WO2020132582A1 - Sting agonists and uses thereof - Google Patents

Sting agonists and uses thereof Download PDF

Info

Publication number
WO2020132582A1
WO2020132582A1 PCT/US2019/068076 US2019068076W WO2020132582A1 WO 2020132582 A1 WO2020132582 A1 WO 2020132582A1 US 2019068076 W US2019068076 W US 2019068076W WO 2020132582 A1 WO2020132582 A1 WO 2020132582A1
Authority
WO
WIPO (PCT)
Prior art keywords
nitrogen
sulfur
oxygen
partially unsaturated
ring
Prior art date
Application number
PCT/US2019/068076
Other languages
French (fr)
Inventor
Silvana Marcel LEIT DE MORADEI
Donna L. Romero
Jennifer Knight
Anthony Clark
Pieter Bos
Original Assignee
Nimbus Titan, 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 date
Application filed by Nimbus Titan, Inc. filed Critical Nimbus Titan, Inc.
Publication of WO2020132582A1 publication Critical patent/WO2020132582A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/16Peri-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the present invention relates to compounds and methods useful for the modulation of STING-dependent type I interferon production.
  • the invention also provides pharmaceutically acceptable compositions comprising compounds of the present invention and methods of using said compositions in the treatment of various disorders.
  • Innate immunity is a rapid nonspecific immune response that fights against environmental insults including, but not limited to, pathogens such as bacteria or viruses.
  • Adaptive immunity is a slower but more specific immune response, which confers long-lasting or protective immunity to the host and involves differentiation and activation of naive T lymphocytes into CD4+ T helper cells and/or CD8+ cytotoxic T cells, to promote cellular and humoral immunity.
  • Antigen presentation cells of the innate immune system such as dendritic cells or macrophages, serve as a critical link between the innate and adaptive immune systems by phagocytosing and processing the foreign antigens and presenting them on the cell surface to the T cells, thereby activating T cell response.
  • STING (stimulator of interferon genes) is an endoplasmic reticulum adaptor that facilitates innate immune signaling (Ishikawa and Barber, Nature 2008, 455(7213):674-678).
  • STING comprises four putative transmembrane regions (Ouyang et al., Immunity (2012) 36, 1073), predominantly resides in the endoplasmic reticulum and is able to activate NF-kB, STAT6, and IRF3 transcription pathways to induce expression of type I interferon (e.g., IFN-a and IFN-13) and exert a potent anti-viral state following expression (Ishikawa and Barber, Nature 2008, 455(7213):674-678; Chen et al., Cell (2011) 147, 436- 446).
  • loss of STING rendered murine embryonic fibroblasts extremely susceptible to negative stranded virus infection, including vesicular stomatitis virus. (Ishikawa and Barber, Nature 2008, 455(7213):674-678).
  • Compounds of the present invention are useful for treating a variety of diseases, disorders or conditions, associated with STING. Such diseases, disorders, or conditions include those described herein.
  • Compounds provided by this invention are also useful for the study of STING and associated proteins in biological and pathological phenomena; the study of STING occurring in bodily tissues; and the comparative evaluation of new STING agonists or other regulators of STING in vitro or in vivo.
  • the terms“agonist,”“binder,”“modulator,” and“ligand” are used interchangeably and describe a compound that binds to, modulates, activates, is a ligand for, or agonist of STING.
  • the present invention provides a compound of formula I:
  • n 0 or 1
  • n 0 or 1
  • Ring A is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
  • Ring A and its substituents is other than
  • Ring B is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
  • L 1 is a covalent bond or a CM O bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 1 -, -C(R’) 2- , -CH(R’)-, -N(R)-, Cy'fL’-Z) , -C(R’)(L’-Z)- -CH(L’-Z)-,
  • each -Cy 1 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R’ is independently hydrogen or C M aliphatic;
  • R’ two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R is independently hydrogen, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or:
  • two R groups on the same nitrogen are optionally taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
  • each L’ is a covalent bond or a CMS bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and
  • each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, napththylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each Z is hydrogen or a CRBN binding moiety
  • L 2 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 2 -, -C(R’) 2 -, -CH(R’)-, -N(R)-, -Cy 2 (L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)- -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2- -S(0) 2 N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0) 2- ;
  • each -Cy 2 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • L 3 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 3 -, -C(R’) 2- , -CH(R’)-, -N(R)-, -Cy 3 (L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)- -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2- , -S(0) 2 N(R)-, -0-, -C(0)-, -0C(0)- -C(0)0-, -S-, -S(0)-, or -S(0) 2- ;
  • each -Cy 3 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R 1 is independently hydrogen, halogen, -CN, -N0 2 , -OR,
  • R 2 is hydrogen or an optionally substituted Ci- 6 aliphatic group
  • each R 3 is independently hydrogen, oxo, halogen, -CN, -NO2, -OR,
  • each R 4 is independently hydrogen, halogen, -CN, -N0 2 , -OR,
  • R 5 is hydrogen or an optionally substituted Ci- 6 aliphatic group
  • each R 6 is independently hydrogen, oxo, halogen, -CN, -N0 2 , -OR,
  • each of q, s, t, and v is independently 0, 1, 2, 3, or 4;
  • each of r and u is independently 0 or 1;
  • each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • the present invention also provides a compound of formula
  • n 0 or 1
  • n 0 or 1
  • p is 0 or 1;
  • L 1 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced
  • each -Cy 1 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R’ is independently hydrogen or C M aliphatic;
  • R’ two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R is independently hydrogen, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or:
  • each L’ is a covalent bond or a CM5 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and
  • each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, napththylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each Z is hydrogen or a CRBN binding moiety
  • L 2 is a covalent bond or a C MO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 2 -, -C(R’) 2 - -CH(R’)-, -N(R)-, -Cy 2 (L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)- -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2- , -S(0) 2 N(R)-, -0-, -C(O)-, -0C(0)- -C(0)0-, -S-, -S(O)-, or -S(0) 2- ;
  • each -Cy 2 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • L 3 is a covalent bond or a C MO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 3 -, -C(R’) 2- , -CH(R’)-, -N(R)-, -Cy 3 (L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)- -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2- -S(0) 2 N(R)-, -0-, -C(0)-, -0C(0)- -C(0)0-, -S-, -S(0)-, or -S(0) 2 -;
  • each -Cy 3 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R 1 is independently hydrogen, halogen, -CN, -N0 2 , -OR,
  • R 2 is hydrogen or an optionally substituted Ci- 6 aliphatic group
  • each R 3 is independently hydrogen, oxo, halogen, -CN, -N0 2 , -OR,
  • each R 4 is independently hydrogen, halogen, -CN, -N0 2 , -OR,
  • R 5 is hydrogen or an optionally substituted Ci- 6 aliphatic group
  • each R 6 is independently hydrogen, oxo, halogen, -CN, -N0 2 , -OR,
  • each of q, s, t, and v is independently 0, 1, 2, 3, or 4;
  • each of r and u is independently 0 or 1;
  • each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10;
  • aliphatic or“aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as "carbocycle,”“cycloaliphatic” or“cycloalkyl”), that has a single point of attachment to the rest of the molecule.
  • aliphatic groups contain 1-6 aliphatic carbon atoms.
  • aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms.
  • “cycloaliphatic” (or“carbocycle” or“cycloalkyl”) refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule.
  • Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
  • bridged bicyclic refers to any bicyclic ring system, i.e. carbocyclic or heterocyclic, saturated or partially unsaturated, having at least one bridge.
  • a“bridge” is an unbranched chain of atoms or an atom or a valence bond connecting two bridgeheads, where a“bridgehead” is any skeletal atom of the ring system which is bonded to three or more skeletal atoms (excluding hydrogen).
  • a bridged bicyclic group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • bridged bicyclic groups are well known in the art and include those groups set forth below where each group is attached to the rest of the molecule at any substitutable carbon or nitrogen atom. Unless otherwise specified, a bridged bicyclic group is optionally substituted with one or more substituents as set forth for aliphatic groups. Additionally or alternatively, any substitutable nitrogen of a bridged bicyclic group is optionally substituted. Exemplary bridged bicyclics include:
  • lower alkyl refers to a C straight or branched alkyl group.
  • exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl.
  • lower haloalkyl refers to a CM straight or branched alkyl group that is substituted with one or more halogen atoms.
  • heteroatom means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3 ,4-di hydro-2//-pyrrol yl ), NH (as in pyrrolidinyl) or NRT (as in N-substituted pyrrolidinyl)).
  • CM bivalent CM (or CM) saturated or unsaturated, straight or branched, hydrocarbon chain
  • CM bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein.
  • alkylene refers to a bivalent alkyl group.
  • An“alkylene chain” is a polymethylene group, i.e., -(03 ⁇ 4 h- , wherein n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3.
  • a substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
  • alkenylene refers to a bivalent alkenyl group.
  • a substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
  • cyclopropylenyl refers to a bivalent cyclopropyl group of the following structure:
  • halogen means F, Cl, Br, or I.
  • aryl used alone or as part of a larger moiety as in“aralkyl,”“aralkoxy,” or “aryloxyalkyl,” refers to monocyclic or bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members.
  • the term“aryl” may be used interchangeably with the term“aryl ring.”
  • “aryl” refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may bear one or more substituents.
  • aryl is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like.
  • heteroaryl and“heteroar-,” used alone or as part of a larger moiety e.g., “heteroaralkyl,” or“heteroaralkoxy,” refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 p electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms.
  • heteroatom refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen.
  • Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl.
  • heteroaryl and“heteroar-”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring.
  • Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3-b]-l,4-oxazin-3(4H)-one.
  • a heteroaryl group may be mono- or bicyclic.
  • the term“heteroaryl” may be used interchangeably with the terms“heteroaryl ring,”“heteroaryl group,” or“heteroaromatic any of which terms include rings that are optionally substituted.
  • the term“heteroaralkyl” refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted.
  • heterocycle As used herein, the terms“heterocycle,”“heterocyclyl,”“heterocyclic radical,” and “heterocyclic ring” are used interchangeably and refer to a stable 5- to 7-membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above.
  • nitrogen includes a substituted nitrogen.
  • the nitrogen may be N (as in 3,4-dihydro- 2 / pyrrolyl), H (as in pyrrolidinyl), or + NR (as in A substituted pyrrolidinyl).
  • a heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted.
  • saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl.
  • heterocycle “heterocyclyl,” “heterocyclyl ring,” “heterocyclic group,”“heterocyclic moiety,” and“heterocyclic radical,” are used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3// indolyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl.
  • a heterocyclyl group may be mono- or bicyclic.
  • heterocyclylalkyl refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.
  • the term“partially unsaturated” refers to a ring moiety that includes at least one double or triple bond.
  • the term“partially unsaturated” is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
  • compounds of the invention may contain“optionally substituted” moieties.
  • the term“substituted,” whether preceded by the term“optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent.
  • an“optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position.
  • Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds.
  • the term“stable,” as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
  • Suitable monovalent substituents on R° are independently halogen, -(CH2)o-2R*, -(haloR*), -(CH 2 )O- 2 OH, -(CH 2 )O- 2 OR ⁇ , -(CH 2 )O-2CH(OR # ) 2 ; -0(haloR # ), -CN, -N 3 , -(CH 2 )o- 2 C(0)R ⁇ , -(CH 2 ) O-2 C(0)OH, -(CH 2 ) O-2 C(0)OR ⁇ , -(CH 2 ) O-2 SR*, -(CH 2 ) O-2 SH, -(CH 2 ) O -2NH 2 , - (CH 2 )O-2NHR ⁇ , -(CH 2 )O-2 R*2, -N0 2 , -SiR*3, -OSiR ⁇ -C
  • Suitable divalent substituents that are bound to vicinal substitutable carbons of an“optionally substituted” group include: -0(CR * 2 ) 2- 3O-, wherein each independent occurrence of R * is selected from hydrogen, CM aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on the aliphatic group of R * include halogen, -R*, -(haloR*), -OH,
  • each R* is unsubstituted or where preceded by“halo” is substituted only with one or more halogens, and is independently CM aliphatic, -CH2PI1, -0(03 ⁇ 4)o-iR1i, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on a substitutable nitrogen of an“optionally substituted” group include -R ⁇ , -NR ⁇ 2 , -C(0)R ⁇ , -C(0)OR ⁇ , -C(0)C(0)R ⁇ ,
  • each R' is independently hydrogen, Ci- 6 aliphatic which may be substituted as defined below, unsubstituted -OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R ⁇ , taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on the aliphatic group of R' are independently halogen, - R ⁇ , -(haloR*), -OH, -OR*, -O(haloR'), -CN, -C(0)OH, -C(0)OR*.
  • each R* is unsubstituted or where preceded by“halo” is substituted only with one or more halogens, and is independently C1-4 aliphatic, -CH 2 Ph, -0(CH 2 )o-iPh, or a 5-6- membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • the term "pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et ah, describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases.
  • Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphor sulfonate, citrate, cyclopentanepropionate, digluconate, dodecyl sulfate, ethanesulfonate, formate, fiimarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, peroxine sodium
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N C 1-4 alky lh salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. For example, it will be appreciated by those skilled in the art that the compounds of this invention may exist in tautomeric
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13 C- or 14 C-enriched carbon are within the scope of this invention.
  • Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present invention
  • an agonist or activator has an EC50 and/or effective concentration of less than about 50 mM, less than about 1 mM, less than about 500 nM, less than about 100 nM, less than about 10 nM, or less than about 1 nM.
  • a compound of the present invention may be tethered to a detectable moiety. It will be appreciated that such compounds are useful as imaging agents. One of ordinary skill in the art will recognize that a detectable moiety may be attached to a provided compound via a suitable substituent.
  • suitable substituent refers to a moiety that is capable of covalent attachment to a detectable moiety.
  • moieties are well known to one of ordinary skill in the art and include groups containing, e.g., a carboxylate moiety, an amino moiety, a thiol moiety, or a hydroxyl moiety, to name but a few. It will be appreciated that such moieties may be directly attached to a provided compound or via a tethering group, such as a bivalent saturated or unsaturated hydrocarbon chain. In some embodiments, such moieties may be attached via click chemistry.
  • such moieties may be attached via a 1,3 -cycloaddition of an azide with an alkyne, optionally in the presence of a copper catalyst.
  • Methods of using click chemistry are known in the art and include those described by Rostovtsev et al, Angew. Chem. Int. Ed. 2002, 44, 2596-99 and Sun etal. , Bioconjugate Chem., 2006, F7, 52-57.
  • the term“detectable moiety” is used interchangeably with the term "label” and relates to any moiety capable of being detected, e.g., primary labels and secondary labels.
  • Primary labels such as radioisotopes (e.g., tritium, 32 P, 33 P, 35 S, or 14 C), mass-tags, and fluorescent labels are signal generating reporter groups which can be detected without further modifications.
  • Detectable moieties also include luminescent and phosphorescent groups.
  • secondary label refers to moieties such as biotin and various protein antigens that require the presence of a second intermediate for production of a detectable signal.
  • the secondary intermediate may include streptavidin-enzyme conjugates.
  • antigen labels secondary intermediates may include antibody-enzyme conjugates.
  • fluorescent label refers to moieties that absorb light energy at a defined excitation wavelength and emit light energy at a different wavelength.
  • fluorescent labels include, but are not limited to: Alexa Fluor dyes (Alexa Fluor 350, Alexa Fluor 488, Alexa Fluor 532, Alexa Fluor 546, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 633, Alexa Fluor 660 and Alexa Fluor 680), AMCA, AMCA-S, BODIPY dyes (BODIPY FL, BODIPY R6G, BODIPY TMR, BODIPY TR, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 576/589, BODIPY 581/591, BODIPY 630/650, BODIPY 650/665), Carboxyrhodamine 6G, carboxy
  • mass-tag refers to any moiety that is capable of being uniquely detected by virtue of its mass using mass spectrometry (MS) detection techniques.
  • mass-tags include electrophore release tags such as N-[3-[4’-[(p- Methoxytetrafluorobenzyl)oxy]phenyl]-3-methylglyceronyl]isonipecotic Acid, 4’-[2, 3,5,6- Tetrafluoro-4-(pentafluorophenoxyl)]methyl acetophenone, and their derivatives.
  • mass-tags include, but are not limited to, nucleotides, dideoxynucleotides, oligonucleotides of varying length and base composition, oligopeptides, oligosaccharides, and other synthetic polymers of varying length and monomer composition.
  • nucleotides dideoxynucleotides
  • oligonucleotides of varying length and base composition oligopeptides, oligosaccharides
  • other synthetic polymers of varying length and monomer composition.
  • a large variety of organic molecules, both neutral and charged (biomolecules or synthetic compounds) of an appropriate mass range (100-2000 Daltons) may also be used as mass-tags.
  • measurable affinity and“measurably inhibit,” as used herein, means a measurable change in an STING activity between a sample comprising a compound of the present invention, or composition thereof, and STING, and an equivalent sample comprising STING, in the absence of said compound, or composition thereof.
  • the present invention provides a compound of formula I:
  • n 0 or 1
  • n 0 or 1
  • p is 0 or 1;
  • Ring A is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
  • Ring A and its substituents is other than
  • Ring B is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
  • L 1 is a covalent bond or a CM O bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 1 -, -C(R’) 2- - CH(R’)-, -N(R)-, Cy'fL’-Z) , -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2- -S(0) 2 N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0) 2- ;
  • each -Cy 1 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R’ is independently hydrogen or C M aliphatic;
  • R’ two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R is independently hydrogen, or an optionally substituted group selected from Ci- 6 aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or:
  • each L’ is a covalent bond or a CM S bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and
  • each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, napththylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each Z is hydrogen or a CRBN binding moiety
  • L 2 is a covalent bond or a CM O bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 2 -, -C(R’) 2- - CH(R’)-, -N(R)-, -Cy 2 (L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2 -, -S(0) 2 N(R)-, -0-, -C(0)-, -0C(0)-, -C(0)0- -S-, -S(0)-, or -S(0) 2- ;
  • each -Cy 2 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • L 3 is a covalent bond or a C O bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 3 -, -C(R’) 2- - CH(R’)-, -N(R)-, -Cy 3 (L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2 -, -S(0) 2 N(R)-, -0-, -C(0)-, -0C(0)-, -C(0)0-, -S-, -S(0)-, or -S(0) 2- ;
  • each -Cy 3 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R 1 is independently hydrogen, halogen, -CN, -N0 2 , -OR,
  • R 2 is hydrogen or an optionally substituted Ci- 6 aliphatic group
  • each R 3 is independently hydrogen, oxo, halogen, -CN, -N0 2 , -OR,
  • each R 4 is independently hydrogen, halogen, -CN, -N0 2 , -OR,
  • R 5 is hydrogen or an optionally substituted Ci- 6 aliphatic group
  • each R 6 is independently hydrogen, oxo, halogen, -CN, -N0 2 , -OR,
  • each of q, s, t, and v is independently 0, 1, 2, 3, or 4;
  • each of r and u is independently 0 or 1;
  • each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • n is 0 or 1. In some embodiments, n is 0 In some embodiments, m is 1.
  • p is 0 or 1. In some embodiments, p is 0 In some embodiments, m is 1.
  • m, n, and p are independently selected from those depicted in Table 1, below.
  • Ring A is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and
  • Ring A and its substituents is other than
  • Ring A is phenyl. In some embodiments, Ring A is a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring. In some embodiments, Ring A is a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur. In some embodiments, Ring A is a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, Ring A is an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, Ring A and its substituents is
  • Ring A is selected from those depicted in Table 1 and 2, below.
  • Ring In some embodiments, Ring A is
  • Ring A is . In some embodiments, Ring A is
  • Ring A is . In some embodiments, Ring A i . In some embodiments, Ring A is . In some embodiments,
  • Ring A is . In some embodiments, Ring A is * . In some embodiments, Ring
  • Ring A is / . In some embodiments, Ring A is . In some embodiments, Ring A is , some embodiments, Ring A is
  • Ring In some embodiments, Ring A is n
  • Ring A is In some embodiments, Ring
  • Ring A is . In some embodiments, Ring . In
  • Ring A is K 1 In some embodiments, Ring A is / . In some embodiments,
  • Ring A is ⁇ . In some embodiments, Ring A is ⁇ . In some
  • Ring some embodiments, Ring some
  • Ring A is ' . In some embodiments, Ring In some
  • Ring A is r . In some embodiments, Ring In
  • Ring B is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • Ring B is phenyl. In some embodiments, Ring B is a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring. In some embodiments, Ring B is a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur. In some embodiments, Ring B is a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, Ring B is an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • Ring B is selected from those depicted in Table 1 and 2, below.
  • Ring In some embodiments, Ring B is
  • Ring B is H P-
  • Ring In some embodiments, Ring . In some embodiments, Ring B is
  • Ring In some embodiments, Ring B is
  • Ring B In some embodiments, Ring B is
  • Ring B is . In some embodiments, Ring B
  • Ring some embodiments, Ring B is
  • Ring B is . In some embodiments, Ring B is
  • Ring some embodiments, Ring B is
  • Ring B is . In some embodiments, Ring B is
  • Ring B is ⁇ 0 3 ⁇ 4 SO me embodiments, Ring B is In some embodiments, Ring some embodiments, Ring B is . , g . In some embodiments, Ring B is In some embodiments, Ring B is . In some embodiments, Ring B is .
  • Ring B is In some embodiments, Ring B is
  • Ring Bis ' In some embodiments, Ring B
  • Ring In some embodiments, Ring B is . n some embodiments, Ring
  • L 1 is a covalent bond or a Ci-io bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 1 -, -C(R’) 2 -, -CH(R’)-, -N(R)-, -Cy'iL’- Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2- , - S(0) 2 N(R)-, -0- -C(0)- -OC(O)-, -C(0)0- -S-, -S(0)- or -S(0) -.
  • L 1 is a covalent bond.
  • L 1 is a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 1 -, -C(R’) 2 -, - CH(R’)-, -N(R)-, Cy'(L’-Z) , -CiR’XL’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -
  • L 1 is selected from those depicted in Table 1 and 2, below.
  • each -Cy 1 - is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 1 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 1 - is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • -Cy 1 - is selected from those depicted in Table 1 and 2, below.
  • each R’ is independently hydrogen or CM aliphatic; or two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring; or two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each R’ is independently hydrogen. In some embodiments, each R’ is independently C aliphatic.
  • two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring.
  • two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • R’ is selected from those depicted in Table 1 and 2, below.
  • each R is independently hydrogen, or an optionally substituted group selected from C aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or two R groups on the same nitrogen are taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • each R is independently hydrogen.
  • R is an optionally substituted group selected from CM aliphatic.
  • R is phenyl.
  • R is naphthalenyl.
  • R is an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • R is a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • R is 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • R is a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • two R groups on the same nitrogen are taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • R is selected from those depicted in Table 1 and 2, below.
  • each L’ is a covalent bond or a Ci-is bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the
  • each L’ is a covalent bond.
  • each L’ is a Ci-15 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and independently replaced by -Cy’-,
  • L’ is selected from those depicted in Table 1 and 2, below.
  • L’ is In some embodiments, L’ is
  • L’ is 0 In some embodiments,
  • L’ is 0 In some embodiments, L’ is
  • L’ is 0 In some embodiments, V is , In some embodiments, L’ is ,
  • L’ is ,
  • L’ In some embodiments, L’ is , In some embodiments, L’ is ,
  • each -Cy’- is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy’- is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • -Cy’- is selected from those depicted in Table 1 and 2, below.
  • each Z is hydrogen or a CRBN binding moiety.
  • each Z is hydrogen. In some embodiments, each Z is a CRBN binding moiety.
  • each Z is selected from those depicted in Table 1 and 2, below.
  • L 2 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 2 -, -C(R’)2-, -CH(R’)-, -N(R)-, -Cy 2 (L’- Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2- - S(0) 2 N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0) 2- [0087] In some embodiments, L 2 is a covalent bond.
  • L 2 is a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 2 -, -C(R’): 2-, -
  • L 2 is selected from those depicted in Table 1 and 2, below.
  • each -Cy 2 - is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 2 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy - is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • -Cy 2 - is selected from those depicted in Table 1 and 2,
  • L 2 is . In some embodiments, L 2 is . In
  • L 2 is In some embodiments, L 2 is , In some embodiments, L 2 is ,
  • L 3 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 3 -, -C(R’)2-, -CH(R’)-, -N(R)-, -Cy 3 (L’- Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2- - S(0) 2 N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0) - [0094] In some embodiments, L 3 is a covalent bond.
  • L 3 is a C MO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 3 -, -C(R’) 2 - -
  • L 3 is selected from those depicted in Table 1 and 2, below.
  • each -Cy 5 - is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 5 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 5 - is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • L 4 is a covalent bond or a Ci-15 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the
  • each -Cy 4 - is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 4 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 4 - is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • -Cy 4 - is selected from those depicted in Table 1 and 2, below.
  • each R 1 is independently hydrogen, halogen, -CN, -
  • each R 1 is independently hydrogen. In some embodiments, each R 1 is independently halogen. In some embodiments, each R 1 is independently -CN. In some embodiments, each R 1 is independently -N0 2 . In some embodiments, each R 1 is independently - OR. In some embodiments, each R 1 is independently -SR. In some embodiments, each R 1 is independently -NR2. In some embodiments, each R 1 is independently -S(0) 2 R. In some embodiments, each R 1 is independently -S(0)(NR)R. In some embodiments, each R 1 is independently -S(0) 2 NR 2 . In some embodiments, each R 1 is independently -S(0)R.
  • each R 1 is independently -S(0) R 2 . In some embodiments, each R 1 is independently -C(0)R. In some embodiments, each R 1 is independently -C(0)0R. In some embodiments, each R 1 is independently -C(0)NR. 2. In some embodiments, each R 1 is independently -C(0)N(R)0R. In some embodiments, each R 1 is independently -0C(0)R. In some embodiments, each R 1 is independently -0C(0)NR 2 . In some embodiments, each R 1 is independently -N(R)C(0)0R. In some embodiments, each R 1 is independently -N(R)C(0)NR 2 . In some embodiments, each R 1 is independently -N(R)C(NR) R2.
  • each R 1 is independently -N(R)S(0) 2 NR 2 . In some embodiments, each R 1 is independently - N(R)S(0) 2 R. In some embodiments, each R 1 is independently -P(0)R 2 . In some embodiments, each R 1 is independently -L’-Z.
  • each R 1 is independently an optionally substituted group selected from Ci- 6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • R 1 is selected from those depicted in Table 1 and 2, below.
  • R 1 is methyl. In some embodiments, R 1 is -C(0)NH 2 . In some embodiments, R 1 is -OMe. In some embodiments, R 1 is In some embodiments,
  • R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some embodiments, R 1 is In some
  • R 1 is In some embodiments, R 1 is -C(0)Me. In some
  • R 1 is -C(OH)Me. In some embodiments, R 1 is ⁇ — / . In some embodiments, R 1 is -CN. In some embodiments, R 1 is bromo. In some embodiments, R 1 is - 0
  • R 1 is H . In certain embodiments,
  • R 1 is H In certain embodiments, R 1 is H . In certain
  • R 1 is ® In certain embodiments, R 1 is 0 . In some embodiments,
  • R 1 is . , . In 0 / — y
  • R is ⁇ .
  • R 1 is methyl. In some embodiments, R 1 is -C(0)0Me.
  • R 2 is hydrogen or an optionally substituted Ci- 6 aliphatic group.
  • each R 2 is hydrogen. In some embodiments, R 2 is an optionally substituted Ci- 6 aliphatic group.
  • R 2 is selected from those depicted in Table 1 and 2, below.
  • R 2 is methyl. In some embodiments, R 2 is ethyl. In some embodiments, R 2 is «-propyl. In some embodiments, R 2 is allyl.
  • each R 3 is independently hydrogen, oxo, halogen, -CN, -NO2, -OR,
  • R 3 is independently hydrogen. In some embodiments, R 3 is independently oxo, halogen. In some embodiments, R 3 is independently -CN. In some embodiments, R 3 is independently -NC . In some embodiments, R 3 is independently -OR. In some embodiments, R 3 is independently -SR. In some embodiments, R 3 is independently -NR2. In some embodiments, R 3 is independently -S(0) 2 R. In some embodiments, R 3 is independently -S(0)(NR)R. In some embodiments, R 3 is independently -S(0) 2 NR 2 In some embodiments, R 3 is independently -S(0)R. In some embodiments, R 3 is independently -S(0)NR 2 . In some embodiments, R 3 is independently -S(0)NR 2 .
  • R 3 is independently -C(0)R. In some embodiments, R 3 is independently -C(0)OR. In some embodiments, R 3 is independently -C(0)NR 2 . In some embodiments, R 3 is independently -C(0)N(R)OR. In some embodiments, R 3 is independently -OC(0)R. In some embodiments, R 3 is independently -OC(0)NR 2 . In some embodiments, R 3 is independently -N(R)C(0)OR. In some embodiments, R 3 is independently -N(R)C(0)NR2. In some embodiments, R 3 is independently -N(R)C(NR) R2. In some embodiments, R 3 is independently -N(R)S(0) 2 NR 2 .
  • R 3 is independently -N(R)S(0) 2 R. In some embodiments, R 3 is independently -P(0)R 2 . In some embodiments, each R 3 is independently -L’-Z. In some embodiments, R 3 is independently an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, two R 3 groups are taken together with their intervening atoms to form a 5-8 membered saturated or partially unsaturated fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
  • R 3 is selected from those depicted in Table 1 and 2, below.
  • R 3 is methyl. In some embodiments, R 3 is ethyl. In some embodiments, R 3 is chloro. In some embodiments, R 3 is -OMe. In some embodiments, R 3 is - CH2CH2OH. In some embodiments, R' is -CH2CH2CH2OH. In some embodiments, R 3 is - CH 2 CH 2 C(0)0H. In some embodiments, R 3 is -CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 3 is -CH 2 CH 2 0CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 3 is CH 2 CH 2 0CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 3 is CH 2 CH 2 0CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 3 is CH 2 CH 2 0CH 2 CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 3 is CH 2 CH 2 0CH 2 CH 2 CH 2 CH 2 C(0)0H. In some embodiments,
  • each R 4 is independently hydrogen, halogen, -CN, - NO2, -OR, -SR, -NR 2 . -S(0) 2 R, -S(0)(NR)R, -S(0) 2 NR 2 , -S(0)R, -S(0)NR 2I -C(0)R, -C(0)OR, -C(0)NR , -C(0)N(R)OR, -OC(0)R, -OC(0)NR 2 , -N(R)C(0)OR, -N(R)C(0)NR 2 , - N(R)C(0)NR 2 , - N(R)C(NR)NR 2 , -N(R)S(0) 2 NR 2 , -N(R)S(0) 2 R, -P(0)R 2 , -L’-Z, or an optionally substituted group selected from Ci- 6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membere
  • each R 4 is independently hydrogen. In some embodiments, each R 4 is independently halogen. In some embodiments, each R 4 is independently -CN. In some embodiments, each R 4 is independently -N0 2 . In some embodiments, each R 4 is independently - OR. In some embodiments, each R 4 is independently -SR. In some embodiments, each R 4 is independently -NR 2 . In some embodiments, each R 4 is independently -S(0) 2 R. In some embodiments, each R 4 is independently -S(0)(NR)R. In some embodiments, each R 4 is independently -S(0) 2 NR 2 . In some embodiments, each R 4 is independently -S(0)R.
  • each R 4 is independently -S(0)NR 2 . In some embodiments, each R 4 is independently -C(0)R. In some embodiments, each R 4 is independently -C(0)OR. In some embodiments, each R 4 is independently -C(0)NR 2 . In some embodiments, each R 4 is independently -C(0)N(R)OR. In some embodiments, each R 4 is independently -OC(0)R. In some embodiments, each R 4 is independently -0C(0)NR 2 . In some embodiments, each R 4 is independently -N(R)C(0)0R. In some embodiments, each R 4 is independently -N(R)C(0)NR 2 . In some embodiments, each R 4 is independently -N(R)C(NR)NR2.
  • each R 4 is independently -N(R)S(0) 2 NR 2 . In some embodiments, each R 4 is independently - N(R)S(0) 2 R. In some embodiments, each R 4 is independently -P(0)R 2 . In some embodiments, each R 4 is independently -L’-Z.
  • each R 4 is independently an optionally substituted group selected from Ci- 6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • R 4 is selected from those depicted in Table 1 and 2, below.
  • R 4 is methyl. In some embodiments, R 4 is -C(0) H 2 . In some embodiments, R 4 is -OMe. In some embodiments, R 4 is . In some embodiments,
  • R 4 is in some embodiments, R 4 is . In some
  • R 4 is In some embodiments, R 4 is -C(0)Me. In some
  • R 4 is -C(OH)Me. In some embodiments, R 4 is ⁇ — / . In some embodiments, R 4 is -CN. In some embodiments, R 4 is bromo. In some embodiments, R 4 is -
  • R 4 is H In certain embodiments,
  • R 4 is H In certain embodiments, R 4 is H In certain
  • R 4 is ⁇ In certain embodiments, R 4 is 0 in some embodiments, R 4 is , In
  • R 4 is In
  • R 4 is -C(0)0Me.
  • R 5 is hydrogen or an optionally substituted Ci- 6 aliphatic group.
  • each R 5 is hydrogen. In some embodiments, R 5 is an optionally substituted Ci- 6 aliphatic group.
  • R 5 is selected from those depicted in Table 1 and 2, below.
  • R 5 is methyl. In some embodiments, R 5 is ethyl. In some embodiments, R 5 is «-propyl. In some embodiments, R 5 is allyl.
  • each R 6 is independently hydrogen, oxo, halogen, -CN,
  • each R 6 is independently hydrogen. In some embodiments, each R 6 is independently oxo. In some embodiments, each R 6 is independently halogen. In some embodiments, each R 6 is independently -CN. In some embodiments, each R 6 is independently - NO2. In some embodiments, each R 6 is independently -OR. In some embodiments, each R 6 is independently -SR. In some embodiments, each R 6 is independently -NR2. In some embodiments, each R 6 is independently -S(0) 2 R. In some embodiments, each R 6 is independently -S(0)( R)R. In some embodiments, each R 6 is independently -S(0) 2 NR. 2 . In some embodiments, each R 6 is independently -S(0)R.
  • each R 6 is independently -S(0)NR 2 . In some embodiments, each R 6 is independently -C(0)R. In some embodiments, each R 6 is independently -C(0)0R. In some embodiments, each R 6 is independently -C(0)NR 2 . In some embodiments, each R 6 is independently -C(0)N(R)0R. In some embodiments, each R 6 is independently -0C(0)R. In some embodiments, each R 6 is independently -0C(0)NR 2 . In some embodiments, each R 6 is independently -N(R)C(0)0R. In some embodiments, each R 6 is independently -N(R)C(0)NR 2 . In some embodiments, each R 6 is independently -N(R)C(NR)NR2.
  • each R 6 is independently -N(R)S(0) 2 NR 2 . In some embodiments, each R 6 is independently -N(R)S(0) 2 R. In some embodiments, each R 6 is independently -P(0)R 2 . In some embodiments, each R 6 is independently -L’-Z.
  • each R 6 is independently an optionally substituted group selected from Ci- 6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • two R 6 groups are optionally taken together with their intervening atoms to form a 5-8 membered saturated or partially unsaturated fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
  • R 6 is selected from those depicted in Table 1 and 2, below.
  • R 6 is methyl. In some embodiments, R 6 is ethyl. In some embodiments, R 6 is chloro. In some embodiments, R 6 is -OMe. In some embodiments, R 6 is - CH2CH2OH. In some embodiments, R 6 is -CH2CH2CH2OH. In some embodiments, R 6 is - CH 2 CH 2 C(0)0H. In some embodiments, R 6 is -CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 6 is -CH 2 CH 2 0CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 6 is CH 2 CH 2 0CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 6 is CH 2 CH 2 0CH 2 CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 6 is CH 2 CH 2 0CH 2 CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 6 is CH 2 CH 2 0CH 2 CH 2 CH 2 CH 2 C(0)0H. In some embodiment
  • each of q, s, t, and v is independently 0, 1, 2, 3, or 4.
  • each of q, s, t, and v is independently 0. In some embodiments, each of q, s, t, and v is independently 1. In some embodiments, each of q, s, t, and v is independently 2. In some embodiments, each of q, s, t, and v is independently 3. In some embodiments, each of q, s, t, and v is independently 4.
  • each of r, u and w is independently 0 or 1.
  • each of r and u is independently 0. In some embodiments, each of r and u is independently 1.
  • each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • each of x is independently 1. In some embodiments, each of x is independently 2. In some embodiments, each of x is independently 3. In some embodiments, each of x is independently 4. In some embodiments, each of x is independently 5. In some embodiments, each of x is independently 6. In some embodiments, each of x is independently 7. In some embodiments, each of x is independently 8. In some embodiments, each of x is independently 9. In some embodiments, each of x is independently 10.
  • each of q, r, s, t, u, v, and x are selected from those depicted in Table 1 and 2, below.
  • the present invention provides a compound of formula I wherein n is 1 , thereby forming a compound of formula I-a:
  • each of Ring A, Ring B, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , L 1 , L 3 , m, p, q, r, s, t, u, and v is as defined above; and ⁇ represents a single or double bond.
  • the present invention provides a compound of formula I wherein m is 1 and L 1 is thereby forming a compound of formula I-b:
  • each of Ring A, Ring B, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , L 2 , L 3 , n, p, q, r, s, t, u, and v is as defined above.
  • the present invention provides a compound of formula I wherein p is 1 and L 3 is , thereby forming a compound of formula I-c:
  • each of Ring A, Ring B, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , L 1 , L 2 , m, n, q, r, s, t, u, and v is as defined above.
  • the present invention provides a compound of formula I wherein R 6 is -L’-Z, thereby forming a compound of formula I-d:
  • each of Ring A, Ring B, R 1 , R 2 , R 3 , R 4 , R 5 , L 1 , L 2 , L 2 , L’, m, n, p, q, r, s, t, u, and v is as defined above.
  • the present invention provides a compound of formula II:
  • n 0 or 1
  • n 0 or 1
  • p is 0 or 1;
  • L 1 is a covalent bond or a C O bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced
  • each -Cy 1 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R’ is independently hydrogen or C M aliphatic;
  • R’ two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R is independently hydrogen, or an optionally substituted group selected from CM aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or:
  • each L’ is a covalent bond or a CM5 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and
  • each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, napththylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each Z is hydrogen or a CRBN binding moiety
  • L 2 is a covalent bond or a CM O bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 2 -, -C(R’) 2- , -CH(R’)-, -N(R)-, -Cy 2 (L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)- -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2- -S(0) 2 N(R)-, -0-, -C(O)-, -OC(O)- -C(0)0-, -S-, -S(O)-, or -S(0) 2 -;
  • each -Cy 2 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • L 3 is a covalent bond or a CM O bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 3 -, -C(R’) 2- , -CH(R’)-, -N(R)-, -Cy 3 (L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2- , -S(0) 2 N(R)-, -0-, -C(0)-, -0C(0)- -C(0)0-, -S-, -S(0)-, or -S(0) 2- ;
  • each -Cy 3 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R 1 is independently hydrogen, halogen, -CN, -N0 2 , -OR,
  • R 2 is hydrogen or an optionally substituted Ci- 6 aliphatic group
  • each R 3 is independently hydrogen, oxo, halogen, -CN, -NO2, -OR,
  • each R 4 is independently hydrogen, halogen, -CN, -N0 2 , -OR,
  • R 5 is hydrogen or an optionally substituted Ci- 6 aliphatic group
  • each R 6 is independently hydrogen, oxo, halogen, -CN, -N0 2 , -OR,
  • each of q, s, t, and v is independently 0, 1, 2, 3, or 4;
  • each of r and u is independently 0 or 1;
  • each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10;
  • each R of the -C(0)NR 2 moiety is other than hydrogen or Ci-4 aliphatic;
  • m is 0 or 1. In some embodiments, m is 0. In some embodiments, m is 1.
  • n is 0 or 1. In some embodiments, n is 0. In some embodiments, m is 1. [00142] As generally described above, p is 0 or 1. In some embodiments, p is 0. In some embodiments, m is 1.
  • m, n, and p are independently selected from those depicted in Table 1, below.
  • L 1 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 1 -, -C(R’)2-, -CH(R’)-, -N(R)-, Cy '(L - Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2- - S(0) 2 N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0) 2 - [00146] In some embodiments, L 1 is a covalent bond.
  • L 1 is a Ci-io bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 1 -, -C(R’) 2- , - CH(R’)-, -N(R)-, -Cy ⁇ L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, - C(0)N(R)-, -N(R)S(0) 2- , -S(0) 2 N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(0)-, or - S(0) 2- .
  • L 1 is selected from those depicted in Table 1 and 2, below.
  • each -Cy 1 - is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 1 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 1 - is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • -Cy 1 - is selected from those depicted in Table 1 and 2, below.
  • each R’ is independently hydrogen or C aliphatic; or two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring; or two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each R’ is independently hydrogen. In some embodiments, each R’ is independently CM aliphatic. In some embodiments, two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring. In some embodimetns, two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • R’ is selected from those depicted in Table 1 and 2, below.
  • each R is independently hydrogen, or an optionally substituted group selected from C aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or two R groups on the same nitrogen are taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • each R is independently hydrogen.
  • R is an optionally substituted group selected from CM aliphatic.
  • R is phenyl.
  • R is naphthalenyl.
  • R is an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • R is a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • R is 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • R is a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • two R groups on the same nitrogen are taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • R is selected from those depicted in Table 1 and 2, below.
  • each L’ is a covalent bond or a Ci- 15 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the
  • each L’ is a covalent bond. In some embodiments, or each L’ is a Ci- 15 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and independently replaced by -Cy’-,
  • L’ is selected from those depicted in Table 1 and 2, below.
  • L’ is 0 In some embodiments, L’ is
  • L’ is 0 . In some embodiments,
  • L’ is 0 In some embodiments, L’ is
  • L’ is 0 In some embodiments, L’ is ,
  • L’ is In some embodiments, L’ is , In some embodiments, L’ is , In some embodiments, In some embodiments, L’ ,
  • L’ In some embodiments, L’ is , In some embodiments,
  • each -Cy’- is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1 -4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy’- is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • -Cy’- is selected from those depicted in Table 1 and 2, below.
  • each Z is hydrogen or a CRBN binding moiety.
  • each Z is hydrogen. In some embodiments, each Z is a CRBN binding moiety.
  • each Z is selected from those depicted in Table 1 and 2, below.
  • L 2 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 2 -, -C(R’) 2 -, -CH(R’)-, -N(R)-, -Cy 2 (L’- Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2- , - S(0) 2 N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0) -
  • L 2 is a covalent bond.
  • L 2 is a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 2 -, -C(R’)2-, -
  • L 2 is selected from those depicted in Table 1 and 2, below.
  • each -Cy 2 - is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 2 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy - is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • -Cy 2 - is selected from those depicted in Table 1 and 2,
  • L 2 is . In some embodiments, L 2 is . In some embodiments, L 2 is . In some embodiments, L 2 is In some embodiments, L 2 is . , . In some embodiments, L 2 is
  • L 3 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 3 -, -C(R’) 2- , -CH(R’)-, -N(R)-, -Cy 3 (L’- Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0) 2- , - S(0) 2 N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0) 2- [00177] In some embodiments, L 3 is a covalent bond.
  • L 2 is a Ci-io bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy 3 -, -C(R’) 2- , -
  • L 3 is selected from those depicted in Table 1 and 2, below.
  • each -Cy 5 - is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 5 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 5 - is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • -Cy 5 - is selected from those depicted in Table 1 and 2, below.
  • L 4 is a covalent bond or a Ci-15 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the
  • each -Cy 4 - is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 4 - is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • each -Cy 4 - is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • -Cy 4 - is selected from those depicted in Table 1 and 2, below.
  • each R 1 is independently hydrogen, halogen, -CN, -
  • each R 1 is independently hydrogen. In some embodiments, each R 1 is independently halogen. In some embodiments, each R 1 is independently -CN. In some embodiments, each R 1 is independently -NO2. In some embodiments, each R 1 is independently - OR. In some embodiments, each R 1 is independently -SR. In some embodiments, each R 1 is independently -NR2. In some embodiments, each R 1 is independently -S(0) 2 R. In some embodiments, each R 1 is independently -S(0)(NR)R. In some embodiments, each R 1 is independently -S(0) 2 NR 2 . In some embodiments, each R 1 is independently -S(0)R. In some embodiments, each R 1 is independently -S(0)NR 2 .
  • each R 1 is independently -C(0)R. In some embodiments, each R 1 is independently -C(0)0R. In some embodiments, each R 1 is independently -C(0)NR 2 In some embodiments, each R 1 is independently -C(0)N(R)0R. In some embodiments, each R 1 is independently -0C(0)R. In some embodiments, each R 1 is independently -0C(0)NR 2 . In some embodiments, each R 1 is independently -N(R)C(0)0R. In some embodiments, each R 1 is independently -N(R)C(0)NR 2 . In some embodiments, each R 1 is independently -N(R)C(NR)NR2.
  • each R 1 is independently -N(R)S(0) 2 NR 2 . In some embodiments, each R 1 is independently - N(R)S(0) 2 R. In some embodiments, each R 1 is independently -P(0)R 2 . In some embodiments, each R 1 is independently -L’-Z.
  • each R 1 is independently an optionally substituted group selected from Ci- 6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • R 1 is selected from those depicted in Table 1 and 2, below.
  • R 1 is methyl. In some embodiments, R 1 is -C(0)NH 2 . In some embodiments, R 1 is -OMe. In some embodiments, R 1 is In some embodiments,
  • R 1 is ⁇ — f . In some embodiments, R 1 is -CN. In some embodiments, R 1 is bromo. In some embodiments, R 1 is -CO2H. In some embodiments, R 1
  • R is 0 .
  • R i:s. .
  • R is 0 .
  • R 1 i is OH . In some embodiments, R 1 is In some embodiments, , . In some embodiments,
  • R 1 is methyl. In some embodiments, R 1 is -C(0)OMe.
  • R 2 is hydrogen or an optionally substituted Ci- 6 aliphatic group.
  • each R 2 is hydrogen. In some embodiments, R 2 is an optionally substituted Ci- 6 aliphatic group.
  • R 2 is selected from those depicted in Table 1 and 2, below.
  • R 2 is methyl. In some embodiments, R 2 is ethyl. In some embodiments, R 2 is «-propyl. In some embodiments, R 2 is allyl.
  • each R 3 is independently hydrogen, oxo, halogen, -CN, -NO2, -OR,
  • R 3 is independently hydrogen. In some embodiments, R 3 is independently oxo, halogen. In some embodiments, R 3 is independently -CN. In some embodiments, R 3 is independently -NO2. In some embodiments, R 3 is independently -OR. In some embodiments, R 3 is independently -SR. In some embodiments, R 3 is independently -NR2. In some embodiments, R 3 is independently -S(0) 2 R. In some embodiments, R 3 is independently -S(0)(NR)R. In some embodiments, R 3 is independently -S(0) 2 NR 2 In some embodiments, R 3 is independently -S(0)R. In some embodiments, R 3 is independently -S(0)NR 2 . In some embodiments, R 3 is independently -S(0)NR 2 .
  • R 3 is independently -C(0)R. In some embodiments, R 3 is independently -C(0)OR. In some embodiments, R 3 is independently -C(0)NR 2 . In some embodiments, R 3 is independently -C(0)N(R)OR. In some embodiments, R 3 is independently -OC(0)R. In some embodiments, R 3 is independently -OC(0)NR 2 . In some embodiments, R 3 is independently -N(R)C(0)OR. In some embodiments, R 3 is independently -N(R)C(0)NR2. In some embodiments, R 3 is independently -N(R)C(NR) R2. In some embodiments, R 3 is independently -N(R)S(0) 2 NR 2 .
  • R 3 is independently -N(R)S(0)2R. In some embodiments, R 3 is independently -P(0)R2. In some embodiments, each R 3 is independently -L’-Z. In some embodiments, R 3 is independently an optionally substituted group selected from Ci- 6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R 3 is independently two R 3 groups are optionally taken together with their intervening atoms to form a 5-8 membered saturated or partially unsaturated fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
  • R 3 is selected from those depicted in Table 1 and 2, below.
  • R 3 is methyl. In some embodiments, R 3 is ethyl. In some embodiments, R 3 is chloro. In some embodiments, R 3 is -OMe. In some embodiments, R 3 is - CH2CH2OH. In some embodiments, R 3 is -CH2CH2CH2OH. In some embodiments, R 3 is - CH 2 CH 2 C(0)0H. In some embodiments, R 3 is -CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 3 is -CH 2 CH 2 0CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 3 is CH 2 CH 2 0CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 3 is CH 2 CH 2 0CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 3 is CH 2 CH 2 0CH 2 CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 3 is CH 2 CH 2 0CH 2 CH 2 CH 2 CH 2 C(0)0H. In some embodiments,
  • each R 4 is independently hydrogen, halogen, -CN, - NO2, -OR, -SR, -NR 2 , -S(0) 2 R, -S(0)(NR)R, -S(0) 2 NR 2 , -S(0)R, -S(0)NR 2 , -C(0)R, -C(0)0R, -C(0)NR 2 , -C(0)N(R)0R, -0C(0)R, -0C(0)NR 2 , -N(R)C(0)0R, -N(R)C(0)NR 2 , - N(R)C(NR)NR 2 , -N(R)S(0) 2 NR 2 , -N(R)S(0) 2 R, -P(0)R 2 , -L’-Z, or an optionally substituted group selected from Ci- 6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5
  • each R 4 is independently hydrogen. In some embodiments, each R 4 is independently halogen. In some embodiments, each R 4 is independently -CN. In some embodiments, each R 4 is independently -N0 2 . In some embodiments, each R 4 is independently - OR. In some embodiments, each R 4 is independently -SR. In some embodiments, each R 4 is independently -NR 2 . In some embodiments, each R 4 is independently -S(0) 2 R. In some embodiments, each R 4 is independently -S(0)(NR)R. In some embodiments, each R 4 is independently -S(0) 2 NR 2 . In some embodiments, each R 4 is independently -S(0)R.
  • each R 4 is independently -S(0)NR 2 . In some embodiments, each R 4 is independently -C(0)R. In some embodiments, each R 4 is independently -C(0)OR. In some embodiments, each R 4 is independently -C(0)NR 2 . In some embodiments, each R 4 is independently -C(0)N(R)OR. In some embodiments, each R 4 is independently -OC(0)R. In some embodiments, each R 4 is independently -OC(0)NR 2 . In some embodiments, each R 4 is independently -N(R)C(0)OR. In some embodiments, each R 4 is independently -N(R)C(0)NR 2 . In some embodiments, each R 4 is independently -N(R)C(NR)NR 2 . In some embodiments, each R 4 is independently -N(R)C(NR)NR 2 .
  • each R 4 is independently -N(R)S(0) 2 NR 2 . In some embodiments, each R 4 is independently - N(R)S(0) 2 R. In some embodiments, each R 4 is independently -P(0)R 2 . In some embodiments, each R 4 is independently -L’-Z.
  • each R 4 is independently an optionally substituted group selected from Ci- 6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • R 4 is selected from those depicted in Table 1 and 2, below.
  • R 4 is methyl. In some embodiments, R 4 is -C(0)NH 2 . In some embodiments, R 4 is -OMe. In some embodiments, R 4 i is, ⁇ 0 ⁇ 0H . In some embodiments,
  • R 4 is ⁇ — f . In some embodiments, R 4 is -CN. In some embodiments, R 4 is bromo. In some embodiments, R 4 is -CO2H. In some embodiments, R 1
  • R 4 is In some embodiments.
  • R 4 is . In some embodiments, R 4 is O . In some embodiments,
  • R 4 is -C(0)0Me.
  • R 5 is hydrogen or an optionally substituted Ci-6 aliphatic group.
  • each R 5 is hydrogen. In some embodiments, R 5 is an optionally substituted Ci- 6 aliphatic group.
  • R 5 is selected from those depicted in Table 1 and 2, below.
  • R 5 is methyl. In some embodiments, R 5 is ethyl. In some embodiments, R 5 is «-propyl. In some embodiments, R 5 is allyl.
  • each R 6 is independently hydrogen, oxo, halogen, -CN, -NOi, -OR,
  • each R 6 is independently hydrogen. In some embodiments, each R 6 is independently oxo. In some embodiments, each R 6 is independently halogen. In some embodiments, each R 6 is independently -CN. In some embodiments, each R 6 is independently - N0 2 . In some embodiments, each R 6 is independently -OR. In some embodiments, each R 6 is independently -SR. In some embodiments, each R 6 is independently -NR 2 . In some embodiments, each R 6 is independently -S(0) 2 R. In some embodiments, each R 6 is independently -S(0)(NR)R. In some embodiments, each R 6 is independently -S(0) 2 NR 2 . In some embodiments, each R 6 is independently -S(0)R.
  • each R 6 is independently -S(0)NR 2 . In some embodiments, each R 6 is independently -C(0)R. In some embodiments, each R 6 is independently -C(0)OR. In some embodiments, each R 6 is independently -C(0)NR 2 . In some embodiments, each R 6 is independently -C(0)N(R)OR. In some embodiments, each R 6 is independently -OC(0)R. In some embodiments, each R 6 is independently -OC(0)NR 2 . In some embodiments, each R 6 is independently -N(R)C(0)OR. In some embodiments, each R 6 is independently -N(R)C(0)NR 2 . In some embodiments, each R 6 is independently -N(R)C(NR)NR 2 . In some embodiments, each R 6 is independently -N(R)C(NR)NR 2 .
  • each R 6 is independently -N(R)S(0) 2 NR 2 . In some embodiments, each R 6 is independently -N(R)S(0) 2 R. In some embodiments, each R 6 is independently -P(0)R 2 . In some embodiments, each R 6 is independently -L’-Z.
  • each R 6 is independently an optionally substituted group selected from Ci- 6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • two R 6 groups are optionally taken together with their intervening atoms to form a 5-8 membered saturated or partially unsaturated fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
  • R 6 is selected from those depicted in Table 1 and 2, below.
  • R 6 is methyl. In some embodiments, R 6 is ethyl. In some embodiments, R 6 is chloro. In some embodiments, R 6 is -OMe. In some embodiments, R 6 is - CH2CH2OH. In some embodiments, R 6 is -CH2CH2CH2OH. In some embodiments, R 6 is - CH 2 CH 2 C(0)0H. In some embodiments, R 6 is -CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 6 is -CH 2 CH 2 0CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 6 is CH 2 CH 2 0CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 6 is CH 2 CH 2 0CH 2 CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 6 is CH 2 CH 2 0CH 2 CH 2 CH 2 CH 2 C(0)0H. In some embodiments, R 6 is CH 2 CH 2 0CH 2 CH 2 CH 2 CH 2 C(0)0H. In some embodiment
  • each of q, s, t, and v is independently 0, 1, 2, 3, or 4.
  • each of q, s, t, and v is independently 0. In some embodiments, each of q, s, t, and v is independently 1. In some embodiments, each of q, s, t, and v is independently 2. In some embodiments, each of q, s, t, and v is independently 3. In some embodiments, each of q, s, t, and v is independently 4.
  • each of r, u and w is independently 0 or 1.
  • each of r and u is independently 0. In some embodiments, each of r and u is independently 1.
  • each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • each of x is independently 1. In some embodiments, each of x is independently 2. In some embodiments, each of x is independently 3. In some embodiments, each of x is independently 4. In some embodiments, each of x is independently 5. In some embodiments, each of x is independently 6. In some embodiments, each of x is independently 7. In some embodiments, each of x is independently 8. In some embodiments, each of x is independently 9. In some embodiments, each of x is independently 10. [00217] In some embodiments, each of q, r, s, t, u, v, and x are selected from those depicted in Table 1 and 2, below.
  • each R of the -C(0)NR 2 moiety is other than hydrogen or CM aliphatic;
  • the present invention provides a compound of formula II wherein n is 1 and L 2 is , ere by forming a compound of formula Il-a:
  • the present invention provides a compound of formula II wherein m is 1 and L 1 is , thereby forming a compound of formula Il-b
  • each of R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , L 2 , L 3 , n, p, q, r, s, t, u, and v is as defined above.
  • the present invention provides a compound of formula II wherein p is 1 and L 3 is , thereby forming a compound of formula II-c:
  • each of R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , L 1 , L 2 , m, n, q, r, s, t, u, and v is as defined above.
  • the present invention provides a compound of formula II wherein R 6 is -L’-Z, thereby forming a compound of formula Il-d:
  • each of R 1 , R 2 , R 3 , R 5 , R 6 , L 1 , L 2 , L 3 , L’, Z, m, n, p, q, r, s, t, u, and v is as defined above.
  • the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • the amount of compound in compositions of this invention is such that is effective to modulate STING, or a mutant thereof, - dependent type I interferon production in a biological sample or in a patient.
  • the amount of compound in compositions of this invention is such that is effective to measurably modulate STING, or a mutant thereof, -dependent type I interferon production in a biological sample or in a patient.
  • a composition of this invention is formulated for administration to a patient in need of such composition.
  • a composition of this invention is formulated for oral administration to a patient.
  • the term“patient,” as used herein, means an animal, preferably a mammal, and most preferably a human.
  • compositions of this invention refers to a nontoxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated.
  • Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropy
  • A“pharmaceutically acceptable derivative” means any non-toxic salt, ester, salt of an ester or other derivative of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an active metabolite or residue thereof.
  • compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • the compositions are administered orally, intraperitoneally or intravenously.
  • Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • a non-toxic parenterally acceptable diluent or solvent for example as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or di-glycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxy ethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include lactose and com starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
  • compositions of this invention may be administered in the form of suppositories for rectal administration.
  • suppositories for rectal administration.
  • suppositories can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, beeswax and polyethylene glycols.
  • compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
  • Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used.
  • compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers.
  • Carriers for topical administration of compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • provided pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride.
  • the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.
  • compositions of this invention may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well- known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents. [00237] Most preferably, pharmaceutically acceptable compositions of this invention are formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, pharmaceutically acceptable compositions of this invention are administered without food. In other embodiments, pharmaceutically acceptable compositions of this invention are administered with food.
  • compositions of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration.
  • provided compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the compound can be administered to a patient receiving these compositions.
  • a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated.
  • the amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
  • the invention provides a method of inducing an immune response in a subject, comprising administering a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof to the subject.
  • the invention provides a method of inducing a STING- dependent type I interferon production in a subject, comprising administering a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof to the subject.
  • the invention provides a method of inducing a STING- dependent cytokine production in a subject, comprising administering a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof to the subject.
  • the invention provides a pharmaceutical composition comprising an effective amount of a compound of the invention, or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof, and a pharmaceutically acceptable carrier.
  • the invention provides a method of treating cell proliferation disorders, including cancers, benign papillomatosis, gestational trophoblastic diseases, and benign neoplastic diseases, such as skin papilloma (warts) and genital papilloma.
  • the invention provides a method of treating a cell proliferation disorder in a subject, comprising administering a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof, to the subj ect.
  • the cell proliferation disorder is cancer.
  • the cancer is brain cancer, leukemia, skin cancer, prostate cancer, thyroid cancer, colon cancer, lung cancer or sarcoma.
  • the cancer is selected from the group consisting of glioma, glioblastoma multiforme, paraganglioma, suprantentorial primordial neuroectodermal tumors, acute myeloid leukemia, myelodysplastic syndrome, chronic myelogenous leukemia, melanoma, breast, prostate, thyroid, colon, lung, central chondrosarcoma, central and periosteal chondroma tumors, fibrosarcoma, and cholangiocarcinoma.
  • the cancer is selected from brain and spinal cancers, cancers of the head and neck, leukemia and cancers of the blood, skin cancers, cancers of the reproductive system, cancers of the gastrointestinal system, liver and bile duct cancers, kidney and bladder cancers, bone cancers, lung cancers, malignant mesothelioma, sarcomas, lymphomas, glandular cancers, thyroid cancers, heart tumors, germ cell tumors, malignant neuroendocrine (carcinoid) tumors, midline tract cancers, and cancers of unknown primary (i.e., cancers in which a metastasized cancer is found but the original cancer site is not known).
  • the cancer is present in an adult patient; in additional embodiments, the cancer is present in a pediatric patient.
  • the cancer is AIDS-related.
  • the cancer is selected from brain and spinal cancers.
  • the cancer is selected from the group consisting of anaplastic astrocytomas, glioblastomas, astrocytomas, and estheosioneuroblastomas (olfactory blastomas).
  • the brain cancer is selected from the group consisting of astrocytic tumor (e.g., pilocytic astrocytoma, subependymal giant-cell astrocytoma, diffuse astrocytoma, pleomorphic xanthoastrocytoma, anaplastic astrocytoma, astrocytoma, giant cell glioblastoma, glioblastoma, secondary glioblastoma, primary adult glioblastoma, and primary pediatric glioblastoma), oligodendroglial tumor (e.g., oligodendroglioma, and anaplastic oligodendroglioma), oligoastrocytic tumor (e.g., oligoastrocytoma, and anaplastic oligoastrocytoma), ependymoma (e.g., myxopapillary ependymoma, and anaplastic aplastic
  • the cancer is selected from cancers of the head and neck, including nasopharyngeal cancers, nasal cavity and paranasal sinus cancers, hypopharyngeal cancers, oral cavity cancers (e.g., squamous cell carcinomas, lymphomas, and sarcomas), lip cancers, oropharyngeal cancers, salivary gland tumors, cancers of the larynx (e.g., laryngeal squamous cell carcinomas, rhabdomyosarcomas), and cancers of the eye or ocular cancers.
  • the ocular cancer is selected from the group consisting of intraocular melanoma and retinoblastoma.
  • the cancer is selected from leukemia and cancers of the blood.
  • the cancer is selected from the group consisting of myeloproliferative neoplasms, myelodysplastic syndromes, myelodysplastic/myeloproliferative neoplasms, acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), chronic myelogenous leukemia (CML), myeloproliferative neoplasm (MPN), post-MPN AML, post-MDS AML, del(5q)- associated high risk MDS or AML, blast-phase chronic myelogenous leukemia, angioimmunoblastic lymphoma, acute lymphoblastic leukemia, Langerans cell histiocytosis, hairy cell leukemia, and plasma cell neoplasms including plasmacytomas and multiple myelomas.
  • Leukemias referenced herein may be acute or chronic.
  • the cancer is selected from skin cancers.
  • the skin cancer is selected from the group consisting of melanoma, squamous cell cancers, and basal cell cancers.
  • the cancer is selected from cancers of the reproductive system.
  • the cancer is selected from the group consisting of breast cancers, cervical cancers, vaginal cancers, ovarian cancers, prostate cancers, penile cancers, and testicular cancers.
  • the cancer is a breast cancer selected from the group consisting of ductal carcinomas and phyllodes tumors.
  • the breast cancer may be male breast cancer or female breast cancer.
  • the cancer is a cervical cancer selected from the group consisting of squamous cell carcinomas and adenocarcinomas.
  • the cancer is an ovarian cancer selected from the group consisting of epithelial cancers.
  • the cancer is selected from cancers of the gastrointestinal system.
  • the cancer is selected from the group consisting of esophageal cancers, gastric cancers (also known as stomach cancers), gastrointestinal carcinoid tumors, pancreatic cancers, gallbladder cancers, colorectal cancers, and anal cancer.
  • the cancer is selected from the group consisting of esophageal squamous cell carcinomas, esophageal adenocarcinomas, gastric adenocarcinomas, gastrointestinal carcinoid tumors, gastrointestinal stromal tumors, gastric lymphomas, gastrointestinal lymphomas, solid pseudopapillary tumors of the pancreas, pancreatoblastoma, islet cell tumors, pancreatic carcinomas including acinar cell carcinomas and ductal adenocarcinomas, gallbladder adenocarcinomas, colorectal adenocarcinomas, and anal squamous cell carcinomas.
  • the cancer is selected from liver and bile duct cancers.
  • the cancer is liver cancer (hepatocellular carcinoma).
  • the cancer is bile duct cancer (cholangiocarcinoma); in instances of these embodiments, the bile duct cancer is selected from the group consisting of intrahepatic cholangiocarcinoma and extrahepatic cholangiocarcinoma.
  • the cancer is selected from kidney and bladder cancers.
  • the cancer is a kidney cancer selected from the group consisting of renal cell cancer, Wilms tumors, and transitional cell cancers.
  • the cancer is a bladder cancer selected from the group consisting of urethelial carcinoma (a transitional cell carcinoma), squamous cell carcinomas, and adenocarcinomas.
  • the cancer is selected from bone cancers.
  • the bone cancer is selected from the group consisting of osteosarcoma, malignant fibrous histiocytoma of bone, Ewing sarcoma, and chordoma.
  • the cancer is selected from lung cancers.
  • the lung cancer is selected from the group consisting of non-small cell lung cancer, small cell lung cancers, bronchial tumors, and pleuropulmonary blastomas.
  • the cancer is selected from malignant mesothelioma.
  • the cancer is selected from the group consisting of epithelial mesothelioma and sarcomatoids.
  • the cancer is selected from sarcomas.
  • the sarcoma is selected from the group consisting of central chondrosarcoma, central and periosteal chondroma, fibrosarcoma, clear cell sarcoma of tendon sheaths, and Kaposi's sarcoma.
  • the cancer is selected from lymphomas.
  • the cancer is selected from the group consisting of Hodgkin lymphoma (e.g., Reed- Stemberg cells), non-Hodgkin lymphoma (e.g., diffuse large B-cell lymphoma, follicular lymphoma, mycosis fungoides, Sezary syndrome, primary central nervous system lymphoma), cutaneous T-cell lymphomas, and primary central nervous system lymphomas.
  • Hodgkin lymphoma e.g., Reed- Stemberg cells
  • non-Hodgkin lymphoma e.g., diffuse large B-cell lymphoma, follicular lymphoma, mycosis fungoides, Sezary syndrome, primary central nervous system lymphoma
  • cutaneous T-cell lymphomas e.g., T-cell lymphomas
  • primary central nervous system lymphomas e.g., cutaneous T-cell lymphomas
  • the cancer is selected from glandular cancers.
  • the cancer is selected from the group consisting of adrenocortical cancer, pheochromocytomas, paragangliomas, pituitary tumors, thymoma, and thymic carcinomas.
  • the cancer is selected from thyroid cancers.
  • the thyroid cancer is selected from the group consisting of medullary thyroid carcinomas, papillary thyroid carcinomas, and follicular thyroid carcinomas.
  • the cancer is selected from germ cell tumors.
  • the cancer is selected from the group consisting of malignant extracranial germ cell tumors and malignant extragonadal germ cell tumors.
  • the malignant extragonadal germ cell tumors are selected from the group consisting of nonseminomas and seminomas.
  • the cancer is selected from heart tumors.
  • the heart tumor is selected from the group consisting of malignant teratoma, lymphoma, rhabdomyosacroma, angiosarcoma, chondrosarcoma, infantile fibrosarcoma, and synovial sarcoma.
  • the cell-proliferation disorder is selected from benign papillomatosis, benign neoplastic diseases and gestational trophoblastic diseases.
  • the benign neoplastic disease is selected from skin papilloma (warts) and genital papilloma.
  • the gestational trophoblastic disease is selected from the group consisting of hydatidiform moles, and gestational trophoblastic neoplasia (e.g., invasive moles, choriocarcinomas, placental-site trophoblastic tumors, and epithelioid trophoblastic tumors).
  • additional therapeutic agents which are normally administered to treat that condition, may be administered in combination with compounds and compositions of this invention.
  • additional therapeutic agents that are normally administered to treat a particular disease, or condition are known as“appropriate for the disease, or condition, being treated.”
  • a provided combination, or composition thereof is administered in combination with another therapeutic agent.
  • combination therapies of the present invention are administered in combination with a monoclonal antibody or an siRNA therapeutic.
  • Those additional agents may be administered separately from a provided combination therapy, as part of a multiple dosage regimen.
  • those agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as part of a multiple dosage regime, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another normally within five hours from one another.
  • the term“combination,”“combined,” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention.
  • a combination of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form.
  • the amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent.
  • the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.
  • the present invention provides a composition comprising a compound of formula I or formula II, and one or more additional therapeutic agents.
  • the therapeutic agent may be administered together with a compound of formula I or formula II, or may be administered prior to or following administration of a compound of formula I or formula II. Suitable therapeutic agents are described in further detail below.
  • a compound of formula I or formula II may be administered up to 5 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5, hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, or 18 hours before the therapeutic agent.
  • a compound of formula I or formula II may be administered up to 5 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5, hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, or 18 hours following the therapeutic agent.
  • the present invention provides a method of treating an inflammatory disease, disorder or condition by administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents.
  • additional therapeutic agents may be small molecules or recombinant biologic agents and include, for example, acetaminophen, non-steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, etodolac (Lodine®) and celecoxib, colchicine (Colcrys®), corticosteroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like, probenecid, allopurinol, febuxostat (Uloric®), sulfasalazine (Azulfidine®), antimalarials such as hydroxychloroquine (Plaquenil®) and chloroquine (Aralen®),
  • NSAIDS non
  • the present invention provides a method of treating rheumatoid arthritis comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from non-steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, etodolac (Lodine®) and celecoxib, corticosteroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like, sulfasalazine (Azulfidine®), antimalarials such as hydroxychloroquine (Plaquenil®) and chloroquine (Aralen®), methotrexate (Rheumatrex®), gold salts such as gold thioglucose (Solganal®), gold thiomalate (Myochrysine®) and auranofm (Ridaura®
  • NSAIDS non-ster
  • the present invention provides a method of treating osteoarthritis comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from acetaminophen, non steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, etodolac (Lodine®) and celecoxib, diclofenac, cortisone, hyaluronic acid (Synvisc® or Hyalgan®) and monoclonal antibodies such as tanezumab.
  • NSAIDS non steroidal anti-inflammatory drugs
  • the present invention provides a method of treating cutaneous lupus erythematosus or systemic lupus erythematosus comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from acetaminophen, non-steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, etodolac (Lodine®) and celecoxib, corticosteroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like, antimalarials such as hydroxychloroquine (Plaquenil®) and chloroquine (Aralen®), cyclophosphamide (Cytoxan®), methotrexate (Rheumatrex®), azathioprine (Imuran®) and anticoagulants such as hepar
  • NSAIDS non-steroidal
  • the present invention provides a method of treating Crohn’s disesase, ulcerative colitis, or inflammatory bowel disease comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from mesalamine (Asacol®) sulfasalazine (Azulfidine®), antidiarrheals such as diphenoxylate (Lomotil®) and loperamide (Imodium®), bile acid binding agents such as cholestyramine, alosetron (Lotronex®), lubiprostone (Amitiza®), laxatives such as Milk of Magnesia, polyethylene glycol (MiraLax®), Dulcolax®, Correctol® and Senokot® and anticholinergics or antispasmodics such as dicyclomine (Bentyl®), anti-TNF therapies, steroids, and antibiotics such as Flagyl or cip
  • the present invention provides a method of treating asthma comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from Singulair®, beta-2 agonists such as albuterol (Ventolin® HFA, Proventil® HFA), levalbuterol (Xopenex®), metaproterenol (Alupent®), pirbuterol acetate (Maxair®), terbutaline sulfate (Brethaire®), salmeterol xinafoate (Serevent®) and formoterol (Foradil®), anticholinergic agents such as ipratropium bromide (Atrovent®) and tiotropium (Spiriva®), inhaled corticosteroids such as prednisone, prednisolone, beclomethasone dipropionate (Beclovent®, Qvar®, and Vanceril®), triamcinolone acet
  • Singulair® beta
  • the present invention provides a method of treating COPD comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from beta-2 agonists such as albuterol (Ventolin® HFA, Proventil® HFA), levalbuterol (Xopenex®), metaproterenol (Alupent®), pirbuterol acetate (Maxair®), terbutaline sulfate (Brethaire®), salmeterol xinafoate (Serevent®) and formoterol (Foradil®), anticholinergic agents such as ipratropium bromide (Atrovent®) and tiotropium (Spiriva®), methylxanthines such as theophylline (Theo-Dur®, Theolair®, Slo-bid®, Uniphyl®, Theo-24®) and aminophylline, inhaled corticosteroids such as pred
  • beta-2 agonists such as
  • the present invention provides a method of treating a hematological malignancy comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from rituximab (Rituxan®), cyclophosphamide (Cytoxan®), doxorubicin (Hydrodaunorubicin®), vincristine (Oncovin®), prednisone, a hedgehog signaling inhibitor, a BTK inhibitor, a JAK/pan-JAK inhibitor, a PI3K inhibitor, a SYK inhibitor, and combinations thereof.
  • the present invention provides a method of treating a solid tumor comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from rituximab (Rituxan®), cyclophosphamide (Cytoxan®), doxorubicin (Hydrodaunorubicin®), vincristine (Oncovin®), prednisone, a hedgehog signaling inhibitor, a BTK inhibitor, a JAK/pan-JAK inhibitor, a PI3K inhibitor, a SYK inhibitor, and combinations thereof.
  • the present invention provides a method of treating a hematological malignancy comprising administering to a patient in need thereof a compound of formula I or formula II, and a Hedgehog (Hh) signaling pathway inhibitor.
  • the hematological malignancy is DLBCL (Ramirez et al“Defining causative factors contributing in the activation of hedgehog signaling in diffuse large B-cell lymphoma” Leuk. Res. (2012), published online July 17, and incorporated herein by reference in its entirety).
  • the present invention provides a method of treating diffuse large B-cell lymphoma (DLBCL) comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from rituximab (Rituxan®), cyclophosphamide (Cytoxan®), doxorubicin (Hydrodaunorubicin®), vincristine (Oncovin®), prednisone, a hedgehog signaling inhibitor, and combinations thereof.
  • rituximab Rostuxan®
  • Cytoxan® cyclophosphamide
  • doxorubicin Hydrodaunorubicin®
  • vincristine Oncovin®
  • prednisone a hedgehog signaling inhibitor
  • the present invention provides a method of treating multiple myeloma comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from bortezomib (Velcade®), and dexamethasone (Decadron®), a hedgehog signaling inhibitor, a BTK inhibitor, a JAK/pan- JAK inhibitor, a TYK2 inhibitor, a PI3K inhibitor, a SYK inhibitor in combination with lenalidomide (Revlimid®).
  • additional therapeutic agents selected from bortezomib (Velcade®), and dexamethasone (Decadron®), a hedgehog signaling inhibitor, a BTK inhibitor, a JAK/pan- JAK inhibitor, a TYK2 inhibitor, a PI3K inhibitor, a SYK inhibitor in combination with lenalidomide (Revlimid®).
  • the present invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I or formula II, and a BTK inhibitor, wherein the disease is selected from inflammatory bowel disease, arthritis, cutaneous lupus erythematosus, systemic lupus erythematosus (SLE), vasculitis, idiopathic thrombocytopenic purpura (ITP), rheumatoid arthritis, psoriatic arthritis, osteoarthritis, Still’s disease, juvenile arthritis, diabetes, myasthenia gravis, Hashimoto’s thyroiditis, Ord’s thyroiditis, Graves’ disease, autoimmune thyroiditis, Sjogren’s syndrome, multiple sclerosis, systemic sclerosis, Lyme neuroborreliosis, Guillain-Barre syndrome, acute disseminated encephalomyelitis, Addison’s disease, opsoclo
  • the present invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I or formula II, and a PI3K inhibitor, wherein the disease is selected from a cancer, a neurodegenative disorder, an angiogenic disorder, a viral disease, an autoimmune disease, an inflammatory disorder, a hormone-related disease, conditions associated with organ transplantation, immunodeficiency disorders, a destructive bone disorder, a proliferative disorder, an infectious disease, a condition associated with cell death, thrombin-induced platelet aggregation, chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), liver disease, pathologic immune conditions involving T cell activation, a cardiovascular disorder, and a CNS disorder.
  • the disease is selected from a cancer, a neurodegenative disorder, an angiogenic disorder, a viral disease, an autoimmune disease, an inflammatory disorder, a hormone-related disease, conditions associated with organ
  • the present invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I or formula II, and a PI3K inhibitor, wherein the disease is selected from benign or malignant tumor, carcinoma or solid tumor of the brain, kidney (e.g., renal cell carcinoma (RCC)), liver, adrenal gland, bladder, breast, stomach, gastric tumors, ovaries, colon, rectum, prostate, pancreas, lung, vagina, endometrium, cervix, testis, genitourinary tract, esophagus, larynx, skin, bone or thyroid, sarcoma, glioblastomas, neuroblastomas, multiple myeloma or gastrointestinal cancer, especially colon carcinoma or colorectal adenoma or a tumor of the neck and head, an epidermal hyperproliferation, psoriasis, prostate hyperplasia, a neoplasi
  • hemolytic anemia aplastic anemia, pure red cell anemia and idiopathic thrombocytopenia
  • cutaneous lupus erythematosus systemic lupus erythematosus
  • rheumatoid arthritis polychondritis
  • sclerodoma a progressive granulamatosis
  • dermatomyositis chronic active hepatitis
  • myasthenia gravis Steven-Johnson syndrome
  • idiopathic sprue autoimmune inflammatory bowel disease (e.g.
  • ulcerative colitis and Crohn's disease endocrine opthalmopathy
  • Grave's disease sarcoidosis, alveolitis, chronic hypersensitivity pneumonitis, multiple sclerosis, pri ary biliary cirrhosis, uveitis (anterior and posterior), keratoconjunctivitis sicca and vernal keratoconjunctivitis, interstitial lung fibrosis, psoriatic arthritis and glomerulonephritis (with and without nephrotic syndrome, e.g.
  • idiopathic nephrotic syndrome or minal change nephropathy, restenosis, cardiomegaly, atherosclerosis, myocardial infarction, ischemic stroke and congestive heart failure, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and cerebral ischemia, and neurodegenerative disease caused by traumatic injury, glutamate neurotoxicity and hypoxia.
  • the present invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I or formula II, and a Bcl-2 inhibitor, wherein the disease is an inflammatory disorder, an autoimmune disorder, a proliferative disorder, an endocrine disorder, a neurological disorder, or a disorder associated with transplantation.
  • the disorder is a proliferative disorder, lupus, or lupus nephritis.
  • the proliferative disorder is chronic lymphocytic leukemia, diffuse large B-cell lymphoma, Hodgkin’s disease, small-cell lung cancer, non-small-cell lung cancer, myelodysplastic syndrome, lymphoma, a hematological neoplasm, or solid tumor.
  • the disease is an autoimmune disorder, an inflammatory disorder, a proliferative disorder, an endocrine disorder, a neurological disorder, or a disorder associated with transplantation.
  • the compounds and compositions, according to the method of the present invention may be administered using any amount and any route of administration effective for treating or lessening the severity of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, an endocrine disorder, a neurological disorder, or a disorder associated with transplantation.
  • the exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like.
  • Compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage.
  • the expression "dosage unit form" as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated.
  • the specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
  • patient means an animal, preferably a mammal, and most preferably a human.
  • compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated.
  • the compounds of the invention may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • the oral compositions can also include
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • injectable formulations can be sterilized, for example, by filtration through a bacterial- retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • a compound of the present invention In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide- polyglycolide.
  • the rate of compound release can be controlled.
  • biodegradable polymers include poly(orthoesters) and poly(anhydrides).
  • Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
  • compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl
  • Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
  • the active compounds can also be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • buffering agents include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preserv atives or buffers as may be required.
  • Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention.
  • the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body.
  • Such dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • the invention relates to a method of inhibiting protein kinase activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.
  • the invention relates to a method of activating STING, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.
  • the invention relates to a method of irreversibly inhibiting STING, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.
  • biological sample includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • Activation of STING (or a mutant thereof) activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ-transplantation, biological specimen storage, and biological assays.
  • Another embodiment of the present invention relates to a method of activating STING in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound.
  • the invention relates to a method of activating STING, or a mutant thereof, in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound.
  • the invention relates to a method of reversibly or irreversibly inhibiting one or more of STING, or a mutant thereof, activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound.
  • the present invention provides a method for treating a disorder mediated by STING, or a mutant thereof, in a patient in need thereof, comprising the step of administering to said patient a compound according to the present invention or pharmaceutically acceptable composition thereof.
  • a disorder mediated by STING or a mutant thereof
  • Such disorders are described in detail herein.
  • additional therapeutic agents that are normally administered to treat that condition may also be present in the compositions of this invention.
  • additional therapeutic agents that are normally administered to treat a particular disease, or condition are known as“appropriate for the disease, or condition, being treated.”
  • a compound of the current invention may also be used to advantage in combination with other therapeutic compounds.
  • the other therapeutic compounds are antiproliferative compounds.
  • antiproliferative compounds include, but are not limited to aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule active compounds; alkylating compounds; histone deacetylase inhibitors; compounds which induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibitors; mTOR inhibitors; anti neoplastic antimetabolites; platin compounds; compounds targeting/decreasing a protein or lipid kinase activity and further anti-angiogenic compounds; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; anti androgens; methionine aminopeptidase inhibitors; matrix metalloproteinase inhibitors; bisphosphonates; biological response modifiers; antiprolifer
  • NSC707545 IPI-504, CNFIOIO, CNF2024, CNFIOIO from Conforma Therapeutics; temozolomide (Temodal ® ); kinesin spindle protein inhibitors, such as SB715992 or SB743921 from GlaxoSmithKline, or pentamidine/chlorpromazine from CombinatoRx; MEK inhibitors such as ARRY142886 from Array BioPharma, AZD6244 from AstraZeneca, PD181461 from Pfizer and leucovorin.
  • temozolomide Temodal ®
  • kinesin spindle protein inhibitors such as SB715992 or SB743921 from GlaxoSmithKline, or pentamidine/chlorpromazine from CombinatoRx
  • MEK inhibitors such as ARRY142886 from Array BioPharma, AZD6244 from AstraZeneca, PD181461 from Pfizer and leucovorin.
  • aromatase inhibitor as used herein relates to a compound which inhibits estrogen production, for instance, the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively.
  • the term includes, but is not limited to steroids, especially atamestane, exemestane and formestane and, in particular, non-steroids, especially aminoglutethimide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketokonazole, vorozole, fadrozole, anastrozole and letrozole.
  • Exemestane is marketed under the trade name AromasinTM.
  • Formestane is marketed under the trade name LentaronTM.
  • Fadrozole is marketed under the trade name AfemaTM.
  • Anastrozole is marketed under the trade name ArimidexTM.
  • Letrozole is marketed under the trade names FemaraTM or FemarTM.
  • Aminoglutethimide is marketed under the trade name OrimetenTM
  • a combination of the invention comprising a chemotherapeutic agent which is an aromatase inhibitor is particularly useful for the treatment of hormone receptor positive tumors, such as breast tumors.
  • antiestrogen as used herein relates to a compound which antagonizes the effect of estrogens at the estrogen receptor level.
  • the term includes, but is not limited to tamoxifen, fulvestrant, raloxifene and raloxifene hydrochloride.
  • Tamoxifen is marketed under the trade name NolvadexTM.
  • Raloxifene hydrochloride is marketed under the trade name EvistaTM.
  • Fulvestrant can be administered under the trade name FaslodexTM.
  • a combination of the invention comprising a chemotherapeutic agent which is an antiestrogen is particularly useful for the treatment of estrogen receptor positive tumors, such as breast tumors.
  • anti -androgen as used herein relates to any substance which is capable of inhibiting the biological effects of androgenic hormones and includes, but is not limited to, bicalutamide (CasodexTM).
  • gonadorelin agonist as used herein includes, but is not limited to abarelix, goserelin and goserelin acetate. Goserelin can be administered under the trade name ZoladexTM.
  • topoisom erase I inhibitor includes, but is not limited to topotecan, gimatecan, irinotecan, camptothecian and its analogues, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148.
  • Irinotecan can be administered, e.g. in the form as it is marketed, e.g. under the trademark CamptosarTM.
  • Topotecan is marketed under the trade name HycamptinTM.
  • topoisomerase II inhibitor includes, but is not limited to the anthracyclines such as doxorubicin (including liposomal formulation, such as CaelyxTM), daunorubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones mitoxantrone and losoxantrone, and the podophillotoxines etoposide and teniposide.
  • anthracyclines such as doxorubicin (including liposomal formulation, such as CaelyxTM), daunorubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones mitoxantrone and losoxantrone, and the podophillotoxines etoposide and teniposide.
  • Etoposide is marketed under the trade name EtopophosTM
  • Teniposide is marketed under the trade name VM 26-Bristol
  • Doxorubicin is marketed under the trade name AcriblastinTM or AdriamycinTM
  • Epirubicin is marketed under the trade name FarmorubicinTM.
  • Idarubicin is marketed under the trade name ZavedosTM.
  • Mitoxantrone is marketed under the trade name Novantron.
  • microtubule active agent relates to microtubule stabilizing, microtubule destabilizing compounds and microtublin polymerization inhibitors including, but not limited to taxanes, such as paclitaxel and docetaxel; vinca alkaloids, such as vinblastine or vinblastine sulfate, vincristine or vincristine sulfate, and vinorelbine; discodermolides; cochicine and epothilones and derivatives thereof.
  • Paclitaxel is marketed under the trade name TaxolTM.
  • Docetaxel is marketed under the trade name TaxotereTM.
  • Vinblastine sulfate is marketed under the trade name Vinblastin R.PTM.
  • Vincristine sulfate is marketed under the trade name FarmistinTM.
  • alkylating agent includes, but is not limited to, cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel). Cyclophosphamide is marketed under the trade name CyclostinTM. Ifosfamide is marketed under the trade name HoloxanTM.
  • histone deacetylase inhibitors or “HD AC inhibitors” relates to compounds which inhibit the histone deacetylase and which possess antiproliferative activity. This includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).
  • SAHA suberoylanilide hydroxamic acid
  • antimetabolite includes, but is not limited to, 5-fluorouracil or 5-FU, capecitabine, gemcitabine, DNA demethylating compounds, such as 5-azacytidine and decitabine, methotrexate and edatrexate, and folic acid antagonists such as pemetrexed.
  • Capecitabine is marketed under the trade name XelodaTM.
  • Gemcitabine is marketed under the trade name GemzarTM.
  • platinum compound as used herein includes, but is not limited to, carboplatin, cis-platin, cisplatinum and oxaliplatin.
  • Carboplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark CarboplatTM.
  • Oxaliplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark EloxatinTM.
  • the term "compounds targeting/decreasing a protein or lipid kinase activity; or a protein or lipid phosphatase activity; or further anti-angiogenic compounds” as used herein includes, but is not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, such as a) compounds targeting, decreasing or inhibiting the activity of the platelet-derived growth factor-receptors (PDGFR), such as compounds which target, decrease or inhibit the activity of PDGFR, especially compounds which inhibit the PDGF receptor, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib, SU101, SU6668 and GFB- 111; b) compounds targeting, decreasing or inhibiting the activity of the fibroblast growth factor- receptors (FGFR); c) compounds targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor I (IGF
  • BCR-Abl kinase and mutants, such as compounds which target decrease or inhibit the activity of c-Abl family members and their gene fusion products, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib or nilotinib (AMN107); PD180970; AG957; NSC 680410; PD173955 from ParkeDavis; or dasatinib (BMS-354825); j) compounds targeting, decreasing or inhibiting the activity of members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK, SRC, JAK/pan-JAK, FAK, PDK1, PKB/Akt, Ras/MAPK, PI3K, SYK, BTK and TEC family, and/or members of the cyclin-
  • PKC protein kinase C
  • Raf family of serine/threonine kinases members of the ME
  • c-Met receptor compounds which target, decrease or inhibit the activity of c-Met, especially compounds which inhibit the kinase activity of c-Met receptor, or antibodies that target the extracellular domain of c-Met or bind to HGF
  • PI3K inhibitor includes, but is not limited to compounds having inhibitory activity against one or more enzymes in the phosphatidylinositol-3 -kinase family, including, but not limited to RBKa, RI3Kg, PI3K5, RI3Kb, PI3K-C2a, PI3K-C2p, PI3K- C2y, Vps34, pi 10-a, pi 10-b, p 110-g, pi 10-d, p85-a, r85-b, r55-g, pi 50, plOl, and p87.
  • PI3K inhibitors useful in this invention include but are not limited to ATU-027, SF-1126, DS- 7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib, pictrelisib, PF-4691502, BYL-719, dactolisib, XL-147, XL-765, and idelalisib.
  • BTK inhibitor includes, but is not limited to compounds having inhibitory activity against Bruton’s Tyrosine Kinase (BTK), including, but not limited to AVL-292 and ibrutinib.
  • SYK inhibitor includes, but is not limited to compounds having inhibitory activity against spleen tyrosine kinase (SYK), including but not limited to PRT- 062070, R-343, R-333, Excellair, PRT-062607, and fostamatinib.
  • Bcl-2 inhibitor includes, but is not limited to compounds having inhibitory activity against B-cell lymphoma 2 protein (Bcl-2), including but not limited to ABT-199, ABT-731, ABT-737, apogossypol, Ascenta’s pan-Bcl-2 inhibitors, curcumin (and analogs thereof), dual Bcl-2/Bcl-xL inhibitors (Infinity Pharmaceuticals/Novartis Pharmaceuticals), Genasense (G3139), HA14-1 (and analogs thereof; see W02008118802), navitoclax (and analogs thereof, see US7390799), NH-1 (Shenayng Pharmaceutical University), obatoclax (and analogs thereof, see W02004106328), S-001 (Gloria Pharmaceuticals), TW series compounds (Univ. of Michigan), and venetoclax.
  • the Bcl-2 inhibitor is a small molecule therapeutic.
  • the Bcl-2 inhibitor is a small molecule therapeutic.
  • SYK inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this invention can be found in W02003063794, W02005007623, and W02006078846, the entirety of which are incorporated herein by reference.
  • PI3K inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this invention can be found in W02004019973, W02004089925, W02007016176, US8138347, W02002088112, W02007084786,
  • W02007129161, W02006122806, W02005113554, and W02007044729 the entirety of which are incorporated herein by reference.
  • JAK inhibitory compounds and conditions treatable by such compounds in combination with compounds of this invention can be found in W02009114512, W02008109943, W02007053452, W02000142246, and W02007070514, the entirety of which are incorporated herein by reference.
  • Further anti-angiogenic compounds include compounds having another mechanism for their activity, e.g. unrelated to protein or lipid kinase inhibition e.g. thalidomide (ThalomidTM) and TNP-470.
  • ThilomidTM thalidomide
  • TNP-470 TNP-470.
  • proteasome inhibitors useful for use in combination with compounds of the invention include, but are not limited to bortezomib, disulfiram, epigallocatechin-3-gallate (EGCG), salinosporamide A, carfilzomib, ONX-0912, CEP-18770, and MLN9708.
  • Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase are e.g. inhibitors of phosphatase 1, phosphatase 2A, or CDC25, such as okadaic acid or a derivative thereof.
  • Compounds which induce cell differentiation processes include, but are not limited to, retinoic acid, a- g- or d- tocopherol or a- g- or d-tocotrienol.
  • cyclooxygenase inhibitor as used herein includes, but is not limited to, Cox- 2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib (CelebrexTM), rofecoxib (VioxxTM), etoricoxib, valdecoxib or a 5-alkyl-2- arylaminophenylacetic acid, such as 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenyl acetic acid, lumiracoxib.
  • Cox- 2 inhibitors such as celecoxib (CelebrexTM), rofecoxib (VioxxTM), etoricoxib, valdecoxib or a 5-alkyl-2- arylaminophenylacetic acid, such as 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenyl acetic acid, lumiracoxib.
  • bisphosphonates includes, but is not limited to, etridonic, clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and zoledronic acid.
  • Etridonic acid is marketed under the trade name DidronelTM.
  • Clodronic acid is marketed under the trade name BonefosTM.
  • Tiludronic acid is marketed under the trade name SkelidTM.
  • Pamidronic acid is marketed under the trade name ArediaTM.
  • Alendronic acid is marketed under the trade name FosamaxTM.
  • Ibandronic acid is marketed under the trade name BondranatTM.
  • Risedronic acid is marketed under the trade name ActonelTM.
  • Zoledronic acid is marketed under the trade name ZometaTM.
  • mTOR inhibitors relates to compounds which inhibit the mammalian target of rapamycin (mTOR) and which possess antiproliferative activity such as sirolimus (Rapamune®), everolimus (CerticanTM), CCI-779 and ABT578.
  • heparanase inhibitor refers to compounds which target, decrease or inhibit heparin sulfate degradation.
  • the term includes, but is not limited to, PI-88.
  • biological response modifier refers to a lymphokine or interferons.
  • inhibitor of Ras oncogenic isoforms such as H-Ras, K-Ras, or N-Ras
  • inhibitor of Ras oncogenic isoforms refers to compounds which target, decrease or inhibit the oncogenic activity of Ras; for example, a “famesyl transferase inhibitor” such as L-744832, DK8G557 or R115777 (ZamestraTM).
  • telomerase inhibitor refers to compounds which target, decrease or inhibit the activity of telomerase. Compounds which target, decrease or inhibit the activity of telomerase are especially compounds which inhibit the telomerase receptor, such as telomestatin.
  • methionine aminopeptidase inhibitor refers to compounds which target, decrease or inhibit the activity of methionine aminopeptidase.
  • Compounds which target, decrease or inhibit the activity of methionine aminopeptidase include, but are not limited to, bengamide or a derivative thereof.
  • proteasome inhibitor refers to compounds which target, decrease or inhibit the activity of the proteasome.
  • Compounds which target, decrease or inhibit the activity of the proteasome include, but are not limited to, Bortezomib (VelcadeTM) and MLN 341.
  • matrix metalloproteinase inhibitor or (“MMP” inhibitor) as used herein includes, but is not limited to, collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, e.g. hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat (BB-2516), prinomastat (AG3340), metastat (NSC 683551) BMS-279251, BAY 12-9566, TAA211, MMI270B or AAJ996.
  • MMP matrix metalloproteinase inhibitor
  • FMS-like tyrosine kinase inhibitors which are compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, I-b-D-arabinofuransyl cytosine (ara-c) and bisulfan; ALK inhibitors, which are compounds which target, decrease or inhibit anaplastic lymphoma kinase, and Bcl-2 inhibitors.
  • FMS-like tyrosine kinase receptors are especially compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, such as PKC412, midostaurin, a staurosporine derivative, SU11248 and MLN518.
  • HSP90 inhibitors includes, but is not limited to, compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90; degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteosome pathway.
  • Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90 are especially compounds, proteins or antibodies which inhibit the ATPase activity of HSP90, such as 17-allylamino,17-demethoxygeldanamycin (17AAG), a geldanamycin derivative; other geldanamycin related compounds; radicicol and HDAC inhibitors.
  • antiproliferative antibodies includes, but is not limited to, trastuzumab (HerceptinTM), Trastuzumab-DMl, erbitux, bevacizumab (AvastinTM), rituximab (Rituxan ® ), PR064553 (anti-CD40) and 2C4 Antibody.
  • trastuzumab HerceptinTM
  • Trastuzumab-DMl erbitux
  • bevacizumab AvastinTM
  • rituximab Rasteran ®
  • PR064553 anti-CD40
  • compounds of the current invention can be used in combination with standard leukemia therapies, especially in combination with therapies used for the treatment of AML.
  • compounds of the current invention can be administered in combination with, for example, farnesyl transferase inhibitors and/or other drugs useful for the treatment of AML, such as Daunorubicin, Adriamycin, Ara-C, VP- 16, Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412.
  • the present invention provides a method of treating AML associated with an ITD and/or D835Y mutation, comprising administering a compound of the present invention together with a one or more FLT3 inhibitors.
  • the FLT3 inhibitors are selected from quizartinib (AC220), a staurosporine derivative (e.g.
  • the FLT3 inhibitors are selected from quizartinib, midostaurin, lestaurtinib, sorafenib, and sunitinib.
  • anti-leukemic compounds include, for example, Ara-C, a pyrimidine analog, which is the 2 -alpha-hydroxy ribose (arabinoside) derivative of deoxycytidine. Also included is the purine analog of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine phosphate.
  • Compounds which target, decrease or inhibit activity of histone deacetylase (HD AC) inhibitors such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA) inhibit the activity of the enzymes known as histone deacetylases.
  • HD AC histone deacetylase
  • SAHA suberoylanilide hydroxamic acid
  • HDAC inhibitors include MS275, SAHA, FK228 (formerly FR901228), Trichostatin A and compounds disclosed in US 6,552,065 including, but not limited to, N-hydroxy-3-[4-[[[2-(2-methyl-lH-indol-3-yl)-ethyl]- amino]methyl]phenyl]- 2E-2-propenamide, or a pharmaceutically acceptable salt thereof and N-hydroxy-3-[4-[(2- hydroxyethyl) ⁇ 2-(lH-indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2- propenamide, or a pharmaceutically acceptable salt thereof, especially the lactate salt.
  • Somatostatin receptor antagonists as used herein refer to compounds which target, treat or inhibit the somatostatin receptor such as octreotide, and SOM230.
  • Tumor cell damaging approaches refer to approaches such as ionizing radiation.
  • the term "ionizing radiation” referred to above and hereinafter means ionizing radiation that occurs as either electromagnetic rays (such as X-rays and gamma rays) or particles (such as alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiation therapy and is known in the art. See Heilman, Principles of Radiation Therapy, Cancer, in Principles and Practice of Oncology, Devita et ah, Eds., 4 th Edition, Vol. 1, pp. 248-275 (1993).
  • EDG binders and ribonucleotide reductase inhibitors.
  • EDG binders refers to a class of immunosuppressants that modulates lymphocyte recirculation, such as FTY720.
  • ribonucleotide reductase inhibitors refers to pyrimidine or purine nucleoside analogs including, but not limited to, fludarabine and/or cytosine arabinoside (ara-C), 6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine (especially in combination with ara-C against ALL) and/or pentostatin.
  • Ribonucleotide reductase inhibitors are especially hydroxyurea or 2-hydroxy-lH-isoindole-l ,3-dione derivatives.
  • VEGF vascular endothelial growth factor
  • l-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof l-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine succinate
  • AngiostatinTM EndostatinTM
  • anthranilic acid amides ZD4190; ZD6474; SU5416; SU6668
  • bevacizumab or anti-VEGF antibodies or anti-VEGF receptor antibodies, such as rhuMAb and RHEIFab
  • VEGF aptamer such as Macugon
  • FLT-4 inhibitors, FLT-3 inhibitors VEGFR-2 IgGI antibody
  • Angiozyme RI 4610)
  • Bevacizumab AvastinTM
  • Photodynamic therapy refers to therapy which uses certain chemicals known as photosensitizing compounds to treat or prevent cancers.
  • Examples of photodynamic therapy include treatment with compounds, such as VisudyneTM and porfimer sodium.
  • Angiostatic steroids refers to compounds which block or inhibit angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11-a-epihydrocotisol, cortexolone, 17a-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.
  • angiogenesis such as, e.g., anecortave, triamcinolone, hydrocortisone, 11-a-epihydrocotisol, cortexolone, 17a-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.
  • Implants containing corticosteroids refers to compounds, such as fluocinolone and dexamethasone.
  • chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or miscellaneous compounds or compounds with other or unknown mechanism of action.
  • the compounds of the invention are also useful as co-therapeutic compounds for use in combination with other drug substances such as anti-inflammatory, bronchodilatory or antihistamine drug substances, particularly in the treatment of obstructive or inflammatory airways diseases such as those mentioned hereinbefore, for example as potentiators of therapeutic activity of such drugs or as a means of reducing required dosaging or potential side effects of such drugs.
  • a compound of the invention may be mixed with the other drug substance in a fixed pharmaceutical composition or it may be administered separately, before, simultaneously with or after the other drug substance.
  • the invention includes a combination of a compound of the invention as hereinbefore described with an anti-inflammatory, bronchodilatory, antihistamine or anti-tussive drug substance, said compound of the invention and said drug substance being in the same or different pharmaceutical composition.
  • Suitable anti-inflammatory drugs include steroids, in particular glucocorticosteroids such as budesonide, beclamethasone dipropionate, fluticasone propionate, ciclesonide or mometasone furoate; non-steroidal glucocorticoid receptor agonists; LTB4 antagonists such LY293111, CGS025019C, CP-195543, SC-53228, BIIL 284, ONO 4057, SB 209247; LTD4 antagonists such as montelukast and zafirlukast; PDE4 inhibitors such cilomilast (Ariflo® GlaxoSmithKline), Roflumilast (Byk Gulden), V-11294A (Napp), BAY19-8004 (Bayer), SCH- 351591 (Schering- Plough), Arofylline (Almirall Prodesfarma), PD189659 / PD168787 (Parke- Davis
  • Suitable bronchodilatory drugs include anticholinergic or antimuscarinic compounds, in particular ipratropium bromide, oxitropium bromide, tiotropium salts and CHF 4226 (Chiesi), and glycopyrrolate.
  • Suitable antihistamine drug substances include cetirizine hydrochloride, acetaminophen, clemastine fumarate, promethazine, loratidine, desloratidine, diphenhydramine and fexofenadine hydrochloride, activastine, astemizole, azelastine, ebastine, epinastine, mizolastine and tefenadine.
  • chemokine receptors e.g. CCR-1, CCR-2, CCR-3, CCR-4, CCR-5, CCR-6, CCR-7, CCR-8, CCR-9 and CCR10, CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, particularly CCR-5 antagonists such as Schering-Plough antagonists SC-351125, SCH- 55700 and SCH-D, and Takeda antagonists such as N-[[4-[[[[6,7-dihydro-2-(4-methylphenyl)-5H- benzo-cyclohepten-8-yl]carbonyl]amino]phenyl]-methyl]tetrahydro-N,N-dimethyl-2H-pyran-4- aminium chloride (TAK-770).
  • TAK-770 antagonists such as N-[[4-[[[6,7-dihydro-2-(4-methylphenyl)-5H- benzo-cyclohepten-8-yl]carbony
  • a compound of the current invention may also be used in combination with known therapeutic processes, for example, the administration of hormones or radiation.
  • a provided compound is used as a radiosensitizer, especially for the treatment of tumors which exhibit poor sensitivity to radiotherapy.
  • a compound of the current invention can be administered alone or in combination with one or more other therapeutic compounds, possible combination therapy taking the form of fixed combinations or the administration of a compound of the invention and one or more other therapeutic compounds being staggered or given independently of one another, or the combined administration of fixed combinations and one or more other therapeutic compounds.
  • a compound of the current invention can besides or in addition be administered especially for tumor therapy in combination with chemotherapy, radiotherapy, immunotherapy, phototherapy, surgical intervention, or a combination of these. Long-term therapy is equally possible as is adjuvant therapy in the context of other treatment strategies, as described above. Other possible treatments are therapy to maintain the patient's status after tumor regression, or even chemopreventive therapy, for example in patients at risk.
  • Those additional agents may be administered separately from an inventive compound- containing composition, as part of a multiple dosage regimen.
  • those agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as part of a multiple dosage regime, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another normally within five hours from one another.
  • the term“combination,”“combined,” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention.
  • a compound of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form.
  • the present invention provides a single unit dosage form comprising a compound of the current invention, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • compositions of this invention should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of an inventive compound can be administered.
  • compositions which comprise an additional therapeutic agent that additional therapeutic agent and the compound of this invention may act synergistically. Therefore, the amount of additional therapeutic agent in such compositions will be less than that required in a monotherapy utilizing only that therapeutic agent. In such compositions a dosage of between 0.01 - 1,000 pg/kg body weight/day of the additional therapeutic agent can be administered.
  • the amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent.
  • the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.
  • the compounds of this invention, or pharmaceutical compositions thereof, may also be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters.
  • an implantable medical device such as prostheses, artificial valves, vascular grafts, stents and catheters.
  • Vascular stents for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury).
  • patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a kinase inhibitor.
  • Implantable devices coated with a compound of this invention are another embodiment of the present invention.
  • the crude product was purified by Prep-HPLC with the following conditions (24SHIMADZU (HPLC-01)): Column, SunFire C18 OBD Prep Column, 19 mm X 250 mm; mobile phase, Water (l%HOAc) and ACN (13% Phase B up to 50% in 10 min); Detector, UV/254nm to yield 1.8 mg (1.25%) of 1-108 as a yellow solid.
  • EXAMPLE 4 Synthesis of E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(2-inethyl-2H- indazole-3-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-7-methyl-lH- benzo[d]imidazol-2-yl)-2-methyl-2H-indazole-3-carboxamide, 1-102
  • the filtrate cake was purified by reverse flash chromatography under the following conditions: column, C 18 silica gel; mobile phase, MeOH in water, 10% to 80% gradient in 10 min; detector, UV 254 nm.
  • the crude product (60mg) was purified by Prep-HPLC (column: XBridge Shield RP18 OBD Column, 5um, 19* 150mm; mobile phase A: water (10MMOL/L NH4HCO3), mobile phase B: ACN; flow rate: 25 mL/min; gradient: 25% B to 35% B in 9 min; 254;220 nm; rt: 8.65 min) to afford 1-60 (5.2mg, 0.36% ) as a white solid.
  • EXAMPLE 8 Synthesis of A-(l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-l//-pyrazole-5- carboxamido)-l T-benzo[d]imidazol-l-yl)butyl)-7-(morpholinomethyl)-l/ - benzo[ ⁇ /
  • the crude product was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): Column, XBridge Prep OBD C18 Column, 19*250mm, 5um; mobile phase, water(10MMOL/L NH4HCOV) and ACN (hold 30% PhaseB in 8 min); Detector, UV 254nm to yield 10 mg (17%) of 1-58 as a white solid.
  • 2#SHIMADZU (HPLC-01) Column, XBridge Prep OBD C18 Column, 19*250mm, 5um; mobile phase, water(10MMOL/L NH4HCOV) and ACN (hold 30% PhaseB in 8 min); Detector, UV 254nm to yield 10 mg (17%) of 1-58 as a white solid.
  • EXAMPLE 10 Synthesis of .V-(l-(4-(5-carbamoyl-2-(l-ethyl-3-niethyl-l//-pyrazole-5- carboxamido)-l//-benzo[d]imidazol-l-yl)butyl)-7-methyl-l//-benzo[d]imidazol-2-yl)-4- ethyl-2-methyloxazole-5-carboxamide, 1-54
  • the solution was extracted with 3x15 mL of ethyl acetate and washed with 15 ml of water, and dried over anhydrous sodium sulfate.
  • the mixture was concentrated and purified by reverse flash chromatography under the following conditions: column, C18 silica gel; mobile phase, ACN in water, 10% to 60% gradient in 15 min; detector, UV 254 nm to afford 1-54 (85.5 mg, 67.5%) as a light yellow solid.
  • the mixture was concentrated and purified by reverse flash chromatography under the following conditions: column, C18 silica gel; mobile phase, ACN in water, 10% to 60% gradient in 15 min; detector, UV 254 nm to afford 1-53 (86.8 mg, 46.53%) as a light yellow solid.
  • the crude product was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column 30x 150mm 5um; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 19% B to 29% B in 7 min; 254; 220 nm; Rt: 5.55 min) to yield 4.0 mg (4 %) of 1-38 as a white solid.
  • EXAMPLE 17 Synthesis of .V-(l-(4-(5-carbainoyl-2-(l-ethyl-3-niethyl-l//-pyrazole-5- carboxamido)-l/T-benzo[i/]imidazol-l-yl)butyl)-7-methyl-l//-benzo[i/]imidazol-2-yl)-2,4- dimethyloxazole-5-carboxamide, 1-43
  • the solids were collected by filtration.
  • the crude product was purified by Prep-HPLC with the following conditions (SHIMADZU (HPLC-01)): column, X bridge Prep OBD C18 column, 19 !l! 250mm, 5um; mobile phase, water (l%HOAc) and MeOH (57% phase B up to 67% in 10 min); Detector, UV 254nm to yield 13.7 mg (7.4%) of 1-43 as a white solid.
  • the crude product was purified by Prep-HPLC with the following conditions: Column: XSelect CSHPrep C18 OBD Column, 5um, 19* 150mm; Mobile Phase A: water (10% NH4HCO3), Mobile Phase B:ACN; Flow rate:25 mL/min; Gradient: 10 B to 40 B in 7 min; UV detection at 210/254 nm to yield 1-142 (30 mg, 34.1%) as a white solid.
  • 5UB5TITUTE SHEET (RULE 26) lambda5-phosphanylidene)acetate (4.09 g, 12.23 mmol, 1.2 equiv) was added at RT and the resulting mixture was stirred for 2 hrs. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography with PE/ ethyl acetate (5: 1) to give 19.11 (1.8 g, 70.6%) as a light yellow oil. (ES, m/z): 251 (M+H + ).
  • the crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep Cl 8 column, 30* 150mm, 5um; mobile phase, water (0.1% FA) and acetonitrile (12% acetonitrile up to 37% in 7 min); UV detection at 254/210 nm.
  • the product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 1-143 (72.5 mg, 61.5%) as an off-white solid. (ESI): 390 (M/2+H + ).
  • the crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep Cl 8 column, 30* 150, 5um; mobile phase, water (0.1% FA) and acetonitrile (12% acetonitrile up to 40% in 7 min); UV detection at 254/210 nm.
  • the product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 1-145 (50.4 mg; 51.3%) as an off-white solid. (ES, m/z) 397 (M/2+H + ).
  • EXAMPLE 22 Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl- lH-pyrazole-5-carboxamido)-7-(3-hydroxypropoxy)-lH-benzo[d]imidazol-l-yl)but-2-enyl)- 7-(3-morpholinopropoxy)-lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5- carboxamide, 1-146 .
  • the crude product was purified by Prep- HPLC with the following conditions: Column, XSelect CSH Prep OBD Cl 8 Column 19*250mm, 5um; mobile phase, water (0.1% FA) and ACN (12% ACN up to 33% in 7 min); UV detection at 254/220 nm.
  • the product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 1-146 (21.2 mg, 11.6%) as a white solid.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides compounds, compositions thereof, and methods of using the same for the modulation of STING, and the treatment of STING-mediated disorders.

Description

STING AGONISTS AND USES THEREOF
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit under 35 U.S.C. § 119(e) of U.S. Provisional Application Nos. US 62/784, 136, filed December 21, 2018; US 62/823,275, filed March 25, 2019; and US 62/889,941, filed August 21, 2019; the entire contents of each of which is incorporated herein by reference.
TECHNICAL FIELD OF THE INVENTION
[0002] The present invention relates to compounds and methods useful for the modulation of STING-dependent type I interferon production. The invention also provides pharmaceutically acceptable compositions comprising compounds of the present invention and methods of using said compositions in the treatment of various disorders.
BACKGROUND OF THE INVENTION
[0003] Innate immunity is a rapid nonspecific immune response that fights against environmental insults including, but not limited to, pathogens such as bacteria or viruses. Adaptive immunity is a slower but more specific immune response, which confers long-lasting or protective immunity to the host and involves differentiation and activation of naive T lymphocytes into CD4+ T helper cells and/or CD8+ cytotoxic T cells, to promote cellular and humoral immunity. Antigen presentation cells of the innate immune system, such as dendritic cells or macrophages, serve as a critical link between the innate and adaptive immune systems by phagocytosing and processing the foreign antigens and presenting them on the cell surface to the T cells, thereby activating T cell response.
[0004] STING (stimulator of interferon genes) is an endoplasmic reticulum adaptor that facilitates innate immune signaling (Ishikawa and Barber, Nature 2008, 455(7213):674-678).
It was reported that STING comprises four putative transmembrane regions (Ouyang et al., Immunity (2012) 36, 1073), predominantly resides in the endoplasmic reticulum and is able to activate NF-kB, STAT6, and IRF3 transcription pathways to induce expression of type I interferon (e.g., IFN-a and IFN-13) and exert a potent anti-viral state following expression (Ishikawa and Barber, Nature 2008, 455(7213):674-678; Chen et al., Cell (2011) 147, 436- 446). In contrast, loss of STING rendered murine embryonic fibroblasts extremely susceptible to negative stranded virus infection, including vesicular stomatitis virus. (Ishikawa and Barber, Nature 2008, 455(7213):674-678).
[0005] There remains a need for new therapeutics that are effective as STING agonists for the treatment of diseases, in particular cancer.
SUMMARY OF THE INVENTION
[0006] It has now been found that compounds of this invention, and pharmaceutically acceptable compositions thereof, are effective as STING agonists. Such compounds have the general formula I:
Figure imgf000003_0001
I
or a pharmaceutically acceptable salt thereof, wherein each variable is as defined and described herein.
[0007] Compounds of the present invention, and pharmaceutically acceptable compositions thereof, are useful for treating a variety of diseases, disorders or conditions, associated with STING. Such diseases, disorders, or conditions include those described herein. [0008] Compounds provided by this invention are also useful for the study of STING and associated proteins in biological and pathological phenomena; the study of STING occurring in bodily tissues; and the comparative evaluation of new STING agonists or other regulators of STING in vitro or in vivo.
DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS
1. General Description of Certain Embodiments of the Invention:
[0009] Compounds of the present invention, and compositions thereof, are useful as STING ligands.
[0010] As defined herein, the terms“agonist,”“binder,”“modulator,” and“ligand” are used interchangeably and describe a compound that binds to, modulates, activates, is a ligand for, or agonist of STING.
[0011] In certain embodiments, the present invention provides a compound of formula I:
Figure imgf000004_0001
I
or a pharmaceutically acceptable salt thereof, wherein:
m is 0 or 1;
n is 0 or 1;
p is 0 or 1; wherein m + n + p = 1, 2, or 3;
Ring A is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
wherein Ring A and its substituents is other than
Figure imgf000005_0001
Ring B is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
L1 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy1-, -C(R’)2-, -CH(R’)-, -N(R)-, Cy'fL’-Z) , -C(R’)(L’-Z)- -CH(L’-Z)-,
N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2- -S(0)2N(R)-, -0-, -C(O)-, -OC(O)- -C(0)0-, -S-, -S(O)-, or -S(0)2-;
each -Cy1- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
4
5UB5TITUTE SHEET (RULE 26) each R’ is independently hydrogen or C M aliphatic; or
two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring; or
two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R is independently hydrogen, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or:
two R groups on the same nitrogen are optionally taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each L’ is a covalent bond or a CMS bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and
independently replaced
Figure imgf000006_0001
, -N(R)- -N(R)C(0)-, -C(0)N(R)-, -N(R)C(0)0- -0C(0)N(R)-, -N(R)S(0) - - S(0)2N(R)-, -0-, -C(O)-, -0C(0)-, -C(0)0- -S-, -S(0)-, or -S(0)2-;
each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, napththylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each Z is hydrogen or a CRBN binding moiety;
L2 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy2-, -C(R’)2-, -CH(R’)-, -N(R)-, -Cy2(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)- -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2- -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0)2-;
each -Cy2- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
L3 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy3-, -C(R’)2-, -CH(R’)-, -N(R)-, -Cy3(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)- -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(0)-, -0C(0)- -C(0)0-, -S-, -S(0)-, or -S(0)2-;
each -Cy3- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R1 is independently hydrogen, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, - N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
R2 is hydrogen or an optionally substituted Ci-6 aliphatic group;
each R3 is independently hydrogen, oxo, halogen, -CN, -NO2, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, - N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each R4 is independently hydrogen, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
R5 is hydrogen or an optionally substituted Ci-6 aliphatic group;
each R6 is independently hydrogen, oxo, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each of q, s, t, and v is independently 0, 1, 2, 3, or 4;
each of r and u is independently 0 or 1; and
each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
[0012] In certain embodiments, the present invention also provides a compound of formula
II:
Figure imgf000009_0001
or a pharmaceutically acceptable salt thereof, wherein:
m is 0 or 1;
n is 0 or 1;
p is 0 or 1;
wherein m + n + p = 1, 2, or 3;
L1 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced
Figure imgf000009_0002
-N(L’-Z)-, -N(R)C(0)- -C(0)N(R)-, -N(R)S(0)2- -S(0)2N(R)-, -0-, -C(O)-, -OC(O)- -C(0)0- -S-, -S(O)-, or -S(0)2-; each -Cy1- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R’ is independently hydrogen or C M aliphatic; or
two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring; or
two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R is independently hydrogen, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or:
two R groups on the same nitrogen are taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each L’ is a covalent bond or a CM5 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and
independently replaced
Figure imgf000010_0001
, -N(R)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)C(0)0-, -0C(0)N(R)-, -N(R)S(0)2-, - S(0)2N(R)-, -0-, -C(0)-, -0C(0)-, -C(0)0- -S-, -S(O)-, or -S(0)2- each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, napththylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each Z is hydrogen or a CRBN binding moiety;
L2 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy2-, -C(R’)2- -CH(R’)-, -N(R)-, -Cy2(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)- -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(O)-, -0C(0)- -C(0)0-, -S-, -S(O)-, or -S(0)2-;
each -Cy2- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
L3 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy3-, -C(R’)2-, -CH(R’)-, -N(R)-, -Cy3(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)- -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2- -S(0)2N(R)-, -0-, -C(0)-, -0C(0)- -C(0)0-, -S-, -S(0)-, or -S(0)2-;
each -Cy3- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R1 is independently hydrogen, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
R2 is hydrogen or an optionally substituted Ci-6 aliphatic group;
each R3 is independently hydrogen, oxo, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each R4 is independently hydrogen, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
R5 is hydrogen or an optionally substituted Ci-6 aliphatic group;
each R6 is independently hydrogen, oxo, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, - N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each of q, s, t, and v is independently 0, 1, 2, 3, or 4;
each of r and u is independently 0 or 1;
each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; and wherein
(a) when R1 or R4 is -C(0)NR¾ each R of the -C(0)NR2 moiety is other than hydrogen or CM aliphatic; or
(b) in at least one instance of L1, L2, or L3, a methylene unit is replaced with -C(O)-; or
(c) in at least one instance of L1, L2, or L3, a methylene unit adjacent to the benzimidazole or pyrazole ring is replaced with -Cy1-, -Cy2-, or -Cy3-.
2. Compounds and Definitions:
[0013] Compounds of the present invention include those described generally herein, and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed. Additionally, general principles of organic chemistry are described in“Organic Chemistry”, Thomas Sorrell, University Science Books, Sausalito: 1999, and“March’s Advanced Organic Chemistry”, 5th Ed., Ed.: Smith, M.B. and March, J., John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference.
[0014] The term“aliphatic” or“aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as "carbocycle,"“cycloaliphatic” or“cycloalkyl”), that has a single point of attachment to the rest of the molecule. Unless otherwise specified, aliphatic groups contain 1-6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms. In some embodiments,“cycloaliphatic” (or“carbocycle” or“cycloalkyl”) refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
[0015] As used herein, the term“bridged bicyclic” refers to any bicyclic ring system, i.e. carbocyclic or heterocyclic, saturated or partially unsaturated, having at least one bridge. As defined by IUPAC, a“bridge” is an unbranched chain of atoms or an atom or a valence bond connecting two bridgeheads, where a“bridgehead” is any skeletal atom of the ring system which is bonded to three or more skeletal atoms (excluding hydrogen). In some embodiments, a bridged bicyclic group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Such bridged bicyclic groups are well known in the art and include those groups set forth below where each group is attached to the rest of the molecule at any substitutable carbon or nitrogen atom. Unless otherwise specified, a bridged bicyclic group is optionally substituted with one or more substituents as set forth for aliphatic groups. Additionally or alternatively, any substitutable nitrogen of a bridged bicyclic group is optionally substituted. Exemplary bridged bicyclics include:
Figure imgf000014_0001
Figure imgf000015_0001
[0016] The term“lower alkyl” refers to a C straight or branched alkyl group. Exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl.
[0017] The term“lower haloalkyl” refers to a CM straight or branched alkyl group that is substituted with one or more halogen atoms.
[0018] The term“heteroatom” means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3 ,4-di hydro-2//-pyrrol yl ), NH (as in pyrrolidinyl) or NRT (as in N-substituted pyrrolidinyl)).
[0019] The term "unsaturated," as used herein, means that a moiety has one or more units of unsaturation.
[0020] As used herein, the term“bivalent CM (or CM) saturated or unsaturated, straight or branched, hydrocarbon chain”, refers to bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein.
[0021] The term“alkylene” refers to a bivalent alkyl group. An“alkylene chain” is a polymethylene group, i.e., -(0¾h-, wherein n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3. A substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
[0022] The term“alkenylene” refers to a bivalent alkenyl group. A substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group. [0023] As used herein, the term“cyclopropylenyl” refers to a bivalent cyclopropyl group of the following structure:
Figure imgf000016_0001
[0024] The term“halogen” means F, Cl, Br, or I.
[0025] The term“aryl” used alone or as part of a larger moiety as in“aralkyl,”“aralkoxy,” or “aryloxyalkyl,” refers to monocyclic or bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members. The term“aryl” may be used interchangeably with the term“aryl ring.” In certain embodiments of the present invention,“aryl” refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may bear one or more substituents. Also included within the scope of the term“aryl,” as it is used herein, is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like.
[0026] The terms“heteroaryl” and“heteroar-,” used alone or as part of a larger moiety, e.g., “heteroaralkyl,” or“heteroaralkoxy,” refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 p electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms. The term“heteroatom” refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen. Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. The terms“heteroaryl” and“heteroar-”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring. Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3-b]-l,4-oxazin-3(4H)-one. A heteroaryl group may be mono- or bicyclic. The term“heteroaryl” may be used interchangeably with the terms“heteroaryl ring,”“heteroaryl group,” or“heteroaromatic any of which terms include rings that are optionally substituted. The term“heteroaralkyl” refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted.
[0027] As used herein, the terms“heterocycle,”“heterocyclyl,”“heterocyclic radical,” and “heterocyclic ring” are used interchangeably and refer to a stable 5- to 7-membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above. When used in reference to a ring atom of a heterocycle, the term "nitrogen" includes a substituted nitrogen. As an example, in a saturated or partially unsaturated ring having 0-3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3,4-dihydro- 2 / pyrrolyl), H (as in pyrrolidinyl), or +NR (as in A substituted pyrrolidinyl).
[0028] A heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted. Examples of such saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl. The terms“heterocycle,”“heterocyclyl,” “heterocyclyl ring,” “heterocyclic group,”“heterocyclic moiety,” and“heterocyclic radical,” are used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3// indolyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl. A heterocyclyl group may be mono- or bicyclic. The term“heterocyclylalkyl” refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.
[0029] As used herein, the term“partially unsaturated” refers to a ring moiety that includes at least one double or triple bond. The term“partially unsaturated” is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
[0030] As described herein, compounds of the invention may contain“optionally substituted” moieties. In general, the term“substituted,” whether preceded by the term“optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an“optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. The term“stable,” as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
[0031] Suitable monovalent substituents on a substitutable carbon atom of an“optionally substituted” group are independently halogen; -(CH2)o-4R°; -(CH2)o-4OR°; -0(CH2)o-4R°, -0- (CH2)O-4C(0)OR°; -(CH2)(MCH(OR°)2 ; -('CH2> SR°; -(CH2)o^Ph, which may be substituted with R°; -(Cfbji OiCTbjo-iPh which may be substituted with R°; -CH=CHPh, which may be substituted with R°; -(CH2)o-40(CH2)o-i-pyridyl which may be substituted with R°; -NO2; -CN; -N3; -(CH2)O-4N(R°)2; -(CH2)O-4N(R0)C(0)R°; -N(R°)C(S)R°; -(CH2)„-
4N(RO)C(0)NR°2; -N(R°)C(S)NR°2; -(CH2)O-4N(R°)C(0)OR0;
N(R°)N(R°)C(0)R°; -N(R°)N(R°)C(0)NR°2; -N(R°)N(R°)C(0)0R°; -(CH2)o-4C(0)R0; - C(S)R°; -(CH2), C(0)0R°; -(CH2)O- C(0)SR0; -(CH2)o- C(0)OSiR°3; -(CH2)o-4OC(0)R°; - OC(0)(CH2)O-4SR-, SC(S)SR°; -(CH2)O-ISC(0)R°; -(CH2)O-4C(0)NR°2; -C(S)NR°2; -C(S)SR°; -SC(S)SR°, -(CH2)O-40C(0)NR°2; -C(0)N(0R°)R°; -C(0)C(0)R°; -C(0)CH2C(0)Ro; - C(NOR°)R°; -(CH2), SSR°; -(CH2> S(0)2R0; -(CH2)O^S(0)2OR°; -(CH2)O-4OS(0)2R0; - S(0)2NR°2; -(CH2)O-4S(0)R°; -N(RO)S(0)2NRo 2; -N(RO)S(0)2R°; -N(OR°)R°; -C(NH)NR°2; - P(0)2R°; -P(0)R°2; -0P(0)R°2; -0P(0)(0Ro)2; SiR°3; -(Ci-4 straight or branched alkyl enejO- N(R°)2; or -(C straight or branched alkylene)C(0)0-N(R°)2, wherein each R° may be substituted as defined below and is independently hydrogen, Ci-6 aliphatic, -CH2Ph, -0(CH2)o- iPh, -CH2-(5-6 membered heteroaryl ring), or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below. [0032] Suitable monovalent substituents on R° (or the ring formed by taking two independent occurrences of R° together with their intervening atoms), are independently halogen, -(CH2)o-2R*, -(haloR*), -(CH2)O-2OH, -(CH2)O-2OR·, -(CH2)O-2CH(OR#)2; -0(haloR#), -CN, -N3, -(CH2)o- 2C(0)R·, -(CH2)O-2C(0)OH, -(CH2)O-2C(0)OR·, -(CH2)O-2SR*, -(CH2)O-2SH, -(CH2)O-2NH2, - (CH2)O-2NHR·, -(CH2)O-2 R*2, -N02, -SiR*3, -OSiR\ -C(0)SR* -(CM straight or branched alkylene)C(0)ORe, or -SSR* wherein each R* is unsubstituted or where preceded by“halo” is substituted only with one or more halogens, and is independently selected from CM aliphatic, - CLhPh, -0(CH2)o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0- 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R° include =0 and =S.
[0033] Suitable divalent substituents on a saturated carbon atom of an“optionally substituted” group include the following: =0, =S, =NNR*¾ =NNHC(0)R*, =NNHC(0)OR*, =NNHS(0)2R*, =NR*, =NOR*,— 0(C(R* 2))2-30— , or -S(C(R*2))2-3S-, wherein each independent occurrence of R* is selected from hydrogen, C aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an“optionally substituted” group include: -0(CR* 2)2- 3O-, wherein each independent occurrence of R* is selected from hydrogen, CM aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0034] Suitable substituents on the aliphatic group of R* include halogen, -R*, -(haloR*), -OH,
-OR*, -0(haloR*), -CN, -C(0)OH, -C(0)OR#, -NH2, -NHR*, -NR*2, or -NO2, wherein each R* is unsubstituted or where preceded by“halo” is substituted only with one or more halogens, and is independently CM aliphatic, -CH2PI1, -0(0¾)o-iR1i, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0035] Suitable substituents on a substitutable nitrogen of an“optionally substituted” group include -R, -NR 2, -C(0)R, -C(0)OR, -C(0)C(0)R,
C(0)CH2C(0)R, -S(0)2R, -S(0)2NR2, -C(S)NR2, -C(NH)NR2, or -N(R)S(0) R; wherein each R' is independently hydrogen, Ci-6 aliphatic which may be substituted as defined below, unsubstituted -OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R, taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0036] Suitable substituents on the aliphatic group of R' are independently halogen, - R·, -(haloR*), -OH, -OR*, -O(haloR'), -CN, -C(0)OH, -C(0)OR*. -NH2, -NHR*, -NR*2, or -NO2, wherein each R* is unsubstituted or where preceded by“halo” is substituted only with one or more halogens, and is independently C1-4 aliphatic, -CH2Ph, -0(CH2)o-iPh, or a 5-6- membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0037] As used herein, the term "pharmaceutically acceptable salt" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et ah, describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphor sulfonate, citrate, cyclopentanepropionate, digluconate, dodecyl sulfate, ethanesulfonate, formate, fiimarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like.
[0038] Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N C 1-4 alky lh salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
[0039] Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. For example, it will be appreciated by those skilled in the art that the compounds of this invention may exist in tautomeric
forms including, but not limited to,
Figure imgf000021_0001
tautomeric forms.
[0040] Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention. Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present invention
[0041] As used herein, the term“agonist” is defined as a compound that binds to and /or activates STING with measurable affinity. In certain embodiments, an agonist or activator has an EC50 and/or effective concentration of less than about 50 mM, less than about 1 mM, less than about 500 nM, less than about 100 nM, less than about 10 nM, or less than about 1 nM. [0042] A compound of the present invention may be tethered to a detectable moiety. It will be appreciated that such compounds are useful as imaging agents. One of ordinary skill in the art will recognize that a detectable moiety may be attached to a provided compound via a suitable substituent. As used herein, the term“suitable substituent” refers to a moiety that is capable of covalent attachment to a detectable moiety. Such moieties are well known to one of ordinary skill in the art and include groups containing, e.g., a carboxylate moiety, an amino moiety, a thiol moiety, or a hydroxyl moiety, to name but a few. It will be appreciated that such moieties may be directly attached to a provided compound or via a tethering group, such as a bivalent saturated or unsaturated hydrocarbon chain. In some embodiments, such moieties may be attached via click chemistry. In some embodiments, such moieties may be attached via a 1,3 -cycloaddition of an azide with an alkyne, optionally in the presence of a copper catalyst. Methods of using click chemistry are known in the art and include those described by Rostovtsev et al, Angew. Chem. Int. Ed. 2002, 44, 2596-99 and Sun etal. , Bioconjugate Chem., 2006, F7, 52-57.
[0043] As used herein, the term“detectable moiety” is used interchangeably with the term "label" and relates to any moiety capable of being detected, e.g., primary labels and secondary labels. Primary labels, such as radioisotopes (e.g., tritium, 32P, 33P, 35S, or 14C), mass-tags, and fluorescent labels are signal generating reporter groups which can be detected without further modifications. Detectable moieties also include luminescent and phosphorescent groups.
[0044] The term“secondary label” as used herein refers to moieties such as biotin and various protein antigens that require the presence of a second intermediate for production of a detectable signal. For biotin, the secondary intermediate may include streptavidin-enzyme conjugates. For antigen labels, secondary intermediates may include antibody-enzyme conjugates. Some fluorescent groups act as secondary labels because they transfer energy to another group in the process of nonradiative fluorescent resonance energy transfer (FRET), and the second group produces the detected signal.
[0045] The terms“fluorescent label”,“fluorescent dye”, and“fluorophore” as used herein refer to moieties that absorb light energy at a defined excitation wavelength and emit light energy at a different wavelength. Examples of fluorescent labels include, but are not limited to: Alexa Fluor dyes (Alexa Fluor 350, Alexa Fluor 488, Alexa Fluor 532, Alexa Fluor 546, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 633, Alexa Fluor 660 and Alexa Fluor 680), AMCA, AMCA-S, BODIPY dyes (BODIPY FL, BODIPY R6G, BODIPY TMR, BODIPY TR, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 576/589, BODIPY 581/591, BODIPY 630/650, BODIPY 650/665), Carboxyrhodamine 6G, carboxy-X-rhodamine (ROX), Cascade Blue, Cascade Yellow, Coumarin 343, Cyanine dyes (Cy3, Cy5, Cy3.5, Cy5.5), Dansyl, Dapoxyl, Dialkylaminocoumarin, 4',5'-Dichloro-2',7'-dimethoxy-fluorescein, DM-NERF, Eosin, Erythrosin, Fluorescein, FAM, Hydroxycoumarin, IRDyes (IRD40, IRD 700, IRD 800), JOE, Lissamine rhodamine B, Marina Blue, Methoxycoumarin, Naphthofluorescein, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, PyMPO, Pyrene, Rhodamine B, Rhodamine 6G, Rhodamine Green, Rhodamine Red, Rhodol Green, 2',4',5',7'-Tetra-bromosulfone- fluorescein, Tetramethyl-rhodamine (TMR), Carboxytetramethylrhodamine (TAMRA), Texas Red, Texas Red-X.
[0046] The term“mass-tag” as used herein refers to any moiety that is capable of being uniquely detected by virtue of its mass using mass spectrometry (MS) detection techniques. Examples of mass-tags include electrophore release tags such as N-[3-[4’-[(p- Methoxytetrafluorobenzyl)oxy]phenyl]-3-methylglyceronyl]isonipecotic Acid, 4’-[2, 3,5,6- Tetrafluoro-4-(pentafluorophenoxyl)]methyl acetophenone, and their derivatives. The synthesis and utility of these mass-tags is described in United States Patents 4,650,750, 4,709,016, 5,360,8191, 5,516,931, 5,602,273, 5,604,104, 5,610,020, and 5,650,270. Other examples of mass- tags include, but are not limited to, nucleotides, dideoxynucleotides, oligonucleotides of varying length and base composition, oligopeptides, oligosaccharides, and other synthetic polymers of varying length and monomer composition. A large variety of organic molecules, both neutral and charged (biomolecules or synthetic compounds) of an appropriate mass range (100-2000 Daltons) may also be used as mass-tags.
[0047] The terms“measurable affinity” and“measurably inhibit,” as used herein, means a measurable change in an STING activity between a sample comprising a compound of the present invention, or composition thereof, and STING, and an equivalent sample comprising STING, in the absence of said compound, or composition thereof.
3. Description of Exemplary Embodiments: [0048] As described above, in certain embodiments, the present invention provides a compound of formula I:
Figure imgf000024_0001
I
or a pharmaceutically acceptable salt thereof, wherein:
m is 0 or 1;
n is 0 or 1;
p is 0 or 1;
wherein m + n + p = 1, 2, or 3;
Ring A is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
wherein Ring A and its substituents is other than
Figure imgf000025_0001
Ring B is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
L1 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy1-, -C(R’)2- -CH(R’)-, -N(R)-, Cy'fL’-Z) , -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2- -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0)2-;
each -Cy1- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R’ is independently hydrogen or C M aliphatic; or
two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring; or
two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R is independently hydrogen, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or:
two R groups on the same nitrogen are taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each L’ is a covalent bond or a CMS bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and
independently replaced
Figure imgf000026_0001
, -N(R)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)C(0)0-, -0C(0)N(R)-, -N(R)S(0)2- - S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0)2- each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, napththylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each Z is hydrogen or a CRBN binding moiety;
L2 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy2-, -C(R’)2- -CH(R’)-, -N(R)-, -Cy2(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(0)-, -0C(0)-, -C(0)0- -S-, -S(0)-, or -S(0)2-;
each -Cy2- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
L3 is a covalent bond or a C O bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy3-, -C(R’)2- -CH(R’)-, -N(R)-, -Cy3(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(0)-, -0C(0)-, -C(0)0-, -S-, -S(0)-, or -S(0)2-;
each -Cy3- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R1 is independently hydrogen, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C( R)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
R2 is hydrogen or an optionally substituted Ci-6 aliphatic group;
each R3 is independently hydrogen, oxo, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, - N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each R4 is independently hydrogen, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
R5 is hydrogen or an optionally substituted Ci-6 aliphatic group;
each R6 is independently hydrogen, oxo, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each of q, s, t, and v is independently 0, 1, 2, 3, or 4;
each of r and u is independently 0 or 1; and
each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
[0049] As generally described above, m is 0 or 1. In some embodiments, m is 0. In some embodiments, m is 1. [0050] As generally described above, n is 0 or 1. In some embodiments, n is 0 In some embodiments, m is 1.
[0051] As generally described above, p is 0 or 1. In some embodiments, p is 0 In some embodiments, m is 1.
[0052] As generally described above, m + n + p = 1, 2, or 3. In some embodiments, m + n + p = 1. In some embodiments, m + n + p = 2. In some embodiments, m + n + p = 3
[0053] In some embodiments, m, n, and p are independently selected from those depicted in Table 1, below.
[0054] As generally described above, Ring A is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and
sulfur, wherein Ring A and its substituents is other than
Figure imgf000029_0001
[0055] In some embodiments, Ring A is phenyl. In some embodiments, Ring A is a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring. In some embodiments, Ring A is a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur. In some embodiments, Ring A is a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, Ring A is an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, Ring A and its substituents is
Figure imgf000030_0001
[0056] In some embodiments, Ring A is selected from those depicted in Table 1 and 2, below.
[0057] In some embodiments, Ring
Figure imgf000030_0002
In some embodiments, Ring A is
In some embodiments, Ring A is
Figure imgf000030_0003
. In some embodiments, Ring A is
In some embodiments, Ring A is
Figure imgf000030_0005
. In some embodiments, Ring A i
Figure imgf000030_0004
. In some embodiments, Ring A is
Figure imgf000030_0006
. In some embodiments,
. K\ . . H
Ring A is . In some embodiments, Ring A is * . In some embodiments, Ring
Figure imgf000030_0007
, . In some embodiments, Ring
S
HGN'
A is / . In some embodiments, Ring A is
Figure imgf000030_0008
. In some embodiments, Ring A is
Figure imgf000031_0001
, some embodiments, Ring A is
Figure imgf000031_0002
. In some embodiments, Ring
Figure imgf000031_0003
In some embodiments, Ring A is
Figure imgf000031_0004
n
some embodiments, Ring A is In some embodiments, Ring
Figure imgf000031_0005
\ some embodiments, Ring A is
Figure imgf000031_0006
. In some embodiments, Ring
Figure imgf000031_0007
. In
Figure imgf000031_0008
some embodiments, Ring A is K 1 In some embodiments, Ring A is / . In some
H ¾ fN ' H ¾ yGN'
embodiments, Ring A is ^ . In some embodiments, Ring A is ^ . In some
embodiments, Ring
Figure imgf000031_0009
. In some embodiments, Ring
Figure imgf000031_0010
some
H y7'N'
embodiments, Ring A is ' . In some embodiments, Ring
Figure imgf000031_0011
In some
N¾/ embodiments, Ring A is r . In some embodiments, Ring
Figure imgf000031_0012
In
Figure imgf000032_0003
[0058] As generally described above, Ring B is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[0059] In some embodiments, Ring B is phenyl. In some embodiments, Ring B is a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring. In some embodiments, Ring B is a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur. In some embodiments, Ring B is a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, Ring B is an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[0060] In some embodiments, Ring B is selected from those depicted in Table 1 and 2, below.
[0061] In some embodiments, Ring
Figure imgf000032_0001
In some embodiments, Ring B is
Figure imgf000032_0002
, In some embodiments, Ring B is H P-
V T
' . In some embodiments, Ring
Figure imgf000033_0001
. In some embodiments, Ring B is
. In some embodiments, Ring
Figure imgf000033_0002
In some embodiments, Ring B is
0—
Figure imgf000033_0003
. In some embodiments, Ring B
Figure imgf000033_0004
In some embodiments, Ring B is
¾_/°'N
Figure imgf000033_0005
. In some embodiments, Ring B is . In some embodiments, Ring B
0
s ^ ' Cc . In some embodiments, Ring
Figure imgf000033_0006
some embodiments, Ring B is
. In some embodiments, Ring B is
Figure imgf000033_0007
. In some embodiments, Ring B is
In some embodiments, Ring
Figure imgf000033_0009
some embodiments, Ring B is
Figure imgf000033_0008
KJ In some embodiments, Ring B is
Figure imgf000033_0010
. In some embodiments, Ring B is
In some embodiments, Ring B is \^0 ¾ SOme embodiments, Ring B is
Figure imgf000033_0011
In some embodiments, Ring
Figure imgf000033_0012
some embodiments, Ring B is
Figure imgf000034_0001
. , g . In some embodiments, Ring B is
Figure imgf000034_0002
In some embodiments, Ring B is
Figure imgf000034_0003
. In some embodiments, Ring B is
\ H
H In some embodiments, Ring B is
Figure imgf000034_0004
In some embodiments, Ring B is
Figure imgf000034_0009
. , g , g
Figure imgf000034_0005
, some embodiments, Ring
H * \ssN'
some embodiments, Ring Bis ' . In some embodiments, Ring B
e embodiments, Ring
Figure imgf000034_0007
In some embodiments, Ring B is
Figure imgf000034_0006
. n some embodiments, Ring
Figure imgf000034_0008
[0062] As generally described above, L1 is a covalent bond or a Ci-io bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy1-, -C(R’)2-, -CH(R’)-, -N(R)-, -Cy'iL’- Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2-, - S(0)2N(R)-, -0- -C(0)- -OC(O)-, -C(0)0- -S-, -S(0)- or -S(0) -.
[0063] In some embodiments, L1 is a covalent bond. In some embodiments, L1 is a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy1-, -C(R’)2-, - CH(R’)-, -N(R)-, Cy'(L’-Z) , -CiR’XL’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -
C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(0)-, or - S(0)2-.
[0064] In some embodiments, L1 is selected from those depicted in Table 1 and 2, below.
[0065] In some embodiments,
Figure imgf000035_0001
[0066] As generally defined above, each -Cy1- is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0067] In some embodiments, each -Cy1- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some emboidments, each -Cy1- is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0068] In some embodiments, -Cy1- is selected from those depicted in Table 1 and 2, below.
[0069] As generally described above, each R’ is independently hydrogen or CM aliphatic; or two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring; or two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [0070] In some embodiments, each R’ is independently hydrogen. In some embodiments, each R’ is independently C aliphatic. In some embodiments, two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring. In some embodimetns, two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0071] In some embodiments, R’ is selected from those depicted in Table 1 and 2, below.
[0072] As generally described above, each R is independently hydrogen, or an optionally substituted group selected from C aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or two R groups on the same nitrogen are taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[0073] In some embodiments, each R is independently hydrogen. In some embodiments, R is an optionally substituted group selected from CM aliphatic. In some embodiments, R is phenyl. In some embodiments, R is naphthalenyl. In some embodiments, R is an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, two R groups on the same nitrogen are taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[0074] In some embodiments, R is selected from those depicted in Table 1 and 2, below.
[0075] As generally defined above, each L’ is a covalent bond or a Ci-is bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the
chain are optionally and independently replaced
Figure imgf000037_0001
-C(R’)2-, -CH(R’)-, -N(R)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)C(0)0- -0C(0)N(R)-, - N(R)S(0)2- -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0) - [0076] In some embodiments, each L’ is a covalent bond. In some embodiments, or each L’ is a Ci-15 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and independently replaced by -Cy’-,
Figure imgf000037_0002
-N(R)C(0)0-, -OC(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, - S-, -S(O)-, or -S(0)2-.
[0077] In some embodiments, L’ is selected from those depicted in Table 1 and 2, below.
[0078] In some embodiments, L’ is
Figure imgf000037_0003
In some embodiments, L’ is
Figure imgf000037_0004
0 . In some embodiments, L’ is 0 In some embodiments,
Figure imgf000037_0005
,
Figure imgf000038_0003
. ,
Figure imgf000038_0004
,
Figure imgf000038_0005
some embodiments, L’ is 0 In some embodiments, L’ is
Figure imgf000038_0001
In some embodiments, L’ is 0 In some embodiments, V is
Figure imgf000038_0002
, In some embodiments, L’ is
Figure imgf000039_0007
,
some embodiments,
Figure imgf000039_0001
In some embodiments, L’
Figure imgf000039_0002
, is
Figure imgf000039_0008
,
some embodiments, L’
Figure imgf000039_0003
In some embodiments, L’ is
Figure imgf000039_0004
, In some embodiments, L’ is
Figure imgf000039_0005
,
In some embodiments,
Figure imgf000039_0006
[0079] As generally defined above, each -Cy’- is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0080] In some embodiments, each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodments, each -Cy’- is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0081] In some embodiments, -Cy’- is selected from those depicted in Table 1 and 2, below.
[0082] As generally described above, each Z is hydrogen or a CRBN binding moiety.
[0083] In some embodiments, each Z is hydrogen. In some embodiments, each Z is a CRBN binding moiety.
[0084] In some embodiments, each Z is selected from those depicted in Table 1 and 2, below.
Figure imgf000040_0001
[0086] As generally described above, L2 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy2-, -C(R’)2-, -CH(R’)-, -N(R)-, -Cy2(L’- Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2- - S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0)2- [0087] In some embodiments, L2 is a covalent bond. In some embodiments, L2 is a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy2-, -C(R’): 2-, -
CH(R’)-, -N(R)-, -Cy2(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -
C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or S(0)2-.
[0088] In some embodiments, L2 is selected from those depicted in Table 1 and 2, below.
[0089] As generally defined above, each -Cy2- is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0090] In some embodiments, each -Cy2- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some emboidments, each -Cy - is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0091] In some embodiments, -Cy2- is selected from those depicted in Table 1 and 2,
Figure imgf000041_0001
embodiments, L2 is
Figure imgf000041_0003
. In some embodiments, L2 is
Figure imgf000041_0002
. In
Figure imgf000041_0004
In some embodiments, L2 is
Figure imgf000041_0005
In some embodiments, L2 is
Figure imgf000041_0006
, In some embodiments, L2 is
Figure imgf000042_0001
,
[0093] As generally described above, L3 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy3-, -C(R’)2-, -CH(R’)-, -N(R)-, -Cy3(L’- Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2- - S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0) - [0094] In some embodiments, L3 is a covalent bond. In some embodiments, L3 is a C MO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy3-, -C(R’)2- -
CH(R , -N(R)-, -Cy3(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -
C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or S(0)2-.
[0095] In some embodiments, L3 is selected from those depicted in Table 1 and 2, below.
[0096] As generally defined above, each -Cy5- is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0097] In some embodiments, each -Cy5- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some emboidments, each -Cy5- is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0098] In some embodiments, -Cy5- is selected from those depicted in Table 1 and 2, below. [0099] In some embodiments,
Figure imgf000042_0002
[00100] As generally described above, L4 is a covalent bond or a Ci-15 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the
chain are optionally and independently replaced
Figure imgf000043_0001
-C(R’)2-, -CH(R’)-, -N(R)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)C(0)0-, -OC(0)N(R)-, - N(R)S(0) - -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0) - [00101] As generally defined above, each -Cy4- is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00102] In some embodiments, each -Cy4- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodments, each -Cy4- is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00103] In some embodiments, -Cy4- is selected from those depicted in Table 1 and 2, below.
[00104] As generally described above, each R1 is independently hydrogen, halogen, -CN, -
N02, -OR, -SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R,
-C(0)NR2, -C(0)N(R)OR, -OC(0)R, -OC(0)NR2, -N(R)C(0)OR, -N(R)C(0) R2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00105] In some embodiments, each R1 is independently hydrogen. In some embodiments, each R1 is independently halogen. In some embodiments, each R1 is independently -CN. In some embodiments, each R1 is independently -N02. In some embodiments, each R1 is independently - OR. In some embodiments, each R1 is independently -SR. In some embodiments, each R1 is independently -NR2. In some embodiments, each R1 is independently -S(0)2R. In some embodiments, each R1 is independently -S(0)(NR)R. In some embodiments, each R1 is independently -S(0)2NR2. In some embodiments, each R1 is independently -S(0)R. In some embodiments, each R1 is independently -S(0) R2. In some embodiments, each R1 is independently -C(0)R. In some embodiments, each R1 is independently -C(0)0R. In some embodiments, each R1 is independently -C(0)NR.2. In some embodiments, each R1 is independently -C(0)N(R)0R. In some embodiments, each R1 is independently -0C(0)R. In some embodiments, each R1 is independently -0C(0)NR2. In some embodiments, each R1 is independently -N(R)C(0)0R. In some embodiments, each R1 is independently -N(R)C(0)NR2. In some embodiments, each R1 is independently -N(R)C(NR) R2. In some embodiments, each R1 is independently -N(R)S(0)2NR2. In some embodiments, each R1 is independently - N(R)S(0)2R. In some embodiments, each R1 is independently -P(0)R2. In some embodiments, each R1 is independently -L’-Z. In some embodiments, each R1 is independently an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00106] In some embodiments, R1 is selected from those depicted in Table 1 and 2, below.
[00107] In some embodiments, R1 is methyl. In some embodiments, R1 is -C(0)NH2. In some embodiments, R1 is -OMe. In some embodiments, R1 is
Figure imgf000044_0001
In some embodiments,
R1 is
Figure imgf000044_0003
In some embodiments, R1 is
Figure imgf000044_0002
In some
embodiments, R1 is
Figure imgf000044_0004
In some embodiments, R1 is -C(0)Me. In some
Figure imgf000044_0005
embodiments, R1 is -C(OH)Me. In some embodiments, R1 is \— / . In some embodiments, R1 is -CN. In some embodiments, R1 is bromo. In some embodiments, R1 is - 0
\ LM N/n°^0 /n0H
CO2H. In certain embodiments, R1 is H . In certain embodiments,
Figure imgf000045_0001
R1 is H In certain embodiments, R1 is H . In certain
Figure imgf000045_0002
embodiments, R1 is ® In certain embodiments, R1 is 0 . In some
embodiments, R1 is
Figure imgf000045_0003
. , . In 0 /— y
N NH
, . 'xf \ / some embodiments,
Figure imgf000045_0004
. In some embodiments, R is ^ . In
some embodiments,
Figure imgf000045_0005
,
some embodiments, R1 is methyl. In some embodiments, R1 is -C(0)0Me.
[00108] As generally described above, R2 is hydrogen or an optionally substituted Ci-6 aliphatic group.
[00109] In some embodiments, each R2 is hydrogen. In some embodiments, R2 is an optionally substituted Ci-6 aliphatic group.
[00110] In some embodiments, R2 is selected from those depicted in Table 1 and 2, below.
[00111] In some embodiments, R2 is methyl. In some embodiments, R2 is ethyl. In some embodiments, R2 is «-propyl. In some embodiments, R2 is allyl.
[00112] As generally described above, each R3 is independently hydrogen, oxo, halogen, -CN, -NO2, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0) R2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)OR, - C(0)NR2, -C(0)N(R)OR, -OC(0)R, -OC(0)NR2, -N(R)C(0)OR, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2 R2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or two R3 groups are optionally taken together with their intervening atoms to form a 5-8 membered saturated or partially unsaturated fused ring having 0- 2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
[00113] In some embodiments, R3 is independently hydrogen. In some embodiments, R3 is independently oxo, halogen. In some embodiments, R3 is independently -CN. In some embodiments, R3 is independently -NC . In some embodiments, R3 is independently -OR. In some embodiments, R3 is independently -SR. In some embodiments, R3 is independently -NR2. In some embodiments, R3 is independently -S(0)2R. In some embodiments, R3 is independently -S(0)(NR)R. In some embodiments, R3 is independently -S(0)2NR2 In some embodiments, R3 is independently -S(0)R. In some embodiments, R3 is independently -S(0)NR2. In some embodiments, R3 is independently -C(0)R. In some embodiments, R3 is independently -C(0)OR. In some embodiments, R3 is independently -C(0)NR2. In some embodiments, R3 is independently -C(0)N(R)OR. In some embodiments, R3 is independently -OC(0)R. In some embodiments, R3 is independently -OC(0)NR2. In some embodiments, R3 is independently -N(R)C(0)OR. In some embodiments, R3 is independently -N(R)C(0)NR2. In some embodiments, R3 is independently -N(R)C(NR) R2. In some embodiments, R3 is independently -N(R)S(0)2NR2. In some embodiments, R3 is independently -N(R)S(0)2R. In some embodiments, R3 is independently -P(0)R2. In some embodiments, each R3 is independently -L’-Z. In some embodiments, R3 is independently an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, two R3 groups are taken together with their intervening atoms to form a 5-8 membered saturated or partially unsaturated fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
[00114] In some embodiments, R3 is selected from those depicted in Table 1 and 2, below.
[00115] In some embodiments, R3 is methyl. In some embodiments, R3 is ethyl. In some embodiments, R3 is chloro. In some embodiments, R3 is -OMe. In some embodiments, R3 is - CH2CH2OH. In some embodiments, R' is -CH2CH2CH2OH. In some embodiments, R3 is - CH2CH2C(0)0H. In some embodiments, R3 is -CH2CH2CH2C(0)0H. In some embodiments, R3 is -CH2CH20CH2CH2CH2C(0)0H. In some embodiments, R3 is CH2CH20CH2CH2CH2CH2C(0)0H. In some embodiments, R3 is
CH2CH20CH2CH20CH2C(0)0H
[00116] As generally described above, each R4 is independently hydrogen, halogen, -CN, - NO2, -OR, -SR, -NR2. -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2I -C(0)R, -C(0)OR, -C(0)NR , -C(0)N(R)OR, -OC(0)R, -OC(0)NR2, -N(R)C(0)OR, -N(R)C(0)NR2, - N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00117] In some embodiments, each R4 is independently hydrogen. In some embodiments, each R4 is independently halogen. In some embodiments, each R4 is independently -CN. In some embodiments, each R4 is independently -N02. In some embodiments, each R4 is independently - OR. In some embodiments, each R4 is independently -SR. In some embodiments, each R4 is independently -NR2. In some embodiments, each R4 is independently -S(0)2R. In some embodiments, each R4 is independently -S(0)(NR)R. In some embodiments, each R4 is independently -S(0)2NR2. In some embodiments, each R4 is independently -S(0)R. In some embodiments, each R4 is independently -S(0)NR2. In some embodiments, each R4 is independently -C(0)R. In some embodiments, each R4 is independently -C(0)OR. In some embodiments, each R4 is independently -C(0)NR2. In some embodiments, each R4 is independently -C(0)N(R)OR. In some embodiments, each R4 is independently -OC(0)R. In some embodiments, each R4 is independently -0C(0)NR2. In some embodiments, each R4 is independently -N(R)C(0)0R. In some embodiments, each R4 is independently -N(R)C(0)NR2. In some embodiments, each R4 is independently -N(R)C(NR)NR2. In some embodiments, each R4 is independently -N(R)S(0)2NR2. In some embodiments, each R4 is independently - N(R)S(0)2R. In some embodiments, each R4 is independently -P(0)R2. In some embodiments, each R4 is independently -L’-Z. In some embodiments, each R4 is independently an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00118] In some embodiments, R4 is selected from those depicted in Table 1 and 2, below.
[00119] In some embodiments, R4 is methyl. In some embodiments, R4 is -C(0) H2. In some embodiments, R4 is -OMe. In some embodiments, R4 is
Figure imgf000048_0001
. In some embodiments,
R4 is
Figure imgf000048_0003
in some embodiments, R4 is
Figure imgf000048_0002
. In some
embodiments, R4 is
Figure imgf000048_0004
In some embodiments, R4 is -C(0)Me. In some
Figure imgf000048_0005
embodiments, R4 is -C(OH)Me. In some embodiments, R4 is \— / . In some embodiments, R4 is -CN. In some embodiments, R4 is bromo. In some embodiments, R4 is -
0
Figure imgf000048_0006
CO2H. In certain embodiments, R4 is H In certain embodiments,
Figure imgf000048_0007
R4 is H In certain embodiments, R4 is H In certain
^o/YOH ^o^Y°Bu embodiments, R4 is ^ In certain embodiments, R4 is 0 in some embodiments, R4 is
Figure imgf000049_0001
, In
some embodiments,
Figure imgf000049_0002
In some embodiments, R4 is
Figure imgf000049_0003
In
Figure imgf000049_0004
some embodiments, R4 is -C(0)0Me.
[00120] As generally described above, R5 is hydrogen or an optionally substituted Ci-6 aliphatic group.
[00121] In some embodiments, each R5 is hydrogen. In some embodiments, R5 is an optionally substituted Ci-6 aliphatic group.
[00122] In some embodiments, R5 is selected from those depicted in Table 1 and 2, below.
[00123] In some embodiments, R5 is methyl. In some embodiments, R5 is ethyl. In some embodiments, R5 is «-propyl. In some embodiments, R5 is allyl.
[00124] As generally described above, each R6 is independently hydrogen, oxo, halogen, -CN,
NO2, OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0) R2, -C(0)R, -C(0)0R, - C(0) R2, -C(0)N(R)0R, -0C(0)R, -0C(0) R2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or two R6 groups are optionally taken together with their intervening atoms to form a 5-8 membered saturated or partially unsaturated fused ring having 0- 2 heteroatoms independently selected from nitrogen, oxygen and sulfur. [00125] In some embodiments, each R6 is independently hydrogen. In some embodiments, each R6 is independently oxo. In some embodiments, each R6 is independently halogen. In some embodiments, each R6 is independently -CN. In some embodiments, each R6 is independently - NO2. In some embodiments, each R6 is independently -OR. In some embodiments, each R6 is independently -SR. In some embodiments, each R6 is independently -NR2. In some embodiments, each R6 is independently -S(0)2R. In some embodiments, each R6 is independently -S(0)( R)R. In some embodiments, each R6 is independently -S(0)2NR.2. In some embodiments, each R6 is independently -S(0)R. In some embodiments, each R6 is independently -S(0)NR2. In some embodiments, each R6 is independently -C(0)R. In some embodiments, each R6 is independently -C(0)0R. In some embodiments, each R6 is independently -C(0)NR2. In some embodiments, each R6 is independently -C(0)N(R)0R. In some embodiments, each R6 is independently -0C(0)R. In some embodiments, each R6 is independently -0C(0)NR2. In some embodiments, each R6 is independently -N(R)C(0)0R. In some embodiments, each R6 is independently -N(R)C(0)NR2. In some embodiments, each R6 is independently -N(R)C(NR)NR2. In some embodiments, each R6 is independently -N(R)S(0)2NR2. In some embodiments, each R6 is independently -N(R)S(0)2R. In some embodiments, each R6 is independently -P(0)R2. In some embodiments, each R6 is independently -L’-Z. In some embodiments, each R6 is independently an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, two R6 groups are optionally taken together with their intervening atoms to form a 5-8 membered saturated or partially unsaturated fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
[00126] In some embodiments, R6 is selected from those depicted in Table 1 and 2, below.
[00127] In some embodiments, R6 is methyl. In some embodiments, R6 is ethyl. In some embodiments, R6 is chloro. In some embodiments, R6 is -OMe. In some embodiments, R6 is - CH2CH2OH. In some embodiments, R6 is -CH2CH2CH2OH. In some embodiments, R6 is - CH2CH2C(0)0H. In some embodiments, R6 is -CH2CH2CH2C(0)0H. In some embodiments, R6 is -CH2CH20CH2CH2CH2C(0)0H. In some embodiments, R6 is CH2CH20CH2CH2CH2CH2C(0)0H. In some embodiments, R6 is
CH2CH20CH2CH20CH2C(0)0H
[00128] As generally described above, each of q, s, t, and v is independently 0, 1, 2, 3, or 4.
[00129] In some embodiments, each of q, s, t, and v is independently 0. In some embodiments, each of q, s, t, and v is independently 1. In some embodiments, each of q, s, t, and v is independently 2. In some embodiments, each of q, s, t, and v is independently 3. In some embodiments, each of q, s, t, and v is independently 4.
[00130] As generally described above, each of r, u and w is independently 0 or 1.
[00131] In some embodiments, each of r and u is independently 0. In some embodiments, each of r and u is independently 1.
[00132] As generally described above, each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
[00133] In some embodiments, each of x is independently 1. In some embodiments, each of x is independently 2. In some embodiments, each of x is independently 3. In some embodiments, each of x is independently 4. In some embodiments, each of x is independently 5. In some embodiments, each of x is independently 6. In some embodiments, each of x is independently 7. In some embodiments, each of x is independently 8. In some embodiments, each of x is independently 9. In some embodiments, each of x is independently 10.
[00134] In some embodiments, each of q, r, s, t, u, v, and x are selected from those depicted in Table 1 and 2, below.
[00135] In some embodiments, the present invention provides a compound of formula I wherein n is 1
Figure imgf000051_0001
, thereby forming a compound of formula I-a:
Figure imgf000052_0001
I-a
or a pharmaceutically acceptable salt thereof, wherein:
each of Ring A, Ring B, R1, R2, R3, R4, R5, R6, L1, L3, m, p, q, r, s, t, u, and v is as defined above; and ~ represents a single or double bond.
[00136] In some embodiments, the present invention provides a compound of formula I wherein m is 1 and L1 is
Figure imgf000052_0002
thereby forming a compound of formula I-b:
Figure imgf000053_0001
or a pharmaceutically acceptable salt thereof, wherein:
each of Ring A, Ring B, R1, R2, R3, R4, R5, R6, L2, L3, n, p, q, r, s, t, u, and v is as defined above.
[00137] In some embodiments, the present invention provides a compound of formula I wherein p is 1 and L3 is
Figure imgf000053_0002
, thereby forming a compound of formula I-c:
Figure imgf000054_0001
or a pharmaceutically acceptable salt thereof, wherein:
each of Ring A, Ring B, R1, R2, R3, R4, R5, R6, L1, L2, m, n, q, r, s, t, u, and v is as defined above.
[00138] In some embodiments, the present invention provides a compound of formula I wherein R6 is -L’-Z, thereby forming a compound of formula I-d:
Figure imgf000054_0002
I-d
or a pharmaceutically acceptable salt thereof, wherein:
each of Ring A, Ring B, R1, R2, R3, R4, R5, L1, L2, L2, L’, m, n, p, q, r, s, t, u, and v is as defined above.
[00139] As described above, in certain embodiments, the present invention provides a compound of formula II:
Figure imgf000055_0001
II
or a pharmaceutically acceptable salt thereof, wherein:
m is 0 or 1;
n is 0 or 1;
p is 0 or 1;
wherein m + n + p = 1, 2, or 3;
L1 is a covalent bond or a C O bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced
Figure imgf000055_0002
-N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(O)-, -OC(O)- -C(0)0-, -S-, -S(O)-, or -S(0)2-;
each -Cy1- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R’ is independently hydrogen or C M aliphatic; or
two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring; or
two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R is independently hydrogen, or an optionally substituted group selected from CM aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or:
two R groups on the same nitrogen are taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each L’ is a covalent bond or a CM5 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and
independently replaced
Figure imgf000056_0001
, -N(R)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)C(0)0-, -0C(0)N(R)-, -N(R)S(0)2- - S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0)2- each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, napththylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each Z is hydrogen or a CRBN binding moiety;
L2 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy2-, -C(R’)2-, -CH(R’)-, -N(R)-, -Cy2(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)- -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2- -S(0)2N(R)-, -0-, -C(O)-, -OC(O)- -C(0)0-, -S-, -S(O)-, or -S(0)2-;
each -Cy2- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
L3 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy3-, -C(R’)2-, -CH(R’)-, -N(R)-, -Cy3(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(0)-, -0C(0)- -C(0)0-, -S-, -S(0)-, or -S(0)2-;
each -Cy3- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R1 is independently hydrogen, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0) R2, -N(R)C(0)0R, -N(R)C(0)NR2, - N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
R2 is hydrogen or an optionally substituted Ci-6 aliphatic group;
each R3 is independently hydrogen, oxo, halogen, -CN, -NO2, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, - N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each R4 is independently hydrogen, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
R5 is hydrogen or an optionally substituted Ci-6 aliphatic group;
each R6 is independently hydrogen, oxo, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each of q, s, t, and v is independently 0, 1, 2, 3, or 4;
each of r and u is independently 0 or 1;
each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; and wherein
(a) when R1 or R4 is -C(0)NR2, each R of the -C(0)NR2 moiety is other than hydrogen or Ci-4 aliphatic; or
(b) in at least one instance of L1, L2, or L3, a methylene unit is replaced with -C(O)-; or
(c) in at least one instance of L1, L2, or L3, a methylene unit adjacent to the benzimidazole or pyrazole ring is replaced with -Cy1-, -Cy2-, or -Cy3-; or
(d) the compound is other than
Figure imgf000059_0001
[00140] As generally described above, m is 0 or 1. In some embodiments, m is 0. In some embodiments, m is 1.
[00141] As generally described above, n is 0 or 1. In some embodiments, n is 0. In some embodiments, m is 1. [00142] As generally described above, p is 0 or 1. In some embodiments, p is 0. In some embodiments, m is 1.
[00143] As generally described above, m + n + p = 1 or 2. In some embodiments, m + n + p = 1. In some embodiments, m + n + p = 2. In some embodiments, m + n + p = 3
[00144] In some embodiments, m, n, and p are independently selected from those depicted in Table 1, below.
[00145] As generally described above, L1 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy1-, -C(R’)2-, -CH(R’)-, -N(R)-, Cy '(L - Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2- - S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0)2- [00146] In some embodiments, L1 is a covalent bond. In some embodiments, L1 is a Ci-io bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy1-, -C(R’)2-, - CH(R’)-, -N(R)-, -Cy^L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, - C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(0)-, or - S(0)2-.
[00147] In some embodiments, L1 is selected from those depicted in Table 1 and 2, below.
[00148] In some embodiments,
Figure imgf000060_0001
[00149] As generally defined above, each -Cy1- is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00150] In some embodiments, each -Cy1- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some emboidments, each -Cy1- is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [00151] In some embodiments, -Cy1- is selected from those depicted in Table 1 and 2, below.
[00152] As generally described above, each R’ is independently hydrogen or C aliphatic; or two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring; or two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00153] In some embodiments, each R’ is independently hydrogen. In some embodiments, each R’ is independently CM aliphatic. In some embodiments, two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring. In some embodimetns, two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00154] In some embodiments, R’ is selected from those depicted in Table 1 and 2, below.
[00155] As generally described above, each R is independently hydrogen, or an optionally substituted group selected from C aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or two R groups on the same nitrogen are taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00156] In some embodiments, each R is independently hydrogen. In some embodiments, R is an optionally substituted group selected from CM aliphatic. In some embodiments, R is phenyl. In some embodiments, R is naphthalenyl. In some embodiments, R is an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, two R groups on the same nitrogen are taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00157] In some embodiments, R is selected from those depicted in Table 1 and 2, below.
[00158] As generally defined above, each L’ is a covalent bond or a Ci-15 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the
chain are optionally and independently replaced
Figure imgf000062_0001
-C(R’)2- -CH(R’)-, -N(R)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)C(0)0-, -OC(0)N(R)-, - N(R)S(0) - -S(0)2N(R)-, -0-, -C(0)- -OC(O)-, -C(0)0- -S-, -S(O)-, or -S(0) -.
[00159] In some embodiments, each L’ is a covalent bond. In some embodiments, or each L’ is a Ci-15 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and independently replaced by -Cy’-,
Figure imgf000062_0002
-N(R)C(0)0- -0C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -
S-, -S(O)-, or -S(0)2-
[00160] In some embodiments, L’ is selected from those depicted in Table 1 and 2, below.
Figure imgf000063_0001
[00161] In some embodiments, L’ is 0 In some embodiments, L’ is
Figure imgf000063_0002
0 . In some embodiments, L’ is 0 . In some embodiments,
Figure imgf000063_0003
some embodiments, L’ is 0 In some embodiments, L’ is
Figure imgf000063_0004
some embodiments, L’ is 0 In some embodiments, L’ is
Figure imgf000064_0001
,
In some embodiments, L’ is
Figure imgf000064_0002
In some embodiments, L’ is
Figure imgf000064_0003
, In some embodiments, L’ is
Figure imgf000064_0004
, In some embodiments,
Figure imgf000064_0005
In some embodiments, L’
Figure imgf000064_0006
Figure imgf000064_0008
,
some embodiments, L’
Figure imgf000064_0007
In some embodiments, L’ is
Figure imgf000065_0001
, In some
embodiments,
Figure imgf000065_0002
[00162] As generally defined above, each -Cy’- is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1 -4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00163] In some embodiments, each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodments, each -Cy’- is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00164] In some embodiments, -Cy’- is selected from those depicted in Table 1 and 2, below.
[00165] As generally described above, each Z is hydrogen or a CRBN binding moiety.
[00166] In some embodiments, each Z is hydrogen. In some embodiments, each Z is a CRBN binding moiety.
[00167] In some embodiments, each Z is selected from those depicted in Table 1 and 2, below.
Figure imgf000065_0003
[00169] As generally described above, L2 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy2-, -C(R’)2-, -CH(R’)-, -N(R)-, -Cy2(L’- Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2-, - S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0) -
[00170] In some embodiments, L2 is a covalent bond. In some embodiments, L2 is a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy2-, -C(R’)2-, -
CH(R’)-, -N(R)-, -Cy2(L’-Z)-, -CiR’XL’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)
C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(0)-, or S(0)2-.
[00171] In some embodiments, L2 is selected from those depicted in Table 1 and 2, below.
[00172] As generally defined above, each -Cy2- is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00173] In some embodiments, each -Cy2- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some emboidments, each -Cy - is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00174] In some embodiments, -Cy2- is selected from those depicted in Table 1 and 2,
Figure imgf000066_0001
embodiments, L2 is
Figure imgf000066_0003
. In some embodiments, L2 is
Figure imgf000066_0002
. In
Figure imgf000066_0004
In some embodiments, L2 is
Figure imgf000067_0001
In some embodiments, L2 is
Figure imgf000067_0002
. , . In some embodiments, L2 is
Figure imgf000067_0003
,
[00176] As generally described above, L3 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy3-, -C(R’)2-, -CH(R’)-, -N(R)-, -Cy3(L’- Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2-, - S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0)2- [00177] In some embodiments, L3 is a covalent bond. In some embodiments, L2 is a Ci-io bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy3-, -C(R’)2-, -
CH(R’)-, -N(R)-, -Cy3(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, - C(0)N(R)-, -N(R)S(0)2- -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0- -S-, -S(O)-, or -
S(0)2-.
[00178] In some embodiments, L3 is selected from those depicted in Table 1 and 2, below.
[00179] As generally defined above, each -Cy5- is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00180] In some embodiments, each -Cy5- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some emboidments, each -Cy5- is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [00181] In some embodiments, -Cy5- is selected from those depicted in Table 1 and 2, below. [00182] In some embodiments,
Figure imgf000068_0001
[00183] As generally described above, L4 is a covalent bond or a Ci-15 bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the
chain are optionally and independently replaced
Figure imgf000068_0002
-C(R’)2- -CH(R’)-, -N(R)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)C(0)0- -OC(0)N(R)-, - N(R)S(0)2- -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0) - [00184] As generally defined above, each -Cy4- is independently an optionally substituted bivalent ring selected from phenyl ene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00185] In some embodiments, each -Cy4- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4- 7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodments, each -Cy4- is independently an optionally substituted 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00186] In some embodiments, -Cy4- is selected from those depicted in Table 1 and 2, below.
[00187] As generally described above, each R1 is independently hydrogen, halogen, -CN, -
NO2, -OR, -SR, - R2, -S(0)2R, -S(0)( R)R, -S(0)2NR2, -S(0)R, -S(0) R2, -C(0)R, -C(0)0R,
-C(0)NR2, -C(0)N(R)OR, -OC(0)R, -OC(0)NR2, -N(R)C(0)OR, -N(R)C(0) R2, - N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. [00188] In some embodiments, each R1 is independently hydrogen. In some embodiments, each R1 is independently halogen. In some embodiments, each R1 is independently -CN. In some embodiments, each R1 is independently -NO2. In some embodiments, each R1 is independently - OR. In some embodiments, each R1 is independently -SR. In some embodiments, each R1 is independently -NR2. In some embodiments, each R1 is independently -S(0)2R. In some embodiments, each R1 is independently -S(0)(NR)R. In some embodiments, each R1 is independently -S(0)2NR2. In some embodiments, each R1 is independently -S(0)R. In some embodiments, each R1 is independently -S(0)NR2. In some embodiments, each R1 is independently -C(0)R. In some embodiments, each R1 is independently -C(0)0R. In some embodiments, each R1 is independently -C(0)NR2 In some embodiments, each R1 is independently -C(0)N(R)0R. In some embodiments, each R1 is independently -0C(0)R. In some embodiments, each R1 is independently -0C(0)NR2. In some embodiments, each R1 is independently -N(R)C(0)0R. In some embodiments, each R1 is independently -N(R)C(0)NR2. In some embodiments, each R1 is independently -N(R)C(NR)NR2. In some embodiments, each R1 is independently -N(R)S(0)2NR2. In some embodiments, each R1 is independently - N(R)S(0)2R. In some embodiments, each R1 is independently -P(0)R2. In some embodiments, each R1 is independently -L’-Z. In some embodiments, each R1 is independently an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00189] In some embodiments, R1 is selected from those depicted in Table 1 and 2, below.
[00190] In some embodiments, R1 is methyl. In some embodiments, R1 is -C(0)NH2. In some embodiments, R1 is -OMe. In some embodiments, R1 is
Figure imgf000069_0001
In some embodiments,
Figure imgf000069_0002
-C(OH)Me. In some embodiments, R1 is \— f . In some embodiments, R1 is -CN. In some embodiments, R1 is bromo. In some embodiments, R1 is -CO2H. In some embodiments, R1
Figure imgf000070_0001
is 0 . In some embodiments, R , i:s. . In some embodiments,
Figure imgf000070_0002
R1 i is OH . In some embodiments, R1 is In some embodiments,
Figure imgf000070_0003
, . In some embodiments,
R1 is methyl. In some embodiments, R1 is -C(0)OMe.
[00191] As generally described above, R2 is hydrogen or an optionally substituted Ci-6 aliphatic group.
[00192] In some embodiments, each R2 is hydrogen. In some embodiments, R2 is an optionally substituted Ci-6 aliphatic group.
[00193] In some embodiments, R2 is selected from those depicted in Table 1 and 2, below.
[00194] In some embodiments, R2 is methyl. In some embodiments, R2 is ethyl. In some embodiments, R2 is «-propyl. In some embodiments, R2 is allyl.
[00195] As generally described above, each R3 is independently hydrogen, oxo, halogen, -CN, -NO2, -OR,
SR, -NR , -S(0)2R, -S(0)(NR)R, -S(0)2 R2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)OR, - C(0)NR2, -C(0)N(R)OR, -OC(0)R, -OC(0)NR2, -N(R)C(0)OR, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R , -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or two R3 groups are optionally taken together with their intervening atoms to form a 5-8 membered saturated or partially unsaturated fused ring having 0- 2 heteroatoms independently selected from nitrogen, oxygen and sulfur. [00196] In some embodiments, R3 is independently hydrogen. In some embodiments, R3 is independently oxo, halogen. In some embodiments, R3 is independently -CN. In some embodiments, R3 is independently -NO2. In some embodiments, R3 is independently -OR. In some embodiments, R3 is independently -SR. In some embodiments, R3 is independently -NR2. In some embodiments, R3 is independently -S(0)2R. In some embodiments, R3 is independently -S(0)(NR)R. In some embodiments, R3 is independently -S(0)2NR2 In some embodiments, R3 is independently -S(0)R. In some embodiments, R3 is independently -S(0)NR2. In some embodiments, R3 is independently -C(0)R. In some embodiments, R3 is independently -C(0)OR. In some embodiments, R3 is independently -C(0)NR2. In some embodiments, R3 is independently -C(0)N(R)OR. In some embodiments, R3 is independently -OC(0)R. In some embodiments, R3 is independently -OC(0)NR2. In some embodiments, R3 is independently -N(R)C(0)OR. In some embodiments, R3 is independently -N(R)C(0)NR2. In some embodiments, R3 is independently -N(R)C(NR) R2. In some embodiments, R3 is independently -N(R)S(0)2NR2. In some embodiments, R3 is independently -N(R)S(0)2R. In some embodiments, R3 is independently -P(0)R2. In some embodiments, each R3 is independently -L’-Z. In some embodiments, R3 is independently an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R3 is independently two R3 groups are optionally taken together with their intervening atoms to form a 5-8 membered saturated or partially unsaturated fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
[00197] In some embodiments, R3 is selected from those depicted in Table 1 and 2, below.
[00198] In some embodiments, R3 is methyl. In some embodiments, R3 is ethyl. In some embodiments, R3 is chloro. In some embodiments, R3 is -OMe. In some embodiments, R3 is - CH2CH2OH. In some embodiments, R3 is -CH2CH2CH2OH. In some embodiments, R3 is - CH2CH2C(0)0H. In some embodiments, R3 is -CH2CH2CH2C(0)0H. In some embodiments, R3 is -CH2CH20CH2CH2CH2C(0)0H. In some embodiments, R3 is CH2CH20CH2CH2CH2CH2C(0)0H. In some embodiments, R3 is
CH2CH20CH2CH20CH2C(0)0H.
[00199] As generally described above, each R4 is independently hydrogen, halogen, -CN, - NO2, -OR, -SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, -C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, - N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00200] In some embodiments, each R4 is independently hydrogen. In some embodiments, each R4 is independently halogen. In some embodiments, each R4 is independently -CN. In some embodiments, each R4 is independently -N02. In some embodiments, each R4 is independently - OR. In some embodiments, each R4 is independently -SR. In some embodiments, each R4 is independently -NR2. In some embodiments, each R4 is independently -S(0)2R. In some embodiments, each R4 is independently -S(0)(NR)R. In some embodiments, each R4 is independently -S(0)2NR2. In some embodiments, each R4 is independently -S(0)R. In some embodiments, each R4 is independently -S(0)NR2. In some embodiments, each R4 is independently -C(0)R. In some embodiments, each R4 is independently -C(0)OR. In some embodiments, each R4 is independently -C(0)NR2. In some embodiments, each R4 is independently -C(0)N(R)OR. In some embodiments, each R4 is independently -OC(0)R. In some embodiments, each R4 is independently -OC(0)NR2. In some embodiments, each R4 is independently -N(R)C(0)OR. In some embodiments, each R4 is independently -N(R)C(0)NR2. In some embodiments, each R4 is independently -N(R)C(NR)NR2. In some embodiments, each R4 is independently -N(R)S(0)2NR2. In some embodiments, each R4 is independently - N(R)S(0)2R. In some embodiments, each R4 is independently -P(0)R2. In some embodiments, each R4 is independently -L’-Z. In some embodiments, each R4 is independently an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
[00201] In some embodiments, R4 is selected from those depicted in Table 1 and 2, below.
[00202] In some embodiments, R4 is methyl. In some embodiments, R4 is -C(0)NH2. In some embodiments, R4 is -OMe. In some embodiments, R4 i is, ^0^0H . In some embodiments,
Figure imgf000073_0002
-C(OH)Me. In some embodiments, R4 is \— f . In some embodiments, R4 is -CN. In some embodiments, R4 is bromo. In some embodiments, R4 is -CO2H. In some embodiments, R1
Figure imgf000073_0001
is o In some embodiments, R4 is In some embodiments.
Figure imgf000073_0003
R4 is . In some embodiments, R4 is O . In some embodiments,
R4 is -C(0)0Me.
[00203] As generally described above, R5 is hydrogen or an optionally substituted Ci-6 aliphatic group.
[00204] In some embodiments, each R5 is hydrogen. In some embodiments, R5 is an optionally substituted Ci-6 aliphatic group.
[00205] In some embodiments, R5 is selected from those depicted in Table 1 and 2, below.
[00206] In some embodiments, R5 is methyl. In some embodiments, R5 is ethyl. In some embodiments, R5 is «-propyl. In some embodiments, R5 is allyl. [00207] As generally described above, each R6 is independently hydrogen, oxo, halogen, -CN, -NOi, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)OR, - C(0)NR2, -C(0)N(R)OR, -OC(0)R, -OC(0)NR2, -N(R)C(0)OR, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or two R6 groups are optionally taken together with their intervening atoms to form a 5-8 membered saturated or partially unsaturated fused ring having 0- 2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
[00208] In some embodiments, each R6 is independently hydrogen. In some embodiments, each R6 is independently oxo. In some embodiments, each R6 is independently halogen. In some embodiments, each R6 is independently -CN. In some embodiments, each R6 is independently - N02. In some embodiments, each R6 is independently -OR. In some embodiments, each R6 is independently -SR. In some embodiments, each R6 is independently -NR2. In some embodiments, each R6 is independently -S(0)2R. In some embodiments, each R6 is independently -S(0)(NR)R. In some embodiments, each R6 is independently -S(0)2NR2. In some embodiments, each R6 is independently -S(0)R. In some embodiments, each R6 is independently -S(0)NR2. In some embodiments, each R6 is independently -C(0)R. In some embodiments, each R6 is independently -C(0)OR. In some embodiments, each R6 is independently -C(0)NR2. In some embodiments, each R6 is independently -C(0)N(R)OR. In some embodiments, each R6 is independently -OC(0)R. In some embodiments, each R6 is independently -OC(0)NR2. In some embodiments, each R6 is independently -N(R)C(0)OR. In some embodiments, each R6 is independently -N(R)C(0)NR2. In some embodiments, each R6 is independently -N(R)C(NR)NR2. In some embodiments, each R6 is independently -N(R)S(0)2NR2. In some embodiments, each R6 is independently -N(R)S(0)2R. In some embodiments, each R6 is independently -P(0)R2. In some embodiments, each R6 is independently -L’-Z. In some embodiments, each R6 is independently an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, two R6 groups are optionally taken together with their intervening atoms to form a 5-8 membered saturated or partially unsaturated fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen and sulfur.
[00209] In some embodiments, R6 is selected from those depicted in Table 1 and 2, below.
[00210] In some embodiments, R6 is methyl. In some embodiments, R6 is ethyl. In some embodiments, R6 is chloro. In some embodiments, R6 is -OMe. In some embodiments, R6 is - CH2CH2OH. In some embodiments, R6 is -CH2CH2CH2OH. In some embodiments, R6 is - CH2CH2C(0)0H. In some embodiments, R6 is -CH2CH2CH2C(0)0H. In some embodiments, R6 is -CH2CH20CH2CH2CH2C(0)0H. In some embodiments, R6 is CH2CH20CH2CH2CH2CH2C(0)0H. In some embodiments, R6 is
CH2CH20CH2CH20CH2C(0)0H
[00211] As generally described above, each of q, s, t, and v is independently 0, 1, 2, 3, or 4.
[00212] In some embodiments, each of q, s, t, and v is independently 0. In some embodiments, each of q, s, t, and v is independently 1. In some embodiments, each of q, s, t, and v is independently 2. In some embodiments, each of q, s, t, and v is independently 3. In some embodiments, each of q, s, t, and v is independently 4.
[00213] As generally described above, each of r, u and w is independently 0 or 1.
[00214] In some embodiments, each of r and u is independently 0. In some embodiments, each of r and u is independently 1.
[00215] As generally described above, each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
[00216] In some embodiments, each of x is independently 1. In some embodiments, each of x is independently 2. In some embodiments, each of x is independently 3. In some embodiments, each of x is independently 4. In some embodiments, each of x is independently 5. In some embodiments, each of x is independently 6. In some embodiments, each of x is independently 7. In some embodiments, each of x is independently 8. In some embodiments, each of x is independently 9. In some embodiments, each of x is independently 10. [00217] In some embodiments, each of q, r, s, t, u, v, and x are selected from those depicted in Table 1 and 2, below.
[00218] As generally defined above, (a) when R1 or R4 is -C(0)NR2, each R of the -C(0)NR2 moiety is other than hydrogen or CM aliphatic; or
(b) in at least one instance of L1, L2, or L3, a methylene unit is replaced with -C(O)-; or
(c) in at least one instance of L1, L2, or L3, a methylene unit adjacent to the benzimidazole or pyrazole ring is replaced with -Cy1-, -Cy2-, or -Cy3-.
[00219] In some embodiments, the present invention provides a compound of formula II wherein n is 1 and L2 is
Figure imgf000076_0001
, ereby forming a compound of formula Il-a:
Figure imgf000076_0002
Il-a
or a pharmaceutically acceptable salt thereof, wherein:
each of R1, R2, R3, R4, R5, R6, L1, L3, m, p, q, r, s, t, u, and v is as defined above; and = represents a single or double bond.
[00220] In some embodiments, the present invention provides a compound of formula II wherein m is 1 and L1 is
Figure imgf000076_0003
, thereby forming a compound of formula Il-b
Figure imgf000077_0001
or a pharmaceutically acceptable salt thereof, wherein:
each of R1, R2, R3, R4, R5, R6, L2, L3, n, p, q, r, s, t, u, and v is as defined above.
[00221] In some embodiments, the present invention provides a compound of formula II wherein p is 1 and L3 is
Figure imgf000077_0002
, thereby forming a compound of formula II-c:
Figure imgf000077_0003
or a pharmaceutically acceptable salt thereof, wherein:
each of R1, R2, R3, R4, R5, R6, L1, L2, m, n, q, r, s, t, u, and v is as defined above. [00222] In some embodiments, the present invention provides a compound of formula II wherein R6 is -L’-Z, thereby forming a compound of formula Il-d:
Figure imgf000078_0001
or a pharmaceutically acceptable salt thereof, wherein:
each of R1, R2, R3, R5, R6, L1, L2, L3, L’, Z, m, n, p, q, r, s, t, u, and v is as defined above.
[00223] Exemplary compounds of the invention are set forth in Table 1 and 2, below.
Table 1. Exemplary Compounds
Figure imgf000078_0002
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0002
Table 2. Exemplary Compounds
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
, Formulation and Administration
Pharmaceutically Acceptable Compositions [00224] According to another embodiment, the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The amount of compound in compositions of this invention is such that is effective to modulate STING, or a mutant thereof, - dependent type I interferon production in a biological sample or in a patient. In certain embodiments, the amount of compound in compositions of this invention is such that is effective to measurably modulate STING, or a mutant thereof, -dependent type I interferon production in a biological sample or in a patient. In certain embodiments, a composition of this invention is formulated for administration to a patient in need of such composition. In some embodiments, a composition of this invention is formulated for oral administration to a patient.
[00225] The term“patient,” as used herein, means an animal, preferably a mammal, and most preferably a human.
[00226] The term“pharmaceutically acceptable carrier, adjuvant, or vehicle” refers to a nontoxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, polyethylene glycol and wool fat.
[00227] A“pharmaceutically acceptable derivative” means any non-toxic salt, ester, salt of an ester or other derivative of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an active metabolite or residue thereof.
[00228] Compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term“parenteral” as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
[00229] For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxy ethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
[00230] Pharmaceutically acceptable compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and com starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
[00231] Alternatively, pharmaceutically acceptable compositions of this invention may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.
[00232] Pharmaceutically acceptable compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
[00233] Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used.
[00234] For topical applications, provided pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Carriers for topical administration of compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, provided pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
[00235] For ophthalmic use, provided pharmaceutically acceptable compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.
[00236] Pharmaceutically acceptable compositions of this invention may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well- known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents. [00237] Most preferably, pharmaceutically acceptable compositions of this invention are formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, pharmaceutically acceptable compositions of this invention are administered without food. In other embodiments, pharmaceutically acceptable compositions of this invention are administered with food.
[00238] The amount of compounds of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration. Preferably, provided compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the compound can be administered to a patient receiving these compositions.
[00239] It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
Uses of Compounds and Pharmaceutically Acceptable Compositions
[00240] In certain embodiments, the invention provides a method of inducing an immune response in a subject, comprising administering a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof to the subject.
[00241] In certain embodiments, the invention provides a method of inducing a STING- dependent type I interferon production in a subject, comprising administering a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof to the subject.
[00242] In certain embodiments, the invention provides a method of inducing a STING- dependent cytokine production in a subject, comprising administering a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof to the subject. [00243] In some embodiments, the invention provides a pharmaceutical composition comprising an effective amount of a compound of the invention, or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof, and a pharmaceutically acceptable carrier.
[00244] In certain aspects, the invention provides a method of treating cell proliferation disorders, including cancers, benign papillomatosis, gestational trophoblastic diseases, and benign neoplastic diseases, such as skin papilloma (warts) and genital papilloma.
[00245] In one aspect, the invention provides a method of treating a cell proliferation disorder in a subject, comprising administering a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof, to the subj ect.
[00246] In certain embodiments, the cell proliferation disorder is cancer.
[00247] In certain embodiments, the cancer is brain cancer, leukemia, skin cancer, prostate cancer, thyroid cancer, colon cancer, lung cancer or sarcoma. In another embodiment the cancer is selected from the group consisting of glioma, glioblastoma multiforme, paraganglioma, suprantentorial primordial neuroectodermal tumors, acute myeloid leukemia, myelodysplastic syndrome, chronic myelogenous leukemia, melanoma, breast, prostate, thyroid, colon, lung, central chondrosarcoma, central and periosteal chondroma tumors, fibrosarcoma, and cholangiocarcinoma.
[00248] In certain embodiments, the cancer is selected from brain and spinal cancers, cancers of the head and neck, leukemia and cancers of the blood, skin cancers, cancers of the reproductive system, cancers of the gastrointestinal system, liver and bile duct cancers, kidney and bladder cancers, bone cancers, lung cancers, malignant mesothelioma, sarcomas, lymphomas, glandular cancers, thyroid cancers, heart tumors, germ cell tumors, malignant neuroendocrine (carcinoid) tumors, midline tract cancers, and cancers of unknown primary (i.e., cancers in which a metastasized cancer is found but the original cancer site is not known). In particular embodiments, the cancer is present in an adult patient; in additional embodiments, the cancer is present in a pediatric patient. In particular embodiments, the cancer is AIDS-related.
[00249] In a further embodiment, the cancer is selected from brain and spinal cancers. In particular embodiments, the cancer is selected from the group consisting of anaplastic astrocytomas, glioblastomas, astrocytomas, and estheosioneuroblastomas (olfactory blastomas). In particular embodiments, the brain cancer is selected from the group consisting of astrocytic tumor (e.g., pilocytic astrocytoma, subependymal giant-cell astrocytoma, diffuse astrocytoma, pleomorphic xanthoastrocytoma, anaplastic astrocytoma, astrocytoma, giant cell glioblastoma, glioblastoma, secondary glioblastoma, primary adult glioblastoma, and primary pediatric glioblastoma), oligodendroglial tumor (e.g., oligodendroglioma, and anaplastic oligodendroglioma), oligoastrocytic tumor (e.g., oligoastrocytoma, and anaplastic oligoastrocytoma), ependymoma (e.g., myxopapillary ependymoma, and anaplastic ependymoma); medulloblastoma, primitive neuroectodermal tumor, schwannoma, meningioma, atypical meningioma, anaplastic meningioma, pituitary adenoma, brain stem glioma, cerebellar astrocytoma, cerebral astorcytom a/ al i gn an t glioma, visual pathway and hypothalmic glioma, and primary central nervous system lymphoma. In specific instances of these embodiments, the brain cancer is selected from the group consisting of glioma, glioblastoma multiforme, paraganglioma, and suprantentorial primordial neuroectodermal tumors (sPNET).
[00250] In specific embodiments, the cancer is selected from cancers of the head and neck, including nasopharyngeal cancers, nasal cavity and paranasal sinus cancers, hypopharyngeal cancers, oral cavity cancers (e.g., squamous cell carcinomas, lymphomas, and sarcomas), lip cancers, oropharyngeal cancers, salivary gland tumors, cancers of the larynx (e.g., laryngeal squamous cell carcinomas, rhabdomyosarcomas), and cancers of the eye or ocular cancers. In particular embodiments, the ocular cancer is selected from the group consisting of intraocular melanoma and retinoblastoma.
[00251] In specific embodiments, the cancer is selected from leukemia and cancers of the blood. In particular embodiments, the cancer is selected from the group consisting of myeloproliferative neoplasms, myelodysplastic syndromes, myelodysplastic/myeloproliferative neoplasms, acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), chronic myelogenous leukemia (CML), myeloproliferative neoplasm (MPN), post-MPN AML, post-MDS AML, del(5q)- associated high risk MDS or AML, blast-phase chronic myelogenous leukemia, angioimmunoblastic lymphoma, acute lymphoblastic leukemia, Langerans cell histiocytosis, hairy cell leukemia, and plasma cell neoplasms including plasmacytomas and multiple myelomas. Leukemias referenced herein may be acute or chronic. [00252] In specific embodiments, the cancer is selected from skin cancers. In particular embodiments, the skin cancer is selected from the group consisting of melanoma, squamous cell cancers, and basal cell cancers.
[00253] In specific embodiments, the cancer is selected from cancers of the reproductive system. In particular embodiments, the cancer is selected from the group consisting of breast cancers, cervical cancers, vaginal cancers, ovarian cancers, prostate cancers, penile cancers, and testicular cancers. In specific instances of these embodiments, the cancer is a breast cancer selected from the group consisting of ductal carcinomas and phyllodes tumors. In specific instances of these embodiments, the breast cancer may be male breast cancer or female breast cancer. In specific instances of these embodiments, the cancer is a cervical cancer selected from the group consisting of squamous cell carcinomas and adenocarcinomas. In specific instances of these embodiments, the cancer is an ovarian cancer selected from the group consisting of epithelial cancers.
[00254] In specific embodiments, the cancer is selected from cancers of the gastrointestinal system. In particular embodiments, the cancer is selected from the group consisting of esophageal cancers, gastric cancers (also known as stomach cancers), gastrointestinal carcinoid tumors, pancreatic cancers, gallbladder cancers, colorectal cancers, and anal cancer. In instances of these embodiments, the cancer is selected from the group consisting of esophageal squamous cell carcinomas, esophageal adenocarcinomas, gastric adenocarcinomas, gastrointestinal carcinoid tumors, gastrointestinal stromal tumors, gastric lymphomas, gastrointestinal lymphomas, solid pseudopapillary tumors of the pancreas, pancreatoblastoma, islet cell tumors, pancreatic carcinomas including acinar cell carcinomas and ductal adenocarcinomas, gallbladder adenocarcinomas, colorectal adenocarcinomas, and anal squamous cell carcinomas.
[00255] In specific embodiments, the cancer is selected from liver and bile duct cancers. In particular embodiments, the cancer is liver cancer (hepatocellular carcinoma). In particular embodiments, the cancer is bile duct cancer (cholangiocarcinoma); in instances of these embodiments, the bile duct cancer is selected from the group consisting of intrahepatic cholangiocarcinoma and extrahepatic cholangiocarcinoma.
[00256] In specific embodiments, the cancer is selected from kidney and bladder cancers. In particular embodiments, the cancer is a kidney cancer selected from the group consisting of renal cell cancer, Wilms tumors, and transitional cell cancers. In particular embodiments, the cancer is a bladder cancer selected from the group consisting of urethelial carcinoma (a transitional cell carcinoma), squamous cell carcinomas, and adenocarcinomas.
[00257] In specific embodiments, the cancer is selected from bone cancers. In particular embodiments, the bone cancer is selected from the group consisting of osteosarcoma, malignant fibrous histiocytoma of bone, Ewing sarcoma, and chordoma.
[00258] In specific embodiments, the cancer is selected from lung cancers. In particular embodiments, the lung cancer is selected from the group consisting of non-small cell lung cancer, small cell lung cancers, bronchial tumors, and pleuropulmonary blastomas.
[00259] In specific embodiments, the cancer is selected from malignant mesothelioma. In particular embodiments, the cancer is selected from the group consisting of epithelial mesothelioma and sarcomatoids.
[00260] In specific embodiments, the cancer is selected from sarcomas. In particular embodiments, the sarcoma is selected from the group consisting of central chondrosarcoma, central and periosteal chondroma, fibrosarcoma, clear cell sarcoma of tendon sheaths, and Kaposi's sarcoma.
[00261] In specific embodiments, the cancer is selected from lymphomas. In particular embodiments, the cancer is selected from the group consisting of Hodgkin lymphoma (e.g., Reed- Stemberg cells), non-Hodgkin lymphoma (e.g., diffuse large B-cell lymphoma, follicular lymphoma, mycosis fungoides, Sezary syndrome, primary central nervous system lymphoma), cutaneous T-cell lymphomas, and primary central nervous system lymphomas.
[00262] In specific embodiments, the cancer is selected from glandular cancers. In particular embodiments, the cancer is selected from the group consisting of adrenocortical cancer, pheochromocytomas, paragangliomas, pituitary tumors, thymoma, and thymic carcinomas.
[00263] In specific embodiments, the cancer is selected from thyroid cancers. In particular embodiments, the thyroid cancer is selected from the group consisting of medullary thyroid carcinomas, papillary thyroid carcinomas, and follicular thyroid carcinomas.
[00264] In specific embodiments, the cancer is selected from germ cell tumors. In particular embodiments, the cancer is selected from the group consisting of malignant extracranial germ cell tumors and malignant extragonadal germ cell tumors. In specific instances of these embodiments, the malignant extragonadal germ cell tumors are selected from the group consisting of nonseminomas and seminomas.
[00265] In specific embodiments, the cancer is selected from heart tumors. In particular embodiments, the heart tumor is selected from the group consisting of malignant teratoma, lymphoma, rhabdomyosacroma, angiosarcoma, chondrosarcoma, infantile fibrosarcoma, and synovial sarcoma.
[00266] In specific embodiments, the cell-proliferation disorder is selected from benign papillomatosis, benign neoplastic diseases and gestational trophoblastic diseases. In particular embodiments, the benign neoplastic disease is selected from skin papilloma (warts) and genital papilloma. In particular embodiments, the gestational trophoblastic disease is selected from the group consisting of hydatidiform moles, and gestational trophoblastic neoplasia (e.g., invasive moles, choriocarcinomas, placental-site trophoblastic tumors, and epithelioid trophoblastic tumors).
Combination Therapies
[00267] Depending upon the particular condition, or disease, to be treated, additional therapeutic agents, which are normally administered to treat that condition, may be administered in combination with compounds and compositions of this invention. As used herein, additional therapeutic agents that are normally administered to treat a particular disease, or condition, are known as“appropriate for the disease, or condition, being treated.”
[00268] In certain embodiments, a provided combination, or composition thereof, is administered in combination with another therapeutic agent.
[00269] Examples of agents the combinations of this invention may also be combined with include, without limitation: treatments for Alzheimer’s Disease such as Aricept® and Excel on®; treatments for HIV such as ritonavir; treatments for Parkinson’s Disease such as L- DOPA/carbidopa, entacapone, ropinrole, pramipexole, bromocriptine, pergolide, trihexephendyl, and amantadine; agents for treating Multiple Sclerosis (MS) such as beta interferon (e.g., Avonex® and Rebif®), Copaxone®, and mitoxantrone; treatments for asthma such as albuterol and Singulair®; agents for treating schizophrenia such as zyprexa, risperdal, seroquel, and haloperidol; anti-inflammatory agents such as corticosteroids, TNF blockers, IL-1 RA, azathioprine, cyclophosphamide, and sulfasalazine; immunomodulatory and immunosuppressive agents such as cyclosporin, tacrolimus, rapamycin, my cophenol ate mofetil, interferons, corticosteroids, cyclophophamide, azathioprine, and sulfasalazine; neurotrophic factors such as acetylcholinesterase inhibitors, MAO inhibitors, interferons, anti-convulsants, ion channel blockers, riluzole, and anti -Parkinsonian agents; agents for treating cardiovascular disease such as beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers, and statins; agents for treating liver disease such as corticosteroids, cholestyramine, interferons, and anti-viral agents; agents for treating blood disorders such as corticosteroids, anti-leukemic agents, and growth factors; agents that prolong or improve pharmacokinetics such as cytochrome P450 inhibitors (i.e., inhibitors of metabolic breakdown) and CYP3A4 inhibitors (e.g., ketokenozole and ritonavir), and agents for treating immunodeficiency disorders such as gamma globulin.
[00270] In certain embodiments, combination therapies of the present invention, or a pharmaceutically acceptable composition thereof, are administered in combination with a monoclonal antibody or an siRNA therapeutic.
[00271] Those additional agents may be administered separately from a provided combination therapy, as part of a multiple dosage regimen. Alternatively, those agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as part of a multiple dosage regime, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another normally within five hours from one another.
[00272] As used herein, the term“combination,”“combined,” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention. For example, a combination of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form.
[00273] The amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent. Preferably the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent. [00274] In one embodiment, the present invention provides a composition comprising a compound of formula I or formula II, and one or more additional therapeutic agents. The therapeutic agent may be administered together with a compound of formula I or formula II, or may be administered prior to or following administration of a compound of formula I or formula II. Suitable therapeutic agents are described in further detail below. In certain embodiments, a compound of formula I or formula II may be administered up to 5 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5, hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, or 18 hours before the therapeutic agent. In other embodiments, a compound of formula I or formula II may be administered up to 5 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5, hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, or 18 hours following the therapeutic agent.
[00275] In another embodiment, the present invention provides a method of treating an inflammatory disease, disorder or condition by administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents. Such additional therapeutic agents may be small molecules or recombinant biologic agents and include, for example, acetaminophen, non-steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, etodolac (Lodine®) and celecoxib, colchicine (Colcrys®), corticosteroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like, probenecid, allopurinol, febuxostat (Uloric®), sulfasalazine (Azulfidine®), antimalarials such as hydroxychloroquine (Plaquenil®) and chloroquine (Aralen®), methotrexate (Rheumatrex®), gold salts such as gold thioglucose (Solganal®), gold thiomalate (Myochrysine®) and auranofm (Ridaura®), D-penicillamine (Depen® or Cuprimine®), azathioprine (Imuran®), cyclophosphamide (Cytoxan®), chlorambucil (Leukeran®), cyclosporine (Sandimmune®), leflunomide (Arava®) and “anti-TNF” agents such as etanercept (Enbrel®), infliximab (Remicade®), golimumab (Simponi®), certolizumab pegol (Cimzia®) and adalimumab (Humira®), “anti-IL-1” agents such as anakinra (Kineret®) and rilonacept (Arcalyst®), canakinumab (Ilaris®), anti-Jak inhibitors such as tofacitinib, antibodies such as rituximab (Rituxan®), “anti-T-cell” agents such as abatacept (Orencia®), “anti-IL-6” agents such as tocilizumab (Actemra®), diclofenac, cortisone, hyaluronic acid (Synvisc® or Hyalgan®), monoclonal antibodies such as tanezumab, anticoagulants such as heparin (Calcinparine® or Liquaemin®) and warfarin (Coumadin®), antidiarrheals such as diphenoxylate (Lomotil®) and loperamide (Imodium®), bile acid binding agents such as cholestyramine, alosetron (Lotronex®), lubiprostone (Amitiza®), laxatives such as Milk of Magnesia, polyethylene glycol (MiraLax®), Dulcolax®, Correctol® and Senokot®, anticholinergics or antispasmodics such as dicyclomine (Bentyl®), Singulair®, beta-2 agonists such as albuterol (Ventolin® HFA, Proventil® HFA), levalbuterol (Xopenex®), metaproterenol (Alupent®), pirbuterol acetate (Maxair®), terbutaline sulfate (Brethaire®), salmeterol xinafoate (Serevent®) and formoterol (Foradil®), anticholinergic agents such as ipratropium bromide (Atrovent®) and tiotropium (Spiriva®), inhaled corticosteroids such as beclomethasone dipropionate (Beclovent®, Qvar®, and Vanceril®), triamcinolone acetonide (Azmacort®), mometasone (Asthmanex®), budesonide (Pulmocort®), and flunisolide (Aerobid®), Afviar®, Symbicort®, Dulera®, cromolyn sodium (Intal®), methylxanthines such as theophylline (Theo-Dur®, Theolair®, Slo-bid®, Uniphyl®, Theo-24®) and aminophylline, IgE antibodies such as omalizumab (Xolair®), nucleoside reverse transcriptase inhibitors such as zidovudine (Retrovir®), abacavir (Ziagen®), abacavir/lamivudine (Epzicom®), abacavir/lamivudine/zidovudine (Trizivir®), didanosine (Videx®), emtricitabine (Emtriva®), lamivudine (Epivir®), lamivudine/zidovudine (Combivir®), stavudine (Zerit®), and zalcitabine (Hivid®), non-nucleoside reverse transcriptase inhibitors such as delavirdine (Rescriptor®), efavirenz (Sustiva®), nevairapine (Viramune®) and etravirine (Intelence®), nucleotide reverse transcriptase inhibitors such as tenofovir (Viread®), protease inhibitors such as amprenavir (Agenerase®), atazanavir (Reyataz®), darunavir (Prezista®), fosamprenavir (Lexiva®), indinavir (Crixivan®), lopinavir and ritonavir (Kaletra®), nelfmavir (Viracept®), ritonavir (Norvir®), saquinavir (Fortovase® or Invirase®), and tipranavir (Aptivus®), entry inhibitors such as enfuvirtide (Fuzeon®) and maraviroc (Selzentry®), integrase inhibitors such as raltegravir (Isentress®), doxorubicin (Hydrodaunorubicin®), vincristine (Oncovin®), bortezomib (Velcade®), and dexamethasone (Decadron ®) in combination with lenalidomide (Revlimid ®), or any combination(s) thereof.
[00276] In another embodiment, the present invention provides a method of treating rheumatoid arthritis comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from non-steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, etodolac (Lodine®) and celecoxib, corticosteroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like, sulfasalazine (Azulfidine®), antimalarials such as hydroxychloroquine (Plaquenil®) and chloroquine (Aralen®), methotrexate (Rheumatrex®), gold salts such as gold thioglucose (Solganal®), gold thiomalate (Myochrysine®) and auranofm (Ridaura®), D-penicillamine (Depen® or Cuprimine®), azathioprine (Imuran®), cyclophosphamide (Cytoxan®), chlorambucil (Leukeran®), cyclosporine (Sandimmune®), leflunomide (Arava®) and“anti-TNF” agents such as etanercept (Enbrel®), infliximab (Remicade®), golimumab (Simponi®), certolizumab pegol (Cimzia®) and adalimumab (Humira®),“anti-IL-1” agents such as anakinra (Kineret®) and rilonacept (Arcalyst®), antibodies such as rituximab (Rituxan®),“anti-T-cell” agents such as abatacept (Orencia®) and“anti-IL-6” agents such as tocilizumab (Actemra®).
[00277] In some embodiments, the present invention provides a method of treating osteoarthritis comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from acetaminophen, non steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, etodolac (Lodine®) and celecoxib, diclofenac, cortisone, hyaluronic acid (Synvisc® or Hyalgan®) and monoclonal antibodies such as tanezumab.
[00278] In some embodiments, the present invention provides a method of treating cutaneous lupus erythematosus or systemic lupus erythematosus comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from acetaminophen, non-steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, etodolac (Lodine®) and celecoxib, corticosteroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like, antimalarials such as hydroxychloroquine (Plaquenil®) and chloroquine (Aralen®), cyclophosphamide (Cytoxan®), methotrexate (Rheumatrex®), azathioprine (Imuran®) and anticoagulants such as heparin (Calcinparine® or Liquaemin®) and warfarin (Coumadin®).
[00279] In some embodiments, the present invention provides a method of treating Crohn’s disesase, ulcerative colitis, or inflammatory bowel disease comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from mesalamine (Asacol®) sulfasalazine (Azulfidine®), antidiarrheals such as diphenoxylate (Lomotil®) and loperamide (Imodium®), bile acid binding agents such as cholestyramine, alosetron (Lotronex®), lubiprostone (Amitiza®), laxatives such as Milk of Magnesia, polyethylene glycol (MiraLax®), Dulcolax®, Correctol® and Senokot® and anticholinergics or antispasmodics such as dicyclomine (Bentyl®), anti-TNF therapies, steroids, and antibiotics such as Flagyl or ciprofloxacin.
[00280] In some embodiments, the present invention provides a method of treating asthma comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from Singulair®, beta-2 agonists such as albuterol (Ventolin® HFA, Proventil® HFA), levalbuterol (Xopenex®), metaproterenol (Alupent®), pirbuterol acetate (Maxair®), terbutaline sulfate (Brethaire®), salmeterol xinafoate (Serevent®) and formoterol (Foradil®), anticholinergic agents such as ipratropium bromide (Atrovent®) and tiotropium (Spiriva®), inhaled corticosteroids such as prednisone, prednisolone, beclomethasone dipropionate (Beclovent®, Qvar®, and Vanceril®), triamcinolone acetonide (Azmacort®), mometasone (Asthmanex®), budesonide (Pulmocort®), flunisolide (Aerobid®), Afviar®, Symbicort®, and Dulera®, cromolyn sodium (Intal®), methylxanthines such as theophylline (Theo-Dur®, Theolair®, Slo-bid®, Uniphyl®, Theo-24®) and aminophylline, and IgE antibodies such as omalizumab (Xolair®).
[00281] In some embodiments, the present invention provides a method of treating COPD comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from beta-2 agonists such as albuterol (Ventolin® HFA, Proventil® HFA), levalbuterol (Xopenex®), metaproterenol (Alupent®), pirbuterol acetate (Maxair®), terbutaline sulfate (Brethaire®), salmeterol xinafoate (Serevent®) and formoterol (Foradil®), anticholinergic agents such as ipratropium bromide (Atrovent®) and tiotropium (Spiriva®), methylxanthines such as theophylline (Theo-Dur®, Theolair®, Slo-bid®, Uniphyl®, Theo-24®) and aminophylline, inhaled corticosteroids such as prednisone, prednisolone, beclomethasone dipropionate (Beclovent®, Qvar®, and Vanceril®), triamcinolone acetonide (Azmacort®), mometasone (Asthmanex®), budesonide (Pulmocort®), flunisolide (Aerobid®), Afviar®, Symbicort®, and Dulera®,
[00282] In another embodiment, the present invention provides a method of treating a hematological malignancy comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from rituximab (Rituxan®), cyclophosphamide (Cytoxan®), doxorubicin (Hydrodaunorubicin®), vincristine (Oncovin®), prednisone, a hedgehog signaling inhibitor, a BTK inhibitor, a JAK/pan-JAK inhibitor, a PI3K inhibitor, a SYK inhibitor, and combinations thereof.
[00283] In another embodiment, the present invention provides a method of treating a solid tumor comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from rituximab (Rituxan®), cyclophosphamide (Cytoxan®), doxorubicin (Hydrodaunorubicin®), vincristine (Oncovin®), prednisone, a hedgehog signaling inhibitor, a BTK inhibitor, a JAK/pan-JAK inhibitor, a PI3K inhibitor, a SYK inhibitor, and combinations thereof.
[00284] In another embodiment, the present invention provides a method of treating a hematological malignancy comprising administering to a patient in need thereof a compound of formula I or formula II, and a Hedgehog (Hh) signaling pathway inhibitor. In some embodiments, the hematological malignancy is DLBCL (Ramirez et al“Defining causative factors contributing in the activation of hedgehog signaling in diffuse large B-cell lymphoma” Leuk. Res. (2012), published online July 17, and incorporated herein by reference in its entirety).
[00285] In another embodiment, the present invention provides a method of treating diffuse large B-cell lymphoma (DLBCL) comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from rituximab (Rituxan®), cyclophosphamide (Cytoxan®), doxorubicin (Hydrodaunorubicin®), vincristine (Oncovin®), prednisone, a hedgehog signaling inhibitor, and combinations thereof.
[00286] In another embodiment, the present invention provides a method of treating multiple myeloma comprising administering to a patient in need thereof a compound of formula I or formula II, and one or more additional therapeutic agents selected from bortezomib (Velcade®), and dexamethasone (Decadron®), a hedgehog signaling inhibitor, a BTK inhibitor, a JAK/pan- JAK inhibitor, a TYK2 inhibitor, a PI3K inhibitor, a SYK inhibitor in combination with lenalidomide (Revlimid®).
[00287] In another embodiment, the present invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I or formula II, and a BTK inhibitor, wherein the disease is selected from inflammatory bowel disease, arthritis, cutaneous lupus erythematosus, systemic lupus erythematosus (SLE), vasculitis, idiopathic thrombocytopenic purpura (ITP), rheumatoid arthritis, psoriatic arthritis, osteoarthritis, Still’s disease, juvenile arthritis, diabetes, myasthenia gravis, Hashimoto’s thyroiditis, Ord’s thyroiditis, Graves’ disease, autoimmune thyroiditis, Sjogren’s syndrome, multiple sclerosis, systemic sclerosis, Lyme neuroborreliosis, Guillain-Barre syndrome, acute disseminated encephalomyelitis, Addison’s disease, opsoclonus-myoclonus syndrome, ankylosing spondylosis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, autoimmune gastritis, pernicious anemia, celiac disease, Goodpasture’s syndrome, idiopathic thrombocytopenic purpura, optic neuritis, scleroderma, primary biliary cirrhosis, Reiter’s syndrome, Takayasu’s arteritis, temporal arteritis, warm autoimmune hemolytic anemia, Wegener’s granulomatosis, psoriasis, alopecia universalis, Behcet’s disease, chronic fatigue, dysautonomia, membranous glomerulonephropathy, endometriosis, interstitial cystitis, pemphigus vulgaris, bullous pemphigoid, neuromyotonia, scleroderma, vulvodynia, a hyperproliferative disease, rejection of transplanted organs or tissues, Acquired Immunodeficiency Syndrome (AIDS, also known as HIV), type 1 diabetes, graft versus host disease, transplantation, transfusion, anaphylaxis, allergies (e.g., allergies to plant pollens, latex, drugs, foods, insect poisons, animal hair, animal dander, dust mites, or cockroach calyx), type I hypersensitivity, allergic conjunctivitis, allergic rhinitis, and atopic dermatitis, asthma, appendicitis, atopic dermatitis, asthma, allergy, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, cholecystitis, chronic graft rejection, colitis, conjunctivitis, Crohn’s disease, cystitis, dacryoadenitis, dermatitis, dermatomyositis, encephalitis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, Henoch- Schonlein purpura, hepatitis, hidradenitis suppurativa, immunoglobulin A nephropathy, interstitial lung disease, laryngitis, mastitis, meningitis, myelitis myocarditis, myositis, nephritis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumonitis, pneumonia, polymyositis, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendonitis, tonsillitis, ulcerative colitis, uveitis, vaginitis, vasculitis, or vulvitis, B-cell proliferative disorder, e.g., diffuse large B cell lymphoma, follicular lymphoma, chronic lymphocytic lymphoma, chronic lymphocytic leukemia, acute lymphocytic leukemia, B-cell prolymphocytic leukemia, lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia, splenic marginal zone lymphoma, multiple myeloma (also known as plasma cell myeloma), non-Hodgkin’s lymphoma, Hodgkin’s lymphoma, plasmacytoma, extranodal marginal zone B cell lymphoma, nodal marginal zone B cell lymphoma, mantle cell lymphoma, mediastinal (thymic) large B cell lymphoma, intravascular large B cell lymphoma, primary effusion lymphoma, Burkitt lymphoma/leukemia, or lymphomatoid granulomatosis, breast cancer, prostate cancer, or cancer of the mast cells (e.g., mastocytoma, mast cell leukemia, mast cell sarcoma, systemic mastocytosis), bone cancer, colorectal cancer, pancreatic cancer, diseases of the bone and joints including, without limitation, rheumatoid arthritis, seronegative spondyloarthropathies (including ankylosing spondylitis, psoriatic arthritis and Reiter’s disease), Behcet’s disease, Sjogren’s syndrome, systemic sclerosis, osteoporosis, bone cancer, bone metastasis, a thromboembolic disorder, (e.g., myocardial infarct, angina pectoris, reocclusion after angioplasty, restenosis after angioplasty, reocclusion after aortocoronary bypass, restenosis after aortocoronary bypass, stroke, transitory ischemia, a peripheral arterial occlusive disorder, pulmonary embolism, deep venous thrombosis), inflammatory pelvic disease, urethritis, skin sunburn, sinusitis, pneumonitis, encephalitis, meningitis, myocarditis, nephritis, osteomyelitis, myositis, hepatitis, gastritis, enteritis, dermatitis, gingivitis, appendicitis, pancreatitis, cholocystitus, agammaglobulinemia, psoriasis, allergy, Crohn’s disease, irritable bowel syndrome, ulcerative colitis, Sjogren’s disease, tissue graft rejection, hyperacute rejection of transplanted organs, asthma, allergic rhinitis, chronic obstructive pulmonary disease (COPD), autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome), autoimmune alopecia, pernicious anemia, glomerulonephritis, dermatomyositis, multiple sclerosis, scleroderma, vasculitis, autoimmune hemolytic and thrombocytopenic states, Goodpasture’s syndrome, atherosclerosis, Addison’s disease, Parkinson’s disease, Alzheimer’s disease, diabetes, septic shock, cutaneous lupus erythematosus, systemic lupus erythematosus (SLE), rheumatoid arthritis, psoriatic arthritis, juvenile arthritis, osteoarthritis, chronic idiopathic thrombocytopenic purpura, Waldenstrom macroglobulinemia, myasthenia gravis, Hashimoto’s thyroiditis, atopic dermatitis, degenerative joint disease, vitiligo, autoimmune hypopituitarism, Guillain-Barre syndrome, Behcet’s disease, scleraderma, mycosis fungoides, acute inflammatory responses (such as acute respiratory distress syndrome and ischemia/reperfusion injury), and Graves’ disease. [00288] In another embodiment, the present invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I or formula II, and a PI3K inhibitor, wherein the disease is selected from a cancer, a neurodegenative disorder, an angiogenic disorder, a viral disease, an autoimmune disease, an inflammatory disorder, a hormone-related disease, conditions associated with organ transplantation, immunodeficiency disorders, a destructive bone disorder, a proliferative disorder, an infectious disease, a condition associated with cell death, thrombin-induced platelet aggregation, chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), liver disease, pathologic immune conditions involving T cell activation, a cardiovascular disorder, and a CNS disorder.
[00289] In another embodiment, the present invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I or formula II, and a PI3K inhibitor, wherein the disease is selected from benign or malignant tumor, carcinoma or solid tumor of the brain, kidney (e.g., renal cell carcinoma (RCC)), liver, adrenal gland, bladder, breast, stomach, gastric tumors, ovaries, colon, rectum, prostate, pancreas, lung, vagina, endometrium, cervix, testis, genitourinary tract, esophagus, larynx, skin, bone or thyroid, sarcoma, glioblastomas, neuroblastomas, multiple myeloma or gastrointestinal cancer, especially colon carcinoma or colorectal adenoma or a tumor of the neck and head, an epidermal hyperproliferation, psoriasis, prostate hyperplasia, a neoplasia, a neoplasia of epithelial character, adenoma, adenocarcinoma, keratoacanthoma, epidermoid carcinoma, large cell carcinoma, non-small-cell lung carcinoma, lymphomas, (including, for example, non-Hodgkin’s Lymphoma (NHL) and Hodgkin’ s lymphoma (also termed Hodgkin’s or Hodgkin’s disease)), a mammary carcinoma, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, or a leukemia, diseases include Cowden syndrome, Lhermitte-Dudos disease and Bannayan-Zonana syndrome, or diseases in which the PI3K/PKB pathway is aberrantly activated, asthma of whatever type or genesis including both intrinsic (non- allergic) asthma and extrinsic (allergic) asthma, mild asthma, moderate asthma, severe asthma, bronchitic asthma, exercise-induced asthma, occupational asthma and asthma induced following bacterial infection, acute lung injury (ALI), adult/acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary, airways or lung disease (COPD, COAD or COLD), including chronic bronchitis or dyspnea associated therewith, emphysema, as well as exacerbation of airways hyperreactivity consequent to other drug therapy, in particular other inhaled drug therapy, bronchitis of whatever type or genesis including, but not limited to, acute, arachidic, catarrhal, croupus, chronic or phthinoid bronchitis, pneumoconiosis (an inflammatory, commonly occupational, disease of the lungs, frequently accompanied by airways obstruction, whether chronic or acute, and occasioned by repeated inhalation of dusts) of whatever type or genesis, including, for example, aluminosis, anthracosis, asbestosis, chalicosis, ptilosis, siderosis, silicosis, tabacosis and byssinosis, Loffler's syndrome, eosinophilic, pneumonia, parasitic (in particular metazoan) infestation (including tropical eosinophilia), bronchopulmonary aspergillosis, polyarteritis nodosa (including Churg-Strauss syndrome), eosinophilic granuloma and eosinophil- related disorders affecting the airways occasioned by drug-reaction, psoriasis, contact dermatitis, atopic dermatitis, alopecia areata, erythema multiforma, dermatitis herpetiformis, scleroderma, vitiligo, hypersensitivity angiitis, urticaria, bullous pemphigoid, lupus erythematosus, pemphisus, epidermolysis bullosa acquisita, conjunctivitis, keratoconjunctivitis sicca, and vernal conjunctivitis, diseases affecting the nose including allergic rhinitis, and inflammatory disease in which autoimmune reactions are implicated or having an autoimmune component or etiology, including autoimmune hematological disorders (e.g. hemolytic anemia, aplastic anemia, pure red cell anemia and idiopathic thrombocytopenia), cutaneous lupus erythematosus, systemic lupus erythematosus, rheumatoid arthritis, polychondritis, sclerodoma, Wegener granulamatosis, dermatomyositis, chronic active hepatitis, myasthenia gravis, Steven-Johnson syndrome, idiopathic sprue, autoimmune inflammatory bowel disease (e.g. ulcerative colitis and Crohn's disease), endocrine opthalmopathy, Grave's disease, sarcoidosis, alveolitis, chronic hypersensitivity pneumonitis, multiple sclerosis, pri ary biliary cirrhosis, uveitis (anterior and posterior), keratoconjunctivitis sicca and vernal keratoconjunctivitis, interstitial lung fibrosis, psoriatic arthritis and glomerulonephritis (with and without nephrotic syndrome, e.g. including idiopathic nephrotic syndrome or minal change nephropathy, restenosis, cardiomegaly, atherosclerosis, myocardial infarction, ischemic stroke and congestive heart failure, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and cerebral ischemia, and neurodegenerative disease caused by traumatic injury, glutamate neurotoxicity and hypoxia. [00290] In some embodiments the present invention provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I or formula II, and a Bcl-2 inhibitor, wherein the disease is an inflammatory disorder, an autoimmune disorder, a proliferative disorder, an endocrine disorder, a neurological disorder, or a disorder associated with transplantation. In some embodiments, the disorder is a proliferative disorder, lupus, or lupus nephritis. In some embodiments, the proliferative disorder is chronic lymphocytic leukemia, diffuse large B-cell lymphoma, Hodgkin’s disease, small-cell lung cancer, non-small-cell lung cancer, myelodysplastic syndrome, lymphoma, a hematological neoplasm, or solid tumor.
[00291] In some embodiments, the disease is an autoimmune disorder, an inflammatory disorder, a proliferative disorder, an endocrine disorder, a neurological disorder, or a disorder associated with transplantation.
[00292] The compounds and compositions, according to the method of the present invention, may be administered using any amount and any route of administration effective for treating or lessening the severity of an autoimmune disorder, an inflammatory disorder, a proliferative disorder, an endocrine disorder, a neurological disorder, or a disorder associated with transplantation. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like. Compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression "dosage unit form" as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts. The term“patient”, as used herein, means an animal, preferably a mammal, and most preferably a human.
[00293] Pharmaceutically acceptable compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated. In certain embodiments, the compounds of the invention may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
[00294] Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
[00295] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables. [00296] Injectable formulations can be sterilized, for example, by filtration through a bacterial- retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
[00297] In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide- polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
[00298] Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
[00299] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.
[00300] Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
[00301] The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
[00302] Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preserv atives or buffers as may be required. Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
[00303] According to one embodiment, the invention relates to a method of inhibiting protein kinase activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.
[00304] According to another embodiment, the invention relates to a method of activating STING, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound. In certain embodiments, the invention relates to a method of irreversibly inhibiting STING, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.
[00305] The term“biological sample”, as used herein, includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
[00306] Activation of STING (or a mutant thereof) activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ-transplantation, biological specimen storage, and biological assays.
[00307] Another embodiment of the present invention relates to a method of activating STING in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound.
[00308] According to another embodiment, the invention relates to a method of activating STING, or a mutant thereof, in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound. According to certain embodiments, the invention relates to a method of reversibly or irreversibly inhibiting one or more of STING, or a mutant thereof, activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound. In other embodiments, the present invention provides a method for treating a disorder mediated by STING, or a mutant thereof, in a patient in need thereof, comprising the step of administering to said patient a compound according to the present invention or pharmaceutically acceptable composition thereof. Such disorders are described in detail herein.
[00309] Depending upon the particular condition, or disease, to be treated, additional therapeutic agents that are normally administered to treat that condition, may also be present in the compositions of this invention. As used herein, additional therapeutic agents that are normally administered to treat a particular disease, or condition, are known as“appropriate for the disease, or condition, being treated.”
[00310] A compound of the current invention may also be used to advantage in combination with other therapeutic compounds. In some embodiments, the other therapeutic compounds are antiproliferative compounds. Such antiproliferative compounds include, but are not limited to aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule active compounds; alkylating compounds; histone deacetylase inhibitors; compounds which induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibitors; mTOR inhibitors; anti neoplastic antimetabolites; platin compounds; compounds targeting/decreasing a protein or lipid kinase activity and further anti-angiogenic compounds; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; anti androgens; methionine aminopeptidase inhibitors; matrix metalloproteinase inhibitors; bisphosphonates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; proteasome inhibitors; compounds used in the treatment of hematologic malignancies; compounds which target, decrease or inhibit the activity of Flt-3; Hsp90 inhibitors such as 17-AAG (17-allylaminogeldanamycin, NSC330507), 17 -DMAG ( 17-dimethyl aminoethyl amino- 17 -demethoxy-geldanamycin,
NSC707545), IPI-504, CNFIOIO, CNF2024, CNFIOIO from Conforma Therapeutics; temozolomide (Temodal®); kinesin spindle protein inhibitors, such as SB715992 or SB743921 from GlaxoSmithKline, or pentamidine/chlorpromazine from CombinatoRx; MEK inhibitors such as ARRY142886 from Array BioPharma, AZD6244 from AstraZeneca, PD181461 from Pfizer and leucovorin. The term "aromatase inhibitor" as used herein relates to a compound which inhibits estrogen production, for instance, the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively. The term includes, but is not limited to steroids, especially atamestane, exemestane and formestane and, in particular, non-steroids, especially aminoglutethimide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketokonazole, vorozole, fadrozole, anastrozole and letrozole. Exemestane is marketed under the trade name Aromasin™. Formestane is marketed under the trade name Lentaron™. Fadrozole is marketed under the trade name Afema™. Anastrozole is marketed under the trade name Arimidex™. Letrozole is marketed under the trade names Femara™ or Femar™. Aminoglutethimide is marketed under the trade name Orimeten™ A combination of the invention comprising a chemotherapeutic agent which is an aromatase inhibitor is particularly useful for the treatment of hormone receptor positive tumors, such as breast tumors.
[00311] The term "antiestrogen" as used herein relates to a compound which antagonizes the effect of estrogens at the estrogen receptor level. The term includes, but is not limited to tamoxifen, fulvestrant, raloxifene and raloxifene hydrochloride. Tamoxifen is marketed under the trade name Nolvadex™. Raloxifene hydrochloride is marketed under the trade name Evista™. Fulvestrant can be administered under the trade name Faslodex™. A combination of the invention comprising a chemotherapeutic agent which is an antiestrogen is particularly useful for the treatment of estrogen receptor positive tumors, such as breast tumors.
[00312] The term "anti -androgen" as used herein relates to any substance which is capable of inhibiting the biological effects of androgenic hormones and includes, but is not limited to, bicalutamide (Casodex™). The term "gonadorelin agonist" as used herein includes, but is not limited to abarelix, goserelin and goserelin acetate. Goserelin can be administered under the trade name Zoladex™.
[00313] The term "topoisom erase I inhibitor" as used herein includes, but is not limited to topotecan, gimatecan, irinotecan, camptothecian and its analogues, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148. Irinotecan can be administered, e.g. in the form as it is marketed, e.g. under the trademark Camptosar™. Topotecan is marketed under the trade name Hycamptin™. [00314] The term "topoisomerase II inhibitor" as used herein includes, but is not limited to the anthracyclines such as doxorubicin (including liposomal formulation, such as Caelyx™), daunorubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones mitoxantrone and losoxantrone, and the podophillotoxines etoposide and teniposide. Etoposide is marketed under the trade name Etopophos™ Teniposide is marketed under the trade name VM 26-Bristol Doxorubicin is marketed under the trade name Acriblastin™ or Adriamycin™ Epirubicin is marketed under the trade name Farmorubicin™. Idarubicin is marketed under the trade name Zavedos™. Mitoxantrone is marketed under the trade name Novantron.
[00315] The term "microtubule active agent" relates to microtubule stabilizing, microtubule destabilizing compounds and microtublin polymerization inhibitors including, but not limited to taxanes, such as paclitaxel and docetaxel; vinca alkaloids, such as vinblastine or vinblastine sulfate, vincristine or vincristine sulfate, and vinorelbine; discodermolides; cochicine and epothilones and derivatives thereof. Paclitaxel is marketed under the trade name Taxol™. Docetaxel is marketed under the trade name Taxotere™. Vinblastine sulfate is marketed under the trade name Vinblastin R.P™. Vincristine sulfate is marketed under the trade name Farmistin™.
[00316] The term "alkylating agent" as used herein includes, but is not limited to, cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel). Cyclophosphamide is marketed under the trade name Cyclostin™. Ifosfamide is marketed under the trade name Holoxan™.
[00317] The term "histone deacetylase inhibitors" or "HD AC inhibitors" relates to compounds which inhibit the histone deacetylase and which possess antiproliferative activity. This includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).
[00318] The term "antineoplastic antimetabolite" includes, but is not limited to, 5-fluorouracil or 5-FU, capecitabine, gemcitabine, DNA demethylating compounds, such as 5-azacytidine and decitabine, methotrexate and edatrexate, and folic acid antagonists such as pemetrexed. Capecitabine is marketed under the trade name Xeloda™. Gemcitabine is marketed under the trade name Gemzar™.
[00319] The term "platin compound" as used herein includes, but is not limited to, carboplatin, cis-platin, cisplatinum and oxaliplatin. Carboplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark Carboplat™. Oxaliplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark Eloxatin™.
[00320] The term "compounds targeting/decreasing a protein or lipid kinase activity; or a protein or lipid phosphatase activity; or further anti-angiogenic compounds" as used herein includes, but is not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, such as a) compounds targeting, decreasing or inhibiting the activity of the platelet-derived growth factor-receptors (PDGFR), such as compounds which target, decrease or inhibit the activity of PDGFR, especially compounds which inhibit the PDGF receptor, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib, SU101, SU6668 and GFB- 111; b) compounds targeting, decreasing or inhibiting the activity of the fibroblast growth factor- receptors (FGFR); c) compounds targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor I (IGF-IR), such as compounds which target, decrease or inhibit the activity of IGF-IR, especially compounds which inhibit the kinase activity of IGF-I receptor, or antibodies that target the extracellular domain of IGF-I receptor or its growth factors; d) compounds targeting, decreasing or inhibiting the activity of the Trk receptor tyrosine kinase family, or ephrin B4 inhibitors; e) compounds targeting, decreasing or inhibiting the activity of the Axl receptor tyrosine kinase family; f) compounds targeting, decreasing or inhibiting the activity of the Ret receptor tyrosine kinase; g) compounds targeting, decreasing or inhibiting the activity of the Kit/SCFR receptor tyrosine kinase, such as imatinib; h) compounds targeting, decreasing or inhibiting the activity of the C-kit receptor tyrosine kinases, which are part of the PDGFR family, such as compounds which target, decrease or inhibit the activity of the c-Kit receptor tyrosine kinase family, especially compounds which inhibit the c-Kit receptor, such as imatinib; i) compounds targeting, decreasing or inhibiting the activity of members of the c-Abl family, their gene-fusion products (e.g. BCR-Abl kinase) and mutants, such as compounds which target decrease or inhibit the activity of c-Abl family members and their gene fusion products, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib or nilotinib (AMN107); PD180970; AG957; NSC 680410; PD173955 from ParkeDavis; or dasatinib (BMS-354825); j) compounds targeting, decreasing or inhibiting the activity of members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK, SRC, JAK/pan-JAK, FAK, PDK1, PKB/Akt, Ras/MAPK, PI3K, SYK, BTK and TEC family, and/or members of the cyclin-
175
5U B5TITUTE SH EET (RU LE 26) dependent kinase family (CDK) including staurosporine derivatives, such as midostaurin; examples of further compounds include UCN-01, safmgol, BAY 43-9006, Bryostatin 1, Perifosine; llmofosine; RO 318220 and RO 320432; GO 6976; Isis 3521; LY333531/LY379196; isochinoline compounds; FTIs; PD184352 or QAN697 (a P13K inhibitor) or AT7519 (CDK inhibitor); k) compounds targeting, decreasing or inhibiting the activity of protein-tyrosine kinase inhibitors, such as compounds which target, decrease or inhibit the activity of protein-tyrosine kinase inhibitors include imatinib mesylate (Gleevec™) or tyrphostin such as Tyrphostin A23/RG- 50810; AG 99; Tyrphostin AG 213; Tyrphostin AG 1748; Tyrphostin AG 490; Tyrphostin B44; Tyrphostin B44 (+) enantiomer; Tyrphostin AG 555; AG 494; Tyrphostin AG 556, AG957 and adaphostin (4-{[(2,5- dihydroxyphenyl)methyl]amino}-benzoic acid adamantyl ester; NSC 680410, adaphostin); 1) compounds targeting, decreasing or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases (EGFRi ErbB2, ErbB3, ErbB4 as homo- or heterodimers) and their mutants, such as compounds which target, decrease or inhibit the activity of the epidermal growth factor receptor family are especially compounds, proteins or antibodies which inhibit members of the EGF receptor tyrosine kinase family, such as EGF receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, CP 358774, ZD 1839, ZM 105180; trastuzumab (Herceptin™), cetuximab (Erbitux™), Iressa, Tarceva, OSI-774, Cl-1033, EKB-569, GW-2016, El. l, E2.4, E2.5, E6.2, E6.4, E2.11, E6.3 or E7.6.3, and 7H-pyrrolo-[2,3-d]pyrimidine derivatives; m) compounds targeting, decreasing or inhibiting the activity of the c-Met receptor, such as compounds which target, decrease or inhibit the activity of c-Met, especially compounds which inhibit the kinase activity of c-Met receptor, or antibodies that target the extracellular domain of c-Met or bind to HGF, n) compounds targeting, decreasing or inhibiting the kinase activity of one or more JAK family members (JAK1/JAK2/JAK3/TYK2 and/or pan-JAK), including but not limited to PRT-062070, SB-1578, baricitinib, pacritinib, momelotinib, VX-509, AZD-1480, TG-101348, tofacitinib, and ruxolitinib; o) compounds targeting, decreasing or inhibiting the kinase activity of PI3 kinase (PI3K) including but not limited to ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib, pictrelisib, PF-4691502, BYL-719, dactolisib, XL-147, XL-765, and idelalisib; and; and q) compounds targeting, decreasing or inhibiting the signaling effects of hedgehog protein (Hh) or smoothened receptor (SMO) pathways, including but not limited to cyclopamine, vismodegib, itraconazole, erismodegib, and IPI-926 (saridegib).
[00321] The term“PI3K inhibitor” as used herein includes, but is not limited to compounds having inhibitory activity against one or more enzymes in the phosphatidylinositol-3 -kinase family, including, but not limited to RBKa, RI3Kg, PI3K5, RI3Kb, PI3K-C2a, PI3K-C2p, PI3K- C2y, Vps34, pi 10-a, pi 10-b, p 110-g, pi 10-d, p85-a, r85-b, r55-g, pi 50, plOl, and p87. Examples of PI3K inhibitors useful in this invention include but are not limited to ATU-027, SF-1126, DS- 7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib, pictrelisib, PF-4691502, BYL-719, dactolisib, XL-147, XL-765, and idelalisib.
[00322] The term“BTK inhibitor” as used herein includes, but is not limited to compounds having inhibitory activity against Bruton’s Tyrosine Kinase (BTK), including, but not limited to AVL-292 and ibrutinib.
[00323] The term“SYK inhibitor” as used herein includes, but is not limited to compounds having inhibitory activity against spleen tyrosine kinase (SYK), including but not limited to PRT- 062070, R-343, R-333, Excellair, PRT-062607, and fostamatinib.
[00324] The term“Bcl-2 inhibitor” as used herein includes, but is not limited to compounds having inhibitory activity against B-cell lymphoma 2 protein (Bcl-2), including but not limited to ABT-199, ABT-731, ABT-737, apogossypol, Ascenta’s pan-Bcl-2 inhibitors, curcumin (and analogs thereof), dual Bcl-2/Bcl-xL inhibitors (Infinity Pharmaceuticals/Novartis Pharmaceuticals), Genasense (G3139), HA14-1 (and analogs thereof; see W02008118802), navitoclax (and analogs thereof, see US7390799), NH-1 (Shenayng Pharmaceutical University), obatoclax (and analogs thereof, see W02004106328), S-001 (Gloria Pharmaceuticals), TW series compounds (Univ. of Michigan), and venetoclax. In some embodiments the Bcl-2 inhibitor is a small molecule therapeutic. In some embodiments the Bcl-2 inhibitor is a peptidomimetic.
[00325] Further examples of BTK inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this invention can be found in W02008039218 and WO2011090760, the entirety of which are incorporated herein by reference.
[00326] Further examples of SYK inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this invention can be found in W02003063794, W02005007623, and W02006078846, the entirety of which are incorporated herein by reference. [00327] Further examples of PI3K inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this invention can be found in W02004019973, W02004089925, W02007016176, US8138347, W02002088112, W02007084786,
W02007129161, W02006122806, W02005113554, and W02007044729 the entirety of which are incorporated herein by reference.
[00328] Further examples of JAK inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this invention can be found in W02009114512, W02008109943, W02007053452, W02000142246, and W02007070514, the entirety of which are incorporated herein by reference.
[00329] Further anti-angiogenic compounds include compounds having another mechanism for their activity, e.g. unrelated to protein or lipid kinase inhibition e.g. thalidomide (Thalomid™) and TNP-470.
[00330] Examples of proteasome inhibitors useful for use in combination with compounds of the invention include, but are not limited to bortezomib, disulfiram, epigallocatechin-3-gallate (EGCG), salinosporamide A, carfilzomib, ONX-0912, CEP-18770, and MLN9708.
[00331] Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase are e.g. inhibitors of phosphatase 1, phosphatase 2A, or CDC25, such as okadaic acid or a derivative thereof.
[00332] Compounds which induce cell differentiation processes include, but are not limited to, retinoic acid, a- g- or d- tocopherol or a- g- or d-tocotrienol.
[00333] The term cyclooxygenase inhibitor as used herein includes, but is not limited to, Cox- 2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib (Celebrex™), rofecoxib (Vioxx™), etoricoxib, valdecoxib or a 5-alkyl-2- arylaminophenylacetic acid, such as 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenyl acetic acid, lumiracoxib.
[00334] The term "bisphosphonates" as used herein includes, but is not limited to, etridonic, clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and zoledronic acid. Etridonic acid is marketed under the trade name Didronel™. Clodronic acid is marketed under the trade name Bonefos™. Tiludronic acid is marketed under the trade name Skelid™. Pamidronic acid is marketed under the trade name Aredia™. Alendronic acid is marketed under the trade name Fosamax™. Ibandronic acid is marketed under the trade name Bondranat™. Risedronic acid is marketed under the trade name Actonel™. Zoledronic acid is marketed under the trade name Zometa™. The term "mTOR inhibitors" relates to compounds which inhibit the mammalian target of rapamycin (mTOR) and which possess antiproliferative activity such as sirolimus (Rapamune®), everolimus (Certican™), CCI-779 and ABT578.
[00335] The term "heparanase inhibitor" as used herein refers to compounds which target, decrease or inhibit heparin sulfate degradation. The term includes, but is not limited to, PI-88. The term "biological response modifier" as used herein refers to a lymphokine or interferons.
[00336] The term "inhibitor of Ras oncogenic isoforms", such as H-Ras, K-Ras, or N-Ras, as used herein refers to compounds which target, decrease or inhibit the oncogenic activity of Ras; for example, a "famesyl transferase inhibitor" such as L-744832, DK8G557 or R115777 (Zamestra™). The term "telomerase inhibitor" as used herein refers to compounds which target, decrease or inhibit the activity of telomerase. Compounds which target, decrease or inhibit the activity of telomerase are especially compounds which inhibit the telomerase receptor, such as telomestatin.
[00337] The term "methionine aminopeptidase inhibitor" as used herein refers to compounds which target, decrease or inhibit the activity of methionine aminopeptidase. Compounds which target, decrease or inhibit the activity of methionine aminopeptidase include, but are not limited to, bengamide or a derivative thereof.
[00338] The term "proteasome inhibitor" as used herein refers to compounds which target, decrease or inhibit the activity of the proteasome. Compounds which target, decrease or inhibit the activity of the proteasome include, but are not limited to, Bortezomib (Velcade™) and MLN 341.
[00339] The term "matrix metalloproteinase inhibitor" or ("MMP" inhibitor) as used herein includes, but is not limited to, collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, e.g. hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat (BB-2516), prinomastat (AG3340), metastat (NSC 683551) BMS-279251, BAY 12-9566, TAA211, MMI270B or AAJ996.
[00340] The term "compounds used in the treatment of hematologic malignancies" as used herein includes, but is not limited to, FMS-like tyrosine kinase inhibitors, which are compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, I-b-D-arabinofuransyl cytosine (ara-c) and bisulfan; ALK inhibitors, which are compounds which target, decrease or inhibit anaplastic lymphoma kinase, and Bcl-2 inhibitors.
[00341] Compounds which target, decrease or inhibit the activity of FMS-like tyrosine kinase receptors (Flt-3R) are especially compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, such as PKC412, midostaurin, a staurosporine derivative, SU11248 and MLN518.
[00342] The term "HSP90 inhibitors" as used herein includes, but is not limited to, compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90; degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteosome pathway. Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90 are especially compounds, proteins or antibodies which inhibit the ATPase activity of HSP90, such as 17-allylamino,17-demethoxygeldanamycin (17AAG), a geldanamycin derivative; other geldanamycin related compounds; radicicol and HDAC inhibitors.
[00343] The term "antiproliferative antibodies" as used herein includes, but is not limited to, trastuzumab (Herceptin™), Trastuzumab-DMl, erbitux, bevacizumab (Avastin™), rituximab (Rituxan®), PR064553 (anti-CD40) and 2C4 Antibody. By antibodies is meant intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibodies fragments so long as they exhibit the desired biological activity.
[00344] For the treatment of acute myeloid leukemia (AML), compounds of the current invention can be used in combination with standard leukemia therapies, especially in combination with therapies used for the treatment of AML. In particular, compounds of the current invention can be administered in combination with, for example, farnesyl transferase inhibitors and/or other drugs useful for the treatment of AML, such as Daunorubicin, Adriamycin, Ara-C, VP- 16, Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412. In some embodiments, the present invention provides a method of treating AML associated with an ITD and/or D835Y mutation, comprising administering a compound of the present invention together with a one or more FLT3 inhibitors. In some embodiments, the FLT3 inhibitors are selected from quizartinib (AC220), a staurosporine derivative (e.g. midostaurin or lestaurtinib), sorafenib, tandutinib, LY-2401401, LS-104, EB-10, famitinib, NOV-110302, NMS-P948, AST-487, G-749, SB-1317, S-209, SC-110219, AKN-028, fedratinib, tozasertib, and sunitinib. In some embodiments, the FLT3 inhibitors are selected from quizartinib, midostaurin, lestaurtinib, sorafenib, and sunitinib.
[00345] Other anti-leukemic compounds include, for example, Ara-C, a pyrimidine analog, which is the 2 -alpha-hydroxy ribose (arabinoside) derivative of deoxycytidine. Also included is the purine analog of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine phosphate. Compounds which target, decrease or inhibit activity of histone deacetylase (HD AC) inhibitors such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA) inhibit the activity of the enzymes known as histone deacetylases. Specific HDAC inhibitors include MS275, SAHA, FK228 (formerly FR901228), Trichostatin A and compounds disclosed in US 6,552,065 including, but not limited to, N-hydroxy-3-[4-[[[2-(2-methyl-lH-indol-3-yl)-ethyl]- amino]methyl]phenyl]- 2E-2-propenamide, or a pharmaceutically acceptable salt thereof and N-hydroxy-3-[4-[(2- hydroxyethyl){2-(lH-indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2- propenamide, or a pharmaceutically acceptable salt thereof, especially the lactate salt. Somatostatin receptor antagonists as used herein refer to compounds which target, treat or inhibit the somatostatin receptor such as octreotide, and SOM230. Tumor cell damaging approaches refer to approaches such as ionizing radiation. The term "ionizing radiation" referred to above and hereinafter means ionizing radiation that occurs as either electromagnetic rays (such as X-rays and gamma rays) or particles (such as alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiation therapy and is known in the art. See Heilman, Principles of Radiation Therapy, Cancer, in Principles and Practice of Oncology, Devita et ah, Eds., 4th Edition, Vol. 1, pp. 248-275 (1993).
[00346] Also included are EDG binders and ribonucleotide reductase inhibitors. The term “EDG binders” as used herein refers to a class of immunosuppressants that modulates lymphocyte recirculation, such as FTY720. The term“ribonucleotide reductase inhibitors” refers to pyrimidine or purine nucleoside analogs including, but not limited to, fludarabine and/or cytosine arabinoside (ara-C), 6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine (especially in combination with ara-C against ALL) and/or pentostatin. Ribonucleotide reductase inhibitors are especially hydroxyurea or 2-hydroxy-lH-isoindole-l ,3-dione derivatives.
[00347] Also included are in particular those compounds, proteins or monoclonal antibodies of VEGF such as l-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, l-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine succinate; Angiostatin™; Endostatin™; anthranilic acid amides; ZD4190; ZD6474; SU5416; SU6668; bevacizumab; or anti-VEGF antibodies or anti-VEGF receptor antibodies, such as rhuMAb and RHEIFab, VEGF aptamer such as Macugon; FLT-4 inhibitors, FLT-3 inhibitors, VEGFR-2 IgGI antibody, Angiozyme (RPI 4610) and Bevacizumab (Avastin™).
[00348] Photodynamic therapy as used herein refers to therapy which uses certain chemicals known as photosensitizing compounds to treat or prevent cancers. Examples of photodynamic therapy include treatment with compounds, such as Visudyne™ and porfimer sodium.
[00349] Angiostatic steroids as used herein refers to compounds which block or inhibit angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11-a-epihydrocotisol, cortexolone, 17a-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.
[00350] Implants containing corticosteroids refers to compounds, such as fluocinolone and dexamethasone.
[00351] Other chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or miscellaneous compounds or compounds with other or unknown mechanism of action.
[00352] The compounds of the invention are also useful as co-therapeutic compounds for use in combination with other drug substances such as anti-inflammatory, bronchodilatory or antihistamine drug substances, particularly in the treatment of obstructive or inflammatory airways diseases such as those mentioned hereinbefore, for example as potentiators of therapeutic activity of such drugs or as a means of reducing required dosaging or potential side effects of such drugs. A compound of the invention may be mixed with the other drug substance in a fixed pharmaceutical composition or it may be administered separately, before, simultaneously with or after the other drug substance. Accordingly the invention includes a combination of a compound of the invention as hereinbefore described with an anti-inflammatory, bronchodilatory, antihistamine or anti-tussive drug substance, said compound of the invention and said drug substance being in the same or different pharmaceutical composition.
[00353] Suitable anti-inflammatory drugs include steroids, in particular glucocorticosteroids such as budesonide, beclamethasone dipropionate, fluticasone propionate, ciclesonide or mometasone furoate; non-steroidal glucocorticoid receptor agonists; LTB4 antagonists such LY293111, CGS025019C, CP-195543, SC-53228, BIIL 284, ONO 4057, SB 209247; LTD4 antagonists such as montelukast and zafirlukast; PDE4 inhibitors such cilomilast (Ariflo® GlaxoSmithKline), Roflumilast (Byk Gulden), V-11294A (Napp), BAY19-8004 (Bayer), SCH- 351591 (Schering- Plough), Arofylline (Almirall Prodesfarma), PD189659 / PD168787 (Parke- Davis), AWD-12- 281 (Asia Medica), CDC-801 (Celgene), SelCID(TM) CC-10004 (Celgene), VM554/UM565 (Vernalis), T-440 (Tanabe), KW-4490 (Kyowa Hakko Kogyo); A2a agonists; A2b antagonists; and beta-2 adrenoceptor agonists such as albuterol (salbutamol), metaproterenol, terbutaline, salmeterol fenoterol, procaterol, and especially, formoterol and pharmaceutically acceptable salts thereof. Suitable bronchodilatory drugs include anticholinergic or antimuscarinic compounds, in particular ipratropium bromide, oxitropium bromide, tiotropium salts and CHF 4226 (Chiesi), and glycopyrrolate.
[00354] Suitable antihistamine drug substances include cetirizine hydrochloride, acetaminophen, clemastine fumarate, promethazine, loratidine, desloratidine, diphenhydramine and fexofenadine hydrochloride, activastine, astemizole, azelastine, ebastine, epinastine, mizolastine and tefenadine.
[00355] Other useful combinations of compounds of the invention with anti-inflammatory drugs are those with antagonists of chemokine receptors, e.g. CCR-1, CCR-2, CCR-3, CCR-4, CCR-5, CCR-6, CCR-7, CCR-8, CCR-9 and CCR10, CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, particularly CCR-5 antagonists such as Schering-Plough antagonists SC-351125, SCH- 55700 and SCH-D, and Takeda antagonists such as N-[[4-[[[6,7-dihydro-2-(4-methylphenyl)-5H- benzo-cyclohepten-8-yl]carbonyl]amino]phenyl]-methyl]tetrahydro-N,N-dimethyl-2H-pyran-4- aminium chloride (TAK-770).
[00356] The structure of the active compounds identified by code numbers, generic or trade names may be taken from the actual edition of the standard compendium "The Merck Index" or from databases, e.g. Patents International (e.g. IMS World Publications).
[00357] A compound of the current invention may also be used in combination with known therapeutic processes, for example, the administration of hormones or radiation. In certain embodiments, a provided compound is used as a radiosensitizer, especially for the treatment of tumors which exhibit poor sensitivity to radiotherapy. [00358] A compound of the current invention can be administered alone or in combination with one or more other therapeutic compounds, possible combination therapy taking the form of fixed combinations or the administration of a compound of the invention and one or more other therapeutic compounds being staggered or given independently of one another, or the combined administration of fixed combinations and one or more other therapeutic compounds. A compound of the current invention can besides or in addition be administered especially for tumor therapy in combination with chemotherapy, radiotherapy, immunotherapy, phototherapy, surgical intervention, or a combination of these. Long-term therapy is equally possible as is adjuvant therapy in the context of other treatment strategies, as described above. Other possible treatments are therapy to maintain the patient's status after tumor regression, or even chemopreventive therapy, for example in patients at risk.
[00359] Those additional agents may be administered separately from an inventive compound- containing composition, as part of a multiple dosage regimen. Alternatively, those agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as part of a multiple dosage regime, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another normally within five hours from one another.
[00360] As used herein, the term“combination,”“combined,” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention. For example, a compound of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form. Accordingly, the present invention provides a single unit dosage form comprising a compound of the current invention, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
[00361] The amount of both an inventive compound and additional therapeutic agent (in those compositions which comprise an additional therapeutic agent as described above) that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Preferably, compositions of this invention should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of an inventive compound can be administered. [00362] In those compositions which comprise an additional therapeutic agent, that additional therapeutic agent and the compound of this invention may act synergistically. Therefore, the amount of additional therapeutic agent in such compositions will be less than that required in a monotherapy utilizing only that therapeutic agent. In such compositions a dosage of between 0.01 - 1,000 pg/kg body weight/day of the additional therapeutic agent can be administered.
[00363] The amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent. Preferably the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.
[00364] The compounds of this invention, or pharmaceutical compositions thereof, may also be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters. Vascular stents, for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury). However, patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a kinase inhibitor. Implantable devices coated with a compound of this invention are another embodiment of the present invention.
EXEMPLIFICATION
[00365] As depicted in the Examples below, in certain exemplary embodiments, compounds are prepared according to the following general procedures. It will be appreciated that, although the general methods depict the synthesis of certain compounds of the present invention, the following general methods, and other methods known to one of ordinary skill in the art, can be applied to all compounds and subclasses and species of each of these compounds, as described herein. EXAMPLE 1. Synthesis of (E)-l-(4-(5-carbamoyl-2-(l /-pyrrole-2-carboxamido)-l /- benzo[d]imidazol-l-yl)but-2-enyl)-7-methyl-2-(l/ -pyrrole-2-carboxamido)-l - benzo [d] imidazole-5-carboxamide, 1-108
Figure imgf000187_0001
Figure imgf000188_0001
[00366] Synthesis of compound 1.1. Into a 100-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of argon, was placed methyl 4-chloro-3-methylbenzoate (10 g, 54.2 mmol, 1.00 eq) and H2SO4 (15.0 mL, 281 mmol, 5.20 eq). The reaction was stirred for 20 min at 0°C in a water/ice bath. To this was added HNO3 (10.0 mL, 223 mmol, 4.12 eq). The reaction was stirred for an additional 60 min while the temperature was maintained at 0°C in a water/ice bath. The reaction was then quenched by the addition of 50 mL of water. The solution was extracted with 3x100 mL of EA and concentrated under vacuum. The residue was applied onto a silica gel column with EA: PA=1 : 10 to yield 11 g (88.4%) of 1.1 as an off-white solid.
[00367] Synthesis of compound 1.2. Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of argon, was placed 1.1 (10 g, 43.6 mmol, 1.0 eq) and NH4OH (100 mL). The reaction was stirred for 15 hr at 30°C in an oil bath. The solids were collected by filtration. The mixture was concentrated under vacuum to yield 8 g (85.6%) of 1.2 as an off-white solid. (ES, m/z): 213/215 (M-H) .
[00368] Synthesis of compound 1.3. Into a 50-mL 3 -necked round-bottom flask purged and maintained with an inert atmosphere of argon, was placed 1.2 (1 g, 4.66 mmol, 1 eq) in DMSO (10 mL), (E)-tert- butyl 4-aminobut-2-enylcarbamate (0.9 g, 4.19 mmol, 1.00 eq) and DIEA (1.8 g, 0.01 mol, 3.00 eq). The reaction was stirred for 15 h at 100°C in an oil bath. Then the reaction was quenched by the addition of 50 mL of water/ice. The solids were collected by filtration to yield 1.1 g (64.8%) of 1.3 as a yellow solid. (ES, m/z): 365 (M+H)+.
[00369] Synthesis of compound 1.4. Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of argon, was placed 1.3 (1.05 g, 2.88 mmol, 1.00 eq) and 1, 4- dioxane/HCl (11 mL, 4 M). The reaction was stirred for 5 h at 20°C. The mixture was concentrated under vacuum to yield 0.8 g (crude) of 1.4 as a red solid. (ES, m/z) 384 (M+H)+.
[00370] Synthesis of compound 1.5. Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of argon, was placed 1.4 (5 g, 23.0 mmol, 1.00 eq) and ammonia (50 mL). The reaction was stirred for 15 h at 30°C in an oil bath. The solids were collected by filtration. The mixture was concentrated under vacuum to yield 4.0 g (86.0%) of 1.5 as an off-white solid. (ES, m/z): 199 (M-H)'.
[00371] Synthesis of compound 1.6. Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of argon, was placed 1.5 (780 mg, 2.95 mmol, 1.00 eq) in DMSO (10 mL), 4-chloro-3-nitrobenzamide (590 mg, 2.95 mmol, 1.00 eq) and DIEA (1.15 g, 8.85 mmol, 3.00 eq). The reaction was stirred for 15 h at 100°C in an oil bath. Then the reaction was quenched by the addition of 50 mL of water/ice. The solids were collected by filtration. The mixture was concentrated under vacuum to yield 750 mg (59.3%) of 1.6 as a yellow solid. (ES, m/z): 429 (M+H)+.
[00372] Synthesis of compound 1.7. Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of argon, was placed 1.6 (730 mg, 1.70 mmol, 1.00 eq) in HOAc (10 mL) and Zn (890 mg, 13.6 mmol, 8.00 eq) was added. The reaction was stirred for 5 h at 20°C. Then the solids were filtered off. The mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography with dichloromethane/methanol (5: 1) to yield 450 mg (71.7%) of 1.7 as a yellow solid. (ES, m/z): 369 (M+H)+.
[00373] Synthesis of compound 1.8. Into a 50 mL round-bottom flask was placed 1.7 (100 mg, 0.27 mmol, 1.0 eq), BrCN (71.9 mg, 0.68 mmol, 2.5 eq) and EtOH (2 mL).
The mixture was stirred for 15 h at 20 °C under N2 atmosphere. The precipitated solids were collected by filtration and washed with MeOH (2 mL). The product was re-crystallized from DMF to afford 1.8 (11.5 mg) as a light pink solid.
[00374] Synthesis of compound 1-108. Into a 8-mL vial purged and maintained with an inert atmosphere of nitrogen, was placed l/7-pyrrole-2-carboxylic acid (132.7 mg, 1.19 mmol, 5 eq) in DMF (4 mL), DIEA (308.9 mg, 2.39 mmol, 10 eq), HO AT (162.6 mg, 1.19 mmol, 5 eq), EDC(185.5 mg, 1.19 mmol, 5 eq), and 1.8 (100 mg, 0.24 mmol, 1 eq). The reaction was stirred overnight at 60° C in an oil bath. Then it was concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions (24SHIMADZU (HPLC-01)): Column, SunFire C18 OBD Prep Column, 19 mm X 250 mm; mobile phase, Water (l%HOAc) and ACN (13% Phase B up to 50% in 10 min); Detector, UV/254nm to yield 1.8 mg (1.25%) of 1-108 as a yellow solid. (ES, m/z): 605 (M+H)+, 603 (M-H) ; lU-NMR (dg-DMSO, 300 MHz , ppm) d 12.55 (s, 2H), 11.30 (s, 2H), 8.10-7.74 (m, 4H), 7.68 (d, 1H), 7.34 (d, 4H), 6.80 (d, 4H), 6.11 (s, 3H), 5.54 (s, 1H), 5.10 (s, 2H), 4.89 (s, 2H), 2.50 (s, 3H).
EXAMPLE 2. Synthesis of E)-l-(4-(5-carbamoyl-2-(l-methyl-l//-pyrrole-2-carboxamido)- l//-benzo[d]imidazol-l-yl)but-2-enyl)-7-methyl-2-(l-methyl-l//-pyrrole-2-carboxamido)- l/ -benzo[< ]imidazole-5-carboxamide, 1-107
Figure imgf000190_0001
1-107 [00375] Synthesis of compound 1-107. Into an 8-mL vial purged and maintained with an inert atmosphere of nitrogen, was placed 1 -methyl- 17/-pyrrole-2-carboxylic acid (149.5 mg, 1.19 mmol, 5 eq) in DMF (4 mL). Then EDC (185.5 mg, 1.19 mmol, 5 eq), HOAT (162.6 mg, 1.19 mmol, 5 eq), DIEA (308.9 mg, 2.39 mmol, 10 eq) and 1.8 (100 mg, 0.24 mmol, 1 eq) were added. The reaction was stirred for 1 hr at 60° C in an oil bath. The reaction was stirred overnight while the temperature was maintained at 60° C in an oil bath. The mixture was concentrated under vacuum. The residue was applied onto a prep TLC with dichloromethane/methanol (10: 1). The crude product was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): Column, SunFire Cl 8 OBD; mobile phase, Water (l%HOAc) and ACN (25% Phase B up to 55% in 9 min); Detector, UV/254nm to yield 3.2 mg (2.12%) of 1-107 as a yellow solid. (ES, m/z): 633 (M+H)+, 631 (M-H)-, H-NMR (DMSO,400MHz. ppm):o 12.55 (s, 2H), 7.86 (d, 4H), 7.68 (d, 1H), 7.53-7.11 (m, 4H), 6.88 (s, 3H), 5.95 (d, 3H), 5.53 (s, 1H), 4.96 (s, 2H), 4.78 (s, 2H), 3.92 (s, 6H).
EXAMPLE 3. Synthesis of (E)-/V-(5-carbamoyl-l-(4-(5-carbainoyl-2-(pyrazolo[l,5- fl]pyridine-2-carboxamido)-l/ -benzo[d]imidazol-l-yl)but-2-enyl)-7-methyl-lH- benzo [d] imidazol-2-yl)pyrazolo [1 ,5- ] pyridine-2-carboxamide, 1-104
Figure imgf000191_0001
[00376] Synthesis of compound 1-104. To a stirred mixture of 1.8 (100 mg, 0.24 mmol, 1 eq) and pyrazolo[l,5-a]pyridine-2-carboxylic acid (193.7 mg, 1.19 mmol, 5 eq) in DMF (4 mL) were added HOAT (162.6 mg, 1.19 mmol, 5 eq) and DIEA (308.9 mg, 2.39 mmol, 10 eq) at room temperature. The mixture was stirred for overnight at 60°C. The reaction was quenched with brine (20 mL) at room temperature. The precipitated solids were collected by filtration and washed with water (2x5 mL). The crude product (100 mg) was purified by recrystallization from DMSO (15 mL) to afford 1-104 (29.0 mg, 17.17%) as a light brown solid. (ES, m/z): 707 (M+H)+, 705 (M- H)\ ¾-NMR (400 MHz, DMSO-i 6, ppm): 512.86-12.84 (m, 1H), 8.70-8.36 (m, 3H), 8.04-6.90 (m, 16H), 6.08-6.04 (m, 1H), 5.63-5.57 (m, 1H), 5.23-4.90 (m, 4H), 2.50 (s, 3H).
EXAMPLE 4. Synthesis of E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(2-inethyl-2H- indazole-3-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-7-methyl-lH- benzo[d]imidazol-2-yl)-2-methyl-2H-indazole-3-carboxamide, 1-102
Figure imgf000192_0001
[00377] Synthesis of compound 1-102. Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 2-methyl -2/7-indazole-3 -carboxylic acid (168 mg, 1.00 mmol, 5.00 eq), DIEA (247 mg, 2.40 mmol, 10.0 eq) and HATU (364 mg, 1.00 mmol, 5.00 eq) in DMF (3 mL). The reaction was stirred for 30 min at 25°C. Then 1.8 (100 mg, 0.24 mmol, 1.00 eq) was added. The reaction was stirred for an additional 15 h at 25°C. The reaction was then quenched by the addition of 10 mL of water. The solids were collected by filtration. The crude product was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): column, sun fire C18 OBD Prep column, 100*5m, 19*250mm; mobile phase, water (1% HOAC) and MeOH (50% Phase B up to 90% in 10 min; detector, UV 254 nm) to yield 2.0 mg (1.42%) of 1-102 as an off-white solid. (ES, m/z): 735 (M+H)+; 'H-NMR (400 MHz, DMSO -d6, ppm): 513.05-13 04(d, 2H), 8.48-6.93(m, 17H), 6.12-6.08 (d, 1H), 5.74- 5.70 (d, 1H), 5.11-4.91 (m, 4H), 4.64-4.63 (d, 6H), 2.50 (s, 3H). EXAMPLE 5. Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-l -imidazole-2-carboxamido)- l//-benzo [d] imidazol-1 -yl)but-2-enyl)-2-(l -ethyl-3-methyl- l//-pyrazole-5-carboxamido)-7- methyl-l/f-benzo[d]imidazole-5-carboxamide, 1-96
Figure imgf000193_0001
Figure imgf000194_0001
[00378] Synthesis of compound 5.1. To a stirred mixture of 1.4 (6.1 g, 16.74 mmol, 1 eq) in HOAc (100 mL) was added Zn (10.9 g, 167.40 mmol, 10 eq) at room temperature. The mixture was stirred for 1 h at room temperature. The mixture was concentrated under vacuum. The mixture was diluted with ethanol (200 mL). The mixture was basified to pH 8 with saturated NaiCCh (aq.). The organic layer was dried over anhydrous NaiSCE. After filtration, the filtrate was concentrated under reduced pressure to yield 5.1 (5.3 g, 94.67%) as a light brown solid. (ES, m/z): 335 (M+H)+.
[00379] Synthesis of compound 5.2. To a stirred mixture of 5.1 (5.2 g, 15.55 mmol, 1 eq) in MeOH (100 mL) was added BrCN (2.0 g, 19.02 mmol, 1.2 eq) at room temperature. The mixture was stirred for overnight at room temperature. The mixture was concentrated under vacuum. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silical gel; mobile phase, CH3CN in water, 10% to 100% gradient in 60 min; detector, UV 254 nm to yield 5.2 (3.8 g, 66.71%) as a light yellow solid. (ES, m/z): 360 (M+H)+.
[00380] Synthesis of compound 5.3. To a stirred mixture of l-ethyl-3-methyl-lH-pyrazole-5 carboxylic acid (1.6 g, 10.57 mmol, 1 eq) and HATU (6.0 g, 15.86 mmol, 1.50 eq) in DMF (100 mL) were added DIEA (4.2 g, 31.72 mmol, 3 eq) and 5.2 (3.8 g, 10.57 mmol, 1 eq) at room temperature. The mixture was stirred for overnight at room temperature. The reaction was quenched with brine (500 mL) at room temperature. The mixture was extracted with EtOAc (2 x 300 mL). The combined organic layers were washed with brine (3x100 mL), dried over anhydrous Na2S04. After filtration, the fdtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silical gel; mobile phase, CLLCN in water, 10% to 100% gradient in 60 min; detector, UV 254 nm to yield 5.3 (3 g, 57.26%) as a white solid. (ES, m/z): 496 (M+H)+.
[00381] Synthesis of compound 5.4. To a stirred mixture of 5.3 (3 g, 6.05 mmol, 1 eq) in DCM (60 mL) was added TFA (12 mL) at room temperature. The mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure to yield 24.4 (2.3 g, 77.17%) as a light brown solid. (ES, m/z): 396 (M+H)+.
[00382] Synthesis of compound 5.5. To a stirred mixture of 5.4 (2.3 g, 4.514 mmol, 1 eq) and 4-chloro-3-nitrobenzamide (1.0 g, 5.417 mmol, 1.2eq) in DMF (60 mL) was added K2CO3 (1.2 g, 9.029 mmol, 2 eq) at room temperature. The mixture was stirred for overnight at 70°C. The reaction was quenched with brine (400 mL) at room temperature. The precipitated solids were collected by filtration and washed with water (2x20 mL) and MeOH (3x30 mL). The solid was dried under vacuum to yield 5.5 (2.3 g, 91.05%) as a yellow solid. (ES, m/z): 560 (M+H)+.
[00383] Synthesis of compound 5.6. To a stirred mixture of 5.5 (2.3 g, 4.11 mmol, 1 eq) in HOAc (40 mL) was added Zn (2.6 g, 41.10 mmol, 10 eq) at room temperature. The mixture was stirred for 1 h at room temperature. After filtration, the filter cake was washed with MeOH (2x20 mL). The filtrate was concentrated under reduced pressure until most of the solvent was removed, then the mixture was quenched with water (100 mL) and basified with K2CO3 (aq.) until PH=9. The precipitated solids were collected by filtration and washed with water (3x20 mL). The filter cake was washed with MeOFLDMF (100: 1) until no TM left in the filter cake (detected by TLC). The filtrate was concentrated under reduced pressure to yield 5.6 (1.8 g, 82.69%) as a light pink solid. (ES, m/z): 530 (M+H)+.
[00384] Synthesis of compound 5.7. To a stirred mixture of 5.6 (1.7 g, 3.36 mmol, 1 eq) in MeOH (80 mL) was added BrCN (391.6 mg, 3.70 mmol, 1.1 eq) at room temperature. The mixture was stirred for overnight at room temperature. The precipitated solids were collected by filtration and washed with MeOH (2x10 mL). The solid was dried under vacuum and purified by reverse flash chromatography with the following conditions: column, C18 silical gel; mobile phase, CEfCN in water, 10% to 100% gradient in 30 min; detector, UV 254 nm to yield 5.7 (11.8 g) as a white solid. (ES, m/z): 555.4 (M+H)+.
[00385] Synthesis of compound 1-96. Into a 8-mL sealed tube, was placedl-ethyl-lH-prrrole- 2-carboxylic acid (38.6 mg, 0.28 mmol, 1.5 eq) in DMF (4 mL), HBTU (91 mg, 0.24 mmol, 1.3 eq), DIEA (118.5 mg, 0.92 mmol, 5 eq), 5.7 (100 mg, 0.18 mmol, 1 eq). The reaction was stirred for overnight at 85°C in an oil bath. The mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): column: sun fire Cl 8 OBD Prep column, lOOA, 5 pm, 19 mm X 250 mm; mobile phase A: water (0.1%HOAC), mobile phase B: ACN; flow rate: 25 mL/min; gradient: 45% B to 55% A in 10 min; detector, UV 254/220 nm; rt: 8.75 min to yield 7.7 mg (6.33%) of 1-96 as a yellow solid. (ES, m/z): 677 (M+H)+, 675 (M-Hf, H-NMR (400MHz,DMSO- /6, /i/w;): d 12.83 (s, 2H), 7.95 (s, 1H), 7.86-7.84 (d, 2H), 7.73 (s, 1H), 7.48 (s, 3H), 7.36-7.34 (d, 3H), 6.98 (s, 1H), 6.52 (s, 1H), 5.99- 5.95 (d, 1H), 5.52-5.48 (d, 1H), 4.98 (s, 2H), 4.81 (s, 2H), 4.55 -4.27 (m, 4H), 2.51-2.50 (t, 3H), 2.13 (s, 3H), 1.28-1.24 (m, 7H).
EXAMPLE 6. Synthesis of (£)-l-(4-(5-carbamoyl-2-(l-ethyl-l//-pyrrole-2-carboxamido)- l /-benzo [d] imidazol-1 -yl)but-2-enyl)-2-(l -ethyl-3-methyl- l/ -pyrazole-5-carboxamido)-7- methyl-l/f-benzo[d]imidazole-5-carboxamide, 1-95
Figure imgf000197_0001
O
5.7 1-95
[00386] Synthesis of compound 1-95. Into an 8-mL sealed tube, was placed 1-ethyl-lH- pyrrole-2-carboxylic acid (37.6 mg, 0.27 mmol, 1.5 eq) in NMP (2 mL), HATU (88.9 mg, 0.23 mmol, 1.30 eq), DIEA (116 mg, 0.9 mmol, 5 eq) and 5.7 (100 mg, 0.18 mmol, 1 eq). The reaction was stirred for overnight at 50°C. The mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): column: sun fire Cl 8 OBD Prep column , lOOA, 5 pm, 19 mm*250 mm; mobile phase A:water(0.1%FA), mobile phase B: ACN; flow rate: 25 mL/min; gradient: 35% B to 45% B in 8 min; detector, UV 254/220 nm; rt: 7.95 min to yield 9.1 mg (7.5%) of 1-95 as a white solid. (ES, m/z): 676 (M+H)+, 674 (M-H)-, 1H-NMR (400 MHz, DMSO-r¾, ppm): d 12.59 (s, 2H), 7.92-7.87 (d, 4H), 7.85 (s, 1H), 7.67-7.66 (d, 2H), 7.50 (s, 1H), 7.35-7.33 (d, 1H), 6.94 (s, 1H), 6.85 (s, 1H), 5.97 (s, 1H), 5.54-5.50 (d, 1H), 4.99 (s, 2H), 4.78 (s, 2H), 4.53-4.51 (d, 2H), 4.44-4.42 (d, 2H), 2.52 (s, 3H), 2.13 (s, 3H), 1.28-1.16 (t, 6H).
EXAMPLE 7. Synthesis of (£)-Ar^V-(l,l'-(but-2-ene-l,4-diyl)bis(5-carbamoyl-7-methoxy-l / -benzo[d]imidazole-2,l-diyl))bis(4-ethyl-2-methyloxazole-5-carboxamide), 1-60
Figure imgf000198_0001
7.1
Figure imgf000198_0003
Figure imgf000198_0002
[00387] Synthesis of compound 7.1. To a stirred mixture of ethyl 3-oxopentanoate (9.00 mL, 1.00 eq) in CHCh (20 ml) was added SO2CI2 (5.40 mL, 1.50 eq) dropwise at 0°C under air atmosphere. The mixture was refluxed overnight. The mixture was washed with saturated aq. NaHCCh and dried over anhydrous MgSCL. Then it was concentrated under reduced pressure to yield 8.1 (10.88 g) as a yellow oil.
[00388] Synthesis of compound 7.2. To a stirred mixture of 7.1 (10.88 g, 60.92 mmol, 1.00 eq) in AcOH (20 mL) was added CH3CONH2 (7.21 g, 2.00 eq) in portions at room temperature under air atmosphere. The mixture was stirred overnight at 130°C. Then it was concentrated under reduced pressure, diluted with water (50 mL), and extracted with diethyl ether (3x70 mL). The mixture was washed with 3x 120 mL of aq. NaHC03 solution and concentrated under vacuum. The residue was purified by silica gel column chromatogaphy and eluted with PE/EA (10: 1) to afford 8.2 (1.58 g, 14.16%) as a yellow oil. (ES, m/z): 184 (M+H)+.
[00389] Synthesis of compound 7.3. To a stirred reaction mixture of 7.2 (1.50 g, 8.19 mmol, 1.00 eq) in THF: MeOH (1 : 1) was added LiOH (1.67 g, 41.75 mmol, 5.00 eq) solution dropwise at 0°C under air atmosphere. The mixture was stirred for 2h at room temperature and concentrated under vacuum. The mixture was adjusted to pH=5 with HC1 (1M). The precipitated solids were collected by filtration and washed with water (3x30 mL) to yield 7.3 (800 mg, 62.99%) as a light yellow solid. (ES, m/z): 154 (M-H) .
[00390] Synthesis of compound 1-60. To a stirred mixture of 7.3 (300 mg, 1.93 mmol, 1 eq) and HATU (514.6 mg, 1.35 mmol, 0.7 eq) in NMP (15 ml) were added DIEA (174.9 mg, 1.35 mmol, 0.7 eq) and 13.6 (179.62 mg, 0.387 mmol, 0.2 eq) in portions at room temperature under nitrogen atmosphere. The final reaction mixture was irradiated with microwave radiation for 3 h at 160°C. Then cooled to RT and quenched by TEO. The filtrate cake was purified by reverse flash chromatography under the following conditions: column, C 18 silica gel; mobile phase, MeOH in water, 10% to 80% gradient in 10 min; detector, UV 254 nm. The crude product (60mg) was purified by Prep-HPLC (column: XBridge Shield RP18 OBD Column, 5um, 19* 150mm; mobile phase A: water (10MMOL/L NH4HCO3), mobile phase B: ACN; flow rate: 25 mL/min; gradient: 25% B to 35% B in 9 min; 254;220 nm; rt: 8.65 min) to afford 1-60 (5.2mg, 0.36% ) as a white solid. (ES, m/z): 739 (M+H)+, 737 (M-H) ; ¾-NMR (400 MHz, DMSO-de, ppm): d 8.21-7.83 (d, 2H), 7.63-7.33 (d, 1H), 7.32-7.08 (d, 3H), 5.78-5.18 (d, 2H), 5.00-4.84 (d, 4H), 3.80-3.77 (t, 6H), 2.81-2.78 (t, 4H), 2.67-2.49 (d, 6H), 2.39-2.38 (m, 1H), 1.49-1.39 (m, 1H), 1.19-1.14 (m, 6H), 1.07-1.03 (m, 1H).
EXAMPLE 8. Synthesis of A-(l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-l//-pyrazole-5- carboxamido)-l T-benzo[d]imidazol-l-yl)butyl)-7-(morpholinomethyl)-l/ - benzo[</|imidazol-2-yl)-2,4-dimethyloxazole-5-carboxainide, 1-59
[00391] Synthesis of compound 8.2. To a stirred reaction of 8.1 (15.00 g, 74.783 mmol, 1.00 eq) and DIEA (29.00 g, 224.35 mmol, 3.00 eq) in DMSO (200 mL) was added tert- butyl N-{ 4- aminobutyl)carbamate (21.12 g, 112.18 mmol, 1.50 eq) dropwise/ in portions at RT. The reaction was stirred at 100°C under nitrogen overnight. The mixture was washed with 3 xlOO mL of water. The aqueous layer was extracted with EtOAc (3x100 mL) and filtered to yield 8.2 (18 g, 68.30%) as a yellow solid. (ES, m/z): 353(M+H)+.
[00392] Synthesis of compound 8.3. Into a 1000-mL 3-necked round-bottom flask, was placed
8.2 (40.00 g, 113.51 mmol, 1.00 eq) in HOAc (400 mL) and Zn (74.94 g, 1135.10 mmol, 10.00 eq). The reaction was stirred for 2 h at room temperature. The solids were filtered off and the mixture was concentrated under vacuum to yield 32 g (87.44%) of 8.3 as a black solid. (ES, m/z): 323 (M+H)+.
[00393] Synthesis of compound 8.4. Into a 500-mL 3-necked round-bottom flask, was placed
8.3 (40.00 g, 124.07 mmol, 1.00 eq) in MeOH (400 mL) and BrCN (15.1 g, 148.88 mmol, 1.20 eq). The reaction was stirred for 3 h at room temperature. The mixture was concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): column, C18 silica gel; mobile phase, ACN/H20=15% increasing to ACN/FbO=60% within 15 min; Detector, UV: 254nm to yield 32 g (74.24%) of 8.4 as a white solid. (ES, m/z): 348 (M+H) .
[00394] Synthesis of compound 8.5. Into a 500-mL 3-necked round-bottom flask, was placed
8.4 (28.00 g, 80.59 mmol, 1.00 eq) in DMF (280 mL), DIEA (20.8 g, 161.19 mmol, 2.00 eq), 1- ethyl-3- ethyl-l //-pyrazole-5-carboxylic acid (14.9 g, 96.713 mmol, 1.20 eq) and HATU (45.9 g, 120.89 mmol, 1.50 eq). The reaction was stirred overnight at room temperature. The solution was diluted with 900 mL of FLO. The solids were collected by filtration. The crude product was recrystallized from EA/PE in the ratio of 1 : 1 to yield 32 g (82.11%) of 8.5 as an off-white solid. (ES, m/z): 484 (M+H)+.
[00395] Synthesis of compound 8.6. Into a 500-mL 3-necked round-bottom flask, was placed
8.5 (32.00 g, 66.17 mmol, 1 eq) in MeOH (200.00 mL) and a solution of HC1 (4M) in dioxane (100.00 mL). The reaction was stirred for 3 h at room temperature. The mixture was concentrated under vacuum. The crude product was re-crystallized from EA/PE in the ratio of 1 : 1 to yield 22 g (86.70%) of 8.6 as an off-white solid. (ES, m/z): 384 (M+H)+. [00396] Synthesis of compound 8.7. Into a 50-mL round-bottom flask, was placed 4-[(2- fluoro-3-nitrophenyl)methyl] morpholine (689.15 mg, 2.869 mmol, 1.10 eq) in DMF (10 mL), K2CO3 (720.84 mg, 5.22 mmol, 2.00 eq) and 8.6 (1.00 g, 2.61 mmol, 1.00 eq). The reaction was stirred for 3 h at 70°C. The reaction was then quenched by the addition of 15 mL of water. The solution was extracted with 3x15 mL of ethyl acetate. The mixture was washed with 1 xl 5 ml of water. The solids were filtered off. The mixture was concentrated and purified by Flash-Prep- HPLC under the following conditions (IntelFlash-1): column, C18 silica gel; mobile phase, ACN/H20=15% increasing to ACN/H2O=40% within 20 min; Detector, UV 254 nm to yield 1.14 g (72.41%) of 8.7 as a white solid. (ES, m/z): 604 (M+H)+.
[00397] Synthesis of compound 8.8. Into an 8-mL round-bottom flask, was placed 8.7 (400.00 mg, 0.663 mmol, 1.00 eq) in HOAc (5.00 mL) and Zn (433.40 mg, 6.626 mmol, 10.00 eq) was added portionwise. The reaction was stirred for 1 h at room temperature. The mixture was filtered, and the filter cake was purified by Flash-Prep-HPLC under the following conditions (IntelFlash- 1): column, Cl 8 silica gel; mobile phase, ACN/H2O=20% increasing to ACN/H2O=40% within 20 min; Detector, UV 254 nm to yield 150 mg (39.46%) of 8.8 as a white solid. (ES, m/z): 574 (M+H)+.
[00398] Synthesis of compound 8.9. Into an 8-mL round-bottom flask, was placed 8.8 (150.00 mg, 0.261 mmol, 1.00 eq) in MeOH (2.00 mL) and BrCN (33.29 mg, 0.314 mmol, 1.20 eq). The reaction was stirred overnight at RT. Then it was concentrated and the crude product was purified by Flash-Prep-HPLC under the following conditions (IntelFlash-1): column, C18 silica gel; mobile phase, ACN/H2O=20% increasing to ACN/H2O=40% within 20 min; Detector, UV 254 nm to yield 140 mg (89.43%) of 8.9 as a white solid. (ES, m/z): 585 (M+H)+.
[00399] Synthesis of compound 1-59. Into an 8-mL round-bottom flask, was placed 8.9 (140.00 mg, 0.234 mmol, 1.00 eq) in DMF (3.00 mL). Then dimethyl-1, 3-oxazole-5-carboxylic acid (36.30 mg, 0.257 mmol, 1.10 eq), DIEA (60.44 mg, 0.468 mmol, 2.00 eq) and HATU (133.37 mg, 0.351 mmol, 1.50 eq) were added. Then the reaction mixture was irradiated with microwave radiation for 1 hr at 140°C in microwave. The reaction was then quenched by the addition of 15 mL of water. The solution was extracted with 3x15 mL of ethyl acetate. The combined extracts were washed with 15 ml of water. The solids were filtered off The mixture was concentrated. The crude product was purified by Flash-Prep-HPLC under the following conditions (IntelFlash-1): column, C18 silica gel; mobile phase, ACN/H20=15% increasing to ACN/H2O=40% within 15 min; Detector, UV 254 nm to yield 14 mg (8.29%) of 1-59 as a white solid. LCMS (ES, m/z): [M- H]- = 720.1; ^-NMR (400 MHz, VMSO-de, ppm): d 12.84-12.83 (d, 2H), 8.09-7.90 (d, 2H), 7.80- 7.73 (m, 1H), 7.75-7.47 (m, 2H), 7.46-7.27 (s, 1H), 7.21-6.99 (m, 2H), 6.68-6.47 (s, 1H), 4.69- 4.53 (m, 2H), 4.51-4.45 (t, 2H), 4.37-4.20 (m, 2H), 3.67-3.54 (t, 2H), 3.47-3.37 (m, 4H), 2.49-2.32 (m, 7H), 2.30-2.21 (d, 3H), 2.18-2.11 (t, 3H), 1.97-1.82 (m, 4H), 1.39-1.25 (m, 3H).
EXAMPLE 9. Synthesis of A-(5-carbamoyl-l-(4-(2-(2,4-dimethyloxazole-5-carboxamido)-7- (morpholinomethyl)-l /-benzo[d]imidazol-l-yl)butyl)-l.ff-benzo[d]imidazol-2-yl)-2,4- dimethyloxazole-5-carboxamide, 1-58
Figure imgf000203_0001
Figure imgf000204_0001
[00400] Synthesis of compound 9.1. Into a 100-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 8.4 (1.00 g, 2.88 mmol, 1.00 eq), dimethyl-1, 3- oxazole-5-carboxylic acid (0.61 g, 0.004 mmol, 1.50 eq), HATU (2.19 g, 0.006 mmol, 2.00 eq), DIEA (0.74 g, 0.006 mmol, 2.00 eq), DMF (10.00 mL). The reaction was stirred for 12 hr at room temperature. The reaction was then quenched by the addition of 20 mL of water. The solution was extracted with 3x10 mL of ethyl acetate and washed with 20 ml of brine and dried over anhydrous sodium sulfate. The mixture was concentrated in vacuo and purified by silica gel column chromatography with ethyl acetate/petroleum ether (1 : 1) to yield 0.8 g (59%) of 9.1 as a white solid. (ES, m/z): 471 (M+H) .
[00401] Synthesis of compound 9.2. Into a 100-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 9.1 (750.00 mg, 1.594 mmol, 1.0 eq), MeOH (10 mL), HCl/dioxane (1.00 mL, 4 mol/L). The reaction was stirred for 12 hr at room temperature and then concentrated in vacuo to yield 550 mg (93%) of 9.2 as a solid. (ES, m/z): 371 (M+H)+
[00402] Synthesis of compound 9.3. Into a 25-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 9.2 (550.00 mg, 1.49 mmol, 1.00 eq), 4-[(2- fluoro-3-nitrophenyl) methyl] morpholine (356.71 mg, 1.49 mmol, 1.00 eq), K2CO3 (615.63 mg, 4.45 mmol, 3.00 eq) and DMF (10 mL). The reaction was stirred for 12 hr at 70 °C in an oil bath. Then the reaction was quenched by the addition of 10 mL of water. The solution was extracted with 3x10 mL of ethyl acetate. The mixture was washed with 10 ml of brine, dried over anhydrous sodium sulfate. Then it was concentrated in vacuo. The residue was purified by silica gel column chromatography with ethyl acetate/petroleum ether (1 :5) to yield 250 mg (29%) of 9.3 as a solid. (ES, m/z) 591 (M+H)+ [00403] Synthesis of compound 9.4. Into a 100-mL round-bottom flask, was placed 9.3 (220.00 mg, 0.372 mmol, 1 eq), MeOH (10 mL), Pd/C (7.93 mg, 0.074 mmol, 0.20 eq) under ¾ (g). The reaction was stirred for 3 hr at 25 °C. The solids were fdtered off. The mixture was concentrated in vacuo to yield 110 mg (53%) of 9.4 as a white solid. (ES, m/z): 561 (M+H)+.
[00404] Synthesis of compound 9.5. Into a 50-mL round-bottom flask, was placed 9.4 (110.00 mg, 0.196 mmol, 1.0 eq), MeOH (10 mL), BrCN (27.02 mg, 0.255 mmol, 1.30 eq). The reaction was stirred for 3 hr at 25 °C. Then the solids were filtered off. The mixture was concentrated under vacuum to yield 50 mg (43.51%) of 9.5 as a solid. (ES, m/z): 586 (M+H)+
[00405] Synthesis of compound 1-58. Into a 50-mL round-bottom flask, was placed dimethyl- 1, 3-oxazole-5-carboxylic acid (32.61 mg, 0.231 mmol, 3 eq), DMF (5 mL), HATU (44.13 mg, 0.116 mmol, 1.5 eq), DIEA (49.74 mg, 0.385 mmol, 5 eq), and 9.5 (45 mg, 0.077 mmol, 1 eq). The reaction was stirred for 3 hr at 25 °C. The reaction was then quenched by the addition of 15 mL of water. The solution was extracted with 3x15 mL of ethyl acetate and washed with 15 ml of water, dried over anhydrous sodium sulfate and concentrated in vacuo. The crude product was purified by Flash-Prep-HPLC under the following conditions (IntelFlash-1): Column, C18; mobile phase, ACN: ¾0= 10 increasing to ACN: ¾O=50 within 20 min; Detector, UV 254nm. The crude product was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): Column, XBridge Prep OBD C18 Column, 19*250mm, 5um; mobile phase, water(10MMOL/L NH4HCOV) and ACN (hold 30% PhaseB in 8 min); Detector, UV 254nm to yield 10 mg (17%) of 1-58 as a white solid. LC-MS (EX, m/z) [M+H]+=710.3; 1H-NMR: (400 MHz, DMSO-d6,ppm) d 12.71-12.64 (d, 2H), 7.98-7.94 (d, 2H), 7.75 -7.32 (m, 4H), 7.16-7.03 (m, 2H), 4.49 (s, 2H), 4.28 (s, 2H), 3.59-3.44 (m, 6H), 2.43-2.35 (m, 12H), 2.27 (s, 4H), 1.88 (s, 4H).
EXAMPLE 10. Synthesis of .V-(l-(4-(5-carbamoyl-2-(l-ethyl-3-niethyl-l//-pyrazole-5- carboxamido)-l//-benzo[d]imidazol-l-yl)butyl)-7-methyl-l//-benzo[d]imidazol-2-yl)-4- ethyl-2-methyloxazole-5-carboxamide, 1-54
Figure imgf000206_0001
[00406] Synthesis of compound 10.1. Into a 500-mL 3-necked round-bottom flask, was placed 8.6 (20.00 g, 52.157 mmol, 1.00 eq) in DMF (200 mL), K2C0 (14.4 g, 104.31 mmol, 2.00 eq) and 2-fluoro-l -methyl-3 -nitrobenzene (8.9 g, 57.37 mmol, 1.10 eq). The reaction was stirred overnight at 70°C in an oil bath. The reaction was diluted with 600 mL of H2O. The solution was extracted with 3x500 mL of ethyl acetate and concentrated in vacuo. The crude product was recrystallized from EA/PE in the ratio of 1 : 1 to yield 18 g (66.6%) of 10.1 as a yellow solid. (ES, m/z): 519 (M+H)+.
[00407] Synthesis of compound 10.2. Into a 500-mL 3-necked round-bottom flask, was placed 10.1 (18.00 g, 34.71 mmol, 1.00 eq) in HOAc (180 mL) and Zn (68.1 g, 1041.31 mmol, 30.00 eq). The reaction was stirred for 2 h at RT. The solids were filtered off. The mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography with dichloromethane/methanol (20: 1) to yield 8.3 g (48.94%) of 10.2 as an off-white solid. (ES, m/z) 489 (M+H)+.
[00408] Synthesis of compound 10.3. Into a 500-mL 3-necked round-bottom flask, was placed 10.2 (18.00 g, 36.84 mmol, 1.00 eq) in MeOH (200 mL) and BrCN (4.68 g, 44.21 mmol, 1.20 eq). The reaction was stirred for 3 h at room temperature and then concentrated in vacuo. The crude product was recrystallized from EA/PE in the ratio of 1 : 1 to yield 10 g (52.91%) of 10.3 as a white solid. (ES, m/z): 514 (M+H) .
[00409] Synthesis of compound 1-54. To a stirred reaction of 4-ethyl-2-methyl-l,3-oxazole-5- carboxylic acid (45.39 mg, 0.293 mmol, 1 eq), HATU (223.04 mg, 0.586 mmol, 2 eq) and DIEA (118.92 mg, 1.172 mmol, 4 eq) in NMP (3 ml) were added 10.3 (150 mg, 0.293 mmol, 1 eq) at RT under nitrogen. The final reaction mixture was irradiated with microwave radiation for lh at 140 °C. Then the reaction was quenched by the addition of 15 mL of water. The solution was extracted with 3x15 mL of ethyl acetate and washed with 15 ml of water, and dried over anhydrous sodium sulfate. The mixture was concentrated and purified by reverse flash chromatography under the following conditions: column, C18 silica gel; mobile phase, ACN in water, 10% to 60% gradient in 15 min; detector, UV 254 nm to afford 1-54 (85.5 mg, 67.5%) as a light yellow solid. (ES, m/z): 651 (M+H)+, 649 (M-H)\ 1H-NMR (300 MHz, DMSO -d6 ,ppm) d 12.81 (s, 1H), 12.58 (s, 1H), 7.97 (s, 2H), 7.75-7.74 (d, 1H), 7.57-7.55 (d, 1H), 7.40-7.39 (d, 1H), 7.33 (s, 1H), 7.09-7.06 (t, 1H), 6.99-6.98 (d, 1H), 6.59 (s, 1H), 4.58-4.56 (q, 2H), 4.37-4.27 (m, 4H), 2.89-2.84 (q, 2H), 2.61 (s, 3H), 2.36 (s, 3H), 2.1 1 (s, 3H), 1.88 (s, 4H), 1.33-1.31 (t, 3H), 1.07-1.02 (t, 3H).
EXAMPLE 11. Synthesis of iV-(l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-l//-pyrazole-5- carboxamido)-l//-benzo[d]imidazol-l-yl)butyl)-7-methyl-l//-benzo[d]imidazol-2-yl)-3- methylisothiazole-4-carboxamide, 1-53
Figure imgf000207_0001
[00410] Synthesis of compound 1-53. To a stirred solution of 3 -methyl- l,2-thiazole-4-
206
5UB5TITUTE SHEET (RULE 26) carboxylic acid (50.17 mg, 0.350 mmol, 1.2 eq), HATU (222.09 mg, 0.584 mmol, 2 eq) and DIEA (150.98 mg, 1.168 mmol, 4 eq) in NMP (10 ml) were added 10.3 (150 mg, 0.292 mmol, 1 eq) at RT under nitrogen. The reaction mixture was irradiated with microwave radiation for lh at 140 °C. The reaction was then quenched by the addition of 15 mL of water. The solution was extracted with 3x15 mL of ethyl acetate and washed with 15 ml of water. The solids were filtered off. The mixture was concentrated and purified by reverse flash chromatography under the following conditions: column, C18 silica gel; mobile phase, ACN in water, 10% to 60% gradient in 15 min; detector, UV 254 nm to afford 1-53 (86.8 mg, 46.53%) as a light yellow solid. (ES, m/z): 639 (M+H)+, 637 (M-H)-; ¾-NMR (300 MHz, mA$Q-d6,ppm) d 12.83-12.72 (d, 2H), 9.42 (s, 1H), 7.98-7.97 (d, 2H), 7.81-7.72 (m, 1H), 7.59-7.58 (d, 1H), 7.40-7.34 (m, 2H), 7.10-7.09 (t, 1H), 7.00- 6.99 (d, 1H), 6.61 (s, 1H), 4.59-4.58 (q, 2H), 4.43 (s, 2H), 4.30 (s, 2H), 2.70 (s, 3H), 2.61 (s, 3H), 2.10-2.08 (d, 3H), 1.95-1.86 (d, 4H), 1.33-1.29 (t, 3H).
EXAMPLE 12. Synthesis of N-(l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)butyl)-7-methyl-lH-benzo[d]imidazol-2-yl)-2,4- dimethylthiazole-5-carboxamide, 1-52
Figure imgf000208_0001
[00411] Synthesis of compound 1-52. To a stirred solution of 3 -methyl- l,2-thiazole-4- carboxylic acid (50.17 mg, 0.350 mmol, 1.2 eq), HATU (222.09 mg, 0.584 mmol, 2 eq) and DIEA (150.98 mg, 1.168 mmol, 4 eq) in NMP (10 ml) were added 10.3 (150 mg, 0.292 mmol, 1 eq) at RT under nitrogen. The reaction mixture was irradiated with microwave radiation for lh at 140 °C. The reaction was then quenched by the addition of 15 mL of water. The solution was extracted with 3x15 mL of ethyl acetate and washed with 15 ml of water. The solids were filtered off. The mixture was concentrated and purified by reverse flash chromatography under the following conditions: column, C18 silica gel; mobile phase, ACN in water, 10% to 60% gradient in 15 min; detector, UV 254 nm to afford 1-52 (86.8 mg, 46.53%) as a light yellow solid. (ES, m/z): 639 (M+H)+, 637 (M-H)-; Ή-NMII (300 MHz, DMSO-i¾ ppm) d 12.83-12.72 (d, 2H), 9.42 (s, 1H), 7.98-7.97 (d, 2H), 7.81-7.72 (m, 1H), 7.59-7.58 (d, 1H), 7.40-7.34 (m, 2H), 7.10-7.09 (t, 1H), 7.00- 6.99 (d, 1H), 6.61 (s, 1H), 4.59-4.58 (q, 2H), 4.43 (s, 2H), 4.30 (s, 2H), 2.70 (s, 3H), 2.61 (s, 3H), 2.10-2.08 (d, 3H), 1.95-1.86 (d, 4H), 1.33-1.29 (t, 3H).
EXAMPLE 13. Synthesis of (£)-AVV-(l,r-(but-2-ene-l,4-diyl)bis(5-carbamoyl-7-methoxy- l//-benzo[d]imidazole-2,l-diyl))bis(2,4-dimethyloxazole-5-carboxamide), 1-48
Figure imgf000209_0001
[00412] Synthesis of compound 13.2. To a solution of 13.1 (10 g, 43.48 mmol, 1 eq) in ethanol (150 mL) was added fe/7-butyl A-[(2A)-4-aminobut-2-en-l-yl]carbamate hydrochloride (11.58 g, 52.176 mmol, 1.2 eq) and DIEA (16.826 g, 130.44 mmol, 3 eq). The reaction was stirred overnight at 120°C. The reaction mixture was cooled to RT and the solids were collected by filtration to yield 8 g (48.43%) of 13.2 as an orange solid. (ES, m/z): 381 (M+H) .
[00413] Synthesis of compound 13.3. To a solution of 13.2 (8 g, 21 mmol, 1 eq) in methanol (100 mL) was added 4M HC1/ 1,4-dioxane (4 mol/L, 20 mL). The mixture was stirred overnight at RT and the concentrated in vacuo to yield 6 g (90%) of 13.3 as an off-white solid. (ES, m/z)·. 281 (M+H)+.
[00414] Synthesis of compound 13.4. To a solution of 13.3 (3.6 g, 12.85 mmol, 1 eq) in DMSO (50 mL) was added DIEA (8.29 g, 64.2 mmol, 5 eq) and 4-chloro-3-methyl-5-nitrobenzamide (2.955 g, 12.85 mmol, 1 eq). The reaction was stirred overnight at 80°C. The mixture was cooled to RT and diluted with water. The solids were collected by filtration to yield 4 g (66%) of 13.4 as a brown solid. (ES, m/z): 475 (M+H)+.
[00415] Synthesis of compound 13.5. To a solution of 13.4 (4 g, 8.431 mmol, 1 eq) in cone. HC1 (50 mL) was added tin(II)chloride dihydrate (19.08 g, 84.31 mmol, 10 eq) at 0°C. The reaction was stirred overnight at RT. The pH of the reaction was adjusted to pH 9 with sat. sodium bicarbonate and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated in vacuo to yield 4 g (crude) of 15.4 as a brown solid. (ES, m/z): 415 (M+H)+.
[00416] Synthesis of compound 13.6. To a solution of 13.5 (400 mg, 0.965 mmol, 1 eq) in methanol (10 mL) and water (10 mL) was added cyanagen bromide (202.65 mg, 1.93 mmol, 2 eq). The reaction was stirred overnight at 70°C. The mixture was cooled to room temperature and concentrated in vacuo to yield 500 mg (crude) of 13.6 as a yellow solid. (ES, m/z): 465 (M+H)+.
[00417] Synthesis of compound 1-48. Into a 8-mL round-bottom flask, was placed 13.6 (128.00 mg, 0.276 mmol, 1.00 eq) in NMP (2.00 mL). Then DIEA (71.23 mg, 0.551 mmol, 2.00 eq), dimethyl-1, 3-oxazole-5-carboxylic acid (46.67 mg, 0.331 mmol, 1.20 eq) and HATU (157.17 mg, 0.413 mmol, 1.50 eq) were added. The reaction was irradiated with microwave radiation 1 h at 140 °C. Cooled to RT and the crude product was purified by Flash-Prep-HPLC under the following conditions (IntelFlash-1): column, C18 silica gel; mobile phase, ACN: H20=15% increasing to ACN: H2O=60% within 20 min; Detector, UV 254 nm to yield 55.2 mg (28.18%) of 1-48 as an off-white solid. (ES, m/z): 711 (M+H)+, 709 (M-H) , ¾-NMR (400 MHz, D20, ppm): d 7.52 (s, 2H), 6.78 (s, 2H), 5.57-4.85(d, 6H), 3.13(s, 6H), 2.55 (s, 6H), 2.21 (s, 6H). EXAMPLE 14. Synthesis of l-(4-(2-(3-ethyl-l-methyl-l//-pyrazole-4-carboxamido)-7- methyl-l//-benzo[d]imidazol-l-yl)butyl)-2-(l-ethyl-3-methyl-li/-pyrazole-5-carboxamido)- l//-benzo[d]imidazole-5-carboxamide, 1-47
Figure imgf000211_0001
[00418] Synthesis of compound 1-47. A solution of 3-ethyl-l-methyl-l /-pyrazole-5- carboxylic acid (54.03 mg, 0.350 mmol, 1.2 eq.) in NMP (7.5 mL) was treated with HATU (222.09 mg, 0.584 mmol, 2 eq.) and DIEA (113.24 mg, 0.876 mmol, 3 eq.) for 30 min at RT followed by the addition of 10.3 (150 mg, 0.292 mmol, 1 eq.) in portions at RT. The final reaction mixture was irradiated with microwave radiation for 1 h at 140 0 C. The mixture cooled to RT then diluted with ethyl acetate (150 mL), washed with 5x50mL of brine, and concentrated under vacuum. The crude product (72 mg) was purified by Prep-HPLC with the following conditions: Column, Cl 8 silica gel; mobile phase, CLLCN/Water (in NH4HCO3) =0: 100% increasing to CH3CN/Water=100%:0 within 15 min; Detector 254nm to yield 1-47 43.8 mg (23.08%) as a white solid. (ES, m/z): 650 (M+H)+, 648 (M-H)-; ¾-NMR (300 MHz, mASO-d6,pprn) ό 12.82 (s, 1H), 12.56 (s, 1H), 8.01- 7.95 (m, 3H), 7.76-7.74 (d, 1H), 7.58-7.56 (d, 1H), 7.34-7.32 (d, 2H), 7.04-6.95 (t, 1H), 6.95-6.93 (d, 1H), 6.61 (s, 1H), 4.58-4.57 (q, 2H), 4.37-4.28 (t, 4H), 3.97-3.68 (t, 3H), 2.88-2.87 (q, 2H), 2.58 (s, 3H), 2.10 (s, 3H), 1.91-1.90 (q, 2H), 1.83-1.82 (q, 2H), 1.31-1.29 (t, 3H), 1.15-1.10 (t, 3H).
EXAMPLE 15. Synthesis of 2-(l-ethyl-3-methyl-l//-pyrazole-5-carboxamido)-l-(4-(7- methyl-2-(oxetane-3-carboxamido)-l//-benzo[d]imidazol-l-yl)butyl)-l//- benzo[</| imidazole- 5-carboxamide, 1-45
Figure imgf000212_0001
[00419] Synthesis of compound 1-45. To a solution of ox etane-3 -carboxylic acid (23.85 mg, 0.234 mmol, 1.20 eq) in DMF (1 mL) were added HATU (148.06 mg, 0.389 mmol, 2.00 eq), DIEA (75.49 mg, 0.584 mmol, 3.0 eq). Then 10.3 (100.00 mg, 0.195 mmol, 1.00 eq) was added in portions at RT. The mixture was stirred for 1 h at RT. Then diluted with 20 mL of water, extracted with 3x20 mL of ethyl acetate and the organic layers were combined and concentrated in vacuo. The residue was purified by reverse flash chromatography under the following conditions: column, Cl 8 silica gel; mobile phase, MeOH in water, 10% to 50% gradient in 10 min; detector, UV 254 nm to yield 26.7mg (16.6%) of 1-45 as a light brown solid. (ES, m/z): 598 (M+H)+; 1H-NMR (300 MHz, DMSO -d6,ppm) d 12.81 (s, 1H), 12.58 (s, 1H), 8.00-7.93 (m, 2H), 7.76-7.75 (d, 1H), 7.54- 7.53 (d, 1H), 7.38-7.35 (d, 2H), 7.09-7.05 (t, 1H), 6.96-6.95 (d, 1H), 6.62 (s, 1H), 4.72-4.53 (m, 6H), 4.34-4.23 (m, 4H), 3.82-3.68 (m, 1H), 2.54 (s, 3H), 2.13 (s, 3H), 1.82-1.73 (m, 4H), 1.32- 1.30 (t, 3H).
EXAMPLE 16. Synthesis of (£)-A-(5-carbamoyl-l-(4-(5-carbamoyl-2-(2,4-dimethyloxazole- 5-carboxamido)-l//-benzo[d]imidazol-l-yl)but-2-enyl)-7-methyl-l//-benzo[d]imidazol-2- yl)-2,4-dimethyloxazole-5-carboxamide, 1-38
Figure imgf000213_0001
[00420] Synthesis of compound 1-38. To a solution of dimethyl-1, 3-oxazole-5-carboxylic acid (121.41 mg, 0.860 mmol, 6.00 eq) in NMP (3 mL) were added HATU (327.11 mg, 0.860 mmol, 6 eq), DIEA (370.62 mg, 2.868 mmol, 20 eq) and 1.8 (60.00 mg, 0.143 mmol, 1.00 eq) in portions at RT. The mixture was stirred for lh at 140°C under microwave radiation. Then it was diluted with water (18 mL). The precipitated solids were collected by filtration and washed with water (3x1 mL). The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column 30x 150mm 5um; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 19% B to 29% B in 7 min; 254; 220 nm; Rt: 5.55 min) to yield 4.0 mg (4 %) of 1-38 as a white solid. (ES, m/z): 665 (M+H)+, 663 (M-H)-; 1H-NMR (300 MHz, DMSO-< p/w) d 12.71 (s, 2H), 8.02-7.83 (m, 4H), 7.72-7.71 (d, 1H), 7.52-7.45 (m, 2H), 7.44-7.31 (d, 2H), 5.88-5.87 (d, 1H), 5.54-5.52 (d, 1H), 4.95 (s, 2H), 4.80-4.79 (d, 2H), 2.50-2.30 (m, 15H).
EXAMPLE 17. Synthesis of .V-(l-(4-(5-carbainoyl-2-(l-ethyl-3-niethyl-l//-pyrazole-5- carboxamido)-l/T-benzo[i/]imidazol-l-yl)butyl)-7-methyl-l//-benzo[i/]imidazol-2-yl)-2,4- dimethyloxazole-5-carboxamide, 1-43
Figure imgf000214_0001
[00421] Synthesis of compound 1-43. Into a 8-mL round-bottom flask purged and maintained under an inert atmosphere of nitrogen, was placed dimethyl-1, 3-oxazole-5-carboxylic acid (49.00 mg, 0.347 mmol, 1.19 eq) in MP (3.00 mL), HATU (333.00 mg, 0.876 mmol, 3.00 eq), DIEA (123.00 mg, 0.952 mmol, 3.26 eq) and 10.3 (150.00 mg, 0.292 mmol, 1.00 eq). The reaction mixture was irradiated with microwave radiation for 1 h at 140 °C. Then the reaction was quenched by the addition of 10 mL of water. The solids were collected by filtration. The crude product was purified by Prep-HPLC with the following conditions (SHIMADZU (HPLC-01)): column, X bridge Prep OBD C18 column, 19!l!250mm, 5um; mobile phase, water (l%HOAc) and MeOH (57% phase B up to 67% in 10 min); Detector, UV 254nm to yield 13.7 mg (7.4%) of 1-43 as a white solid. (ES, m/z): 637 (M+H)+, 635 (M-H)'; LH-NMR (300 MHz, DMSO- ppm) d 12.66- 12.57(m, 2H), 7.97-7.95 (d, 2H), 7.77-7.74 (d, 1H), 7.54-7.53 (d, 1H), 7.40-7.37 (d, 1H), 7.32 (s, 1H), 7.11-7.06 (t, 1H), 7.00-6.97 (d, 1H), 6.58 (s, 1H), 4.61-4.54(m, 2H), 4.38-4.27 (m, 4H), 2.60 (s, 3H), 2.36 (s, 6H), 2.1 1 (s, 3H),1.86(s, 4H), 1.33-1.29(t, 3H).
EXAMPLE 18. Synthesis of l-[(2E)-4-[5-carbamoyl-2-[2-(3-hydroxypropyl)-5- methylpyrazole-3-amido]-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-(morpholin-4-ylmethyl)-l,3-benzodiazole-5-carboxamide, 1-142
Figure imgf000215_0001
Boc
Figure imgf000215_0002
Boc
Figure imgf000216_0001
1-142
[00422] Synthesis of compound 18.2. Into a 250 mL 3-necked round-bottom flask, was placed methyl 4-chloro-3-methylbenzoate (25 g, 135.9 mmol, 1.00 equiv) in H2SO4 (36 mL) at -20 °C under an ice/salt bath. Then HNO3 (6.03 mL, 135.9 mmol, 1.00 equiv) was added dropwise and the resulting solution was stirred for lh at -20 °C. The resulting solution was then diluted with 500 mL of ice water. The resulting solution was extracted with 3x200 mL of ethyl acetate and concentrated under vacuum to yield 18.2 (30 g, crude) as yellow oil. (ES, m/z): 230 (M+H+).
[00423] Synthesis of compound 18.3. To a stirred solution of 18.2 (48 g, 209.6 mmol, 1.00 equiv) in CCL (500 mL) were added NBS (37.3 g, 209.6 mmol, 1.00 equiv) and AP3N (3.44 g, 20.96 mmol, 0.10 equiv) in portions at room temperature. The resulting mixture was stirred overnight at 85 °C. The resulting mixture was cooled to room temperature and filtered, the filter cake was washed with THF (3x50 mL). The filtrate was concentrated under reduced pressure to yield 18.3 (70 g, crude) as yellow oil. (ES, m/z): 308 (M+LL).
[00424] Synthesis of compound 18.4. To a stirred solution of 18.3 (70 g, crude) in DCM (700 mL) were added DIEA (108.2 g, 838.4 mmol) and morpholine (18.23 g, 209.6 mmol) at room temperature. The resulting mixture was stirred overnight at room temperature. The resulting mixture was washed with water (3 x 150 mL), dried over anhydrous NaiSCL. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (10: 1 to 2: 1) to afford 18.4 (28 g) as an off-white oil. (ES, m/z): 315 (M+H+).
[00425] Synthesis of compound 18.5. Into a 250 mL sealed tube was added 18.4 (28 g, 89.17 mmol, 1.00 equiv) in NEL (g) in CLLOH (300 mL) at room temperature. The resulting mixture was stirred overnight at 50 °C. The resulting mixture was cooled to room temperature and filtered, the filter cake was washed with CH3OH (3 x 30 mL). The filtrate was concentrated under reduced pressure. The product was precipitated by the addition of CH2CI2 to yield 18.5 (9.6 g, 36%) as an off-white solid. (ES, m/z): 300 (M+EL).
[00426] Synthesis of compound 18.6. To a stirred solution of tert- butyl Ar-[(2A)-4-aminobut- 2-en-l-yl] carbamate (5.90 g, 31.72 mmol, 1.00 equiv) in DMSO (100 mL) were added DIEA (12.28 g, 95.16 mmol, 3.00 equiv) and 18.5 (9.48 g, 31.72 mmol, 1.00 equiv) at room temperature. The resulting mixture was stirred for 5 h at 100 °C. The resulting mixture was cooled to room temperature, diluted with water and extracted with EtOAc (3 x 300 mL). The combined organic layers were washed with water (3 x 150 mL), dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. The product was precipitated by the addition of CH2CI2 to yield 18.6 (6.9 g, 48.4%) as a yellow solid. (ES, m/z): 450 (M+H+).
[00427] Synthesis of compound 18.7. Into a 100-mL round-bottom flask, was placed 18.6 (4 g, 8.9 mmol, 1.00 equiv) in MeOH (30 mL) and NH3.H2O (10 mL), then NaiSiCL (9.29 g, 53.4 mmol, 6.00 equiv) was added. The resulting solution was stirred for 2 hr at room temperature.
The solids were filtered out. The resulting mixture was concentrated. The residue was applied onto a silica gel column with dichloromethane/methanol (15: 1) to yield 1.1 g (29.5%) of 18.7 as an off-white solid. (ES, m/z) 420 (M+H+).
[00428] Synthesis of compound 18.8. To a stirred solution of 18.7 (4.40 g, 10.5 mmol, 1.00 equiv) in methanol (40 mL) was added cyanogen bromide (1.32 g, 12.6 mmol, 1.20 equiv) at room temperature. The resulting mixture was stirred for 1.5 h at room temperature. The resulting mixture was concentrated in vacuo and the resulting precipitated solids were collected by fdtration and washed with MeOH (3 x 10 mL). The filtrate was concentrated under reduced pressure to give 18.8 (3.3 g, 70.8%) as an off-white solid. (ES, m/z): 445 (M+H+).
[00429] Synthesis of compound 18.9. Into a 25-mL round-bottom flask, was placed 4-ethyl- 2-methyl-l,3-oxazole-5-carboxylic acid (261.9 mg, 1.69 mmol, 1.5 equiv) in DMF (5.0 mL), HATU (642.2 mg, 1.69 mmol, 1.5 equiv) and DIEA (437.3 mg, 3.39 mmol, 3 equiv) were added under nitrogen, followed by addition of 18.8 (500 mg, 1.13 mmol, 1.00 equiv) at room temperature. The resulting solution was stirred for 2 hrs at 85 °C. The resulting solution was cooled to room temperature, diluted with water and extracted with 3 x 20 mL of ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated in vacuo. The residue was applied onto a silica gel column with EA/PE (1 :2) to give of 18.9 (480 mg, 73.1%) as a brown yellow solid. (ES, m/z): 582 (M+H+).
[00430] Synthesis of compound 18.10. Into a 25-mL round-bottom flask, was placed 18.9 (480 mg, 0.826 mmol, 1.00 equiv) in 4 M HCl-l,4-dioxane (5.0 mL) at 0 °C. The resulting solution was stirred for 2 hrs at room temperature. The resulting mixture was concentrated to yield 700 mg (crude) of 18.10 as a brown crude solid. (ES, m/z): 482 (M+H+).
[00431] Synthesis of compound 18.11. Into a 25-mL round-bottom flask, 18.10 (500 mg, 1.00 equiv) in DMSO (5.0 mL), then DIEA (402.5 mg, 3.12 mmol, 3.00 equiv) and 4-fluoro-3- nitrobenzamide (191.2 mg, 1.04 mmol, 1.00 equiv) were added. The resulting solution was stirred for 2 hrs at 60 °C. The resulting solution was cooled to room temperature, diluted with water and extracted with 4x10 mL of ethyl acetate. The organic layers was combined and concentrated. The residue was applied onto a silica gel column with dichloromethane/methanol (10: 1) to afford of 18.11 (300 mg, 44.7%) as a brown solid. (ES, m/z ): 646 (M+H+).
[00432] Synthesis of compound 18.12. Into a 25-mL round-bottom flask, was placed 18.11 (300 mg, 0.465 mmol, 1.00 equiv) in methanol (5.0 mL) and ammonia (1.0 mL), then T^SiCL (485.4mg, 2.79 mmol, 6.00 equiv) was added. The resulting solution was stirred for 2 hr at room temperature. The solids were filtered out and the filtrate was concentrated in vacuo. The residue was applied onto a silica gel column with dichloromethane/methanol (5: 1) to provide) of 18.12 (200 mg, 69.9%) as a brown yellow solid. (ES, m/z): 616 (M+H+).
[00433] Synthesis of compound 18.13. Into a 25-mL round-bottom flask, was placed 18.12 (200 mg, 0.325 mmol, 1.00 equiv) in MeOH (5.0 mL), then BrCN (275.3 mg, 2.60 mmol, 8.00 equiv) was added. The resulting solution was stirred for 2 hr at room temperature. The resulting mixture was concentrated in vacuo to give 200 mg (crude) of 18.13 as a brown yellow solid. (ES, m/z): 641 (M+H+).
[00434] Synthesis of compound 18.14. Into a 25-mL round-bottom flask, was placed 2-[3- [(tert-butyldimethylsilyl)oxy]propyl]-5-methylpyrazole-3-carboxylic acid (139.8mg, 0.468 mmol, 1.50 equiv), HATU (177.8 mg, 0.468 mmol, 1.50 equiv) and DIEA (241.6 mg, 1.873 mmol, 6.00 equiv) in DMF (2.0 mL), Then a solution of 18.3 (200 mg, 0.312 mmol, 1.00 equiv) were added and the resulting solution was stirred for 2 hr at 85 °C. The resulting solution was cooled to room temperature, diluted with water and extracted with 3 x 20 mL of ethyl acetate. The combined organic layers were dried over sodium sulfate and concentrated in vacuo. The residue was applied onto a silica gel column with dichloromethane/methanol (20: 1 to 1 : 1) to give of 18.14 (60 mg, 20.9%) as a brown yellow solid. (ES, m/z): 921 (M+H+).
[00435] Synthesis of compound 1-142. Into a 25-mL round-bottom flask, was placed 18.14 (100 mg, 0.109 mmol, 1.00 equiv) in 4 M HCl-l,4-dioxane (2.0 mL). The resulting solution was stirred for 2 hr at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column: XSelect CSHPrep C18 OBD Column, 5um, 19* 150mm; Mobile Phase A: water (10% NH4HCO3), Mobile Phase B:ACN; Flow rate:25 mL/min; Gradient: 10 B to 40 B in 7 min; UV detection at 210/254 nm to yield 1-142 (30 mg, 34.1%) as a white solid. (ES, m/z): 807 (M+H+); 'H-NMR (400 MHz, DMSO -d6) d 12.79 (s, 2H), 8.01 - 7.96 (m, 2H), 7.93 (d, 2H), 7.72 (dd, 1H), 7.55 (d, 1H), 7.43 (d, 1H), 7.32 (s, 2H), 6.57 (s, 1H), 5.93 (dt, 1H), 5.42 (dd, 1H), 5.25 - 5.13 (m, 2H), 4.81 (d, 2H), 4.60 (t, 2H), 4.50 (t, 1H), 3.44 (td, 8H), 2.84 (q, 2H) , 2.42 (s, 3H), 2.18 (d, 7H), 1.90 (p, 2H), 1.04 (t, 3H).
EXAMPLE 19. Synthesis of 4-[2-[5-([5-carbamoyl-l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazol-2- yl]carbamoyl)-3-methylpyrazol-l-yl]ethoxy]butanoic acid, 1-143
Figure imgf000220_0001
Figure imgf000221_0001
19.13 .
Figure imgf000222_0001
[00436] Synthesis of compound 19.1. To a stirred solution of methyl 4-fluoro-3-nitrobenzoate (10.0 g, 50.217 mmol, 1 equiv) and tert-butyl N-[(2E)-4-aminobut-2-en-l-yl]carbamate hydrochloride (12.30 g, 55.238 mmol, 1.1 equiv) in DMF (100 mL) was added K2CO3 (20.82 g, 150.650 mmol, 3.0 equiv). The resulting mixture was stirred for 4h at 70 °C. The reaction was cooled to rt and diluted with water. The precipitated solids were collected by filtration to yield 19.1(17 g, 92.65%) as a yellow solid. LC-MS (ES, m/z ): 753 (2M+Na+).
[00437] Synthesis of compound 19.2. To a stirred solution of 19.1 (10 g, 27.368 mmol, 1 equiv) in DMF (80 mL) was added SnCE^FEO (30.88 g, 136.842 mmol, 5.0 equiv). The resulting mixture was stirred overnight at 35 °C. The reaction was diluted with water and basified to pH 9 with saturated NaHCCE (aq ). The mixture was extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous INfeSCE and concentrated under reduced pressure. This resulted in 19.2 (8.0 g, 87.15%) as an off-white solid. LC-MS (ES, m/z): 336 (M+H+).
[00438] Synthesis of compound 19.3. To a stirred solution of 19.2 (8 g, 23.852 mmol, 1 equiv) in MeOH (100 mL) was BrCN (7.58 g, 71.555 mmol, 3.0 equiv). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. This resulted in 19.3 (8 g, 93.17%) as an off-white solid. LC-MS (ES, m/z): 361 (M+H+).
[00439] Synthesis of compound 19.4. To a stirred solution of 19.3 (8 g, 22.22 mmol, 1.00 equiv) and 1 -ethyl-3 -methyl- lH-pyrazole-5 -carboxylic acid (5.13 g, 33.33 mmol, 1.5 equiv) in DMF (50 mL) were added HATU (12.67 g, 33.33 mmol, 1.5 equiv) and DIEA (8.6 g, 66.66 mmol, 3.00 equiv). The resulting mixture was stirred for 4h at room temperature. The reaction was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous NaiSCL and concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (45% ACN up to 54% in 10 min); Detector, UV 254/220 nm. This resulted in 19.4 (9.5 g, 86.2%) of as an off-white solid. LC-MS (ES, m/z): 497 (M+H+).
[00440] Synthesis of compound 19.5. To a solution of 19.4 (2 g, 4.028 mmol, 1 equiv) in methanol (30 mL), tetrahydrofuran (15 mL) and water (5 mL) was added lithium hydroxide
(385.85 mg, 16.111 mmol, 4.00 equiv). The resulting mixture was stirred at 60 °C for 12 hrs. The mixture was concentrated under reduced pressure. The residue was diluted with water, then adjusted to pH 3 with 1 M HC1. The solids were collected by fdtration to yield 19.5 (746 mg, 38.4%) as an off-white solid. (ES, m/z): 483 (M+H+).
[00441] Syntheis of compound 19.6. To a solution of 19.5 (746 mg, 1.546 mmol, 1 equiv) in dimethyl formamide (5 mL) was added ammonium chloride (413.48 mg, 7.730 mmol, 5.00 equiv), DIEA (1998.08 mg, 15.460 mmol, 10.00 equiv) and HATU (1175.66 mg, 3.092 mmol, 2.00 equiv) under nitrogen. The resulting mixture was stirred at RT overnight. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, water (0.1% FA) and ACN (13% ACN up to 65% in 7 min); UV detection at 254/220 nm. This resulted in 19.6 (400 mg, 53.7%) as a white solid. (ES, m/z): 482 (M+H+).
[00442] Synthesis of compound 19.7. To a solution of 19.6 (100 mg, 0.207mmol) in dichloromethane (3 mL) was added trifluoroacetic acid (0.5 mL). The resulting mixture was stirred at RT for 2h. The mixture was concentrated under reduced pressure. This resulted in 19.7 (60 mg, 76%) as a brown yellow solid. (ES, m/z): 382 (M+H+).
[00443] Synthesis of compound 19.8. To a stirred mixture of l9.7 (1.5 g, 3.932 mmol, 1 equiv) and 4-fluoro-3-nitrobenzamide (0.72 g, 3.932 mmol, 1 equiv) in dimethyl formamide (20 mL) was added potassium carbonate (1.63 g, 11.797 mmol, 3.00 equiv). The resulting mixture was stirred for 6 h at 80 °C under nitrogen atmosphere. The mixture was cooled to room temperature, diluted with water and extracted with dichloromethane. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 19.8 (2.2 g, 58.33%) as a yellow solid. (ES, m/z): 546 (M+H+).
[00444] Synthesis of compound 19.9. To a stirred solution of 19.8 (2.2 g, 4.037 mmol, 1 equiv) in acetic acid (20 mL) was added zinc powder (0.6 g, 9.173 mmol, 5 equiv). The resulting mixture was stirred for 3 h at 50 °C. The mixture was cooled to room temperature. The solids were filtered out and the filtrate was concentrated under reduced pressure. The pH value of the solution was adjusted to 8 with sat. sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure to yield 19.9 (2.2 g, crude) as a brown oil. (ES, m/z) 516 (M+H+).
[00445] Synthesis of compound 19.10. To a stirred solution of 19.9 (2.2 g crude, 4.037 mmol, 1 equiv) in methanol (20 mL) was added cyanogen bromide (0.636 g, 6.056 mmol, 1.5 equiv) under nitrogen. The resulting mixture was stirred overnight at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, water (0.1% FA) and ACN (28% ACN up to 40% in 10 min); UV detection at 254/220 nm to yield 19.10 (1 g) as a brown solid. (ES, m/z): 541 (M+H+).
[00446] Synthesis of compound 19.11. To a stirred solution of dimethylsulfoxide (1.59 g, 20.4 mmol, 2 equiv) in dichloromethane (20 mL) was added dropwise oxalyl chloride (1.94 g, 15.287 mmol, 1.5 equiv) at -78 °C under nitrogen atmosphere. After 30 min, 2-(2- (benzyloxy)ethoxy)ethanol (2 g, 10.2 mmol, 1 equiv) was added via syringe at -78 °C. The resulting mixture was stirred for 1 h at -78 °C. To the above mixture was added dropwise triethylamine (5.16 g, 51 mmol, 5 equiv) at -78 °C. After addition, the resulting mixture was warmed up to room temperature and stirred for additional lh at RT. Then methyl 2-(triphenyl-
223
5UB5TITUTE SHEET (RULE 26) lambda5-phosphanylidene)acetate (4.09 g, 12.23 mmol, 1.2 equiv) was added at RT and the resulting mixture was stirred for 2 hrs. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography with PE/ ethyl acetate (5: 1) to give 19.11 (1.8 g, 70.6%) as a light yellow oil. (ES, m/z): 251 (M+H+).
[00447] Synthesis of compound 19.12. To a solution of 19.11 (800 mg, 3.196 mmol, 1 equiv) in methanol (10 mL) was added palladium on carbon (10%, 200 mg) under nitrogen. Hydrogen (g, 3 atm) was introduced in and degassed three times. The resulting mixture was stirred at RT for 2h. The solids were filtered out and washed with MeOH. The filtrate was concentrated under vacuum. This resulted in 19.12 (400 mg, 77.3%) of as a colorless oil. (ES, m/z): 163 (M+H+).
[00448] Synthesis of compound 19.13. To a stirred solution of 19.12 (400 mg, 2.466 mmol, 1 equiv), benzyl 5-methyl-2H-pyrazole-3-carboxylate (639.98 mg, 2.96 mmol, 1.2 equiv) and triphenylphosphine (970.32 mg, 3.699 mmol, 1.5 equiv) in tetrahydrofuran (20 mL) was added dropwise diisopropyl azodicarboxylate (748.06 mg, 3.699 mmol, 1.5 equiv) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography with PE/ ethyl acetate (3 : 1) to 19.13 (600 mg, 67.5%) as a light yellow oil. (ES, m/z): 361 (M+H+).
[00449] Synthesis of compound 19.14. To a solution of 19.13 (600 mg, 1.665 mmol, 1 equiv) in methanol (20 mL) was added palladium on carbon (10%, 200 mg) under nitrogen. Hydrogen (g, 3 atm) was introduced in and degassed three times. The resulting mixture was stirred at RT for 2h. The solid was filtered out and washed with MeOH. The filtrate was concentrated under vacuum. This resulted in 19.14 (400 mg, 88.9%) as a light yellow oil. (ES, m/z): 271 (M+H+).
[00450] Synthesis of compound 19.15. To a stirred solution of 19.14 (224.99 mg, 0.832 mmol,
3 equiv) and 19.10 (150 mg, 0.277 mmol, 1 equiv) in N,N-dimethylformamide (5 mL) were added HATU (211.01 mg, 0.555 mmol, 2 equiv) and DIEA (179.31 mg, 1.387 mmol, 5 equiv) under nitrogen. The reaction mixture was irradiated with microwave radiation for lh at 120 °C. The resulting mixture was concentrated under vacuum. The residue was purified by reverse flash with the following conditions: column, C18; mobile phase, water (10% NH4HCO3) and ACN (20% ACN up to 50% in 15 min); UV detection at 254/220 nm to yield 19.15 (120 mg, 54.6%) as a light yellow solid. (ES, m/z): 793 (M+H ).
[00451] Synthesis of compound 1-143. To a stirred solution of methyl 19.15 (120 mg, 0.151 mmol, 1 equiv) in a mixed methanol (10 mL) and water (2 mL) was added lithium hydroxide (18.12 mg, 0.757 mmol, 5 equiv). The resulting mixture was stirred overnight at room temperature. The mixture was concentrated under reduced pressure. The mixture was diluted with water and acidified to pH 3 with 1 N hydrochloric acid. The mixture was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep Cl 8 column, 30* 150mm, 5um; mobile phase, water (0.1% FA) and acetonitrile (12% acetonitrile up to 37% in 7 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 1-143 (72.5 mg, 61.5%) as an off-white solid. (ESI): 390 (M/2+H+). 1H NMR (400 MHz, DMSO-d6) d 12.80 (br s, 2H), 11.98 (br s, 1H), 8.03-7.92 (m, 4H), 7.70 (dd, 2H), 7.43 (dd, 2H), 7.31 (br s, 2H), 6.57 (s, 1H), 6.56 (s, 1H), 6.01- 5.92 (m, 2H), 4.84-4.83 (m, 4H), 4.70 (t, 2H), 4.54 (q, 2H), 3.68 (t, 2H), 3.33 (t, 2H), 2.18-2.13 (m, 8H), 1.67-1.60 (m, 2H), 1.33 (t, 3H).
EXAMPLE 20. Synthesis of (2-[2-[5-([5-carbamoyl-l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazol-2- yl]carbamoyl)-3-methylpyrazol-l-yl]ethoxy]ethoxy)acetic acid, 1-144
Figure imgf000226_0001
Figure imgf000227_0001
[00452] Synthesis of compound 20.1. To a stirred solution of fert-butyl 2-(2-(2- hydroxyethoxy)ethoxy)acetate (440 mg, 2 mmol, 1 equiv), benzyl 5-methyl-2H-pyrazole-3- carboxylate (518.4 mg, 2.4 mmol, 1.2 equiv) and triphenylphosphine (786 mg, 3 mmol, 1.5 equiv) in tetrahydrofuran (20 mL) was added dropwise diisopropyl azodicarboxylate (606 mg, 3 mmol, 1.5 equiv) at 0 °C under nitrogen atmosphere. After addition, the resulting mixture was stirred for 2 h at room temperature at room temperature. The mixture was concentrated under vacuum and the residue was purified by silica gel column chromatography with PE/ethyl acetate (3: 1) to give 20.1 (550 mg, 65.8%) as a light yellow oil. (ES, m/z): 441 (M+Na+).
[00453] Synthesis of compound 20.2. To a solution of 20.1 (550 mg, 1.314 mmol, 1 equiv) in methanol (20 mL) was added palladium on carbon (10%, 200 mg) under nitrogen. Hydrogen (g) was introduced in and degassed three times. The resulting mixture was stirred at RT for 2h. The solids were filtered out and the filtrate was concentrated under vacuum. This resulted in 20.2 (380 mg, 88%) as a light yellow oil. (ES, m/z): 329 (M+H+).
[00454] Synthesis of compound 20.3. To a stirred solution of 20.2 (182.23 mg, 0.555 mmol, 3 equiv) and 19.3 (100 mg, 0.185 mmol, 1 equiv) in N,N-dimethylformamide (5 mL) were added HATU (140.67 mg, 0.37 mmol, 2 equiv) and DIEA (119.54 mg, 0.925 mmol, 5 equiv) under nitrogen. The reaction mixture was irradiated with microwave radiation for lh at 120 °C. The resulting mixture was concentrated under vacuum. The residue was purified by reverse flash with the following conditions: column, C18; mobile phase, water (10% NTLHCCh) and ACN (20% ACN up to 50% in 15 min); UV detection at UV 254/220 nm. This resulted in 20.3 (50 mg, 31.8%) as a light yellow solid. (ES, m/z): 851 (M+H+).
[00455] Synthesis of compound 1-144. To a stirred solution of 20.3 (50 mg, 0.059 mmol, 1 equiv) in dichloromethane (5 mL) was added trifluoroacetic acid (1 mL) at 0 °C. The resulting mixture was stirred for lh at room temperature and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep C18 column, 30% 50mm, 5um; mobile phase, water (0.1% FA) and acetonitrile (15% acetonitrile up to 40% in 7 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 1-144 (24.7 mg, 52.9%) as an off- white solid. (ES, m/z): 398 (M/2+H+). ¾ NMR (400 MHz, DMSO-d6) d 12.81 (brs, 2H), 12.65 (br s, 1H), 8.03-7.92 (m, 4H), 7.71 (d, 2H), 7.43 (dd, 2H), 7.31 (br s, 2H), 6.58 (s, 1H), 6.56 (s, 1H), 6.02-5.91 (m, 2H), 4.92-4.83 (m, 4H), 4.70 (t, 2H), 4.54 (q, 2H), 3.96 (s, 2H), 3.73 (t, 2H), 3.61-3.50 (m, 4H), 2.13 (s, 6H), 1.27 (t, 3H).
EXAMPLE 21. Synthesis of 5-[2-[5-([5-carbamoyl-l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazol-2- yl]carbamoyl)-3-methylpyrazol-l-yl]ethoxy]pentanoic acid, 1-145
Figure imgf000229_0001
[00456] Synthesis of compound 21.1. To a stirred solution of dimethylsulfoxide (1.59 g, 20.4 mmol, 2 equiv) in dichloromethane (20 mL) was added dropwise oxalyl chloride (1.94 g, 15.287 mmol, 1.5 equiv) at -78 °C under nitrogen atmosphere. After 30 min, 3-(2- (benzyloxy)ethoxy)propan-l-ol (2.142 g, 10.2 mmol, 1 equiv) was added at -78 °C and the resulting mixture was stirred for 1 hr at -78 °C. To the above mixture was added dropwise triethylamine (5.16 g, 51 mmol, 5 equiv) at -78 °C via syringe. The resulting mixture was warmed to room temperature and stirred for additional 2 hrs. Methyl 2-(triphenyl-lambda5- phosphanylidene)acetate (4.09 g, 12.23 mmol, 1.2 equiv) was added and stirred for 2 hrs. The reaction was quenched with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography with PE/ ethyl acetate (5: 1) to give 21.1 (1.7 g, 63.2%) as a light yellow oil. (ES, m/z): 265 (M+H+).
[00457] Synthesis of compound 21.2. To a solution of 21.1 (900 mg, 3.405 mmol, 1 equiv) in methanol (20 mL) was added palladium on carbon (10%, 200 mg) under nitrogen. Then hydrogen (g, ~3 atm) was introduced in and degassed three times. The resulting mixture was stirred at RT for 2 hrs. The solids were filtered out and the filtrate was concentrated under vacuum. This resulted in 21.2 (450 mg, 75%) as a light yellow oil. (ES, m/z): 177 (M+E1+).
[00458] Synthesis of compound 21.3. To a stirred solution of 21.2 (450 mg, 2.554 mmol, 1 equiv), benzyl 5-methyl-2H-pyrazole-3-carboxylate (662.67 mg, 3.064 mmol, 1.2 equiv) and triphenylphosphine (1 g, 3.831 mmol, 1.50 equiv) in tetrahydrofuran (20 mL) was added dropwise diisopropyl azodicarboxylate (774.58 mg, 3.831 mmol, 1.50 equiv) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography with PE/ ethyl acetate (3 : 1) to give 21.3 (900 mg, 94.1%) as a light yellow oil. (ES, m/z): 375 (M+H+).
[00459] Synthesis of compound 21.4. To a solution of 21.3 (900 mg, 2.404 mmol, 1 equiv) in methanol (15 mL) was added palladium on carbon (10%, 200 mg) under nitrogen. Then hydrogen (g, ~3 atm) was introduced in and degassed three times. The resulting mixture was stirred at RT for 2h. The solids were filtered out and washed with MeOH and the filtrate was concentrated under vacuum. This resulted in 21.4 (600 mg, 87.8%) as a light yellow oil. (ES, m/z): 285 (M+H+).
[00460] Synthesis of compound 21.5. To a stirred solution of 21.4 (236.67 mg, 0.832 mmol, 3 equiv) and 19.3 (150 mg, 0.277 mmol, 1 equiv) in N,N-dimethylformamide (5 mL) were added HATU (211.01 mg, 0.555 mmol, 2 equiv) and DIEA (179.31 mg, 1.387 mmol, 5 equiv) under nitrogen. The final reaction mixture was irradiated with microwave radiation for lh at 120 °C. The resulting mixture was concentrated under vacuum and the residue was purified by reverse flash with the following conditions: column, C 18; mobile phase, water (10% NH4HCO3) and ACN (20% ACN up to 50% in 15 min); UV detection at 254/220 nm to yield 21.5 (100 mg, 44.7%) as an off-white solid. (ES, m/z): 807 (M+TC).
[00461] Synthesis of compound 1-145. To a stirred solution of 21.5 (100 mg, 0.124 mmol, 1 equiv) in a mixed methanol (5 mL), tetrahydrofuran (5 mL) and water (2 mL) was added lithium hydroxide (14.88 mg, 0.62 mmol, 5 equiv). The resulting mixture was stirred overnight at room temperature. The mixture was concentrated under reduced pressure. The mixture was diluted with water and acidified to pH 3 with 1 N hydrochloric acid. The mixture was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep Cl 8 column, 30* 150, 5um; mobile phase, water (0.1% FA) and acetonitrile (12% acetonitrile up to 40% in 7 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 1-145 (50.4 mg; 51.3%) as an off-white solid. (ES, m/z) 397 (M/2+H+). ¾ NMR (300 MHz, DMSO-d6) d 12.80 (br s, 1H), 8.03-7.93 (m, 4H), 7.72 (d, 2H), 7.44 (d, 2H), 7.32 (br s, 2H), 6.58 (s, 1H), 6.57 (s, 1H), 6.03-5.92 (m, 2H), 4.85-4.84 (m, 4H), 4.70 (t, 2H), 4.55 (q, 2H), 3.68 (t, 2H), 3.32 (t, 2H), 2.17-2.14 (m, 8H), 1.44- 1.42 (m, 4H), 1.29 (t, 3H).
EXAMPLE 22. Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl- lH-pyrazole-5-carboxamido)-7-(3-hydroxypropoxy)-lH-benzo[d]imidazol-l-yl)but-2-enyl)- 7-(3-morpholinopropoxy)-lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5- carboxamide, 1-146
Figure imgf000232_0001
.
Figure imgf000233_0001
1-146
[00462] Synthesis of compound 22.1. To a stirred solution of tert-butyl N-[(2E)-4-aminobut- 2-en-l-yl] carbamate (0.93 g, 4.993 mmol, 1.00 equiv) in n-butyl alcohol (20 mL) was added sodium bicarbonate (0.84 g, 9.999 mmol, 2.00 equiv) and DIEA (3.22 g, 24.914 mmol, 4.99 equiv). After 10 min, 3-[3-[(tert-butyldimethylsilyl)oxy]propoxy]-4-chloro-5-nitrobenzamide (1.94 g, 4.988 mmol, 1.00 equiv) was added and the resulting mixture was stirred overnight at 120oC under nitrogen. The mixture was cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure to yield 22.1 (2.5 g, 76.5%) as a yellow solid. (ES, m/z): 539 (M+H+).
[00463] Synthesis of compound 22.2. To a stirred solution of 22.1 (2.40 g, 4.455 mmol, 1.00 equiv) in dimethyl formamide (40 mL) was added tin (II) chloridedihydrate (5.03 g, 22.275 mmol, 5.00 equiv). The resulting mixture was stirred overnight at room temperature. The mixture was diluted with water. The pH value of the solution was adjusted to 9 with sat. sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure to afford 22.2 (1.8 g, 54%) as a brown oil. (ES, m/z): 395 (M+H+).
[00464] Synthesis of compound 22.3. To a stirred solution of 22.2 (1.80 g, 4.563 mmol, 1.00 equiv) in methanol (20 mL) was added cyanogen bromide (0.726 g, 6.845 mmol, 1.5 equiv). The resulting mixture was stirred for 3 h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, water (0.1% FA) and ACN (48% ACN up to 59% in 10 min); UV detection at 254/220 nm. This resulted in 22.3 (1.3 g, 67.85%) as a brown oil. (ES, m/z): 420 (M+H+).
[00465] Synthesis of compound 22.4. To a stirred mixture of 22.3 (1.30 g, 3.099 mmol, 1.00 equiv) and 2-ethyl-5-methylpyrazole-3 -carboxylic acid (954.8 mg, 6.2 mmol, 2 equiv) in dimethyl formamide (20 mL) was added HATU (2.356 g, 6.2 mmol, 2.00 equiv) and DIEA (2.00 g, 15.495 mmol, 5.00 equiv) under nitrogen. The resulting mixture was stirred overnight at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, water (0.1% FA) and ACN (41% ACN up to 50% in 10 min); UV detection at 254/220 nm to yield 22.4 (1.2 g, 62.7%) as a brown oil. LC-MS (ES, m/z): 692 (M+H+).
[00466] Synthesis of compound 22.5. To a stirred mixture of 22.4 (1.2 g, 1.302 mmol, 1.00 equiv) in methanol (15 mL) and water (5 mL) was added lithium hydroxide (156.24 mg, 6.51 mmol, 5.00 equiv). The resulting mixture was stirred overnight at room temperature. The mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 22.5 (500 mg, 69.16%) of as a white solid. (ES, m/z): 556 (M+H+).
[00467] Synthesis of compound 22.6. To a solution of 22.5 (500 mg, 0.9 mmol, 1 equiv) in di chi orom ethane (10 mL) was added trifluoroacetic acid (3 mL) at 0 °C. The resulting solution was stirred overnight at room temperature. After completion, the mixture was concentrated under reduced pressure to give 22.6 (500 mg, crude) as a brown oil. (ES, m/z): 456 (M+H+).
[00468] Synthesis of compound 22.7. To a solution of 22.6 (500 g crude, 0.9 mmol, 1 equiv) in n-butyl alcohol (20 mL) was added sodium bicarbonate (151.2 mg, 1.8 mmol, 2 equiv) and DIEA (580.5 mg, 4.5 mmol, 5 equiv). After 10 min, 4-chloro-3-[3-(morpholin-4-yl)propoxy]-5- nitrobenzamide (309.6 mg, 0.9 mmol, 1 equiv) was added and the resulting solution was stirred overnight at 120 °C. The mixture was cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, water (10% NH4HCO3) and ACN (34% ACN up to 49% in 10 min); UV detection at 254/220 nm. This resulted in 22.7 (200 mg, 29%) as a yellow oil. (ES, m/z) 382 (M/2+TE).
[00469] Synthesis of compound 22.8. To a solution of 22.7 (200 mg, 0.262 mmol, 1 equiv) in MeOH (10 mL), water (5 mL) and ammonium hydroxide (2 mL) was added sodium hyposulfite (456.7 mg, 2.62 mmol, 10 equiv) at room temperature. The resulting mixture was stirred overnight at ambient temperature. After starting material was consumed completely, the mixture was concentrated under vacuum. The residue was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 22.8 (150 mg, 78.2%) as a brown solid. (ES, m/z): 367 (M/2+H+).
[00470] Synthesis of compound 22.9. To a solution of 22.8 (150 mg, 0.205 mmol, 1.00 equiv) in methanol (20 mL) was added cyanogen bromide (43.05 mg, 0.41 mmol, 2 equiv). The resulting mixture was stirred for 3 h at room temperature. After completion, the reaction mixture was concentrated under vacuum to give 22.9 (200 mg, crude) as a brown solid. (ES, m/z): 758 (M+H+). [00471] Synthesis of compound 1-146. To a solution of 22.9 (200 mg crude, 0.205 mmol, 1.00 equiv) and 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (38.13 mg, 0.246 mmol, 1.2 equiv) in dimethyl formamide (10 mL) was added DIEA (132.25 mg, 1.025 mmol, 5.00 equiv) and HATU (93.48 mg, 0.246 mmol, 1.2 equiv). The resulting mixture was stirred for 3 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep- HPLC with the following conditions: Column, XSelect CSH Prep OBD Cl 8 Column 19*250mm, 5um; mobile phase, water (0.1% FA) and ACN (12% ACN up to 33% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 1-146 (21.2 mg, 11.6%) as a white solid. (ES, m/z): 448 (M/2+FC); 1H-NMR (400 MHz, DMSO-d6) d 12.74 (br s, 2H), 8.02-7.93 (m, 2H), 7.65 (s, 2H), 7.40-7.25 (m, 4H), 6.51 (s, 1H), 5.88-5.70 (m, 2H), 4.97-4.86 (m, 4H), 4.62-4.48 (m, 3H), 4.08-3.90 (m, 4H), 3.60-3.35 (m, 8H), 2.89 (q, 2H), 2.42 (s, 3H), 2.37-2.12 (m, 7H), 1.74-1.58 (m, 4H), 1.29 (t, 3H), 1.08 (t, 3H).
EXAMPLE 23. Synthesis of 7-(2-aminoethoxy)-l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-l,3-benzodiazole-5-carboxamide, 1-147
Figure imgf000236_0001
23.1
Figure imgf000237_0001
23.7
Figure imgf000238_0001
23.8 1-147
[00472] Synthesis of compound 23.1. To a solution of 4-chloro-3-hydroxy-5-nitrobenzamide (1.08 g, 5 mmol, 1 equiv) and 2-bromoethan-l-ol (1.23 g, 10 mmol, 2 equiv) in dimethyl formamide (20 mL) was added potassium carbonate (2.07 g, 15 mmol, 3 equiv). The resulting mixture was stirred at lOOoC overnight. The mixture was cooled to RT, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with dichloromethane/methanol (10/1) to give 23.1 (700 mg, 53.85%) of 4-chloro-3-(2-hydroxyethoxy)-5-nitrobenzamide a yellow solid. (ES, m/z): 261 (M+H+).
[00473] Synthesis of compound 23.2. To a stirred solution of methyl (E)-l-(4-aminobut-2- en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate hydrochloride (1.14 g, 2.19 mmol, 1.0 equiv) in n-BuOH (20 mL) was added sodium bicarbonate (368 mg, 4.38 mmol, 2.0 equiv) and DIEA (1.41 g, 10.95 mmol, 5.0 equiv). After 10 min, 23.1 (570 mg, 2.19 mmol, 1.0 equiv) was added. The resulting mixture was stirred overnight at 120 °C under nitrogen. The mixture was cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 23.2 (770 mg, 56.6%) as a red solid. (ES, m/z): 621 (M+H+).
[00474] Synthesis of compound 23.3. To a stirred solution of 23.2 (770 mg, 1.13 mmol, 1.0 equiv) in methanol (20 mL) and water (5 mL) was added sodium dithionite (1.96 g, 11.3 mmol, 10.0 equiv) ammonia water (2 mL). The resulting mixture was stirred at RT for 2h. The mixture was diluted with water, extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. This resulted in 23.3 (587 mg, 88%) as an off- red solid. (ES, m/z): 591 (M+H+).
[00475] Synthesis of compound 23.4. To a solution of 23.3 (587 mg, 0.99 mmol, 1.0 equiv) in methanol (10 mL) was added cyanogen bromide (210 mg, 2 mmol, 2 equiv). The resulting mixture was stirred at RT for overnight. The resulting mixture was concentrated under reduced pressure to yield 23.4 (650 mg, crude) as a yellow solid. (ES, m/z): 616 (M+H+).
[00476] Synthesis of compound 23.5. To a solution of 4-ethyl-2-methyloxazole-5-carboxylic acid (232.5 mg, 1.5 mmol, 1.5 equiv), DIEA (645 mg, 5 mmol, 5.0 equiv) and HATU (570 mg, 1.5 mmol, 1.5 equiv) in dimethyl formamide (5 mL) was added 23.4 (650 mg crude, 0.99 mmol, 1.0 equiv). The resulting mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (39% ACN up to 51% in 10 min); Detector, UV 254/220 nm to yield 23.5 (385 mg, 51.13%) as a yellow solid. (ES, m/z): 753 (M+H+).
[00477] Synthesis of compound 23.6. To a solution of 23.5 (385 mg, 0.511 mmol, 1.0 equiv) in methanol (20 mL) and water (4 mL) was added lithium hydroxide (61.32 mg, 2.555 mmol, 5 equiv). The resulting mixture was stirred at 70 °C for 2 hour. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration to yield 23.6 (210 mg, 55.7%) of as a yellow solid. (ES, m/z): 739 (M+H+).
[00478] Synthesis of compound 23.7. To a solution of 23.6 (100 mg, 0.14 mmol, 1.0 equiv), DIEA (87 mg, 0.68 mmol, 5.0 equiv) and HATU (77 mg, 0.20 mmol, 1.5 equiv) in dimethyl formamide (5 mL) was added ammonium chloride (36 mg, 0.68 mmol, 5 equiv). The reaction mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (26% ACN up to 38% in 10 min); Detector, UV 254/220 nm. The crude product (70 mg) was purified by Prep-HPLC with the following conditions: Column SunFire prep C18 Column 30*150mm, 5um; mobile phase, Water (0.1% FA) and CAN (18% ACN up to 40% in 7 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 23.7 (33.3 mg, 32.23%) as a white solid. (ES, m/z): 738 (M+H+); ¾ NMR (400 MHz, DMSO-d6) d 12.74 (br s, 2H), 8.03-7.91 (m, 3H), 7.70 (dd, 1H), 7.64 (s, 1H), 7.47-7.32 (m, 4H), 6.55 (s, 1H), 6.09-5.97 (m, 1H), 5.88-5.79 (m, 1H), 5.02-4.78 (m, 4H), 4.53 (q, 2H), 4.08 (t, 2H), 3.65 (t, 2H), 2.83 (q, 2H), 2.40 (s, 3H), 2.12 (s, 3H), 1.27 (t, 3H), 1.02 (t, 3H).
[00479] Synthesis of compound 23.8. Into a 25-mL round-bottom flask, was placed 23.7 (20.00 mg, 0.027 mmol, 1.00 equiv) and TEA (13.72 mg, 0.136 mmol, 5 equiv) in THF (2 mL) at the temperature, then MsCl (31.05 mg, 0.271 mmol, 10 equiv) was added in protions. The resulting solution was stirred for 16 hr at room temperature. The reaction was monitored by LCMS. The reaction mixture was used directly in the next step. (ES, m/z): 816 (M+H+).
[00480] Synthesis of compound 1-147. N¾ (gas) was bubbled into THF (5 mL) at 0 °C for 30 min. 23.8 was added and stirred for 16 hr at 50 °C in a sealed tube. Then resulting mixture was cooled to room temperature and concentrated in vacuo. The crude product was purified by Prep- HPLC with the following conditions: Column: XSelect CSH Prep C18 OBD Column,, 5um, 19* 150mm ; Mobile Phase A: water (10% NH4HCO3), Mobile Phase B:ACN; Flow rate:25 mL/min; Gradient: 10 B to 40 B in 7 min; UV detection at 210/254 nm. This resulted in 1-147 (1.8 mg, 10%) as a white solid. (ES, m/z): 737 (M+H+).
EXAMPLE 24. Synthesis of (2E)-4-(5-[[(3E)-ll,21-dicarbamoyl-25-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1[69].0[2326].0|1327]]heptacosa-3,7,9,ll,13(27),19,21,23(26),24- nonaen-7-yl]carbamoyl]-3-methylpyrazol-l-yl)but-2-enoic acid, 1-148
Figure imgf000240_0001
Figure imgf000241_0001
[00481] Synthesis of compound 24.1. To a solution of 4-ethyl-2-methyloxazole-5-carboxylic acid (32.55 mg, 0.21 mmol, 1.0 equiv) and DIEA (135.45 mg, 1.05 mmol, 5.0 equiv) and HATU (119.7 mg, 0.315 mmol, 1.5 equiv) in dimethyl formamide (5 mL) was added (E)-l,15-diamino- 8,9, 16, 19-tetrahydro-7H-6, 10-dioxa-2, 14, 15 a, 19a-tetraazacyclopentadeca[3 ,2, 1 -cd: 8,9, 10- c'd']diindene-4, 12-dicarboxamide (100 mg, 0.21 mmol, 1.0 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 120 °C. The reaction was cooled to RT and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, water (0.1% FA) and ACN (37% ACN up to 51% in 10 min); UV detection at 254/220 nm. This resulted in 24.1 (30 mg, 23.3%) as a yellow solid. (ES, in :): 614 (M+H+).
[00482] Synthesis of compound 24.2. To a solution of l,4-dioxane-2,5-diol (600 mg, 4.996 mmol, 1.00 equiv) in dichloromethane (20 mL) was added tert-butyl 2-(triphenyl4ambda5- phosphanylidene)acetate (1.88 g, 4.996 mmol, 1.00 equiv). The resulting solution was stirred overnight at room temperature. The mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography with PE/ ethyl acetate (7: 1) to give 24.2 (650 mg, 82.3%) as a yellow solid. (ES, m/z): 159 (M+H+).
[00483] Synthesis of compound 24.3. To a solution of 24.2 (650.00 mg, 4.109 mmol, 1.00 equiv), (4-methoxyphenyl)methyl 5-methyl-2H-pyrazole-3-carboxylate (1.214 g, 4.931 mmol, 1.20 equiv) and PPh3 (1.616 g, 6.163 mmol, 1.50 equiv) in tetrahydrofuran (30 mL) was added dropwise DIAD (1246.25 mg, 6.163 mmol, 1.50 equiv) at 0 °C under nitrogen. The resulting mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography with PE/ ethyl acetate (20: 1) to afford 24.3 (300 mg, 19%) as a yellow solid. (ES, m/z): 409 (M+Na+).
[00484] Synthesis of compound 24.4. To a solution of 24.3 (300.00 mg, 0.776 mmol, 1.00 equiv) in tetrahydrofuran (10 mL) and water (2 mL) was added lithium hydroxide (18.6 mg, 0.776 mmol, 1 equiv). The resulting solution was stirred for lh at room temperature. The mixture was concentrated under reduced pressure. The pH value of the solution was adjusted to 3 with 1 N hydrochloric acid. The mixture was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. This resulted in 24.4 (100 mg, 48.4%) as colorless oil. (ES, m/z): 267 (M+H+).
[00485] Synthesis of compound 24.5. To a solution of 24.1 (70 mg, 0.1142 mmol, 1.00 equiv. and 24.4 (36.45 mg, 0.137 mmol, 1.2 equiv) in dimethyl formamide (4 mL) was added HATU (52 mg, 0.137 mmol, 1.2 equiv) and DIEA (73.53 mg, 0.57 mmol, 5.00 equiv) under nitrogen. The final reaction mixture was irradiated with microwave radiation for lh at 120 °C. The mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (31% ACN up to 43% in 10 min); UV detection at 254/220 nm to yield 24.5 (46 mg, 47.6%) as a yellow solid. (ES, m/z): 848 (M+H+).
[00486] Synthesis of compound 1-148. To a solution of24.5 (46 mg, 0.0543 mmol, l .OO equiv) in dichloromethane (5 mL) was added trifluoroacetic acid (1 mL). The resulting mixture was stirred for 2h at RT. The resulting mixture concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, water (0.1% FA) and ACN (21% ACN up to 33% in 10 min); UV detection at 254/220 nm. The crude product (14 mg) was purified by Prep-HPLC with the following conditions: Column XSelect CSH Prep C18 OBD Column 19*250mm, 5um; mobile phase, mobile phase, water (10% NH4HCO +O.I % NIT3Ή2O) and methanol (33% methanol up to 45% in 10 min). The product- containing fractions were collected and evaporated partially and lyophilized overnight to afford 1.6 mg of 1-148 as a white solid. (ES, m/z): 404 (M/2+H+).
EXAMPLE 25. Synthesis of (3Z)-7,27-bis(2-ethyl-5-methylpyrazole-3-amido)-17-(2,2,2- trifluoroacetyl)- 14,20-dioxa- 1,6,8,17,26- pentaazapentacyclo[19.6.1.1[6,9].0[25,281.0[13,29]]nonacosa-3,7,9,ll,13(29),21,23,25(28),26- nonaene-11,23-dicarboxamide, 1-149
Figure imgf000243_0001
Figure imgf000244_0001
Synthesis of compound 25.1.
[00487] To a stirred mixture of 2,2'-azanediyldiethanol (10.5 g, 0.1 mmol, 1 equiv) in tetrahydrofuran (200 mL) was added dropwise methyltrifluoroacetate (12.8 g, 0.1 mmol, 1 equiv) at 0 °C. The resulting solution was stirred overnight at room temperature. The mixture was concentrated under reduced pressure to yield 25.1 (24 g, crude) as a colorless oil. (ES, m/z): 202 (M+H+).
[00488] Synthesis of compound 25.2. To a stirred solution of 25.1 (5 g, 25 mmol, 1 equiv), 4- chloro-3-hydroxy-5-nitrobenzamide (10.8 g, 50 mmol, 2 equiv) and triphenylphosphine (20 g, 75 mmol, 3 equiv) in anhydrous tetrahydrofuran (200 mL) and was added dropwise diisopropyl azodicarboxylate (15 g, 75 mmol, 3 equiv) at 0 °C under nitrogen. The resulting solution was stirred overnight at room temperature. The mixture was concentrated under vacuum. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, water (10% NH4HC03) and ACN (20% ACN up to 50% in 15 min); UV detection at 254/220 nm to yield 25.2 (5 g, crude) as a dark yellow solid. (ES, m/z): 598 (M+H+).
[00489] Synthesis of compound 25.3. To a stirred solution of 25.2 (1.79 g, 3 mmol, 1 equiv) in dimethylsulfoxide (15 mL) and was added (E)-but-2-ene-l, 4-diamine dihydrochloride (711 mg, 4.5 mmol, 1.5 equiv) and DIEA (2.322 g, 18 mmol, 6 equiv). The final reaction mixture was irradiated with microwave radiation for 2 h at 120 °C. The mixture was cooled to room temperature and diluted with water. The precipitated solids were collected by filtration. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, water (10% NH4HC03) and ACN (30% ACN up to 50% in 15 min); UV detection at 254/220 nm. This resulted in 25.3 (560 mg, 30.6%) as an orange solid. (ES, m/z ): 612 (M+H+).
[00490] Synthesis of compound 25.4. To a stirred solution of 25.3 (560 mg, 0.917 mmol, 1 equiv) in dimethyl formamide (10 mL) and was added tin(II)chloridedihydrate (2.068 g, 9.17 mmol, 10 equiv). The mixture was cooled to room temperature and diluted with water. The pH value of the solution was adjusted to 9 with sat. sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure to yield 25.4 (600 mg, crude) as a light yellow oil. 5 (ES, m/z): 552 (M+H+);
[00491] Synthesis of compound 25.5. To a stirred solution of 25.4 (600 mg crude, 0.917 mmol, 1.00 equiv) in methanol (10 mL) and was added cyanogen bromide (486 mg, 4.585 mmol, 5 equiv). The resulting solution was stirred overnight at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, water (10% NH4HCO3) and ACN (20% ACN up to 40% in 10 min); UV detection at 254/220 nm. This resulted in 25.5 (200 mg, 36.2%) as a light yellow solid. (ES, m/z): 602 (M+H+).
[00492] Synthesis of compound 1-149. To a stirred solution of 25.5 (200 mg, 0.33 mmol, 1 equiv) and 2-ethyl-5-methylpyrazole-3-carboxylic acid (113 mg, 0.73 mmol, 2.2 equiv) in dimethyl formamide (5 mL) was added DIEA (213 mg, 1.65 mmol, 5 equiv) and HATU (314 mg, 8.25 mmol, 2.5 equiv) at room temperature under nitrogen. The final reaction mixture was irradiated with microwave radiation for lh at 120 °C. The mixture was cooled to RT and concentrated under reduced pressure. The crude product was pre-purified by reverse flash with the following conditions: Column, C18; mobile phase, water (10% NH4HC03) and ACN (20% ACN up to 40% in 20 min); UV detection at 254/220 nm. The crude product (40 mg) was re-purified by Prep-HPLC with the following conditions: Column, XSelect CSH Prep C18 OBD Column, 5um, 19*150 mm; mobile phase, water (0.1% FA ) and ACN (33% ACN up to 50% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 1-149 (8.8 mg, 12.1%) as an off-white solid. (ES, m/z): 437 (M/2+fT); ¾ NMR (400 MHz, DMSO-d6) d 7.67 (s, 2H), 7.40 (s, 2H), 6.49 (s, 1H), 6.48 (s, 1H), 5.90-5.70 (m, 2H), 5.08-4.90 (m, 4H), 4.60-4.35 (m, 8H), 4.18-4.05 (m, 4H), 1.95 (s, 3H), 1.94 (s, 3H), 1.23 (t, 6H).
EXAMPLE 26. Synthesis of (3Z)-7,27-bis(2-ethyl-5-methylpyrazole-3-amido)-14,20-dioxa- l,6,8,17,26-pentaazapentacyclo[19.6.1.1[69].0[25281.0|13291]nonacosa-
3, 7, 9, 11, 13(29), 21, 23, 25(28), 26-nonaene-ll, 23-dicarboxamide trifluoroacetate, Z-21
Figure imgf000246_0001
[00493] Synthesis of compound Z-21. To a stirred mixture of 1-149 (40 mg, 0.0458 mmol, 1 equiv) in dimethyl formamide (3 mL) and tetrahydrofuran (3 mL) was added ammonium hydroxide (3 mL). The resulting solution was stirred for 3 h at room temperature. The mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Prep OBD C18 Column, 5um, 30* 150mm; mobile phase, water (0.05% TFA) and ACN (15% ACN up to 35% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford Z-21 (2.8 mg, 7.87%) as an off-white solid. (ES, m/z)·. 778 (M+H+); 'H NMR (400 MHz, DMSO-d6) d 12.90 (br s, 2H), 8.76 (br s, 1H), 8.06 (s, 2H), 7.74 (s, 2H), 7.58-7.40 (m, 4H), 6.50 (s, 2H), 5.98-5.89 (m, 2H), 5.10-4.91 (m, 4H), 4.72-4.45 (m, 8H), 3.72-3.58 (m, 4H), 1.94 (s, 6H), 1.27 (t, 6H). EXAMPLE 27. ¾ NMR and Mass Spectral Data for Exemplary Compounds
[00494] ' H NMR and MS data for additional exemplary compounds which were prepared using similar synthetic methodology and routes are provided in Table 3, below.
Table 3. 1H NMR and Mass Spectral Data for Exemplary Compounds.
Figure imgf000247_0001
Figure imgf000248_0001
Figure imgf000249_0001
Figure imgf000250_0001
Figure imgf000251_0001
Figure imgf000252_0001
Figure imgf000253_0001
Figure imgf000254_0001
Figure imgf000255_0001
Figure imgf000256_0001
Figure imgf000257_0001
Figure imgf000258_0001
Figure imgf000259_0001
Figure imgf000260_0001
Figure imgf000261_0001
Figure imgf000262_0001
Figure imgf000263_0001
Figure imgf000264_0001
Figure imgf000265_0001
Figure imgf000266_0001
EXAMPLE 28. Synthesis of l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)- 7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-
Figure imgf000267_0001
[00495] Step 1. To a stirred mixture of l-[(2E)-4-aminobut-2-en-l-yl]-2-(2-ethyl-5- methylpyrazole-3-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazole-5-carboxamide
(38.2, 500 mg, 0.953 mmol, 1.00 equiv) and 4-chloro-3-methoxy-5-nitrobenzamide (219.78 mg, 0.953 mmol, 1 equiv) in dimethylsulfoxide (8 mL) was added DIEA (615.88 mg, 4.765 mmol, 5 equiv). The final reaction mixture was irradiated with microwave radiation for 6h at 120°C. The mixture was cooled to room temperature. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (lO oMLHCCh) and ACN (33% ACN up to 45% in 10 min); Detector, UV 254/220 nm. This resulted in 300 mg (43.85%) of 1- [(2E)-4-[(4-carbamoyl-2-methoxy-6-nitrophenyl)amino]but-2-en-l-yl]-2-(2-ethyl-5- methylpyrazole-3-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazole-5-carboxamide
(28.2) as a brown solid. LC-MS (ES, m/z): 719 (M+H+).
[00496] Step 2. To a stirred mixture of l-[(2E)-4-[(4-carbamoyl-2-methoxy-6- nitrophenyl)amino]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-7-[3-(morpholin-4- yl)propoxy]-l,3-benzodiazole-5-carboxamide (28.2, 300 mg, 0.417 mmol, 1 equiv) in methanol (10 mL) and water (5 mL) was added sodium dithionite (726.69 mg, 4.174 mmol, 10 equiv) and ammonia water (1 mL). The resulting solution was stirred for 3h at room temperature. The mixture was diluted with water, extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure to yield 120 mg (41.82%) of l-[(2E)-4-[(2-amino-4-carbamoyl-6-methoxyphenyl)amino]but-2-en-l-yl]-2-(2- ethyl-5-methylpyrazole-3-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazole-5- carboxamide (28.3) as a brown oil. LC-MS (ES, m/z): 689 (M+H+).
[00497] Step 3. To a stirred solution of l-[(2E)-4-[(2-amino-4-carbamoyl-6- methoxyphenyl)amino]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-7-[3-(morpholin-4- yl)propoxy]-l,3-benzodiazole-5-carboxamide (28.3, 100 mg, 0.145 mmol, 1 equiv) in methanol (5 mL) was added cyanogen bromide (30.45 mg, 0.29 mmol, 2 equiv). The resulting mixture was stirred for 3h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (10%NH4HCO3) and ACN (23% ACN up to 41% in 10 min); Detector, UV 254/220 nm to yield 40 mg (38.64%) of l-[(2E)-4-(2-amino-5-carbamoyl-7-methoxy-l,3-benzodiazol-l- yl)but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3- benzodiazole-5-carboxamide (28.4) as a brown solid. LC-MS (ES, m/z): 714 (M+H+).
[00498] Step 4. To a stirred mixture of l-[(2E)-4-(2-amino-5-carbamoyl-7-methoxy-l,3- benzodiazol-l-yl)but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-7-[3-(morpholin-4- yl)propoxy]-l,3-benzodiazole-5-carboxamide (28.4, 40 mg, 0.056 mmol, 1 equiv) and 4-ethyl-2- methyl-l,3-oxazole-5-carboxylic acid (8.69 mg, 0.056 mmol, 1 equiv) in dimethyl formamide (5 mL) was added HATU (21.31 mg, 0.056 mmol, 1 equiv) and DIEA (36.21 mg, 0.280 mmol, 5 equiv). The resulting mixture was stirred for 3 h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XSelect CSH Prep C18 OBD Column, 5um, 19* 150mm; mobile phase, Water (10%NH4HC03+0.1%NH3 H20) and ACN (26% ACN up to 32% in 7 min); UV detection at 254/220 nm to yield 7.7 mg (16.18%) of l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5- methylpyrazole-3-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2- (4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-methoxy-l,3-benzodiazole-5-carboxamide (1-150) as a white solid. LC-MS (ES, m/z): 851 (M+H+); ¾ NMR: (400 MHz, DMSO- 6) 512.48 (br s, 2H), 7.99 (s, 2H), 7.64 (d, 2H), 7.41-7.28 (m, 4H), 6.56 (s, 1H), 5.91-5.76 (m, 2H), 5.04-4.88 (m, 4H), 4.55 (q, 2H), 3.95 (t, 2H), 3.72 (s, 3H), 3.54-3.45 (m, 4H), 2.82 (q, 2H), 2.39 (s, 3H), 2.33-2.11 (m, 9H), 1.74-1.60 (m, 2H), 1.31 (t, 3H), 1.02 (t, 3H).
EXAMPLE 29. Synthesis of (3E)-25-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(2-ethyl-5- m ethyl pyrazole-3-amido)-N21- [2- [2-(2-hydroxyethoxy)ethoxy] ethyl]- 14, 18-dioxa- 1,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3, 7, 9, 11, 13(27), 19, 21, 23(26), 24-nonaene-ll, 21-dicarboxamide. 1-151
Figure imgf000269_0001
29.1 1-151
[00499] To a solution of (3E)-21-carbamoyl-7-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-25-(2- ethyl-5-methylpyrazole-3-amido)-14, 18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylic acid (29.1, 50 mg, 0.067 mmol, 1.00 equiv) and 2- [2-(2-aminoethoxy)ethoxy] ethanol (29.81 mg, 0.200 mmol, 3.00 equiv) in dimethyl formamide (5 mL) was added HATU (37.98 mg, 0.1 mmol, 1.50 equiv) and DIEA (43.04 mg, 0.333 mmol, 5.00 equiv). The resulting mixture was stirred for 2 h at RT. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridre Prep OBD C18 Column, 19*250mm, 5um; mobile phase, water (10% NH4HCO3+0.1%NH3 H2O) and methanol (55% methanol up to 80% in 9 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 14.3 mg (24.35%) of (3E)-25-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(2-ethyl-5- methylpyrazole-3-amido)-N21-[2-[2-(2-hydroxyethoxy)ethoxy]ethyl]-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3, 7, 9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l l, 21 -dicarboxamide (1-151) as a white solid. LC-MS (ES, m/z): 882 (M+H+); H-NMR (400 MHz, DMSO-t¾): d 12.85 (br s, 1H), 12.78 (br s, 1H), 8.53 (t, 1H), 7.98 (s, 1H), 7.69 (d, 2H), 7.50 (d, 2H), 7.36 (s, 1H), 6.55 (s, 1H), 5.62-5.48 (m, 2H), 4.95- 4.84 (m, 4H), 4.64-4.52 (m, 3H), 4.49-4.38 (m, 4H), 3.62-3.44 (m, 12H), 2.83 (q, 2H), 2.38 (s, 3H), 2.36-2.28 (m, 2H), 2.11 (s, 3H), 1.27 (t, 3H), 1.03 (t, 3H). EXAMPLE 30. Synthesis of (3E)-25-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(2-ethyl-5- methylpyrazole-3-amido)-N21-[2-(2-hydroxyethoxy)ethyl]-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3, 7, 9, 11, 13(27), 19, 21, 23(26), 24-nonaene-ll, 21-dicarboxamide. 1-152
Figure imgf000270_0001
[00500] To a solution of (3E)-21-carbamoyl-7-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-25-(2- ethyl-5-methylpyrazole-3-amido)-14, 18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylic acid (29.1, 50 mg, 0.067 mmol, 1.00 equiv) and 2-(2-aminoethoxy)ethanol (21.01 mg, 0.200 mmol, 3.00 equiv) in dimethyl formamide (5 mL) was added HATU(37.98 mg, 0.100 mmol, 1.50 equiv) and DIEA (43.04 mg, 0.333 mmol, 5.00 equiv). The resulting mixture was stirred for 2 h at RT. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XSelect CSH Prep C18 OBD Column, 19*250mm, 5um; mobile phase, water (10% NH4HCO +0. 1 %NHVH20) and methanol (55% methanol up to 85% in 7 min); The product- containing fractions were collected and evaporated partially and lyophilized overnight to afford 6.2 mg (11.11%) of (3E)-25-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(2-ethyl-5- methylpyrazole-3-amido)-N21-[2-(2-hydroxyethoxy)ethyl]-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3, 7, 9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l l, 21-dicarboxamide (1-152) as a white solid. LC- MS (ES, m/z): 838 (M+H+); H-NMR: (400 MHz, DMSO-tfc): d 12.45 (br s, 2H), 8.50 (t, 1H), 7.99 (s, 1H), 7.69 (d, 2H), 7.50 (d, 2H), 7.37 (s, 1H), 6.55 (s, 1H), 5.59-5.48 (m, 2H), 4.95-4.86 (m, 4H), 4.64-4.52 (m, 3H), 4.49-4.38 (m, 4H), 3.62-3.44 (m, 8H), 2.84 (q, 2H), 2.38 (s, 3H), 2.36- 2.28 (m, 2H), 2.09 (s, 3H), 1.28 (t, 3H), 1.03 (t, 3H).
EXAMPLE 31. Synthesis of (3E)-25-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(2-ethyl-5- methylpyrazole-3-amido)-N21-(2-hydroxyethyl)-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3, 7, 9, 11, 13(27), 19, 21, 23(26), 24-nonaene-ll, 21-dicarboxamide. 1-153
Figure imgf000271_0001
29.1 1-153
[00501] To a solution of (3E)-21-carbamoyl-7-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-25-(2- ethyl-5-methylpyrazole-3-amido)-14, 18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylic acid (29.1, 50.00 mg, 0.067 mmol, 1.00 equiv) and ethanolamine (12.20 mg, 0.200 mmol, 3.00 equiv) in dimethyl formamide (5 mL) was added HATU (37.98 mg, 0.100 mmol, 1.50 equiv) and DIEA (43.04 mg, 0.333 mmol, 5.00 equiv). The resulting mixture was stirred for 2 h at RT. The reaction was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (38% ACN up to 48% in 10 min); Detector, UV 254/220 nm. The crude product (40 mg) was purified by Prep-HPLC with the following conditions: Column, XBridge Shield RP18 OBD Column, 19*250mm, lOum; mobile phase, water (10% NH4HCO3+0.1%NH3 H2O) and methanol (50% methanol up to 80% in 9 min); The product- containing fractions were collected and evaporated partially and lyophilized overnight to afford 10.2 mg (19.2%) of (3E)-25-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(2-ethyl-5- methylpyrazole-3-amido)-N21-(2-hydroxyethyl)-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3, 7, 9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l l, 21 -dicarboxamide (1-153) as a white solid. LC-MS (ES, m/z): 794 (M+H+); H-MVIR: (300 MHz, DMSO-<¾): d 12.55 (br s, 2H), 8.44 (t, 1H), 7.97 (s, 1H), 7.69 (d, 2H), 7.51 (d, 2H), 7.41 (s, 1H), 6.54 (s, 1H), 5.58-5.51 (m, 2H), 4.95-4.86 (m, 4H), 4.74 (t, 1H), 4.62-4.41 (m, 6H), 3.61-3.52 (m, 2H), 3.42-3.35 (m, 2H), 2.84 (q, 2H), 2.38 (s, 3H), 2.36-2.28 (m, 2H), 2.08 (s, 3H), 1.27 (t, 3H), 1.03 (t, 3H).
EXAMPLE 32. Synthesis of 4-(5-[[(3E)-ll,21-dicarbamoyl-25-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3,7,9,ll513(27),19,21,23(26),24-nonaen-7-yl]carbamoyl]-3-methylpyrazol-l-yl)butanoic acid. 1-154
Figure imgf000272_0001
[00502] Step 1. To a solution of 3-methyl-lH-pyrazole-5-carboxylic acid (32.1, 6.3 g, 50 mmol, 1 equiv) and K2CO3 (13.8 g, 100 mmol, 2 equiv) in DMF (200 mL) was added BnBr (8.075 g, 47.5 mmol, 0.95 equiv). The resulting mixture was stirred at RT overnight. The mixture was diluted with water, and extracted with EtOAc. The combined organic layers were washed with brine, dried over NaiSCri and concentrated under reduced pressure. The residue was applied on a silica gel column with PE/EtOAc (3/1) to yield 7 g (68.22%) of benzyl 3-methyl-lH-pyrazole-5-carboxylate (32.2) as a white solid.
[00503] Step 2. To a stirred mixture of benzyl 3-methyl-lH-pyrazole-5-carboxylate (32.2, 1 g, 4.63 mmol, 1.00 equiv) and tert-butyl 4-hydroxybutanoate (740.8 mg, 4.63 mmol, 1 equiv) and triphenylphosphine (1.82 g, 6.945 mmol, 1.50 equiv) in tetrahydrofuran (20 mL) was added dropwise DIAD (1.4 g, 6.945 mmol, 1.50 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred overnight at 0°C under nitrogen atmosphere. The mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography with PE/ethyl acetate (10/1) to give 1 g (60.35%) of benzyl l-(4-tert-butoxy-4- oxobutyl)-3 -methyl- lH-pyrazole-5-carboxylate (32.3) as a white solid. LC-MS (ES, m/z): 359 (M+H+);
[00504] Step 3. To a solution of benzyl l-(4-tert-butoxy-4-oxobutyl)-3 -methyl- lH-pyrazole-5- carboxylate (32.3, 1 g, 2.79 mmol, 1.00 equiv) in methanol (20 mL) was added palladium carbon (10%, 200 mg). To the above hydrogen (g) was introduced in. The resulting mixture was stirred at RT for 2 h. The reaction mixture was filtered. The filtrate was concentrated under reduced pressure to yield 600 mg (80.15%) of l-(4-tert-butoxy-4-oxobutyl)-3-methyl-lH-pyrazole-5-carboxylic acid (32.4) as an off-white oil. LC-MS (ES, m/z): 269 (M+H+).
[00505] Step 4. To a solution of methyl (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5- carboxamido)-7-(2-morpholinoethoxy)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l -ethyl-3 - methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate (32.5, 70 mg, 0.114 mmol, 1.00 equiv) and l-(4-tert-butoxy-4-oxobutyl)-3-methyl-lH-pyrazole-5-carboxylic acid (32.4, 36.67 mg, 0.1368 mmol, 1.2 equiv) in dimethyl formamide (5 mL) was added HATU (52 mg, 0.1368 mmol, 1.2 equiv) and DIEA (73.53 mg, 0.57 mmol, 5.00 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 120°C. The mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (38% ACN up to 50% in 10 min); Detector, UV 254/220 nm to yield 30 mg (30.46%) of tert-butyl 4-(5- [[(3E)-1 l,21-dicarbamoyl-25-(4-ethyl-2 -methyl-1, 3-oxazole-5-amido)-14,18-dioxa-l, 6, 8, 24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9, 11, 13(27), 19, 21, 23(26), 24-nonaen-7-yl]carbamoyl]-3-methylpyrazol-l-yl)butanoate (32.6) as a yellow solid. LC-MS (ES, m/z): 864 (M+FE).
[00506] Step 5. To a solution of tert-butyl 4-(5-[[(3E)-l l,21-dicarbamoyl-25-(4-ethyl-2- methyl- 1 ,3 -oxazole-5-amido)-l 4, 18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9, 11, 13(27), 19, 21, 23(26), 24-nonaen-7-yl]carbamoyl]-3-methylpyrazol-l-yl)butanoate (32.6, 30 mg, 0.0347 mmol, 1.00 equiv) in dichloromethane (5 mL) was added trifluoroacetic acid (1 mL). The resulting mixture was stirred for 2h at RT. The resulting mixture concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1%FA) and ACN (23% ACN up to 35% in 10 min); Detector, UV 254/220 nm. The crude product (8 mg) was purified by Prep-HPLC with the following conditions: Column XSelect CSH Prep C18 OBD Column 19*250mm, 5um; mobile phase, mobile phase, Water (10%N¾HCO3H). l%NH3 H2O) and methanol (43% methanol up to 65% in 10 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 2.7 mg (9.64%) of 4-(5-[[(3E)-l l,21-dicarbamoyl-25-(4-ethyl-2- methyl-l,3-oxazole-5-amido)-14, 18-dioxa-l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A
[23,26] .0A[ 13 ,27]]heptacosa-3 ,7,9, 11,13 (27), 19,21 ,23 (26),24-nonaen-7-yl]carbamoyl]-3 - methylpyrazol-l-yl)butanoic acid (1-154) as a white solid. LC-MS (ES, m/z): 808 (M+H+); 1H NMR (400 MHz, DMSO -d6) d 12.28 (br s, 3H), 7.99 (s, 2H), 7.71 (s, 2H), 7.54-7.31 (m, 4H), 6.57 (s, 1H), 5.62-5.48 (m, 2H), 5.02-4.82 (m, 4H), 4.61-4.38 (m, 6H), 2.84 (q, 2H), 2.41-2.24 (m, 6H), 2.22-1.98 (m, 6H), 1.03 (t, 3H).
EXAMPLE 33. (E)-A-(5-carbamoyl-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5- carboxamido)-7-(2-hydroxyethoxy)-l//-benzo[</]iiiiidazol-l-yl)but-2-en-l-yl)-7-methoxy- l//-benzo imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-155
Figure imgf000275_0001
33.3
Figure imgf000276_0001
[00507] Step 1. To a stirred solution of 4-chloro-3-hydroxy-5-nitrobenzamide (33.1, 5.00 g, 23.086 mmol, 1.00 equiv) and 2-bromoethanol (5.77 g, 46.172 mmol, 2.00 equiv) in DMF (50.00 mL) in a 100 mL 3-necked round-bottom flask, CS2CO3 (7.52 g, 23.086 mmol, 1.00 equiv) was added in portions at 50°C under nitrogen atmosphere. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.5% NH4HCO3), 10% to 50% gradient in 10 min; detector, UV 254 nm to yield 4-chloro- 3-(2-hydroxyethoxy)-5-nitrobenzamide (33.2, 3.5 g, 58.17%) (ES, m/z): [M+H]+ 261.0 as ayellow solid. [00508] Step 2. To a stirred solution of 4-chloro-3 -(2-hydroxy ethoxy)-5-nitrobenzamide (33.2, 1.80 g, 6.906 mmol, 1.00 equiv) and 4-[[(2E)-4-aminobut-2-en-l-yl]amino]-3-methoxy-5- nitrobenzamide (33.x, 2.90 g, 10.347 mmol, 1.50 equiv) in DMSO (20 mL), DIEA (3.58 g, 27.700 mmol, 4.01 equiv) was added. The mixture was stirred for overnight at 120 °C under nitrogen atmosphere. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.5 % NH3Ή2O), 10% to 50% gradient in 10 min; detector, UV 254 nm to yield 4-[[(2E)-4-[[4-carbamoyl-2-(2-hydroxyethoxy)- 6-nitrophenyl]amino]but-2-en-l-yl]amino]-3-methoxy-5-nitrobenzamide (33.3, 1.5g,43.05%)
(ES, m/z): [M+H]+ 505.2 as a yellow solid.
[00509] Step 3. To a stirred solution of 4-[[(2E)-4-[[4-carbamoyl-2-(2-hydroxyethoxy)-6- nitrophenyl]amino]but-2-en-l-yl]amino]-3-methoxy-5-nitrobenzamide (33.3, 1.50 g, 2.974 mmol, 1.00 equiv) and NazS204 (5.90 g, 33.887 mmol, 11.40 equiv) in MeOH (37.50 mL) in a 100 mL 3-necked round-bottom flask, ammonia (9.00 mL) was added in portions at room temperature under nitrogen atmosphere. The resulting mixture was filtered and the filter cake was washed with MeOH (3x5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (CH2Cl2:MeOH=5:l) to afford 3-amino-4-[[(2E)-4-[[2-amino-4-carbamoyl-6-(2- hydroxyethoxy)phenyl]amino]but-2-en-l-yl]amino]-5-methoxybenzamide (33.4, 260 mg,
19.67%) (ES, m/z): [M+H]+ 445.2 as a yellow solid.
[00510] Step 4. A solution of 3-amino-4-[[(2E)-4-[[2-amino-4-carbamoyl-6-(2- hydroxyethoxy)phenyl]amino]but-2-en-l-yl]amino]-5-methoxybenzamide (33.4, 260.00 mg, 0.585 mmol, 1.00 equiv) and BrCN (185.87 mg, 1.755 mmol, 3.00 equiv) in MeOH (2.00 mL) in an 8 mL vial was stirred for 2h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure to yield 2-amino-l-[(2E)-4-[2-amino-5- carbamoyl-7-(2-hydroxyethoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-7-methoxy-l,3- benzodiazole-5-carboxamide (33.5, 240 mg, 82.97%) (ES, m/z): [M+H]+ 495.2 as a white solid.
[00511] Into an 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed 4-ethyl-2 -methyl-1, 3-oxazole-5-carboxylic acid (125.50 mg, 0.809 mmol, 4.00 equiv), DIEA (261.36 mg, 2.022 mmol, 10.00 equiv), HATU (461.34 mg, 1.213 mmol, 6.00 equiv), 2- amino- 1 -[(2E)-4-[2-amino-5-carbamoyl-7-(2-hydroxyethoxy)- 1 ,3 -benzodiazol- 1 -yl]but-2-en- 1 - yl]-7-methoxy-l,3-benzodiazole-5-carboxamide (33.5, 100.00 mg, 0.202 mmol, 1.00 equiv) in NMP (2.00 mL). The resulting mixture was irradiated for 1.5h at 140 °C. The resulting mixture was washed with 3x5 mL of water. The crude product was purified by Prep-TLC with (DCM:MeOH=35: l). The crude product (60 mg) was purified by Prep-HPLC with the following conditions (Column: XBridge Prep Phenyl OBD Column, 19x 150 mm 5 um 13 nm ; Mobile Phase A: water (0.05% NH3H2O), Mobile Phase B: MeOH-HPLC; Flow rate: 25 mL/min; Gradient: 45% B to 60% B in 15 min; 254;220 nm; RT 13.10 min) to afford 1-155 (3.9 mg, 1.29%) as a white solid. LC-MS (ES, m .-): [M+H]+: 769.4 (ES, m/z): [M-H] :767.2
[00512] H-NMR: (400 MHz, DMSO-de, ppm ) 512.69 (s, 2H), 7.94 (s, 2H), 7.63 (s, 2H), 7.31 (s, 4H), 5.89-5.81 (m, 2H), 4.95-4.86 (m, 4H), 4.04-4.01 (t, 2H), 3.76 (s, 3H), 3.62-3.58 (m, 2H), 2.82-2.77 (m, 4H), 2.40-2.35 (d, 6H), 1.01-0.93 (t, 6H).
EXAMPLE 34. (E)- V-(5-carbamoyl-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5- carboxamido)-7-hydroxy- 1/i-benzo [d\ ini idazol- l-yl)but-2-en- l-yl)-7-methoxy-l/T- benzo[i/]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-12
Figure imgf000278_0001
[00513] Into a 8-mL sealed tube, was placed l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-methoxy-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-methoxy-l,3-benzodiazole-5-carboxamide (1-60, 140.00 mg, 0.190 mmol, 1.00 equiv), DCE (3.00 mL), and BCE in DCM (1 mL, 4 mol/L). The resulting solution was stirred for 12h at 80 °C under nitrogen atmosphere. The reaction was then quenched by the addition of 15 mL of water. The solids were collected by filtration. The crude product (8 mg) was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): Column, XBridge Prep C18 OBD Column, 19x150mm 5um; mobile phase, water (10 mmol/L NH4HCO3) and EtOH- (hold 58% Phase B in 7 min); Detector, UV to yield 5.6 mg of l-[(2E)-4-[5-carbamoyl-2-(4-ethyl- 2-methyl- 1 ,3 -oxazole-5-amido)-7-hydroxy- 1 ,3 -benzodiazol- 1 -yl]but-2-en- 1 -yl]-2-(4-ethyl-2- methyl-l,3-oxazole-5-amido)-7-methoxy-l,3-benzodiazole-5-carboxamide (1-12) as a white solid. LC-MS (ES, m/z): [M+H]+ = 725.3; 1H-NMR: (400 MHz, DMSO-d6, /¾wi): 57.90-7.95 (m, 1H), 57.75-7.82 (m, 1H), 57.62 (s, 1H), 57.40-7.42 (d, 1H), 57.31 (s, 1H), 57.11-7.18 (m,3H), 55.82-5.86 (m, 2H), 54.86-4.91 (m, 4H), 53.69-3.80 (d, 3H), 52.79-2.82 (t, 4H), 52.38-2.39 (d, 6H), 50.98-1.07 (m, 6H), 50.82-0.89 (m, 1H).
EXAMPLE 35. Synthesis of ([6-carbamoyl-3-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-
1.3-oxazole-5-amido)-7-methoxy-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-
1.3-oxazole-5-amido)-l ,3-benzodiazol-4-yl] oxy)acetic acid. 1-156
Figure imgf000279_0001
1-156 [00514] Step 1. To a stirred solution of tert-butyl 2-bromoacetate (38.76 mg, 0.199 mmol, 1.20 equiv) and DIEA (128.40 mg, 0.993 mmol, 6.00 equiv) in DMSO (2.00 mL) was added 1-[(2E)- 4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-hydroxy-l,3-benzodiazol-l-yl]but- 2-en- 1 -yl] -2-(4-ethyl -2-methyl- 1 ,3 -oxazole-5-amido)-7-m ethoxy- 1 ,3-benzodiazole-5- carboxamide (1-12, 120.00 mg, 0.166 mmol, 1.00 equiv) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3h at room temperature. The mixture was concentrated. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, NH4HC03 in water, 10% to 50% gradient in 10 min; detector, UV 254 nm to yield tert-butyl 2-([6-carbamoyl-3-[(2E)-4-[5-carbamoyl-2-(4-ethyl- 2-methyl- 1 ,3 -oxazole-5-amido)-7-m ethoxy- 1 ,3 -benzodiazol- 1 -yl]but-2-en- 1 -yl]-2-(4- ethyl -2 - methyl-1, 3-oxazole-5-amido)-l,3-benzodiazol-4-yl]oxy)acetate (35.1, 60 mg, 43.20%) as a white solid.
[00515] Step 2. To a stirred solution of tert-butyl 2-([6-carbamoyl-3-[(2E)-4-[5-carbamoyl-2- (4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-methoxy-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4- ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazol-4-yl]oxy)acetate (35.1, 60.00 mg, 0.072 mmol, 1.00 equiv) in DCM (10 mL) was added TFA (1.00 mL) in portions at room temperature under nitrogen atmosphere. The mixture was stirred for lh at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product (60 mg) was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): Column, XBridge Prep OBD C18 Column, 19*250mm, 5um; mobile phase, water (10MMOL/L NH4HC03) and ACN (hold 30% PhaseB in 14 min); Detector, UV 254nm.) to afford ([6-carbamoyl-3-[(2E)-4-[5- carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-methoxy-l,3-benzodiazol-l-yl]but-2-en- l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazol-4-yl]oxy)acetic acid (1-156, 4.6mg,8.22%) as a white solid. LC-MS (ES, m/z): 783 (M+H+); H- MR: (400 MHz, DMSO- d6). d 12.63 ( s, 1H), 7.98 (d, 2H), 7.79-7.60 (m, 2H), 7.31-7.12 (d, 4H), 5.97 (d, 2H), 5.07-4.85 (t, 4H), 4.43 (s, 2H), 3.80 (d, 3H), 2.78-2.68 (m, 4H), 2.49-2.35 (m, 6H), 1.00-0.79 (m, 6H).
EXAMPLE 36. (£)-2-((5-carbamoyl-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5- carboxamido)-7-(2-hydroxyethoxy)-l//-benzo[r/]imidazol-l-yl)but-2-en-l-yl)-2-(4-ethyl-2- methyloxazole-5-carboxamido)-l /-benzo[i/]imidazol-7-yl)oxy)acetic acid, 1-157
Figure imgf000281_0001
Figure imgf000282_0001
36.8 1-157
[00516] Step 1. To a stirred mixture of 2-(2-bromoethoxy)oxane (1.26 g, 6.002 mmol, 1.30 equiv) and K2C03(1.28 g, 9.234 mmol, 2.00 equiv) in DMF (20 mL) was added 4-chloro-3- hydroxy-5-nitrobenzamide (36.1, 1.00 g, 4.617 mmol, 1.00 equiv) at 25 °C under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 100 °C under nitrogen atmosphere. The resulting mixture was diluted with water (100 mL). The resulting mixture was extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine (2 x 100 mL), dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CTLCh / MeOH (80: 1) to afford 4-chloro-3-nitro-5-[2-(oxan-2-yloxy)ethoxy]benzamide (36.2, 1.1 g, 69.10%) as a yellow oil. LC-MS (ES, m/z): 345 (M+fT).
[00517] Step 2. A solution of 4-chloro-3-nitro-5-[2-(oxan-2-yloxy)ethoxy]benzamide (2.00 g, 5.801 mmol, 1.00 equiv), tert-butyl N-[(2E)-4-aminobut-2-en-l-yl]carbamate hydrochloride (36.2, 1.55 g, 6.962 mmol, 1.20 equiv) and DIEA (3.75 g, 29.007 mmol, 5.00 equiv) in DMSO (15 mL) was stirred for 2 h at 120 °C under nitrogen atmosphere. The resulting mixture was diluted with water (200 mL). The resulting mixture was extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with brine (2x200 mL), and dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 10% to 70% gradient in 30 min; detector, UV 254 nm to yield tert-butyl N-[(2E)-4-([4-
281
5U B5TITUTE SH EET (RU LE 26) carbamoyl-2-nitro-6-[2-(oxan-2-yloxy)ethoxy]phenyl]amino)but-2-en-l-yl]carbamate (36.3, 1.5g, 52.28%) as a yellow solid. LC-MS (ES, m/z): 495 (M+H+).
[00518] Step 3. A solution of tert-butyl N-[(2E)-4-([4-carbamoyl-2-nitro-6-[2-(oxan-2- yloxy)ethoxy]phenyl]amino) but-2-en-l-yl]carbamate (36.3, 3.00 g, 6.066 mmol, 1.00 equiv) and 4 N HC1 in 1,4-dioxane (15 mL) in DCM (30 mL) was stirred for 1 h at 25 °C. The precipitated solids were collected by filtration and washed with CH2CI2 (5 x 15 mL) to yield 4-[[(2E)-4- aminobut-2-en-l-yl]amino]-3-(2-hydroxyethoxy)-5-nitrobenzamide hydrochloride (36.4, 2. lg, 99.83%) as a yellow crude solid. LC-MS (ES, m/z): 311 (M+H+).
[00519] Step 4. A solution of 4-[[(2E)-4-aminobut-2-en-l-yl]amino]-3-(2-hydroxyethoxy)-5- nitrobenzamide hydrochloride (36.4, 1.00 g, 2.884 mmol, 1.00 equiv), butyl 2-(5 -carbarn oyl-2- chloro-3-nitrophenoxy)acetate (1.14 g, 3.461 mmol, 1.20 equiv) and DIEA (5.59 g, 43.256 mmol, 15.00 equiv) in DMSO (20 mL) was stirred for 1 h at 120 °C under nitrogen atmosphere. The resulting mixture was diluted with water (100 mL). The resulting mixture was extracted with EtOAc (3 x 150 mL). The combined organic layers were washed with brine (1 x 100 mL), and dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 10% to 50% gradient in 25 min; detector, UV 254/210 nm to yield butyl 2-(5-carbamoyl-2-[[(2E)-4-[[4-carbamoyl-2-(2-hydroxyethoxy)-6- nitrophenyl]amino]but-2-en-l-yl]amino]-3-nitrophenoxy)acetate (36.5, 530 mg, 30.40%) as a yellow solid. LC-MS (ES, m/z): 605 (M+H+).
[00520] Step 5. A solution of butyl 2-(5-carbamoyl-2-[[(2E)-4-[[4-carbamoyl-2-(2- hydroxyethoxy)-6-nitrophenyl]amino]but-2-en-l-yl]amino]-3-nitrophenoxy)acetate (36.5, 430.00 mg, 0.711 mmol, 1.00 equiv), ammonia (8.00 mL), Na2S204 (1238.33 mg, 7.112 mmol, 10.00 equiv) and H2O (4 mL) in MeOH (16 mL) was stirred for 2 h at 25 °C. The resulting mixture was diluted with water (30 mL), extracted with EtOAc (6 x 100 mL). The combined organic layers were washed with brine (1x30 mL), and dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, ACN in water, 0% to 50% gradient in 30 min; detector, UV 254/210 nm to yield butyl 2-(3-amino-2-[[(2E)- 4-[[2-amino-4-carbamoyl-6-(2-hydroxyethoxy)phenyl]amino]but-2-en-l-yl]amino]-5-
282
5U B5TITUTE SH EET (RU LE 26) carbamoylphenoxy)acetate (36.6, 100 mg, 25.82%) as a yellow solid. LC-MS (ES, m/z): 545 (M+H+).
[00521] Step 6. A solution of butyl 2-(3-amino-2-[[(2E)-4-[[2-amino-4-carbamoyl-6-(2- hy droxy ethoxy )phenyl] amino]but-2-en- 1 -yl] amino] -5 -carbarn oylphenoxy)acetate (36.6, 100.00 mg, 0.184 mmol, 1.00 equiv) and BrCN (58.35 mg, 0.551 mmol, 3.00 equiv) in MeOH (10 mL) was stirred for 16 h at 25 °C. The resulting mixture was concentrated under vacuum to yield butyl 2-([2-amino-3-[(2E)-4-[2-amino-5-carbamoyl-7-(2-hydroxyethoxy)-l,3-benzodiazol-l-yl]but-2- en-l-yl]-6-carbamoyl-l,3-benzodiazol-4-yl]oxy)acetate (36.7, 110 mg, 100.75%) as a yellow crude solid. LC-MS (ES, m/z): 595 (M+H+).
[00522] Step 7. Into a 10-mL sealed tube, was placed butyl 2-([2-amino-3-[(2E)-4-[2-amino-5- carbamoyl-7-(2-hydroxyethoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-6-carbamoyl-l,3- benzodiazol-4-yl]oxy)acetate (36.7, 90.00 mg, 0.151 mmol, 1.00 equiv), 4-ethyl-2-m ethyl- 1,3- oxazole-5-carboxylic acid (117.42 mg, 0.757 mmol, 5.00 equiv), HOBt (102.26 mg, 0.757 mmol, 5.00 equiv), EDC (117.48 mg, 0.757 mmol, 5.00 equiv), DMAP (92.45 mg, 0.757 mmol, 5.00 equiv) and DMF (5 mL). The final reaction mixture was irradiated with microwave radiation for 1 h at 120 °C. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 0% to 50% gradient in 25 min; detector, UV 254/210 nm to yield 2-([3-[(2E)-4-[7-(2-butoxy-2-oxoethoxy)-5-carbamoyl-2-(4- ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-6-carbamoyl-2-(4- ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazol-4-yl]oxy)ethyl 4-ethyl-2-methyl-l,3- oxazole-5-carboxylate (36.8, 90 mg, 59.11%) as a yellow solid. LC-MS (ES, m/z): 1006 (M+H+).
[00523] Step 8. Into a 25-mL round-bottom flask containing a solution of 2-([3-[(2E)-4-[7-(2- butoxy-2-oxoethoxy)-5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazol- l-yl]but-2-en-l-yl]-6-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazol-4- yl]oxy)ethyl 4-ethyl -2-methyl- 1, 3 -oxazole-5-carboxylate (36.8, 90.00 mg, 0.089 mmol, 1.00 equiv) in DCM (5 mL) was added TFA (1 mL) 0 °C via a syringe, then the reaction mixture was stirred at room temperature for 4h. The reaction mixture was concentrated and the residue was dissolved in THF (5 mL) and FLO (1.00 mL), LiOH (10.71 mg, 0.447 mmol, 5.00 equiv) was added. The resulting solution was stirred for 1 h at 25 °C. The resulting mixture was concentrated in vacuo. The crude product was purified by Prep-HPLC with the following conditions (SHIMADZU): Column, XSelect CSH Prep C18 OBD Column, 5um, 19* 150mm; mobile phase, water (0.1% NH4HCO3 + 0.1% NH3Ή2O) and ACN (10% Phase B up to 25% in 7 min); UV detection at 254/210 nm to yield 22.2 mg (30.53%) of ([6-carbamoyl-3-[(2E)-4-[5-carbamoyl-2- (4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(2-hydroxyethoxy)-l,3-benzodiazol-l-yl]but-2-en-l- yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazol-4-yl]oxy)acetic acid (1-157) as a white solid. LC-MS (ES, m/z): 813 (M+H+);
Figure imgf000285_0001
d 12.70 (br s, 2H), 8.01-7.95 (m, 2H), 7.65-7.63 (m, 2H), 7.32-7.25 (m, 4H), 6.04-5.91 (m, 2H), 5.00-4.92 (m, 4H), 4.66 (s, 2H), 4.02 (t, J= 4.8 Hz, 2H), 3.59 (t, J= 4.7 Hz, 2H), 2.83-2.75 (m, 4H), 2.39 (d, J= 5.4 Hz, 6H), 1.04 - 0.92 (m, 6H).
EXAMPLE 37. Synthesis of l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5- amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5- methylpyrazole-3-amido)-7-methoxy-l,3-benzodiazole-5-carboxamide. 1-114
Figure imgf000285_0002
NHBoc
37.7
37.6
Figure imgf000286_0001
Figure imgf000287_0001
1-114
[00524] Step 1. A solution of methyl 4-chloro-3-methoxy-5-nitrobenzoate (37.1, 10 g, 40.82 mmol, 1 equiv) in NH4OH (100 mL) was stirred overnight at 35 °C. The mixture was cooled to room temperature. The solid was collected by fdtration and dried in an oven at 45 °C to give 7.2 g (76.7%) of 4-chloro-3 -m ethoxy-5 -nitrobenzamide (37.2) as a white solid. LC-MS (ES, m/z): 231 (M+H+).
[00525] Step 2. To a solution of 4-chloro-3-methoxy-5-nitrobenzamide (37.2, 3 g, 13.04 mmol, 1.0 equiv) in DCE (80 mL) was added dropwise BBn (9.82 g, 39.12 mmol, 3 equiv) at 0 °C. The resulting mixture was stirred at 50 °C overnight. The mixture was cooled to RT and diluted with water. The solid was collected by fdtration and dried in an oven at 45 °C to give 2 g (71%) of 4- chloro-3-hydroxy-5-nitrobenzamide (37.3) as an off-white solid. LC-MS (ES, m/z): 217 (M+H+). [00526] Step 3. To a solution of 4-chloro-3-hydroxy-5-nitrobenzamide (37.3, 1.404 g, 6.5 mmol, 1.0 equiv), 3-morpholinopropan-l-ol (1.885 g, 13 mmol, 2 equiv) and PPI13 (3.406 g, 13 mmol, 2 equiv) in THF (40 mL) was added dropwise DIAD (2.626 g, 13 mmol, 2 equiv) at 0 °C. The resulting mixture was stirred at 50 °C overnight. The mixture was cooled to RT and concentrated under reduced pressure. The residue was applied on a silica gel column with DCM/MeOH (20/1) to afford 1.7 g (76%) of 4-chloro-3-(3-morpholinopropoxy)-5- nitrobenzamide (37.4) as an off-white solid. LC-MS (ES, m/z): 344 (M+H+).
[00527] Step 4. To a stirred solution of 4-chloro-3-[3-(morpholin-4-yl)propoxy]-5- nitrobenzamide (37.4, 6.2 g, 18.06 mmol, lequiv) in n-butyl alcohol (60 mL) was added sodium bicarbonate (13.02 g, 36.12 mmol, 2 equiv) and DIEA (11.64 g, 90.3 mmol, 5 equiv). After 10 min, was added tert-butyl N-[(2E)-4-aminobut-2-en-l-yl]carbamate (4.2 g, 19.86 mmol, 1.10 equiv). The resulting solution was stirred overnight at 120 °C. The mixture was cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with dichloromethane/methanol (10/1) to give 4.8 g (53.93%) of tert-butyl N- [(2E)-4-([4-carbamoyl-2-[3-(morpholin-4-yl)propoxy]-6-nitrophenyl]amino)but-2-en-l- yl]carbamate (37.5) as a dark yellow solid. LC-MS (ES, m/z): 494 (M+H+).
[00528] Step 5. To a stirred solution of tert-butyl N-[(2E)-4-([4-carbamoyl-2-[3-(morpholin-4- yl)propoxy]-6-nitrophenyl]amino)but-2-en-l-yl]carbamate (37.5, 4.80 g, 9.94 mmol, 1 equiv) in methanol (40 mL) and water (8 mL) was added sodium dithionite (10 g, 8.75 mmol, 5 equiv) and ammonia water (5 mL). The resulting solution was stirred for 3h at room temperature. The mixture was diluted with water, extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure to yield 3.74 g (81.1%) of tert-butyl N-[(2E)-4-([2-amino-4-carbamoyl-6-[3-(morpholin-4- yl)propoxy]phenyl]amino)but-2-en-l-yl]carbamate (37.6) as a light yellow solid. LC-MS (ES, m/z): 464 (M+H+).
[00529] Step 6. To a stirred solution of tert-butyl N-[(2E)-4-([2-amino-4-carbamoyl-6-[3- (morpholin-4-yl)propoxy]phenyl]amino)but-2-en-l-yl]carbamate (37.6, 3.74 g, 8.08 mmol, 1 equiv) in methanol (50 mL) was added cyanogen bromide (1.7 g, 16.16 mmol, 2 equiv). The resulting solution was stirred for 3 hours at room temperature. The resulting mixture was concentrated to yield 4 g (crude) of tert-butyl N-[(2E)-4-[2-amino-5-carbamoyl-7-[3-(morpholin-
4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate (37.7) as a dark yellow oil. LC-MS (ES, m/z): 489 (M+H+).
[00530] Step 7. To a stirred solution of tert-butyl N-[(2E)-4-[2-amino-5-carbamoyl-7-[3- (morpholin-4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate (37.7, 2.2 g, 4.51 mmol, 1 equiv) and 4-ethyl-2-methyl-l,3-oxazole-5-carboxamide (839 mg, 5.41 mmol, 1.2 equiv) in dimethyl formamide (15 mL) and was added DIEA (5.8 g, 45.1 mmol, 10 equiv) and HATU (2.055 g, 5.41 mmol, 1.2 equiv). The resulting solution was stirred overnight at RT. The resulting mixture was concentrated under vacuum. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (10%MTdTCO3) and ACN (60% ACN up to 80% in 7 min); UV detection at 254/220 nm to yield 1.5 g (53.23%) of tert-butyl N-[(2E)-4- [5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3- benzodiazol-l-yl]but-2-en-l-yl]carbamate (37.8) as a light yellow solid. LC-MS- (ES, m/z): 626 (M+H+);
[00531] Step 8. To a stirred solution of tert-butyl N-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-
1.3-oxazole-5-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l- yl]carbamate (37.8,600 mg, 0.96 mmol, 1 equiv) in dichloromethane (10 mL) was added trifluoroacetic acid (3 mL). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under vacuum to yield 560 mg (crude) of l-[(2E)-4-aminobut-2-en-l- yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazole-
5-carboxamide (37.9) as a brown oil. LC-MS (ES, m/z): 526 (M+H+).
[00532] Step 9. To a stirred solution of l-[(2E)-4-aminobut-2-en-l-yl]-2-(4-ethyl-2-m ethyl-
1.3-oxazole-5-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazole-5-carboxamide (37.9, 500 mg, 0.97 mmol, 1 equiv) in dimethylsulfoxide (8 mL) was added 4-chloro-3-methoxy-5- nitrobenzamide (370 mg, 1.2 mmol, 1.2 equiv) and DIEA (1.25 g, 9.7 mmol, 10 equiv). The final reaction mixture was irradiated with microwave radiation for 4h at 120°C. The mixture was cooled to room temperature. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water ( KAoNFLHCCh) and ACN (31% ACN up to 42% in 10 min); Detector, UV 254/220 nm. This resulted in 286 mg (41%) of l-[(2E)-4-[(4-carbamoyl-
288
5U B5TITUTE SH EET (RU LE 26) 2-methoxy-6-nitrophenyl)amino]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-[3- (morpholin-4-yl)propoxy]-l,3-benzodiazole-5-carboxamide (37.10) as a yellow solid. LC-MS- (ES, m/z): 720 (M+H+).
[00533] Step 10. To a stirred solution of l-[(2E)-4-[(4-carbamoyl-2-methoxy-6- nitrophenyl)amino]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-[3-(morpholin-4- yl)propoxy]-l,3-benzodiazole-5-carboxamide (37.10, 286 mg, 0.397 mmol, 1 equiv) in methanol (20 mL) and water (5 mL) was added sodium dithionite (2.27 g, 1.986 mmol, 5 equiv) and ammonia water (2 mL). The resulting solution was stirred for 3h at room temperature. The mixture was diluted with water, extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure to yield 175 mg (63.89%) of l-[(2E)-4-[(2-amino-4-carbamoyl-6-methoxyphenyl)amino]but-2-en-l-yl]-2-(4- ethyl-2-methyl-l,3-oxazole-5-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazole-5- carboxamide (37.11) as a light yellow solid. LC-MS (ES, m/z): 690 (M+E1+).
[00534] Step 11. To a stirred solution of l-[(2E)-4-[(2-amino-4-carbamoyl-6- methoxyphenyl)amino]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-[3- (morpholin-4-yl)propoxy]-l,3-benzodiazole-5-carboxamide (37.11, 175 mg, 0.254 mmol, 1 equiv) in methanol (10 mL) was added cyanogen bromide (53.34 mg, 0.508 mmol, 2 equiv). The resulting solution was stirred for 3 hours at room temperature. The resulting mixture was concentrated. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (10%NH4HCO3) and ACN (21% ACN up to 32% in 10 min); Detector, UV 254/220 nm. This resulted in 70 mg (38.55%) of 2-amino-l-[(2E)-4-[5-carbamoyl- 2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazol-l- yl]but-2-en-l-yl]-7-methoxy-l,3-benzodiazole-5-carboxamide (37.12) as a yellow solid. LC-MS (ES, m/z): 715 (M+H+).
[00535] Step 12. To a stirred solution of2-amino-l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl- l,3-oxazole-5-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l-yl]-7- methoxy-l,3-benzodiazole-5-carboxamide (37.12, 70 mg, 0.098 mmol, 1.00 equiv) and 2-ethyl-5- methylpyrazole-3 -carboxylic acid (22.79 mg, 0.147 mmol, 1.5 equiv) in dimethyl formamide (5 mL) and was added DIEA (63.21 mg, 0.49 mmol, 5 equiv) and HATU (41 mg, 0.108 mmol, 1.10 equiv). The resulting solution was stirred for 5h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Prep OBD C18 Column, 30* 150mm, 5um; mobile phase, Water (10%NH4HC03+0.1%N¾ H20) and ACN (20% ACN up to 40% in 7 min); UV detection at 254/220 nm to yield 11 mg (13.2%) of l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2- ethyl-5-methylpyrazole-3-amido)-7-methoxy-l,3-benzodiazole-5-carboxamide (1-114) as a white solid. LC-MS (ES, m/z): 851 (M+H+); ¾ NMR (400 MHz, DMSO -d6) d 13.70 (br s, 2H), 7.97 (s, 2H), 7.65 (dd, 2H), 7.39-7.27 (m, 4H), 6.50 (s, 1H), 5.84-5.77 (m, 2H), 4.99-4.79 (m, 4H), 4.53 (q, 2H), 3.95 (t, 2H), 3.72 (s, 3H), 3.54-3.45 (m, 4H), 2.88 (q, 2H), 2.43 (s, 3H), 2.29-2.15 (m, 6H), 2.13 (s, 3H), 1.71-1.62 (m, 2H), 1.28 (t, 3H), 1.09 (t, 3H).
EXAMPLE 38. Synthesis of l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5- amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-7-[3- (morpholin-4-yl)propoxy]-l,3-benzodiazole-5-carboxamide. 1-113
Figure imgf000291_0001
290
5UB5TITUTE SHEET (RULE 26)
Figure imgf000292_0001
[00536] Step 1. To a stirred mixture of tert-butyl N-[(2E)-4-[2-amino-5-carbamoyl-7-[3- (morpholin-4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate (37.5, 2 g, 4.093 mmol, 1 equiv) and 2-ethyl-5-methylpyrazole-3-carboxylic acid (0.63 g, 4.093 mmol, 1 equiv) in dimethyl formamide (20 mL) was added HATU (1.56 g, 4.093 mmol, 1 equiv) and DIEA (2.645 g, 20.467 mmol, 5 equiv). The resulting solution was stirred overnight at RT. The resulting mixture was concentrated under vacuum. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (W/oMLHCCb) and ACN (58% ACN up to 76% in 10 min); UV detection at 254/220 nm to yield 1.3 g (50.14%) of tert-butyl N-[(2E)- 4-[5-carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3- benzodiazol-l-yl]but-2-en-l-yl]carbamate (38.1) as a brown solid. LC-MS (ES, m/z): 625 (M+H+).
[00537] Step 2. To a stirred solution of tert-butyl N-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5- methylpyrazole-3-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l- yl] carbarn ate (38.1, 600 mg, 0.962 mmol, 1 equiv) in dichloromethane (10 mL) was added trifluoroacetic acid (2 mL). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under vacuum to yield 550 mg (crude) of l-[(2E)-4-aminobut-2-en-l- yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazole-5- carboxamide (38.2) as a brown oil. LC-MS (ES, m/z): 525 (M+H+).
[00538] Step 3. To a stirred solution of l-[(2E)-4-aminobut-2-en-l-yl]-2-(2-ethyl-5- methylpyrazole-3-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazole-5-carboxamide
(38.2, 230 mg, 0.438 mmol, 1 equiv) and potassium carbonate (181.77 mg, 1.315 mmol, 3 equiv) in dimethyl formamide (10 mL) was added 4-fluoro-3-nitrobenzamide (80.72 mg, 0.438 mmol, 1 equiv). The resulting mixture was stirred for 3 h at 70 °C. The mixture was cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure to yield 190 mg (54.61%) of l-[(2E)-4-[(4-carbamoyl-2- nitrophenyl)amino]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-7-[3-(morpholin-4- yl)propoxy]-l,3-benzodiazole-5-carboxamide (38.3) as a yellow solid. LC-MS (ES, m/z): 689 (M+LL).
[00539] Step 4. To a stirred mixture of l-[(2E)-4-[(4-carbamoyl-2-nitrophenyl)amino]but-2-en- l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazole-5- carboxamide (38.3, 190 mg, 0.276 mmol, 1 equiv) in methanol (10 mL) and water (2 mL) was added sodium dithionite (480.3 mg, 2.759 mmol, 10 equiv) and ammonia water (0.5 mL). The resulting solution was stirred for 2h at room temperature. The mixture was diluted with water, extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure to yield 100 mg (39.66%) of l-[(2E)-4- [(2-amino-4-carbamoylphenyl)amino]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-7-[3- (morpholin-4-yl)propoxy]-l,3-benzodiazole-5-carboxamide (38.4) as a brown oil. LC-MS (ES, m/z): 659 (M+H+).
[00540] Step 5. To a stirred solution of l-[(2E)-4-[(2-amino-4-carbamoylphenyl)amino]but-2- en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3- benzodiazole-5-carboxamide (38.4, 100 mg, 0.152 mmol, 1 equiv) in methanol (5 mL) was added cyanogen bromide (31.92 mg, 0.304 mmol, 2 equiv). The resulting solution was stirred for 2 hours at room temperature. The resulting mixture was concentrated. The cmde product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (10%NH4HCO3) and ACN (38% ACN up to 46% in 10 min); UV detection at 254/220 nm to yield 50 mg (48%) of l-[(2E)-4-(2-amino-5-carbamoyl-l,3-benzodiazol-l-yl)but-2-en-l-yl]-2-(2- ethyl-5-methylpyrazole-3-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazole-5- carboxamide (38.5) as a dark yellow oil. LC-MS (ES, m/z): 684 (M+H+).
[00541] Step 6. To a stirred mixture of l-[(2E)-4-(2-amino-5-carbamoyl-l,3-benzodiazol-l- yl)but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3- benzodiazole-5-carboxamide (38.5, 50 mg, 0.073 mmol, 1.00 equiv) and 4-ethyl-2-methyl-l,3- oxazole-5-carboxylic acid (11.35 mg, 0.073 mmol, 1 equiv) in dimethyl formamide (5 mL) was added HATU (27.80 mg, 0.073 mmol, 1 equiv) and DIEA (47.25 mg, 0.366 mmol, 5 equiv). The resulting solution was stirred for 5h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Prep C18 OBD Column, 19* 150mm, 5um; mobile phase, Water (10%NH4HC03+0.1%NH3 H20) and ACN (21% ACN up to 27% in 7 min); UV detection at 254/220 nm to yield 18.1 mg (30.24%) of l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-7- [3-(morpholin-4-yl)propoxy]-l,3-benzodiazole-5-carboxamide (1-113) as a white solid. LC-MS (ES, m/z): 821 (M+H+); ¾ NMR (400 MHz, DMSO -d6) d 12.12 (br s, 2H), 8.03-7.91 (m, 3H), 7.70 (dd, 1H), 7.63 (s, 1H), 7.46-7.28 (m, 4H), 6.56 (s, 1H), 6.08-5.98 (m, 1H), 5.79-5.68 (m, 1H), 4.94 (d, 2H), 4.79 (d, 2H), 4.55 (q, 2H), 3.99 (t, 2H), 3.54-3.44 (m, 4H), 2.81 (q, 2H), 2.41 (s, 3H), 2.33- 2.12 (m, 9H), 1.76-1.64 (m, 2H), 1.31 (t, 3H), 1.02 (t, 3H).
EXAMPLE 39: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(l-methyl-lH- indazole-3-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-methyl-lH- benzo[d]imidazol-2-yl)-l-methyl-lH-indazole-3-carboxamide, 1-103
Figure imgf000295_0001
[00542] Synthesis of 39.2. Into a 100-mL 3 -necked round-bottom flask purged and maintained with an inert atmosphere of argon, was placed methyl 4-chloro-3-methylbenzoate, 39.1 (10 g, 54.2 mmol, 1.00 equiv), H2SO4 (15.0 mL, 281 mmol, 5.20 equiv). The resulting solution was stirred for 20 min at 0 °C in a water/ice bath. To this was added HNC (10.0 mL, 223 mmol, 4.12 equiv). The resulting solution was allowed to react for an additional 60 min while the temperature was maintained at 0 °C in a water/ice bath. The reaction was then quenched by the addition of 50 mL of water. The resulting solution was extracted with 3x100 mL of EA concentrated under vacuum. The residue was applied onto a silica gel column with EA:PA=1 : 10 to yield 11 g (88.4%) of 39.2 as an off-white solid.
[00543] Synthesis of 39.3. Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of argon, was placed 39.2 (10 g, 43.6 mmol, 1.00 equiv), NELOH (100 mL). The resulting solution was stirred for 15 hr at 30 °C in an oil bath. The solids were collected by filtration. The resulting mixture was concentrated under vacuum to yield 8 g (85.6%) of 39.3 as an off-white solid.
[00544] Synthesis of 39.4. Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of argon, was placed 4-chloro-3 -methyl-5 -nitrobenzamide (1 g, 4.66 mmol, 1 equiv), DMSO (10 mL), tert-butyl (E)-(4-aminobut-2-en-l-yl)carbamate (0.9 g, 1.00 equiv), DIEA (1.8 g, 0.01 mol, 3.00 equiv). The resulting solution was stirred for 15 hr at 100 °C in an oil bath. The reaction was then quenched by the addition of 50 mL of water/ice. The solids were collected by filtration to yield 1.1 g (64.8%) of 39.4 as a yellow solid.
[00545] Synthesis of 39.5. Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of argon, was placed 39.4 (1.05 g, 2.88 mmol, 1.00 equiv), 1,4-dioxane/HCl (11 mL, 4M). The resulting solution was stirred for 5 hr at 20 °C. The resulting mixture was concentrated under vacuum to yield 0.8 g of 39.5 as a red solid.
[00546] Synthesis of 39.6. Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of argon, was placed methyl 4-chloro-3-nitrobenzoate (5 g, 23.0 mmol, 1.00 equiv), NELOH (50 mL). The resulting solution was stirred for 15 hr at 30 °C in an oil bath. The solids were collected by fdtration. The resulting mixture was concentrated under vacuum to yield 4.0 g (86.0%) of 39.6 as an off-white solid.
[00547] Synthesis of 39.7. Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of argon, was placed 39.5 (780 mg, 2.95 mmol, 1.00 equiv), DMSO (10 mL), 39.6 (590 mg, 2.95 mmol, 1.00 equiv), DIEA (1.15 g, 8.85 mmol, 3.00 equiv). The resulting solution was stirred for 15 hr at 100 °C in an oil bath. The reaction was then quenched by the addition of 50 mL of water/ice. The solids were collected by filtration. The resulting mixture was concentrated under vacuum to yield 750 mg (59.3%) of 39.7 as a yellow solid.
[00548] Synthesis of 39.8. Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of argon, was placed 39.7 (730 mg, 1.70 mmol, 1.00 equiv), HOAc (10 mL), Zn (890 mg, 13.6 mmol, 8.00 equiv). The resulting solution was stirred for 5 hr at 20 °C. The solids were filtered out. The resulting mixture was concentrated under vacuum. The residue was applied onto a silica gel column with dichloromethane/methanol (5:1) to yield 450 mg (71.7%) of 39.8 as a yellow solid.
[00549] Synthesis of 39.9. Into a 50 mL round-bottom flask was placed 39.8 (100 mg, 0.27 mmol, 1.00 equiv), BrCN (71.9 mg, 0.68 mmol, 2.5 equiv) and EtOH (2 mL). The resulting mixture was stirred for 15 h at 20
°C under N2 atmosphere. The precipitated solids were collected by filtration and washed methan ol (2 mL). The product was re-crystallized from DMF to afford 39.9 (11.5 mg) as a light pink solid. LCMS: (ES, m z): [M+H]+ 419; H- MR: (400MHz, DMSO , ppm): 57.50-8.00 (m, 6H), 57.07-7.46 (m, 4H), 56.50-7.00 (m, 3H), 55.80 (d, 1H), 55.31 (d, 1H), 54.66-4.83 (m, 4H), 52.25 (s, 3H).
[00550] Synthesis of 1-103: Into a 10-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed 1 -methyl- lH-indazole-3 -carboxylic acid (168 mg, 0.96 mmol, 5.00 equiv), DMF (3 mL), EDC (148 mg, 0.96 mmol, 5.00 equiv), HOAt (130 mg, 0.96 mmol, 5.00 equiv), DIEA (247 mg, 1.91 mmol, 10.0 equiv) and 39.1 (80 mg, 0.19 mmol, 1.00 equiv). The resulting solution was stirred for 15 hr at 60 °C. The reaction was then quenched by the addition of 10 mL of water. The solids were collected by filtration. The crude product (0.1 g) was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): Column, SunFire C 18 OBD Prep Column, 100A, 5 A pm, 19*250mm; mobile phase, Water (1% HAC) and ACN (55% Phase B up to 80% in 8 min) to yield 17.2 mg (12.2% yield) of 1-103 as an off- white solid. LCMS: (ES, m/z): [M+Hf 735; 1H-NMR: (400MHz, DMSO , ppm) 513.04 (d, 2H), 57.06-8.58 (m, 17H), 56.10 (s, 1H), 55.71 (s, 1H), 55.22 (s, 2H), 55.00 (s, 2H), 54.27 (d, 6H), 52.50 (s, 3H). Example 40: Synthesis of l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)butyl)-N-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3- dioxoisoindolin-4-yl)oxy)acetamido)ethyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxamide. 1-98
Figure imgf000298_0001
Figure imgf000299_0001
Figure imgf000300_0001
[00551] Synthesis of 40.2. To a stirred solution of methyl 4-fluoro-3-nitrobenzoate, 40.1, (10.0 g, 50.217 mmol, 1 equiv) and tert-butyl N-[(2E)-4-aminobut-2-en-l-yl]carbamate hydrochloride (12.30 g, 55.238 mmol, 1.1 equiv) in DMF (100 mL) was added K2CO3 (20.82 g, 150.650 mmol, 3.0 equiv). The resulting mixture was stirred for 4h at 70°C. The reaction was cooled to rt and diluted with water. The precipitated solids were collected by filtration. This resulted in 17 g (92.65%) of methyl 4-[[(2E)-4-[[(tert-butoxy)carbonyl]amino]but-2-en-l-yl]amino]-3- nitrobenzoateas a yellow solid. LC-MS (ES, m/z): 753 (2M+Na+).
[00552] Synthesis of 40.3. To a stirred solution of methyl 4-[[(2E)-4-[[(tert- butoxy)carbonyl]amino]but-2-en-l-yl]amino]-3-nitrobenzoate, 40.2, (10 g, 27.368 mmol, 1 equiv) in DMF (80 mL) was added SnCL 2¾0 (30.88 g, 136.842 mmol, 5.0 equiv). The resulting mixture was stirred overnight at 35°C. The reaction was with water and basified to pH 9 with saturated NaHCCh (aq.). The mixture was extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SC>4 and concentrated under reduced pressure. This resulted in 8.0 g (87.15%) of methyl 3-amino-4-[[(2E)-4-[[(tert-butoxy)carbonyl]amino]but-2-en-l- yl]amino]benzoateas an off-white solid. LC-MS (ES, m/z): 336 (M+H+).
[00553] Synthesis of 40.4. To a stirred solution of methyl 3-amino-4-[[(2E)-4-[[(tert- butoxy)carbonyl]amino]but-2-en-l-yl]amino]benzoate, 40.3, (8 g, 23.852 mmol, 1 equiv) in MeOH (100 mL) was BrCN (7.58 g, 71.555 mmol, 3.0 equiv). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. This resulted in 8 g (93.17%) of methyl 2-amino-l-[(2E)-4-[[(tert-butoxy)carbonyl]amino]but-2-en-l-yl]-lH- l,3-benzodiazole-5-carboxylate as an off-white solid. LC-MS (ES, m/z): 361 (M+H+).
[00554] Synthesis of 40.5. To a stirred solution of methyl 2-amino- l-[(2E)-4-[[(tert- butoxy)carbonyl]amino]but-2-en-l-yl]-lH-l,3-benzodiazole-5-carboxylate, 40.4, (8 g, 22.22 mmol, 1.00 equiv) and l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (5.13 g, 33.33 mmol, 1.5 equiv) in DMF (50 mL) were added HATU (12.67 g, 33.33 mmol, 1.5 equiv) and DIEA (8.6 g, 66.66 mmol, 3.00 equiv). The resulting mixture was stirred for 4h at room temperature. The reaction was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (45% ACN up to 54% in 10 min); Detector, UV 254/220 nm. This resulted in 9.5 g (86.2%) of methyl l-[(2E)-4-[[(tert-butoxy)carbonyl]amino]but-2-en-l-yl]-2-(l- ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxylateas an off-white solid. LC-MS (ES, m/z) 497 (M+H+). [00555] Synthesis of 40.6. To a stirred solution of methyl l-[(2E)-4-[[(tert- butoxy)carbonyl]amino]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxylate, 40.5, (9.5 g, 19.131 mmol, 1.00 equiv) in DCM (50 mL) were added TFA (10 mL). The resulting mixture was stirred for lh at room temperature. The mixture was concentrated under reduced pressure. This resulted in 9.5 g crude of methyl l-[(2E)-4-aminobut- 2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxylate as an off-white solid. LC-MS (ES, m/z): 397 (M+H+).
[00556] Synthesis of 40.7. To a stirred solution of methyl l-[(2E)-4-aminobut-2-en-l-yl]-2-(l- ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxylate, 40.6, (2 g, 5.05 mmol, 1.00 equiv) and 4-chloro-3-nitrobenzamide (1.515 g, 7.575 mmol, 1.50 equiv) in DMF (20 mL) was added K2CO3 (1.62 g, 11.754 mmol, 3.00 equiv). The resulting mixture was stirred overnight at 80°C. The reaction was cooled to rt and diluted with water. The precipitated solids were collected by fdtration. This resulted in 2 g (70.72%) of methyl l-[(2E)-4-[(4-carbamoyl-2- nitrophenyl)amino]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxylate as a yellow solid. LC-MS (ES, m/z): 561 (M+H+).
[00557] Synthesis of 40.8. To a stirred solution of methyl l-[(2E)-4-[(4-carbamoyl-2- nitrophenyl)amino]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxylate, 40.7, (700 mg, 1.249 mmol, 1 equiv) in DMF (20 mL) was added SnCh 2H2O (1.41 g, 6.244 mmol, 5.0 equiv). The resulting mixture was stirred overnight at 35°C. The reaction was diluted with water and basified to pH 9 with saturated NaHCCh (aq.). The mixture was extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2S04 and concentrated under reduced pressure. This resulted in 600 mg (90.56%) of methyl l-[(2E)-4-[(2-amino-4-carbamoylphenyl)amino]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxylate as an off-white solid. LC-MS (ES, m/z) 531 (M+H+).
[00558] Synthesis of 40.9. To a stirred solution of methyl l-[(2E)-4-[(2-amino-4- carbamoylphenyl)amino]but-2-en- 1 -yl] -2-(l -ethyl -3 -methyl - 1 H-pyrazole-5 -amido)- 1 H- 1 , 3 - benzodiazole-5-carboxylate, 40.8, (600 mg, 1.131 mmol, 1.00 equiv) in MeOH (20 mL) was added BrCN (360 mg, 3.393 mmol, 3 equiv). The resulting mixture was stirred for 4h at room temperature. The mixture was concentrated under reduced pressure. This resulted in 650 mg crude of methyl l-[(2E)-4-(2-amino-5-carbamoyl-lH-l,3-benzodiazol-l-yl)but-2-en-l-yl]-2-(l-ethyl-3- methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxylate as a light yellow solid. LC-MS (ES, m/z): 556 (M+H+).
[00559] Synthesis of 40.10 To a stirred solution of methyl l-[(2E)-4-(2-amino-5-carbamoyl- lH-l,3-benzodiazol-l-yl)but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxylate, 40.10, (650 mg, 1.170 mmol, 1.00 equiv) and l-ethyl-3-methyl-lH- pyrazole-5-carboxylic acid (360.73 mg, 2.340 mmol, 2.0 equiv) in DMF (10 mL) were added HATU (889.67 mg, 2.340 mmol, 2.0 equiv) and DIEA (453.61 mg, 3.510 mmol, 3.0 equiv). The resulting mixture was stirred for 4h at room temperature. The reaction was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2S04 and concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, Cl 8; mobile phase, water (0.1% FA) and ACN (56% ACN up to 64% in 10 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 700 mg (80.32%) of methyl 1-[(2E)- 4-[5-carbamoyl-2-(l -ethyl -3-methyl-lH-pyrazole-5-amido)- lH-1, 3-benzodiazol- l-yl]but-2-en-l - yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxylate as a white solid. LC-MS (ES, m/z): 692 (M+H+); ¾ NMR: ¾ NMR (400 MHz, DMSO- ) d 12.87 (s, 1H), 12.81 (s, 1H), 8.07-7.94 (m, 3H), 7.76-7.69 (m, 2H), 7.47-7.42 (m, 2H), 7.33 (s, 1H), 6.58 (s, 1H), 6.53 (s, 1H), 5.93-5.91 (m, 2H), 4.89-4.81 (m, 4H), 4.54 (q, 4H), 3.86 (s, 3H), 2.13 (s, 6H), 1.29 (t, 6H).
[00560] Synthesis of 40.11. To a solution of methyl l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3- amido)-l,3-benzodiazole-5-carboxylate, 4.10, (500 mg, 0.723 mmol, 1 equiv) in methanol (20 mL) and dichloromethane (10 mL) was added palladium on carbon (10%, 100 mg). To the above hydrogen (g) was introduced in. The resulting mixture was stirred at r.t for 2h. The solid was filtered out. The filtrate was concentrated under vacuum. This resulted in 400 mg (79.77%) of methyl l-[4-[5-carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]butyl]-2- (2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5-carboxylate as an off-white solid. LC- MS: (ES, m/z): 694 (M+H+). [00561] Synthesis of 40.12. To a stirred solution of methyl l-[4-[5-carbamoyl-2-(l-ethyl-3- methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]butyl]-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxylate, 40.11, (50 mg, 0.072 mmol, 1 equiv) in methanol (5 mL), tetrahydrofuran (5 mL) and water (2 mL) was added lithium hydroxide (6.90 mg, 0.288 mmol, 4 equiv). The resulting mixture was stirred for 2h at 60°C. The mixture was cooled to RT and concentrated under reduced pressure. The residue was diluted with water and acidified to pH 3 with 1 N hydrochloric acid. The precipitated solids were collected by filtration. This resulted in 32.4 mg (66.14%) of l-[4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-l-yl]butyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxylic acid as a white solid.LC-MS: (ES, m/z): 680 (M+H+); 'H NMR: ' ft NMR (400 MHz, DMSO-r/e) d 12.85 (br s, 3H), 8.08 (s, 1H), 7.99-7.96 (m, 2H), 7.86-7.76 (m, 2H), 7.58-7.55 (m, 2H), 7.33 (s, 1H), 6.60 (s, 2H), 4.57 (q, 4H), 4.23 (t, 4H), 2.10 (s, 6H), 1.94- 1.88 (m, 4H), 1.30 (t, 6H).
[00562] Synthesis of 40.13. To a stirred solution of l-[4-[5-carbamoyl-2-(l-ethyl-3-methyl- lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]butyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazole-5-carboxylic acid, 40.12, (200 mg, 0.294 mmol, 1.0 equiv) and tert- butyl N-(2-aminoethyl)carbamate (141.42 mg, 0.883 mmol, 3.0 equiv) in DMF (10 mL) were added HATU (167.81 mg, 0.441 mmol, 1.5 equiv) and DIEA (152.11 mg, 1.177 mmol, 4.0 equiv). The resulting mixture was stirred for 2h at room temperature. The reaction was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SCE and concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (47% ACN up to 57% in 8 min); Detector, UV 254/220 nm. This resulted in 200 mg (82.70%) of tert-butyl N-[2-[(l-[4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazol-l-yl]butyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-5- yl)formamido]ethyl]carbamate as an off-white solid. LC-MS (ES, m/z): 822 (M+H+).
[00563] Synthesis of 40.14 To a stirred solution of tert-butyl N-[2-[(l-[4-[5-carbamoyl-2-(l- ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]butyl]-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-5-yl)formamido]ethyl]carbamate, 4.13, (80 mg, 0.097 mmol, 1.00 equiv) in DCM (10 mL) was added HC1 in 1,4-dioxane (4 mol/L, 4.00 mL). The resulting mixture was stirred for 30 min at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD Column 30* 150mm, 5um; mobile phase, water (0.1% FA) and ACN (25% CAN up to 42% in 7 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 31.4 mg (42.02%) of N-(2-aminoethyl)-l-[4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazol-l-yl]butyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5- carboxamide as a white solid. LC-MS (ES, m/z): 722 (M+H+); 'H NMR (400 MHz, DMSO-t/ό) ό 8.71 (s, 1H), 8.70 (s, 0.8H), 7.97-7.96 (m, 3H), 7.76 (d, 2H), 7.54 (t, 2H), 7.34 (s, 1H), 6.59 (s, 2H), 4.59-4.54 (m, 4H), 4.27-4.26 (m, 4H), 3.46-3.44 (m, 2H), 2.89 (t, 2H), 2.10 (s, 6H), 1.94-1.87 (m, 4H), 1.30 (t, 6H).
[00564] Synthesis of 1-98. To a stirred solution of N-(2-aminoethyl)-l-[4-[5-carbamoyl-2-(l- ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]butyl]-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide hydrochloride, 40.14, (65 mg, 0.086 mmol, 1.00 equiv) and 2-[[2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-isoindol-4- yl]oxy]acetic acid (42.72 mg, 0.129 mmol, 1.50 equiv) in DMF (5 mL) were added HATU (48.89 mg, 0.129 mmol, 1.50 equiv) and DIEA (33.24 mg, 0.257 mmol, 3.00 equiv). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 OBD Column, lOum, 19*250mm; mobile phase, water (0.1% FA) and ACN (30% ACN up to 50% in 7 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 22.3 mg (25%) of l-[4-(5-[[2-(2-[[2- (2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-isoindol-4- yl]oxy]acetamido)ethyl]carbamoyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazol-l-yl)butyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5- carboxamide as a white solid. LC-MS (ES, m/z): 519 (M/2+H+); ' H NMR (400 MHz, DMSO-t/») S 12.80 (br s, 2H), 11.15 (s, 1H), 8.48 (s, 1H), 8.20-8.12 (m, 1H), 7.97-7.92 (m, 3H), 7.76-7.72 (m, 2H), 7.67-7.66 (m, 1H), 7.54-7.51 (m, 2H), 7.43-7.34 (m, 3H), 6.60 (s, 2H), 5.10 (q, 1H), 4.80 (s, 2H), 4.56 (q, 4H), 4.30-4.21 (m, 4H), 3.45-3.36 (m, 4H), 2.90-2.88 (m, 1H), 2.59-2.50 (m, 2H), 2.10 (s, 6H), 2.03-2.00 (m, 1H), 1.87-1.86 (m, 4H), 1.32 (t, 6H). Example 41: Synthesis of (E)-l-(4-(2-benzamido-5-carbamoyl-lH-benzo[d]imidazol-l- yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-7-methyl-lH- benzo [d] imidazole-5-carboxamide, 1-97
Figure imgf000306_0001
Figure imgf000307_0001
41.7 I -97
Synthesis of 41.1
[00565] To a stirred mixture of tert-butyl N-[(2E)-4-[(4-carbamoyl-2-methyl-6- nitrophenyl)amino]but-2-en-l-yl]carbamate, 39.4 (6 g, 16.74 mmol, 1 equiv) in HOAc (100 mL) in a 250 mL 3-necked round bottom flask, was added Zn (10.9 g, 167.40 mmol, 10 equiv) at room temperature. The resulting mixture was stirred for lh at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The resulting mixture was diluted with ethanol (100 mL). The mixture was basified to pH=8 with saturated Na2CC>3 (aq.). The organic layer was dried over anhydrous NaiSCL. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 41.1 (5.2 g, 94.67%) as a light brown solid.
Synthesis of 41.2
[00566] To a stirred mixture of 41.1 (5.2 g, 15.55 mmol, 1 equiv) in MeOH (100 mL) in a 250 mL 3-necked round bottom flask, was added BrCN (2.0 g, 19.02 mmol, 1.2 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, CH3CN in water, 10% to 100% gradient in 60 min; detector, UV 254 nm. This resulted in 41.2 (3.8 g, 66.71%) as a light yellow solid.
Synthesis of 41.3
[00567] To a stirred mixture of l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (1629.9 mg, 10.57 mmol, 1 equiv) and HATU (6029.9 mg, 15.86 mmol, 1.5 equiv) in DMF (100 mL) in a 250 mL 3-necked round bottom flask, were added DIEA (4099.2 mg, 31.72 mmol, 3 equiv) and 41.2 (3800 mg, 10.57 mmol, 1 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature under nitrogen atmosphere. The reaction was quenched with brine (100 mL) at room temperature. The resulting mixture was extracted with EtOAc (2x100 mL). The combined organic layers were washed with brine (3x100 mL), dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 10% to 100% gradient in 60 min; detector, UV 254 nm. This resulted in 41.3 (3100 mg, 57.26%) as a white solid.
Synthesis of 41.4
[00568] To a stirred mixture of 41.3 (3080 mg, 6.05 mmol, 1 equiv) in DCM (60 mL) in a 250 mL 3-necked round bottom flask, was added TFA (12 mL) at room temperature. The resulting mixture was stirred for 2h at room temperature. The resulting mixture was concentrated under reduced pressure. This resulted in 41.4 (3.2 g) as a light brown solid.
Synthesis of 41.5
[00569] To a stirred mixture of 41.4 (3.20 g, 6.281 mmol, 1 equiv) and 4-chloro-3- nitrobenzamide (1511.76 mg, 7.537 mmol, 1.2 equiv) in in a 250 mL 3-necked
Figure imgf000308_0001
round bottom flask, was added K2CO3 (1736.08 mg, 12.562 mmol, 2 equiv) at room temperature. The resulting mixture was stirred for overnight at 70 degrees C under nitrogen atmosphere. The reaction was quenched with brine (100 mL) at room temperature. The precipitated solids were collected by filtration and washed with water (2x20 mL) and MeOH (3x30 mL). The resulting solid was dried under vacuum. This resulted in 41.5 (2300 mg) as a yellow solid.
Synthesis of 41.6
[00570] To a stirred mixture of 41.5 (2.28 g, 4.11 mmol, 1 equiv) in HOAc (40 mL) in a 100 mL 3-necked round-bottom flask, was added Zn (2688.4 mg, 41.10 mmol, 10 equiv) at room temperature. The resulting mixture was stirred for lh at room temperature under nitrogen atmosphere. After filtration, the filter cake was washed with MeOH (2x20 mL). The filtrate was concentrated under reduced pressure until most of the solvent was removed, then the mixture was quenched with water (50 mL), basified with saturated K2CO3 (aq.) until pH=9. The precipitated solids were collected by filtration and washed with water (3x20 mL). The filter cake was washed with MeOH/DMF (100: 1) until no product left in the filter cake (detected by TLC). The filtrate was concentrated under reduced pressure. This resulted in 41.6 (1800 mg, 82.69%) as a light pink solid.
Synthesis of 41.7
[00571] To a stirred mixture of 41.6 (1780 mg, 3.36 mmol, 1 equiv) in MeOH (80 mL) in a 250 mL 3-necked round bottom flask, was added BrCN (391.6 mg, 3.70 mmol, 1.1 equiv) at room temperature. The resulting mixture was stirred for overnight at room temperature. The precipitated solids were collected by filtration and washed with MeOH (2x10 mL). The resulting solid was dried under vacuum. This resulted in a crude product of 41.7 (1200 mg, 64.38%) as a light pink solid. 41.7 (crude, 60 mg) was purified by Prep-HPLC with the following conditions: Column: Kinetex EVO C18 Column, 30*150 mm, 5 um; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 20% B to 40% B in 7 min; 254;220 nm; RT7.20 min. This resulted in 41.7 (11.8 mg) as a white solid. LC-MS: (ES, m/z): [M+H]+ 555.4; 1H-NMR: (400MHz, DMSO -d6, ppm)·. 512.87 (s, 1H), 7.95-7.86 (m, 3H), 7.78 (s, 1H), 7.67-
7.66 (d, 1H), 7.52 (s, 1H), 7.36-7.29 (m, 3H), 6.54-6.51 (d, 1H), 6.04-5.98 (m, 1H), 5.44-5.40 (t, 1H), 5.00 (s, 2H), 4.72 (s, 2H), 4.56-4.51 (m, 2H), 2.53-2.51 (m, 3H), 2.15 (s, 3H), 1.29-1.25 (t, 3H).
Synthesis of 1-97
[00572] To a stirred mixture of benzoic acid (44.04 mg, 0.361 mmol, 1 equiv) and HATU (205.67 mg, 0.541 mmol, 1.5 equiv) in DMF (4 mL) in an 8 mL vial, DIEA (139.82 mg, 1.082 mmol, 3 equiv) and 41.7 (200 mg, 0.361 mmol, 1 equiv) were added at room temperature. The resulting mixture was stirred for overnight at 80 °C under nitrogen atmosphere. The reaction was quenched with water (5 mL) at room temperature. The precipitated solids were collected by filtration and washed with water (2x5 mL). After concentration under reduced pressure, the residue was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD C18 Column 19*250 mm, 5 um; Mobile Phase A: water (0.05%NH3 H20), Mobile Phase B: ACN; Flow rate: 20 mL/min; Gradient: 30% B to 50% B in 8 min; 254/220 nm; Rt: 7.77 min) to 1-97 (9.9 mg, 4.17%) as a white solid. LC-MS: (ES, m/z): [M+H]+ 659.3; lH-NMR:(400MHz, DMSO- ek,ppm): 512.85 (s, 2H), 58.16-8.18 (d, 2H), 57.99-7.84 (m, 4H), 57.72-7.28 (m, 9H), 56.53 (s, 1H), 56.00-5.96 (d, 1H), 55.60-5.56 (d, 1H), 54.99-4.89 (m, 4H), 54.53-4.52 (d, 2H), 52.10-2.08 (d, 3H), 51.29-1.25 (t, 3H).
Example 42: Synthesis of (E)-8-ethyl-10,18-dimethyl-8,ll,12,13,14,15,28,31- octahydrobenzo[4,5]imidazo[l,2-a]benzo[4,5]imidazo[2,l-p]dipyrazolo[5,l-e:4',3'- l][l,3,6,15,17]pentaazacyclohenicosine-7,20(6H,21H)-dione, I-H6
Figure imgf000310_0001
42.2 42.3
Figure imgf000311_0001
[00573] Synthesis of 42.1. To a solution of l-fluoro-2- nitrobenzene (1.5 g, 10.64 mmol, 1 equiv) in DMSO (40 mL) was added (E)-tert-butyl 4- aminobut-2-enylcarbamate hydrochloride (3.54 g, 15.96 mmol, 1.5 equiv) and
K2CO3 (4.4 g, 31.929 mmol, 3 equiv) at r.t. The resulting solution was stirred overnight at 70°C under nitrogen. The mixture was cooled to r.t, diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over Na SCE and concentrated under reduced pressure. The residue was applied on a silica gel column with PE/EtOAc (3/1). This resulted in 2.5 g (76.52%) of (E)-tert-butyl 4-(2-nitrophenylamino)but-2- enylcarbamate as a yellow solid. LC-MS (ES, m/z): 330 (M+Na+).
[00574] Synthesis of 42.2. To a solution of (E)-tert-butyl 4-(2-nitrophenylamino)but-2- enylcarbamate, 42.1, (2.5 g, 8.14 mmol, 1 equiv) in DCM (40 mL) was added TFA (8 mL). The resulting solution was stirred overnight at room temperature. The mixture was concentrated under reduced pressure to give 2 g cmde of (E)-/Vi-(2-nitrophenyl)but-2-ene- 1,4- diamine as a brown oil. LC-MS (ES, m/z) 208 (M+H+).
[00575] Synthesis of 42.3. To a solution of (E)-A -(2-nitrophenyl)but-2-ene- 1,4-diamine, 42.2, (2 g crude, 8.14 mmol, 1 equiv) and l-fluoro-2- nitrobenzene (1.15 g, 8.14 mmol, 1 equiv) in DMSO (10 mL) was added K2CO3 (3.37 g, 24.42 mmol, 3 equiv). The resulting solution was stirred overnight at 70°C. The mixture was cooled to r.t, diluted with water, extracted with EtOAc. The combined organic layers were washed with brine, dried over NaiSCri and concentrated under reduced pressure. The residue was applied on a silica gel column with PE/EtOAc (5/1). This resulted in 1.5 g (56.18%) of (E)- N1, A -bis(2 -nitrophenyl)but-2-ene-l, 4-diamine as a yellow solid. LC-MS (ES, m å): 351 (M+Na+).
[00576] Synthesis of 42.4. To a solution of (E)-Ai,A#-bis(2-nitrophenyl)but-2-ene- 1,4- diamine, 42.3, (1.5 g, 4.57 mmol, 1 equiv) in DMF (20 mL) was added SnCh^ThO (10.33 g, 45.7 mmol, 10 equiv). The resulting solution was stirred overnight at room temperature. The mixture was diluted with water. The pH value of the solution was adjusted to 9 with sat. NaHCCri solution and extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2S04 and concentrated under reduced pressure. This resulted in 0.8 g (65.36%) of (E)- Ari,/V7 -(but-2-ene-l,4-diyl)dibenzene-l, 2-diamine as a yellow solid. LC-MS (ES, m/z): 269 (M+H+).
[00577] Synthesis of 42.5. To a solution of (E)-/V7,Ai'-(but-2-ene-l,4-diyl)dibenzene- 1,2- diamine (800 mg, 2.985 mmol, 1 equiv) in EtOH (20 mL) was added BrCN (950 mg, 8.955 mmol, 3 equiv). The resulting solution was stirred overnight at room temperature. The resulting mixture was concentrated under vacuum. This resulted in 1 g crude of (E)-l,T-(but-2-ene-l,4-diyl)bis(lH- benzo[d]imidazol-2-amine) as a yellow solid. LC-MS (ES, m/z): 319 (M+H+).
[00578] Synthesis of 1-116. To a solution of (E)-l, T-(but-2-ene-l,4-diyl)bis(lH- benzo[d]imidazol-2-amine), 42.5, (100 mg, 0.314 mmol, 1 equiv) and 4-(5-(5-carboxy-3-methyl- lH-pyrazol-l-yl)pentyl)-l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (109.4 mg, 0.314 mmol, 1 equiv) in NMP (4 mL) was added HATU (298.3 mg, 0.785 mmol, 2.5 equiv) and DIEA (202.53 mg, 1.57 mmol, 5 equiv). The final reaction mixture was irradiated with microwave radiation at 140°C for 1 hour. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Prep OBD C18 Column 30* 150mm, 5um; mobile phase, Water (0.01 NH4HCO3) and ACN (34% ACN up to 64% in 8 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 11.7 mg (6.79%) of (31E)-5-ethyl-7,16-dimethyl-2,5,6, 14, 15,20,22,29,34,41- decaazaheptacyclo[32.7.0.0A[4,8].0A[14,18].0A[21,29].0A[23,28].0A[35,40]]hentetraconta-
311
5UB5TITUTE SHEET (RULE 26) l(41),4(8),6,15,17,21,23(28),24,26,31,35,37,39-tridecaene-3,19-dione as an off-white solid. LC- MS (ESI): 631 (M+H+); 1H- MR (400 MHz, DMSO-i/g) d 12.71 (s, 1H), 12.67 (s, 1H), 7.49-7.47 (m, 2H), 7.41-7.34 (m, 2H), 7.22-7.17 (m, 4H), 6.53 (s, 1H), 5.95-5.89 (m, 1H), 5.69-5.65 (m, 1H), 4.95-4.84 (m, 4H), 4.72 (t, 2H), 4.45 (q, 2H), 2.74 (t, 2H), 2.18 (s, 3H), 2.15 (s, 3H), 1.74-1.65 (m, 2H), 1.48-1.35 (m, 2H), 1.32-1.21 (m, 5H).
Example 43: Synthesis of(E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-N-isopentyl-lH-benzo [d] imidazole-5-carboxamide, 1-94
Figure imgf000313_0001
[00579] Synthesis of 43.1. To a stirred solution of methyl l-[(2E)-4-[5-carbamoyl-2-(l-ethyl- 3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl- lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxylate, 40.10, (700 mg, 1.012 mmol, 1.00 equiv) in MeOH (20 mL), THF (20 mL) and H2O (8 mL) was added LiOH (96.94 mg, 4.048 mmol, 4.00 equiv). The resulting mixture was stirred for 2h at 60°C. The mixture was concentrated under reduced pressure. The residue was diluted with water and acidified to pH 3 with IN HC1 (aq.). The precipitated solids were collected by filtration to give 600 mg (85.6%) of l-[(2E)-4-[5-carbamoyl-
2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-
3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxylic acid as a white solid. LC-MS (ES, m/z) 678 (M+H+); ¾ NMR (300 MHz, DMSO-flfe) d 12.97-12.82 (m, 3H), 8.07-7.97 (m, 3H), 7.79-7.71 (m, 2H), 7.47-7.45 (m, 2H), 7.34 (s, 1H), 6.57 (s, 1H), 6.53 (s, 1H), 5.93-5.91 (m, 2H), 4.89-4.81 (m, 4H), 4.55 (q, 4H), 2.13 (s, 6H), 1.28 (t, 6H).
[00580] Synthesis of 1-94. To a solution of l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazole-5-carboxylic acid, 43.1, (50 mg, 0.074 mmol, 1 equiv) and 3- methylbutan-1 -amine (12.88 mg, 0.148 mmol, 2 equiv) in DMF (5 mL) was added HATU (42.08 mg, 0.111 mmol, 1.5 equiv) and DIEA (47.68 mg, 0.369 mmol, 5 equiv). The reaction mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 OBD Column 19*250mm, lOum; mobile phase, Water (0.1% FA) and ACN (30% ACN up to 51% in 10 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 21.8 mg (39.5%) of (E)-l- (4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l- yl)but-2-enyl)-2-( 1 -ethyl -3 -methyl - 1 H-pyrazole-5 -carboxamido)-N-i sopentyl - 1 H- benzo[d]imidazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 747 (M+H ); 1HNMR (400 MHz, DMSO-t/s) ό 12.81 (br s, 2H), 8.40 (s, 1H), 7.97-7.94 (m, 3H), 7.73-7.65 (m, 2H), 7.44 (d, 2H), 7.34 (s, 1H), 6.55 (s, 2H), 5.95-5.91 (m, 2H), 4.89-4.80 (m, 4H), 4.53 (q, 4H), 3.32-3.27 (m, 2H), 2.12 (s, 6H), 1.68-1.52 (m, 1H), 1.48-1.41 (m, 2H), 1.28 (d, 6H), 0.91 (t, 6H).
Example 44: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-N-pentyl-lH-benzo [d] imidazole-5-carboxamide, 1-93
Figure imgf000315_0001
[00581] To a stirred solution of l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH- l,3-benzodiazole-5-carboxylic acid, 43.1, (50.00 mg, 0.074 mmol, 1.00 equiv) and pentan-1- amine (19.29 mg, 0.221 mmol, 3.00 equiv) in DMF (5.00 mL) were added HATU (42.08 mg, 0.111 mmol, 1.50 equiv) and DIEA (28.61 mg, 0.221 mmol, 3.00 equiv). The resulting mixture was stirred overnight at 85°C under nitrogen. The mixture was cooled to rt and concentrated under reduced pressure. The residue was purified by Flash with the following conditions: Column, Sunfire Prep OBD C18 Column 19*250mm, lOum; mobile phase, water (0.1% FA) and ACN (38% ACN up to 45% in 10 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 30.2 mg (54.71%) of l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l- yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-N-pentyl-lH-l,3-benzodiazole-5- carboxamide as a white solid. LC-MS (ES, m/z): 747 (M+H+); ¾ NMR (300 MHz, DMSO-t/») d 12.81 (br s, 2H), 8.42 (t, 1H), 8.01-7.95 (m, 3H), 7.74-7.66 (m, 2H), 7.44 (d, 2H), 7.35 (s, 1H), 6.56 (s, 2H), 5.97-5.93 (m, 2H), 4.84-4.83 (m, 4H), 4.54 (q, 4H), 3.35-3.22 (m, 2H), 2.13 (s, 6H), 1.56-1.51 (m, 2H), 1.32-1.26 (m, 10H), 0.85 (t, 3H).
Example 45: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-N-(3-morpholinopropyl)-lH-benzo[d]imidazole-5-carboxamide, 1-92
Figure imgf000316_0001
43.1 1-92
[00582] To a solution of l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxylic acid, 43.1, (50 mg, 0.074 mmol, 1 equiv) and 3-(morpholin-4- yl)propan-l -amine (12.77 mg, 0.089 mmol, 1.20 equiv) in DMF (5 mL) was added HATU (42.08 mg, 0.111 mmol, 1.5 equiv) and DIEA (47.68 mg, 0.369 mmol, 5 equiv). The reaction mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 OBD Column 19*250mm, lOum; mobile phase, Water (0.1% FA) and ACN (12% ACN up to 31% in 10 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 23.4 mg (37.32%) of 1- [(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but- 2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-N-[3-(morpholin-4-yl)propyl]-lH-l,3- benzodiazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 804 (M+H+); 1HNMR (400 MHz, DMSO-i e) ό 12.78 (br s, 2H), 8.46 (s, 1H), 7.97-7.94 (m, 3H), 7.72-7.64 (m, 2H), 7.45-7.33 (m, 3H), 6.55 (s, 2H), 5.95-5.91 (m, 2H), 4.89-4.80 (m, 4H), 4.53 (q, 4H), 3.58-3.51 (m, 4H), 3.32- 3.27 (m, 2H), 2.40-2.30 (m, 6H), 2.12 (s, 6H), 1.74-1.65 (m, 2H), 1.28 (t, 6H).
Example 46: Synthesis of (E)-N-(3-(2-naphthamido)propyl)-l-(4-(5-carbamoyl-2-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-91
Figure imgf000317_0001
Synthesis of 46.1
[00583] To a solution of naphthalene-2-carboxylic acid (500 mg, 2.904 mmol, 1.00 equiv) and tert-butyl N-(3-aminopropyl)carbamate (607.18 mg, 3.485 mmol, 1.20 equiv) in DCM (10 mL) was added HATU (1.656 g, 4.356 mmol, 1.50 equiv) and DIEA (1.88 g, 14.519 mmol, 5.00 equiv). The resulting mixture was stirred at RT for 2 h. The mixture was diluted with water, extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SC>4 and concentrated under reduced pressure. The residue was applied on a silica gel column with PE/EtOAc (5/1). This resulted in 750 mg (78.64%) of tert-butyl N-[3-[(naphthalen-2-yl)formamido]propyl]carbamate as an off-white solid. LC-MS (ES, m/z): 329 (M+H+).
Synthesis of 46.2
[00584] To a stirred solution of tert-butyl N-[3-[(naphthalen-2- yl)formamido]propyl]carbamate, 46.1, (750 mg, 2.284 mmol, 1.00 equiv) and DCM (10 mL) was added TFA (2 mL). The reaction mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. This resulted in 600 mg crude of N-(3- aminopropyl)naphthalene-2-carboxamide as a brown solid. LC-MS (ES, m/z): 229 (M+H+).
Synthesis of 1-91
[00585] To a solution of l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxylic acid, 43.1, (100 mg, 0.148 mmol, 1 equiv) and N-(3- aminopropyl)naphthalene-2-carboxamide 46.2, (67.37 mg, 0.295 mmol, 2.00 equiv) in DMF (5 mL) was added HATU (84.16 mg, 0.221 mmol, 1.5 equiv) and DIEA (95.35 mg, 0.738 mmol, 5 equiv). The reaction mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 OBD Column 19*250mm, lOum; mobile phase, Water (0.1% FA) and ACN (40% ACN up to 50% in 10 min); UV detection at 254/220 nm. The product- containing fractions were collected and evaporated partially and lyophilized overnight to afford 31 mg (23.6%) of 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-[(2E)-4-[2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-5-([3-[(naphthalen-2-yl)formamido]propyl]carbamoyl)-lH-l,3-benzodiazol- l-yl]but-2-en-l-yl]-lH-l,3-benzodiazole-5-carboxamide as an off-white solid. LC-MS (ES, m/z): 445 (M/2+FC); 1HNMR (400 MHz, DMSO -d6) d 12.78 (br s, 2H), 8.70 (s, 1H), 8.58-8.45 (m, 2H), 8.02-7.93 (m, 7H), 7.73-7.59 (m, 4H), 7.45-7.42 (m, 2H), 7.32 (s, 1H), 6.56 (s, 2H), 5.95-5.91 (m, 2H), 4.89-4.80 (m, 4H), 4.54 (q, 4H), 3.41-3.34 (m, 4H), 2.12 (s, 6H), 1.86-1.80 (m, 2H), 1.28 (t, 6H).
Example 47: Synthesis of tert-butyl (E)-(2-(l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamido)ethyl)carbamate, 1-90
Figure imgf000319_0001
[00586] To a solution of l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxylic acid, 43.1, (100 mg, 0.148 mmol, 1 equiv) and tert-butyl 2- aminoethylcarbamate (28.42 mg, 0.178 mmol, 1.2 equiv) in DMF (5 mL) was added HATU (84.36 mg, 0.222 mmol, 1.5 equiv) and DIEA (95.46 mg, 0.74 mmol, 5 equiv). The reaction mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: XBridge Prep C18 OBD Column 19* 150mm, 5um; Water (0.01% NH4HCO3 + 0.1% NH3Ή2O) and ACN (28% ACN up to 45% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 21.2 mg (17.5%) of (E)-tert-butyl 2-(l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazol-l-yl)but-2-enyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxamido)ethylcarbamate as an off-white solid. LC-MS (ES, m/z): 820 (M+H+); 1HNMR (400 MHz, DMSO -d6) S 12.81 (s, 2H), 8.42 (s, 1H), 7.98-7.94 (m, 3H), 7.73- 7.65 (m, 2H), 7.46-7.28 (m, 3H), 6.88-6.80 (m, 1H), 6.55 (s, 2H), 5.95-5.91 (m, 2H), 4.89-4.80 (m, 4H), 4.54 (q, 4H), 3.28-3.21 (m, 2H), 3.18-3.02 (m, 2H), 2.12 (s, 6H), 1.37 (s, 9H), 1.28 (t, 6H). Example 48: Synthesis of tert-butyl N-[3-([l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-5-yl]formamido)propyl]carbamate, 1-89
Figure imgf000320_0001
43.1 1-89
[00587] To a solution of l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxylic acid, 43.1, (100 mg, 0.148 mmol, 1 equiv) and tert-butyl N-(3- aminopropyl)carbamate (51.42 mg, 0.295 mmol, 2.00 equiv) in DMF (5 mL) was added HATU (67.32 mg, 0.177 mmol, 1.2 equiv) and DIEA (95.35 mg, 0.738 mmol, 5 equiv). The reaction mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 OBD Column 19*250mm, lOum; mobile phase, Water (0.1% FA) and ACN (41% ACN up to 45% in 10 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 24.4 mg (18.79%) of tert-butyl N-[3-([l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazol-5-yl]formamido)propyl]carbamate as an off-white solid. LC-MS (ES, m/z): 834 (M+H+); 1HNMR (300 MHz, DMSO^) d 12.81 (br s, 2H), 8.39 (s, 1H), 7.98-7.94 (m, 3H), 7.73- 7.65 (m, 2H), 7.46-7.28 (m, 3H), 6.88-6.80 (m, 1H), 6.55 (s, 2H), 5.95-5.91 (m, 2H), 4.89-4.80 (m, 4H), 4.54 (q, 4H), 3.28-3.21 (m, 2H), 3.02-2.97 (m, 2H), 2.12 (s, 6H), 1.65-1.59 (m, 2H), 1.38 (s, 9H), 1.28 (t, 6H). Example 49: Synthesis of (E)-N-(3-benzamidopropyl)-l-(4-(5-carbamoyl-2-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-88
Figure imgf000321_0001
Synthesis of 49.1
[00588] To a stirred mixture of tert-butyl 3 -aminopropyl carbamate (870 mg, 5 mmol, 1 equiv) and DIEA (1.935 g, 15 mmol, 3 equiv) in DCM (20 mL) was added dropwise benzoyl chloride (840 mg, 6 mmol, 1.2 equiv) at 0°C. The resulting mixture was stirred for 1 h at 0°C. The mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over NaiSCri and concentrated under reduced pressure. This resulted in 0.9 g (64.75%) of tert-butyl 3-benzamidopropylcarbamate as a white solid. LC-MS (ES, m/z): 279 (M+H+).
Synthesis of 49.2
[00589] To a stirred solution of tert-butyl 3-benzamidopropylcarbamate, 49.1, (900 mg, 3.237 mmol, 1 equiv) in DCM (10 mL) was added TFA (2 mL) at room temperature. The resulting mixture was stirred for 2 h at ambient temperature. The mixture was concentrated under vacuum to provide 700 mg crude of N-(3-aminopropyl)benzamide trifluoroacetate as a brown oil. LC-MS (ES, m å): 179 (M+H+).
Synthesis of 1-88
[00590] To a stirred solution of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxylic acid, 43.1, (100 mg, 0.148 mmol, 1 equiv) in DMF (10 mL) was added DIEA (94.72 mg, 0.74 mmol, 5 equiv), N-(3-aminopropyl)benzamide, 49.2, (31.6 mg, 0.178 mmol, 1.2 equiv) and HATU (67.74 mg, 0.178 mmol, 1.2 equiv). The resulting mixture was stirred for 2 h at room temperature. The mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over NaiSCE and concentrated under reduced pressure. The crude product was purified by reverse phase flash with the following conditions: Column, SunfirePrep OBD C18 Column 19*250mm, lOum; mobile phase, water (0.1% FA) and ACN (35% ACN up to 42% in 12 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 33.5 mg (27%) of (E)-N-(3-benzamidopropyl)-l-(4-(5-carbamoyl-2-(l-ethyl- 3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-2-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide as a white solid. LC- MS (ES, m/z): 420 (M/2+H+); 1H-NMR (400 MHz, DMSO- ) ό 12.80 (br s, 2H), 8.54-8.47 (m, 2H), 7.97-7.94 (m, 3H), 7.86-7.84 (m, 2H), 7.73-7.67 (m, 2H), 7.55-7.33 (m, 6H), 6.57 (s, 2H), 5.93-5.89 (m, 2H), 4.85-4.81 (m, 4H), 4.53 (q, 4H), 3.30-3.21 (m, 4H), 2.13 (s, 6H), 1.82-1.75 (m, 2H), 1.28 (t, 6H).
Example 50: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-N-(l-((2-(2,6-dioxopiperidin-3-yl)- l,3-dioxoisoindolin-4-yl)oxy)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)-2-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-87
Figure imgf000323_0001
[00591] To a stirred solution of l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH- l,3-benzodiazole-5-carboxylic acid, 43.1, (100 mg, 0.148 mmol, 1 equiv) and N-(2-[2-[2-(2- aminoethoxy)ethoxy]ethoxy]ethyl)-2-[[2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH- isoindol-4-yl]oxy]acetamide (89.68 mg, 0.177 mmol, 1.2 equiv) in DMF (10 mL) were added HATU (67.32 mg, 0.177 mmol, 1.2 equiv) and DIEA (76.28 mg, 0.590 mmol, 4.0 equiv). The resulting mixture was stirred for 2h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 OBD Column, lOum, 19*250mm; mobile phase, water (0.1% FA) and ACN (28% ACN up to 35% in 15 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 22.3 mg (13%) l-[(2E)-4-(5-[[2-(2-[2-[2-(2-[[2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-
322
5U B5TITUTE SH EET (RU LE 26) isoindol-4-yl]oxy]acetamido)ethoxy]ethoxy]ethoxy)ethyl]carbamoyl]-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl)but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazole-5-carboxamide as a white solid.
LC-MS (ES, m/z): 581 (M/2+H+); ¾ NMR (400 MHz, DMSO -d6) S 12.80 (BR s, 2H), 11.15 (s, 1H), 8.48 (s, 1H), 8.02-7.94 (m, 4H), 7.81-7.77 (m, 1H), 7.70-7.66 (m, 2H), 7.49-7.33 (m, 5H), 6.55 (s, 2H), 5.92 (s, 2H), 5.11 (q, 1H), 4.83-4.78 (m, 6H), 4.53 (q, 4H), 3.53-3.41 (m, 16H), 2.90-2.89 (m, 1H), 2.62-2.50 (m, 2H), 2.12 (s, 6H), 2.05-2.01 (m, 1H), 1.27(t, 6H).
Example 51: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-N-(4-(2-((2-(2,6-dioxopiperidin-3- yl)-l,3-dioxoisoindolin-4-yl)oxy)acetamido)butyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-86
Figure imgf000324_0001
[00592] To a stirred solution of l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH- l,3-benzodiazole-5-carboxylic acid, 43.1, (50.0 mg, 0.074 mmol, 1 equiv) and N-(4-aminobutyl)- 2-[[2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-isoindol-4-yl]oxy]acetamide (32.66 mg, 0.081 mmol, 1.1 equiv) in DMF (5 mL) were added HATU (61.71 mg, 0.162 mmol, 2.2 equiv) and DIEA (47.68 mg, 0.369 mmol, 5.0 equiv). The resulting mixture was stirred for 2h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep Cl 8 OBD Column, lOum, 19*250mm; mobile phase, water (0.1% FA) and ACN (33% ACN up to 39% in 11 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 31.6 mg (40.33%) of l-[(2E)-4-(5-[[4-(2-[[2-(2,6- dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-isoindol-4-yl]oxy]acetamido)butyl]carbamoyl]-
2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl)but-2-en-l-yl]-2-(l-ethyl-
3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide as an off-white solid. LC-MS (ES, m/z): 532 (M/2+H+); ¾ NMR (400 MHz, DMSO -d6) S 12.82 (s, 2H), 11.13 (s, 1H), 8.44-8.42 (m, 1H), 8.03-7.94 (m, 4H), 7.83-7.81 (m, 1H), 7.79-7.65 (m, 2H), 7.49-7.34 (m, 5H), 6.55 (s, 2H), 5.94-5.90 (m, 2H), 5.11 (q, 1H), 4.83-4.78 (m, 6H), 4.54 (q, 4H), 3.27-3.19 (m,
4H), 2.89-2.85 (m, 1H), 2.56-2.50 (m, 2H), 2.12 (s, 6H), 2.11-2.04 (m, 1H), 1.60-1.50 (m, 4H), 1.26 (t, 6H).
Example 52: Synthesis of (E)-N-(2-aminoethyl)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-85
Figure imgf000326_0001
[00593] To a solution of (E)-tert-butyl 2-(l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole- 5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxamido)ethylcarbamate, 1-90, (50 mg, 0.060 mmol, 1 equiv) in DCM (5 mL) was added TFA (1 mL). The reaction mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD Column 30* 150mm, 5um; mobile phase, water (0.1% FA) and ACN (28% ACN up to 45% in 7min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 25.3 mg (58.56%) of (E)-N-(2-aminoethyl)-l-(4-(5- carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2- enyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 720 (M+H+); ¾-NMR (300 MHz, DMSO -d6): S 8.74 (s, 1H), 8.39 (s, 1H), 8.05-7.99 (m, 3H), 7.73-7.70 (m, 2H), 7.47-7.35 (m, 3H), 6.58 (s, 2H), 5.99-5.88 (m, 2H), 4.89-4.80 (m, 4H), 4.55 (q, 4H), 3.56-3.45 (m, 2H), 3.00-2.87 (m, 2H), 2.14 (s, 6H), 1.29 (t, 6H).
Example 53: Synthesis of (E)-N-(3-aminopropyl)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl- lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl- lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-84
Figure imgf000327_0001
[00594] To a solution of tert-butyl N-[3-([l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-5-yl]formamido)propyl]carbamate, 1-89, (50 mg, 0.060 mmol, 1 equiv) in DCM (5 mL) was added TFA (1 mL). The reaction mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD Column 30* 150mm, 5um; mobile phase, Water (0.1% FA) and ACN (10% ACN up to 35% in 7min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 17.9 mg (38.28%) of N-(3-aminopropyl)-l-[(2E)-4-[5- carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]- 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide as an off-white solid. LC-MS (ES, m/z): 734 (M+H+); 1HNMR (400 MHz, DMSO-</6) d 8.71 (s, 1H), 7.98-7.94 (m, 3H), 7.73-7.68 (m, 2H), 7.46-7.30 (m, 3H), 6.56 (s, 2H), 5.89-5.84 (m, 2H), 4.86-4.80 (m, 4H), 4.54 (q, 4H), 3.38-3.31 (m, 2H), 2.84-2.78 (m, 2H), 2.12 (s, 6H), 1.88-1.72 (m, 2H), 1.28 (t, 6H).
Example 54: Synthesis of (E)-N-(3-biphenyl-4-ylcarboxamidopropyl)-l-(4-(5-carbamoyl-2- (l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-2-(l- ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-83
Figure imgf000328_0001
Synthesis of 54.1
[00595] To a solution of biphenyl-4-carboxylic acid (500 mg, 2.904 mmol, 1.00 equiv) and tert- butyl 3-aminopropylcarbamate (505.3 mg, 3.485 mmol, 1.20 equiv) in DCM (10 mL) was added HATU (1.656 g, 4.356 mmol, 1.50 equiv) and DIEA (1.88 g, 14.519 mmol, 5.00 equiv). The resulting mixture was stirred at RT for 2 h. The mixture was diluted with water, extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SC>4 and concentrated under reduced pressure. The residue was applied on a silica gel column with PE/EtOAc (5/1). This resulted in 870 mg (84.63%) of tert-butyl 3-biphenyl-4-ylcarboxamidopropylcarbamate as an off- white solid. LC-MS (ES, m/z): 355 (M+H+).
Synthesis of 54.2
[00596] To a stirred solution of tert-butyl 3-biphenyl-4-ylcarboxamidopropylcarbamate, 54.1, (870 mg, 2.458 mmol, 1.00 equiv) and DCM (10 mL) was added TFA (2 mL). The reaction mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. This resulted in 700 mg crude of N-(3-aminopropyl)biphenyl-4-carboxamide as a brown solid. LC-MS-PH (ES, m/z): 255 (M+H+).
Synthesis of 1-83
[00597] To a solution of l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxylic acid, 43.1, (100 mg, 0.148 mmol, 1 equiv) and N-(3- aminopropyl)biphenyl-4-carboxamide (74.93 mg, 0.295 mmol, 2.00 equiv) in DMF (5 mL) was added HATU (84.16 mg, 0.221 mmol, 1.5 equiv) and DIEA (95.35 mg, 0.738 mmol, 5 equiv). The reaction mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselec CSH OBD Column 30* 150mm, 5um; mobile phase, Water (0.1% FA) and ACN (30% ACN up to 60% in 10 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 32.9 mg (24.9%) of (E)-N-(3-biphenyl-4-ylcarboxamidopropyl)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 914 (M+H+); 1HNMR (300 MHz, DMSO-t/6) ό 12.82 (br s, 2H), 8.59-8.49 (m, 2H), 8.02- 7.88 (m, 5H), 7.83-7.69 (m, 6H), 7.52-7.33 (m, 6H), 6.56 (s, 2H), 5.95-5.91 (m, 2H), 4.92-4.78 (m, 4H), 4.54 (q, 4H), 2.12 (s, 6H), 1.86-1.75 (m, 2H), 1.28 (t, 6H).
Example 55: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-N-(8-(2-((2-(2,6-dioxopiperidin-3- yl)-l,3-dioxoisoindolin-4-yl)oxy)acetamido)octyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-82
Figure imgf000330_0001
[00598] To a stirred solution of l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH- l,3-benzodiazole-5-carboxylic acid, 43.1, (70.00 mg, 0.103 mmol, 1.00 equiv) and N-(8- aminooctyl)-2-[[2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-isoindol-4- yl]oxy] acetamide (71.04 mg, 0.155 mmol, 1.50 equiv) inDMF (5.00 mL) were added DIEA (40.05 mg, 0.310 mmol, 3.00 equiv) and HATU (58.91 mg, 0.155 mmol, 1.50 equiv). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 OBD Column 19*250mm, lOum; mobile phase, water (0.1% FA) and ACN (30% ACN up to 50% in 7 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 55.2 mg (47.79%) of l-[(2E)-4-(5-[[8-(2-[[2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-isoindol-4-
329
5U B5TITUTE SH EET (RU LE 26) yl]oxy]acetamido)octyl]carbamoyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazol-l-yl)but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxamideas a white solid. LC-MS (ES, m/z): 1118.85 (M+H+) and 560 (M/2+H-); ¾ NMR (400 MHz, DMSO-<&) d 12.80 (br s, 2H), 11.12 (s, 1H), 8.41-8.49 (m, 1H), 7.97-7.94 (m, 4H), 7.82-7.80 (m, 1H), 7.78-7.70 (m, 2H), 7.67-7.33 (m, 5H), 6.55 (s, 2H), 5.94- 5.91 (m, 2H), 5.12 (q, 1H), 4.83-4.77 (m, 6H), 4.53 (q, 4H), 3.27-3.25 (m, 2H), 3.17-3.12 (m, 2H), 2.90-2.89 (m, 1H), 2.61-2.54 (m, 2H), 2.12 (s, 6H), 2.12-2.05 (m, 1H), 1.52-1.44 (m, 4H),1.29- 1.23 (m, 14H).
Example 56: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-N-(2-(2-((2-(2,6-dioxopiperidin-3- yl)-l,3-dioxoisoindolin-4-yl)oxy)acetamido)ethyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-81
Figure imgf000331_0001
Figure imgf000332_0001
[00599] To a solution of 2-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yloxy)acetic acid (66.4 mg, 0.2 mmol, 1 equiv) and (E)-N-(2-aminoethyl)-l-(4-(5-carbamoyl-2-(l -ethyl-3 - methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-2-(l -ethyl-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-85, (144 mg, 0.2 mmol, 1 equiv) in DMF (5 mL) was added HATU (114 mg, 0.3 mmol, 1.5 equiv) and DIEA (77.4 mg, 0.6 mmol, 3 equiv). The resulting mixture was stirred at RT for 2 h. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 OBD Column 19*250mm, lOum; mobile phase, Water (0.1% FA) and ACN (30% ACN up to 37% in 9 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 60.5 mg (29.28%) of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-N-(2-(2-(2-(2,6-dioxopiperidin-3-yl)-l,3- dioxoisoindolin-4-yloxy)acetamido)ethyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)- lH-benzo[d]imidazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 1034 (M+H+); ¾ NMR (300 MHz, DMSO-tfe) d 12.78 (s, 1H), 12.63 (br s, 2H), 11.13 (br s, 1H), 8.48 (s, 1H), 8.20 (s, 1H), 8.04-7.92 (m, 3H), 7.75-7.61 (m, 3H), 7.44-7.35 (m, 5H), 6.56 (s, 2H), 5.95-5.90 (m, 2H), 5.10 (q, 1H), 4.89-4.72 (m, 6H), 4.53 (q, 4H), 3.75-3.40 (m, 4H), 2.86-2.80 (m, 1H), 2.68-2.51 (m, 2H), 2.19-1.99 (m, 7H), 1.26 (t, 6H).
Example 57: Synthesis of (E)-N,N'-(but-2-ene-l,4-diylbis(lH-benzo[d]imidazole-l,2-diyl))bis(l- ethyl-3-methyl-lH-pyrazole- 5-carboxamide), 1-80
Figure imgf000333_0001
42.5 1-80
[00600] To a stirred solution of l-[(2E)-4-(2-amino-lH-l,3-benzodiazol-l-yl)but-2-en-l-yl]- lH-l,3-benzodiazol-2-amine (160 mg, 0.503 mmol, 1 equiv) and l-ethyl-3-methyl-lH-pyrazole- 5-carboxylic acid, 42.5, (232.4 mg, 1.509 mmol, 3 equiv) in NMP (8 ml) was added HATU (573.42 mg, 1.509 mmol, 3 equiv) and DIEA (649.50 mg, 5.025 mmol, 10 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 140oC. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, XBridge Shield RP18 OBD Column, 19*250mm, lOum; mobile phase, Water (0.1% FA) and ACN (30% ACN up to 65% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 41.3 mg of (E)-N,N'-(l,l'-(but-2-ene-l,4-diyl)bis(lH- benzo[d]imidazole-2,l-diyl))bis(l-ethyl-3-methyl-lH-pyrazole-5-carboxamide) as an off-white solid. LC-MS (ES, m/z): 591 (M+H+); 1H NMR (300 MHz, DMSO-d6) d 12.69 (s, 2H), 7.49 (d, 2H), 7.39 (d, 2H), 7.22-7.11 (m, 4H), 6.55 (s, 2H), 5.94-5.89 (m, 2H), 4.85-4.80 (m, 4H), 4.54 (q, 4H), 2.13 (s, 6H), 1.28 (t, 6H).
Example 58: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-N-(3-(2-((2-(2,6-dioxopiperidin-3- yl)-l,3-dioxoisoindolin-4-yl)oxy)acetamido)propyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-79
Figure imgf000334_0001
[00601] To a stirred solution of N-(3-aminopropyl)-l-[(2E)-4-[5-carbamoyl-2-(l -ethyl-3 - methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide hydrochloride, 1-84, (60.00 mg, 0.078 mmol, 1.00 equiv) and 2-[[2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-isoindol-4- yl]oxy]acetic acid (38.82 mg, 0.117 mmol, 1.50 equiv) in DMF (10 mL) were added HATU (44.43 mg, 0.117 mmol, 1.50 equiv) and DIEA (50.33 mg, 0.389 mmol, 5.00 equiv). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD Column 30* 150mm, 5um; mobile phase, water (0.1% FA) and ACN (25% ACN up to 42% in 7 min); Detector, UV 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 50.4 mg (61.74%) l-[(2E)-4-(5-[[3-(2-[[2- (2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-isoindol-4-
333
5U B5TITUTE SH EET (RU LE 26) yl]oxy]acetamido)propyl]carbamoyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazol-l-yl)but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 1048.3 (M+H+); 1H NMR (400 MHz, DMSO- ,) d 12.80 (br s, 2H), 11.15 (s, 1H), 8.45-8.40 (m, 1H), 8.07-8.05 (m, 1H), 7.98- 7.94 (m, 3H), 7.83-7.80 (m, 1H), 7.73-7.70 (m, 2H), 7.67-7.40 (m, 4H), 7.33 (s, 1H), 6.55 (s, 2H), 5.92-5.90 (m, 2H), 5.12 (q, 1H), 4.83-4.80 (m, 6H), 4.56-4.51 (m, 4H), 3.34-3.20 (m, 4H), 2.98- 2.90 (m, 1H), 2.61-2.50 (m, 2H), 2.13 (s, 6H), 2.13-2.05 (m, 1H), 1.73-1.70 (m, 2H), 1.28 (t, 6H).
Example 59: Synthesis of N,N'-(butane-l,4-diylbis(lH-benzo[d]imidazole-l,2-diyl))bis(l- ethyl-3-methyl-lH-pyrazole-5-carboxamide), 1-77
Figure imgf000335_0001
[00602] To a solution of l-ethyl-N-[l-[(2E)-4-[2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3-benzodiazol-2-yl]-3-methyl-lH-pyrazole-5- carboxamide, 1-80, (100 mg, 0.169 mmol, 1.00 equiv) in MeOH (20 ml) and NMP (5 ml) was added Pd/C (50 mg). To the above ¾ (g) was introduced in. The mixture was stirred 4 h at room temperature. The solid was fdtered. The filtrate was concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, XB ridge Shield RP 18 OBD Column, 19*250mm, lOum; mobile phase, Water (0.1% FA) and ACN (34% ACN up to 70% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 43 mg (41%) of l-ethyl-N-(l-[4-[2-(l- ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]butyl]-lH-l,3-benzodiazol-2- yl)-3-methyl-lH-pyrazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 593 (M+H+); 'H NMR (300 MHz, DMSO-tfc) d 12.68 (s, 2H), 7.54-7.46 (m, 4H), 7.23-7.17 (m, 4H), 6.59 (s, 2H), 4.56 (q, 4H), 4.33-4.21 (m, 4H), 2.10 (s, 6H), 1.96-1.80 (m, 4H), 1.29 (t, 6H).
Example 60: Synthesis of (E)-l,l'-(but-2-ene-l,4-diyl)bis(2-(l-ethyl-lH-imidazole-2- carboxamido)-7-methoxy-lH-benzo[d]imidazole-5-carboxamide), 1-76
Figure imgf000336_0001
Figure imgf000337_0001
[00603] Synthesis of 60.1. Into a 250-mL round-bottom flask was placed a solution of 4- chloro-3 -m ethoxy-5 -nitrobenzamide (8.5 g, 36.860 mmol, 1 equiv) in EtOH (50 mL). To the solution was added tert-butyl N-[(2E)-4-aminobut-2-en-l-yl]carbamate hydrochloride (9.85 g, 44.232 mmol, 1.2 equiv) and DIEA (14.29 g, 110.581 mmol, 3 equiv). The resulting solution was stirred overnight at 120 degrees C. The reaction mixture was cooled to room temperature and the solids were collected by filtration to give the product (8 g crude) as an orange solid. LCMS (ESI): M+H+ = 381.3.
Synthesis of 60.2. Into a 500 mL round-bottom flask were added tert-butyl N-[(2E)-4-[(4- carbamoyl-2-methoxy-6-nitrophenyl)amino]but-2-en-l-yl]carbamate, 60.1, (8 g, 21 mmol, 1 equiv) in MeOH (100 mL). To the above mixture was added HC1 (20 mL, 658.238 mmol, 86.34 equiv) at room temperature. The resulting mixture was stirred overnight at 40 degrees C. The resulting mixture was concentrated under reduced pressure. The crude product (3.6 g) was used in the next step directly without further purification. LCMS (ESI): M+H+ = 281.1.
[00604] Synthesis of 60.3. Into a 250-mL round-bottom flask, was placed a solution of 4- [[(2E)-4-aminobut-2-en-l-yl]amino]-3-methoxy-5-nitrobenzamide, 60.2, (3.6 g, 12.85 mmol, 1 equiv) and 4-chloro-3-methoxy-5 -nitrobenzamide (3.6 , 15.42 mmol, 1.2 equiv) in DMSO (50 mL). To the solution was added DIEA (8.29 g, 64.2 mmol, 5 equiv). The resulting solution was stirred overnight at 80degrees C. The mixture was cooled to room temperature and dropped into 500mL of water. The solids were collected by filtration to give the product (4 g crude) as a brown solid. LCMS (ESI): M+H+= 475.3.
[00605] Synthesis of 60.4. Into a 250-mL round-bottom flask, was placed a solution of 4- [[(2E)-4-[(4-carbamoyl-2-methoxy-6-nitrophenyl)amino]but-2-en-l-yl]amino]-3-methoxy-5- nitrobenzamide, 60.3, (4 g, 8.431 mmol, 1 equiv) in HC1 (50 mL). To the solution was added SnCh.2H20 (9.51 g, 42.156 mmol, 5 equiv) at 0 degrees C. The resulting solution was stirred for 2 hours at room temperature. The mixture was basified to pH 9 with NaiCC . The solids were separated by filtration and the water layers were collected. The product in water (500 mL) was used directly for the next step. LCMS (ESI): M+H+ = 415.3.
[00606] Synthesis of 60.5. Into a 25-mL round-bottom flask, was placed a solution of 3-amino-
4-[[(2E)-4-[(2-amino-4-carbamoyl-6-methoxyphenyl)amino]but-2-en-l-yl]amino]-5- methoxybenzamide, 60.4, (400 mg, 0.965 mmol, 1 equiv) in water (200 mL). To the solution was added carbononitridic bromide (1022.25 mg, 9.651 mmol, 10.00 equiv). The resulting solution was stirred overnight at 70degrees C. The resulting mixture was concentrated under reduced pressure. The mixture was purified by Flash to give the product (150 mg) as a yellow solid. LCMS (ESI): M+H+ = 465.3.
[00607] Synthesis of 1-76. To a solution of (E)-l,T-(but-2-ene-l,4-diyl)bis(2-amino-7- methoxy-lH-benzo[d]imidazole-5-carboxamide), 60.5, (80 mg, 0.172 mmol, 1 equiv) and 1-ethyl- lH-imidazole-2-carboxylic acid (72.24 mg, 0.516 mmol, 3 equiv) in NMP (5 mL) was added HATU (196.08 mg, 0.516 mmol, 3 equiv) and DIEA (133.56 mg, 1.033 mmol, 6 equiv). The final reaction mixture was irradiated with microwave radiation for 1 hour at 140°C. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH F-Phenyl OBD Column 19* 150mm, 5um; mobile phase, Water (0.1% FA) and ACN (10% ACN up to 25% in 8 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 9.9 mg (8.12%) of (E)-l, l'-(but-2-ene-l,4-diyl)bis(2-(l-ethyl-lH-imidazole-2- carboxamido)-7-methoxy-lH-benzo[d]imidazole-5-carboxamide) as a brown yellow solid. LCMS (ESI, m/z) 709 (M+H+); 'H-NMR (400 MHz, DMSO- L) d 13.17 (br s, 2H), 8.06 (s, 2H), 7.90 (s, 2H), 7.74-7.70 (m, 4H), 7.52-7.35 (m, 4H), 5.85-5.81 (m, 2H), 5.14-5.02 (m, 4H), 4.63 (q, 4H), 3.75 (s, 6H), 1.36 (t, 6H).
Example 61: Synthesis of l-ethyl-3-methyl-4-(5-(3-methyl-5-((l-propyl-lH- benzo[d]imidazol-2-yl)carbamoyl)-lH-pyrazol-l-yl)pentyl)-N-(l-propyl-lH- benzo[d]imidazol-2-yl)-lH-pyrazole-5-carboxamide, 1-74
Figure imgf000339_0001
Figure imgf000340_0001
Synthesis of 61.1
[00608] To a stirred solution of 3-methyl-lH-pyrazole-5-carboxylic acid (4 g, 31.746 mmol, 1 equiv) in dimethyl formamide (100 mL) was added potassium carbonate (8.76 g, 63.492 mmol, 2 equiv) and (bromomethyl)benzene
(6.477 g, 38.1 mmol, 1.2 equiv). The resulting solution was stirred overnight at room temperature. The mixture was diluted with water and extracted with dichloromethane. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography with PE/ethyl acetate (3/1) to give 4.7 g (68.54%) of benzyl 3-methyl-lH-pyrazole-5-carboxylate as a white solid. LCMS (ES, m/z): 217 (M+H+).
Synthesis of 61.2
[00609] To a solution of benzyl 3-methyl-lH-pyrazole-5-carboxylate, 61.1, (4.7 g, 21.76 mmol, 1 equiv), pent-4-yn-l-ol (2.19 g, 26.112 mmol, 1.2 equiv) and PPI13 (8.55 g, 32.64 mmol, 1.5 equiv) in tetrahydrofuran (100 mL) was added DIAD (6.593 g, 32.64 mmol, 1.5 equiv) at 0°C. The resulting solution was stirred overnight at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography with PE/ethyl acetate (5/1) to give 4.5 g (73.34%) of benzyl 3-methyl-l-(pent-4-ynyl)-lH-pyrazole-5- carboxylate as a white solid. LC-MS (ES, m/z): 283 (M+H+). Synthesis of 61.3
[00610] To a stirred mixture of l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (3.08 g, 20 mmol, 1 equiv) in di chi orom ethane (100 mL) was added dropwise oxalyl di chloride (3.5 mL, 40 mmol, 2 equiv) at 0°C. The resulting mixture was stirred for 2h at r.t. The mixture was concentrated under reduced pressure. To the crude product was added ethanol (30 mL). The resulting mixture was stirred for lh at r.t. The mixture was concentrated under reduced pressure. The residue was diluted with water and extracted with dichloromethane. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 2.2 g (60.44%) of ethyl l-ethyl-3-methyl-lH-pyrazole-5-carboxylate as a light yellow oil. LC-MS (ES, m/z): 183 (M+H+).
Synthesis of 61.4
[00611] To a stirred solution of ethyl l-ethyl-3-methyl-lH-pyrazole-5-carboxylate, 61.3, (2.2 g, 12.088 mmol, 1 equiv) in dimethyl formamide (40 mL) was added
NIS (3.26 g, 14.5 mmol, 1.2 equiv). The resulting solution was stirred for 72h at 90°C. The mixture was cooled to r.t, diluted with water and extracted with dichloromethane. The combined organic layers were washed with sat. sodium sulfite solution, brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography with PE/ethyl acetate (10/1) to give 2.5 g (68.54%) of ethyl l-ethyl-4- iodo-3-methyl-lH-pyrazole-5-carboxylate as a colorless oil. LCMS (ES, m/z): 309 (M+H+). Synthesis of 61.5
[00612] To a solution of ethyl 1 -ethyl -4-iodo-3-methyl-lH-pyrazole-5-carboxylate, 61.4, (2.5 g, 8.116 mmol, 1 equiv) and benzyl 3-methyl-l-(pent-4-ynyl)-lH-pyrazole-5-carboxylate (2.289 g, 8.116 mmol, 1.0 equiv) in dimethyl formamide (20 mL) was added copper(I) iodide (310 mg, 1.623 mmol, 0.2 equiv) and Pd(PPli3)Cl2 (570 mg, 0.811 mmol, 0.1 equiv). The resulting solution was stirred overnight at 100°C. The mixture was cooled to r.t and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography with PE/ethyl acetate (3/1) to give 1.25 g (33.34%) of ethyl 4-(5-(5-(benzyloxycarbonyl)-3-methyl-lH-pyrazol- l-yl)pent-l-ynyl)-l-ethyl-3-methyl-lH-pyrazole-5-carboxylate as a colorless oil. LC-MS (ES, m/z): 463 (M+H+).
Synthesis of 61.6 [00613] To a stirred solution of ethyl 4-(5-(5-(benzyloxycarbonyl)-3-methyl-lH-pyrazol-l- yl)pent-l-ynyl)-l-ethyl-3-methyl-lH-pyrazole-5-carboxylate, 61.5, (1 g, 2.165 mmol, 1 equiv) in ethanol (20 mL) was added palladium on carbon (200 mg). To the above hydrogen (g) was introduced in. The resulting mixture was stirred at r.t for 2h. The solid was filtered out. The filtrate was concentrated under vacuum. This resulted in 0.6 g (73.71%) of l-(5-(5-(ethoxycarbonyl)-l- ethyl-3-methyl-lH-pyrazol-4-yl)pentyl)-3-methyl-lH-pyrazole-5-carboxylic acid as a white solid. LC-MS (ES, m å): 377 (M+H+).
Synthesis of 61.7
[00614] To a stirred mixture of l-(5-(5-(ethoxycarbonyl)-l-ethyl-3-methyl-lH-pyrazol-4- yl)pentyl)-3-methyl-lH-pyrazole-5-carboxylic acid, 61.6, (600 mg, 1.6 mmol, 1 equiv) in methanol (20 mL) and water (4 mL) was added lithium hydroxide (115.2 mg, 4.8 mmol, 3 equiv). The resulting mixture was stirred at r.t for 2 hours. The mixture was concentrated under reduced pressure. The residue was diluted with water, then adjusted to pH 3 with 1 N HC1. The solid was collected by filtration. This resulted in 400 mg (71.84%) of 4-(5-(5-carboxy-3-methyl-lH-pyrazol- l-yl)pentyl)-l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid as a white solid. LCMS: (ES, m/z): 349 (M+H+).
Synthesis of 61.8
[00615] To a solution of l-fluoro-2-nitrobenzene (2.82 g, 20 mmol, 1.0 equiv) in dimethyl formamide (50 mL) was added potassium carbonate (5.52 g, 40 mmol, 2 equiv) and propan-1- amine (5.9 g, 100 mmol, 5 equiv). The resulting mixture was stirred at 50°C for 4 h. The mixture was cooled to r.t, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with PE/ethyl acetate (3/1) to give 2.5 g (69.44%) of 2-nitro-N-propylbenzenamine as a yellow solid. LC-MS (ES, m/z): 181 (M+H+); Synthesis of 61.9
[00616] To a solution of 2-nitro-N-propylbenzenamine, 61.8, (2 g, 11.098 mmol, 1 equiv) in methanol (30 mL) was added palladium on carbon (400 mg). To the above hydrogen (g) was introduced in. The mixture was stirred for 2h at room temperature. The solid was filtered. The filtrate was concentrated under reduced pressure. This resulted in 2 g (crude) of/V^-propylbenzene- 1, 2-diamine as an off-white solid. LC-MS (ES, m/z): 151 (M+H+); Synthesis of 61.10
[00617] To a solution of /V;-propylbenzene- 1,2-diamine, 61.9, (2 g crude, 11.098 mmol, 1 equiv) in methanol (20 mL) was added cyanogen bromide (1.748 g, 16.647 mmol, 1.5 equiv). The resulting solution was stirred for 3h at room temperature. The mixture was concentrated. This resulted in 2.1 g (crude) of l-propyl-lH-benzo[d]imidazol-2-amine as a brown solid. LC-MS (ES, m/z): 176 (M+H+);
Synthesis of 1-74
[00618] To a solution of 4-(5-(5-carboxy-3-methyl-lH-pyrazol-l-yl)pentyl)-l-ethyl-3-methyl- lH-pyrazole-5-carboxylic acid (100 mg, 0.287 mmol, 1.00 equiv) in l-methylpyrrolidin-2-one (5 mL) was added l-propyl-lH-benzo[d]imidazol-2-amine, 61.10, (100.45 mg, 0.574 mmol, 2.00 equiv), HATU (239.7 mg, 0.631 mmol, 2.2 equiv) and DIEA (185.1 mg, 1.435 mmol, 5 equiv). The final reaction mixture was irradiated with microwave radiation at 140°C for lh. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH Prep C18 OBD Column 19*250mm, 5um; mobile phase, Water (0.1% FA) and ACN (90% ACN up to 95% in 7 min); Detector, UV 254/210 nm. This resulted in 43.8 mg (23%) of 1 -ethyl-3 -methyl-4- (5-(3-methyl-5-(l-propyl-lH-benzo[d]imidazol-2-ylcarbamoyl)-lH-pyrazol-l-yl)pentyl)-N-(l- propyl-lH-benzo[d]imidazol-2-yl)-lH-pyrazole-5-carboxamide as an off-white solid. LC-MS (ES, m/z): 663 (M+H+); Ti NMR ^OO MHz, DMSO-nfe) d 12.71 (s, 1H), 12.63 (s, 1H), 7.59-7.42 (m, 4H), 7.29-7.18 (m, 4H), 6.60 (s, 1H), 4.61-4.49 (m, 4H), 4.10 (q, 4H), 2.74 (t, 2H), 2.14 (s, 3H), 2.06 (s, 3H), 1.84-1.63 (m, 6H), 1.60-1.42 (m, 2H), 1.38-1.22 (m, 5H), 0.86 (t, 6H).
Example 62: Synthesis of l-ethyl-3-methyl-N-(l-methyl-lH-benzo[d]imidazol-2-yl)-4-(5-(3- methyl-5-((l-methyl-lH-benzo[d]imidazol-2-yl)carbamoyl)-lH-pyrazol-l-yl)pentyl)-lH- pyrazole-5-carboxamide, 1-73
Figure imgf000344_0001
[00619] To a stirred solution of 4-[5-(5-carboxy-3-methyl-lH-pyrazol-l-yl)pentyl]-l-ethyl-3- methyl-lH-pyrazole-5-carboxylic acid, 61.7, (100 mg, 0.287 mmol, 1 equiv) in 1- methylpyrrolidin-2-one (5 mL) was added l-methyl-lH-l,3-benzodiazol-2-amine (84.49 mg, 0.574 mmol, 2 equiv), DIEA (185.48 mg, 1.435 mmol, 5 equiv) and HATU (240.10 mg, 0.631 mmol, 2.2 equiv). The resulting mixture was irradiated with microwave radiation at 140°C for lh. The mixture was concentrated under reduced pressure. The crude product was purified by Prep- HPLC with the following conditions: Column, XBridge Prep C18 OBD Column, 19* 150mm, 5um; mobile phase, Water (10% NH4HCO3+0.1%NH3.H20) and ACN (47% ACN up to 77% in 7 min); Detector, UV 254/220 nm. This resulted in 54.1 mg (31.07%) of l-ethyl-3-methyl-N-(l- methyl-lH-l,3-benzodiazol-2-yl)-4-(5-[3-methyl-5-[(l-methyl-lH-l,3-benzodiazol-2- yl)carbamoyl]-lH-pyrazol-l-yl]pentyl)-lH-pyrazole-5-carboxamide as an off-white solid. LC- MS (ES, m/z): 607 (M+H+); ¾ NMR (300 MHz, DMSO-i&) ό 12.66 (s, 1H), 12.63 (s, 1H), 7.54- 7.39 (m, 4H), 7.30-7.19 (m, 4H), 6.67 (s, 1H), 4.64-4.53 (m, 4H), 3.65 (s, 3H), 3.61 (s, 3H), 2.79 (t, 2H), 2.17 (s, 3H), 2.11 (s, 3H), 1.84-1.79 (m, 2H), 1.61-1.51 (m, 2H), 1.36-1.32 (m, 5H).
Example 63: Synthesis of (E)-l-ethyl-N-(l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-(morpholinomethyl)-lH- benzo[d]imidazol-2-yl)-3-methyl-lH-pyrazole-5-carboxamide, 1-72
Figure imgf000345_0001
Synthesis of 63.1
[00620] To a solution of 2-fluoro-l -methyl-3 -nitrobenzene (15.5 g, 100 mmol, 1 equiv) in CCU (200 ml) was added AIBN (1.64 g, 10 mmol, 0.1 equiv) and NBS (21.36 g, 120 mmol, 1.2 equiv). The resulting mixture was stirred at 85°C overnight. The mixture was cooled to rt and concentrated. The residue was applied on a silica gel column with PE/EtOAc (5/1). This resulted in 20 g crude of l-(bromomethyl)-2-fluoro-3 -nitrobenzene as a white solid. LC-MS (ES, m/z): 234/236 (M+TE). Synthesis of 63.2
[00621] To a solution of l-(bromomethyl)-2-fluoro-3-nitrobenzene, 63.1, (20 g crude, 100 mmol, 1 equiv) in DCM (200 ml) was added DIEA (38.7 g, 300 mmol, 3 equiv) and morpholine (43.5 g, 50 mmol, 0.5 equiv). The resulting mixture was stirred at RT overnight. The mixture was diluted with water and extracted with DCM. The combined organic layers were washed with brine, dried over NazSCE and concentrated under reduced pressure. The residue was applied on a silica gel column with PE/EtOAc (1/1) to give 9 g (37.5%) of 4-(2-fluoro-3-nitrobenzyl)morpholine as a yellow solid. LC-MS (ES, m/z): 241 (M+EC).
Synthesis of 63.3
[00622] To a stirred mixture of l-fluoro-2-nitrobenzene (2.82 g, 20 mmol, 1 equiv) and (E)- tert-butyl 4-aminobut-2-enylcarbamate hydrochloride (5.328 g, 24 mmol, 1.2 equiv) in DMF (50 ml) was added K2CO3 (5.52 g, 40 mmol, 2 equiv). The resulting mixture was stirred for 3 h at 8O0C. The mixture was cooed to room temperature and diluted with water. The precipitated solids were collected by filtration. This resulted in 4 g (65.15 %) of (E)-tert-butyl 4-(2- nitrophenylamino)but-2-enylcarbamate as a red solid. LC-MS (ES, m/z): 330 (M+Na+);
Synthesis of 63.4
To a stirred solution of (E)-tert-butyl 4-(2-nitrophenylamino)but-2-enylcarbamate, 63.3, (1.5 g, 4.886 mmol, 1 equiv) in DCM (20 mL) was added TFA (5 mL). The resulting mixture was stirred for 3 h at room temperature. The resulting mixture was concentrated under reduced pressure. This resulted in 1.2 g crude of (E)-A;-(2-nitrophenyl)but-2-ene- 1,4-diamine as a brown oil. LC-MS (ES, m/z): 208 (M+H ).
Synthesis of 63.5
[00623] To a stirred solution of 4-(2-fluoro-3-nitrobenzyl)morpholine, 63.2, (1.17 g, 4.886 mmol, 1 equiv) and (E)-/V;-(2-nitrophenyl)but-2-ene- 1,4-diamine, 63.6, (1.2 g crude, 4.886 mmol, 1 equiv) in DMF (30 mL) was added K2CO3 (2.02 g, 14.658 mmol, 3 equiv). The resulting mixture was stirred overnight at 70°C under nitrogen atmosphere. The mixture was cooled to room temperature and diluted with water. The precipitated solids were collected by filtration. This resulted in 0.95 g (45.54%) of (E)-Af/-(2-(morphol i nomethyl )-6-nitrophenyl)-N4-(2- nitrophenyl)but-2-ene- 1,4-diamine as a yellow solid. LC-MS (ES, m/z): 428 (M+FE).
Synthesis of 63.6 [00624] To a stirred solution of (E)-A7-(2-(morphol i nomethyl )-6-nitrophenyl)-N4-(2- nitrophenyl)but-2-ene- 1,4-diamine, 63.6, (0.95 g, 2.225 mmol, 1 equiv) in DMF (20 mL) was added SnCh 2H2O (5.03 g, 22.25 mmol, 10 equiv). The resulting mixture was stirred overnight at 35°C. The mixture was cooled to RT and diluted with water. The pH value of the solution was adjusted to 9 with sat.NaHCCb solution and extracted with EtOAc. The combined organic layers were washed with brine, dried over NaiSCE and concentrated under reduced pressure. This resulted in 500 mg (61%) of (E)-Af/-(4-(2-aminophenylainino)but-2-enyl)-6-
(morpholinomethyl)benzene-l, 2-diamine as a yellow solid. LC-MS (ES, m/z): 368 (M+H+); Synthesis of 63.7
[00625] To a stirred solution of (E)-A -(4-(2-aminophenylamino)but-2-enyl)-6-
(morpholinomethyl)benzene-l, 2-diamine (500 mg, 1.362 mmol, 1 equiv) in MeOH (20 mL) was added BrCN (288.75 mg, 2.724 mmol, 2 equiv). The resulting mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, Cl 8; mobile phase, Water (10 MMOL/L NH4HCO3) and ACN (28% ACN up to 40% in 10 min); Detector, UV 254/220 nm. This resulted in 200 mg (35.21%) of (E)-l-(4-(2-amino-lH-benzo[d]imidazol-l-yl)but-2-enyl)-7- (morpholinom ethyl)- lH-benzo[d]imidazol -2-amine as a yellow solid. LC-MS (ES, m/z): 418 (M+H+);
Synthesis of 1-72
[00626] To a stirred mixture of (E)-l-(4-(2-amino-lH-benzo[d]imidazol-l-yl)but-2-enyl)-7- (morpholinom ethyl)- lH-benzo[d]imidazol -2-amine, 63.7, (120 mg, 0.287 mmol, 1 equiv) and 1- ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (88.4 mg, 0.547 mmol, 2.2 equiv) in NMP (6 mL) was added HATU (272.65 mg, 0.718 mmol, 2.5 equiv) and DIEA (185.11 mg, 1.435 mmol, 5.0 equiv). The final reaction mixture was irradiated with microwave radiation for 1 h at 140°C. The mixture was cooled to RT and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD column, 30* 150mm, 5um; mobile phase, water (0.1% FA) and ACN (20% ACN up to 50% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 25.7 mg (13%) of (E)-l-ethyl-N-(l-(4-(2-(l-ethyl-3-methyl- lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-7-(morpholinomethyl)-lH-
346
5UB5TITUTE SHEET (RULE 26) benzo[d]imidazol-2-yl)-3-methyl-lH-pyrazole-5-carboxamide as an off-white solid. LC-MS (ES, m/z): 690 (M+H+); ¾ MR (400 MHz, DMSO-flfe) d 12.71 (br s, 2H), 7.50 (dd, 2H), 7.40 (d, 1H), 7.25-7.11 (m, 3H), 6.99 (d, 1H), 6.54 (s, 1H), 6.53 (s, 1H), 6.05-5.95 (m, 1H), 5.42-5.33 (m, 1H), 5.19 (d, 2H), 4.78 (d, 2H), 4.56-4.51 (m, 4H), 3.54 (s, 6H), 2.22-2.11 (m, 10H), 1.27 (t, 6H).
Example 64: Synthesis of tert-butyl (E)-(3-(l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-N-methyl-lH-benzo[d]imidazole-5- carboxamido)propyl)carbamate, 1-68
[00627] To a stirred solution of l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH- l,3-benzodiazole-5-carboxylic acid, 43.1, (300 mg, 0.443 mmol, 1 equiv) and HATU (252.47 mg, 0.664 mmol, 1.5 equiv) in DMF (10 mL) were added DIEA (171.63 mg, 1.328 mmol, 3.0 equiv) and tert-butyl N-[3-(methylamino)propyl]carbamate (250.02 mg, 1.328 mmol, 3.0 equiv). The resulting mixture was stirred for 2 h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD Column 30* 150mm, 5um; mobile phase, water (0.1% FA) and ACN (21% ACN up to 55% in 8 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 20.7 mg (5.51%) of tert-butyl N-[3-(l-[l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazol-5-yl]-N-methylformamido)propyl]carbamateas a white solid. LC-MS (ES, m/z)·. 848 (M+H+) and 870 (M+Na+); ¾ NMR (400 MHz, DMSO -d6) d 12.81 (br s, 1H), 12.76 (br s, 1H), 7.97-7.94 (m, 2H), 7.70 (d, 1H), 7.49-7.32 (m, 4H), 7.16 (d, 1H), 6.90-6.70 (m, 1H), 6.58-6.56 (m, 2H), 5.93-5.91 (m, 2H), 4.85-4.84 (m, 4H), 4.54 (q, 4H), 3.20-3.19 (m, 2H), 2.97-2.67 (m, 5H), 2.13 (s, 6H), 1.72-1.70 (m, 2H), 1.47-1.18 (m, 14H).
Example 65: Synthesis of 8-ethyl-10,18-dimethyl-8,ll,12,13,14,15,28,29,30,31- decahydrobenzo[4,5]imidazo[l,2-a]benzo[4,5]imidazo[2,l-p]dipyrazolo[5,l-e:4',3'- l][l,3,6,15,17]pentaazacyclohenicosine-7,20(6H,21H)-dione, 1-117
Figure imgf000349_0001
[00628] To a solution of (31E)-5-ethyl-7,16-dimethyl-2,5,6, 14,15,20,22,29,34,41- decaazaheptacyclo [32.7.0.0A[4,8].0A[14,18].0A[21,29].0A[23,28].0A[35,40]]hentetraconta- 1(41), 4(8), 6, 15, 17, 21, 23(28), 24, 26, 31,35,37,39-tridecaene-3, 19-dione, 1-116, (0.116 g, 0.184 mmol, 1 equiv) in methanol (20 mL) was added palladium carbon (10%, 50 mg). To the above hydrogen (g) was introduced in. The resulting mixture was stirred at RT for 2 h. The reaction mixture was filtered. The filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Prep C18 OBD Column 19* 150 mm, 5um; mobile phase, Water (10% NH4HCO3 + 0.1% NH3 H2O) and ACN (30% ACN up to 80% in 8 min); Detector, UV 254/220 nm. This resulted in 35.1 mg (30.16%) of 5-ethyl- 7, 16-dimethyl-2,5,6,14, 15,20,22,29,34,41- decaazaheptacyclo[32.7.0.0A[4,8].0A[14,18].0A[21,29].0A[23,28].0A[35,40]]hentetraconta- 1(41), 4(8), 6, 15, 17,21 ,23 (28),24,26,35,37,39-dodecaene-3 , 19-dione as an off-white solid. LC- MS(ES, m/z): 633 (M+H+); ¾ NMR: (300 MHz, DMSO -d6) d 12.77 (br s, 1H), 12.74 (br s, 1H), 7.68-7.52 (m, 4H), 7.36-7.22 (m, 4H), 6.56 (s, 1H), 4.76 (q, 2H), 4.46 (t, 2H), 4.41-4.14 (m, 4H), 2.82 (t, 2H), 2.16 (s, 3H), 2.09 (s, 3H), 2.01-1.76 (m, 6H), 1.59-1.29 (m, 7H).
Example 66: Synthesis of (E)-8-ethyl-10,18-dimethyl-7,20-dioxo-
6,7,8,ll,12,13,14,15,20,21,28,31-dodecahydrobenzo[4,5]imidazo[l,2- a]benzo[4,5]imidazo[2,l-p]dipyrazolo[5,l-e:4',3'-l][l,3,6,15,17]pentaazacyclohenicosine- 3,24-dicarboxylic acid (1-119)
Figure imgf000350_0001
Synthesis of 66.1 [00629] To a stirred solution of methyl 4-fluoro-3-nitrobenzoate (2 g, 10.0 mmol, 1 equiv) in DMF (20 mL) was added K2CO3 (4.14 g, 30.0 mmol, 3 equiv) and (E)-tert-butyl 4-aminobut-2- enylcarbamate hydrochloride (2.45 g, 11.0 mmol, 1.1 equiv). The resulting solution was stirred overnight at 70 °C. The reaction was cooled to RT and diluted with water. The precipitated solids were collected by filtration. This resulted in 3.5 g (81.9%) of (E)- methyl 4-(4-(tert-butoxycarbonylamino)but-2-enylamino)-3-nitrobenzoate as a yellow solid. LC- MS (ES, m/z): 310 (M-56)+.
Synthesis of 66.2
[00630] To a stirred solution of (E)-m ethyl 4-(4-(tert-butoxycarbonylamino)but-2-enylamino)- 3-nitrobenzoate 66.1 (3.0 g, 8.21 mmol 1 equiv) in DCM (30 mL) was added TFA (6 mL). The resulting solution was stirred for 4 hours at room temperature. The mixture was concentrated. This resulted in 2.5 g (crude) of (E)-methyl 4-(4-aminobut-2-enylamino)-3-nitrobenzoate as a brown oil. LC-MS (ES, m/z) 266 (M+H+).
Synthesis of 66.3
[00631] To a stirred solution of (E)-methyl 4-(4-aminobut-2-enylamino)-3-nitrobenzoate, 66.2, (2.5 g crude, 8.21 mmol, 1 equiv) in DMF (40 mL) was added K2CO3 (3.4 g, 24.63 mmol, 3 equiv) and methyl 4-fluoro-3-nitrobenzoate (1.8 g, 9.03 mmol, 1.1 equiv). The resulting solution was stirred overnight at 70°C. The reaction was cooled to RT and diluted with water. The precipitated solids were collected by filtration. This resulted in 3.5 g (83.4%) of (E)- dimethyl 4,4'-(but-2-ene-l,4-diylbis(azanediyl))bis(3-nitrobenzoate) as a yellow solid. LC-MS (ES, m/z): 445 (M+H+).
Synthesis of 66.4
[00632] To a solution of (E)-dimethyl 4,4'-(but-2-ene-l,4-diylbis(azanediyl))bis(3- nitrobenzoate), 66.3, (2 g, 4.500 mmol, 1.00 equiv) in AcOH (40 mL) was added Zn (2.88 g, 45 mmol, 10.00 equiv). The resulting solution was stirred overnight at RT. The solids were filtered out. The resulting mixture was concentrated under reduced pressure. This resulted in 2 g crude of (E)-dimethyl 4,4'-(but-2-ene-l,4-diylbis(azanediyl))bis(3-aminobenzoate) as a brown yellow solid. LC-MS (ES, m/z): 769 (2M+H+). Synthesis of 66.5
[00633] To a stirred solution of (E)-dimethyl 4,4'-(but-2-ene-l,4-diylbis(azanediyl))bis(3- aminobenzoate), 66.4, (2 g crude, 4.5 mmol, 1 equiv) in MeOH (30 mL) was added BrCN (2.385 g, 22.5 mmol, 5 equiv). The resulting solution was stirred overnight at room temperature. The mixture was concentrated under reduced pressure. This resulted in 2.1 g crude of (E)-dimethyl l, T-(but-2-ene-l,4-diyl)bis(2-amino-lH-benzo[d]imidazole-5-carboxylate) as an off-white solid. LC-MS (ES, m/z) 452 (M+H20+).
Synthesis of 66.6
[00634] To a stirred solution of (E)-dimethyl l,l'-(but-2-ene-l,4-diyl)bis(2-amino-lH- benzo[d]imidazole-5-carboxylate), 66.5, (500 mg, 1.15 mmol, 1 equiv) and 4-[5-(5-carboxy-3- methyl-lH-pyrazol-l-yl)pentyl]-l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid. 61.7, (315 mg, 1.27 mmol, 1.1 equiv) in NMP (6 mL) was added DIEA (754 mg, 5.8 mmol, 5 equiv) and HATU (961.4 mg, 2.53 mmol, 2.2 equiv). The final reaction mixture was irradiated with microwave radiation for 1 hour at 140 °C. The mixture was cooled to RT and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (38% ACN up to 48% in 10 min); Detector, UV 254/220 nm. This resulted in 300 mg (35%) of 25,38-dimethyl - (3 lE)-5-ethyl-7, 16-dimethyl-3,19-dioxo-
2,5,6, 14,15,20,22,29,34,41,decaazaheptacyclo[32.7.0.0A[4,8].0A[14,18].0A[21,29].0A[23,28].0A[3 5,40]]hentetraconta-l(41),4(8),6,15,17,21,23(28),24,26,31,35(40),36,38-tridecaene-25,38- dicarboxylate as a light yellow solid. LC-MS (ES, m/z): 747 (M+H+).
Synthesis of 1-119
[00635] To a stirred solution of 25,38-dimethyl (31E)-5-ethyl-7,16-dimethyl-3, 19-dioxo- 2,5,6, 14,15,20,22,29,34,41- decaazaheptacyclo[32.7.0.0A[4,8].0A[14,18].0A[21,29].0A[23,28].0A[35,40]]hentetraconta. l(41),4 (8),6, 15,17,21,23(28),24,26,31,35(40),36,38-tridecaene-25,38-dicarboxylate, 66.6, (100 mg, 0.13 mmol, 1 equiv) in MeOH (5 mL), THF (5 mL) and ¾0 (2 mL) was added Li OH (31.2 mg, 1.3 mmol, 10 equiv). The resulting solution was stirred for 4 h at 60°C. The resulting mixture was cooled to RT and concentrated under reduced pressure. The pH value of the solution was adjusted to 3 with HC1 (1 mol/L). The solids were collected by filtration. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD Column, 30*150 mm, 5um; mobile phase, Water (0.1% FA) and ACN (30% ACN up to 63% in 8 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 17.9 mg of (31E)-5-ethyl-7, 16-dimethyl-3, 19-dioxo- 2,5,6, 14,15,20,22,29,34,41decaazaheptacyclo[32.7.0.0A[4,8].0A[14, 18].0A[21,29].0A[23,28].0A[3 5,40]]hentetraconta-l(41),4(8),6,15,17,21,23(28),24,26,31,35(40),36,38-tridecaene-25,38- dicarboxylic acid as a white solid. LC-MS (ES, m/z): 719 (M+H+); 'H NMR (400 MHz, DMSO- <k) d 12.93-12.81 (m, 4H), 8.05 (s, 2H), 7.82 (dd, 2H), 7.49 (d, 1H), 7.43 (d, 1H), 6.54 (s, 1H), 6.00-5.90 (m, 1H), 5.80-5.65 (m, 1H), 4.95-4.85 (m, 4H), 4.71 (t, 2H), 4.45 (q, 2H), 2.75 (t, 2H), 2.16 (s, 3H), 2.09 (s, 3H), 1.78-1.65 (m, 2H), 1.49-1.40 (m, 2H), 1.37-1.23 (m, 5H).
Example 67: Synthesis of (E)-N,N'-(but-2-ene-l,4-diylbis(5-cyano-lH-benzo[d]imidazole- l,2-diyl))bis(l-ethyl-3-methyl-lH-pyrazole-5-carboxamide), 1-67
Figure imgf000353_0001
Figure imgf000354_0001
Synthesis of 67.1
[00636] To a stirred solution of 4-chloro-3-nitrobenzamide (10 g, 50 mmol, 1 equiv) in dimethyl sulfoxide (200 mL) was added (E)-tert-butyl 4-aminobut-2-enylcarbamate hydrochloride (13.32 g, 60 mmol, 1.2 equiv) and potassium carbonate (20.7 g, 150 mmol, 3 equiv) at r.t. The resulting solution was stirred overnight at 70oC under nitrogen. The mixture was cooled to r.t and diluted with water. The precipitated solids were collected by filtration. This resulted in 7 g (40%) of (E)-tert-butyl 4-(4-carbamoyl-2-nitrophenylamino)but-2-enylcarbamate as a yellow solid. LC- MS (ES, m/z): 295 (M-56+H+).
Synthesis of 67.2
[00637] To a stirred solution of (E)-tert-butyl 4-(4-carbamoyl-2-nitrophenylamino)but-2- enylcarbamate, 67.1, (3.5 g, 10 mmol, 1 equiv) in dichloromethane (40 mL) was added 2,2,2- trifluoroacetic acid (5 mL). The resulting solution was stirred overnight at room temperature. The mixture was concentrated under reduced pressure to give 2.8 g (crude) of (E)-4-(4-aminobut-2- enylamino)-3-nitrobenzamide as a brown oil. LC-MS (ES, m/z): 251 (M+H+).
Synthesis of 67.3
[00638] To a stirred solution of (E)-4-(4-aminobut-2-enylamino)-3-nitrobenzamide, 67.2, (2.8 g crude, 10 mmol, 1 equiv) and 4-chloro-3-nitrobenzamide (2.4 g, 12 mmol, 1.2 equiv) in dimethyl sulfoxide (40 mL) was added potassium carbonate (4.14 g, 30 mmol, 3 equiv). The resulting solution was stirred overnight at 70°C under nitrogen. The mixture was cooled to r.t and diluted with water. The precipitated solids were collected by fdtrati on. This resulted in 3.2 g (77.3%) of (E)-4,4'-(but-2-ene-l,4-diylbis(azanediyl))bis(3-nitrobenzamide) as a yellow solid. LC-MS (ES, m/z): 415 (M+H+).
Synthesis of 67.4
[00639] To a solution of (E)-4,4'-(but-2-ene-l,4-diylbis(azanediyl))bis(3-nitrobenzamide), 67.3, (2 g, 4.83 mmol, 1 equiv) in dimethyl formamide (20 mL) was added tin( II )chloridedihydrate (10.9 g, 48.3 mmol, 10 equiv). The resulting mixture was stirred overnight at r.t. The mixture was cooled to room temperature and diluted with water. The pH value of the solution was adjusted to 9 with sat. sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. This resulted in 0.8 g (63.5%) of (E)-4,4'-(but-2-ene-l,4- diylbis(azanediyl))bis(3-aminobenzamide) as a yellow solid. LC-MS (ES, m/z): 355 (M+H+). Synthesis of 67.5
[00640] To a stirred solution of (E)-4,4'-(but-2-ene-l,4-diylbis(azanediyl))bis(3- aminobenzamide) (0.8 g, 2.26mmol, 1 equiv) in in methanol (20 mL) was added cyanogen bromide (711.9 mg, 6.78 mmol, 3 equiv). The resulting solution was stirred overnight at room temperature. The resulting mixture was concentrated under vacuum. This resulted in 1.5 g (crude) of (E)-1,T- (but-2-ene-l,4-diyl)bis(2-amino-lH-benzo[d]imidazole-5-carboxamide) as a brown solid. LC-MS (ES, m/z): 405 (M+H+).
Synthesis of 67.6
[00641] To a solution of (E)-l, T-(but-2-ene-l,4-diyl)bis(2-amino-lH-benzo[d]imidazole-5- carboxamide), 67.5, (40.4 mg, 0.1 mmol, 1 equiv) and l-ethyl-3-methyl-lH-pyrazole-5-
354
5UB5TITUTE SHEET (RULE 26) carboxylic acid (33.88 mg, 0.22 mmol, 2.2 equiv) in NMP (5 mL) was added HATU (95 mg, 0.25 mmol, 2.5 equiv) and DIEA (64.5 mg, 0.5 mmol, 5 equiv). The final reaction mixture was irradiated with microwave radiation for 1 h at 140oC. The mixture was cooled to RT and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD column, 30* 150mm, 5um; mobile phase, water (0.1% FA) and ACN (20% ACN up to 50% in 8 min); UV detection at 254/220 nm. The product- containing fractions were collected and evaporated partially and lyophilized overnight to afford 5 mg (7.39%) of (E)-l,l'-(but-2-ene-l,4-diyl)bis(2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxamide) as an off-white solid. LC-MS (ES, m/z): 677 (M+H+); 1H NMR (400 MHz, DMSO-d6) d 12.78 (br s, 2H), 8.03-7.92 (m, 4H), 7.72 (dd, 2H), 7.44 (d, 2H), 7.33 (s, 2H), 6.55 (s, 2H), 5.95-5.92 (m, 2H), 4.86-4.81 (m, 4H), 4.53 (q, 4H), 2.16 (s, 6H), 1.28 (t, 6H).
Synthesis of 1-67
[00642] To a stirred solution of (E)-l,T-(but-2-ene-l,4-diyl)bis(2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide), 67.6, (100 mg, 0.148 mmol, lequiv) and Et3N (149.48 mg, 1.48 mmol, 10 equiv) in DMF (5 mL) and THF (5 mL) was added dropwise TFAA (155.4 mg, 0.74 mmol, 5 equiv) at 0°C. The resulting solution was stirred overnight at room temperature. The mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Shield RP18 OBD column, 19*250mm, lOum; mobile phase, water (0.01% NH4HCO3) and ACN (53% ACN up to 58% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 19.7 mg of (E)-N,N'-( \ , 1 '-(but-2-ene-l ,4- diyl)bis(5-cyano-lH-benzo[d]imidazole-2,l-diyl))bis(l-ethyl-3-methyl-lH-pyrazole-5- carboxamide) as off-white solid. LC-MS (ES, m/z): 641 (M+H+); 'H NMR: ' H NMR (400 MHz, DMSO -d6) d 12.90 (s, 2H), 7.77 (s, 2H), 7.55 (d, 4H), 6.55 (s, 2H), 5.95-5.91 (m, 2H), 4.92-4.81 (m, 4H), 4.54 (q, 4H), 2.15 (s, 6H), 1.30 (t, 6H).
Example 68: Synthesis of (E)-N-(3-aminopropyl)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl- lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl- lH-pyrazole-5-carboxamido)-N-methyl-lH-benzo[d]imidazole-5-carboxamide, 1-66
Figure imgf000357_0001
[00643] To a solution of N-[3-(l-[l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH- l,3-benzodiazol-5-yl]-N-methylformamido)propyl]carbamate, 1-68, (150 mg, 0.177 mmol, 1.0 equiv) in DCM (10 mL) was added TFA (2 mL). The resulting mixture was stirred for 1 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD Column 30* 150mm, 5um; mobile phase, water (0.1% FA) and ACN (10% ACN up to 30% in 7 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 39.7 mg (42.40%) of N-(3-aminopropyl)-l-[(2E)-4-[5- carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]- 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-N-methyl-lH-l,3-benzodiazole-5-carboxamide as a white solid LC-MS (ES, m/z): 748 (M+H+); Tf NMR (300 MHz, DMSO-^e) d 8.41 (s, 1H), 7.98- 7.90 (m, 2H), 7.72-7.69 (m, 1H), 7.51-7.34 (m, 4H), 7.18-7.16 (m, 1H), 6.58-6.56 (m, 2H), 5.93- 5.92 (m, 2H), 4.85-4.84 (m, 4H), 4.52 (q, 4H), 3.47-3.45 (m, 2H), 2.89-2.70 (m, 5H), 2.13 (s, 6H), 1.83-1.82 (m, 2H), 1.28 (t, 6H).
Example 69: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-N-(3-(2-((2-(2,6-dioxopiperidin-3- yl)-l,3-dioxoisoindolin-4-yl)oxy)acetamido)propyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-N-methyl-lH-benzo[d]imidazole-5-carboxamide, 1-65
Figure imgf000358_0001
[00644] To a stirred solution of 2-[[2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH- isoindol-4-yl]oxy]acetic acid, 1-66, (93.30 mg, 0.281 mmol, 3.0 equiv) and HATU (71.18 mg, 0.187 mmol, 2.0 equiv) in DMF (5 mL) was added DIEA (60.49 mg, 0.468 mmol, 5.0 equiv) and N-(3-aminopropyl)-l-[(2E)-4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-N-methyl-lH-l,3- benzodiazole-5-carboxamide (70 mg, 0.094 mmol, 1.0 equiv). The resulting mixture was stirred for 2 h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Prep OBD C18 Column, 30*150mm, 5um; mobile phase, water (0.1% FA) and CFbCN (20% CFbCN up to 45% in 8 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 45.9 mg (46.2%) of l-[(2E)-4-(5-[[3-(2-
357
5U B5TITUTE SH EET (RU LE 26) [[2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-isoindol-4- yl]oxy]acetamido)propyl](methyl)carbamoyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH- l,3-benzodiazol-l-yl)but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 1062 (M+H+); 'H NMR (400
Figure imgf000359_0001
S 12.50 (br s, 2H), 11.10 (s, 1H), 8.10-7.32 (m, 11H), 7.16-7.14 (m, 1H), 6.58- 6.55 (m, 2H), 5.91-5.90 (m, 2H), 5.12-5.09 (m, 1H), 4.83-4.51 (m, 10H), 3.34-2.84 (m, 8H), 2.61- 2.49 (m, 2H), 2.13 (s, 6H), 2.04-2.03 (m, 1H), 1.79-1.76 (m, 2H), 1.27 (t, 6H).
Example 70: Synthesis of N,N'-((propane-l,3-diylbis(oxy))bis(5-cyano-l-propyl-lH- benzo[d]imidazole-7,2-diyl))bis(l-ethyl-3-methyl-lH-pyrazole-5-carboxamide), 1-70
Figure imgf000359_0002
70.5
70.6
Figure imgf000360_0001
Synthesis of 70.1
[00645] A solution of ethyl methyl 4-chloro-3 -m ethoxy-5 -nitrobenzoate (14 g, 57.14 mmol, 1 equiv) in NH3Ή2O (500 mL) was stirred at 35°C overnight. The reaction mixture was cooled to r.t. The solids were collected by filtration, washed with water and dried in an oven at 40°C to give 10.4 g (79%) of 4-chloro-3-methoxy-5-nitrobenzamide as a light yellow solid. LC-MS (ES, m/z): 231 (M+H+).
Synthesis of 70.2
[00646] To a solution of 4-chloro-3-methoxy-5-nitrobenzamide, 70.1, (3.7 g, 16.045 mmol, 1 equiv) in DCE (250 mL) was added BBr3 (12.06 g, 48.135 mmol, 3 equiv) under nitrogen. The resulting mixture was stirred at 50°C overnight. The reaction mixture was cooled to r.t and quenched with ice-water. The solids were collected by filtration and the cake was washed with water and dried in an oven at 40°C to give 1.88 g (54%) of 4-chloro-3 -hydroxy-5 -nitrobenzamide as an off-white solid. LC-MS (ES, m/z): 215 (M-H+).
Synthesis of 70.3
[00647] To a solution of 4-chloro-3-hydroxy-5-nitrobenzamide, 70.2, (6.00 g, 27.703 mmol, 1.00 equiv) in l-methyl-2-pyrrolidinone (90 mL) was added 1,3-dibromopropane (2.80 g, 13.852 mmol, 0.50 equiv) and DIEA (14.32 g, 110.814 mmol, 4.00 equiv). The resulting mixture was stirred at 100°C overnight under nitrogen. The reaction mixture was cooled to r.t and diluted with water. The solids were collected by filtration and the cake was washed with water and dried in an oven at 40°C to give rise to 4.99 g (38%) of 3-[3-(5-carbamoyl-2-chloro-3-nitrophenoxy)propoxy]- 4-chloro-5-nitrobenzamide as a light yellow solid.
LC-MS (ES, m/z): 473 (M+H+). Synthesis of 70.4
[00648] To a solution of 3-[3-(5-carbamoyl-2-chloro-3-nitrophenoxy)propoxy]-4-chloro-5- nitrobenzamide, 70.3, (1.419 g, 2.999 mmol, 1 equiv) in dimethylsulfoxide (20 mL) was added potassium carbonate (2.072 g, 14.993 mmol, 5 equiv) and propan- 1 -amine (886.27 mg, 14.993 mmol, 5.00 equiv). The resulting mixture was irradiated with microwave radiation at 120°C for 30 min. The reaction mixture was cooled to r.t and diluted with water. The solids were collected by filtration and the cake was washed with water and dried in an oven at 40°C to give 1.473 g (94.37%) of 3-[3-[5-carbamoyl-3-(hydroperoxyamino)-2-(propylamino)phenoxy]propoxy]-5-nitro-4- (propylamino) benzamide as an orange solid. LC-MS (ES, m/z): 519 (M+H+).
Synthesis of 70.5
[00649] To a solution of 3-[3-[5-carbamoyl-3-(hydroperoxyamino)-2- (propylamino)phenoxy]propoxy]-5-nitro-4-(propylamino)benzamide, 70.4, (1.938 g, 3.723 mmol, 1 equiv) in 1 -methyl -2 -pyrrolidinone (20 mL) and methanol (100 mL) was added palladium on activated carbon (10%, 500 mg) under nitrogen. Then hydrogen (g) was introduced in and the resulting mixture was degassed three times and stirred at RT for 2 h. The reaction mixture was filtered. The filtrate was concentrated under reduced pressure. This resulted in 2 g (crude) of 3- amino-5-[3-[3-amino-5-carbamoyl-2-(propylamino)phenoxy]propoxy]-4-(propylamino) benzamide as orange oil. LC-MS (ES, m/z): 459 (M+H+).
Synthesis of 70.6
[00650] To a stirred solution of 3-amino-5-[3-[3-amino-5-carbamoyl-2-(propylamino) phenoxy]propoxy]-4-(propylamino)benzamide, 70.5, (2 g crude, 3.723 mmol, 1 equiv) in methanol (40 mL) was added cyanogen bromide (1.172 g, 11.169 mmol, 3 equiv). The resulting mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (37% ACN up to 50% in 7 min); UV detection at 254/220 nm. This resulted in 648 mg (34.26%) of 2-amino-7-[3-[(2-amino-5-carbamoyl-l-propyl-lH-l,3- benzodiazol-7-yl)oxy]propoxy]-l-propyl-lH-l,3-benzodiazole-5-carboxamide as a light yellow solid. LC-MS (ES, m/z): 509 (M+H+); Synthesis of 70.7
[00651] To a solution of 2-amino-7-[3-[(2-amino-5-carbamoyl-l-propyl-lH-l,3-benzodiazol- 7-yl)oxy]propoxy]-l-propyl-lH-l,3-benzodiazole-5-carboxamide, 70.6, (100 mg, 0.197 mmol, 1 equiv) in NMP (4 mL) was added l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (121.25 mg, 0.786 mmol, 4.00 equiv), DIEA (254.12 mg, 1.966 mmol, 10 equiv) and HATU (373.81 mg, 0.983 mmol, 5 equiv). The resulting mixture was irradiated with microwave radiation at 140°C for 1 h. The mixture was concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (37% ACN up to 56% in 8 min); Detector, UV 254/220 nm. This resulted in 90 mg (58.62%) of 7-(3- [[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-propyl-lH-l,3-benzodiazol-7- yl]oxy] propoxy)-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-propyl-lH-l,3-benzodiazole-5- carboxamide as a light yellow solid. LC-MS (ES, m/z): 781 (M+FC).
Synthesis of 1-70
[00652] To a solution of 7-(3-[[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l- propyl-lH-l,3-benzodiazol-7-yl]oxy]propoxy)-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l- propyl-lH-l,3-benzodiazole-5-carboxamide, 70.7, (90 mg, 0.115 mmol, 1 equiv) in DMF (4 mL) was added dropwise sulfurooyl dichloride (41.13 mg, 0.346 mmol, 3.00 equiv) at 0°C. The resulting mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, SunFire Prep C18 OBD Column 19* 150mm, 5um; mobile phase, water (0.1% FA) and ACN (67% ACN up to 85% in 7 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 20.4 mg (24%) of N-[5-cyano-7-(3-[[5-cyano-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- l-propyl-lH-l,3-benzodiazol-7-yl]oxy]propoxy)-l-propyl -lH-l,3-benzodiazol-2-yl]-l-ethyl-3- methyl-lH-pyrazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 745 (M+FL); 'H NMR (400 MHz, DMSO -d6) S 12.97 (s, 2H), 7.52 (s, 2H), 7.42 (s, 2H), 6.64 (s, 2H), 4.60 (q, 4H), 4.43 (t, 4H), 4.31 (t, 4H), 2.50-2.38 (m, 2H), 2.17 (s, 6H), 1.83-1.72 (m, 4H), 1.35 (t, 6H), 0.88 (t, 6H). Example 71: Synthesis of (31E)-5-ethyl-7,16-dimethyl-3,19-dioxo-2,5,6,14,15,20,22,29,34,41- decaazaheptacyclo[32.7.0.0A[4,8].0A[14,18].0A[21,29].0A[23,28].0A[35,40]]hentetraconta- l(41),4(8),6,15,17,21,23(28),24,26,31,35(40),36,38-tridecaene-25,38-dicarbonitrile (1-120)
Figure imgf000363_0001
Synthesis of 70.1
[00653] To a solution of (E)-l, T-(but-2-ene-l,4-diyl)bis(2-amino-lH-benzo[d]imidazole-5- carboxamide) (101 mg, 0.25 mmol, 1 equiv) and 4-(5-(5-carboxy-3-methyl-lH-pyrazol-l- yl)pentyl)-l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid, 70.1, (87 mg, 0.25 mmol, 1 equiv) in l-methylpyrrolidin-2-one (5 mL) was added HATU (209 mg, 0.55 mmol, 2.2 equiv) and DIEA (161.25 mg, 1.25 mmol, 5 equiv). The final reaction mixture was irradiated with microwave radiation at 140°C for 1 hour. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Prep OBD C18 Column 19* 150mm, 5um; mobile phase, Water (0.01%NH4HC03+0.1%NH3 H20) and ACN (24% ACN up to 36% in 10 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 21.2 mg (6.79%) of (31E)-5-ethyl-7,16-dimethyl-3, 19-dioxo- 2,5,6, 14,15,20,22,29,34,41-decaazaheptacyclo
[32.7.0.0L[4,8].0L[14, 18].0A[21,29].0A[23,28].0A[35,40]]hentetraconta-
1(41), 4(8), 6, 15, 17, 21, 23(28), 24, 26, 31,35, 37, 39-tridecaene-25, 38-dicarboxamide as an off-white solid. LC-MS (ESI): 717 (M+H+); 1H- MR (400 MHz, DMSO-t¾) d 12.85 (br s, 2H), 8.04-7.91
(m, 4H), 7.75 (dd, 2H), 7.48-7.31 (m, 4H), 6.52 (s, 1H), 5.98-5.91 (m, 1H), 5.77-5.68 (m, 1H),
4.95-4.84 (m, 4H), 4.71 (t, 2H), 4.46 (q, 2H), 2.75 (t, 2H), 2.16 (s, 3H), 2.09 (s, 3H), 1.79-1.66 (m, 2H), 1.48-1.38 (m, 2H), 1.32-1.21 (m, 5H).
Synthesis of 1-120
[00654] To a solution of (31E)-5-ethyl-7,16-dimethyl-3,19-dioxo-2,5,6, 14,15,20,22,29,34,41- decaazaheptacyclo[32.7.0.0A[4,8].0A[14,18].0A[21,29].0A[23,28].0A[35,40]]hentetraconta- 1(41), 4(8), 6, 15, 17, 21, 23(28), 24, 26, 31,35(40), 36, 38-tridecaene-25, 38-dicarboxamide, 70.1, (200 mg, 0.279 mmol, 1.00 equiv) and triethylamine (282.1 mg, 2.79 mmol, 10 equiv) in dimethyl formamide (5 mL) and tetrahydrofuran (5 mL) was added dropwise trifluoroacetic acid anhydride (351.54 mg, 1.674 mmol, 6 equiv) at 0°C. The resulting mixture was stirred overnight at RT. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridre Prep OBD C18 Column, 19* 150mm, 5um; mobile phase, water (0.1%FA) and methanol (43% methanol up to 60% in 8 min); The product- containing fractions were collected and evaporated partially and lyophilized overnight to afford 39 mg (20.56%) of (31E)-5-ethyl-7,16-dimethyl-3,19-dioxo-2,5,6,14, 15,20,22,29,34,41- decaazaheptacyclo[32.7.0.0A[4,8].0A[14,18].0A[21,29].0A[23,28].0A[35,40]]hentetraconta- l(41),4(8),6,15,17,21,23(28),24,26,31,35(40),36,38-tridecaene-25,38-dicarbonitrile as a white solid. LC-MS (ES, m/z): 341 (M/2+H+); H-NMR: (400 MHz, DMSO-^e): d 12.89 (br s, 2H), 7.76 (s, 2H), 7.69 (dd, 2H), 7.54 (dd, 2H), 6.55 (s, 1H), 5.98-5.92 (m, 1H), 5.79-5.67 (m, 1H),
4.95-4.82 (m, 4H), 4.49-4.38 (m, 4H), 4.69 (t, 2H), 4.44 (q, 2H), 2.79-2.71 (m, 2H), 2.16 (s, 3H), 2.09 (s, 3H), 1.76-1.65 (m, 2H), 1.46-1.37 (m, 2H), 1.32-1.21 (m, 5H).
Example 72: Synthesis of (E)-N-(4-((2-amino-2-oxoethyl)(methyl)amino)-l-(4-(2-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-lH- benzo[d]imidazol-2-yl)-l-ethyl-3-methyl-lH-pyrazole-5-carboxamide, 1-64
Figure imgf000365_0001
Synthesis of 72.1
[00655] Into a 100-mL round-bottom flask, was placed a solution of N-tert-butyl-2-[(3-fluoro- 2-nitrophenyl)(methyl)amino]acetamide (2 g, 7.060 mmol, 1 equiv) in DMSO (30 mL). To the solution was added tert-butyl N-[(2E)-4-aminobut-2-en-l- yl] carbarn ate hydrochloride (3.14 g, 14.119 mmol, 2 equiv) and DIEA (2.74 g, 21.179 mmol, 3 equiv). The resulting solution was stirred overnight at 120 degrees C. LCMS showed the reaction was complete, and the mixture was quenched with water, extracted with EA. The organic layer was dried with NazSCL, concentrated to afford the product (2.1 g crude) as a brown oil. LCMS (ESI): M+H+= 450.4.
Synthesis of 72.2
[00656] Into a 250-mL round-bottom flask, was placed a solution of tert-butyl N-[(2E)-4-[(3- [[(tert-butylcarbamoyl)methyl](methyl)amino]-2-nitrophenyl)amino]but-2-en-l- yljcarbamate, (2 g, 4.449 mmol, 1 equiv) in DCM (40 mL). To the solution was added HC1 in 1,4- dioxane 72.1, (10 mL). The resulting solution was stirred overnight at room temperature. LCMS showed the reaction was complete and the mixture was quenched with water. The pH value of the solution was adjusted to 9 with sodium carbonate (aq) (1 mol/L) and extracted with DCM. The organic layer was dried with NaiSCL, concentrated to afford the product (2.1 g crude) as a brown solid. LCMS (ESI): M+H+= 350.3.
Synthesis of 72.3
[00657] Into a 40-mL tube, was placed a solution of 2-[(3-[[(2E)-4-aminobut-2-en-l-yl]amino]- 2-nitrophenyl)(methyl)amino]-N-tert-butylacetamide, 72.2, (1 g, 2.862 mmol, 1 equiv) and 1- fluoro-2-nitrobenzene (0.44 g, 0.003 mmol, 1.1 equiv) in DMSO (10 mL). To the solution was added DIEA (1.11 g, 0.009 mmol, 3 equiv). The resulting solution was stirred overnight at 120degrees C. LCMS showed the reaction was complete, and the mixture was quenched with water, extracted with EA. The organic layer was dried with NaiSCL and concentrated to afford solid (1 g crude). LCMS (ESI): M+H = 471.4.
Synthesis of 72.4
[00658] Into a 100-mL round-bottom flask, was placed 2-[(3-[[(2E)-4-[(2- aminophenyl)amino]but-2-en-l-yl]amino]-2-nitrophenyl)(methyl)amino]-N-tert-butylacetamide, 72.3 (1 g, 2.270 mmol, 1 equiv)N-tert-butyl-2-[methyl(2-nitro-3-[[(2E)-4-[(2- nitrophenyl)amino]but-2-en-l - yl]amino]phenyl)amino]acetamide (800.00 mg, 1.700 mmol, 1.00 equiv) in AcOH (15 mL). To the solution was added Zn (556.05 mg, 8.501 mmol, 5.00 equiv). The resulting solution was stirred overnight at room temperature. The solids were filtered out. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1/1) to give the product (480 mg) as a brown solid. LCMS (ESI): M+H+= 411.5.
Synthesis of 72.5
[00659] Into a 100-mL round-bottom flask, was placed a solution of 2-[(2-amino-3-[[(2E)-4- [(2-aminophenyl)amino]but-2-en-l-yl]amino]phenyl)(methyl)amino]-N-tert-butylacetamide (480 mg, 1.169 mmol, 1 equiv) in MeOH (10 mL). To the solution was added BrCN (371.50 mg, 3.507 mmol, 3 equiv). The resulting solution was stirred overnight at room temperature. The resulting mixture was concentrated under vacuum to give the product (500 mg crude) as a yellow solid. LCMS (ESI): M+H+ = 461.4.
Synthesis of 72.6
[00660] Into a 100-mL round-bottom flask, was placed a solution of 2-([2-amino-l-[(2E)-4-(2- amino-lH-l,3-benzodiazol-l-yl)but-2-en-l-yl]-lH-l,3-benzodiazol-4-yl](methyl)amino)-N-tert- butylacetamide, 72.5, (500 mg, 1.303 mmol, 1 equiv) and l-ethyl-3-methyl-lH-pyrazole-5- carboxylic acid (1004.17 mg, 6.513 mmol, 5 equiv) in DMF (20 mL). To the solution was added HATU (2476.61 mg, 6.513 mmol, 5 equiv) and DIEA (1346.91 mg, 10.422 mmol, 8 equiv). The resulting solution was stirred overnight at room temperature. The solution was purified by Prep- TLC with EA/PE=1/1 to give the product (200 mg) as a yellow solid. LCMS (ESI): M+H+= 733.6. Synthesis of 1-64
[00661] Into a 50-mL round-bottom flask, was placed N-(4-[[(tert- butylcarbamoyl)methyl](methyl)amino]-l-[(2E)-4-[2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3-benzodiazol-2-yl)-l-ethyl-3-methyl-lH- pyrazole-5-carboxamide, 72.6, (150 mg, 0.205 mmol, 1 equiv) in TFA (2 mL) and methanesulfonic acid (2 mL). The resulting solution was stirred for 3 hr at room temperature. The resulting mixture was concentrated. The pH value of the solution was adjusted to 9 with NaiCCL (1 mol/L). The resulting solution was extracted with 3x30 mL of dichloromethane The crude product (100 mg) was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD Column 30* 150mm 5um, n; mobile phase, Water (0.1%FA) and ACN (31% PhaseB up to 55% in 8 min); Flow Rate, 30 mL/min; Detector, UV254nm. This resulted in 27.8 mg (20.1%) of N-[4- [(carbamoylmethyl)(methyl)amino]-l-[(2E)-4-[2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH- l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3-benzodiazol-2-yl]-l-ethyl-3-methyl-lH-pyrazole-5- carboxamide as an off-white solid. LC-MS (ESI): M+H+ = 677.2. 'H-NMR (400 MHz, CD3OD) d 7.46 (d, 1H), 7.33 (d, 1H), 7.20-7.16 (m, 2H), 7.12 (t, 1H), 6.96 (d, 1H), 6.85 (d, 1H), 6.58 (s, 2H), 5.86 (s, 2H), 5.00-4.80 (m, 4H), 4.65-4.55 (m, 4H), 3.81 (s, 2H), 3.00 (s, 3H), 2.20 (s, 6H), 1.40-1.25 (m, 6H).
Example 73: Synthesis of l-(((lr,4r)-4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)cyclohexyl)methyl)-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-63
Figure imgf000368_0001
73.2 73.3
Figure imgf000369_0001
Synthesis of 73.1
[00662] To a stirred solution of 4-chloro-3-nitrobenzamide (1.00 g, 4.986 mmol, 1.00 equiv) and K2C03 (2.07 g, 14.957 mmol, 3.00 equiv) in DMF (20 ml) was added tert-butyl N-[4- (aminomethyl)cyclohexyl]carbamate (1.14 g, 4.986 mmol, 1.00 equiv). The resulting mixture was stirred overnight at 80oC. The reaction was cooled to RT and diluted with water. The precipitated solids were collected by filtration. This resulted in 1.3 g (66.44%) of tert-butyl N-[(lr,4r)-4-[[(4- carbamoyl-2-nitrophenyl)amino]methyl]cyclohexyl]carbamate as a yellow solid. LC-MS (ES, m/z): 415 (M+Na+).
Synthesis of 73.2
[00663] To a solution of tert-butyl N-[(lr,4r)-4-[[(4-carbamoyl-2- nitrophenyl)amino]methyl]cyclohexyl]carbamate, 73.1 (1.3 g, 3.312 mmol, 1.00 equiv) in DCM (40 ml) was added TFA (8 ml). The resulting mixture was stirred for 1 h at room temperature. The mixture was concentrated under vacuum. This resulted in 1 g crude of 3-nitro-4-([[(lr,4r)-4- aminocyclohexyl]methyl]amino)benzamide as a brown oil. LC-MS (ES, m/z): 293 (M+H+). Synthesis of 73.3
[00664] To a stirred solution of 3-nitro-4-([[(lr,4r)-4- aminocyclohexyl]methyl]amino)benzamide, 73.2, (1 g crude, 3.312 mmol, 1.00 equiv) and K2C03 (1.37 g, 9.934 mmol, 3.00 equiv) in DMF (20 ml) was added 4-chloro-3-nitrobenzamide (797 mg, 3.973 mmol, 1.20 equiv). The resulting mixture was stirred overnight at 80oC. The reaction was cooled to RT and diluted with water. The precipitated solids were collected by filtration. This resulted in 876 mg (58%) of 3-nitro-4-([[(lr,4r)-4-[(4-carbamoyl-2- nitrophenyl)amino]cyclohexyl]methyl]amino)benzamide as a yellow solid. LC-MS (ES, m/z): 457 (M+H+).
Synthesis of 73.4
[00665] To a stirred solution of 3-nitro-4-([[(lr,4r)-4-[(4-carbamoyl-2- nitrophenyl)amino]cyclohexyl]methyl]amino)benzamide, 73.3, (876 mg, 1.921 mmol, 1 equiv) in AcOH (20 mL), MeOH (20 ml) and NMP (20 mL) and Zn (1.23 g, 19.21 mmol, 10 equiv). The resulting mixture was stirred overnight at RT. The mixture was filtered. The filtrate was concentrated under reduced pressure. This resulted in 900 mg crude of l-(2-amino-3-[[(2E)-4-[(2- aminophenyl)amino]but-2-en-l-yl]amino]phenyl)ethan-l-one as a dark yellow oil. LC-MS (ES, m/z): 397 (M+H+).
Synthesis of 73.5
[00666] To a solution of 3-amino-4-([[(lr,4r)-4-[(2-amino-4- carbamoylphenyl)amino]cyclohexyl]methyl]amino)benzamide (900 mg crude, 1.921 mmol, 1.00 equiv) in MeOH (10 ml) was added BrCN (407.25 mg, 3.842 mmol, 2.00 equiv). The resulting mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, C18; mobile phase, Water (10 MMOL/L NH4HCO3) and ACN (28% ACN up to 40% in 10 min); Detector, UV 254/220 nm. This resulted in 300 mg (35%) of 2-amino-l-[(lr,4r)-4-[(2-amino-5-carbamoyl-lH-l,3- b enzodi azol - 1 -yl)m ethyl ] cy cl ohexyl ] - 1 H- 1 , 3 -b enzodi azol e- 5 - carboxamide as a brown solid. LC-MS (ES, m/z): 447 (M+H+).
Synthesis of 1-63
[00667] To a stirred solution of 2-amino-l-[(lr,4r)-4-[(2-amino-5-carbamoyl-lH-l,3- b enzodi azol - 1 -yl)m ethyl ] cy cl ohexyl ] - 1 H- 1 , 3 -b enzodi azol e- 5 -carb oxami de,
73.5, (165 mg, 0.370 mmol, 1 equiv) and l-ethyl-3-methyl-lH-pyrazole-5- carboxylic acid (113.94 mg, 0.739 mmol, 2 equiv) in NMP (5 ml) was added HATU (337.21 mg, 0.887 mmol, 2.4 equiv) and DIEA (238.80 mg, 1.848 mmol, 5 equiv). The final rea ction mixture was irradiated with microwave radiation for 1 h at 140°C. The mixture was cooled to RT and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD column, 30* 150mm, 5um; mobile phase, water (0.1% FA) and ACN (20% ACN up to 50% in 8 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 61.3 mg (22.39%) of 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-[(lr,4r)-4- [[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l- yl]methyl]cyclohexyl]-lH-l,3-benzodiazole-5-carboxamide as an off-white solid. LC-MS (ES, m/z): 719 (M+H+); ¾ NMR (400 MHz, DMSO- ) 5 12.85 (br s, 2H), 8.09-7.95 (m, 4H), 7.86- 7.66 (m, 4H), 7.33 (d, 2H), 6.72 (s, 1H), 6.61 (s, 1H), 4.71-4.59 (m, 5H), 4.20 (d, 2H), 2.45-2.12 (m, 9H), 1.94-1.81 (m, 4H), 1.48-1.35 (m, 8H).
Example 74: Synthesis of (E)-N,N'-(but-2-ene-l,4-diylbis(4-((2-amino-2- oxoethyl)(methyl)amino)-lH-benzo[d]imidazole-l,2-diyl))bis(l-ethyl-3-methyl-lH- pyrazole-5-carboxamide), 1-62
Figure imgf000371_0001
72.2 74.1
Figure imgf000372_0001
Synthesis of 74.1
[00668] To a solution of (E)-2-((3-((4-aminobut-2-en-l-yl)amino)-2- nitrophenyl)(methyl)amino)-N-(tert-butyl)acetamide, 72.2, (1 g, 2.865 mmol, 1 equiv) in DMSO (20 mL) was added N-(tert-butyl)-2-((3-fluoro-2-nitrophenyl)(methyl)amino)acetamide (1.2 g, 4.298 mmol, 1.5 equiv) and DIEA (1.1 g, 8.598 mmol, 3 equiv). The resulting mixture was stirred at 120°C overnight. The mixture was cooled to RT, diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SC>4 and concentrated under reduced pressure. This resulted in 800 mg (45.62%) of (E)-2,2'-(((but-2-ene-l,4- diylbis(azanediyl))bis(2-nitro-3,l-phenylene))bis(methylazanediyl))bis(N-(tert-butyl)acetamide) as a red solid. LC-MS (ES, m/z): 613 (M+H+).
Synthesis of 74.2
[00669] To a stirred solution of N-tert-butyl-2-[(3-[[(2E)-4-[(3-[[(tert- butylcarbamoyl)methyl](methyl)amino]-2-nitrophenyl)amino]but-2-en-l-yl]amino]-2- nitrophenyl)(methyl)amino]acetamide, 74.1, (770.00 mg, 1.257 mmol, 1.00 equiv) in AcOH (10 mL) was added Zn (0.76 g, 11.663 mmol, 5 equiv). The reaction mixture was stirred at room temperature overnight. The solids were filtered out. The filtrate was concentrated under reduced pressure. The pH value of the solution was adjusted to 8 with sat.NaHCCb solution and extracted with DCM. The combined organic layers were washed with brine, dried over NaiSCE and concentrated under reduced pressure. The resulted in 600 mg (86.38%) of 2-[(2-amino-3-[[(2E)-4-[(2-amino-3-[[(tert- butylcarbamoyl)methyl](methyl)amino]phenyl)amino]but-2-en-l- yl]amino]phenyl)(methyl)amino]-N-tert-butylacetamide as a light brown solid. LC-MS (ES, m/z): 553 (M+H+).
Synthesis of 74.3
[00670] To a stirred solution of 2-[(2-amino-3-[[(2E)-4-[(2-amino-3-[[(tert- butylcarbamoyl)methyl](methyl)amino]phenyl)amino]but-2-en-l- yl]amino]phenyl)(methyl)amino]-N-tert-butylacetamide, 74.2, (600 mg, 1.085 mmol, 1 equiv) in MeOH (10 mL) was added BrCN (344.91 mg, 3.256 mmol, 3 equiv). The resulting mixture was stirred at RT for 2 hours. The mixture was concentrated under reduced pressure. This resulted in 700 mg crude of 2-([2-amino-l-[(2E)-4-(2-amino-4-[[(tert- butylcarbamoyl)methyl](methyl)amino]-lH-l,3-benzodiazol-l-yl)but-2-en-l-yl]-lH-l,3- benzodiazol-4-yl](methyl)amino)-N-tert-butylacetamide as a light brown solid. LC-MS (ES, m/z): 603 (M+H ).
Synthesis of 74.4
[00671] To a stirred solution of 2-([2-amino-l-[(2E)-4-(2-amino-4-[[(tert- butylcarbamoyl)methyl](methyl)amino]-lH-l,3-benzodiazol-l-yl)but-2-en-l-yl]-lH-l,3-
372
5UB5TITUTE SHEET (RULE 26) benzodiazol-4-yl](methyl)amino)-N-tert-butylacetamide, 74.3, (700 mg crude, 1.085 mmol, 1.00 equiv) in DMD (20 mL) was added HATU (1614.81 mg, 4.247 mmol, 4.00 equiv), DIEA (686.11 mg, 5.309 mmol, 5.00 equiv) and l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (491.06 mg, 3.185 mmol, 3.00 equiv). The reaction mixture was stirred at RT overnight. The mixture was concentrated under vacuum, diluted with water and extracted with CH2CI2. The combined organic layers layers were washed with brine, dried over anhydrous Na2SC>4 and concentrated under reduced pressure. This resulted in 1 g (86.10%) of N-(4-[[(tert- butylcarbamoyl)methyl](methyl)amino]-l-[(2E)-4-(4-[[(tert- butylcarbamoyl)methyl](methyl)amino]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazol-l-yl)but-2-en-l -yl]-lH-l, 3-benzodiazol-2-yl)-l -ethyl -3-methyl-lH-pyrazole-5- carboxamide as a light dark yellow solid. LC-MS (ES, m/z): 875 (M+TE).
Synthesis of 1-62
[00672] To a stirred solution of N-(4-[[(tert-butylcarbamoyl)methyl](methyl)amino]-l-[(2E)-4- (4-[[(tert-butylcarbamoyl)methyl](methyl)amino]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- lH-l,3-benzodiazol-l-yl)but-2-en-l-yl]-lH-l,3-benzodiazol-2-yl)-l-ethyl-3-methyl-lH- pyrazole-5-carboxamide, 74.4, (200 mg, 0.229 mmol, 1.00 equiv) in TFA (5 mL) was added methanesulfonic acid (5 mL). The resulting solution was stirred for 1 h at room temperature. The mixture was concentrated under vacuum. The residue was basified to pH 8 with saturated sat. Na2CC>3 solution and extracted with CH2CI2. The combined organic layers were washed with brine, dried over Na2SC>4 and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Atlantis Prep T3 OBD Column, 19* 150mm, 5um; mobile phase, Water (0.1%FA) and ACN (37% ACN up to 55% in 8 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 66.9 mg (38.34%) of N-[4- [(carbamoylmethyl)(methyl)amino]-l-[(2E)-4-[4-[(carbamoylmethyl)(methyl)amino]-2-(l-ethyl- 3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3-benzodiazol-2- yl]-l-ethyl-3-methyl-lH-pyrazole-5-carboxamide as awhite solid. LCMS (ES, m/z): 763 (M+H+); 1H NMR (300 MHz, CF3COOD) d 8.14-8.04 (m, 2H), 7.93-7.81 (m, 4H), 7.36 (s, 2H), 6.45-6.38 (m, 2H), 5.36 (s, 4H), 5.08-4.90 (m, 8H), 3.77 (s, 6H), 2.64 (s, 6H), 1.75 (t, 6H). Example 75: Synthesis of (E)-l-ethyl-N-(l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-4-(l-hydroxyethyl)-lH- benzo [d] imidazol-2-yl)-3-methyl- lH-pyrazole-5-carboxamide, 1-61
Br
NO? Tributyl(1 -ethoxyvinyl)tin HCI/1 ,4-dioxane
Pd(PPh3)2CI2/1 ,4-dioxane DCM
F
Figure imgf000375_0002
Figure imgf000375_0001
75.1 75.2
Figure imgf000375_0003
Synthesis of 75.1
[00673] To a stirred solution of l-bromo-3-fluoro-2 -nitrobenzene (500 mg, 2.273 mmol, 1 equiv) and tributyl(l -ethoxyethenyl)stannane (1.23 g, 3.409 mmol, 1.5 equiv) in THF (20 mL) was added Pd(PPh3)2Ch (159.52 mg, 0.227 mmol, 0.1 equiv). The resulting mixture was stirred for 2h at 70 °C under nitrogen atmosphere. The reaction was cooled to rt, diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous NaiSCE and concentrated under reduced pressure. This resulted in 500 mg crude of 1 -(1 -ethoxy ethenyl)- 3-fluoro-2-nitrobenzeneas a black oil. LC-MS (ES, m/z): 212 (M+H+).
Synthesis of 75.2
[00674] To a stirred solution of 1-(1 -ethoxy ethenyl)-3-fluoro-2-nitrobenzene, 75.1 (500 mg, 2.368 mmol, 1.0 equiv) in DCM (10 mL) was added HC1 (4mol/L, 5 mL). The resulting mixture was stirred for lh at room temperature. The mixture was concentrated under reduced pressure. This resulted in 300 mg (69.19%) of l-(3-fluoro-2-nitrophenyl)ethan-l-one as a black oil. LC-MS (ES, m/z): 184 (M+H+).
Synthesis of 75.3
[00675] To a stirred solution of l-(3-fluoro-2-nitrophenyl)ethan-l-one, 75.2 (1.00 g, 5.460 mmol, 1.00 equiv) and tert-butyl N-[(2E)-4-aminobut-2-en-l-yl]carbamate hydrochloride (1.22 g, 5.460 mmol, 1.00 equiv) in DMF (20 mL) was added K2CO3 (2.26 g, 16.381 mmol, 3 equiv). The resulting mixture was stirred for 2 h at 70 °C. The reaction was cooled to rt, diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2S04 and concentrated under reduced pressure. The residue was applied on a silica gel column with PE/EtOAc (2/1). This resulted in 600 mg (31.65%) of tert-butyl N-[(2E)-4-[(3-acetyl-2- nitrophenyl)amino]but-2-en-l-yl]carbamate as a light yellow solid. LC-MS (ES, m/z): 372 (M+Na+).
Synthesis of 75.4
[00676] To a stirred solution of tert-butyl N-[(2E)-4-[(3-acetyl-2-nitrophenyl)amino]but-2-en- l-yl]carbamate, 75.3 (600 mg, 1.717 mmol, 1 equiv) in DCM (10 mL) was added TFA (3 mL). The resulting mixture was stirred for 1 h at room temperature. The mixture was concentrated under vacuum. This resulted in 500 mg crude of l-(3-[[(2E)-4-aminobut-2-en-l-yl]amino]-2- nitrophenyl)ethan-l-one as a light yellow solid. LC-MS (ES, m/z): 250 (M+H+).
Synthesis of 75.5
[00677] To a stirred solution of l-(3-[[(2E)-4-aminobut-2-en-l-yl]amino]-2-nitrophenyl)ethan- 1-one, 75.4 (500 mg, 2.006 mmol, 1 equiv) and l-fluoro-2-nitrobenzene (339.63 mg, 2.407 mmol, 1.2 equiv) in DMF (10 mL) was added DIEA (777.73 mg, 6.018 mmol, 3.0 equiv). The resulting mixture was stirred for 8 h at 70 °C. The reaction was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous NaiSCL and concentrated under reduced pressure. The residue was applied on a silica gel column with PE/EtOAc (1/1). This resulted in 300 mg (40.38%) of l-(2-nitro-3-[[(2E)-4-[(2- nitrophenyl)amino]but-2-en-l-yl]amino]phenyl)ethan-l-one as a dark yellow solid. LC-MS (ES, m/z) 371(M+H+).
Synthesis of 75.6
[00678] To a stirred solution of l-(2-nitro-3-[[(2E)-4-[(2-nitrophenyl)amino]but-2-en-l- yl]amino]phenyl)ethan-l-one, 75.5 (400 mg, 1.080 mmol, 1 equiv) in acetic acid (20 mL) were added zinc (706.11 mg, 10.8 mmol, 10.0 equiv). The resulting mixture was stirred for 4 h at room temperature. The mixture was filtered. The filtrate was concentrated under reduced pressure. This resulted in 300 mg (89.49%) of l-(2-amino-3-[[(2E)-4-[(2-aminophenyl)amino]but-2-en-l- yl]amino]phenyl)ethan-l-one as a dark yellow solid. LC-MS (ES, m/z): 310 (M+H+).
Synthesis of 75.7
[00679] To a stirred solution of l-(2-amino-3-[[(2E)-4-[(2-aminophenyl)amino]but-2-en-l- yl]amino]phenyl)ethan-l-one, 75.6 (300 mg, 0.966 mmol, 1 equiv) in MeOH (20 mL) were added BrCN (512 mg, 4.83 mmol, 5 equiv). The resulting mixture was stirred for 4h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, Cl 8; mobile phase, Water (10MMOL/L H4HCO3) and ACN (21% ACN up to 31% in 10 min); Detector, UV 254/220 nm. This resulted in 200 mg (57.41%) of l-[2-amino-l-[(2E)-4-(2-amino-lH-l,3-benzodiazol-l-yl)but-2-en-l-yl]-lH-l,3- benzodiazol-4-yl]ethan-l-one as a black solid. LC-MS (ES, m/z): 361 (M+EL).
Synthesis of 75.8 [00680] To a stirred solution of l-[2-amino-l-[(2E)-4-(2-amino-lH-l,3-benzodiazol-l-yl)but-
2-en-l-yl]-lH-l,3-benzodiazol-4-yl]ethan-l-one, 75.7 (200 mg, 0.555 mmol, 1 equiv) and 1-ethyl-
3-methyl-lH-pyrazole-5-carboxylic acid (342.20 mg, 2.220 mmol, 4.0 equiv) in DMF (5 mL) were added HATU (843.97 mg, 2.220 mmol, 4.0 equiv) and DIEA (573.74 mg, 4.439 mmol, 8.0 equiv). The resulting mixture was stirred for 4h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (24% ACN up to 40% in 10 min); Detector, UV 254/220 nm. This resulted in 150 mg (42.7%) ofN-[4-acetyl-l-[(2E)-4-[2-(l- ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3- benzodiazol-2-yl]-l-ethyl-3-methyl-lH-pyrazole-5-carboxamide as an off-white solid. LC-MS (ES, m/z): 317 (M/2+FE).
Synthesis of 1-61
[00681] To a stirred solution of N-[4-acetyl-l-[(2E)-4-[2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3-benzodiazol-2-yl]-l-ethyl-3-methyl-lH- pyrazole-5-carboxamide, 75.8 (70.0 mg, 0.111 mmol, 1.0 equiv) in MeOH (10 mL) were added NaBFE (12.22 mg, 0.332 mmol, 3.0 equiv). The resulting mixture was stirred for lh at room temperature. The reaction was quenched with water and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 column, 30* 150mm, 5um; mobile phase, water (0.1% FA) and ACN (48% ACN up to 64% in 8 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 30.7 mg (44%) of l-ethyl-N-[l-[(2E)-4-[2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-4-(l-hydroxyethyl)-lH-l,3- benzodiazol-2-yl]-3-methyl-lH-pyrazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 635 (M+FE); ¾ NMR (300 MHz, DMSO -d6) d 12.67 (s, 1H), 12.18 (s, 1H), 7.49 (d, 1H), 7.38 (d, 1H), 7.29 (d, 1H), 7.22-7.04 (m, 4H), 6.56-6.51 (m, 2H), 5.90-5.83 (m, 3H), 5.12-5.11 (m, 1H), 4.91-4.83 (m, 4H), 4.54 (q, 4H), 2.13 (s, 6H), 1.42 (d, 3H), 1.27 (t, 6H).
Example 76: Synthesis of (E)-N,N'-(but-2-ene-l,4-diylbis(4-(l-hydroxyethyl)-lH- benzo[d]imidazole-l,2-diyl))bis(l-ethyl-3-methyl-lH-pyrazole-5-carboxamide), 1-57
Figure imgf000379_0001
Synthesis of 76.1 [00682] To a stirred solution of l-(3-[[(2E)-4-aminobut-2-en-l-yl]amino]-2-nitrophenyl)ethan-
1-one, 75.4 (1.00 g, 4.012 mmol, 1.00 equiv) and l-(3-fluoro-2-nitrophenyl)ethan-l-one (734.70 mg, 4.012 mmol, 1.00 equiv) in DMF (20 mL) were added DIEA (1.56 g, 12.035 mmol, 3.00 equiv). The resulting mixture was stirred for 2h at 70 °C under nitrogen atmosphere. The reaction was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous NaiSCL and concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, Cl 8; mobile phase, Water (10MMOL/L NH4HCO3) and ACN (51% ACN up to 62% in 10 min); Detector, UV 254/220 nm. This resulted in 700 mg (42.31%) of l-(3-[[(2E)-4-[(3-acetyl-2-nitrophenyl)amino]but-2-en-l- yl]amino]-2-nitrophenyl)ethan-l-one as a dark yellow solid. LC-MS (ES, m/z): 413 (M+FE). Synthesis of 76.2
[00683] To a stirred solution of l-(3-[[(2E)-4-[(3-acetyl-2-nitrophenyl)amino]but-2-en-l- yl]amino]-2-nitrophenyl)ethan-l-one, 76.1 (700.00 mg, 1.697 mmol, 1.00 equiv) in acetic acid (20 mL) was added zinc (1.11 g, 16.974 mmol, 10 equiv) at room temperature. The resulting mixture was stirred for 4h at room temperature. The mixture was filtered. The filtrate was concentrated under reduced pressure. This resulted in 600 mg crude of l-(3-[[(2E)-4-[(3-acetyl-2- aminophenyl)amino]but-2-en-l-yl]amino]-2-aminophenyl)ethan-l-one as a black oil. LC-MS (ES, m/z) 353 (M+H+).
Synthesis of 76.3
[00684] To a stirred solution l-(3-[[(2E)-4-[(3-acetyl-2-aminophenyl)amino]but-2-en-l- yl]amino]-2-aminophenyl)ethan-l-one, 76.2 (600 mg, 1.702 mmol, 1.00 equiv) in MeOH (10 mL) was BrCN (1.08 g, 10.215 mmol, 6.00 equiv). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. This resulted in 500 mg (72.98%) of l-[l-[(2E)-4-(4-acetyl-2-amino-lH-l,3-benzodiazol-l-yl)but-2-en-l-yl]-2-amino- lH-l,3-benzodiazol-4-yl]ethan-l-one as a black oil. LC-MS (ES, m/z): 2403 (M+FL).
Synthesis of 76.4
[00685] To a stirred solution of l-[l-[(2E)-4-(4-acetyl-2-amino-lH-l,3-benzodiazol-l-yl)but-
2-en-l-yl]-2-amino-lH-l,3-benzodiazol-4-yl]ethan-l-one, 76.3 (500 mg, 1.242 mmol, 1.00 equiv) and l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (287.30 mg, 1.864 mmol, 1.50 equiv) in DMF (10 mL) were added HATU (708.58 mg, 1.864 mmol, 1.50 equiv) and DIEA (802.84 mg, 6.212 mmol, 5.00 equiv). The resulting mixture was stirred for 5h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, C18; mobile phase, Water (0.5% FA) and ACN (37% ACN up to 47% in 10 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to give 300 mg (36%) of N-[4-acetyl-l-[(2E)- 4-[4-acetyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]- lH-l,3-benzodiazol-2-yl]-l-ethyl-3-methyl-lH-pyrazole-5-carboxamide as an off-white solid. LC-MS (ES, m z): 338.5 (M/2+H+).
Synthesis of 1-57
[00686] To a stirred solution of N-[4-acetyl-l-[(2E)-4-[4-acetyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3-benzodiazol-2-yl]-l-ethyl-3- methyl-lH-pyrazole-5-carboxamide, 76.4 (100 mg, 0.148 mmol, 1 equiv) in MeOH (20 mL) was added NaBFC (54.57 mg, 1.482 mmol, 10 equiv). The resulting mixture was stirred for lh at room temperature. The reaction was quenched with water and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Atlantis Prep T3 OBD Column, 19* 150mm, 5um; mobile phase, water (0.1% FA) and ACN (50% ACN up to 73% in 8 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 54.1 mg (54%) of 1 -ethyl -N-[l-[(2E)-4- [2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-4-(l-hydroxyethyl)-lH-l,3-benzodiazol-l-yl]but-2- en-l-yl]-4-(l-hydroxyethyl)-lH-l,3-benzodiazol-2-yl]-3-methyl-lH-pyrazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 701 (M+Na+); Ή NMR (400 MHz, DMSC ) d 12.20 (s, 2H), 7.31 (d, 2H), 7.16-7.07 (m, 4H), 6.59 (s, 2H), 5.93-5.89 (m, 4H), 5.13-5.12 (m, 2H), 4.88-4.84 (m, 4H), 4.54 (q, 4H), 2.15 (s, 6H), 1.44 (d, 6H), 1.28 (t, 6H).
Example 77: Synthesis of (E)-8-ethyl-10,18-dimethyl-8,ll,12,13,14,15,28,29,32,33- decahydrobenzo[4,5]imidazo[l,2-a]benzo[4,5]imidazo[2,l-p]dipyrazolo[5,l-e:4',3'- l][l,3,6,15,17]pentaazacyclotricosine-7,20(6H,21H)-dione, 1-121
Figure imgf000382_0001
Synthesis of 77.1
[00687] To a stirred solution of (E)-dimethyl hex-3-enedioate (8.6 g, 50 mmol, 1 equiv) in tetrahydrofuran (150 mL) were added in several portions lithium aluminium hydride (3.8 g, 100 mmol, 2 equiv) at 0°C. The reaction mixture was stirred for 4h at 0°C. The mixture was quenched with ice-water and extracted with dichloromethane. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 3.2 g (55.17%) of (E)-hex-3-ene-l,6-diol as a colorless oil. LCMS (ES, m/z): 117 (M+H+).
Synthesis of 77.2
[00688] To a stirred solution of (E)-hex-3-ene-l,6-diol, 77.1 (3.2 g, 27.59 mmol, 1 equiv) in dichloromethane (100 mL) was added carbon tetrabromide (13.74 g, 41.385 mmol, 1.5 equiv) and triphenyl phosphine (10.84 g, 41.385 mmol, 1.5 equiv). The reaction mixture was stirred overnight at r.t. The mixture was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography with PE/ethyl acetate (10/1) to give 7 g (crude) of (E)-l,6- dibromohex-3-ene as a light yellow oil. LCMS (ES, m/z): 241 (M+H+).
Synthesis of 77.3
[00689] To a stirred solution of (E)-l,6-dibromohex-3-ene, 77.2 (7 g crude, 27.59 mmol, 1 equiv) in dimethyl formamide (150 mL) was added potassium l,3-dioxoisoindolin-2-ide (10.21 g, 55.18 mmol, 2 equiv). The reaction mixture was stirred at 90°C overnight. The reaction mixture was cooled to r.t, diluted with water. The precipitated solids were collected by filtration. This resulted in 6 g (58.14%) of (E)-2,2'-(hex-3-ene-l,6-diyl)diisoindoline-l,3-dione as a yellow solid. LCMS (ES, m/z): 375 (M+H+).
Synthesis of 77.4
[00690] To a solution of (E)-2,2'-(hex-3-ene-l,6-diyl)diisoindoline-l,3-dione, 77.3 (6 g, 16 mmol, 1 equiv) in ethanol (30 mL) was added hydrazine hydrate (80%, 5.6 g, 160 mmol, 10 equiv). The reaction mixture was stirred for 2h at 80°C. The reaction mixture was cooled to r.t. The solids were filtered out. The filtrate was concentrated under vacuum. This resulted in 0.96 g (52.63%) of (E)-hex-3-ene- 1,6-diamine as a yellow oil. LCMS (ES, m/z): 115 (M+H+).
Synthesis of 77.5
[00691] To a stirred solution of (E)-hex-3-ene-l, 6-diamine, 77.4 (440 mg, 3.826 mmol, 1 equiv) in dimethyl formamide (10 mL) was added potassium carbonate (1.58 g, 11.478 mmol, 3 equiv) and l-fluoro-2-nitrobenzene (1.079 g, 7.652 mmol, 2 equiv). The resulting solution was stirred for 3 hours at 80°C. The mixture was cooled to r.t, diluted with water and extracted with dichloromethane. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography with PE/ethyl acetate (3/1) to give 600 mg (44%) of (E)-NL,N6-bis(2- nitrophenyl)h ex-3 -ene- 1,6-diamine as a yellow solid. LCMS (ES, m/z): 357 (M+H+).
Synthesis of 77.6
[00692] To a stirred solution of (E)-N1,N6-bis(2-nitrophenyl)hex-3-ene-l, 6-diamine, 77.5 (600 mg, 1.685 mmol, 1 equiv) in acetic acid (10 mL) was added zinc powder (1.08 g, 16.85 mmol, 10 equiv). The resulting solution was stirred overnight at room temperature. The solids were filtered out. The filtrate was concentrated under reduced pressure. The pH value of the solution was adjusted to 8 with sat. sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The resulted in 300 mg (60.12%) of (E)-N1,N1'-(hex-3-ene-l,6- diyl)dibenzene-l, 2-diamine as a yellow solid. LCMS (ES, m/z): 297 (M+H+).
Synthesis of 77.7
[00693] To a solution of (E)-N1,Nll-(hex-3 -ene- l,6-diyl)dibenzene- 1,2-diamine, 77.6 (300 mg, 1.014 mmol, 1 equiv) in methanol (10 mL) was added cyanogen bromide (425.88 mg, 4.056 mmol, 4.00 equiv). The resulting mixture was stirred at r.t for 2 hours. The mixture was concentrated under reduced pressure. The residue was diluted with water and extracted with dichloromethane. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 160 mg (45.61%) of (E)- l, T-(hex-3-ene-l,6-diyl)bis(lH-benzo[d]imidazol-2-amine) as a brown solid. LC-MS (ES, m/z): 347 (M+H+).
Synthesis of 1-121
[00694] To a stirred mixture of (E)-l,l'-(hex-3-ene-l,6-diyl)bis(lH-benzo[d]imidazol-2- amine), 77.7 (100 mg, 0.289 mmol, 1 equiv) and 4-(5-(5-carboxy-3-methyl-lH-pyrazol-l- yl)pentyl)-l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid, 61.7 (100.58 mg, 0.289 mmol, 1.00 equiv) in l-methylpyrrolidin-2-one (5 mL) was added DIEA (283.8 mg, 2.2 mmol, 10 equiv) and HATU (250.8 mg, 0.66 mmol, 3 equiv). The resulting mixture was irradiated with microwave radiation at 140°C for lh. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column: Sunfire Prep C18 Column 30* 150mm, 5um; mobile phase, Water (0.1%FA) and ACN (69% ACN up to 70% in 8 min); Detector, UV 254/220 nm. This resulted in 12.2 mg (6.41%) of (32E)-5-ethyl -7, 16-dimethyl -
383
5UB5TITUTE SHEET (RULE 26)
Figure imgf000385_0001
MS (ES, m/z): 659 (M+H+); ¾ NMR: (400 MHz, CD30D) d 7.56-7.19 (m, 8H), 6.69 (s, 1H),
5.62-5.37 (m, 2H), 4.79 (t, 2H), 4.52 (q, 2H), 4.25 (t, 2H), 2.86 (t, 2H), 2.54-2.42 (m, 4H), 2.26 (s,
3H), 2.20 (s, 3H), 1.95-1.88 (m, 2H), 1.66-1.57 (m, 2H), 1.46-1.35 (m, 5H).
Example 78: Synthesis of (E)-N-(4-acetyl-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-lH-benzo[d]imidazol-2-yl)-l-ethyl- 3-methyl-lH-pyrazole-5-carboxamide, 1-56
Figure imgf000385_0002
75.7 1-56
[00695] To a stirred solution of l-[2-amino-l-[(2E)-4-(2-amino-lH-l,3-benzodiazol-l-yl)but-
2-en-l-yl]-lH-l,3-benzodiazol-4-yl]ethan-l-one, 75.7 (200 mg, 0.555 mmol, 1 equiv) and 1-ethyl-
3-methyl-lH-pyrazole-5-carboxylic acid (342.20 mg, 2.22 mmol, 4.0 equiv) in DMF (5 mL) were added HATU (843.97 mg, 2.22 mmol, 4.0 equiv) and DIEA (573.74 mg, 4.439 mmol, 8 equiv). The resulting mixture was stirred for 4h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 column, 30* 150mm, 5um; mobile phase, water (0.1% FA) and ACN (45% ACN up to 70% in 8 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 56.8 mg (16.18%) of N-[4-acetyl-l-[(2E)-4-[2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but- 2-en-l-yl]-lH-l,3-benzodiazol-2-yl]-l-ethyl-3-methyl-lH-pyrazole-5-carboxamide as an off- white solid. LC-MS (ES, m/z): 633 (M+H+);’H NMR (400 MHz, DMSO-de) d 12.67 (br s, 2H), 7.88 (d, 1H), 7.71 (d, 1H), 7.50 (d, 1H), 7.40 (d, 1H), 7.31-7.27 (m, 1H), 7.22-7.13 (m, 2H), 6.62 (s, 1H), 6.51 (s, 1H), 5.98-5.87 (m, 2H), 4.88-4.84 (m, 4H), 4.54 (q, 4H), 2.68 (s, 3H), 2.16 (s, 3H), 2.12 (s, 3H), 1.30 (t, 6H).
Example 79: Synthesis of l-(4-(2-(l,3-dimethyl-2-oxo-2,3-dihydro-lH-imidazole-4- carboxamido)-7-methyl-lH-benzo[d]imidazol-l-yl)biityl)-2-(l-ethyl-3-methyl-lH-pyrazole- 5-carboxamido)-lH-benzo [d] imidazole-5-carboxamide, 1-55
Figure imgf000386_0001
Figure imgf000387_0001
Synthesis of 79.1
[00696] A solution of methyl 4-chloro-3-nitrobenzoate (25.00 g, 115.961 mmol, 1.00 equiv) in NH3.H20 (500.00 mL) was stirred in a 1 L round-bottom flask for 12h at 50 degrees C under nitrogen atmosphere. The precipitated solids were collected by filtration and washed with water (3x100 mL). The residue was dissolved in ethyl acetate (300 mL). This resulted in 4-chloro-3- nitrobenzamide (16 g, 68.79%) as a yellow solid.
Synthesis of 79.2
[00697] To a stirred solution of 4-chloro-3-nitrobenzamide, 79.1 (15.00 g, 74.783 mmol, 1.00 equiv) and DIEA (29.00 g, 224.349 mmol, 3.00 equiv) in DMSO (200.00 mL) in a 500 mL 3- necked round-bottom flask, tert-butyl N-(4-aminobutyl)carbamate (21.12 g, 112.175 mmol, 1.50 equiv) was added dropwise in portions at lOOoC under nitrogen atmosphere. After reacting for 1 overnight, the resulting mixture was washed with 3x100 mL of water. The aqueous layer was extracted with EtOAc (3x100 mL). This resulted in tert-butyl N-[4-[(4-carbamoyl-2- nitrophenyl)amino]butyl]carbamate (18 g, 68.30%) as a yellow solid.
Synthesis of 79.3
[00698] Into a 1000-mL 3-necked round-bottom flask, was placed 79.2 (40.00 g, 113.510 mmol, 1.00 equiv), HOAc (400.00 mL) and Zn (74245.94 mg, 1135.103 mmol, 10.00 equiv). The resulting solution was stirred for 2h at room temperature under nitrogen atmosphere. The solids were filtered out. The resulting mixture was concentrated under vacuum. This resulted in 32 g (87.44%) of tert-butyl N-[4-[(2-amino-4-carbamoylphenyl)amino]butyl]carbamate as a black solid.
Synthesis of 79.4
[00699] Into a 500-mL 3-necked round-bottom flask, was placed tert-butyl N-[4-[(2-amino-4- carbamoylphenyl)amino]butyl]carbamate, 79.3 (40.00 g, 124.066 mmol, 1.00 equiv), MeOH (399.99 mL), BrCN (15769.44 mg, 148.879 mmol, 1.20 equiv). The resulting solution was stirred for 3h at room temperature. The resulting mixture was concentrated under vacuum. The crude product was purified by FI ash-Prep -HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN/H20=15% increasing to ACN/H2O=60% within 15 min; Detector, UV: 254nm. This resulted in 32 g (74.24%) of tert-butyl N-[4-(2-amino-5-carbamoyl- lH-l,3-benzodiazol-l-yl)butyl]carbamate as a white solid.
Synthesis of 79.5
[00700] Into a 500-mL 3-necked round-bottom flask, was placed tert-butyl N-[4-(2-amino-5- carbamoyl-lH-l,3-benzodiazol-l-yl)butyl]carbamate, 79.4 (28.00 g, 80.594 mmol, 1.00 equiv), DMF (280.00 mL), DIEA (20.832 g, 161.189 mmol, 2.00 equiv), l-ethyl-3-methyl-lH-pyrazole- 5-carboxylic acid (14.91 g, 96.713 mmol, 1.20 equiv), HATU (45.967 g, 120.891 mmol, 1.50 equiv). The resulting solution was stirred for 1 overnight at room temperature. The resulting solution was diluted with 900 mL of FLO. The solids were collected by filtration. The crude product was re-crystallized from EA/PE in the ratio of 1 : 1. This resulted in 32 g (82.11%) of tert- butyl N-[4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l- yl]butyl]carbamate as an off-white solid.
Synthesis of 79.6 [00701] Into a 500-mL 3-necked round-bottom flask, was placed tert-butyl N-[4-[5-carbamoyl-
2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]butyl]carbamate, 79.5
(32.00 g, 1 equiv), MeOH (200.00 mL), a solution of HC1 (4M) in dioxane (100.00 mL). The resulting solution was stirred for 3h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The crude product was re-crystallized from EA/PE in the ratio of 1 : 1. This resulted in 22 g (86.70%) of l-(4-aminobutyl)-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide as an off-white solid.
Synthesis of 79.7
[00702] Into a 500-mL 3-necked round-bottom flask, was placed l-(4-aminobutyl)-2-(l -ethyl -
3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide, 79.6 (20.00 g, 52.157 mmol, 1.00 equiv), DMF (200.00 mL), K2CO3 (14416.83 mg, 104.314 mmol, 2.00 equiv), 2- fluoro-1 -methyl-3 -nitrobenzene (8900.15 mg, 57.373 mmol, 1.10 equiv). The resulting solution was stirred for 1 overnight at 70 degrees C in an oil bath under nitrogen atmosphere. The resulting solution was diluted with 600 mL of H2O. The resulting solution was extracted with 3x500 mL of ethyl acetate concentrated under vacuum. The crude product was re-crystallized from EA/PE in the ratio of 1 :1. This resulted in 18 g (66.55%) of 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l- [4-[(2-methyl-6-nitrophenyl)amino]butyl]-lH-l,3-benzodiazole-5-carboxamide as a yellow solid. Synthesis of 79.8
[00703] Into a 500-mL 3-necked round-bottom flask, was placed 2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-l-[4-[(2-methyl-6-nitrophenyl)amino]butyl]-lH-l,3-benzodiazole-5- carboxamide, 79.8 (18.00 g, 34.710 mmol, 1.00 equiv), HOAc (180.00 mL), Zn (68110.99 mg, 1041.309 mmol, 30.00 equiv). The resulting solution was stirred for 2h at room temperature under nitrogen atmosphere. The solids were fdtered out. The resulting mixture was concentrated under vacuum. The residue was applied onto a silica gel column with dichloromethane/methanol (20: 1). This resulted in 8.3 g (48.94%) of l-[4-[(2-amino-6-methylphenyl)amino]butyl]-2-(l-ethyl-3- methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide as an off-white solid.
Synthesis of 79.9
[00704] Into a 500-mL 3-necked round-bottom flask, was placed 79.8 (8.3 g, 16.987 mmol, 1.00 equiv), MeOH (85 mL), BrCN (2.16 g, 20.385 mmol, 1.20 equiv). The resulting solution was stirred for 3h at room temperature. The resulting mixture was concentrated under vacuum. The cmde product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN/FhO=15% increasing to ACN/FhO=60% within 15 min; Detector, UV: 254nm yielding 7.3 g as a white solid.
Synthesis of 1-55
[00705] Into a 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed NMP (3.00 mL), l,3-dimethyl-2-oxo-2,3-dihydro-lH-imidazole-4-carboxylic acid (54.6 mg, 0.350 mmol, 1.20 equiv), HATU (332.7 mg, 0.875 mmol, 3.00 equiv), DIEA (123.4 mg, 0.955 mmol, 3.27 equiv), and 79.9 (150 mg, 0.292 mmol, 1.00 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 140 degrees C. The reaction was then quenched by the addition of 15 mL of water. The resulting solution was extracted with 3x15 mL of ethyl acetate. The resulting mixture was washed with 1x15 ml of water. The mixture was dried over anhydrous sodium sulfate. The solids were filtered out. The resulting mixture was concentrated. The crude product (75 mg) was purified by Flash-Prep-HPLC with the following conditions (IntelFl ash- 1 ) : Column, silica gel; mobile phase, CH3CN/water=l/l increasing to CH3CN/water=2/l within 20 min resulting in 55.1 mg (28.95%) of 1-55 as an off-white solid. LC-MS: (ES, m/z): [M+H]+ :652.2; H-NMR: (300 MHz, DMSO-de, />/wi) 12.50-12.82 (d, 2H), 7.96-7.98 (d, 2H), 7.75-7.78 (m, 1H), 7.56-7.59 (d, 1H), 7.30-7.35 (m, 3H), 7.03-7.08 (m, 1H), 6.94-6.96 (d, 1H), 6.61 (s, 1H), 4.53-4.61 (m, 2H), 4.27-4.37 (m,4H), 3.47 (s, 3H) , 3.09 (s, 3H) , 2.57 (s, 3H) , 2.11-2.17 (d, 3H), 1.75-1.91(m, 4H), 1.28-1.33 (t, 3H).
Example 80: Synthesis of tert-butyl (E)-(3-(2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazol-l-yl)but-2-en-l-yl)-N-methyl-lH-benzo[d]imidazole-5- carboxamido)propyl)carbamate, 1-51
Figure imgf000391_0001
1-51 Synthesis of 80.1
[00706] To a solution of methyl l-[(2E)-4-aminobut-2-en-l-yl]-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxylate (2.2 g, 5.549 mmol, 1 equiv) and DMF (30 mL) was added K2CO3 (2.3 g, 16.647 mmol, 3 equiv) and l-fluoro-2 -nitrobenzene (0.94 g, 6.659 mmol, 1.2 equiv). The reaction mixture was stirred for 4 h at 70°C. The mixture was cooled to room temperature and diluted with water. The precipitated solids were collected by filtration. This resulted in 1.5 g (52.23%) of methyl 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-[(2E)-4-[(2- nitrophenyl)amino]but-2-en-l-yl]-lH-l,3-benzodiazole-5-carboxylate as a yellow solid. LC-MS (ES, m/z): 518 (M+H+).
Synthesis of 80.2
[00707] To a solution of methyl 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-[(2E)-4-[(2- nitrophenyl)amino]but-2-en-l-yl]-lH-l,3-benzodiazole-5-carboxylate, 80.1 (1.5 g, 2.898 mmol, 1 equiv) in AcOH (20 mL) was added Zn (941.85 mg, 14.49 mmol, 5 equiv). The resulting solution was stirred overnight at room temperature. The solids were filtered out. The filtrate was concentrated under reduced pressure. This resulted in 1.2 g (84.92%) of methyl l-[(2E)-4-[(2- aminophenyl)amino]but-2-en-l-yl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxylate as a brown solid. LC-MS (ES, m/z): 488 (M+H+).
Synthesis of 80.3
[00708] To a solution of methyl l-[(2E)-4-[(2-aminophenyl)amino]but-2-en-l-yl]-2-(l-ethyl- 3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxylate, 80.2 (1 g, 2.051 mmol, 1 equiv) in MeOH (20 mL) was added BrCN (652.2 mg, 6.153 mmol, 3 equiv). The resulting solution was stirred overnight at room temperature. The resulting mixture was concentrated under vacuum. This resulted in 1.2 g crude of (E)-methyl l-(4-(2-amino-lH-benzo[d]imidazol-l-yl)but- 2-enyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate as a brown oil. LC-MS (ES, m/z): 513 (M+H+).
Synthesis of 80.4
[00709] To a solution of (E)-methyl l-(4-(2-amino-lH-benzo[d]imidazol-l-yl)but-2-enyl)-2- (l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate, 80.3 (1 g, 1.951 mmol, 1 equiv) and l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (360.55 mg, 2.341 mmol, 1.2 equiv) in DMF (15 mL) was added HATU (1.11 g, 2.927 mmol, 1.5 equiv) and DIEA (1.26 g, 9.775 mmol, 5 equiv). The resulting solution was stirred overnight at room temperature. The mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2S04 and concentrated under reduced pressure. The residue was applied on a silica gel column with PE/EtOAc (1/1). This resulted in 800 mg (63.21%) of methyl 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-[(2E)-4-[2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3-benzodiazole-5-carboxylate as a light yellow solid. LC-MS (ES, m/z): 649 (M+H+).
Synthesis of 80.5
[00710] To a solution of methyl 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-[(2E)-4-[2-(l- ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3- benzodiazole-5-carboxylate, 80.5 (800 mg, 1.233 mmol, 1 equiv) in MeOH (10 mL), THF (10 mL) and H2O (4 mL) was added LiOH (147.67 mg, 6.166 mmol, 5.00 equiv). The resulting mixture was stirred at 70°C overnight. The mixture was concentrated under reduced pressure. The pH value of the solution was adjusted to 3 with IN HC1. The solid was collected by filtration. This resulted in 650 mg (83.05%) of 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-[(2E)-4-[2-(l-ethyl-3- methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3-benzodiazole-5- carboxylic acid as an off-white solid. LC-MS (ES, m/z): 635 (M+H+).
Synthesis of 1-51
[00711] To a solution of 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-[(2E)-4-[2-(l-ethyl-3- methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3-benzodiazole-5- carboxylic acid, 80.5 (63.4 mg, 0.1 mmol, 1 equiv) and tert-butyl N-[3-
(methylamino)propyl]carbamate (22.56 mg, 0.12 mmol, 1.2 equiv) in DMF (4 mL) was added HATU (57 mg, 0.15 mmol, 1.5 equiv) and DIEA (64.5 mg, 0.5 mmol, 5 equiv). The resulting solution was stirred overnight at room temperature. The mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2S04 and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 OBD Column 30*150mm, 5um; mobile phase, Water (0.1% FA) and ACN (42% ACN up to 62% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 36.5 mg (45.4%) of tert-butyl N-(3-[l-[2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-[(2E)- 4-[2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3- benzodiazol-5-yl]-N-methylformamido]propyl)carbamate as a white solid. LC-MS (ES, m/z): 805 (M+H+); 1HNMR (400 MHz, DMSO-c/e) d 12.65 (br s, 2H), 7.51-7.37 (m, 4H), 7.21-7.12 (m, 3H), 6.83-6.57 (m, 3H), 6.02-5.87 (m, 2H), 4.86-4.80 (m, 4H), 4.57-4.51 (m, 4H), 3.29-3.11 (m, 2H), 3.09-2.72 (m, 5H), 2.13 (s, 6H), 1.76-1.62 (m, 2H), 1.51-1.11 (m, 15H).
Example 81: Synthesis of (E)-N-(3-aminopropyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazol-l-yl)but-2-en-l-yl)-N-methyl-lH-benzo[d]imidazole-5-carboxamide, 1-50
Figure imgf000394_0001
1-51 1-50
[00712] To a solution of tert-butyl N-(3-[l-[2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l- [(2E)-4-[2-(l -ethyl-3 -methyl- lH-pyrazole-5-amido)- 1H- 1 ,3 -benzodiazol- 1 -yl]but-2-en- 1 -yl]- lH-l,3-benzodiazol-5-yl]-N-methylformamido]propyl)carbamate, 1-51, (100 mg, 0.124 mmol, 1 equiv) and DCM (10 mL) was added TFA (2 mL). The resulting mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 OBD Column 30* 150mm, 5um; mobile phase, Water (0.1% FA) and ACN (11% ACN up to 36% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 20.6 mg (23.56%) of (E)-N-(3-aminopropyl)-2-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)- lH-benzo[d]imidazol-l-yl)but-2-enyl)-N-methyl-lH-benzo[d]imidazole-5-carboxamide as a brown oil. LC-MS (ES, m/z): 705 (M+H+); ^MR ^OO MHz, DMSO-i/e) ό 8.40 (br s, 1H), 7.51- 7.38 (m, 4H), 7.21-7.12 (m, 3H), 6.57 (s, 2H), 6.02-5.87 (m, 2H), 4.86-4.80 (m, 4H), 4.57-4.51 (m, 4H), 3.29-3.11 (m, 2H), 3.09-2.72 (m, 5H), 2.13 (s, 6H), 1.76-1.62 (m, 2H), 1.28 (t, 6H).
Example 82: Synthesis of (E)-N-(3-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)oxy)acetamido)propyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-l-(4-(2-(l- ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-N- methyl-lH-benzo[d]imidazole-5-carboxamide, 1-49
Figure imgf000395_0001
[00713] To a solution of 2-[[2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-isoindol-4- yl]oxy]acetic acid (47.14 mg, 0.142 mmol, 1.00 equiv) and (E)-N-(3-aminopropyl)-2-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)- lH-benzo[d]imidazol-l-yl)but-2-enyl)-N-methyl-lH-benzo[d]imidazole-5-carboxamide, 1-50, (100 mg, 0.142 mmol, 1 equiv) DMF (6 was added HATU (107.89 mg, 0.284 mmol, 2 equiv)
Figure imgf000395_0002
and DIEA (183.37 mg, 1.419 mmol, 10.00 equiv). The resulting mixture was stirred at RT for 2 h. The mixture was concentrated under reduced pressure. The crude product was purified by Prep- HPLC with the following conditions: Column, Sunfire Prep C18 OBD Column 30*150mm, 5um; mobile phase, Water (0.1% FA) and ACN (32% ACN up to 48% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 16 mg (11%) of N-(5-[[3-(2-[[2-(2,6-dioxopiperidin-3-yl)-l,3- dioxo-2,3-dihydro-lH-isoindol-4-yl]oxy]acetamido)propyl](methyl)carbamoyl]-l-[(2E)-4-[2-(l- ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3- benzodiazol-2-yl)-l-ethyl-3-methyl-lH-pyrazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 1019 (M+H+); LHNMR (400 MHz, DMSO-<¾) d 12.67 (br s, 2H), 11.13 (s, 1H), 8.19-7.61 (m, 2H), 7.50-7.11 (m, 9H), 6.55 (s, 2H), 5.95-5.81 (m, 2H), 5.10 (dd, 1H), 4.82-4.51 (m, 10H), 3.32-2.97 (m, 4H), 2.96-2.81 (m, 4H), 2.62-2.51 (m, 2H), 2.12 (s, 6H), 2.11-1.98 (m, 1H), 1.83- 1.72 (m, 2H), 1.28 (t, 6H).
Example 83: Synthesis of l-(4-(2-(5,6-dimethoxybenzo[b]thiophene-2-carboxamido)-7- methyl-lH-benzo[d]imidazol-l-yl)butyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)- lH-benzo[d]imidazole-5-carboxamide, 1-41
Figure imgf000396_0001
[00714] Into a 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed NMP (2.00 mL), 79.9 (150.00 mg, 2.924 mmol, 1.20 equiv), 5,6-dimethoxy-l- benzothiophene-2-carboxylic acid (57.80 mg, 2.437 mmol, 1.00 equiv), HATU (184.8 mg, 4.873 mmol, 2.00 equiv), DIEA (62.74 mg, 4.873 mmol, 2.00 equiv). The resulting mixture was irradiated with microwave radiation for 1 h at 140 degrees C under nitrogen atmosphere. The resulting mixture was washed with 1x20 mL of water. The resulting solution was extracted with 3x20 mL of ethyl acetate and the organic layers combined. The crude product was purified by Flash-Prep-HPLC with the following condition: Column: XBridge Prep OBD Cl 8 Column, 30x 150 mm 5 um; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 40 B to 50 B in 7 min; 254; 220 nm; RT1 : 6.57 min resulting in 1-41 (18.6 mg) as a yellow solid. LC-MS: (ES, m/z): [M+H]+ 734.5; 1H-NMR:(300 MHz, DMSO, ppm): 512.69-12.77 (brs, 2H) , 57.90-7.98 (m, 3H), 57.75-7.78 (d, 1H), 57.58-7.61 (d, 1H), 57.32- 7.40(m, 4H), 57.07-7.12 (m, 1H), 56.98-7.00(d, 1H), 56.64 (s, 1H), 54.57-4.59(d, 2H), 54.45(s, 2H), 54.33(s, 2H), 53.80-3 83(d, 6H), 52.61 (d, 3H), 52.03 (s, 3H), 51.89-1.99 (m, 4H), 1.28-1.33 (m, 3H).
Example 84: Synthesis of l-(4-(2-(3,4-dimethoxybenzamido)-7-methyl-lH- benzo[d]imidazol-l-yl)butyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo [d] imidazole-5-carboxamide, 1-40
Figure imgf000397_0001
79.9 1-40
[00715] Into a 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed NMP (2 mL), 79.9 (150.00 mg, 0.2924 mmol, 1.00 equiv), 3,4-dimethoxybenzoic acid (44.30 mg, 0.2924 mmol, 1.00 equiv), HATU (184.8 mg, 0.5848 mmol, 2.00 equiv), and DIEA (62.74 mg, 0.5848 mmol, 2.00 equiv). The resulting mixture was irradiated with microwave radiation for lh at 140 degrees C. The resulting mixture was washed with 1x20 mL of water. The resulting solution was extracted with 3x20 mL of ethyl acetate and the organic layers combined. The crude product was purified by Prep-HPLC with the following condition Column: Kinetex EVO C18 Column, 30*150 mm, 5 um; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 31 B to 41 B in 10 min; 254; 220 nm; RT1 : 10.02 min. This resulted in l-[4-[2-(3,4-dimethoxybenzamido)-7-methyl-lH- l,3-benzodiazol-l-yl]butyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5- carboxamide (13.8 mg) as a white solid. LC-MS:(ES, m/z): [M+H]+ 678.6; 1H-NMR:(300 MHz, DMSO ,ppm): 512.68-12.79 (m, 2H), 57.95-7.98 (d, 2H), 57.72-7.77 (m, 3H), 57.52-7.58 (d, 1H), 57.37-7.39 (d, 1H), 57.29 (s, 1H), 57.05-7.10 (m, lH), 56.96-6.99(d, 1H), 56.88-6.90(d, 1H), 56.60 (s, 1H), 54.53-4.58(m, 2H), 54.47 (s, 2H), 54.3 l(s, 2H), 53.78(s, 3H), 53.71(s, 3H), 52.61(s, 3H), 52.07 (s, 3H), 51.91(s, 4H), 51.23-1.31 (m, 3H).
Example 85: Synthesis of N-(l-[4-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- lH-l,3-benzodiazol-l-yl]butyl]-7-methyl-lH-l,3-benzodiazol-2-yl)-4,5,6,7-tetrahydro-2,l- benzoxazole-3-carboxamide, 1-39
Figure imgf000398_0001
[00716] Into a 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed NMP (2 mL) ,l-[4-(2-amino-7-methyl-lH-l,3-benzodiazol-l-yl)butyl]-2-(l-ethyl-3- methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide, 79.9 (150.00 mg, 0.3509 mmol, 1.20 equiv),4,5,6,7-tetrahydro-2,l-benzoxazole-3-carboxylic acid (40.60 mg, 0.2924 mmol, 1.00 equiv), HATU (184.8 mg, 0.5848 mmol, 2.00 equiv), and DIEA (62.74 mg, 0.5848 mmol, 2.00 equiv).The resulting mixture was irradiated with microwave radiation for 1 h at 140 degrees C. The resulting mixture was washed with 1 x 20 mL of water. The resulting solution was extracted with 3 x 20 mL of ethyl acetate and the organic layers combined. The crude product was purified by Prep-HPLC with the following condition Column: Kinetex EVO Cl 8 Column, 30* 150 mm, 5 um; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 30 B to 50 B in 7 min; 254; 220 nm; RT1: 6.82 min. This resulted in N-(l-[4- [5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]butyl]-7-
397
5UB5TITUTE SHEET (RULE 26) methyl-lH-l,3-benzodiazol-2-yl)-4,5,6,7-tetrahydro-2, l-benzoxazole-3 -carboxamide (84.8 mg) as a white solid. LC-MS: (ES, in :): [M+H]+ 663.5; 1H-NMR: (300 MHz, DMSO ,ppm): 512.76- 12.81 (m, 2H), 57.95-7.97 (d, 2H), 57.74-7.77 (m, 1H), 57.57-7.59 (d, 1H), 57.41-7.43 (d, 1H), 57.33 (s, 1H), 57.10-7.15 (m, 1H), 57.05-7.05 (d, 1H), 56.54-6.56(d, 1H), 54.53-4.60(m, 2H), 54.39(s, 2H), 54.27(s, 2H), 52.73-2.79(m, 2H), 52.60-2.63 (d, 5H), 52.08 (s, 3H), 51.88 (s, 4H), 51.49-1.60 (m, 4H), 51.25-1.33 (t, 3H).
Example 86: Synthesis of 2-(l-(5-(5-((5-carbamoyl-l-methyl-lH-benzo[d]imidazol-2- yl)carbamoyl)-l-ethyl-3-methyl-lH-pyrazol-4-yl)pentyl)-3-methyl-lH-pyrazole-5- carboxamido)-N-(3-(2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)oxy)acetamido)propyl)-N,l-dimethyl-lH-benzo[d]imidazole-5-carboxamide, 1-37
Figure imgf000399_0001
Figure imgf000400_0001
Synthesis of 86.1
[00717] To a solution of methyl 4-fluoro-3-nitrobenzoate (4.00 g, 20 mmol, 1.0 equiv) in DMF (20 mL) was added DIEA (7.78 g, 60 mmol, 3 equiv) and Me E/THF (2 mol/L, 12 mL, 24 mmol, 1.2 equiv). The resulting mixture was stirred at RT for 4 h. The mixture was diluted with water, extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SC>4 and concentrated under reduced pressure. The residue was applied on a silica gel column with PE/EtOAc (1/1). This resulted in 3 g (71%) of methyl 4-(methylamino)-3-nitrobenzoate as a yellow solid. LC-MS (ES, m/z): 211 (M+H+).
Synthesis of 86.2
[00718] To a solution of methyl 4-(methylamino)-3-nitrobenzoate, 86.1 (2 g, 9.5 mmol, 1.0 equiv) in MeOH (50 mL) and DCM (20 mL) was added Pd/C (400 mg). To the above ¾ (g) was introduced in. The mixture was stirred 4 h at room temperature. The solid was filtered. The filtrate was concentrated under reduced pressure. This resulted in 1.3 g (75.8%) of methyl 3-amino-4- (methylamino)benzoate as an off-white solid. LC-MS (ES, m/z): 181 (M+H+);
Synthesis of 86.3
[00719] To a solution of methyl 3-amino-4-(methylamino)benzoate, 86.1 (1.3 g, 7.2 mmol, 1.0 equiv) in MeOH (50 mL) was added BrCN (2.3 g, 21.6 mmol, 3.0 equiv). The resulting mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. This resulted in 1.2 g (81%) of methyl 2-amino-l-methyl-lH-l,3-benzodiazole-5-carboxylate as an off-white solid. LC-MS (ES, m/z): 206 (M+H+).
Synthesis of 86.4
[00720] To a solution of methyl 2-amino-l-methyl-lH-l,3-benzodiazole-5-carboxylate, 86.3 1.2 g, 5.85 mmol, 1.0 equiv) in MeOH (30 mL) and ¾0 (6 mL) was added LiOH (702 mg, 29.25 mmol, 5.0 equiv). The resulting mixture was stirred at 70°C overnight. The mixture was concentrated under reduced pressure. The pH value of the solution was adjusted to 3 with IN HC1. The solid was collected by filtration. This resulted in 1.0 g (90%) of 2-amino- 1 -methyl- 1H- 1,3- benzodiazole-5-carboxylic acid as an off-white solid. LC-MS (ES, m/z): 192 (M+H+). Synthesis of 86.5
[00721] To a solution of 2-amino-l-methyl-lH-l,3-benzodiazole-5-carboxylic acid, 86.4 (810 mg, 4.24 mmol, 1.0 equiv) and DIEA (2.73 g, 21.2 mmol, 5.0 equiv) in DMF (20 mL) was added tert-butyl 3-(methylamino)propyl carbamate (800 mg, 4.24 mmol, 1.0 equiv) and HATU (2.43 g, 6.36 mmol, 1.5 equiv). The resulting mixture was stirred at RT overnight. The mixture was diluted with water, extracted with CHCh/z-PrOH (3/1). The combined organic layers were washed with brine, dried over Na2SC>4 and concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (41% ACN up to 51% in 10 min); Detector, UV 254/220 nm. This resulted in 460 mg (30%) of tert-butyl -N-[3-[l-(2-amino-l-methyl-lH-l,3-benzodiazole-5-yl)-N- methylformamido]propyl)carbamate as a brown oil. LC-MS (ES, m/z): 362 (M+H+).
Synthesis of 86.6
[00722] To a solution of l-(5-(5-(ethoxycarbonyl)-l-ethyl-3-methyl-lH-pyrazol-4-yl)pentyl)- 3-methyl-lH-pyrazole-5-carboxylic acid, 86.5 (376 mg, 1.0 mmol, 1.0 equiv) and DIEA (645 mg, 5.0 mmol, 5.0 equiv) in NMP (8 mL) was added methyl tert-butyl -N-[3-[l-(2-amino-l-methyl- lH-l,3-benzodiazole-5-yl)-N-methylformamido]propyl)carbamate (360 mg, 1.0 mmol, 1.0 equiv) and HATU (570 mg, 1.5 mmol, 1.5 equiv). The final reaction mixture was irradiated with microwave radiation at 140°C for 1 hour. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (35% ACN up to 45% in 10 min); Detector, UV 254/220 nm. This resulted in 580 mg (81%) of ethyl 4-(5-(5-((5- ((3-((tert-butoxycarbonyl)amino)propyl)(methyl)carbamoyl)-l-methyl-lH-l,3-benzodiazol-2- yl)carbamoyl)-3-methyl-lH-pyrazol-l-yl)pentyl)-l-ethyl-3-methyl-lH-pyrazole-5-carboxylate as a brown oil. LC-MS (ES, m/z): 720 (M+H+).
Synthesis of 86.7
[00723] To a solution of ethyl 4-(5-(5-((5-((3-((tert- butoxycarbonyl)amino)propyl)(methyl)carbamoyl)-l-methyl-lH-l,3-benzodiazol-2- yl)carbamoyl)-3-methyl-lH-pyrazol-l-yl)pentyl)-l-ethyl-3-methyl-lH-pyrazole-5-carboxylate, 86.6 ( 580 mg, 0.81 mmol, 1.0 equiv) in MeOH (10 mL) and H2O (2 mL) was added LiOH (96 mg, 4.03 mmol, 5.0 equiv). The resulting mixture was stirred at 70°C overnight. The mixture was concentrated under reduced pressure. The pH value of the solution was adjusted to 3 with IN HC1. The solid was collected by filtration. This resulted in 300 mg (54%) of 4-(5-(5-((5-((3-((tert- butoxycarbonyl)amino)propyl)(methyl)carbamoyl)-l-methyl-lH-l,3-benzodiazol-2- yl)carbamoyl)-3-methyl-lH-pyrazol-l-yl)pentyl)-l-ethyl-3-methyl-lH-pyrazole-5-carboxylate carboxylic acid as a brown oil. LC-MS (ES, m/z): 692 (M+H+).
Synthesis of 86.8
[00724] To a solution of 4-(5-(5-((5-((3-((tert- butoxycarbonyl)amino)propyl)(methyl)carbamoyl)-l-methyl-lH-l,3-benzodiazol-2- yl)carbamoyl)-3-methyl-lH-pyrazol-l-yl)pentyl)-l-ethyl-3-methyl-lH-pyrazole-5-carboxylate carboxylic acid, 86.7 (300 mg, 0.43 mmol, 1.0 equiv) and DIEA (280 mg, 2.17 mmol, 5.0 equiv) in NMP (6 mL) was added 2-amino-l-methyl-lH-l,3-benzodiazole-5-carboxamide (99 mg, 0.52 mmol, 1.2 equiv) and HATU (247 mg, 0.65 mmol, 1.5 equiv). The final reaction mixture was irradiated with microwave radiation at 140°C for 1 hour. The mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by Flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (30% ACN up to 40% in 10 min); Detector, UV 254/220 nm. This resulted in 193 mg (52%) of tert- butyl-N-3-(l-(2-l-(5-(5-(5-carbamoyl-l-methyl-lH-l,3-benzodiazol-2-ylcarbamoyl)-l-ethyl-3- methyl-lH-pyrazol-4-yl)pentyl)-3-methyl-lH-pyrazole-5-amido)-l-methyl-lH-l,3- benzodiazole-5-yl)-N-methylformamido)propylcarbamate as a brown oil. LC-MS (ES, m/z): 865 (M+H+).
Synthesis of 86.9
[00725] To a solution of tert-butyl-N-3-(l-(2-l-(5-(5-(5-carbamoyl-l-methyl-lH-l,3- benzodiazol-2-ylcarbamoyl)-l-ethyl-3-methyl-lH-pyrazol-4-yl)pentyl)-3-methyl-lH-pyrazole-5- amido)-l-methyl-lH-l,3-benzodiazole-5-yl)-N-methylformamido)propylcarbamate, 86.8 in DCM (10 mL) was added TFA (3 mL). The resulting mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. This resulted in 140 mg (82%) of tert-butyl N- 3-(aminopropyl-(2-(l-(5-(5-(5-carbamoyl-l-methyl-lH-l,3-benzodiazol-2-ylcarbamoyl)-l-ethyl- 3-methyl-lH-pyrazol-4-yl)pentyl)-3-methyl-lH-pyrazole-5-amino)-N,l-dimethyl-lH-l,3- benzodiazole-5-carboxmide as a brown oil. LC-MS (ES, m/z): 764 (M+H+). Synthesis of 1-37
[00726] To a solution of 2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-isoindolin- 4-yl)oxy)acetic acid (80 mg, 0.24 mmol, 1.3 equiv) and DIEA (118 mg, 0.92 mmol, 5.0 equiv) in DMF (8 mL) was added N-3-(aminopropyl-(2-(l-(5-(5-(5-carbamoyl-l-methyl-lH-l,3- benzodiazol-2-ylcarbamoyl)-l-ethyl-3-methyl-lH-pyrazol-4-yl)pentyl)-3-methyl-lH-pyrazole-5- amino)-N,l-dimethyl-lH-l,3-benzodiazole-5-carboxmide, 86.9 (140 mg, 0.18 mmol, 1.0 equiv) and HATU (105 mg, 0.28 mmol, 1.5 equiv). The resulting mixture was stirred at RT for 2 h. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Prep Phenyl OBD Column 19* 150mm, 5um, 13nm; mobile phase, Water (0.1% FA) and MeOH (60% MeOHup to 75% in 7 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 24.7 mg (13%) of 2-(4-(5-(5-((5-((3-(2-((2-(2,6-dioxopiperidin-3- yl)-l,3-dioxo-2,3-dihydro-lH-isoindol-4-yl)oxy)acetamino)propyl)(methyl)-carbamoyl)-l- methyl-lH-l,3-benzodiazol-2-yl)carbamoyl)-3-methyl-lH-pyrazol-l-yl)pentyl)-l-ethyl-3- methyl-lH-pyrazole-5-amino)-l-methyl-lH-l,3-benzodiazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 540 (M/2+H+); 1H NMR (400 MHz, DMSO-t¾) d 12.78 (s, 1H), 12.68 (br s, 1H), 11.13 (s, 1H), 8.14-7.54 (m, 5H), 7.52-7.16 (m, 7H), 6.65 (s, 1H), 5.10 (q, 1H), 4.89-4.50 (m, 6H), 3.71-3.35 (m, 8H), 3.08-2.84 (m, 6H), 2.82-2.73 (m, 2H), 2.70-2.55 (m, 2H), 2.19-2.00 (m, 7H), 1.86-1.73 (m, 4H), 1.62-1.51 (m, 2H), 1.41-1.27 (m, 5H).
Example 87: Synthesis of l-(2-(l-(2-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)acetyl)piperidin-4-yl)ethyl)-2-(l-ethyl-3-methyl- lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-35
Figure imgf000404_0001
Figure imgf000405_0001
Synthesis of 87.1 [00727] To a solution of 4-chloro-3-nitrobenzamide (600 mg, 2.991 mmol, 1.00 equiv) in dimethyl formamide (20 mL) was added potassium carbonate (1.24 g, 8.974 mmol, 3.00 equiv) and tert-butyl 4-(2-aminoethyl)piperidine-l-carboxylate (683.03 mg, 2.991 mmol, 1.00 equiv). The resulting mixture was stirred at 80°C overnight. The mixture was cooled to r.t, diluted with water. The solids were collected by filtration. This resulted in 1.05 g (89.44%) of tert-butyl 4-[2- [(4-carbamoyl-2-nitrophenyl)amino]ethyl]piperidine-l-carboxylate as a yellow solid. LC-MS (ES, m/z): 393 (M+H+);
Synthesis of 87.2
[00728] To a solution of tert-butyl 4-[2-[(4-carbamoyl-2-nitrophenyl)amino]ethyl]piperidine-l- carboxylate, 87.1 (1.05 g, 2.675 mmol, 1.00 equiv) in methanol (40 mL) was added palladium on carbon (500 mg). To the above hydrogen (g) was introduced in. The mixture was stirred for 2h at room temperature. The solid was filtered. The filtrate was concentrated under reduced pressure. This resulted in 900 mg (92.80%) of tert-butyl 4-[2-[(2-amino-4- carbamoylphenyl)amino]ethyl]piperidine-l-carboxylate as a black solid. LC-MS (ES, m/z): 363 (M+H+);
Synthesis of 87.3
[00729] To a stirred solution of tert-butyl 4-[2-[(2-amino-4- carbamoylphenyl)amino]ethyl]piperidine-l-carboxylate, 87.2 (900 mg, 2.483 mmol, 1.00 equiv) in MeOH (15 mL) was added cyanogen bromide (3.58 g, 33.76 mmol, 2 equiv). The resulting mixture was stirred at RT for 2 h. The mixture was concentrated under reduced pressure. This resulted in 1.08 g (crude) of tert-butyl 4-[2-(2-amino-5-carbamoyl-lH-l,3-benzodiazol-l- yl)ethyl]piperidine-l-carboxylate as a brown solid. LC-MS (ES, m/z): 386 (M-H+);
Synthesis of 87.4
[00730] To a stirred solution of tert-butyl 4-[2-(2-amino-5-carbamoyl-lH-l,3-benzodiazol-l- yl)ethyl]piperidine-l-carboxylate, 87.3 (50.00 mg, 0.129 mmol, 1.00 equiv) in dimethyl formamide (5 mL) was added l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (19.89 mg, 0.129 mmol, 1.00 equiv), DIEA (83.39 mg, 0.645 mmol, 5.00 equiv) and HATU (58.88 mg, 0.155 mmol, 1.20 equiv). The resulting mixture was stirred at RT overnight. The reaction mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep C18 column, 30* 150mm, 5um; mobile phase, Water (0.1% FA) and ACN (37% ACN up to 60% in 7 min); Detector, UV 254/220 nm. This resulted in 13 mg (19.24%) of tert-butyl 4-[2-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH- l,3-benzodiazol-l-yl]ethyl]piperidine-l-carboxylate as a white solid. LC-MS (ES, m/z): 524 (MAID); ¾ MR (400 MHz, DMSO-i&) d 12.84 (s, 1H), 8.06-7.95 (m, 2H), 7.80 (dd, 1H), 7.55 (d, 1H), 7.34 (s, 1H), 6.63 (s, 1H), 4.63 (q, 2H), 4.25 (t, 2H), 3.96-3.82 (m, 2H), 2.78-2.55 (m, 2H), 2.19 (s, 3H), 1.82-1.67 (m, 4H), 1.56-1.32 (m, 13H), 1.17-1.02 (m, 2H).
Synthesis of 87.5
[00731] To a stirred solution of tert-butyl 4-[2-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole- 5-amido)-lH-l,3-benzodiazol-l-yl]ethyl]piperidine-l-carboxylate, 87.6 (50 mg, 0.095 mmol, 1.00 equiv) in dichloromethane (5 mL) was added trifluoroacetic acid (1 mL). The resulting mixture was stirred at RT for 2 h. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep C18 column, 30*150, 5um; mobile phase, Water (0.1% FA) and ACN (5% ACN up to 30% in 5 min); Detector, UV 254/220 nm. This resulted in 26.6 mg (59.33%) of 2-(l -ethyl-3 -methyl- lH-pyrazole- 5-amido)-l-[2-(piperidin-4-yl)ethyl]-lH-l,3-benzodiazole-5-carboxamide as a light pink solid. LC-MS (ES, m/z): 424 (M+H+); ¾ NMR (400 MHz, DMSO -d6) d 8.03-7.95 (m, 2H), 7.81 (d, 1H), 7.55 (d, 1H), 7.33 (s, 1H), 6.62 (s, 1H), 4.62 (q, 2H), 4.25 (t, 2H), 3.21-3.12 (m, 2H), 2.71- 2.62 (m, 2H), 2.19 (s, 3H), 1.97-1.89 (m, 2H), 1.73-1.68 (m, 2H), 1.61-1.47 (m, 1H) , 1.41-1.25 (m, 5H).
Synthesis of 87.6
[00732] To a stirred solution of 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-[2-(piperidin-4- yl)ethyl]-lH-l,3-benzodiazole-5-carboxamide, 87.5 (300 mg, 0.708 mmol, 1.00 equiv) in dimethyl formamide (10 mL) was added DIEA (1830.98 mg, 14.167 mmol, 20.00 equiv), 2-[(4- carbamoyl-2-nitrophenyl)amino]acetic acid (203.31 mg, 0.850 mmol, 1.20 equiv) and HATU (404.00 mg, 1.063 mmol, 1.50 equiv). The resulting mixture was stirred at RT for 2 h. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (35% ACN up to 50% in 7 min); Detector, UV 254/220 nm. This resulted in 400 mg (87.59%) of l-[2-(l-[2-[(4- carbamoyl-2-nitrophenyl)amino]acetyl]piperidin-4-yl)ethyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazole-5-carboxamide as a yellow solid. LC-MS (ES, m/z): 643 (M-H+); Synthesis of 87.7
[00733] To a solution of (5-carbamoyl-2-[[2-(4-[2-[5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]ethyl]piperidin-l-yl)-2- oxoethyl]amino]phenyl)azinic acid, 87.6 (200 mg, 0.309 mmol, 1.00 equiv) in methanol (20 mL) was added palladium on carbon (50 mg). To the above hydrogen (g) was introduced in. The mixture was stirred for 2h at room temperature. The solid was filtered. The filtrate was concentrated under reduced pressure. This resulted in 150 mg (78.90%) of l-[2-(l-[2-[(2-amino- 4-carbamoylphenyl)amino]acetyl]piperidin-4-yl)ethyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazole-5-carboxamide as a light yellow solid. LC-MS (ES, m/z): 615 (M+H+);
Synthesis of 87.8
[00734] To a stirred solution of l-[2-(l-[2-[(2-amino-4- carbamoylphenyl)amino]acetyl]piperidin-4-yl)ethyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- lH-l,3-benzodiazole-5-carboxamide, 87.7 (150.00 mg, 0.244 mmol, 1.00 equiv) in methanol (10 mL) was added cyanogen bromide (77.54 mg, 0.732 mmol, 3.00 equiv). The resulting mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (35% ACN up to 46% in 7 min); Detector, UV 254/220 nm. This resulted in 115 mg (73.67%) of 2-amino-l-[2-(4-[2-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-l-yl]ethyl]piperidin-l-yl)-2-oxoethyl]-lH-l,3-benzodiazole-5- carboxamide as a brown yellow solid. LC-MS (ES, m/z): 638 (M-H+);
Synthesis of 1-35
[00735] To a stirred solution of 2-amino-l-[2-(4-[2-[5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]ethyl]piperidin-l-yl)-2-oxoethyl]-lH-l,3- benzodiazole-5-carboxamide, 87.8 (115 mg, 0.18 mmol, 1.00 equiv) in dimethyl formamide (5 mL) was added l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (41.57 mg, 0.270 mmol, 1.5 equiv), DIEA (116.17 mg, 0.899 mmol, 5.00 equiv) and HATU (102.53 mg, 0.270 mmol, 1.50 equiv). The resulting mixture was irradiated with microwave radiation at 120°C for lh. The reaction mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep C18 column, 30*150mm, 5um; mobile phase, Water (0.1% FA) and ACN (18% ACN up to 46% in 7 min); Detector, UV 254/220 nm. This resulted in 41.5 mg (28.09%) of l-[2-(4-[2-[5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]ethyl]piperidin-l-yl)-2-oxoethyl]-2-(l-ethyl-3- methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide; formic acid as a white solid. LC-MS (ES, m/z): 776 (M+H+); Tl NMR (400 MHz, DMSO-tfc) d 12.82 (br s, 2H), 8.03-7.96 (m, 4H), 7.83 (d, 1H), 7.76 (d, 1H), 7.58 (d, 1H), 7.45-7.32 (m, 3H), 6.66 (s, 1H), 6.59 (s, 1H), 5.20- 5.10 (m, 2H), 4.66-4.55 (m, 4H), 4.33-4.24 (m, 3H), 4.06-4.00 (m, 1H), 3.15-3.09 (m, 1H), 2.67- 2.56 (m, 1H), 2.17 (s, 3H), 2.10 (s, 3H), 2.00-1.91 (m, 2H), 1.78-1.64 (m, 3H), 1.38-1.29 (m, 6H), 1.27-1.03 (m, 2H).
Example 88: Synthesis of l-(4-(2-(2,5-dimethylthiophene-3-carboxamido)-7-methyl-lH- benzo[d]imidazol-l-yl)butyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo [d] imidazole-5-carboxamide, 1-34
Figure imgf000410_0001
[00736] Into a 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed NMP (2.00 mL), 79.9 (150 mg, 0.292 mmol, 1.00 equiv), 2,5-dimethylthiophene-3- carboxylic acid (273.71 mg, 1.752 mmol, 6.00 equiv), HATU (666.28 mg, 1.752 mmol, 6.00 equiv), DIEA (226.47 mg, 1.752 mmol, 6.00 equiv). The resulting mixture was stirred for 1 h at 140°C under microwave monitor. The resulting mixture was washed with 1x20 mL of water. The resulting solution was extracted with 3x20 mL of ethyl acetate and the organic layers combined. The crude product was purified by Prep-HPLC with the following condition (Column: XBridge Prep OBD C18 Column, 30x 150mm 5um; Mobile Phase A: water, Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 10% B to 40% B in 20 min; 254; 220 nm; RT1 :6.15 min). This resulted in l-[4-[2-(2, 5-dimethylthiophene-3-amido)-7-methyl-lH-l,3-benzodiazol-l-yl]butyl]-2-(l- ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide (45.3mg, 23.80%) as an off-white solid. LC-MS: (ES, m/z): [M+H]+: 652.5, [M-H] : 650.5; 'H-NMR:(300 MHz, DMSO-d 6, ppm) 612.64-12.80 (d, 2H), 67.95-7.97 (d, 2H), 67.75-7.77 (m, 1H), 67.56-7.59 (d, 1H), 57.32-7.38 (m, 2H), 57.18 (s, 1H), 57.04-7.07 (m, 1H), 56.95-6.98 (d, 1H), 56.60(s, 1H), 54.54- 4.61 (m, 2H), 54.28-4.54(m, 4H), 52.60-2.73(m, 6H), 52.26 (s, 3H), 52.07-2.11 (d, 3H), 51.85- 1.87 (d, 4H), 51.28-1.33 (t, 3H).
Example 89: Synthesis of l-(4-(2-(2,5-dimethylfuran-3-carboxamido)-7-methyl-lH- benzo[d]imidazol-l-yl)butyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo [d] imidazole-5-carboxamide, 1-33
Figure imgf000411_0001
79.9 1-33
[00737] Into a 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed NMP (2.00 mL), 79.9 (150.00 mg, 0.292 mmol, 1.00 equiv), 2,5-dimethylfuran-3- carboxylic acid (245.57 mg, 1.752 mmol, 6.00 equiv), HATU (666.28 mg, 1.752 mmol, 6.00 equiv), DIEA (226.47 mg, 1.752 mmol, 6.00 equiv). The resulting mixture was stirred for lh at 140°C under microwave monitor. The resulting mixture was washed with 1x20 mL of water. The resulting solution was extracted with 3x20 mL of ethyl acetate and the organic layers combined. The crude product was purified by Flash-Prep-HPLC with the following condition (Column: XBridge Prep OBD C18 Column, 30x 150mm 5um; Mobile Phase A: water, Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 10% B to 40% B in 20 min; 254; 220 nm; RT1 :5.73 min.). This resulted in l-[4-[2-(2,5-dimethylfuran-3-amido)-7-methyl-lH-l,3-benzodiazol-l-yl]butyl]- 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide (44.1 mg, 23.75%) as an off-white solid. LC-MS: (ES, m å) [M+H]+: 636.5, [M-H] : 634.5; 1H-NMR: (400 MHz, DMSO-d 6, ppm) 512.82 (s, 1H), 512.58 (s, 1H), 57.96-7.98 (d, 2H), 57.76-7.78 (d, 1H), 57.57-7.59 (d, 1H), 57.34-7.36 (d, 2H), 57.04-7.07 (m, 1H), 56.95-6.97(d, 1H), 56.60 (s, 1H), 56.31 (s, 1H), 54.56-4.59(t, 2H), 54.28-4.35(d, 4H), 52.60 (s, 3H), 52.51 (s, 3H), 52.12-2.15 (d, 6H), 51.84-1.91 (d, 4H), 51.30-1.34 (t, 3H).
Example 90: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(4-ethyl-2- methyloxazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-methyl-lH- benzo [d] imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-32
Figure imgf000412_0001
39.9 1-32
[00738] To a stirred solution of 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (133.48 mg, 0.860 mmol, 6.00 equiv) in NMP (5 mL) in an 8 mL sealed tube, HATU (327.11 mg, 0.860 mmol, 6 equiv) and DIEA (370.62 mg, 2.868 mmol, 20 equiv) and 2-amino-l-[(2E)-4-(2-amino-5- carbamoyl-lH-l,3-benzodiazol-l-yl)but-2-en-l-yl]-7-methyl-lH-l,3-benzodiazole-5- carboxamide, 39.9 (60.00 mg, 0.143 mmol, 1.00 equiv) were added in portions at room temperature under air atmosphere. After purged and maintained nitrogen atmosphere, the resulting mixture was stirred for lh at 140°C under microwave monitor. The resulting mixture was diluted with water (16 mL). The precipitated solids were collected by filtration and washed with water (3x1 mL). The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD Cl 8 Column 19*250mm, 5um;Mobile Phase A: water(10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 23% B to 35% B in 7 min; 254;220 nm; Rt: 6.72 min) to afford (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(4-ethyl-2- methyloxazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-methyl-lH- benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide (10.8 mg, 10.50%) as a white solid. LC-MS: (ES, m/z) [M+H]+:693.5 (ES, m/z): [M-H] :691.4; ¾-NMR: (300 MHz, DMSO-d6) 512.72 (s, 2H), 57.95-7.97 (d, 2H), 57.84-7.87 (d, 2H), 57.71-7.74 (m, 1H), 57.49 (s, 1H), 57.41-7.44 (d, 1H), 57.29-7.33 (d, 2H), 55.86-5.92 (d, 1H), 55.42-5.55 (d, 1H), 54.95 (s, 2H), 54.77-4.79 (d, 2H), 52.76-2.84 (m, 4H), 52.49 (s, 3H), 52.31-2.40 (m, 6H), 50.98-1.04 (m, 6H).
Example 91: Synthesis of (E)-N,N'-(hex-3-ene-l,6-diylbis(5-carbamoyl-lH- benzo[d]imidazole-l,2-diyl))bis(4-ethyl-2-methyloxazole-5-carboxamide), 1-31
Figure imgf000413_0001
Synthesis of 91.1
[00739] To a stirred solution of (3 E)-hex-3-ene- 1,6-diamine, 77,4 (570 mg, 5 mmol, 1.00 equiv) and 4-chloro-3-nitrobenzamide (2 g, 10 mmol, 2.00 equiv) in dimethyl formamide (20 ml) was added potassium carbonate (2.07 g, 15 mmol, 3.00 equiv). The resulting mixture was stirred overnight at 80°C. The reaction was cooled to r.t and diluted with water. The precipitated solids were collected by fdtration. This resulted in 1 g (45.25%) of 4-[[(3E)-6-[(4-carbamoyl-2- nitrophenyl) amino]hex-3-en-l-yl]amino]-3-nitrobenzamide as a yellow solid. LC-MS (ES, m/z): 443 (M+EE).
Synthesis of 91.2
[00740] To a solution of 4-[[(3E)-6-[(4-carbamoyl-2-nitrophenyl)amino]hex-3-en-l-yl]amino]- 3-nitrobenzamide, 91.1 (1 g, 2.26 mmol, 1.00 equiv) in concentrated hydrochloric acid (10 mL) was added tin( II )chloridedihydrate (5.1 g, 22.6 mmol, 10 equiv). The resulting mixture was stirred at r.t overnight. The mixture was diluted with water. The pH value of the solution was adjusted to 9 with sat. sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. This resulted in 0.32 g (37%) of 3-amino-4-[[(3E)-6-[(2- amino-4-carbamoylphenyl) amino]hex-3-en-l-yl]amino]benzamide as a brown solid. LC-MS (ES, m/z) 383 (M+H+).
Synthesis of 91.3
[00741] To a solution of 3-amino-4-[[(3E)-6-[(2-amino-4-carbamoylphenyl)amino]hex-3-en- l-yl]amino]benzamide, 91.2 (0.32 g, 0.838 mmol, 1.00 equiv) in methanol (10 mL) was added cyanogen bromide (264 mg, 2.514 mmol, 3 equiv). The resulting mixture was stirred at r.t. overnight. The resulting mixture was concentrated under vacuum. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (27% ACN up to 37% in 10 min); Detector, UV 254/220 nm. This resulted in 110 mg (30.39%) of 2-amino-l-[(3E)-6-(2-amino-5-carbamoyl-lH-l,3-benzodiazol-l-yl)hex-3-en-l-yl]- lH-l,3-benzodiazole-5-carboxamideas a brown solid. LC-MS (ES, m/z): 433 (M+H+).
Synthesis of 1-31
[00742] To a stirred solution of 4-ethyl-2-methyloxazole-5-carboxylic acid (78.9 mg, 0.509 mmol, 2.20 equiv) and HATU (211.00 mg, 0.555 mmol, 2.40 equiv) in l-methylpyrrolidin-2-one (5 mL) were added DIEA (298.84 mg, 2.312 mmol, 10.00 equiv) and 2-amino- l-[(3E)-6-(2-amino- 5-carbamoyl-lH-l,3-benzodiazol-l-yl)hex-3-en-l-yl]-lH-l,3-benzodiazole-5-carboxamide, 91.3 (100.00 mg, 0.231 mmol, 1.00 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 140°C. The mixture was cooled to RT and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, X Bridge Prep Phenyl OBD Column, 19* 150mm, 5um; mobile phase, water (0.1% FA) and methanol (50% methanol up to 53% in 8 min); UV detection at 254/210 nm. This resulted in 13.5 mg (8.28%) of (E)-N,N'-(l,T-(hex-3-ene-l,6-diyl)bis(5-carbamoyl-lH-benzo[d]imidazole-2,l-diyl))bis(4-ethyl- 2-methyloxazole-5-carboxamide) as an off-white solid. LC-MS (ES, m/z): 707 (M+H_);'H NMR (400 MHz, DMSO- s) d 12.66 (br s, 2H), 8.06-7.88 (m, 4H), 7.85-7.63 (m, 2H), 7.55-7.18 (m, 4H), 5.58-5.32 (m, 2H), 4.08 (t, 4H), 2.95 (t, 4H), 2.49-2.28 (m, 10H), 1.33-1.09 (m, 8H).
Example 92: Synthesis of N-(3-aminopropyl)-2-(l-(5-(5-((5-carbamoyl-l-methyl-lH- benzo[d]imidazol-2-yl)carbamoyl)-l-ethyl-3-methyl-lH-pyrazol-4-yl)pentyl)-3-methyl-lH- pyrazole-5-carboxamido)-N,l-dimethyl-lH-benzo[d]imidazole-5-carboxamide, 1-29
Figure imgf000415_0001
[00743] To a solution of tert-butyl-N-3-(l-(2-l-(5-(5-(5-carbamoyl-l-methyl-lH-l,3- benzodiazol-2-ylcarbamoyl)-l-ethyl-3-methyl-lH-pyrazol-4-yl)pentyl)-3-methyl-lH-pyrazole-5- amido)-l-methyl-lH-l,3-benzodiazole-5-yl)-N-methylformamido)propylcarbamate, 86.8 (100 mg, 0.116 mmol, 1 equiv) in dichloromethane (10 mL) was added trifluoroacetic acid (3 mL). The resulting mixture was stirred for 2h at RT. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD Column 30* 150mm, 5um; mobile phase, Water (0.1% FA) and ACN (18% ACN up to 28% in 8 min); UV detection at 254/210 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 18.5 mg (20.95%) of N-(3-aminopropyl)- 2-(l-(5-(5-(5 -carbamoyl- 1 -methyl- lH-benzo[d]imidazol-2-ylcarbamoyl)- 1 -ethyl -3 -methyl- 1H- pyrazol-4-yl)pentyl)-3-methyl-lH-pyrazole-5-carboxamido)-N,l-dimethyl-lH- benzo[d]imidazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 383 (M/2+FC); 1H NMR (400 MHz, DMSO-di) d 8.02-7.93 (m, 2H), 7.79 (dd, 1H), 7.57-7.42 (m, 3H), 7.38-7.25 (m, 2H), 6.67 (s, 1H), 4.67-4.52 (m, 4H), 3.68-3.38 (m, 8H), 2.95-2.58 (m, 7H), 2.16 (s, 3H), 2.09 (s, 3H), 1.93-1.75 (m, 4H), 1.61-1.50 (m, 2H), 1.47-1.27 (m, 5H).
Example 93: Synthesis of 1-28
Figure imgf000416_0001
Figure imgf000417_0001
Synthesis of 93.1
[00744] To a solution of 4-chloro-3-nitrobenzamide (1 g, 4.986 mmol, 1.00 equiv) in dimethyl formamide (30 mL) was added potassium carbonate (2.067 g, 14.957 mmol, 3.00 equiv) and tert- butyl 4-(2-aminoethyl)piperidine-l-carboxylate (1.068 g, 4.986 mmol, 1.00 equiv). The resulting mixture was stirred at 80°C overnight. The mixture was cooled to r.t, diluted with water. The solids were collected by filtration. This resulted in 1.1 g (58.30%) of tert-butyl 4-[[(4-carbamoyl-2- nitrophenyl)amino]methyl]piperidine-l-carboxylate as a yellow solid. LC-MS (ES, m/z): 279 (M- 100+H+).
Synthesis of 93.2
[00745] To a solution of tert-butyl 4-[[(4-carbamoyl-2-nitrophenyl)amino]methyl]piperidine-l- carboxylate, 93.1 (1.10 g, 2.907 mmol, 1.00 equiv) in methanol (30 mL) was added palladium on carbon (500 mg). To the above hydrogen (g) was introduced in. The mixture was stirred for 2h at room temperature. The solid was filtered. The filtrate was concentrated under reduced pressure. This resulted in 960 mg (94.78%) of tert-butyl 4-[[(2-amino-4- carbamoylphenyl)amino]methyl]piperidine-l-carboxylate as a black solid. LC-MS (ES, m/z): 349 (M+H+);
Synthesis of 93.3
[00746] To a stirred solution of tert-butyl 4-[[(2-amino-4- carbamoylphenyl)amino]methyl]piperidine-l-carboxylate, 93.2 (960 mg, 2.755 mmol, 1.00 equiv) in methanol (30 mL) was added cyanogen bromide (875.46 mg, 8.265 mmol, 3.00 equiv). The resulting solution was stirred for 3h at room temperature. The mixture was concentrated under reduced pressure. This resulted in 1.1 g (crude) of tert-butyl 4-[(2-amino-5- carbamoyl-lH-l,3-benzodiazol-l-yl) methyl]piperidine-l-carboxylate as a black solid. LC-MS (ES, m/z): 374 (M+H+);
Synthesis of 93.4
[00747] To a stirred solution of tert-butyl 4-[(2-amino-5-carbamoyl-lH-l,3-benzodiazol-l- yl)methyl]piperidine-l-carboxylate, 93.3 (100 mg, 0.268 mmol, LOO equiv) in dimethyl formamide (5 mL) was added l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (41.28 mg, 0.268 mmol, 1.00 equiv), DIEA (173.04 mg, 1.339 mmol, 5.00 equiv) and HATU (122.18 mg, 0.321 mmol, 1.20 equiv). The resulting mixture was stirred at RT overnight. The reaction mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH Prep C18 OBD Column 19*250mm, 5um; mobile phase, Water (0.1% FA) and ACN (14% ACN up to 38% in 7 min); Detector, UV 254/210 nm. This resulted in 10.4 mg (7.62%) oftert-butyl 4-[[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole- 5-amido)-lH-l,3-benzodiazol-l-yl]methyl]piperidine-l-carboxylate as a light brown solid. LC- MS (ES, m/z): 510 (M+H+); 1H NMR (300 MHz, DMSO-tfc) d 12.84 (s, 1H), 8.03-7.85 (m, 2H), 7.79 (d, 1H), 7.60 (d, 1H), 7.32 (s, 1H), 6.64 (s, 1H), 4.62 (q, 2H), 4.11-3.91 (m, 4H), 2.78-2.58 (m, 2H), 2.20-2.03 (m, 4H), 1.56-1.17 (m, 16H).
Synthesis of 93.5
[00748] To a stirred solution of tert-butyl 4-[[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-l-yl]methyl]piperidine-l-carboxylate, 93.4 (50 mg, 0.098 mmol, 1.00 equiv) in dichloromethane (5 mL) was added trifluoroacetic acid (1 mL). The resulting mixture was stirred at RT for 2 h. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep Cl 8 column, 30* 150mm, 5um; mobile phase, Water (0.1% FA) and ACN (6% ACN up to 20% in 7 min); Detector, UV 254/210 nm. This resulted in 15.9 mg (35.58%) of 2-(l -ethyl-3 -methyl- lH-pyrazole- 5-amido)-l-[(piperidin-4-yl)methyl]-lH-l,3-benzodiazole-5-carboxamide as a white solid. LC- MS (ES, m/z): 410 (M+H+); ¾ NMR (400 MHz, DMSO-i/e) d 8.03-7.98 (m, 2H), 7.80 (d, 1H), 7.58 (d, 1H), 7.33 (s, 1H), 6.67 (s, 1H), 4.62 (q, 2H), 4.13 (d, 2H), 3.21-3.12 (m, 2H), 2.72-2.58 (m, 2H), 2.22-2.12 (m, 4H), 1.73-1.64 (m, 2H), 1.55-1.36 (m, 5H).
Synthesis of 93.6
[00749] To a stirred solution of 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-[(piperidin-4- yl)methyl]-lH-l,3-benzodiazole-5-carboxamide, 93.5 (420 mg, 1.026 mmol, 1.00 equiv) in dimethyl formamide (10 mL) was added DIEA (2651.18 mg, 20.513 mmol, 20.00 equiv), 2-[(4- carbamoyl-2-nitrophenyl)amino]acetic acid (294.39 mg, 1.231 mmol, 1.20 equiv) and HATU (584.98 mg, 1.538 mmol, 1.5 equiv). The resulting mixture was stirred at RT for 2h. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (28% ACN up to 47% in 7 min); Detector, UV 254/220 nm. This resulted in 440 mg (68.02%) of l-[(l-[2-[(4-
418
5U B5TITUTE SH EET (RU LE 26) carbamoyl-2-nitrophenyl)amino]acetyl]piperidin-4-yl)methyl]-2-(l-ethyl-3-methyl-lH-pyrazole- 5-amido)-lH-l,3-benzodiazole-5-carboxamide as a yellow solid. LC-MS (ES, m/z): 629 (M-H+);
Synthesis of 93.7
[00750] To a solution of l-[(l-[2-[(4-carbamoyl-2-nitrophenyl)amino]acetyl]piperidin-4- yl)methyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide,
93.6 (200 mg, 0.317 mmol, 1.00 equiv) in methanol (10 mL) was added palladium on carbon (50 mg). To the above hydrogen (g) was introduced in. The mixture was stirred for 2h at room temperature. The solid was filtered. The filtrate was concentrated under reduced pressure. This resulted in 180 mg (94.49%) of l-[(l-[2-[(2-amino-4-carbamoylphenyl)amino]acetyl]piperidin-4- yl)methyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide as a yellow solid. LC-MS (ES, m/z): 601 (M+H+);
Synthesis of 93.8
[00751] To a stirred solution of l-[(l-[2-[(2-amino-4- carbamoylphenyl)amino]acetyl]piperidin-4-yl)methyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazole-5-carboxamide, 93.7 (180 mg, 0.300 mmol, 1.00 equiv) in methanol (10 mL) was added cyanogen bromide (95.22 mg, 0.899 mmol, 3.00 equiv). The resulting mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. This resulted in 200 mg (crude) of 2-amino-l-[2-(4-[[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- lH-l,3-benzodiazol -l-yl]methyl]piperidin-l-yl)-2-oxoethyl]-lH-l,3-benzodiazole-5- carboxamide as an orange solid. LC-MS (ES, m/z): 626 (M+H+);
Synthesis of 1-28
[00752] To a stirred solution of 2-amino-l-[2-(4-[[5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]methyl]piperidin-l-yl)-2-oxoethyl]-lH-l,3- benzodiazole-5-carboxamide, 93.8 (100 mg, 0.16 mmol, 1.00 equiv) in dimethyl formamide (5 mL) was added l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (29.57 mg, 0.192 mmol, 1.20 equiv), DIEA (103.2 mg, 0.8 mmol, 5.00 equiv) and HATU (72.96 mg, 0.192 mmol, 1.20 equiv). The resulting mixture was irradiated with microwave radiation at 120°C for lh. The reaction mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Prep OBD C18 Column, 30* 150mm, 5um; mobile phase, Water (0.01% MLHCCb+O. L/oMLTLO) and ACN (15% ACN up to 35% in 7 min); Detector, UV 254/220 nm. This resulted in 33.4 mg (27.4%) of l-[(l-[2-[5-carbamoyl-2-(l- ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]acetyl]piperidin-4-yl)methyl]-2- (l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 762 (M+H+); ¾ NMR (400 MHz, DMSC ) d 12.86 (br s, 2H), 8.08-7.91 (m, 4H), 7.82 (dd, 1H), 7.76 (dd, 1H), 7.62 (d, 1H), 7.44-7.32 (m, 3H), 6.65 (s, 2H), 5.15 (q, 2H), 4.66-4.57 (m, 4H), 4.31-4.12 (m, 4H), 3.21-3.11 (m, 1H), 2.67-2.58 (m, 1H), 2.33-2.25 (m, 1H), 2.17 (s, 3H), 2.13 (s, 3H), 1.81-1.66 (m, 2H), 1.52-1.45 (m, 1H), 1.38-1.18 (m, 7H).
Example 94: Synthesis of (S)-l-(2-(l-(2-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)acetyl)pyrrolidin-3-yl)ethyl)-2-(l-ethyl-3-methyl- lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-27
Figure imgf000421_0001
Figure imgf000422_0001
94.11
Figure imgf000423_0001
94.14
Synthesis of 94.1
[00753] To a solution of 2-[(3R)-l-[(tert-butoxy)carbonyl]pyrrolidin-3-yl]acetic acid (2.00 g, 8.723 mmol, 1.00 equiv) and ammonium chloride (2.33 g, 43.616 mmol, 5 equiv) in dimethyl formamide (30 mL) were added HATU (6.63 g, 17.446 mmol, 2 equiv) and DIEA (4.51 g, 34.892 mmol, 4 equiv). The resulting solution was stirred overnight at room temperature. The mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 900 mg (45.25%) of tert-butyl (3R)-3-(carbamoylmethyl)pyrrolidine-l-carboxylate as a light yellow solid. LC-MS (ES, m/z): 227 (M-H+).
Synthesis of 94.2
[00754] To a solution of tert-butyl (3R)-3-(carbamoylmethyl)pyrrolidine-l-carboxylate, 94.1 (900 mg, 3.942 mmol, 1.00 equiv) in tetrahydrofuran (20 mL) was added boranetetrahydrofurancomplexsolution (1 mol/L, 19.71 mL, 19.710 mmol, 5.00 equiv) at 0°C. The resulting solution was stirred overnight at room temperature. The mixture was quenched with methanol and concentrated under vacuum. This resulted in 800 mg (94.45%) of tert-butyl (3S)-3- (2-aminoethyl)pyrrolidine-l-carboxylate as a yellow oil. LC-MS (ES, m/z): 215 (M+H+).
Synthesis of 94.3
[00755] To a solution of tert-butyl (3S)-3-(2-aminoethyl)pyrrolidine-l-carboxylate, 94.2 (800 mg, 3.733 mmol, 1.00 equiv) and 4-chloro-3-nitrobenzamide (898.50 mg, 4.480 mmol, 1.2 equiv) in dimethyl formamide (15 mL) were added potassium carbonate (1.548 g, 1 1.199 mmol, 3 equiv). The resulting solution was stirred overnight at 80oC. The mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography with PE/ethyl acetate (1/1) to afford 460 mg (31.59%) of tert-butyl (3S)-3-[2-[(4-carbamoyl-2-nitrophenyl)amino]ethyl]pyrrolidine-l- carboxylate as a yellow solid. LC-MS (ES, m/z): 279 (M+H+-100).
Synthesis of 94.4
[00756] To a solution of tert-butyl (3S)-3-[2-[(4-carbamoyl-2- nitrophenyl)amino]ethyl]pyrrolidine-l-carboxylate, 94.3 (460 mg, 1.216 mmol, 1.00 equiv) in methanol (15 mL) was added palladium on carbon (50 mg). To the above hydrogen (g) was introduced in. The resulting solution was stirred for 2h at room temperature. The solids were filtered out. The filtrate was concentrated under vacuum. This resulted in 500 mg (crude) of tert- butyl tert-butyl (3S)-3-[2-[(2-amino-4-carbamoylphenyl)amino]ethyl]pyrrolidine-l- carboxylate as a brown oil. LC-MS (ES, m/z): 349 (M+H+).
Synthesis of 94.5
[00757] To a solution of tert-butyl tert-butyl (3 S)-3-[2-[(2-amino-4- carbamoylphenyl)amino]ethyl]pyrrolidine-l-carboxylate, 94.4 (348 mg, 1 mmol, 1.00 equiv) in methanol (15 mL) was added cyanogen bromide (210 mg, 2 mmol, 2 equiv). The resulting solution was stirred for 3 hours at room temperature. The resulting mixture was concentrated under vacuum to give 400 mg (crude) of tert-butyl (3S)-3-[2-(2-amino-5-carbamoyl-lH-l, 3- benzodiazol-l-yl)ethyl]pyrrolidine-l-carboxylate as a brown oil. LC-MS (ES, m/z): 372 (M-H+).
[00758] Synthesis of 94.6
To a solution of tert-butyl (3S)-3-[2-(2-amino-5-carbamoyl-lH-l,3-benzodiazol-l- yl)ethyl]pyrrolidine-l-carboxylate, 94.5 (50 mg, 0.134 mmol, 1.00 equiv) and 1 -ethyl -3-methyl - lH-pyrazole-5-carboxylic acid (26.83 mg, 0.174 mmol, 1.3 equiv) in dimethyl formamide (5 mL) were added DIEA (51.91 mg, 0.402 mmol, 3.00 equiv) and HATU (76.36 mg, 0.201 mmol, 1.5 equiv). The resulting solution was stirred overnight at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep C18 column, 30* 150mm, 5um; mobile phase, Water (0.1%FA) and ACN (30% ACN up to 58% in 7 min); Detector, UV 254/210 nm. This resulted in 6.1 mg (8.94%) of tert-butyl (3S)-3-[2-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- lH-l,3-benzodiazol-l-yl]ethyl]pyrrolidine-l-carboxylate as a white solid. LC-MS (ESI): 510 (M+H+); ¾-NMR (400 MHz, DMSO-ifc) d 12.76 (br s, 1H), 8.03-7.95 (m, 2H), 7.82 (d, 1H), 7.58 (d, 1H), 7.34 (s, 1H), 6.67 (d, 1H), 4.63 (q, 2H), 4.32-4.15 (m, 2H), 3.52-3.40 (m, 1H), 3.40-3.25 (m, 1H), 3.20-3.05 (m, 1H), 2.93-2.80 (m, 1H), 2.25-2.06 (m, 4H), 2.05-1.93 (m, 1H), 1.90-1.75 (m, 2H), 1.62-1.42 (m, 1H), 1.40-1.29 (m, 12H).
[00759] Synthesis of 94.7
To a solution of tert-butyl (3S)-3-[2-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)- lH-l,3-benzodiazol-l-yl]ethyl]pyrrolidine-l-carboxylate, 94.6 (50.00 mg, 0.098 mmol, 1.00 equiv) in dichloromethane (5 mL) was added tifluoroacetic aid (1 mL). The resulting solution was stirred for lh at room temperature. The mixture was concentrated. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep C18 column, 30* 150mm, 5um; mobile phase, Water (0.1%FA) and ACN (5% ACN up to 21% in 7 min); Detector, UV 254/210 nm. This resulted in 7 mg (15.63%) of 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-[2- [(3R)-pyrrolidin-3-yl]ethyl]-lH-l,3-benzodiazole-5-carboxamide as a pink solid. LC-MS (ESI): 410 (M+H+); 1H-NMR (400 MHz, DMSO -d6) d 8.05-7.98 (m, 2H), 7.82 (dd, 1H), 7.59 (d, 1H),
7.35 (s, 1H), 6.68 (s, 1H), 4.62 (q, 2H), 4.38-4.20 (m, 2H), 3.35-3.26 (m, 1H), 3.22-3.13 (m, 1H), 3.06-2.95 (m, 1H), 2.81-2.70 (m, 1H), 2.25-2.01 (m, 5H), 1.96-1.81 (m, 2H), 1.60-1.49 (m, 1H),
1.35 (t, 3H).
Synthesis of 94.8
[00760] To a stirred mixture of 4-amino-3-nitrobenzoic acid (5 g, 27.47 mmol, 1 equiv) in DMF (30 mL) was added MLCl (7.30 g, 137.36 mmol, 5 equiv), DIEA (35.68 g, 274.7 mmol, 10 equiv) and HATU (15.6 g, 41.2 mmol, 1.5 equiv). The resulting solution was stirred overnight at room temperature. The mixture was diluted with water. The solids were collected by filtration. This resulted in 4.1 g (82%) of 4-amino-3-nitrobenzamide as a yellow solid. LC-MS (ES, m/z): 182 (M+H+).
Synthesis of 94.9
[00761] To a stirred solution of 4-amino-3-nitrobenzamide, 94.8 (4.1 g, 22.53 mmol, 1 equiv) in acetic acid (100 mL) was added zinc (7.21 g, 112.65 mmol, 5 equiv). The resulting solution was stirred for 4 hour at room temperature. The solids were filtered out. The resulting mixture was concentrated. This resulted in 4 g (crude) of 3,4-diaminobenzamide as purple oil. LC-MS (ES, m/z): 152 (M+H+).
Synthesis of 94.10
[00762] To a stirred solution of 3,4-diaminobenzamide, 94.9 (4 g crude, 22.53 mmol, 1 equiv) in methanol (50 mL) was added carbononitridic bromide (3.58 g, 33.795 mmol, 1.5 equiv). The resulting solution was stirred overnight at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0. 1%FA) and ACN (30% ACN up to 42% in 10 min); Detector, UY 254/220 nm. This resulted in 2.4 g (60.6%) of 2-amino-lH-l,3- benzodiazole-5-carboxamide as a light yellow solid. LC-MS (ES, m/z): 177 (M+H+).
Synthesis of 94.11
[00763] To a stirred solution of 2-amino-lH-l,3-benzodiazole-5-carboxamide, 94.10 (1.3 g, 7.378 mmol, 1 equiv) in DMF (20 mL) and addded DIEA (1.90 g, 14.76 mmol, 2 equiv) and ethyl 2-bromoacetate (1.22 g, 7.378 mmol, 1 equiv). The resulting solution was stirred for 3 hours at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (10% NH4HCO3) and ACN (40% ACN up to 50% in 10 min); Detector, UV 254/220 nm. This resulted in 850 mg (44%) of ethyl 2-(2-amino-5-carbamoyl-lH-l,3-benzodiazol-l-yl)acetate and ethyl 2- (2-amino-6-carbamoyl-lH-benzo[d]imidazol-l-yl)acetate as a light yellow solid. LC-MS (ES, m/z): 263 (M+H+); Synthesis of 94.12
[00764] To a stirred solution of ethyl 2-(2-amino-5-carbamoyl-lH-l,3-benzodiazol-l-yl)acetate and ethyl 2-(2-amino-6-carbamoyl-lH-benzo[d]imidazol-l-yl)acetate, 94.11 (700 mg, 2.67 mmol, 1 equiv) and 3-methyl-lH-pyrazole-5-carboxylic acid (493.00 mg, 3.2 mmol, 1.2 equiv) in DMF (10 mL) was added DIEA (1.74 mg, 13.35 mmol, 5 equiv) and HATU (1.5 g, 4 mmol, 1.5 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 100°C. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (10% NH4HCO3) and ACN (30% ACN up to 40% in 8 min); Detector, UV 254/220 nm. This resulted in 360 mg (66.7%) of ethyl 2- [5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]acetate and ethyl 2-(6-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l- yl)acetate as a yellow solid. LC-MS (ES, m/z): 299 (M+H+).
Synthesis of 94.13 & 94.14
[00765] To a stirred solution of ethyl 2-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-l-yl]acetate and ethyl 2-(6-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)acetate, 94.12 (120 mg, 0.4 mmol, 1 equiv) in MeOH (5 mL), THF (5 mL) and LhO (2 mL) was added LiOH (28.8 mg, 1.2 mmol, 3 equiv). The resulting solution was stirred for 2 hour at room temperature. The resulting mixture was concentrated. The pH value of the solution was adjusted to 3 with IN HC1. The solids were collected by filtration. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD Column 30* 150mm 5um, mobile phase, Water (0.1%FA) and ACN (40% ACN up to 60% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 11.2 mg (7.57%) of 2-[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l- yl]acetic acid as a white solid and 15 mg (10.14%) of 2-[6-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]acetic acid as a white solid. 94.13: LC-MS (ES, m/z):371 (M+H+); ¾ NMR: ¾ NMR (400 MHz, DMSO-t 6) d 13.26 (br s, 1H), 12.87 (s, 1H), 8.05-7.91 (m, 2H), 7.80 (dd, 1H), 7.54 (dd, 1H), 7.34 (br s, 1H), 6.64 (s, 1H), 5.00 (s, 2H), 4.60 (q, 2H), 2.18 (s, 3H), 1.35 (t, 3H). 94.14: LC-MS (ES, m/z): 371 (M+H+); ¾NMR: Tl NMR ^OO MHz, DMSO -d6) d 12.86 (br s, 1H), 8.05-7.91 (m, 2H), 7.80 (dd, 1H), 7.52 (d, 1H), 7.38 (br s, 1H), 6.64 (s, 1H), 4.97 (s, 2H), 4.59 (q, 2H), 2.17 (s, 3H), 1.34 (t, 3H).
Synthesis of 1-27
[00766] To a solution of 2-(2-ethyl-5-methylpyrazole-3-amido)-l-[2-[(3R)-pyrrolidin-3- yl]ethyl]-l,3-benzodiazole-5-carboxamide, 94.7 (100.00 mg, 0.244 mmol, 1.00 equiv) and [5- carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]acetic acid, 94.13 (108.53 mg, 0.293 mmol, 1.20 equiv) in dimethyl formamide (10 mL) were added DIEA (94.69 mg, 0.733 mmol, 3.00 equiv) and HATU (139.28 mg, 0.366 mmol, 1.50 equiv). The resulting solution was stirred for 4h at room temperature. The mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep C18 column, 30* 150, 5um; mobile phase, Water (0.1%FA) and ACN (22% ACN up to 36% in 7 min); Detector, UV 254/210 nm. This resulted in 19.7 mg (9.79%) of l-[2-[(3S)-3-[2-[5- carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]ethyl]pyrrolidin-l-yl]-2- oxoethyl]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5-carboxamide asan off-white solid. LC-MS (ESI): 762 (M+H+); ^-NMR (400 MHz, CD OD) d 8.08-7.95 (m, 2H), 7.91-7.79 (m, 2H), 7.62-7.50 (m, 1H), 7.43-7.30 (m, 1H), 6.83-6.52 (m, 2H), 5.15-4.95 (m, 1H), 4.70-4.40 (m, 5H), 4.40-4.25 (m, 2H), 3.95-3.55 (m, 2H), 3.54-3.42 (m, 1H), 3.30-3.10 (m, 1H), 2.40-2.20 (m, 2H), 2.20-1.96 (m, 8H), 1.95-1.63 (m, 1H), 1.45-1.25 (m, 6H).
Example 95: Synthesis of (R)-l-((l-(2-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)acetyl)pyrrolidin-3-yl)methyl)-2-(l -ethyl-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-26
Figure imgf000429_0001
Figure imgf000430_0001
Synthesis of 95.1
[00767] To a solution of 4-chloro-3-nitrobenzamide (3 g, 14.96 mmol, 1 equiv) and tert-butyl (3S)-3-(aminomethyl)pyrrolidine-l-carboxylate (2.8 g, 14.96 mmol, 1 equiv) in dimethyl formamide (60 mL) was added potassium carbonate (6.2 g, 44.88 mmol, 3 equiv). The resulting mixture was stirred at 80oC overnight. The mixture was cooled to r.t and diluted with water. The precipitated solids were collected by filtration. This resulted in 2.7 g (49.58%) of tert-butyl (3S)- 3-[[(4-carbamoyl-2-nitrophenyl)amino]methyl]pyrrolidine-l-carboxylate as a yellow oil. LC-MS (ES, m/z): 387 (M+Na+);
Synthesis of 95.2
[00768] To a solution of tert-butyl (3S)-3-[[(4-carbamoyl-2- nitrophenyl)amino]methyl]pyrrolidine-l-carboxylate (2.7 g, 7.42 mmol, 1 equiv) in methanol (30 mL) was added palladium on carbon (500 mg). To the above hydrogen (g) was introduced in. The resulting mixture was stirred at RT for 4 h. The mixture was filtered. The filtrate was concentrated under reduced pressure. This resulted in 2.2 g (88.77%) of 3-amino-4-([[(3R)- pyrrolidin-3-yl]methyl]amino)benzamide as a brown oil. LC-MS (ES, m/z): 235 (M-100+LL); Synthesis of 95.3
[00769] To a solution of 3-amino-4-([[(3R)-pyrrolidin-3-yl]methyl]amino)benzamide (835 mg, 2.5 mmol, 1 equiv) in methanol (10 mL) was added cyanogen bromide (397.5 mg, 3.75 mmol, 1.5 equiv). The resulting solution was stirred for 3h at room temperature. The mixture was concentrated. This resulted in 1 g (crude) of tert-butyl (3S)-3-[(2-amino-5-carbamoyl-lH-l,3- benzodiazol-l-yl)methyl]pyrrolidine-l-carboxylate as a brown solid. LC-MS (ES, m/z): 360 (M+H+);
Synthesis of 95.4
[00770] To a solution of tert-butyl (3S)-3-[(2-amino-5-carbamoyl-lH-l,3-benzodiazol-l- yl)methyl]pyrrolidine-l-carboxylate, 95.3 (60 mg, 0.167 mmol, 1 equiv) and l-ethyl-3-m ethyl - lH-pyrazole-5-carboxylic acid (25.72 mg, 0.167 mmol, 1 equiv) in dimethyl formamide (5 mL) was added HATU (76.2 mg, 0.2 mmol, 1.2 equiv) and DIEA (107.72 mg, 0.835 mmol, 5 equiv). The reaction mixture was stirred at RT for 2h. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Shield RP18 OBD Column 30* 150mm, 5um; mobile phase, Water (0.01% NH4HCO3) and ACN (27% ACN up to 49% in 7 min); Detector, UV 220/254 nm. This resulted in 6.9 mg (8.35%) of tert-butyl (3S)-3-[[5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-amido)-lH-l,3-benzodiazol-l-yl]methyl]pyrrolidine-l-carboxylate as a white solid. LC-MS (ES, m/z): 496 (M+H+); ¾ NMR (400 MHz, DMSO-i e) d 12.86 (s, 1H), 8.08-7.96 (m, 2H), 7.81 (d, 1H), 7.62 (d, 1H), 7.35 (s, 1H), 6.68 (s, 1H), 4.63 (q, 2H), 4.25 (d, 2H), 3.51-3.35 (m, 2H), 3.29-3.12 (m, 2H), 2.89-2.74 (m, 1H), 2.18 (s, 3H), 1.97-1.86 (m, 1H), 1.81-1.66 (m, 1H), 1.45-1.31 (m, 12H).
Synthesis of 95.5
[00771] To a solution of tert-butyl (3R)-3-[[5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazol-l-yl]methyl]pyrrolidine-l-carboxylate, 95.4 (50 mg, 0.101 mmol, 1 equiv) in dichloromethane (8 mL) was added trifluoroacetic acid (2 mL). The reaction mixture was stirred at RT for 2h. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH Prep Cl 8 OBD Column 19* 150mm, 5um; mobile phase, Water (0.1% LA) and ACN (5% ACN up to 17% in 7 min); Detector, UV 254/220 nm. This resulted in 9.9 mg (8.35%) of 2-(l-ethyl-3-methyl-lH-
430
5UB5TITUTE SHEET (RULE 26) pyrazole-5-amido)-l-[[(3S)-pyrrolidin-3-yl]methyl]-lH-l,3-benzodiazole-5-carboxamide as an off-white solid. LC-MS (ES, m/z): 396 (M+H+); ¾ NMR (400 MHz, DMSO -d6) d 8.08-7.96 (m, 2H), 7.82 (dd, 1H), 7.66 (d, 1H), 7.35 (s, 1H), 6.75 (s, 1H), 4.61 (q, 2H), 4.35-4.22 (m, 2H), 3.31- 3.16 (m, 2H), 3.12-2.97 (m, 2H), 2.94-2.79 (m, 2H), 2.19 (s, 3H), 2.01-1.91 (m, 1H), 1.80-1.66 (m, 1H), 1.36 (t, 3H).
Synthesis of 95.6
[00772] To a solution of 2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-l-[[(3S)-pyrrolidin-3- yl]methyl]-lH-l,3-benzodiazole-5-carboxamide, 95.5 (1 g, 2.532 mmol, 1 equiv) and 2-[(4- carbamoyl-2-nitrophenyl)amino]acetic acid (729 mg, 3.04 mmol, 1.2 equiv) in dimethyl formamide (20 mL) was added HATU (1.443 g, 3.798 mmol, 1.5 equiv) and DIEA ( 3.27 g, 25.32mmol, 10 equiv). The resulting mixture was stirred at RT overnight. The mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (45% ACN up to 55% in 10 min); Detector, UV 254/210 nm. This resulted in 750 mg (48%) of l-[[(3R)-l-[2-[(4-carbamoyl-2-nitrophenyl)amino]acetyl]pyrrolidin-3- yl]methyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide as a yellow solid. LC-MS (ES, m/z): 617 (M+H+);
Synthesis of 95.7
[00773] To a solution of l-[[(3R)-l-[2-[(4-carbamoyl-2-nitrophenyl)amino]acetyl]pyrrolidin- 3-yl]methyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide, 95.6 (700 mg, 1.22 mmol, 1 equiv) in methanol (30 mL) was added palladium on carbon (200 mg). To the above hydrogen(g) was introduced in. The mixture was stirred for 2h at room temperature. The solid was filtered. The filtrate was concentrated under reduced pressure. This resulted in 350 mg (48.96%) of l-[[(3R)-l-[2-[(2-amino-4- carbamoylphenyl)amino]acetyl]pyrrolidin-3-yl]methyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-lH-l,3-benzodiazole-5-carboxamide as an off-white solid. LC-MS (ES, m/z): 587 (M+H+); Synthesis of 95.8
[00774] To a solution of l-[[(3R)-l-[2-[(2-amino-4-carbamoylphenyl)amino]acetyl]pyrrolidin- 3-yl]methyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide, 95.7 (350 mg, 0.6 mmol, 1 equiv) in methanol (10 mL) was added cyanogen bromide (127.2 mg, 1.2 mmol, 2 equiv). The resulting mixture was stirred for 2h at RT. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (38% ACN up to 48% in 10 min); Detector, UV 254/220 nm. This resulted in 200 mg (54%) of l-[[(3R)-l-[2-(2- amino-5-carbamoyl-lH-l,3-benzodiazol-l-yl)acetyl]pyrrolidin-3-yl]methyl]-2-(l-ethyl-3- methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazole-5-carboxamide as a yellow solid. LC-MS (ES, m/z): 612 (M+H+);
Synthesis of 1-26
[00775] To a solution of l-[[(3R)-l-[2-(2-amino-5-carbamoyl-lH-l,3-benzodiazol-l- yl)acetyl]pyrrolidin-3-yl]methyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxamide, 95.8 (50 mg, 0.082 mmol, 1.00 equiv) in l-methylpyrrolidin-2-one (5 mL) was added l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (18.87 mg, 0.123 mmol, 1.5 equiv), HATU (46.74 mg, 0.123 mmol, 1.5 equiv) and DIEA (52.89 mg, 0.41 mmol, 5 equiv). The final reaction mixture was irradiated with microwave radiation at 140°C for lh. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH Prep C18 OBD Column 19*250mm, 5um; mobile phase, Water (0.1% FA) and ACN (33% ACN up to 50% in 7 min); Detector, UV 254/220 nm. This resulted in 7.3 mg (12%) of l-[2-[(3R)-3-[[5- carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3-benzodiazol-l- yl]methyl]pyrrolidin-l-yl]-2-oxoethyl]-2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-lH-l,3- benzodiazole-5-carboxamide as a light brown solid. LC-MS (ES, m/z): 747 (M-FT); 1 H NMR (400 MHz, DMSO- ) S 12.78 (br s, 2H), 8.11-7.94 (m, 4H), 7.92-7.64 (m, 3H), 7.61-7.28 (m, 3H), 6.78-6.59 (m, 2H), 5.14-4.88 (m, 2H), 4.68-4.52 (m, 4H), 4.44-4.25 (m, 2H), 3.98-3.49 (m, 4H), 2.18 (s, 3H), 2.13 (s, 3H), 2.09-1.70 (m, 3H), 1.42-1.23 (m, 6H). Example 96: Synthesis of 2-(l-(5-(5-((5-carbamoyl-lH-benzo[d]imidazol-2-yl)carbamoyl)-l- ethyl-3-methyl-lH-pyrazol-4-yl)pentyl)-3-methyl-lH-pyrazole-5-carboxamido)-N-(3-(2-((2- (2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)oxy)acetamido)propyl)-N-methyl-lH- benzo [d] imidazole-5-carboxamide, 1-25
Figure imgf000434_0001
96.6
Figure imgf000435_0001
Synthesis of 96.1
[00776] To a stirred solution of methyl 4-fluoro-3-nitrobenzoate (4 g, 20 mmol, 1 equiv) in dimethyl formamide (40 mL) was added (2,4-dimethoxyphenyl)methanamine (5.01 g, 30 mmol, 1.5 equiv) and DIEA (7.74 g, 60 mmol, 3 equiv). The resulting mixture was stirred overnight at RT. The mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with PE/ethyl acetate (3/1) to give 4.58 g (66%) of methyl 4-(2,4- dimethoxybenzylamino)-3-nitrobenzoate as a yellow solid. LC-MS (ES, m/z): 347 (M+H+).
Synthesis of 96.2
[00777] To a stirred solution of methyl 4-(2,4-dimethoxybenzylamino)-3-nitrobenzoate, 96.1 (2 g, 5.775 mmol, 1 equiv) in acetic acid (40 mL) was added zinc (1.88 g, 28.875 mmol, 5 equiv). The resulting mixture was stirred for 4 h at room temperature. The mixture was filtered. The filtrate was concentrated under reduced pressure. This resulted in 2 g (crude) of methyl 3-amino-4-[[(2,4- dimethoxyphenyl)methyl]amino]benzoate as a black solid. LC-MS (ES, m/z): 317 (M+H+); Synthesis of 96.3
[00778] To a stirred solution of methyl 3-amino-4-[[(2,4- dimethoxyphenyl)methyl]amino]benzoate, 96.2 (2 g crude, 5.775 mmol, 1.00 equiv) in methanol (30 mL) was added cyanogen bromide (1.224 g, 11.55 mmol, 2 equiv). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (10%NH4HCO3) and ACN (31% ACN up to 44% in 10 min); Detector, UV 254/220 nm. This resulted in 1.6 g (81.22%) of methyl 2-amino-l-[(2,4-dimethoxyphenyl)methyl]-lH-l,3- benzodiazole-5-carboxylate as an off-white solid. LC-MS (ES, m/z): 342 (M+H+).
Synthesis of 96.4
[00779] To a solution of methyl 2-amino-l-[(2,4-dimethoxyphenyl)methyl]-lH-l,3- benzodiazole-5-carboxylate, 96.3 (1 g, 2.929 mmol, 1 equiv) in methanol (30 mL) and water (8 mL) was added lithium hydroxide (281.28 mg, 11.72 mmol, 4.00 equiv). The resulting mixture was stirred for 2h at 65°C. The mixture was cooled to RT and concentrated under vacuum. The residue was acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration. This resulted in 700 mg (72.84%) of 2- amino-l-[(2,4-dimethoxyphenyl)methyl]-lH-l,3-benzodiazole-5-carboxylic acid as a yellow solid. LC-MS (ES, m/z): 328 (M+H+).
Synthesis of 96.5
[00780] To a stirred solution of 2-amino-l-[(2,4-dimethoxyphenyl)methyl]-lH-l,3- benzodiazole-5-carboxylic acid, 96.4 (1.20 g, 3.666 mmol, 1 equiv) in dimethyl formamide (15 mL) was added HATU (2.09 g, 5.499 mmol, 1.50 equiv), DIEA (2.37 g, 18.338 mmol, 5 equiv) and tert-butyl N-[3-
(methylamino)propyl]carbamate (1.38 g, 7.332 mmol, 2 equiv). The resulting mixture was stirred for 2 h at room temperature. The reaction was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 1.2 g (65.78%) of tert-butyl N-[3-(l-[2-amino-l-[(2,4-dimethoxyphenyl)methyl]-lH-l,3-benzodiazol-5-yl]-N- methylformamido)propyl]carbamate as a brown solid. LC-MS (ES, m/z): 498 (M+H+).
Synthesis of 96.6
[00781] To a solution of l-[5-[5-(ethoxycarbonyl)-l-ethyl-3-methyl-lH-pyrazol-4-yl]pentyl]- 3-methyl-lH-pyrazole-5-carboxylic acid, 96.5 (1.09 g, 2.894 mmol, 1.20 equiv) and tert-butyl N- [3-(l-[2-amino-l-[(2,4-dimethoxyphenyl)methyl]-lH-l,3-benzodiazol-5-yl]-N- methylformamido)propyl]carbamate (1.20 g, 2.412 mmol, 1.00 equiv)in l-methylpyrrolidin-2- one (15 mL) was added
HATU (1.38 g, 3.629 mmol, 1.50 equiv) and DIEA (1.56 g, 12.07 mmol, 5 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 140°C. The mixture was cooled to room temperature. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (31% ACN up to 42% in 10 min); Detector, UV 254/220 nm. This resulted in 1.1 g (53.28%) of ethyl 4-[5-[5-([5-[(3-[[(tert- butoxy)carbonyl]amino]propyl)(methyl)carbamoyl]-l-[(2,4-dimethoxyphenyl)methyl]-lH-l,3- benzodiazol-2-yl]carbamoyl)-3-methyl-lH-pyrazol-l-yl]pentyl]-l-ethyl-3-methyl-lH-pyrazole- 5-carboxylate as a brown solid. LC-MS (ES, m/z): 856 (M+H+).
Synthesis of 96.7
[00782] To a solution of ethyl 4-[5-(5-[[5-([3-[(tert- butoxycarbonyl)amino]propyl](methyl)carbamoyl)-l-[(2,4-dimethoxyphenyl)methyl]-l,3- benzodiazol-2-yl]carbamoyl]-3-methylpyrazol-l-yl)pentyl]-2-ethyl-5-methylpyrazole-3- carboxylate, 96.6 (1 g, 1.168 mmol, 1 equiv) in methanol (20 mL) and water (5 mL) was added lithium hydroxide (112.2 mg, 4.672 mmol, 4.00 equiv). The resulting mixture was stirred for 2h at 65°C. The mixture was cooled to RT and concentrated under vacuum. The residue was acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (28% ACN up to 39% in 10 min); Detector, UV 254/220 nm. This resulted in 600 mg (62%) of 4-[5-(5-[[5-([3-[(tert- butoxycarbonyl)amino]propyl](methyl)carbamoyl)-l-[(2,4-dimethoxyphenyl)methyl]-l,3- benzodiazol-2-yl]carbamoyl]-3-methylpyrazol-l-yl)pentyl]-2-ethyl-5-methylpyrazole-3- carboxylic acid as a light yellow solid. LC-MS (ES, m/z): 828 (M+FL).
Synthesis of 96.8
[00783] To a solution of 2-amino-l-[(2,4-dimethoxyphenyl)methyl]-l,3-benzodiazole-5- carboxamide (177.37 mg, 0.543 mmol, 1 equiv) and 4-[5-(5-[[5-([3-[(tert- butoxycarbonyl)amino]propyl](methyl)carbamoyl)-l-[(2,4-dimethoxyphenyl)methyl]-l,3- benzodiazol-2-yl]carbamoyl]-3-methylpyrazol-l-yl)pentyl]-2-ethyl-5-methylpyrazole-3- carboxylic acid, 96.7 (450.00 mg, 0.543 mmol, 1 equiv) in l-methylpyrrolidin-2-one (10 mL) was added HATU (309.98 mg, 0.815 mmol, 1.50 equiv) and DIEA (351.21 mg, 2.717 mmol, 5 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 140 °C. The mixture was cooled to room temperature. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (21% ACN up to 32% in 10 min); Detector, UV 254/220 nm. This resulted in 200 mg (32.38%) of tert-butyl N-(3- [l-[2-(2-[5-[5-([5-carbamoyl-l-[(2,4-dimethoxyphenyl)methyl]-l,3-benzodiazol-2- yl]carbamoyl)-l-ethyl-3-methylpyrazol-4-yl]pentyl]-5-methylpyrazole-3-amido)-l-[(2,4- dimethoxyphenyl)methyl]-l,3-benzodiazol-5-yl]-N-methylformamido]propyl)carbamate as a off- white solid. LC-MS (ES, m/z): 569 (M/2+H+).
Synthesis of 96.9
[00784] To a stirred solution of tert-butyl N-(3-[l-[2-(2-[5-[5-([5-carbamoyl-l-[(2,4- dimethoxyphenyl)methyl]-l,3-benzodiazol-2-yl]carbamoyl)-l-ethyl-3-methylpyrazol-4- yl]pentyl]-5-methylpyrazole-3-amido)-l-[(2,4-dimethoxyphenyl)methyl]-l,3-benzodiazol-5-yl]- N-methylformamido]propyl)carbamate, 96.8 (200 mg, 0.176 mmol, 1 equiv) in dichloromethane (10 mL) was added trifluoroacetic acid (3 mL). The resulting mixture was stirred overnight at room temperature. The mixture was concentrated under vacuum. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8; mobile phase, Water (0.1% FA) and ACN (11% ACN up to 31% in 16 min); Detector, UV 254/220 nm. This resulted in 50 mg (38.65%) of N-(3-aminopropyl)-2-[2-(5-[5-[(5-carbamoyl-lH-l,3-benzodiazol-2- yl)carbamoyl]-l-ethyl-3-methylpyrazol-4-yl]pentyl)-5-methylpyrazole-3-amido]-N-methyl-lH- l,3-benzodiazole-5-carboxamide as a light yellow oil. LC-MS (ES, m/z): 736 (M+H+).
Synthesis of 1-25
[00785] To a solution of [[2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindol-4-yl]oxy]acetic acid (22.58 mg, 0.068 mmol, 1 equiv) and N-(3-aminopropyl)-2-[2-(5-[5-[(5-carbamoyl-lH-l,3- benzodiazol-2-yl)carbamoyl]-l-ethyl-3-methylpyrazol-4-yl]pentyl)-5-methylpyrazole-3-amido]- N-methyl-lH-l,3-benzodiazole-5-carboxamide, 96.9 (50 mg, 0.068 mmol, 1 equiv) in dimethyl formamide (5 mL) was added HATU (38.75 mg, 0.102 mmol, 1.5 equiv) and DIEA (43.91 mg, 0.340 mmol, 5 equiv). The resulting mixture was stirred for 2 h at room temperature. The mixture was concentrated under vacuum. The residue was purified by reverse flash chromatography with the following conditions: column, C18; mobile phase, Water (0.1% FA) and ACN (28% ACN up to 38% in 12 min); Detector, UV 254/220 nm. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire C18 OBD Prep Column, 19*250mm, 5um; mobile phase, Water (0.1%FA) and ACN (25% ACN up to 45% in 7 min); UV detection at 254/220 nm. This resulted in 4.9 mg (6.87%) of 2-[4-(5-[5-[(5-[[3-(2-[[2-(2,6-dioxopiperidin-3-yl)-l,3- dioxoisoindol-4-yl]oxy]acetamido)propyl](methyl)carbamoyl]-lH-l,3-benzodiazol-2- yl)carbamoyl]-3-methylpyrazol-l-yl]pentyl)-2-ethyl-5-methylpyrazole-3-amido]-lH-l,3- benzodiazole-5-carboxamideas a white solid. LC-MS (ES, m/z): 1050 (M+H+); 1H NMR: 1H NMR (400 MHz, DMSO-d6) d 12.51 (br s, 2H), 11.12 (s, 1H), 8.24 (s, 1H), 8.18-7.58 (m, 5H), 7.53-6.85 (m, 7H), 5.19-5.11 (m, 1H), 4.94-4.33 (m, 6H), 3.06-2.85 (m, 8H), 2.82-2.55 (m, 4H), 2.25-1.99 (m, 7H), 1.87-1.45 (m, 5H), 1.42-0.82 (m, 7H).
Example 97: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-N-(2-(2-(2-methoxyethoxy)ethoxy)ethyl)-lH-benzo[d]imidazole-5- carboxamide, 1-23
Figure imgf000440_0001
[00786] To a solution of l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5- carboxylic acid, 43.1 (100.00 mg, 0.148 mmol, 1.00 equiv) and l-[2-(2-aminoethoxy)ethoxy]-2- methoxyethane (72.25 mg, 0.443 mmol, 3.00 equiv) in dimethyl formamide (5 mL) was added DIEA (95.35 mg, 0.738 mmol, 5 equiv) and HATU (72.93 mg, 0.192 mmol, 1.3 equiv). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Prep OBD C18 Column, 30* 150mm, 5um; mobile phase, Water ( 10%NH4HCO3 +0.1 %NH3.H20) and ACN (22% ACN up to 27% in 7 min); UV detection at 254/220 nm. This resulted in 30 mg (24.66%) of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-N-(2-(2-(2-methoxyethoxy)ethoxy)ethyl)-lH-benzo[d]imidazole-5- carboxamide as a white solid. LC-MS (ES, m/z): 823 (M+H+); 1H NMR (400 MHz, DMSO-d6) d 12.75 (br s, 2H), 8.45 (t, 1H), 8.02-7.91 (m, 3H), 7.72-7.66 (m, 2H), 7.43 (dd, 2H), 7.31 (s, 1H), 6.55 (s, 2H), 5.98-5.92 (m, 2H), 4.89-4.81 (m, 4H), 4.54 (q, 4H), 3.59-3.50 (m, 8H), 3.47-3.38 (m, 4H), 3.21 (s, 3H), 2.13 (s, 6H), 1.29 (t, 6H).
Example 98: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-N-(3-hydroxypropyl)-lH-benzo[d]imidazole-5-carboxamide, 1-22
Figure imgf000441_0001
[00787] To a solution of l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5- carboxylic acid, 43.1 (100 mg, 0.148 mmol, 1.00 equiv) and 3-aminopropan-l-ol (33.25 mg, 0.443 mmol, 3 equiv) in dimethyl formamide (6 mL) was added HATU (72.93 mg, 0.192 mmol, 1.3 equiv) and DIEA (95.35 mg, 0.738 mmol, 5 equiv). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XSelect CSH Prep Cl 8 OBD Column, 5um, 19* 150mm; mobile phase: Water (0.1% FA) and ACN (22% ACN up to 34% in 7 min); UV detection at 254/220 nm. This resulted in 43.9 mg (40.46%) of (E)-l-(4-(5-carbamoyl- 2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-2-(l- ethyl-3-methyl-lH-pyrazole-5-carboxamido)-N-(3-hydroxypropyl)-lH-benzo[d]imidazole-5- carboxamide as an off-white solid. LC-MS (ES, m/z): 735 (M+H+); 'H NMR 400 MHz, DMSO- d6) d 12.79 (br s, 2H), 8.41 (t, 1H), 8.01-7.95 (m, 3H), 7.73-7.65 (m, 2H), 7.45-7.42 (m, 2H), 7.38- 7.25 (m, 1H), 6.56 (s, 2H), 5.96-5.89 (m, 2H), 4.94-4.81 (m, 4H), 4.62-4.47 (m, 5H), 3.56-3.44 (m, 4H), 2.33 (s, 6H), 1.74-1.65 (m, 2H), 1.68 (t, 6H).
Example 99: Synthesis of N,N'-((cyclopropane-l,2-diylbis(ethane-2,l-diyl))bis(5- carbamoyl-lH-benzo [d] imidazole-1, 2-diyl))bis(4-ethyl-2-methyloxazole-5-carboxamide), I- 21
Figure imgf000442_0001
Synthesis of 99.1 [00788] To a stirred solution of 2,2'-(cyclopropane-l,2-diyl)diethanamine (3 g crude, 11.29 mmol, 1 equiv) in dimethyl formamide (40 mL) was added potassium carbonate (4.67 g, 33.87 mmol, 3 equiv) and 4-fluoro-3-nitrobenzamide (2.08 g, 11.29 mmol, 1 equiv). The resulting solution was stirred for 4h at 70°C. The reaction was cooled to RT and diluted with water. The precipitated solids were collected by filtration. This resulted in 3.5 g (68%) of 4,4'- (2,2'-(cyclopropane-l,2-diyl)bis(ethane-2, l-diyl))bis(azanediyl)bis(3-nitrobenzamide) as a yellow solid. LC-MS (ES, m/z): 457 (M+H+);
Synthesis of 99.2
[00789] To a stirred solution of 4,4'-(2,2'-(cyclopropane-l,2-diyl)bis(ethane-2, l- diyl))bis(azanediyl)bis(3-nitrobenzamide), 99.1 (3.5 g, 7.68 mmol, 1.00 equiv) in acetic acid (40 mL) was zincpowder (4.99 g, 76.8 mmol, 10.00 equiv). The resulting solution was stirred overnight at RT. The solids were filtered out. The resulting mixture was concentrated under reduced pressure. This resulted in 3.6 g (crude) of 4,4'-(2,2'-(cyclopropane-l,2- diyl)bis(ethane-2,l-diyl))bis(azanediyl)bis(3-aminobenzamide) as a brown solid. LC-MS (ES, m/z): 397 (M+H+);
Synthesis of 99.3
[00790] To a stirred solution of 4,4'-(2,2'-(cyclopropane-l,2-diyl)bis(ethane-2,l- diyl))bis(azanediyl)bis(3-aminobenzamide), 99.2 (3.6 g crude, 7.68 mmol, 1.00 equiv) in methanol (30 mL) was added cyanogen bromide (3.225 g, 30.72 mmol, 4 equiv). The resulting solution was stirred overnight at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (21% ACN up to 43% in 10 min); Detector, UV 254/220 nm. This resulted in 1 g (29.15%) of l,T-(2,2'-(cyclopropane-l,2-diyl)bis(ethane-2, l-diyl))bis(2- amino-lH-benzo[d]imidazole-5-carboxamide) as a brown oil. LC-MS (ES, m/z): 447 (M+H+); Synthesis of 1-21
[00791] To a stirred mixture of 2-amino-l-(2-[2-[2-(2-amino-5-carbamoyl-l,3-benzodiazol-l- yl)ethyl]cyclopropyl]ethyl)-l,3-benzodiazole-5-carboxamide, 99.3 (50 mg, 0.112 mmol, 1.00 equiv) and 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (34.75 mg, 0.224 mmol, 2 equiv) in 1- methylpyrrolidin-2-one (4 mL) was added HATU (93.63 mg, 0.2464 mmol, 2.20 equiv) and DIEA (72.24 mg, 0.56 mmol, 5.00 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 140oC. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Shield RP18 OBD Column 19*250mm, lOum; mobile phase, Water (0.1% FA) and ACN (35% ACN up to 37% in 7 min); Detector, UV 254/210 nm. This resulted in 9 mg (11.16%) of l-[2-(2-[2-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3- benzodiazol-l-yl]ethyl]cyclopropyl)ethyl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3- benzodiazole-5-carboxamide as a purple solid. LC-MS (ES, m/z): 721 (M+H+); 1H NMR: (400 MHz, DMSO-d6) 512.68 (br s, 2H), 8.01-7.93 (m, 4H), 7.76 (dd, 2H), 7.46 (d, 2H), 7.31 (s, 2H), 4.19 (t, 4H), 2.94 (q, 4H), 2.43 (s, 6H), 1.78-1.51 (m, 4H), 1.12 (t, 6H), 0.56-0.45 (m, 2H), 0.18- 0.10 (m, 2H).
Example 100: Synthesis of(E)-N-(5-carbamoyl-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-7-(morpholinomethyl)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-methoxy- lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-110
Boc Boc Boc
Figure imgf000444_0001
Boc
Figure imgf000445_0001
Synthesis of 100.1
[00792] To a stirred solution/mixture of tert-butyl A'-[(2/'/)-4-[(4-carbamoyl-2-methoxy-6- nitrophenyl)amino]but-2-en-l-yl]carbamate (6.00 g, 15.773 mmol, 1.00 eq. ) in acetic acid was added Zn (20.63 g, 315.400 mmol, 20.00 eq.) in portions at ice bath. The resulting mixture was stirred for 1.5 h at room temperature. The resulting mixture was filtered; the filter cake was washed with MeOH (3x10 mL). The filtrate was concentrated under reduced pressure. This resulted in tert- butyl ;V-[(2/',)-4-[(2-amino-4-carbamoyl-6-methoxyphenyl)atnino]but-2-en-l -yl]carbamate (3 g, 54.28%) as a light yellow solid. LC-MS (ES, m/z): 351 (M+H+).
Synthesis of 100.2
[00793] A solution of tert-butyl /V-[(2//)-4-[(2-amino-4-carbamoyl-6- methoxyphenyl)amino]but-2-en-l-yl] carbamate, 100.1 (3.00 g, 8.561 mmol, 1.00 eq.) and BrCN (1.09 g, 10.273 mmol, 1.20 eq.) in MeOH (30.00 mL) was stirred for 2 h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was re-crystallized from ethyl acetate / MeOH (10: 1, 50 mL) to afford tert-butyl /V-[(2£)-4-(2-amino-5-carbamoyl-7- methoxy-1, 3-benzodiazol-l-yl)but-2-en-l-yl]carbamate (3.1 g, 96.45%) as a yellow solid. LC- MS (ES, m/z): 376 (M+H+).
Synthesis of 100.3
[00794] A solution of 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (1.53 g, 9.861 mmol, 1.20 eq.) in DMF was treated with DIEA (3.19 g, 24.692 mmol, 3 eq.) and HATU (6.26 g, 16.464 mmol, 2.00 eq.) for 30 min at room temperature followed by the addition of tert-butyl N-[(2E)-A- (2-amino-5-carbamoyl-7-methoxy-l, 3-benzodiazol-l-yl)but-2-en-l-yl]carbamate, 100.2 (3.09 g, 8.231 mmol, 1.00 eq.) at room temperature. The resulting mixture was stirred for overnight at 85 degree C. The mixture was cooled to room temperature. The resulting mixture was diluted with ethyl acetate (150 mL). The combined organic layers were washed with brine (3x50 mL), dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (3: 1) to afford tert-butyl Ar-[(2A)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l, 3-oxazole-5-amido)-7-methoxy-l,3- benzodiazol-l-yl]but-2-en-l-yl]carbamate (2.1 g, 49.78%) as a light yellow solid. LC-MS (ES, m/z): 513 (M+H+).
Synthesis of 100.4
[00795] To a stirred solution of tert-butyl /V-[(2£)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l, 3- oxazole-5-amido)-7-methoxy-l, 3-benzodiazol-l-yl]but-2-en-l-yl]carbamate, 100.3 (2.10 g, 4.097 mmol, 1.00 eq.) in MeOH (21.00 mL) was added HC1 in 1, 4-dioxane (11.00 mL) dropwise at 0 0 C. The resulting mixture was stirred overnight at room temperature. The reaction was monitored by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product was re-crystallized from MeOH/EtOAc (1 : 10, 60 mL) to afford 1 -[(2//)-4-aminobut-2-en- l-yl]-2-(4-ethyl-2-methyl-l, 3-oxazole-5-amido)-7-methoxy-l, 3-benzodiazole-5-carboxamide (1.7 g, 98.60%) as a yellow solid. LC-MS (ES, m/z): 412 (M+H+).
Synthesis of 100.5
[00796] To a stirred solution of 1 -[(2//)-4-aminobut-2-en-l -yl]-2-(4-ethyl-2-methyl-l , 3- oxazole-5-amido)-7-methoxy-l, 3-benzodiazole-5-carboxamide, 100.4 (1.80 g, 4.364 mmol, 1.00 eq.) and DIEA (1.13 g, 8.728 mmol, 2 eq.) in DMSO (20.00 mL) were added 4-[(2-fluoro-3- nitrophenyl)methyl]morpholine (1.10 g, 4.579 mmol, 1.05 eq.) at room temperature. The resulting mixture was stirred for overnight at 60 degree C. The mixture was cooled to room temperature. The product was precipitated by the addition of water. The precipitated solids were collected by filtration and washed with water (3x10 mL). The crude product was re-crystallized from PE/EtOAc (8: 1 mL) to afford 2-(4-ethyl-2-methyl-l, 3-oxazole-5-amido)-7-methoxy-l-[(2£)-4-[[2- (morpholin-4-ylmethyl)-6-nitrophenyl]amino]but-2-en-l-yl]-l, 3-benzodiazole-5- carboxamide(2.1g, 83.30%) as a light yellow solid. LC-MS (ES, m/z): 633 (M+H+).
Synthesis of 100.6
[00797] To a stirred mixture of 2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-methoxy-l-[(2£)- 4-[[2-(morpholin-4-ylmethyl)-6-nitrophenyl]amino]but-2-en-l-yl]-l,3-benzodiazole-5- carboxamide, 100.5 (50.00 mg, 0.079 mmol, 1.00 eq.) in MeOH (1.00 mL) and ammonia (0.10 mL, 3.523 mmol, 44.58 eq.) was added NaiS Cfi (68.80 mg, 0.395 mmol, 5.00 eq.) in portions at degree C. The resulting mixture was stirred for additional 1 h at room temperature. The resulting mixture was filtered; the filter cake was washed with MeOH (3x20 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, Cix silica gel; mobile phase, MeOH in water, 5% to 65% gradient in 30 min; detector, UV 254 nm. This resulted in l -[(2//)-4-[[2-amino-6-(morpholin-4- ylmethyl)phenyl]amino]but-2-en-l-yl]-2-(4-ethyl -2 -methyl-1, 3-oxazole-5-amido)-7-methoxy-l, 3-benzodiazole-5-carboxamide (680mg, 37.57%) as a white solid. LC-MS (ES, m/z): 603 (M+H+). Synthesis of 100.7 [00798] To a stirred mixture of l-[(2E)-4-[[2-amino-6-(morpholin-4- ylmethyl)phenyl]amino]but-2-en-l-yl]-2-(4-ethyl -2 -methyl-1, 3-oxazole-5-amido)-7-methoxy- l,3-benzodiazole-5-carboxamide, 100.6 (665.00 mg, 1.103 mmol, 1.00 eq.) in MeOH (8.00 mL) was added BrCN (140.24 mg, 1.324 mmol, 1.20 eq.) at room temperature. The resulting mixture was stirred overnight at room temperature. The resulting mixture was concentrated under vacuum. The crude product was re-crystallized from EtOAc/MeOH (8: 1, 50 mL) to afford l-[(2£)-4-[2- amino-7-(morpholin-4-ylmethyl)-l, 3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l, 3- oxazole-5-amido)-7-methoxy-l, 3-benzodiazole-5-carboxamide(214 mg, 30.90%) as a yellow solid. LC-MS (ES, m/z): 628 (M+H+).
Synthesis of 1-110
[00799] A solution of 2-ethyl-5-methylpyrazole-3 -carboxylic acid (19.16 mg, 0.124 mmol, 1.20 eq.) in DMF (3.00 mL) was added DIEA (40.15 mg, 0.311 mmol, 3.00 eq.) and HATU (78.75 mg, 0.207 mmol, 2.00 eq.) for 30 min at room temperature followed by the addition of l-[(2£)-4-[2- amino-7-(morpholin-4-ylmethyl)-l, 3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l, 3- oxazole-5-amido)-7-methoxy-l, 3-benzodiazole-5-carboxamide, 100.7 (65.00 mg, 0.104 mmol, 1.00 eq.) at room temperature. The resulting mixture was stirred for overnight at 85 degree C. The mixture was allowed to cool down to room temperature. The resulting mixture was diluted with ethyl acetate (20mL). The combined organic layers were washed with brine (3x15 mL), dried over anhydrous NaiSCL. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeOH in water, 10% to 50% gradient in 10 min; detector, UV 254 nm. The crude product (15 mg) was purified by Prep-HPLC with the following conditions (Column: XBridge Prep OBD Cis Column, 30x 150mm 5um; Mobile Phase A: Water(0.1 % FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 10 B to 20 B in 6 min; 254; 220 nm) to afford 2- (4-ethyl-2-methyl-l, 3-oxazole-5-amido)-l-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-7- (morpholin-4-ylmethyl)-l, 3-benzodiazol-l-yl]but-2-en-l-yl]-7-methoxy-l, 3-benzodiazole-5- carboxamide(7mg, 6.32%) as a white solid. LC-MS (ES, m/z): 764 (M+H+). 'H-NMR: (400 MHz, DMSO -d6) d 12.78 (s, 1H), 12.71 (s, 1H), 7.96 (s, 1H), 7.64 (d, J = 1.3 Hz, 1H), 7.50 (d, J = 7.9 Hz, 1H), 7.34 (d, J = 1.3 Hz, 1H), 7.12 (t, J = 7.8 Hz, 1H), 6.99 (d, J = 7.4 Hz, 1H), 6.50 (s, 1H), 5.77 (d, J = 16.1 Hz, 1H), 5.39 (d, J = 16.3 Hz, 1H), 5.17 (s, 2H), 5.05 (s, 1H), 4.86 (s, 2H), 4.51 (q, J = 7.0 Hz, 2H), 4.31 (s, OH), 3.89 (s, 1H), 3.81 (s, 3H), 3.62 (s, 1H), 3.23 (s, 1H), 2.79 (q, J = 7.5 Hz, 2H), 2.39 (s, 3H), 2.18 (s, 3H), 2.12 (s, 3H), 1.26 (t, J = 7.1 Hz, 3H), 1.00 (t, J = 7.5 Hz, 3H).
Example 101: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-N-(2-(2-hydroxyethoxy)ethyl)-lH-benzo[d]imidazole-5-carboxamide, 1-20
Figure imgf000449_0001
[00800] To a solution of l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5- carboxylic acid, 43.1 (100 mg, 0.148 mmol, 1.00 equiv) and 2-(2-aminoethoxy)ethanol (46.54 mg, 0.443 mmol, 3 equiv) in dimethyl formamide (5 mL) was added DIEA (95.35 mg, 0.738 mmol, 5 equiv) and HATU (72.93 mg, 0.192 mmol, 1.3 equiv). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep C18 column 30* 150mm, 5um; mobile phase, Water (0.1% FA) and ACN (27% ACN up to 30% in 7 min); UV detection at 254/220 nm. This resulted in 27.4 mg (24.65%) of (E)-l-(4-(5-carbamoyl-2-(l-ethyl- 3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-2-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-N-(2-(2-hydroxyethoxy)ethyl)-lH-benzo[d]imidazole-5- carboxamide as a white solid. LC-MS(ES, m/z): 765 (M+H+); 1H NMR (4300 MHz, DMSO-t/s) d 12.79 (br s, 2H), 8.45 (t, 1H), 8.03-7.92 (m, 3H), 7.76-7.65 (m, 2H), 7.50-7.30 (m, 3H), 6.56 (s, 2H), 5.98-5.91 (m, 2H), 4.90-4.82 (m, 4H), 4.68-4.51 (m, 5H), 3.59-3.41 (m, 8H), 2.13 (s, 6H), 1.28 (t, 6H). Example 102: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-N-(2-(2-(2-hydroxyethoxy)ethoxy)ethyl)-lH-benzo[d]imidazole-5- carboxamide, 1-19
Figure imgf000450_0001
[00801] To a solution of l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5- carboxylic acid, 43.1 (100.00 mg, 0.148 mmol, 1.00 equiv) and 2-(2-(2- aminoethoxy)ethoxy)ethanol (66.16 mg, 0.443 mmol, 3 equiv) in dimethyl formamide (5 mL) was added DIEA (95.35 mg, 0.738 mmol, 5 equiv) and HATU (72.93 mg, 0.192 mmol, 1.3 equiv). The resulting mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 OBD column 30* 150mm, 5um; mobile phase, Water (0.1% FA) and ACN (23% ACN up to 37% in 7 min); UV detection at 254/220 nm. This resulted in 35.4 mg (29.6%) of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazol-l-yl)but-2-enyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-N-(2-(2- (2-hydroxyethoxy)ethoxy)ethyl)-lH-benzo[d]imidazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 809 (M+H+); ¾ NMR (400 MHz, DMSO -d6) d 12.80 (br s, 2H), 8.47 (t, 1H), 8.02-7.93 (m, 3H), 7.77-7.68 (m, 2H), 7.48-7.30 (m, 3H), 6.56 (s, 2H), 5.98-5.92 (m, 2H), 4.89-4.81 (m, 4H), 4.68-4.51 (m, 5H), 3.61-3.41 (m, 12H), 2.13 (s, 6H), 1.29 (t, 6H).
Example 103: Synthesis of N,N'-((propane-l,3-diylbis(oxy))bis(l-allyl-5-carbamoyl-lH- benzo [d] imidazole-7, 2-diyl))bis(4-ethyl-2-methyloxazole-5-carboxamide), 1-18
Figure imgf000451_0001
[00802] To a solution of 5,5'-(propane-l,3-diylbis(oxy))bis(4-chloro-3-nitrobenzamide), 70.3 (2.5 g, 5.3 mmol, 1 equiv) in 1 -methyl pyrrolidin-2-one (20 mL) was added prop-2-en-l -amine (3.02 g, 53 mmol, 10 equiv). The final reaction mixture was irradiated with microwave radiation for 1 hour at 140°C. The mixture was cooled to r.t and diluted with water. The precipitated solids were collected by filtration. This resulted in 2 g (73.42%) of 5,5'-(propane- l,3-diylbis(oxy))bis(4-(allylamino)-3-nitrobenzamide) as an orange solid. LC-MS (ES, m/z) 515 (M+H+).
Synthesis of 103.2
[00803] To a solution of 5,5'-(propane-l,3-diylbis(oxy))bis(4-(allylamino)-3-nitrobenzamide), 103.1 (620 mg, 1.2 mmol, 1 equiv) in concentrated hydrochloric acid (10 mL) was added tin( II )chloridedihydrate (2.71 g, 12 mmol, 10 equiv). The resulting mixture was stirred at r.t overnight. The mixture was diluted with water. The pH value of the solution was adjusted to 9 with sat. sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. This resulted in 320 mg (58.74%) of 5,5'-(propane-l,3- diylbis(oxy))bis(4-(allylamino)-3-aminobenzamide) as a yellow solid. LC-MS (ES, m/z): 455 (M+H+);
Synthesis of 103.3
[00804] To a solution of 5,5'-(propane-l,3-diylbis(oxy))bis(4-(allylamino)-3- aminobenzamide), 103.2 (300 mg, 0.66 mmol, 1 equiv) in methanol (10 mL) was added cyanogen bromide (346.5 mg, 3.3 mmol, 5 equiv). The resulting mixture was stirred at r.t. overnight. The resulting mixture was concentrated under vacuum. This resulted in 390 mg (crude) of 7,7'- (propane-l,3-diylbis(oxy))bis(l-allyl-2-amino-lH-benzo[d]imidazole-5-carboxamide) as a brown solid. LC-MS (ES, m/z): 505 (M+H+);
Synthesis of 1-18
[00805] To a solution of 2-amino-7-(3-[[2-amino-6-carbamoyl-3-(prop-2-en-l-yl)-l,3- benzodiazol-4-yl]oxy]propoxy)-l-(prop-2-en-l-yl)-l,3-benzodiazole-5-carboxamide, 103.3 (50 mg, 0.1 mmol, 1.00 equiv) and 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (31 mg, 0.2 mmol, 2 equiv) in l-methylpyrrolidin-2-one (3.00 mL) and was added DIEA (129 mg, 1 mmol, 10.00 equiv) and HATU (76 mg, 0.2 mmol, 2 equiv). The resulting mixture was irradiated with microwave radiation at 140oC for lh. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Prep OBD C18 Column, 30* 150mm, 5um; mobile phase, Water (10%NH4HC03+0.1%NH3.H20) and ACN (25% ACN up to 40% in 7 min); Detector, UV 254/220 nm. This resulted in 10.1 mg (12.98%) of 7-(3-[[6-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-3-(prop-2-en-l-yl)- l,3-benzodiazol-4-yl]oxy]propoxy)-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l-(prop-2-en-l- yl)-l,3-benzodiazole-5-carboxamide as an off-white solid. LC-MS (ES, m/z): 779 (M+H+); 1H MR: (400 MHz, DMSO-d6) d 7.97 (s, 2H), 7.68 (s, 2H), 7.52-7.34 (m, 4H), 6.12-6.02 (m, 2H), 5.12-4.86 (m, 8H), 4.50-4.35 (m, 4H), 2.96 (q, 4H), 2.49-2.35 (m, 8H), 1.19 (t, 6H).
Example 104: Synthesis of (E)-N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)-l-(4-(5-carbamoyl-2- (l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2- (l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-17
Figure imgf000453_0001
[00806] To a solution of l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5- carboxylic acid, 43.1 (100 mg, 0.148 mmol, 1.00 equiv) and tert-butyl N-[2-[2-(2- aminoethoxy)ethoxy]ethyl]carbamate (109.92 mg, 0.443 mmol, 3 equiv) in dimethyl formamide (5 mL) was added HATU (72.93 mg, 0.192 mmol, 1.3 equiv) and DIEA (95.35 mg, 0.738 mmol, 5 equiv). The resulting mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The residue was purified by reverse flash with the following conditions: column, C18; mobile phase, Water (0.1% FA) and ACN (41% ACN up to 51% in 10 min); UV detection at 254/220 nm. This resulted in 95 mg (61.69%) of (E)-tert-butyl 2-(2-(2-(l- (4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l- yl)but-2-enyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5- carboxamido)ethoxy)ethoxy)ethylcarbamate as an off-white solid. LC-MS (ES, m/z): 909 (M+TE).
Synthesis of 1-17
[00807] To a solution of (E)-tert-butyl 2-(2-(2-(l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamido)ethoxy)ethoxy)ethylcarbamate, 104.1 (95.00 mg, 0.105 mmol, 1.00 equiv) in dichloromethane (10 ml) was added trifluoroacetic acid (2 mL). The resulting mixture was stirred for 2 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XSelect CSH Prep C18 OBD Column, 5um, 19*150mm; mobile phase: Water (0.1% FA) and ACN (10% ACN up to 32% in 7 min); UV detection at 254/220 nm. This resulted in 44.8 mg (50%) of (E)-N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)-l-(4-(5-carbamoyl- 2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-2-(l- ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide as an off- white solid. LC-MS (ES, m/z): 807 (M+H+); Ή NMR (400 MHz, DMSO-tE) d 8.50 (t, 1H), 8.02- 7.91 (m, 3H), 7.76-7.65 (m, 2H), 7.46-7.30 (m, 3H), 6.56 (s, 2H), 5.95-5.91 (m, 2H), 4.92-4.81 (m, 4H), 4.54 (q, 4H), 3.63-3.41 (m, 10H), 2.86 (t, 2H), 2.13 (s, 6H), 1.27 (t, 6H). Example 105: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-N-(2,5,8,ll,14-pentaoxahexadecan-16-yl)-lH-benzo[d]imidazole-5- carboxamide, 1-16
Figure imgf000455_0001
[00808] To a solution of l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5- carboxylic acid, 43.1 (100.00 mg, 0.148 mmol, 1.00 equiv) and 2,5,8,11, 14-pentaoxahexadecan- 16-amine (111.2 mg, 0.443 mmol, 3.00 equiv) in dimethyl formamide (5 mL) was added DIEA (95.35 mg, 0.738 mmol, 5 equiv) and HATU (72.93 mg, 0.192 mmol, 1.3 equiv). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. The cmde product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 Column, 30*150mm, 5um; mobile phase, Water (0.1%FA) and ACN (25% ACN up to 55% in 7 min); UV detection at 254/220 nm. This resulted in 46.6 mg (34.6%) of (E)-N- (2,5,8,l l, 14-pentaoxahexadecan-16-yl)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-enyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 933 (M+Na+); 1H NMR (400 MHz, DMSO-d6) d 12.79 (br s, 2H), 8.46 (t, 1H), 8.02-7.91 (m, 3H), 7.77-7.68 (m, 2H), 7.44 (dd, 2H), 7.32 (s, 1H), 6.56 (s, 2H), 5.98-5.92 (m, 2H), 4.89-4.81 (m, 4H), 4.54 (q, 4H), 3.62-3.35 (m, 20H), 3.22 (s, 3H), 2.15 (s, 6H), 1.29 (t, 6H).
Example 105: Synthesis of (E)-N-(5-carbamoyl-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-5-((3-hydroxypropyl)carbamoyl)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)- lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-15
Figure imgf000456_0001
106.2 1-15
Synthesis of 106.1
[00809] To a solution of 4-ethyl-2-methyloxazole-5-carboxylic acid (126 mg, 0.82 mmol, 1.2 equiv) and DIEA (442 mg, 3.4 mmol, 5.0 equiv) and HATU (388 mg, 1.02 mmol, 1.5 equiv) in 1- methylpyrrolidin-2-one (6 mL) was added methyl (E)-l-(4-(2-amino-5-carbamoyl-lH- benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxylate, 40.9 (380 mg, 0.68 mmol, 1.0 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 140°C.The resulting mixture was cooled to RT and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (28% ACN up to 51% in 10 min); Detector, UV 254/220 nm. This resulted in 300 mg (64%) of methyl (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5-carboxamido)-lH- benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxylate as a brown oil. LC-MS (ES, m/z): 693 (M+EC);
Synthesis of 106.2
[00810] To a solution of methyl (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxylate (300 mg, 0.43 mmol, 1.0 equiv) in methanol (10 mL) and water (2 mL) was added lithium hydroxide (52 mg, 2.17 mmol, 5.0 equiv). The resulting mixture was stirred for 4h at 70oC. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (23% ACN up to 33% in 9 min); Detector, UV 254/220 nm. This resulted in 90 mg (30.83%) of 4 (E)-
1-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-
2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5- carboxylic acid as a brown oil. LC-MS (ES, m/z): 679 (M+H+).
Synthesis of 1-15
[00811] To a solution of 4 (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5-carboxamido)- lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxylic acid, 106.2 (50 mg, 0.07 mmol, 1.0 equiv), DIEA (48 mg, 0.37 mmol, 5.0 equiv) and HATU (40 mg, 0.11 mmol, 1.5 equiv) in dimethyl formamide (4 mL) was added 3-aminopropan-l-ol (7 mg, 0.1 mmol, 1.3 equiv). The final reaction mixture was irradiated with microwave radiation at 120°C for lh. The mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (33% ACN up to 41% in 9 min); Detector, UV 254/220 nm. The crude product (30 mg) was purified by Prep-HPLC with the following conditions: Column, SunFire C18 OBD Prep Column, lOOA, 5um, 19* 150mm; mobile phase, Water (0.1% FA) and ACN(25% ACN up to 50% in 7 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 13.6 mg (26.4%) of (E)-N-(5-carbamoyl-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-5-((3- hydroxypropyl)carbamoyl)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-lH-benzo[d]imidazol-2- yl)-4-ethyl-2-methyloxazole-5-carboxamide as an off-white solid. LC-MS (ES, m/z): 736 (M+H+); 1H NMR (400 MHz, DMSO-t/e) 5 12.81 (s, 1H), 12.71 (s, 1H), 8.40 (t, 1H), 8.04-7.90 (m, 3H), 7.78-7.65 (m, 2H), 7.43 (d, 2H), 7.32 (s, 1H), 6.55 (s, 1H), 5.98-5.87 (m, 2H), 4.88-4.80 (m, 4H), 4.61-4.47 (m, 3H), 3.48 (t, 2H), 3.30-3.22 (m, 2H), 2.85 (q, 2H), 2.41 (s, 3H), 2.13 (s, 3H), 1.76- 1.65 (m, 2H), 1.28 (t, 3H), 1.05 (t, 3H).
Example 107: Synthesis of (E)-N-(5-carbamoyl-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-(3-hydroxypropoxy)-lH- benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-14
Figure imgf000458_0001
107.2 107.3
Figure imgf000459_0001
Synthesis of 107.1
[00812] To a stirred solution of tert-butyl N-[(2E)-4-[(2-nitrophenyl) amino] but-2-en-l-yl] carbamate (2.718 g, 8.843 mmol, 1 equiv) in dimethyl formamide (40 mL) was added tin( II )chloridedihydrate (9.977 g, 44.214 mmol, 5.00 equiv). The resulting mixture was stirred overnight at 30°C. The mixture was cooled to room temperature and diluted with water. The pH value of the solution was adjusted to 9 with sat. sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. This resulted in 2.2 g (89.8%) of tert-butyl N-[(2E)-4-[(2- aminophenyl) amino] but-2-en-l-yl] carbamate as a brown oil. LCMS: (ES, m/z): 278 (M+H+).
Synthesis of 107.2
[00813] To a stirred solution of tert-butyl N-[(2E)-4-[(2-aminophenyl) amino] but-2-en-l-yl] carbamate, 107.1 (2.2 g, 7.942 mmol, 1 equiv) in methanol (20 mL) was added cyanogen bromide (1.668 g, 15.884 mmol, 2 equiv). The resulting mixture was stirred at r.t overnight. The resulting mixture was concentrated under vacuum. This resulted in 2.4 g (crude) of tert-butyl N-[(2E)-4-(2- amino-lH-l,3-benzodiazol-l-yl) but-2-en-l-yl] carbamate as a light brown oil. LCMS: (ES, m/z): 303 (M+H+).
Synthesis of 107.3
[00814] To a stirred solution of (E)-tert-butyl 4-(2-amino-lH-benzo[d]imidazol-l-yl)but-2- enylcarbamate, 107.2 (1.51 g, 5 mmol, 1 equiv) and l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (924 mg, 6 mmol, 1.2 equiv) in N,N-dimethylformamide (20 mL) were added DIEA (3.225 g, 25 mmol, 5 equiv) and HATU (2.28 g, 6 mmol, 1.2 equiv). The resulting mixture was stirred for 2h at room temperature. The reaction was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 1.5 g (68.5%) of tert-butyl N-[(2E)-4-[2-(2- ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate as a pink solid. LC-MS (ES, m/z): 439 (M+H+).
Synthesis of 107.4
[00815] To a stirred solution of tert-butyl N-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)- l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate, 107.3 (1 g, 2.283 mmol, 1 equiv) in 1,2- dichloroethane (15 mL) was added hydrogen chloride in 1,4-dioxane (8 mL). The resulting mixture was stirred for 2h at room temperature. The precipitated solids were collected by filtration. This resulted in 800 mg (93.35%) of N-[l-[(2E)-4-aminobut-2-en-l-yl]-l,3-benzodiazol-2-yl]-2-ethyl- 5-methylpyrazole-3-carboxamide hydrochloride as a white solid. LC-MS (ES, m/z): 339 (M+H+).
Synthesis of 107.5
[00816] To a stirred solution of N-[l-[(2E)-4-aminobut-2-en-l-yl]-l,3-benzodiazol-2-yl]-2- ethyl-5-methylpyrazole-3-carboxamide, 107.4 (600 mg, 1.601 mmol, 1 equiv) and 3-[3-[(tert- butyldimethylsilyl)oxy]propoxy]-4-chloro-5-nitrobenzamide (684.74 mg, 1.761 mmol, 1.10 equiv) in n-butyl alcohol (20 mL) were added sodiumbicarbonate (403.37 mg, 4.802 mmol, 3.00 equiv) and DIEA (1.03 g, 8.003 mmol, 5.00 equiv). The resulting solution was stirred overnight at 120°C. The mixture was cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (10%NH4HC(T) and ACN (40% ACN up to 50% in 12 min); Detector, UV 254/220 nm. This resulted in 350 mg (31.7%) of N-[1-[(2E)- 4-[(2-[3-[(tert-butyldimethylsilyl)oxy]propoxy]-4-carbamoyl-6-nitrophenyl)amino]but-2-en-l- yl]-l,3-benzodiazol-2-yl]-2-ethyl-5-methylpyrazole-3-carboxamide as a light yellow solid. LC- MS (ES, m/z) 713 (M+Na+).
Synthesis of 107.6
[00817] To a stirred solution of N-[l-[(2E)-4-[(2-[3-[(tert-butyldimethylsilyl)oxy]propoxy]-4- carbamoyl-6-nitrophenyl)amino]but-2-en-l-yl]-l,3-benzodiazol-2-yl]-2-ethyl-5-methylpyrazole- 3 -carboxamide, 107.5 (350 mg, 0.49 mmol, 1 equiv) in acetic acid (10 mL) was added zinc (156.8 mg, 2.45 mmol, 5 equiv). The resulting mixture was stirred for 4 h at room temperature. The mixture was filtered. The filtrate was concentrated under reduced pressure. This resulted in 300 mg (crude) of N-[l-[(2E)-4-[[2-amino-4-carbamoyl-6-(3-hydroxypropoxy)phenyl]amino]but-2- en-l-yl]-l,3-benzodiazol-2-yl]-2-ethyl-5-methylpyrazole-3-carboxamide as a brown solid. LC- MS (ES, m/z) 547 (M+H+).
Synthesis of 107.7
[00818] To a stirred solution of N-[l-[(2E)-4-[[2-amino-4-carbamoyl-6-(3- hydroxypropoxy)phenyl]amino]but-2-en-l-yl]-l,3-benzodiazol-2-yl]-2-ethyl-5-methylpyrazole-
460
5UB5TITUTE SHEET (RULE 26) 3 -carboxamide, 107.6 (300 mg crude, 0.49 mmol, 1 equiv) in methanol (10 mL) was added cyanogen bromide (102.9 mg, 0.98 mmol, 2 equiv). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (10%NH HCO3) and ACN (20% ACN up to 40% in 10 min); Detector, UV 254/220 nm. This resulted in 100 mg (35.68%) of 2-amino-l-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-7-(3-hydroxypropoxy)-l,3-benzodiazole-5-carboxamide as a brown solid. LC-MS (ES, m/z): 572 (M+Ef).
Synthesis of 107.8
[00819] To a stirred solution of 2-amino-l-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)- l,3-benzodiazol-l-yl]but-2-en-l-yl]-7-(3-hydroxypropoxy)-l,3-benzodiazole-5-carboxamide, 107.7 (100 mg, 0.175 mmol, 1 equiv) and 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (54.28 mg, 0.35 mmol, 2 equiv) in N,N-dimethylformamide (5 mL) were added DIEA (113.05 mg, 0.875 mmol, 5 equiv) and HATU (133.03 mg, 0.35 mmol, 2 equiv). The resulting mixture was stirred for 2 h at room temperature. The reaction was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (W/oMEHCCh) and ACN (38% ACN up to 48% in 10 min); Detector, UV 254/220 nm. This resulted in 50 mg (33.79%) of 3-[[6-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-3-[(2E)-4-[2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazol-4-yl]oxy]propyl
4-ethyl-2-methyl-l,3-oxazole-5-carboxylate as an off-white solid. LC-MS (ES, m/z): 846 (M+H+). Synthesis of 1-14
[00820] To a stirred solution of 3-[[6-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-3- [(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3- benzodiazol-4-yl]oxy]propyl 4-ethyl-2-methyl-l,3-oxazole-5-carboxylate, 107.8 (50 mg, 0.059 mmol, 1 equiv) in methanol (10 mL) and water (2 mL) was added lithium hydroxide (5.66 mg, 0.236 mmol, 4.00 equiv). The resulting mixture was stirred for 2h at room temperature. The mixture was concentrated under vacuum. The residue was acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration. The crude product was purified by Prep-
461
5UB5TITUTE SHEET (RULE 26) HPLC with the following conditions: Column, XBridge Prep OBD C18 Column, 30* 150mm, 5um; mobile phase, Water (lOVo BHCC ) and ACN (20% ACN up to 40% in 7 min); UV detection at 254/220 nm. This resulted in 24.6 mg (58.72%) of 2-(4-ethyl-2-methyl-l,3-oxazole- 5-amido)-l-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]- 7-(3-hydroxypropoxy)-l,3-benzodiazole-5-carboxamide as an off-white solid. LC-MS (ESI): 709 (M+H+); ^NMR (400 MHz, DMSO-c¾) d 12.65 (br s, 2H), 7.95 (s, 1H), 7.64 (s, 1H), 7.49 (d, 1H), 7.36-7.34 (m, 3H), 7.21-7.13 (m, 2H), 6.53 (s, 1H), 5.95-5.92 (m, 1H), 5.76-5.75 (m, 1H), 4.91 (d, 2H), 4.81 (d, 2H), 4.57-4.50 (m, 3H), 4.11 (t, 2H), 3.52-3.50 (m, 2H), 2.83 (q, 2H), 2.40 (s, 3H), 2.12 (s, 3H), 1.80-1.77 (m, 2H), 1.26 (t, 3H), 1.03 (t, 3H).
Example 108: Synthesis of (E)-N-(l-(4-(5-((3-aminopropyl)carbamoyl)-2-(l-ethyl-3-methyl- lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-5-carbamoyl-lH- benzo [d] imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-13
Figure imgf000464_0001
1-13
Synthesis of 108.1
[00821] To a solution of 4 (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5-carboxamido)- lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxylic acid, 108.1 (265 mg, 0.39 mmol, 1.0 equiv), DIEA (252 mg, 1.95 mmol, 5.0 equiv) and HATU (223 mg, 0.59 mmol, 1.5 equiv) in dimethyl formamide (5 mL) was added tert-butyl (3-aminopropyl)carbamate (204 mg, 1.17 mmol, 3.0 equiv). The final reaction mixture was irradiated with microwave radiation at 120oC for lh. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (43% ACN up to 53% in 10 min); Detector, UV 254/220 nm. This resulted in 120 mg (36.85%) of tert-butyl (E)-(3-(l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5-carboxamido)- lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxamido)propyl)carbamate as a brown oil. LC-MS (ES, m/z): 418 (M/2+H+);
Synthesis of 1-13
[00822] To a solution of tert-butyl (E)-(3-(l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxamido)propyl)carbamate, 108.1 (120 mg, 0.144 mmol, 1.0 equiv) in dichloromethane (10 mL) was added trifluoroacetic acid (2 mL). The resulting mixture was stirred at RT for 2h. The mixture concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, SunFire C18 OBD Prep Column, lOOA, 5um, 19* 150mm; mobile phase, Water (0.1% FA) and CAN (15% ACN up to 38% in 7 min); Detector, ETV 254/220 nm. This resulted in 27.9 mg (26.36%) of (E)-N-(l-(4- (5-((3-aminopropyl)carbamoyl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazol-l-yl)but-2-en-l-yl)-5-carbamoyl-lH-benzo[d]imidazol-2-yl)-4-ethyl-2- methyloxazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 735 (M+H+); 1H NMR (400 MHz, DMSO-d6) d 8.66 (t, 1H), 8.05-7.89 (m, 3H), 7.78-7.69 (m, 2H), 7.43 (dd, 2H), 7.33 (s, 1H), 6.56 (s, 1H), 6.00-5.88 (m, 2H), 4.90-4.81 (m, 4H), 4.54 (q, 2H), 3.341-3.35 (m, 2H), 2.91-2.84 (m, 4H), 2.41 (s, 3H), 2.14 (s, 3H), 1.90-1.81 (m, 2H), 1.28 (t, 3H), 1.05 (t, 3H).
Example 109: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-(3-hydroxypropoxy)- lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-9
Figure imgf000465_0001
Figure imgf000466_0001
Synthesis of 109.1 [00823] To a stirred solution of methyl l-[(2E)-4-aminobut-2-en-l-yl]-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazole-5-carboxylate, 40.6 (1 g, 2.5 mmol, 1.20 equiv) in n- butyl alcohol (20 mL) and was added sodium bicarbonate (420 mg, 5 mmol, 2 equiv) and DIEA (1.60 g, 12.5 mmol, 5 equiv). After 10 min, was added 3-[3-[(tert- butyldimethylsilyl)oxy]propoxy]-4-chloro-5-nitrobenzamide (816 mg, 2.1 mmol, 1 equiv). The resulting solution was stirred overnight at 120°C. The mixture was cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (10%NH4HCC>3) and ACN (30% ACN up to 50% in 12 min); Detector, UY 254/220 nm. This resulted in 450 mg (24%) of methyl l-[(2E)-4-[(2-[3-[(tert- butyldimethylsilyl)oxy]propoxy]-4-carbamoyl-6-nitrophenyl)amino]but-2-en-l-yl]-2-(2 -ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazole-5-carboxylate as a dark yellow solid. LC-MS (ES, m/z): 749 (M+H+).
Synthesis of 109.2
[00824] To a stirred solution of methyl l-[(2E)-4-[(2-[3-[(tert- butyldimethylsilyl)oxy]propoxy]-4-formyl-6-nitrophenyl)amino]but-2-en-l-yl]-2-(2,5- dimethylpyrazole-3-amido)-l,3-benzodiazole-5-carboxylate, 109.1 (450 mg, 0.6 mmol, 1 equiv) in acetic acid (10 mL) was added zinc powder (240 mg, 3.61 mmol, 6 equiv). The resulting solution was stirred for lh at 50oC. The resulting mixture was cooled to RT. The solids were filtered out. The filtrate was concentrated under reduced pressure. This resulted in 500 mg (crude) of methyl l-[(2E)-4-[(2-amino-6-[3-[(tert-butyldimethylsilyl)oxy]propoxy]-4- formylphenyl)amino]but-2-en-l-yl]-2-(2,5-dimethylpyrazole-3-amido)-l,3-benzodiazole-5- carboxylate as a light yellow solid. LC-MS (ES, m/z): 719 (M+H+).
Synthesis of 109.3
[00825] To a stirred solution of methyl l-[(2E)-4-[(2-amino-6-[3-[(tert- butyldimethylsilyl)oxy]propoxy]-4-carbamoylphenyl)amino]but-2-en-l-yl]-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazole-5-carboxylate, 109.2 (500 mg crude, 0.6 mmol, 1 equiv) in methanol (10 mL) was cyanogen bromide (126 mg, 1.2 mmol, 2 equiv). The resulting solution was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (10% NH4HC03) and ACN (30% ACN up to 48% in 10 min); Detector, UV 254/220 nm. This resulted in 300 mg (79.36%) of methyl l-[(2E)-4-[2-amino-5-carbamoyl-7- (3-hydroxypropoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-
1.3-benzodiazole-5-carboxylate as a light yellow solid. LC-MS (ES, m/z): 630 (M+H+).
Synthesis of 109.4
[00826] To a stirred solution of methyl l-[(2E)-4-[2-amino-5-carbamoyl-7-(3- hydroxypropoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-
1.3-benzodiazole-5-carboxylate, 109.3 (300 mg, 0.477 mmol, 1 equiv) and 4-ethyl-2-methyl-l,3- oxazole-5-carboxylic acid (74 mg, 0.477 mmol, 1 equiv) in dimethyl formamide (5 mL) was added DIEA (310 mg, 2.385 mmol, 5.00 equiv) and HATU (218 mg, 0.572 mmol, 1.20 equiv). The final reaction mixture was irradiated with microwave radiation for 1 h at 120°C. The reaction was cooled to RT and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (31% ACN up to 46% in 10 min); UV detection at 254/210 nm. This resulted in 230 mg (62.9%) of methyl l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(3- hydroxypropoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-
1.3-benzodiazole-5-carboxylate as a yellow solid. LC-MS (ES, m/z): 767 (M+H+).
Synthesis of 109.5
[00827] To a stirred solution of methyl l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-(3-hydroxypropoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazole-5-carboxylate, 109.4 (230 mg, 0.3 mmol, 1 equiv) in methanol (10 mL) and water (2 mL) was added lithium hydroxide (28.8 mg, 1.2 mmol, 4 equiv). The resulting solution was stirred for 4h at 70oC. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration. This resulted in 120 mg (53.12%) of 1- [(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(3-hydroxypropoxy)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5- carboxylic acid as a yellow solid. LC-MS (ES, m/z): 753 (M+H+).
Synthesis of 1-9 [00828] To a stirred solution of 2-amino-l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-7-(3-hydroxypropoxy)-l,3- benzodiazole-5-carboxamide, 109.5 (90.00 mg, 0.120 mmol, 1.00 equiv) and ammonium chloride (732.5 mg, 0.6 mmol, 5 equiv) in dimethyl formamide (5 mL) was added DIEA (154.8 mg, 1.2 mmol, 10 equiv) and HATU (91.2 mg, 0.24 mmol, 2 equiv). The resulting solution was stirred for 4h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (26% ACN up to 38% in 10 min); Detector, UV 254/220 nm. The crude product (40 mg) was purified by Prep-HPLC with the following conditions: Column, XBridge Prep OBD C18 Column, 30*150mm, 5um; mobile phase, Water (10%NH4HCO3) and ACN (25% ACN up to 40% in 7 min); UV detection at 254/220 nm. This resulted in 14.9 mg (15.3%) of 1- [(2E)-4-[5-carbamoyl-2-(2-ethyl-5-methylpyrazole-3 -amido)- 1 ,3 -benzodiazol- 1 -yl]but-2-en- 1 - yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(3-hydroxypropoxy)-l,3-benzodiazole-5- carboxamide as a white solid. LC-MS (ES, m/z): 752 (M+H+); 1H NMR: (400 MHz, DMSO-d6) d 12.72 (br s, 2H), 8.03-7.92 (m, 3H), 7.70 (dd, 1H), 7.64 (s, 1H), 7.40 (d, 1H), 7.39-7.28 (m, 3H), 6.54 (s, 1H), 6.03-5.93 (m, 1H), 5.81-5.71 (m, 1H), 4.99-4.81 (m, 4H), 4.66-4.49 (m, 3H), 4.12 (t, 2H), 3.49 (t, 2H), 2.83 (q, 2H), 2.41(s, 3H), 2.12 (s, 3H), 1.84-1.74 (m, 2H), 1.27 (t, 3H), 1.03 (t, 3H).
Example 110: Synthesis of (E)-N-(5-carbamoyl-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-methoxy-lH-benzo[d]imidazol-2- yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-8
Figure imgf000470_0001
Synthesis of 110.1 [00829] Into a 50-mL 3-necked round-bottom flask, was placed 107.4 (2.50 g, 7.387 mmol, 1.00 equiv), DMSO (30.00 mL), DIEA (9.55 g, 73.874 mmol, 10.00 equiv) and 4-chloro-3- methoxy-5-nitrobenzamide (3.41 g, 14.788 mmol, 2.00 equiv). The resulting solution was stirred for 1 overnight at 100 degrees C under nitrogen atmosphere. The resulting mixture was washed with 1x30 ml of ThO. The resulting solution was extracted with 3x30 mL of ethyl acetate concentrated. The residue was applied onto a silica gel column with ethyl acetate/hexane (1 :5). This resulted in 1.7 g (43.21%) of N-[l-[(2E)-4-[(4-carbamoyl-2-methoxy-6- nitrophenyl)amino]but-2-en- 1 -yl] - 1.
Synthesis of 110.2
[00830] To a stirred solution of 110.1 (1.70 g, 3.192 mmol, 1.00 equiv) in HOAc (30.00 mL, 523.546 mmol) in a 100 mL round-bottom flask, Zn (4.18 g, 63.842 mmol, 20.00 equiv) was added in portions at 0 degrees C under air atmosphere. The resulting mixture was stirred for lh at room temperature under nitrogen atmosphere. Reaction liquid filtrated by MeOH. This resulted in N-[l- [(2E)-4-[(2-amino-4-carbamoyl-6-methoxyphenyl)amino]but-2-en-l-yl]-l,3-benzodiazol-2-yl]- 2-ethyl-5-methylpyrazole-3-carboxamide 1.2 g (92%) as a yellow solid.
Synthesis of 110.3
[00831] Into a 50 mL round-bottom flask were added 110.2 (1.20 g, 2.388 mmol, 1.00 equiv), MeOH (20.00 mL) and BrCN (379.36 mg, 3.582 mmol, 1.50 equiv) at room temperature. The resulting mixture was stirred for 1 h at room temperature under nitrogen atmosphere. The filtrate was concentrated under reduced pressure. The resulting mixture was concentrated under vacuum to afford 800 mg (80%) of 110.3 as an off-white solid.
Synthesis of 1-8
[00832] To a stirred solution of 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (58.82 mg, 0.379 mmol, 2.00 equiv) in DMF (2.00 mL) in an 8 mL vial, DIEA (73.49 mg, 0.569 mmol, 3.00 equiv) and HATU (144.14 mg, 0.379 mmol, 2.00 equiv) was added at room temperature. The mixture was stirred for 30 min at room temperature under nitrogen atmosphere. To the above mixture was placed 2-amino-l-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol- l-yl]but-2-en-l-yl]-7-methoxy-l,3-benzodiazole-5-carboxamide, 110.3 (100.00 mg, 0.190 mmol, 1.00 equiv) in DMF (lmL) dropwise portions at room temperature. The resulting mixture was stirred for additional lh at 85 degrees C under nitrogen atmosphere. The crude product (50 mg) was purified by Prep-HPLC with the following conditions (Column: SunFire Cl 8 OBD Prep Column, lOOA, 5 um, 19 mm X 250 mm; Mobile Phase A: water(0.1%FA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient:40 B to 70 B in 7 min; 254;220 nm; RTT5.82 min;) to afford 2- (4-ethyl-2-methyl-l,3-oxazole-5-amido)-l-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-7-methoxy-l,3-benzodiazole-5-carboxamide(14.3mg,95%) as a white solid. LC-MS: (ES, m/z) [M+H]+ 665.5
1H-NMR: (400 MHz, DMSO-d6) 512.66-12.71 (d, 2H), 57.95 (s, 1H), 57.64 (s, 1H), 57.48- 7.50 (d, 1H), 57.32-7.35 (m, 3H), 57.10-7.20 (m, 2H), 56.53 (s, 1H), 55.91-5.95 (m, 1H), 55.71- 5.75 (m, 1H), 54.80-4.90 (m, 4H), 54.50-4.55 (m, 2H), 53.78 (s, 3H), 52.81-2.86(m, 2H), 52.40 (s, 3H), 52.12(s, 3H), 51.23-1.28 (m, 3H), 51.02-1.06 (t, 3H).
Example 111: Synthesis of (E)-N-(5-carbamoyl-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-methyl-lH-benzo[d]imidazol-2- yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-7
Figure imgf000473_0001
1-7
111.4 111.5
Synthesis of 111.1 [00833] Into a 50-mL 3-necked round-bottom flask, was placed N-[l-[(2E)-4-aminobut-2-en- l-yl]-l,3-benzodiazol-2-yl]-2-ethyl-5-methylpyrazole-3-carboxamide, 107.4 (1.00 g, 2.955 mmol, 1.00 equiv), DMSO (10.00 mL), methyl 4-fluoro-3 -methyl-5 -nitrobenzoate (0.63 g, 2.955 mmol, 1.00 equiv) and DIEA (3.82 g, 29.55mmol, 10.00 equiv). The resulting solution was stirred for 1 overnight at room temperature under nitrogen atmosphere. The resulting mixture was washed with 1x10 mL of H2O. The resulting mixture was concentrated. The residue was applied onto a silica gel column with ethyl acetate/hexane (1 :5). This resulted in 1.2 g (76.40%) of methyl 4-[[(2E)-4- [2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]amino]-3-methyl-5- nitrobenzoate as a solid. LC-MS (ES, m/z): 531 [M+H]+.
Synthesis of 111.2
[00834] Into a 50-mL 3-necked round-bottom flask, was placed methyl 4-[[(2E)-4-[2-(2-ethyl- 5-methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]amino]-3-methyl-5- nitrobenzoate, 111.1 (1.19 g, 2.239 mmol, 1.00 equiv), HOAc (20.00 mL), Zn (2.93 g, 44.795 mmol, 20.01 equiv). The resulting solution was stirred for 2h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated. The resulting mixture was washed with 1x30 ml of MeOH. The solids were filtered out. This resulted in 0.8 g (71.25%) of methyl 3-amino-4- [[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]amino]-5- methylbenzoate as a solid. LC-MS (ES, m/z): 502 [M+H]+.
Synthesis of 111.3
[00835] Into a 50-mL 3-necked round-bottom flask, was placed methyl 3-amino-4-[[(2E)-4-[2- (2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]amino]-5- methylbenzoate (0.80 g, 1.595 mmol, 1.00 equiv), MeOH (15.00 mL), BrCN (253.40 mg, 2.392 mmol, 1.50 equiv). The resulting solution was stirred for 1 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated. The crude product was purified by Flash- Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN/H20=15% increasing to ACN/H2O=60% within 15 min. The product was obtained. This resulted in 0.6 g (71.44%) of methyl 2-amino-l-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)- l,3-benzodiazol-l-yl]but-2-en-l-yl]-7-methyl-l,3-benzodiazole-5-carboxylate as a solid. LC-MS (ES, m/z): 527 [M+H]+.
Synthesis of 111.4 [00836] Into a 50-mL 3-necked round-bottom flask, was placed 4-ethyl -2 -methyl- 1,3 -oxazole- 5-carboxylic acid (0.35 g, 2.256 mmol, 1.98 equiv), DMF (10.00 mL), DIEA (0.44 g, 3.418 mmol, 3.00 equiv), HATU (0.87 g, 2.288 mmol, 2.01 equiv), methyl 2-amino-l-[(2E)-4-[2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-7-methyl-l,3-benzodiazole-5- carboxylate (0.60 g, 1.139 mmol, 1.00 equiv). The resulting solution was stirred for 2 h at 85°C under nitrogen atmosphere. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN/H20=15% increasing to ACN/H2O=60% within 15min. The product was obtained. This resulted in 290 mg (38.35%) of methyl 2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l-[(2E)-4-[2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-7-methyl-l,3-benzodiazole-5- carboxylate as a solid. LC-MS (ES, m/z): 664 [M+H]+.
Synthesis of 111.5
[00837] Into a 50-mL 3-necked round-bottom flask, was placed methyl 2-(4-ethyl-2-methyl- l,3-oxazole-5-amido)-l-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-l- yl]but-2-en-l-yl]-7-methyl-l,3-benzodiazole-5-carboxylate (270.00 mg, 0.407 mmol, 1.00 equiv), NaOH (97.62 mg, 2.441 mmol, 6.00 equiv), THF(3 mL), MeOH(3 mL) and H20 (1 mL). The resulting solution was stirred for 2 h at room temperature under nitrogen atmosphere. The pH value of the solution was adjusted to 5 with HC1 (1 mol/L). The solids were collected by filtration. This resulted in 190 mg (71.89%) of 2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l-[(2E)-4-[2-(2-ethyl- 5-methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-7-methyl-l,3-benzodiazole-5- carboxylic acid as a solid. LC-MS (ES, m/z): 650 [M+H]+.
Synthesis of 1-7
[00838] Into a 50-mL 3-necked round-bottom flask, was placed 2-(4-ethyl -2-methyl- 1,3- oxazole-5-amido)-l-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2- en-l-yl]-7-methyl-l,3-benzodiazole-5-carboxylic acid (152.00 mg, 0.234 mmol, 1.00 equiv), DMF (5.00 mL), DIEA (90.71 mg, 0.702 mmol, 3.00 equiv), HATU (177.91 mg, 0.468 mmol, 2.00 equiv), ammonium chloride (62.57 mg, 1.170 mmol, 5.00 equiv). The resulting solution was stirred for 1 h at room temperature. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN/H20=15% increasing to ACN/H2O=60% within 15min. This resulted in 28.1 mg (18.51%) of 2-(4-ethyl-2-
474
5UB5TITUTE SHEET (RULE 26) methyl-l,3-oxazole-5-amido)-l-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-7-methyl-l,3-benzodiazole-5-carboxamide as a white solid. LC- MS: (ES, in r): [M+H]+ 649.1 , [M-H] 647.0; 1H-NMR: (300 MHz, DMSO -d6, ppm) 512.68- 12.75 (d, 2H), 57.85-7.86 (d, 2H), 57.47-7.52 (m, 2H), 57.37-7.40 (m, 2H), 57.13-7.32 (m, 2H), 56.52 (s, 1H), 55.91-5.96 (m, 1H), 55.49-5.57 (m, 1H), 54.80-4.97 (t, 4H), 54.50-4.57 (m, 2H), 52.74-2.86 (m, 2H), 52.52 (s, 3H), 52.47 (s, 3H), 52.11-2.13 (d, 3H), 51.25-1.29 (t, 3H), 50.92- 1.07 (m, 3H).
Example 112: Synthesis of (E)-N-(5-carbamoyl-l-(4-(2-(2,4-dimethyloxazole-5- carboxamido)-7-(morpholinomethyl)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-lH- benzo [d] imidazol-2-yl)-2, 4-dimethyl oxazole-5-carboxamide, 1-5
Figure imgf000476_0001
Figure imgf000477_0001
1-5
Synthesis of 112.1
[00839] Into a 1-L 3 -necked round-bottom flask, was placed 2-fluoro-l -methyl-3 -nitrobenzene (40.00 g, 257.852 mmol, 1.00 equiv), tetrachloromethane (500 mL), NBS (55.07 g, 309.422 mmol, 1.20 equiv), AP3N (2.12 g, 12.910 mmol, 0.05 equiv). The resulting solution was stirred for 1 overnight at 85 degrees C in an oil bath under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 :60). This resulted in 11.7 g (19.39%) of l-(bromomethyl)-2-fluoro-3- nitrobenzene as a yellow liquid.
Synthesis of 112.2
[00840] Into a 250-mL 3-necked round-bottom flask, was placed morpholine (6.53 g, 74.992 mmol, 1.50 equiv), DCM (120.00 mL), DIEA (12.92 g, 99.990 mmol, 2.00 equiv), 1- (bromomethyl)-2-fluoro-3-nitrobenzene, 112.1 (11.70 g, 49.995 mmol, 1.00 equiv). The resulting solution was stirred for 2 hours at room temperature under nitrogen atmosphere. The resulting solution was extracted with 3x200 mL of ethyl acetate dried over anhydrous sodium sulfate and concentrated under vacuum. The crude product was re-crystallized from EA/PE in the ratio of 1 : 100. This resulted in 9 g (74.93%) of 4-[(2-fluoro-3-nitrophenyl) methyl] morpholine as a yellow solid.
Synthesis of 112.3
[00841] To a stirred solution of 4-[(2-fluoro-3-nitrophenyl)methyl]morpholine, 112.2 (3.00 g, 12.488 mmol, 1.00 equiv) in DMSO (30 mL) in a 100-mL 3-necked round- bottom flask, tert- butyl N-[(2E)-4-aminobut-2-en-l-yl]carbamate hydrochloride(2.78 g, 12.488 mmol, 1.00 equiv) and DIEA(4.84 g, 37.463 mmol, 3.00 equiv) were added in portions at room temperature. The resulting mixture was stirred for overnight at 100 degrees C under nitrogen atmosphere. The resulting mixture was extracted with EtOAc (3x50 mL). The combined organic layers were washed with water (3x60 mL), dried over anhydrous Na SCL. After filtration, the filtrate was concentrated under reduced pressure to afford tert- butylN-[(2E)-4-([2-[(morpholin-4-yl)methyl]-6- nitrophenyl]amino)but-2-en-lyl]carbamate(2.56g,47.41%) as an off-white solid.
Synthesis of 112.4
[00842] Into a 250-mL 3-necked round-bottom flask, was placed tert-butyl N-[(2E)-4-[[2- (morpholin-4-ylmethyl)-6-nitrophenyl]amino]but-2-en-l-yl]carbamate, 112.3 (2.80 g, 6.888 mmol, 1.00 equiv), EtOH (60.00 mL), NaBLL (1303.04 mg, 34.442 mmol, 5.00 equiv), Cu(acac)2 (1.00 g, 3.820 mmol, 0.5 equiv) at 0°C. The resulting solution was stirred for 25 min at 25 degrees C under nitrogen atmosphere. The pH value of the solution was adjusted to 7 with ELCl. The solids were filtered out. The resulting mixture was concentrated under vacuum. This resulted in tert-butyl N-[(2E)-4-[[2-amino-6-(morpholin-4-ylmethyl) phenyl] amino] but-2-en-l-yl] carbamate (2 g, 77.12%) as a yellow solid.
Synthesis of 112.5
[00843] Into a 8-mL sealed tube, was placed tert-butyl N-[(2E)-4-[[2-amino-6-(morpholin-4- ylmethyl) phenyl] amino] but-2-en-l-yl] carbamate (2.00 g, 5.312 mmol, 1.00 equiv), MeOH (20 mL), BrCN (0.74 g, 6.986 mmol, 1.30 equiv). The resulting solution was stirred for 2h at 25 degrees C under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. This resulted in 112.5 (2.6 g, 95.24%) as a yellow solid.
Synthesis of 112.6
[00844] To a stirred mixture of tert-butyl N-[(2E)-4-[2-amino-7-(morpholin-4-ylmethyl)-l,3- benzodiazol-l-yl]but-2-en-l-yl]carbamate, 112.5 (2.56 g, 6.376 mmol, 1.00 equiv) and TFA (25.00 mL) in DCM (75.00 mL) in a 250 mL round-bottom flask in portions at room temperature under nitrogen atmosphere. The resulting solution was stirred for 2h at 25 degrees C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. This resulted in 112.6 (2.5 g, 98.17%) as a yellow solid.
Synthesis of 112.7
[00845] To a stirred solution of l-[(2E)-4-aminobut-2-en-l-yl]-7-(morpholin-4-ylmethyl)-l,3- benzodiazol-2-amine, 112.6 (2.46 g, 8.162 mmol, 1.00 equiv) and 4-fluoro-3-nitrobenzamide (2.25 g, 12.243 mmol, 1.50 equiv) in DMSO (25 mL) in a 100-mL round-bottom flask, DIEA (10.55 g, 81.621 mmol, 10.00 equiv) was added in portions at 60 degrees C under nitrogen atmosphere. The resulting solution was stirred for 4h at 60 degrees C under nitrogen atmosphere. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, MeOH in water, 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 4-[[(2E)-4-[2-amino-7-(morpholin-4-ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]amino]-3- nitrobenzamide (1.88 g, 49.48%) as a yellow solid.
Synthesis of 112.8
[00846] To a stirred solution of 4-[[(2E)-4-[2-amino-7-(morpholin-4-ylmethyl)-l,3- benzodiazol-l-yl]but-2-en-l-yl]amino]-3-nitrobenzamide, 112.7 (1.00 g, 2.148 mmol, 1.00 equiv) in HOAc (10.00 mL) in a 50 mL round-bottom flask, Zn (2.81 g, 42.963 mmol, 20.00 equiv) was added in portions at room temperature under nitrogen atmosphere. The resulting solution was stirred for 2h at 25 degrees C under nitrogen atmosphere. The resulting mixture was filtered. The filter cake was washed with MeOH (3x10 mL). The filtrate was concentrated under reduced pressure. This resulted in 3-amino-4-[[(2E)-4-[2-amino-7-(morpholin-4-ylmethyl)-l,3- benzodiazol-l-yl]but-2-en-l-yl]amino]benzamide (890 mg, 95.13%) as an off-white solid.
Synthesis of 112.9
[00847] To a stirred solution of 3-amino-4-[[(2E)-4-[2-amino-7-(morpholin-4-ylmethyl)-l,3- benzodiazol-l-yl]but-2-en-l-yl]amino]benzamide, 112.8 (890.00 mg, 2.043 mmol, 1.00 equiv) and BrCN (649.34 mg, 6.130 mmol, 3.00 equiv) in MeOH (10.00 mL) in portions at room temperature under nitrogen atmosphere. The resulting solution was stirred for 2h at 25 degrees C under nitrogen atmosphere. The resulting mixture was filtered. The filter cake was washed with MeOH (3x10 mL). The filtrate was concentrated under reduced pressure. This resulted in 2-amino- 1 -[(2E)-4-[2-amino-7-(morpholin-4-ylmethyl)- 1 ,3 -benzodiazol- 1 -yl]but-2-en- 1 -yl]- 1 ,3- benzodiazole-5-carboxamide (600 mg, 63.75%) as an off-white solid.
Synthesis of 1-5
[00848] Into a 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed 2-amino-l-[(2E)-4-[2-amino-7-(morpholin-4-ylmethyl)-l,3-benzodiazol-l-yl]but-2- en-l-yl]-l,3-benzodiazole-5-carboxamide, 112.9 (300.00 mg, 0.651 mmol, 1.00 equiv), NMP (5.00 mL), DIEA(505.14 mg, 3.908 mmol, 6.00 equiv), HATU(990.74 mg, 2.606 mmol, 4.00 equiv), dimethyl-1, 3-oxazole-5-carboxylic acid(551.58 mg, 3.908 mmol, 6.00 equiv). The resulting mixture was stirred for 1.5 h at 140 degrees C under microwave monitor. The resulting mixture was washed with 1x20 mL of water. The crude product (300 mg) was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, HCOOH (0.5%) in water, 10% to 50% gradient in 35 min; detector, UV 254 nm. The crude product (80 mg) was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): Column, XBridge Prep OBD C18 Column, 19*250mm, 5um; mobile phase, water (10 mmol/L NH4HCO3) and ACN (hold 30% Phase B in 8 min); Detector, UV 254nm. RT = 9.2 min) to afford 1-5 (10.2mg, 2.17%) as a white solid. LC-MS: (ES, m/z): [M+H]+:707.4, [M- H]':705.4; 1H-NMR: (300 MHz, DMSO-de, ppm ) 512.80-12.49 (brs, 2H), 57.97-7.93 (d, 2H), 57.73-57.69 (d, 1H), 57.51-7.41 (m, 2H), 57.32 (s, 1H), 57.14-6.99 (m, 2H), 55.94-5.89(d, 1H), 55.44-5.39 (t, 1H), 55.17(s, 2H), 54.78-4.77 (d, 2H), 53.45(s, 6H), 52.49(s, 6H), 52.45(s, 6H), 52.28-2.20 (m, 4H).
Example 113: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-(3- (ethyl(methyl)amino)propoxy)-lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5- carboxamide, 1-3
Figure imgf000481_0001
113.2 113.3
Figure imgf000482_0001
113.2 113.3
Figure imgf000483_0001
Synthesis of 113.1
[00849] To a solution of methyl l-[(2E)-4-aminobut-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole- 3-amido)-l,3-benzodiazole-5-carboxylate, 40.6 (600 mg, 1.513 mmol, 1 equiv) in n-butyl alcohol (15 mL) and was added sodium bicarbonate (254.28 mg, 3.027 mmol, 2 equiv) and DIEA (978.00 mg, 7.567 mmol, 5 equiv). After 10 min, was added 4-chloro-3-[3-(morpholin-4-yl)propoxy]-5- nitrobenzamide (520.26 mg, 1.513 mmol, 1.00 equiv). The resulting solution was stirred overnight at 120°C. The mixture was cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (W/oNLLHCCh) and ACN (30% ACN up to 46% in 12 min); Detector, UV 254/220 nm. This resulted in 420 mg (39.51%) of methyl 1- [(2E)-4-([4-carbamoyl-2-[3-(morpholin-4-yl)propoxy]-6-nitrophenyl]amino)but-2-en-l-yl]-2-(2- ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5-carboxylate as a dark yellow solid. LC-MS (ES, m/z): 704 (M+H+).
Synthesis of 113.2
[00850] To a solution of methyl l-[(2E)-4-([4-carbamoyl-2-[3-(morpholin-4-yl)propoxy]-6- nitrophenyl]amino)but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5- carboxylate, 113.1 (420 mg, 0.597 mmol, 1 equiv) in in methanol (15 mL) and water (4 mL) was added sodium dithionite (1.04 g, 5.97 mmol, 10 equiv) and ammonia water (1 mL). The resulting solution was stirred for 2h at room temperature. The mixture was diluted with water, extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. This resulted in 350 mg (87.11%) of methyl l-[(2E)-4- ([2-amino-4-carbamoyl-6-[3-(morpholin-4-yl)propoxy]phenyl]amino)but-2-en-l-yl]-2-(2-ethyl- 5-methylpyrazole-3-amido)-l,3-benzodiazole-5-carboxylate as a brown solid. LC-MS (ES, m/z): 674 (M+H+).
Synthesis of 113.3
[00851] To a solution of methyl l-[(2E)-4-([2-amino-4-carbamoyl-6-[3-(morpholin-4- yl)propoxy]phenyl]amino)but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3- benzodiazole-5-carboxylate, 113.2 (350 mg, 0.519 mmol, 1 equiv) in methanol (15 mL) was cyanogen bromide (109 mg, 1.038 mmol, 2 equiv). The resulting solution was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (10% NH4HCOV) and ACN (27% ACN up to 48% in 10 min); Detector, UV 254/220 nm. This resulted in 275 mg (76%) of methyl l-[(2E)-4-[2-amino-5-carbamoyl-7-[3-(morpholin-4-yl)propoxy]-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5- carboxylate as a light yellow solid. LC-MS (ES, m/z): 699 (M+H+).
Synthesis of 113.4
[00852] To a solution of methyl l-[(2E)-4-[2-amino-5 -carbamoyl -7- [3 -(morpholin-4- yl)propoxy]- 1 ,3-benzodiazol- 1 -yl]but-2-en- 1 -yl]-2-(2-ethyl-5-m ethyl pyrazole-3 -amido)- 1,3-
483
5UB5TITUTE SHEET (RULE 26) benzodiazole-5-carboxylate, 113.3 (275 mg, 0.394 mmol, 1 equiv) and 4-ethyl-2-m ethyl- 1,3- oxazole-5-carboxylic acid (73.3 mg, 0.473 mmol, 1.2 equiv) in dimethyl formamide (5 mL) was added HATU (179.7 mg, 0.473 mmol, 1.2 equiv) and DIEA (254.13 mg, 1.97 mmol, 5 equiv). The final reaction mixture was irradiated with microwave radiation for 1 h at 120°C. The reaction was cooled to RT and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (31% ACN up to 46% in 10 min); UV detection at 254/210 nm. This resulted in 220 mg (66.86%) of methyl l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-[3- (morpholin-4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3- amido)-l,3-benzodiazole-5-carboxylate as a yellow solid. LC-MS (ES, m/z): 418 (M/2+EC). Synthesis of 113.5
[00853] To a solution of methyl l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5- amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazole-5-carboxylate, 113.4 (220 mg, 0.263 mmol, 1 equiv) in methanol (10 mL) and water (2 mL) was added lithium hydroxide (25.25 mg, 1.052 mmol, 4 equiv). The resulting solution was stirred for 4h at 70°C. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration. The resulting solid was dried under infrared light. This resulted in 120 mg (55.48%) of l-[(2E)-4-[5- carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5- carboxylic acid as a light yellow solid. LC-MS (ES, m/z): 822 (M+H+).
Synthesis of 1-3
[00854] To a solution of l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7- [3-(morpholin-4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5-methylpyrazole- 3-amido)-l,3-benzodiazole-5-carboxylic acid, 113.5 (120 mg, 0.146 mmol, 1 equiv) and ammonium chloride (39.04 mg, 0.73 mmol, 5 equiv) in dimethyl formamide (8 mL) was added HATU (83.28 mg, 0.22 mmol, 1.50 equiv) and DIEA (94.36 mg, 0.73 mmol, 5 equiv). The resulting solution was stirred for 4h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (28% ACN up to 40% in 10 min); Detector, UV 254/220 nm. The crude product (60 mg) was purified by Prep-HPLC with the following conditions: Column, XBridge Prep OBD C18 Column, 30* 150mm, 5um; mobile phase, Water (10%NH4HC0 +0.1%NH3 H20) and ACN (20% ACN up to 30% in 7 min); UV detection at 254/220 nm. This resulted in 26.4 mg (21%) of l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole- 5-amido)-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 821 (M+H+); H-NMR: 1HNMR (400 MHz, DMSO- M d 12.78 (br s, 2H), 8.03-7.92 (m, 3H), 7.75-7.65 (m, 2H), 7.45-7.27 (m, 4H), 6.54 (s, 1H), 6.08-5.97 (m, 1H), 5.81- 5.70 (m, 1H), 5.00-4.80 (m, 4H), 4.52 (q, 2H), 4.00 (t, 2H), 3.55-3.45 (m, 4H), 2.89 (q, 2H), 2.42 (s, 3H), 2.34-2.12 (m, 9H), 1.75-1.66 (m, 2H), 1.28 (t, 3H), 1.09 (t, 3H).
Example 114: Synthesis of (E)-12-carbamoyl-l,15-bis(4-ethyl-2-methyloxazole-5- carboxamido)-8,9,16,19-tetrahydro-7H-6,10-dioxa-2,14,15a,19a- tetraazacyclopentadeca[3,2,l-cd:8,9,10-c'd']diindene-4-carboxylic acid, 1-78
Figure imgf000486_0001
Figure imgf000487_0001
Synthesis of 114.1
[00855] To a stirred solution of methyl 4-chloro-3-m ethoxy-5 -nitrobenzoate (10 g, 40.82 mmol, 1 equiv) in dichloromethane (150 mL) were added dropwise boron tribromide (51 g, 204.1 mmol, 5 equiv) at 0°C. The reaction mixture was stirred overnight at 0°C. The mixture was concentrated under reduced pressure. This resulted in 10 g (crude) of 4-chloro-3 -hydroxy-5 -nitrobenzoic acid as a brown solid. LCMS (ES, m/z): 218 (M+H+).
Synthesis of 114.2
[00856] To a stirred solution of 4-chloro-3-hydroxy-5-nitrobenzoic acid, 114.1 (10 g crude, 40.82 mmol, 1 equiv) in methanol (100 mL) was added dropwise concentrated sulfuric acid (5 mL, 98%) at 0°C. The reaction mixture was stirred overnight at 60°C. The mixture was cooled to r.t and concentrated under reduced pressure. The crude product was diluted with water and extracted with dichloromethane. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 8 g (84.84%) of methyl 4-chloro-3 -hydroxy-5 -nitrobenzoate as a light yellow solid. LCMS (ES, m/z): 230 (M-H+).
Synthesis of 114.3
[00857] To a stirred solution of methyl 4-chloro-3-hydroxy-5-nitrobenzoate, 114.2 (3.37 g, 14.6 mmol, 1 equiv), triphenyl phosphine (7.65 g, 29.2 mmol, 2 equiv) and 4-chloro-3-(3- hydroxypropoxy)-5-nitrobenzamide (4 g, 14.6 mmol, 1 equiv) in tetrahydrofuran (100 mL) was added dropwise diisopropyl azodicarboxylate (2.95 g, 14.6 mmol, 2 equiv) at 0°C. The reaction mixture was stirred at 0°C overnight. The mixture was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography with PE/ethyl acetate (1/1) to give 4.5 g (63.3%) of methyl 3-(3-(5-carbamoyl-2-chloro-3-nitrophenoxy)propoxy)-4-chloro-5- nitrobenzoate as a yellow solid. LCMS (ES, m/z): 510 (M+Na+).
Synthesis of 114.4
[00858] To a solution of methyl 3-(3-(5-carbamoyl-2-chloro-3-nitrophenoxy)propoxy)-4- chloro- 5 -nitrobenzoate, 114.3 (2.5 g, 5.133 mmol, 1 equiv) in dimethylsulfoxide (20 mL) was added DIEA (6.621 g, 51.33 mmol, 10 equiv) and (E)-but-2-ene- 1,4-diamine dihydrochloride (816 mg, 5.133 mmol, 1 equiv). The resulting mixture was irradiated with microwave radiation at 120°C for 2h. The reaction mixture was cooled to r.t, diluted with water. The precipitated solids were collected by filtration. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (31% ACN up to 41% in 10 min); Detector, UV 254/220 nm. This resulted in 0.8 g (31.18%) of methyl (15E)-22-carbamoyl-l l,20-dinitro-2,6-dioxa-13,18-diazatricyclo[17.4.0.0A[7,12]]tricosa- l(23),7(12),8,10, 15,19,21-heptaene-9-carboxylate as an orange red solid. LCMS (ES, m/z): 502 (M+H+).
Synthesis of 114.5
[00859] To a stirred solution of methyl (15E)-22-carbamoyl-l l,20-dinitro-2,6-dioxa-13,18- diazatricyclo[17.4.0.0A[7,12]]tricosa-l(23),7(12),8, 10,15,19,21-heptaene-9-carboxylate, 114.4
(800 mg, 1.6 mmol, 1 equiv) in methanol (20 mL) was added sodiumhyposulfite (2.784 g, 16 mmol, 10 equiv) and ammonia water (2 mL). The reaction mixture was stirred for 2h at r.t. The resulting mixture was diluted with water and extracted with ethyl
acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 500 mg (70.86%) of methyl (15E)-l l,20-diamino-22-carbamoyl-2,6-dioxa-13, 18-diazatricyclo[17.4.0.0A[7,12]]tricosa- l(23),7(12),8,10, 15,19,21-heptaene-9-carboxylate as a yellow solid. LCMS (ES, m/z): 442 (M+TE).
Synthesis of 114.6
[00860] To a stirred solution of methyl (15E)-l l,20-diamino-22-carbamoyl-2,6-dioxa-13,18- diazatricyclo[17.4.0.0A[7,12]]tricosa-l(23),7(12),8, 10,15,19,21-heptaene-9-carboxylate, 114.5
(500 mg, 1.136 mmol, 1 equiv) in methanol (10 mL) was added cyanogen bromide (596.4 mg, 5.68 mmol, 5.00 equiv). The resulting mixture was stirred at r.t for 2 hours. The mixture was concentrated under reduced pressure. The residue was diluted with water and extracted with dichloromethane. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The resulted in 450 mg (80.64%) of methyl (3E)-7,25-diamino-21-carbamoyl-14, 18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l l-carboxylate as a yellow solid. LCMS (ES, m/z): 492 (M+H+).
Synthesis of 114.7
[00861] To a solution of methyl (3E)-7,25-diamino-21-carbamoyl-14, 18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l l-carboxylate, 114.6 (450 mg, 0.918 mmol, 1 equiv) and 4-ethyl-2-methyloxazole-5-carboxylic acid (284.58 mg, 1.836 mmol, 2.00 equiv) in dimethyl formamide (8 mL) was added DIEA (592.11 mg, 4.59 mmol, 5 equiv) and HATU (697.68 mg, 1.836 mmol, 2 equiv). The resulting mixture was stirred at 100°C for 2 hours. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.01%NH4HC03+0.1%NH3 H20) and methanol (81% methanol up to 91% in 10 min); Detector, UV 254/220 nm. This resulted in 500 mg (71.22%) of methyl (3E)-21-carbamoyl-7,25-bis(4-ethyl-2-methyl-l,3-oxazole-5-amido)- 14,18-dioxa-l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
488
5UB5TITUTE SHEET (RULE 26) 3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylate as a brown solid. LC-MS (ES, m/z): 384 (M/2+EG).
Synthesis of 1-78
[00862] To a stirred mixture of methyl (3E)-21-carbamoyl-7,25-bis(4-ethyl-2-methyl-l,3- oxazole-5-amido)- 14, 18-dioxa- 1 ,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylate, 114.7 (50 mg, 0.065 mmol, 1 equiv) in methanol (10 mL) and water (2 mL) was added lithium hydroxide (4.68 mg, 0.195 mmol, 3 equiv). The resulting mixture was stirred at 70°C for 4 hours. The mixture was concentrated under reduced pressure. The residue was diluted with water, then adjusted to pH 3 with 1 N HC1. The solid was collected by filtration. The crude product was purified by Prep-HPLC with the following conditions: Column: XSelect CSHPrep C18 OBD Column, 19* 150mm, 5um; mobile phase, Water (0.01%NH4HC03+0.1%NH3 H20) and ACN (18% ACN up to 33% in 8 min); Detector, UV 254/220 nm. This resulted in 5.9 mg (12.1%) of (3E)-21-carbamoyl-7,25-bis(4-ethyl-2-methyl- 1 ,3-oxazole-5-amido)- 14, 18-dioxa- 1 ,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylic acid as a white solid. LC-MS (ES, m/z): 752 (M+H+); ¾ NMR: (400 MHz, DMSO-t¾) d 12.58 (br s, 2H), 7.99 (s, 1H), 7.81 (s, 1H), 7.70 (s, 1H), 7.52 (d, 2H), 7.34 (d, 1H), 5.66-5.45 (m, 2H), 4.97-4.82 (m, 4H), 4.45-4.31 (m, 4H), 2.83 (q, 4H), 2.39 (s, 6H), 2.36-2.22 (m, 2H), 1.04 (t, 6H).
Example 115: Synthesis of (E)-N,N'-(12-carbamoyl-4-(4-hydroxypiperidine-l-carbonyl)- 8,9,16,19-tetrahydro-7H-6,10-dioxa-2,14,15a,19a-tetraazacyclopentadeca[3,2,l-cd:8,9,10- c'd']diindene-l,15-diyl)bis(4-ethyl-2-methyloxazole-5-carboxamide), 1-130
Figure imgf000491_0001
1-78 1-130
[00863] To a stirred mixture of (3E)-21-carbamoyl-7,25-bis(4-ethyl-2-methyl-l,3-oxazole-5- amido)-14,18-dioxa-l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylic acid, Example 115 (50 mg, 0.067 mmol, 1 equiv) and piperidin-4-ol (10.1 mg, 0.1 mmol, 1.5 equiv) in dimethyl formamide (5 mL) was added DIEA (25.8 mg, 0.2 mmol, 3 equiv) and HATU (30.5 mg, 0.08 mmol, 1.2 equiv). The resulting mixture was stirred at 30°C for 2 hours. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep C18 Column, 30*150mm, 5um; mobile phase, Water (0.1%FA) and ACN (17% ACN up to 46% in 8 min); Detector, UV 254/220 nm. This resulted in 11.8 mg (21.12%) of (3E)-7,25- bis(4-ethyl-2-methyl-l,3-oxazole-5-amido)-21-(4-hydroxypiperidine-l-carbonyl)-14,18-dioxa- l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3,7,9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l 1-carboxamide as a white solid. LC-MS (ES, m/z): 835 (M+EC); 1HNMR (400 MHz, DMSO-t e) d 12.74 (br s, 2H), 7.98 (s, 1H), 7.70 (s, 1H), 7.50 (s, 1H), 7.36 (s, 1H), 7.21 (s, 1H), 7.00 (s, 1H), 5.57-5.46 (m, 2H), 4.92-4.81 (m, 5H), 4.46-4.38 (m, 4H), 4.06-3.45 (m, 3H), 3.26-3.13 (m, 2H), 2.82 (q, 4H), 2.39 (s, 6H), 2.34-2.22 (m, 2H), 1.88- 1.71 (m, 2H), 1.48-1.32 (m, 2H), 1.03 (t, 6H).
Example 116: Synthesis of (E)-l,15-bis(4-ethyl-2-methyloxazole-5-carboxamido)-8,9,16,19- tetrahydro-7H-6,10-dioxa-2,14,15a,19a-tetraazacyclopentadeca[3,2,l-cd:8,9,10- c'd']diindene-4, 12-dicarboxamide, 1-129 H.N^^'Boc nCI/ 1.4-dioxane (4 mol/L) c|HH2N / ^ ^NH2HCI
DCM
Figure imgf000492_0001
Synthesis of 116.1
[00864] To a stirred solution of (E)-tert-butyl 4-aminobut-2-enylcarbamate (3 g, 16.13 mmol, 1 equiv) in dichloromethane (30 mL) were added hydrochloric acid in 1,4-dioxane (4 mol/L, lOmL) at 0°C. The reaction mixture was stirred for 4h at r.t. The mixture was concentrated under reduced pressure. This resulted in 2.5 g (98%) of (E)-but-2-ene- 1,4-diamine dihydrochloride as a white solid. LCMS (ES, m/z): 87 (M+H+).
Synthesis of 116.2
[00865] To a stirred solution of 5,5'-(propane-l,3-diylbis(oxy))bis(4-chloro-3-nitrobenzamide), 116.1 (1.187 g, 2.516 mmol, 1 equiv) in dimethyl sulfoxide (10 mL) was added DIEA (3.246 g, 25.16 mmol, 10 equiv) and (E)-but-2-ene-l, 4-diamine dihydrochloride (400 mg, 2.516 mmol, 1 equiv). The resulting mixture was irradiated with microwave radiation at 120°C for 2h. The reaction mixture was cooled to r.t, diluted with water. The precipitated solids were collected by filtration. This resulted in 600 mg (49%) of (15E)-11,20- dinitro-2,6-dioxa-13,18-diazatricyclo[17.4.0.0A[7, 12]]tricosa-l(19),7,9,l l, 15,20,22-heptaene- 9, 22-dicarboxamide as an orange red solid. LCMS(ES, m/z): 487 (M+EE).
Synthesis of 116.3
[00866] To a stirred solution of (15E)-l l,20-dinitro-2,6-dioxa-13,18- diazatricyclo[17.4.0.0L[7, 12]]tricosa-l(19), 7, 9, 11, 15, 20, 22 -heptaene-9, 22-dicarboxamide, 116.2 (600 mg, 1.235 mmol, 1 equiv) in methanol (20 mL) was added sodiumhyposulfite (2.15 g, 12.35 mmol, 10 equiv) and ammonia water (2 mL). The reaction mixture was stirred for 2h at r.t. The resulting mixture was diluted with water and extracted with ethylacetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 300 mg (57%) of (15E)-l l,20-diamino-2,6-dioxa-13,18- diazatricyclo[17.4.0.0A[7,12]]tricosa-l(19), 7, 9, 11, 15, 20, 22 -heptaene-9, 22-dicarboxamide as a yellow solid. LCMS (ES, m/z): 427 (M+H+).
Synthesis of 116.4
[00867] To a solution of (15E)-l l,20-diamino-2,6-dioxa-13,18- diazatricyclo[17.4.0.0A[7,12]]tricosa-l(19), 7, 9, 11, 15, 20, 22 -heptaene-9, 22-dicarboxamide,
116.3 (300 mg, 0.704 mmol, 1 equiv) in methanol (10 mL) was added cyanogen bromide (369.6 mg, 3.52 mmol, 5.00 equiv). The resulting mixture was stirred at r.t for 2 hours. The mixture was concentrated under reduced pressure. The residue was diluted with water and extracted with dichloromethane. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 180 mg (53.73%) of (3E)-7,25-diamino-14, 18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3, 7, 9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l l, 21 -dicarboxamide as a brown solid. LC-MS (ES, m/z): 477 (M+H+).
Synthesis of 116.5 [00868] To a stirred mixture of (3E)-7,25-diamino-14, 18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3, 7, 9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l l, 21 -dicarboxamide, 116.4 (180 mg, 0.378 mmol, 1 equiv) and 4-ethyl-2-methyloxazole-5-carboxylic acid (117.18 mg, 0.756 mmol, 2.00 equiv) in dimethyl formamide (5 mL) was added DIEA (243.81 mg, 1.89 mmol, 5 equiv) and HATU (287.28 mg, 0.756 mmol, 2 equiv). The resulting mixture was stirred at 100°C for 2 hours. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 19*250mm, 5um; mobile phase, Water (0.01%NH4HC03+0.1%1S1H3 H20) and methanol (40% methanol up to 68% in 15 min); Detector, UV 254/220 nm. This resulted in 33.6 mg (11.85%) of (E)- 1, 15 -bi s(4-ethyl-2- methyloxazole-5-carboxamido)-8,9, 16, 19-tetrahydro-7H-6, 10-dioxa-2, 14, 15 a, 19a- tetraazacyclopentadeca[3,2, l-cd:8,9,10-c'd']diindene-4, 12-dicarboxamide as a white solid. LC- MS (ES, m/z): 751 (M+H+); 1H NMR: (400 MHz, DMSO-t/e) d 12.65 (br s, 2H), 7.98 (s, 2H), 7.70 (s, 2H), 7.52 (s, 2H), 7.31 (s, 2H), 5.54-5.39 (m, 2H), 4.95-4.82 (m, 4H), 4.49-4.35 (m, 4H), 2.82 (q, 4H), 2.42-2.27 (m, 8H), 1.04 (t, 6H).
Example 117: Synthesis of (E)-N,N'-(12-carbamoyl-4-(4-(2-hydroxyethyl)piperazine-l- carbonyl)-8,9, 16,19-tetr ahydro-7H-6,l 0-dioxa-2, 14,15a, 19a-tetraazacy clopentadeca [3,2,1- cd:8,9,10-c'd']diindene-l,15-diyl)bis(4-ethyl-2-methyloxazole-5-carboxamide), 1-128
Figure imgf000494_0001
[00869] To a stirred mixture of (3E)-21-carbamoyl-7,25-bis(4-ethyl-2-methyl-l,3-oxazole-5- amido)-14,18-dioxa-l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3, 7, 9(27), 10, 12, 19(26), 20, 22, 24-nonaene-l l -carboxylic acid (50 mg, 0.067 mmol, 1 equiv) and 2- (piperazin-l-yl)ethanol, 1-78, (13 mg, 0.1 mmol, 1.5 equiv) in dimethyl formamide (5 mL) was added DIEA (25.8 mg, 0.2 mmol, 3 equiv) and HATU (30.5 mg, 0.08 mmol, 1.2 equiv). The resulting mixture was stirred at 30°C for 2 hours. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XSelect CSH Prep C18 OBD Column, 19* 150mm, 5um; mobile phase, Water (0.1%F A) and ACN (11% ACN up to 32% in 7 min); Detector, UV 254/220 nm. This resulted in 14 mg (24.22%) of (3E)-7,25-bis(4-ethyl -2-methyl- 1 ,3 -oxazole-5-amido)-21 -[4-(2-hydroxy ethyl )piperazine- 1 - carbonyl]- 14, 18-dioxa- 1 ,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l 1-carboxamide as a white solid. LC-MS (ES, m/z): 864 (M+EC); TlNMR (400 MHz, DMSO-<¾) <5 12.76 (s, 1H), 12.72 (s, 1H), 7.98 (s, 1H), 7.69 (s, 1H), 7.53 (s, 1H), 7.36 (s, 1H), 7.23 (s, 1H), 7.02 (s, 1H), 5.57-5.48 (m, 2H), 4.94-4.62 (m, 5H), 4.47-4.38 (m, 4H), 3.74-3.41 (m, 6H), 2.83 (q, 4H), 2.79-2.54 (m, 6H), 2.39 (s, 6H), 2.35-2.22 (m, 2H), 1.03 (t, 6H).
Example 118: Synthesis of (E)-l,15-bis(4-ethyl-2-methyloxazole-5-carboxamido)-N-(2-(2-(2- hydroxyethoxy)ethoxy)ethyl)-8,9, 16,19-tetrahy dro-7H-6,l 0-dioxa-2, 14, 15a,l 9a- tetraazacyclopentadeca[3, 2, l-cd:8, 9, 10-c'd']diindene-4, 12-dicarboxamide, 1-127
Figure imgf000495_0001
1-127
[00870] To a stirred mixture of (3E)-21-carbamoyl-7,25-bis(4-ethyl-2-methyl-l,3-oxazole-5- amido)-14,18-dioxa-l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
494
5UB5TITUTE SHEET (RULE 26) 3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylic acid (30 mg, 0.04 mmol, 1 equiv) and 2- (2-(2-aminoethoxy)ethoxy)ethanol (9 mg, 0.06 mmol, 1.5 equiv) in dimethyl formamide (5 mL) was added DIEA (15.48 mg, 0.12 mmol, 3 equiv) and HATU (18.24 mg, 0.048 mmol, 1.2 equiv). The resulting mixture was stirred at 30°C for 2 hours. The mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XSelect CSH Prep C18 OBD Column, 19* 150mm, 5um; mobile phase, Water (0.1%F A) and ACN (20% ACN up to 38% in 7 min); Detector, UV 254/220 nm. This resulted in 4.3 mg (12.17%) of (3E)-7,25-bis(4-ethyl-2-methyl-l,3-oxazole-5-amido)-N21-[2-[2-(2- hydroxyethoxy)ethoxy]ethyl]-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3, 7, 9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l l, 21 -dicarboxamide as a white solid. LC-MS (ES, m/z): 905 (M+Na+); 1HNMR (400 MHz, DMSO-t/e) S 12.77 (s, 2H), 8.53 (t, 1H), 7.98 (s, 1H), 7.69 (d, 2H), 7.51 (d, 2H), 7.37 (s, 1H), 5.56-5.48 (m, 2H), 4.92-4.83 (m, 4H), 4.41 (t, 1H), 4.49- 4.38 (m, 4H), 3.65-3.42 (m, 12H), 2.82 (q, 4H), 2.39 (s, 6H), 2.36-2.22 (m, 2H), 1.05 (t, 6H).
Example 119: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(4-ethyl-2- methyloxazole-5-carboxamido)-7-(3-hydroxypropoxy)-lH-benzo[d]imidazol-l-yl)but-2-en- l-yl)-7-methoxy-lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-115
Figure imgf000496_0001
Figure imgf000497_0001
1-115
Synthesis of 119.1
[00871] To a stirred solution of 4-chloro-3-methoxy-5-nitrobenzamide (16.00 g, 69.384 mmol, 1.00 equiv) and 1 M BBr3 in DCE (180.00 mL) in portions at 50°C under nitrogen atmosphere. The mixture was stirred for overnight at 50°C under nitrogen atmosphere. The precipitated solids were collected by filtration and washed with water (3x200 mL). This resulted in 4-chloro-3- hydroxy-5-nitrobenzamide (15.2 g) (ES, m/z): [M+H]+ 217.0 as an off-white solid.
Synthesis of 119.2 [00872] To a stirred solution of 4-chloro-3-hydroxy-5-nitrobenzamide, 119.1 (2.00 g, 9.259 mmol, 1.00 equiv) in DMSO (25.00 mL) were added 3-bromopropanol (1.67 g, 12.037 mmol, 1.3 equiv) and K2CO3 (2.56 g, 18.519 mmol, 2 equiv) in portions at room temperature. The resulting mixture was stirred for overnight at 100°C under nitrogen atmosphere. The resulting mixture was extracted with EtOAc (3x30 mL). The combined organic layers were washed with brine (3x30 mL), dried over anhydrous NaiSCL. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 4-chloro-3-(3-hydroxypropoxy)-5-nitrobenzamide (2 g, 70.97%) (ES, m/z): [M+H]+ 275.0 as a yellow solid.
Synthesis of 119.3
[00873] To a stirred solution of 4-chloro-3-(3-hydroxypropoxy)-5-nitrobenzamide, 119.2 (2.00 g, 7.282 mmol, 1.00 equiv) in n-BuOH (30.00 mL) in a 100 mL 3-necked round-bottom flask, NaHCCb (1.22 g, 14.563 mmol, 2 equiv) and DIEA (5.65 g, 43.690 mmol, 6 equiv) and 4-[[(2E)- 4-aminobut-2-en-l-yl]amino]-3-methoxy-5-nitrobenzamide (2.04 g, 7.278 mmol, 1.00 equiv) were added in portions at room temperature. The resulting mixture was stirred for overnight at 120°C under nitrogen atmosphere. The resulting mixture was filtered and the filter cake was washed with n-BuOH (3x30 mL). The filtrate was stirred for 10 min at room temperature in water (100 mL). The resulting mixture was filtered, the filter cake was washed with EtOH (3x30 mL). The filtrate was concentrated under reduced pressure. The crude product was re-crystallized from EtO Ac/PE (1 : 10, 2 mL) to afford 4-[[(2E)-4-[[4-carbamoyl-2-(3-hydroxypropoxy)-6- nitrophenyl]amino]but-2-en-l-yl]amino]-3-methoxy-5-nitrobenzamide (2 g, 47.68%) (ES, m/z): [M+H]+ 519.2 as an off-white solid.
Synthesis of 119.4
[00874] To a stirred solution of 4-[[(2E)-4-[[4-carbamoyl-2-(3-hydroxypropoxy)-6- nitrophenyl]amino]but-2-en-l-yl]amino]-3-methoxy-5-nitrobenzamide, 119.3 (1.90 g, 3.857 mmol, 1.00 equiv) in MeOH (40.00 mL) in a 100 mL 3-necked round-bottom flask, NaiSiCri (6.72 g, 38.597 mmol, 10.01 equiv), ammonia (1.64 g, 96.435 mmol, 25 equiv) and H2O (16.00 mL) were added in portions at room temperature. The resulting mixture was stirred for lh at room temperature. The resulting mixture was filtered and the filter cake was washed with MeOH (3x10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (CH2Cl2:MeOH=5: l) to afford 3-amino-4-[[(2E)-4-[[2-amino-4-carbamoyl-6-(3-
497
5UB5TITUTE SHEET (RULE 26) hydroxypropoxy)phenyl]amino]but-2-en-l-yl]amino]-5-methoxybenzamide (278 mg, 13.83%) (ES, m/z): [M+H]+ 459.2 as an off-white solid.
Synthesis of 119.5
[00875] To a stirred solution of 3-amino-4-[[(2E)-4-[[2-amino-4-carbamoyl-6-(3- hydroxypropoxy)phenyl]amino]but-2-en-l-yl]amino]-5-methoxybenzamide, 119.4 (268.00 mg, 0.584 mmol, 1.00 equiv) in MeOH (5.00 mL) in a 20 mL vial, BrCN (61.91 mg, 0.584 mmol, 1 equiv) was added in portions at room temperature. The resulting mixture was stirred for lh at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was precipitated by the addition of 2 mL MeOH. This resulted in 2- amino-l-[(2E)-4-[2-amino-5-carbamoyl-7-(3-hydroxypropoxy)-l,3-benzodiazol-l-yl]but-2-en-l- yl]-7-methoxy-l,3-benzodiazole-5-carboxamide(260 mg, 87.47%) (ES, m/z): [M+H]+509.2 as a white solid.
Synthesis of 1-115
[00876] To a stirred solution of 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (122.04 mg, 0.787 mmol, 4.00 equiv) in NMP (3.00 mL) in an 8 mL sealed tube, HATU (299.08 mg, 0.787 mmol, 4 equiv), DIEA (304.98 mg, 2.360 mmol, 12 equiv) and 2-amino-l-[(2E)-4-[2-amino-5- carbamoyl-7-(3-hydroxypropoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-7-methoxy-l,3- benzodiazole-5-carboxamide, 119.5 (100.00 mg, 0.197 mmol, 1.00 equiv) were added at room temperature. The resulting mixture was irradiated for 1.5 h at 140°C under nitrogen atmosphere. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, MeOH in water, 10% to 50% gradient in 10 min; detector, UV 254 nm. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge Shield RP18 OBD Column, 19*250 mm, 10 um; Mobile Phase A: water(10 mmol/L NH4HCO +0.1 %NHVH20), Mobile Phase B: ACN; Flow rate: 20 mL/min; Gradient: 33% B to 37% B in 7 min; 254/220 nm; RT: 6.8 min) to afford 1-115 (25.7 mg, 12.76%) as a white solid. LC-MS: (ES, m/z) [M+H]+ :783.4 [M-H] : 781.3; ¾-NMR: (400 MHz, DMSO-de , ppm) 512.71 (s, 2H), 57.97 (s, 2H), 57.64-7.63 (d, 2H), 57.35-7.32 (m, 4H), 55.81-5.78 (m, 2H), 54.91-4.87 (m, 4H), 54.57-4.54 (t, 1H), 54.09-4.06 (t, 2H), 53.78 (s, 3H), 53.49-3.45 (m, 2H), 52.84-2.77 (m, 4H), 52.40-2.39 (d, 6H), 51.75-1.65 (m, 2H), 51.04-0.91 (t, 6H). Example 120: Synthesis of (E)-l-(4-ethyl-2-methyloxazole-5-carboxamido)-15-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-8,9,16,19-tetrahydro-7H-6,10-dioxa-2,14,15a,19a- tetraazacyclopentadeca[3, 2, l-cd:8, 9, 10-c'd']diindene-4, 12-dicarboxamide, 1-126
Figure imgf000500_0001
120.4 120.5
Figure imgf000501_0001
1-126 Synthesis of 120.1
[00877] To a stirred solution of tert-butyl N-[(2E)-4-aminobut-2-en-l-yl]carbamate (0.93 g, 4.993 mmol, 1.00 equiv) in n-butyl alcohol (20 mL) was added sodium bicarbonate (0.84 g, 9.999 mmol, 2.00 equiv) and DIEA (3.22 g, 24.914 mmol, 4.99 equiv). After 10 min, was added 3-[3- [(tert-butyldimethylsilyl)oxy]propoxy]-4-chloro-5-nitrobenzamide (1.94 g, 4.988 mmol, 1.00 equiv). The resulting mixture was stirred overnight at 120°C under nitrogen. The mixture was cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 2.5g (76.46%) of tert-butyl N-[(2E)-4-[(2- [3-[(tert-butyldimethylsilyl)oxy]propoxy]-4-carbamoyl-6-nitrophenyl)amino]but-2-en-l- yl] carbarn ate as a yellow solid. LC-MS (ES, m/z): 539 (M+H+).
Synthesis of 120.2
[00878] To a stirred solution of tert-butyl N-[(2E)-4-[(2-[3-[(tert- butyldimethylsilyl)oxy]propoxy]-4-carbamoyl-6-nitrophenyl)amino]but-2-en-l-yl]carbamate, 120.1 (2.40 g, 4.455 mmol, 1.00 equiv) in dimethyl formamide (40 mL) was added tin(II)chloride dihydrate (5.03 g, 22.275 mmol, 5.00 equiv). The resulting mixture was stirred overnight at room temperature. The mixture was diluted with water. The pH value of the solution was adjusted to 9 with sat. sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. This resulted in 1.8 g (54.01%) of tert-butyl N-[(2E)-4-[[2- amino-4-carbamoyl-6-(3-hydroxypropoxy)phenyl]amino]but-2-en-l-yl]carbamate as a brown oil. LC-MS (ES, m/z): 395 (M+H+).
Synthesis of 120.3
[00879] To a stirred solution of tert-butyl N-[(2E)-4-[[2-amino-4-carbamoyl-6-(3- hydroxypropoxy)phenyl]amino]but-2-en-l-yl]carbamate, 120.2 (1.80 g, 4.563 mmol, 1.00 equiv) in methanol (20 mL) was added cyanogen bromide (0.726 g, 6.845 mmol, 1.5 equiv). The resulting mixture was stirred for 3 h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (48% ACN up to 59% in 10 min); Detector, UV 254/220 nm. This resulted in 1.3 g (67.85%) of tert-butyl N-[(2E)-4-[2-amino-5-
501
5UB5TITUTE SHEET (RULE 26) carbamoyl-7-(3-hydroxypropoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate as a brown oil. LC-MS (ES, m/z): 420 (M+H+).
Synthesis of 120.4
[00880] To a stirred mixture of tert-butyl N-[(2E)-4-[2-amino-5-carbamoyl-7-(3- hydroxypropoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate, 120.3 (1.30 g, 3.099 mmol, 1.00 equiv) and 2-ethyl-5-methylpyrazole-3 -carboxylic acid (954.8 mg, 6.2 mmol, 2 equiv) in dimethyl formamide (20 mL) was added HATU (2.356 g, 6.2 mmol, 2.00 equiv) and DIEA (2.00 g, 15.495 mmol, 5.00 equiv). The resulting mixture was stirred overnight at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (41% ACN up to 50% in 10 min); Detector, UV 254/220 nm. This resulted in 1.2 g (62.7%) of 3-([3-[(2E)-4- [(tert-butoxycarbonyl)amino]but-2-en-l-yl]-6-carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)- l,3-benzodiazol-4-yl]oxy)propyl 2-ethyl-5-methylpyrazole-3-carboxylate as a brown oil. LC-MS (ES, m/z): 692 (M+H+).
Synthesis of 120.5
[00881] To a stirred mixture of 3-([3-[(2E)-4-[(tert-butoxycarbonyl)amino]but-2-en-l-yl]-6- carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-4-yl]oxy)propyl 2-ethyl-5- methylpyrazole-3-carboxylate, 120.4 (1.2 g, 1.302 mmol, 1.00 equiv) in methanol (15 mL) and water (5 mL) was added lithium hydroxide (156.24 mg, 6.51 mmol, 5.00 equiv). The resulting mixture was stirred overnight at room temperature. The mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 500 mg (69.16%) of tert-butyl N-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-7-(3- hydroxypropoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate as a white solid. LC-MS (ES, m/z): 556 (M+H+).
Synthesis of 120.6
[00882] To a stirred mixture of tert-butyl N-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5- methylpyrazole-3-amido)-7-(3-hydroxypropoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate, 120.5 (500 mg, 0.900 mmol, 1.00 equiv) and 4-chloro-3-hydroxy-5-nitrobenzamide (243.62 mg, 1.125 mmol, 1.25 equiv) and triphenylphosphine (354.03 mg, 1.350 mmol, 1.50 equiv) in tetrahydrofuran (20 mL) was added dropwise DIAD (272.94 mg, 1.350 mmol, 1.50 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred overnight at 50°C under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (33% ACN up to 43% in 10 min); Detector, UV 254/220 nm. This resulted in 600 mg (82.27%) of tert-butyl N- [(2E)-4-[5-carbamoyl-7-[3-(5-carbamoyl-2-chloro-3-nitrophenoxy)propoxy]-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate as a yellow solid. LC- MS (ES, m/z): 754 (M+H+).
Synthesis of 120.7
[00883] To a stirred solution of tert-butyl N-[(2E)-4-[5-carbamoyl-7-[3-(5-carbamoyl-2- chloro-3-nitrophenoxy)propoxy]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-l- yl]but-2-en-l-yl]carbamate, 120.6 (600 mg, 0.797 mmol, 1.00 equiv) in dichloromethane (20 mL) was added trifluoroacetic acid (5 mL). The resulting mixture was stirred overnight at room temperature. The mixture was concentrated under vacuum. This resulted in 600 mg (crude) of 1- [(2E)-4-aminobut-2-en-l-yl]-7-[3-(5-carbamoyl-2-chloro-3-nitrophenoxy)propoxy]-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazole-5-carboxamide as a brown oil. LC-MS (ES, m/z): 654 (M+LL).
Synthesis of 120.8
[00884] To a stirred solution of l-[(2E)-4-aminobut-2-en-l-yl]-7-[3-(5-carbamoyl-2-chloro-3- nitrophenoxy)propoxy]-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazole-5-carboxamide, 120.7 (600 mg crude, 0.797 mmol, 1.00 equiv) in dimethylsulfoxide (20 mL) was added DIEA (514 mg, 3.985 mmol, 5.00 equiv). The final reaction mixture was irradiated with microwave radiation for 6h at 120°C. The mixture was cooled to room temperature. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (31% ACN up to 42% in 10 min); Detector, UV 254/220 nm. This resulted in 344 mg (69.83%) of (15E)-19-(2-ethyl-5-methylpyrazole-3-amido)-l l-nitro-2,6-dioxa-13,18,20- tri azatetracyclo[16.6.1.0L[7, 12] 0L[21, 25]]pentacosa-l(25), 7, 9, 11, 15, 19,21, 23-octaene-9, 23- dicarboxamide as a brown oil. LC-MS (ES, m/z): 618 (M+FL). Synthesis of 120.9
[00885] To a stirred solution of (15E)-19-(2-ethyl-5-methylpyrazole-3-amido)-l l-nitro-2,6- dioxa-13,18,20-triazatetracyclo[16.6.1.0A[7, 12].0A[21,25]]pentacosa-l(25),7,9,l l, 15,19,21,23- octaene-9, 23 -dicarboxamide, 120.8 (344 mg, 0.557 mmol, 1.00 equiv) in dimethyl formamide (10 mL) was added tin( II )chloridedihydrate (628 mg, 2.785 mmol, 5.00 equiv). The resulting mixture was stirred overnight at room temperature. The mixture was diluted with water. The pH value of the solution was adjusted to 9 with sat. sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. This resulted in 157 mg (48%) of (E)-14-amino-l-(l-ethyl- 3 -methyl- 1 H-pyrazole-5-carboxamido)-8,9, 16,19-tetrahydro-7H, 15H-6, 10-dioxa-2, 15,19a- triazabenzo[9, 10]cyclopentadeca[l, 2, 3-cd]indene-4, 12-dicarboxamide as a brown oil. LC-MS (ES, m/z): 588 (M+H+).
Synthesis of 120.10
[00886] To a stirred solution of (E)-14-amino-l-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-8,9, 16,19-tetrahydro-7H, 15H-6, 10-dioxa-2, 15,19a- triazabenzo[9, 10]cyclopentadeca[l, 2, 3-cd]indene-4, 12-dicarboxamide, 120.9 (157 mg, 0.267 mmol, 1.00 equiv) in methanol (10 mL) was added cyanogen bromide (42 mg, 0.4 mmol, 1.5 equiv). The resulting mixture was stirred for 2h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (28% ACN up to 38% in 10 min); Detector, UV 254/220 nm. This resulted in 80 mg (48.88%) of (3Z)-25-amino-7- (2-ethyl-5-methylpyrazole-3-amido)-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3, 7, 9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l l, 21 -dicarboxamide as a brown oil. LC-MS (ES, m/z): 613 (M+H+).
Synthesis of 1-126
[00887] To a stirred mixture of (3Z)-25-amino-7-(2-ethyl-5-methylpyrazole-3-amido)-14,18- dioxa-l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3, 7, 9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l l, 21-dicarboxamide, 120.10 (80.00 mg, 0.131 mmol, 1.00 equiv) and 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (20.26 mg, 0.131 mmol, 1.00 equiv) in dimethyl formamide (8 mL) was added HATU (49.65 mg, 0.131 mmol, 1.00 equiv) and DIEA (84.38 mg, 0.653 mmol, 5.00 equiv). The resulting mixture was stirred for 3h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XSelect CSH Prep Cl 8 OBD Column,, 19*250mm, 5um; mobile phase, Water (10%NH4HC03+0.1%NH3 H20) and methanol (60% methanol up to 93% in 9 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 24 mg (24.43%) of (3Z)-25-(4-ethyl-2- methyl-l,3-oxazole-5-amido)-7-(2-ethyl-5-methylpyrazole-3-amido)-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3 ,7,9, 11 , 13 (27), 19,21 ,23 (26),24-nonaene- 11 ,21 -dicarboxamide as an off-white solid. LC-MS (ES, m/z): 750 (M+H+); 1H NMR: (400 MHz, DMSO -d6) d 12.81 (br s, 2H), 7.99 (s, 2H), 7.70 (s, 2H), 7.52 (s, 2H), 7.37 (s, 2H), 6.55 (s, 1H), 5.57-5.48 (m, 2H), 5.02-4.81 (m, 4H), 4.65-4.38 (m, 6H), 2.84 (q, 2H), 2.49-2.29 (m, 5H), 2.09 (s, 3H), 1.27 (t, 3H), 1.03 (t, 3H).
Example 121: Synthesis of (E)-l-(4-ethyl-2-methyloxazole-5-carboxamido)-15-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-8,9,16,19-tetrahydro-7H-6,10-dioxa-2,14,15a,19a- tetraazacyclopentadeca[3, 2, l-cd:8, 9, 10-c'd']diindene-4, 12-dicarboxamide, 1-112
Figure imgf000506_0001
70.2 121.1
Figure imgf000507_0001
1-112 Synthesis of 121.1
[00888] To a solution of 4-chloro-3 -hydroxy-5 -nitrobenzamide (1.08 g, 5 mmol, 1 equiv), triphenylphosphine (1.965 g, 7.5 mmol, 1.5 equiv) and 2-morpholinoethan-l-ol (786 mg, 6 mmol, 1.2 equiv) in tetrahydrofuran (30 mL) was added dropwise DIAD (1.515 g, 7.5 mmol, 1.5 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred at 50°C overnight. The mixture was cooled to RT and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with dichloromethane/methanol (10/1) to give 700 mg (41.17%) of 4-chloro-3-(2-morpholinoethoxy)-5 -nitrobenzamide a yellow solid. LC-MS (ES, m/z): 340 (M+H+);
Synthesis of 121.2
[00889] To a solution of 4-chloro-3-(2-morpholinoethoxy)-5-nitrobenzamide, 121.1 (700 mg, 2.13 mmol, 1.0 equiv) in dimethylsulfoxide (10 mL) was added methyl (E)-l-(4-aminobut-2-en- l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate hydrochloride (880 mg, 2.1 mmol, 1.0 equiv) and DIEA (2.75 g, 21.3 mmol, 10.0 equiv). The final reaction mixture was irradiated with microwave radiation at 120°C for 2h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (42% ACN up to 52% in 10 min); Detector, UV 254/220 nm. This resulted in 440 mg (27%) of methyl (E)-l-(4-((4-carbamoyl-2-(2-morpholinoethoxy)-6-nitrophenyl)amino)but-2- en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate as an off-yellow solid. LC-MS (ES, m/z): 690 (M+H+);
Synthesis of 121.3
[00890] To a stirred solution of methyl (E)-l-(4-((4-carbamoyl-2-(2-morpholinoethoxy)-6- nitrophenyl)amino)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxylate, 121.2 (440 mg, 0.49 mmol, 1.0 equiv) in methanol (20 mL) and water (5 mL) was added sodium dithionite (1.03 g, 4.95 mmol, 10.0 equiv) and ammonia water (2 mL). The resulting mixture was stirred at RT for 2h. The mixture was diluted with water, extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. This resulted in 300 mg (88%) of methyl (E)-l-(4-((2- amino-4-carbamoyl-6-(2-morpholinoethoxy)phenyl)amino)but-2-en-l-yl)-2-(l-ethyl-3-methyl-
507
5UB5TITUTE SHEET (RULE 26) lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate an off-yellow solid. LC-MS (ES, m/z): 660 (M+H+);
Synthesis of 121.4
[00891] To a solution of methyl (E)-l-(4-((2-amino-4-carbamoyl-6-(2- morpholinoethoxy)phenyl)amino)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxylate, 121.3 (300 mg, 0.46 mmol, 1.0 equiv) in methanol (10 mL) was added cyanogen bromide (96.5 mg, 0.91 mmol, 2.0 equiv). The resulting mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. This resulted in 350 mg (crude) of methyl (E)-l-(4-(2-amino-5-carbamoyl-7-(2-morpholinoethoxy)- lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxylate as a brown solid. LC-MS (ES, m/z): 685 (M+H+).
Synthesis of 121.5
[00892] To a solution of 4-ethyl-2-methyloxazole-5-carboxylic acid (107 mg, 0.69 mmol, 1.5 equiv) and DIEA (296.7 mg, 2.3 mmol, 5.0 equiv) and HATU (262.2 mg, 0.69 mmol, 1.5 equiv) in dimethyl formamide (5 mL) was added (E)-l-(4-(2-amino-5-carbamoyl-7-(2- morpholinoethoxy)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole- 5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate, 121.4 (350 mg crude, 0.46 mmol, 1.0 equiv). The resulting mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (38% ACN up to 48% in 10 min); Detector, UV 254/220 nm. This resulted in 200 mg (52.89%) of methyl (E)-l- (4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5-carboxamido)-7-(2-morpholinoethoxy)-lH- benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxylate as an off-yellow solid. LC-MS (ES, m/z): 822 (M+H+).
Synthesis of 121.6
[00893] To a solution of methyl (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5- carboxamido)-7-(2-morpholinoethoxy)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l -ethyl-3 - methyl- lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate, 121.5 (200 mg, 0.24 mmol, 1.0 equiv) in methanol (20 mL) and water (4 mL) was added lithium hydroxide (29 mg, 1.2 mmol, 5.0 equiv). The resulting mixture was stirred at 70°C for 2 hrs. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration. This resulted in 100 mg (61.88%) of (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5- carboxamido)-7-(2-morpholinoethoxy)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l -ethyl-3 - methyl- lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylic acid as a yellow solid. LC-MS (ES, m/z): 808 (M+H+).
Synthesis of 1-112
[00894] To a solution of (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5-carboxamido)- 7-(2-morpholinoethoxy)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylic acid, 121.6 (100 mg, 0.12 mmol, 1.0 equiv), DIEA (80 mg, 0.62 mmol, 5.0 equiv) and HATE! (68 mg, 0.18 mmol, 1.5 equiv) in dimethyl formamide (5 mL) was added ammonium chloride (64 mg, 1.2 mmol, 10 equiv). The reaction mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (22% ACN up to 36% in 10 min); Detector, UV 254/220 nm. The crude product (80 mg) was purified by Prep-HPLC with the following conditions: Column SunFire Prep C18 Column 30* 150mm, 5um; mobile phase, Water (0.1% FA) and CAN (15% ACN up to 36% in 7 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 35.6 mg (36.76%) of (E)-N-(5-carbamoyl-l-(4-(5- carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2- en-l-yl)-7-(2-morpholinoethoxy)-lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5- carboxamide as a white solid. LC-MS (ES, m/z): 807 (M+H+); 1H NMR (400 MHz, DMSO-tfe) d 8.07-7.91 (m, 3H), 7.75-7.62 (m, 2H), 7.49-7.32 (m, 4H), 6.54 (s, 1H), 6.09-5.97 (m, 1H), 5.73- 5.61 (m, 1H), 5.02-4.78 (m, 4H), 4.68-4.45 (m, 4H), 3.62-3.32 (m, 10H), 2.81 (q, 2H), 2.40 (s, 3H), 2.13 (s, 3H), 1.26 (t, 3H), 1.02 (t, 3H).
Example 122: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-(2-hydroxyethoxy)- lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-111
Figure imgf000511_0001
Figure imgf000512_0001
1-111
Synthesis of 122.1
[00895] To a solution of 4-chloro-3 -hydroxy-5 -nitrobenzamide (1.08 g, 5 mmol, 1 equiv) and 2-bromoethan-l-ol (1.23 g, 10 mmol, 2 equiv) in dimethyl formamide (20 mL) was added potassium carbonate (2.07 g, 15 mmol, 3 equiv). The resulting mixture was stirred at 100°C overnight. The mixture was cooled to RT, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with dichloromethane/methanol (10/1) to give 700 mg (53.85%) of 4-chloro-3- (2-hydroxyethoxy)-5-nitrobenzamide a yellow solid. LC-MS (ES, m/z): 261 (M+H+).
Synthesis of 122.2
[00896] To a stirred solution of methyl (E)-l-(4-aminobut-2-en-l-yl)-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate hydrochloride, 40.6 (1.14 g, 2.19 mmol, 1.0 equiv) in n-BuOH (20 mL) was added sodium bicarbonate (368 mg, 4.38 mmol, 2.0 equiv) and DIEA (1.41 g, 10.95 mmol, 5.0 equiv). After 10 min, was added 4-chloro-3-(2- hydroxyethoxy)-5-nitrobenzamide, 122.1 (570 mg, 2.19 mmol, 1.0 equiv). The resulting mixture was stirred overnight at 120°C under nitrogen. The mixture was cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 770 mg (56.6%) of methyl (E)-l-(4-((4-carbamoyl-2-(2-hydroxyethoxy)-6- nitrophenyl)amino)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxylate as a red solid. LC-MS (ES, m/z): 621 (M+H+). Synthesis of 122.3
[00897] To a stirred solution of methyl (E)-l-(4-((4-carbamoyl-2-(2-hydroxyethoxy)-6- nitrophenyl)amino)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxylate, 122.2 (770 mg, 1.13 mmol, 1.0 equiv) in methanol (20 mL) and water (5 mL) was added sodium dithionite (1.96 g, 11.3 mmol, 10.0 equiv) ammonia water (2 mL). The resulting mixture was stirred at RT for 2h. The mixture was diluted with water, extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. This resulted in 587 mg (88%) of methyl (E)-l-(4-((2- amino-4-carbamoyl-6-(2-hydroxyethoxy)phenyl)amino)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate an off- red solid. LC-MS (ES, m/z): 591 (M+H+).
Synthesis of 122.4
[00898] To a solution of methyl (E)-l-(4-((2-amino-4-carbamoyl-6-(2- hydroxyethoxy)phenyl)amino)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)- lH-benzo[d]imidazole-5-carboxylate, 122.3 (587 mg, 0.99 mmol, 1.0 equiv) in methanol (10 mL) was added cyanogen bromide (210 mg, 2 mmol, 2 equiv). The resulting mixture was stirred at RT for overnight. The resulting mixture was concentrated under reduced pressure. This resulted in 650 mg (crude) of methyl (E)-l-(4-(2-amino-5-carbamoyl-7-(2-hydroxyethoxy)-lH- benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxylate as a yellow solid. LC-MS (ES, m/z): 616 (M+H+).
Synthesis of 122.5
[00899] To a solution of 4-ethyl-2-methyloxazole-5-carboxylic acid (232.5 mg, 1.5 mmol, 1.5 equiv), DIEA (645 mg, 5 mmol, 5.0 equiv) and HATU (570 mg, 1.5 mmol, 1.5 equiv) in dimethyl formamide (5 mL) was added methyl (E)-l-(4-(2-amino-5-carbamoyl-7-(2-hydroxyethoxy)-lH- benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxylate, 122.4 (650 mg crude, 0.99 mmol, 1.0 equiv). The resulting mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (39% ACN up to 51% in 10 min); Detector, UV 254/220 nm. This resulted in 385 mg (51.13%) of methyl (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-
512
5UB5TITUTE SHEET (RULE 26) 5-carboxamido)-7-(2-hydroxyethoxy)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3- methyl- lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate as a yellow solid. LC-MS (ES, m/z): 753 (M+H+).
Synthesis of 122.6
[00900] To a solution of methyl (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5- carboxamido)-7-(2-hydroxyethoxy)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate, 122.5 (385 mg, 0.511 mmol, 1.0 equiv) in methanol (20 mL) and water (4 mL) was added lithium hydroxide (61.32 mg, 2.555 mmol, 5 equiv). The resulting mixture was stirred at 70°C for 2 hour. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration. This resulted in 210 mg (55.7%) of (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5- carboxamido)-7-(2-hydroxyethoxy)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylic acid as a yellow solid. LC-MS (ES, m/z): 739 (M+H+).
Synthesis of 1-111
[00901] To a solution of (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5-carboxamido)- 7-(2-hydroxyethoxy)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylic acid, 122.6 (100 mg, 0.14 mmol, 1.0 equiv), DIEA (87 mg, 0.68 mmol, 5.0 equiv) and HATU (77 mg, 0.20 mmol, 1.5 equiv) in dimethyl formamide (5 mL) was added ammonium chloride (36 mg, 0.68 mmol, 5 equiv). The reaction mixture was stirred at RT overnight. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (26% ACN up to 38% in 10 min); Detector, UV 254/220 nm. The crude product (70 mg) was purified by Prep-HPLC with the following conditions: Column SunFire prep C18 Column 30* 150mm, 5um; mobile phase, Water (0.1% FA) and CAN (18% ACN up to 40% in 7 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 33.3 mg (32.23%) of (E)-N-(5-carbamoyl-l-(4-(5- carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2- en-l-yl)-7-(2-hydroxyethoxy)-lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5- carboxamide as a white solid. LC-MS (ES, m/z): 738 (M+H+); 1H NMR (400 MHz, DMSO-t/s) d 12.74 (br s, 2H), 8.03-7.91 (m, 3H), 7.70 (dd, 1H), 7.64 (s, 1H), 7.47-7.32 (m, 4H), 6.55 (s, 1H), 6.09-5.97 (m, 1H), 5.88-5.79 (m, 1H), 5.02-4.78 (m, 4H), 4.53 (q, 2H), 4.08 (t, 2H), 3.65 (t, 2H), 2.83 (q, 2H), 2.40 (s, 3H), 2.12 (s, 3H), 1.27 (t, 3H), 1.02 (t, 3H).
Example 123: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(4-ethyl-2- methyloxazole-5-carboxamido)-7-(3-morpholinopropoxy)-lH-benzo[d]imidazol-l-yl)biit-2- en-l-yl)-7-methoxy-lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, I- 160
Figure imgf000515_0001
Figure imgf000516_0001
Synthesis of 123.1
[00902] To a stirred solution of 4-chloro-3-hydroxy-5-nitrobenzamide (2.00 g, 9.234 mmol, 1.00 equiv) in DMSO (25.00 mL) in a 50 mL 3-necked round-bottom flask, 4-(3- bromopropyl)morpholine (2.88 g, 13.852 mmol, 1.5 equiv) and Cs2CO3(9.03 g, 27.703 mmol, 3 equiv) were added at room temperature. The resulting mixture was stirred for 2h at 80°C under nitrogen atmosphere. The resulting mixture was extracted with EtOAc (3x25 mL). The combined organic layers were washed with brine (3x25 mL), dried over anhydrous NaiSCL. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeOH in water, 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 4-chloro-3-[3- (morpholin-4-yl)propoxy]-5-nitrobenzamide (1 g, 28.67%) (ES, m/z): [M+H] 344.1 as an off- white solid.
Synthesis of 123.2
[00903] To a stirred solution of 4-chloro-3-(3-hydroxypropoxy)-5-nitrobenzamide, 123.1 (980 mg, 3.641 mmol, 1.00 equiv) in i-PrOH (20.00 mL) in a 50 mL 3-necked round-bottom flask, NaHCC (0.61 g, 7.282 mmol, 2 equiv), DIEA (2.82 g, 21.845 mmol, 6 equiv) and 4-[[(2E)-4- aminobut-2-en-l-yl]amino]-3-methoxy-5-nitrobenzamide (1.02 g, 3.639 mmol, 1.00 equiv) were added in portions at room temperature. The resulting mixture was stirred for overnight at 120°C under nitrogen atmosphere. The resulting mixture was filtered, and the filter cake was washed with i-PrOH (3x10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (CTLCHMeOHMO: 1) to afford 4-[[(2E)-4-[[4-carbamoyl-2-(3-hydroxypropoxy)-6- nitrophenyl]amino]but-2-en-l-yl]amino]-3-methoxy-5-nitrobenzamide (370 mg, 19.15%) (ES, m/z): [M+H]+ 588.2 as an off-white solid.
Synthesis of 123.3
[00904] To a stirred solution of 4-[[(2E)-4-([4-carbamoyl-2-[3-(morpholin-4-yl)propoxy]-6- nitrophenyl]amino)but-2-en-l-yl]amino]-3-methoxy-5-nitrobenzamide, 123.2 (360.00 mg, 0.613 mmol, l .OO equiv) in MeOH (9.00 mL) in a 20 mL vial, Na2S2C>4 (1066.71 mg, 6.127 mmol, 10.00 equiv) and ammonia (260.85 mg, 15.317 mmol, 25 equiv) and TbO (7.20 mL) were added in portions at room temperature. The resulting mixture was stirred for lh at room temperature under nitrogen atmosphere. The resulting mixture was filtered, and the filter cake was washed with MeOH (3x3 mL). The filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (CH2Cl2:MeOH=5:l) to afford 3-amino-4-[[(2E)-4-([2-amino-4-carbamoyl-6-[3- (morpholin-4-yl)propoxy]phenyl]amino)but-2-en-l-yl]amino]-5-methoxybenzamide (75 mg, 11.60%) (ES, m/z): [M+H]+ 528.3 as an off-white solid.
Synthesis of 123.4
[00905] To a stirred solution of 3-amino-4-[[(2E)-4-([2-amino-4-carbamoyl-6-[3-(morpholin- 4-yl)propoxy]phenyl]amino)but-2-en-l-yl]amino]-5-methoxybenzamide, 123.3 (70.00 mg, 0.133 mmol, 1.00 equiv) in MeOH (3.00 mL) in an 8 mL vial, BrCN(14.05 mg, 0.133 mmol, 1 equiv) was added in portions at room temperature. The resulting mixture was stirred for lh at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was precipitated by the addition of 2 mL MeOH. This resulted in 2- amino-l-[(2E)-4-[2-amino-5-carbamoyl-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazol-l- yl]but-2-en-l-yl]-7-methoxy-l,3-benzodiazole-5-carboxamide(75 mg) (ES, m/z): [M+H]+ 578.3 as an off-white solid.
Synthesis of 1-160
[00906] To a stirred solution of 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid, 123.4 (94.01 mg, 0.606 mmol, 5.00 equiv) in NMP (2.00 mL) in an 8 mL sealed tube, HATU (184.31 mg, 0.485 mmol, 4 equiv), DIEA (187.94 mg, 1.454 mmol, 12 equiv) and 2-amino- l-[(2E)-4-[2-amino-5-
516
5UB5TITUTE SHEET (RULE 26) carbamoyl-7-[3-(morpholin-4-yl)propoxy]-l,3-benzodiazol-l-yl]but-2-en-l-yl]-7-methoxy-l,3- benzodiazole-5-carboxamide (70.00 mg, 0.121 mmol, 1.00 equiv) were added at room temperature. The resulting mixture was irradiated for 1.5 h at 140°C under nitrogen atmosphere. The resulting mixture was diluted with water (5 mL). The precipitated solids were collected by filtration and washed with water (3x1 mL). The crude product was purified by Prep-HPLC with the following conditions (Column: SunFire Cl 8 OBD Prep Column lOOA, 5 pm, 19x250 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 17% B to 33% B in 7 min; 254/220 nm; RT: 6.63 min) to afford 1-160 (3.0 mg, 2.78%) as a white solid. LC-MS (ES, in r): [M+H]+: 852.5; 1H-NMR:(300 MHz, DMSO-de, ppm ) 58.05-7.97 (brs, 2H), 7.65-7.62 (m, 2H), 7.45-7.13 (m, 4H), 5.85-5.71 (m, 2H), 4.95-4.80 (m, 4H), 4.01-3.88 (m, 2H), 3.75-3.70 (d, 3H), 3.49-3.46 (m, 4H), 2.89-2.71 (m, 4H), 2.41-2.39 (d, 6H), 2.28-2.13 (m, 6H), 1.71-1.55 (m, 2H), 1.09-0.91 (m, 6H).
Example 124: Synthesis of (E)-N,N'-(12-carbamoyl-4-(4-(2-hydroxyethyl)piperidine-l- carbonyl)-8,9,16,19-tetrahydro-7H-6,10-dioxa-2,14,15a,19a-tetraazacyclopentadeca[3,2,l- cd:8,9,10-c'd']diindene-l,15-diyl)bis(4-ethyl-2-methyloxazole-5-carboxamide), 1-161
Figure imgf000518_0001
[00907] To a stirred solution of (3E)-21-carbamoyl-7,25-bis(4-ethyl-2-methyl-l,3-oxazole-5- amido)-14,18-dioxa-l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylic acid, 1-78 (100 mg, 0.133 mmol, 1 equiv) and 4-piperidineethanol (51.56 mg, 0.399 mmol, 3 equiv) in N,N-dimethylformamide (6 mL) were added HATU (101.16 mg, 0.266 mmol, 2 equiv) and DIEA (85.96 mg, 0.665 mmol, 5 equiv). The resulting mixture was stirred for 2 h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep C18 column, 30*150, 5um; mobile phase, water (0.1%FA) and acetonitrile (25% acetonitrile up to 40% in 8 min); Detector, UV: 254 nm. This resulted in 10.4 mg (9.06%) of (3E)-7,25-bis(4-ethyl-2-methyl-l,3-oxazole-5-amido)-21-[4-(2-hydroxyethyl)piperidine-l- carbonyl]- 14, 18-dioxa- 1 ,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l 1-carboxamide as an off-white solid. LC-MS (ESI): 863 (M+EC). ¾ NMR: ¾ NMR (400 MHz, DMSO-i/e) d 12.80 (s, 1H), 12.70 (s, 1H), 7.98 (s, 1H), 7.69 (s, 1H), 7.50 (s, 1H), 7.36 (s, 1H), 7.21 (s, 1H), 6.99 (s, 1H), 5.55-5.50 (m, 2H), 4.86- 4.85 (m, 4H), 4.47-4.37 (m, 4H), 3.49-3.38 (m, 6H), 2.83 (q, 4H), 2.39 (s, 6H), 2.27-2.26 (m, 2H), 1.79-1.69 (m, 3H), 1.44-1.39 (m, 2H), 1.14-1.01 (m, 8H).
Example 125: Synthesis of (E)-l,15-bis(4-ethyl-2-methyloxazole-5-carboxamido)-N-(2- hydroxyethyl)-8,9,l 6, 19-tetrahydro-7H-6,l 0-dioxa-2, 14,15a, 19a- tetraazacyclopentadeca[3, 2, l-cd:8, 9, 10-c'd']diindene-4, 12-dicarboxamide, 1-162
Figure imgf000519_0001
[00908] To a stirred mixture of (3Z)-21-carbamoyl-7,25-bis(4-ethyl-2-methyl-l,3-oxazole-5- amido)-14,18-dioxa-l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3 ,7,9, 11 , 13 (27), 19,21 ,23 (26),24-nonaene- 11 -carboxylic acid, 1-78 (100 mg, 0.133 mmol, 1 equiv) and ethanolamine (24.4 mg, 0.4 mmol, 3 equiv) in N,N-dimethylformamide (6 mL) were added HATU (101.16 mg, 0.266 mmol, 2 equiv) and DIEA (85.96 mg, 0.665 mmol, 5 equiv). The resulting mixture was stirred for 2 h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep C18 column, 30* 150mm, 5um; mobile phase, Water (0.1%FA) and ACN (25% ACN up to 46% in 7 min); Detector, 254/220 nm. This resulted in 21.9 mg (20.74 %) of (3Z)-7,25-bis(4-ethyl-2-methyl-l,3-oxazole-5-amido)-Nl l-(2-hydroxyethyl)-14,18-dioxa- l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3 ,7,9, 11 , 13 (27), 19,21 ,23 (26),24-nonaene- 11 ,21 -dicarboxamide as an off-white solid. LC-MS (ES, m/z): 795 (M+H+); Tl NMR: 1HNMR (400 MHz, DMSO -d6) d 12.76 (br s, 2H), 8.44 (t, 1H), 7.98 (s, 1H), 7.72-7.68 (m, 2H), 7.54-7.50 (m, 2H), 7.35 (s, 1H), 5.56-5.42 (m, 2H), 5.05-4.60 (m, 5H), 4.49-4.38 (m, 4H), 3.62-3.40 (m, 4H), 2.82 (q, 4H), 2.46-2.25 (m, 8H), 1.02 (t, 6H).
Example 126: Synthesis of (E)-N,N'-(4-carbamoyl-12-(piperazine-l-carbonyl)-8,9,16,19- tetrahydro-7H-6,10-dioxa-2,14,15a,19a-tetraazacyclopentadeca[3,2,l-cd:8,9,10- c'd']diindene-l,15-diyl)bis(4-ethyl-2-methyloxazole-5-carboxamide), 1-163
Figure imgf000520_0001
[00909] To a solution of (E)-4-carbamoyl-l,15-bis(4-ethyl-2-methyloxazole-5-carboxamido)- 8,9, 16, 19-tetrahydro-7H-6, 10-dioxa-2, 14, 15 a, 19a-tetraazacyclopentadeca[3 ,2, 1 -cd: 8,9, 10- c'd']diindene-12-carboxylic acid, 1-78 (100 mg, 0.13 mmol, 1.0 equiv) and piperazine (22.36 mg, 0.26 mmol, 2 equiv) in dimethyl formamide (5 mL) was added DIEA (86 mg, 0.67 mmol, 5.0 equiv) and HATU (76 mg, 0.2 mmol, 1.5 equiv). The resulting mixture was stirred at 50°C for 2 h. The reaction was cooled to RT and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire prep C18 Column 30* 150mm, 5um; mobile phase, Water (0.1%FA) and ACN (15% ACN up to 42% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 11.4 mg (11%) of (E)-N,N'-(4-carbamoyl-12-(piperazine-l- carbonyl)-8,9, 16, 19-tetrahydro-7H-6, 10-dioxa-2, 14, 15 a, 19a-tetraazacyclopentadeca[3 ,2, 1 - cd:8,9,10-c'd']diindene-l, 15-diyl)bis(4-ethyl-2-methyloxazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 411 (M/2+H+); ¾ NMR (400 MHz, DMSC ) d 7.60 (s, 1H), 7.42 (s, 1H), 7.19 (s, 1H), 7.01 (s, 1H), 5.48-5.31 (m, 2H), 4.97-4.69 (m, 4H), 4.45-4.28 (m, 4H), 3.88-3.35 (m, 6H), 3.22-3.11 (m, 2H), 2.80-2.64 (m, 4H), 2.35 (s, 6H), 2.28-2.18 (m, 2H), 0.94 (t, 6H).
Example 127: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(4-ethyl-2- methyloxazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7- (morpholinomethyl)-lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, I- 164
Figure imgf000521_0001
Boc
Figure imgf000522_0001
Synthesis of 127.1
[00910] Into a 250-mL 3-necked round-bottom flask, was placed methyl 4-chloro-3- methylbenzoate (12.00 g, 64.998 mmol, 1.00 equiv), H2SO4 (25.00 mL). To the above mixture HNO3 (25.00 mL) was added. The resulting solution was stirred for 2h at 50 degrees C under nitrogen atmosphere. The resulting solution was diluted with 50 mL of H2O. The solids were collected by fdtration. This resulted in 10 g (67.00%) cmde of 127.1 as a light yellow solid. Synthesis of 127.2
[00911] Into a 500-mL 3-necked round-bottom flask, was placed 127.1 (10.00 g, 43.550 mmol, 1.00 equiv), CCL (150.00 mL), NBS (9.3 g, 52.260 mmol, 1.20 equiv). The resulting solution was stirred for 12h at 80 degrees C under nitrogen atmosphere. The solids were fdtered out. The resulting mixture was concentrated under vacuum. This resulted in 7.9 g crude of methyl 3- (bromomethyl)-4-chloro-5-nitrobenzoate as brown oil.
Synthesis of 127.3
[00912] Into a 250-mL 3-necked round-bottom flask, was placed 127.2 (7.9 g, 25.607 mmol, 1.00 equiv), DCM (105.00 mL), DIEA (19.857 g, 153.642 mmol, 6.00 equiv), morpholine (1115.46 mg, 12.803 mmol, 0.50 equiv). The resulting solution was stirred for 12h at 25 degrees C under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The crude product was purified by FI ash-Prep -HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACNiFbO =10% increasing to ACN:FhO=50% within 15 min; Detector, UV 254 nm. This resulted in 3.6 g (38.12%) of 127.3 as brown oil. LC-MS (ES, m/z): 315 [M+H]+.
Synthesis of 127.4
[00913] Into a 500-mL 3-necked round-bottom flask, was placed methyl 4-chloro-3- (morpholin-4-ylmethyl)-5-nitrobenzoate, 127.3 (3.60 g, 11.121 mmol, 1.00 equiv), ammonia (200.00 mL). The resulting solution was stirred for 12h at 50 degrees C. The solids were collected by filtration. This resulted in 3.3 g (99.01%) of 4-chloro-3-(morpholin-4-ylmethyl)-5- nitrobenzamide as a yellow solid. LC-MS (ES, m/z): 300 [M+H]+.
Synthesis of 127.5
[00914] Into a 250-mL 3-necked round-bottom flask, was placed 127.4 (3.3 g, 11.544 mmol, 1.00 equiv), DMSO (33 mL), DIEA (8.95 g, 69.267 mmol, 6 equiv), tert-butyl N-[(2E)-4- aminobut-2-en-l-yl]carbamate (3.225 g, 17.317 mmol, 1.5 equiv). The resulting solution was stirred for 12h at 100 degrees C under nitrogen atmosphere. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACINfFhO =10% increasing to ACN:H2O=50% within 20 min; Detector, UV 254 nm. This resulted in 1.9 g of 127.5 as a yellow solid. LC-MS (ES, m/z): 450 [M+H]+.
Synthesis of 127.6
[00915] Into a 100-mL 3-necked round-bottom flask, was placed tert-butyl N-[(2E)-4-[[4- carbamoyl-2-(morpholin-4-ylmethyl)-6-nitrophenyl]amino]but-2-en-l-yl]carbamate, 127.5 (1.90 g, 4.227 mmol, 1.00 equiv), DCM (30.00 mL) and TFA (10 mL). The resulting solution was stirred for 12h at 25 degrees C. The resulting mixture was concentrated under vacuum. This resulted in 1.6 g of 127.6 as a brown oil. LC-MS (ES, m/z): 350 [M+H]+.
Synthesis of 127.7
[00916] Into a 100-mL 3-necked round-bottom flask, was placed 4-[[(2E)-4-aminobut-2-en-l- yl]amino]-3-(morpholin-4-ylmethyl)-5-nitrobenzamide, 127.6 (640.00 mg, 1.832 mmol, 1.00 equiv), DMSO (9 mL), DIEA (1.42 g, 10.991 mmol, 6.00 equiv), 4-fluoro-3-nitrobenzamide (505.91 mg, 2.748 mmol, 1.50 equiv). The resulting solution was stirred for 12h at 60 degrees C under nitrogen atmosphere. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN:H2O=10% increasing to ACN:FhO=50% within 20 min; Detector, UV 254 nm to yield 127.6 (410 mg) as a yellow solid. LC-MS (ES, m/z): 514[M+H]+.
Synthesis of 127.8
[00917] Into a 50-mL 3-necked round-bottom flask, was placed 127.7 (380.00 mg, 0.740 mmol, 1.00 equiv), EtOH (5.00 mL), N2H4Ή2O (555.00 mg, 11.100 mmol, 15 equiv), Pd/C (23.63 mg, 0.222 mmol, 0.3 equiv). The resulting solution was stirred for 30 min at 25 degrees C under nitrogen atmosphere. The solids were filtered out. The resulting mixture was concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN:H20 = 10% increasing to ACN:H2O=50% within 20 min; Detector, UV 254 nm to yield 210 mg of a white solid. LC-MS (ES, m/z): 454 [M+H]+.
Synthesis of 127.9
[00918] Into a 50-mL 3-necked round-bottom flask, was placed 3-amino-4-[[(2E)-4-[(2-amino- 4-carbamoylphenyl)amino]but-2-en-l-yl]amino]-5-(morpholin-4-ylmethyl)benzamide, 127.8 (200.00 mg, 0.441 mmol, 1.00 equiv), MeOH (5.00 mL), BrCN (280.25 mg, 2.646 mmol, 6 equiv). The resulting solution was stirred for 5h at 25 degrees C under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. This resulted in 305 mg (135.10%) crude of 2-amino-l- [(2E)-4-(2-amino-5-carbamoyl-l,3-benzodiazol-l-yl)but-2-en-l-yl]-7-(morpholin-4-ylmethyl)- l,3-benzodiazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 504 [M+H] .
Synthesis of 1-164 [00919] Into a 50-mL 3-necked round-bottom flask, was placed 4-ethyl -2 -methyl- 1,3 -oxazole- 5-carboxylic acid (169.46 mg, 1.092 mmol, 5.00 equiv), DMF (15.00 mL), HATU (830.58 mg, 2.184 mmol, 10.00 equiv), DIEA (564.64 mg, 4.369 mmol, 20.00 equiv), 2-amino-l-[(2E)-4-(2- amino-5-carbamoyl-l,3-benzodiazol-l-yl)but-2-en-l-yl]-7-(morpholin-4-ylmethyl)-l,3- benzodiazole-5-carboxamide (110.00 mg, 0.218 mmol, 1.00 equiv). The resulting solution was stirred for 12h at 25 degrees C under nitrogen atmosphere. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN:H2O=10% increasing to ACN:H2O=50% within 25 min; Detector, UV 254 nm. The crude product was purified by Prep-HPFC with the following condition (Column: Xselect CSH OBD Column 30* 150 mm 5 urn, n; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mF/min; Gradient: 13% B to 28% B in 7 min; 254/220 nm; RT:6.97 min). This resulted in 0.8 mg (0.47%) of 1-164 as an off-white solid. LC-MS: (ES, m/z): [M+H]+ 778.5; 1H-
NMR: (400 MHz, DMSO -d6, ppm) 512.69-12.89 (brs, 2H), 58.27 (s, 1H), 57.90-8.00 (m, 4H), 57.72-7.74 (d, 1H), 57.57 (s, 1H), 57.43-7.44 (d, 1H), 57.33-7.37 (m, 1H), 55.95-5.99 (d, 1H),
55.40-5.43 (d, 1H), 55.18 (s, 2H), 54.78-4.83 (m, 2H), 53.48-3.50 (m, 6H), 52.80-2.82 (d, 4H),
52.42 (s, 6H), 52.24 (s, 4H), 51.01-1.02 (d, 6H).
Example 128: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(l-(3-hydroxypropyl)- 3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-lH- benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-165
Figure imgf000525_0001
Figure imgf000526_0001
128.3 1-165
Synthesis of 128.1
[00920] To a stirred solution of methyl 2-amino-l-[(2E)-4-[2-amino-5-(methoxycarbonyl)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazole-5-carboxylate, 66.5 (500.00 mg, 1.150 mmol, 1.00 equiv) and 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (178.00 mg, 1.150 mmol, 1.00 equiv) in dimethyl formamide (5.00 mL) was added DIEA (742.00 mg, 5.750 mmol, 5.00 equiv) and HATU (481.00 mg, 1.270 mmol, 1.10 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 120°C. The mixture was cooled to RT and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (WANTLdTCCri) and ACN (30% ACN up to 60% in 10 min); Detector, UV 254/220 nm. This resulted in 200 mg (33.4%) of methyl 2-amino-l-[(2E)-4-[2-(4- ethyl-2-methyl-l,3-oxazole-5-amido)-5-(methoxycarbonyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]- l,3-benzodiazole-5-carboxylate as a light yellow solid. LC-MS (ES, m/z): 572 (M+H+).
Synthesis of 128.2 [00921] To a stirred solution of methyl 2-amino-l-[(2E)-4-[2-(4-ethyl-2-methyl-l,3-oxazole-5- amido)-5-(methoxycarbonyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodi azole-5- carboxylate, 128.1 (200 mg, 0.35 mmol, 1.00 equiv) and 2-[3-[(tert- butyldimethylsilyl)oxy]propyl]-5-methylpyrazole-3-carboxylic acid (132.00 mg, 0.440 mmol, 1.20 equiv) in dimethyl formamide (5 mL) was added DIEA (239.00 mg, 1.850 mmol, 5.00 equiv) and HATU (183.00 mg, 0.480 mmol, 1.30 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 120°C. The mixture was cooled to RT and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (10% NH4HCC>3) and ACN (25% ACN up to 51% in 10 min); Detector, UV 254/220 nm. This resulted in 200 mg (67.3%) of methyl l-[(2E)-4-[2-(2-[3-[(tert- butyldimethylsilyl)oxy]propyl]-5-methylpyrazole-3-amido)-5-(methoxycarbonyl)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazole-5- carboxylate as a light yellow solid. LC-MS (ES, m/z): 852 (M+TG).
Synthesis of 128.3
[00922] To a stirred solution of methyl l-[(2E)-4-[2-(2-[3-[(tert- butyldimethylsilyl)oxy]propyl]-5-methylpyrazole-3-amido)-5-(methoxycarbonyl)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazole-5- carboxylate, 128.2 (200.00 mg, 0.234 mmol, 1.00 equiv) in methanol (20 mL) and water (4 mL) was added lithium hydroxide (34.00 mg, 1.4 mmol, 6.00 equiv). The resulting solution was stirred for 4 h at 70 °C. The mixture was cooled to RT and concentrated under reduced pressure. The residue was acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration. This resulted in 150 mg (90.4%) of 1- [(2E)-4-[5-carboxy-2-[2-(3-hydroxypropyl)-5-methylpyrazole-3-amido]-l,3-benzodiazol-l- yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazole-5-carboxylic acid as an off-white solid. LC-MS (ES, m/z): 710 (M+H+).
Synthesis of 1-165
[00923] To a stirred solution of l-[(2E)-4-[5-carboxy-2-[2-(3-hydroxypropyl)-5- methylpyrazole-3-amido]-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole- 5-amido)-l,3-benzodiazole-5-carboxylic acid, 128.3 (150 mg, 0.211 mmol, 1.00 equiv) and ammonium chloride (57 mg, 1.055 mmol, 5 equiv) in dimethyl formamide (6 mL) was added DIEA (272.2 mg, 2.11 mmol, 10.00 equiv) and HATU (160.36 mg, 0.422 mmol, 2 equiv). The resulting solution was stirred for 4 h at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (22% ACN up to 43% in 10 min); Detector, UV 254/220 nm. The crude product (60 mg) was purified by Prep-HPLC with the following conditions: Column, Sunfire prep C18 column, 30* 150mm, 5um; mobile phase, Water (0.1%FA) and ACN (10% ACN up to 38% in 7 min); Detector, UV 254/210 nm. This resulted in 18.8 mg (12.6%) of l-[(2E)-4-[5-carbamoyl-2-[2-(3-hydroxypropyl)-5-methylpyrazole-3-amido]- l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3- benzodiazole-5-carboxamide as a white solid.LC-MS (ES, m/z): 708 (M+FE); TlNMR: Tl NMR (400 MHz, DMSO-t/e) d 12.69 (br s, 2H), 8.03-7.92 (m, 4H), 7.71 (dd, 2H), 7.43 (dd, 2H), 7.32 (s, 2H), 6.57 (s, 1H), 5.98-5.88 (m, 2H), 4.92-4.81 (m, 4H), 4.68-4.45 (m, 3H), 3.41 (t, 2H), 2.85 (q, 2H), 2.41 (s, 3H), 2.13 (s, 3H), 1.97-1.87 (m, 2H), 1.07 (t, 3H).
Example 129: Synthesis of (E)-l-(4-ethyl-2-methyloxazole-5-carboxamido)-15-(l-(2-hydroxy ethyl)-3-methyl-lH-pyrazole-5-carboxamido)-8,9,16,19-tetrahydro-7H-6,10-dioxa-2,14,15a, 19a-tetraazacyclopentadeca[3, 2, l-cd:8, 9, 10-c'd']diindene-4, 12-dicarboxamide, 1-166
Figure imgf000529_0001
Synthesis of 129.1
[00924] To a solution of l-(2-((tert-butyldimethylsilyl)oxy)ethyl)-3-methyl-lH-pyrazole-5- carboxylic acid, 32.5 (21 mg, 0.075 mmol, 1.5 equiv) and DIEA (33 mg, 0.25 mmol, 5.0 equiv) and HATU (30 mg, 0.075 mmol, 1.5 equiv) in dimethyl formamide (3 mL) was added methyl (E)- l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5-carboxamido)-7-(2-morpholinoethoxy)-lH- benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxylate (30 mg, 0.05 mmol, 1.0 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 120°C. The reaction was cooled to RT and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (41% ACN up to 52% in 10 min); Detector, UV 254/220 nm. This resulted in 20 mg (45.45%) of (E)-15-(l-(2- ((tert-butyldimethylsilyl)oxy)ethyl)-3-methyl-lH-pyrazole-5-carboxamido)-l-(4-ethyl-2- methyloxazole-5-carboxamido)-8,9, 16, 19-tetrahydro-7H-6, 10-dioxa-2, 14, 15 a, 19a- tetraazacyclopentadeca[3,2, l-cd:8,9,10-c'd']diindene-4, 12-dicarboxamide as a yellow solid. LC- MS (ES, m/z): 880 (M+H+). Synthesis of 1-166
[00925] To a solution of (E)-15-(l-(2-((tert-butyldimethylsilyl)oxy)ethyl)-3-methyl-lH- pyrazole-5-carboxamido)-l-(4-ethyl-2-methyloxazole-5-carboxamido)-8,9,16, 19-tetrahydro-7H- 6, 10-dioxa-2,14, 15a, 19a-tetraazacyclopentadeca[3,2,l-cd:8,9, 10-c'd']diindene-4, 12- di carboxamide, 129.1 (20 mg, 0.0227 mmol, 1.0 equiv) in dichloromethane (5 mL) was added trifluoroacetic acid (1 mL). The resulting mixture was stirred for 2h at RT. The resulting mixture concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (24% ACN up to 33% in 10 min); Detector, UV 254/220 nm. The crude product (6.8 mg) was purified by Prep- HPLC with the following conditions: Column XSelect CSH Prep C18 OBD Column 19*250mm, 5um; mobile phase, Water (0.1%FA) and ACN (20% ACN up to 35% in 10 min); The product- containing fractions were collected and evaporated partially and lyophilized overnight to afford 0.3 mg (1.73%) of (E)-l-(4-ethyl-2-methyloxazole-5-carboxamido)-15-(l-(2-hydroxyethyl)-3- methyl- lH-pyrazole-5-carboxamido)-8,9, 16, 19-tetrahydro-7H-6, 10-dioxa-2, 14, 15a, 19a- tetraazacyclopentadeca[3,2, l-cd:8,9,10-c'd']diindene-4, 12-dicarboxamide as a white solid. LC- MS (ES, m/z): 766 (M+H+); 1H NMR (400 MHz, DMSO-t4) d 7.66 (s, 2H), 7.47 (s, 2H), 6.54 (s, 1H), 5.61-5.45 (m, 2H), 4.95-4.82 (m, 4H), 4.57 (t, 2H), 4.44-4.38 (m, 4H), 3.80-3.75 (m, 2H), 2.81 (q, 2H), 2.41-2.27 (m, 5H), 2.07 (s, 3H), 1.00 (t, 3H).
Example 130: Synthesis of (E)-4-carbamoyl-l-(4-ethyl-2-methyloxazole-5-carboxamido)-15- (l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-8,9,16,19-tetrahydro-7H-6,10-dioxa- 2,14,15a,19a-tetraazacyclopentadeca[3,2,l-cd:8,9,10-c'd']diindene-12-carboxylic acid, 1-167
NH2
Figure imgf000530_0001
120.3 130.1
Figure imgf000531_0001
Figure imgf000532_0001
Synthesis of 130.1
[00926] To a stirred solution of 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (2.03 g, 13.114 mmol, 2 equiv) in DMF (30.00 mL) was added HATU (4.98 g, 13.114 mmol, 2 equiv), DIEA (4.23 g, 32.785 mmol, 5 equiv) and tert-butyl N-[(2E)-4-[2-amino-5-carbamoyl-7-(3-hydroxypropoxy)- l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate, 120.3 (2.75 g, 6.557 mmol, 1.00 equiv). The resulting solution was stirred overnight at room temperature. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (61% ACN up to 70% in 10 min); Detector, UV 254/220 nm. This resulted in 2.8 g (61.54%) of (E)-3-(3-(4-(tert- butoxycarbonylamino)but-2-enyl)-6-carbamoyl-2-(4-ethyl-2-methyloxazole-5-carboxamido)-3H- benzo[d]imidazol-4-yloxy)propyl 4-ethyl-2-methyloxazole-5-carboxylate as a yellow solid. LCMS (ES, m/z): 694 (M+H+).
Synthesis of 130.2
[00927] To a stirred solution of (E)-3-(3-(4-(tert-butoxycarbonylamino)but-2-enyl)-6- carbamoyl-2-(4-ethyl-2-methyloxazole-5-carboxamido)-3H-benzo[d]imidazol-4-yloxy)propyl 4- ethyl-2-methyloxazole-5-carboxylate, 130.1 (2.80 g, 4.04 mmol, 1.00 equiv) in methanol (15 mL) and water (5 mL) was added lithium hydroxide (484.8 mg, 20.2 mmol, 5.00 equiv). The resulting mixture was stirred overnight at room temperature. The mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 1.7 g (75.56%) of tert-butyl N-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(3- hydroxypropoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate as a yellow solid. LCMS (ES, m/z): 557 (M+H+). Synthesis of 130.3
[00928] To a stirred solution of tert-butyl N-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-(3-hydroxypropoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate, 130.2 (1.3 g, 2.33 mmol, 1.00 equiv), 4-chloro-3-hydroxy-5-nitrobenzoate (594.97 mg, 2.57 mmol, 1.10 equiv) and triphenylphosphine (918.86 mg, 3.3 mmol, 1.5 equiv) in tetrahydrofuran (50 mL) was added dropwise DIAD (708.39 mg, 3.3 mmol, 1.5 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred overnight at 0°C under nitrogen atmosphere. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (38% ACN up to 48% in 10 min); Detector, UV 254/220 nm. This resulted in 0.9 g (50.1%) of methyl 3-[3-([3- [(2E)-4-[(tert-butoxycarbonyl)amino]but-2-en-l-yl]-6-carbamoyl-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-l,3-benzodiazol-4-yl]oxy)propoxy]-4-chloro-5-nitrobenzoate as a yellow solid. LCMS (ES, m/z): 770 (M+H+).
Synthesis of 130.4
[00929] To a stirred solution of methyl 3-[3-([3-[(2E)-4-[(tert-butoxycarbonyl)amino]but-2-en- l-yl]-6-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazol-4- yl]oxy)propoxy]-4-chloro-5-nitrobenzoate, 130.3 (0.90 g, 1.169 mmol, 1.00 equiv) in dichloromethane (10 mL) was added trifluoroacetic acid (3 mL). The resulting mixture was stirred overnight at room temperature. The mixture was concentrated under vacuum. This resulted in 0.83 g (crude) of methyl 3-[3-([3-[(2E)-4-aminobut-2-en-l-yl]-6-carbamoyl-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-l,3-benzodiazol-4-yl]oxy)propoxy]-4-chloro-5-nitrobenzoate as red oil. LC MS (ES, m/z): 670 (M+H+).
Synthesis of 130.5
[00930] To a stirred solution of methyl 3-[3-([3-[(2E)-4-aminobut-2-en-l-yl]-6-carbamoyl-2- (4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazol-4-yl]oxy)propoxy]-4-chloro-5- nitrobenzoate, 130.4 (0.83 g crude, 1.169 mmol, 1.00 equiv) in DMSO (10 mL) was added DIEA (1.5 g, 11.69 mmol, 10 equiv). The final reaction mixture was irradiated with microwave radiation for 2h at 100°C. The mixture was cooled to room temperature and diluted with water. The precipitated solids were collected by filtration. This resulted in 0.6 g (81.08%) of methyl (15E)- 23 -carbamoyl- 19-(4-ethyl-2-methyl-l, 3-oxazole-5-amido)-l l-nitro-2,6-dioxa-13, 18,20- triazatetracyclo[16.6.1.0A[7,12].0A[21,25]]pentacosa-l(25),7,9,l l,15, 19,21,23-octaene-9- carboxylate as a dark yellow solid. LC-MS (ES, m/z): 634 (M+H+).
Synthesis of 130.6
[00931] To a stirred solution of methyl (15E)-23-carbamoyl-19-(4-ethyl-2 -methyl-1, 3-oxazole- 5-amido)-l l-nitro-2,6-dioxa-13,18,20-triazatetracyclo[16.6.1.0A[7,12].0A[21,25]]pentacosa- l(25),7,9,l l, 15,19,21,23-octaene-9-carboxylate, 130.5 (0.60 g, 0.946 mmol, 1.00 equiv) in dimethyl formamide (10 mL) was added tin(II)chloride dihydrate (1.07 g, 4.73 mmol, 5.00 equiv). The resulting mixture was stirred overnight at room temperature. The mixture was diluted with water. The pH value of the solution was adjusted to 9 with sat. sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. This resulted in 0.5 g (87.56%) of methyl (15E)-l l-amino-23-carbamoyl-19-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-2,6-dioxa-13,18,20- triazatetracyclo[16.6.1.0A[7,12].0A[21,25]]pentacosa-l(25),7,9,l l,15, 19,21,23-octaene-9- carboxylate as a red solid. LC-MS (ES, m/z): 604 (M+H+).
Synthesis of 130.7
[00932] To a stirred solution of methyl (15E)-l l-amino-23-carbamoyl-19-(4-ethyl-2-methyl- l,3-oxazole-5-amido)-2,6-dioxa-13,18,20-triazatetracyclo[16.6.1.0A[7,12].0A[21,25]]pentacosa- l(25),7,9,l l, 15,19,21,23-octaene-9-carboxylate, 130.6 (0.50 g, 0.828 mmol, 1.00 equiv) in methanol (10 mL) was added cyanogen bromide (130.41 mg, 1.242 mmol, 1.5 equiv). The resulting mixture was stirred for 2h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (35% ACN up to 45% in 10 min); Detector, UV 254/220 nm. This resulted in 0.36 g (69.23%) of methyl (3E)-7-amino-21- carbamoyl-25-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-14, 18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylate as a red solid. LC-MS (ES, m/z): 629 (M+H+).
Synthesis of 130.8
[00933] To a stirred solution of methyl (3E)-7-amino-21-carbamoyl-25-(4-ethyl-2-methyl-l,3- oxazole-5-amido)- 14, 18-dioxa- 1 ,6, 8,24-
533
5UB5TITUTE SHEET (RULE 26) tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3 ,7,9(27), 10, 12, 19(26),20,22,24-nonaene- 11 -carboxylate, 130.7 (360 mg, 0.573 mmol, 1.00 equiv) in dimethyl formamide (8 mL) was added DIEA (740.12 mg, 5.727 mmol, 10.00 equiv), 2-ethyl-5-methylpyrazole-3-carboxylic acid (264.86 mg, 1.718 mmol, 3.00 equiv) and HATU (435.48 mg, 1.145 mmol, 2.00 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 120°C. The reaction was cooled to RT and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (30% ACN up to 41% in 10 min); Detector, UV 254/220 nm. This resulted in 260 mg (59.32%) of methyl (3E)-21-carbamoyl-25-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-(2-ethyl-5-methylpyrazole-3-amido)-14, 18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylate as a dark orange solid. LC-MS (ES, m/z): 765 (M+H+).
Synthesis of 1-167
[00934] To a stirred solution of methyl (3E)-21-carbamoyl-25-(4-ethyl-2-methyl-l,3-oxazole-
5-amido)-7-(2-ethyl-5-methylpyrazole-3-amido)-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylate, 130.8 (40 mg, 0.052 mmol, 1.00 equiv) in methanol (10 mL) and water (2 mL) and lithium hydroxide (5 mg, 0.208 mmol, 4 equiv). The resulting mixture was stirred for 4h at 60°C. The resulting mixture was concentrated under vacuum. The mixture was diluted with water and acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration. The cmde product was purified by Prep-HPLC with the following conditions: Column, XSelect CSH Prep C18 OBD Column,, 19*250mm, 5um; mobile phase, Water (10%NH4HC03+0.1% H3 H20) and methanol (48% methanol up to 68% in 9 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 6.9 mg (17.69%) of (3E)-21-carbamoyl-25-(4-ethyl-2-methyl-l,3-oxazole-5- amido)-7-(2-ethyl-5-methylpyrazole-3-amido)-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylic acid as a white solid. LC-MS (ES, m/z): 751 (M+H+); ¾ NMR: (400 MHz, DMSO -d6) d 12.70 (br s, 3H), 7.99 (s, 1H), 7.81 (s, 1H), 7.70 (s, 1H), 7.51 (d, 2H), 7.36 (s, 1H), 6.56 (s, 1H), 5.59-5.49 (m, 2H), 5.00-4.86 (m, 4H), 4.54 (q, 2H), 4.42-4.38 (m, 4H), 2.85 (q, 2H), 2.45-2.21 (m, 5H), 2.08 (s, 3H), 1.27 (t, 3H), 1.03 (t, 3H).
Example 131: Synthesis of (E)-12-carbamoyl-l-(4-ethyl-2-methyloxazole-5-carboxamido)-15
-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-8,9,16,19-tetrahydro-7H-6,10-dioxa-2,14,l
5a,19a-tetraazacyclopentadeca[3,2,l-cd:8,9,10-c'd']diindene-4-carboxylic acid, 1-168
Figure imgf000536_0001
Figure imgf000537_0001
Synthesis of 131.1
[00935] To a stirred mixture of tert-butyl N-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5- methylpyrazole-3-amido)-7-(3-hydroxypropoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate, 120.5 (500 mg, 0.900 mmol, 1.00 equiv) and methyl 4-chloro-3-hydroxy-5-nitrobenzoate (260 mg, 1.125 mmol, 1.25 equiv) and triphenylphosphine (354.03 mg, 1.350 mmol, 1.50 equiv) in tetrahydrofuran (20 mL) was added dropwise DIAD (272.94 mg, 1.350 mmol, 1.50 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred overnight at 0 °C under nitrogen atmosphere. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (39% ACN up to 50% in 10 min); Detector, UV 254/220 nm. This resulted in 560 mg (81%) of methyl 3-[3-([3-[(2E)-4-[(tert-butoxycarbonyl)amino]but-2-en-l-yl]-6-carbamoyl-2- (2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-4-yl]oxy)propoxy]-4-chloro-5- nitrobenzoate as a yellow solid. LC-MS (ES, m/z): 769 (M+H+).
Synthesis of 131.2
[00936] To a solution of methyl 3-[3-([3-[(2E)-4-[(tert-butoxycarbonyl)amino]but-2-en-l-yl]- 6-carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-4-yl]oxy)propoxy]-4- chloro-5-nitrobenzoate, 131.1 (560 mg, 0.728 mmol, 1.00 equiv) in dichloromethane (20 mL) was added trifluoroacetic acid (5 mL). The resulting mixture was stirred overnight at room temperature. The mixture was concentrated under vacuum. This resulted in 530 mg (crude) of methyl 3-[3-([3- [(2E)-4-aminobut-2-en-l-yl]-6-carbamoyl-2-(2-ethyl-5-methylpyrazole-3-amido)-l,3- benzodiazol-4-yl]oxy)propoxy]-4-chloro-5-nitrobenzoate as a brown oil. LC-MS (ES, m/z): 669 (M+H+).
Synthesis of 131.3
[00937] To a solution of methyl 3-[3-([3-[(2E)-4-aminobut-2-en-l-yl]-6-carbamoyl-2-(2-ethyl- 5-methylpyrazole-3-amido)-l,3-benzodiazol-4-yl]oxy)propoxy]-4-chloro-5-nitrobenzoate, 131.2 (530 mg crude, 0.728 mmol, 1.00 equiv) in dimethylsulfoxide (10 mL) was added DIEA (470 mg, 3.64 mmol, 5.00 equiv). The final reaction mixture was irradiated with microwave radiation for 2h at 100°C. The mixture was cooled to room temperature and diluted with water. The precipitated solids were collected by filtration. This resulted in 415 mg (84.41%) of methyl (15E)-23- carbamoyl-19-(2-ethyl-5-methylpyrazole-3-amido)-l l-nitro-2,6-dioxa-13, 18,20- triazatetracyclo[16.6.1.0A[7,12].0A[21,25]]pentacosa-l(25),7,9,l l,15, 19,21,23-octaene-9- carboxylate as a yellow solid. LC-MS (ES, m/z): 633 (M+H+).
Synthesis of 131.4
[00938] To a solution of methyl (15E)-23-carbamoyl-19-(2-ethyl-5-methylpyrazole-3-amido)- l l-nitro-2,6-dioxa-13, 18,20-triazatetracyclo[16.6.1.0A[7, 12].0A[21,25]]pentacosa- l(25),7,9,l l, 15,19,21,23-octaene-9-carboxylate, 131.3 (415 mg, 0.656 mmol, 1.00 equiv) in dimethyl formamide (10 mL) was added tin(II)chloride dihydrate (740 mg, 3.28 mmol, 5.00 equiv). The resulting mixture was stirred overnight at room temperature. The mixture was diluted with water. The pH value of the solution was adjusted to 9 with sat. sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. This resulted in 356 mg (85.91%) of methyl (15E)-l l-amino-23-carbamoyl-19-(2-ethyl-5-methylpyrazole-3-amido)-2,6-dioxa- 13,18,20-triazatetracyclo[16.6.1.0A[7,12].0A[21,25]]pentacosa-l(25),7,9,l l,15, 19,21,23-octaene- 9-carboxylate as a light yellow solid. LC-MS (ES, m/z): 603 (M+H+).
Synthesis of 131.5 [00939] To a solution of methyl (15E)-l l-amino-23-carbamoyl-19-(2-ethyl-5-methylpyrazole- 3-amido)-2,6-dioxa-13, 18,20-triazatetracyclo[16.6.1.0A[7, 12].0A[21,25]]pentacosa- l(25),7,9,l l, 15,19,21,23-octaene-9-carboxylate, 131.4 (356.00 mg, 0.591 mmol, 1.00 equiv) in methanol (10 mL) was added cyanogen bromide (94 mg, 0.8865 mmol, 1.5 equiv). The resulting mixture was stirred for 2h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (33% ACN up to 44% in 10 min); Detector, UV 254/220 nm. This resulted in 265 mg (71.62%) of methyl (3E)-7-amino-21- carbamoyl-25-(2-ethyl-5-methylpyrazole-3-amido)-14, 18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylate as a brown oil. LC-MS (ES, m/z): 628 (M+FC);
Synthesis of 131.6
[00940] To a solution of methyl (3E)-7-amino-21-carbamoyl-25-(2-ethyl-5-methylpyrazole-3- amido)-14,18-dioxa-l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3 ,7,9(27), 10, 12, 19(26),20,22,24-nonaene- 11 -carboxylate, 131.5 (265.00 mg, 0.423 mmol, 1.00 equiv) and 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (196.7 mg, 1.269 mmol, 3.00 equiv) in dimethyl formamide (8 mL) was added HATU (241 mg, 0.635 mmol, 1.50 equiv) and DIEA (272.8 mg, 2.115 mmol, 5.00 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 120°C. The reaction was cooled to RT and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (29% ACN up to 39% in 10 min); Detector, UV 254/220 nm. This resulted in 240 mg (74.16%) of methyl (3E)-21-carbamoyl-7-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-25-(2-ethyl-5-methylpyrazole-3 -amido)- 14, 18-dioxa- 1 ,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylate(290mg,71.04%) as a brown solid. LC-MS (ES, m/z): 765 (M+H+).
Synthesis of 1-168
[00941] To a solution of methyl (3E)-21-carbamoyl-7-(4-ethyl-2-methyl-l,3-oxazole-5- amido)-25-(2-ethyl-5-methylpyrazole-3-amido)-14,18-dioxa-l, 6, 8, 24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylate, 131.6 (40 mg, 0.052 mmol, 1.00 equiv) in methanol (10 mL) and water (2 mL) and lithium hydroxide (5 mg, 0.208 mmol, 4 equiv). The resulting mixture was stirred for 4h at 60°C. The resulting mixture was concentrated under vacuum. The mixture was diluted with water and acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration. The crude product was purified by Prep-HPLC with the following conditions: Column, XSelect CSH Prep C18 OBD Column, 19*250mm, 5um; mobile phase, Water (10%NH4HC03+0.1% H3 H20) and methanol (50% methanol up to 73% in 9 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 7.7 mg (19.74%) of (3E)-21-carbamoyl-7-(4-ethyl-2-methyl-l,3-oxazole-5- amido)-25-(2-ethyl-5-methylpyrazole-3-amido)-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylic acid as a light brown solid. LC-MS (ES, m/z): 751 (M+H+); H-NMR: (400 MHz, DMSO-c¾): d 12.16 (br s, 3H), 7.99 (s, 1H), 7.81 (s, 1H), 7.70 (s, 1H), 7.51 (d, 2H), 7.36 (s, 1H), 6.55 (s, 1H), 5.62-5.51 (m, 2H), 5.01-4.86 (m, 4H), 4.54 (q, 2H), 4.51-4.33 (m, 4H), 2.85 (q, 2H), 2.49-2.27 (m, 5H), 2.10 (s, 3H), 1.28 (t, 3H), 1.04 (t, 3H).
Example 132: Synthesis of (E)-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-7- (morpholinomethyl)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-(3-hydroxypropyl)-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-169
Figure imgf000540_0001
132.3 132.4 NHBoc
Figure imgf000541_0002
132.5 132.6
Figure imgf000541_0001
Figure imgf000542_0001
Synthesis of 132.1
[00942] Into a 500-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 2-fluoro-l-methyl-3-nitrobenzene (18 g, 116.033 mmol, 1 equiv), CC14 (200 ml), NBS (24.78 g, 139.240 mmol, 1.20 equiv), AIBN (1.91 g, 11.632 mmol, 0.10 equiv). The resulting solution was stirred for 12h at 85°C under nitrogen atmosphere. The solids were filtered out. The resulting mixture was concentrated under vacuum. This resulted in 26.6 g (97.96%) of l-(bromomethyl)-2-fluoro-3 -nitrobenzene as a yellow solid.
Synthesis of 132.2
[00943] Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed l-(bromomethyl)-2-fluoro-3-nitrobenzene, 132.1 (26.6 g, 29.553 mmol, 1 equiv, 26%), ACN (80 mL), morpholine (2.57 g, 29.553 mmol, 1.00 equiv), K2CO3 (8.17 g, 59.105 mmol, 2.00 equiv). The resulting solution was stirred for 12h at 25°C under nitrogen atmosphere. The reaction was then quenched by the addition of 400 mL water. The resulting solution was extracted with 3x400 mL of ethyl acetate. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 : 1). This resulted in 7 g (98.60%) of 4-[(2- fluoro-3-nitrophenyl)methyl]morpholine as a white solid.
Synthesis of 132.3
[00944] Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 4-[(2-fluoro-3-nitrophenyl)methyl]morpholine, 132.2 (7 g, 29.138 mmol, 1 equiv), DMF (50 mL), tert-butyl N-[(2E)-4-aminobut-2-en-l-yl]carbamate (5.43 g, 29.154 mmol, 1.00 equiv), K2CO3 (12.08 g, 87.415 mmol, 3 equiv). The resulting solution was stirred for 12h at 25°C. The reaction was then quenched by the addition of 300 mL water. The resulting solution was extracted with 3x300 mL of ethyl acetate concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 : 1). This resulted in 8 g (67.54%) of tert-butyl N-[(2E)-4-([2-[(morpholin-4-yl)methyl]-6-nitrophenyl]amino)but-2- en-l-yl]carbamate as a white solid.
Synthesis of 132.4
[00945] Into a 500-mL 3-necked round-bottom flask, was placed tert-butyl N-[(2E)-4-([2- [(morpholin-4-yl)methyl]-6-nitrophenyl]amino)but-2-en-l-yl]carbamate, 132.3 (8 g, 19.681 mmol, 1 equiv), THF (75 ml), MeOH (75 ml), H20 (30 ml), Na2S20 (34.27 g, 196.833 mmol, 10.00 equiv). The resulting solution was stirred for 2h at 25°C. The solids were filtered out. The pH value of the solution was adjusted to 8 with NaHCCL. The resulting solution was extracted with 3x200 mL of ethyl acetate and the organic layers combined and concentrated. This resulted in 2 g (26.99%) of tert-butyl N-[(2E)-4-([2-amino-6-[(morpholin-4-yl)methyl]phenyl]amino)but- 2-en-l-yl] carbamate as a white solid.
Synthesis of 132.5
[00946] Into a 100-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed tert-butyl N-[(2E)-4-([2-amino-6-[(morpholin-4- yl)methyl]phenyl]amino)but-2-en-l-yl]carbamate, 132.4 (2.4 g, 5.291 mmol, 1 equiv, 83%), MeOH (20 mL), BrCN (0.56 g, 5.291 mmol, 1 equiv). The resulting solution was stirred for 2h at 25°C. The resulting mixture was concentrated under vacuum yielding 1.6 g (75.32%) of a white solid.
Synthesis of 132.6
[00947] Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (0.74 g, 4.782 mmol, 1.2 equiv), DMF(10 ml) , DIEA (1.55 g, 11.955 mmol, 3 equiv), HATU (2.27 g, 5.977 mmol, 1.5 equiv), tert-butyl N-[(2E)-4-[2-amino-7-[(morpholin-4-yl)methyl]-lH-l,3- benzodiazol-l-yl]but-2-en-l-yl]carbamate, 132.5 (1.6 g, 3.985 mmol, 1 equiv). The resulting solution was stirred for 12h at 25°C. The reaction was then quenched by the addition of 50 mL water. The resulting solution was extracted with 3x50 mL of ethyl acetate and the organic layers combined and concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN/FhO=10% increasing to ACN/FhO=60% within 10 min; Detector 254/220nm to yield 679 mg (31.69%) of a white solid.
Synthesis of 132.7
[00948] Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed tert-butyl N-[(2E)-4-[2-(l-ethyl-3-methyl-lH-pyrazole-5- amido)-7-[(morpholin-4-yl)methyl]-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate, 132.6 (679 mg, 1.263 mmol, 1 equiv), DCM (7 mL), TFA (2.35 mL). The resulting solution was stirred for 2h at 25°C. The resulting mixture was concentrated under vacuum. This resulted in 552 mg (99.90%) of N-[l-[(2E)-4-aminobut-2-en-l-yl]-7-[(morpholin-4-yl)methyl]-lH-l,3-benzodiazol- 2-yl]-l-ethyl-3-methyl-lH-pyrazole-5-carboxamide as a white solid.
Synthesis of 132.8
[00949] Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed N-[l-[(2E)-4-aminobut-2-en-l-yl]-7-[(morpholin-4- yl)methyl]-lH-l,3-benzodiazol-2-yl]-l-ethyl-3-methyl-lH-pyrazole-5-carboxamide, 132.7 (552 mg, 1.262 mmol, 1 equiv), DMF (10 mL), 4-fluoro-3-nitrobenzamide (278.75 mg, 1.514 mmol, 1.20 equiv), K2CO3 (871.78 mg, 6.308 mmol, 5.00 equiv). The resulting solution was stirred for 12h at 70°C. The reaction was then quenched by the addition of 50 mL water. The resulting solution was extracted with 3x50 mL of ethyl acetate and the organic layers combined and concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN/H2O=10% increasing to ACN/H2O=60% within 10 min; Detector 254/220nm. This resulted in 500 mg (65.87%) of N-[l-[(2E)-4-[(4-carbamoyl-2-nitrophenyl)amino]but-2-en-l-yl]-7-[(morpholin-4- yl)methyl]-lH-l,3-benzodiazol-2-yl]-l-ethyl-3-methyl-lH-pyrazole-5-carboxamide as a white solid.
Synthesis of 132.9
[00950] Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed N-[l-[(2E)-4-[(4-carbamoyl-2-nitrophenyl)amino]but-2-en-l- yl]-7-[(morpholin-4-yl)methyl]-lH-l,3-benzodiazol-2-yl]-l-ethyl-3-methyl-lH-pyrazole-5-
543
5UB5TITUTE SHEET (RULE 26) carboxamide, 132.8 (500 mg, 0.831 mmol, 1 equiv), HO Ac (5 mL), Zn (1.09 g, 16.664 mmol, 20.05 equiv). The resulting solution was stirred for 2h at 25°C. The reaction was then quenched by the addition of 50 mL water. The resulting solution was extracted with 3x50 mL of ethyl acetate and the organic layers combined. The solids were fdtered out. The resulting mixture was concentrated under vacuum. This resulted in 475 mg (99.98%) of N-[l-[(2E)-4-[(2-amino-4- carbamoylphenyl)amino]but-2-en-l-yl]-7-[(morpholin-4-yl)methyl]-lH-l,3-benzodiazol-2-yl]-l- ethyl-3-methyl-lH-pyrazole-5-carboxamide as a white solid.
Synthesis of 132.10
[00951] Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed N-[l-[(2E)-4-[(2-amino-4-carbamoylphenyl)amino]but-2-en- l-yl]-7-[(morpholin-4-yl)methyl]-lH-l,3-benzodiazol-2-yl]-l-ethyl-3-methyl-lH-pyrazole-5- carboxamide, 132.9 (475 mg, 0.831 mmol, 1 equiv), MeOH (3 mL), BrCN (88.01 mg, 0.831 mmol, 1 equiv). The resulting solution was stirred for 2h at 25°C. The resulting mixture was concentrated under vacuum. The crude product was purified by FI ash-Prep -HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN/H2O=10% increasing to ACN/H2O=60% within 10 min; Detector 254/220nm. This resulted in 280 mg (40.10%) of 2- amino-l-[(2E)-4-[2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-7-[(morpholin-4-yl)methyl]-lH- l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3-benzodiazole-5-carboxamide as a white solid. Synthesis of 132.11
[00952] Into a 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid (86.81 mg, 0.563 mmol, 1.2 equiv), DMF (2 mL), DIEA (181.94 mg, 1.408 mmol, 3 equiv), HATU (267.63 mg, 0.704 mmol, 1.5 equiv), 2-amino-l-[(2E)-4-[2-(l-ethyl-3-methyl-lH-pyrazole-5-amido)-7-[(morpholin-4- yl)methyl]-lH-l,3-benzodiazol-l-yl]but-2-en-l-yl]-lH-l,3-benzodiazole-5-carboxamide, 132.10 (280 mg, 0.469 mmol, 1 equiv). The resulting solution was stirred for 12h at 25 °C. The reaction was then quenched by the addition of 10 mL water. The resulting solution was extracted with 3x10 mL of ethyl acetate and the organic layers combined and concentrated under vacuum. The crude product was purified by FI ash-Prep -HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN/H2O=10% increasing to ACN/H2O=60% within 10 min; Detector 254/220nm. This resulted in 219 mg (63.70%) of (E)-2-(l-ethyl-3-methyl-lH-pyrazole-
544
5UB5TITUTE SHEET (RULE 26) 5-carboxamido)-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-7-(morpholinomethyl)- lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-lH-benzo[d]imidazole-5-carboxamide as a white solid. LC-MS: (ES, m/z): [M+H]+ 733.4; ¾-NMR: (400 MHz, DMSO ,ppm): d 12.79 (s, 2H), 7.90-8.00 (m, 2H), 7.68-7.74 (d, 1H), 7.42-7.55 (m, 2H), 7.32(s, 1H), 7.10-7.18 (m, 1H), 6.92-7.00 (d, 1H), 6.52-6.61 (d, 2H), 5.92-6.05 (d, 1H), 5.32-5.45 (d, 1H), 5.19 (s, 2H), 4.80 (s, 2H), 4.48-4.58 (d, 4H), 3.43 (s, 6H), 2.10-2.20 (m, 10H), 1.20-1.30 (m, 6H).
Synthesis of 1-169
[00953] Into a 20-mL round-bottom flask, was placed 2-[3-[(tert- butyldimethylsilyl)oxy]propyl]-5-methylpyrazole-3-carboxylic acid (180.07 mg, 0.603 mmol, 4.00 equiv), DMF (10.00 mL), DIEA (155.95 mg, 1.207 mmol, 8.00 equiv), HATU (229.40 mg, 0.603 mmol, 4.00 equiv) and 2-amino-l-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-7- (morpholin-4-ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazole-5-carboxamide, 132.11 (90.00 mg, 0.151 mmol, 1.00 equiv). The solution was stirred overnight at 85 °C under nitrogen atmosphere. To the above mixture was added H2O (3.00 mL), NaOH (60.33 mg, 1.508 mmol, 10.00 equiv). The resulting solution was stirred for another 1 overnight at room temperature. The resulting solution was extracted with 3x10 mL of ethyl acetate and the organic layers combined and concentrated. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN/H20=15% increasing to ACN/H2O=60% within 15 min. The crude product was purified by Prep-HPLC with the following condition (Column: Sunfire prep C18 column, 30*150 nm, 5 um; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 15% B to 34% B in 8 min; 254;220 nm; RT: 8.02 min). This resulted in 1.7 mg (1.54%) of2-(2-ethyl-5-methylpyrazole- 3-amido)-l-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-7-(morpholin-4-ylmethyl)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazole-5-carboxamide as a brown solid. LC-MS: (ES, m/z) [M+H]+ 763.5 , [M-H]' 761.3; 1H-NMR: (400 MHz, DMSO -d^ppm) d 8.38 (s, 1H), 7.98 (s, 1H), 7.93 (s, 1H), 7.69-7.72 (d, 1H), 7.49-7.51 (d, 1H), 7.42-7.44 (d, 1H), 7.31 (s, 1H), 7.12-7.13 (d, 1H), 6.97-6.99 (d, 1H), 6.57 (s, 1H), 6.52 (s, 1H), 5.95-5.99 (d, 1H), 5.38-5.42 (d, 1H), 5.19 (s, 2H), 4.80-4.81 (d, 2H), 4.46-4.61 (m, 5H), 3.43 (s, 8H), 2.17 (s, 4H), 2.12-2.14 (d, 6H), 1.82-1.91 (m, 2H), 1.20-1.28 (t, 3H). Example 133: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(2,4-dimethyloxazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-(morpholinomethyl)-lH- benzo [d] imidazol-2-yl)-2, 4-dimethyl oxazole-5-carboxamide, 1-170
Figure imgf000547_0001
127.9
1-170
[00954] Into a 25-mL 3-necked round-bottom flask, was placed dimethyl- 1, 3 -oxazole-5- carboxylic acid (112.10 mg, 0.794 mmol, 4.00 equiv), DMF (10.00 mL), HATU (302.03 mg, 0.794 mmol, 4.00 equiv), DIEA (205.32 mg, 1.589 mmol, 8.00 equiv), 2-amino- 1- [(2E)-4-(2-amino-5- carbamoyl- 1,3 -benzodiazol- 1 -yl)but-2-en- 1 -yl] -7-(morpholin-4-ylmethyl)- 1 ,3-benzodiazole-5- carboxamide (110.00 mg, 0.199 mmol, 1.00 equiv). The resulting solution was stirred for 12h at 25 degrees C under nitrogen atmosphere. The residue was purified by Prep-HPLC with the following condition (Column: XBridge Prep OBD Cl 8 Column, 19*250 mm, 5 um; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 18% B to 29% B in 10 min; 254;220 nm; RT:9.22 min). This resulted in 7.3 mg (3.36%) of l-[(2E)-4-[5- carbamoyl-2-(2,4-dimethyl-l,3-oxazole-5-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2,4- dimethyl-l,3-oxazole-5-amido)-7-(morpholin-4-ylmethyl)-l,3-benzodiazole-5-carboxamide as a white solid. LC-MS: (ES, m/z): [M+H]+ 750.4, [M-H]' 748.2
1H-NMR: (400 MHz, OMSO-ek, ppm) d 12.73 (s, 2H), 7.92-7.97 (m, 4H), 7.70-7.72 (d, 1H), 7.55 (s, 1H), 7.41-7.43 (d, 1H), 7.33-7.37 (d, 2H), 5.80-5.91 (m, 1H), 5.40-5.44 (d, 1H), 5.18-5.24 (m, 2H), 4.77-4.78 (d, 2H), 3.45-3.46 (d, 6H), 2.40-2.41 (d, 6H), 2.28-2.32 (d, 6H), 2.20-2.25 (d, 4H). Example 134: Synthesis of (E)-l-(4-ethyl-2-methyloxazole-5-carboxamido)-15-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-N-(2-(2-(2-hydroxyethoxy)ethoxy)ethyl)-8,9,16,19- tetrahydro-7H-6,10-dioxa-2,14,15a,19a-tetraazacyclopentadeca[3,2,l-cd:8,9,10- c'd']diindene-4, 12-dicarboxamide, 1-171
Figure imgf000548_0001
[00955] To a stirred solution of (3E)-21-carbamoyl-25-(4-ethyl-2-methyl-l,3-oxazole-5- amido)-7-(2-ethyl-5-methylpyrazole-3-amido)-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9, 11, 13 (27), 19, 21,23 (26), 24-nonaene- 11 -carboxylic acid, 1-167, (50.00 mg, 0.053 mmol, 1.00 equiv) in dimethyl formamide (5 mL) was added DIEA (68.86 mg, 0.533 mmol, 10.00 equiv), HATU (40.52 mg, 0.107 mmol, 2.00 equiv) and 2- [2-(2-aminoethoxy)ethoxy] ethanol (39.74 mg, 0.266 mmol, 5.00 equiv). The resulting solution was stirred for 3h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (38% ACN up to 51%) in 10 min); Detector, UV 254/220 nm. The crude product (20 mg) was purified by Prep-HPLC with the following conditions: Column, XBridge Prep OBD Cl 8 Column, 19*250mm, 5um; mobile phase, Water (10 o H4HC03+0.1%NH3 H20) and methanol (55%o methanol up to 80%o in 9 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 6.9 mg (15.11%) of (3E)-25-(4-ethyl-2-methyl-l,3-
547
5UB5TITUTE SHEET (RULE 26) oxazole-5-amido)-7-(2-ethyl-5-methylpyrazole-3-amido)-Nl l-[2-[2-(2- hydroxyethoxy)ethoxy]ethyl]-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3, 7, 9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l l, 21 -dicarboxamide as a white solid. LC-MS (ES, m/z): 882 (M+H+); ¾ NMR: (400 MHz, DMSO -d6) d 12.65 (br s, 2H), 8.54 (t, 1H), 7.98 (s, 1H), 7.69 (d, 2H), 7.51 (d, 2H), 7.37 (s, 1H), 6.55 (s, 1H), 5.57-5.48 (m, 2H), 4.95-4.86 (m, 4H), 4.58- 4.52 (m, 3H), 4.50-4.42 (m, 4H), 3.57-3.53 (m, 6H), 3.48-3.32 (m, 6H), 2.85 (q, 2H), 2.38-2.28 (m, 5H), 2.08 (s, 3H), 1.27 (t, 3H), 1.03 (t, 3H).
Example 135: Synthesis of (E)-N-(l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-car boxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-(morpholinomethyl)-lH-benzo[d]imi dazol-2-yl)-2,4-dimethyloxazole-5-carboxamide, 1-172
Figure imgf000549_0001
Figure imgf000550_0001
Synthesis of 135.1
[00956] To a stirred solution of dimethyl-1, 3-oxazole-5-carboxylic acid, 112.7 (284.97 mg, 2.019 mmol, 2.00 equiv) and HATU (767.79 mg, 2.019 mmol, 2.00 equiv) and DIEA(521.95 mg, 4.039 mmol, 4.00 equiv) in DMF (5.00 mL) was added 4-[[(2E)-4-[2-amino-7-(morpholin-4- ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]amino]-3-nitrobenzamide(470.00 mg, 1.010 mmol, 1.00 equiv) in portions at 85 degrees C under nitrogen atmosphere. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, NH4HC03 in water, 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in N-[l- [(2E)-4-[(4-carbamoyl-2-nitrophenyl)amino]but-2-en-l-yl]-7-(morpholin-4-ylmethyl)-l,3- benzodiazol-2-yl]-2,4-dimethyl-l,3-oxazole-5-carboxamide (385 mg, 64.78%) as a yellow solid. Synthesis of 135.2
[00957] To a stirred solution of N-[l-[(2E)-4-[(4-carbamoyl-2-nitrophenyl)amino]but-2-en-l- yl]-7-(morpholin-4-ylmethyl)-l,3-benzodiazol-2-yl]-2,4-dimethyl-l,3-oxazole-5-carboxamide. 135.2 (380.00 mg, 0.646 mmol, 1.00 equiv) in HO Ac (5.00 mL) was added Zn (844.52 mg, 12.911 mmol, 20.00 equiv) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3h and filtered, the filter cake was washed with MeOH (3x10 mL). The filtrate was concentrated under reduced pressure. This resulted in N-[l-[(2E)-4-[(2-amino-4- carbamoylphenyl)amino]but-2-en-l-yl]-7-(morpholin-4-ylmethyl)-l,3-benzodiazol-2-yl]-2,4- dimethyl-l,3-oxazole-5-carboxamide (200 mg, 55.46%) as a red solid. Synthesis of 135.3
[00958] To a stirred solution of N-[l-[(2E)-4-[(2-amino-4-carbamoylphenyl)amino]but-2-en-l- yl]-7-(morpholin-4-ylmethyl)-l,3-benzodiazol-2-yl]-2,4-dimethyl-l,3-oxazole-5-carboxamide, 135.2 (800.00 mg, 1.432 mmol, 1.00 equiv) in MeOH (10.00 mL) was added BrCN (227.52 mg, 2.148 mmol, 1.50 equiv) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2h at room temperature and concentrated under reduced pressure. The precipitated solids were collected by filtration and washed with MeOH (3x5 mL). This resulted in 2-amino-l-[(2E)-4-[2-(2,4-dimethyl-l,3-oxazole-5-amido)-7-(morpholin-4-ylmethyl)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazole-5-carboxamide (220 mg, 26.32%) as a off- white solid.
Synthesis of 1-172
[00959] Into a 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed 2-amino-l-[(2E)-4-[2-(2,4-dimethyl-l,3-oxazole-5-amido)-7-(morpholin-4- ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazole-5-carboxamide, 135.3 (160.00 mg, 0.274 mmol, 1.00 equiv) in DMF (2.00 mL), DIEA (708.60 mg, 5.483 mmol, 20.00 equiv) and HATU (625.41 mg, 1.645 mmol, 6.00 equiv) were added and followed by the addition of 2- ethyl-5-methylpyrazole-3-carboxylic acid (253.58 mg, 1.645 mmol, 6.00 equiv). The resulting mixture was stirred for 3 h at 85 degrees C. The resulting mixture was washed with 1x20 mL of water. The crude product (100 mg) was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, HCOOH in water, 10% to 50% gradient in 35 min; detector, UV 254 nm. The crude product (60 mg) was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): Column, XBridge Prep OBD C18 Column, 19*250mm, 5um; mobile phase, water (10 mmol/L NH4HCO3) and ACN (hold 30% PhaseB in 14 min); Detector, UV 254nm.) to afford l-[(2E)-4-[2-(2,4-dimethyl-l,3-oxazole-5- amido)-7-(morpholin-4-ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2-ethyl-5- methylpyrazole-3-amido)-l,3-benzodiazole-5-carboxamide(13.1mg,6.64%) as a white solid. LC- MS (ES, m/z): 720 (M+H+); H-NMR: (400 MHz, DMSO-d6): d 12.71 (s, 2H), 7.97 (d, 2H), 7.72 (d, 1H), 7.51-7.42 (m, 2H), 7.32 (s, 1H), 7.13-7.08 (m, 1H), 6.97 (d, 1H), 6.55 (s, 1H), 6.00 (d, 1H), 5.42-5.35 (m, 1H), 5.18 (s, 2H), 4.78 (s, 2H), 4.57-4.50 (m, 2H), 3.43 (s, 6H), 2.51 (s, 3H), 2.34 (s, 3H), 2.14 (d, 7H), 1.30 (t, 3H). Example 136: Synthesis of (E)-3-(5-((5-carbamoyl-l-(4-(5-carbamoyl-2-(4-ethyl-2- methyloxazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-lH- benzo[d]imidazol-2-yl)carbamoyl)-3-methyl-lH-pyrazol-l-yl)propanoic acid, 1-173
Figure imgf000552_0001
Synthesis of 136.1
[00960] To a stirred mixture of 2-amino-l-[(2E)-4-(2-amino-5-carbamoyl-l,3-benzodiazol-l- yl)but-2-en-l-yl]-l,3-benzodiazole-5-carboxamide, 67.5 (170 mg, 0.420 mmol, 1 equiv) and 4-
551
5UB5TITUTE SHEET (RULE 26) ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (65.00 mg, 0.420 mmol, 1 equiv) in dimethyl formamide (4 mL) was added DIEA (271 mg, 2.1 mmol, 5.00 equiv) and HATU (175 mg, 0.460 mmol, 1.10 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 120°C. The mixture was cooled to RT and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (10%NH4HCO3) and ACN (30% ACN up to 50% in 10 min); Detector, UV 254/220 nm. This resulted in 150 mg (66%) of 2-amino-l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5- amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazole-5-carboxamide as a light yellow solid. LC-MS (ES, m/z): 542 (M+H+).
Synthesis of 136.2
[00961] To a stirred solution of 2-amino-l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazole-5-carboxamide, 136.1 (130 mg, 0.240 mmol, 1.00 equiv) and 2-(3-methoxy-3-oxopropyl)-5-methylpyrazole-3- carboxylic acid (65 mg, 0.290 mmol, 1.20 equiv) in dimethyl formamide (4 mL) was added DIEA (155 mg, 1.200 mmol, 5 equiv) and HATU (118 mg, 0.312 mmol, 1.30 equiv). The final reaction mixture was irradiated with microwave radiation for lh at 120°C. The mixture was cooled to RT and concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (10%NH4HCO3) and ACN (26% ACN up to 46% in 10 min); Detector, UV 254/220 nm. This resulted in 100 mg (56.82%) of methyl 3- [5-([5-carbamoyl-l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazol-2-yl]carbamoyl)-3-methylpyrazol-l- yl]propanoate as a light yellow solid. LC-MS (ES, m/z): 736 (M+H+).
Synthesis of 1-173
[00962] To a stirred solution of 3-[5-([5-carbamoyl-l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2- methyl-l,3-oxazole-5-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazol-2- yl]carbamoyl)-3-methylpyrazol-l-yl]propanoate, 136.2 (100 mg, 0.136 mmol, 1 equiv) in methanol (10 mL) and water (2 mL) was added lithium hydroxide (13 mg, 0.544 mmol, 4 equiv). The resulting solution was stirred for 2h at room temperature. The mixture was concentrated under reduced pressure. The residue was acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD Column, 30* 150mm, 5um; mobile phase, Water (0.1%FA) and ACN (70% ACN up to 80% in 7 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 16.4 mg (16.7%) of 3-[5-([5-carbamoyl-l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazol-2-yl]carbamoyl)-3- methylpyrazol-l-yl]propanoic acid as a white solid. LC-MS (ES, m/z): 361 (M/2+TE); 1H NMR: 1H NMR (400 MHz, DMSO-<¾) 6 10.89 (br s, 1H), 8.03-7.92 (m, 3H), 7.71 (dd, 2H), 7.69-6.90 (m, 5H), 6.56 (s, 1H), 6.04-5.82 (m, 2H), 4.90-4.58 (m, 6H), 2.85 (q, 2H), 2.70 (t, 2H), 2.41 (s, 3H), 2.15 (s, 3H), 1.09 (t, 3H).
Example 137: Synthesis of (E)-l-(4-ethyl-2-methyloxazole-5-carboxamido)-15-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-N-(2-(2-hydroxyethoxy)ethyl)-8,9,16,19-tetrahydro- 7H-6,10-dioxa-2, 14, 15a, 19a-tetraazacyclopentadeca[3, 2, l-cd:8, 9, 10-c'd']diindene-4, 12- dicarboxamide, 1-174
Figure imgf000554_0001
1-167 1-174
[00963] To a stirred solution of (3E)-21-carbamoyl-25-(4-ethyl-2-methyl-l,3-oxazole-5- amido)-7-(2-ethyl-5-methylpyrazole-3-amido)-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9, 11, 13(27), 19, 21, 23(26), 24-nonaene- 11 -carboxylic acid, 1-167 (50.00 mg, 0.053 mmol, 1.00 equiv) in dimethyl formamide (5 mL) was added DIEA (68.86 mg, 0.533 mmol, 10.00 equiv), HATU (40.52 mg, 0.107 mmol, 2.00 equiv) and 2-(2-aminoethoxy)ethanol (28.01 mg, 0.266 mmol, 5.00 equiv). The resulting solution was stirred for 3h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (37% ACN up to 50% in 10 min); Detector, UY 254/220 nm. The crude product (22 mg) was purified by Prep- HPLC with the following conditions: Column, XBridge Prep OBD C18 Column, 19*250mm, 5um; mobile phase, Water (10%NHdTC03+0.1%NH3 H20) and methanol (50% methanol up to 72% in 10 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 6.9 mg (14.64%) of (3E)-25-(4-ethyl-2-methyl-l,3-oxazole-5- amido)-7-(2-ethyl-5-methylpyrazole-3-amido)-Nl l-[2-(2-hydroxyethoxy)ethyl]-14, 18-dioxa- l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3 ,7,9, 11 , 13 (27), 1 ,21 ,23 (26),24-nonaene- 11 ,21 -dicarboxamide as an off-white solid. LC-MS (ES, m/z): 838 (M+H+); Tl NMR: (400 MHz, DMSO-i¾) d 12.65 (br s, 2H), 8.50 (t, 1H), 7.98 (s, 1H), 7.69 (d, 2H), 7.49 (d, 2H), 7.36 (s, 1H), 6.54 (s, 1H), 5.57-5.48 (m, 2H), 4.88-4.85 (m, 4H), 4.61 (t, 1H), 4.53 (q, 2H), 4.51-4.40 (m, 4H), 3.57-3.43 (m, 8H), 2.85 (q, 2H), 2.39 (s, 3H), 2.31-2.28 (m, 2H), 2.08 (s, 3H), 1.27 (t, 3H), 1.03 (t, 3H).
Example 138: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(4-ethyl-2- methyloxazole-5-carboxamido)-7-methoxy-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-(2- (dimethylamino)ethoxy)-lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5- carboxamide, 1-175
Figure imgf000555_0001
Figure imgf000556_0001
Synthesis of 138.1
[00964] To a stirred solution of 4-chloro-3-hydroxy-5-nitrobenzamide (2.00 g, 9.234 mmol, 1.00 equiv) and
Figure imgf000556_0002
g, 9.234 mmol, 1.00 equiv) in DMF (20.00 mL) was 2-bromoethanol (11.45 g, 92.34 mmol, 10.00 equiv) in portions. The resulting mixture was stirred at 100 degrees C under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, NH4HCO3 in water, 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 4-chloro-3-(2-hydroxyethoxy)-5-nitrobenzamide (1.5 g, 62.32%) as a yellow solid. Synthesis of 138.2
[00965] To a stirred solution of 4-chloro-3-(2-hydroxyethoxy)-5-nitrobenzamide, 138.1 (1.40 g, 5.372 mmol, 1.00 equiv) in DCM(10.00 mL) were added Et3N(2.17 g, 21.445 mmol, 3.99 equiv) and methanesulfonyl chloride(3.69 g, 32.230 mmol, 6.00 equiv) in portions at 0 degrees C. The resulting mixture was stirred for 1 h at room temperature. The resulting mixture was concentrated under vacuum. The resulting mixture was extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with water (3x5 mL), dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 2-(5-carbamoyl-2-chloro-3- nitrophenoxy)ethyl methanesulfonate (1.6g, 70.35%) as a off-white solid.
Synthesis of 138.3
[00966] To a stirred solution of dimethylamine (0.26 g, 5.767 mmol, 1.22 equiv) and NaH (0.17 g, 7.085 mmol, 1.50 equiv) in DMF (1.00 mL) was added 2-(5-carbamoyl-2-chloro-3- nitrophenoxy)ethyl methanesulfonate, 138.2 (1.60 g, 4.724 mmol, 1.00 equiv) in portions at room temperature under nitrogen atmosphere. The reaction was stirred for 3h at room temperature. The resulting mixture was quenched by ice water and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with water (3x10 mL), dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 4-chloro-3-[2- (dimethylamino)ethoxy]-5-nitrobenzamide (1.1 g, 80.94%) as a red solid.
Synthesis of 138.4
[00967] To a stirred solution of 4-chloro-3-[2-(dimethylamino)ethoxy]-5-nitrobenzamide, 138.3 (1.10 g, 3.823 mmol, 1.00 equiv) and DIEA (1.48 g, 11.451 mmol, 3.00 equiv) in DMSO (10.00 mL) were added 4-[[(2E)-4-aminobut-2-en-l-yl]amino]-3-methoxy-5-nitrobenzamide (1.61 g, 5.735 mmol, 1.50 equiv) in portions. The reaction mixture was stirred overnight at 120 degrees C under nitrogen atmosphere. The mixture was cooled to room temperature, diluted with water and exacted with EtOAc (3 x 30 mL). The organic layer was concentrated. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, MeOH in water, 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 4-[[(2E)-4-([4-carbamoyl-2-[2-(dimethylamino)ethoxy]-6-nitrophenyl]amino)but-2-en-l- yl]amino]-3-methoxy-5-nitrobenzamide (540 mg, 26.57%) as a off-white solid. Synthesis of 138.5
[00968] To a stirred solution of 4-[[(2E)-4-([4-carbamoyl-2-[2-(dimethylamino)ethoxy]-6- nitrophenyl]amino)but-2-en-l-yl]amino]-3-methoxy-5-nitrobenzamide, 138.4 (540.00 mg, 1.016 mmol, 1.00 equiv) and ammonia (1.00 mL) in MeOH (1.00 mL, 24.699 mmol, 24.31 equiv) was added Na2S2C>4 (530.65 mg, 3.048 mmol, 3.00 equiv) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2h at room temperature. Then the solid was filtered, the filter cake was washed with MeOH (3x5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (CH2C12 /MeOH 5: 1) to afford 3-amino- 4-[[(2E)-4-([2-amino-4-carbamoyl-6-[2-(dimethylamino)ethoxy]phenyl]amino)but-2-en-l- yl]amino]-5-methoxybenzamide (220 mg, 45.92%) as a off-white solid.
Synthesis of 138.6
[00969] To a stirred solution of 3-amino-4-[[(2E)-4-([2-amino-4-carbamoyl-6-[2- (dimethylamino)ethoxy]phenyl]amino)but-2-en-l-yl]amino]-5-methoxybenzamide, 138.5
(220.00 mg, 0.467 mmol, 1.00 equiv) in MeOH (5.00 mL) was added BrCN (148.25 mg, 1.400 mmol, 3.00 equiv) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2h at room temperature and then concentrated under reduced pressure. This resulted in 2-amino-l-[(2E)-4-[2-amino-5-carbamoyl-7-[2-(dimethylamino)ethoxy]-l,3- benzodiazol-l-yl]but-2-en-l-yl]-7-methoxy-l,3-benzodiazole-5-carboxamide (150 mg, 61.64%) as a off-white solid.
Synthesis of 1-175
[00970] Into a 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed 2-amino-l-[(2E)-4-[2-(2,4-dimethyl-l,3-oxazole-5-amido)-7-(morpholin-4- ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazole-5-carboxamide, 138.6 (160.00 mg, 0.274 mmol, 1.00 equiv), DIEA (743.37 mg, 5.752 mmol, 20.00 equiv), HATU (656.09 mg, 1.726 mmol, 6.00 equiv), 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (267.72 mg, 1.726 mmol, 6.00 equiv) in DMF (2.00 mL). The resulting mixture was stirred for 3 h at 85 degrees C. The resulting mixture was washed with 1x20 mL of water. The crude product (60 mg) was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, HCOOH in water, 10% to 50% gradient in 35 min; detector, UV 254 nm. The crude product (60 mg) was purified by Prep-HPLC with the following conditions (2#SH1MADZU (HPLC-01)): Column, XBridge Prep OBD Cl 8 Column, 19*250mm, 5um; mobile phase, water (10MMOL/L NH4HCOV) and ACN (hold 30% PhaseB in 14 min); Detector, UY 254nm.) to afford l-[(2E)-4- [5-carbamoyl-7-[2-(dimethylamino)ethoxy]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-methoxy-l,3- benzodiazole-5-carboxamide (2.5 mg, 2.18%) as a white solid. LC-MS (ES, m/z):796 (M+H+); H-NMR: (400 MHz, DMSO -d6): d 12.60 (s, 1H), 7.63 (s, 2H),7.62(s, 2H), 7.27 (s, 4H), 5.82 (d, 2H), 4.88 (d, 4H), 4.02-4.00 (m, 2H), 3.71 (s, 3H), 2.80-2.71 (m, 4H), 2.50 (m, 8H), 2.04 (s, 6H), 1.02 (m, 6H).
Example 139: Synthesis of (E)-l-(4-ethyl-2-methyloxazole-5-carboxamido)-15-(l-ethyl-3-methyl- lH-pyrazole-5-carboxamido)-N-(2-hydroxyethyl)-8,9,16,19-tetrahydro-7H-6,10-dioxa-2,14,15a,19a- tetraazacyclopentadeca[3, 2, l-cd:8, 9, 10-c'd']diindene-4, 12-dicarboxamide, 1-176
Figure imgf000559_0001
[00971] To a stirred solution of (3E)-21-carbamoyl-25-(4-ethyl-2-methyl-l,3-oxazole-5- amido)-7-(2-ethyl-5-methylpyrazole-3-amido)-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9, 11, 13(27), 19, 21, 23(26), 24-nonaene- 11 -carboxylic acid, 1-167 (100.00 mg, 0.107 mmol, 1.00 equiv) in dimethyl formamide (5 mL) was added DIEA (137.72 mg, 1.066 mmol, 10.00 equiv), HATU (81.03 mg, 0.213 mmol, 2.00 equiv) and ethanolamine (19.53 mg, 0.320 mmol, 3.00 equiv). The resulting solution was stirred for 2h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (36% ACN up to 47% in 10 min); Detector, UV 254/220 nm. The crude product (30 mg) was purified by Prep- HPLC with the following conditions: Column: XBridge Shield RP18 OBD C18 Column, 19*250mm, lOum; mobile phase, Water (10% NH4HCO3 + O. /oNThTBO) and methanol (53% methanol up to 77% in 9 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 13.7 mg (14.3%) of (3E)-25-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-(2-ethyl-5-methylpyrazole-3-amido)-Nl l-(2-hydroxyethyl)-14,18-dioxa- l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3 ,7,9, 11 , 13 (27), 19,21 ,23 (26),24-nonaene- 11 ,21 -dicarboxamide as an off-white solid. LC-MS (ES, m/z): 794 (M+H+); 1H MR: (400 MHz, DMSO-i¾) d 12.79 (br s, 2H), 8.47 (t, 1H), 7.99 (s, 1H), 7.69 (d, 2H), 7.51 (d, 2H), 7.36 (s, 1H), 6.55 (s, 1H), 5.61-5.50 (m, 2H), 4.89-4.86 (m, 4H), 4.56- 4.41 (m, 7H), 3.54 (t, 2H), 3.38-3.33 (m, 2H), 2.85 (q, 2H), 2.39 (s, 3H), 2.31-2.28 (m, 2H), 2.08 (s, 3H), 1.27 (t, 3H), 1.03 (t, 3H).
Example 140: Synthesis of (E)-l-(4-ethyl-2-methyloxazole-5-carboxamido)-15-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-12-(4-(2-hydroxyethyl)piperazine-l-carbonyl)- 8,9,16,19-tetrahydro-7H-6,10-dioxa-2,14,15a,19a-tetraazacyclopentadeca[3,2,l-cd:8,9,10- c'd']diindene-4-carboxamide, 1-177
Figure imgf000560_0001
[00972] To a stirred solution of (3E)-21-carbamoyl-25-(4-ethyl-2-methyl-l,3-oxazole-5- amido)-7-(2-ethyl-5-methylpyrazole-3-amido)-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9, 11, 13(27), 19, 21, 23(26), 24-nonaene- 11 -carboxylic acid, 1-167 (50.00 mg, 0.053 mmol, 1.00 equiv) in dimethyl formamide (5 mL) was added DIEA (68.86 mg, 0.533 mmol, 10.00 equiv), HATU (40.52 mg, 0.107 mmol, 2.00 equiv) and 1-piperazineethanol (20.81 mg, 0.160 mmol, 3.00 equiv). The resulting solution was stirred for 2h at room temperature. The reaction was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (36% ACN up to 49% in 10 min); Detector, UV 254/220 nm. The crude product (26 mg) was purified by Prep- HPLC with the following conditions: Column: XBridge Prep OBD C18 Column, 19*250mm, 5um; mobile phase, Water (10%NH4HC03+0.1%NH3 H20) and ACN (16% ACN up to 43% in 9 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 9.8 mg (20.78%) of (3E)-7-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-25-(2- ethyl-5-methylpyrazole-3 -amido)-21 -[4-(2-hydroxyethyl)piperazine- 1 -carbonyl] -14, 18-dioxa- l,6,8,24-tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa- 3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxamide as an off-white solid. LC-MS (ES, m/z): 863 (M+H+); 1H NMR: (400 MHz, DMSO -d6) d 12.77 (br s, 2H), 7.98 (s, 1H), 7.70 (s, 1H), 7.50 (s, 1H), 7.36 (s, 1H), 7.21 (s, 1H), 7.00 (s, 1H), 6.55 (s, 1H), 5.57-5.51 (m, 2H), 4.87-4.54 (m, 4H), 4.52-4.50 (m, 2H), 4.45-4.30 (m, 5H), 3.58-3.46 (m, 6H), 2.85 (q, 2H), 2.49-2.28 (m, 11H), 2.08 (s, 3H), 1.27 (t, 3H), 1.03 (t, 3H).
Example 141: Synthesis of (E)-l-(4-ethyl-2-methyloxazole-5-carboxamido)-15-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-4-(4-(2-hydroxyethyl)piperazine-l-carbonyl)- 8,9,16,19-tetrahydro-7H-6,10-dioxa-2,14,15a,19a-tetraazacyclopentadeca[3,2,l-cd:8,9,10- c'd']diindene-12-carboxamide, 1-178
Figure imgf000561_0001
[00973] To a solution of (3E)-21-carbamoyl-7-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-25-(2- ethyl-5-methylpyrazole-3-amido)-14, 18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9(27),10, 12,19(26),20,22,24-nonaene-l l-carboxylic acid, 29.1 (50 mg, 0.067 mmol, 1.00 equiv) and l-piperazineethanol(26.01 mg, 0.200 mmol, 3.00 equiv) in dimethyl formamide (5 mL) was added HATU (37.98 mg, 0.100 mmol, 1.50 equiv) and DIEA (43.04 mg, 0.333 mmol, 5.00 equiv). The resulting mixture was stirred for 2 h at RT. The reaction was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (36% ACN up to 49% in 10 min); Detector, UV 254/220 nm. The crude product (46 mg) was purified by Prep-HPLC with the following conditions: Column, XBridge Prep OBD C18 Column, 19*250mm, 5um; mobile phase, water (10% NH4HCO3+O. l %NHrH20) and methanol (47% methanol up to 70% in 10 min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 12.9 mg (22.45%) of (3E)-25-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(2-ethyl-5- methylpyrazole-3-amido)-21-[4-(2-hydroxyethyl)piperazine-l-carbonyl]-14,18-dioxa-l,6,8,24- tetraazapentacyclo[17.6.1.1A[6,9].0A[23,26].0A[13,27]]heptacosa-
3,7,9, 11, 13(27), 19, 21, 23(26), 24-nonaene-l 1-carboxamide as a white solid. LC-MS (ES, m/z): 863 (M+H+); H-NMR: (400 MHz, DMSO-de): d 12.86 (s, 1H), 12.71 (s, 1H), 7.99 (t, 1H), 7.70 (s, 1H), 7.50 (s, 1H), 7.36 (s, 1H), 7.21 (s, 1H), 7.01 (s, 1H), 6.55 (s, 1H), 5.65-5.51 (m, 2H), 4.97- 4.86 (m, 4H), 4.64-4.38 (m, 7H), 3.70-3.35 (m, 6H), 2.85 (q, 2H), 2.49-2.25 (m, 11H), 2.10 (s, 3H), 1.27 (t, 3H), 1.03 (t, 3H).
Example 142: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-(2-hydroxyethyl)-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxamide, 1-179
Figure imgf000563_0001
Synthesis of 142.1
[00974] To a solution of l-(2-((tert-butyldimethylsilyl)oxy)ethyl)-3-methyl-lH-pyrazole-5- carboxylic acid, 40.9 (123 mg, 0.43 mmol, 1.2 equiv), DIEA (232 mg, 1.8 mmol, 5.0 equiv) and HATU (2.5 mg, 0.54 mmol, 1.5 equiv) in dimethyl formamide (5 mL) was added methyl (E)-l-(4- (2-amino-5-carbamoyl- lH-benzo[d]imidazol- 1 -yl)but-2-en- 1 -yl)-2-(l -ethyl-3 -methyl- 1 H- pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate (200 mg, 0.36 mmol, 1.0 equiv). The resulting mixture was stirred at RT overnight. The reaction was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (41% ACN up to 51% in 10 min); Detector, UV 254/220 nm. This resulted in 100 mg (39.23%) of methyl (E)-l-(4-(5-carbamoyl-2-(l-(2- hydroxyethyl)-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)- 2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate as an off-white solid. LC-MS (ES, m/z): 708
Figure imgf000564_0001
Synthesis of 142.2
[00975] To a solution of methyl methyl (E)-l-(4-(5-carbamoyl-2-(l-(2-hydroxyethyl)-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3- methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate, 142.1 (100 mg, 0.141 mmol, 1.0 equiv) in methanol (10 mL) and water (2 mL) was added lithium hydroxide (13.6 mg, 0.564 mmol, 4 equiv). The resulting mixture was stirred for 4h at 70°C. The mixture was cooled to RT and concentrated under reduced pressure. The residue was acidified to pH 3 with IN hydrochloric acid. The precipitated solids were collected by filtration. The crude product was purified by reverse flash with the following conditions: Column, C18; mobile phase, Water (0.1% FA) and ACN (21% ACN up to 31% in 10 min); Detector, UV 254/220 nm. This resulted in 50 mg (51.12%) of (E)-l-(4-(5-carbamoyl-2-(l-(2-hydroxyethyl)-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxylic acid as a white solid. LC-MS (ES, m/z): 694 (M+H+).
Synthesis of 1-179
[00976] To a solution of (E)-l-(4-(5-carbamoyl-2-(l-(2-hydroxyethyl)-3-methyl-lH-pyrazole- 5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxylic acid, 142.1 (50 mg, 0.072 mmol, 1.0 equiv) and DIEA (46.44 mg, 0.36 mmol, 5.0 equiv) and HATU (41.04 mg, 0.108 mmol, 1.5 equiv) in dimethyl formamide (4 mL) was added ammonium chloride (20 mg, 0.36 mmol, 5 equiv). The resulting mixture was stirred at RT for 4h. The mixture was concentrated under reduced pressure. The crude product was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (0.1% FA) and ACN (32% ACN up to 43% in 10 min); Detector, UV 254/220 nm. The crude product (24 mg) was purified by Prep-HPLC with the following conditions: Column, XBridge Prep C18 OBD Column, 5um, 19* 150mm; mobile phase, Water (W/oMTCCh + 0.1% NH3Ή2O) and ACN (15% ACN up to 30% in 8min); The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 5.1 mg (10.22%) of (E)-l- (4-(5-carbamoyl-2-(l-(2-hydroxyethyl)-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-lH- benzo[d]imidazole-5-carboxamide as a white solid. LC-MS (ES, m/z): 693 (M+H+); 1H NMR (400 MHz, DMSO-i/e) d 12.76 (br s, 2H), 8.01-7.92 (m, 4H), 7.71 (d, 2H), 7.44 (dd, 2H), 7.32 (s, 2H), 6.57 (s, 2H), 6.03-5.92 (m, 2H), 4.95-4.76 (m, 5H), 4.68-4.51 (m, 4H), 3.82-3.71 (m, 2H), 2.13 (s, 6H), 1.29 (t, 3H).
Example 143: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(l-(3-hydroxypropyl)- 3-methyl-lH-pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-methyl- lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-180
Figure imgf000565_0001
143.3
143.2
Figure imgf000566_0001
Figure imgf000567_0001
143.11 1-180
Synthesis of 143.1
[00977] Into a 250-mL 3-necked round-bottom flask, was placed methyl 4-chloro-3- methylbenzoate (12.00 g, 64.998 mmol, 1.00 equiv), H2SO4 (25.00 mL). HNO3 (25.00 mL) was added dropwise at 0°C to the above mixture. The resulting solution was stirred for 2h at room temperature under nitrogen atmosphere. The resulting solution was diluted with 50 mL of H2O. The solids were collected by fdtration yielding 11 g of a light yellow solid, which was used directly into the next step without further purification.
Synthesis of 143.2
[00978] Into a 500-mL 3 -necked round-bottom flask, was placed methyl 143.1 (11.00 g, 47.905 mmol, 1.00 equiv), ammonia (250.00 mL). The resulting solution was stirred for 12h at 50 degrees C under nitrogen atmosphere. The solids were collected by filtration yielding 8.5 g (76.84%) of a yellow solid, which was used directly into the next step without further purification.
Synthesis of 143.3
[00979] Into a 250-mL 3-necked round-bottom flask, was placed 143.2 (5.00 g, 23.298 mmol, 1.00 equiv), DMSO (31.64 mL), DIEA (18066.71 mg, 139.788 mmol, 6.00 equiv), tert-butyl (E)- (4-aminobut-2-en-l-yl)carbamate hydrochloride (7793.181 mg, 34.947 mmol, 1.50 equiv). The resulting solution was stirred for 12h at 100 degrees C under nitrogen atmosphere. The residue was purified by reverse flash chromatography with the following conditions: column, C 18 silica gel; mobile phase, MeCN in water, 5% to 90% gradient in 50 min; detector, UV 254 nm. This resulted in 2 g (23.56%) of an off-white solid. LC-MS (ES, m/z): 364 [M+H]+.
Synthesis of 143.4
[00980] Into a 100-mL 3-necked round-bottom flask, was placed tert-butyl 143.3 (2.00 g, 5.488 mmol, 1.00 equiv), HO Ac (30.00 mL), Zn (5384.90 mg, 82.327 mmol, 15.00 equiv). The resulting solution was stirred for 12h at 25 degrees C. The solids were fdtered out. The resulting mixture was concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN:H20 =10% increasing to ACN:H2O=50% within 15 min; Detector, UV 254 nm yielding 1.1 g (59.93%) of an off-white solid. LC-MS (ES, m/z): 334[M+H]+.
Synthesis of 143.5
[00981] Into a 100-mL 3-necked round-bottom flask, was placed 143.4 (1.10 g, 2.990 mmol, 1.00 equiv), MeOH (20.00 mL), BrCN (0.95 g, 8.971 mmol, 3.00 equiv). The resulting solution was stirred for 2h at 25 degrees C under nitrogen atmosphere. The resulting mixture was concentrated under vacuum yielding 1.3 g of an off-white solid. LC-MS (ES, m/z): 360 [M+H]+.
Synthesis of 143.6
[00982] Into a 100-mL 3-necked round-bottom flask, was placed 143.5 (1.30 g, 3.617 mmol, 1 equiv), 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (1683.49 mg, 10.851 mmol, 3.00 equiv) in DMF (26 mL), DIEA (2804.71 mg, 21.701 mmol, 6.00 equiv) and HATU (4125.69 mg, 10.851 mmol, 3.00 equiv) were added. The resulting solution was stirred for 2h at 70 degrees C under nitrogen atmosphere. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN:H20 =10% increasing to ACN:H2O=50% within 15 min; Detector, UV 254 nm yielding 0.7 g of a white solid.
Synthesis of 143.7
[00983] Into a 50-mL 3-necked round-bottom flask, was placed 143.6 (0.68 g, 1.369 mmol, 1.00 equiv), DCM (9.00 mL), TFA (3 mL). The resulting solution was stirred for 12h at 25 degrees C. The resulting mixture was concentrated under vacuum to yield 300 mg.
Synthesis of 143.8
[00984] Into a 500-mL 3-necked round-bottom flask, was placed LCl (8.669 g, 162.066 mmol, 3.00 equiv), DMF (200.00 mL), DIEA (4.189 g, 324.132 mmol, 6.00 equiv), HATU (61622.26 mg, 162.066 mmol, 3.00 equiv), 4-fluoro-3-nitrobenzoic acid (10.00 g, 54.022 mmol, 1.00 equiv). The resulting solution was stirred for 12h at 25 degrees C. The crude product 8 g was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, Cl 8 silica gel; mobile phase, ACN:H2O=10% increasing to ACN:H2O=50% within 15 min; Detector, UV 254 nm to yield 7.3 g.
Synthesis of 143.9
[00985] Into a 50-mL 3-necked round-bottom flask, was placed 143.8 (290.00 mg, 1.575 mmol, 1.00 equiv), DMSO (9.00 mL), DIEA (1221.35 mg, 9.450 mmol, 6.00 equiv), and 143.7 (312.21 mg, 0.788 mmol, 0.50 equiv). The resulting solution was stirred for 12 hr at 60 degrees C. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN:H2O=10% increasing to ACN:H2O=50% within 15 min; Detector, UY 254 nm to yield 180 mg.
Synthesis of 143.10
[00986] Into a 50-mL 3-necked round-bottom flask, was placed 143.9 (170.00 mg, 0.285 mmol, 1.00 equiv), AcOH (10.00 mL), Zn (280.04 mg, 4.281 mmol, 15.00 equiv). The resulting solution was stirred for 12h at 25 degrees C under nitrogen atmosphere. The solids were filtered out. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN:H2O=10% increasing to ACN:H2O=50% within 15 min; Detector, UV 254 nm to yield 500 mg, which was used directly into the next step without further purification.
Synthesis of 143.11
[00987] Into a 50-mL 3-necked round-bottom flask, was placed 143.10 (500.00 mg, 0.942 mmol, 1.00 equiv), MeOH (10.00 mL), BrCN (598.89 mg, 5.654 mmol, 6.00 equiv). The resulting solution was stirred for 12h at 25 degrees C. The resulting mixture was concentrated under vacuum. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, silica gel; mobile phase, ACN:H2O=10% increasing to ACN:H2O=60% within 30 min; Detector, UV 254 nm to yield 120 mg (22.46%) of a white solid.
Synthesis of 1-180
[00988] Into a 50-mL 3-necked round-bottom flask, was placed l-(3-((tert- butyldimethylsilyl)oxy)propyl)-3-methyl-lH-pyrazole-5-carboxylic acid (386.77 mg, 1.296 mmol, 6.00 equiv), DMF (15.00 mL), HATU (492.74 mg, 1.296 mmol, 6.00 equiv), DIEA (418.72 mg, 3.240 mmol, 15.00 equiv) and 143.11 (120.00 mg, 0.216 mmol, 1.00 equiv). The solution was stirred overnight at 85 °C under nitrogen atmosphere. Then NaOH (4319.35 mg, 107.992 mmol, 500.00 equiv) and H2O (5 mL) were added. The resulting solution was stirred for another 12h at 25 degrees C. The crude product was purified by Prep-HPLC with the following conditions: Column: XBridge Shield RP18 OBD Column, 30*150 mm, 5 um; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 22% B to 32% B in 7 min; 254; 220 nm; RT: 6.55 min. This resulted in the desired crude product. The crude product was purified by Prep-HPLC with the following conditions Column: XBridge Shield RP18 OBD Column, 30* 150 mm, 5 um; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 22% B to 32% B in 7 min; 254;220 nm; RT:6.45 min. LC- MS: (ES, in :): [M+H]+ 722.4, [M-H] 720.2; ^-NMR^OO MHz, DMSO -d6, ppm) 512.30 (s, 2H), 57.85-7.98 (m, 4H), 57.68-7.72 (d, 1H), 57.40-7.49 (m, 2H), 57.20-7.35 (m, 2H), 56.42-6.41 (m, 1H), 55.85-5.97 (d, 1H), 55.45-5.60 (m, 1H), 54.76-5.09 (m, 5H), 54.42-4.60 (m, 4H), 53.38- 3.40 (m, 3H), 52.70-2.88 (m, 2H), 52.40 (s, 3H), 52.13 (s, 3H), 51.86-1.90 (t, 2H), 51.01-1.06 (t,
3H).
Example 144: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(4-ethyl-2- methyloxazole-5-carboxamido)-7-(2-(3-hydroxyazetidin-l-yl)-2-oxoethoxy)-lH- benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-methoxy-lH-benzo[d]imidazol-2-yl)-4-ethyl-2- methyloxazole-5-carboxamide, 1-181
Figure imgf000570_0001
144.1
144.2
Figure imgf000571_0001
1-181
Synthesis of 144.1
[00989] Into a 250-mL 3-necked round-bottom flask, was placed azetidin-3-ol hydrochloride (5.00 g, 45.641 mmol, 1.00 equiv) in THF (50.00 mL, 617.150 mmol, 13.52 equiv) was added imidazole (6214.27 mg, 91.283 mmol, 2.00 equiv) and TBDPSC1 (13799.50 mg, 50.205 mmol, 1.10 equiv). The resulting solution was stirred overnight at room temperature. The resulting solution was diluted with 100 mL of H20. The resulting solution was extracted with 3x100 mL of ethyl acetate concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 : 1). This resulted in 6 g (42.20%) of 3-[(tert- butyldiphenylsilyl)oxy]azetidine as yellow oil.
Synthesis of 144.2
[00990] To a stirred solution of 3-[(tert-butyldiphenylsilyl)oxy]azetidine, 141.1 (500.00 mg, 1.605 mmol, 1.00 equiv) and Et3N (406.06 mg, 4.013 mmol, 2.50 equiv) in DCM (10.00 mL) was added chloroacetyl chloride (217.54 mg, 1.926 mmol, 1.20 equiv) in portions at 0 degrees C under air atmosphere. The resulting mixture was stirred overnight at room temperature under nitrogen atmosphere. The residue was purified by silica gel column chromatography, eluted with PE/EtOAc (5: 1) to afford l-[3-[(tert-butyldiphenylsilyl)oxy]azetidin-l-yl]-2-chloroethanone (370mg, 59.41%) as a yellow oil.
Synthesis of 144.3
[00991] To a stirred solution of l-[3-[(tert-butyldiphenylsilyl)oxy]azetidin-l-yl]-2- chloroethanone, 141.1 (136.00 mg, 0.351 mmol, 1.00 equiv) and Cs2C03 (456.84 mg, 1.402 mmol, 4.00 equiv) in DMF(2.00 mL) was added l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-hydroxy-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-methoxy-l,3-benzodiazole-5-carboxamide, 1-12 (170.00 mg, 0.235 mmol, 0.67 equiv) in portions at 50 degrees C under nitrogen atmosphere. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, FA in water, 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 7-(2-[3-[(tert- butyldiphenylsilyl)oxy]azetidin-l-yl]-2-oxoethoxy)-l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2- methyl-l,3-oxazole-5-amido)-7-methoxy-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2- methyl-1, 3-oxazole-5-amido)-l,3-benzodiazole-5-carboxamide (100 mg, 26.51%) as a off-white solid.
Synthesis of 1-181
[00992] Into a 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed 7-(2-[3-[(tert-butyldiphenylsilyl)oxy]azetidin-l-yl]-2-oxoethoxy)-l-[(2E)-4-[5- carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-methoxy-l,3-benzodiazol-l-yl]but-2-en- l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazole-5-carboxamide, 144.3 (120.00 mg, 0.111 mmol, 1.00 equiv) in MeOH (1.00 mL),TFA (1.00 mL) was added. The resulting mixture was stirred for 1 h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product (60 mg) was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, NH4HC03 in water, 10% to 50% gradient in 35 min; detector, UV 254 nm. The crude product (60 mg) was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): Column, XBridge Prep OBD C18 Column, 19*250mm, 5um; mobile phase, water(10MMOL/L NH4HC03) and ACN (hold 30% PhaseB in 14 min); Detector, UV 254nm.) to afford l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl- l,3-oxazole-5-amido)-7-methoxy-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-[2-(3-hydroxyazetidin-l-yl)-2-oxoethoxy]-l,3-benzodiazole-5-carboxamide (7.2mg,5.35%) as a off-white solid. LC-MS (ES, m/z): 838 (M+H+); ¾NMR (400 MHz, DMSO- <k) S 12.69 (s, 2H), 7.96-7.92(d, 2H), 7.64 (s, 2H), 7.40-7.25 (m, 4H), 5.97-5.78(m, 3H), 4.99-4.67 (d, 4H), 4.62-4.23 (m, 4H), 4.04-3.58 (m, 6H), 3.33-2.74(m, 4H), 2.39 (s, 6H), 0.90(s, 6H).
Example 145: Synthesis of (E)-N-(5-carbamoyl-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH- pyrazole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-(morpholinomethyl)- lH-benzo[d]imidazol-2-yl)-2,4-dimethyloxazole-5-carboxamide, 1-182
Figure imgf000574_0001
145.4 145.5 .
H2N
Figure imgf000575_0001
Synthesis of 148.1
[00993] To a stirred solution of dimethyl-1, 3-oxazole-5-carboxylic acid (0.71 g, 5.061 mmol, 1.50 equiv) and HATU (1.92 g, 5.061 mmol, 1.50 equiv) and DIEA (1.31 g, 10.123 mmol, 3.00 equiv) in DMF (15.00 mL) was added tert-butyl N-[(2E)-4-[2-amino-5-carbamoyl-7-(morpholin- 4-ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate (1.50 g, 3.374 mmol, 1.00 equiv) in portions at 85 degrees C under nitrogen atmosphere. The precipitated solids were collected by filtration and washed with water (3x20 mL). The residue was purified by Prep-TLC (CH2C12 / MeOH 5: 1) to afford tert-butyl N-[(2E)-4-[5-carbamoyl-2-(2, 4-dimethyl- 1,3-oxazol e-5-amido)-7- (morpholin-4-ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate (1.4 g, 73.09%) as a dark yellow solid.
Synthesis of 145.2
[00994] To a stirred solution of tert-butyl N-[(2E)-4-[5-carbamoyl-2-(2,4-dimethyl-l,3- oxazole-5-amido)-7-(morpholin-4-ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]carbamate,
145.1 (1.40 g, 2.466 mmol, 1.00 equiv) in MeOH (10.00 mL) was added HCl(gas)in 1,4-dioxane (10.00 mL) in portions at 0 degrees C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The precipitated solids were collected by filtration and washed with MeOH (3x10 mL). This resulted in l-[(2E)-4-aminobut-2-en-l-yl]-2-(2, 4-dimethyl - l,3-oxazole-5-amido)-7-(morpholin-4-ylmethyl)-l,3-benzodiazole-5-carboxamide (1.1 g, 95.40%) as a yellow solid. Synthesis of 145.3
[00995] To a stirred solutionof l-[(2E)-4-aminobut-2-en-l-yl]-2-(2,4-dimethyl-l,3-oxazole-5- amido)-7-(morpholin-4-ylmethyl)-l,3-benzodiazole-5-carboxamide, 145.2 (1.30 g, 2.781 mmol, 1.00 equiv) and DIEA (7.19 g, 55.611 mmol, 20.00 equiv) in DMSO (15.00 mL) was added 4- fluoro-3-nitrobenzamide (0.61 g, 3.337 mmol, 1.20 equiv) in portions at 60 degrees C under nitrogen atmosphere. The resulting mixture was washed with 3x20 mL of water. The precipitated solids were collected by filtration and washed with PE (3x5 mL). This resulted in l-[(2E)-4-[(4- carbamoyl-2-nitrophenyl)amino]but-2-en-l-yl]-2-(2,4-dimethyl-l,3-oxazole-5-amido)-7- (morpholin-4-ylmethyl)-l,3-benzodiazole-5-carboxamide (1.6 g, 91.10%) as a yellow solid. Synthesis of 145.4
[00996] To a stirred solution of l-[(2E)-4-[(4-carbamoyl-2-nitrophenyl)amino]but-2-en-l-yl]- 2-(2,4-dimethyl-l,3-oxazole-5-amido)-7-(morpholin-4-ylmethyl)-l,3-benzodiazole-5- carboxamide, 145.4 (800.00 mg, 1.267 mmol, 1.00 equiv) in HAC(10.00 mL) was added Zn (0.83 g, 12.689 mmol, 10.02 equiv) in portions at room temperature under nitrogen atmosphere. Desired product could be detected by LCMS.The resulting mixture was filtered, the filter cake was washed with MeOH (3x10 mL). The filtrate was concentrated under reduced pressure. This resulted in 1- [(2E)-4-[(2-amino-4-carbamoylphenyl)amino]but-2-en-l-yl]-2-(2,4-dimethyl-l,3-oxazole-5- amido)-7-(morpholin-4-ylmethyl)-l,3-benzodiazole-5-carboxamide (600 mg, 78.74%) as a yellow solid.
Synthesis of 145.5
[00997] To a stirred solution of l-[(2E)-4-[(2-amino-4-carbamoylphenyl)amino]but-2-en-l-yl]- 2-(2,4-dimethyl-l,3-oxazole-5-amido)-7-(morpholin-4-ylmethyl)-l,3-benzodiazole-5- carboxamide, 145.4 (800.00 mg, 1.330 mmol, 1.00 equiv) in MeOH (10.00 mL) was added BrCN (422.51 mg, 3.989 mmol, 3.00 equiv) in portions at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, MeOH in water, 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 1-[(2E)- 4-(2 -amino-5 -carbamoyl- 1 ,3-benzodiazol- 1 -yl)but-2-en- 1 -yl]-2-(2, 4-dimethyl- 1 ,3 -oxazole-5- amido)-7-(morpholin-4-ylmethyl)-l,3-benzodiazole-5-carboxamide(220mg,26.40%) as a off- white solid. Synthesis of 1-182
[00998] Into a 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed 2-ethyl-5-methylpyrazole-3-carboxylic acid (73.80 mg, 0.479 mmol, 3.00 equiv) ,HATU (182.02 mg, 0.479 mmol, 3.00 equiv),5,6-dimethoxy-l-benzothiophene-2- carboxylic acid (57.80 mg, 1.00 equiv), DIEA (123.74 mg, 0.957 mmol, 6.00 equiv),l-[(2E)-4-(2- amino-5-carbamoyl-l,3-benzodiazol-l-yl)but-2-en-l-yl]-2-(2,4-dimethyl-l,3-oxazole-5-amido)- 7-(morpholin-4-ylmethyl)-l,3-benzodiazole-5-carboxamide, 145.5 (100.00 mg, 0.160 mmol, 1.00 equiv). The resulting mixture was stirred for 2 h at 85 degrees C.The resulting mixture was washed with 1x20 mL of water. The resulting solution was extracted with 3x20 mL of ethyl acetate and the organic layers combined. The crude product was purified by Flash-Prep-HPLC the following conditions (2#SHIMADZU (HPLC-01)): Column, XBridge Prep OBD C18 Column, 19*250mm, 5um; mobile phase, water(10MMOL/L NH4HC03) and ACN (hold 30% PhaseB in 14 min); Detector, UV 254nm.) . This resulted in l-[(2E)-4-[5-carbamoyl-2-(2-ethyl-5-methylpyrazole-3- amido)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(2,4-dimethyl-l,3-oxazole-5-amido)-7- (morpholin-4-ylmethyl)-l,3-benzodiazole-5-carboxamide(45.9mg,37.71%) as a white solid. LC- MS (ES, m/z): 763 (M+H+); H-NMR: (400 MHz, DMSO-d6): d 12.80 (s, 2H), 7.98-7.91 (m, 4H), 7.72 (d, 1H), 7.50 (d, 1H), 7.32 (s, 2H), 6.56 (d, 1H), 5.98 (d, 1H), 5.45 (s, 2H), 4.81 (s, 2H), 4.56 (d, 2H), 3.46 (s, 6H), 2.41-2.30 (d, 6H), 2.15 (s, 7H), 1.28 (t, 3H).
Example 146: Synthesis of butyl (E)-2-((5-carbamoyl-l-(4-(5-carbamoyl-2-(4-ethyl-2- methyloxazole-5-carboxamido)-7-(morpholinomethyl)-lH-benzo[d]imidazol-l-yl)but-2-en- l-yl)-2-(4-ethyl-2-methyloxazole-5-carboxamido)-lH-benzo[d]imidazol-7-yl)oxy)acetate, I- 183
Figure imgf000577_0001
Figure imgf000578_0001
1-183
Synthesis of 146.1
[00999] To a stirred solution of 4-[[(2E)-4-aminobut-2-en-l-yl]amino]-3-(morpholin-4- ylmethyl)-5-nitrobenzamide, 127.6 (2.30 g, 6.583 mmol, 1.00 equiv)in DMSO (8.00 mL) were added DIEA (8.51 g, 65.829 mmol, 10.00 equiv) and butyl 2-(5-carbamoyl-2-chloro-3- nitrophenoxy)acetate(2.18 g, 6.583 mmol, 1.00 equiv) in portions at 120 degrees under nitrogen atmosphere through microwave heated for lh. The resulting mixture was extracted with EtOAc (3x30 mL). The combined organic layers were washed with brine (2x10 mL), dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by reverse phase flash with the following conditions(Column: XBridge Prep OBD C18 Column, 30x 150mm 5um; Mobile Phase A:Water(10MMOL/L NH4HCO3+0.1%NH3 H20), Mobile Phase B:ACN; Flow rate:60 mL/min; Gradient: 10 B to 40 B in 7 min; 210/254 nm; RTF6.62; RT2:; Injection Volumn: ml; Number Of Runs:;) to afford butyl 2-(5-carbamoyl-2-[[(2E)-4-[[4-carbamoyl-2-(morpholin-4-ylmethyl)-6- nitrophenyl]amino]but-2-en-l-yl]amino]-3-nitrophenoxy)acetate(550 mg, 12.98%)as a yellow solid. LC-MS (ES, m/z): 644 (M+H+).
Synthesis of 146.2
[001000] To a stirred solution of butyl 2-(5-carbamoyl-2-[[(2E)-4-[[4-carbamoyl-2- (morpholin-4-ylmethyl)-6-nitrophenyl]amino]but-2-en-l-yl]amino]-3- nitrophenoxy)acetate(250.00 mg, 0.388 mmol, 1.00 equiv) in methanol (40.00 mL), ammonia (10.00 mL, 352.309 mmol, 839.87 equiv) were added Na2S204 (676.24 mg, 3.884 mmol, 10.00 equiv) in portions at room temperature under air atmosphere. The aqueous layer was extracted with EtOAc (3x30 mL). The resulting mixture was concentrated under vacuum. This resulted in butyl 2-(3-amino-2-[[(2E)-4-[[2-amino-4-carbamoyl-6-(morpholin-4- ylmethyl)phenyl]amino]but-2-en-l-yl]amino]-5-carbamoylphenoxy)acetate(180 mg, 73.52%)as a yellow solid. LC-MS (ES, m/z): 584 (M+H+).
Synthesis of 146.3
[001001] To a stirred solution of butyl 2-(3-amino-2-[[(2E)-4-[[2-amino-4-carbamoyl-6- (morpholin-4-ylmethyl)phenyl]amino]but-2-en-l-yl]amino]-5-carbamoylphenoxy)acetate, 146.2 (200.00 mg, 0.343 mmol, 1.00 equiv)in MeOH(10.00 mL, 246.989 mmol, 720.83 equiv)were added BrCN(362.94 mg, 3.427 mmol, 10.00 equiv)in portions at room temperature under air atmosphere. The reaction was quenched with sat. NaOH(aq.) at 0 degrees C. The residue/crude product was purified by reverse phase flash with the following conditions(Column: XBridge Prep OBD C18 Column, 30x 150mm 5um; Mobile Phase A:Water(10MMOL/L NH4HCO3+0.1%NH3 H20), Mobile Phase B:ACN; Flow rate:40 mL/min; Gradient: 10 B to 40 B in 7 min; 210/254 nm; RTF6.62; RT2:; Injection Volumn: ml; Number Of Runs:;) to afford butyl 2-([2-amino-3-[(2E)-4-[2-amino-5-carbamoyl-7-(morpholin-4-ylmethyl)-l,3-benzodiazol- l-yl]but-2-en-l-yl]-6-carbamoyl-l,3-benzodiazol-4-yl]oxy)acetate(45 mg, 23.03%) as a yellow oil. LC-MS (ES, m/z): 634 (M+H ).
Synthesis of 1-183
[001002] To a stirred solution of butyl 2-([2-amino-3-[(2E)-4-[2-amino-5-carbamoyl-7- (morpholin-4-ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-6-carbamoyl-l,3-benzodiazol-4- yl]oxy)acetate, 146.3 (75.00 mg, 0.118 mmol, 1.00 equiv)and 4-ethyl-2-methyl-l,3-oxazole-5- carboxylic acid (36.72 mg, 0.237 mmol, 2.00 equiv)in DMF (5 mL) were added DMAP (86.75 mg, 0.710 mmol, 6 equiv) and HOBT (95.95 mg, 0.710 mmol, 6 equiv), EDCI (136.13 mg, 0.710 mmol, 6 equiv)dropwise at 120 degrees C under nitrogen atmosphere. The final reaction mixture was irradiated with microwave radiation for 1 h at 120 degrees C. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with ACN/H20 (2:5) to afford butyl 2-([6-carbamoyl-3-[(2E)-4-[5- carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(morpholin-4-ylmethyl)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazol-4- yl]oxy)acetate (90 mg, 83.75%)as a yellow solid. LC-MS (ES, m/z): 908 (M+H+/H NMR (400 MHz, DMSO- ) d 12.81 - 12.72 (m, 2H), 7.96 (s, 2H), 7.91 (d , J = 11.4 Hz, 3H), 7.68 (s, 1H), 7.53 (s, 1H), 7.34 (s, 3H), 5.91 (d , J = 14.7 Hz, 1H), 5.53 (s, 1H), 5.15 (s, 2H), 4.98 (s, 2H), 4.71 (s, 2H), 3.45 (s, 10H), 2.83 - 2.74 (m, 4H), 2.40 (s, 6H), 2.19 (s, 4H), 1.35 (s, 9H), 0.99 (t, J= 7.5 Hz, 6H).
Example 147: Synthesis of methyl (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5- carboxamido)-7-(morpholinomethyl)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(4-ethyl-2- methyloxazole-5-carboxamido)-7-methyl-lH-benzo[d]imidazole-5-carboxylate, 1-184
Figure imgf000580_0001
o 147.2
Figure imgf000581_0001
Synthesis of 147.1
[001003] To a stirred solution of 4-[[(2E)-4-aminobut-2-en-l-yl]amino]-3-(morpholin-4- ylmethyl)-5-nitrobenzamide hydrochloride, 127.6 (1.00 g, 2.592 mmol, 1.00 equiv)and methyl 4- fluoro-3 -methyl-5 -nitrobenzoate(0.66 g, 3.096 mmol, 1.19 equiv)in DMSO (10.00 mL) was added DIEA (1.34 g, 10.368 mmol, 4.00 equiv) dropwise at 60 degrees C under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 60 degrees C under nitrogen atmosphere. The resulting mixture was extracted with CHC13 (3x50 mL). The combined organic layers were washed with water (2x50 mL), dried over anhydrous Na2S04. After fdtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2C12/MeOH (80: 1) to afford methyl 4-[[(2E)-4-[[4-carbamoyl-2-(morpholin-4-ylmethyl)- 6-nitrophenyl]amino]but-2-en-l-yl]amino]-3-methyl-5-nitrobenzoate (1 g, 71.12%)as a red solid. LC-MS (ES, m/z): 543 (M+H+)
Synthesis of 147.2
[001004] To a stirred solution of methyl 4-[[(2E)-4-[[4-carbamoyl-2-(morpholin-4- ylmethyl)-6-nitrophenyl]amino]but-2-en-l-yl]amino]-3-methyl-5-nitrobenzoate, 147.1 (900.00 mg, 1.659 mmol, 1.00 equiv) in methanol (15.00 mL) was added Na2S204 (1444.08 mg, 8.294 mmol, 5.00 equiv)dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The resulting mixture was extracted with CHC13(3xl00 mL). The combined organic layers were washed with water(2xl00mL), dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. This resulted in methyl 3-amino-4-[[(2E)-4-[[2-amino-4-carbamoyl-6- (morpholin-4-ylmethyl)phenyl]amino]but-2-en-l-yl]amino]-5-methylbenzoate (500 mg, 62.46%) as a yellow solid. The crude product mixture was used in the next step directly without further purification. LC-MS (ES, m/z): 483 (M+H+).
Synthesis of 147.3
[001005] To a stirred solution of methyl 3-amino-4-[[(2E)-4-[[2-amino-4-carbamoyl-6- (morpholin-4-ylmethyl)phenyl]amino]but-2-en-l-yl]amino]-5-methylbenzoate (500.00 mg, 1.036 mmol, 1.00 equiv)in MeOH(10.00 mL)was added BrCN(548.72 mg, 5.180 mmol, 5.00 equiv)dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The residue was purified by silica gel column chromatography, eluted with H20/CAN (2:5)to afford methyl 2-amino- l-[(2E)-4- [2- amino-5-carbamoyl-7-(morpholin-4-ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-7-methyl- l,3-benzodiazole-5-carboxylate (250 mg, 45.30%) as a yellow solid. LC-MS (ES, m/z): 533 (M+H+).
Synthesis of 1-184
[001006] To a stirred solution of 2-amino-l-[(2E)-4-(2-amino-5-carbamoyl-7-methyl-l,3- benzodiazol-l-yl)but-2-en-l-yl]-7-(morpholin-4-ylmethyl)-l,3-benzodiazole-5-carboxamide, 147.3 (500.00 mg, 0.966 mmol, 1.00 equiv)and 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (599.52 mg, 3.864 mmol, 4.00 equiv) in DMF (8.00 mL) were added DIEA (998.80 mg, 7.728 mmol, 8.00 equiv) and HATU (1469.22 mg, 3.864 mmol, 4.00 equiv) dropwise at 120 degrees C under nitrogen atmosphere. The final reaction mixture was irradiated with microwave radiation for 1 h at 120 degrees C. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with ACN/H20(2:5) to afford methyl l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2 -methyl-1, 3-oxazole-5-amido)-7-(morpholin-4- ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl -2 -methyl-1, 3-oxazole-5-ami do)-7- methyl-l,3-benzodiazole-5-carboxylate (610 mg, 78.26%) as a red solid.
LC-MS (ES, m/z): 807 (M+H+); ¾ NMR (400 MHz, DMSO -d6) d 12.80 (s, 2H), 7.98 (s, 2H), 7.92 (s, 1H), 7.57 (s, 2H), 7.32 (s, 1H), 5.57 (d, J= 15.3 Hz, 2H), 5.46 (d, J= 16.6 Hz, 2H), 4.92 (s, 2H), 3.86 (s, 3H), 3.45 (d, J = 15.5 Hz, 6H), 2.78 (t, J= 7.4 Hz, 4H), 2.41 (d, J= 4.8 Hz, 9H), 2.21 (s, 4H), 1.01 (dt, J= 12.9, 7.5 Hz, 6H). Example 148: Synthesis of methyl (E)-l-(4-(5-carbamoyl-2-(4-ethyl-2-methyloxazole-5- carboxamido)-7-(morpholinomethyl)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(4-ethyl-2- methyloxazole-5-carboxamido)-lH-benzo[d]imidazole-5-carboxylate, 1-185
Figure imgf000583_0001
Synthesis of 148.1
[001007] To a stirred solution of 4-[[(2E)-4-aminobut-2-en-l-yl]amino]-3-(morpholin-4- ylmethyl)-5-nitrobenzamide hydrochloride, 127.6 (1.00 g, 2.592 mmol, 1.00 equiv) and methyl 4- fluoro-3-nitrobenzoate (0.62 g, 3.110 mmol, 1.20 equiv) in DMSO(20.00 mL) was added DIEA (1.34 g, 10.368 mmol, 4.00 equiv) dropwise at 60 degrees C under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 60 degrees C under nitrogen atmosphere. The resulting mixture was extracted with CHC13 (3x100 mL). The combined organic layers were washed with water (2x100 mL), dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2/MeOH (80: 1) to afford methyl 4-[[(2E)-4-[[4-carbamoyl-2-(morpholin-4-ylmethyl)- 6-nitrophenyl]amino]but-2-en-l-yl]amino]-3-nitrobenzoate (1 g, 73.01%) as a red solid. LC-MS (ES, m/z): 529 (M+H+).
Synthesis of 148.2
[001008] To a stirred solution of methyl 4-[[(2E)-4-[[4-carbamoyl-2-(morpholin-4- ylmethyl)-6-nitrophenyl]amino]but-2-en-l-yl]amino]-3-nitrobenzoate, 148.1 (1.00 g, 1.892 mmol, 1.00 equiv) in methanol (20.00 mL)was added Na2S204 (1.65 g, 9.477 mmol, 5.01 equiv) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2h at room temperature under nitrogen atmosphere. The resulting mixture was extracted with CHC13 (3x100 mL). The combined organic layers were washed with water (2x100 mL), dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. This resulted in methyl 3-amino-4-[[(2E)-4-[[2-amino-4-carbamoyl-6-(morpholin-4- ylmethyl)phenyl]amino]but-2-en-l-yl]amino]benzoate (450mg, 50.76%) as a yellow solid. The crude product mixture was used in the next step directly without further purification. LC-MS (ES, m/z): 469 (M+H+).
Synthesis of 148.3
[001009] To a stirred solution of methyl 3-amino-4-[[(2E)-4-[[2-amino-4-carbamoyl-6- (morpholin-4-ylmethyl)phenyl]amino]but-2-en-l-yl]amino]benzoate, 148.2 (450.00 mg, 0.960 mmol, 1.00 equiv) in MeOH (10.00 mL)was added BrCN (508.63 mg, 4.802 mmol, 5.00 equiv) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeOH in water, 10% to 50% gradient in 10 min; detector, UV 254 nm. to afford methyl 2-amino- l-[(2E)-4-
583
5UB5TITUTE SHEET (RULE 26) [2-amino-5-carbamoyl-7-(morpholin-4-ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3- benzodiazole-5-carboxylate(250 mg,50.20%)as a yellow solid. LC-MS (ES, m/z): 519 (M+H+).
Synthesis of 1-185
[001010] To a stirred solution of methyl 2-amino-l-[(2E)-4-[2-amino-5-carbamoyl-7- (morpholin-4-ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazole-5-carboxylate, 148.3 (550.00 mg, 1.061 mmol, 1.00 equiv) and 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (658.22 mg, 4.242 mmol, 4.00 equiv)in DMF(8.00 mL)were added DIEA (1096.60 mg, 8.485 mmol, 8 equiv) and HATU (1613.08 mg, 4.242 mmol, 4 equiv) dropwise at 120 degrees C under nitrogen atmosphere. The final reaction mixture was irradiated with microwave radiation for 1 h at 120 degrees C. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with ACN/H20(2:5) to afford methyl 1- [(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(morpholin-4-ylmethyl)-l,3- benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l,3-benzodiazole-5- carboxylate (530mg, 63.03%) as a red solid. LC-MS (ES, m/z): 793 (M+H+).
¾ NMR (400 MHz, DMSO -d6) d 12.79 (s, 2H), 8.07 (s, 1H), 7.96 (s, 1H), 7.90 (s, 1H), 7.76 (d, J = 8.4 Hz, 1H), 7.57 (s, 1H), 7.47 (d, J= 8.5 Hz, 1H), 7.31 (s, 1H), 5.99 (d, J= 15.6 Hz, 1H), 5.42 (d, J= 16.0 Hz, 1H), 5.17 (s, 2H), 4.78 (s, 2H), 3.87 (s, 3H), 3.47 (s, 6H), 2.80 (p , J= 7.7 Hz, 4H), 2.41 (d, J= 4.9 Hz, 6H), 2.24 (s, 4H), 1.01 (t, J= 7.5 Hz, 6H).
Example 149: Synthesis of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-N-propyl-lH-benzo[d]imidazole-5-carboxamide, Z-ll
Figure imgf000586_0001
[001011] To a solution of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazole-5-carboxylic acid, 43.1 (50 mg, 0.074 mmol, 1 equiv) and propan- 1 -amine (6.55 mg, 0.111 mmol, 1.5 equiv) in dimethyl formamide (5 mL) was added HATU (33.75 mg, 0.089 mmol, 1.2 equiv) and DIEA (47.73 mg, 0.37 mmol, 5.00 equiv). The resulting mixture was stirred at 85°C for 2h. The mixture was cooled to r.t and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Sunfire Prep Cl 8 OBD Column 19*250mm; mobile phase, Water (0.1% FA) and ACN (25% ACN up to 40% in 16 min); Detector, UV 254/210 nm. This resulted in 12.0 mg (22.6%) of (E)-l-(4-(5-carbamoyl-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)- lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-2-(l-ethyl-3-methyl-lH-pyrazole-5-carboxamido)-N- propyl-lH-benzo[d]imidazole-5-carboxamide as an off-white solid. LC-MS (ES, m/z): 719 (AMT ); ^ MR (400 MHz, DMSO -d6) d 12.81 (br s, 2H), 8.42 (t, 1H), 8.04-7.95 (m, 3H), 7.74- 7.66 (m, 2H), 7.45 (dd, 2H), 7.34 (s, 1H), 6.57 (s, 2H), 6.02-5.94 (m, 2H), 4.95-4.80 (m, 4H), 4.55 (q, 4H), 3.31-3.21 (m, 2H), 2.14 (s, 6H), 1.64-51 (m, 2H), 1.31 (t, 6H), 0.92 (t, 3H).
Example 150: Synthesis of benzyl (3-(2-(l-(5-(5-((l-(3-(((benzyloxy)carbonyl)amino)propyl)- 5-carbamoyl-lH-benzo[d]imidazol-2-yl)carbamoyl)-l-ethyl-3-methyl-lH-pyrazol-4- yl)pentyl)-3-methyl-lH-pyrazole-5-carboxamido)-5-carbamoyl-lH-benzo[d]imidazol-l- yl)propyl)carbamate, 1-139
Figure imgf000587_0001
Synthesis of 150.1
[001012] To a stirred mixture of 4-chloro-3-nitrobenzamide (3 g, 14.957 mmol, 1 equiv) and benzyl N-(3-aminopropyl)carbamate hydrochloride (4.39 g, 17.948 mmol, 1.2 equiv) in DMF (60 mL) was added K2C03 (6.20 g, 44.870 mmol, 3 equiv). The resulting mixture was stirred overnight at 80oC. The mixture was cooed to room temperature and diluted with water. The precipitated solids were collected by filtration. This resulted in 4 g (71.89 %) of benzyl 3-(4- carbamoyl-2-nitrophenylamino)propyl carbamate as a red solid. LC-MS (ES, m/z): 373 (M+H+). Synthesis of 150.2 [001013] To a stirred solution of benzyl 3-(4-carbamoyl-2- nitrophenylamino)propylcarbamate, 150.1 (4 g, 10.75 mmol, 1 equiv) in DMF (50 mL) was added SnCh 2H20 (12.12 g, 53.75 mmol, 5.00 equiv). The resulting mixture was stirred overnight at 35°C. The mixture was cooled to RT and diluted with water. The pH value of the solution was adjusted to 9 with sat.NaHCCf solution and extracted with EtOAc. The combined organic layers were washed with brine, dried over NaiSCE and concentrated under reduced pressure. This resulted in 2.6 g (70.73 %) of benzyl 3-(2-amino-4-carbamoylphenylamino)propylcarbamate as a yellow solid. LC-MS (ES, m/z): 343 (M+H+).
Synthesis of 150.3
[001014] To a stirred solution of benzyl 3-(2-amino-4- carbamoylphenylamino)propylcarbamate, 150.2 (2.6 g, 7.6 mmol, 1 equiv) in MeOH (40 mL) was added BrCN (1.611 g, 15.2 mmol, 2 equiv). The resulting mixture was stirred for 3 h at room temperature. The mixture was concentrated under vacuum. The crude product was purified by Flash with the following conditions: Column, C18; mobile phase, Water (0.01% NH4HCO3) and ACN (26% ACN up to 36% in 10 min); Detector, UV 254/220 nm. This resulted in 1.8 g (64.54%) of benzyl 3-(2-amino-5-carbamoyl-lH-benzo[d]imidazol-l-yl)propylcarbamate as a yellow solid. LC-MS (ES, m/z): 368 (M+H+).
Synthesis of 1-139
[001015] To a stirred mixture of 4-(5-(5-carboxy-3-methyl-lH-pyrazol-l-yl)pentyl)-l-ethyl- 3-methyl-lH-pyrazole-5-carboxylic acid, 150.3 (100 mg, 0.287 mmol, 1 equiv) and benzyl 3-(2- amino-5-carbamoyl-lH-benzo[d]imidazol-l-yl)propylcarbamate (210.91 mg, 0.574 mmol, 2.00 equiv) in NMP (5 mL) was added HATU (218.27 mg, 0.574 mmol, 2.00 equiv) and DIEA (185.48 mg, 1.435 mmol, 5.00 equiv). The final reaction mixture was irradiated with microwave radiation for 1 h at 140°C. The mixture was cooled to RT and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH F-Phenyl OBD Column, 19* 150mm, 5um; mobile phase, water (0.1% FA) and ACN (40% ACN up to 75% in 9 min); UV detection at 254/220 nm. The product-containing fractions were collected and evaporated partially and lyophilized overnight to afford 57.6 mg (19.2%) of benzyl N-[3-(2- [l-[5-(5-[[l-(3-[[(benzyloxy)carbonyl]amino]propyl)-5-carbamoyl-lH-l,3-benzodiazol-2- yl]carbamoyl]-l-ethyl-3-methyl-lH-pyrazol-4-yl)pentyl]-3-methyl-lH-pyrazole-5-amido]-5- carbamoyl-lH-l,3-benzodiazol-l-yl)propyl]carbamate as a white solid. LC-MS (ES, m/z): 1047 (M+H+); 1H NMR (400 MHz, DMSO-fife) d 12.88 (br s, 1H), 12.79 (br s, 1H), 8.03-7.95 (m, 4H), 7.79 (d, 2H), 7.53 (dd, 2H), 7.39-7.20 (m, 14H), 6.69 (s, 1H), 4.98 (s, 2H), 4.96 (s, 2H), 4.62-4.52 (m, 4H), 4.26-4.15 (m, 4H), 3.15-3.05 (m, 4H), 2.75 (t, 2H), 2.11 (s, 3H), 2.07 (s, 3H), 1.96-1.75 (m, 6H), 1.60-1.50 (m, 2H), 1.38-1.23 (m, 5H).
Example 151: Synthesis of benzyl (3-(5-carbamoyl-2-(l-(5-(5-((5-carbamoyl-l-propyl-lH-be nzo[d]imidazol-2-yl)carbamoyl)-l-ethyl-3-methyl-lH-pyrazol-4-yl)pentyl)-3-methyl-lH-pyr azole-5-carboxamido)-lH-benzo[d]imidazol-l-yl)propyl)carbamate, 1-138
Figure imgf000589_0001
Figure imgf000590_0001
Synthesis of 151.1
[001016] To a stirred solution of l-[5-[5-(ethoxycarbonyl)-l-ethyl-3-methyl-lH-pyrazol-4- yl]pentyl]-3-methyl-lH-pyrazole-5-carboxylic acid, 150.3 (500 mg, 1.33 mmol, 1 equiv) in NMP (5 mL) and was added benzyl N-[3-(2-amino-5-carbamoyl-lH-l,3-benzodiazol-l- yl)propyl] carbamate (536 mg, 1.46 mmol, 1.1 equiv), DIEA (514.71 mg, 3.99 mmol, 3 equiv) and HATU (606.48 mg, 1.596 mmol, 1.2 equiv). The final reaction mixture was irradiated with microwave radiation for 1 hour at 140°C. The mixture was cooled to RT and concentrated. The crude produc was purified by reverse flash with the following conditions: Column, Cl 8; mobile phase, Water (10% NH4HCO3) and ACN (30% ACN up to 52% in 10 min); Detector, UV 254/220 nm. This resulted in 600 mg (62.22%) of ethyl 4-[5-(5-[[l-(3- [[(benzyloxy)carbonyl]amino]propyl)-5-carbamoyl-lH-l,3-benzodiazol-2-yl]carbamoyl]-3- methyl-lH-pyrazol-l-yl)pentyl]-l-ethyl-3-methyl-lH-pyrazole-5-carboxylate as a light yellow solid. LC-MS (ES, m/z): 726 (M+H+).
Synthesis of 151.2
[001017] To a stirred solution of ethyl 4-[5-(5-[[l-(3-[[(benzyloxy)carbonyl]amino]propyl)- 5-carbamoyl-lH-l,3-benzodiazol-2-yl]carbamoyl]-3-methyl-lH-pyrazol-l-yl)pentyl]-l-ethyl-3- methyl-lH-pyrazole-5-carboxylate, 151.1 (600 mg, 0.828 mmol, 1 equiv) in MeOH (16 mL) and H2O (4 mL) was added LiOH (59.62 mg, 2.484 mmol, 3 equiv). The resulting solution was stirred for 3h at 60°C. The mixture was cooled to RT and concentrated under vacuum. The residue was diluted with water. The pH value of the solution was adjusted to 3 with IN HC1. The solids were collected by fdtration. This resulted in 500 mg (86.66%) of 4-[5-(5-[[l-(3- [[(benzyloxy)carbonyl]amino]propyl)-5-carbamoyl-lH-l,3-benzodiazol-2-yl]carbamoyl]-3-
589
5UB5TITUTE SHEET (RULE 26) methyl-lH-pyrazol-l-yl)pentyl]-l-ethyl-3-methyl-lH-pyrazole-5-carboxylic acid as a light yellow solid. LC-MS (ES, m/z): 698 (M+H+).
Synthesis of 1-138
[001018] To a stirred solution of 4-[5-(5-[[l-(3-[[(benzyloxy)carbonyl]amino]propyl)-5- carbamoyl-lH-l,3-benzodiazol-2-yl]carbamoyl]-3-methyl-lH-pyrazol-l-yl)pentyl]-l-ethyl-3- methyl- lH-pyrazole-5-carboxylic acid, 151.2 (100 mg, 0.143 mmol, 1 equiv) and 2-amino-l- propyl-lH-l,3-benzodiazole-5-carboxamide (35 mg, 0.158 mmol, 1.1 equiv) in DMF (5 mL) was added DIEA (91 mg, 0.715 mmol, 5 equiv) and HATU (64 mg, 0.172 mmol, 1.2 equiv). The final reaction mixture was irradiated with microwave radiation for lhour at 140°C. The mixture was cooled to RT and concentrated. The crude product was purified by Prep-HPLC with the following conditions: Column, Xselect CSH OBD Column 30* 150mm 5um; mobile phase, Water (0.1%FA) and ACN (33% ACN up to 43% in 10 min); UV detection at 254/220 nm. This resulted in 46.0 mg (35.82%) of benzyl N-(3-[5-carbamoyl-2-[l-(5-[5-[(5-carbamoyl-l-propyl-lH-l,3-benzodiazol- 2-yl)carbamoyl]-l-ethyl-3-methyl-lH-pyrazol-4-yl]pentyl)-3-methyl-lH-pyrazole-5-amido]-lH- l,3-benzodiazol-l-yl]propyl)carbamate as a white solid. LC-MS (ES, m/z): 899 (M+H+); 'H MR: 1H NMR (400 MHz, DMSO-t¾) d 12.84 (br s, 2H), 8.01-7.93 (m, 4H), 7.81-7.77 (m, 2H), 7.53 (dd, 2H), 7.39-7.28 (m, 8H), 6.70 (s, 1H), 4.98 (s, 2H), 4.68-4.50 (m, 4H), 4.22 (t, 2H), 4.11 (t, 2H), 3.11-3.08 (m, 2H), 2.76 (t, 2H), 2.12 (s, 3H),2.08 (s, 3H), 1.98-1.88 (m, 2H), 1.85-1.70 (m, 4H), 1.60-1.51 (m, 2H), 1.40-1.28 (m, 5H), 0.86 (t, 3H).
Example 152: Synthesis of (E)-N,N'-(but-2-ene-l,4-diylbis(5-carbamoyl-7-hydroxy-lH- benzo[d]imidazole-l,2-diyl))bis(4-ethyl-2-methyloxazole-5-carboxamide), 1-11
Figure imgf000592_0001
[001019] Into a 8-mL sealed tube, was placed l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl- l,3-oxazole-5-amido)-7-methoxy-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-methoxy-l,3-benzodiazole-5-carboxamide, 1-60 (100.00 mg, 0.135 mmol, 1.00 equiv), DCE (2.00 mL), boron tribromide (339.11 mg, 1.354 mmol, 10.00 equiv). The resulting solution was stirred for 12h at 85 degrees C. The reaction was then quenched by the addition of 20 mL of water. The solids were collected by filtration. The crude product (40 mg) was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): Column, XBridge Prep C18 OBD Column, 19x150mm 5um; mobile phase, water (10 mmol/L NH4HCO3) and EtOH- (45% Phase B up to 75% in 7 min); Detector, UV, RT = 6.28 min to yield 2.6 mg (5.41%) of a white solid. LC-MS:(ES, m/z): [M+H]+ = 711.2; H-NMR:(400 MHz, DMSO-de, ppm) 512.48-12.65 (brs, 2H), 512.31-12.43 (brs, 2H), 57.72-7.89 (d, 2H), 57.40-7.41 (d, 2H), 57.08-7.19 (m, 4H), 55.92 (s, 2H), 54.91-4.97 (d, 4H), 52.74-2.87 (m, 4H), 52.31-2.34 (m, 6H), 50.92-1.03 (m, 6H).
Example 153: Synthesis of (E)-N-(5-carbamoyl-l-(4-(2-(4-ethyl-2-methyloxazole-5- carboxamido)-7-(morpholinomethyl)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-lH- benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-10
Figure imgf000593_0001
[001020] Into a 8-mL sealed tube purged and maintained with an inert atmosphere of nitrogen, was placed 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (300 mg, 2.008 mmol, 4.00 equiv), NMP (5.00 mL), DIEA (389.23 mg, 3.012 mmol, 6.00 equiv), HATU (763.41 mg, 2.008 mmol, 4.00 equiv), 2-amino-l-[(2E)-4-[2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(morpholin- 4-ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3-benzodiazole-5-carboxamide, 112.9 (300.00 mg, 0.502 mmol, 1.00 equiv). The resulting mixture was stirred for 1.5 h at 140 degrees C under nitrogen atmosphere. The resulting mixture was washed with 1x20 mL of water. The crude product (300 mg) was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, HCOOH in water, 10% to 50% gradient in 35 min; detector, UV 254 nm. The crude product (60 mg) was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): Column, XBridge Prep OBD C18 Column, 19*250mm, 5um; mobile phase, water(10 mmol/L NH4HCO3) and ACN (hold 30% Phase B in 8 min); Detector, UV 254 nm.) to afford 2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-l-[(2E)-4-[2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-(morpholin-4-ylmethyl)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-l,3- benzodiazole-5-carboxamide (10.9 mg, 4.43%) as a white solid. LC-MS: (ES, m/z): [M+H]+:
735.5, [M-H] : 733.5; H-NMR: (300 MHz, DMSO-d6, ppm) 612.63-12.45 (brs, 2H), 67.97-7.94 (d, 2H), 67.73-7.70 (d, 1H), 67.51-7.41 (m, 2H), 67.31 (s, 1H), 67.14-7.08 (m, 1H), 67.01-6.99(d, 1H), 66.00-5.95(d, 1H), 65.42-5.37(d, 1H), 65.17(s, 2H), 64.77(s, 2H), 63.44(s, 6H), 62.84-2.76 (m, 4H), 62.44 (s, 6H), 62.31-2.11 (m, 4H), 61.10-1.00 (m, 6H). Example 154: Synthesis of (E)-N-(5-carbamoyl-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-7-(morpholinomethyl)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-methyl- lH-benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-6
Figure imgf000594_0001
Figure imgf000595_0001
Synthesis of 154.1
[001021] To a stirred solution of methyl 4-fluoro-3 -methyl -5 -nitrobenzoate (228.97 mg, 1.074 mmol, 1 equiv) in DMSO (10.00 mL) in a 20 mL vial, DIEA (1388.29 mg, 10.742 mmol, 10 equiv) and (E)-N-(l-(4-aminobut-2-en-l-yl)-7-(morpholinomethyl)-lH-benzo[d]imidazol-2- yl)-l -ethyl-3 -methyl- lH-pyrazole-5-carboxamide (470.00 mg, 1.074 mmol, 1.00 equiv) were added in portions at room temperature. The resulting mixture was stirred for 1 h at room temperature under nitrogen atmosphere. The resulting mixture was extracted with EtOAc (2x100 mL). The combined organic layers were washed with water (3x20 mL), dried over anhydrous Na2S04. After filtration, the filtrate was concentrated under reduced pressure. This resulted in methyl 4-[[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-7-(morpholin-4-ylmethyl)-l,3- benzodiazol-l-yl]but-2-en-l-yl]amino]-3-methyl-5-nitrobenzoate (400mg, 54.91%) (ES, m/z): [M+H]+ 631.3 as an off-white solid.
Synthesis of 154.2
[001022] To a stirred solution of 154.1 (390.00 mg, 0.618 mmol, 1.00 equiv) in THF (6.00 mL) and MeOH (2.00 mL) in an 25mL round-bottom flask, NaOH (74.20 mg, 1.855 mmol, 3 equiv) and H2O (2.00 mL) were added dropwise at 0°C. The resulting mixture was stirred for lh at room temperature under nitrogen atmosphere. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeOH in water, 10% to 50% gradient in 10 min; detector, UV 254 nm to afford 188mg (49.30%) (ES, m/z): [M+H]+ 617.3 as an off-white solid. Synthesis of 154.3
[001023] To a stirred solution of 154.2 (188.00 mg, 0.305 mmol, 1.00 equiv) in DMF (2.00 mL) in an 8-mL vial, HATU (231.83 mg, 0.610 mmol, 2 equiv) and DIEA (118.20 mg, 0.915 mmol, 3 equiv) and NH4CI (81.54 mg, 1.524 mmol, 5.00 equiv) were added in portions at room temperature. The resulting mixture was stirred for lh at room temperature under nitrogen atmosphere. The residue was purified by Prep-TLC (CH2CI2 / MeOH = 20: 1) to afford 154.3 (130mg, 65.10%) (ES, m/z): [M+H]+ 616.3 as an off-white solid.
Synthesis of 154.4
[001024] To a stirred solution of N-[l-[(2E)-4-[(4-carbamoyl-2-methyl-6- nitrophenyl)amino]but-2-en- 1 -yl] -
7-(morpholin-4-ylmethyl)-l,3-benzodiazol-2-yl]-2-ethyl-5-methylpyrazole-3-carboxamide, 154.3 (130.00 mg, 0.211 mmol, 1.00 equiv) in HOAc(2.00 mL) in an 8-mL vial, Zn(276.21 mg, 4.223 mmol, 20 equiv) was added at 0°C. The resulting mixture was stirred for lh at room temperature under nitrogen atmosphere. The resulting mixture was filtered, and the filter cake was washed with MeOH (3x3 mL). The filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (CH2C12 / MeOH = 10: 1) to afford 154.4 (58mg, 37.38%).
Synthesis of 154.5
[001025] To a stirred solution of N-[l-[(2E)-4-[(2-amino-4-carbamoyl-6- methylphenyl)amino]but-2-en-l-yl]-
7-(morpholin-4-ylmethyl)-l,3-benzodiazol-2-yl]-2-ethyl-5-methylpyrazole-3-carboxamide, 154.4 (58.00 mg, 0.099 mmol, 1.00 equiv) in MeOH(2.00 mL) in an 8-mL vial, BrCN (10.49 mg, 0.099 mmol, 1 equiv) were added at room temperature. The resulting mixture was stirred for lh at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum to afford 154.5 (49 mg, 63.20%) (ES, m/z): [M+H]+ 611.3 as an off-white solid.
Synthesis of 1-6
[001026] To a stirred solution of 4-ethyl-2-methyl-l,3-oxazole-5-carboxylic acid (14.94 mg, 0.096 mmol, 1.20 equiv) in MP (0.5 mL) in an 8-mL vial, HATU (61.01 mg, 0.160 mmol, 2 equiv) and DIEA (31.11 mg, 0.241 mmol, 3 equiv) and 154.5 (49.00 mg, 0.080 mmol, 1.00 equiv) were added in portions at room temperature. The resulting mixture was stirred for 2h at 85°C in an oil bath under nitrogen atmosphere. To the above mixture was added Na2C03 (sat. aq.) (0.5 mL). The resulting mixture was stirred for additional 3h at 60 degrees C under nitrogen atmosphere. The resulting mixture was extracted with EtOAc (2 x 50mL). The combined organic layers were washed with brine (3x10 mL), dried over anhydrous NaiSCE. After fdtration, the fdtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: SunFire C18 OBD Prep Column, lOOA, 5 pm, 19 mm x 250 mm; Mobile Phase A: water (0.1%FA), Mobile Phase B: ACN; Flow rate: 20 mL/min; Gradient: 12 B to 65 B in 8 min; 254/220 nm; RTF7.54) to afford 1-6 (4.6 mg, 7.49%) as a white solid; LC-MS: (ES, m/z): [M+H]+748.5 [M-H] 746.5; H-NMR: (400 MHz, DMSO-d6) 512.77 (s, 2H), 57.86- 7.88 (m, 2H), 57.50-7.53 (m, 2H), 57.29 (s, 1H), 57.13-7.17 (m, 1H), 57.02-7.04 (d, 1H), 56.49 (s, 1H), 55.55-5.59 (d, 1H), 55.44-5.48 (d, 1H), 55.14 (s, 2H), 54.92 (s, 2H), 54.50-4.55 (m, 2H), 53.43-3 46(d, 6H), 52.78-2.83 (m, 2H), 52.55(s, 3H), 52.41-2.46 (d, 3H), 52.19-2.30 (d, 4H), 52.13 (s, 3H), 51.25-1.29 (t, 3H), 50.98-1.08 (t, 3H).
Example 155: Synthesis of (E)-N-(5-carbamoyl-l-(4-(2-(l-ethyl-3-methyl-lH-pyrazole-5- carboxamido)-lH-benzo[d]imidazol-l-yl)but-2-en-l-yl)-7-(2-morpholinoethoxy)-lH- benzo[d]imidazol-2-yl)-4-ethyl-2-methyloxazole-5-carboxamide, 1-186
Figure imgf000597_0001
Figure imgf000598_0001
Synthesis of 155.1
[001027] Into a 50-mL 3-necked round-bottom flask, was placed 2-(4-ethyl-2-m ethyl- 1,3- oxazole-5-amido)-l-[(2E)-4-[2-(2-ethyl-5-methylpyrazole-3-amido)-l,3-benzodiazol-l-yl]but-2- en-l-yl]-7-methoxy-l,3-benzodiazole-5-carboxamide, 1-8 (440.00 mg, 0.662 mmol, 1.00 equiv), DCE (10.00 ml), BBn (497.48 mg, 1.986 mmol, 3.00 equiv). The resulting solution was stirred for lh at room temperature under nitrogen atmosphere. The resulting mixture was concentrated. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN/H20=15% increasing to ACN/H2O=60% within 15 min; Detector UV 254 nm to afford 140 mg (32.50%) of an off-white solid. LC-MS (ES, m/z): 651 [M+H]+.
Synthesis of 1-186
[001028] Into a 50-mL 3-necked round-bottom flask, was placed 155.1 (100.00 mg, 0.154 mmol, 1.00 equiv), DMF (5.00 mL), 4-(2-bromoethyl)morpholine (29.83 mg, 0.154 mmol, 1.00 equiv). The resulting solution was stirred for 1 overnight at 30 degrees C under nitrogen atmosphere. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN/H20=15% increasing to ACN/H2O=60% within 15 min. The crude product (50 mg) was purified by Prep-HPLC with the following conditions: Column: Sunfire prep Cl 8 column, 30* 150, 5 um; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 13% B to 30% B in 8 min; 254;220 nm; RT1 :5.97 min. This resulted in 1.1 mg (0.94%) of the desired product as an off-white solid. LC-MS: (ES, m/z): [M+H]+ 764.5, [M-H]' 762.4; 'H-NMR: (300 MHz, DMSO -d6, ppm) 512.69-12.79 (m, 2H), 7.92 (s, 1H), 7.64 (s, 1H), 7.48-7.50 (d, 1H), 7.33-7.35 (d, 3H), 7.08-7.21 (m, 2H), 6.53 (s, 1H), 5.77-5.99 (m, 2H), 4.94-4.96 (d, 2H), 4.80-4.87 (t, 2H), 4.51-4.56 (t, 2H), 4.07-4.10 (t, 2H), 3.48 (s, 4H), 2.84-2.90 (m, 2H), 2.41 (s, 4H), 2.30 (s, 5H), 2.12 (s, 3H), 1.24- 1.29 (t, 3H), 1.05-1.10 (t, 3H).
Example 156: Synthesis of (E)-N,N'-(but-2-ene-l,4-diylbis(5-carbamoyl-7-(2- hydroxyethoxy)-lH-benzo[d]imidazole-l,2-diyl))bis(4-ethyl-2-methyloxazole-5- carboxamide), 1-2
Figure imgf000599_0001
[001029] Into a 20-mL sealed tube was placed l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl- l,3-oxazole-5-amido)-7-hydroxy-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3- oxazole-5-amido)-7-hydroxy-l,3-benzodiazole-5-carboxamide, 1-11 (100.00 mg, 0.141 mmol, 1.00 equiv), DMF (5.00 mL), 2-bromoethanol (105.50 mg, 0.844 mmol, 6.00 equiv), CS2CO3 (183.38 mg, 0.563 mmol, 4.00 equiv), Nal (42.18 mg, 0.281 mmol, 2.00 equiv). The resulting solution was stirred for 12h at 80 degrees C under nitrogen atmosphere. The solids were fdtered out. The crude product was purified by Flash-Prep-HPLC with the following conditions (IntelFlash-1): Column, C18 silica gel; mobile phase, ACN:H20=15% increasing to ACN:H20=60°/O within 15 min; the crude product was purified by Prep-HPLC with the following conditions (2#SHIMADZU (HPLC-01)): Column: SunFire C18 OBD Prep Column, IOOA, 5 pm, 19 mm X 250 mm; Mobile Phase A: water(0.1%FA), Mobile Phase B: ACN; Flow rate: 20 mL/min; Gradient:23 B to 30 B in 10.6 min; 254/220 nm; RT1 : 10.38 min. This resulted in 3.0 mg (2.52%) of l-[(2E)-4-[5-carbamoyl-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)-7-(2- hydroxyethoxy)-l,3-benzodiazol-l-yl]but-2-en-l-yl]-2-(4-ethyl-2-methyl-l,3-oxazole-5-amido)- 7-(2-hydroxyethoxy)-l,3-benzodiazole-5-carboxamide formic acid as a white solid. LC-MS: (ES, m/z): [M+H]+ = 799.4; [M-H]- = 797.4. ¾-NMR: (300 MHz, DMSO, ppm) 512.70-12.88 (brs, 2H), 58.41 (s, 1H), 57.96-8.19 (m,2H), 57.64 (s, 2H), 57.10-7.34 (m, 4H), 55.71-5.85 (d, 2H), 54.94-5.08 (m, 6H), 53.90-3.98 (t, 4H), 53.54 (s, 4H), 52.65-2.84 (m, 4H), 52.40 (s, 6H), 50.90- 1.04 (t , 6H).
Example 157: Synthesis of (E)-N,N'-(but-2-ene-l,4-diylbis(5-carbamoyl-7-methoxy-lH- benzo[d]imidazole-l,2-diyl))bis(4,5,6,7-tetrahydrobenzo[c]isoxazole-3-carboxamide), 1-1
Figure imgf000600_0001
[001030] Into a 10-mL sealed tube, was placed 4,5,6,7-tetrahydro-2,l-benzoxazole-3- carboxylic acid (194.34 mg, 1.163 mmol, 6.00 equiv), NMP (5.00 mL), DIEA (500.85 mg, 3.875 mmol, 20.00 equiv), HATU (442.05 mg, 1.163 mmol, 6.00 equiv), 2-amino-l-[(2E)-4-(2-amino- 5-carbamoyl-7-methoxy-l,3-benzodiazol-l-yl)but-2-en-l-yl]-7-methoxy-l,3-benzodiazole-5- carboxamide, 60.5 (90.00 mg, 0.194 mmol, 1.00 equiv). The resulting solution was stirred for lh at 140°C. The reaction progress was monitored by M.W. The resulting mixture was washed with 20 mL of H2O. The solids were collected by filtration. This resulted in 22.0 mg (14.89%) ofN-[5- carbamoyl-l-[(2E)-4-[5-carbamoyl-7-methoxy-2-(4,5,6,7-tetrahydro-2,l-benzoxazole-3-amido)- l,3-benzodiazol-l-yl]but-2-en-l-yl]-7-methoxy-l,3-benzodiazol-2-yl]-4,5,6,7-tetrahydro-2,l-
599
5UB5TITUTE SHEET (RULE 26) benzoxazole-3 -carboxamide as a off-white solid. LC-MS: (ES, m/z): [M+H]+ 763.4; 1H-NMR: (300 MHz, CF COOD,/¾p/w) 58.57 (s, 2H), 58.17 (s, 2H), 57.02 (s, 2H), 56.18 (s, 4H), 54.40-4.62 (m, 6H), 53.47-3.71 (m, 9H), 52.46 (s, 8H).
EXAMPLE 158. STING Surface Plasmon Resonance (SPR) Protocol (human).
[001031] Surface plasmon resonance experiments were performed on Biacore™ T200 (GE Healthcare). Briefly, recombinant STING (H232R, aal49-341) was chemically biotinylated using Sulfo-NHS-LC-LC-Biotin kits (Thermofisher) and captured on the streptavidin surface in the buffer of 20mM Tris.HCl pH 7.5, 150 mM NaCl at 10°C to give final capture levels of7000-10000 RU. Kinetics study of the compound binding was performed in the buffer of 20mM Tris.HCl pH 7.5, 150 mM NaCl, 0.005% tween-20, 5% DMSO, at 10°C. For multi-cycle kinetics study, compounds were series diluted in the buffer and injected over the STING surface for 60s followed by a dissociation time of 60-200s. Data were corrected for DMSO excluded volume effects. All data were double-referenced for blank injections and reference surface using standard processing procedures. Data were fitted using a 1 : 1 binding model to determine KD, kd and ka as appropriate.
[001032] Results for exemplary compounds tested in the SPR assay are provided in Table 4, below. Compounds having a Kd of less than or equal to 10 nM are designated“A.” Compounds having a Kd of greater than 10 nM and less than or equal to 500 nM are designated“B.” Compounds having a Kd of greater than 500 nM and less than or equal to 100 mM are designated“C” Compounds having a Kd of greater than 100 mM are designated“D.”
Table 4. hSTING SPR Assay Results.
Figure imgf000601_0001
Figure imgf000602_0001
Figure imgf000603_0001
EXAMPLE 159. 293T-luc2p-h232R agonist assay
[001033] HEK293T cells (ATCC CRL-3216) were stably co-transfected with plasmids expressing full length human STING (NM_198282 with the Histidine at position 232 mutated to an Arginine) (Genewiz) and the enzyme firefly luciferase driven by the interferon stimulated response element promoter (pISRE-luc2p) (Promega). Transfections were prepared using a cell suspension with Fugene ® 6 following the manufacturer’s instructions (3 : 1 Fugene® 6 :DNA). Stable clones were selected in media containing puromycin and hygromycin. 25 microliters of cell suspension containing 20,000 cells was dispensed into a 384 well plate containing 125 nanoliters of a compound. Control wells expected to generate maximal activation of STING were treated with 2’,3’-cyclic-GAMP. Plates were sealed and incubated for 16 hours at 37° C. The expression of firefly luciferase was processed using Steady-Glo luciferase assay system (Promega) following the manufacturer’s instructions and was analyzed using a Perkin Elmer EnVision 2104 luminescence plate reader. EC50 was generated by non-linear regression plot of log compound concentration versus luminescence value. Percent activity was determined using the following equation: (Tope - Low)*100/(TopR - Low) where Tope is the top luminescence value of compound; Low is luminescence value of low control and TopR is the top luminescence value of the reference compound.
[001034] Results of exemplary compounds tested in the 293T-luc2p-h232R agonist assay are provided in Table 5, below. Compounds having an EC50 of less than or equal to 40 nM are designated“A.” Compounds having an EC50 of greater than 40 nM and less than or equal to 500 nM are designated“B.” Compounds having an EC50 of greater than 500 nM and less than or equal to 30 mM are designated“C” Compounds having an EC50 of greater than 30 mM are designated “D”
Table 5. 293T-luc2p-h232R Agonist Assay Results
Figure imgf000604_0001
Figure imgf000605_0001
Figure imgf000606_0001
Figure imgf000607_0001
Figure imgf000608_0001
Figure imgf000609_0001
Figure imgf000610_0001
Figure imgf000611_0001
EXAMPLE 160. 293T-luc2p-mouse STING agonist assay.
[001035] HEK293T cells (ATCC CRL-3216) were stably co-transfected with plasmids expressing full length mouse STING (NM_028261) and the enzyme firefly luciferase driven by the interferon stimulated response element promoter (pISRE-luc2p) (Promega). Transfections were prepared using a cell suspension with Fugene ® 6 following the manufacturer’s instructions (3 : 1 Fugene® 6 :DNA). Stable clones were selected in media containing puromycin and hygromycin. 25 microliters of cell suspension containing 20,000 cells was dispensed into a 384 well plate containing 125 nanoliters of a compound. Control wells expected to generate maximal activation of STING were treated with 2’,3’-cyclic-GAMP. Plates were sealed and incubated for 16 hours at 37° C. The expression of firefly luciferase was processed using Steady-Glo luciferase assay system (Promega) following the manufacturer’s instructions and was analyzed using a Perkin Elmer EnVision 2104 luminescence plate reader. EC50 was generated by non-linear regression plot of log compound concentration versus luminescence value. Percent activity was determined using the following equation: (Tope - Low)* 100/(TopR - Low) where Tope is the top luminescence value of compound; Low is luminescence value of low control and TopR is the top luminescence value of the reference compound.
[001036] Results of exemplary compounds tested in the 293T-luc2p-mouse STING agonist assay are provided in Table 6, below. Compounds having an EC 50 of less than or equal to 10 nM are designated“A.” Compounds having an EC50 of greater than 10 nM and less than or equal to 500 nM are designated“B.” Compounds having an EC50 of greater than 500 nM and less than or equal to 30 mM are designated“C.” Compounds having an EC50 of greater than 30 mM are designated“D.” Table 6. 293T-luc2p-Mouse STING Agonist Assay Results.
Figure imgf000612_0001
Figure imgf000613_0001
Figure imgf000614_0001
Figure imgf000615_0001
Figure imgf000616_0001
Figure imgf000617_0001
Figure imgf000618_0001
EXAMPLE 161. Human STING SPA Binding Assay.
[001037] A radioligand binding assay was used to measure interactions of compound binding to STING protein by competition with [3H]2’,3’-cGAMP. Test compound was pre-incubated with 0.2 pg/well His-tagged human STING (NM_198282) protein spanning residues 149-341 for 10 minutes at room temperature in a Perkin-Elmer Isoplate. 150 nM of the ['H]2’.3’-cGAMP probe along with 100 pg/well PVT Cu-His-tag SPA beads were then added to the plate and the reaction mixture was incubated at room temperature for another 45 minutes. Following incubation, the plates was placed in counting cassettes and counted on a Microbeta microplate counter for radiometric detection. Compound binding IC50 was calculated using non-linear regression of the dose-response curve. Compounds having an IC50 of less than or equal to 1 pM (<1 pM) are designated“A”. Compounds having an IC50 between 1 pM and less than or equal to 10 pM (1 pM < x < 10 pM) are designated“B”. Compounds having an IC50 between 10 pM and less than or equal to 100 pM (1 pM < x < 100 pM) are designated“C”. Compounds having an IC50 of greater than 100 pM (> 100 pM) are designated“D”.
Table 7. Human STING SPA Binding ICso Results.
Figure imgf000618_0002
Figure imgf000619_0001
Figure imgf000620_0001
Figure imgf000621_0001
Figure imgf000622_0001
Figure imgf000623_0001
Figure imgf000624_0001
Figure imgf000625_0001
EXAMPLE 162. Mouse STING SPA Binding Assay.
[001038] A radioligand binding assay was used to measure interactions of compound binding to mouse STING protein by competition with [3H]2’,3’-cGAMP. Test compound was pre incubated with 0.04 pg/well His-tagged mouse STING (NM_028261) protein spanning residues 149-341 for 10 minutes at room temperature in a Perkin-Elmer Isoplate. 20 nM of the [3H]2’,3’- cGAMP probe along with 20 pg/well PVT Cu-His-tag SPA beads were then added to the plate and the reaction mixture was incubated at room temperature for another 45 minutes. Following incubation, the plates were placed in counting cassettes and counted on a Microbeta microplate counter for radiometric detection. Compound binding IC50 was calculated using non-linear regression of the dose-response curve. Compounds having an IC50 of less than or equal to 1 pM (<1 pM) are designated“A”. Compounds having an IC50 between 1 pM and less than or equal to 10 pM (1 pM < x < 10 pM) are designated“B”. Compounds having an IC50 between 10 pM and less than or equal to 100 pM (1 pM < x < 100 pM) are designated“C”. Compounds having an IC50 of greater than 100 pM (> 100 pM) are designated“D”.
Table 8. Mouse STING SPA Binding ICso Results.
Figure imgf000625_0002
Figure imgf000626_0001
Figure imgf000627_0001
Figure imgf000628_0001
Figure imgf000629_0001
[001039] While we have described a number of embodiments of this invention, it is apparent that our basic examples may be altered to provide other embodiments that utilize the compounds and methods of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims rather than by the specific embodiments that have been represented by way of example.

Claims

CLAIMS We claim:
1. A compound of formula I:
Figure imgf000630_0001
I
or a pharmaceutically acceptable salt thereof, wherein:
m is 0 or 1;
n is 0 or 1;
p is 0 or 1;
wherein m + n + p = 1, 2, or 3;
Ring A is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
wherein Ring A and its substituents is other than
Figure imgf000631_0001
Ring B is phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, phosphorous, silicon and sulfur, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-14 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
L1 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy1-, -C(R’)2- -CH(R’)-, -N(R)-, Cy'fL’-Z) , -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2- -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0)2-;
each -Cy1- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R’ is independently hydrogen or C M aliphatic; or
two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring; or
two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R is independently hydrogen, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or:
two R groups on the same nitrogen are optionally taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each L’ is a covalent bond or a CMS bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and
independently replaced
Figure imgf000632_0001
, -N(R)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)C(0)0-, -0C(0)N(R)-, -N(R)S(0)2- - S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0)2- each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, napththylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroaryl ene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each Z is hydrogen or a CRBN binding moiety;
L2 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy2-, -C(R’)2- -CH(R’)-, -N(R)-, -Cy2(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(0)-, -0C(0)-, -C(0)0- -S-, -S(0)-, or -S(0)2-;
each -Cy2- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
L3 is a covalent bond or a C O bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy3-, -C(R’)2- -CH(R’)-, -N(R)-, -Cy3(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-, -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(0)-, -0C(0)-, -C(0)0-, -S-, -S(0)-, or -S(0)2-;
each -Cy3- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R1 is independently hydrogen, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C( R)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
R2 is hydrogen or an optionally substituted Ci-6 aliphatic group;
each R3 is independently hydrogen, oxo, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, - N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each R4 is independently hydrogen, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
R5 is hydrogen or an optionally substituted Ci-6 aliphatic group;
each R6 is independently hydrogen, oxo, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each of q, s, t, and v is independently 0, 1, 2, 3, or 4;
each of r and u is independently 0 or 1; and
each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
2. The compound of claim 1, wherein the compound is of formula I-a:
Figure imgf000635_0001
I-a
or a pharmaceutically acceptable salt thereof.
3. The compound of claim 1, wherein the compound is of formula I-b:
Figure imgf000635_0002
or a pharmaceutically acceptable salt thereof.
4. The compound of claim 1, wherein the compound is of formula I-c:
Figure imgf000636_0001
or a pharmaceutically acceptable salt thereof.
5. The compound of claim 1, wherein the compound is of formula I-d:
Figure imgf000637_0001
I-d or a pharmaceutically acceptable salt thereof.
6. A compound of formula II:
Figure imgf000637_0002
or a pharmaceutically acceptable salt thereof, wherein: m is 0 or 1;
n is 0 or 1;
p is 0 or 1;
wherein m + n + p = 1, 2, or 3;
L1 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced
Figure imgf000638_0001
-N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0-, -S-, -S(O)-, or -S(0)2-;
each -Cy1- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R’ is independently hydrogen or C M aliphatic; or
two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 3-7 membered saturated or partially unsaturated carbocyclylene ring; or
two instances of R’ on the same carbon are optionally taken together with the carbon to form an optionally substituted bivalent 4-7 membered saturated or partially unsaturated heterocyclylene ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R is independently hydrogen, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, naphthalenyl, an 8-10 membered bicyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 7-12 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or: two R groups on the same nitrogen are taken together with the nitrogen to form an optionally substituted 4-7 membered monocyclic saturated, partially unsaturated, or heteroaryl ring having, in addition to the nitrogen, 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each L’ is a covalent bond or a C S bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one to four methylene units of the chain are optionally and
independently replaced
Figure imgf000639_0001
, -N(R)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)C(0)0-, -0C(0)N(R)-, -N(R)S(0)2-, - S(0)2N(R)-, -0-, -C(0)-, -0C(0)-, -C(0)0- -S-, -S(0)-, or -S(0)2-;
each -Cy’- is independently an optionally substituted bivalent ring selected from phenylene, napththylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each Z is hydrogen or a CRBN binding moiety;
L2 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy2-, -C(R’)2-, -CH(R’)-, -N(R)-, -Cy2(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-
-N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2- -S(0)2N(R)-, -0-, -C(O)-, -OC(O)- -C(0)0- -S-, -S(0)-, or -S(0)2-;
each -Cy2- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
L3 is a covalent bond or a CMO bivalent saturated or unsaturated, straight or branched hydrocarbon chain, wherein one or two methylene units of the chain are optionally and independently replaced by -Cy3-, -C(R’)2- -CH(R’)-, -N(R)-, -Cy3(L’-Z)-, -C(R’)(L’-Z)-, -CH(L’-Z)-,
638
5U B5TITUTE SH EET (RULE 26) -N(L’-Z)-, -N(R)C(0)-, -C(0)N(R)-, -N(R)S(0)2-, -S(0)2N(R)-, -0-, -C(O)-, -OC(O)-, -C(0)0- -S-, -S(O)-, or -S(0)2-;
each -Cy3- is independently an optionally substituted bivalent ring selected from phenylene, 3-7 membered saturated or partially unsaturated carbocyclylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R1 is independently hydrogen, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, - N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
R2 is hydrogen or an optionally substituted Ci-6 aliphatic group;
each R3 is independently hydrogen, oxo, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each R4 is independently hydrogen, halogen, -CN, -N02, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, -
N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
R5 is hydrogen or an optionally substituted Ci-6 aliphatic group;
each R6 is independently hydrogen, oxo, halogen, -CN, -NO2, -OR,
SR, -NR2, -S(0)2R, -S(0)(NR)R, -S(0)2NR2, -S(0)R, -S(0)NR2, -C(0)R, -C(0)0R, - C(0)NR2, -C(0)N(R)0R, -0C(0)R, -0C(0)NR2, -N(R)C(0)0R, -N(R)C(0)NR2, - N(R)C(NR)NR2, -N(R)S(0)2NR2, -N(R)S(0)2R, -P(0)R2, -L’-Z, or an optionally substituted group selected from Ci-6 aliphatic, phenyl, a 3-7 membered saturated or partially unsaturated monocyclic carbocyclic ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, and a 3-7 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur;
each of q, s, t, and v is independently 0, 1, 2, 3, or 4;
each of r and u is independently 0 or 1;
each of x is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; and wherein
(a) when R1 or R4 is -C(0) R¾ each R of the -C(0)NR2 moiety is other than hydrogen or CM aliphatic; or
(b) in at least one instance of L1, L2, or L3, a methylene unit is replaced with -C(O)-; or
(c) in at least one instance of L1, L2, or L3, a methylene unit adjacent to the benzimidazole or pyrazole ring is replaced with -Cy1-, -Cy2-, or -Cy3-; or
(d) the compound is other than
Figure imgf000642_0001
7. The compound of claim 6, wherein the compound is of formula Il-a:
Figure imgf000642_0002
or a pharmaceutically acceptable salt thereof.
8. The compound of claim 6, wherein the compound is of formula Il-b:
Figure imgf000643_0001
or a pharmaceutically acceptable salt thereof.
9. The compound of claim 6, wherein the compound is of formula II-c:
Figure imgf000643_0002
or a pharmaceutically acceptable salt thereof.
10. The compound of claim 6, wherein the compound is of formula Il-d:
Figure imgf000644_0001
or a pharmaceutically acceptable salt thereof.
11. The compound of any one of the preceding claims, wherein said compound is selected from any one of the compounds depicted in Table 1 or Table 2, or a pharmaceutically acceptable salt thereof.
12. A pharmaceutical composition comprising a compound of any one of the preceding claims, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
13. A pharmaceutical composition comprising an effective amount of a compound of any one of the preceding claims, or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof, and a pharmaceutically acceptable carrier.
14. A method of inducing a STING-dependent type I interferon production in a subj ect, comprising administering a therapeutically effective amount of a compound of any one of the preceding claims, or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof, to the subject.
15. A method of inducing a STING-dependent cytokine production in a subject, comprising administering a therapeutically effective amount of a compound of any one of the preceding claims, or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof, to the subject.
16. A method of treating a cell proliferation disorder, comprising administering a therapeutically effective amount of a compound of any one of the preceding claims, or a pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof, to the subject.
17. The method according to claim 16, wherein the cell proliferation disorder is cancer, benign papillomatosis, gestational trophoblastic diseases, or a benign neoplastic disease.
18. The method of claim 16, wherein the cell proliferation disorder is cancer.
19. The method of claim 17, wherein the benign neoplastic disease is skin papilloma or genital papilloma.
20. The method of claim 18, wherein the cancer is brain cancer, leukemia, skin cancer, prostate cancer, thyroid cancer, colon cancer, lung cancer, or sarcoma.
21. The method of claim 18, wherein the cancer is selected from the group consisting of glioma, glioblastoma multiforme, paraganglioma, suprantentorial primordial neuroectodermal tumors, acute myeloid leukemia, myelodysplastic syndrome, chronic myelogenous leukemia, melanoma, breast, prostate, thyroid, colon, lung, central chondrosarcoma, central and periosteal chondroma tumors, fibrosarcoma, and cholangiocarcinoma.
PCT/US2019/068076 2018-12-21 2019-12-20 Sting agonists and uses thereof WO2020132582A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201862784136P 2018-12-21 2018-12-21
US62/784,136 2018-12-21
US201962823275P 2019-03-25 2019-03-25
US62/823,275 2019-03-25
US201962889941P 2019-08-21 2019-08-21
US62/889,941 2019-08-21

Publications (1)

Publication Number Publication Date
WO2020132582A1 true WO2020132582A1 (en) 2020-06-25

Family

ID=71100373

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/068076 WO2020132582A1 (en) 2018-12-21 2019-12-20 Sting agonists and uses thereof

Country Status (1)

Country Link
WO (1) WO2020132582A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021014365A1 (en) * 2019-07-22 2021-01-28 Lupin Limited Macrocyclic compounds as sting agonists and methods and uses thereof
WO2021026009A1 (en) * 2019-08-02 2021-02-11 Mersana Therapeutics, Inc. Bis-[n-((5-carbamoyl)-1h-benzo[d]imidazol-2-yl)-pyrazol-5-carboxamide] derivatives and related compounds as sting (stimulator of interferon genes) agonists for the treatment of cancer
CN112920172A (en) * 2021-02-01 2021-06-08 厦门大学 Interferon-stimulated protein targeted compound, radioactive marker thereof, and preparation methods and applications of interferon-stimulated protein targeted compound and radioactive marker
CN114163420A (en) * 2021-10-26 2022-03-11 中山大学附属第一医院 Endoplasmic reticulum Golgi matrix targeting small molecule, conjugate and application thereof
WO2022166741A1 (en) * 2021-02-06 2022-08-11 正大天晴药业集团股份有限公司 Macrocyclic compound containing benzoheterocycle and acting as egfr kinase inhibitor, and pharmaceutical composition and use thereof
US11596692B1 (en) * 2018-11-21 2023-03-07 Incyte Corporation PD-L1/STING conjugates and methods of use
WO2024032782A1 (en) * 2022-08-12 2024-02-15 Jacobio Pharmaceuticals Co.Ltd. Vaccine adjuvants and uses thereof
DE102022131737A1 (en) 2022-11-30 2024-06-06 Technische Universität München, Körperschaft des öffentlichen Rechts Pyrrole compound and process for producing a pyrrole compound

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170240556A1 (en) * 2012-06-27 2017-08-24 4Sc Discovery Gmbh Bifluorodioxalane-amino-benzimidazole kinase inhibitors for the treatment of cancer, autoimmuneinflammation and cns disorders
WO2017175147A1 (en) * 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides useful as protein modulators

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170240556A1 (en) * 2012-06-27 2017-08-24 4Sc Discovery Gmbh Bifluorodioxalane-amino-benzimidazole kinase inhibitors for the treatment of cancer, autoimmuneinflammation and cns disorders
WO2017175147A1 (en) * 2016-04-07 2017-10-12 Glaxosmithkline Intellectual Property Development Limited Heterocyclic amides useful as protein modulators

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11596692B1 (en) * 2018-11-21 2023-03-07 Incyte Corporation PD-L1/STING conjugates and methods of use
WO2021014365A1 (en) * 2019-07-22 2021-01-28 Lupin Limited Macrocyclic compounds as sting agonists and methods and uses thereof
WO2021026009A1 (en) * 2019-08-02 2021-02-11 Mersana Therapeutics, Inc. Bis-[n-((5-carbamoyl)-1h-benzo[d]imidazol-2-yl)-pyrazol-5-carboxamide] derivatives and related compounds as sting (stimulator of interferon genes) agonists for the treatment of cancer
US11155567B2 (en) 2019-08-02 2021-10-26 Mersana Therapeutics, Inc. Sting agonist compounds and methods of use
US20220064189A1 (en) * 2019-08-02 2022-03-03 Mersana Therapeutics, Inc. Sting agonist compounds and methods of use
US11939343B2 (en) 2019-08-02 2024-03-26 Mersana Therapeutics, Inc. Sting agonist compounds and methods of use
CN112920172A (en) * 2021-02-01 2021-06-08 厦门大学 Interferon-stimulated protein targeted compound, radioactive marker thereof, and preparation methods and applications of interferon-stimulated protein targeted compound and radioactive marker
WO2022166741A1 (en) * 2021-02-06 2022-08-11 正大天晴药业集团股份有限公司 Macrocyclic compound containing benzoheterocycle and acting as egfr kinase inhibitor, and pharmaceutical composition and use thereof
CN114163420A (en) * 2021-10-26 2022-03-11 中山大学附属第一医院 Endoplasmic reticulum Golgi matrix targeting small molecule, conjugate and application thereof
CN114163420B (en) * 2021-10-26 2023-01-24 中山大学附属第一医院 Endoplasmic reticulum Golgi matrix targeting small molecule, conjugate and application thereof
WO2024032782A1 (en) * 2022-08-12 2024-02-15 Jacobio Pharmaceuticals Co.Ltd. Vaccine adjuvants and uses thereof
DE102022131737A1 (en) 2022-11-30 2024-06-06 Technische Universität München, Körperschaft des öffentlichen Rechts Pyrrole compound and process for producing a pyrrole compound

Similar Documents

Publication Publication Date Title
WO2020132582A1 (en) Sting agonists and uses thereof
EP3262049B1 (en) Tyk2 inhibitors and uses thereof
AU2013207972B2 (en) IRAK inhibitors and uses thereof
WO2020010210A1 (en) Mertk degraders and uses thereof
AU2020407200A1 (en) Irak degraders and uses thereof
WO2017004133A1 (en) Irak inhibitors and uses thereof
WO2015164374A1 (en) Irak inhibitors and uses thereof
CA2940666A1 (en) Tyk2 inhibitors and uses thereof
WO2012097013A1 (en) Irak inhibitors and uses thereof
WO2017004134A1 (en) Irak inhibitors and uses thereof
WO2014110114A1 (en) Irak inhibitors and uses thereof
EP3049086A1 (en) Irak inhibitors and uses thereof
EP2872144A1 (en) Irak inhibitors and uses thereof
EP4232425A1 (en) Ctps1 inhibitors and uses thereof
WO2014194245A2 (en) Cdk8 inhibitors and uses thereof
WO2020132549A1 (en) Sting heterocycle agonists and uses thereof
AU2022256074A1 (en) Cbl-b modulators and uses thereof
EP4291235A1 (en) Hpk1 antagonists and uses thereof
EP4121043A1 (en) Mdm2 degraders and uses thereof
WO2014194242A2 (en) Flt3 inhibitors and uses thereof
WO2020132566A1 (en) Sting pyrazole agonists and uses thereof
WO2021158516A1 (en) IRE1α MODULATORS AND USES THEREOF
EP4232059A1 (en) Double degraders and uses thereof
WO2023205475A1 (en) Ctps1 inhibitors and uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19899715

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19899715

Country of ref document: EP

Kind code of ref document: A1