (s, 2H), 3.88 (s, 1H), 3.47 (s, 2H), 2.71 — 2.69 (m, 2H), 2.47 — 2.37 (m, 4H), 2.07 — 2.05 (m, 2H), 1.07 — 0.99 (m, 12H); LCMS Calculated for C24H34N402: ; Observed: 411.10(M + 1)*. tic Scheme for Compound 477, Compound 477-lsomer—l, Compound 477- Isomer-II and Compound 478, Compound 478—lsomer-l, Compound 478—Isomer-II xx 4M HCI/DioxaneHCI O HN ,0‘ (Boc)20 ,.\\ , __,.\ / NH —> HN/fi o Step-1 N 1 Step-3 ve streochemistry - 5 R ~ RAIAII' .o“ .~“‘ .u“ Yatlon N/mfio ‘ N N/Q MeOH,H20' 2:)“ A/ij/QAH NHBOC Step-4 Step-5 HATU, Pyridine, 9a_b 6a-b - ACN Step-6 R “x Donane.HCl. ‘N ~‘ N —. NVGA; Step-7 N NH2 Compound-477 8. 47B R = l>—>q‘ Hofl Compound-477 nd-478 Compoundlsomer-l Compoundlsomer-l Compoundlsomer-ll Compoundlsomer-ll a b Step 1: Synthesis of tert—butyl (2R,5S)-2,5-dimethylpiperazinecarboxylate (2): To a stirred solution of compound 1 (0.5 g, 1 eq) in DCM (15 mL) at 0 °C, Boc-anhydride (0.478 g, 0.5 eq) dissolved in DCM was added drop wise. The reaction mixture was stirred at room temperature for 24 h. the reaction progress was monitored by TLC. After completion, the reaction mixture was diluted with water and ted with DCM. The organic layers were ted, washed with water and brine, dried over NaZSO4 and evaporated to afford the desired compound 2.
Step 2: Synthesis of tert—butyl (2R,5S)(4-(methoxycarbonyl)benzyl)-2,5- dimethylpiperazinecarboxylate (4): To a stirred solution of nd 2 (0.85 g, 1 eq) and compound 3 (0.91 g, 1 eq) in ACN (10 mL), potassium carbonate (1.65 g, 3 eq) was added.
The reaction mixture was d at room temperature for 16 h. The ss of reaction was monitored by TLC. After completion, the reaction e was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over anhydrous NaZSO4, filtered and concentrated under reduced re to provide a crude residue which was purified by silica gel column chromatography to afford compound 4.
Step 3: Synthesis of methyl 4-(((2S,5R)-2,5-dimethylpiperazin yl)methyl)benzoate hydrochloride (5): To a stirred solution of Boc compound 4 (0.6 g, 1 eq) in 1,4-dioxane (2 mL) was added 4M HCI in dioxane (1 mL) reaction was stirred at room temperature for 1 h. After completion of reaction, the reaction mixture was concentrated and the ing residue was triturated with n-pentane and dried under vacuum to give the desired nd 5 as HCI salt.
Step 4: Synthesis of nd 6a for compound 477: To a d solution of amine compound 5 (1 eq) and aldehyde (1.2 eq) in DCM (10 vol.), acetic acid (6 eq) was added and stirred at room ature for 30 min. To this, sodium triacetoxyborohydride (STAB) (3 eq) was added at room temperature. The resulting reaction mixture was stirred at room temperature for overnight; the reaction progress was monitored by TLC and LCMS.
After completion, the reaction mixture was quenched with sat. NaH003 solution and extracted with DCM. The organic layers were separated, washed with water and brine, dried over NaZSO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the desired compound 6a.
Step 4: Synthesis of compound 6b for compound 478: To a solution of compound (1 eq) in ethanol (10 vol.), TEA (3 eq) and 2,2—dimethyloxirane (1.5 eq) were added and the reaction mixture was heated at 80 °C for 12 h. The progress of reaction was monitored by TLC. After completion, the reaction mixture was d to cool, concentrated to give a crude compound which was purified by silica gel column chromatography to afford the d nd 6b.
No Structure 1 N 0/ 2 HOflAN/w‘fio/N 6b Step 5: Synthesis of compounds 7a—b: To stirred solution of ester compound in Methanol: Water (1 :1) was added NaOH (1.5 eq) at room temperature. The above mixture was heated to 90 °C for 5h. The progress of the reaction was monitored by TLC. After completion of reaction, the reaction mixture was concentrated and the ing residue partioned between diethyl ether and water. The aqueous layer was neutralized to pH = 7 using 1N HCI at 0 °C. The solid obtained was filtered, washed with water and dried under vacuum to provide the desired compound.
W0 20182’119362 18 Step 6: sis of compounds 9a—b: To a stirred solution of acid compound (1 eq) and amine (1.2 eq) in ACN, pyridine (6 eq) and HATU (1.5 eq) were added at room temperature. The reaction mixture was d at 90 °C for overnight; the reaction progress was monitored by TLC and LCMS. After completion, the reaction mixture was concentrated and resulting residue was portioned between water and ethyl acetate. The organic layers were separated, washed with water and 1% HCI to remove traces of pyridine, dried over Na2SO4 and concentrated. The crude residue was purified by silica gel column chromatography to provide the desired compound.
No Structure Step 7: sis of N-(2-aminophenyI)(((2S,5R)(cyc|opropy|methy|)-2,5- dimethylpiperazin-1—yl)methyl)benzamide (Compound 477): To a stirred solution of Boc compound 9a (, 1 eq) in 1,4-dioxane (5vol.), 4M HCI in dioxane (5 vol.) was added. The resulting reaction mass was stirred at room temperature for 1 h. The progress of the reaction was monitored TLC. After completion, the reaction mixture was trated under reduced pressure. The residue was basified with sat. NaHCOs solution and extracted with ethyl e. The organic layers were separated, washed with water and brine, dried over NazSO4 and concentrated. The crude residue was purified by silica gel column chromatography /prep. HPLC to provide the desired compound. 1H NMR (400 MHz, DMSO-d6) 6 9.61 (s, 1H), 7.93 (d, J: 8.0 Hz, 2H), 7.42 (d, J = 8.0 Hz, 2H), 7.16 (d, J: 7.8 Hz, 1H), 6.97 (t, J: 7.6 Hz, 1H), 6.78 (d, J: 7.6 Hz, 1H), 6.60 (t, J: 7.6 Hz, 1H), 4.88 (s, 2H), 4.07 (d, J: 13.8 Hz, 1H), 3.10 (d, J: 13.8 Hz, 1H), 2.97 (dd, J: 11.5, 2.8 Hz, 1H), 2.59 — 2.56 (m, 2H), 2.42 — 2.29 (m, 1H), 2.31 — 2.19 (m, 1H), 2.10 — 1.97 (m, 2H), 1.77 (t, J: 10.6 Hz, 1H), 1.10 (d, J: 6.0 Hz, 3H), 0.89 — 0.74 (m, 4H), 0.53 — 0.36 (m, 2H), 0.07 — 0.05 (m, 2H); LCMS Calculated for C24H32N40: ; Observed: 393.30 (M + 1)+.
Step 6: Synthesis of compound 9 compound omer-I, compound 477- lsomer-ll and nd 478-lsomer-l, compound 478-lsomer-ll: To a d solution of W0 20182’119362 18 compound 7 (1 eq) and compound 8 (1.2 eq) in DMF (5 mL), DIPEA (3 eq) was added and stirred for 10 min. To this, HATU (0.534 g, 1.5 eq) was added and the reaction mixture was stirred at room temperature for overnight, the reaction progress was monitored by TLC and LCMS. After completion of reaction, the reaction mixture was portioned between ethyl acetate and water. The organic layers were separated, washed with water and brine, dried over NaZSO4 and ated to get the crude product which was purified by silica gel column chromatography to afford the desired compound.
Step 7: Synthesis of N—(2-aminophenyl)((4—(cyclopropylmethyl)-2,5— dimethylpiperazin-1—yl)methyl)benzamide (Compound 477—lsomer-ll): To a stirred solution of Boc compound 9a (1 eq) in 1,4-dioxane (5 vol.), 4M HCI in dioxane (5 vol.) was added. The resulting reaction mass was stirred at room temperature for 1 h. The progress of the reaction was monitored TLC. After completion, the reaction mixture was concentrated under reduced pressure. The residue was ed with sat. NaHC03 solution and extracted with ethyl acetate. The organic layers were ted, washed with water and brine, dried over NaZSO4 and concentrated. The crude residue was ed by chiral prep. HPLC using column YMC ART CELLULOSE-SC, 250 mm x 4.6 mm, 5pm column and delivered as Compound 477-lsomer-l as free base (RT is 17.19) and Compound 477-lsomer-ll as free base (RT is 25.46), where the absolute stereochemistry has yet to be confirmed.
Compound omer-ll as free base, 1H NMR (400 MHz, 6) 6 9.60 (s, 1H), 7.92 (d, J: 7.8 Hz, 2H), 7.40 (d, J: 7.9 Hz, 2H), 7.14 (d, J: 7.8 Hz, 1H), 7.00 — 6.91 (m, 1H), 6.76 (d, J: 8.0 Hz, 1H), 6.63 — 6.54 (m, 1H), 4.87 (s, 2H), 4.06 (d, J: 13.8 Hz, 1H), 3.09 (d, J: 13.7 Hz, 1H), 2.97 (d, J: 11.2 Hz, 1H), 2.35 - 2.26 (m, 4H), 2.06 — 2.04 (m, 1H), 1.77 (t, J: 10.6 Hz,1H), 1.20 — 1.05 (m, 3H), 0.85 — 0.83 (m, 4H), 0.46 - 0.40 (m, 2H), 0.05 — 0.04 (m, 2H), 1H merged in solvent peak; LCMS Calculated for C24H32N40: 392.26; Observed: 393.35 (M + 1)+.
Compound 477-lsomer-l as free base, 1H NMR (400 MHz, 6) 6 9.61 (s, 1H), 7.91 (d, J: 7.2 Hz, 2H), 7.41 (d, J: 8.0 Hz, 2H), 7.15 (d, J: 7.6 Hz,1H),7.01 — 6.91 (m, 1H), 6.76 (d, J: 8.0 Hz, 1H), 6.58 (t, J: 7.5 Hz, 1H), 4.87 (s, 2H), 4.06 (d, J: 13.8 Hz, 1H), 3.08 (d, J: 13.8 Hz, 1H), 2.98 — 2.95 (m, 1H), 2.32 — 2.24 (m, 2H), 2.10 — 1.98 (m, 2H), 1.77 — 1.73 (m, 1H), 1.24 — 1.03 (m, 3H), 0.88 — 0.75 (m, 4H), 0.52 — 0.35 (m, 2H), 0.11 — W0 20182’119362 18 0.03 (m, 2H), 2H merged in solvent peak; LCMS Calculated for C24H32N4O: 392.26; Observed: 393 (M + 1)+.
Step 7: sis of N-(2—aminophenyl)(((2S,5R)(2-hydroxy methylpropyI)-2,5-dimethylpiperazinyl)methyl)benzamide (Compound 478): To a stirred solution of Boc compound 9b (1 eq) in 1,4-dioxane (5 vol.), 4M HCI in dioxane (5 vol.) was added. The resulting reaction mass was stirred at room temperature for 1 h. The progress of the reaction was red TLC. After completion, the reaction mixture was concentrated under reduced pressure. The residue was basified with sat. NaH003 solution and extracted with ethyl acetate. The organic layers were separated, washed with water and brine, dried over NaZSO4 and concentrated. The crude residue was ed by silica gel column chromatography /prep. HPLC to provide the desired compound. 1H NMR (400 MHz, DMSO-d6) 6 9.61 (s, 1H), 7.93 (d, J: 8.0 Hz, 2H), 7.42 (d, J = 8.0 Hz, 2H), 7.20 — 7.13 (m, 1H), 6.98 — 6.95 (m, 1H), 6.78 (d, J: 8.0 Hz, 1H), 6.64 — 6.55 (m, 1H), 4.88 (s, 2H), 4.06 — 3.94 (m, 2H), 3.18 — 3.05 (m, 2H), 2.39 - 2.27 (m, 2H), 2.11 — 2.00 (m, 1H), 1.94— 1.91 (m, 1H), 1.80 (t, J: 10.4 Hz, 1H), 1.07— 1.05 (m, 9H), 0.85 (d, J: 6.4 Hz, 3H), 1H merged in solvent peak; LCMS Calculated for C24H34N402: ; Observed: 411.25 (M + 1)+.
Step 7: Synthesis of N—(2-aminophenyl)((4—(2-hydroxymethylpropyl)-2,5— ylpiperazin-1—yl)methyl)benzamide (Compound 478—lsomer-l): To a stirred solution of Boc compound 9b(1 eq) in 1,4-dioxane (5 vol.), 4M HCI in dioxane (5 vol.) was added. The resulting on mass was stirred at room temperature for 1 h. The progress of the reaction was monitored TLC. After completion, the reaction mixture was concentrated under reduced pressure. The residue was basified with sat. NaH003 on and ted with ethyl acetate. The organic layers were separated, washed with water and brine, dried over NagSO4 and trated. The crude residue was purified by chiral prep. HPLC using column YMC Chiral AMYLOSE-SA, 250 mm x 4.6 mm, 5pm column and red as Compound 478-lsomer-l as free base (RT is 10.52) and Compound 478-lsomer-ll as free base (RT is 13.77), where the absolute stereochemistry has yet to be confirmed.
Compound 478-lsomer-l as free base 1H NMR (400 MHz, DMSO-dB) 6 9.62 (s, 1H), 7.93 (d, J: 7.8 Hz, 2H), 7.42 (d, J: 7.8 Hz, 2H), 7.22 — 7.12 (m, 1H), 7.01 — 6.92 (m, 1H), 6.78 — 6.76 (m, 1H), 6.61 — 6.57 (m, 1H), 4.88 (s, 2H), 4.06 — 3.95 (m, 2H), 3.20 — 3.05 (m, 2H), 2.43 — 2.34 (m, 1H), 2.26 — 2.24 (m, 1H), 2.08 — 2.03 (m, 1H), 1.95 — 1.91 (m, 1H), 1.83 — 1.77 (m, 1H), 1.10 — 1.02 (m, 9H), 0.84 (d, J: 6.1 Hz, 3H), 1H merged in solvent peak; LCMS Calculated for C24H34N402: 410.27; Observed: 411.15 (M + 1)+.
Compound 478-lsomer-ll as free base 1H NMR (400 MHz, DMSO-d6) 6 9.61 (s, 1H), 7.92 (d, J: 7.8 Hz, 2H), 7.41 (d, J: 7.8 Hz, 2H), 7.15 (d, J: 7.6 Hz, 1H), 7.00 — 6.91 (m, 1H), 6.77 (d, J: 8.0 Hz, 1H), 6.58 (t, J: 7.6 Hz, 1H), 4.88 (s, 2H), 4.03 - 3.99 (m, 2H), 3.15 — 3.08 (m, 2H), 2.52 - 2.49 (m, 1 H), 2.38 — 2.36 (m, 1H), 2.26 — 2.24 (m, 1 H), 2.06 — 2.05 (m, 1H), 1.95 — 1.91 (m, 1H), 1.80 — 1.78 (m, 1H), 1.09 — 1.02 (m, 9H), 0.84 (d, J: 6.0 Hz, 3H); LCMS Calculated for C24H34N402: 410.27; Observed: 411.15 (M + 1)+.
Synthetic Scheme for Compound 356 and Compound 359 /\©\002Me HZND NaOH _> — COZMG COOH NaH THFrf 12h MeOH:HzO HAT” D'PEA Step 1 DMF RT step 2 Step 3 R-x (X=Br/| )/ O RCHO Boc‘N A'kY'a“ N DioxaneHCI HN \ HN~ Fmoc Step4 NFmoc WNQOH\ St5e Fmo Piperidine. DMF WN“IQ— Compound-356 Compound-359 Compound-356 Compound-359 Step 1: Synthesis of tert-butyl 4-(4-(methoxycarbonyl)benzylidene)-2,2- ylpiperidine-1—carboxylate (3): To a stirred solution of compound 2 (3 g, 1.2 eq) in dry THF (20 mL) at 0 °C, NaH (60 %, 0.506 g, 1.2 eq) was added slowly and stirred at same temperature for 30 min. To this solution, compound 1 (2 g, 1 eq) dissolved in dry THF was added slowly. The ing reaction mixture was stirred at room temperature for 12 h. The progress of the reaction was monitored by TLC. After completion, the reaction was quenched with water and extracted with ethyl acetate. The organic layers were separated, dried over NaZSO4 and trated. The crude residue was purified by silica gel column chromatography to provide the e of desired compound 3 (Cis/Trans mixture).
Step 2: Synthesis of 4—((1-(tert—butoxycarbonyI)-2,2-dimethylpiperidin e)methyl)benzoic acid (4): To stirred solution of mixture of compound 3 (2.5 g, 1 eq) in Methanol: Water (1:1, 20 mL), NaOH (0.417 g, 1.5 eq) was added at room temperature. The above mixture was at room temperature for 2 h. The progress of the reaction was monitored by TLC. After completion of reaction, the reaction mixture was concentrated and the resulting residue was neutralized to pH = 7 using 1N HCI at 0 °C. The solid obtained was filtered, washed with water and dried under vacuum to provide the mixture of desired compound.
Step 3: Synthesis of compound 6: To a stirred on of compound 4 (1 g, 1 eq) and amine 5 (0.956 g, 1 eq) in DMF, DIPEA (1.24 mL, 2.5 eq) was added and stirred for 10 min. To this, HATU (1.65 g 1.5 eq) was added and the reaction mixture was stirred at room temperature for ght, the reaction progress was monitored by TLC and LCMS. After completion, the reaction mixture was portioned between ethyl acetate and water. The organic layers were separated, washed with water and brine, dried over NaZSO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the mixture of desired compound.
Step 4: Synthesis of compound 7: To a stirred solution of compound 6 (1.2 g, 1 eq) in oxane, 4M HCI in dioxane was added. The resulting reaction mass was d at room temperature for 1 h. After completion, the reaction mixture was concentrated under reduced pressure and the resulting residue was triturated with diethyl ether and dried under vacuum to give the mixture of title compound 7 as HCI salt.
Step 5: Synthesis of compound 8: To a stirred solution of amine compound 7 (1 eq) and corresponding aldehyde (1.5 eq) in DCM, acetic acid (6 eq) was added and stirred at room temperature for 30 min. To this, sodium triacetoxyborohydride (STAB) (3 eq) was added at room temperature. The resulting on mixture was stirred at room temperature for overnight; the reaction progress was monitored by TLC and LCMS. After completion, the reaction mixture was quenched with sat. NaHCOs solution and extracted with DCM. The organic layers were separated, washed with water and brine, dried over NaZSO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the e of desired nd.
Step 6: Synthesis of compound 9: A mixture of nd 7 (0.32 g, 1 eq) and % piperidine in DMF (2 mL) was stirred at room temperature for 15 min. The progress of the on was monitored by TLC. After completion, the reaction mixture diluted with ice cold water. The solid obtained was filtered, washed with water and dried under vacuum to provide the mixture of desired compound 9.
W0 20182’119362 18 Step 7: Synthesis of N-(2-aminophenyl)—4-((1-(cyc|opropy|methyl)-2,2- dimethylpiperidinyl)methyl)benzamide (Compound 356): To a stirred solution of compound 9a (0.05 g, 1 eq) in methanol (5 mL), 10% Pd/C (10% w/w of substrate, 30 mg) was added and the reaction mixture was stirred under hydrogen atmosphere (balloon re) at room temperature for 1 h. The progress of the reaction was monitored by TLC.
After completion, the reaction mixture was filtered h a pad of celite, the filtrate was evaporated under reduced pressure and the resulting residue was triturated with diethyl ether and ane and then dried under vacuum to afford the title compound. 1H NMR (400 MHz, DMSO-d6) 6 9.59 (s, 1H), 7.90 (d, J: 7.2 Hz, 2H), 7.28 (d, J = 7.2 Hz, 2H), 7.15 (d, J: 8.0 Hz, 1H), 6.96 (t, J: 7.6 Hz, 1H), 6.78 (d, J: 8.0 Hz, 1H), 6.59 (t, J: 7.6 Hz, 1 H), 4.88 (s, 2H), 2.92 — 2.90 (m, 2H), 2.62 — 2.59 (m, 2H), 2.33 — 2.32 (m, 1H), 2.16—2.14 (m, 2H), 1.75 — 1.72 (m, 1H), 1.55— 1.51 (m, 1H), 1.32— 1.07 (m, 5H), 0.80 — 0.78(m, 4H), 0.45 — 0.34 (m, 2H), 0.07 — 0.04 (m, 2H); LCMS Calculated for N30: 391.26; Observed: 391.95 (M + 1)+.
Step 7: Synthesis of N-(2-aminophenyI)((2,2-dimethylpiperidin y|)methy|)benzamide und 359): To a stirred solution of compound 9b (0.08 g, 1 eq) in methanol (5 mL), 10% Pd/C (10% w/w of substrate, 40 mg) was added and the reaction mixture was stirred under hydrogen atmosphere (balloon pressure) at room temperature for 1 h. The progress of the reaction was monitored by TLC. After completion, the reaction mixture was filtered through a pad of celite, the filtrate was evaporated under reduced pressure and the resulting residue was ated with l ether and ane and then dried under vacuum to afford the title compound. 1H NMR (400 MHz, DMSO-d6) 6 9.63 (s, 1H), 8.41 (s, 1H), 7.92 (d, J: 7.8 Hz, 2H), 7.31 (d, J: 7.8 Hz, 2H), 7.16 (d, J: 7.8 Hz, 1H), 6.97 (t, J: 7.6 Hz, 1H), 6.78 (d, J: 7.9 Hz, 1H), 6.60 (t, J: 7.5 Hz, 1H), 4.87 — 4.84 (m, 2H), 3.36 — 3.27 (m, 1H), 2.99 — 2.82 (m, 3H), 2.56 — 2.49 (m, 2H), 2.05 — 1.90 (m, 1 H), 1.66 — 1.48 (m, 2H), 1.24 — 1.10 (m, 6H); LCMS Calculated for C21H27N30: ; Observed: 338.10(M + 1)+.
Synthetic scheme for Compound 357: COzMe HCI / 4M HCI/ dioxane COZMe —> COzMe NaH THF rt 12h Step2 4 Step 1 N,Boc NaOH MeOH: H20 KN Step4 HO COOH HATU DIPEA DMF rt TEA EtOH ' ' Step 5 7o 00, 12 h Step 3 Ho)k/WW4M HCI/ dioxane H0)k/mum-'2 H2 pdyc Step 6 MeOH Step 7 HO>|/\N N Compound-357 Step 2: Synthesis of methyl 4-((2,2-dimethylpiperidinylidene)methyl)benzoate (4): To a stirred on of compound 3 (0.85 g, 1 eq) in 1,4-dioxane (5 mL), 4M HCI in e (10 mL) was added. The resulting reaction mass was stirred at room temperature for 1 h. After completion, the reaction e was concentrated under reduced pressure and the ing residue was triturated with l ether and dried under vacuum to give the mixture of title compound 4 (Cis/Trans mixture) as HCI salt.
Step 3: Synthesis of methyl 4-((1-(2—hydroxy-2—methylpropyl)-2,2- dimethylpiperidinylidene)methy|)benzoate (5): To a solution of compound 4 (0.35 g, 1 eq) in ethanol (10 mL), TEA (0.567 mL, 3 eq) and 2,2—dimethyloxirane (0.42 mL, 3.5 eq) were added and the reaction mixture was heated at 70 °C for 12 h. The progress of on was monitored by TLC. After completion, the on e was allowed to cool, concentrated to afford the mixture of desired compound 5.
Step 4: Synthesis of 4-((1-(2—hydroxy-2—methylpropyl)-2,2—dimethylpiperidin ylidene)methyl)benzoic acid (6): To stirred solution of ester compound 5 (0.4 g, 1 eq) in Methanol: Water (1:1, 10 mL), NaOH (0.073 g, 1.5 eq) was added at room temperature. The above mixture was heated to 70 °C for 3 h. The progress of the reaction was monitored by TLC. After completion of reaction, the reaction mixture was concentrated and the resulting residue was washed with diethyl ether followed by treatment with water. The aqueous layer W0 20182’119362 18 was neutralized to pH = 7 using 1N HCI at 0 °C. The solid obtained was filtered, washed with water and dried under vacuum to provide the mixture of desired compound 6.
Step 5: Synthesis of tert—butyl (2—(4-((1-(2-hydroxymethylpropyl)-2,2— dimethylpiperidinylidene)methyl)benzamido)phenyl)carbamate (8): To a stirred solution of compound 6 (0.38 g, 1 eq) and compound 7 (0.299 g, 1.2 eq) in DMF (7 mL), DIPEA (0.515 mL, 2.5 eq) was added and stirred for 10 min. To this, HATU (0.683 g, 1.5 eq) was added and the on mixture was stirred at room temperature for overnight, the reaction progress was monitored by TLC and LCMS. After completion, the reaction mixture was portioned between ethyl acetate and water. The organic layers were separated, washed with water and brine, dried over NaZSO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the mixture of desired compound 8.
Step 6: Synthesis of N-(2—aminophenyl)—4-((1-(2—hydroxymethylpropyl)-2,2- ylpiperidinylidene)methyl)benzamide (9): To a stirred solution of compound 8 (0.5 g, 1 eq) in 1,4-dioxane, 4M HCI in dioxane was added. The resulting reaction mass was stirred at room temperature for 1 h. After completion, the reaction mixture was concentrated under d pressure and the resulting residue was triturated with diethyl ether and dried under vacuum to give the mixture of title compound 9 as HCI salt.
] Step 7: Synthesis of N-(2-aminophenyI)((1-(2-hydroxymethylpropyl)-2,2- dimethylpiperidinyl)methyl)benzamide (Compound 357): To a d solution of compound 9 (0.09 g, 1 eq) in ol (5 mL), 10% Pd/C (10% w/w of substrate, 40 mg) was added and the reaction mixture was stirred under en atmosphere (balloon pressure) at room temperature for 1 h. The progress of the reaction was monitored by TLC.
After completion, the reaction e was filtered through a pad of celite, the filtrate was evaporated under d pressure and the resulting e was triturated with diethyl ether and n—pentane and then dried under vacuum to afford the title compound. 1H NMR (400 MHz, DMSO-d6) 6 9.59 (s, 1H), 7.89 (d, J: 7.9 Hz, 2H), 7.28 (d, J = 7.9 Hz, 2H), 7.15 (d, J: 7.9 Hz, 1H), 7.01 — 6.92 (m, 1H), 6.77 (d, J: 7.2 Hz, 1H), 6.64 — 6.55 (m, 1H), 4.88 (s, 2H), 3.90 — 3.88 (m, 1H), 2.92 — 2.89 (m, 1H), 2.31 — 2.28 (m, 1H), 1.81 - 1.78 (m, 2H), 1.45 - 1.29 (m, 4H), 1.28 — 1.01 (m, 10H), 0.98 (s, 3H), 2H merged in solvent peak; LCMS Calculated for C25H35N302: 409.27; Observed: 410.15(M + 1)+.
Synthetic Scheme for compound 379: Boc\ N0:0 Br/\©\ A,triethl hos hite \/OO;p/\©\ —3> 4M HCI/Dioxane COzMe 120 °C 30 min Ste'p-1 r COzMe NaH, THF, 80 °C, Step 3 1 2 12 h Step 2 ol>< CENHZ NaOH Water:MeOH C EtaN, EtOH, 90C' 3 h HATU, DIPEA 60°C 12h I N DMF RT 6 St9" 5 Step4 H0>—/ Steps 4M HCI/Dioxane g<~N Dioxane RT MeOH Step 7 Step 8 Compound-379 Step 1:Synthesis of methyl 4-((diethoxyphosphoryl)methyl)benzoate (2): A mixture of compound 1 (10 g, 1 eq) and yl ite (8.96 mL, 1.2 eq) was heated in sealed tube at 120 °C for 30 min. The progress of the reaction was monitored by TLC. After completion, the reaction mixture was trated under reduced pressure to afford the crude compound 2.
Step 2: Synthesis of tert—butyl 4—(4-(methoxycarbonyl)benzylidene)azepane-1 — ylate (4): To a stirred solution of compound 2 (0.5 g, 1 eq) in dry THF (10 mL) at 0 °C, NaH (60 %, 0.063 g, 1.5 eq) was added slowly and stirred at same ature for 30 min. To this solution, compound 3 (0.261 g, 0.7 eq) dissolved in dry THF was added slowly.
The resulting reaction mixture was stirred at 80 °C for 12 h. The progress of the reaction was monitored by TLC. After completion, the reaction was ed with water and extracted with ethyl acetate. The organic layers were separated, washed with water and dried over NaZSO4 and concentrated. The crude residue was purified by silica gel column chromatography to provide the desired compound.
Step 3: Synthesis of methyl 4-(azepanylidenemethyl)benzoate hydrochloride (5): To a stirred solution of Boc compound 4 (2.4 g, 1 eq) in 1,4-dioxane (10 mL) ,4M HCI in dioxane (4 mL) was added and the reaction was stirred at room temperature for 1 h. After completion, the reaction mixture was trated and the resulting residue was triturated with n-pentane and dried under vacuum to give the desired compound 5 as HCI salt.
Step 4: Synthesis of methyl 4-((1-(2-hydroxymethylpropyl)azepan-4— ylidene)methyl)benzoate (6): To a solution of compound 5 (1.6 g, 1 eq) in ethanol (20 mL), TEA (2.75 mL, 3 eq) and 2,2-dimethyloxirane (0.47 g, 1 eq) were added at room temperature and the reaction mixture was heated at 60 °C for 12 h. The ss of reaction was monitored by TLC. After completion, the reaction mixture was allowed to cool, concentrated to give a crude compound which was ed by silica gel column chromatography.
Step 5: Synthesis of 4-((1-(2-hydroxy-2—methylpropyl)azepan ylidene)methyl)benzoic acid (7): To stirred solution of ester compound 6 (1.9 g, 1 eq) in Methanol: Water (1:1, 10 mL), NaOH (0.36 g, 1.5 eq) was added at room temperature. The above mixture was heated to 70 °C for 12 h. The progress of the reaction was monitored by TLC. After completion of reaction, the reaction mixture was concentrated and the resulting residue was washed with diethyl ether followed by treatment with water. The aqueous layer was neutralized to pH = 7 using 1N HCI at 0 °C. The solid obtained was filtered, washed with water and dried under vacuum to provide the desired compound 7.
] Step 6: sis of tert—butyl (2—(4-((1-(2-hydroxymethylpropyl)azepan ylidene)methyl)benzamido)phenyl)carbamate (8): To a stirred solution of compound 7 (1.6 g, 1 eq) and tert—butyl (2—aminophenyl)carbamate (1.1 g, 1 eq) in DMF (10 mL), DIPEA (2.28 mL, 2.5 eq) was added and stirred for 10 min. To this, HATU (3 g, 1.5 eq) was added and the on mixture was stirred at room temperature for overnight, the reaction progress was monitored by TLC and LCMS. After completion, the reaction mixture was partitioned between ethyl acetate and water. The organic layers were separated, washed with water and brine, dried over NaZSO4 and ated to get the crude product which was purified by silica gel column chromatography to afford the d compound 8.
Step 7: Synthesis of N-(2-aminophenyl)—4-((1-(2-hydroxymethylpropyl)azepan- 4-ylidene)methyl)benzamide (9): To a d on of Boc nd 8 (1 g, 1 eq) in 1,4- dioxane (5 mL), 4M HCI in dioxane (2 mL) was added and stirred at room ature for 1 h. After completion, the reaction mixture was concentrated and the resulting residue was triturated with n-pentane and dried under vacuum to provide the desired compound 9 as HCI salt.
Step 8: Synthesis of N—(2-aminophenyl)((1—(2-hydroxymethylpropyl)azepan- 4-y|)methyl)benzamide dihydrochloride (Compound-379): To a stirred solution of 9 (0.2 g, 1 eq) in ol (10 mL), 10% Pd/C (20 mg) was added and the reaction mixture was stirred under hydrogen atmosphere (balloon pressure) at room temperature for 5 h. The progress of the reaction was monitored by TLC. After tion, the reaction mixture was ed through a pad of celite, the filtrate was evaporated under reduced pressure and the resulting residue was triturated with diethyl ether and n-pentane and then dried under vacuum to afford the title nd. 1H NMR (400 MHz, DMSO-dB) 6 10.13 (s, 1H), 9.40 (s, 1H), 8.01 (d, J: 7.8 Hz, 2H), 7.39 (d, J: 8.0 Hz, 1 H), 7.32 (d, J: 7.6 Hz, 2H), 7.18 — 7.16 (m, 2H), 7.05 — 7.03 (m 1H), 3.58 — 3.00 (m, 7H), 2.61 — 2.57 (m, 2H), 1.93 — 1.61 (m, 7H), 1.23 (s, 6H); LCMS Calculated for C24H33N302 (free base): 395.26; Observed: 396.30 (M + 1)+. tic Scheme for Compound 181 and Compound 472: COOMe NaOH —2 Hm? ater K2003 ACN, rt M Step 1 Step 2 L j 0 FmocHN 20 % Piperidine in DMF NH2 HO N Step 4 HATU DIPEA FmOCHN DMF, rt muonmocHN Compound-181 % Piperidine in DMF Step 5 Compound- 472 Step 1: Synthesis of methyl 4—((4-(2-hydroxy—2—methylpropyl)piperidin yl)methyl)benzoate (3): To a d on of compound 1 (1 g, 1 eq) and compound 2 (1.02 g, 1eq) in ACN (20 mL), potassium carbonate (2.6 g, 3 eq) was added. The reaction mixture was stirred at room temperature for 16 h. The progress of reaction was monitored by TLC. After completion, the reaction e was diluted with water and ted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over anhydrous NaZSO4, filtered and concentrated under reduced pressure to afford the crude product which was purified by silica gel column chromatography to afford the desired compound 3.
Step 2: Synthesis of 4-((4-(2-hydroxymethylpropyl)piperidin yl)methyl)benzoic acid and 4-((4-(2-methylpropenyl)piperidinyl)methyl)benzoic acid (4 and 4a): To stirred solution of ester compound 3 (0.3 g, 1 eq) in Methanol: Water (1:1, 10 mL), NaOH (0.02 g, 5eq) was added at room temperature. The above mixture was heated to W0 20182’119362 18 70 0C for 12 h. The progress of the on was monitored by TLC. After completion of reaction, the reaction mixture was concentrated and the resulting residue was washed with diethyl ether followed by treatment with water. The aqueous layer was neutralized to pH = 7 using 1N HCI at 0 °C. The solid obtained was filtered, washed with water and dried under vacuum to afford the mixture of compound 4 and 4a.
Step 3: Synthesis of (9H-fluorenyl)methyl (2-(4-((4-(2-hydroxy methylpropyl)piperidinyl)methyl)benzamido)phenyl)carbamate and (9H-fluorenyl)methyl ((4-(2-methylpropenyl)piperidinyl)methyl)benzamido) pheny|)carbamate (6 and 6a): To a stirred solution of compound 4 and 4a (0.3 g, 1 eq) and compound 5 (0.339 g, 1 eq) in DMF (10 mL), DIPEA (0.45 mL, 2.5 eq) was added and stirred for 10 min. To this, HATU (0.586 g, 1.5 eq) was added and the reaction mixture was stirred at room temperature for overnight, the reaction progress was monitored by TLC and LCMS. After completion, the reaction mixture was portioned between ethyl e and water. The organic layers were separated, washed with water and brine, dried over NaZSO4 and evaporated to get the crude product which was purified by silica gel column tography to afford the desired compound 6 (0.1 g, 16.14%) and compound 6a (0.13 g, 21%).
Step 4: sis of N-(2-aminophenyl)((4-(2-hydroxy methylpropyl)piperidinyl)methyl)benzamide (Compound 181): A solution of compound 6 (0.1 g, 1 eq) in 20% piperidine in DMF (2 mL) was stirred at room temperature for 15 min.
The progress of the reaction was monitored by TLC. After completion, the reaction mixture diluted with ice cold water. The solid obtained was ed, washed with water; pentane and dried under vacuum to provide the desired compound. 1H NMR (400 MHz, DMSO-d6) 6 9.61 (s, 1H), 8.16 (s, 1H), 7.92 (d, J: 7.6 Hz, 2H), 7.40 (d, J: 7.6 Hz, 2H), 7.14 (d, J: 7.6 Hz, 1H), 6.95 (t, J: 7.6 Hz, 1H), 6.76 (d, J: 7.6, Hz, 1H), 6.58 (t, J: 7.2 Hz, 1H), 3.51 (s, 2H), 2.76 —2.72 (m, 2H), 1.96 (t, J: 10.8 Hz, 2H), 1.70 — 1.68 (m, 2H), 1.43 — 1.40 (m, 1H), 1.30— 1.09 (m, 4H), 1.07 (s, 6H); LCMS Calculated for N302 (free base): 381.24; Observed: 382.20 (M + 1)+.
Step 5: Synthesis of N-(2-aminophenyl)((4-(2-methylpropenyl)piperidin yl)methyl)benzamide und 472): A solution of compound 6a (0.13 g, 1 eq) in 20% piperidine in DMF (2 mL) was stirred at room temperature for 15 min. The progress of the reaction was monitored by TLC. After completion, the reaction mixture d with ice cold water. The solid ed was filtered, washed with water; e and dried under vacuum to provide the desired compound. 1H NMR (400 MHz, DMSO-d6) 6 9.51 (s, 1H), 7.92 (d, J: 7.8 Hz, 2H), 7.40 (d, J = 7.8 Hz, 2H), 7.15 (d, J: 7.7 Hz, 1H), 5.95 (t, J: 7.5 Hz, 1H), 5.75 (d, J: 7.9 Hz, 1H), 6.58 (t, J: 7.5 Hz, 1H), 4.95 - 4.87 (m, 1H), 4.71 — 4.50 (m, 1H), 3.50 (s, 2H), 2.81 — 2.71 (m, 2H), 2.13 — 2.00 (m, 1H), 2.02 — 1.86 (m, 2H), 1.67 — 1.44 (m, 7H), 1.30 — 1.24 (m, 1H), 1.17 — 1.02 (m, 1H); LCMS Calculated for C23H29N30 (free base): 363.23; Observed: 364.20 (M + 1)".
] Synthetic Scheme for nd 238 and Compound 241: Boc—N?<>NH BOC‘N NaOH Boc\N NaH DMF MeOHH20 Step 1 Step 2 RN Boc\ Orv/Q350% TFAm DCM >—CHo HNQCNfir“N/Q —> HATU DIPEA NNQC Step 4 HN~P Alkylalion or DMF Red. amination Step 5 Step 3 Com 0 nd-241: P=H 6a: P=6Fmoc for Compound-238 7b: Pp: lll=moc 6b: P= Boc, for nd-241 :N/CIELWPiperidine DMF O RNWBC R‘NQC\ N/QH Step6 N NHz Compound-238 R=V/‘§\ H Compound-238 Compound-241 ] Step 1: Synthesis of tert-butyl 6-(4-(methoxycarbonyl)benzyI)-2,6- diazaspiro[3.3]heptanecarboxylate (3): To a stirred solution of compound 1 (1 g, 1 eq) in dry DMF (10 mL) at 0 °C, NaH (60 %, 0.123 g, 1.5 eq) was added slowly and stirred at same temperature for 30 min. To this solution, compound 2 (0.47 g, 1 eq) was added slowly. The resulting reaction mixture was d at room temperature for 1 h. The progress of the reaction was monitored by TLC. After completion, the reaction was quenched with water and extracted with ethyl acetate. The organic layers were separated, washed with water and dried over Na2SO4 and concentrated. The crude residue was purified by silica gel column chromatography to provide the desired compound.
Step 2: Synthesis of 4-((6-(tert—butoxycarbonyl)-2,6-diazaspiro[3.3]heptan y|)methyl)benzoic acid (4): To d solution of ester compound 3 (0.53 g, 1 eq) in Methanol: Water (1:1, 8 mL), NaOH (0.091 g, 5eq) was added at room temperature. The above mixture was heated to 70 °C for 12 h. The progress of the reaction was monitored by TLC. After tion of reaction, the reaction mixture was concentrated and the resulting residue was washed with l ether followed by treatment with water. The aqueous layer was neutralized to pH = 7 using 1N HCI at 0 °C. The solid obtained was filtered, washed with water and dried under vacuum to afford the title compound 4.
Step 3: Synthesis of compound 6: To a stirred solution of acid nd 4 (1 eq) and corresponding amine 5 (1 eq) in DMF (10 mL), DIPEA (2.5 eq) was added and stirred for 10 min. To this, HATU (1.5 eq) was added and the reaction mixture was stirred at room temperature for ght, the reaction progress was monitored by TLC and LCMS. After completion, the reaction mixture was portioned between ethyl acetate and water. The organic layers were separated, washed with water and brine, dried over NaZSO4 and evaporated to get the crude product which was purified by silica gel column tography to afford the desired compound.
Step 4: sis of compound 7b and Compound 241 : A stirred solution of Compound 6a or 6b (0.01 g, 1 eq) in 50% TFA/DCM (0.5 mL) was stirred at room temperature for 15 min. The progress of the reaction was monitored by TLC. After completion, the reaction mixture was concentrated and the resulting residue was purified by basic resin (sp- carbonate) to afford the desired nd.
‘Fmoc TFA 7b 4—((2,6-diazaspiro[3.3]heptanyl)methyl)-N-(2-aminophenyl)benzamide (Compound-241): Compound delivered as TFA salt. 1H NMR (400 MHz, DMSO-d6) 6 9.63 (s, 1H), 8.69 (s, 1H), 7.93 (d, .1: 7.6 Hz, 2H), 7.36 (d, J: 7.6 Hz, 2H), 7.15 (d, J: 8.0 Hz, 1H), 6.97 (t, J: 7.6 Hz, 1H), 6.78 (d, J: 8.0 Hz, 1H), 6.60 (1, .1: 7.2 Hz, 1H), 4.88 (s, 2H), 4.07 - 4.05 (m, 4H), 3.67 — 3.62 (m, 2H), W0 20182’119362 18 3.43 - 3.32 (m, 4H); LCMS Calculated for C19H22N40 (free base): 322.18; Observed: 322.85 (M + 1)+.
Step 5: Synthesis of compound (9H-fluorenyl)methyl (2-(4-((6- (cyclopropylmethyl)—2,6-diazaspiro[3.3]heptanyl)methyl)benzamido)phenyl)carbamate (8): To a stirred solution of amine compound 7b (1 eq) and corresponding aldehyde (1.2 eq) in DCM, acetic acid (6 eq) was added and stirred at room temperature for 30 min. To this, sodium triacetoxyborohydride (STAB) (3 eq) was added at room temperature. The resulting reaction mixture was stirred at room temperature for ght; the reaction progress was monitored by TLC and LCMS. After completion, the reaction mixture was quenched with sat.
NaH003 solution and extracted with DCM. The organic layers were separated, washed with water and brine, dried over NaZSO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the desired compound.
Step 6: Synthesis of N-(2-aminophenyI)((6-(cyclopropylmethyl)-2,6- piro[3.3]heptany|)methy|)benzamide (Compound—238): A solution of compound 8 (0.04 g, 1 eq) in 20% piperidine in DMF (1 mL) was stirred at room temperature for 15 min.
The progress of the reaction was monitored by TLC. After completion, the on mixture diluted with ice cold water and extracted with 10% MeOH/DCM. The organic layers were separated, washed with sat. NaHCOs solution and brine, dried over NaZSO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the desired compound. 1H NMR (400 MHz, 6) 6 9.61 (s, 1H), 7.91 (d, J: 6.4 Hz, 2H), 7.35 (d, J = 6.4 Hz, 2H),7.15 (d, J: 6.0 Hz, 1H), 6.97 — 6.95 (m, 1H), 6.78 (d, J: 7.6 Hz, 1H), 6.59 — 6.57 (m, 1 H), 4.88 (s, 2H), 3.58 — 3.56 (m, 2H), 3.24 — 3.21 (m, 8H), 2.22 — 2.20 (m, 2H), 1.24 — 1.22 (m, 1 H), 0.39 — 0.31 (m, 2H), 0.05 — 0.04 (m, 2H); LCMS Calculated for N40 (free base): ; Observed: 376.95 (M + 1)+. 2017/0681 18 Synthetic Scheme for Compound 176: O HzN I o 2 NHBoc TFA DCM OH—> N EDCI, HOBt,DMF, DC'V' EN NHBoc ACOH STAB NHBoc OHC Step3 72:21:“ 3 Compound-176 Step 1: Synthesis of tert-butyl (2-(4-formylbenzamido)phenyl)carbamate (3): To a stirred solution of compound 1 (0.5 g, 1 eq) and compound 2 (0.693 g, 1.2 eq) in DMF (5 mL), HOBt (0.45 g, 1 eq) and EDCI HCI (0.64 g, 1 eq) were added. The resulting reaction mixture was stirred at 70 °C for 4h; the reaction progress was monitored by TLC. After completion, the reaction mixture was quenched with water and extracted with ethyl acetate.
The organic layers were separated, washed with sat. NaHC03 solution and brine; dried over NaZSO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the desired compound 3.
Step 2: Synthesis of tert-butyl (2-(4-(azetidinylmethyl)benzamido) phenyl)carbamate (5) To a stirred solution of compound 3 (0.3 g, 1 eq) and compound 4 (0.06 g, 1.2 eq) in DCM (6 mL), acetic acid (0.317 g, 6 eq) was added and stirred at room ature for 30 min. To this, sodium triacetoxyborohydride (STAB) (0.561 g, 3 eq) was added at room temperature. The resulting reaction mixture was d at room temperature for ght; the on progress was monitored by TLC and LCMS. After completion, the reaction mixture was quenched with sat. NaHCOs solution and extracted with DCM. The organic layers were separated, washed with water and brine, dried over NaZSO4 and ated to get the crude product which was purified by silica gel column chromatography to afford the desired compound 5.
Step 3: Synthesis of N-(2—aminophenyl)—4-(azetidinylmethyl)benzamide (Compound 176): A mixture of compound 5 (0.07 g, 1 eq) and 50% TFA/DCM (2 mL) was d at room temperature for 15 min. The progress of the on was monitored by TLC.
After completion, the reaction mixture was concentrated and the resulting residue was triturated with n-pentane, diethyl ether and dried under vacuum to give the desired compound as TFA salt. 1H NMR (400 MHz, DMSO-d6) 6 10.28 (s, 1H), 9.93 (s, 1H), 8.03 (d, J: 7.6 Hz, 2H), 7.58 (d, J: 8.0 Hz, 2H), 7.23 (d, J: 7.6 Hz, 1H), 7.08 (t, J: 7.2 Hz, 1H), 6.95 (d, J: 8.0 Hz, 1H), 6.81 (t, J: 7.2 Hz, 1H), 4.44 — 4.42 (m, 2H), 4. 11 — 3.99 (m, 4H), 2.46 — 2.25 (m, 2H); LCMS Calculated for C17H19N30 (free base): 281.15; Observed: 282.05 (M + 1)+.
] Synthetic Scheme for Compound 171, Compound 172, Compound 174 and nd 175: R NaOH R 0K2003MeCN /l\|l —> | rt R1/ MeOHIH2O, 70 OC, R1/N Step-1' Step-2 4M HCI in Dioxan- R ”/Q ONQ 20% P?preridine 5 ””2 in DMF Amide formation Step--4 R /1 Step--3 6a-d Compounds-171, 172, 174 & 175 R=Boc, Compound-171i172l174 , Compound-175 Compound-171 Compound-172 Compound-174 Compound-175 Step 1: Synthesis of Compound 3: To a stirred solution of respective amine 2 (1 eq) in ACN at 0 °C, potassium carbonate (3 eq) was added. To this, compound 1 (1 eq) was added and the on e was stirred at room temperature for 16 h. The progress of reaction was monitored by TLC. After completion, the reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over anhydrous NaZSO4, ed and concentrated under reduced pressure to afford the crude product which was purified by silica gel column chromatography to afford the desired compound 3a-d.
WO 19362 18 Step 2: Synthesis of nd 4: To stirred solution of corresponding ester compound 3a-d (1 eq) in Methanol: Water (1:1), NaOH (1.5 eq) was added at room temperature. The above e was heated to 70 °C for 12 h. The ss of the reaction was monitored by TLC. After completion of reaction, the reaction mixture was trated and the resulting residue was washed with diethyl ether followed by ent with water.
The aqueous layer was neutralized to pH = 7 using 1N HCI at 0 0C. The solid obtained was filtered, washed with water and dried under vacuum to afford the mixture of compound 4a-d.
Step 3: Synthesis of Compound 6a—c: To a stirred solution of respective acid compound 4a-c (1 eq) and respective amine 5 (1.1 eq) in ACN, pyridine (5 eq) and HATU (1.5 eq) was added at room temperature. After stirring the reaction mixture at 80 °C for overnight, the reaction progress was monitored by TLC and LCMS. After completion, the reaction mixture was concentrated and resulting residue was portioned between water and ethyl acetate. The organic layers were separated, washed with water and 1% HCI to remove traces of pyridine, dried over NaZSO4 and concentrated. The crude residue was purified by silica gel column chromatography to provide the desired compound 6a-c.
Step 3: Synthesis of Compound 6d: To a stirred solution of compound 4d (1 eq) and respective amine 5 (1 eq) in DMF, DIPEA (2.5 eq) was added and stirred for 10 min. To this, HATU (1.5 eq) was added and the reaction mixture was stirred at room temperature for overnight, the reaction progress was monitored by TLC and LCMS. After completion, the reaction mixture was portioned n ethyl acetate and water. The organic layers were separated, washed with water and brine, dried over NaZSO4 and ated to get the crude product which was purified by silica gel column chromatography to afford the desired compound 6d.
] Step 4: Synthesis of minophenyl)—4-(piperidiny|methyl)benzamide (Compound 171): To a stirred on of Boc compound 6a (1 eq) in 1,4-dioxane (5 vol), 4M HCI in dioxane (5 vol) was added. The resulting reaction e was stirred at room temperature for 1 h. The progress of the reaction was monitored by TLC. After completion, the reaction mixture was concentrated and the resulting residue was triturated with n- pentane and dried under vacuum to give desired compound as HCI salt. 1H NMR (400 MHz, DMSO-d6) 6 10.28 (s, 2H), 8.13 (d, J: 7.6 Hz, 2H), 7.74 (d, J = 8.0 Hz, 2H), 7.41 (d, J: 7.6 Hz, 1H), 7.30 — 7.22 (m, 3H), 4.35 (d, J: 4.4 Hz, 2H), 3.30 — 3.27 (m, 2H), 2.88 — 2.85 (m, 2H), 1.85 — 1.65 (m, 5H), 1.37 — 1.35 (m, 1H); LCMS Calculated for C19H23N30 (free base): 309.18; Observed: 309.90(M + 1)+.
Step 4: sis of N-(2-aminophenyl)((4,4—dimethylpiperidin-1 - yl)methyl)benzamide (Compound 172): To a stirred solution of Boc compound 6b (1 eq) in 1,4-dioxane(5 vol), 4M HCI in dioxane (5 vol), was added. The resulting reaction mixture was W0 20182’119362 18 stirred at room temperature for 1 h. The progress of the reaction was monitored by TLC.
After tion, the reaction mixture was concentrated and the resulting e was triturated with n-pentane and dried under vacuum to give desired compound as HCI salt. 1H NMR (400 MHz, DMSO-d6) 6 10.32 (s, 1H), 10.10 (bs, 1H), 8.13 (d, J: 8.0 Hz, 2H), 7.74 (d, J: 7.6 Hz, 2H), 7.42 (d, J: 8.0 Hz, 1H), 7.24 - 7.16 (m, 3H), 4.40 (d, J: .2 Hz, 2H), 3.24 — 3.00 (m, 4H), 1.71 — 1.65 (m, 2H), 1.52 — 1.48 (m, 2H), 1.01 (s, 3H), 0.96 (s, 3H); LCMS Calculated for C21H27N3O (free base): 337.22; Observed: 337.88 (M + 1)+.
Step 4: Synthesis of 4-((3-azaspiro[5.5]undecanyl)methyI)-N-(2- aminophenyl)benzamide (Compound 174): To a stirred solution of Boc compound 6c (1 eq) in 1,4-dioxane (5 vol), 4M HCI in dioxane(5 vol), was added. The resulting reaction mixture was stirred at room temperature for 1 h. The progress of the reaction was monitored by TLC.
After completion, the reaction mixture was concentrated and the resulting residue was triturated with ane and dried under vacuum to give desired compound as HCI salt. 1H NMR (400 MHz, DMSO-dB) 6 10.58 (s, 1H), 10.42 (s, 1H), 8.14 (d, J: 8.0 Hz, 2H), 7.76 (d, J: 8.0 Hz, 2H), 7.48 (d, J: 7.7 Hz, 1H), 7.34 — 7.15 (m, 3H), 4.37 (d, J: 5.2 Hz, 2H), 3.13 — 2.96 (m, 4H), 1.78 — 1.68 (m, 2H), 1.65 — 1.55 (m, 2H), 1.49 - 1.47 (m, 2H), 1.38 — 1.34 (m, 6H), 1.22 — 1.19 (m, 2H); 90.28%; LCMS Calculated for C24H31N30 (free base): 377.25; Observed: 378.01 (M + 1)+.
Step 4: Synthesis of 4-((2-oxaazaspiro[3.5]nonanyl)methyl)-N-(2- aminophenyl)benzamide und 175): A mixture of compound 6d (10 mg, 1 eq) and % piperidine in DMF (0.5 mL) was stirred at room temperature for 15 min. The progress of the on was monitored by TLC. After completion, the reaction mixture d with ice cold water. The solid obtained was filtered, washed with water; pentane and dried under vacuum to provide the desired compound. 1H NMR (400 MHz, 6) 6 9.61 (s, 1H), 7.93 (d, J: 8.0 Hz, 2H), 7.40 (d, J = 8.0 Hz, 2H), 7.16 (d, J: 7.6 Hz, 1H), 7.01 — 6.92 (m, 1H), 6.77 (d, J: 7.2 Hz, 1H), 6.59 (t, J = 7.2 Hz, 1 H), 4.88 (s, 2H), 4.30 — 4.25 (m, 4H), 3.48 (s, 2H), 2.27 — 2.25 (m, 4H), 1.79 — 1.75 (m, 4H); LCMS Calculated for C21H25N302 (free base): 351.19; Observed: 351.80 (M + 1)+.
Synthetic Scheme for Compound 354: Boc—NO=O BACL P—>Ph3 W —H’a PW DMF 0 C 30 mIn COQMe Toluene reflux 002Me 002Me DMF 65 °C overnight Boo/NmC02Me Step 2 Step 1 2 Step 3 4 / NHCbZ NaOH 6©:NHNHCbZ / NHCbZ _—) Boo/m 6.HATU P H 4M HCI in dioxane M OH:H Oe CO H ridine N N —> 2 2 yBoc’ 1,4»dioxane:MeOH (1:1), Hm“HCI ‘” 4 0 ACN 80 00 16 h 7 o n, 4 h Step 5 Step 6 s E / NHCbz NH2 N H H2, Pd/C, n, 1h 11 7 , —. MeOH titanium tetra- isopropoxide O O N\© STAB DCE 60 °C,16h Step 8 Step 7 nd-354 OH CHO PCC, DCM, RT @ Step 9 Step 1: Synthesis of methyl 4-((bromotriphenyI—l5-phosphanyl)methyl)benzoate (2): To a d solution of compound 1 (50 g, 1 eq) in toluene (500 mL), triphenyl phosphine (55.5 g, 1 eq) was added and the reaction mixture was heated at reflux for 17 h.
After 17 h, the reaction e was d to cool to room temperature, the precipitate was filtered, washed with toluene followed by hexane and dried under vacuum to afford the title compound 2.
Steps 2 and 3: Synthesis of tert—butyl 4—(4-(methoxycarbonyl)benzylidene) piperidine-1—carboxylate (4): To a stirred solution of compound 2 (100 g, 1 eq) in DMF (500 mL) at 0 °C, NaH (60 %, 207 g, 1.1 eq) was added slowly and stirred at same temperature for 30 min. To this solution, tert-butyl 4-oxopiperidinecarboxylate (10.75 g, 1.1 eq) was added at 0 °C. The resulting reaction mixture was stirred at 65 °C for overnight. The progress of the on was monitored by TLC. After completion, the reaction was quenched with water and extracted with ethyl acetate. The organic layers were separated, washed with water and dried over NaZSO4 and concentrated. The crude residue was ed by silica gel column chromatography to provide the title compound 4.
Step 4: Synthesis of 4-((1-(tert-butoxycarbonyl)piperidinylidene)methyl)benzoic acid (5): To stirred solution of ester compound 4 (1 g, 1 eq) in Methanol: Water (1:1, 20 mL), NaOH (0.181 g, 1.5 eq) was added at room temperature. The above e was heated to 70 °C for 12 h. The progress of the reaction was monitored by TLC. After completion of reaction, the reaction mixture was concentrated and the resulting residue was washed with W0 20182’119362 18 diethyl ether ed by treatment with water. The aqueous layer was neutralized to pH = 7 using 1N HCI at 0 °C. The solid obtained was filtered, washed with water and dried under vacuum to provide the title compound 5.
Step 5: Synthesis of tert-butyl 4-(4-((2-(((benzyloxy)carbonyl)amino)phenyl) carbamoyl)benzylidene)piperidinecarboxylate (7): To a stirred solution of acid compound (0.85 g, 1 eq) and amine 6 (0.716 g, 1.1 eq) in ACN (16 mL), pyridine (1.05 mL, 5 eq) and HATU (1.53 g, 1.5 eq) was added at room temperature. The reaction mixture was stirred at 80 °C for 16 h; the reaction progress was monitored by TLC. After completion, the reaction mixture was concentrated and resulting residue was diluted with water and extracted with ethyl acetate. The organic layers were separated, washed with water and 1% HCI to remove traces of ne, dried over NaZSO4 and concentrated. The crude residue was purified by silica gel column chromatography to provide the title compound 7.
Step 6: Synthesis of benzyl (2-(4-(piperidinylidenemethyl)benzamido) )carbamate hydrochloride (8): To a stirred on of Boc compound 7 (1 g, 1 eq) in 1,4-dioxane (10 mL), 4M HCI in dioxane (4 mL) was added and the reaction was stirred at room temperature for 1 h. After completion, the reaction e was concentrated and the resulting e was triturated with diethyl ether; acetonitrile and dried under vacuum to give the title compound 8 as HCI salt.
Step 9: Synthesis of (3r,5r,7r)-adamantanecarbaldehyde (11): To a stirred on of compound 10 (1 g, 1 eq) in DCM (10 mL) at 0 °C, PCC (1.42 g, 1.1 eq) was added portion wise. The ing reaction mass was stirred at room temperature for 2 h.
The progress of the reaction was monitored by TLC. After completion, the resulting mixture was filtered over a pad of celite. The filtrate was washed with water; the organic layer was separated; dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide the title compound 11.
] Step 7: Synthesis of benzyl (2-(4-((1-(((3r,5r,7r)-adamantan-1—yl)methyl) piperidin- 4-ylidene)methyl)benzamido)phenyl)carbamate (9): To a stirred solution of compound 8 (0.3 g, 1 eq) and compound 11 (0.155 g, 1.5 eq) in DCE (8 mL) titanium tetra-isopropoxide (Ti(O/Pr)4)(1.04 g, 6 eq) was added at room temperature. After 5 min. STAB (0.298 g, 3 eq) was added and the mixture was heated at 60 0C for 16 h. The progress of the reaction was monitored by TLC and LCMS. After tion, the reaction mixture was diluted with DCM and the resulting mixture was ed over a pad of celite. The filtrate was concentrated and the resulting e was purified by silica gel column chromatography to provide the title compound 9.
Step 8: Synthesis of 4-((1-(((3r,5r,7r)-adamantanyl)methyl)piperidin yl)methyl)-N-(2-aminophenyl)benzamide und 354): To a stirred solution of compound 9 (0.11 g, 1 eq) in methanol (3 mL), 10% Pd/C (50 mg) was added and the reaction mixture was stirred under hydrogen atmosphere (balloon pressure) at room temperature for 1 h. The progress of the reaction was monitored by TLC. After completion, the reaction mixture was ed through a pad of celite, the filtrate was ated under reduced pressure and the resulting residue was purified by prep. HPLC to afford the title compound. 1H NMR (400 MHz, DMSO-d6) 6 9.58 (s, 1H), 7.89 (d, J: 8.0 Hz, 2H), 7.28 (d, J = 7.6 Hz, 2H), 7.15 (d, J: 7.6 Hz, 1H), 6.96 (t, J: 8.0 Hz, 1H), 6.77 (d, .1: 7.2 Hz, 1H), 6.59 (t, J: 7.2 Hz, 1 H), 4.88 (s, 2H), 2.68 — 2.65 (m, 2H), 2.58 — 2.56 (m, 2H), 2.13 — 2.02 (m 2H), 1.90 — 1.83 (m, 5H), 1.67 - 1.57 (m, 6H), 1.50 — 1.40 (m, 9H), 1.27 — 1.21 (m, 2H); LCMS Calculated for C30H39N30 (free base): ; ed: 458.43 (M + 1)+.
Synthetic Scheme for Compound 169: Br PPh3 NaH Ph3P/ Boc —> —*, —> Toluene reflux Bump/O DMF- 0 “c, 30 m'” COzMe COgMe 002m DMF, 65 °c, ght Step 1 Step 2 1 2 3 Step 3 / 4M HCIIn dioxane A/Br / BocN N COzMe 1 4,-MeOHdioxane:(5: 1), COZMe CSzhCOg, DMF, 60°C, i 002Me 4 n 4 h Step 5 5 Step 4 LiOH THF Mm F: :NHBoc me NHB°¢ NOF 4M HCI in dioxane 1 4-dioxane co H—’ , .
MeOHv H201 11,5 h £7 HATU DIPEA 11, 4 h Step 6 DMF rt 30 h Step 8 S1:ep 7 / NH2 HCI N H2, PdiC.
O MeOH 9 Step 9 Compound-169 Step 1: Synthesis of methyl 4—((bromotriphenyl—l5-phosphanyl)methyl)benzoate (2): To a stirred solution of compound 1 (50 g, 1 eq) in toluene (500 mL), triphenyl phosphine (55.5 g, 1 eq) was added and the reaction mixture was heated at reflux for 17 h. After 17 h, the reaction mixture was allowed to cool to room temperature, the precipitate was filtered, washed with toluene followed by hexane and dried under vacuum to afford the title compound 2.
Step 2 and 3: Synthesis of tert-butyl 3-(4-(methoxycarbonyl) benzylidene)azetidinecarboxylate (4): To a d solution of compound 2 (70 g, 1 eq) in dry DMF (350 mL) at 0 °C, NaH (60 % in mineral oil, 7.9 g, 1.4 eq) was added slowly and stirred at same ature for 30 min. To this solution, tert—butyl 3—oxoazetidine—1 — carboxylate (24.4 g, 1 eq) was added at 0 °C. The resulting reaction mixture was stirred at 65 °C for overnight. The progress of reaction was monitored by TLC. After completion, the reaction mixture was cooled to 0 0C and ed with sat. NH4CI solution, the precipitate was filtered. The residue was taken in acetonitrile, d for 10 min and again filtered. The solid washed with itrile. The crude product was purified by silica gel column chromatography to afford the compound 4.
Step 4: Synthesis of methyl 4-(azetidinylidenemethyl)benzoate hydrochloride (5) To a stirred solution of Boc nd 4 (26 g, 1 eq) in 1,4-dioxane: methanol (30 mL: 20 mL) mixture 4M HCI in e (150 mL) was added and the reaction was stirred at room temperature for 4 h. After completion, the reaction mixture was concentrated and the resulting e was triturated with n-pentane, diethyl ether and dried under vacuum to give the d compound 5 as HCI salt.
Step 5: Synthesis of methyl 4-((1-(cyclopropylmethy|)azetidin ylidene)methyl)benzoate (6): To a stirred solution of compound 5 (1.5 g, 1 eq) in DMF (20 mL), cesium carbonate (5.09 g, 2.5 eq) was added and stirred at room temperature for 10 min. To this solution, (bromomethy|)cyc|opropane (0.847 g, 1 eq) was added. The ing reaction mixture was stirred at 60 °C for 16 h. The progress of the reaction was monitored by TLC. After completion, the reaction was quenched with ice cold water and extracted with ethyl acetate. The organic layers were separated, washed with water and dried over NaZSO4 and concentrated. The crude residue was purified by silica gel column chromatography to provide the d compound 6.
Step 6: Synthesis of 4—((1-(cyclopropy|methyl)azetidin—3-ylidene)methy|)benzoic acid (7): To stirred solution of ester compound 6 (0.53 g, 1 eq) in MeOH:THF (1 :3) mixture, aqueous LiOH (0.258 g, 3 eq, dissolved in 0.75 mL of water) was added. The reaction mixture was stirred at room temperature for 5 h. The progress of the on was monitored by TLC. After completion, the reaction e was concentrated and the resulting residue was taken in water and acidified to pH = 6 using 2N HCI . The solid obtained was filtered, washed with water and dried under vacuum to provide the desired compound 7.
Step 7: Synthesis of tert-butyl (2-(4-((1-(cyclopropylmethyl)azetidin y|idene)methy|)benzamido)—5-f|uoropheny|)carbamate (8): To a stirred solution of compound 7 (0.4 g, 1 eq) and tert-butyl (2-aminofluorophenyl)carbamate (0.407 g, 1.1 eq) in DMF (10 mL), DIPEA (0.845 g, 4 eq) was added and stirred for 10 min. To this, HATU (0.116 g, 1.5 eq) was added and the reaction mixture was stirred at room temperature for overnight, the reaction ss was monitored by TLC. After completion, the on mixture was diluted with water and extracted with ethyl acetate. The organic layers were separated, W0 20182’119362 18 washed with water and brine, dried over NaZSO4 and evaporated to get the crude product which was purified by silica gel column chromatography to afford the desired compound 8.
Step 8: Synthesis of N-(2-aminofluorophenyI)((1- (cyclopropylmethyl)azetidinylidene)methyl)benzamide dihydrochloride (9): To a stirred solution of Boc compound 8 (0.18 g, 1 eq) in 1,4-dioxane (1 mL), 4M HCI in dioxane (3 mL) and the reaction was stirred at room ature for 4 h. After completion, the reaction mixture was concentrated and the ing residue was ated with diethyl ether, acetonitrile and dried under vacuum to give the desired compound 9 as HCI salt.
Step 9: Synthesis of N—(2-aminofluorophenyI)((1—(cyclopropylmethyl) azetidinyl)methyl)benzamide (Compound-169): To a stirred solution of 9 (0.14 g, 1 eq) in methanol (10 mL), 10% Pd/C (30 mg) was added and the reaction e was stirred under hydrogen atmosphere (balloon pressure) at room temperature for 2 h. The progress of the reaction was red by TLC. After completion, the on e was filtered through a pad of celite, the filtrate was evaporated under reduced pressure and the resulting residue was triturated with diethyl ether and acetonitrile and then dried under vacuum to afford the title compound. 1H NMR (400 MHz, DMSO-d6) 6 9.52 (s, 1H), 7.89 (d, J: 8.0 Hz, 2H), 7.29 (d, J = 7.6 Hz, 2H), 7.11 — 7.07 (m, 1H), 6.55 — 6.51 (m, 1H), 6.37 — 6.33 (m, 1H), 5.22 (s, 2H), 3.27 (t, J: 6.8 Hz, 2H), 2.88 — 2.85 (m, 2H), 2.80 (t, J: 6.4 Hz, 2H), 2.64 — 2.61 (m, 1 H), 2.21 — 2.20 (m, 2H), 0.68 — 0.66 (m, 1 H), 0.40 — 0.29 (m, 2H), 0.06 — 0.04 (m, 2H); LCMS Calculated for C21H24FN30 (free base): 353.19; Observed: 353.90 (M + 1)+. ] tic Scheme for Compound 161, Compound 162, Compound 163: Br PPh3 BrPhaP NaH PhsP/ 0\ Toluene, reflux, 17h 0\ DMF 0 “,C c’ Step 1 30 min 1 0 O OO\—>DMF 65 “,C ght Step 2 Step 3 R-XorR-CHO Rm:\ 4M HCI'In dioxane NaOH, Water:MeOH RIO/mo}: 1 xane: MeOH Alkylation OR 90°C 3 h (4)1) rt 4 h OO\ Red. amination Step 6 StepN4 Step 5 /©:NHBDCH2R%N HCI HCI NHBoc 4M HCIIn dioxane NH? NH? H2 Pd/C HATUDPledine 14—dioxane: MeOH ACN, 850C rt 4 h FStep 9 3‘94) 7 (Step 8 Compound161 to 163 I:Boc anhydride F,TFA DCM H Rane “f”: y Ni 02N DMAP, THF, 02N Step 110 Step 12 NB0‘32 step1o c NHBoo NHBoc Raga; >m: var nd-181 Compound-162 Compound-183 Step 1: Synthesis of methyl omotriphenyI—l5-phosphanyl)methyl)benzoate (2): To a stirred solution of compound 1 (50 g, 1 eq) in toluene (500 mL), triphenyl phosphine (55.5 g, 1 eq) was added and the reaction mixture was heated at reflux for 17 h.
After 17 h, the reaction mixture was allowed to cool to room temperature, the precipitate was filtered, washed with toluene ed by hexane and dried under vacuum to afford the title compound 2.
Steps 2 and 3: Synthesis of tert—butyl 4—(4-(methoxycarbonyl) benzylidene)piperidinecarboxy|ate (4): To a stirred solution of compound 2 (100 g, 1 eq) in DMF (500 mL) at 0 °C, NaH (60 %, 207 g, 1.1 eq) was added slowly and stirred at same temperature for 30 min. To this solution, tert-butyl 4-oxopiperidinecarboxylate (10.75 g, 1.1 eq) was added at 0 °C. The resulting reaction mixture was stirred at 65 0C for overnight.
The progress of the reaction was monitored by TLC. After completion, the on was quenched with water and extracted with ethyl acetate. The organic layers were separated, washed with water and dried over NaZSO4 and concentrated. The crude residue was purified by silica gel column tography to provide the title compound 4.
Step 4: Synthesis of methyl 4-(piperidinylidenemethy|)benzoate hydrochloride (5): To a stirred solution of Boc compound 4 (11 g, 1 eq) in 1,4-dioxane:methanol (4:1, 200 mL) mixture, 4M HCI in dioxane (120 mL) was added and the reaction was stirred at room temperature for 3 h. After completion, the reaction mixture was concentrated and the resulting residue was triturated with l ether and dried under vacuum to give the title compound 5 as HCI salt.
] Step 5: Synthesis of compound 6a: To a on of compound 5 (0.5 g, 1 eq) in ethanol (10 mL), TEA (0.8mL, 3 eq) and 2,2-dimethyloxirane (0.203 g, 1.5 eq) were added at room temperature and the reaction mixture was heated at 60 °C for 12 h. The progress of reaction was monitored by TLC. After completion, the on mixture was allowed to cool, concentrated to give a crude compound which was purified by silica gel column chromatography.
Step 5: Synthesis of compounds 6b and 6c: To a stirred solution of compound 5 (3.5 g, 1 eq) in DMF (35 mL), cesium carbonate (10.7 g, 2.5 eq) was added and stirred at room temperature for 10 min. To this on, methyl)cyclopropane (1.6 mL, 1.2 eq) was added. The resulting reaction mixture was stirred at 70 °C for 16 h. The ss of the reaction was monitored by TLC. After completion, the reaction was quenched with water and extracted with ethyl acetate. The organic layers were separated, washed with water and dried over NaZSO4 and concentrated. The crude residue was purified by silica gel column chromatography to provide the desired compound.
Step 6: Synthesis of compound 7a—c: To stirred solution of ester compound 6 (1 eq) in Methanol: Water (1:1), NaOH (1.5 eq) was added at room temperature. The above mixture was heated to 70 °C for 12 h. The progress of the on was monitored by TLC.
After completion of reaction, the reaction mixture was concentrated and the resulting residue was washed with diethyl ether followed by treatment with water. The aqueous layer was neutralized to pH = 7 using 1N HCI at 0 °C. The solid obtained was filtered, washed with water and dried under vacuum to provide the desired compound. 1 H%U®YOH 0 7a Step 7: Synthesis of compound 8a—c: To a stirred solution of acid compound 7 (g, 1 eq) and tert-butyl (2-aminofluorophenyl)carbamate (1.1 eq) in ACN, ne (5 eq) and HATU (1.5 eq) was added at room temperature. After stirring the reaction mixture at 80 °C for overnight, the reaction ss was monitored by TLC and LCMS. After completion, the reaction mixture was concentrated and resulting residue was portioned between water and ethyl e. The organic layers were separated, washed with water and 1% HCI to remove traces of pyridine, dried over NaZSO4 and concentrated. The crude residue was ed by silica gel column chromatography to provide the desired compound.
Step 8: Synthesis of compound 9a—c: To a stirred solution of Boc compound 8 (1 eq) in 1,4-dioxane, 4M HCI in dioxane was added and the reaction was stirred at room temperature for 1 h. After completion, the reaction mixture was trated and the resulting residue was triturated with n-pentane and dried under vacuum to give the desired compound as HCI salt. 2 10%» O “(1 3 HCI 0 ”f1 Step 10: Synthesis of compound B: To a stirred solution of compound A (5 g, 1 eq) in THF (100 mL), DMAP (0.312 g, 0.08 eq) and Boc anhydride (17.4 g, 2.5 eq) dissolved in THF was added. The resulting reaction mixture was d at room temperature for 16 h.
The progress of the reaction was monitored by TLC. After completion, the reaction was quenched with sat. NaHC03 on and extracted with ethyl acetate. The organic layers were separated, washed with water and dried over NaZSO4 and concentrated to provide the desired compound B.
] Step 11: Synthesis of tert—butyl (5-fluoro—2-nitrophenyl)carbamate (C): To a stirred solution of compound B (11 g, 1 eq) in DCM (110 mL) at 0 °C, TFA (3.5 mL, 1.5 eq) was added. The resulting reaction mixture was stirred at room temperature for 1 h. The progress of the reaction was monitored by TLC. After completion, the reaction was quenched with sat. NaHCOa solution and extracted with ethyl acetate. The organic layers were separated, washed with water and dried over NaZSO4 and concentrated to provide the desired compound C.
Step 12: sis of tert-butyl (2-aminofluorophenyl)carbamate (D): To a stirred solution of compound C (4 g, 1 eq) in dry THF (100 mL) under argon atmosphere, Raney Ni (2 g) was added and the reaction mixture was stirred under en atmosphere on pressure) at room temperature for ght. The progress of the reaction was monitored by TLC. After completion, the reaction mixture was filtered through a pad of celite, the filtrate was evaporated under reduced pressure and the resulting residue was ated with diethyl ether and n-pentane and then dried under vacuum to afford the title compound Step 9: Synthesis of minofluorophenyI)((1-(2-hydroxy methylpropyl)piperidinyl)methyl)benzamide (Compound 161): To a stirred solution of 9c (0.05 g, 1 eq) in ol (1 mL), methanolic HCI (1 mL) and 10% Pd/C (5 mg) was added and the reaction e was stirred under hydrogen atmosphere (balloon pressure) at room temperature for 4 h. The progress of the reaction was monitored by TLC. After completion, the reaction mixture was filtered through a pad of celite, the filtrate was evaporated under W0 20182’119362 18 reduced pressure and the resulting residue was triturated with diethyl ether and ane and then dried under vacuum to afford the title compound. ] 1H NMR (400 MHz, DMSO-d6) 6 9.84 (s, 1H), 9.18 (bs, 1H), 7.98 (d, J: 7.6 Hz, 2H), 7.33 (d, J: 7.6 Hz, 2H), 7.22 — 7.20 (m, 1H), 6.76 — 6.74 (m, 1H), 6.59 — 6.57 (m, 1H), 3.57 — 3.54 (m, 2H), 3.29 — 3.25 (m, 1 H), 3.16 — 3.14 (m, 2H), 3.05 — 2.89 (m, 4H), 2.72-2.65 (m, 1H), 2.60 (d, J: 5.9 Hz, 1H), 1.78 — 1.68 (m, 4H), 1.25 (s, 6H); LCMS Calculated for C23H30FN302 (free base): 399.23; Observed: 399.95 (M + 1)+.
Step 9: Synthesis of N-(2-aminofluorophenyI)((1-(cyclopropylmethyl) piperidiny|)methy|)benzamide (Compound 163): To a d solution of 9b (0.05 g, 1 eq) in methanol (1 mL), 10% Pd/C (5 mg) was added and the reaction mixture was stirred under hydrogen atmosphere (balloon pressure) at room ature for 4 h. The progress of the reaction was monitored by TLC. After completion, the reaction mixture was filtered through a pad of celite, the filtrate was evaporated under d pressure and the resulting residue was triturated with diethyl ether and n-pentane and then dried under vacuum to afford the title compound. 1H NMR (400 MHz, DMSO-dB) 6 10.19 (s, 1H), 9.80 (s, 1H), 7.97 (d, J: 8.0 Hz, 2H), 7.33 (d, J: 7.6 Hz, 2H), 7.21 (t, J: 7.6 Hz, 1H), 6.74 — 6.71 (m, 1H), 6.57 - 6.55 (m, 1H), 3.57 — 3.44 (m, 2H), 2.92 — 2.73 (m, 4H), 2.67 — 2.63 (m, 2H), 1.87 — 1.70 (m, 3H), 1.64 — 1.49 (m, 2H), 1.19 — 1.02 (m, 1H), 0.63 — 0.61 (m, 2H), 0.45 — 0.31 (m, 2H); LCMS Calculated for C23H23FN3O (free base): 381.22; Observed: 381.95 (M + 1)*.
Step 9: Synthesis of N N—(2-aminofluorophenyl)((1—neopentylpiperidin-4— yl)methyl)benzamide (Compound-162): To a stirred solution of 9 (0.1 g, 1 eq) in methanol (5 mL), 10% Pd/C (10 mg) was added and the reaction mixture was stirred under en atmosphere (balloon pressure) at room temperature for 4 h. The progress of the reaction was monitored by TLC. After completion, the reaction mixture was filtered through a pad of celite, the filtrate was evaporated under reduced re and the resulting residue was purified by combiflash and SFC chromatography to afford the desired compound. 1H NMR (400 MHz, DMSO-d6) 6 9.51 (s, 1H), 7.89 (d, J: 8.0 Hz, 2H), 7.28 (d, J = 8.0 Hz, 2H), 7.10 (t, J: 8.4 Hz, 1H), 6.55 — 6.52 (m, 1H), 6.38 — 6.33 (m, 1H), 5.20 (s, 2H), 2.74 —2.70 (m, 2H), 2.58 — 2.56 (m, 2H), 2.10 (t, J: 11.2 Hz, 2H), 1.49 — 1.46 (m, 3H), 1.24— 1.22 (m, 2H), 1.04 —1.02 (m, 1H), 0.82 (s, 9H), 1H merged in t peak; LCMS Calculated for FN30 (free base): 397.25; Observed: 398.37 (M + 1)+.
] Synthetic Scheme for Compound 146 and Compound 147: 4M HCI in dioxane K2003, ACN, 0°C-rt, 16h Step-2 HU Step-1 3 Boc\NI) R—X OR R-CHO Rs NaOH —> N —* R‘N/fi OH H—, Alkylation or MeOH:H20 N HATU, pyridine, Red. amination Step-4 CH3CN, reflux Step-3 - - Step-5 R N/fi 4M HCI'In dioxane R K/N H HN\ Step--6 NOK/HC| NHQ BOG HCI 6a-b Compound-146 Compound-147 Compound-146 Compound-147 Step 1: Synthesis of tert—butyl 4—(4-(methoxycarbonyl)benzy|)piperazine-1 - carboxylate (2): To a stirred solution of tert-butyl piperazinecarboxylate (2.92 g, 1.2 eq) and potassium carbonate (3.33 g, 3 eq) in ACN (25 mL), compound 1 (3 g, 1 eq) was added.
The reaction mixture was d at room temperature for 16 h. The progress of reaction was monitored by TLC. After completion, the on mixture was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over anhydrous , filtered and trated under reduced re to e a crude residue which was purified by silica gel column chromatography to afford compound 2.
Step 2: Synthesis of methyl 4-(piperaziny|methyl)benzoate hydrochloride (3): To a stirred solution of Boc compound 2 (4 g, 1 eq) in 1,4-dioxane (2 mL), 4M HCI in dioxane was added and reaction was stirred at room temperature for 1 h. After completion of reaction, the reaction mixture was concentrated and the resulting residue was triturated with n-pentane and dried under vacuum to give the desired compound 3.
Step 3: Synthesis of nd 4a: To a solution of compound 3 (1 eq) in 5 vol of ethanol was added TEA (3 eq) followed by 2,2-dimethyloxirane (2.5 eq) at room ature and the reaction mixture was heated at 90 °C for 4h. The progress of reaction was monitored by TLC. After completion, the reaction e was allowed to cool, concentrated to give a crude compound which was purified by silica gel column chromatography.
Step 3: Synthesis of compound 4b: To a stirred solution of compound 3 (1 eq) and cesium carbonate (3 eq) in DMF (10 vol), ponding alkyl halide (1.1 eq) was added. The reaction mixture was heated at 80 °C for 12 h. The progress of reaction was monitored by TLC. After completion, the reaction mixture was poured into ice-water and extracted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over anhydrous NaZSO4, filtered and concentrated under reduced re to provide a crude residue which was purified by silica gel column chromatography 1 0 Step 4: Synthesis of compound 5a—b: To stirred solution of ester compound in Methanol: Water (1:1) was added NaOH (1.5 eq) at room temperature. The above mixture was heated to 90 0C for 5h. The ss of the reaction was monitored by TLC. After completion of on, the reaction mixture was concentrated and the resulting e was ved in water and washed with diethyl ether. The s layer was neutralized to pH = 7 using 1N HCI at 0 °C. The solid obtained was filtered, washed with water and dried under vacuum to provide the desired compound.
Step 5: Synthesis of compound 6a—b: To a stirred solution of acid compound (1 eq) and amine (1.1 eq) in ACN (10 vol.), pyridine (5eq) and HATU (1.5 eq) was added at room temperature. After stirring the reaction mixture at 80 °C for overnight, the reaction progress was monitored by TLC and LCMS. After completion, the reaction e was concentrated and resulting residue was partitioned between water and ethyl acetate. The organic layers were separated, washed with water and 1% HCI to remove traces of ne, dried over NaZSO4 and concentrated. The crude residue was purified by silica gel column chromatography to provide the desired compound.
WO 19362 18 ] Step 6: Synthesis of N-(2-aminophenyI)((4-(2-hydroxy methylpropyl)piperazinyl)methyl)benzamide (Compound 146): To a stirred solution of Boc compound 6a (1 eq) in 1,4-dioxane (5 vol.) 4M HCI in dioxane (5 vol)at room temperature.
After completion of reaction, the reaction mixture was concentrated and the resulting residue was triturated with n-pentane and dried under vacuum to provide the desired compound. 1H NMR (400 MHz, DMSO-d6): 6 10.65 (s, 1H), 8.22 (d, J: 7.8 Hz, 2H), 7.82 (d, J: 7.8 Hz, 2H), 7.63 — 7.56 (m, 1H), 7.48 (d, J: 7.5 Hz, 1H), 7.41 — 7.28 (m, 2H), 4.49 (s, 2H), 3.76 — 3.66 (m, 4H), 3.62 — 3.56 (m, 4H), 3.21 — 3.16 (m, 2H), 1.26 (s, 6H); LCMS Calculated for 022H30N402 for free base: ; Observed: 382.90 (M + 1)+.
Step 6: Synthesis of N-(2-aminophenyI)((4-(cyclopropylmethyl)piperazin yl)methyl)benzamide (Compound 147): To a stirred solution of Boc compound 6b (1 eq) in 1,4-dioxane (5 vol.) 4M HCI in dioxane (5 vol.)at room temperature. After completion of reaction, the reaction mixture was concentrated and the resulting residue was triturated with n-pentane and dried under vacuum to provide the desired compound. 1H NMR (400 MHz, DMSO-d6):611.72(s, 1H), 10.53 (s, 1H), 8.18 (d, J: 7.8 Hz, 2H), 7.80 (d, J: 7.8 Hz, 2H), 7.56 — 7.49 (m, 1H), 7.42 — 7.34 (m, 1 H), 7.31 — 7.30 (m 2H), 4.42 (s, 2H), 3.72 — 3.70 (m, 2H), 3.55 — 3.44 (m, 6H), 3.10 — 3.02 (m, 2H), 1.11 — 1.09 (m, 1H), 0.63 — 0.62 (m, 2H), 0.41 -0.39 (m, 2H); LCMS ated for szH23N4O for free base: 364.23; Observed: 365.15 (M + 1)+.
Synthetic Scheme for Compound-555 \/0FAQ 0'/\©\ O,NaOH MeOH:H20((),1:1 BOO, 002Me BOON 60 0C, 3 h H2N5 BocN NHFmoc \ HATU, DIPEA o Nal—l, THF, 00C: to StePZ DMF 12h. rt 1 r.t, 12h Step 1 Step 3 i)>—/O :HQ —.4M HCI'In dioxane CchOOH, DCM, 0'”HN GHQ4 30 min NHFmoc Step 4 NHFmOC ii) STAB, 12 h, r.t., Step 5 be, 0 D: o % piperidine N N N N in DMF, 0.5 h H NHFmoc NH2 Step 6 Compound-555 W0 20182’119362 18 Step 1: Synthesis of tert—butyl (Z)(4-(methoxycarbonyl)benzylidene)pyrrolidine - 1-carboxylate (3): Step 1a: Synthesis of methyl ethoxyphosphoryl)methyl)benzoate (2): A mixture of methyl 4-(bromomethyl)benzoate (10 g, 43.66 mmol, 1 eq) and triethyl phosphite (10.8 g, 65.50 mmol, 1.5 eq) was heated in seal tube at 130 °C for 12 h. The progress of the reaction was monitored by TLC. After completion, the reaction mixture was concentrated under reduced pressure. The crude residue was purified by silica gel column chromatography to afford the title compound 2.
Step1 b: Synthesis of tert-butyl (Z)(4—(methoxycarbonyl)benzylidene) idine-1—carboxy|ate (3): To a stirred solution of compound 2 (17 g, 59.39 mmol, 1.1 eq) in anhydrous THF (100 mL) at 0 °C, NaH (3.88 g, 60% w/win mineral oil, 80.98 mmol, 1.5 eq) was added under N2 atmosphere. After stirring the reaction e for 30 min, a solution of compound 1 (10 g, 53.99 mmol, 1 eq) in THF was added at 0 °C. The reaction e was then stirred at room temperature for 12 h. The progress of reaction was monitored by TLC. After completion, the reaction e was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over anhydrous NaZSO4, filtered, and concentrated under reduced pressure to afford a crude residue which was purified by silica gel column chromatography to afford the title compound Step 2: Synthesis of (Z)—4-((1-(tert-butoxycarbonyl)pyrroIidin-3—ylidene)methyl) benzoic acid (4): To a stirred on of compound 3 (4 g, 12.62 mmol, 1 eq) in methanol: water (1:1, 20 mL), NaOH (0.757 g, 18.92 mmol, 1.5 eq) was added and the reaction mixture was stirred at 60 0C for 3 h. The progress of reaction was monitored by TLC. After completion, methanol was d under reduced pressure and reaction mixture was acidified with 2N HCI up to pH ~ 5, during which a solid itated. The obtained solid was filtered, washed with water, and dried under reduced pressure to afford the title compound 4.
Step 3: Synthesis of tert-butyl (4-((2-((((9H-fluorenyl)methoxy)carbonyl) amino)phenyl)carbamoyl)benzylidene)pyrrolidinecarboxy|ate (6): Step 3a: Synthesis of (9H-fluorenyl)methyl (2-aminophenyl)carbamate (5): To a d solution of benzene-1,2-diamine (5 g, 46.29 mmol, 1 eq) in DMF (20 mL), a solution of FmocOSu (15.60 g, 46.29 mmol, 1 eq) in DMF (50 mL) was added slowly. The on mixture was stirred at room temperature for 2 h. The progress of the reaction was monitored by TLC. After completion, the on mixture was quenched with water. The precipitated solid was collected by filtration and dried under reduced pressure. The crude compound was purified by silica gel column chromatography to afford the title compound 5.
W0 20182’119362 18 Step 3b: tert-butyl (Z)-3—(4-((2-((((9H-f|uorenyl)methoxy)carbonyl)amino) phenyl)carbamoyl)benzylidene)pyrro|idine—1-carboxylate (6): To a stirred solution of compound 4 (3.8 g, 12.5 mmol, 1 eq) and compound 5 (4.96 g, 15.04 mmol, 1.2 eq) in DMF (20 mL), DIPEA (5.39 mL, 31.35 mmol, 2.5 eq) was added and stirred for 10 min. To this solution, HATU (7.15 g, 18.31 mmol, 1.5 eq) was added slowly and the reaction mixture was stirred at room temperature for 12 h. The progress of the reaction was monitored by TLC.
After completion, the reaction e was d with water and ted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over anhydrous NaZSO4, filtered, and concentrated under reduced re. The crude product was purified by column chromatography to afford the title compound 6.
Step 4: Synthesis of (9H-fluorenyl)methyl (Z)—(2-(4—(pyrrolidin y|idenemethyl)benzamido)pheny|)carbamate hydrochloride (7): To a stirred solution of compound 6 (2.1 g, 3.41 mmol, 1 eq) in 1,4 dioxane (5 mL) at 0 °C, 4M HCI in dioxane (15 mL) was added and the reaction mixture was d at room temperature for 3 h. The progress of the on was monitored by TLC. After completion, the reaction mixture was concentrated under reduced pressure. The residue was diluted with saturated NaHCOs solution and extracted with ethyl acetate. The ed organic extracts were washed with water, brine, dried over anhydrous NaZSO4, filtered, and concentrated under reduced pressure to afford the compound 7 as HCI salt.
Step-5: Synthesis of (9H-fluorenyl)methyl (2-(4-((1-(cyclopropylmethyl) pyrrolidinyl)methyl)benzamido)phenyl)carbamate (8): To a stirred solution of amine compound 7 (0.2 g, 0.362 mmol, 1 eq) and cyclopropanecarbaldehyde (0.03 g, 0.435 mmol, 1.2 eq) in DCM (10 mL), acetic acid (0.065 g, 1.086 mmol, 3 eq) was added and stirred at room temperautre for 30 min. To this solution, sodium triacetoxyborohydride (STAB) (0.115 g, 0.543 mmol, 1.5 eq) was added and ng was continued at room temperature for 12 h.
The reaction progress was red by TLC and LCMS. After completion, the reaction mixture was diluted with saturated NaHCOa solution and extracted with DCM. The combined organic extracts were washed with water, brine, dried over anhydrous Na2804, ed, and concentrated under d pressure. The crude product was ed by column chromatography to afford the title compound 8.
Step-6: Synthesis of N-(2—aminophenyl)—4-((1-(cyclopropylmethyl)pyrrolidin yl)methy|)benzamide (Compound-555): A solution of compound 8 (0.1 g, 0.175 mmol, 1 eq) in 20% piperidine in DMF (3 mL) was a stirred at room ature for 30 min. The progress of the reaction was monitored by TLC. After completion, the reaction mixture was quenched with ice cold water. The itated solid was collected by filtration, then the solid was washed with water, pentane, and concentrated under reduced pressure. The residue was ed by silica gel column chromatography followed by atory TLC to afford nd nd-555. 1H NMR (400 MHz, DMSO-dB) 6 9.60 (s, 1H), 7.91 (d, J: 7.6 Hz, 2H), 7.34 (d, J = 8.0 Hz, 2H), 7.12 (d, J: 7.6 Hz, 1H), 6.94 (t, J: 7.2 Hz, 1H), 6.75 (d, J: 7.6 Hz, 1H), 6.56 (t, J: 7.2 Hz, 1H), 4.86 (s, 2H), 3.53 — 3.47 (m, 1H), 3.17 — 3.15 (m, 2H), 2.97 - 2.95 (m 2H), 2.80 — 2.74 (m, 2H), 2.00 — 1.84 (m, 1H), 1.67 — 1.50 (m, 1H), 0.88 — 0.77 (m, 1H), 0.56 - 0.54 (m, 2H), 0.33 — 0.32 (m, 2H), 2H merged in solvent peak; LCMS 350.05 (M + 1)+.
Synthetic Scheme for Compound 556: COzMe Toluene reflux 002Me COZMe DMF, 65 °C, overnight Step 1 Step 2 1 2 3 Step 3 A/Br 4M HCIIn dioxane Boo/Nm —>k©/\©\/ Cone 1 4,-dioxane:MeOH (:5 1), mcone 002Me 032003 DMF ”4h 00CtoSOoC,4h 8 Step 4 Step 5 LiOH km1©wa NHBoc N cm A» 4M HCIIn dioxane COZH HATU DIPEA Step 6 1 4,-dioxane:MeOH (1:1), DMF, rt, 30 h rt 4 h Step 7 Step 8 nd-556 Step 1: Synthesis of methyl 4-((bromotripheny|—:5-phosphanyl) methyl)benzoate (2): To a stirred solution of compound 1 (75 g, 326 mmol, 1 eq) in toluene (1 L) triphenyl phosphine (85.5 g, 326 mmol, 1 eq) was added and the reaction mixture was heated at reflux for 7 h. After 7 h, the reaction mixture was allowed to cool to room temperature. The precipitate formed was filtered, washed with toluene followed by hexane, and dried under vacuum to afford compound 2.
Step 2 & 3: Synthesis of tert—butyl 4-(4-(methoxycarbonyl)benzylidene)piperidine - 1-carboxylate (5): To a stirred solution of compound 2 (50 g, 102 mmol, 1 eq) in anhydrous DMF (500 mL) at 0 °C, NaH (4.9 g, 60% W/Win mineral oil, 122 mmol, 1.2 eq) was added under N2 atmosphere. After stirring the reaction mixture for 30 min, a solution of compound 4 (24.3 g, 122 mmol, 1.2 eq) in DMF was added and the reaction mixture was then heated at 65 °C for 12 h. The ss of reaction was monitored by TLC. After completion, the reaction mixture was cooled to 0 °C, diluted with ice-cold water under stirring and the W0 20182’119362 18 itate formed was ed. The solid obtained was taken in ethyl acetate, stirred for 10 min and the resulting mixture was filtered. The filtrate was washed with brine and water. The organic layer was ted, dried over anhydrous , and evaporated under reduced pressure to obtain the product, which was purified by silica gel column chromatography to afford compound 5.
Step 4: Synthesis of methyl 4-(piperidinylidenemethyl)benzoate hydrochloride (6): To a d solution of compound 5 (30 g, 90.6 mmol, 1 eq) in 1,4 dioxane: MeOH (210 mL: 40 mL) at 0 °C, 4M HCI in dioxane (85 mL) was added and the reaction mixture was stirred at room temperature for 4 h. The progress of the reaction was monitored by TLC.
After completion, the reaction mixture was diluted with diethyl ether. The solid obtained was filtered and washed with diethyl ether. The residue was dried under reduced pressure to afford the title compound 6 as a hydrochloride salt.
Step 5: Synthesis of methyl 4-((1-(cyc|opropylmethyl)piperidinylidene)methyl) te (8): To a stirred solution of compound 6 (12 g, 45 mmol) in DMF (250 mL) at 0 °C, ropyl methylene bromide 7 (5 mL, 50 mmol) and 032003 (29.3 g, 90 mmol) were added. The reaction mixture was stirred at 60 0C for 4 h. The reaction progress was monitored by TLC and LCMS. After completion, the reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over ous Na2SO4, filtered, and trated under reduced pressure. The residue was purified by silica gel column chromatography to afford nd 8.
Step 6: Synthesis of 4-((1-(cyc|opropylmethyl)piperidinylidene)methyl)benzoic acid (9): To a stirred solution of compound 8 (18.5 g, 64.91 mmol, 1 eq) in methanol (300 mL), aqueous LiOH (4.1 g, 97.36 mmol in 60 mL water) was added and the reaction mixture was stirred at room ature for 12 h. The progress of reaction was monitored by TLC.
After completion, ol was removed under reduced pressure and reaction mixture was acidified with 2N HCI until pH = 3-4. The resulting solid obtained was filtered, washed with 2N HCI (2 L) & dried. The solid was further washed with diethyl ether (1 L), azeotroped with toluene (3 X 500 mL), and dried under vacuum to afford compound 9.
Step 7: Synthesis of tert-butyl (2-(4-((1-(cyclopropylmethyl)piperidinylidene) methyl)benzamido)phenyl)carbamate (11): Step 7a: Synthesis of tert-butyl (2-aminophenyl)Carbamate (10): To a stirred solution of benzene—1,2-diamine (54 g, 500 mmol, 1 eq) in THF (500 mL), (Boc)20 (109.09 g, 500 mmol) in 150 mL THF was added slowly at 0 °C. The reaction mixture was slowly warmed to room temperature and stirred for 12 h. The progress of the reaction was red by TLC. After completion, the reaction mixture was concentrated and resulting residue was purified by silica gel column chromatography to afford compound 10.
W0 20182’119362 18 ] Step 7b: Synthesis of tert-butyl (2-(4-((1-(cyclopropy|methyl)piperidin y|idene)methyl)benzamido)pheny|)carbamate (11): To a stirred solution compound 9 (8.8 g, 32.47 mmol, 1 eq) and compound 10 (8.1 g, 38.96 mmol, 1.2 eq) in DMF (100 mL), DIPEA (23 mL, 129.8 mmol, 4 eq) was added and stirred for 10 min. To this solution, HATU (18.5 g, 48.70 mmol, 1.5 eq) was added slowly and the reaction mixture was stirred at room temperature for 12 h. The progress of the on was monitored by TLC. After completion, the reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with water, brine, dried over anhydrous NaZSO4, filtered and concentrated in vacuo. The residue was purified by column chromatography to afford compound 11 .
Step 8: Synthesis of N—(2-aminophenyI)((1-(cyclopropylmethyl)piperidin y|idene)methyl)benzamide (12): To a stirred solution of compound 11 (7 g, 15.18 mmol, 1 eq) in 1,4 dioxanezMeOH (21 mL: 7 mL) at 0 °C, 4M HCI in dioxane (21 mL) was added and the reaction mixture was stirred at room ature for 4 h. The progress of the reaction was monitored by TLC. After completion, the reaction mixture was diluted with 1,4 dioxane, stirred for 15 min and ed. The e was taken in diethyl ether, stirred for 15 min and again filtered. The solid nd was dried under reduced pressure to afford compound 12 as HCI salt.
Step 9: Synthesis of N-(2-aminophenyl)((1-(cyc|opropy|methyl)piperidin yl)methyl)benzamide und-556): To a stirred solution of 12 (0.1 g, 0.216 mmol, 1 eq) in methanol (10 mL), 10% Pd/C (50 mg) was added and the reaction mixture was stirred under hydrogen atmosphere on pressure) at room ature for 2 h. The progress of the reaction was monitored by TLC. After completion, the reaction mixture was filtered through a pad of celite and the filtrate was concentrated under d pressure. The crude product was purified by silica gel column chromatography to afford Compound-556. 1H NMR (400 MHz, DMSO-d6) 6 9.58 (s, 1H), 7.90 (d, .1: 8.4 Hz, 2H), 7.29 (d, J = 8.4 Hz, 2H), 7.15 (d, J: 8.0 Hz, 1H), 6.96 (t, J: 8.0 Hz, 1H), 6.77 (d, .1: 8.4 Hz, 1H), 6.59 (t, J: 7.6 Hz, 1H), 4.85 (brs, 2 H), 3.01 — 2.93 (m, 2H), 2.59 — 2.57 (m, 2H), 2.25 - 2.23 (m, 2H), 2.01 — 1.95 (m, 2H), 1.58 — 1.54 (m, 3H), 1.26 — 1.23 (m, 2H), 0.82 — 0.81 (m, 1 H), 0.48 - 0.45 (m, 2H), 0.09 — 0.05 (m, 2H); LCMS: 364.15 (M +1)+.
HDAC enzyme inhibition The HDAC activity inhibition assay is performed as follows to ine the y of a test compound to inhibit HDAC enzymatic activity. Serial dilutions of HDAC inhibitors are prepared in HDAC assay buffer (25 mM Tris/HCI, pH 8.0, 137 mM NaCl, 2.7 mM KCI, 1 mM MgCI2, pH 8) in 96—well assay plates (Fisher scientific, #07-200—309) and pre-incubated for 2 hours at room temperature in the presence of 125ug/ml BSA and purified HDAC1 (BPS W0 20182’119362 18 Bioscience, San Diego, CA, #50051), HDAC2 (BPS Bioscience, #50053), or HDAC3/NcoR2 (BPS Bioscience, #50003) at concentrations of 1.25, 1.32, and 0.167 ug/mL, respectively.
Following pre-incubation, Fluor-de-LysTM substrate (Enzo Life es, Plymouth Meeting, PA, BML-KI104-0050) is added to a final concentration of 10 [M and plates are further incubated for 30 minutes at room ature. The enzymatic reaction is stopped by addition of Trichostatin A (Sigma-Aldrich, St Louis, MO, #T8552, final concentration: 100 nM) and trypsin (MP Biomedicals, Solon, OH, #02101179) are added to reach a final concentration of 100ug/mL. After a 15 minute incubation at room temperature, fluorescence is recorded using a Spectramax M2 fluorometer (Molecular Devices, ale, CA) with excitation at 365nm and emission at 460 nm. |C50 values are calculated by using a sigmoidal dose-response (variable slope) equation in GraphPad Prism® 5 for Windows (Graph Pad re, La Jolla, CA).
Acid stability determination A 100uM solution of test compound is prepared by on of a 10 mM DMSO stock solution in a 0.01 M on of HCI in deionized water. Immediately after mixing, an aliquot (100 uL) is sampled and analyzed by HPLC/UV. The area under the compound peak is determined and used as the time zero reference point. The der of the acid sample is incubated at 50 °C and s were taken after 2, 4, and 24 or 30 hours of incubation.
These are analyzed by the same HPLC/UV method and the area of the peak corresponding to the test compound is measured. Percent remaining at a given time point is then calculated as the ratio of the area under the peak after incubation to that at time zero times 100. In those embodiments where a 30 hour time point is recorded, the percent remaining at 24 hours is obtained by olation of the t remaining versus time curve assuming a ecular process, Le. a monoexponential decay.
Brain penetration studies Test compounds are prepared at either 0.5 mg/ml or 5 mg/ml in 30% hydroxypropyl-B-cyclodextrin, 100 mM sodium acetate pH 5.5, 5% DMSO. Rats or C57/BL6/J mice are dosed so. at 5 mg/kg or 50 mg/kg, or iv at 5 mg/kg. Animals are euthanized at pre-dose, 5, 15, 30 min, 1, 2 and 4 hours post-dose and plasma and brain obtained. Three animals per dose per time points are used. The levels of compound in the plasma and brain are determined by standard LC/MS/MS methods. Brain/plasma ratio (BPR) is calculated as the ratio of the rain)/Cmax(plasma). ln-cell deacetylase inhibition assay (DAC assay) GM lymphoblastoid cells line) cells are seeded in 96-well plates at an appropriate density (100,000 cells/well) in 90 [LL RPM|1640 medium containing 10% v/v fetal bovine serum (FBS), 1% v/v penicillin/streptomycin, and 1% v/v L—glutamine. Compound dilutions are made in 100% DMSO followed by parallel dilution in media with 2% DMSO. 10 ul of the compound dilutions are added to the cells to achieve the desired concentrations.
The final concentration of DMSO in each well is 0.2%. The cells are ted for 4h at 37 °C with 5% 002. After incubation, the cells are fuged down and the supernatant removed. The cell pellets are washed with 100 uL phosphate-buffered saline (PBS) and then lysed with 45 uL lysis buffer (HDAC assay buffer at pH 8.0 (25 mM Tris/HCI, 137 mM NaCl, 2.7 mM KCI, 1 mM MgCIZ) + 1% v/v lgepal CA-630). To te the reaction, the HDAC substrate Kl-104 (Enzo Life Sciences, Farmingdale, NY) is added to a final concentration of 50 uM. The reaction is stopped after 30 min incubation by addition of 50 uL developer (6 mg/mL trypsin in HDAC assay buffer). The reaction is allowed to develop for 30 min at room temperature and the fluorescence signal is detected using a fluorometer (Spectramax M2, Molecular Devices, Sunnyvale, CA) with excitation and emission wavelengths of 360 nm and 470 nm respectively. The data are fitted to a sigmoidal dose response equation with variable slope in GraphPad Prism 5.0 (GraphPad Software, La Jolla, CA) to determine IC50. Bottom and top of the curve are fixed to the average fluorescence response of control wells with no cells and cells but no compound respectively.
Cell proliferation assay HCT116 cells (5000 cells/well) in 80 uL McCoy’s 5A medium containing 10% v/v FBS, 1% v/v penicillin/streptomycin and 1% v/v L-glutamine are incubated in 96-well plates with nds at various concentrations for 72h at 37 °C in a 5% 002 atmosphere. The compound dilutions are made in 100% DMSO followed by parallel dilutions in media. The final concentration of DMSO in each well is 0.05%. After 72h, 20uL of Cell titer 96 aqueous one solution (Promega ation, Madison, WI) are added to the cells and the plate is incubated at 37 °C for another 4h. The absorbance at 490nm is then ed on a 96-well plate reader (Spectramax M2, Molecular Devices, Sunnyvale, CA). Data analysis is performed in Microsoft Excel (Microsoft Corp, Redmond, WA).( (O.D. sample — average O.D. ve l)/(average O.D. negative control - average O.D. positive control))*100, where O.D. is the measured absorbance, O.D. positive control is the ance from cells ted with trichostatin A at 5 uM and OD. negative control is the absorbance measured from cells incubated t any compound, is plotted against compound concentration and an IC5O is determined by graphical olation of the concentration required for 50% inhibition of cell growth.
Effect of HDAC inhibitors on frataxin (FXN) mRNA expression Method: mRNA fication of compound-treated iPSC derived neuronal cells Neuronal stem cells were cultured in Neurobasal A medium (Life technologies 022) supplemented with N2, B27 (Life technologies #17502-048 and #17504-044), amine (Life technologies #25030081), supplemented with 20ng/ml EGF (R&D Systems # 236-EG) and 20ng/ml bFGF (BioPioneer # HRP-0011). Neuronal differentiation was initiated by ng growth factors and ing cells in Neurobasal A with N2 and B27. Cells were allowed to differentiate for 16 days. HDAC inhibitory compound was then added and incubate for 24h. RNA isolation was performed using the RNeasy Plus mini kit (QlAgen #74134) using a QIAcube instrument per manufacturer’s instructions. qRT-PCR was performed using chript One-Step SYBR Green qRT-PCR Kit (Quanta Biosciences 170— 8893BR) with the ing ions: 20 minutes at 50 °C, 5 minutes at 95 °C, and then 40 cycles of 20 seconds at 95 °C, 20 seconds at 55 °C, 30 s at 72 °C. The primer sequences to detect expression of FXN were: 5’-CAGAGGAAACGCTGGACTCT-3’ and ’-AGCCAGATTTGCTTGTTTGG-3’.
Data for compounds for iPSC fold inductions and cLogP are shown in Table 3.
Data for additional compounds for iPSC fold inductions and cLogP are shown in Table 4 The ranges ed for cLogP refer to the following. A < 1, 1 < B < 2, 2 < C < 3, 3 < D < 5, E >5.
NA refers to “not available.” Table 3 Fold 477(racenflc) cLogP lnducfion 478(racenuc) 480 2.0 146(Tn-ch 479(Free 147(Tn-ch base) 171(DFHCD 172(DFHCD 1.7 base) 174(DFHCD 481(Free 354(Free 1.6 base) base) 489(Free 175(Free 1.5 J> base) base) 490(Hee 241(TrFTFA) 1.4 base) 491(Free 176(DFTFA) 1.7 base) 4784son1erl 163 1.6 (Free base) 478-isomer ll 162 1.5 (Free base) 484(Hee 357 UDOUJ 1.6 base) 359(Free base) UU 492(Free base) 379(DFHCD 483(Hee 181(Free l“ U1 (3 base) base) 4774somerH 472(Free 4774son1erl base) 487(Free 485(Tfi-HCD base) W0 20182’119362 18 488(Free base) Table 4 Fold Con1pound cLogP 555(Free 1.11 C base) 556 (Free base) Protocol for com ound it in he atoc es To assess the stability and metabolism of RGFP compounds in hepatocytes.
This assay was designed to evaluate the metabolism of RGFP compounds, following their incubation with human, monkey, dog and rat hepatocytes by monitoring either parent drug disappearance or metabolite appearance using HPLC.
Equipment: Applied Biosystem Triple Quadrupole LC/MS/MS; Ice bucker, timer; 96 well plates; Falcon, Cat# 353072; 96 well plates shaker; s pipettes: 10uL, 20 uL, 200 (1L, and 1000 (1L; Test tubes: Catalog # VWR 47729-572, 13x 100 mm Procedure: Turn on the water-bath heater to 37°C. Take out the KHB buffer and make sure it is at room temp before use. Prepare 2.5 mM concentration of RGFP compound in DMSO stock. Add 10 uL of above DMSO stock to 2490 uL KHB ; final concentration of RGFP compound will be 10 (1M. rm 45ml anitro HT Medium to 37 °C in a sterile 50 mL conical tube. Add 1.0 mL Torpedo Antibiotic Mix per 45 mL anitro HT medium.
Transfer 13 mL of warm HT medium with Antibiotic Mix into a 15 mL l tube. Carefully remove the hepatocyte vials from liquid nitrogen (liquid phase). Immediately e the vial into a 37 °C water bath. Shake gently until the ice melts entirely. Do not keep the cells in 37 °C water bath longer than necessary. Immediately empty contents of the vial into 13 mL of pre-warmed anitro HT Medium with antibiotics. Rinse the vial with the HT media that you have just transferred the hepatocytes to, in order to ensure complete transfer. Centrifuge the cell suspension at 600 RPM for 5 minutes at room temperature. Discard the supernatant by either pouring in one motion (do not pour partially and re-invert centrifuge tube) or aspirating using a vacuum pump. Add 1.0 mL of KHB (at room temperature) buffer to the tube of hepatocyte pellet. Loosen the cell pellet by gently swirling the centrifuge tube.
Transfer 100 uL of above solution to a different tube and add 900 uL of KHB buffer to count the cells. ine the total cell count and the number of viable cells using the Trypan Blue exclusion method. Once the cell count is obtained, ly the number by 10 (attributing to the dilution factor). Now add required volume of KHB buffer to the tube containing 2017/0681 18 hepatocytes such that the final count will be 2 million cells/mL. Dispense 50 uL of 2 million ml to a 96 well plate and then add 50 uL of DMSO stock to respective wells (such that, the concentration of RGFP compounds is 5 uM and number of cells are 100000 in each well). Place the plates on a shaker in a 37 °C incubator with 5% 002. Separate plates for each time point are advisable (Time points: 0h, 1h, 2h, and 6 h). After each time point, add 100 uL of quenching solution.
Quenching solution is an acetonitrile solution containing RGFP531 (10 uM) internal standard, 0.1% formic acid and phenylglyoxol (400 uM). The formic acid and phenylglyoxal is used for the identification and quantification of CPD as mentioned above.
Pipette up and down a few times to ensure a complete stop of reaction. Transfer all the solution into a 1.5 mL tube, vortex thoroughly, and centrifuge at 14000 RPM at 4 °C for 5 minutes to precipitate cell . er the 150 uL of supernatant to vials for analysis using HPLC.
Effect of nds on long term memory for object recognition Rats or C57BL/6J male mice are handled 1-2 min for 5 days and habituated to the experimental apparatus 5 min a day for 4 consecutive days in the absence of objects.
During the training trial, rats or mice are placed in the experimental apparatus with two identical objects and allowed to explore these s for 3 min, which does not result in short- or long-term memory nko, et al., 2009). Immediately following training, rats or mice receive subcutaneous injections of either vehicle (20% glycerol, 20% PEG 400, 20% propylene glycol, and 100 mM sodium acetate, pH 5.4), reference compound 1, RGFP109, class I HDAC inhibitor, (3, 10, 30 mg/kg), reference compound 2, RGFP136 (3, 10, 30 mg/kg), or a test compound disclosed herein (3, 10, 30 mg/kg). 24-h later rats or mice are tested for memory retention (5 min) using the object recognition memory task (OHM), in which a familiar object is replaced with a novel one. All training and testing trials are videotaped and analyzed by individuals blind to the treatment condition and the genotype of subjects. A rat or mouse is scored as exploring an object when its head was oriented toward the object within a ce of 1 cm or when the nose is touching the object. The relative exploration time is recorded and expressed by a discrimination index [DI = (tnovel — tfamiliar)/(tnovel + tfamiliar) x 100]- A number of embodiments have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the disclosure. Accordingly, other embodiments are within the scope of the ing claims.

Claims (1)

What is Claimed is:
1. A compound having a structure of formula (I), or a pharmaceutically acceptable salt thereof: wherein ring A is a 4—7 membered monocyclic heterocycloalkyl ring or a 7-12 membered spiro heterocycloalkyl ring, wherein ring A contains one nitrogen ring atom and optionally contains one additional ring atom independently selected from O, N, and S; R1 is H, C1_6alkyl, 02.6alkenyl, C1_6hydroxyalkyl, C(O)C1.6alkyl, Co_3alkylene-Cg
NZ755543A 2016-12-22 2017-12-22 Histone deacetylase inhibitors NZ755543A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US62/437,767 2016-12-22

Publications (1)

Publication Number Publication Date
NZ755543A true NZ755543A (en)

Family

ID=

Similar Documents

Publication Publication Date Title
US10968180B2 (en) Piperidine derivatives as HDAC1/2 inhibitors
DK2680694T3 (en) HISTONDEACETYLASE INHIBITORS
EP2580212B1 (en) Urea derivatives and their therapeutic use in the treatment of, inter alia, diseases of the respiratory tract
CA3048005A1 (en) Histone deacetylase inhibitors
AU2014239677B2 (en) Histone deacetylase inhibitors
US11981680B2 (en) Substituted thienopyrroles as PAD4 inhibitors
KR20070091677A (en) Heterocyclic compounds as ccr2b antagonists
US10308608B2 (en) Histone deacetylase inhibitors
JP2008504301A (en) Condensed pyrimidine derivatives as CXCR3 receptor modulators and compositions thereof useful for the prevention and treatment of inflammatory and immunoregulatory disorders and diseases
NZ717552A (en) Piperidine urea derivatives
US20100311776A1 (en) Novel sEH Inhibitors and their Use
KR20190027877A (en) Inhibitors of Tryptophan 2,3-dioxygenase (Tryptophan 2,3-dioxygenase)
NZ755543A (en) Histone deacetylase inhibitors
MX2011006087A (en) (4-tert-butylpiperazin-2-yl)(piperazin-1-yl)methanone-n-carboxam ide derivatives.
US20100311775A1 (en) Novel sEH Inhibitors and Their Use