WO2022251359A1 - Bicyclic inhibitors of alk5 and methods of use - Google Patents

Bicyclic inhibitors of alk5 and methods of use Download PDF

Info

Publication number
WO2022251359A1
WO2022251359A1 PCT/US2022/030925 US2022030925W WO2022251359A1 WO 2022251359 A1 WO2022251359 A1 WO 2022251359A1 US 2022030925 W US2022030925 W US 2022030925W WO 2022251359 A1 WO2022251359 A1 WO 2022251359A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
membered heterocycle
compound
carbocycle
fibrosis
Prior art date
Application number
PCT/US2022/030925
Other languages
French (fr)
Inventor
Christina OWENS
Steven D.E. Sullivan
Paul Allegretti
Svitlana KULYK
Erik FENSTER
Jennifer KOZAK
Mandy Loo
Adam D. Hughes
Original Assignee
Theravance Biopharma R&D Ip, Llc
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 Theravance Biopharma R&D Ip, Llc filed Critical Theravance Biopharma R&D Ip, Llc
Publication of WO2022251359A1 publication Critical patent/WO2022251359A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms 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
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • fibrotic diseases such as systemic sclerosis (SSc), sclerodermatous graft vs. host disease, nephrogenic system fibrosis, and radiation-induced fibrosis, as well as cardiac, pulmonary, skin, liver, bladder and kidney fibrosis, constitute a major health problem.
  • SSc systemic sclerosis
  • sclerodermatous graft vs. host disease nephrogenic system fibrosis
  • radiation-induced fibrosis as well as cardiac, pulmonary, skin, liver, bladder and kidney fibrosis
  • cardiac, pulmonary, skin, liver, bladder and kidney fibrosis constitute a major health problem.
  • Activated myofibroblasts may be responsible for replacing normal tissues with nonfunctional fibrotic tissue. Therefore, signaling pathways responsible for stimulating profibrotic reactions in myofibroblasts have potential as targets for development of therapies to treat fibrotic diseases.
  • TGF- ⁇ extracellular matrix
  • BMP bone morphogenic protein
  • TGF- ⁇ signaling is typically initiated by binding of a TGF- ⁇ ligand to a TGF- ⁇ RII. This in turn may recruit and phosphorylate TGF- ⁇ RI, also known as the activin receptor-like kinase 5 (ALK5). Once phosphorylated, ALK5 typically adopts an active conformation and is free to associate with and phosphorylate Smad2 or Smad3. Once phosphorylated, Smad 2 and 3 proteins then may form heterodimeric complexes with Smad4 which can translocate across the nuclear membrane and modulate Smad-mediated gene expression, including, for example, the production of collagen.
  • TGF- ⁇ RII also known as the activin receptor-like kinase 5
  • ALK5 activin receptor-like kinase 5
  • Smad2 and 3 proteins then may form heterodimeric complexes with Smad4 which can translocate across the nuclear membrane and modulate Smad-mediated gene expression, including, for example, the production of collagen.
  • ALK5 is believed to be the most relevant of the activin-like kinases (ALKs) in the fibrotic process (Rosenbloom, et al., Fibrosis: Methods and. Protocols, Methods in Molecular Biology, 2017, Vol. 1627, Chapter 1, pp. 1-21).
  • ALK5 activin-like kinases
  • ALK5 inhibitors have been associated with ventricular or cardiac remodeling in preclinical safety studies resulting from significant systemic exposure from oral administration.
  • the present disclosure provides these and other related advantages.
  • One objective of the present disclosure is to deliver a potent ALK5 inhibitor locally with minimal systemic exposure in. order to address any unintended and unwanted systemic side effects of ALK5 inhibition during treatment.
  • the present disclosure provides inhaled, long-acting and lung-selective ALK5 inhibitors for the treatment of idiopathic pulmonary fibrosis.
  • Compounds of the present disclosure may be used to treat other diseases, including, but not limited to, pulmonary fibrosis, liver fibrosis, renal glomerulosclerosis, and cancer.
  • Compounds of the present disclosure may be used as a monotherapy or co- dosed with other therapies, whether delivered by inhalation, orally, intravenously, subcutaneously, or topically.
  • X and ⁇ are each independently selected from CH and N;
  • A is a 9- or 10-membered bicyclic heteroaryl group selected from:
  • B is selected from phenyl, pyridyl, and thiazolyl
  • R A is selected from:
  • R B is independently selected at each occurrence from halogen, -CN, -NH 2 , -NHCH 3 , -NHCH 2 CH 3 , -C(O)CH 3 , -OH-,OC H 3 , -OCH 2 CH 3 , -CH 3 , -CH 2 CH 3 , -CH(CH 3 ) 2 , -C(CH 3 ) 3 , -CH 2 F, -CHF 2 , -CF 3 , C 3 .4 carbocycle, and 3- to 4-membered heterocycle; n is an integer from 0 to 3;
  • R 2 and R 3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R 1 ;
  • X and Y are each independently selected from CH and N;
  • A is a 9- or 10-membered bicyclic heteroaryl group selected from:
  • B is selected from phenyl, pyridyl, and thiazolyl
  • R A is selected from:
  • R B is independently selected at each occurrence from halogen, -CN, -NH 2 , -NHCH 3 , -NHCH 2 CH 3 , -C(O)CH 3 , -OH, -OCH 3 , -OCH 2 CH 3 , -CH 3 , -CH 2 CH 3 , -CH(CH 3 ) 2 , -C(CH 3 ) 3 , -CH 2 F, -CHF 2 , -CF 3 , C 3-4 carbocycle, and 3- to 4-membered heterocycle; n is an integer from 0 to 3;
  • R 2 and R 3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R 1 ;
  • X may be CH and Y may be N. In some embodiments, X is N and Y is CH. In some embodiments, X and Y are each N.
  • B may be selected from and . In some embodiments, B is . In some embodiments, B is
  • a compound of Formula (I) may be selected from and
  • A may be selected from and . In some embodiments, A is selected from and . In some embodiments, A is selected from and
  • A is selected from. and . In some embodiments, A is selected from , and . In some embodiments, A is selected from , and .
  • R A may be selected from:
  • C 10 aryl, C 3-12 cycloalkyl, and C 5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -CN, -OR 1 , -SR 1 , -CH 2 N(R 1 ) 2 , -N(R 1 ) 2 , -NR 2 R 3 , -C(O)R 1 , -CH 2 C(O)OR 1 , -C(O)OR 1 , -OC(O)R 1 , -NR 1 C(O)R 1 , -NR 1 C(O)OR 1 , -NR 1 C(O)N(R 1 ) 2 , -NR 1 C(O)NR 2 R 3 , -C(O)N(R 1 ) 2 , -C(O)NR 2 R 3 , O, R 1 , C 1-6 alkyl, and C 1-6 haloalkyl; and
  • each C 3-12 carbocycle and 3- to 12-membered heterocycle in R A is independently optionally substituted with one or more substituents selected from halogen, -CN, -OR 1 , -SR 1 , -CH 2 N(R 1 ) 2 , -N(R 1 ) 2 , -NR 2 R 3 , -C(O)R 1 , -CH 2 C(O)OR 1 , -C(O)OR 1 , -OC(O)R 1 , -NR 1 C(O)R 1 , -NR 1 C(O)OR 1 , -NR 1 C(O)N(R 1 ) 2 , -NR 1 C(O)NR 2 R 3 , -C(O)N(R 1 ) 2 , -C(O)NR 2
  • R A may be selected from:
  • R A is selected from -C(O)OR 3 , -NR 1 C(O)R 1 , and -NR 1 C(O)N(R 1 ) 2 .
  • R 1 is independently selected at each occurrence from hydrogen; and C 1-6 alkyl and C 0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -NH 2 and -CH 3 .
  • R 5 is independently selected at each occurrence from C 1-6 alkyl and C 0-3 alkyl-(3- to 12-membered heterocycle).
  • R A is selected from -NH-(3- to 12-membered heterocycle) and -NH-(C 3-12 carbocycle), each of which is optionally substituted with C 0-3 alkyl-(3- to 12- membered heterocycle), wherein the C 0-3 alkyl-(3- to 12-membered heterocycle) is optionally substituted by one or more substituents selected from halogen, -NH 2 , -NHCH 3 , -N(CH 3 ) 2 , -NHCH 2 CH 3 , -CH 2 CH 2 N(CH 3 ) 2 , -CH 3 , and -CF 3 .
  • R A is selected from:
  • R 1 is independently selected at each occurrence from hydrogen; and C 1-6 alkyl and C 0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by -NH 2 ; and
  • R 5 is independently selected at each occurrence from C1-6 alkyl and C 0-3 alkyl-(3- to 12-membered heterocycle).
  • A is selected from and
  • B is selected from and ;
  • R A is selected from:
  • B is selected from and ;
  • R A is selected from -NH-(3- to 12-membered heterocycle) and -NH-(C 3-12 carbocycle), each of which is optionally substituted with C 0-3 alkyl-(3- to 12-membered heterocycle), wherein the C 0-3 alkyl-(3- to 12- membered heterocycle) is optionally substituted by one or more substituents selected from halogen, -NH 2 , -NHCH 3 , -N(CH 3 ) 2 , -NHCH 2 CH 3 , -CH 2 CH 2 N(CH 3 ) 2 , -CH 3 , and -CF 3 .
  • A is selected from , and
  • B is selected from , and ;
  • R A is selected from:
  • A is selected from and
  • B is selected from and
  • R A is selected from:
  • the present disclosure provides a compound of Formula (I) wherein B is
  • the present disclosure provides a substantially pure stereoisomer of a compound or salt disclosed herein. In certain aspects, the present disclosure provides a substantially pure stereoisomer of a compound disclosed herein. The stereoisomer may be provided in at least 90% enantiomeric excess. [0024] In certain aspects, the present disclosure provides a compound selected from Table 1, or a pharmaceutically acceptable salt thereof. In certain aspects, the present disclosure provides a compound selected from Table la, or a pharmaceutically acceptable salt thereof.
  • the present disclosure provides a conjugate of the formula: wherein:
  • A' is an antibody construct or targeting moiety
  • L 1 is a linker
  • D' is a compound or salt disclosed herein; and p is an integer from 1 to 20.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound disclosed herein and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition may be formulated for inhalation.
  • the present disclosure provides a method of inhibiting ALK5, comprising contacting ALK5 with an effective amount of a compound or salt disclosed herein.
  • the present disclosure provides a method of treating an ALK5 -mediated disease or condition in a subject, comprising administering to the subject a therapeutically effective amount of a compound or salt disclosed herein.
  • the disease or condition may be selected from fibrosis, alopecia, and cancer.
  • the disease or condition is fibrosis.
  • the present disclosure provides a method of treating fibrosis, comprising administering to a patient a therapeutically effective amount of a compound or salt disclosed herein.
  • the fibrosis may be selected from systemic sclerosis, nephrogenic systemic fibrosis, organ-specific fibrosis, fibrosis associated with cancer, cystic fibrosis, and fibrosis associated with an autoimmune disease.
  • the organ-specific fibrosis is selected from cardiac fibrosis, kidney fibrosis, pulmonary fibrosis, liver fibrosis, portal vein fibrosis, skin fibrosis, bladder fibrosis, intestinal fibrosis, peritoneal fibrosis, myelofibrosis, oral submucous fibrosis, and retinal fibrosis.
  • the organ-specific fibrosis is intestinal fibrosis.
  • the pulmonary fibrosis is selected from idiopathic pulmonary fibrosis (IPF), familial pulmonary' fibrosis (FPF), interstitial lung fibrosis, fibrosis associated with asthma, fibrosis associated with chronic obstructive pulmonary disease (COPD), silica -induced fibrosis, asbestos -induced fibrosis and. chemotherapy -induced lung fibrosis.
  • the pulmonary fibrosis is idiopathic pulmonary fibrosis (IPF).
  • the pulmonary fibrosis was induced by a viral infection.
  • the disease or condition may be cancer, optionally wherein the cancer is selected from breast cancer, coion cancer, prostate cancer, lung cancer, hepatocellular carcinoma, glioblastoma, melanoma, and pancreatic cancer.
  • the cancer is lung cancer, optionally non-small cell lung cancer.
  • a method of the subject disclosure may further comprise administering a second therapeutic agent.
  • the second therapeutic agent is an immunotherapeutic agent, such as a PD-1 inhibitor or a CTLA-4 inhibitor.
  • the immunotherapeutic agent is selected from pembrolizumab and durvalumab.
  • a method of the present disclosure may further comprise administering an effective amount of radiation.
  • the compound or salt disclosed herein may be administered by inhalation.
  • the present disclosure provides a compound disclosed herein for use in treating fibrosis. In certain aspects, the present disclosure provides the use of a compound disclosed herein for the manufacture of a medicament for treating fibrosis.
  • C x-y or “C x -C y ” when used in conjunction with a chemical moiety, such as alkyl, alkenyl, or alkynyl, is meant to include groups that contain from x to y carbons in the chain.
  • C x-y alkyl refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups, that contain from x to y carbons in the chain.
  • Alkyl refers to substituted or unsubstituted saturated hydrocarbon groups, including linear and branched alkyl groups.
  • An alkyl group may contain from one to twelve carbon atoms (e.g., C 1-12 alkyl), such as one to eight carbon atoms (C 1-8 alkyl) or one to six carbon atoms (C 1-6 alkyl).
  • alkyl groups include methyl, ethyl, n-propyi, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, hexyl, septyl, octyl, nonyl, and decyl.
  • An alkyl group is attached to the rest of the molecule by a single bond. Unless stated otherwise specifically in the specification, an alkyl group is optionally substituted by one or more substituents such as those substituents described herein.
  • Haloalkyl refers to an alkyl group that is substituted by one or more halogens.
  • exemplary haloalkyl groups include trifluoromethyl, di fluoromethyl, trichloro methyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl, 3- bromo-2-fluoropropyl , and 1,2-dibromoethyl.
  • alkenyl refers to substituted or unsubstituted, hydrocarbon groups, including linear and branched alkenyl groups, containing at least one double bond.
  • An alkenyl group may contain from two to twelve carbon atoms (e.g., C 2-12 alkenyl), such as two to eight carbon atoms (C 2-8 alkenyl) or two to six carbon atoms (C 2-6 alkenyl).
  • Exemplary alkenyl groups include ethenyl (i.e., vinyl), prop-1-enyl, but-1-enyl, pent-1-enyl, penta- 1, 4-dienyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group is optionally substituted by one or more substituents such as those substituents described herein.
  • Alkynyl refers to substituted or unsubstituted hydrocarbon groups, including linear and branched alkynyl groups, containing at least one triple bond.
  • An alkynyl group may contain from two to twelve carbon atoms (e.g., C 2-12 alkynyl), such as two to eight carbon atoms (C 2-8 alkynyl) or two to six carbon atoms (C 2-6 alkynyl).
  • Exemplary alkynyl groups include ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like.
  • an alkynyl group is optionally substituted by one or more substituents such as those substituents described herein.
  • Alkylene or “alkylene chain” refers to substituted or unsubstituted divalent saturated hydrocarbon groups, including linear alkylene and branched alkylene groups, that contain from one to twelve carbon atoms (e.g., C 1-12 alkylene), such as one to eight carbon atoms (C 1-8 alkylene) or one to six carbon atoms (C 1-6 alkylene).
  • Exemplary alkylene groups include methylene, ethylene, propylene, and n-butylene.
  • alkenylene and alkynylene refer to alkylene groups, as defined above, which comprise one or more carbon-carbon double or triple bonds, respectively.
  • alkylene, alkenylene or alkynylene chain can be through one carbon or any two carbons of the chain.
  • an alkylene, alkenylene, or alkynylene group is optionally substituted by one or more substituents such as those substituents described herein.
  • Heteroalkyl refers to substituted or unsubstituted alkyl, alkenyl and alkynyl groups, respectively, in which one or more, such as 1, 2 or 3, of the carbon atoms are replaced with a heteroatom, such as O, N, P, Si, S, or combinations thereof. Any nitrogen, phosphorus, and sulfur heteroatoms present in the chain may optionally be oxidized, and any nitrogen heteroatoms may optionally be quaternized. If given, a numerical range refers to the chain length in total. For example, a 3- to 8-membered heteroalkyl group has a chain length of 3 to 8 atoms.
  • Connection to the rest of the molecule may be through either a heteroatom or a carbon in the heteroalkyl, heteroalkenyl or heteroalkynyl chain.
  • a heteroalkyl, heteroalkenyl, or heteroalkynyl group is optionally substituted by one or more substituents such as those substituents described herein.
  • Heteroalkylene refers to substituted or unsubstituted alkylene, alkenylene and. alkynylene groups, respectively, in which one or more, such as 1, 2, or 3, of the carbon atoms are replaced with a heteroatom, such as 0, N, P, Si, S, or combinations thereof. Any nitrogen, phosphorus, and sulfur heteroatoms present in the chain may optionally be oxidized, and any nitrogen heteroatoms may optionally be quaternized. If given, a numerical range refers to the chain length in total. For example, a 3- to 8-membered heteroalkylene group has a chain length of 3 to 8 atoms.
  • the points of attachment of the heteroalkylene, heteroalkenylene or heteroalkynylene chain to the rest of the molecule can be through either one heteroatom or one carbon, or any two heteroatoms, any two carbons, or any one heteroatom and any one carbon in the heteroalkylene, heteroalkenylene or heteroalkynylene chain.
  • a heteroalkylene, heteroalkenylene, or heteroalkynylene group is optionally substituted by one or more substituents such as those substituents described herein.
  • “Carbocycle” refers to a saturated, unsaturated, or aromatic ring in which each atom of the ring is a carbon atom.
  • Carbocycle may include C 3-10 monocyclic rings, C 5-12 bicyclic rings, C 6-12 spirocyclic rings, and ( C 6-12 bridged rings. Each ring of a bicyclic carbocycle may be selected from saturated, unsaturated, and aromatic rings.
  • the carbocycle is a C 6-12 aryl group, such as C 6-10 aryl.
  • the carbocycle is a C 6-12 cycloalkyl group.
  • the carbocycle is a C 6-12 cycloalkenyl group.
  • cycloalkenyl refers to a non-aromatic ring containing at least one double bond, wherein each atom of the ring is a carbon atom.
  • an aromatic ring e.g., phenyl
  • a saturated or unsaturated ring e.g., cyclohexane, cyclopentane, or cyclohexene. Any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits, are included in the definition of carbocycle.
  • Exemplary carbocycles include cyclopentyl, cyclohexyl, cyclohexenyl, adamantyl, phenyl, indanyl, and naphthyl. Unless stated otherwise specifically in the specification, a carbocycle is optionally substituted by one or more substituents such as those substituents described herein.
  • Heterocycle refers to a saturated, unsaturated, or aromatic ring comprising one or more heteroatoms, for example 1, 2, or 3 heteroatoms selected from O, S and N. Heterocycles include 3- to 10- membered monocyclic rings, 6- to 12-membered bicyclic rings, 6- to 12-membered spirocyclic rings, and 6- to 12-membered bridged rings. Each ring of a bicyclic heterocycle may be selected from saturated, unsaturated, and aromatic rings. The heterocycle may be attached to the rest of the molecule through any atom of the heterocycle, valence permitting, such as a carbon or nitrogen atom of the heterocycle.
  • the heterocycle is a 5- to 10-membered heteroaryl group, such as 5- or 6-membered heteroaryl.
  • the heterocycle is a 3- to 12-membered heterocycloalkyl group.
  • a heterocycle e.g., pyridyl
  • heterocycles include pyrrolidinyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, piperidinyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, thiophenyl, oxazolyl, thiazolyl, morpholinyl, indazolyl, indolyl, and quinolinyl.
  • a heterocycle is optionally substituted by one or more substituents such as those substituents described herein.
  • Heteroaryl refers to a 5- to 12-membered aromatic ring that comprises at least one heteroatom, such as 1, 2, or 3 heteroatoms, selected from O, S, and N.
  • the heteroaryl ring may be selected from monocyclic or bicyclic — including fused, spirocyclic and bridged ring systems — wherein at least one of the rings in the ring system, is aromatic.
  • the heteroatom(s) in the heteroaryl may optionally be oxidized.
  • One or more nitrogen atoms, if present, are optionally quaternized.
  • heteroaryl may be attached to the rest of the molecule through any atom of the heteroaryl, valence permitting, such as a carbon or nitrogen atom of the heteroaryl.
  • heteroaryl groups include, but are not limited to, azepinyl, benzimidazolyl, benzisothiazolyl, benzisoxazolyl, benzofuranyl, benzothiazolyl, benzothiophenyl, benzoxazolyl, furanyl, imidazolyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, purinyl, pyrazinyl, pyrazolidinyl, pyrazolyl, pyridazinyl, pyridazolyl, pyridyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl,
  • a waved line drawn across a bond or a dashed bond are used interchangeably herein to denote where a bond, disconnection or attachment occurs.
  • R a is attached to the para position of a fluorophenyl ring through a single bond. If R a is 2-pyridine as in , then R a may be depicted as or
  • substituted refers to moieties having substituents replacing a hydrogen on one or more carbons or heteroatoms of the structure. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance 'with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and iron-aromatic substituents of organic compounds.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • heteroatoms such as nitrogen may have any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
  • R 2 and R 3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R 1 .
  • R 2 and R 3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R 1 .
  • bivalent substituent groups are specified herein by their conventional chemical formulae, written from left to right, they are intended to encompass the isomer that would result from writing the structure from right to left, e.g., -CH 2 O- is also intended to encompass to -OCH 2 -
  • Compounds of the present disclosure also include crystall ine and amorphous forms of those compounds, pharmaceutically acceptable salts, and active metabolites having the same type of activity, including, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrat.es), conformational polymorphs, amorphous forms of the compounds, and mixtures thereof.
  • the compounds described herein may exhibit their natural isotopic abundance, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. All isotopic variations of the compounds of the present disclosure, whether radioactive or not, are encompassed within the scope of the present disclosure.
  • hydrogen has three naturally occurring isotopes, denoted 1 H (protium), 2 H (deuterium), and 3 H (tritium). Protium is the most abundant isotope of hydrogen in nature.
  • Enriching for deuterium may afford certain therapeutic advantages, such as increased in vivo half-life and/or exposure, or may provide a compound useful for investigating in vivo routes of drag elimination and metabolism.
  • isotopes that may be incorporated into compounds of the present disclosure include, but are not limited to, 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 35 S, 36 CI, and 18 F.
  • compounds of Formula (I) enriched in tritium or carbon- 14, which can be used, for example, in tissue distribution studies; compounds of the disclosure enriched in deuterium especially at a site of metabolism, resulting, for example, in compounds having greater metabolic stability; and compounds of Formula (I) enriched in a positron emitting isotope, such as 11 C, 18 F, 15 O and 13 N, which can be used, for example, in Positron Emission Topography (PET) studies.
  • Isotopically-enriched compounds may be prepared by conventional techniques well known to those skilled in the art.
  • Certain compounds described herein contain one or more asymmetric centers and can thus give rise to enantiomers, diastereomers, and other stereoisomeric forms, the asymmetric centers of which can be defined, in terms of absolute stereochemistry, as (R)- or (S)-.
  • the carbon atoms in order to optimize the therapeutic activity of the compounds of the disclosure, e.g., to treat fibrosis, it may be desirable that the carbon atoms have a particular configuration (e.g., (R,R), (5,5), (S,R), or (R,S)') or are enriched in a stereoisomeric form having such configuration.
  • the compounds of the disclosure may be provided as racemic mixtures.
  • the disclosure relates to racemic mixtures, pure stereoisomers (e.g., enantiomers and diastereoisomers), stereoisomer -enriched mixtures, and the like, unless otherwise indicated.
  • pure stereoisomers e.g., enantiomers and diastereoisomers
  • stereoisomer -enriched mixtures and the like, unless otherwise indicated.
  • a chemical structure is depicted herein without any stereochemistry, it is understood that all possible stereoisomers are encompassed by such structure.
  • a particular stereoisomer is shown or named herein, it will be understood by those skilled in the art that minor amounts of other stereoisomers may be present in the compositions of the disclosure unless otherwise indicated, provided that the utility of the composition as a whole is not eliminated by the presence of such other isomers.
  • stereoisomers may be obtained by numerous methods that are known in the art, including preparation using chiral synthons or chiral reagents, resolution using chiral chromatography using a suitable chiral stationary phase or support, or by chemically converting them into diastereoisomers, separating the diastereoisomers by conventional means such as chromatography or recrystallization, then regenerating the original stereoisomer.
  • tautomer refers to each of two or more isomers of a compound that exist in equilibrium and which readily interconvert. For example, one skilled in the art would readily understand that 1,2,3-triazole exists in two tautomeric forms:
  • pharmaceutically acceptable refers to a material that is not biologically or otherwise unacceptable when used in the subject compositions and methods.
  • pharmaceutically acceptable carrier refers to a material — such as an adjuvant, excipient, glidant, sweetening agent, diluent, preservative, dye, colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent or emulsifier— -that can be incorporated into a composition and administered to a patient without causing unacceptable biological effects or interacting in an unacceptable manner with other components of the composition.
  • Such pharmaceutically acceptable materials typically have met the required standards of toxicological and manufacturing testing, and include those materials identified as suitable inactive ingredients by the U.S. Food and Drug Administration.
  • salts and “pharmaceutically acceptable salt” refer to a salt prepared from a base or an acid.
  • Pharmaceutically acceptable salts are suitable for administration to a patient, such as a mammal (for example, salts having acceptable mammalian safety for a given dosage regime). Salts can be formed from inorganic bases, organic bases, inorganic acids and organic acids.
  • a compound contains both a basic moiety, such as an amine, pyridine or imidazole, and an acidic moiety, such as a carboxylic acid, or tetrazole, zwitterions may be formed and are included within the term “salt” as used herein.
  • Salts derived from inorganic bases include ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium., sodium, and zinc salts, and the like.
  • Salts derived from organic bases include salts of primary, secondary and tertiary amines, including substituted amines, cyclic amines, naturally-occurring amines and the like, such as arginine, betaine, caffeine, choline, N,N' -dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N- ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobro
  • Salts derived from inorganic acids include salts of boric, carbonic, hydrohalic (hydrobromic, hydrochloric, hydrofluoric or hydroiodic), nitric, phosphoric, sulfamic, and sulfuric acids.
  • Saits derived from organic acids include salts of aliphatic hydroxyl acids (for example, citric, gluconic, glycolic, lactic, lactobionic, malic, and tartaric acids), aliphatic monocarboxylic acids (for example, acetic, butyric, formic, propionic and trifluoroacetic acids), amino acids (for example, aspartic and glutamic acids), aromatic carboxylic acids (for example, benzoic, p-chlorobenzoic, diphenylacetic, gentisic, hippuric, and triphenylacetic acids), aromatic hydroxyl acids (for example, o- hydroxy benzoic, p-hydroxybenzoic, 1-hydroxynaphthalene-2-carboxylic and 3-hydroxynaphthalene-2- carboxylic acids), ascorbic, dicarboxylic acids (for example, fumaric, maleic, oxalic and succinic acids), glucoronic, mandelic, mucic, nicotinic,
  • a therapeutically effective amount refers to that amount of a compound described herein that is sufficient to affect treatment when administered to a subject in need thereof.
  • a therapeutically effective amount for treating pulmonary fibrosis is an amount of compound needed, to, for example, reduce, suppress, eliminate, or prevent the formation of fibrosis in a subject, or to treat the underlying cause of pulmonary fibrosis.
  • the therapeutically effective amount may vary depending upon the intended treatment application (in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the specific dose will vary depending on the particular compound chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • the term “effective amount” refers to an amount sufficient to obtain a desired result, which may not necessarily be a therapeutic result.
  • an “effective amount” may be the amount needed to inhibit an enzyme.
  • treating refers to an approach for obtaining beneficial or desired results with respect to a disease, disorder, or medical condition (such as pulmonary' fibrosis) in a subject, including but not limited, to the following: (a) preventing the disease or medical condition from occurring, e.g., preventing the reoccurrence of the disease or medical condition or prophylactic treatment of a subject that is pre-disposed to the disease or medical condition; (b) ameliorating the disease or medical condition, e.g., eliminating or causing regression of the disease or medical condition in a subject; (c) suppressing the disease or medical condition, e.g., slowing or arresting the development of the disease or medical condition in a subject; or (d) alleviating symptoms of the disease or medical condition in a subject.
  • “treating pulmonary fibrosis” would include preventing fibrosis from occurring, ameliorating fibrosis, suppressing fibrosis, and alleviating the symptoms of fibrosis (for example, increasing oxygen levels in blood or improved lung function tests). Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject, notwithstanding that the subject may still be afflicted with the underlying disorder [0064]
  • a “therapeutic effect”, as that term is used herein, encompasses a therapeutic benefit and/or a prophylactic benefit as described above.
  • a prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
  • antagonists are used interchangeably, and they refer to a compound having the ability to inhibit a biological function (e.g., activity, expression, binding, protein-protein interaction) of a target protein (e.g., ALK5). Accordingly, the terms “antagonist” and “inhibitor” are defined in the context of the biological role of the target protein. While preferred antagonists herein specifically interact with (e.g., bind to) the target, compounds that inhibit a biological activity of the target protein by interacting with other members of the signal transduction pathway of which the target protein is a member are also specifically included within this definition.
  • selective inhibition refers to the ability of a biologically active agent to preferentially reduce the target signaling activity as compared to off-target signaling activity, via direct or indirect interaction with the target.
  • an antibody refers to an immunoglobulin molecule that specifically binds to, or is immunologically reactive toward, a specific antigen.
  • An antibody may be, for example, polyclonal, monoclonal, genetically engineered, or an antigen binding fragment thereof, and further may be, for example, murine, chimeric, humanized, heteroconjugate, bispecific, a diabody, a triabody, or a tetrabody.
  • An antigen binding fragment includes an antigen binding domain and may be in the form, of, for example, a Fab’, F(ab’) 2 , Fab, Fv, rlgG, scFv, hcAbs (heavy chain antibodies), a single domain antibody, V HH , V NAR , sdAb, or nanobody.
  • antigen binding domain refers to a region of a molecule that binds to an antigen.
  • An antigen binding domain may be an antigen-binding portion of an antibody or an antibody fragment.
  • An antigen binding domain may be one or more fragments of an antibody that retain the ability to specifically bind to an antigen.
  • An antigen binding domain can be an antigen binding fragment and may recognize a single antigen, two antigens, three antigens or more.
  • “recognize” with regard to antibody interactions refers to the association or binding between an antigen binding domain of an antibody or portion thereof and an antigen.
  • an “ antibody construct” refers to a molecule, e.g., a protein, peptide, antibody or portion thereof, that contains an antigen binding domain and an Fc domain (e.g., an Fc domain from within the Fc region).
  • An antibody construct may recognize, for example, one antigen or multiple antigens.
  • a “targeting moiety” refers to a structure that has a selective affinity for a target molecule relative to other non-targel molecules.
  • the targeting moiety binds to a target molecule.
  • a targeting moiety may include an antibody, a peptide, a ligand, a receptor, or a binding portion thereof.
  • the target biological molecule may be a biological receptor or other structure of a cell, such as a tumor antigen.
  • subject and “patient” refer to an animal, such as a mammal, for example a human.
  • the methods described herein can be useful in both human therapeutics and veterinary applications.
  • the subject is a mammal, and in some embodiments, the subject is human.
  • “Mammal” includes humans and both domestic animals such as laboratory animals and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.
  • Prodrug is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein (e.g., compound of Formula (I)).
  • prodrug refers to a precursor of a biologically active compound that is pharmaceutically acceptable.
  • a prodrug is inactive when administered to a subject but is converted in vivo to an active compound, for example, by hydrolysis.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp.
  • prodrug is also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a mammalian subject.
  • Prodrags of an active compound are typically prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound.
  • Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively.
  • Examples of prodrags include, but are not limited to, acetate, formate and benzoate derivatives of a hydroxy functional group, or acetamide, formamide and. benzamide derivatives of an amine functional group in the active compound, and. the like.
  • in vivo refers to an event that takes place in a subject's body.
  • in vitro refers to an event that takes places outside of a subject's body.
  • an in vitro assay encompasses any assay run outside of a subject.
  • in vitro assays encompass cell-based, assays in which cells alive or dead are employed.
  • In vitro assays also encompass a cell-free assay in which no intact cells are employed.
  • the disclosure is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, esterification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the disclosure includes compounds produced by a process comprising administering a compound of this disclosure to a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radiolabeled compound of the disclosure in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to a human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.
  • an animal such as rat, mouse, guinea pig, monkey, or to a human
  • Lung function tests include tests to check how well the lungs work. Spirometry, for example, measures the amount of air the lungs can hold as well as how forcefully one can empty air from, the lungs.
  • Forced expiratory volume is a measure of the amount of air a person can exhale during a forced breath. FEV1, for example, is the amount of air a person can force from their lungs in one second.
  • Forced vital capacity FVC is the total amount of air exhaled during an FEV test.
  • the ratio of FEV1/FVC also known as Index of Air Flow or Tiffeneau-Pinelli Index, is a measurement used to assess the health of a patient’s lung function.
  • a ratio of ⁇ 80% indicates an obstructive defect is present in the lungs, such as chronic obstructive pulmonary disease (COPD).
  • COPD chronic obstructive pulmonary disease
  • a ratio of > 80% indicates a restrictive defect is present in the lungs, such as pulmonary fibrosis.
  • the ratio of > 80% in restrictive lung disease results from both FEV1 and FVC being reduced but that the decline in FVC is more than that of FEV 1, resulting in a higher than 80% value.
  • transforming growth factor - ⁇ may also be referred to as TGF- ⁇ , transforming growth factor beta-1, or TGF-beta-1. It is also cleaved into latency-associated peptide (LAP).
  • LAP latency-associated peptide
  • TGF- ⁇ receptor II may also be referred to as TpRII, type II TGF- ⁇ receptor, TGF- ⁇ RII, TGF-beta receptor type-2, TGFR-2, TGF-beta type II receptor, transformi ng growth factor-beta receptor type II, TGF-beta receptor type II or TbetaR-II.
  • TGF- ⁇ receptor I may also be referred to as TpRI, type I TGF -p receptor, TGF- ⁇ RI, TGF- beta receptor type-1, TGFR-1 , activin A receptor type Il-like protein kinase of 53kD, activin receptor-like kinase 5, ALK-5, ALK5, serine/threonine -protein kinase receptor R4, SKR4, TGF-beta type I receptor, transforming growth factor-beta receptor type I, TGF-beta receptor type I, transforming growth factor beta receptor I, TGF -beta receptor 1, or TbetaR -I.
  • the present disclosure provides compounds that are capable of selectively binding to and/or modulating ALK5.
  • the compounds modulate ALK5 by binding to or interacting with one or more amino acids and/or one or more metal ions. The binding of these compounds may disrupt ALK5 downstream signaling.
  • X and Y are each independently selected from CH and N;
  • A is a 9- or 10-membered bicyclic heteroaryl group
  • B is selected from phenyl, pyridyl, and thiazolyl
  • R A is selected from:
  • R B is independently selected at each occurrence from halogen, -CN, -NH 2 , -NHCH 3 , -NHCH 2 CH 3 , -C(O)CH 3 , -OH, -OCH 3 , -OCH 2 CH 3 , -CH 3 , -CH 2 CH 3 , -CH(CH 3 ) 2 , -C(CH 3 ) 3 , -CH 2 F, -CHF 2 , -CF 3 , C3-4 carbocycle, and 3- to 4-membered heterocycle; n is an integer from 0 to 3;
  • R 2 and R 3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R 1 ;
  • R 4 is independently selected at each occurrence from C 1-3 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, CH 2 -(C 3-12 cycloalkyl), CH 2 -(C 5-12 cycloalkenyl), C 2-3 alkyl-(C 3-12 carbocycle), and C 1-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO 2 , -NH 2 , -NHCH 3 , -N(CH 3 ) 2 , -NHCH 2 CH 3 , -CH 2 CH 2 N(CH 3 ) 2 , -C(O)CH 3 , -C(O)OH, -C(O)OCH 3 ,
  • the present disclosure provides a compound of Formula (I): or a pharmaceutically acceptable salt thereof, wherein:
  • X and ⁇ are each independently selected from CH and N;
  • A is a 9- or 10-membered bicyclic heteroaryl group
  • B is selected from phenyl, pyridyl, and thiazolyl
  • R B is independently selected at each occurrence from halogen, -CN, -NH 2 , -NHCH 3 , -NHCH 2 CH,, -C(O)CH 3 , -OH, -OCH 3 , -OCH 2 CH 3 , -CH 3 , -CH 2 CH 3 , -CH(CH 3 ) 2 , -C(CH 3 ) 3 , -CH 2 F, -CHF 2 , -CF 3 , C 3-4 carbocycle, and 3- to 4-membered heterocycle; n is an integer from 0 to 3; R 1 is independently selected at each occurrence from hydrogen; and C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, 1- to 6-membered heteroalkyl, C 0-3 alkyl-(C 3-12 carbocycle), and C 0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from
  • R 2 and R 3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R 1 .
  • the present disclosure provides a compound of Formula (I): or a pharmaceutically acceptable salt thereof, wherein:
  • X and Y are each independently selected from CH and N;
  • A is a 9- or 10-membered bicyclic heteroaryl group selected from:
  • B is selected from phenyl, pyridyl, and thiazolyl
  • R A is selected from:
  • R B is independently selected at each occurrence from halogen, -CN, -NH 2 , -NHCH 3 , -NHCH 2 CH 3 , -C(O)CH 3 , -OH, -OCH 3 , -OCH 2 CH 3 , -CH 3 , -CH 2 CH 3 , -CH(CH 3 ) 2 , -C(CH 3 ) 3 , -CH 2 F, -CHF 2 , -CF 3 , C 3 .4 carbocycle, and 3- to 4-membered heterocycle; n is an integer from 0 to 3;
  • R 2 and R 3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R 1 ;
  • R 4 is independently selected at each occurrence from C 1-3 alkyl, CM alkenyl, CM alkynyl, CH 2 -(C 3-12 cycloalkyl), CH 2 -( C 5-12 cycloalkenyl), C 2-3 alkyl-( C 3-12 carbocycle), and C 1-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO 2 , -NH 2 , -NHCH 3 , -N(CH 3 ) 2 , -NHCH 2 CH 3 , -CH 2 CH 2 N(CH 3 ) 2 , -C(O)CH 3 , -C(O)OH, -C(O)OCH 3 ,
  • X and Y are N, such as X is CH and Y is N. In some embodiments, X is N and Y is CH. In some embodiments, X and Y are each N. In some embodiments, the compound of Formula (I) is a compound of Formula (I-A), (I-B), or (1-C):
  • B is phenyl, optionally wherein n is 2 or 3, such as or .
  • B is pyridyl, optionally wherein n is 1, such as
  • the compound of Formula (I) is a compound of Formula (I-D), (I-E), or (I-F):
  • the compound of Formula (I) is a compound of Formula (I-G), (I-H), or (I-I): or
  • R B is independently selected at each occurrence from halogen and C 1-6 alkyl, such as R B is independently selected at each occurrence from Cl, F, and -CH 3 .
  • n is an integer from 1 to 3, such as n is 1 or 2.
  • n is 2 and R B independently selected at each occurrence from Cl and F.
  • n is 1, and R B is -CH 3 .
  • B is phenyl, n is 2, and R B is independently selected at each occurrence from Cl and. F.
  • B is pyridyl, n is 1, and R B is -CH 3 .
  • A is a 9- or 10-membered bicyclic heteroaryi group substituted by at least one R A .
  • A comprises at least one ring nitrogen atom, and optionally further comprises one or more heteroatoms selected from N, S, and O.
  • A is selected from , and . In some embodiments, A is selected from , and . In some embodiments, A is selected from and . In some embodiments, A is selected from , and . In some embodiments, A is selected from and . In some embodiments, A is selected from and . In some embodiments, A is selected from and . In some embodiments, A is selected from and .
  • the compound of Formula (I) is a compound of Formula (I-J): , such as or
  • the compound of Formula (I) is a compound of Formula (I-K): , such as or
  • the compound of Formula (I) is a compound of Formula (I-L): , such as or
  • the compound of Formula (I) is a compound of Formula (I-M): , such as or
  • the compound of Formula (I) is a compound of Formula (I-N): , such as or
  • the compound of Formula (I) is a compound of Formula (I-O): , such as or
  • the compound of Formula (I) is a compound of Formula (I-P): , such as or
  • the compound of Formula (I) is a compound of Formula (I-Q) : , such as or
  • the compound of Formula (I) is a compound of Formula (I-R): , such as or
  • the compound of Formula (I) is a compound of Formula (I-S): , such as or
  • the compound of Formula (I) is a compound of Formula (I-T): , such as or
  • the compound of Formula (I) is a compound of Formula (I-U): , such as or
  • the compound of Formula (I) is a compound of Formula (I-V): , such as or
  • the compound of Formula (I) is a compound of Formula (I-W): , such as or
  • the compound of Formula (I) is a compound of Formula (I-X): , such as or
  • the compound of Formula (I) is a compound of Formula (I-Y): such as or
  • the compound of Formula (I) is a compound of Formula (I-Z): , such as or
  • the compound of Formula (I) is a compound of Formula (I-AA): , such as or
  • the compound of Formula (I) is a compound of Formula (I-BB): , such as or
  • the compound of Formula (I) is a compound of Formula (I-CC): , such as or
  • references herein to “a compound of Formula (I)” implicitly also include the compound of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-F), (LG), (I-H), (I-I), (I-J), (I-K), (I-L), (I-M), (I-N), (I-O), (I-P), (I-Q), (I-A), (I-B), (I-C), (I-D), (I-E), (I-F), (LG), (I-H), (I-I), (I-J), (I-K), (I-L), (I-M), (I-N), (I-O), (I-P), (I-Q), (I-
  • R 2 and R 3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R 1 .
  • R A is selected from:
  • R 2 and R 3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R 1 ;
  • R A is selected from:
  • C 10 aryl, C 3-12 cycloalkyl, and C 5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -CN, -OR 1 , -SR 1 , -CH 2 N(R 1 ) 2 , -N(R 1 ) 2 , -NR 2 R 3 , -C(O)R 1 , -CH 2 C(O)OR 1 , -C(O)OR 1 , -OC(O)R 1 , -NR 1 C(O)R 1 , -NR 1 C(O)OR 1 , -NR 1 C(O)N(R 1 ) 2 , -NR 1 C(O)NR 2 R 3 , -C(O)N(R 1 ) 2 , -C(O)NR 2 R 3 , O, R 1 , C 1-6 alkyl, and C 1-6 haloalkyl; and
  • each C 3-12 carbocycle and 3- to 12-membered heterocycle in R A is independently optionally substituted with one or more substituents selected from halogen, -CN, -OR 1 , -SR 1 , -CH 2 N(R 1 ) 2 , -N(R 1 ) 2 , -NR 2 R 3 , -C(O)R 1 , -CH 2 C(O)OR 1 , -C(O)OR 1 , -OC(O)R 1 , -NR 1 C(O)R 1 , -NR 1 C(O)OR 1 , -NR 1 C(O)N(R 1 ) 2 , -NR 1 C(O)NR 2 R 3 , -C(O)N(R 1 ) 2 , -C(O)NR 2
  • R A is selected from:
  • R A is selected from:
  • R A is selected from -C(O)OR 5 , -NR 1 C(O)R 1 , and -NR 1 C(O)N(R 1 ) 2 .
  • R A is -NHC(O)R 1 , such as -NHC(O)-CH 2 -piperazinyl.
  • R A is 3- to 12-membered heterocycle, such as 4- to 6-membered heterocycle.
  • R A is 8- to 11-membered spirocyclic heterocycle, such as 9-membered spirocyclic heterocycle.
  • R A is phenyl substituted with -CH 2 NH 2 .
  • R A is -NH-(3- to 12-membered heterocycle), wherein the 3- to 12-membered heterocycle is substituted with R 1 , such as R A is -NH-(pyridyl)- amino -piperidyl.
  • R A is -C(O)OR 1 , such as -C(O)O-(C 5-10 carbocycle).
  • R A is -NH- (phenyl), wherein the phenyl is substituted with 3- to 6-membered heterocycle.
  • R A is 3- to 6-membered heterocycle substituted by R 1 , such as pyrazolyl substituted by -CH 2 CH 2 NH 2 or thiophenyl substituted by -CH 2 NH 2 .
  • R A is -NH-(C 3-12 carbocycle), optionally substituted by -NHz, such as -NH-(indane)-NH 2 .
  • R A is selected from -NH-(3- to 12-membered heterocycle) and -NH-(C 3-12 carbocycle), each of which is optionally substituted with C 0-3 alkyl-(3- to 12-membered heterocycle), wherein the Co.3 aIkyl-(3- to 12-membered heterocycle) is optionally substituted by one or more substituents selected from halogen, -NH 2 , -NHCH 3 , -N(CH 3 ) 2 , -NHCH 2 CH 3 , -CH 2 CH 2 N(CH 3 ) 2 , -CH 3 , and -CF 3 .
  • R 1 is independently selected at each occurrence from hydrogen; and aClk 1 y -6 l and C 0-3 alkyl -(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -NH 2 and -CH 3 .
  • R A is -NR'R 4 , wherein R‘ is hydrogen and R 4 is C 1-3 alkyl-(3- to 12-membered heterocycle); or C 1-3 alkyl substituted by -C(O)OCH 3 or -C(O)OCH 2 CH 3 .
  • R A is -NR 1 R 4 , wherein R 1 is hydrogen and R 4 is C 1-3 alkyI-(3- to 12- membered heterocycle).
  • R A is -NR 1 R 4 , wherein R 1 is hydrogen and R 4 is C 1-3 alkyl substituted by -C(O)OCH 3 or -C(O)OCH 2 CH 3 .
  • R 5 is independently selected at each occurrence from C 1-6 alkyl and C 0-3 alkyl -(3 - to 12-membered heterocycle).
  • R A may be -C(O)OR 5 , wherein R 5 is selected from C al 1 k -6 yl and C 0-3 alkyl-(3- to 12-membered heterocycle).
  • R A is -C(O)OR 5 , wherein R 5 is C 0-3 aIkyl-(3- to 12-membered heterocycle), wherein the 3 - to 12-membered heterocycle is optionally substituted with halogen, -NH 2 , -NHCH 3 , -N(CH 3 ) 2 , -NHCH 2 CH 3 , -CH 2 CH 2 N(CH 3 ) 2 , -CH 3 , -CH 2 CH 3 , C 3-12 carbocycle, and 3- to 6-membered heterocycle.
  • R A is -C(O)OR 5 , wherein R 5 is C 1-6 alkyl. [0100]
  • R A is selected from and .
  • R A is selected from , and
  • R A is selected from , and . In some embodiments, R A is selected from , and . In some embodiments, R A is selected from , and . In some embodiments, R A is selected from , and . In some embodiments, R A is selected from , and , In some embodiments, R A is selected from , and
  • R A is selected from:
  • R 5 is independently selected at each occurrence from C 1-6 alkyl and C 0-3 alkyl-(3- to 12-membered heterocycle).
  • A is selected from and.
  • B is selected from , and ;
  • R A is selected from:
  • a is B is selected from , and
  • R A is selected from -NH-(3- to 12-membered heterocycle) and -NH-(C 3-12 carbocycle), each of which is optionally substituted with C 0-3 alkyl-(3- to 12-membered heterocycle), wherein the C 0-3 alkyl-(3- to 12- membered heterocycle) is optionally substituted by one or more substituents selected from halogen, -NH 2 , -NHCH 3 , -N(CH 3 ) 2 , -NHCH 2 CH 3 , -CH 2 CH 2 N(CH 3 ) 2 , -CH 3 , and -CF 3 .
  • A is selected from , and
  • R A is selected from:
  • A is selected from and
  • B is selected from and ;
  • R A is selected from:
  • the present disclosure provides a compound of Formula (I-G): or a pharmaceutically acceptable salt thereof, wherein:
  • A is a 9- or 10-membered bicyclic heteroaryl group selected from: , and
  • R A is selected from:
  • R B is -CH 3 ;
  • R 2 and R 3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R 1 ;
  • A is a 9- or 10-membered bicyclic heteroaryl group selected from:
  • R A is selected from:
  • R B is -CH 3 ;
  • A is a 9- or 10-membered bicyclic heteroaryl group selected from:
  • R A is selected from:
  • R B is independently selected at each occurrence from halogen
  • R 2 and R 3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R 1 ;
  • a compound of Formula (I) is provided as a substantially pure stereoisomer.
  • the stereoisomer is provided in at least 80% enantiomeric excess, such as at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or at least 99.9% enantiomeric excess.
  • the present disclosure provides a soft ALK5 inhibitor.
  • the term “soft drug” or “soft ALK5 inhibitor” refers to a biologically active compound that is converted upon entering the systemic circulation into a predictable metabolite that exhibits reduced biological activity relative to the parent compound.
  • a soft drug in some embodiments, exerts its desired therapeutic effect locally at the target organ or tissue, then is rapidly converted to a predictable metabolite designed to be less active than the parent soft drug upon entering the systemic circulation, thus reducing systemic exposure to the biologically active compound. Accordingly, soft drugs have a lower potential for undesired side effects relative to non-soft drug compounds having comparable biological activity.
  • a soft drag of the present disclosure exhibits good stability at the intended site of action (e.g., the lung), is rapidly metabolized upon entering systemic circulation, and displays more functional activity than the corresponding metabolite.
  • a soft drug provided herein exhibits an ALK5 pK i of greater than or equal to 9, while the corresponding soft drag metabolite exhibits an ALK5 pK i of 9 or less, such as 8 or less (assessed according to the assay provided in Example 21).
  • the difference in pK; of the soft drug and the corresponding soft drug metabolite is at least 0.5, such as at least 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or at least 2.0.
  • a soft drag provided herein exhibits a BEAS2B pIC 50 of greater than or equal to 7, while the corresponding soft drug metabolite exhibits a BEAS2B pIC 50 of 6 or less (assessed according to the assay provided in Example 22).
  • the difference in pIC 50 of the soft drug and the corresponding soft drug metabolite is at least 1.0, such as at least 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or at least 2.0.
  • the soft drug and corresponding soft drug metabolite exhibit similar ALK5 pK i values, but the soft drag is more active in cells (e.g., the soft drag exhibits a BEAS2B pIC 50 of at least 1.0 greater than the soft drug metabolite).
  • the present disclosure provides a soft ALK5 inhibitor comprising an ester.
  • the ester inhibits ALK5 activity, while the corresponding carboxylic acid of the ester exhibits reduced ALK5 inhibitory activity.
  • the difference in ALK5 pK i of the ester and corresponding acid may be at least 1.0.
  • a soft drag ester of the present disclosure is administered, to the lung, for example, by inhalation, and inhibits the activity of ALK5 in the lung. However, upon exiting the lung, the ester may be readily hydrolyzed to the corresponding carboxylic acid, thus reducing systemic exposure to the ester.
  • the present disclosure provides a conjugate comprising a compound disclosed herein linked, e.g., covalently linked, either directly or through a linker to an antibody construct or targeting moiety, thereby forming a conjugate.
  • the linker may be a non-cleavable linker or a cleavable linker.
  • a conjugate may be represented by the formula: wherein A' is an antibody construct or targeting moiety; L 1 is a linker; D' is a compound or salt disclosed herein, such as a compound of Formula (I); and p is an integer from 1 to 20. In some embodiments, p is an integer from 1 to 10, such as from 1 to 8, 2 to 8, 1 to 6, 3 to 5, or from 1 to 3.
  • a conjugate is represented by the formula: wherein A' is an antibody construct or targeting moiety; D' is a compound or salt disclosed herein, such as a compound of Formula (I); and p is an integer from 1 to 20. In some embodiments, p is an integer from 1 to 10, such as from 1 to 8, 2 to 8, 1 to 6, 3 to 5, or from 1 to 3.
  • a compound or salt of the present disclosure such as a compound of Formula (I) may be attached to A' via a linker, L 1 , or directly attached to A' without an intermediate linker.
  • the compound or salt is covalently attached to an A' or L 1 . It will be understood by the skilled person that not all compounds of the present disclosure are meant to be attached to L 1 or A', only those that have suitable attachment sites. A compound or salt disclosed herein that does not have a suitable attachment site may be modified to introduce an attachment site.
  • L 1 or D' is bound to A' via a terminus of an amino acid sequence or via a side chain of an amino acid, such as the side chain of lysine, serine, threonine, cysteine, tyrosine, aspartic acid, glutamine, a non-natural amino acid residue, or glutamic acid residue.
  • L 1 or D' is bound to A' via one or more glycans or short peptide tags of four to six amino acids.
  • L 1 or D' may be conjugated to A' via any suitable functional group, such as a thiol, an amine, an amide, an alcohol, a ketone, a carboxylic acid, or an ester.
  • a linker may be attached to a compound or salt of the present disclosure at any available position.
  • a compound of Formula (I) may comprise a linker L 1 to A' in place of R A or through substituent R A :
  • linker L 1 may be covalently bound to any suitable atom for attachment, such as a substitutable nitrogen, carbon, sulfur, phosphorous or oxygen of a compound.
  • L 1 may be a cleavable or non- cleavable linker.
  • the linker may further be bound to A'.
  • L 1 does not affect the binding of the active portions of the conjugate to the binding target(s).
  • Covalent linkages may be formed by reaction between a functional group on the linker with a functional group on the compound, and by reaction between a functional group on the linker with a functional group on A'.
  • linker includes (i) unattached forms of the linker comprising a functional group capable of covalently attaching the linker to a compound disclosed herein and a functional group capable of covalently attaching the linker to an antibody construct or targeting moiety; (ii) partially attached forms of the linker bound to a compound disclosed herein, wherei n the linker comprises a functional group capable of covalently attachi ng the linker to an antibody construct or targeting moiety; (iii) partially attached forms of the linker bound to an antibody construct or targeting moiety, wherein the linker comprises a functional group capable of covalently attaching the linker to a compound disclosed herein; and (iv) fully attached forms of the linker bound to both an antibody construct or targeting moiety and a compound disclosed herein.
  • Linker L 1 may be short, flexible, rigid, cleavable (e.g., by a lysosomal enzyme), non- cleav able, hydrophilic, or hydrophobic.
  • a linker may contain segments having different characteristics, such as flexible segments and rigid segments.
  • a linker may be chemically stable to extracellular environments, for example, in the bloodstream, or may include moieties that are not stable or are selectively stable.
  • a linker comprises a moiety that is selectively cleaved, for example, selectively cleaved in cells, a particular organ, or in plasma.
  • a linker may be sensitive to enzymes, such as proteases.
  • a linker may be insensitive to intracellular processes or proteases.
  • a linker may be acid-labile, protease-sensitive or photolabile.
  • a linker comprises a peptide, succinimide, maleimide, polyethylene glycol, alkylene, alkenylene, alkynyiene, disulfide, hydrazone, polyether, polyester, polyamide, arninobenzyl -carbamate, or a combination thereof.
  • the present disclosure provides a compound of Formula (I), or a compound disclosed in Table 1, wherein the compound is covalently bound to A', optionally via linker L 1 .
  • the antibody construct is an antibody.
  • the present disclosure provides a compound of Formula (I), or a compound disclosed in Table 1, wherein the compound is covalently bound to a linker, L 1 , to form a compound-linker.
  • A' or L 1 may be covalently attached to any position of the compound, valence permitting.
  • a linker L 1 disclosed herein may comprise from about 10 to about 500 atoms, such as 10 to 400 atoms, 10 to 300 atoms, 30 to 400 atoms, or 30 to 300 atoms.
  • the targets of the antibody, antibody construct, or targeting moiety may depend on the desired therapeutic applications of the conjugate.
  • the targets are molecules present on the surfaces of cells into which it is desirable to deliver an ALK5 inhibitor, such as T cells, and the antibodies, in some embodiments, internalize upon binding to the target.
  • an ALK5 inhibitor such as T cells
  • the conjugates are intended to stimulate the immune system by reducing TGF- ⁇ activity
  • the delivery of ALK5 inhibitors to T cells can activate CD4 + and/or CD8 + T cell activity and inhibit regulatory T cell activity, both of which contribute to immune tolerance of tumors.
  • antibodies, antibody constructs, or targeting moieties (A') that bind to T cell surface molecules in the conjugates of the present disclosure are useful for the treatment of various cancers, such as those described herein below.
  • A' binds to CD4 + T cells, CD8 + T cells, T REG cells, or any combination thereof.
  • A' binds to a pan T cell surface molecule, such as CD1, CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD25, CD28, CD70, CD71 , CD103, CD184, Tim3, LAG3, CTLA4, or
  • PD1 examples include OKT6, OKT11, OKT3, OKT4, OKT8, 7D4, OKT9, CD28.2, UCHT1, M290, FR70, pembrolizumab, nivolumab, cemiplimab, and dostarlimab.
  • An antibody, antibody construct, or targeting moiety disclosed herein may comprise an antigen bindi ng domain that specifically binds to a tumor antigen or antigen associated with the pathogenesis of fibrosis.
  • the antigen binding domain specifically binds to an antigen on a T cell, a B cell, a stellate cell, an endothelial cell, a tumor cell, an APC, a fibroblast cell, a fibrocyte cell, or a cell associated with the pathogenesis of fibrosis.
  • the antigen binding domain targets CTLA4, PD-1, OX40, LAG-3, GITR, GARP, CD25, CD27, PD-LL TNFR.2, ICOS, 41BB, CD70, CD73, CD38 or VTCN1.
  • the antigen binding domain targets PDGFRp, integrin ⁇ v ⁇ l, integrin ⁇ v ⁇ 3, integrin ⁇ v ⁇ 6, ⁇ v ⁇ 8, endosialin, FAP, ADAM12, LRRC15, MMP14, PDPN, CDH11, F2RL2, ASGR1, or ASGR2.
  • reaction times and conditions are intended to be approximate, e.g. taking place at about atmospheric pressure within a temperature range of about -10 °C to about 110 °C over a period of about 1 to about 24 hours; reactions left to run overnight average a period of about 16 hours.
  • a compound of Formula Id or Formula lg may be prepared according to Scheme 1.
  • ethanone la can be reacted with DMF-DMA at elevated temperatures to give intermediate lb, which can be reacted with hydrazine monohydrate to provide pyrazoie 1c.
  • le may be subjected to one or more coupling reactions, and optionally one or more protecting group manipulations, to provide a pyrazoie of Formula Id.
  • oxidation of la can provide dione le, which can be converted to imidazole If in the presence of urotropine and ammonium acetate.
  • If may be subjected to one or more coupling reactions, and optionally one or more protecting group manipulations, to provide an imidazole of Formula lg.
  • a compound of Formula 2c may be prepared according to Scheme 2.
  • alkyne 2a can be converted to triazole 2b in the presence of a suitable azide, such as TMS-N 3 .
  • 2b may be subjected to one or more coupling reactions, and optionally one or more protecting group manipulations, to provide a triazole of Formula 2c.
  • a compound of Formula 3c, Formula 3e or Formula 3h may be prepared according to Scheme 3.
  • bromide 3a can be subjected to a C-N coupling reaction — optionally a Pd-catalyzed coupling reaction such as a Buchwald -Hartwig amination — -with an acyclic primary or secondary amine (3b) or a cyclic secondary amine (3d) to provide an amine of Formula 3c or Formula 3e, respectively.
  • R A substituent may proceed via a Suzuki reaction, either in one step using boronic ester 3f or in two steps — wherein heteroaryl bromide 3a is first converted to the corresponding boronic ester, their coupled to a suitable halide (e.g., R A X) — to give a compound of Formula 311.
  • a suitable halide e.g., R A X
  • a compound of Formula 4j may be prepared according to Scheme 4.
  • bromide 4a can be subjected to a coupling reaction— -optionally a Suzuki -Miyaura coupling— -with boronic ester 4b to provide 4c.
  • Halogenation of 4c can provide heteroaryl iodide 4d, which may be converted to boronic ester 4f and coupled to a suitable heteroaryl halide (e.g., 4g) to afford 4h.
  • 4h may be subjected to one or more coupling reactions, and optionally one or more protecting group manipulations, to provide a compound of Formula 4j.
  • a compound of the present disclosure for example, a compound, of a formula given in Table 1, is synthesized according to one of the general routes outlined in Schemes 1-4, Examples 1- 20, or by methods generally known in the art.
  • exemplary compounds may include, but are not limited to, a compound or salt thereof selected from Table 1.
  • exemplary compounds may include, but are not limited to, a compound, a stereoisomer thereof, or salt thereof selected from Table 1.
  • exemplary compounds may include, but are not limited to, a compound, a stereoisomer thereof, or salt thereof selected, from Table 1a.
  • the present disclosure provides a method of inhibiting TGFp signaling, comprising contacting a cell with an effective amount of a compound disclosed herein, such as a compound of Formula (I).
  • a method of inhibiting ALK5 comprising contacting ALK5 with an effective amount of a compound disclosed herein. Inhibition of ALK5 or TGFp signaling can be assessed by a variety of methods known in the art.
  • Non-limiting examples include a showing of (a) a decrease in kinase activity of ALK5; (b) a decrease in binding affinity between the TGF ⁇ /TGF ⁇ -RII complex and ALK5; (c) a decrease in the levels of phosphorylated intracellular signaling molecules downstream in the TGFp signaling pathway, such as a decrease in pSMAD2 or pSMAD3 levels; (d) a decrease in binding of ALK5 to downstream signaling molecules, such as SMAD2 and SMAD3; and/or (e) an increase in ATP levels or a decrease in ADP levels. Kits and commercially available assays can be utilized for determining one or more of the above.
  • the present disclosure provides a method of treating an A L K5- mediated disease or condition in a subject, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein.
  • the disease or condition is selected from fibrosis and cancer.
  • the disease or condition is pulmonary fibrosis, such as idiopathic pulmonary fibrosis or virus-induced fibrosis.
  • the disease or condition is intestinal fibrosis.
  • the disease or condition is alopecia.
  • the disease is a neurodegenerative disease, such as Alzheimer’s disease.
  • the present disclosure provides a method of reversing symptoms of aging. For example, the method may enhance neurogenesis, reduce neuroinflammation, improve cognitive performance, regenerate liver tissue, and reduce pl6 levels.
  • the present disclosure provides a method of treating fibrosis, comprising administering to a patient an effective amount of a compound disclosed herein.
  • the fibrosis is mediated by ALK5.
  • the fibrosis is selected from systemic sclerosis, systemic fibrosis, organ-specific fibrosis, kidney fibrosis, pulmonary fibrosis, liver fibrosis, portal vein fibrosis, skin fibrosis, bladder fibrosis, intestinal fibrosis, peritoneal fibrosis, myelofibrosis, oral submucous fibrosis, and retinal fibrosis.
  • the fibrosis is pulmonary fibrosis, such as idiopathic pulmonary fibrosis (IPF), familial pulmonary fibrosis (FPF), interstitial lung fibrosis, fibrosis associated with asthma, fibrosis associated with chronic obstructive pulmonary disease (COPD), silica-induced fibrosis, asbestos- induced fibrosis or chemotherapy-induced lung fibrosis.
  • the fibrosis is idiopathic pulmonary fibrosis (IPF).
  • the fibrosis is TGF- ⁇ -mediated pulmonary fibrosis.
  • the patient has been diagnosed with acute respiratory distress syndrome (ARDS).
  • ARDS acute respiratory distress syndrome
  • the fibrosis is acute fibrosis.
  • the fibrosis is chronic fibrosis.
  • the present disclosure provides a method of treating pulmonary fibrosis induced by a viral infection, comprising administering to a patient an effective amount of a compound disclosed herein.
  • the pulmonary fibrosis may be induced by an erythrovirus, a dependovirus, a papillomavirus, a polyomavirus, a mastadenovirus, an alphaherpesvirinae, a varicellovirus, a gammaherpesvirinae, a betaherpesvirinae, a roseolovirus, an orthopoxvirus, a parapoxvirus, a moliuscipoxvirus, an orthohepadnavirus, an enterovirus, a rhinovirus, a hepatovirus, an aphthovirus, a calicivirus, an astrovirus, an alpha virus, a rubivirus, a flavi virus, a Hepatitis C vims, a reovirus, an orbivirus, a rotavirus, an influenzavirus A, an influenzavirus B, an influenzavirus C, a paramyxovirus, a morbilli virus, a rub
  • the fibrosis is virus-induced fibrosis, such as virus-induced pulmonary fibrosis. In some embodiments, the fibrosis is selected from EBV-induced pulmonary fibrosis, CMV-induced pulmonary fibrosis, herpesvirus- induced pulmonary fibrosis and coronavirus-induced pulmonary fibrosis.
  • the fibrosis is selected from EBV-induced pulmonary fibrosis, CMV-induced pulmonary fibrosis, HHV-6-induced pulmonary fibrosis, HHV-7-induced pulmonary fibrosis, HHV-8-induced pulmonary fibrosis, H5N1 virus- induced pulmonary fibrosis, SARS-CoV-induced pulmonary fibrosis, MERS-CoV-induced pulmonary fibrosis and SARS-CoV-2-induced pulmonary fibrosis.
  • the pulmonary fibrosis is coronavirus-induced pulmonary fibrosis.
  • the pulmonary fibrosis is SARS-CoV-2- induced pulmonary fibrosis.
  • the pulmonary fibrosis is COVID-19-induced pulmonary fibrosis.
  • the present disclosure provides a method of treating acute lung injury (ALI), comprising administering to a patient an effective amount of a compound disclosed herein.
  • the present disclosure provides a method of treating acute respiratory distress syndrome (ARDS), comprising administering to a patient an effective amount of a compound disclosed herein.
  • the ARDS may be in the early acute injury phase or the fibroproliferative phase.
  • the ARDS is fibroproliferative ARDS.
  • the present disclosure provides a method of treating fibrosis resulting from. ARDS, comprising administering to a patient an effective amount of a compound disclosed herein.
  • the fibrosis resulting from ARDS may be pulmonary fibrosis.
  • the present disclosure provides a method of treating fibrosis resulting from ALI, comprising administering to a patient an effective amount of a compound disclosed herein.
  • the fibrosis resulting from ALI may be pulmonary fibrosis.
  • the present disclosure provides a method of treating intestinal fibrosis, comprising administering to a patient an effective amount of a compound disclosed herein.
  • the intestinal fibrosis is mediated by ALK5.
  • the compound is administered in an amount effective to delay progression of, reduce the i ncidence of, or reduce the degree of one or more characteristics associated with intestinal fibrosis.
  • the compound is admi nistered, either in a single dose or over multiple doses, in an amount effective to reverse established fibrosis.
  • the present disclosure provides a method of treating cancer, comprising administering to a patient an effective amount of a compound disclosed herein.
  • the cancer is mediated by ALK5.
  • the cancer is selected from breast cancer, coion cancer, prostate cancer, lung cancer, hepatocellular carcinoma, glioblastoma, melanoma, and pancreatic cancer.
  • the cancer is lung cancer, such as non-small cell lung cancer.
  • the present disclosure provides a method of treating cancer, such as non-small cell lung cancer, comprising administering to a patient an effective amount of a compound disclosed herein and an immunotherapeutic agent.
  • the cancer is stage III non-small cell lung cancer.
  • the method further comprises administering radiation to the patient.
  • the immunotherapeutic agent is a PD- 1 inhibitor or a CTLA-4 inhibitor.
  • the immunotherapeutic agent is selected from atezolizumab, a velum ab, nivolumab, pembrolizumab, durvaiumab, BGB-A317, tremelimumab, and ipilimumab.
  • the immunotherapeutic agent is selected from pembrolizumab and durvaiumab.
  • the compounds described herein, including compounds of Formula (I), are ALK5 inhibitors that limit the activity of TGFp.
  • TGFP is one of several factors involved in the initiation and development of fibrotic diseases throughout the body.
  • the compounds of the disclosure are expected to be useful for the treatment, prevention and/or reduction of fibrosis in a patient by administering a therapeutically effective amount of a compound disclosed herein.
  • the compound By inhibiting ALK5, the compound is expected to potentiate the formation of fibrosis in areas of the body that suffer from excessive deposition of the extracellular matrix. Those areas are described below.
  • SSc Systemic sclerosis
  • TGF- ⁇ Transforming growth factor p
  • the present disclosure provides a method of treating SSc, comprising administering to a subject an effective amount of a compound disclosed herein.
  • Multifocal fibrosclerosis (MF) and. idiopathic multifocal fibrosclerosis (IMF) are disorders characterized by fibrous lesions at varying sites and include retroperitoneal fibrosis, mediastinal fibrosis and Riedel’s thyroiditis. Both multifocal fibrosclerosis and idiopathic multifocal fibrosclerosis are considered to be an outcome of IgG-; -associated fibrosis/disease and TGF- ⁇ is believed to be one factor involved in the initiation and development of fibrosis (Pardali, E., et al., Int. J. Mol. Set., 18, 2157, pp. 1-22).
  • the present disclosure provides a method of treating multifocal fibrosclerosis or idiopathic multifocal fibrosclerosis, comprising administering to a subject an effective amount of a compound disclosed herein.
  • the present disclosure provides a method of treating nephrogenic systemic fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein.
  • Nephrogenic systemic fibrosis is a rare disease occurring mainly in people with advanced kidney failure with or without dialysis.
  • Kelly et al. J. Am. Acad. Dermatol., 2008, 58, 6, pp. 1025- 1030
  • TGF- ⁇ as well as Smad 2/3
  • Sclerodermatous graft-versus-host disease is a prevalent complication of allogeneic hematopoietic stem cell graft appearing two to three months after allogeneic bone marrow transplantation.
  • the present disclosure provides a method of treating sclerodermatous GVHD, comprising administering to a subject an effective amount of a compound disclosed herein.
  • Cardiac fibrosis refers to the abnormal thickening of heart valves due to the abnormal proliferation of cardiac fibroblasts resulting in excess deposition of ECM in heart muscle. Fibroblasts secrete collagen, which serves as structural support for the heart. However, when collagen is excessively secreted in the heart, wall and valve thickening can result in tissue build -up on the tricuspid and pulmonary valves. This in turn causes loss of flexibility and ultimately valvular dysfunction leading to heart failure.
  • a specific type of cardiac fibrosis is hypertension-associated cardiac fibrosis as described by J. Diez (J. Clin. Hypertens. , 2007, July 9(7), pp. 546-550).
  • the present disclosure provides a method of treating various forms of cardiac fibrosis, such as hypertension-associated cardiac fibrosis, post -myocardial infarction or Chagas disease-induced myocardial fibrosis, comprising admi ddlingring to a subject an effective amount of a compound disclosed herein.
  • Renal fibrosis encompasses a variety of disorders associated with the aberrant expression and activity of TGF- ⁇ , including, but not limited to, diabetic and hypertensive nephropathy, urinary tract obstruction- induced kidney fibrosis, inflammatory/autoimmune-induced kidney fibrosis, aristolochic acid nephropathy, progressive kidney fibrosis, and. polysystic kidney disease.
  • fibrosis involves an excess accumulation of the ECM, which in turn causes loss of function when normal tissue is replaced with scar tissue (Wynn, T.A., J Clin Invest., 2007, 117, pp. 524-529).
  • the present disclosure provides a method of treating renal fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein.
  • a fibrotic disease that has been particularly difficult to treat is idiopathic pulmonary fibrosis (IPF).
  • IPF is a chronic, progressive and fatal fibrotic lung disease with survival only improved by lung transplantation.
  • Current oral therapies such as nintedanib and pirfenidone have been shown to slow the progression of the disease, but have adverse effects that lead to discontinuation and lack of compliance by the patient. Although there are other therapies in development targeting various pathways, an unmet need remains for patients with IPF.
  • ALK5 is an important and known component in the fibrotic disease pathway, the efficacy of ALK5 inhibitors in IPF have not been realized due to systemic adverse effects, especially in the heart.
  • one of the goals of this disclosure is to develop ALK5 inhibitors with high lung selectivity and rapid clearance.
  • One exemplary embodiment of this disclosure is to treat patients with idiopathic pulmonary fibrosis with a compound described herein, for example, by once or twice daily administration of inhalable ALK5 inhibitor having minimal systemic exposure.
  • the inhaled ALK5 inhibitor may be administered as a monotherapy or co- dosed with other orally available IPF therapies.
  • the present disclosure provides a method of treating idiopathic pulmonary fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein.
  • the compound is administered by inhalation.
  • Familial pulmonary fibrosis is a hereditary disease where two or more family members have confirmed IPF.
  • the present disclosure provides a method of treating familial pulmonary fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein.
  • Pulmonary fibrosis is a typical clinical feature associated with viral infection, such as SARS and COVID-19.
  • SARS-mediated TGF- ⁇ signaling has been shown to promote fibrosis and block apoptosis of SARS-CoV-infected host cells (Zhao, X. et al. J. Biol Chem., 2008, 283(6), pp. 3272-3280). Increased TGF- p expression was similarly observed in patients infected with SARS-CoV-2, ultimately leading to the development of pulmonary fibrosis.
  • TGF ⁇ P signaling mediated by SARS-CoV-2 can promote fibroblast proliferation and myofibroblast differentiation and block host cell apoptosis. (Xiong, Y.
  • the present disclosure provides a method of treating pulmonary fibrosis induced by a viral infection, comprising administering to a subject an effective amount of a compound disclosed herein.
  • the pulmonary fibrosis is induced by SARS-CoV or SARS-CoV-2.
  • the compound is administered by inhalation.
  • PH pulmonary hypertension
  • Pulmonary hypertension is a progressive disease characterized by high blood pressure in the lungs.
  • the World Health Organization (WHO) has defined five classifications of PH (WHO Group I: Pulmonary arterial hypertension (PAH); WHO Group II: Pulmonary hypertension due to left heart disease; WHO Group III: Pulmonary hypertension due to lung disease and/or hypoxia; WHO Group IV: Chronic thromboembolic pulmonary hypertension (CTEPH); and WHO Group V: Pulmonary hypertension with unclear multifactorial mechanisms).
  • TGF- ⁇ signaling has been implicated in the pathogenesis of PH.
  • inhibition of ALK5 in a monocrotaline (MCT) model of severe PH was shown to attenuate the development of PH and reduce pulmonary vascular remodeling in a dose-dependent manner, namely by reducing RV systolic pressure, reducing RV diastolic pressure, increasing cardiac output and reducing RV hypertrophy (Zaiman, A. L.; et al., Am. J. Respir. Crit. Care Med., 2008, 177, pp. 896-905).
  • Compounds of the present disclosure are expected to inhibit TGF- ⁇ signaling in lung tissue and prevent, halt, slow, or reverse the progression of PH, particularly in WHO Group III PH.
  • the present disclosure provides a method of treating pulmonary hypertension, comprising administering to a subject an effective amount of a compound disclosed herein.
  • the pulmonary hypertension may be WHO Group III pulmonary hypertension, such as pulmonary fibrosis-related pulmonary hypertension (PH-PF) or interstitial lung disease-related pulmonary hypertension (PH-ILD).
  • the compound is administered by inhalation.
  • interstitial lung diseases include, but are not limited to, (1) interstitial pneumonia caused by bacteria, viruses, or fungi; (2) nonspecific interstitial pneumonitis usually associated with autoimmune conditions such as rheumatoid arthritis or scleroderma; (3) hypersensitivity pneumonitis caused by inhalation of dust, mold, or other irritants; (4) cryptogenic organizing pneumonia; (5) acute interstitial pneumonitis; (6) desquamative interstitial pneumonitis; (7) sarcoidosis; (8) drag-induced interstitial lung disease; and (9) progressive fibrosing interstitial lung disease (PF-ILD).
  • the present disclosure provides a method of treating an interstitial lung disease, comprising administering to a subject an effective amount of a compound disclosed herein.
  • TGF transforming growth factor
  • activin-A transforming growth factor-beta(l) and activin-A have been implicated in airway remodeling in asthma (Kariyawasam, H.H., J Allergy Clin Immunol., 2009, September, 12.4(3), pp. 454-462).
  • the present disclosure provides a method of treating asthma, comprising administering to a subject an effective amount of a compound disclosed herein.
  • COPD chronic obstructive pulmonary disease
  • Fibrosis associated with emphysema is known and research has demonstrated TGF- ⁇ 1 involvement in chronic sinus disease, pulmonary fibrosis, asthma, and COPD (Yang, Y.C., et al., Allergy, 2012, 67, pp. 1193 -1202).
  • the present disclosure provides a method of treating COPD, comprising administering to a subject an effective amount of a compound disclosed herein.
  • Other types of lung injury that result in fibrosis include silica-induced pneumoconiosis (silicosis), asbestos-induced pulmonary fibrosis (asbestosis), and chemotherapeutic agent-induced pulmonary fibrosis.
  • the present disclosure provides a method of treating injury-induced fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein.
  • the present disclosure provides a method of treating liver fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein.
  • Fibrosis develops in the liver when it is repeatedly or continuously damaged, for example, in patients with chronic hepatitis.
  • TGF- ⁇ signaling participates in all stages of disease progression, from, initial liver injury through inflammation and fibrosis, to cirrhosis and cancer (Fabregat, I., et al., The FEES J., 2016, 283(12), pp. 2219-2232).
  • a related condition involves fibrosis resulting from idiopathic non-cirrhotic portal hypertension (INCPH).
  • IOCPH idiopathic non-cirrhotic portal hypertension
  • This disease is of uncertain etiology characterized by periportal fibrosis and involvement of small and medium branches of the portal vein.
  • small portal veins and skin findings are similar between patients with scleroderma and INCPH (Nakanuma, Y., Hepatol. Res., 2009, 39, pp. 1023-1031).
  • TGF- ⁇ Transforming growth factor-p
  • connective tissue growth factor which are fibrosis-related and vascular endothelial growth factors, respectively
  • TGF- ⁇ Transforming growth factor-p
  • connective tissue growth factor connective tissue growth factor
  • EndMT endothelial mesenchymal transition
  • Kitao et al. based on these findings (Kitao, A., et al., Am. J. Pathol., 2009, 175, pp. 616-626).
  • the increase of TGF- ⁇ in sera may act as a potent inducer of EndMT.
  • the present disclosure provides a method of treating INCPH, comprising administering to a subject an effective amount of a compound disclosed herein.
  • liver fibrosis include alcoholic and non-alcoholic liver fibrosis, hepatitis C -induced liver fibrosis, primary biliary cirrhosis or cholangitis, and parasite-induced liver fibrosis (schistosomiasis).
  • the present disclosure provides a method of treating alcoholic liver fibrosis, nonalcoholic liver fibrosis, hepatitis C -induced liver fibrosis, primary biliary cirrhosis, primary biliary cholangitis, or parasite- induced liver fibrosis (schistosomiasis), comprising administering to a subject an effective amount of a compound disclosed herein.
  • PBC Primary biliary cholangitis
  • PSC primary sclerosing cholangitis
  • Liver biopsies of patients with PBC or PSC typically reveal inflammation and fibrosis.
  • the present disclosure provides a method, of treating primary biliary cholangitis or primary sclerosing cholangitis, comprising administering to a subject an effect amount of a compound described herein.
  • the present disclosure provides a method of treating liver fibrosis, optionally in a subject that suffers from PBC or PSC, comprising administering to the subject an effective amount of a compound described herein.
  • Fibrotic skin conditions include, but are not limited to, hypertrophic scarring, keloids, and localized or systemic sclerosis (scleroderma).
  • TGF- ⁇ is a potent stimulus of connective tissue accumulation and has been implicated in the pathogenesis of scleroderma and other fibrotic disorders (Lakos, G., et al., Am. J. Pathol., 2004, 165(1), pp. 203-217). Lakos et.al.
  • the present disclosure provides a method of treating skin fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein.
  • Intestinal fibrosis is a common complication of inflammatory bowel disease (IBD) and is a serious clinical problem.
  • TGF- ⁇ has been implicated as a major driving factor of intestinal fibrosis.
  • TGF- pl signaling contributes to stricture formation in fibrostenotic Crohn’s disease by inducing insulin-like growth factor I (IGF-I) and mechano-growth factor (MGF) production in intestinal smooth muscle.
  • IGF-I insulin-like growth factor I
  • MMF mechano-growth factor
  • the present disclosure provides a method of treating intestinal fibrosis, comprising administering to a subject an effective amount of a compound described herein, for example, by once or twice daily administration of an oral ALK5 inhibitor having minimal systemic exposure.
  • the subject suffers from inflammatory bowel disease, such as Crohn’s disease or colitis.
  • the degree of therapeutic efficacy may be with respect to a starting condition of the subject (e.g., a baseline Mayo score, baseline Lichtiger score, or severity or incidence of one or more symptoms), or with respect to a reference population (e.g., an untreated population, or a population treated with a different agent).
  • Severity of intestinal fibrosis may be assessed using any suitable method, such as delayed enhancement MRI, ultrasound elastography, shear wave elastography, magnetization MRI, or by the direct detection of macromolecules such as collagen.
  • treatment with a compound of the present disclosure reduces the severity of the fibrosis, such as from severe fibrosis to moderate or mild fibrosis.
  • the treatment increases intestinal tissue elasticity, reduces tissue stiffness, and/or reduces collagen levels. In some embodiments, the treatment prevents myofibroblast accumulation, inhibits expression of pro-fibrotic factors, and/or inhibits accumulation of fibrotic tissue.
  • organ-specific fibrosis or fibrotic diseases involving the TGF- ⁇ pathway include, but are not limited to, radiation-induced fibrosis (various organs), bladder fibrosis, intestinal fibrosis, peritoneal sclerosis, diffuse fasciitis, Dupuytren’s disease, myelofibrosis, oral submucous fibrosis, and retinal fibrosis.
  • the present disclosure provides a method of treating radiation-induced fibrosis, bladder fibrosis, intestinal fibrosis, peritoneal sclerosis, diffuse fasciitis, Dupuytren’s disease, myelofibrosis, oral submucous fibrosis, or retinal fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein.
  • the compounds described herein may also be used to treat patients systemically.
  • Diseases that may be treated systemically include, for example, oncologic diseases such as glioblastoma, pancreatic cancer and hepatocellular carcinoma, breast cancer metastasized to lungs, non-small cell lung cancer, small cell lung cancer, cystic fibrosis, and metastasis of other forms of primary cancer subtypes. Some of the forgoing diseases may also be treated locally as well.
  • Alopecia includes alopecia totalis, alopecia uni versalis, androgenetic alopecia, alopecia areata, diffuse alopecia, postpartum alopecia, and traction alopecia.
  • the present disclosure provides a method of reversing one or more symptoms of aging, comprising administering to a subject an ALK5 inhibitor.
  • the method may further comprise administering an activator of the MAPK pathway, such as oxytocin.
  • the method may be effective in one or more of enhancing neurogenesis in the hippocampus, reducing neuroinflammation, improving cognitive ability, reducing liver adiposity, reducing liver fibrosis, and decreasing the number of p16 + cells.
  • a method described herein increases stem cell activity .
  • the increase in stem cell activity may allow the subject to generate new muscle fibers and/or to form new neurons in the hippocampus.
  • Treatment with an ALK5 inhibitor, such as a compound described herein may prevent or slow the onset of age-related diseases, such as Alzheimer’s disease, (see Mehdipour, M. et al. Aging 2018, 10, 5628-5645).
  • the present disclosure provides a pharmaceutical composition.
  • the pharmaceutical composition may comprise a compound disclosed herein, such as a compound of Formula (I), and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition is formulated for oral administration.
  • the pharmaceutical composition is formulated for inhalation.
  • the pharmaceutical composition comprises a compound disclosed herein and an additional therapeutic agent. Non-limiting examples of such therapeutic agents are described herein below.
  • Pharmaceutical compositions typically include at least one pharmaceutically acceptable carrier, diluent or excipient and at least one compound of Formula (I), or a compound provided in Table 1 — described herein as the active agent.
  • the active agent may be provided in any form suitable for the particular mode of administration, such as a free base, a free acid, or a pharmaceutically acceptable salt.
  • the methods and pharmaceutical compositions of the present disclosure include the use of N-oxides, crystalline forms (e.g., polymorphs), as well as metabolites of these compounds having similar activity. All tautomers of the compounds described herein are included within the scope of the present disclosure. Additionally, the compounds described herein encompass unsolvated, as well as solvated, forms with pharmaceutically acceptable solvents such as water, ethanol and the like.
  • Suitable routes of administration include, but tire not limited to, oral, intravenous, rectal, vaginal, aerosol, pulmonary, nasal, transmucosal, topical, transdermal, otic, ocular, and parenteral modes of administration.
  • parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections.
  • a compound described herein is administered in a local rather than systemic manner, for example, via injection of the compound directly into an organ, often in a depot preparation or sustained, release formulation.
  • a long acting formulation is administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection.
  • a compound described herein is provided in the form of a rapid release formulation, an extended release formulation, or an intermediate release formulation.
  • a compound described herein is provided in the form of a nebulized formulation.
  • a compound described herein is administered locally to the lungs by inhalation.
  • Compounds of the present disclosure are effective over a wide dosage range. For example, in the treatment of adult humans, dosages from 0.01 to 1000 mg, 0.5 to 100 mg, 1 to 50 mg, or from 5 to 40 mg per day may be administered to a subject in need thereof. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
  • a compound of the present disclosure may be administered in a single dose.
  • a compound of the disclosure is administered in multiple doses, such as about once, twice, three times, four times, five times, six times, or more than six times per day.
  • dosing is about once a month, once every two weeks, once a week, or once every other day.
  • a compound of the disclosure and an additional therapeutic agent are administered together about once per day to about 6 times per day.
  • the administration continues for more than about 6, 1.0, 14, 28 days, two months, six months, or more than about one year.
  • a dosing schedule is maintained as long as necessary.
  • a compound of the present disclosure may be administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
  • compositions of the present disclosure typically contain a therapeutically effective amount of a compound of the present disclosure.
  • a pharmaceutical composition may contain more than a therapeutically effective amount, e.g., bulk compositions, or less than a therapeutically effective amount, e.g., individual unit doses designed for coadministration to achieve a therapeutically effective amount.
  • compositions of the present disclosure contain from about 0.01 to about 95% by weight of the active agent; including, for example, from about 0.05 to about 30% by weight; and from about 0.1 % to about 10% by weight of the active agent.
  • any conventional carrier or excipient may be used in the pharmaceutical compositions of the present disclosure.
  • the choice of a particular carrier or excipient, or combinations of carriers or excipients, will depend on the mode of administration being used to treat a particular patient or type of medical condition or disease slate. Additionally, the earners or excipients used in the pharmaceutical compositions of this disclosure may be commercially-available. Conventional formulation techniques are described in Remington: The Science and Practice of Pharmacy, 20th Edition, Lippincott Williams & White, Baltimore, Maryland (2000); and H.C. Ansel et at, Pharmaceutical Dosage Forms and Drag Delivery Systems, 7th Edition, Lippincott Williams & White, Baltimore, Maryland (1999).
  • Representative examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, the following: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, such as microcrystalline cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and.
  • cellulose acetate powdered, tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical compositions.
  • oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil
  • compositions are typically prepared by thoroughly and i ntimately mixing or blending the active agent with a p.barmaceutically-acceptable carrier and one or more optional ingredients. The resulting uniformly blended mixture can then be shaped or loaded into tablets, capsules, pills and the like using conventional procedures and equipment.
  • the pharmaceutical composition is suitable for inhaled administration.
  • Pharmaceutical compositions for inhaled administration are typically in. the form of an aerosol or a powder.
  • Such compositions are generally administered, using inhaler delivery devices, such as a dry powder inhaler (DPI), a metered-dose inhaler (MDI), a nebulizer inhaler, or a similar delivery device.
  • DPI dry powder inhaler
  • MDI metered-dose inhaler
  • nebulizer inhaler or a similar delivery device.
  • the pharmaceutical composition is administered by inhalation using a dry powder inhaler.
  • dry powder inhalers typically administer the pharmaceutical composition as a free- flowing powder that is dispersed, in a patient's air-stream during inspiration.
  • the therapeutic agent is typically formulated with a suitable excipient such as lactose, starch, mannitol, dextrose, polylactic acid (PL A), polylactide-co-glycolide (PLGA) or combinations thereof.
  • the therapeutic agent is micronized and combined with a suitable carrier to form a composition suitable for inhalation.
  • a representative pharmaceutical composition for use in a dry powder inhaler comprises lactose and a micronized form of a compound disclosed herein.
  • a dry powder composition can be made, for example, by combining dry milled lactose with the therapeutic agent and then dry blending the components. The composition is then typically loaded into a dry powder dispenser, or into inhalation cartridges or capsules for use with a dry powder delivery device.
  • Dry powder inhaler delivery devices suitable for administering therapeutic agents by inhalation are described in the art and examples of such devices are commercially available.
  • representative dry powder inhaler delivery devices or products include Aeolizer (Novartis); Airmax (TV AX); ClickHaler (Innovata Biomed); Diskhaler (GlaxoSmithKline); Diskus/Accuhaler (GlaxoSmithKline); Eliipta (GlaxoSmithKline); Easyhaler (Orion Pharma); Eclipse (Aventis); FlowCaps (Hovione); Handihaler (Boehringer Ingelheim); Pulvinal (Chiesi); Rotahaier (GlaxoSmithKline); SkyeHaler/Certihaler (SkyePharma); Twisthaler (Schering-Plough); Turbuhaler (AstraZeneca); Ultrahaler (Aventis); and the like.
  • a pharmaceutical composition of the present disclosure may be administered by inhalation, using a metered -dose inhaler.
  • metered -dose inhalers typically discharge a measured amount of a therapeutic agent using a compressed propellant gas.
  • pharmaceutical compositions administered using a metered-dose inhaler typically comprise a solution or suspension of the therapeutic agent in a liquefied propellant.
  • Any suitable liquefied propellant may be employed, including hydrofluoroalkanes (HFAs), such as 1,1,1,2-tetrafluoroethane (HF A 134a) and 1,1,1,2,3,3,3-heptafluoro-n-propane, (HF A 227); and chlorofluorocarbons, such as CCI3F.
  • HFAs hydrofluoroalkanes
  • HF A 134a 1,1,1,2-tetrafluoroethane
  • HF A 227 1,1,1,2,3,3,3-heptafluoro-n-propane
  • the propellant is a hydrofluoroalkane.
  • the hydrofluoroalkane formulation contains a co-solvent, such as ethanol or pentane, and/or a surfactant, such as sorbitan trioleate, oleic acid, lecithin, and glycerin.
  • a representative pharmaceutical composition for use in a metered-dose inhaler comprises from about 0.01% to about 5% by weight of a compound of the present disclosure; from about 0% to about 2.0%' by weight ethanol; and from about 0% to about 5% by weight surfactant; with the remainder being an HF A propellant.
  • Such compositions are typically prepared by adding chilled or pressurized hydrofluoroalkane to a suitable container containing the therapeutic agent, ethanol (if present) and the surfactant (if present). To prepare a suspension, the therapeutic agent is micronized. and then combined with the propellant. The composition is then loaded into an aerosol canister, which typically forms a portion of a metered-dose inhaler device.
  • Metered-dose inhaler devices suitable for administering therapeutic agents by inhalation are described in the art and examples of such devices are commercially available.
  • representative metered-dose inhaler devices or products include AeroBid Inhaler System (Forest Pharmaceuticals); Atrovent Inhalation Aerosol (Boehringer Ingelheim); Flovent (GlaxoSmithKline); Maxair Inhaler (3M); Proventil Inhaler (Schering); Serevent Inhalation Aerosol (GlaxoSmithKline); and the like.
  • a pharmaceutical composition of the present disclosure may be administered by inhalation using a nebulizer inhaler.
  • nebulizer devices typically produce a stream of high velocity air that causes the pharmaceutical composition to spray as a mist that is carried into the patient's respiratory tract.
  • the therapeutic agent when formulated for use in a nebulizer inhaler, can be dissolved in a suitable carrier to form a solution.
  • the therapeutic agent can be micronized or nanomilled and combined with a suitable carrier to form a suspension.
  • a representative pharmaceutical composition for use in a nebulizer inhaler comprises a solution or suspension comprising from about 0.05 pg/mL to about 20 mg/mL of a compound of the present disclosure and excipients compatible with nebulized formulations.
  • the solution has a pH of about 3 to about 8.
  • Nebulizer devices suitable for administering therapeutic agents by inhalation are described in the art and examples of such devices are commercially available.
  • representative nebulizer devices or products include the Respimat® Softmist lM Inhalaler (Boehringer Ingelheim); the AERx® Pulmonary Delivery System (Aradigm Corp.); the PARI LC Plus® Reusable Nebulizer or PARI eF1ow®rapid Nebulizer System (Pari GmbH); and. the like.
  • a pharmaceutical composition of the present disclosure may be prepared in a dosage form intended for oral administration.
  • Suitable pharmaceutical compositions for oral administration may be in the form of capsules, tablets, pills, lozenges, cachets, dragees, powders, granules; or as a solution or a suspension in an aqueous or non-aqueous liquid: or as an oil-in-water or water-in-oil liquid, emulsion; or as an elixir or syrup; and the like; each containing a predetermined amount of a compound of the present disclosure as an active ingredient.
  • the pharmaceutical compositions of the disclosure will typically comprise the active agent and one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate.
  • such solid dosage forms may also comprise: fillers or extenders, binders, humectants, solution retarding agents, absorption accelerators, wetting agents, absorbents, lubricants, coloring agents, and buffering agents. Release agents, wetting agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the present pharmaceutical compositions.
  • Alternative formulations may include controlled release formulations, liquid dosage forms for oral administration, transdermal patches, and parenteral formulations. Conventional excipients and methods of preparation of such alternative formulations are described, for example, in the reference by Remington, supra. [0194] The following non-limiting examples illustrate representative pharmaceutical compositions of the present disclosure. [0195] Dry Powder Composition
  • a micronized compound of the present disclosure (1 g) is blended with milled lactose (25 g). This blended mixture is then loaded into individual blisters of a peelable bl ister pack in an amount sufficient to provide between about 0.1 mg to about 4 mg of the compound per dose. The contents of the blisters are administered using a dry powder inhaler.
  • a micronized compound of the present disclosure (1 g) is blended with milled lactose (20 g) to form a bulk composition having a weight ratio of compound to milled lactose of 1 :20.
  • the blended composition is packed into a dry powder inhalation device capable of delivering between about 0.1 mg to about 4 mg of the compound per dose.
  • a micronized compound of the present disclosure (10 g) is dispersed in a solution prepared by dissolving lecithin (0.2 g) in demineralized water (200 mL). The resulting suspension is spray dried and then micronized to form a micronized composition comprising particles having a mean diameter less than about 1.5 pm. The micronized composition is then loaded into metered-dose inhaler cartridges containing pressurized 1,1, 1,2- tetrafluoroethane in an amount sufficient to provide about 0.1 mg to about 4 mg of the compound per dose when administered by the metered dose inhaler.
  • a representative nebulizer composition is as follows.
  • a compound of the present disclosure (2 g of free -base equivalents) is dissolved in a solution containing 80 ml, reverse -osmosis water, 0.1-1% by weight of anhydrous citric acid, and 0.5- 1.5 equivalents of hydrochloric acid, followed by addition of sodium hydroxide to adjust the pH to 3.5 to 5.5. Thereafter, between 4-6% by weight of D-mannitol is added and solution q.s. to 100 ml.,. The solution is then filtered through a 0.2 pm filter and stored at room temperature prior to use. The solution is administered using a nebulizer device that provides about 0.1 mg to about 4 mg of the compound per dose.
  • the present disclosure provides a kit comprising one or more unit doses of a compound or pharmaceutical composition described herein, optionally wherein the kit further comprises instructions for using the compound or pharmaceutical composition.
  • the kit comprises a carrier, package, or container that is compartmentalized to receive one or more containers, such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers may be formed from, a variety of materials, such as glass or plastic.
  • the articles of manufacture provided herein may contain packaging materials.
  • Packaging materials for use in packaging pharmaceutical products include those found in, e.g., U.S. Pat. Nos. 5,323,907, 5,052,558 and 5,033,252.
  • Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
  • the container/ s) may include one or more compounds described herein, optionally in a composition or in combination with another agent as disclosed herein.
  • the container(s) may optionally have a sterile access port (for example, the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • kits may optionally comprise a compound with an identifying description or label or instructions relating to its use in the methods described herein.
  • a kit includes one or more containers, each with one or more of various materials (such as reagents, optionally in concentrated form, and/or devices) desirable from a commercial and user standpoint for use of a compound described herein.
  • materials include, but are not limited to, buffers, diluents, filters, needles, syringes, carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use.
  • a set of instructions will also typically be included.
  • a label is optionally on or associated with the container.
  • a label is on a container when letters, numbers or other characters forming the label are attached, molded, or etched onto the container itself, a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert.
  • a label is used to indicate that the contents are to be used for a specific therapeutic application.
  • the label indicates directions for use of the contents, such as in the methods described herein.
  • the pharmaceuticai composition is presented in a pack or dispenser device which contains one or more unit dosage forms containing a compound provided herein.
  • the pack may contain metal or plastic foil, such as a blister pack.
  • the pack or dispenser device is accompanied by instructions for administration.
  • the pack or dispenser is accompanied with a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration.
  • a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration.
  • Such notice for example, is the labeling approved by the U.S. Food and. Drug Administration for prescription drugs, or the approved product insert.
  • compositions containing a compound provided herein formulated in a compatible pharmaceutical carrier are prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • the compounds and pharmaceutical compositions of the disclosure may be used in combination with one or more therapeutic agents which act by the same mechanism or by a different mechanism to treat a disease.
  • the one or more agents may be administered sequentially or simultaneously, in separate compositions or in the same composition.
  • Useful classes of agents for combination therapy include, but are not limited to, compounds used to treat cardiac, kidney, pulmonary, liver, skin, immunological and oncological conditions.
  • an ALK5 inhibitor and a second therapeutic agent can be administered sequentially, wherein the two agents are introduced into a subject at two different time points.
  • the two time points can be separated by more than 2 hours, 1 or more days, 1 or more weeks, 1 or more months, or according to any intermittent regimen schedule disclosed herein.
  • the ALK5 inhibitor and the second therapeutic agent are administered simultaneously.
  • the two agents may form, part of the same composition, or the two agents may be provided in one or more unit doses.
  • the ALK5 inhibitor or the second therapeutic agent are administered parenterally, orally, inhalatively, intraperitoneally, intravenously, intraarterially, transdermally, intramuscularly, liposomally, via local delivery by catheter or stent, subcutaneously, intraadiposally, or intrathecally.
  • a therapeutically effective amount of a combination of an ALK5 inhibitor and a second therapeutic agent refers to a combination of an ALK5 inhibitor and a second therapeutic agent, wherein the combination is sufficient to affect the intended application, including but not limited to, disease treatment, as defined herein.
  • Also contemplated in the subject methods is the use of a sub-therapeutic amount of an ALK5 inhibitor and a second therapeutic agent in combination for treating an intended disease condition.
  • the individual components of the combination though present in sub- therapeutic amounts, synergistically yield an efficacious effect and/or reduced a side effect in an intended application.
  • the amount of the ALK5 inhibitor and the second therapeutic agent administered may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • Measuring an immune response and/or the inhibition of biological effects of ALK5 can comprise performing an assay on a biological sample, such as a sample from a subject. Any of a variety of samples may be selected, depending on the assay. Examples of samples include, but are not limited to blood samples (e.g. blood plasma or serum), exhaled breath condensate samples, bronchoalveolar lavage fluid, sputum samples, urine samples, and tissue samples.
  • blood samples e.g. blood plasma or serum
  • exhaled breath condensate samples e.g. blood plasma or serum
  • bronchoalveolar lavage fluid e.g. bronchoalveolar lavage fluid
  • sputum samples e.g. urine samples, and tissue samples.
  • a subject being treated with an ALK5 inhibitor and a second therapeutic agent may be monitored to determine the effectiveness of treatment, and the treatment regimen may be adjusted, based on the subject's physiological response to treatment. For example, if inhibition of a biological effect of ALK5 inhibition is above or below a threshold, the dosing amount or frequency may be decreased or increased, respectively. Alternatively, the treatment regimen may be adjusted with respect to an immune response.
  • the methods can further comprise continuing the therapy if the therapy is determined to be efficacious.
  • the methods can comprise maintaining, tapering, reducing, or slopping the administered amount of a compound or compounds in the therapy if the therapy is determined to be efficacious.
  • the methods can comprise increasing the administered amount of a compound or compounds in the therapy if it is determined not to be efficacious. Alternatively, the methods can comprise stopping therapy if it is determined not to be efficacious. In some embodiments, treatment with an ALK5 inhibitor and a second therapeutic agent is discontinued if inhibition of the biological effect is above or below a threshold, such as in a lack of response or an adverse reaction.
  • the biological effect may be a change in any of a variety of physiological indicators.
  • Specific agents that may be used in combination with the compounds disclosed herein include, but are not limited to, OFEV® (nintedanib) and Esbriet* (pirfenidone).
  • a compound disclosed herein is administered in combination with pirfenidone, optionally wherein the pirfenidone is administered by inhalation.
  • the present disclosure provides a method of treating fibrosis, such as idiopathic pulmonary fibrosis, in a subject, comprising administering to the subject an ALK5 inhibitor, such as a compound disclosed in Table 1 , and nintedanib or pirfenidone.
  • the present disclosure provides a method of treating cancer, such as lung cancer, in a subject, comprising administering to the subject an ALK5 inhibitor, such as a compound disclosed in Table 1, and nintedanib or pirfenidone.
  • an ALK5 inhibitor such as a compound disclosed in Table 1, and nintedanib or pirfenidone.
  • the present disclosure provides a method for treating a proliferative disorder (e.g., lung cancer) in a subject in need thereof, comprising administering to said subject an ALK5 inhibitor and an immunotherapeutic agent.
  • a proliferative disorder e.g., lung cancer
  • an immunotherapeutic agent e.g., TGF- ⁇ has been shown to regulate lymphocyte differentiation, suppress T cell proliferation and to enhance tumor growth.
  • TGF- ⁇ has been shown to prevent optimal activation of the immune system in immunotherapy-resistant patients (see Loffek, S. J. Oncolo. 2018, 1 -9; incorporated herein by reference in its entirety).
  • the present inventors expect that inhibition of ALK5 may enhance the efficacy of a particular immunotherapy .
  • an immunotherapeutic agent such as durvaiumab or pembrolizumab
  • an ALK5 inhibitor such as a compound of the present disclosure
  • the combination is expected to produce a synergistic effect.
  • a synergistic combination is also expected for a triple combination of radiation therapy, immunotherapy, and ALK5 inhibition.
  • the ALK5 inhibitor even when administered, locally (e.g., to the lung by inhalation), may stimulate both local and systemic immune responses, allowing for the treatment of primary or metastatic tumors in tissues beyond the site of the local deli very.
  • an inhaled ALK5 inhibitor may be administered in combination with an immunotherapeutic agent to treat melanoma, renal cell carcinoma, colon cancer, or breast cancer.
  • the ALK5 inhibitor and the immunotherapeutic agent are administered sequentially or simultaneously. In some embodiments, the ALK5 inhibitor and the immunotherapeutic agent are more effective in treating the proliferative disorder than either agent alone. In some embodiments, the ALK5 inhibitor and the immunotherapeutic agent yield a synergistic effect in treating the proliferative disorder. The synergistic effect may be a therapeutic effect that is greater than either agent used alone in comparable amounts under comparable conditions. The synergistic effect may be a therapeutic effect that is greater than results expected by adding the effects of each agent alone. In some embodiments, the proliferative disorder is a cancer condition. In some embodiments, the cancer condition is lung cancer, such as non-small cell lung cancer.
  • immunotherapeutic agent refers to any agent that induces, enhances, suppresses or otherwise modifies an immune response. This includes the administration of an active agent to, or any type of intervention or process performed on, the subject, with the objective of modifying an immune response.
  • An immunotherapeutic agent may, for example, increase or enhance the effectiveness or potency of an existing immune response in a subject, for example, by stimulating mechanisms that enhance the endogenous host immune response or overcoming mechanisms that suppress the endogenous host immune response.
  • Immuno response refers to the action of a cell of the immune system including, for example, B lymphocytes, T lymphocytes, natural killer (NK) cells, macrophages, eosinophils, mast cells, myeloid-derived suppressor cells, dendritic cells and neutrophils and soluble macromolecules produced by any of these cells or the liver (including antibodies, cytokines and complement), that results in selective targeting, binding to, damage to, destruction of, and/or elimination of invading pathogens, cells or tissues infected with pathogens, cancerous or other abnormal cells, or, in cases of autoimmunity or pathological inflammation, normal human cells or tissues, from the body of a subject.
  • B lymphocytes B lymphocytes, T lymphocytes, natural killer (NK) cells
  • macrophages eosinophils
  • mast cells myeloid-derived suppressor cells
  • dendritic cells and neutrophils dendritic cells and neutrophils and soluble macromolecules produced by any of these cells or the
  • an immunotherapeutic agent may comprise a PD-1 inhibitor.
  • an immunotherapeutic agent may comprise a CTLA-4 inhibitor.
  • an immunotherapeutic agent may comprise a B7 inhibitor.
  • a PD-1 inhibitor suitable for use in the subject methods can be selected from a variety of types of molecules.
  • the PD-1 inhibitor can be a biological or chemical compound, such as an organic or inorganic molecule, peptide, peptide mimetic, antibody or an antigenbinding fragment of an antibody.
  • Some exemplary classes of agents suitable for use in the subject methods are detailed in the sections below.
  • a PD-1 inhibitor for use in the present disclosure can be any PD-1 inhibitor that is known in the art, and can include any entity that, upon administration to a patient, results in inhibition of the PD-1 pathway in the patient.
  • a PD-1 inhibitor can inhibit PD-1 by any biochemical mechanism, including disruption of any one or more of PD-1/PD-L1, PD1/PD-L2 and PD-L1/CD80 interactions.
  • the PD-1 inhibitor is a molecule that inhibits the binding of PD-1 to its ligand binding partners.
  • the PD-1 ligand binding partners are PD-L1 and/or PD-L2.
  • a PD-1 inhibitor is a molecule that inhibits the binding of PD-L1 to its binding partners.
  • PD-L1 binding partners are PD1 and/or CD80.
  • the PD-1 inhibitor is a molecule that inhibits the binding of PD-L2. to its binding partners.
  • a PD-L2 binding partner is PD1.
  • the inhibitor may be an antibody, an antigen binding fragment thereof, an irnmunoadhesin, a fusion protein or oligopeptide.
  • the PD-1 inhibitor is an anti-PD-1 antibody.
  • the anti-PD-1 antibody is capable of inhibiting binding between PD-1 and PD-L1.
  • the anti-PD-1 antibody is capable of inhibiting binding between PD-1 and PD-L2.
  • the PD- 1 inhibitor is an anti-PD-L1 antibody.
  • the anti-PD-L1 antibody is capable of inhibiting binding between PD-L1 and PD-1 and/or between PD-L1 and CD80.
  • the PD-1 inhibitor is an anti-PD-L2 antibody. In some further embodiments, the anti-PD-L2 antibody is capable of inhibiting binding between PD-1 and PD-L2. In yet another embodiment, the PD-1 inhibitor is nivolumab or pembrolizumab. In some embodiments, the PD-1 inhibitor is selected from atezohzumab, avelumab, nivolumab, pembrolizumab, durvalumab, and BGB-A317.
  • Inhibition of the PD-1 pathway can enhance the immune response to cancerous cells in a patient.
  • the interaction between PD-1 and PD-L1 impairs T cell response as manifested by a decrease in tumor-infiltrating lymphocytes (TILs) and a decrease in T-cell receptor mediated proliferation, resulting in T cell anergy, exhaustion or apoptosis, and immune evasion by the cancerous cells.
  • TILs tumor-infiltrating lymphocytes
  • This immune suppression can be reversed by inhibiting the local interaction between PD-L1 and PD-1 using a PD-1 inhibitor, including, for example, an anti-PD-1 and/or an anti- PD-L1 Ab.
  • a PD-1 inhibitor may improve or restore antitumor T-cell functions.
  • Anti-PD-1 antibodies suitable for use in the disclosure can be generated using methods well known in the art.
  • Exemplary PD-1 inhibitors include, bitt are not limited to: nivolumab (BMS936558), pembrolizumab (MK-347.5), pidilizumab (CT-OU), AMP-224, AMP-514, BMS-936559, RG7446 (MPDL3280A), MDX- 1106 (Medarex Inc.), MSB0010718C, MEDI4736, and HenGtui mAB005 (WO 15/085847).
  • Further PD-1 antibodies and other PD-1 inhibitors include those described in WO 04/056875, WO 06/121168, WO
  • anti-PD-1 antibodies are commercially available, for example from ABCAM (AB137132), BIOLEGEND (EH12.2H7, RMP 1-14) and AFFYMETRIX EBIOSCIENCE (J105, 1116, M1H4).
  • CTLA-4 inhibitors A CTLA-4 inhibitor suitable for use in the subject methods can be selected from a variety of types of molecules.
  • the CTLA-4 inhibitor can be a biological or chemical compound, such as an organic or inorganic molecule, peptide, peptide mimetic, antibody or an antigen-binding fragment of an antibody.
  • Some exemplary classes of agents suitable for use in the subject methods are detailed in the sections below.
  • a CTLA-4 inhibitor for use in the present disclosure can be any CTLA-4 inhibitor that is known in the art, and can include any entity that, upon administration to a patient, results in inhibition of the CTLA-4 pathway in the patient.
  • a CTLA-4 inhibitor can inhibit CTLA-4 by any biochemical mechanism, including disruption of either one or both of CTLA-4/CD80 and CTLA -4/CD86 interactions.
  • the CTLA-4 inhibitor is a molecule that inhibits the binding of CTLA-4 to its ligand binding partners.
  • the CTLA-4 ligand binding partners are CD80 and/or CD86.
  • a CTLA-4 inhibitor is a molecule that inhibits the binding of CD80 to its binding partners.
  • a CD80 binding partner is CTLA-4.
  • the CTLA-4 inhibitor is a molecule that inhibits the binding of CD86 to its binding partners.
  • a CD86 binding partner is CTLA-4.
  • the inhibitor may be an antibody, an antigen binding fragment thereof, an immunoadhesin, a fusion protein or oligopeptide.
  • the CTLA-4 inhibitor is an anti-CTLA-4 antibody.
  • the anti-CTLA-4 antibody is capable of inhibiting binding between CTLA-4 and CD80.
  • the anti-CTLA-4 antibody is capable of inhibiting binding between CTLA-4 and CD86.
  • the CTLA-4 inhibitor is an anti-CD80 antibody.
  • the anti-CD80 antibody is capable of inhibiting binding between CTLA-4 and CD80.
  • the CTLA-4 inhibitor is an anti-CD86 antibody.
  • the anti-CD86 antibody is capable of inhibiting binding between CTLA-4 and CD86.
  • the CTLA-4 inhibitor is tremeiimumab or ipilimumab.
  • CTLA-4 pathway can enhance the immune response to cancerous cells in a patient.
  • This immune suppression can be reversed by inhibiting the local interaction between CD80 or CD86 and CTLA-4 using a CTLA-4 inhibitor, including, for example, an anti-CTLA-4 Ab, anti-CD80 Ab or an antiCD86 Ab.
  • a CTLA-4 inhibitor may improve or restore antitumor T-cell functions.
  • Anti-CTLA-4 antibodies suitable for use in the disclosure can be generated using methods well known in the art.
  • CTLA-4 inhibitors include but are not limited to tremeiimumab and ipilimumab (also known as 10D1 or MDX-010). Further CTLA-4 antibodies and other CTLA-4 inhibitors include those described in WO 98/042752, WO 00/037504, WO 01/014424 and WO 04/035607; LT. S. Pub. Nos.
  • a pharmaceutical composition comprising a compound of the disclosure or a pharmaceutically acceptable salt thereof and one or more other therapeutic agents.
  • the therapeutic agent may be selected from the classes of agents specified above and from the lists of specific agents described above.
  • the pharmaceutical composition is suitable for delivery to the lungs.
  • the pharmaceutical composition is suitable for inhaled or nebulized administration.
  • the pharmaceutical composition is a dry powder or a liquid, composition.
  • the disclosure provides a method of treating a disease or disorder in a mammal comprising administering to the mammal a compound of the disclosure or a pharmaceutically acceptable salt thereof and one or more other therapeutic agents.
  • the agents When used in combination therapy, the agents may be formulated in a single pharmaceutical composition, or the agents may be provided in separate compositions that are administered simultaneously or at separate times, by the same or by different routes of administration. Such compositions can be packaged separately or may be packaged together as a kit. The two or more therapeutic agents in the kit may be administered by the same route of administration or by different routes of administration.
  • reaction mixtures were worked up as described specifically in each preparation; commonly, reaction mixtures were purified by extraction and other purification methods such as temperature- and solvent-dependent crystallization, and precipitation.
  • reaction mixtures were routinely purified by preparative HPLC, typically using Microsorb C18 and Microsorb BDS column packings and conventional eluents.
  • Progress of reactions was typically monitored by liquid chromatography mass spectrometry (LCMS). Characterization of isomers was typically done by Nuclear Overhauser effect spectroscopy (NOE). Characterization of reaction products was routinely carried out by mass spectrometry and/or 1 H-NMR spectroscopy.
  • Example 1 Synthesis of5-(3-(6-niethylpyridin-2-yl)-1H-pyrazol-4-yl)-N-(4-(piperazin-1-yl)phenyl)- 1H-benzo[d]imidazol-2-amine (580) .
  • Step A Preparation of tert-butyl 2-bromo-6-iodo-1H-benzo[d]imidazole-1-carboxylate (1-2).
  • 1-1 (6.50 g, 20.1 mmol) and BocaO (5.30 g, 24.2 mmol) in MeCN (150 mL) was added DMAP (491 mg, 4.03 mmol) at 25 °C.
  • DMAP 491 mg, 4.03 mmol
  • the mixture was stirred at 25 °C for 3 h.
  • the reaction was poured into ice water (1,000 mL) and was extracted with EtOAc (3 x 250 mL).
  • Step B Preparation of 2-methyl-6-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl)pyridine (1-5).
  • 1-3 30.0 g, 176 mmol
  • H 2 O 25 mL
  • 1-4 58.6 g, 211 mmol
  • Pd(dppf)Cl 2 6.40 g, 8.80 mmol
  • K 3 PO 4 55.8 g, 263 mmol
  • Step C Preparation of 2-(4-iodo-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl)-6-methylpyridine (1- 6).
  • K 2 CO 3 (17.0 g, 123.3 mmol) in CH 2 Cl 2 (300 mL)
  • ICI 32.3 g, 123.3 mmol
  • CH 2 Cl 2 100 mL
  • the mixture was stirred at 25 °C for 12 h.
  • the reaction mixture was poured into Na 2 S 2 O 3 (300 mL, 2.0 M) at 0 °C.
  • Step D Preparation of (3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4- yDboronic acid (1-8).
  • 1-6 (14.0 g, 29.8 mmol) and 1-7 (16.6 g, 89.4 mmol) in THF (180 ml.,) was added i-PrMgCl (44.7 ml.,, 89.4 mmol) at 0 °C. After addition, the mixture was stirred for 1 h at 0 °C then warmed to 25 °C and stirred for 15 h. The reaction was quenched with water (100 mL).
  • Step E Preparation of tert-butyl 2-bromo-6-(3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)- 1H-pyrazol-4-yl)-1H-benzo[d]imidazole-1-carboxylate (1-9). This reaction was run as 4 smaller reaction batches that were combined after the reactions were complete.
  • Step F Preparation of 5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)-N-(4-(piperazin-l-yl)phenyl)- 1H-benzo[d]imidazol-2-amine (580).
  • Example 2 Synthesis of N-(isoindolin-5-yl)-6-(5-(6-methylpyridin-2-yl)-lH-imidazol-4- yl)benzo[d]oxazoI-2-amine (517) .
  • Step A Preparation of 2-(1H-imidazol-5-yl)-6-methylpyridine (2-3).
  • 2-2 3.00 g, 24.8 mmol
  • EtOH 50 mL
  • p-toluenesulfonylmethyl isocyanide 2-1, 4.84 g, 24.8 mmol
  • KCN 0.23 g, 4.95 mmol
  • the solvent was removed under reduced pressure and the residue was partitioned between H 2 O (50 ml.,) and EtOAc (75 mL).
  • Step B Preparation of 2-methyI-6-(1-((2-(trimethylsilyl)ethoxy)methyl)-1H-imidazol-5-yl)pyridine (2-4).
  • SEM-C1 (1.69 mL, 9.43 mmol) was added dropwise at 0 °C and the mixture was allowed to warm to 23 °C and was stirred for 1 h.
  • Step C Preparation of 2-(4-bromo-1-((2-(trimethyIsilyl)ethoxy)methyI)-1H-imidazol-5 -yI)-6- methylpyridine (2-5).
  • a solution of NBS (2.51 g, 14.1 mmol) in DMF (15.0 mL) was added dropwise at 0 °C over 30 min to a solution of 2-4 (4.54, 15.7 mmol) in DMF (32.0 mL). The reaction mixture was stirred at 23 °C for an additional 45 min, then was quenched with sat.
  • Step D Preparation of 6-(5-(6-methylpyridin-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyi)-1H- imidazol-4-yl)benzo[d]oxazoI-2-amine (2-7).
  • 2-6 (3.11 g, 11.9 mmol)
  • Pd(dppf)Ch (0.795 g, 1.09 mmol)
  • K 2 CO 3 (4.50 g, 32.6) mmol) were added to a solution of 2-5 (4.00 g, 10.9 mmol) in dioxane/H 2 O (8:1 , 36.0 mL) under nitrogen and the mixture was stirred, at 100 °C for 18 h.
  • Step E Preparation of N-(isoindolin-5-yl)-6-(5-(6-methylpyridin-2-yl)-1H-imidazol-4- yl)benzo[d]oxazoi-2-amine (517).
  • Example 3 Synthesis of 5-(5-(5-chloro-2-fluorophenyl)-1H-imidazol-4-yl)-N-( 1, 2,3,4- tetrahydroisoquinolin-7-yl)benzo[d]oxazol-2-amine (259).
  • Step A Preparation of 5-(5-chloro-2-fluorophenyl)-1H-imidazole (3-2).
  • the reaction was completed in 2 batches of 5 g each. 3-1 (5.00 g, 31.5 mmol) and p-toluenesulfonylmethyl isocyanide (2-1, 6.15 g, 31.5 mmol) were dissolved in EtOH (50 mL).
  • NaCN 154 mg, 3.15 mmol
  • the solvent was removed under reduced pressure and the crude residue was diluted with H 2 O (50 mL) and CH 2 Cl 2 (100 mL).
  • Step B Preparation of 5-(5-chloro-2-fluorophenyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H- imidazole (3-3).
  • 3-2 5.50 g, 28.1 mmol
  • THF 60 ml.,
  • SEMC1 6.20 g, 33.7 mmol
  • the reaction mixture was quenched with sat. aq.
  • Step C Preparation of 4-bromo-5-(5-chloro-2-fluorophenyl)-l-((2-(trimethylsilyl)ethoxy)methyl) - 1H-imidazole (3-4).
  • NBS NBS (2.51 g, 14.1 mmol)
  • the reaction mixture was diluted with H 2 O (100 mL) and extracted with EtOAc (3 x 50 mL). The combined organics were washed with brine (2 x 20 mL), dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure.
  • the crude residue was purified via flash chromatography (10-20% EtOAc/hexanes) to afford 3-4 (2.7 g) as a brown oil.
  • Step D Preparation of 5-(5-(5-chioro-2-fluorophenyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H- imidazol-4-yl)benzo[d]oxazol-2-amine (3-6).
  • This reaction was performed in 2 batches of 400 mg each. 3-4 (400 mg, 0.990 mmol) and 3-5 (308 mg, 1.18 mmol) were charged in dioxane (8.00 mL).
  • K 3 PO 4 419 mg, 1.98 mmol
  • Step E Preparation of 5-(5-(5-chloro-2-fluorophenyl)-1H-imidazol-4-yl)-N-(1, 2,3,4- tetrahydroisoquinolin-7-yl)benzo[d]oxazol-2-amine (259).
  • a mixture of 3-6 (23.0 mg, 0.0500 mmol), Cs 2 CO 3 (41.0 mg, 0.125 mmol), and 3-7 (19.0 mg, 0.0600 mmol) was suspended in a solution of BrettPhos (2.68 mg, 5.00 ⁇ mol) and BrettPhos Pd G4 (4.60 mg, 5.00 ⁇ mol) in 1,4-dioxane (0.250 mL).
  • Example 4 Synthesis of N-(6-(5-(6-methylpyridin-2-yl)-1H-imidazol-4-yl)benzo[d]thiazol-2-yl)-2- (piperazin- 1 -yl)acetamide (636).
  • Step A Preparation of 2-methyl-6-(l-((2-(trimethylsilyl)ethoxy)methyl)-1H-imidazol-4-yl)pyridine (4-2).
  • 4-1 4.484 g, 28.168 mmol
  • sodium hydride 1.352 g, 33.802 mmol
  • Step B Preparation of 2-(5-bromo-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-imidazol-4-yl)-6- methylpyridine (4-3).
  • a solution of NBS (3.985 g, 22.387 mmol) in 23 mL of DMF was added dropwise at 0 °C for 30 min to a solution of 4-2 (7.2. g, 24.871 mmol) in 51 mL of DMF and the reaction mixture was stirred at rt for 45 min.
  • the reaction was quenched with NaHSOs (sat), extracted with EtOAc and the organic layer was washed with water, dried over MgSQs, filtered and evaporated to dryness.
  • Step C Preparation of 6-(4-(6-methylpyridin-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-imidazol- 5-yl)benzo[d]thiazol-2-amine (4-5).
  • 4-4 (3.299 g, 11.945 mmol)
  • Pd(dppf)Cl 2 (0.795 g, 1.086 mmol)
  • K 2 CO 3 4.502 g, 32.577 mmol) were added to a solution of 4-3 (4 g, 10.859 mmol) in 36 mL of dioxane/ H 2 O (8:1) under nitrogen and the mixture was stirred at 100 °C overnight.
  • Step D Preparation of 2-chloro-N-(6-(4-(6-methylpyridin-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)- 1H-imidazol-5-yl)benzo[d]thiazol-2-yl)acetamide (4-6).
  • DIPEA 96 ⁇ L, 0.548 mmol
  • DCM 914 ⁇ L
  • chloroacetyl chloride 15.29 ⁇ L, 0.192 mmol. The reaction was stirred and allowed to warm to rt overnight.
  • Step E Preparation of N-(6-(5-(6-methylpyridin-2-yl)-1H-imidazol-4-yl)benzo[d]thiazol-2-yl)-2- (piperazin- 1-yl)acetamide (636).
  • a solution of 4-6 2.4 mg, 0.047 mmol
  • acetonitrile 467 ⁇ L
  • tert-butyl piperazine- 1- carboxylate 17.39 mg, 0.093 mmol
  • DIPEA 32.6 pL, 0.187 mmol
  • Example 5 Synthesis of 1-[6-[5-(6-methyl-2-pyridyl)-1H-pyrazol-4-yl]-[1,2,4]triazolo[1,5-a]pyridin- 2-yl] -3-(3-pyridyl)urea (52).
  • Step A Preparation of 2-methyl-6-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-5-yl)pyridine (5-3).
  • 5-1 (10.0 g, 58.5 mmol) in dioxane (150 mL) and H 2 O (15 mL) was added 5-2 (19.5 g, 70.2 mmol), Pd(dppf)Cl 2 (2.1 g, 2.92 mmol), and K 3 PO 4 (18.6 g, 87.7 mmol).
  • the reaction was then stirred at 80 °C for 4 h under N2 atmosphere.
  • the mixture was filtered thru a pad of celite and concentrated in vacuum.
  • Step B Preparation of 2-(4-iodo4-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-5-yl)-6-methylpyridine (5- 4).
  • 5-3 6.0 g, 24.7 mmol
  • K 2 CO 3 10.2 g, 74.0 mmol
  • DCM 130 mL
  • IC1 IC1 (19.4 g, 74.0 mmol) in DCM (50 mL) dropwise at -50 °C.
  • the mixture was warmed and stirred at 10 °C for 16 h.
  • Step C Preparation of 2-methyl-6-(1-(tetrahydro-2H-pyran-2-yl)-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1H-pyrazol-5 -yl)pyridine (5-6).
  • 5-4 7.5 g, 20.31 mmol
  • 5-5 (11.3 g, 60.94 mmol) in THE (150 mL) was added i-PrMgCl (30.5 mL, 60.94 mmol) at -5 to 0 °C. After addition, the mixture was stirred at 15 °C for 16 h. The mixture was quenched with sat. NH4CI aq (200 mL) and extracted with EA (150 mL x 3). The organic layer was concentrated in vacuum to obtain crude 5-6 (7.0 g) as a yellow oil.
  • Step D Preparation of 6-(5-(6-methylpyridin-2-yl)- 1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)- [1,2,4]triazolo[1,5-a]pyridin-2-amine (5-8).
  • 5-6 4.0 g, 18.77 mmol
  • 5-7 7.0 g, 17.06 mmol
  • Pd(dppf)Cl 2 850 mg, 1.16 mmol
  • K 2 CO 3 5.2 g, 37.54 mmol.
  • Step E Preparation of 1-[6-[5-(6-methyl-2-pyridyl)-1H-pyrazol-4-yl]-[1 ,2,4]triazolo[1,5-a]pyridin-2- yl]-3-(3-pyridyl)urea (52).
  • a mixture of 5-8 (20 mg, 0.053 mmol) and DIPEA (2.7.9 pL, 0.160 mmol) in DME (266 ⁇ L) was added 4-8 (12.80 mg, 0.017 mmol). The reaction mixture was allowed to stir for 1 h at 70 °C. The reaction mixture was concentrated under vacuum.
  • Step A Preparation of 7 -bromo-5 -iodo-lH-indazole (6-2).
  • 6-1 5.0 g, 23.57 mmol
  • DCM:H 2 O 100 mL:100 mL
  • NaNO 2 8.12 g, 117.9 mmol
  • CH 2 I 2 3.80 mL, 47.14 mmol
  • AcOH 26.9 mL, 471.4 mmol
  • Step B Preparation of 7-bromo-5-iodo-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazole (6-3).
  • Step C Preparation of 7-bromo-5-(3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazole (6-4).
  • 6-3 600 mg, 1.32 mmol
  • DMA 6.0 mL
  • 5-6 537 mg, 1.45 mmol
  • folio wed by the addition of K 3 PO 4 (560 mg, 2.64 mmol) and water (1.0 mL).
  • Step D Preparation of [4- [5--[3 -(6-methylpyridin-2-yl)- 1H-pyrazol -4-yl]-2H-indazol-7 - yl] thiophen -2- yl]methanamine (574).
  • Example 7 Synthesis of N-[5-(2,5-dihydro- lH-pyrrol-3-yl)pyridin- 2-yI]-5--[3-(6--methylpyridin-2-- yl)-1H-pyrazol-4-yl]-1H-indazol-3-amine (348).
  • Step A Preparation of tert-butyl 3 -(6- aminopyridin- 3-yl)-2,5-dihydro- 1H -pyrrole- 1 -carboxylate- - methane (7-3).
  • Step B Preparation of 5-(5-(6-methylpyridin-2-yI)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)- IH-indazole (7-5).
  • 5-4 (4.00 g, 10.8 mmol) in 1,4-dioxane (40.0 L) was added 7-4 (4.48 g, 13.0 mmol) and the reaction was purged with argon for 15 minutes followed by addition of K3PO4 (4.60 g, 2 eq., 21.7 mmol) in water (8.00 mL) and further purged with argon for 5 min.
  • Step C Preparation of 3-bromo-5-(5-(6-methylpyridm-2-yl)-l-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)-1H-indazole (7-6).
  • NBS NBS (1.19 g, 6.68 mmol) portionwise at 0 °C.
  • the resulting mixture was allowed to stir at 25 °C for 30 min.
  • the reaction mixture was diluted with ice cold water (100 mL).
  • the solid precipitate was filtered and washed, with water (3 times) followed by a pentane wash.
  • the resulting light yellow solid (7-6, 2.30 g) was used in the next step without further purification.
  • Step D Preparation of 3-bromo-5-(5-(6-methylpyridin-2-yl)-l-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)-1-(tetrahydro-2H--pyran-2-yl)-1H-indazole (7-7).
  • DCM dimethyl sulfoxide
  • DHP 1,3-bis(trimethoxysilyl)
  • pTSA-H 2 O 200 mg, 1.05 mmol
  • Step E Preparation of N-[5-(2,5-dihydro-1H-pyrrol-3-yl)pyridin-2-yl]-5-[3-(6-methylpyridin-2-yl)- 1H-pyrazol-4-yl]-1H-indazol-3-amine (348).
  • Step A Preparation of 5-(3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)- 1H-indazole (8-2).
  • 8-1 1.0 g, 5.07 mmol
  • H 2 O 5 mL
  • K 3 PO 4 2.15 g, 10.14 mmol
  • the reaction was purged with argon for 5 minutes followed by addition of PdCl 2 (dppf)DCM (414 mg, 0.507 mmol).
  • the reaction was allowed, to stir at 110 °C for 12 h.
  • the reaction mixture was cooled to 25 °C, diluted with water, and extracted with EtOAc.
  • Step B Preparation of 1-(4-bromophenyl)-5-(3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl) 1H-pyrazol-4-yl)-1H-indazole (8-5).
  • Step C Preparation of 1- [4-(2,5-dihydro-1H-pyrrol-3-yl)phenyl]-5-[3 -(6-methylpyridin-2-yl)- 1H- pyrazol-4-yl]indazole. (594).
  • Example 9 Synthesis of [4- [6-[3--(6-methylpyridin-2-yl)- 1H-pyrazol-4-yl]- 1H-indazol-4- yl]phenyl] methanamine (107).
  • Step A Preparation of 7-bromo-5-iodo-1-(tetrahydro-2H-pyran-2-yI)-1H-indazoIe (9-2).
  • DHP 1.97 mL, 23.3 mmol
  • pTSA- H 2 O 295 mg, 1.55 mmol
  • the reaction mixture was stirred at 55 °C for 6 h.
  • the reaction mixture was diluted with ammonium chloride solution (20 mb) and DCM (40 mb). The organic layer was washed with brine, dried over sodium, sulfate, and concentrated in vacuum.
  • Step B Preparation of 4-bromo-6-(3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (9-3).
  • Step C Preparation of [4-[6-[3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl]-1H-indazol-4- yl]phenyl]methanamine (107).
  • Example 11 Synthesis of [4-[6-[3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl]-1H-indazol-3- yljphenyljmethanamine (5).
  • Step A Preparation of 6-(3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)- 1H-indazoIe (10-2).
  • 10-1 500 mg, 2.53 mmol
  • 1,4-dioxane 8.0 mL
  • H 2 O 2.0 mL
  • K 3 PO 4 1.07 g, 5.06 mmol
  • the reaction mixture was purged with N2 for 15 minutes followed by addition of Pd(dppf)Cl 2 (412 mg, 0.50 mmol).
  • Step B Preparation of 3-bromo-6-(3-(6-methylpyridin-2-yl)-l.-(tetrahydro-2H-pyran-2-yi)-1H- pyrazol-4-yl)-1H- indazole (10-3).
  • DMF 10 ml.
  • NBS 273 mg, 1.53 mmol
  • the reaction mixture was diluted with water and extracted using EtOAc (3x).
  • Step C Preparation of [4-[6-[3--(6-methylpyridin-2-yl)- 1H-pyrazol-4-yl]-1H-indazol-3- yl]phenyl]methanamine (5).
  • the reaction mixture was concentrated under vacuum.
  • Step A Preparation of tert -butyl 2-(4-(3-fluoro-4-nitrophenyl)-l-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-5-yi)-6-methylpyridine (11-2).
  • Step B Preparation of N-(4-bromophenyl)-5-(5-(6-methylpyridin-2-yl)-l-(tetrahydro-2H-pyran-2-yl)- 1H-pyrazol-4-yi)-2-nitroaniline (11-4).
  • Step C Preparation of N1 -(4 -bromophenyl)- 5-(5-(6-methyIpyridin-2-yl)- 1 -(tetrahydro-2H-pyran--2- yl)-1H-pyrazol-4-yl)benzene- 1,2 -diamine (11-5).
  • 11-4 58 mg, 0.109 mmol
  • acetone/EbO 6:1, 2.56 mL
  • NH 4 CI 233 mg, 4.36 mmol
  • Zn 143 mg, 2.18 mmol
  • Step D Preparation of 1 -(4-bromophenyl) -6 (5 -(6-methylpyridin-2-yl)- 1 - (tetrahydro -2H- pyran -2 -yl) - 1H-pyrazol-4-yl)-1H-benzo[d][1,2,3]triazole (11-6).
  • Sodium nitrite 0.055 g, 0.790 mmol
  • 11-5 0.040 g, 0.079 mmol
  • DCM 2 mL
  • Step E Preparation of 6-[5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl]-1-[4-(1,2,3,6-tetrahydropyridin- 4-yl)phenyl]benzotriazole (200).
  • Example 12 Synthesis of 5-[3-(6-methyl-2-pyridyl)-1H-pyrazol-4-yl]-N-(5-piperazin-1-yl-2- pyridyi)-1 ,2-benzothiazoi-3-amine (415).
  • Step A Preparation of 5--(3-(6--methylpyridin-2-y])- 1-(tetrahydro-2H-pyran- 2-yl)- 1H-pyrazoI-4- yl)benzo[d]isothiazol-3 -amine (12-2).
  • 5-6 500 mg, 1.35 mmol
  • 12-1 401 mg, 1.76 mmol
  • K 3 PO 4 572 mg, 2.70 mmol
  • the reaction mixture was purged with N 2 for 15 minutes followed by addition of catalyst Pd(dppf)Cl 2 -DCM (110 mg, 0.135 mmol).
  • Step B Preparation of 5 -[3 -(6 -methyl- 2 -pyridyl)- 1H-pyrazol-4-yI]-N-(5-piperazin-1-yl-2-pyridyI)- 1,2-benzothiazol -3-amine (415).
  • a mixture of 12-2 (25.6 mg, 0.065 mmol), 12-3 (18.66 mg, 0.055 mmol), BrettPhos Pd G4 (5.02 mg, 5.45 ⁇ mol), cesium carbonate (71.1 mg, 0.218 mmol), and BrettPhos (2.93 mg, 5.45 ⁇ mol) in 1 ,4-dioxane (273 ⁇ L) was heated to 85 °C for 16 h.
  • Example 13 Synthesis of 4-((4-isopropylpiperazin- 1-yl)methyl)-2-(4-(5-(6-methylpyridin-2-yl)-1H pyrazol-4-yl)quinolin-6-yl)oxazole (60) and (4-isopropylpiperazin-1-yl)-[2-[4-[3-(6-methyl-2-pyridyl)-1H- pyrazol-4-yl]-6-quinolyl]oxazol-4-yl]methanone (441).
  • Step A Preparation of (Z)-2-(6-bromoqumolin-4-yl)-3-(dimethylamino)-l-(6-methylpyridm-2 yl)prop-2-en-1-one (13-2).
  • 13-1 14 g, 41.0 mmol
  • Step B Preparation of 6-bromo-4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinoline (13-3).
  • Step C Preparation of methyl 4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinoline-6-carboxylate (13-4).
  • 13-3 6.50 g, 17.8 mmol
  • PPh 3 934 mg, 3.60 mmol
  • NaOAc 2.20 g, 26.7 mmol
  • MeOH 130 mL
  • DMF 26 ml.
  • Pd(OAc) 2 (1.20 g, 5.30 mmol) under N 2 atmosphere.
  • the reaction was stirred at 80 °C for 16 h under CO (50 psi).
  • the mixture was filtered and concentrated under reduced pressure.
  • the residue was purified by flash chromatography (0-5% MeOH/CH 2 Cl 2 ) to afford 13-4 (3.0 g) as a yellow solid.
  • Step D Preparation of (4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinolin-6-yl)methanol (13-5).
  • UAIH4 330 mg, 8.70 mmol
  • 13-4 (1.50 g, 4.40 mmol
  • THF 30 mL
  • the mixture was warmed to 10 °C and stirred for 4 h.
  • the reaction was quenched with H 2 O ( 1 mL) and the suspension was filtered and washed with EtOAc (20 mL). The solid was dried to obtain 13-5 (1.80 g) as a yellow solid.
  • Step E Preparation of 4-(5-(6-methylpyridin-2-yi)-1H-pyrazol-4-yi)quinoline-6-carbalde.hyde (13-6). To a suspension of 13-5 (500 mg, 1.26 mmol) in DMF (8.0 mL) was added. MnO 2 (3.05 g, 31.6 mmol) and the resulting mixture was stirred at 23 °C for 40 h. The crude reaction was filtered through Celite, washing with DMF (10 mL). The filtrate was concentrated under reduced pressure and the residue was purified via flash chromatography (0-10% MeOH/DCM) to afford 13-6 (168 mg) as a white solid. [M+H] + calcd for C 19 H 14 N 4 O 315.1, found 315.
  • Step F Preparation of methyl 2-(4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinolin-6-yl)oxazole- 4-carboxylate (13-8).
  • a suspension of 1-serine methyl ester (13-7, 26.7 mg, 0.172 mmol) and K 2 CO 3 (47.5 mg, 0.344 mmol) in DMF (200 p.L) was added a solution of 13-6 (54 mg, 0.172. mmol) in DMA (277 pl.,) and the mixture was stirred at 23 °C for 15 h.
  • Step G Preparation of 2-(4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinolin-6-yl)oxazole-4- carboxylic acid (13-9).
  • 13-8 15.8 mg, 0.038 mmol
  • MeOH 32.0 ⁇ L
  • water 32.0 ⁇ L
  • LiOH 1.6 mg, 0.067 mmol
  • the reaction was warmed to 23 °C and was stirred, for 1 h.
  • the solvents were removed under reduced pressure and. the residue was purified by preparative HPLC chromatography to obtain the bis-TFA salt of 13-9 as a white solid (14.1 mg).
  • [M+H] + calcd for C 22 H 15 N 5 O3 398.1, found 398.2.
  • Step H Preparation of (4-isopropylpiperazin-1-yl)(2-(4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4- yl)quinolin-6-yl)oxazol-4-yl)methanone (441).
  • Step I Preparation of 4-((4-isopropylpiperazin-l-yl)methyl)-2-(4-(5-(6-methylpyridin-2-yl)-1H- pyrazol-4-yl)quinolin-6-yl)oxazole (60).
  • Li AIH4 10 pL, 10.0 pmol, 1.0 M in THE
  • the reaction was quenched with a solution of AcOH/H 2 O (1:1, 100 pL) and the resulting suspension was filtered.
  • the filtrate was purified by preparative HPLC chromatography to obtain the bis-TFA salt of the title compound (0.6 mg). [M+H] + calcd for C 29 H 31 N 7 O 494.3, found 494.0.
  • Example 14 Synthesis of 4-(5-(6-methyipyridin-2-yi)-1H-pyrazol-4-yl)-7-(2-azaspiro[3.5]non-6-en- 7-yl)quinoline (551).
  • Step A Preparation of (Z)-2-(7-bromoquinolin-4-yl)-3-(dimethylamino)-1-(6-methylpyridin-2- yl)prop-2-en-1-one (13-2).
  • a solution of 13-1 (32.0 g, 93.8 mmol) in N,N-dimethylformaniide dimethyl acetal (250 mL) was stirred at 80 °C for 6 h. The reaction was concentrated under reduced pressure to afford crude 13-2 as a red oil (32.0 g) which was used directly in the next step.
  • Step B Preparation of 7-bromo-4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinoline (13-3).
  • a solution of 13-2 (32.0 g, 80.8 mmol) and NH 2 NH 2 H 2 O (20.0 g, 404 mmol) in EtOH (300 mL) was stirred at 80 °C for 4 b.
  • the solution was concentrated under reduced pressure and the residue was purified by flash chromatography (0-40% EtOAc/petroIeum ether) and recrystallization (50 mL, of EtOAc) to obtain 13-3 (5.70 g) as a white solid.
  • Step C Preparation of 4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)-7-(2-azaspiro[3.5]non-6-en-7- yl)quinoline (551).
  • 13-3 0.018 g, 0.05 mmol
  • 13-4 (21.0 mg, 0.060 mmol)
  • Pd(dppf)Cl 2 (1.83 mg, 2.500 ⁇ mol) in 1,4-dioxane (0.25 mL) was added a solution of Na 2 CO 3 (0.150 mL, 1.0 M, 0.150 mmol).
  • Example 15 Synthesis of 7-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)-4-(4-(piperidin-3- yl)phenyl)isoquinoline (557).
  • Step A Preparation of 4,7- dibromoisoquinoline (15-2).
  • 15-1 3.0 g, 14.42 mmol
  • acetic acid 30.0 ml
  • NBS 3.08 g, 17.30 mmol
  • the reaction mass was allowed to warm to RT and then stirred for 6 h at 110 °C.
  • the reaction was cooled back to RT and concentrated under reduced pressure to get the crude compound.
  • the crude compound was quenched using saturated NaHCCh solution and extracted with EtOAc.
  • the organic layer was dried over NasSOa, concentrated under vacuum, and purified by silica column chromatography (0-5% EtOAc in heptane), yielding the title compound (2.2 g).
  • Step B Preparation of 4-bromo-7-(5-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)isoquinoline (15-4).
  • 15-2 (3.0 g, 10.45 mmol) in 1,4-dioxane (30 ml.,) and H 2 O (6 mL) was added 15-3 (4.62 g, 12.54 mmol) and Na 2 CO 3 (2.21 g, 20.9 mmol).
  • the reaction mixture was purged with argon for 5 minutes, then Pd(dppf)Cl 2 -DCM added (852 mg, 1.045 mmol).
  • Step C Preparation of 7-(5-(6-methyIpyridin-2-yl)-1H-pyrazoI-4-yl)-4-(4-(piperidin-3- yl)phenyl)isoquinoline (557).
  • a vial of 15-4 (20 mg, 0.045 mmol), 15-5 (16.30 mg, 0.053 mmol), 2M sodium carbonate in water (66.8 ⁇ L), and Pd(dppf)Cl 2 (3.26 mg, 4.45 ⁇ mol) in degassed 1,4-dioxane (0.5 mL) was heated at 105 °C for 16 h. The reaction mixture was concentrated in vacuum.
  • TFA (0.5 mL) was added to the residue and stirred at 50 °C for 1 h. TFA was removed in vacuum and the residue was petrified by preparative HPLC chromatography using a gradient (5 to 35%) of acetonitrile in water with 0.05% trifluoroacetic acid to yield a TFA salt of the title compound (19.3 mg). [M+H] + calcd for C 29 H 27 N 5 445.23, found 446.2.
  • Example 16 Synthesis of 2-(4-(2-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)thieno['3,2-c]pyridin-7- yl) - 1 H-pyrazol - 1 -ylfethan- 1 -amine (294).
  • Step A Preparation of 7-bromothieno[3,2-c]pyridine (16-2). To a stirred solution of 16-1 (4.00 g,
  • Step B Preparation of 7-bronio-2-(trimethyistannyl)thieno[3,2-c]pyridine (16-3).
  • 16-2 (1.50 g, 7.01 mmol) in anhydrous THF (20.0 mL) was added EDA (5.26 mb, 10.5 mmol) at -78 °C under argon atmosphere.
  • EDA 5.26 mb, 10.5 mmol
  • the reaction was stirred at -78 °C for 1 h followed by addition of MesSnCl (2.09 g,
  • Step C Preparation of 7-bromo-2-(5-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)thieno[3,2-c]pyridine (16-5).
  • 16-4 686 mg, 1.86 mmol
  • toluene 12.0 mL
  • 16-3 700 mg, 1.86 mmol
  • tetralds(triphenylphosphine)palladium(0) 215 mg, 0.186 mmol.
  • Step D Preparation of 2-(4-(2-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)thieno[3,2-c]pyridin-7-yl)- 1 H-pyrazol-l-yl)ethan-1 -amine (294).
  • a vial of 16-5 (20 mg, 0.044 mmol), 16-6 (17.77 mg, 0.053 mmol), 2M sodium carbonate in water (65.9 ⁇ L), and Pd(dppf)Cl 2 (3.21 mg, 4.36 ⁇ mol) in degassed 1,4-dioxane (0.5 mL) was heated at 105 °C for 16 h. The reaction mixture was concentrated in vacuum.
  • Example 17 Synthesis of 2-(5-(6-methylpyridin-2-yl)-l-(tetrahydro-2H-pyran-2-yi)-1H-pyrazol-4 yl)-N-(4-(piperazin-1 -yl)phenyl)tbieno[3,2-c]pyridin-6-amine (664).
  • Step A Preparation of 6 -chloro -2-(trimethylstannyl)thieno[3,2-c]pyridine (17-2).
  • LDA 3.32 mL, 6.63 mmol
  • the reaction mixture was stirred at -78 °C for 30 min, then trimethyltin chloride ( 1.33 g, 6.63 mmol) added dropwise.
  • the reaction mixture was allowed to reach room temperature within 15 min, then stirred for 2h at rt.
  • the reaction mixture was diluted with EtOAc and saturated NH4CI solution and the organics were separated, dried over Na 2 SO 4 and concentrated under vacuo to give the crude product, used as such without further purification.
  • Step B Preparation of 6-chloro-2-(5-(6-methylpyridin-2-yl)-l-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)thieno[3,2-c]pyridine (17-3).
  • Pd(PPh 3 ) 4 (348 mg, 0.301 mmol) was added, and the reaction mixture was heated at 100 °C for 6 h.
  • the reaction mixture was passed through celite and washed with EtOAc (2-3 times).
  • Step C Preparation of 2-(5-(6-niethyIpyridin-2-yl)- ⁇ -(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)-N- (4-(piperazin-1-yl)phenyI)thieno[3,2-c]pyridin-6-amine (664).
  • Example 18 Synthesis of 2-(4-(2-(5 -(6-methylpyridin-2-yl)-1 H-pyrazol -4-yi)thieno[3,2-c]pyridin-7- yl)-1H-pyrazol-1-yl)ethan-1 -amine (603).
  • Step A Preparation of 4-bromothieno[2,3-c]pyridine (18-2).
  • 18-1 (3.0 g, 11.66 mmol)
  • DMSO:diphenyl ether (20 rnL:20 mL) was added and heated at 230 °C for 4h.
  • the reaction mixture was partitioned between EtOAc and water and the organics were separated, dried over Na2SOr and concentrated under vacuum to give a crude residue.
  • the residue was purified by silica column chromatography (0-100% EtOAc :hexanes) to yield the title compound (1.3 g) [M+H] + calcd for C 7 H 4 BrNS 212.92, found 213.9.
  • Step B Preparation of 4-bromo-2-(trimethylstannyl)thieno[2,3-c]pyridine (18-3).
  • 18-2 (980 mg., 4.60 mmol) in dry THF (10 ml) was added LDA (6.9 ml, 13.80 mmol) at -78 °C under nitrogen atmosphere.
  • LDA 6.9 ml, 13.80 mmol
  • the reaction mixture was stirred at -78 °C for 30 min.
  • MesSnCl (1 .37 g, 6.90 mmol in 5 ml THF) was added dropwise and the resulting mixture allowed to reach room temperature within 15 min, then stirred for 1 h.
  • Step C Preparation of 4-bromo-2-(5-(6-methylpyridin-2-yl)-l-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol -4-yi)thieno[2,3-c]pyridine (18-4).
  • To etude 18-3 (2.4 g, 3.18 mmol) in toluene (10 mi) was added 16- 4 (1.17 g, 3.18 mmol) at rt.
  • the reaction mixture was purged with N 2 for 10 min.
  • Pd(PPh 3 ) 4 (367 mg, 0.318 mmol) was added and stirred at 80 °C for 4 h.
  • Step D Preparation of 2-(4-(2-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)thieno[3,2-c]pyridin-7-yl)- 1H-pyrazol-1-yl)ethan-1-amine (603).
  • a vial of 18-4 (20 mg, 0.044 mmol), 16-6 (17.77 mg, 0.053 mmol), 2M sodium carbonate in water (65.9 ⁇ L), Pd(dppf)Cl 2 (3.21 mg, 4.36 ⁇ mol) in degassed 1,4-dioxane (0.5 mL) was heated at 1.05 °C for 1.6 h. The reaction mixture was concentrated in vacuum.
  • TFA (0.5 mL) was added to the residue and stirred at 50 °C for 1 h. TFA was removed in vacuum and the residue was purified by preparative HPLC chromatography using a gradient (2 to 20%) of acetonitrile in water with 0.05% trifluoroacetic acid to yield a TFA salt of the title compound (9.3 mg). [M+H] + calcd for C 21 H 19 N 7 S 401.14, found 402.1.
  • Example 19 Synthesis of 6-(5-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4- yl)-N-(4-(piperazin-1 -yl)phenyl)tbieno[2,3-d]pyrimidin-2-amine (667).
  • Step A Preparation of 6-bromo-2-chlorothieno[2,3-d]pyriinidine (19-2).
  • 19-1 1.0 g, 5.86 mmol
  • THF 10 mL
  • HBr 6 mL , 48% aq.
  • H 2 O 2 1-5 mL, 30% aq.
  • the resulting mixture was stirred for 16 h.
  • Sodium bicarbonate solution (20 mL) was added and the mixture was stirred for 5 min, then diluted with EtOAc. The organic layer was separated, dried over sodium sulfate, and. concentrated under vacuum to get the crude compound.
  • Step B Preparation of 2-chloro-6-(5-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)thieno[2,3-d]pyrimidine (19-4).
  • Step C Preparation of 6-(5-(6-methylpyridin-2-yl)- 1- (tetrahydro -2H-pyran-2-yl)-1 H-pyrazol -4-yl)-N- (4-(piperazin-1-yl)phenyl)thieno[2,3-d]pyrimidin-2-amine (667).
  • Example 20 Synthesis of 1-[6-[4-(4-methylthiazol-2-yl)-1H-imidazol-5-yl]-1,3-benzothiazol-2-yl]- 3-(3-pyridyl)urea (449).
  • Step A Preparation of tert-butyl (6-bromobenzo[d]tliiazol-2-yl)(tert-butoxycarbonyl)carbamate (20- 2).
  • DIPEA 21.4 g, 166 mmol
  • DMAP 2.0 g, 16.6 mmol
  • Boc 2 O 145 g, 663 mmol
  • the reaction mixture was stirred for 2 h at 70 °C.
  • the mixture was concentrated in vacuum to give the residue.
  • the residue was purified by trituration (PE, 100 mL) to afford 20-2 (28.0 g) as a yellow solid.
  • [M+HF calcd for C 17 H 21 BrN 2 O 4 S 429.04, found 429.1.
  • Step B Preparation of tert-butyl (6-((4-methylthiazol-2-yl)ethynyl)benzo[d]thiazol-2-yl)carbamate (20-4).
  • Step C Preparation of tert-butyl (6-(2-(4-methyithiazol -2-yl)-2-oxoacetyl)benzo[d]thiazol-2- ylicarbamate (20-5).
  • 20-4 2.0 g, 5.38 mmol
  • acetone 50 mL
  • MgSOr 2.25 mmol
  • H 2 O 30 mL
  • KMnO.4 3.3 g, 21.0 mmol
  • Step D Preparation of 6-(4-(4-methylthiazol-2-yl)- 1H-imidazol-5-yI)benzo[d]tliiazol-2 -amine (20-7).
  • a solution of 20-5 (2.5 g, 3.10 mmol), 20-6 (1.3 g, 9.30 mmol) and NH 4 OAc (1.4 g, 18.6 mmol) in AcOH (40 mL) was stirred for 2.5 h at 100 °C.
  • the mixture was extracted with EtOAc (400ml, x 3).
  • Step E Preparation of 1-[6-[4-(4-methylthiazol-2-yl)-1H-imidazol-5-yl]-1,3-benzothiazol-2-yl]-3-(3- pyridyl)urea (449).
  • 20-7 (30 mg, 0.086 mmol) and. DIPEA (44.9 , ⁇ L, 0.257 mmol) in DMF (857 ⁇ L) was added 20-8 (20.6 mg, 0.171 mmol).
  • the reaction mixture was allowed to stir for 1 h at 90 °C.
  • the reaction mixture was concentrated under vacuum.
  • Example 21 Biochemical ALK5 (TGF- ⁇ R1) Assay to Measure pKi.
  • Apparent pKi values for compounds of the present disclosure were determined using a recombinant human ALK5 (TGF- ⁇ Rl) protein (Product No. PR9075A or equivalent, Life Technologies) and a commercially -available kinase assay (LANCE® 1 (lanthanide chelate excite) Ultra ULightTM kinase assay. Product Nos. TRF0130-M and TRF02108-M, Perkin Elmer) as described below. [0346] The assays were performed in a 384-well piate (24 columns x 16 wells/rows).
  • An Echo® 5 550 Liquid Handler (Labcyte) was used to prepare various intermediate concentrations of compounds of the present disclosure in 100% DMSO. From the intermediate concentrations, a range of concentrations (from 10 pM to 25 pM corresponding to volumes up to 105 nL) were prepared and ejected into a final assay plate to be used to create individual dose response curves for each of the subject compounds. To a separate column within the assay plate, 105 nL of DMSO in each well was used to establish a maximum assay signal. Additionally, 105 nL of 100 pM SD-208, a selective TGF- ⁇ R1 inhibitor (Catalog #87624, Selleck Chemicals), was used in another column of wells to establish a minimal assay signal.
  • the enzyme mixture consisted of 250 pM ALK5 enzyme and 62.5 nM peptide substrate (LANCE® 5 (lanthanide chelate excite) Ultra ULightTM-DNA Topoisomerase 2-alpha (Th1l 342)) prepared in assay buffer (50 fflM HEPES, 10 mM MgCl 2 , 1 mM EGTA, 0.01% Tween-20, pH 7.5 at room temperature) with 2 mM DTT added prior to use. The plate was then sealed with an adhesive seal and. allowed, to equilibrate for 60 minutes at room temperature.
  • assay buffer 50 fflM HEPES, 10 mM MgCl 2 , 1 mM EGTA, 0.01% Tween-20, pH 7.5 at room temperature
  • pIC 50 values were determined from a 4-parameter robust fit model with GraphPad Prism V5 Software (GraphPad Software, Inc., La Jolla, CA). This model obtains pIC 50 values by fitting the sigmoidal dose- response (variable slope) equation to the data. Results were expressed as pIC 50 (negative logarithm, of IC 50 ) and subsequently converted to pK i (negative logarithm of dissociate constant, K i ) using the Cheng-Prusoff equation. The higher the value of pK i (lower value of Ki), the greater the inhibition of ALK5 activity. Certain compounds disclosed herein exhibited pK i values of greater than 8 or greater than 9 when tested in the biochemical ALK5 assay.
  • Table 2 shows biological activities of selected compounds in a biochemical ALK5 assay. Compound numbers correspond to the numbers and structures provided in Table 1 and Examples 1-20. Table 2 [0351] Example 22: Cellular ALK5 Potency Assay to Measure pIC 50 , Inhibition of TGF- ⁇ Stimulated pSMAD3 Formation in BEAS-2B Cells.
  • TGF- ⁇ -stimulated SM AD3 phosphorylation was measured in BEAS-2B cells, a human lung epithelial cell line. TGF- ⁇ signals through activin receptor-like kinase 5 (ALK5) immediately prior to SMAD3 phosphorylation.
  • ALK5 activin receptor-like kinase 5
  • BEAS-2.B cells were grown using 50% DMEM (Life Technologies) and 50% F-12. (Life Technologies).
  • BEAS-2B cells were seeded at 7,500 cells/well (25 ⁇ L/well) in a 384-well plate and cultured overnight. Before dosing, growth media was aspirated and the wells were rinsed with HBSS Buffer
  • Table 3 shows biological activities of selected compounds in a cellular ALK5 potency assay.
  • Compound numbers correspond to the numbers and structures provided in Table 1 and Examples 1-20.
  • Example 23 Cytotoxicity Measured by Premature Chromosome Condensation [15] (pCCis).
  • ATP cellular adenosine triphosphate
  • Beas2B cells a human lung epithelial cell line.
  • Levels of ATP are correlated with the viability of cells and are often measured to determine tire potential cytotoxicity of compounds.
  • CellTiter-GIo which lyses the cells and produces a luminescent signal proportional to the amount of ATP present, was used to determine the effect of test compound on cell viability.
  • Beas2B cells were grown in 50% DMEM (Life Technologies) and 50% F-12 (Life Technologies) media, supplemented with 10% Fetal Bovine Serum ( ATCC), 25 mM HEPES (Life Technologies), and I x
  • Pen-Strep (Life Technologies). Cells were cultured in a humidified incubator set at 37 °C, 5% CO 2 , and trypsinized using 0.25% Trypsin with 0.5% polyvinylpyrrolidone (PVP).
  • PVP polyvinylpyrrolidone
  • Beas2B cells were seeded at 500 cells/well (25 ⁇ L/well) in a 384- well plate and cultured overnight. Compounds were serially diluted in DMSO, then further diluted with growth media (40 ⁇ L/well) to create a compound plate 6x of the final assay concentration, at 0.6% DMSO. The diluted compounds were then added to the cells (5 ⁇ L/well) and incubated at 37 °C, 5% CO 2 for 48 hours. After the compound incubation, CeilTiter-Glo (Promega) was added directly to the cells (30 ⁇ L/mL).
  • the assay plate was sealed and shaken at 700 rpm for 15 minutes in a darkened environment, then centrifuged for 2 minutes at 1500 rpm to settle the lysate at the bottom of the well.
  • the effect of the compound on cell viability was determined through analysis of dose- dependent quantified changes in ATP from baseline (non -compound treated cells) and wells treated with 60 ⁇ M AT9283, a well -characterized cytotoxic compound. Data are expressed as pCC 15 (negative decadic logarithm CC 15 ) values. Certain compounds disclosed herein exhibited pCC 15 values of less than 6 or less than 5.5 when tested in Beas2B cells.
  • Table 4 shows cytotoxicities of selected compounds in a premature chromosome condensation assay.
  • Compound numbers correspond to the numbers and structures provided in Table 1 and. Examples 1-20.
  • Example 24 In Vitro Human Liver Microsome Intrinsic Clearance (HLM Clint).
  • liver microsomes were used for in vitro determination of hepatic clearance of compounds of the present disclosure.
  • a microsomal incubation cofactor solution was prepared with 100 mM potassium phosphate buffered to pH 7.4 (BD Biosciences, Woburn, MA) supplemented with 2 mM NADPH (Sigma-
  • HLM Cl int data was reported in units of pL/min/mg. See Riley, R.J., et al., Drug Metab. Dispos., 2005, September, 33(9), pp. 1304-1311. Certain compounds disclosed herein exhibited HLM Clint of greater than 50 ⁇ L/min/mg or greater than 100 pL/min/mg.
  • Example 25 Lung PK/PD.
  • mice were challenged via oral aspiration a second time with PBS vehicle or recombinant human TGF- ⁇ 1 protein (0.01 ⁇ g per animal dissolved in 1 part 4 mM HCl and 2 parts 3% glycerol in PBS).
  • animals were deeply anesthetized under isoflurane and euthanized via cervical dislocation.
  • Bronchoalveolar lavage fluid (BALF), plasma and left lung lobes were collected during harvest.
  • BALF Bronchoalveolar lavage fluid
  • BA bioanalytical
  • the lungs were flushed via the trachea with 0.7 mL of PBS 3 times.
  • the BALF which consists almost entirely of tissue-derived macrophages, was immediately centrifuged at 700xg’s for 15 minutes. After centrifugation, the supernatant was removed, the BALF was re-suspended in IX cell lysis buffer, and immediately frozen. Prior to BA submission, the BALF was dethawed and sonicated for 30 minutes on cold water to lyse open the cells
  • MSD Meso-scale Discovery
  • a coating antibody that binds the target protein(s) within the sample.
  • a primary antibody is used to bind the epitope of interest.
  • a secondary -antibody with a SULFO-TAG detection is used to allow for quantification of the epitope of interest.
  • the microplate is read via an electric pulse that causes the SULFO-TAG to emit light, which serves as the final read-out of the assay.
  • BSA bovine serum albumin
  • a one-way ANOVA (fisher’s uncorrected LSD) was used to compare all drug treated groups with the 3% glycerol/TGF- ⁇ group to determine if statistically significant differences are observed.
  • Percent pSMAD3 inhibition was calculated using the vehicle pSMAD3 as a baseline value and displayed as the final readout.
  • Dose-response curves were fitted with a 4-paramater non-linear regression algorithm; the minimum response was set to 0% pSMAD3 inhibition and the maximum response set to 100% pSMAD3 inhibition. Compound potencies were obtained from the regression and reported as ID50s.
  • Certain compounds disclosed herein exhibited (lung AUC 0-t ):(plasma AUC 0-t ) ratios of greater than 10, such as greater than 50, greater than 75, or greater than 100.
  • a compound intended for local delivery to the lung with minimal systemic exposure in some embodiments, exhibits a (lung AUC 0-t ): (plasma AUC 0-t ) ratio of greater than 50.
  • Certain compounds provided in Table 2 having pKi values of greater than 9.5 exhibited a (lung AUC 0-t ):(plasma AUC 0-t ) ratio of greater than 75.
  • Example 26 Cardiac PK/PD.
  • OA oral aspiration
  • mice were challenged via tail-vein intravenous injection with PBS vehicle or recombinant human TGF- ⁇ l protein (1 pg per animal dissolved in 1 part 4 mM HC1 and 2 parts 3% glycerol in PBS).
  • animals were deeply anesthetized under isoflurane and euthanized via cervical dislocation. Plasma, left lung lobes and whole hearts were collected during harvest.
  • Blood plasma was harvested as described above in the Lung PK/PD experiment. Whole hearts were processed in the same manners as left lung lobes in the Lung PK/PD experiment. Left lung lobes were homogenized in 500 ⁇ L of water and submitted for BA Analysis.
  • Heart samples were processed using MSD in the same manner as the left lung lobes above. Data analysis was performed in the same manners as the lung PK/PD experiment. Plasma, lung and heart drug concentrations were quantified.
  • One or more compounds disclosed herein are expected to suppress tumor growth in syngeneic cancer models when administered alone or in combination with an immunotherapeutic agent.
  • Six- to 8-week old BALB/c mice are used for In vivo efficacy studies in accordance with IACUC guidelines.
  • Commercially available 4T1 cells (0.5-2.0 x 1.0 4 cells/mouse) are implanted subcutaneously into the right flanks of BALB/c mice. When the tumor reaches a palpable size of approximately 8-10 mm.
  • CT26 cells 0.5-2.0 x 10 4 cells/mouse
  • a compound of the present disclosure at an appropriate amount and frequency (formulated in 3% glycerol in PBS: pH 4) via oral aspiration or intranasaliy
  • an immunotherapeutic agent e.g., pembrolizumab or durvalumab
  • a compound of the present disclosure and an immunotherapeutic agent each at an appropriate amount and frequency.
  • Body weight is measured twice weekly. Following 2- to 4- weeks of treatment, the lung and liver of each animal is harvested, and the number of metastatic cells in each tissue sample determined using a clonogenic metastasis assay. Cells may be further subjected to one or more of FACS analysis, T-cell function assay, and RNA extraction. It is expected that the animal group treated with one or more of the ALK5 inhibitors disclosed herein exhibits reduction in lung tumor burden. Activation of an immune response by the ALK5 inhibitor may stimulate both local and systemic antitumor T-cell activation, thus a reduction in liver tumor burden may also be observed.
  • a compound of the present disclosure When administered in combination with an immunotherapeutic agent, a compound of the present disclosure, such as a compound provided in Table 1, is expected to produce an increased reduction in lung tumor burden relative to the reduction in tumor burden observed in animals treated with either single agent alone.
  • the compounds described herein are expected to interact synergistically with an immunotherapeutic agent to suppress tumor growth and increase survival.
  • Example 28 Prophylactic study in murine DSS-induced intestinal fibrosis model.
  • One or more compounds disclosed herein are expected to slow, halt or reverse the progression of intestinal fibrosis in a murine colitis model.
  • Six to 8-week old male C57BL/6J mice are tagged and weighed.
  • the drinking water of the animals is treated with 2.5% dextran sulfate sodium (DSS) for 7 days to induce acute colitis, followed by 2 days of normal drinking water.
  • Three, 3 -week cycles of 2.5% DSS treatment (1 week of 2.5% DSS in water; 2 weeks of normal water) are then completed to induce intestinal fibrosis.
  • mice are treated with either vehicle control or a compound of the present disclosure at an appropriate amount and frequency via oral gavage (e.g., once daily).
  • the animals are sacrificed 9 weeks after the first DSS administration, then distal, mid and proximal sections of the colon harvested for histologic analysis, RNA extraction and cytokine measurement.
  • a compound of the present disclosure is expected to decrease ALK5 activity in the colon and to slow or prevent intestinal fibrosis as evidenced by one or more of (1) reduction in the ratio of colon weight to colon length; (2) reduction in deposition of extracellular matrix as observed by histology; (3) reduction in expression of collagen 1 (Colla1) and connective tissue growth factor (Ctgf) in colon tissue; and (4) reduction in production of TGF- ⁇ 1 and IL6 in the colon, relative to vehicle-treated controls.
  • Example 29 Efficacy study in murine DSS-induced intestinal fibrosis model.
  • One or more compounds disclosed herein are expected to slow, halt or reverse the progression of intestinal fibrosis in a murine colitis model.
  • Six to 8-week old male C57BL/6J mice are tagged and weighed.
  • the drinking water of the animals is treated with 2.5% dextran sulfate sodium (DSS) for 7 days to induce acute colitis, followed by 2 days of normal drinking water.
  • Three, 3 -week cycles of 2.5% DSS treatment (1 week of 2.5% DSS in water; 2 weeks of normal water) are then completed to induce intestinal fibrosis.
  • mice are treated with either vehicle control or a compound of the present disclosure at an appropriate amount and frequency via oral gavage (e.g., once daily). Animals are sacrificed at either 6, 9 or 12 weeks after the first DSS cycle, then distal, mid and proximal sections of the colon harvested for histologic analysis, RNA extraction and cytoki ne measurement.
  • a compound of the present disclosure is expected to decrease ALK5 activity in the colon and to slow, halt or reverse intestinal fibrosis as evidenced by one or more of (1) reduction in the ratio of colon weight to colon length; (2) reduction in deposition of extracellular matrix as observed by histology; (3) reduction in expression of collagen 1 (Coll a1) and connective tissue growth factor (Cigf) in colon tissue; and (4) reduction in production of TGF- ⁇ 1 and IL6 in the colon, relative to vehicle - treated controls.
  • Example 30 Efficacy study in adoptive T-cell transfer model of colitis.
  • One or more compounds disclosed herein e.g., a compound provided in Table 1 having an ALK5 pK i value of greater than 9.5, or greater than 10.5 (a measurement reflecting the ability of the compound to inhibit ALK5 activity, measured in accordance with Example 21), are expected to slow, halt or reverse the progression of intestinal fibrosis in an adoptive T-cell transfer model of colitis.
  • Six- to 8 -week old female CB17 SCID mice are tagged and weighed, then administered CD4* CD25" CD62L* naive T cells isolated from the spleens of Balb/C mice (IP; lx 10 6 cells) to induce colitis.
  • mice are treated with either vehicle control or a compound of the present disclosure at an appropriate amount and frequency via oral gavage (e.g., once daily). Animals are sacrificed 45 days after induction of colitis, then distal, mid and proximal sections of the coion harvested for histologic analysis, RNA extraction and cytokine measurement.
  • a compound of the present disclosure is expected to decrease ALK5 activity in the colon and to slow, halt or reverse intestinal fibrosis as evidenced by one or more of (1) reduction in the ratio of colon weight to colon length; (2) reduction in deposition of extracellular matrix as observed by histology; (3) reduction in expression of collagen 1 (Colla1) and connective tissue growth factor (Ctgf) in colon tissue; and (4) reduction in production of TGF- ⁇ 1 and IL6 in the colon, relative to vehicle- treated controls.
  • (1) reduction in the ratio of colon weight to colon length (2) reduction in deposition of extracellular matrix as observed by histology; (3) reduction in expression of collagen 1 (Colla1) and connective tissue growth factor (Ctgf) in colon tissue; and (4) reduction in production of TGF- ⁇ 1 and IL6 in the colon, relative to vehicle- treated controls.
  • Example 31 Efficacy study in monocrotaline model of severe pulmonary hypertension.
  • One or more compounds disclosed herein e.g., a compound provided in Table 1 having an ALK5 pK i value of greater than 9.5, or greater than 10.5 (a measurement reflecting the ability of the compound to inhibit ALK5 activity, measured in accordance with Example 21), are expected to slow, halt or reverse the progression of pulmonary hypertension in a monocrotaline (MCT) model of severe pulmonary hypertension.
  • MCT monocrotaline
  • a compound of the present disclosure is expected to decrease ALK5 activity in the lung and slow, halt or reverse the progression of pulmonary hypertension as evidenced by one or more of (1) reduction in systolic pulmonary arterial pressure; (2) reduction in right ventricular (RV) systolic pressure; (3) reduction in RV diastolic pressure; (4) increase in cardiac output; (5) reduction in RV hypertrophy; (6) reduction in pSmad2 or pSmad3 staining within vascular and/or alveolar cells; (7) reduction in medial thickness; (8) reduction in vascular smooth muscle cell proliferation; (9) reduction in vascular smooth muscle hypertrophy; and (10) reduction in expression of matrix metalloproteinase (MMP)-2 and/or MMP-9
  • MMP matrix metalloproteinase

Abstract

The present disclosure provides inhibitors of activin receptor-like kinase 5 (ALK5). Also disclosed are methods to modulate the activity of ALK5 and methods of treatment of disorders mediated by ALK5.

Description

BICYCLIC INHIBITORS OF ALK5 AND METHODS OF USE
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to United States Provisional Application No. 63/202,062, filed May 26, 2021, which is hereby incorporated, by reference in its entirety.
BACKGROUND
[0002] Human fibrotic diseases such as systemic sclerosis (SSc), sclerodermatous graft vs. host disease, nephrogenic system fibrosis, and radiation-induced fibrosis, as well as cardiac, pulmonary, skin, liver, bladder and kidney fibrosis, constitute a major health problem. These diseases often progress to organ dysfunction with eventual organ failure and death due to lack of treatment available, mainly because the etiologic mechanisms of runaway fibrosis are complex, heterogeneous, and difficult to decipher. Activated myofibroblasts may be responsible for replacing normal tissues with nonfunctional fibrotic tissue. Therefore, signaling pathways responsible for stimulating profibrotic reactions in myofibroblasts have potential as targets for development of therapies to treat fibrotic diseases.
[0003] Normal tissue repair involves fibrotic reactions through homeostatic regulatory mechanisms. Uncontrolled fibrosis, however, may result in excess deposition of the extracellular matrix (ECM) macromolecules in interstitial space that stiffens over time. There are many sites along the molecular pathway leading up to myofibroblast activation, including, but not limited to, transforming growth factor-p (TGF-β) and bone morphogenic protein (BMP) signaling pathways. Of importance in this disclosure is the pathway involving transforming growth factor-β (TGF-β), TGF-β receptor I (TGF-βRI), and TGF-β receptor II (TGF- pRII).
[0004] TGF-β signaling is typically initiated by binding of a TGF-β ligand to a TGF-βRII. This in turn may recruit and phosphorylate TGF-βRI, also known as the activin receptor-like kinase 5 (ALK5). Once phosphorylated, ALK5 typically adopts an active conformation and is free to associate with and phosphorylate Smad2 or Smad3. Once phosphorylated, Smad 2 and 3 proteins then may form heterodimeric complexes with Smad4 which can translocate across the nuclear membrane and modulate Smad-mediated gene expression, including, for example, the production of collagen. Smad proteins are intracellular regulators of transcription and therefore may serve as modulators of TGF-β-regulated genes involving, inter alia, cell cycle arrest in epithelial and hematopoietic cells, control of mesenchymal cell proliferation and differentiation, wound healing, extracellular matrix production, immunosuppression, and carcinogenesis. [0005] ALK5 is believed to be the most relevant of the activin-like kinases (ALKs) in the fibrotic process (Rosenbloom, et al., Fibrosis: Methods and. Protocols, Methods in Molecular Biology, 2017, Vol. 1627, Chapter 1, pp. 1-21). Several small molecules have been developed to inhibit the activity of ALK5 for various therapeutic indications, related mostly to oncology (see Yingling, et al., Nature Reviews: Drug Discovery, December 2004, Vol. 3, pp. 1011-1022). SUMMARY
[0006] One of the main problems with ALK5 inhibitors developed to date is that these molecules have been associated with ventricular or cardiac remodeling in preclinical safety studies resulting from significant systemic exposure from oral administration. In view of the foregoing, a need exists for small molecules that target ALK5 and for use of such compounds in the treatment of various diseases, such as cancer and fibrosis, while limiting adverse side effects. The present disclosure provides these and other related advantages. One objective of the present disclosure is to deliver a potent ALK5 inhibitor locally with minimal systemic exposure in. order to address any unintended and unwanted systemic side effects of ALK5 inhibition during treatment. Therefore, in some aspects, the present disclosure provides inhaled, long-acting and lung-selective ALK5 inhibitors for the treatment of idiopathic pulmonary fibrosis. Compounds of the present disclosure may be used to treat other diseases, including, but not limited to, pulmonary fibrosis, liver fibrosis, renal glomerulosclerosis, and cancer. Compounds of the present disclosure may be used as a monotherapy or co- dosed with other therapies, whether delivered by inhalation, orally, intravenously, subcutaneously, or topically.
[0007] In certain aspects the present disclosure provides a compound of Formula (I):
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof, wherein:
X and ¥ are each independently selected from CH and N;
A is a 9- or 10-membered bicyclic heteroaryl group selected from:
Figure imgf000003_0002
Figure imgf000003_0003
and
Figure imgf000003_0004
B is selected from phenyl, pyridyl, and thiazolyl;
RA is selected from:
-NO2, -CN, -SR1, -NR'R1. -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)OR5, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2;
C2-10 alkenyl and C2-10 alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2 -P(O)(R1)2 =O, =S, =N(R1), C3-12 carbocycle, and 3- to 12-membered heterocycle;
C1-10 alkyl, -N(R1)- C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, C3. 12 carbocycle, and 3- to 12-membered heterocycle;
C9-10 aryl, C3-12 cycloalkyl, and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1-,OC (O)R1, -OC (O)OR1,-OC (O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(RT), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
RB is independently selected at each occurrence from halogen, -CN, -NH2, -NHCH3, -NHCH2CH3, -C(O)CH3, -OH-,OC H3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CH2F, -CHF2, -CF3, C3.4 carbocycle, and 3- to 4-membered heterocycle; n is an integer from 0 to 3;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -SCH3, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -N(CH2CH3)2, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -OC(O)CH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle;
R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1;
R4 is independently selected at each occurrence from C1-3 alkyl, C2-6 alkenyl, C2-6 alkynyl, CH2-(C3-12 cycloalkyl), CH2-(C5-12 cycloalkenyl), C2-3 alkyl-(C3-12 carbocycle), and C1-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH(CH3)2, -C(CH3)3, C3- 12 cycloalkyl, and 3- to 6-membered heterocycle; and
R5 is independently selected at each occurrence from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6- membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6- membered heterocycle.
[0008] In certain aspects the present disclosure provides a compound of Formula (I):
Figure imgf000005_0001
or a pharmaceutically acceptable salt thereof, wherein:
X and Y are each independently selected from CH and N;
A is a 9- or 10-membered bicyclic heteroaryl group selected from:
Figure imgf000005_0002
Figure imgf000006_0001
Figure imgf000006_0002
and
Figure imgf000006_0003
B is selected from phenyl, pyridyl, and thiazolyl;
RA is selected from:
-NO2, -CN, -SR1, -NR'R1. -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)OR5, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2; C2-10 alkenyl and C2-10 alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), C3-12 carbocycle, and 3- to 12-membered heterocycle; C1-10 alkyl, -N(R1)- C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -O(OC)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, C3- 12 carbocycle, and 3- to 12-membered heterocycle;
C9-10 aryl, C3-12 cycloalkyl, and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -O(OC)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R'), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and N(R1)- (C3-12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
RB is independently selected at each occurrence from halogen, -CN, -NH2, -NHCH3, -NHCH2CH3, -C(O)CH3, -OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CH2F, -CHF2, -CF3, C3-4 carbocycle, and 3- to 4-membered heterocycle; n is an integer from 0 to 3;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle;
R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1;
R4 is independently selected at each occurrence from C1-3 alkyl, C2-6 alkenyl, C2-6 alkynyl, CH2-(C3-12 cycloalkyl), CH2-(C5-12 cycloalkenyl), C2-3 alkyl-(C3-12 carbocycle), and C1-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH), -OCH3 , -OOCCH2CH3, -CH(CH3)2, -C(CH3)3, C3-
12 cycloalkyl, and 3- to 6-membered heterocycle; and
R5 is independently selected at each occurrence from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6- membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OHC3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6- membered heterocycle.
[0009] For a compound, of Formula (I), X may be CH and Y may be N. In some embodiments, X is N and Y is CH. In some embodiments, X and Y are each N. [0010] For a compound, of Formula (I), B may be selected from
Figure imgf000008_0001
and . In some
Figure imgf000008_0002
embodiments, B is
Figure imgf000008_0003
. In some embodiments, B is
Figure imgf000008_0004
[0011] A compound of Formula (I) may be selected from
Figure imgf000008_0005
and
Figure imgf000008_0006
[0012] For a compound of Formula (I), A may be selected from
Figure imgf000008_0007
Figure imgf000008_0008
and
Figure imgf000008_0009
. In some embodiments, A is selected from
Figure imgf000008_0010
Figure imgf000008_0011
and . In some embodiments, A is selected from and
Figure imgf000008_0012
Figure imgf000008_0013
. In some embodiments, A is selected from.
Figure imgf000008_0015
and . In some
Figure imgf000008_0014
Figure imgf000008_0016
embodiments, A is selected from
Figure imgf000008_0017
, and
Figure imgf000008_0018
. In some embodiments, A is selected from , and .
Figure imgf000008_0019
Figure imgf000008_0020
[0013] For a compound of Formula (I), RA may be selected from:
-NR1R4, -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3;
C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle;
C10 aryl, C3-12 cycloalkyl, and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
[0014] For a compound of Formula (I), RA may be selected from:
-NR1R4, -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3;
C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, C3-12 carbocycle, and 3- to 12- membered heterocycle; C3-12 cycloalkyl and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
[0015] In some embodiments, for a compound of Formula (I), RA is selected from 3- to 12-membered heterocycle, -N(R‘)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl. In some embodiments, RA is selected from -C(O)OR3, -NR1C(O)R1, and -NR1C(O)N(R1)2. In some embodiments, R1 is independently selected at each occurrence from hydrogen; and CM; alkyl, 1- to 6-membered heteroalkyl, C0- 3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -N(CH3)2, -C(O)OCH2CH3, =O, and -CH3. In some embodiments, R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -NH2 and -CH3. In some embodiments, R5 is independently selected at each occurrence from C1-6 alkyl and C0-3 alkyl-(3- to 12-membered heterocycle).
[0016] In some embodiments, for a compound of Formula (I), RA is selected from -NH-(3- to 12-membered heterocycle) and -NH-(C3-12 carbocycle), each of which is optionally substituted with C0-3 alkyl-(3- to 12- membered heterocycle), wherein the C0-3 alkyl-(3- to 12-membered heterocycle) is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -CH3, and -CF3.
[0017] In some embodiments, for a compound of Formula (I):
RA is selected from:
-C(O)OR5, -NHC(O)NHR1;
C1-10 alkyl and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1 and -CH2NHR1;
C5-12 cycloalkenyl, optionally substituted with -NHR1; and
3- to 12-membered heterocycle, -NH-(3- to 12-membered heterocycle), and -NH-(C3.
12 carbocycle), each of which is independently optionally substituted with one or more substituents selected from halogen, -CH2NHR1, and R1;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by -NH2; and
R5 is independently selected at each occurrence from C1-6 alkyl and C0-3 alkyl-(3- to 12-membered heterocycle).
[0018] In some embodiments, for a compound of Formula (I):
A is selected from and
Figure imgf000010_0001
Figure imgf000010_0002
B is selected from
Figure imgf000010_0003
and
Figure imgf000010_0004
; and
RA is selected from:
-NR'R1. -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3;
C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; C3-12 cycloalkyl and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
[0019] In some embodiments, for a compound of Formula (I):
A is
Figure imgf000011_0001
B is selected from
Figure imgf000011_0002
and
Figure imgf000011_0003
; and
RA is selected from -NH-(3- to 12-membered heterocycle) and -NH-(C3-12 carbocycle), each of which is optionally substituted with C0-3 alkyl-(3- to 12-membered heterocycle), wherein the C0-3 alkyl-(3- to 12- membered heterocycle) is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -CH3, and -CF3.
[0020] In some embodiments, for a compound of Formula (I):
A is selected from
Figure imgf000011_0004
, and
Figure imgf000011_0005
B is selected from , and
Figure imgf000011_0007
; and
Figure imgf000011_0006
RA is selected from:
-NR'R1. -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3;
C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; and 3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3- c12arbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
[0021] In some embodiments, for a compound of Formula (I):
A is selected from
Figure imgf000012_0001
and
Figure imgf000012_0002
B is selected from and and
Figure imgf000012_0003
Figure imgf000012_0004
RA is selected from:
-NR1R4, -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3;
C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; C3-12 cycloalkyl and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
[0022] In some embodiments, the present disclosure provides a compound of Formula (I) wherein B is
.Me
[0023] In certain aspects, the present disclosure provides a substantially pure stereoisomer of a compound or salt disclosed herein. In certain aspects, the present disclosure provides a substantially pure stereoisomer of a compound disclosed herein. The stereoisomer may be provided in at least 90% enantiomeric excess. [0024] In certain aspects, the present disclosure provides a compound selected from Table 1, or a pharmaceutically acceptable salt thereof. In certain aspects, the present disclosure provides a compound selected from Table la, or a pharmaceutically acceptable salt thereof.
[0025] In certain aspects, the present disclosure provides a conjugate of the formula:
Figure imgf000013_0001
wherein:
A' is an antibody construct or targeting moiety;
L1 is a linker;
D' is a compound or salt disclosed herein; and p is an integer from 1 to 20.
[0026] In certain aspects, the present disclosure provides a pharmaceutical composition comprising a compound disclosed herein and a pharmaceutically acceptable carrier. The pharmaceutical composition may be formulated for inhalation.
[0027] In certain aspects, the present disclosure provides a method of inhibiting ALK5, comprising contacting ALK5 with an effective amount of a compound or salt disclosed herein. In certain aspects, the present disclosure provides a method of treating an ALK5 -mediated disease or condition in a subject, comprising administering to the subject a therapeutically effective amount of a compound or salt disclosed herein. In practicing any of the subject methods, the disease or condition may be selected from fibrosis, alopecia, and cancer. In some embodiments, the disease or condition is fibrosis. In some embodiments, the present disclosure provides a method of treating fibrosis, comprising administering to a patient a therapeutically effective amount of a compound or salt disclosed herein. The fibrosis may be selected from systemic sclerosis, nephrogenic systemic fibrosis, organ-specific fibrosis, fibrosis associated with cancer, cystic fibrosis, and fibrosis associated with an autoimmune disease. Optionally, the organ-specific fibrosis is selected from cardiac fibrosis, kidney fibrosis, pulmonary fibrosis, liver fibrosis, portal vein fibrosis, skin fibrosis, bladder fibrosis, intestinal fibrosis, peritoneal fibrosis, myelofibrosis, oral submucous fibrosis, and retinal fibrosis. In some embodiments, the organ-specific fibrosis is intestinal fibrosis. Optionally, the pulmonary fibrosis is selected from idiopathic pulmonary fibrosis (IPF), familial pulmonary' fibrosis (FPF), interstitial lung fibrosis, fibrosis associated with asthma, fibrosis associated with chronic obstructive pulmonary disease (COPD), silica -induced fibrosis, asbestos -induced fibrosis and. chemotherapy -induced lung fibrosis. Optionally, the pulmonary fibrosis is idiopathic pulmonary fibrosis (IPF). In some embodiments, the pulmonary fibrosis was induced by a viral infection.
[0028] In practicing any of the subject methods, the disease or condition, may be cancer, optionally wherein the cancer is selected from breast cancer, coion cancer, prostate cancer, lung cancer, hepatocellular carcinoma, glioblastoma, melanoma, and pancreatic cancer. In some embodiments, the cancer is lung cancer, optionally non-small cell lung cancer. A method of the subject disclosure may further comprise administering a second therapeutic agent. Optionally, the second therapeutic agent is an immunotherapeutic agent, such as a PD-1 inhibitor or a CTLA-4 inhibitor. In some embodiments, the immunotherapeutic agent is selected from pembrolizumab and durvalumab. A method of the present disclosure may further comprise administering an effective amount of radiation. In practicing any of the subject methods, the compound or salt disclosed herein may be administered by inhalation.
[0029] In certain aspects, the present disclosure provides a compound disclosed herein for use in treating fibrosis. In certain aspects, the present disclosure provides the use of a compound disclosed herein for the manufacture of a medicament for treating fibrosis.
INCORPORATION BY REFERENCE
[0030] All publications and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.
DETAILED DESCRIPTION
[0031] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this disclosure belongs.
[0032] Chemical structures are named herein according to IUPAC conventions as implemented in ChemDraw® software (Perkin Elmer, Inc., Cambridge, MA).
[0033] As used in the specification and claims, the singular forms “a”, “an” and “the” include plural references unless the context clearly dictates otherwise.
[0034] The term “Cx-y” or “Cx-Cy” when used in conjunction with a chemical moiety, such as alkyl, alkenyl, or alkynyl, is meant to include groups that contain from x to y carbons in the chain. For example, the term “Cx-y alkyl” refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups, that contain from x to y carbons in the chain.
[0035] “Alkyl" refers to substituted or unsubstituted saturated hydrocarbon groups, including linear and branched alkyl groups. An alkyl group may contain from one to twelve carbon atoms (e.g., C1-12 alkyl), such as one to eight carbon atoms (C1-8 alkyl) or one to six carbon atoms (C1-6 alkyl). Exemplary alkyl groups include methyl, ethyl, n-propyi, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, hexyl, septyl, octyl, nonyl, and decyl. An alkyl group is attached to the rest of the molecule by a single bond. Unless stated otherwise specifically in the specification, an alkyl group is optionally substituted by one or more substituents such as those substituents described herein.
[0036] “Haloalkyl” refers to an alkyl group that is substituted by one or more halogens. Exemplary haloalkyl groups include trifluoromethyl, di fluoromethyl, trichloro methyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl, 3- bromo-2-fluoropropyl , and 1,2-dibromoethyl.
[0037] “Alkenyl” refers to substituted or unsubstituted, hydrocarbon groups, including linear and branched alkenyl groups, containing at least one double bond. An alkenyl group may contain from two to twelve carbon atoms (e.g., C2-12 alkenyl), such as two to eight carbon atoms (C2-8 alkenyl) or two to six carbon atoms (C2-6 alkenyl). Exemplary alkenyl groups include ethenyl (i.e., vinyl), prop-1-enyl, but-1-enyl, pent-1-enyl, penta- 1, 4-dienyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group is optionally substituted by one or more substituents such as those substituents described herein.
[0038] “Alkynyl” refers to substituted or unsubstituted hydrocarbon groups, including linear and branched alkynyl groups, containing at least one triple bond. An alkynyl group may contain from two to twelve carbon atoms (e.g., C2-12 alkynyl), such as two to eight carbon atoms (C2-8 alkynyl) or two to six carbon atoms (C2-6 alkynyl). Exemplary alkynyl groups include ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like.
Unless stated otherwise specifically in the specification, an alkynyl group is optionally substituted by one or more substituents such as those substituents described herein.
[0039] “Alkylene” or “alkylene chain” refers to substituted or unsubstituted divalent saturated hydrocarbon groups, including linear alkylene and branched alkylene groups, that contain from one to twelve carbon atoms (e.g., C1-12 alkylene), such as one to eight carbon atoms (C1-8 alkylene) or one to six carbon atoms (C1-6 alkylene). Exemplary alkylene groups include methylene, ethylene, propylene, and n-butylene. Similarly, “alkenylene” and “alkynylene" refer to alkylene groups, as defined above, which comprise one or more carbon-carbon double or triple bonds, respectively. The points of attachment of the alkylene, alkenylene or alkynylene chain to the rest of the molecule can be through one carbon or any two carbons of the chain. Unless stated otherwise specifically in the specification, an alkylene, alkenylene, or alkynylene group is optionally substituted by one or more substituents such as those substituents described herein.
[0040] “Heteroalkyl”, “heteroalkenyl” and “heteroalkynyl” refer to substituted or unsubstituted alkyl, alkenyl and alkynyl groups, respectively, in which one or more, such as 1, 2 or 3, of the carbon atoms are replaced with a heteroatom, such as O, N, P, Si, S, or combinations thereof. Any nitrogen, phosphorus, and sulfur heteroatoms present in the chain may optionally be oxidized, and any nitrogen heteroatoms may optionally be quaternized. If given, a numerical range refers to the chain length in total. For example, a 3- to 8-membered heteroalkyl group has a chain length of 3 to 8 atoms. Connection to the rest of the molecule may be through either a heteroatom or a carbon in the heteroalkyl, heteroalkenyl or heteroalkynyl chain. Unless stated otherwise specifically in the specification, a heteroalkyl, heteroalkenyl, or heteroalkynyl group is optionally substituted by one or more substituents such as those substituents described herein.
[0041] “Heteroalkylene”, “heteroalkenylene” and “heteroalkynylene” refer to substituted or unsubstituted alkylene, alkenylene and. alkynylene groups, respectively, in which one or more, such as 1, 2, or 3, of the carbon atoms are replaced with a heteroatom, such as 0, N, P, Si, S, or combinations thereof. Any nitrogen, phosphorus, and sulfur heteroatoms present in the chain may optionally be oxidized, and any nitrogen heteroatoms may optionally be quaternized. If given, a numerical range refers to the chain length in total. For example, a 3- to 8-membered heteroalkylene group has a chain length of 3 to 8 atoms. The points of attachment of the heteroalkylene, heteroalkenylene or heteroalkynylene chain to the rest of the molecule can be through either one heteroatom or one carbon, or any two heteroatoms, any two carbons, or any one heteroatom and any one carbon in the heteroalkylene, heteroalkenylene or heteroalkynylene chain. Unless stated otherwise specifically in the specification, a heteroalkylene, heteroalkenylene, or heteroalkynylene group is optionally substituted by one or more substituents such as those substituents described herein. [0042] “Carbocycle” refers to a saturated, unsaturated, or aromatic ring in which each atom of the ring is a carbon atom. Carbocycle may include C3-10 monocyclic rings, C5-12 bicyclic rings, C6-12 spirocyclic rings, and ( C6-12 bridged rings. Each ring of a bicyclic carbocycle may be selected from saturated, unsaturated, and aromatic rings. In some embodiments, the carbocycle is a C6-12 aryl group, such as C6-10 aryl. In some embodiments, the carbocycle is a C6-12 cycloalkyl group. In some embodiments, the carbocycle is a C6-12 cycloalkenyl group. As used herein, “cycloalkenyl” refers to a non-aromatic ring containing at least one double bond, wherein each atom of the ring is a carbon atom. In an exemplary embodiment, an aromatic ring, e.g., phenyl, may be fused to a saturated or unsaturated ring, e.g., cyclohexane, cyclopentane, or cyclohexene. Any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits, are included in the definition of carbocycle. Exemplary carbocycles include cyclopentyl, cyclohexyl, cyclohexenyl, adamantyl, phenyl, indanyl, and naphthyl. Unless stated otherwise specifically in the specification, a carbocycle is optionally substituted by one or more substituents such as those substituents described herein.
[0043] “Heterocycle” refers to a saturated, unsaturated, or aromatic ring comprising one or more heteroatoms, for example 1, 2, or 3 heteroatoms selected from O, S and N. Heterocycles include 3- to 10- membered monocyclic rings, 6- to 12-membered bicyclic rings, 6- to 12-membered spirocyclic rings, and 6- to 12-membered bridged rings. Each ring of a bicyclic heterocycle may be selected from saturated, unsaturated, and aromatic rings. The heterocycle may be attached to the rest of the molecule through any atom of the heterocycle, valence permitting, such as a carbon or nitrogen atom of the heterocycle. In some embodiments, the heterocycle is a 5- to 10-membered heteroaryl group, such as 5- or 6-membered heteroaryl. In some embodiments, the heterocycle is a 3- to 12-membered heterocycloalkyl group. In an exemplary embodiment, a heterocycle, e.g., pyridyl, may be fused to a saturated or unsaturated ring, e.g., cyclohexane, cyclopentane, or cyclohexene. Exemplary heterocycles include pyrrolidinyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, piperidinyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, thiophenyl, oxazolyl, thiazolyl, morpholinyl, indazolyl, indolyl, and quinolinyl. Unless stated otherwise specifically in the specification, a heterocycle is optionally substituted by one or more substituents such as those substituents described herein. [0044] “Heteroaryl” refers to a 5- to 12-membered aromatic ring that comprises at least one heteroatom, such as 1, 2, or 3 heteroatoms, selected from O, S, and N. As used herein, the heteroaryl ring may be selected from monocyclic or bicyclic — including fused, spirocyclic and bridged ring systems — wherein at least one of the rings in the ring system, is aromatic. The heteroatom(s) in the heteroaryl may optionally be oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heteroaryl may be attached to the rest of the molecule through any atom of the heteroaryl, valence permitting, such as a carbon or nitrogen atom of the heteroaryl. Examples of heteroaryl groups include, but are not limited to, azepinyl, benzimidazolyl, benzisothiazolyl, benzisoxazolyl, benzofuranyl, benzothiazolyl, benzothiophenyl, benzoxazolyl, furanyl, imidazolyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, purinyl, pyrazinyl, pyrazolidinyl, pyrazolyl, pyridazinyl, pyridazolyl, pyridyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydroquinolinyl, thiadiazolyi, thiazolyl, and thienyl groups. Unless stated otherwise specifically in the specification, a heteroaryl is optionally substituted by one or more substituents such as those substituents described herein.
[0045] Unless stated otherwise, hydrogen atoms are implied in structures depicted herein as necessary to satisfy the valence requirement.
[0046] A waved line drawn across a bond or a dashed bond are used interchangeably herein to
Figure imgf000017_0001
Figure imgf000017_0002
denote where a bond, disconnection or attachment occurs. For example, in the structure
Figure imgf000017_0003
, Ra is attached to the para position of a fluorophenyl ring through a single bond. If Ra is 2-pyridine as in
Figure imgf000017_0004
, then Ra may be depicted as or
Figure imgf000017_0005
Figure imgf000017_0006
[0047] The term “substituted” refers to moieties having substituents replacing a hydrogen on one or more carbons or heteroatoms of the structure. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance 'with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and iron-aromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this disclosure, heteroatoms such as nitrogen may have any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
[0048] A compound disclosed herein, such as a compound of Formula (I), is optionally substituted by one or more, such as 1 , 2, or 3 substituents selected from: halogen, -NO2, -CN, -OR1, -SR1, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1);
C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, and -N(R1)- C1-10 alkyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), C3-12 carbocycle, and 3- to 12-membered heterocycle; and C3-12 carbocycle, 3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3 , -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle; and
R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1.
[0049] In some embodiments, a compound disclosed herein, such as a compound of Formula (I), is optionally substituted by one or more, such as 1, 2, or 3 substituents selected from: halogen, -NO2, -CN, -OR1, -SR1, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1);
C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, and -N(R1)-C1-10 alkyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, =O, =S, =N(R1), C3-12 carbocycle, and 3- to 12-membered heterocycle; and C3-12 carbocycle, 3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle is independently optionally substituted with one or more substituents selected from halogen, =O, =S, =N(R1), C1-6 alkyl, C1-6 haloalkyl,C2-6 alkenyl, and C2-6 alkynyl;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, H3, --OOCCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle; and
R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1.
[0050] In some embodiments, a compound disclosed herein, such as a compound of Formula (I), is optionally substituted by one or more, such as 1, 2 or 3 substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -NHCH2CH3, =O, -OH, -OCH3, and -OCH2CH3; and C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, 1- to 6- membered heteroalkyl, C3-12 carbocycle, and 3- to 12-membered heterocycle, each of which is optionally substituted by halogen, -CN, -NO2, -NH2, -NHCH3, -NHCH2CH3, =O, -OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, or 3- to 6 -membered heterocycle.
[0051] It will be understood by those skilled in the art that substituents can themselves be substituted, if appropriate. Unless specifically stated as “unsubstituted”, references to chemical moieties herein are understood to include substituted variants. For example, reference to a “heteroaryl” group or moiety implicitly includes both substituted and unsubstituted variants.
[0052] Where bivalent substituent groups are specified herein by their conventional chemical formulae, written from left to right, they are intended to encompass the isomer that would result from writing the structure from right to left, e.g., -CH2O- is also intended to encompass to -OCH2-
[0053] “Optional” or “optionally” means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. For example, an “optionally substituted” group may be either unsubstituted or substituted.
[0054] Compounds of the present disclosure also include crystall ine and amorphous forms of those compounds, pharmaceutically acceptable salts, and active metabolites having the same type of activity, including, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrat.es), conformational polymorphs, amorphous forms of the compounds, and mixtures thereof.
[0055] The compounds described herein may exhibit their natural isotopic abundance, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. All isotopic variations of the compounds of the present disclosure, whether radioactive or not, are encompassed within the scope of the present disclosure. For example, hydrogen has three naturally occurring isotopes, denoted 1H (protium), 2H (deuterium), and 3H (tritium). Protium is the most abundant isotope of hydrogen in nature.
Enriching for deuterium may afford certain therapeutic advantages, such as increased in vivo half-life and/or exposure, or may provide a compound useful for investigating in vivo routes of drag elimination and metabolism. Examples of isotopes that may be incorporated into compounds of the present disclosure include, but are not limited to, 2H, 3H, 13C, 14C, 15N, 18O, 17O, 35S, 36CI, and 18F. Of particular interest are compounds of Formula (I) enriched in tritium or carbon- 14, which can be used, for example, in tissue distribution studies; compounds of the disclosure enriched in deuterium especially at a site of metabolism, resulting, for example, in compounds having greater metabolic stability; and compounds of Formula (I) enriched in a positron emitting isotope, such as 11C, 18F, 15O and 13N, which can be used, for example, in Positron Emission Topography (PET) studies. Isotopically-enriched compounds may be prepared by conventional techniques well known to those skilled in the art.
[0056] As used herein, the phrase “of the formula” or “having the formula” or “having the structure” is not intended to be limiting and is used in the same way that the term “comprising” is commonly used. For example, if one structure is depicted, it is understood that all stereoisomer and tautomer forms are encompassed, unless stated otherwise.
[0057] Certain compounds described herein contain one or more asymmetric centers and can thus give rise to enantiomers, diastereomers, and other stereoisomeric forms, the asymmetric centers of which can be defined, in terms of absolute stereochemistry, as (R)- or (S)-. In some embodiments, in order to optimize the therapeutic activity of the compounds of the disclosure, e.g., to treat fibrosis, it may be desirable that the carbon atoms have a particular configuration (e.g., (R,R), (5,5), (S,R), or (R,S)') or are enriched in a stereoisomeric form having such configuration. The compounds of the disclosure may be provided as racemic mixtures. Accordingly, the disclosure relates to racemic mixtures, pure stereoisomers (e.g., enantiomers and diastereoisomers), stereoisomer -enriched mixtures, and the like, unless otherwise indicated. When a chemical structure is depicted herein without any stereochemistry, it is understood that all possible stereoisomers are encompassed by such structure. Similarly, when a particular stereoisomer is shown or named herein, it will be understood by those skilled in the art that minor amounts of other stereoisomers may be present in the compositions of the disclosure unless otherwise indicated, provided that the utility of the composition as a whole is not eliminated by the presence of such other isomers. Individual stereoisomers may be obtained by numerous methods that are known in the art, including preparation using chiral synthons or chiral reagents, resolution using chiral chromatography using a suitable chiral stationary phase or support, or by chemically converting them into diastereoisomers, separating the diastereoisomers by conventional means such as chromatography or recrystallization, then regenerating the original stereoisomer.
[0058] Additionally, where applicable, all cis-trans or E/Z isomers (geometric isomers), tautomeric forms and topoisomeric forms of the compounds of the disclosure are included within the scope of the disclosure unless otherwise specified. [0059] The term “tautomer”, as used herein, refers to each of two or more isomers of a compound that exist in equilibrium and which readily interconvert. For example, one skilled in the art would readily understand that 1,2,3-triazole exists in two tautomeric forms:
Figure imgf000021_0001
Unless otherwise specified, chemical entities described herein are intended to include all possible tautomers, even when a structure depicts only one of them. For example, even though a single tautomer of a compound of Formula (I-G) may be depicted herein, the disclosure is intended to include all possible tautomers, including:
Figure imgf000021_0002
[0060] The term “pharmaceutically acceptable” refers to a material that is not biologically or otherwise unacceptable when used in the subject compositions and methods. For example, the term “pharmaceutically acceptable carrier” refers to a material — such as an adjuvant, excipient, glidant, sweetening agent, diluent, preservative, dye, colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent or emulsifier— -that can be incorporated into a composition and administered to a patient without causing unacceptable biological effects or interacting in an unacceptable manner with other components of the composition. Such pharmaceutically acceptable materials typically have met the required standards of toxicological and manufacturing testing, and include those materials identified as suitable inactive ingredients by the U.S. Food and Drug Administration.
[0061] The terms “salt” and “pharmaceutically acceptable salt” refer to a salt prepared from a base or an acid. Pharmaceutically acceptable salts are suitable for administration to a patient, such as a mammal (for example, salts having acceptable mammalian safety for a given dosage regime). Salts can be formed from inorganic bases, organic bases, inorganic acids and organic acids. In addition, when a compound contains both a basic moiety, such as an amine, pyridine or imidazole, and an acidic moiety, such as a carboxylic acid, or tetrazole, zwitterions may be formed and are included within the term “salt” as used herein. Salts derived from inorganic bases include ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium., sodium, and zinc salts, and the like. Salts derived from organic bases include salts of primary, secondary and tertiary amines, including substituted amines, cyclic amines, naturally-occurring amines and the like, such as arginine, betaine, caffeine, choline, N,N' -dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N- ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like. Salts derived from inorganic acids include salts of boric, carbonic, hydrohalic (hydrobromic, hydrochloric, hydrofluoric or hydroiodic), nitric, phosphoric, sulfamic, and sulfuric acids. Saits derived from organic acids include salts of aliphatic hydroxyl acids (for example, citric, gluconic, glycolic, lactic, lactobionic, malic, and tartaric acids), aliphatic monocarboxylic acids (for example, acetic, butyric, formic, propionic and trifluoroacetic acids), amino acids (for example, aspartic and glutamic acids), aromatic carboxylic acids (for example, benzoic, p-chlorobenzoic, diphenylacetic, gentisic, hippuric, and triphenylacetic acids), aromatic hydroxyl acids (for example, o- hydroxy benzoic, p-hydroxybenzoic, 1-hydroxynaphthalene-2-carboxylic and 3-hydroxynaphthalene-2- carboxylic acids), ascorbic, dicarboxylic acids (for example, fumaric, maleic, oxalic and succinic acids), glucoronic, mandelic, mucic, nicotinic, orotic, pamoic, pantothenic, sulfonic acids (for example, benzenesulfonic, camphorsulfonic, edisylic, ethanesulfonic, isethionic, methanesulfonic, naphthalenesulfonic, naphthalene-1,5-disulfonic, naphthalene -2,6-disulfonic and p-toluenesulfonic acids), xinafoic acid, and the like.
[0062] The term “therapeutically effective amount” refers to that amount of a compound described herein that is sufficient to affect treatment when administered to a subject in need thereof. For example, a therapeutically effective amount for treating pulmonary fibrosis is an amount of compound needed, to, for example, reduce, suppress, eliminate, or prevent the formation of fibrosis in a subject, or to treat the underlying cause of pulmonary fibrosis. The therapeutically effective amount may vary depending upon the intended treatment application (in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. The specific dose will vary depending on the particular compound chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried. The term “effective amount” refers to an amount sufficient to obtain a desired result, which may not necessarily be a therapeutic result. For example, an “effective amount” may be the amount needed to inhibit an enzyme.
[0063] As used herein, “treating” or “treatment” refers to an approach for obtaining beneficial or desired results with respect to a disease, disorder, or medical condition (such as pulmonary' fibrosis) in a subject, including but not limited, to the following: (a) preventing the disease or medical condition from occurring, e.g., preventing the reoccurrence of the disease or medical condition or prophylactic treatment of a subject that is pre-disposed to the disease or medical condition; (b) ameliorating the disease or medical condition, e.g., eliminating or causing regression of the disease or medical condition in a subject; (c) suppressing the disease or medical condition, e.g., slowing or arresting the development of the disease or medical condition in a subject; or (d) alleviating symptoms of the disease or medical condition in a subject. For example, “treating pulmonary fibrosis” would include preventing fibrosis from occurring, ameliorating fibrosis, suppressing fibrosis, and alleviating the symptoms of fibrosis (for example, increasing oxygen levels in blood or improved lung function tests). Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject, notwithstanding that the subject may still be afflicted with the underlying disorder [0064] A “therapeutic effect”, as that term is used herein, encompasses a therapeutic benefit and/or a prophylactic benefit as described above. A prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
[0065] The terms “antagonist” and “inhibitor” are used interchangeably, and they refer to a compound having the ability to inhibit a biological function (e.g., activity, expression, binding, protein-protein interaction) of a target protein (e.g., ALK5). Accordingly, the terms “antagonist” and “inhibitor” are defined in the context of the biological role of the target protein. While preferred antagonists herein specifically interact with (e.g., bind to) the target, compounds that inhibit a biological activity of the target protein by interacting with other members of the signal transduction pathway of which the target protein is a member are also specifically included within this definition.
[0066] The term “selective inhibition” or “selectively inhibit” refers to the ability of a biologically active agent to preferentially reduce the target signaling activity as compared to off-target signaling activity, via direct or indirect interaction with the target.
[0067] As used herein, the term “antibody” refers to an immunoglobulin molecule that specifically binds to, or is immunologically reactive toward, a specific antigen. An antibody may be, for example, polyclonal, monoclonal, genetically engineered, or an antigen binding fragment thereof, and further may be, for example, murine, chimeric, humanized, heteroconjugate, bispecific, a diabody, a triabody, or a tetrabody. An antigen binding fragment includes an antigen binding domain and may be in the form, of, for example, a Fab’, F(ab’)2, Fab, Fv, rlgG, scFv, hcAbs (heavy chain antibodies), a single domain antibody, VHH, VNAR, sdAb, or nanobody.
[0068] The term “antigen binding domain” as used herein refers to a region of a molecule that binds to an antigen. An antigen binding domain may be an antigen-binding portion of an antibody or an antibody fragment. An antigen binding domain may be one or more fragments of an antibody that retain the ability to specifically bind to an antigen. An antigen binding domain can be an antigen binding fragment and may recognize a single antigen, two antigens, three antigens or more. As used herein, “recognize” with regard to antibody interactions refers to the association or binding between an antigen binding domain of an antibody or portion thereof and an antigen. [0069] An “ antibody construct” refers to a molecule, e.g., a protein, peptide, antibody or portion thereof, that contains an antigen binding domain and an Fc domain (e.g., an Fc domain from within the Fc region). An antibody construct may recognize, for example, one antigen or multiple antigens.
[0070] A “targeting moiety” refers to a structure that has a selective affinity for a target molecule relative to other non-targel molecules. The targeting moiety binds to a target molecule. A targeting moiety may include an antibody, a peptide, a ligand, a receptor, or a binding portion thereof. The target biological molecule may be a biological receptor or other structure of a cell, such as a tumor antigen.
[0071] The terms “subject” and “patient” refer to an animal, such as a mammal, for example a human. The methods described herein can be useful in both human therapeutics and veterinary applications. In some embodiments, the subject is a mammal, and in some embodiments, the subject is human. “Mammal” includes humans and both domestic animals such as laboratory animals and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animals such as wildlife and the like.
[0072] “Prodrug” is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein (e.g., compound of Formula (I)). Thus, the term “prodrug" refers to a precursor of a biologically active compound that is pharmaceutically acceptable. In some aspects, a prodrug is inactive when administered to a subject but is converted in vivo to an active compound, for example, by hydrolysis. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam); Higuchi, T., et al., “Pro-drags as Novel Delivery Systems,” (1987) A.C.S. Symposium. Series, Vol. 14; and Bioreversible Carriers in Drag Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press), each of which is incorporated in full by reference herein). The term “prodrug” is also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a mammalian subject. Prodrags of an active compound, as described herein, are typically prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound. Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrags include, but are not limited to, acetate, formate and benzoate derivatives of a hydroxy functional group, or acetamide, formamide and. benzamide derivatives of an amine functional group in the active compound, and. the like.
[0073] The term “in vivo” refers to an event that takes place in a subject's body.
[0074] The term “in vitro” refers to an event that takes places outside of a subject's body. For example, an in vitro assay encompasses any assay run outside of a subject. In vitro assays encompass cell-based, assays in which cells alive or dead are employed. In vitro assays also encompass a cell-free assay in which no intact cells are employed.
[0075] The disclosure is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, esterification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the disclosure includes compounds produced by a process comprising administering a compound of this disclosure to a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radiolabeled compound of the disclosure in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to a human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.
[0076] Lung function tests include tests to check how well the lungs work. Spirometry, for example, measures the amount of air the lungs can hold as well as how forcefully one can empty air from, the lungs. Forced expiratory volume (FEV) is a measure of the amount of air a person can exhale during a forced breath. FEV1, for example, is the amount of air a person can force from their lungs in one second. Forced vital capacity (FVC) is the total amount of air exhaled during an FEV test. The ratio of FEV1/FVC, also known as Index of Air Flow or Tiffeneau-Pinelli Index, is a measurement used to assess the health of a patient’s lung function. A ratio of < 80% indicates an obstructive defect is present in the lungs, such as chronic obstructive pulmonary disease (COPD). A ratio of > 80% indicates a restrictive defect is present in the lungs, such as pulmonary fibrosis. The ratio of > 80% in restrictive lung disease results from both FEV1 and FVC being reduced but that the decline in FVC is more than that of FEV 1, resulting in a higher than 80% value.
[0077] The term “transforming growth factor -β” may also be referred to as TGF-β, transforming growth factor beta-1, or TGF-beta-1. It is also cleaved into latency-associated peptide (LAP).
[0078] The term “TGF-β receptor II” may also be referred to as TpRII, type II TGF-β receptor, TGF-βRII, TGF-beta receptor type-2, TGFR-2, TGF-beta type II receptor, transformi ng growth factor-beta receptor type II, TGF-beta receptor type II or TbetaR-II.
[0079] The term “TGF-β receptor I” may also be referred to as TpRI, type I TGF -p receptor, TGF-βRI, TGF- beta receptor type-1, TGFR-1 , activin A receptor type Il-like protein kinase of 53kD, activin receptor-like kinase 5, ALK-5, ALK5, serine/threonine -protein kinase receptor R4, SKR4, TGF-beta type I receptor, transforming growth factor-beta receptor type I, TGF-beta receptor type I, transforming growth factor beta receptor I, TGF -beta receptor 1, or TbetaR -I.
[0080] The present disclosure provides compounds that are capable of selectively binding to and/or modulating ALK5. In some embodiments, the compounds modulate ALK5 by binding to or interacting with one or more amino acids and/or one or more metal ions. The binding of these compounds may disrupt ALK5 downstream signaling.
[0081] In certain aspects the present disclosure provides a compound of Formula (I):
Figure imgf000026_0001
or a pharmaceutically acceptable salt thereof, wherein:
X and Y are each independently selected from CH and N;
A is a 9- or 10-membered bicyclic heteroaryl group;
B is selected from phenyl, pyridyl, and thiazolyl;
RA is selected from:
-NO2, -CN, -SR1, -NR'R1. -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)OR5, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2;
C2-10 alkenyl and C2-10 alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), C3-12 carbocycle, and 3- to 12-membered heterocycle;
C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -O(OC)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, C3- 12 carbocycle, and 3- to 12-membered heterocycle;
C9-10 aryl, C3-12 cycloalkyl, and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -O(OC)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
RB is independently selected at each occurrence from halogen, -CN, -NH2, -NHCH3, -NHCH2CH3, -C(O)CH3, -OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CH2F, -CHF2, -CF3, C3-4 carbocycle, and 3- to 4-membered heterocycle; n is an integer from 0 to 3;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -SCH3, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -N(CH2CH3)2, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -OC(O)CH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle;
R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1;
R4 is independently selected at each occurrence from C1-3 alkyl, C2-6 alkenyl, C2-6 alkynyl, CH2-(C3-12 cycloalkyl), CH2-(C5-12 cycloalkenyl), C2-3 alkyl-(C3-12 carbocycle), and C1-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3,
-C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH(CH3)2, -C(CH3)3, C3- 12 cycloalkyl, and 3- to 6-membered heterocycle; and
R5 is independently selected at each occurrence from C1-3 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6- membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6- membered heterocycle.
[0082] In certain aspects, the present disclosure provides a compound of Formula (I):
Figure imgf000028_0001
or a pharmaceutically acceptable salt thereof, wherein:
X and ¥ are each independently selected from CH and N;
A is a 9- or 10-membered bicyclic heteroaryl group;
B is selected from phenyl, pyridyl, and thiazolyl;
RA is selected from: halogen, -NO2, -CN, -OR1, -SR1, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1);
C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, and -N(R1)-C1-10 alkyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -N(R1)2 , -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, (O-)ONC(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), C3-12 carbocycle, and 3- to 12-membered heterocycle; and
C3-12 carbocycle, 3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, (O)-NO(CR1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
RB is independently selected at each occurrence from halogen, -CN, -NH2, -NHCH3, -NHCH2CH,, -C(O)CH3, -OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CH2F, -CHF2, -CF3, C3-4 carbocycle, and 3- to 4-membered heterocycle; n is an integer from 0 to 3; R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH3CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle; and
R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1.
[0083] In certain aspects, the present disclosure provides a compound of Formula (I):
Figure imgf000029_0001
or a pharmaceutically acceptable salt thereof, wherein:
X and Y are each independently selected from CH and N;
A is a 9- or 10-membered bicyclic heteroaryl group selected from:
Figure imgf000029_0002
Figure imgf000029_0003
and
Figure imgf000029_0004
B is selected from phenyl, pyridyl, and thiazolyl;
RA is selected from:
-NO2, -CN, -SR1, -NR1R4, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)OR5, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2;
C2-10 alkenyl and C2-10 alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR1 -SR1, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), C3-12 carbocycle, and 3- to 12-membered heterocycle;
C1-10 alkyl, -N(R1)- C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)zR1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -PCO(R1)2, C3- 12 carbocycle, and 3- to 12-membered heterocycle;
C9-10 aryl, C3-12 cycloalkyl, and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CHZC(O)OR1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)z, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)z, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -PCOXOR1)2 -PCOXR1^, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -O(CO)R1, -O(CO)OR1, -O(CO)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(RT), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
RB is independently selected at each occurrence from halogen, -CN, -NH2, -NHCH3, -NHCH2CH3, -C(O)CH3, -OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CH2F, -CHF2, -CF3, C3.4 carbocycle, and 3- to 4-membered heterocycle; n is an integer from 0 to 3;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle;
R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1;
R4 is independently selected at each occurrence from C1-3 alkyl, CM alkenyl, CM alkynyl, CH2-(C3-12 cycloalkyl), CH2-( C5-12 cycloalkenyl), C2-3 alkyl-( C3-12 carbocycle), and C1-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3,
-C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH(CH3)2, -C(CH3)3, C3- 12 cycloalkyl, and 3- to 6-membered heterocycle; and
R5 is independently selected at each occurrence from CM alkyl, CM alkenyl, CM alkynyl, 1- to 6- membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6- membered heterocycle.
[0084] In some embodiments, for a compound of Formula (I), at least one of X and Y is N, such as X is CH and Y is N. In some embodiments, X is N and Y is CH. In some embodiments, X and Y are each N. In some embodiments, the compound of Formula (I) is a compound of Formula (I-A), (I-B), or (1-C):
Figure imgf000031_0001
[0085] In some embodiments, for a compound of Formula (I), B is phenyl, optionally wherein n is 2 or 3, such as
Figure imgf000031_0002
or
Figure imgf000031_0003
. In some embodiments, B is pyridyl, optionally wherein n is 1, such as
. In some embodiments, B is thiazolyl, optionally wherein n is 1, such as . In some
Figure imgf000031_0004
Figure imgf000031_0005
embodiments, the compound of Formula (I) is a compound of Formula (I-D), (I-E), or (I-F):
Figure imgf000031_0006
In some embodiments, the compound of Formula (I) is a compound of Formula (I-G), (I-H), or (I-I):
Figure imgf000032_0001
or
Figure imgf000032_0002
[0086] In some embodiments, for a compound of Formula (I), RB is independently selected at each occurrence from halogen and C1-6 alkyl, such as RB is independently selected at each occurrence from Cl, F, and -CH3. Optionally, n is an integer from 1 to 3, such as n is 1 or 2. In some embodiments, n is 2 and RB independently selected at each occurrence from Cl and F. In some embodiments, n is 1, and RB is -CH3. In some embodiments, B is phenyl, n is 2, and RB is independently selected at each occurrence from Cl and. F. In some embodiments, B is pyridyl, n is 1, and RB is -CH3.
[0087] In some embodiments, for a compound of Formula (I), A is a 9- or 10-membered bicyclic heteroaryi group substituted by at least one RA. In some embodiments, A comprises at least one ring nitrogen atom, and optionally further comprises one or more heteroatoms selected from N, S, and O.
[0088] In some embodiments, A is selected from
Figure imgf000032_0003
Figure imgf000032_0004
Figure imgf000032_0005
, and
Figure imgf000032_0006
. In some embodiments, A is selected from
Figure imgf000032_0007
Figure imgf000032_0008
Figure imgf000032_0009
, and
Figure imgf000032_0010
. In some embodiments, A is selected from
Figure imgf000032_0011
Figure imgf000033_0001
and
Figure imgf000033_0002
. In some embodiments, A is selected from
Figure imgf000033_0003
Figure imgf000033_0004
, and
Figure imgf000033_0005
. In some embodiments, A is selected from
Figure imgf000033_0006
and
Figure imgf000033_0007
. In some embodiments, A is selected from
Figure imgf000033_0008
and
Figure imgf000033_0009
. In some embodiments, A is selected from
Figure imgf000033_0010
and
Figure imgf000033_0011
. In some embodiments, A is selected from
Figure imgf000033_0012
and
Figure imgf000033_0013
[0089] In some embodiments, the compound of Formula (I) is a compound of Formula (I-J):
Figure imgf000033_0014
, such as
Figure imgf000033_0015
or
Figure imgf000033_0016
In some embodiments, the compound of Formula (I) is a compound of Formula (I-K):
Figure imgf000033_0017
, such as
Figure imgf000033_0018
or
Figure imgf000033_0019
In some embodiments, the compound of Formula (I) is a compound of Formula (I-L):
Figure imgf000033_0020
, such as
Figure imgf000033_0021
or
Figure imgf000033_0022
In some embodiments, the compound of Formula (I) is a compound of Formula (I-M):
Figure imgf000034_0001
, such as
Figure imgf000034_0002
or
Figure imgf000034_0003
In some embodiments, the compound of Formula (I) is a compound of Formula (I-N):
Figure imgf000034_0004
, such as
Figure imgf000034_0005
or
Figure imgf000034_0006
In some embodiments, the compound of Formula (I) is a compound of Formula (I-O):
Figure imgf000034_0007
, such as
Figure imgf000034_0008
or
Figure imgf000034_0009
In some embodiments, the compound of Formula (I) is a compound of Formula (I-P):
Figure imgf000034_0010
, such as
Figure imgf000034_0011
or
Figure imgf000034_0012
In some embodiments, the compound of Formula (I) is a compound of Formula (I-Q) : , such as or
Figure imgf000034_0013
Figure imgf000034_0014
Figure imgf000034_0015
In some embodiments, the compound of Formula (I) is a compound of Formula (I-R):
Figure imgf000034_0016
, such as
Figure imgf000034_0017
or
Figure imgf000034_0018
In some embodiments, the compound of Formula (I) is a compound of Formula (I-S):
Figure imgf000035_0001
, such as
Figure imgf000035_0002
or
Figure imgf000035_0003
In some embodiments, the compound of Formula (I) is a compound of Formula (I-T):
Figure imgf000035_0004
, such as
Figure imgf000035_0005
or
Figure imgf000035_0006
In some embodiments, the compound of Formula (I) is a compound of Formula (I-U):
Figure imgf000035_0007
, such as
Figure imgf000035_0008
or
Figure imgf000035_0009
In some embodiments, the compound of Formula (I) is a compound of Formula (I-V):
Figure imgf000035_0010
, such as
Figure imgf000035_0011
or
Figure imgf000035_0012
In some embodiments, the compound of Formula (I) is a compound of Formula (I-W):
Figure imgf000035_0013
, such as
Figure imgf000035_0014
or
Figure imgf000035_0015
In some embodiments, the compound of Formula (I) is a compound of Formula (I-X): , such as
Figure imgf000035_0016
Figure imgf000035_0017
or
Figure imgf000035_0018
In some embodiments, the compound of Formula (I) is a compound of Formula (I-Y): such as
Figure imgf000036_0002
or
Figure imgf000036_0001
Figure imgf000036_0003
In some embodiments, the compound of Formula (I) is a compound of Formula (I-Z):
Figure imgf000036_0004
, such as
Figure imgf000036_0005
or
Figure imgf000036_0006
In some embodiments, the compound of Formula (I) is a compound of Formula (I-AA): , such as
Figure imgf000036_0008
or
Figure imgf000036_0007
Figure imgf000036_0009
In some embodiments, the compound of Formula (I) is a compound of Formula (I-BB): , such as or
Figure imgf000036_0010
Figure imgf000036_0011
Figure imgf000036_0012
In some embodiments, the compound of Formula (I) is a compound of Formula (I-CC):
Figure imgf000036_0013
, such as
Figure imgf000036_0014
or
Figure imgf000036_0015
[0090] References herein to “a compound of Formula (I)” implicitly also include the compound of Formula (I-A), (I-B), (I-C), (I-D), (I-E), (I-F), (LG), (I-H), (I-I), (I-J), (I-K), (I-L), (I-M), (I-N), (I-O), (I-P), (I-Q), (I-
R), (I-S), (I-T), (I-U), (I-V), (I-W), (I-X), (I-Y), (I-Z), (I-AA), (I-BB), and (I-CC). The skilled person will understand that the context of a passage may exclude one or more of Formulas (I-A), (I-B), (I-C), (I-D), (I-E), (I-F), (I-G), (I-H), (I-I), (I-J), (I-K), (I-L), (I-M), (I-N), (I-O), (I-P), (I-Q), (I-R), (I-S), (I-T), (I-U), (I-V), (I-
W), (I-X), (I-Y), (I-Z), (I-AA), (I-BB), and (I-CC), for example, if the passage provides an embodiment of a particular variable (e.g., Y) different than what is depicted in the formula. [0091] In some embodiments, for a compound of Formula (I):
RA is selected from: halogen, -NO2, -CN, -OR1, -SR1, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =s, =N(R1); C1-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, and -N(R1)-C1-10 alkyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), C3-12 carbocycle, and 3- to 12- membered heterocycle; and C3-12 carbocycle, 3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH), OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle; and
R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1.
[0092] In some embodiments, for a compound of Formula (I):
RA is selected from:
-NO2, -CN, -SR1, -NR1R4, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)OR5, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -P(O)(OR1)2, -P(O)( R1)2; C2-10 alkenyl and C2-10 alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), C3-12 carbocycle, and 3- to 12-membered heterocycle; C1-10 alkyl, -N(R1)- C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, C3-12 carbocycle, and 3- to 12-membered heterocycle;
C9-10 aryl, C3-12 cycloalkyl, and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and =N(R1)-C3-12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, (O)R1, -OC(O)OR1-,OC-OC (O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, H3,- -OOCCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle;
R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1;
R4 is independently selected at each occurrence from C1-3 alkyl, C2-6 alkenyl, C2-6 alkynyl, CH2-(C3-12 cycloalkyl), CH2-(C5-12 cycloalkenyl), C2-3 alkyl-(C3-12 carbocycle), and C1-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH(CH3)2, -C(CH3)3, C3-
12 cycloalkyl, and 3- to 6-membered heterocycle; and
R5 is independently selected at each occurrence from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6- membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6- membered heterocycle.
[0093] In some embodiments, for a compound of Formula (I), RA is selected from:
-NR1R4, -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3; C1-10 alkyl, -N(R1)- C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle;
C10 aryl, C3-12 cycloalkyl, and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, =O, R1, C1-6 alkyl, andC1-6 haloalkyl.
[0094] In some embodiments, for a compound of Formula (I), RA is selected from:
-NR'R1. -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3; C1-10 alkyl, -N(R1)- C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, C3-12 carbocycle, and 3- to 12- membered heterocycle; C3-12 cycloalkyl and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
[0095] In some embodiments, for a compound of Formula (I), RA is selected from:
-NHR4, -NR2R3, -C(O)OR5, -OC(O)R1, -NHC(O)R1, -NHC(O)N(R1)2, -NHC(O)NR2R3; C1-10 alkyl, -NH- C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2NHR1, -NHR1, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NHC(O)R1, -NHC(O)N(R1)2, -NHC(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; C3-12 cycloalkyl and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2NHR1, -NHR1, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NHC(O)R1, -NHC(O)NHR1, -NHC(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -NH-(3- to 12-membered heterocycle), and -NH-(C3-12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2NHR1, -NHR1, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NHC(O)R1, -NHC(O)NHR1, -NHC(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
[0096] In some embodiments, for a compound of Formula (I), RA is selected from 3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, NR1C (O)N(R1)2, -NR1C(O)NR2R3 =O, R1, C1-6 aLkyl, and C1-6 haloalkyl. In some embodiments, RA is selected from -C(O)OR5, -NR1C(O)R1, and -NR1C(O)N(R1)2. In some embodiments, RA is -NHC(O)R1, such as -NHC(O)-CH2-piperazinyl. In some embodiments, RA is 3- to 12-membered heterocycle, such as 4- to 6-membered heterocycle. In some embodiments, RA is 8- to 11-membered spirocyclic heterocycle, such as 9-membered spirocyclic heterocycle. In some embodiments, RA is phenyl substituted with -CH2NH2. In some embodiments, RA is -NH-(3- to 12-membered heterocycle), wherein the 3- to 12-membered heterocycle is substituted with R1, such as RA is -NH-(pyridyl)- amino -piperidyl. In some embodiments, RA is -C(O)OR1, such as -C(O)O-(C5-10 carbocycle). In some embodiments, RA is -NH- (phenyl), wherein the phenyl is substituted with 3- to 6-membered heterocycle. In some embodiments, R ts phenyl, wherein the phenyl is substituted with 3- to 12-membered heterocycle, such as RA is phenyl substituted by 2,5-dihydro-1H-pyrroIyL In some embodiments, RA is 3- to 6-membered heterocycle substituted by R1, such as pyrazolyl substituted by -CH2CH2NH2 or thiophenyl substituted by -CH2NH2. In some embodiments, RA is -NH-(C3-12 carbocycle), optionally substituted by -NHz, such as -NH-(indane)-NH2. In some embodiments, RA is selected from -NH-(3- to 12-membered heterocycle) and -NH-(C3-12 carbocycle), each of which is optionally substituted with C0-3 alkyl-(3- to 12-membered heterocycle), wherein the Co.3 aIkyl-(3- to 12-membered heterocycle) is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -CH3, and -CF3.
[0097] In some embodiments, for a compound of Formula (I), R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0- 3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -N(CH3)2, -C(O)OCH2CH3, =O, and -CH3. In some embodiments, R1 is independently selected at each occurrence from hydrogen; and aClk1y-6l and C0-3 alkyl -(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -NH2 and -CH3.
[0098] In some embodiments, for a compound of Formula (I), RA is -NR'R4, wherein R‘ is hydrogen and R4 is C1-3 alkyl-(3- to 12-membered heterocycle); or C1-3 alkyl substituted by -C(O)OCH3 or -C(O)OCH2CH3. In some embodiments, RA is -NR1R4, wherein R1 is hydrogen and R4 is C1-3 alkyI-(3- to 12- membered heterocycle). In some embodiments, RA is -NR1R4, wherein R1 is hydrogen and R4 is C1-3 alkyl substituted by -C(O)OCH3 or -C(O)OCH2CH3.
[0099] In some embodiments, for a compound of Formula (I), R5 is independently selected at each occurrence from C1-6 alkyl and C0-3 alkyl -(3 - to 12-membered heterocycle). For example, RA may be -C(O)OR5, wherein R5 is selected from C al1k-6yl and C0-3 alkyl-(3- to 12-membered heterocycle). In some embodiments, RA is -C(O)OR5 , wherein R5 is C0-3 aIkyl-(3- to 12-membered heterocycle), wherein the 3 - to 12-membered heterocycle is optionally substituted with halogen, -NH2 , -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -CH3, -CH2CH3, C3-12 carbocycle, and 3- to 6-membered heterocycle. In some embodiments, RA is -C(O)OR5, wherein R5 is C1-6 alkyl. [0100] In some embodiments, for a compound of Formula (I), RA is selected from
Figure imgf000042_0001
Figure imgf000042_0002
Figure imgf000042_0003
and
Figure imgf000042_0004
. In some embodiments, RA is selected from
Figure imgf000042_0005
Figure imgf000042_0006
Figure imgf000042_0007
, and
. In some embodiments, RA is selected from
Figure imgf000042_0008
Figure imgf000042_0009
Figure imgf000043_0001
, and
Figure imgf000043_0002
. In some embodiments, RA is selected from
Figure imgf000043_0003
Figure imgf000043_0004
, and
Figure imgf000043_0005
. In some embodiments, RA is selected from
Figure imgf000043_0006
Figure imgf000043_0007
, and
Figure imgf000043_0008
. In some embodiments, RA is selected from
Figure imgf000043_0009
Figure imgf000043_0010
, and
Figure imgf000043_0011
, In some embodiments, RA is selected from
Figure imgf000043_0012
Figure imgf000043_0013
Figure imgf000043_0014
, and
Figure imgf000043_0015
[0101] In some embodiments, for a compound of Formula (I):
RA is selected from:
-C(O)OR5, -NHC(O)NHR1; C1-10 alkyl and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1 and -CH2NHR1;
C5-12 cycloalkenyl optionally substituted with -NHR1; and
3- to 12-membered heterocycle, -NH-(3- to 12-membered heterocycle), and -NH-(C3- 12 carbocycle), each of which is independently optionally substituted with one or more substituents selected from halogen, -CH2NHR1, and R1; R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by -NH2; and
R5 is independently selected at each occurrence from C1-6 alkyl and C0-3 alkyl-(3- to 12-membered heterocycle).
[0102] In some embodiments, for a compound of Formula (I):
A is selected from
Figure imgf000044_0001
and.
Figure imgf000044_0002
B is selected from
Figure imgf000044_0003
, and
Figure imgf000044_0004
; and
RA is selected from:
-NR'R1. -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3; C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; C3-12 cycloalkyl and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
[0103] In some embodiments, for a compound of Formula (I):
A is
Figure imgf000044_0005
B is selected from
Figure imgf000045_0001
, and and
Figure imgf000045_0002
RA is selected from -NH-(3- to 12-membered heterocycle) and -NH-(C3-12 carbocycle), each of which is optionally substituted with C0-3 alkyl-(3- to 12-membered heterocycle), wherein the C0-3 alkyl-(3- to 12- membered heterocycle) is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -CH3, and -CF3.
[0104] In some embodiments, for a compound of Formula (I):
A is selected from
Figure imgf000045_0003
, and
Figure imgf000045_0004
B is selected from
Figure imgf000045_0005
, and
Figure imgf000045_0006
and
RA is selected from:
-NR1R4, -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3; C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
[0105] In some embodiments, for a compound of Formula (I):
A is selected from
Figure imgf000045_0007
and
Figure imgf000045_0008
B is selected from
Figure imgf000045_0009
and
Figure imgf000045_0010
; and
RA is selected from:
-NR1R4, -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, NR1C(O)NR2R3; C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, - NR1C(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; C3-12 cycloalkyl and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
[0106] In certain aspects, the present disclosure provides a compound of Formula (I-G):
Figure imgf000046_0001
or a pharmaceutically acceptable salt thereof, wherein:
A is a 9- or 10-membered bicyclic heteroaryl group selected from:
Figure imgf000046_0002
Figure imgf000046_0003
, and
Figure imgf000046_0004
RA is selected from:
-NHR4, -NR2R3, -C(O)0R5, -O(OC)R1, -NHC(O)R1, -NHC(O)N(R1)2, -NHC(O)NR2R3; C1-10 alkyl, -NH-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2NHR1, -NHR1, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NHC(O)R1, -NHC(O)N(R1)2, -NHC(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; C3-12 cycloalkyl and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2NHR1, -NHR1, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NHC(O)R1, -NHC(O)NHR1, -NHC(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -NH-(3- to 12-membered heterocycle), and -NH-(C3- 12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2NHR1, -NHR1, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NHC(O)R1, -NHC(O)NHR1, -NHC(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl;
RB is -CH3;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle;
R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1;
R4 is independently selected at each occurrence from C1-3 alkyl, C2-6 alkenyl, C2-6 alkynyl, CH2-(C3-12 cycloalkyl), CH2-(C5-12 cycloalkenyl), C2-3 alkyl-(C3-12 carbocycle), and C1-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 cycloalkyl, and 3- to 6-membered heterocycle; and
R5 is independently selected at each occurrence from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6- membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6-membered heterocycle. [0107] In certain aspects, the present disclosure provides a compound of Formula (I-H):
Figure imgf000047_0001
or a pharmaceutically acceptable salt thereof, wherein:
A is a 9- or 10-membered bicyclic heteroaryl group selected from:
Figure imgf000048_0001
Figure imgf000048_0002
and
Figure imgf000048_0003
RA is selected from:
-NHR4, -NR2R3, -C(O)OR5, -OC(O)R1, -NHC(O)R1, -NHC(O)N(R1)2,
-NHC(O)NR2R3; C1-10 alkyl, -NH-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2NHR1, -NHR1, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NHC(O)R1, -NHC(O)N(R1)2, -NHC(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; C3-12 cycloalkyl and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2NHR1, -NHR1, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NHC(O)R1, -NHC(O)NHR1, -NHC(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -NH-(3- to 12-membered heterocycle), and -NH-(C3- 12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2NHR1, -NHR1, -NR2R3, -CH2C(O)OR1, -C(O)OR1, (O)R-O1, C -NHC(O)R1, -NHC(O)NHR1, -NHC(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl;
RB is -CH3;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle; R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1;
R4 is independently selected at each occurrence from C1-3 alkyl, C2-6 alkenyl, C2-6 alkynyl, CH2-(C3-12 cycloalkyl), CH2-(C5-12 cycloalkenyl), C2-3 alkyl-(C3-12 carbocycle), and C1-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 cycloalkyl, and 3- to 6-membered heterocycle; and
R5 is independently selected at each occurrence from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6- membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6-membered heterocycle. [0108] In certain aspects, the present disclosure provides a compound of Formula (I-E):
Figure imgf000049_0001
or a pharmaceutically acceptable salt thereof, wherein:
A is a 9- or 10-membered bicyclic heteroaryl group selected from:
Figure imgf000049_0002
, and
Figure imgf000049_0003
Figure imgf000049_0004
RA is selected from:
-NHR4, -NR2R3, -C(O)OR5, -OC(O)R1, -NHC(O)R1, -NHC(O)N(R1)2, -NHC(O)NR2R3; C1-10 alkyl, -NH-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2NHR1, -NHR1, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NHC(O)R1, -NHC(O)N(R1)2, -NHC(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; C3-12 cycloalkyl and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2NHR1, -NHR1, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NHC(O)R1, -NHC(O)NHR1, - NHC(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -NH-(3- to 12-membered heterocycle), and -NH-(C3- 12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, OR1, -CH2NHR1, -NHR1, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NHC(O)R1, -NHC(O)NHR1, -NHC(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl;
RB is independently selected at each occurrence from halogen;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle;
R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1;
R4 is independently selected at each occurrence from C1-3 alkyl, C2-6 alkenyl, C2-6 alkynyl, CH2-(C3-12 cycloalkyl), CH2-(C5-12 cycloalkenyl), C2-3 alkyl-(C3-12 carbocycle), and C1-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 cycloalkyl, and 3- to 6-membered heterocycle; and
R5 is independently selected at each occurrence from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6- membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6-membered heterocycle. [0109] In some embodiments, a compound of Formula (I) is provided as a substantially pure stereoisomer. In some embodiments, the stereoisomer is provided in at least 80% enantiomeric excess, such as at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or at least 99.9% enantiomeric excess. [0110] In some embodiments, the present disclosure provides a soft ALK5 inhibitor. As used herein, the term “soft drug” or “soft ALK5 inhibitor” refers to a biologically active compound that is converted upon entering the systemic circulation into a predictable metabolite that exhibits reduced biological activity relative to the parent compound. A soft drug, in some embodiments, exerts its desired therapeutic effect locally at the target organ or tissue, then is rapidly converted to a predictable metabolite designed to be less active than the parent soft drug upon entering the systemic circulation, thus reducing systemic exposure to the biologically active compound. Accordingly, soft drugs have a lower potential for undesired side effects relative to non-soft drug compounds having comparable biological activity. In some embodiments, a soft drag of the present disclosure exhibits good stability at the intended site of action (e.g., the lung), is rapidly metabolized upon entering systemic circulation, and displays more functional activity than the corresponding metabolite.
[0111] In some embodiments, a soft drug provided herein exhibits an ALK5 pKi of greater than or equal to 9, while the corresponding soft drag metabolite exhibits an ALK5 pKi of 9 or less, such as 8 or less (assessed according to the assay provided in Example 21). In some embodiments, the difference in pK; of the soft drug and the corresponding soft drug metabolite is at least 0.5, such as at least 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or at least 2.0. In some embodiments, a soft drag provided herein exhibits a BEAS2B pIC50 of greater than or equal to 7, while the corresponding soft drug metabolite exhibits a BEAS2B pIC50 of 6 or less (assessed according to the assay provided in Example 22). In some embodiments, the difference in pIC50 of the soft drug and the corresponding soft drug metabolite is at least 1.0, such as at least 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or at least 2.0. In some embodiments, the soft drug and corresponding soft drug metabolite exhibit similar ALK5 pKi values, but the soft drag is more active in cells (e.g., the soft drag exhibits a BEAS2B pIC50 of at least 1.0 greater than the soft drug metabolite).
[0112] In some embodiments, the present disclosure provides a soft ALK5 inhibitor comprising an ester. In some embodiments, the ester inhibits ALK5 activity, while the corresponding carboxylic acid of the ester exhibits reduced ALK5 inhibitory activity. For example, the difference in ALK5 pKi of the ester and corresponding acid may be at least 1.0. In some embodiments, a soft drag ester of the present disclosure is administered, to the lung, for example, by inhalation, and inhibits the activity of ALK5 in the lung. However, upon exiting the lung, the ester may be readily hydrolyzed to the corresponding carboxylic acid, thus reducing systemic exposure to the ester.
Figure imgf000051_0001
[0113] In some embodiments, the present disclosure provides a conjugate comprising a compound disclosed herein linked, e.g., covalently linked, either directly or through a linker to an antibody construct or targeting moiety, thereby forming a conjugate. The linker may be a non-cleavable linker or a cleavable linker. A conjugate may be represented by the formula:
Figure imgf000052_0001
wherein A' is an antibody construct or targeting moiety; L1 is a linker; D' is a compound or salt disclosed herein, such as a compound of Formula (I); and p is an integer from 1 to 20. In some embodiments, p is an integer from 1 to 10, such as from 1 to 8, 2 to 8, 1 to 6, 3 to 5, or from 1 to 3.
[0114] In some embodiments, a conjugate is represented by the formula:
Figure imgf000052_0002
wherein A' is an antibody construct or targeting moiety; D' is a compound or salt disclosed herein, such as a compound of Formula (I); and p is an integer from 1 to 20. In some embodiments, p is an integer from 1 to 10, such as from 1 to 8, 2 to 8, 1 to 6, 3 to 5, or from 1 to 3.
[0115] Accordingly, a compound or salt of the present disclosure, such as a compound of Formula (I), may be attached to A' via a linker, L1, or directly attached to A' without an intermediate linker. In some embodiments, the compound or salt is covalently attached to an A' or L1. It will be understood by the skilled person that not all compounds of the present disclosure are meant to be attached to L1 or A', only those that have suitable attachment sites. A compound or salt disclosed herein that does not have a suitable attachment site may be modified to introduce an attachment site.
[0116] In some embodiments, L1 or D' is bound to A' via a terminus of an amino acid sequence or via a side chain of an amino acid, such as the side chain of lysine, serine, threonine, cysteine, tyrosine, aspartic acid, glutamine, a non-natural amino acid residue, or glutamic acid residue. In some embodiments, L1 or D' is bound to A' via one or more glycans or short peptide tags of four to six amino acids. L1 or D' may be conjugated to A' via any suitable functional group, such as a thiol, an amine, an amide, an alcohol, a ketone, a carboxylic acid, or an ester.
[0117] A linker may be attached to a compound or salt of the present disclosure at any available position. For example, a compound of Formula (I) may comprise a linker L1 to A' in place of RA or through substituent RA:
Figure imgf000052_0003
[0118] Compounds are typically depicted herein in their unconjugated form, though it will be understood by the skilled person that linker L1 may be covalently bound to any suitable atom for attachment, such as a substitutable nitrogen, carbon, sulfur, phosphorous or oxygen of a compound. L1 may be a cleavable or non- cleavable linker. The linker may further be bound to A'. In some embodiments, L1 does not affect the binding of the active portions of the conjugate to the binding target(s). Covalent linkages may be formed by reaction between a functional group on the linker with a functional group on the compound, and by reaction between a functional group on the linker with a functional group on A'. As used herein in the context of conjugates, the term “linker” includes (i) unattached forms of the linker comprising a functional group capable of covalently attaching the linker to a compound disclosed herein and a functional group capable of covalently attaching the linker to an antibody construct or targeting moiety; (ii) partially attached forms of the linker bound to a compound disclosed herein, wherei n the linker comprises a functional group capable of covalently attachi ng the linker to an antibody construct or targeting moiety; (iii) partially attached forms of the linker bound to an antibody construct or targeting moiety, wherein the linker comprises a functional group capable of covalently attaching the linker to a compound disclosed herein; and (iv) fully attached forms of the linker bound to both an antibody construct or targeting moiety and a compound disclosed herein.
[0119] Linker L1 may be short, flexible, rigid, cleavable (e.g., by a lysosomal enzyme), non- cleav able, hydrophilic, or hydrophobic. A linker may contain segments having different characteristics, such as flexible segments and rigid segments. A linker may be chemically stable to extracellular environments, for example, in the bloodstream, or may include moieties that are not stable or are selectively stable. In some embodiments, a linker comprises a moiety that is selectively cleaved, for example, selectively cleaved in cells, a particular organ, or in plasma. A linker may be sensitive to enzymes, such as proteases. A linker may be insensitive to intracellular processes or proteases. A linker may be acid-labile, protease-sensitive or photolabile. In some embodiments, a linker comprises a peptide, succinimide, maleimide, polyethylene glycol, alkylene, alkenylene, alkynyiene, disulfide, hydrazone, polyether, polyester, polyamide, arninobenzyl -carbamate, or a combination thereof.
[0120] In some aspects, the present disclosure provides a compound of Formula (I), or a compound disclosed in Table 1, wherein the compound is covalently bound to A', optionally via linker L1. In some embodiments, the antibody construct is an antibody. In some embodiments, the present disclosure provides a compound of Formula (I), or a compound disclosed in Table 1, wherein the compound is covalently bound to a linker, L1, to form a compound-linker. A' or L1 may be covalently attached to any position of the compound, valence permitting. A linker L1 disclosed herein may comprise from about 10 to about 500 atoms, such as 10 to 400 atoms, 10 to 300 atoms, 30 to 400 atoms, or 30 to 300 atoms.
[0121] The targets of the antibody, antibody construct, or targeting moiety may depend on the desired therapeutic applications of the conjugate. Typically, the targets are molecules present on the surfaces of cells into which it is desirable to deliver an ALK5 inhibitor, such as T cells, and the antibodies, in some embodiments, internalize upon binding to the target. For applications in which the conjugates are intended to stimulate the immune system by reducing TGF-β activity, it may be desirable to generate antibodies, antibody constructs, or targeting moieties that bind to T cell surface molecules. Not wishing to be bound by any particular theory, it is believed that the delivery of ALK5 inhibitors to T cells can activate CD4+ and/or CD8+ T cell activity and inhibit regulatory T cell activity, both of which contribute to immune tolerance of tumors. Accordingly, antibodies, antibody constructs, or targeting moieties (A') that bind to T cell surface molecules in the conjugates of the present disclosure are useful for the treatment of various cancers, such as those described herein below. In some embodiments. A' binds to CD4+ T cells, CD8+ T cells, TREG cells, or any combination thereof. In some embodiments, A' binds to a pan T cell surface molecule, such as CD1, CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD25, CD28, CD70, CD71 , CD103, CD184, Tim3, LAG3, CTLA4, or
PD1. Examples of antibodies that bind to T cell surface molecules and are believed to be internalizing include OKT6, OKT11, OKT3, OKT4, OKT8, 7D4, OKT9, CD28.2, UCHT1, M290, FR70, pembrolizumab, nivolumab, cemiplimab, and dostarlimab.
[0122] An antibody, antibody construct, or targeting moiety disclosed herein may comprise an antigen bindi ng domain that specifically binds to a tumor antigen or antigen associated with the pathogenesis of fibrosis. In some embodiments, the antigen binding domain specifically binds to an antigen on a T cell, a B cell, a stellate cell, an endothelial cell, a tumor cell, an APC, a fibroblast cell, a fibrocyte cell, or a cell associated with the pathogenesis of fibrosis. In some embodiments, the antigen binding domain targets CTLA4, PD-1, OX40, LAG-3, GITR, GARP, CD25, CD27, PD-LL TNFR.2, ICOS, 41BB, CD70, CD73, CD38 or VTCN1. In some embodiments, the antigen binding domain targets PDGFRp, integrin αvβl, integrin αvβ3, integrin αvβ6, αvβ8, endosialin, FAP, ADAM12, LRRC15, MMP14, PDPN, CDH11, F2RL2, ASGR1, or ASGR2.
[0123] The chemical entities described herein can be synthesized according to one or more illustrative schemes herein and/or techniques known in the art. Materials used herein are either commercially available or prepared by synthetic methods generally known in the art. These schemes are not limited to the compounds listed in the examples or by any particular substituents, which are employed for illustrative purposes.
Although various steps are described and depicted in Schemes 1-4 and Examples 1-20, the steps in some cases may be performed in a different order than the order shown in Schemes 1-4 and Examples 1-20. Various modifications to these synthetic reaction schemes may be made and will be suggested to one skilled in the art having referred to the present disclosure. Numberings or R groups in each scheme typically have the same meanings as those defined elsewhere herein unless otherwise indicated.
[0124] Unless specified to the contrary, the reactions described herein take place at atmospheric pressure, generally within a temperature range from -10 °C to 200 °C. Further, except as otherwise specified, reaction times and conditions are intended to be approximate, e.g. taking place at about atmospheric pressure within a temperature range of about -10 °C to about 110 °C over a period of about 1 to about 24 hours; reactions left to run overnight average a period of about 16 hours.
[0125] In general, compounds of the disclosure may be prepared by the following reaction schemes:
Figure imgf000055_0001
[0126] In some embodiments, a compound of Formula Id or Formula lg may be prepared according to Scheme 1. For example, ethanone la can be reacted with DMF-DMA at elevated temperatures to give intermediate lb, which can be reacted with hydrazine monohydrate to provide pyrazoie 1c. Optionally, le may be subjected to one or more coupling reactions, and optionally one or more protecting group manipulations, to provide a pyrazoie of Formula Id. Alternatively, oxidation of la can provide dione le, which can be converted to imidazole If in the presence of urotropine and ammonium acetate. Optionally, If may be subjected to one or more coupling reactions, and optionally one or more protecting group manipulations, to provide an imidazole of Formula lg.
Figure imgf000055_0002
[0127] In some embodiments, a compound of Formula 2c may be prepared according to Scheme 2. For example, alkyne 2a can be converted to triazole 2b in the presence of a suitable azide, such as TMS-N3. Optionally, 2b may be subjected to one or more coupling reactions, and optionally one or more protecting group manipulations, to provide a triazole of Formula 2c.
Figure imgf000056_0001
[0128] In some embodiments, a compound of Formula 3c, Formula 3e or Formula 3h may be prepared according to Scheme 3. For example, bromide 3a can be subjected to a C-N coupling reaction — optionally a Pd-catalyzed coupling reaction such as a Buchwald -Hartwig amination — -with an acyclic primary or secondary amine (3b) or a cyclic secondary amine (3d) to provide an amine of Formula 3c or Formula 3e, respectively. Alternatively, installation of a desired RA substituent may proceed via a Suzuki reaction, either in one step using boronic ester 3f or in two steps — wherein heteroaryl bromide 3a is first converted to the corresponding boronic ester, their coupled to a suitable halide (e.g., RAX) — to give a compound of Formula 311.
Figure imgf000056_0002
[0129] In some embodiments, a compound of Formula 4j may be prepared according to Scheme 4. For example, bromide 4a can be subjected to a coupling reaction— -optionally a Suzuki -Miyaura coupling— -with boronic ester 4b to provide 4c. Halogenation of 4c can provide heteroaryl iodide 4d, which may be converted to boronic ester 4f and coupled to a suitable heteroaryl halide (e.g., 4g) to afford 4h. Optionally, 4h may be subjected to one or more coupling reactions, and optionally one or more protecting group manipulations, to provide a compound of Formula 4j.
[0130] In some embodiments, a compound of the present disclosure, for example, a compound, of a formula given in Table 1, is synthesized according to one of the general routes outlined in Schemes 1-4, Examples 1- 20, or by methods generally known in the art. In some embodiments, exemplary compounds may include, but are not limited to, a compound or salt thereof selected from Table 1. In some embodiments, exemplary compounds may include, but are not limited to, a compound, a stereoisomer thereof, or salt thereof selected from Table 1.
Table 1
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
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_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
[0131] In some embodiments, exemplary compounds may include, but are not limited to, a compound, a stereoisomer thereof, or salt thereof selected, from Table 1a.
Figure imgf000195_0002
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
[0132] Methods
[0133] In some aspects, the present disclosure provides a method of inhibiting TGFp signaling, comprising contacting a cell with an effective amount of a compound disclosed herein, such as a compound of Formula (I). In some embodiments, the present disclosure provides a method of inhibiting ALK5, comprising contacting ALK5 with an effective amount of a compound disclosed herein. Inhibition of ALK5 or TGFp signaling can be assessed by a variety of methods known in the art. Non-limiting examples include a showing of (a) a decrease in kinase activity of ALK5; (b) a decrease in binding affinity between the TGFβ/TGFβ-RII complex and ALK5; (c) a decrease in the levels of phosphorylated intracellular signaling molecules downstream in the TGFp signaling pathway, such as a decrease in pSMAD2 or pSMAD3 levels; (d) a decrease in binding of ALK5 to downstream signaling molecules, such as SMAD2 and SMAD3; and/or (e) an increase in ATP levels or a decrease in ADP levels. Kits and commercially available assays can be utilized for determining one or more of the above.
[0134] In some aspects, the present disclosure provides a method of treating an A L K5- mediated disease or condition in a subject, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein. In some embodiments, the disease or condition is selected from fibrosis and cancer. In some embodiments, the disease or condition is pulmonary fibrosis, such as idiopathic pulmonary fibrosis or virus-induced fibrosis. In some embodiments, the disease or condition is intestinal fibrosis. In some embodiments, the disease or condition is alopecia. In some embodiments, the disease is a neurodegenerative disease, such as Alzheimer’s disease. In some embodiments, the present disclosure provides a method of reversing symptoms of aging. For example, the method may enhance neurogenesis, reduce neuroinflammation, improve cognitive performance, regenerate liver tissue, and reduce pl6 levels.
[0135] In some aspects, the present disclosure provides a method of treating fibrosis, comprising administering to a patient an effective amount of a compound disclosed herein. In some embodiments, the fibrosis is mediated by ALK5. In some embodiments, the fibrosis is selected from systemic sclerosis, systemic fibrosis, organ-specific fibrosis, kidney fibrosis, pulmonary fibrosis, liver fibrosis, portal vein fibrosis, skin fibrosis, bladder fibrosis, intestinal fibrosis, peritoneal fibrosis, myelofibrosis, oral submucous fibrosis, and retinal fibrosis. In some embodiments, the fibrosis is pulmonary fibrosis, such as idiopathic pulmonary fibrosis (IPF), familial pulmonary fibrosis (FPF), interstitial lung fibrosis, fibrosis associated with asthma, fibrosis associated with chronic obstructive pulmonary disease (COPD), silica-induced fibrosis, asbestos- induced fibrosis or chemotherapy-induced lung fibrosis. In some embodiments, the fibrosis is idiopathic pulmonary fibrosis (IPF). In some embodiments, the fibrosis is TGF-β -mediated pulmonary fibrosis. In some embodiments, the patient has been diagnosed with acute respiratory distress syndrome (ARDS). In some embodiments, the fibrosis is acute fibrosis. In some embodiments, the fibrosis is chronic fibrosis.
[0136] In some aspects, the present disclosure provides a method of treating pulmonary fibrosis induced by a viral infection, comprising administering to a patient an effective amount of a compound disclosed herein.
The pulmonary fibrosis may be induced by an erythrovirus, a dependovirus, a papillomavirus, a polyomavirus, a mastadenovirus, an alphaherpesvirinae, a varicellovirus, a gammaherpesvirinae, a betaherpesvirinae, a roseolovirus, an orthopoxvirus, a parapoxvirus, a moliuscipoxvirus, an orthohepadnavirus, an enterovirus, a rhinovirus, a hepatovirus, an aphthovirus, a calicivirus, an astrovirus, an alpha virus, a rubivirus, a flavi virus, a Hepatitis C vims, a reovirus, an orbivirus, a rotavirus, an influenzavirus A, an influenzavirus B, an influenzavirus C, a paramyxovirus, a morbilli virus, a rubulavirus, a pneumovirus, a vesiculovirus, a lyssavirus, a bunyavirus, a hantavirus, a nairovirus, a phlebovirus, a coronavirus, an arenavirus, a BLV-HTLV -retrovirus, a lentivirinae, a spumavirinae or a filovirus. In some embodiments, the fibrosis is virus-induced fibrosis, such as virus-induced pulmonary fibrosis. In some embodiments, the fibrosis is selected from EBV-induced pulmonary fibrosis, CMV-induced pulmonary fibrosis, herpesvirus- induced pulmonary fibrosis and coronavirus-induced pulmonary fibrosis. In some embodiments, the fibrosis is selected from EBV-induced pulmonary fibrosis, CMV-induced pulmonary fibrosis, HHV-6-induced pulmonary fibrosis, HHV-7-induced pulmonary fibrosis, HHV-8-induced pulmonary fibrosis, H5N1 virus- induced pulmonary fibrosis, SARS-CoV-induced pulmonary fibrosis, MERS-CoV-induced pulmonary fibrosis and SARS-CoV-2-induced pulmonary fibrosis. In some embodiments, the pulmonary fibrosis is coronavirus-induced pulmonary fibrosis. In some embodiments, the pulmonary fibrosis is SARS-CoV-2- induced pulmonary fibrosis. In some embodiments, the pulmonary fibrosis is COVID-19-induced pulmonary fibrosis.
[0137] In some aspects, the present disclosure provides a method of treating acute lung injury (ALI), comprising administering to a patient an effective amount of a compound disclosed herein. In some embodiments, the present disclosure provides a method of treating acute respiratory distress syndrome (ARDS), comprising administering to a patient an effective amount of a compound disclosed herein. The ARDS may be in the early acute injury phase or the fibroproliferative phase. In some embodiments, the ARDS is fibroproliferative ARDS. In some embodiments, the present disclosure provides a method of treating fibrosis resulting from. ARDS, comprising administering to a patient an effective amount of a compound disclosed herein. The fibrosis resulting from ARDS may be pulmonary fibrosis. In some embodiments, the present disclosure provides a method of treating fibrosis resulting from ALI, comprising administering to a patient an effective amount of a compound disclosed herein. The fibrosis resulting from ALI may be pulmonary fibrosis.
[0138] In some aspects, the present disclosure provides a method of treating intestinal fibrosis, comprising administering to a patient an effective amount of a compound disclosed herein. In some embodiments, the intestinal fibrosis is mediated by ALK5. In some embodiments, the compound is administered in an amount effective to delay progression of, reduce the i ncidence of, or reduce the degree of one or more characteristics associated with intestinal fibrosis. In some embodiments, the compound is admi nistered, either in a single dose or over multiple doses, in an amount effective to reverse established fibrosis.
[0139] In some aspects, the present disclosure provides a method of treating cancer, comprising administering to a patient an effective amount of a compound disclosed herein. In some embodiments, the cancer is mediated by ALK5. In some embodiments, the cancer is selected from breast cancer, coion cancer, prostate cancer, lung cancer, hepatocellular carcinoma, glioblastoma, melanoma, and pancreatic cancer. In some embodiments, the cancer is lung cancer, such as non-small cell lung cancer. In some aspects, the present disclosure provides a method of treating cancer, such as non-small cell lung cancer, comprising administering to a patient an effective amount of a compound disclosed herein and an immunotherapeutic agent. In some embodiments, the cancer is stage III non-small cell lung cancer. In some embodiments, the method further comprises administering radiation to the patient. In some embodiments, the immunotherapeutic agent is a PD- 1 inhibitor or a CTLA-4 inhibitor. In some embodiments, the immunotherapeutic agent is selected from atezolizumab, a velum ab, nivolumab, pembrolizumab, durvaiumab, BGB-A317, tremelimumab, and ipilimumab. In some embodiments, the immunotherapeutic agent is selected from pembrolizumab and durvaiumab.
[0140] The compounds described herein, including compounds of Formula (I), are ALK5 inhibitors that limit the activity of TGFp. TGFP is one of several factors involved in the initiation and development of fibrotic diseases throughout the body. As such, the compounds of the disclosure are expected to be useful for the treatment, prevention and/or reduction of fibrosis in a patient by administering a therapeutically effective amount of a compound disclosed herein. By inhibiting ALK5, the compound is expected to potentiate the formation of fibrosis in areas of the body that suffer from excessive deposition of the extracellular matrix. Those areas are described below.
[0141] Systemic Fibrotic Diseases
[0142] Systemic sclerosis (SSc) is an autoimmune disorder that affects the skin and internal organs and results in autoantibody production, vascular endothelial activation of small blood vessels, and tissue fibrosis as a result of fibroblast dysfunction. Transforming growth factor p (TGF-β) has been identified as a regulator of pathological fibrogenesis in SSc (Ayers, N.B., et al., Journal of Biomedical Research, 2018, 32(1), pp. 3- 12). According to the authors, “understanding the essential role TGF-β pathways play in the pathology of systemic sclerosis could provide a potential outlet for treatment and a better understanding of this severe disease." In some embodiments, the present disclosure provides a method of treating SSc, comprising administering to a subject an effective amount of a compound disclosed herein.
[0143] Multifocal fibrosclerosis (MF) and. idiopathic multifocal fibrosclerosis (IMF) are disorders characterized by fibrous lesions at varying sites and include retroperitoneal fibrosis, mediastinal fibrosis and Riedel’s thyroiditis. Both multifocal fibrosclerosis and idiopathic multifocal fibrosclerosis are considered to be an outcome of IgG-; -associated fibrosis/disease and TGF-β is believed to be one factor involved in the initiation and development of fibrosis (Pardali, E., et al., Int. J. Mol. Set., 18, 2157, pp. 1-22). In some embodiments, the present disclosure provides a method of treating multifocal fibrosclerosis or idiopathic multifocal fibrosclerosis, comprising administering to a subject an effective amount of a compound disclosed herein.
[0144] In some embodiments, the present disclosure provides a method of treating nephrogenic systemic fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein.
Nephrogenic systemic fibrosis is a rare disease occurring mainly in people with advanced kidney failure with or without dialysis. In a study performed by Kelly et al. (J. Am. Acad. Dermatol., 2008, 58, 6, pp. 1025- 1030), it was shown that TGF-β, as well as Smad 2/3, appear to be associated with fibrosis seen in nephrogenic systemic fibrosis. [0145] Sclerodermatous graft-versus-host disease (GVHD) is a prevalent complication of allogeneic hematopoietic stem cell graft appearing two to three months after allogeneic bone marrow transplantation. The disease results in production of autoantibodies and fibrosis of skin and inner organs. Using a murine cutaneous GVHD model, it has been shown that progression of early skin and lung disease can be inhibited with TGF-β neutralizing antibodies (McCormick, L.L., et al., J. Immunol., 1999, 163, pp. 5693-5699). In some embodiments, the present disclosure provides a method of treating sclerodermatous GVHD, comprising administering to a subject an effective amount of a compound disclosed herein.
[0146] Organ- specific Fibrotic Diseases
[0147] Cardiac fibrosis refers to the abnormal thickening of heart valves due to the abnormal proliferation of cardiac fibroblasts resulting in excess deposition of ECM in heart muscle. Fibroblasts secrete collagen, which serves as structural support for the heart. However, when collagen is excessively secreted in the heart, wall and valve thickening can result in tissue build -up on the tricuspid and pulmonary valves. This in turn causes loss of flexibility and ultimately valvular dysfunction leading to heart failure. A specific type of cardiac fibrosis is hypertension-associated cardiac fibrosis as described by J. Diez (J. Clin. Hypertens. , 2007, July 9(7), pp. 546-550). According to Diez, changes in the composition of cardiac tissue develop in hypertensive patients with left ventricular hypertrophy and lead to structural remodeling of the heart tissue. One change relates to the disruption of the equilibrium between the synthesis and degradation of collagen types I and III molecules, resulting in excessive accumulation of collagen fibers in the heart tissue. Other types of cardiac fibrosis include post-myocardial infarction and Chagas disease-induced myocardial fibrosis. In Chagas disease, transforming growth factor pi ( TGF-β1) has been implicated in Chagas disease physiopathology, where animal models suggest that the TGF-β1-pathway is up-regulated during infection (Araujo -Jorge, T.C., el al., Clin. Pharmacol. Then, 2012, 92(5), pp. 613-621; Curvo, E., Mem Inst Oswaldo Cruz, 2018, Vol.
113(4), e170440, pp. 1-8). In some embodiments, the present disclosure provides a method of treating various forms of cardiac fibrosis, such as hypertension-associated cardiac fibrosis, post -myocardial infarction or Chagas disease-induced myocardial fibrosis, comprising admi nistering to a subject an effective amount of a compound disclosed herein.
[11148] Renal fibrosis encompasses a variety of disorders associated with the aberrant expression and activity of TGF-β, including, but not limited to, diabetic and hypertensive nephropathy, urinary tract obstruction- induced kidney fibrosis, inflammatory/autoimmune-induced kidney fibrosis, aristolochic acid nephropathy, progressive kidney fibrosis, and. polysystic kidney disease. As discussed above, fibrosis involves an excess accumulation of the ECM, which in turn causes loss of function when normal tissue is replaced with scar tissue (Wynn, T.A., J Clin Invest., 2007, 117, pp. 524-529). As early as 2005, ALK5 inhibitors were being studied in models for renal disease (Laping, NJ., Current Opinion in Pharmacology, 2003, 3, pp. 204-208). In some embodiments, the present disclosure provides a method of treating renal fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein. [0149] One fibrotic disease that has been particularly difficult to treat is idiopathic pulmonary fibrosis (IPF). IPF is a chronic, progressive and fatal fibrotic lung disease with survival only improved by lung transplantation. Current oral therapies such as nintedanib and pirfenidone have been shown to slow the progression of the disease, but have adverse effects that lead to discontinuation and lack of compliance by the patient. Although there are other therapies in development targeting various pathways, an unmet need remains for patients with IPF.
[0150] Although ALK5 is an important and known component in the fibrotic disease pathway, the efficacy of ALK5 inhibitors in IPF have not been realized due to systemic adverse effects, especially in the heart. Thus, one of the goals of this disclosure is to develop ALK5 inhibitors with high lung selectivity and rapid clearance. One exemplary embodiment of this disclosure is to treat patients with idiopathic pulmonary fibrosis with a compound described herein, for example, by once or twice daily administration of inhalable ALK5 inhibitor having minimal systemic exposure. The inhaled ALK5 inhibitor may be administered as a monotherapy or co- dosed with other orally available IPF therapies. In some embodiments, the present disclosure provides a method of treating idiopathic pulmonary fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein. In some embodiments, the compound is administered by inhalation.
[0151] Familial pulmonary fibrosis is a hereditary disease where two or more family members have confirmed IPF. In some embodiments, the present disclosure provides a method of treating familial pulmonary fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein.
[0152] Pulmonary fibrosis is a typical clinical feature associated with viral infection, such as SARS and COVID-19. SARS-mediated TGF-β signaling has been shown to promote fibrosis and block apoptosis of SARS-CoV-infected host cells (Zhao, X. et al. J. Biol Chem., 2008, 283(6), pp. 3272-3280). Increased TGF- p expression was similarly observed in patients infected with SARS-CoV-2, ultimately leading to the development of pulmonary fibrosis. TGF~P signaling mediated by SARS-CoV-2 can promote fibroblast proliferation and myofibroblast differentiation and block host cell apoptosis. (Xiong, Y. el al., Emerging Microbes & Infections, 2020, 9( 1), pp. 761-770). Compounds of the present disclosure are expected to inhibit increased TGF-β signaling mediated by viral infection and prevent, halt, slow or reverse the progression of pulmonary fibrosis associated with the infection. Accordingly, in some embodiments, the present disclosure provides a method of treating pulmonary fibrosis induced by a viral infection, comprising administering to a subject an effective amount of a compound disclosed herein. In some embodiments, the pulmonary fibrosis is induced by SARS-CoV or SARS-CoV-2. In some embodiments, the compound is administered by inhalation. [0153] Chronic lung disease, such as interstitial lung disease (ILD), chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF), may lead to pulmonary hypertension (PH). Pulmonary hypertension is a progressive disease characterized by high blood pressure in the lungs. The World Health Organization (WHO) has defined five classifications of PH (WHO Group I: Pulmonary arterial hypertension (PAH); WHO Group II: Pulmonary hypertension due to left heart disease; WHO Group III: Pulmonary hypertension due to lung disease and/or hypoxia; WHO Group IV: Chronic thromboembolic pulmonary hypertension (CTEPH); and WHO Group V: Pulmonary hypertension with unclear multifactorial mechanisms). TGF-β signaling has been implicated in the pathogenesis of PH. Moreover, inhibition of ALK5 in a monocrotaline (MCT) model of severe PH was shown to attenuate the development of PH and reduce pulmonary vascular remodeling in a dose-dependent manner, namely by reducing RV systolic pressure, reducing RV diastolic pressure, increasing cardiac output and reducing RV hypertrophy (Zaiman, A. L.; et al., Am. J. Respir. Crit. Care Med., 2008, 177, pp. 896-905). Compounds of the present disclosure are expected to inhibit TGF-β signaling in lung tissue and prevent, halt, slow, or reverse the progression of PH, particularly in WHO Group III PH. Accordingly, in some embodiments, the present disclosure provides a method of treating pulmonary hypertension, comprising administering to a subject an effective amount of a compound disclosed herein. The pulmonary hypertension may be WHO Group III pulmonary hypertension, such as pulmonary fibrosis-related pulmonary hypertension (PH-PF) or interstitial lung disease-related pulmonary hypertension (PH-ILD). In some embodiments, the compound is administered by inhalation.
[0154] Other types of interstitial lung diseases include, but are not limited to, (1) interstitial pneumonia caused by bacteria, viruses, or fungi; (2) nonspecific interstitial pneumonitis usually associated with autoimmune conditions such as rheumatoid arthritis or scleroderma; (3) hypersensitivity pneumonitis caused by inhalation of dust, mold, or other irritants; (4) cryptogenic organizing pneumonia; (5) acute interstitial pneumonitis; (6) desquamative interstitial pneumonitis; (7) sarcoidosis; (8) drag-induced interstitial lung disease; and (9) progressive fibrosing interstitial lung disease (PF-ILD). In some embodiments, the present disclosure provides a method of treating an interstitial lung disease, comprising administering to a subject an effective amount of a compound disclosed herein.
[0155] Both transforming growth factor (TGF)-beta(l) and activin-A have been implicated in airway remodeling in asthma (Kariyawasam, H.H., J Allergy Clin Immunol., 2009, September, 12.4(3), pp. 454-462). In some embodiments, the present disclosure provides a method of treating asthma, comprising administering to a subject an effective amount of a compound disclosed herein.
[0156] Chronic obstructive pulmonary disease (COPD) is a pulmonary disorder characterized by a poorly reversible and progressive airflow limitation caused by airway inflammation and emphysema, whereas IFF is associated with impaired diffusion capacity (Chilosi, M., et al., Respir. Res., 2012, 13(1), 3, pp. 1-9). Both diseases, however, demonstrate a progressive loss of alveolar parenchyma leading to severe impairment of respiratory function. Fibrosis associated with emphysema is known and research has demonstrated TGF-β 1 involvement in chronic sinus disease, pulmonary fibrosis, asthma, and COPD (Yang, Y.C., et al., Allergy, 2012, 67, pp. 1193 -1202). In some embodiments, the present disclosure provides a method of treating COPD, comprising administering to a subject an effective amount of a compound disclosed herein. [0157] Other types of lung injury that result in fibrosis include silica-induced pneumoconiosis (silicosis), asbestos-induced pulmonary fibrosis (asbestosis), and chemotherapeutic agent-induced pulmonary fibrosis. In some embodiments, the present disclosure provides a method of treating injury-induced fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein.
[0158] In some embodiments, the present disclosure provides a method of treating liver fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein. Fibrosis develops in the liver when it is repeatedly or continuously damaged, for example, in patients with chronic hepatitis. TGF-β signaling participates in all stages of disease progression, from, initial liver injury through inflammation and fibrosis, to cirrhosis and cancer (Fabregat, I., et al., The FEES J., 2016, 283(12), pp. 2219-2232).
[0159] A related condition involves fibrosis resulting from idiopathic non-cirrhotic portal hypertension (INCPH). This disease is of uncertain etiology characterized by periportal fibrosis and involvement of small and medium branches of the portal vein. According to Nakanuma et al., small portal veins and skin findings are similar between patients with scleroderma and INCPH (Nakanuma, Y., Hepatol. Res., 2009, 39, pp. 1023-1031). Transforming growth factor-p (TGF-β) and connective tissue growth factor, which are fibrosis-related and vascular endothelial growth factors, respectively, increase in serum, skin, and the portal vein, suggesting that these could be mechanisms of the portal vein occlusion in INCPH. Moreover, endothelial mesenchymal transition (EndMT) theory was proposed by Kitao et al. based on these findings (Kitao, A., et al., Am. J. Pathol., 2009, 175, pp. 616-626). The increase of TGF-β in sera may act as a potent inducer of EndMT. In some embodiments, the present disclosure provides a method of treating INCPH, comprising administering to a subject an effective amount of a compound disclosed herein.
[0160] Other types of liver fibrosis include alcoholic and non-alcoholic liver fibrosis, hepatitis C -induced liver fibrosis, primary biliary cirrhosis or cholangitis, and parasite-induced liver fibrosis (schistosomiasis). In some embodiments, the present disclosure provides a method of treating alcoholic liver fibrosis, nonalcoholic liver fibrosis, hepatitis C -induced liver fibrosis, primary biliary cirrhosis, primary biliary cholangitis, or parasite- induced liver fibrosis (schistosomiasis), comprising administering to a subject an effective amount of a compound disclosed herein.
[0161] Primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC) are two types of chronic liver disease that often lead to cirrhosis and liver failure. Liver biopsies of patients with PBC or PSC typically reveal inflammation and fibrosis. Inhibition of integrin αvβ6, which has been shown to bind to and activate TGFpl on epithelial cells, suppresses biliary fibrosis in rodents. (Peng, Z-W., et al., Hepatology, 2016, 63, pp. 217-232). Accordingly, inhibition of the TGF-β pathway is also expected to suppress fibrotic processes in both PBC and PSC. Compounds of the present disclosure are expected to inhibit TGF-β signaling in liver tissue and prevent, halt, slow or reverse the progression of PBC and PSC. Thus, in some embodiments, the present disclosure provides a method, of treating primary biliary cholangitis or primary sclerosing cholangitis, comprising administering to a subject an effect amount of a compound described herein. In some embodiments, the present disclosure provides a method of treating liver fibrosis, optionally in a subject that suffers from PBC or PSC, comprising administering to the subject an effective amount of a compound described herein.
[0162] Fibrotic skin conditions include, but are not limited to, hypertrophic scarring, keloids, and localized or systemic sclerosis (scleroderma). As discussed previously, TGF-β is a potent stimulus of connective tissue accumulation and has been implicated in the pathogenesis of scleroderma and other fibrotic disorders (Lakos, G., et al., Am. J. Pathol., 2004, 165(1), pp. 203-217). Lakos et.al. demonstrated that Smad3 functions as a key intracellular signal transducer for profibrotic TGF-β responses in normal skin fibroblasts and found that the targeted disruption of TGF-β/Smad3 signaling modulated skin fibrosis in the mouse model of scleroderma. In some embodiments, the present disclosure provides a method of treating skin fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein.
[0163] Intestinal fibrosis is a common complication of inflammatory bowel disease (IBD) and is a serious clinical problem. TGF-β has been implicated as a major driving factor of intestinal fibrosis. Moreover, TGF- pl signaling contributes to stricture formation in fibrostenotic Crohn’s disease by inducing insulin-like growth factor I (IGF-I) and mechano-growth factor (MGF) production in intestinal smooth muscle. (Lateila, G., Rieder, F., Cure. Opin, Gastroenterol., 2017, 33(4), pp. 239-245). Inhibition of TGF~p signaling could thus slow, halt or reverse the progression of fibrosis in the intestine. However, adverse side effects of concern to patients with IBD — such as inflammation and neoplasia — would likely result from systemic inhibition of TGF-β signaling. One goal of the present disclosure is to develop ALK5 inhibitors with high selectivity for the gastrointestinal tract and rapid clearance. In some embodiments, the present disclosure provides a method of treating intestinal fibrosis, comprising administering to a subject an effective amount of a compound described herein, for example, by once or twice daily administration of an oral ALK5 inhibitor having minimal systemic exposure. In some embodiments, the subject suffers from inflammatory bowel disease, such as Crohn’s disease or colitis. The degree of therapeutic efficacy may be with respect to a starting condition of the subject (e.g., a baseline Mayo score, baseline Lichtiger score, or severity or incidence of one or more symptoms), or with respect to a reference population (e.g., an untreated population, or a population treated with a different agent). Severity of intestinal fibrosis may be assessed using any suitable method, such as delayed enhancement MRI, ultrasound elastography, shear wave elastography, magnetization MRI, or by the direct detection of macromolecules such as collagen. In some embodiments, treatment with a compound of the present disclosure reduces the severity of the fibrosis, such as from severe fibrosis to moderate or mild fibrosis. In some embodiments, the treatment increases intestinal tissue elasticity, reduces tissue stiffness, and/or reduces collagen levels. In some embodiments, the treatment prevents myofibroblast accumulation, inhibits expression of pro-fibrotic factors, and/or inhibits accumulation of fibrotic tissue.
[0164] Other types of organ-specific fibrosis or fibrotic diseases involving the TGF-β pathway include, but are not limited to, radiation-induced fibrosis (various organs), bladder fibrosis, intestinal fibrosis, peritoneal sclerosis, diffuse fasciitis, Dupuytren’s disease, myelofibrosis, oral submucous fibrosis, and retinal fibrosis. In some embodiments, the present disclosure provides a method of treating radiation-induced fibrosis, bladder fibrosis, intestinal fibrosis, peritoneal sclerosis, diffuse fasciitis, Dupuytren’s disease, myelofibrosis, oral submucous fibrosis, or retinal fibrosis, comprising administering to a subject an effective amount of a compound disclosed herein.
[0165] Although one of the goals of this disclosure is to treat fibrotic and pulmonary diseases locally or in a targeted way, the compounds described herein may also be used to treat patients systemically. Diseases that may be treated systemically, include, for example, oncologic diseases such as glioblastoma, pancreatic cancer and hepatocellular carcinoma, breast cancer metastasized to lungs, non-small cell lung cancer, small cell lung cancer, cystic fibrosis, and metastasis of other forms of primary cancer subtypes. Some of the forgoing diseases may also be treated locally as well.
[0166] Other fibrotic diseases that compounds disclosed herein may treat include angioedema, anti -aging, and alopecia. Alopecia includes alopecia totalis, alopecia uni versalis, androgenetic alopecia, alopecia areata, diffuse alopecia, postpartum alopecia, and traction alopecia.
[0167] Other Indications
[0168] In certain aspects, the present disclosure provides a method of reversing one or more symptoms of aging, comprising administering to a subject an ALK5 inhibitor. The method may further comprise administering an activator of the MAPK pathway, such as oxytocin. The method may be effective in one or more of enhancing neurogenesis in the hippocampus, reducing neuroinflammation, improving cognitive ability, reducing liver adiposity, reducing liver fibrosis, and decreasing the number of p16+ cells. In some embodiments, a method described herein increases stem cell activity . The increase in stem cell activity may allow the subject to generate new muscle fibers and/or to form new neurons in the hippocampus. Treatment with an ALK5 inhibitor, such as a compound described herein, may prevent or slow the onset of age-related diseases, such as Alzheimer’s disease, (see Mehdipour, M. et al. Aging 2018, 10, 5628-5645).
[0169] Pharmaceutical Compositions
[0170] In some aspects, the present disclosure provides a pharmaceutical composition. The pharmaceutical composition may comprise a compound disclosed herein, such as a compound of Formula (I), and a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition is formulated for oral administration. In some embodiments, the pharmaceutical composition is formulated for inhalation. In some embodiments, the pharmaceutical composition comprises a compound disclosed herein and an additional therapeutic agent. Non-limiting examples of such therapeutic agents are described herein below. [0171] Pharmaceutical compositions typically include at least one pharmaceutically acceptable carrier, diluent or excipient and at least one compound of Formula (I), or a compound provided in Table 1 — described herein as the active agent. The active agent may be provided in any form suitable for the particular mode of administration, such as a free base, a free acid, or a pharmaceutically acceptable salt. Additionally, the methods and pharmaceutical compositions of the present disclosure include the use of N-oxides, crystalline forms (e.g., polymorphs), as well as metabolites of these compounds having similar activity. All tautomers of the compounds described herein are included within the scope of the present disclosure. Additionally, the compounds described herein encompass unsolvated, as well as solvated, forms with pharmaceutically acceptable solvents such as water, ethanol and the like.
[0172] Suitable routes of administration include, but tire not limited to, oral, intravenous, rectal, vaginal, aerosol, pulmonary, nasal, transmucosal, topical, transdermal, otic, ocular, and parenteral modes of administration. In addition, by way of example only, parenteral delivery includes intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathecal, direct intraventricular, intraperitoneal, intralymphatic, and intranasal injections.
[0173] In certain embodiments, a compound described herein is administered in a local rather than systemic manner, for example, via injection of the compound directly into an organ, often in a depot preparation or sustained, release formulation. In some embodiments, a long acting formulation is administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection. In some embodiments, a compound described herein is provided in the form of a rapid release formulation, an extended release formulation, or an intermediate release formulation. In some embodiments, a compound described herein is provided in the form of a nebulized formulation. In some embodiments, a compound described herein is administered locally to the lungs by inhalation.
[0174] Compounds of the present disclosure are effective over a wide dosage range. For example, in the treatment of adult humans, dosages from 0.01 to 1000 mg, 0.5 to 100 mg, 1 to 50 mg, or from 5 to 40 mg per day may be administered to a subject in need thereof. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
[0175] A compound of the present disclosure may be administered in a single dose. In some embodiments, a compound of the disclosure is administered in multiple doses, such as about once, twice, three times, four times, five times, six times, or more than six times per day. In some embodiments, dosing is about once a month, once every two weeks, once a week, or once every other day. In some embodiments, a compound of the disclosure and an additional therapeutic agent are administered together about once per day to about 6 times per day. In some embodiments, the administration continues for more than about 6, 1.0, 14, 28 days, two months, six months, or more than about one year. In some embodiments, a dosing schedule is maintained as long as necessary. A compound of the present disclosure may be administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
[0176] Pharmaceutical compositions of the present disclosure typically contain a therapeutically effective amount of a compound of the present disclosure. Those skilled in the art will recognize, however, that a pharmaceutical composition may contain more than a therapeutically effective amount, e.g., bulk compositions, or less than a therapeutically effective amount, e.g., individual unit doses designed for coadministration to achieve a therapeutically effective amount.
[0177] Typically, pharmaceutical compositions of the present disclosure contain from about 0.01 to about 95% by weight of the active agent; including, for example, from about 0.05 to about 30% by weight; and from about 0.1 % to about 10% by weight of the active agent.
[0178] Any conventional carrier or excipient may be used in the pharmaceutical compositions of the present disclosure. The choice of a particular carrier or excipient, or combinations of carriers or excipients, will depend on the mode of administration being used to treat a particular patient or type of medical condition or disease slate. Additionally, the earners or excipients used in the pharmaceutical compositions of this disclosure may be commercially-available. Conventional formulation techniques are described in Remington: The Science and Practice of Pharmacy, 20th Edition, Lippincott Williams & White, Baltimore, Maryland (2000); and H.C. Ansel et at, Pharmaceutical Dosage Forms and Drag Delivery Systems, 7th Edition, Lippincott Williams & White, Baltimore, Maryland (1999).
[0179] Representative examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, the following: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, such as microcrystalline cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and. cellulose acetate; powdered, tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical compositions.
[0180] Pharmaceutical compositions are typically prepared by thoroughly and i ntimately mixing or blending the active agent with a p.barmaceutically-acceptable carrier and one or more optional ingredients. The resulting uniformly blended mixture can then be shaped or loaded into tablets, capsules, pills and the like using conventional procedures and equipment.
[0181] In one aspect, the pharmaceutical composition is suitable for inhaled administration. Pharmaceutical compositions for inhaled administration are typically in. the form of an aerosol or a powder. Such compositions are generally administered, using inhaler delivery devices, such as a dry powder inhaler (DPI), a metered-dose inhaler (MDI), a nebulizer inhaler, or a similar delivery device.
[0182] In a particular embodiment, the pharmaceutical composition is administered by inhalation using a dry powder inhaler. Such dry powder inhalers typically administer the pharmaceutical composition as a free- flowing powder that is dispersed, in a patient's air-stream during inspiration. In order to achieve a free-flowing powder composition, the therapeutic agent is typically formulated with a suitable excipient such as lactose, starch, mannitol, dextrose, polylactic acid (PL A), polylactide-co-glycolide (PLGA) or combinations thereof. Typically, the therapeutic agent is micronized and combined with a suitable carrier to form a composition suitable for inhalation.
[0183] A representative pharmaceutical composition for use in a dry powder inhaler comprises lactose and a micronized form of a compound disclosed herein. Such a dry powder composition can be made, for example, by combining dry milled lactose with the therapeutic agent and then dry blending the components. The composition is then typically loaded into a dry powder dispenser, or into inhalation cartridges or capsules for use with a dry powder delivery device.
[0184] Dry powder inhaler delivery devices suitable for administering therapeutic agents by inhalation are described in the art and examples of such devices are commercially available. For example, representative dry powder inhaler delivery devices or products include Aeolizer (Novartis); Airmax (TV AX); ClickHaler (Innovata Biomed); Diskhaler (GlaxoSmithKline); Diskus/Accuhaler (GlaxoSmithKline); Eliipta (GlaxoSmithKline); Easyhaler (Orion Pharma); Eclipse (Aventis); FlowCaps (Hovione); Handihaler (Boehringer Ingelheim); Pulvinal (Chiesi); Rotahaier (GlaxoSmithKline); SkyeHaler/Certihaler (SkyePharma); Twisthaler (Schering-Plough); Turbuhaler (AstraZeneca); Ultrahaler (Aventis); and the like. [0185] A pharmaceutical composition of the present disclosure may be administered by inhalation, using a metered -dose inhaler. Such metered -dose inhalers typically discharge a measured amount of a therapeutic agent using a compressed propellant gas. Accordingly, pharmaceutical compositions administered using a metered-dose inhaler typically comprise a solution or suspension of the therapeutic agent in a liquefied propellant. Any suitable liquefied propellant may be employed, including hydrofluoroalkanes (HFAs), such as 1,1,1,2-tetrafluoroethane (HF A 134a) and 1,1,1,2,3,3,3-heptafluoro-n-propane, (HF A 227); and chlorofluorocarbons, such as CCI3F. In a particular embodiment, the propellant is a hydrofluoroalkane. In some embodiments, the hydrofluoroalkane formulation contains a co-solvent, such as ethanol or pentane, and/or a surfactant, such as sorbitan trioleate, oleic acid, lecithin, and glycerin.
[0186] A representative pharmaceutical composition for use in a metered-dose inhaler comprises from about 0.01% to about 5% by weight of a compound of the present disclosure; from about 0% to about 2.0%' by weight ethanol; and from about 0% to about 5% by weight surfactant; with the remainder being an HF A propellant. Such compositions are typically prepared by adding chilled or pressurized hydrofluoroalkane to a suitable container containing the therapeutic agent, ethanol (if present) and the surfactant (if present). To prepare a suspension, the therapeutic agent is micronized. and then combined with the propellant. The composition is then loaded into an aerosol canister, which typically forms a portion of a metered-dose inhaler device.
[0187] Metered-dose inhaler devices suitable for administering therapeutic agents by inhalation are described in the art and examples of such devices are commercially available. For example, representative metered-dose inhaler devices or products include AeroBid Inhaler System (Forest Pharmaceuticals); Atrovent Inhalation Aerosol (Boehringer Ingelheim); Flovent (GlaxoSmithKline); Maxair Inhaler (3M); Proventil Inhaler (Schering); Serevent Inhalation Aerosol (GlaxoSmithKline); and the like.
[0188] A pharmaceutical composition of the present disclosure may be administered by inhalation using a nebulizer inhaler. Such nebulizer devices typically produce a stream of high velocity air that causes the pharmaceutical composition to spray as a mist that is carried into the patient's respiratory tract. Accordingly, when formulated for use in a nebulizer inhaler, the therapeutic agent can be dissolved in a suitable carrier to form a solution. Alternatively, the therapeutic agent can be micronized or nanomilled and combined with a suitable carrier to form a suspension.
[0189] A representative pharmaceutical composition for use in a nebulizer inhaler comprises a solution or suspension comprising from about 0.05 pg/mL to about 20 mg/mL of a compound of the present disclosure and excipients compatible with nebulized formulations. In one embodiment, the solution has a pH of about 3 to about 8.
[0190] Nebulizer devices suitable for administering therapeutic agents by inhalation are described in the art and examples of such devices are commercially available. For example, representative nebulizer devices or products include the Respimat® SoftmistlM Inhalaler (Boehringer Ingelheim); the AERx® Pulmonary Delivery System (Aradigm Corp.); the PARI LC Plus® Reusable Nebulizer or PARI eF1ow®rapid Nebulizer System (Pari GmbH); and. the like.
[0191] A pharmaceutical composition of the present disclosure may be prepared in a dosage form intended for oral administration. Suitable pharmaceutical compositions for oral administration may be in the form of capsules, tablets, pills, lozenges, cachets, dragees, powders, granules; or as a solution or a suspension in an aqueous or non-aqueous liquid: or as an oil-in-water or water-in-oil liquid, emulsion; or as an elixir or syrup; and the like; each containing a predetermined amount of a compound of the present disclosure as an active ingredient.
[0192] When intended for oral administration in a solid dosage form, the pharmaceutical compositions of the disclosure will typically comprise the active agent and one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate. Optionally or alternatively, such solid dosage forms may also comprise: fillers or extenders, binders, humectants, solution retarding agents, absorption accelerators, wetting agents, absorbents, lubricants, coloring agents, and buffering agents. Release agents, wetting agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the present pharmaceutical compositions.
[0193] Alternative formulations may include controlled release formulations, liquid dosage forms for oral administration, transdermal patches, and parenteral formulations. Conventional excipients and methods of preparation of such alternative formulations are described, for example, in the reference by Remington, supra. [0194] The following non-limiting examples illustrate representative pharmaceutical compositions of the present disclosure. [0195] Dry Powder Composition
[0196] A micronized compound of the present disclosure (1 g) is blended with milled lactose (25 g). This blended mixture is then loaded into individual blisters of a peelable bl ister pack in an amount sufficient to provide between about 0.1 mg to about 4 mg of the compound per dose. The contents of the blisters are administered using a dry powder inhaler.
[0197] Dry Powder Composition
[0198] A micronized compound of the present disclosure (1 g) is blended with milled lactose (20 g) to form a bulk composition having a weight ratio of compound to milled lactose of 1 :20. The blended composition is packed into a dry powder inhalation device capable of delivering between about 0.1 mg to about 4 mg of the compound per dose.
[11199] Metered-Dose Inhaler Composition
[0200] A micronized compound of the present disclosure (10 g) is dispersed in a solution prepared by dissolving lecithin (0.2 g) in demineralized water (200 mL). The resulting suspension is spray dried and then micronized to form a micronized composition comprising particles having a mean diameter less than about 1.5 pm. The micronized composition is then loaded into metered-dose inhaler cartridges containing pressurized 1,1, 1,2- tetrafluoroethane in an amount sufficient to provide about 0.1 mg to about 4 mg of the compound per dose when administered by the metered dose inhaler.
[0201] Nebulizer Composition
[0202] A representative nebulizer composition is as follows. A compound of the present disclosure (2 g of free -base equivalents) is dissolved in a solution containing 80 ml, reverse -osmosis water, 0.1-1% by weight of anhydrous citric acid, and 0.5- 1.5 equivalents of hydrochloric acid, followed by addition of sodium hydroxide to adjust the pH to 3.5 to 5.5. Thereafter, between 4-6% by weight of D-mannitol is added and solution q.s. to 100 ml.,. The solution is then filtered through a 0.2 pm filter and stored at room temperature prior to use. The solution is administered using a nebulizer device that provides about 0.1 mg to about 4 mg of the compound per dose.
[0203] Kits
[11204] In certain aspects, the present disclosure provides a kit comprising one or more unit doses of a compound or pharmaceutical composition described herein, optionally wherein the kit further comprises instructions for using the compound or pharmaceutical composition. In some embodiments, the kit comprises a carrier, package, or container that is compartmentalized to receive one or more containers, such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in a method described herein. Suitable containers include, for example, bottles, vials, syringes, and test tubes. The containers may be formed from, a variety of materials, such as glass or plastic.
[0205] The articles of manufacture provided herein may contain packaging materials. Packaging materials for use in packaging pharmaceutical products include those found in, e.g., U.S. Pat. Nos. 5,323,907, 5,052,558 and 5,033,252. Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment. For example, the container/ s) may include one or more compounds described herein, optionally in a composition or in combination with another agent as disclosed herein. The container(s) may optionally have a sterile access port (for example, the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). Such kits may optionally comprise a compound with an identifying description or label or instructions relating to its use in the methods described herein.
[0206] In some embodiments, a kit includes one or more containers, each with one or more of various materials (such as reagents, optionally in concentrated form, and/or devices) desirable from a commercial and user standpoint for use of a compound described herein. Nonlimiting examples of such materials include, but are not limited to, buffers, diluents, filters, needles, syringes, carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use. A set of instructions will also typically be included. A label is optionally on or associated with the container. For example, a label is on a container when letters, numbers or other characters forming the label are attached, molded, or etched onto the container itself, a label is associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert. In addition, a label is used to indicate that the contents are to be used for a specific therapeutic application. In addition, the label indicates directions for use of the contents, such as in the methods described herein. In certain embodiments, the pharmaceuticai composition is presented in a pack or dispenser device which contains one or more unit dosage forms containing a compound provided herein. The pack may contain metal or plastic foil, such as a blister pack. In some embodiments, the pack or dispenser device is accompanied by instructions for administration. Optionally, the pack or dispenser is accompanied with a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, is the labeling approved by the U.S. Food and. Drug Administration for prescription drugs, or the approved product insert. In some embodiments, compositions containing a compound provided herein formulated in a compatible pharmaceutical carrier are prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
[0207] Combination Therapy
[0208] The compounds and pharmaceutical compositions of the disclosure may be used in combination with one or more therapeutic agents which act by the same mechanism or by a different mechanism to treat a disease. The one or more agents may be administered sequentially or simultaneously, in separate compositions or in the same composition. Useful classes of agents for combination therapy include, but are not limited to, compounds used to treat cardiac, kidney, pulmonary, liver, skin, immunological and oncological conditions.
[0209] In practicing any of the subject methods, an ALK5 inhibitor and a second therapeutic agent can be administered sequentially, wherein the two agents are introduced into a subject at two different time points. The two time points can be separated by more than 2 hours, 1 or more days, 1 or more weeks, 1 or more months, or according to any intermittent regimen schedule disclosed herein.
[0210] In some embodiments, the ALK5 inhibitor and the second therapeutic agent are administered simultaneously. The two agents may form, part of the same composition, or the two agents may be provided in one or more unit doses.
[0211] In some embodiments, the ALK5 inhibitor or the second therapeutic agent are administered parenterally, orally, inhalatively, intraperitoneally, intravenously, intraarterially, transdermally, intramuscularly, liposomally, via local delivery by catheter or stent, subcutaneously, intraadiposally, or intrathecally. As used herein, a therapeutically effective amount of a combination of an ALK5 inhibitor and a second therapeutic agent refers to a combination of an ALK5 inhibitor and a second therapeutic agent, wherein the combination is sufficient to affect the intended application, including but not limited to, disease treatment, as defined herein. Also contemplated in the subject methods is the use of a sub-therapeutic amount of an ALK5 inhibitor and a second therapeutic agent in combination for treating an intended disease condition. The individual components of the combination, though present in sub- therapeutic amounts, synergistically yield an efficacious effect and/or reduced a side effect in an intended application.
[0212] The amount of the ALK5 inhibitor and the second therapeutic agent administered may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
[0213] Measuring an immune response and/or the inhibition of biological effects of ALK5 can comprise performing an assay on a biological sample, such as a sample from a subject. Any of a variety of samples may be selected, depending on the assay. Examples of samples include, but are not limited to blood samples (e.g. blood plasma or serum), exhaled breath condensate samples, bronchoalveolar lavage fluid, sputum samples, urine samples, and tissue samples.
[0214] A subject being treated with an ALK5 inhibitor and a second therapeutic agent may be monitored to determine the effectiveness of treatment, and the treatment regimen may be adjusted, based on the subject's physiological response to treatment. For example, if inhibition of a biological effect of ALK5 inhibition is above or below a threshold, the dosing amount or frequency may be decreased or increased, respectively. Alternatively, the treatment regimen may be adjusted with respect to an immune response. The methods can further comprise continuing the therapy if the therapy is determined to be efficacious. The methods can comprise maintaining, tapering, reducing, or slopping the administered amount of a compound or compounds in the therapy if the therapy is determined to be efficacious. The methods can comprise increasing the administered amount of a compound or compounds in the therapy if it is determined not to be efficacious. Alternatively, the methods can comprise stopping therapy if it is determined not to be efficacious. In some embodiments, treatment with an ALK5 inhibitor and a second therapeutic agent is discontinued if inhibition of the biological effect is above or below a threshold, such as in a lack of response or an adverse reaction. The biological effect may be a change in any of a variety of physiological indicators.
[0215] Specific agents that may be used in combination with the compounds disclosed herein include, but are not limited to, OFEV® (nintedanib) and Esbriet* (pirfenidone). In some embodiments, a compound disclosed herein is administered in combination with pirfenidone, optionally wherein the pirfenidone is administered by inhalation. In some embodiments, the present disclosure provides a method of treating fibrosis, such as idiopathic pulmonary fibrosis, in a subject, comprising administering to the subject an ALK5 inhibitor, such as a compound disclosed in Table 1 , and nintedanib or pirfenidone. In some embodiments, the present disclosure provides a method of treating cancer, such as lung cancer, in a subject, comprising administering to the subject an ALK5 inhibitor, such as a compound disclosed in Table 1, and nintedanib or pirfenidone.
[0216] In some embodiments, the present disclosure provides a method for treating a proliferative disorder (e.g., lung cancer) in a subject in need thereof, comprising administering to said subject an ALK5 inhibitor and an immunotherapeutic agent. TGF-β has been shown to regulate lymphocyte differentiation, suppress T cell proliferation and to enhance tumor growth. Moreover, TGF-β has been shown to prevent optimal activation of the immune system in immunotherapy-resistant patients (see Loffek, S. J. Oncolo. 2018, 1 -9; incorporated herein by reference in its entirety). Not wishing to be bound by any particular theory, the present inventors expect that inhibition of ALK5 may enhance the efficacy of a particular immunotherapy . As such, treatment with an immunotherapeutic agent, such as durvaiumab or pembrolizumab, and an ALK5 inhibitor, such as a compound of the present disclosure, is expected to improve the clinical outcome of a subject with cancer, such as a subject with iron-small cell lung cancer. The combination is expected to produce a synergistic effect. A synergistic combination is also expected for a triple combination of radiation therapy, immunotherapy, and ALK5 inhibition. In addition, the ALK5 inhibitor, even when administered, locally (e.g., to the lung by inhalation), may stimulate both local and systemic immune responses, allowing for the treatment of primary or metastatic tumors in tissues beyond the site of the local deli very. For example, an inhaled ALK5 inhibitor may be administered in combination with an immunotherapeutic agent to treat melanoma, renal cell carcinoma, colon cancer, or breast cancer.
[0217] In some embodiments, the ALK5 inhibitor and the immunotherapeutic agent are administered sequentially or simultaneously. In some embodiments, the ALK5 inhibitor and the immunotherapeutic agent are more effective in treating the proliferative disorder than either agent alone. In some embodiments, the ALK5 inhibitor and the immunotherapeutic agent yield a synergistic effect in treating the proliferative disorder. The synergistic effect may be a therapeutic effect that is greater than either agent used alone in comparable amounts under comparable conditions. The synergistic effect may be a therapeutic effect that is greater than results expected by adding the effects of each agent alone. In some embodiments, the proliferative disorder is a cancer condition. In some embodiments, the cancer condition is lung cancer, such as non-small cell lung cancer.
[0218] The term “inununotherapeutic agent” refers to any agent that induces, enhances, suppresses or otherwise modifies an immune response. This includes the administration of an active agent to, or any type of intervention or process performed on, the subject, with the objective of modifying an immune response. An immunotherapeutic agent may, for example, increase or enhance the effectiveness or potency of an existing immune response in a subject, for example, by stimulating mechanisms that enhance the endogenous host immune response or overcoming mechanisms that suppress the endogenous host immune response.
[0219] “Immune response” refers to the action of a cell of the immune system including, for example, B lymphocytes, T lymphocytes, natural killer (NK) cells, macrophages, eosinophils, mast cells, myeloid-derived suppressor cells, dendritic cells and neutrophils and soluble macromolecules produced by any of these cells or the liver (including antibodies, cytokines and complement), that results in selective targeting, binding to, damage to, destruction of, and/or elimination of invading pathogens, cells or tissues infected with pathogens, cancerous or other abnormal cells, or, in cases of autoimmunity or pathological inflammation, normal human cells or tissues, from the body of a subject.
[0220] In one embodiment, an immunotherapeutic agent may comprise a PD-1 inhibitor. In another embodiment, an immunotherapeutic agent may comprise a CTLA-4 inhibitor. In still another embodiment, an immunotherapeutic agent may comprise a B7 inhibitor.
[0221] Exemplary PD-1 inhibitors: A PD-1 inhibitor suitable for use in the subject methods can be selected from a variety of types of molecules. For example, the PD-1 inhibitor can be a biological or chemical compound, such as an organic or inorganic molecule, peptide, peptide mimetic, antibody or an antigenbinding fragment of an antibody. Some exemplary classes of agents suitable for use in the subject methods are detailed in the sections below. A PD-1 inhibitor for use in the present disclosure can be any PD-1 inhibitor that is known in the art, and can include any entity that, upon administration to a patient, results in inhibition of the PD-1 pathway in the patient. A PD-1 inhibitor can inhibit PD-1 by any biochemical mechanism, including disruption of any one or more of PD-1/PD-L1, PD1/PD-L2 and PD-L1/CD80 interactions.
[0222] In some embodiments, the PD-1 inhibitor is a molecule that inhibits the binding of PD-1 to its ligand binding partners. In a specific aspect, the PD-1 ligand binding partners are PD-L1 and/or PD-L2. In another embodiment, a PD-1 inhibitor is a molecule that inhibits the binding of PD-L1 to its binding partners. In a specific aspect, PD-L1 binding partners are PD1 and/or CD80. In another embodiment, the PD-1 inhibitor is a molecule that inhibits the binding of PD-L2. to its binding partners. In a specific aspect, a PD-L2 binding partner is PD1. The inhibitor may be an antibody, an antigen binding fragment thereof, an irnmunoadhesin, a fusion protein or oligopeptide. [0223] In some embodiments, the PD-1 inhibitor is an anti-PD-1 antibody. In some further embodiments, the anti-PD-1 antibody is capable of inhibiting binding between PD-1 and PD-L1. In another embodiment, the anti-PD-1 antibody is capable of inhibiting binding between PD-1 and PD-L2. In some embodiments, the PD- 1 inhibitor is an anti-PD-L1 antibody. In some embodiments, the anti-PD-L1 antibody is capable of inhibiting binding between PD-L1 and PD-1 and/or between PD-L1 and CD80. In some embodiments, the PD-1 inhibitor is an anti-PD-L2 antibody. In some further embodiments, the anti-PD-L2 antibody is capable of inhibiting binding between PD-1 and PD-L2. In yet another embodiment, the PD-1 inhibitor is nivolumab or pembrolizumab. In some embodiments, the PD-1 inhibitor is selected from atezohzumab, avelumab, nivolumab, pembrolizumab, durvalumab, and BGB-A317.
[0224] Inhibition of the PD-1 pathway can enhance the immune response to cancerous cells in a patient. The interaction between PD-1 and PD-L1 impairs T cell response as manifested by a decrease in tumor-infiltrating lymphocytes (TILs) and a decrease in T-cell receptor mediated proliferation, resulting in T cell anergy, exhaustion or apoptosis, and immune evasion by the cancerous cells. This immune suppression can be reversed by inhibiting the local interaction between PD-L1 and PD-1 using a PD-1 inhibitor, including, for example, an anti-PD-1 and/or an anti- PD-L1 Ab. A PD-1 inhibitor may improve or restore antitumor T-cell functions.
[0225] Anti-PD-1 antibodies suitable for use in the disclosure can be generated using methods well known in the art. Exemplary PD-1 inhibitors include, bitt are not limited to: nivolumab (BMS936558), pembrolizumab (MK-347.5), pidilizumab (CT-OU), AMP-224, AMP-514, BMS-936559, RG7446 (MPDL3280A), MDX- 1106 (Medarex Inc.), MSB0010718C, MEDI4736, and HenGtui mAB005 (WO 15/085847). Further PD-1 antibodies and other PD-1 inhibitors include those described in WO 04/056875, WO 06/121168, WO
07/005874, WO 08/156712, WO 09/014708, WO 09/114335, WO 09/101611, WO 10/036959, WO
10/089411, WO 10/027827, WO 10/077634, WO 11/066342, WO 12/145493, WO 13/019906, WO
13/181452, WO 14/022758, WO 14/100079, WO 14/206107, WO 15/036394, WO 15/085847, WO
15/112900, WO 15/112805, WO 15/112800, WO 15/109124, WO 15/061668, WO 15/048520. WO
15/044900, WO 15/036927, WO 15/035606; U. S. Pub. No. 2015/0071910; and U. S. Pat. Nos. 7,488,802;
7,521,051; 7.595,048; 7,722, 868; 7,794,710; 8,008,449; 8,354,509; 8,383,796; 8,652,465; and 8,735,553; all of which are incorporated herein by reference. Some anti-PD-1 antibodies are commercially available, for example from ABCAM (AB137132), BIOLEGEND (EH12.2H7, RMP 1-14) and AFFYMETRIX EBIOSCIENCE (J105, 1116, M1H4).
[0226] Exemplary CTLA-4 inhibitors: A CTLA-4 inhibitor suitable for use in the subject methods can be selected from a variety of types of molecules. For example, the CTLA-4 inhibitor can be a biological or chemical compound, such as an organic or inorganic molecule, peptide, peptide mimetic, antibody or an antigen-binding fragment of an antibody. Some exemplary classes of agents suitable for use in the subject methods are detailed in the sections below. A CTLA-4 inhibitor for use in the present disclosure can be any CTLA-4 inhibitor that is known in the art, and can include any entity that, upon administration to a patient, results in inhibition of the CTLA-4 pathway in the patient. A CTLA-4 inhibitor can inhibit CTLA-4 by any biochemical mechanism, including disruption of either one or both of CTLA-4/CD80 and CTLA -4/CD86 interactions.
[0227] In some embodiments, the CTLA-4 inhibitor is a molecule that inhibits the binding of CTLA-4 to its ligand binding partners. In a specific aspect, the CTLA-4 ligand binding partners are CD80 and/or CD86. In another embodiment, a CTLA-4 inhibitor is a molecule that inhibits the binding of CD80 to its binding partners. In a specific aspect, a CD80 binding partner is CTLA-4. In another embodiment, the CTLA-4 inhibitor is a molecule that inhibits the binding of CD86 to its binding partners. In a specific aspect, a CD86 binding partner is CTLA-4. The inhibitor may be an antibody, an antigen binding fragment thereof, an immunoadhesin, a fusion protein or oligopeptide.
[0228] In some embodiments, the CTLA-4 inhibitor is an anti-CTLA-4 antibody. In some further embodiments, the anti-CTLA-4 antibody is capable of inhibiting binding between CTLA-4 and CD80. In another embodiment, the anti-CTLA-4 antibody is capable of inhibiting binding between CTLA-4 and CD86. In some embodiments, the CTLA-4 inhibitor is an anti-CD80 antibody. In some embodiments, the anti-CD80 antibody is capable of inhibiting binding between CTLA-4 and CD80. In some embodiments, the CTLA-4 inhibitor is an anti-CD86 antibody. In some further embodiments, the anti-CD86 antibody is capable of inhibiting binding between CTLA-4 and CD86. In yet another embodiment, the CTLA-4 inhibitor is tremeiimumab or ipilimumab.
[0229] Inhibition of the CTLA-4 pathway can enhance the immune response to cancerous cells in a patient. The interaction between CTLA-4 and one of its natural ligands, CD80 and CD86, delivers a negative regulatory signal to T cells. This immune suppression can be reversed by inhibiting the local interaction between CD80 or CD86 and CTLA-4 using a CTLA-4 inhibitor, including, for example, an anti-CTLA-4 Ab, anti-CD80 Ab or an antiCD86 Ab. A CTLA-4 inhibitor may improve or restore antitumor T-cell functions. [0230] Anti-CTLA-4 antibodies suitable for use in the disclosure can be generated using methods well known in the art. Exemplary CTLA-4 inhibitors include but are not limited to tremeiimumab and ipilimumab (also known as 10D1 or MDX-010). Further CTLA-4 antibodies and other CTLA-4 inhibitors include those described in WO 98/042752, WO 00/037504, WO 01/014424 and WO 04/035607; LT. S. Pub. Nos.
2002/0039581, 2002/086014 and 2005/0201994; LT. S. Pat. Nos. 5,811,097; 5.855,887; 5,977.318; 6,051 ,227;
6,207, 156; 6.682,736; 6,984.720; 7, 109,003; 7, 132,281; 7,605,238; 8, 143,379; 8,318,916; 8.435,516;
8,784,815; and 8,883,984; EP Pat. No. 1212422; Hurwitz et al. , PNAS 1998, 95(17): 10067-10071; Camacho et al., J Clin Oncology 2004, 22(145): abstract no. 2505 (antibody CP675206); and Mokyr, et al., Cancer Research 1998, 58:5301-5304; each of which is incorporated herein by reference.
[0231] Also provided herein is a pharmaceutical composition comprising a compound of the disclosure or a pharmaceutically acceptable salt thereof and one or more other therapeutic agents. The therapeutic agent may be selected from the classes of agents specified above and from the lists of specific agents described above. In some embodiments, the pharmaceutical composition is suitable for delivery to the lungs. In some embodiments, the pharmaceutical composition is suitable for inhaled or nebulized administration. In some embodiments, the pharmaceutical composition is a dry powder or a liquid, composition.
[0232] Further, in a method aspect, the disclosure provides a method of treating a disease or disorder in a mammal comprising administering to the mammal a compound of the disclosure or a pharmaceutically acceptable salt thereof and one or more other therapeutic agents.
[0233] When used in combination therapy, the agents may be formulated in a single pharmaceutical composition, or the agents may be provided in separate compositions that are administered simultaneously or at separate times, by the same or by different routes of administration. Such compositions can be packaged separately or may be packaged together as a kit. The two or more therapeutic agents in the kit may be administered by the same route of administration or by different routes of administration.
EXAMPLES
[0234] The following examples are given for the purpose of illustrating various embodiments of the disclosure and are not meant to limit the present disclosure in any fashion. The present examples, along with the methods and compositions described herein, are presently representative of some embodiments, are exemplary, and are not intended as limitations on the scope of the disclosure. Changes therein and other uses which are encompassed within the spirit of the disclosure as defined by the scope of the claims will occur to those skilled in the art.
[0235] The following abbreviations have the following meanings unless otherwise indicated and any other abbreviations used herein and not defined have their standard, generally accepted meaning:
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0002
[0236] Unless noted otherwise, all materials, such as reagents, starting materials and solvents, were purchased from commercial suppliers, such as Sigma-Aldrich, Fluka Riedel-de Haen, and the like, and were used without further purification.
[0237] Reactions were run under nitrogen atmosphere, unless noted otherwise. The progress of reactions was monitored by thin layer chromatography (TLC), analytical high-performance liquid chromatography (anal. HPLC), and mass spectrometry, the details of which are given in specific examples.
[0238] Reactions were worked up as described specifically in each preparation; commonly, reaction mixtures were purified by extraction and other purification methods such as temperature- and solvent-dependent crystallization, and precipitation. In addition, reaction mixtures were routinely purified by preparative HPLC, typically using Microsorb C18 and Microsorb BDS column packings and conventional eluents. Progress of reactions was typically monitored by liquid chromatography mass spectrometry (LCMS). Characterization of isomers was typically done by Nuclear Overhauser effect spectroscopy (NOE). Characterization of reaction products was routinely carried out by mass spectrometry and/or 1H-NMR spectroscopy. For NMR measurement, samples were dissolved in deuterated solvent (CD3OD, CDCl3, or DMSO-d6 ), and 1H-NMR spectra were acquired with a Varian Gemini 2000 instrument (400 MHz) under standard observation conditions. Mass spectrometric identification of compounds was typically conducted using an electrospray ionization method (ESMS) with an Applied Biosystems (Foster City, CA) model API 150 EX instrument or an Agilent (Palo Alto, CA) model 1200 LC/MSD instrament.
[0239] Example 1: Synthesis of5-(3-(6-niethylpyridin-2-yl)-1H-pyrazol-4-yl)-N-(4-(piperazin-1-yl)phenyl)- 1H-benzo[d]imidazol-2-amine (580) .
Figure imgf000221_0001
Figure imgf000222_0001
[0240] Step A: Preparation of tert-butyl 2-bromo-6-iodo-1H-benzo[d]imidazole-1-carboxylate (1-2). To a solution of 1-1 (6.50 g, 20.1 mmol) and BocaO (5.30 g, 24.2 mmol) in MeCN (150 mL) was added DMAP (491 mg, 4.03 mmol) at 25 °C. The mixture was stirred at 25 °C for 3 h. The reaction was poured into ice water (1,000 mL) and was extracted with EtOAc (3 x 250 mL). The combined organics were washed with brine (3 x 100 mL), dried with Na2SO4 and concentrated under reduced pressure to give the crude product which was purified by flash chromatography (1 to 20% EtOAc/petroleum ether) to afford 1-2 (7.2 g) as a white solid.
[0241] Step B: Preparation of 2-methyl-6-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl)pyridine (1-5). To a solution of 1-3 (30.0 g, 176 mmol) in 1,4-dioxane (250 mL) and H2O (25 mL) was added 1-4 (58.6 g, 211 mmol), Pd(dppf)Cl2 (6.40 g, 8.80 mmol), and K3PO4 (55.8 g, 263 mmol). The mixtitre was degassed via vacuum/N2 backfill (3x) then the reaction was heated to 80 °C and stirred for 4 b. The crude mixture was filtered and concentrated under vacuum. The crude residue was purified by flash chromatography (5 to 25% EtOAc/petroleum ether) to afford 1-5 (40.0 g) as a yellow oil. [M+H]+ calcd for C14H17N3O 244.1, found 244.2.
[0242] Step C: Preparation of 2-(4-iodo-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl)-6-methylpyridine (1- 6). To a mixture of 1-5 (10.0 g, 41.1 mmol) and K2CO3 (17.0 g, 123.3 mmol) in CH2Cl2 (300 mL) was added a solution of ICI (32.3 g, 123.3 mmol) in CH2Cl2 (100 mL) dropwise at 0 °C. The mixture was stirred at 25 °C for 12 h. The reaction mixture was poured into Na2S2O3 (300 mL, 2.0 M) at 0 °C. The mixture was extracted with CH2Cl2 (3 x 600 mL). The organic layer was washed with brine (2 x 400 mL), dried over anhydrous Na2SO4, filtered, and concentrated in vacuum to afford the crude residue which was purified by flash chromatography (0 to 16% EtOAc/petroleum ether) to afford 1-6 (11.0 g) as a yellow solid.
[0243] Step D: Preparation of (3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4- yDboronic acid (1-8). To a solution of 1-6 (14.0 g, 29.8 mmol) and 1-7 (16.6 g, 89.4 mmol) in THF (180 ml.,) was added i-PrMgCl (44.7 ml.,, 89.4 mmol) at 0 °C. After addition, the mixture was stirred for 1 h at 0 °C then warmed to 25 °C and stirred for 15 h. The reaction was quenched with water (100 mL). The mixture was diluted with ice water (500 mL) and extracted with EtOAc (3 x 200 mL). The combined organics were washed with brine (2 x 150 mL), dried with Na2SO4 and concentrated under reduced pressure to afford 1-8 (16.0 g) as a yellow oil which was used directly without further purification. [M+H]+ calcd for C14H18BN3O3 288.1, found 288.1.
[0244] Step E: Preparation of tert-butyl 2-bromo-6-(3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)- 1H-pyrazol-4-yl)-1H-benzo[d]imidazole-1-carboxylate (1-9). This reaction was run as 4 smaller reaction batches that were combined after the reactions were complete. To a mixture of 1-8 (1.0 g x 4, 2.36 x 4 mmol) and 1-2 (1.5 g x 4, 5.22 mmol x 4) in 1,4-dioxane (20 mL x 4) and H2O (4 mL x 4) was added NaHCO3 (0.6 g x 4, 7.14 mmol x 4) and Pd(dppf)C12 (0.2 g x 4, 0.27 mmol x 4). The mixture was stirred at 60 °C for 6 h under N2. The 4 reaction mixtures were combined and concentrated under reduced pressure. The residue was purified via preparative HPLC chromatography to obtain 1-9 (1.1 g) as a white solid. [M+H]+ calcd for C26H28BrN5O3540.1, found 540.1.
[0245] Step F: Preparation of 5-(3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)-N-(4-(piperazin-l-yl)phenyl)- 1H-benzo[d]imidazol-2-amine (580). A suspension of 1-9 (36.8 mg, 0.0680 mmol), 1-10 (30.3 mg, 0.109 mmol) , Pd2(dba)3 (12.5 mg, 0.0140 mmol), Xantphos (7.90 mg, 0.0140 mmol), and sodium tert-butoxide (19.7 mg, 0.205 mmol) in 1,4-dioxane (341 μL) was degassed via vacuum/N2 backfill (5x). The suspension was heated to 95 °C for 18 h. The reaction was cooled, filtered through a pad of celite and concentrated under reduced pressure. TFA (0.500 mL) was added and the reaction was heated to 50 °C for 1 h. The reaction was concentrated under reduced pressure and the residue was purified via preparative HPLC chromatography to obtain the bis-TFA salt of the title compound (9.7 mg). [M+H]+ calcd for C26H26N8451.2, found 451.2.
[0246] Example 2: Synthesis of N-(isoindolin-5-yl)-6-(5-(6-methylpyridin-2-yl)-lH-imidazol-4- yl)benzo[d]oxazoI-2-amine (517) .
Figure imgf000223_0001
[0247] Step A: Preparation of 2-(1H-imidazol-5-yl)-6-methylpyridine (2-3). To a solution of 2-2 (3.00 g, 24.8 mmol) in EtOH (50 mL) was added p-toluenesulfonylmethyl isocyanide (2-1, 4.84 g, 24.8 mmol) at 23 °C and the reaction was stirred for 15 min. To this was added KCN (0.323 g, 4.95 mmol) and the mixture was stirred at 23 °C for 1 h. The solvent was removed under reduced pressure and the residue was partitioned between H2O (50 ml.,) and EtOAc (75 mL). The organic phase was dried over MgSO4, filtered and concentrated under reduced pressure. The crude residue was transferred to a pressure reactor (steel bomb) by dissolving in MeOH (20 ml) and a solution of NH3 (20 mL, 7M in MeOH). The reaction was sealed and was heated at 125 °C for 18 h. The reaction mixture was concentrated under reduced pressure and the residue was suspended in EtOAc (40 mL). The beige precipitate was filtered off and the filtrate was concentrated under reduced pressure. The crude residue was purified by flash chromatography (0-10% MeOH/CH2Cl2) to afford 2-3 (1.73 g). [M+H]+ calcd for C9H9N3 160.1, found 160.
[0248] Step B: Preparation of 2-methyI-6-(1-((2-(trimethylsilyl)ethoxy)methyl)-1H-imidazol-5-yl)pyridine (2-4). To a solution of 2-3 (1.00 g, 6.28 mmol) in dry THF ( 19.0 mL) was added NaH (0.302 g, 7.54 mmol) at 0 °C and the mixture was stirred for 15 min. SEM-C1 (1.69 mL, 9.43 mmol) was added dropwise at 0 °C and the mixture was allowed to warm to 23 °C and was stirred for 1 h. The reaction was diluted with H2O (30 mL) and the product was extracted with EtOAc (60 mL), dried, over MgSO4 , filtered and evaporated under reduced pressure to afford 2-4 (1.81 g) which was used without purification. [M+H]+ calcd for C15H23N3OSi 290.2, found 290.
[0249] Step C: Preparation of 2-(4-bromo-1-((2-(trimethyIsilyl)ethoxy)methyI)-1H-imidazol-5 -yI)-6- methylpyridine (2-5). A solution of NBS (2.51 g, 14.1 mmol) in DMF (15.0 mL) was added dropwise at 0 °C over 30 min to a solution of 2-4 (4.54, 15.7 mmol) in DMF (32.0 mL). The reaction mixture was stirred at 23 °C for an additional 45 min, then was quenched with sat. NaHSO3 (30 mL), extracted with EtOAc (75 ml.,) and the organic phase was washed with water (50 ml,), dried over MgSO4, filtered and concentrated under reduced pressure. The crude residue was purified by flash chromatography (0-60% EtOAc/heptane) to afford 2-5 (2.94 g). [M+H]+ calcd for C15H22BrN3OSi 368.1, found 368.
[0250] Step D: Preparation of 6-(5-(6-methylpyridin-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyi)-1H- imidazol-4-yl)benzo[d]oxazoI-2-amine (2-7). 2-6 (3.11 g, 11.9 mmol), Pd(dppf)Ch (0.795 g, 1.09 mmol) and K2CO3 (4.50 g, 32.6) mmol) were added to a solution of 2-5 (4.00 g, 10.9 mmol) in dioxane/H2O (8:1 , 36.0 mL) under nitrogen and the mixture was stirred, at 100 °C for 18 h. The solvent was removed, under reduced pressure and the residue was diluted with H2O (30 ml.,) and the product was extracted with EtOAc (3 x 25 ml.,), dried over MgSO4, filtered and evaporated to dryness. The crude residue was purified by flash chromatography (0-10% MeOH/CH2Cl2) to afford 2-7 (1.30 g). [M+H]+ calcd for C22H23 N5O2Si 422 .2, found 422.1.
[0251] Step E: Preparation of N-(isoindolin-5-yl)-6-(5-(6-methylpyridin-2-yl)-1H-imidazol-4- yl)benzo[d]oxazoi-2-amine (517). To a vial containing 2-7 (21.0 mg g, 0.0500 mmol), CS2CO3 (41.0 mg, 0.125 mmol), and 2-8 (18.0 mg, 0.0600 mmol) was added a solution of BrettPhos (1.34 mg, 2.500 μmol) and BrettPhos Pd G4 (2.30 mg, 2.500 μmol) in 1,4-dioxane (0.250 mL) and the resulting mixture was degassed with N2 via vacuumdSL backfill (5x). The reaction was heated to 110 °C for 16 h. LCMS at this time indicated incomplete conversion of the starting material. An additional portion of BrettPhos Pd G4 (2.301 mg, 2.500 μmol) and BrettPhos (1.342 mg, 2.500 μmol) in 1,4-dioxane (0.15 mL) was added and the reaction was degassed via vacuum/N2 backfill (5x) and heated to 110 °C for 24 h. The reaction was concentrated under reduced pressure and the residue was dissolved in TFA (0.400 mL) and then heated to 55 °C for 2 h. The solvent was removed under reduced pressure and the residue was purified via preparative HPLC chromatography to obtain the bis-TFA salt of the title compound (3.5 mg). [M+H]+ calcd for C24H20N6O 409.2, found 409.1.
[0252] Example 3: Synthesis of 5-(5-(5-chloro-2-fluorophenyl)-1H-imidazol-4-yl)-N-( 1, 2,3,4- tetrahydroisoquinolin-7-yl)benzo[d]oxazol-2-amine (259).
Figure imgf000225_0001
[0253] Step A: Preparation of 5-(5-chloro-2-fluorophenyl)-1H-imidazole (3-2). The reaction was completed in 2 batches of 5 g each. 3-1 (5.00 g, 31.5 mmol) and p-toluenesulfonylmethyl isocyanide (2-1, 6.15 g, 31.5 mmol) were dissolved in EtOH (50 mL). NaCN (154 mg, 3.15 mmol) was added at 23 °C and the reaction was stirred for 3 h. The solvent was removed under reduced pressure and the crude residue was diluted with H2O (50 mL) and CH2Cl2 (100 mL). The organic phase was separated, washed with brine (50 mL), dried over MgSO4 and concentrated under reduced pressure to afford a brown oil (19.5 g crude). This residue was dissolved in MeOH (30 mL ), was charged into a steel bomb, and methanolic NH3 (70 ml.,) was added while cooling at 0 °C. The closed vessel was heated at 125 °C for 16 h. The solvent was removed under reduced pressure and the crude residue was purified via flash chromatography (0-5% MeOH/CH2Cl2) to afford 3-2 (6.0 g) as a brown solid. [M+H]+ calcd for C9H6CIFN2 197.0, found 197.0.
[0254] Step B: Preparation of 5-(5-chloro-2-fluorophenyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H- imidazole (3-3). To a solution of 3-2 (5.50 g, 28.1 mmol) in THF (60 ml.,) at 0 °C was added NaH (1.68 g, 42.1 mmol) and the mixture was stirred at 0 °C for 30 min. SEMC1 (6.20 g, 33.7 mmol) was added dropwise and the mixture was stirred for another 30 min at 23 °C. The reaction mixture was quenched with sat. aq. NH4CI (50 mL) and extracted with EtOAc (2 x 50 mL). The combined organics were dried over Na2SO2 and concentrated under reduced pressure. The crude residue was purified via flash chromatography (0-10% EtOAc/hexanes) to afford 3-3 (4.2 g) as a brown oil. [M+Na]+ calcd for C15H20ClFN2OSi 351.1, found 351.2.
[0255] Step C: Preparation of 4-bromo-5-(5-chloro-2-fluorophenyl)-l-((2-(trimethylsilyl)ethoxy)methyl) - 1H-imidazole (3-4). To a solution of 3-3 (4.19 g, 12.9 mmol) in DMF (50 mL) stirring at 23 °C was added NBS (2.51 g, 14.1 mmol) and the mixture was stirred for 1 h. The reaction mixture was diluted with H2O (100 mL) and extracted with EtOAc (3 x 50 mL). The combined organics were washed with brine (2 x 20 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure. The crude residue was purified via flash chromatography (10-20% EtOAc/hexanes) to afford 3-4 (2.7 g) as a brown oil.
[0256] Step D: Preparation of 5-(5-(5-chioro-2-fluorophenyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H- imidazol-4-yl)benzo[d]oxazol-2-amine (3-6). This reaction was performed in 2 batches of 400 mg each. 3-4 (400 mg, 0.990 mmol) and 3-5 (308 mg, 1.18 mmol) were charged in dioxane (8.00 mL). K3PO4 ( 419 mg, 1.98 mmol) was added followed by water (2.0 mL) and the resulting suspension was purged with N2 for 5 min. To this was added CatacXium A PdG3 (73.0 mg, 0.10 mmol) and purging continued with N2 for 1 min. The reaction was sealed and heated at 100 °C for 8 h. The reaction was cooled to 23 °C and filtered through Celite, washing with EtOAc (20 mL). The filtrate was dried over Na2SO4 and concentrated under reduced pressure. The crude residue was purified via flash chromatography (50-70% EtOAc/hexanes) to afford 3-6 (482 mg). [M+H]+ calcd for C22H24ClFN4O2Si 459.1, found 459.0.
[0257] Step E: Preparation of 5-(5-(5-chloro-2-fluorophenyl)-1H-imidazol-4-yl)-N-(1, 2,3,4- tetrahydroisoquinolin-7-yl)benzo[d]oxazol-2-amine (259). A mixture of 3-6 (23.0 mg, 0.0500 mmol), Cs2CO3 (41.0 mg, 0.125 mmol), and 3-7 (19.0 mg, 0.0600 mmol) was suspended in a solution of BrettPhos (2.68 mg, 5.00 μmol) and BrettPhos Pd G4 (4.60 mg, 5.00 μmol) in 1,4-dioxane (0.250 mL). The resulting mixture was degassed with N2 via vacuum/NL backfill (5x). The reaction was sealed and heated to 110 °C for 16 h. The solvent was removed under reduced pressure and the crude residue was dissolved in TEA (0.400 mL, 5.19 mmol) and the mixture was heated to 60 °C for 1 h. The solvent was removed under reduced pressure and the crude residue was purified by via preparative HPLC chromatography to obtain the TEA salt of the title compound (4.0 mg). [M+H]+ calcd for C25H19CIFN5O 460.1, found 460.1.
[0258] Example 4: Synthesis of N-(6-(5-(6-methylpyridin-2-yl)-1H-imidazol-4-yl)benzo[d]thiazol-2-yl)-2- (piperazin- 1 -yl)acetamide (636).
Figure imgf000227_0001
[0259] Step A: Preparation of 2-methyl-6-(l-((2-(trimethylsilyl)ethoxy)methyl)-1H-imidazol-4-yl)pyridine (4-2). To a solution of 4-1 (4.484 g, 28.168 mmol) in dry THF (47 mL) was added sodium hydride (1.352 g, 33.802 mmol) at 0 °C and the resulting mixture stirred for 15 min. To this, (2-
(chloromethoxy )ethyl)trimethylsilane (7.478 mL, 42.252 mmol) was added dropwise at 0 °C and. the mixture was allowed to stir at rt for 1 h. Water was added and the product was extracted with EtOAc, dried over MgSO4 filtered and evaporated to dryness. Then, the crude material was purified by column chromatography (silica; DCM/MeOH (9:1) in DCM 0/100 to 20/80), yielding the title compound (4.877 g). [M+H]+ calcd for C15H23N3OSi 289.16, found 290.
[0260] Step B: Preparation of 2-(5-bromo-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-imidazol-4-yl)-6- methylpyridine (4-3). A solution of NBS (3.985 g, 22.387 mmol) in 23 mL of DMF was added dropwise at 0 °C for 30 min to a solution of 4-2 (7.2. g, 24.871 mmol) in 51 mL of DMF and the reaction mixture was stirred at rt for 45 min. The reaction was quenched with NaHSOs (sat), extracted with EtOAc and the organic layer was washed with water, dried over MgSQs, filtered and evaporated to dryness. The crude material was purified by column chromatography (silica; ethyl acetate in heptane 0/100 to 40/60), yielding the title compound (3.856 g). [M+HJ- calcd for C15H22BrN3OSi 367.07, found 368.
[0261] Step C: Preparation of 6-(4-(6-methylpyridin-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-imidazol- 5-yl)benzo[d]thiazol-2-amine (4-5). 4-4 (3.299 g, 11.945 mmol), Pd(dppf)Cl2 (0.795 g, 1.086 mmol) and K2CO3 (4.502 g, 32.577 mmol) were added to a solution of 4-3 (4 g, 10.859 mmol) in 36 mL of dioxane/ H2O (8:1) under nitrogen and the mixture was stirred at 100 °C overnight. Then, Pd(dppf)Cl2 (0.795 g, 1.086) mmol) was added and the mixture was stirred, at 100 °C overnight. Then, additional Pd(dppf)Cl2 (0.795 g, 1.086 mmol) was added and the mixture was stirred at 100 ®C overnight. Solvent was evaporated, water was added and the product was extracted with EtOAc, dried over MgSO4 filtered and evaporated to dryness. The crude material was purified by column chromatography (silica; ethyl acetate in heptane 0/100 to 100/0) and (silica; DCM/MeOH (9:1) in DCM 0/100 to 40/60), yielding the title compound (0.589 g). [M+H]+ calcd for C22H27N5OSSi 437.17, found 438.
[0262] Step D: Preparation of 2-chloro-N-(6-(4-(6-methylpyridin-2-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)- 1H-imidazol-5-yl)benzo[d]thiazol-2-yl)acetamide (4-6). To a solution of 4-5 (60 mg, 0.137 mmol) in DIPEA (96 μL, 0.548 mmol) and DCM (914 μL) at 0 °C was added chloroacetyl chloride (15.29 μL, 0.192 mmol). The reaction was stirred and allowed to warm to rt overnight. The reaction mixture was concentrated under vacuum and purified by silica column chromatography (0-100% EtOAc in hexanes), yielding the title compound (24 mg). [M+Hp calcd for C24H28ClN5OrSSi 513.14, found 514.
[0263] Step E: Preparation of N-(6-(5-(6-methylpyridin-2-yl)-1H-imidazol-4-yl)benzo[d]thiazol-2-yl)-2- (piperazin- 1-yl)acetamide (636). To a solution of 4-6 (2.4 mg, 0.047 mmol) in acetonitrile (467 μL) was added tert-butyl piperazine- 1- carboxylate (17.39 mg, 0.093 mmol) and DIPEA (32.6 pL, 0.187 mmol). The reaction was stirred at 100 °C overnight. The reaction mixture was then concentrated under vacuum. The resulting residue was treated with TEA (500 μL) and stirred at 50 °C for 1 h. The reaction mixture was then concentrated under vacuum. The residue was purified by preparative HPI..C chromatography using a gradient (10 to 25%) of acetonitrile in water with 0.05% TEA to obtain the title compound as a white solid (6 mg). [M+H]+ calcd for C22H23N7OS 433.17, found 434.1.
[0264] Example 5: Synthesis of 1-[6-[5-(6-methyl-2-pyridyl)-1H-pyrazol-4-yl]-[1,2,4]triazolo[1,5-a]pyridin- 2-yl] -3-(3-pyridyl)urea (52).
Figure imgf000228_0001
[0265] Step A: Preparation of 2-methyl-6-(1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-5-yl)pyridine (5-3). To a solution of 5-1 (10.0 g, 58.5 mmol) in dioxane (150 mL) and H2O (15 mL) was added 5-2 (19.5 g, 70.2 mmol), Pd(dppf)Cl2 (2.1 g, 2.92 mmol), and K3PO4 (18.6 g, 87.7 mmol). The reaction was then stirred at 80 °C for 4 h under N2 atmosphere. The mixture was filtered thru a pad of celite and concentrated in vacuum. The residue was purified by column chromatography (0-30% of EA in PE) to afford 5-3 (12 g) as yellow oil. [0266] Step B: Preparation of 2-(4-iodo4-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-5-yl)-6-methylpyridine (5- 4). To a mixture of 5-3 (6.0 g, 24.7 mmol) and K2CO3 (10.2 g, 74.0 mmol) in DCM (130 mL) was added a solution of IC1 (19.4 g, 74.0 mmol) in DCM (50 mL) dropwise at -50 °C. The mixture was warmed and stirred at 10 °C for 16 h. The reaction mixture was diluted with sat. Na2SO3 (200 mL) and extracted with DCM (200 mL x 3). The organic layer was washed with brine (50 mL x 2), dried over .bmjSCfo filtered and concentrated in vacuum to give a residue, which was purified by column chromatography to afford 5-4 (7.5 g) as yellow oil. [M+H]+ calcd for C14H16IN3O 370.03, found 369.9.
[0267] Step C: Preparation of 2-methyl-6-(1-(tetrahydro-2H-pyran-2-yl)-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1H-pyrazol-5 -yl)pyridine (5-6). To a solution of 5-4 (7.5 g, 20.31 mmol) and 5-5 (11.3 g, 60.94 mmol) in THE (150 mL) was added i-PrMgCl (30.5 mL, 60.94 mmol) at -5 to 0 °C. After addition, the mixture was stirred at 15 °C for 16 h. The mixture was quenched with sat. NH4CI aq (200 mL) and extracted with EA (150 mL x 3). The organic layer was concentrated in vacuum to obtain crude 5-6 (7.0 g) as a yellow oil.
[0268] Step D: Preparation of 6-(5-(6-methylpyridin-2-yl)- 1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)- [1,2,4]triazolo[1,5-a]pyridin-2-amine (5-8). To a mixture of 5-6 (4.0 g, 18.77 mmol) and 5-7 (7.0 g, 17.06 mmol) in dioxane (100 mL) and H2O (20 mL) was added Pd(dppf)Cl2. (850 mg, 1.16 mmol) and K2CO3 (5.2 g, 37.54 mmol). The reaction mixture was stirred at 100 °C for 2 h under N2. The reaction mixture was concentrated in vacuum and purified, by column chromatography (30%-100% of EA in PE: 0-10%' of MeOH in EA) to afford 5-8 (4.0 g) as a yellow solid. [M+H]+ calcd for C20H21N7O 376.18, found 376.3.
[0269] Step E: Preparation of 1-[6-[5-(6-methyl-2-pyridyl)-1H-pyrazol-4-yl]-[1 ,2,4]triazolo[1,5-a]pyridin-2- yl]-3-(3-pyridyl)urea (52). To a mixture of 5-8 (20 mg, 0.053 mmol) and DIPEA (2.7.9 pL, 0.160 mmol) in DME (266 μL) was added 4-8 (12.80 mg, 0.017 mmol). The reaction mixture was allowed to stir for 1 h at 70 °C. The reaction mixture was concentrated under vacuum. TFA (500 μL) was added and the reaction was heated to 50 °C for 1 h. TFA was removed in vacuum and the residue was purified by preparative HPLC chromatography using a gradient (2 to 40%) of acetonitrile in water with 0.05% tri fluoroacetic acid to yield a TFA salt of the title compound (4.3 mg). [M+H]* calcd for C21H17N9O 412.16, found 412.1. [0270] Example 6: Synthesis of [4-[5-[3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl]-2H-indazol-7- y]]thiophen-2-yl]methanamine (574).
Figure imgf000230_0001
[0271] Step A: Preparation of 7 -bromo-5 -iodo-lH-indazole (6-2). To a stirred solution of 6-1 (5.0 g, 23.57 mmol) in DCM:H2O (100 mL:100 mL) was added NaNO2 (8.12 g, 117.9 mmol) and CH2I2 (3.80 mL, 47.14 mmol), and the resulting mixture stirred for 5 min. To this was added AcOH (26.9 mL, 471.4 mmol) and the resulting reaction mixture stirred at 25 °C for 1 d. A saturated NaHCO3 solution was added to the mixture and the pH was adjusted to 8 -9 followed by extraction with DCM. The organic layer was separated and concentrated to get the crude compound which was purified by silica gel flash column chromatography (25% EtOAc in hexane) to yield 6-2 (3.0 g) as an off-white solid. [M+H]+ caicd for C7H4BrIN2322.86, found 322.7. [0272] Step B: Preparation of 7-bromo-5-iodo-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazole (6-3). In two batches: to a stirred solution of 6-2 (500 mg, 1.55 mmol) in THF (10 mL) at 0 °C was added NaH (124 mg, 3.10 mmol). The reaction mixture was allowed to stir for 30 min and. to this SEM-Cl (0.42 mL, 2.32 mmol) was added. The reaction mixture was allowed to stir at 25 °C for 2 h. Ice cold water was added and the product extracted with DCM. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated to get the crude compound which was purified by silica gel column chromatography (10% EtOAc in hexane) to yield 6-3 (850 mg) as an yellow semi-solid. [M+H]+ caicd for C13H18BrIN2OSi 452.94, found 453.0.
[0273] Step C: Preparation of 7-bromo-5-(3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-indazole (6-4). To a solution of 6-3 (600 mg, 1.32 mmol) in DMA (6.0 mL) was added 5-6 (537 mg, 1.45 mmol) folio wed by the addition of K3PO4 (560 mg, 2.64 mmol) and water (1.0 mL). The reaction mixture was purged with N2 for 5 min followed by the addition of Pd(PPh3)4 (152 mg, 0.13 mmol) and again purged with N2 for 5 min. The reaction mixture was stirred at 500 °C for 2 h. The reaction mixture was diluted with ice-cold water and extracted using EtOAc. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated to get the crude compound which was purified further by silica gei flash column chromatography (25% EtOAc in hexane) to yield 6-4 (330 mg) as a light yellow semi-solid. [M+H]+ calcd for C27H34BrN5O2Si 568.17, found 568.2.
[0274] Step D: Preparation of [4- [5--[3 -(6-methylpyridin-2-yl)- 1H-pyrazol -4-yl]-2H-indazol-7 - yl] thiophen -2- yl]methanamine (574). A mixture of 6-4 (20 mg, 0.035 mmol), 6-5 (17 mg, 0.049 mmol), Pd(dppf)Cl2 (5.15 mg, 7.03 μmol), and Na2CO3 (14.91 mg, 0.141 mmol) in 1,4-dioxane: water (0.500 mL:0.125 mL) was sparged with N2 for 10 min before heating to 100 °C for 16 h. The reaction mixture was filtered through a pad of celite and concentrated under vacuum. TFA (500 μL) was added and the reaction was heated to 60 °C for 1 h. TFA was removed in vacuum and the residue was purified by preparative HPLC chromatography using a gradient (11 to 23%) of acetonitrile in water with 0.05% trifluoroacetic acid to yield a TFA salt of the title compound (6.3 mg). [M+ H]+ calcd for C21H18N6S 387.13, found 387.3.
[0275] Example 7: Synthesis of N-[5-(2,5-dihydro- lH-pyrrol-3-yl)pyridin- 2-yI]-5--[3-(6--methylpyridin-2-- yl)-1H-pyrazol-4-yl]-1H-indazol-3-amine (348).
Figure imgf000231_0001
[0276] Step A: Preparation of tert-butyl 3 -(6- aminopyridin- 3-yl)-2,5-dihydro- 1H -pyrrole- 1 -carboxylate- - methane (7-3). A mixture of 7-1 (0.097 g, 0.330 mmol), Pd(dppf)Cl2 (0.044 g, 0.060 mmol), Na2CO3 (0.095 g, 0.900 mmol) and 7-2 (104 mg, 0.3 mmol) in 1,4-dioxane (1.0 mL): water (0.250 mL) was sparged with N2 for 10 min. before heating to 100 °C for 16 h. The mixture was filtered thru a pad of celite and concentrated in vacuum. Tire residue was purified by column chromatography (0 to 100% EtOAc in hexane) to afford 7-3 (63 mg) as a beige solid. [M+ H]+ calcd for C15H23N3O2278.18, found 278.0.
[0277] Step B: Preparation of 5-(5-(6-methylpyridin-2-yI)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)- IH-indazole (7-5). To a stirred solution of 5-4 (4.00 g, 10.8 mmol) in 1,4-dioxane (40.0 L) was added 7-4 (4.48 g, 13.0 mmol) and the reaction was purged with argon for 15 minutes followed by addition of K3PO4 (4.60 g, 2 eq., 21.7 mmol) in water (8.00 mL) and further purged with argon for 5 min. Pd(dppf)Cl2-DCM (884 mg, 1.08 mmol) subsequently was added. The resulting mixture was stirred at 110 °C for 16 h. The reaction mixture cooled to 25 °C. To this was added. K2CO3 (2.99 g, 21.7 mmol) and MeOH (20.0 mL). The reaction mixture was further heated al 110 °C for 2 h. The reaction mixture was filtered through a celite pad. The filtrate was evaporated under vacuum to get the crude compound. Purification using silica gel chromatography (50% EtOAc in hexane) afforded 7-5 (2.40 g) as an off-white solid. [M+H]+ calcd for C21H21N5O 360.17, found 360.2.
[0278] Step C: Preparation of 3-bromo-5-(5-(6-methylpyridm-2-yl)-l-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)-1H-indazole (7-6). To a stirred solution of 7-5 (2.40 g, 6.68 mmol) in DMF (20.0 mL) was added NBS (1.19 g, 6.68 mmol) portionwise at 0 °C. The resulting mixture was allowed to stir at 25 °C for 30 min. The reaction mixture was diluted with ice cold water (100 mL). The solid precipitate was filtered and washed, with water (3 times) followed by a pentane wash. The resulting light yellow solid (7-6, 2.30 g) was used in the next step without further purification.
[0279] Step D: Preparation of 3-bromo-5-(5-(6-methylpyridin-2-yl)-l-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)-1-(tetrahydro-2H--pyran-2-yl)-1H-indazole (7-7). To a solution of 7-6 (2.30 g, 5.25 mmol) in DCM (40.0 mL) was added DHP (1.44 mL, 15.7 mmol) followed, by addition of pTSA-H2O (200 mg, 1.05 mmol) at 0 °C. The reaction mixture was stirred at 25 °C for 16 h. The reaction mixture was partitioned between water and DCM. The organic layer was separated, washed with brine, dried over sodium sulfate, and concentrated in vacuum.. Purification using silica gel column chromatography (25% EtOAc in hexane) afforded 7-7 (1.40 g) as an off -white solid. [M+H]+ calcd for C26H28BrN5O2522.14, found 522.4.
[0280] Step E: Preparation of N-[5-(2,5-dihydro-1H-pyrrol-3-yl)pyridin-2-yl]-5-[3-(6-methylpyridin-2-yl)- 1H-pyrazol-4-yl]-1H-indazol-3-amine (348). A mixture of 7-7 (40 mg, 0.077 mmol), 7-3 (30.0 mg, 0.115 mmol), BrettP.hos Pd G4 (7.05 mg, 7.66 μmol), cesium carbonate (100 mg, 0.306 mmol), and BrettPhos (4.11 mg, 7.66 μmol) in 1,4-dioxane (383 μL) was sparged with N2 for 5 min and allowed to stir for 16 h at 85 °C. The reaction mixture was filtered thru a pad of celite and concentrated under vacuum. TFA (500 μL) was added and the reaction was heated to 50 °C for 1 h. TFA was removed in vacuum and the residue was purified by preparative HPLC chromatography using a gradient (8 to 21%) of acetonitrile in water with 0.05% tri fluoroacetic acid to yield a TFA salt of the title compound (8.0 mg). [M+H]+ calcd for C25H22N8435.20, found 435.1. [0281] Example 8: Synthesis of l-[4-(2,5-dihydro-1H-pyrrol-3-yl)phenyl]-5-[3-(6-methylpyridin-2-yl)-1H- pyrazol-4-yl]indazole (594).
Figure imgf000233_0001
[0282] Step A: Preparation of 5-(3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)- 1H-indazole (8-2). To a stirred solution of 8-1 (1.0 g, 5.07 mmol) in 1,4-dioxane (25 mL) and H2O (5 mL) was added 5-6 (2.2.4 g, 6.09 mmol) and K3PO4 (2.15 g, 10.14 mmol). The reaction was purged with argon for 5 minutes followed by addition of PdCl2(dppf)DCM (414 mg, 0.507 mmol). The reaction was allowed, to stir at 110 °C for 12 h. The reaction mixture was cooled to 25 °C, diluted with water, and extracted with EtOAc.
The combined organic layer was dried over Na2SO4 and concentrated under vacuum to get the crude compound. The crude compound was purified using silica gel column chromatography (40 % EtOAc in hexane) to yield 8-2 (620 mg) as a light yellow solid. [M+H]+ calcd for C21H21N5O 360.17, found 360.2. [0283] Step B: Preparation of 1-(4-bromophenyl)-5-(3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl) 1H-pyrazol-4-yl)-1H-indazole (8-5). To a stirred solution of 8-2 (400 mg, 1.11 mmol) in 1,4-dioxane (8 mL) were added 8-3 (314 mg, 1.33 mmol) and K3PO4 (470 mg, 2.22 mmol) in a vial. The reaction mixture was degassed with argon for 5 minutes, then 8-4 (63 mg, 0.44 mmol) and CuI (21 mg, 0.11 mmol) added. The reaction mixture was stirred and heated at 100 °C for 16 h. The reaction mixture was filtered through a cehte bed and concentrated under vacuum. Purification of the. crude material was done using silica gel column chromatography (25% EtOAc in hexane) to obtain 8-5 (160 mg) as a light yellow product. [M+H]+ calcd for C27H24BrN5O 514.12, found 360.2.
[0284] Step C: Preparation of 1- [4-(2,5-dihydro-1H-pyrrol-3-yl)phenyl]-5-[3 -(6-methylpyridin-2-yl)- 1H- pyrazol-4-yl]indazole. (594). A mixture, of 8-5 (20 mg, 0.039 mmol), 7-1 (13.77 mg, 0.047 mmol), sodium carbonate (16.48 mg, 0.156 mmol), and PdCl2(dppf) (5.69 mg, 7.78 μmol) in 1 ,4-dioxane (311 μL):water (78 μL) was heated to 100 °C for 16 h. The reaction mixture was concentrated under vacuum. TFA (500 μL) was added, and the reaction was heated to 50 °C for 1 h. TFA was removed in vacuum and the residue was purified by preparative HPbC chromatography using a gradient (15 to 35%) of acetonitrile in water with 0.05% trifluoroacetic acid to yield a TFA salt of the title compound (11.5 mg). [M+H]+ calcd for C26H22N6419.19, found 419.2.
[0285] Example 9: Synthesis of [4- [6-[3--(6-methylpyridin-2-yl)- 1H-pyrazol-4-yl]- 1H-indazol-4- yl]phenyl] methanamine (107).
Figure imgf000234_0001
[0286] Step A: Preparation of 7-bromo-5-iodo-1-(tetrahydro-2H-pyran-2-yI)-1H-indazoIe (9-2). To a solution of 9-1 (5.0 g, 15.5 mmol) in THF (50.0 mb) was added DHP (1.97 mL, 23.3 mmol) followed by pTSA- H2O) (295 mg, 1.55 mmol). The reaction mixture was stirred at 55 °C for 6 h. The reaction mixture was diluted with ammonium chloride solution (20 mb) and DCM (40 mb). The organic layer was washed with brine, dried over sodium, sulfate, and concentrated in vacuum. The crude material was triturated with Et2O and pentane to obtain 9-2 (5.0 g) as an off-white solid. [M+H]+ calcd for C12H12BrIN 2O 405.92, found. 406.0. [0287] Step B: Preparation of 4-bromo-6-(3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole (9-3). To a stirred solution of 9-2 (2.0 g, .91 mmol) in DMA (24.0 mL) and water (6.00 mL) was added 5-6 (1.81 g, 4.91 mmol) and K3PO4 (2.09 g, 9.83 mmol). The reaction was purged with argon for 5 min followed, by addition of Tetrkis (568 mg, 491 μmol). Argon purging was continued for an additional 2 min. The reaction was allowed to stir at 55 °C for 2 h. The reaction mixture was diluted with cold water (50 mL) and EtOAc (50 mL). The organic layer was separated, washed with brine, dried over anhydrous sodium sulfate and concentrated under vacuum. The reaction was purified by column chromatography (25% EtOAc in hexane). Trituration was done using Et2O/pentane to afford, pure 9-3 (900 mg, light yellow). [M+H]+ calcd for C26H28BrN5O2522.14, found 522.4.
[11288] Step C: Preparation of [4-[6-[3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl]-1H-indazol-4- yl]phenyl]methanamine (107). A mixture of 9-3 (34 mg, 0.065 mmol), 9-4 (21.3 mg, 0.085 mmol), Pd(dppf)Cl2 (9.51 mg, 0.013 mmol), and. Na2CO3 (21 mg, 0.195 mmol) in l,4-dioxane:water (0.500 mb:0.125 mb) was sparged with N2 for 10 min before heating to 100 °C for 16 h. The reaction mixture was filtered. through a pad of celite and concentrated under vacuum. TFA (500 μL) was added and the reaction was heated to 50 °C for 1 h. TFA was removed in vacuum and the residue was purified by preparative HPLC chromatography using a gradient (5 to 40%') of acetonitrile in water with 0.05%' trill uoroacetic acid to yield a TFA salt of the title compound (23.6 mg). [M+H]+ calcd for C23H20N6381.17, found 381.1.
[0289] Example 11): Synthesis of [4-[6-[3-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl]-1H-indazol-3- yljphenyljmethanamine (5).
Figure imgf000235_0001
[0290] Step A: Preparation of 6-(3-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)- 1H-indazoIe (10-2). To a stirred solution of 10-1 (500 mg, 2.53 mmol) in 1,4-dioxane (8.0 mL) and H2O (2.0 mL) was added 5-6 (1.4 g, 3.80 mmol) and K3PO4 (1.07 g, 5.06 mmol). The reaction mixture was purged with N2 for 15 minutes followed by addition of Pd(dppf)Cl2 (412 mg, 0.50 mmol). The reaction was allowed to stir at 100 °C for 6 h. The reaction mixture was filtered through a celite bed and then diluted with water and extracted with EtOAc. The organics were concentrated under vacuum to get the crude residue which was purified by silica gel column chromatography (20 to 30% EtOAc in hexane) to afford 10-2 (400 mg) as an off-white solid. [M+H] * calcd for C21H21N5O 360.17, found 360.2.
[0291] Step B: Preparation of 3-bromo-6-(3-(6-methylpyridin-2-yl)-l.-(tetrahydro-2H-pyran-2-yi)-1H- pyrazol-4-yl)-1H- indazole (10-3). To compound 10-2 (550 mg, 1.53 mmol) in DMF (10 ml.,) was added NBS (273 mg, 1.53 mmol) at 25 °C and the resulting mixture stirred, for 1 h. The reaction mixture was diluted with water and extracted using EtOAc (3x). The combined organic layer was washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under vacuum to get the crude residue which was purified by silica gel column chromatography (10 to 20% EtOAc in hexane) to afford 10-3 (510 mg) as an off-white solid. [M+H]+ calcd for C21H20BrN5O 438.09, found. 438.1.
[0292] Step C: Preparation of [4-[6-[3--(6-methylpyridin-2-yl)- 1H-pyrazol-4-yl]-1H-indazol-3- yl]phenyl]methanamine (5). A mixture of 10-3 (20 mg, 0.046 mmol), Pd(dppf)Cb (6.68 mg, 9.13 μmol), Na2CO3 (19.34 mg, 0.183 mmol), and 9-4 (1.6 mg, 0.064 mmol) in 1 ,4-dioxane:water (0.4 ml,:0.100 mL) was sparged for 5 min with N2 before heating to 100 °C for 16 h. The reaction mixture was concentrated under vacuum. TFA (500 μL) was added and the reaction was heated to 50 °C for 1 h. TFA was removed in vacuum and the residue was purified by preparative HPLC chromatography using a gradient (5 to 40%) of acetonitrile in water with 0.05% trifluoroacetic acid to yield a TFA salt of the title compound (11.1 mg). [M+H]+ calcd for C23H20N6 381.17, found 381.1. [0293] Example 11: Synthesis of 6-[5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl]-l-[4-(l, 2,3,6- tetrahydropyridin-4-yl)phenyl]benzotriazole (200).
Figure imgf000236_0001
[0294] Step A: Preparation of tert -butyl 2-(4-(3-fluoro-4-nitrophenyl)-l-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-5-yi)-6-methylpyridine (11-2). A mixture of 5-6 (0.751 g, 2.61 mmol), Pd(dppf)Ch (0.166 g, 0.227 mmol), K2CO3 (0.942 g, 6.82 mmol) and 11-1 (0.500 g, 2.27 mmol) in 8:1 1 ,4-dioxane/water (6.82 mL) was sparged with N? for 10 min. before heating to 100 °C for 16 h. The mixture was filtered thru a pad of celite and concentrated in vacuum. The residue was purified by column (0 to 100% EtOAc in heptane) to afford 11- 2 (570 mg). [M+H]+ calcd for C20H19FN4O3 383.14, found 383.1.
[0295] Step B: Preparation of N-(4-bromophenyl)-5-(5-(6-methylpyridin-2-yl)-l-(tetrahydro-2H-pyran-2-yl)- 1H-pyrazol-4-yi)-2-nitroaniline (11-4). A mixture of 11-2 (120 mg, 0.314 mmol), 11-3 (60 mg, 0.349 mmol), K2CO3 (73 mg, 0.523 mmol), and KF (20 mg, 0.349 mmol) with 5 drops of DMF was heated at 180 °C for 16 h. The reaction mixture was extracted with EtOAc, washed with H2O, brine, dried over MgSO4, filtered and concentrated under reduced pressure to give a crude residue. Purification using silica gel chromatography (0 to 100% EtOAc in heptane) afforded 11-4 (60 mg). [M+H]+ calcd for C26MrN5O3534.11, found 534.2.
[0296] Step C: Preparation of N1 -(4 -bromophenyl)- 5-(5-(6-methyIpyridin-2-yl)- 1 -(tetrahydro-2H-pyran--2- yl)-1H-pyrazol-4-yl)benzene- 1,2 -diamine (11-5). To a mixture of 11-4 (58 mg, 0.109 mmol) in acetone/EbO (6:1, 2.56 mL) was added NH4CI (233 mg, 4.36 mmol) and Zn (143 mg, 2.18 mmol). The mixture was stirred at rt overnight. The mixture was concentrated, diluted with IM NaOH and EtOAc, and filtered through celite. The residue was purified by column chromatography (7:3 heptane:EtOAc) to afford 11-5 (28 mg).
[0297] Step D: Preparation of 1 -(4-bromophenyl) -6 (5 -(6-methylpyridin-2-yl)- 1 - (tetrahydro -2H- pyran -2 -yl) - 1H-pyrazol-4-yl)-1H-benzo[d][1,2,3]triazole (11-6). Sodium nitrite (0.055 g, 0.790 mmol) was added to a solution of 11-5 (0.040 g, 0.079 mmol) in DCM (2 mL). The mixture was cooled to 0 °C and acetic acid (0.045 mL, 0.790 mmol) was added. The reaction mixture was stirred at 25 °C for 16 h. Water was added and the product was extracted with DCM, dried over MgSO4, filtered and evaporated to dryness. The crude material was purified by column chromatography (0 to 50% EtOAC in heptane) to afford 11-6 (30 mg). [M+2H]+ calcd for C26H23BrN6O 516.11, found 516.1.
[0298] Step E: Preparation of 6-[5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl]-1-[4-(1,2,3,6-tetrahydropyridin- 4-yl)phenyl]benzotriazole (200). A mixture of 11-6 (15 mg, 0.029 mmol), Pd(dppf)Cl2 (3.55 mg, 4.85 μmol), Na2CO3 (10.28 mg, 0.097 mmol) and 11-7 (9.0 mg, 0.029 mmol) in 4:1 1 ,4-dioxane/water (0.194 mL:0.048 mL) was sparged with N2 for 10 min. before heating to 100 °C for If) h. The mixture was filtered thru a pad of celite and concentrated in vacuum. TFA (500 μL) was added and the reaction was heated to 50 °C for 1 h. TFA was removed in vacuum and the residue was purified by preparative HPLC chromatography using a gradient (16 to 28%) of acetonitrile in water with 0.05% trifluoroacetic acid to yield a TFA salt of the title compound (4.4 mg). [M+H]+ calcd for C26H21N7O 434.20, found 434.3.
[0299] Example 12: Synthesis of 5-[3-(6-methyl-2-pyridyl)-1H-pyrazol-4-yl]-N-(5-piperazin-1-yl-2- pyridyi)-1 ,2-benzothiazoi-3-amine (415).
Figure imgf000237_0001
[0300] Step A: Preparation of 5--(3-(6--methylpyridin-2-y])- 1-(tetrahydro-2H-pyran- 2-yl)- 1H-pyrazoI-4- yl)benzo[d]isothiazol-3 -amine (12-2). To a stirred solution of 5-6 (500 mg, 1.35 mmol) in dioxane- water (10 mL, 4:1.) was added 12-1 (401 mg, 1.76 mmol) and K3PO4 (572 mg, 2.70 mmol). The reaction mixture was purged with N2 for 15 minutes followed by addition of catalyst Pd(dppf)Cl2-DCM (110 mg, 0.135 mmol). Nitrogen purging was continued for 5 minutes. The reaction was allowed to stir at 100 °C for 6 h. The reaction mixture was filtered through a celite bed, then diluted with water and extracted with EtOAc. The organic portion was concentrated under vacuum to get the crude residue which was petrified by silica gel column chromatography (2-5% MeOH-DCM). It was further enriched by triturating w'ith diethyl ether to afford 12-2 (230 mg) as a white solid. [M+ H]+ calcd for C21H2lN5OS 392.15, found 392.17.
[0301] Step B: Preparation of 5 -[3 -(6 -methyl- 2 -pyridyl)- 1H-pyrazol-4-yI]-N-(5-piperazin-1-yl-2-pyridyI)- 1,2-benzothiazol -3-amine (415). A mixture of 12-2 (25.6 mg, 0.065 mmol), 12-3 (18.66 mg, 0.055 mmol), BrettPhos Pd G4 (5.02 mg, 5.45 μmol), cesium carbonate (71.1 mg, 0.218 mmol), and BrettPhos (2.93 mg, 5.45 μmol) in 1 ,4-dioxane (273 μL) was heated to 85 °C for 16 h. The reaction mixture was filtered through a pad of celite and concentrated under vacuum. TFA (500 μL) was added and the reaction was heated to 50 °C for 1 h. TFA was removed in vacuum and the residue was purified by preparative HPLC chromatography using a gradient (12 to 25%) of acetonitrile in water with 0,05% tri fluoroacetic acid to yield a TFA salt of the title compound (13 mg). [M+H]+ calcd for C25H24N8S 496.18, found 496.1.
[0302] Example 13: Synthesis of 4-((4-isopropylpiperazin- 1-yl)methyl)-2-(4-(5-(6-methylpyridin-2-yl)-1H pyrazol-4-yl)quinolin-6-yl)oxazole (60) and (4-isopropylpiperazin-1-yl)-[2-[4-[3-(6-methyl-2-pyridyl)-1H- pyrazol-4-yl]-6-quinolyl]oxazol-4-yl]methanone (441).
Figure imgf000238_0001
[0303] Step A: Preparation of (Z)-2-(6-bromoqumolin-4-yl)-3-(dimethylamino)-l-(6-methylpyridm-2 yl)prop-2-en-1-one (13-2). A solution of 13-1 (14 g, 41.0 mmol) in N,N-dimethylformamide dimethyl acetal
(140 mL) was stirred at 80 °C for 16 h. The reaction was concentrated under reduced pressure to obtain crude
13-2 (14.0 g) as a red oil, which was used directly in the next step. [M+H]+ calcd for CroHisBrNgO
396.1/398.1, found 398.1.
[0304] Step B: Preparation of 6-bromo-4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinoline (13-3). A solution of 13-2 (14.0 g, 35.3 mmol) and NH2NH2H2O (12.4 g, 247 mmol) in EtOH (120 mL) was stirred at
80 °C for 4 h. The solution was concentrated under reduced pressure and the residue was purified by flash chromatography (0-2% MeOH/CH2Cl2) io obtain 13-3 (7.5 g) as a red oil. [M+H]+ calcd for CisHnBr^
365.0/367.0, found 365.1. [0305] Step C: Preparation of methyl 4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinoline-6-carboxylate (13-4). To a solution of 13-3 (6.50 g, 17.8 mmol), PPh3 (934 mg, 3.60 mmol) and NaOAc (2.20 g, 26.7 mmol) in MeOH (130 mL) and DMF (26 ml.,) was added Pd(OAc)2 (1.20 g, 5.30 mmol) under N2 atmosphere. The reaction was stirred at 80 °C for 16 h under CO (50 psi). The mixture was filtered and concentrated under reduced pressure. The residue was purified by flash chromatography (0-5% MeOH/CH2Cl2) to afford 13-4 (3.0 g) as a yellow solid. [M+H]+ calcd for C20H16N4O2345.1, found 345.1.
[0306] Step D: Preparation of (4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinolin-6-yl)methanol (13-5). UAIH4 (330 mg, 8.70 mmol) was added slowly to a solution of 13-4 (1.50 g, 4.40 mmol) in THF (30 mL) at - 20 °C. The mixture was warmed to 10 °C and stirred for 4 h. The reaction was quenched with H2O ( 1 mL) and the suspension was filtered and washed with EtOAc (20 mL). The solid was dried to obtain 13-5 (1.80 g) as a yellow solid. [M+H]+ calcd for C19H16N4O 317.1, found 317.0.
[0307] Step E: Preparation of 4-(5-(6-methylpyridin-2-yi)-1H-pyrazol-4-yi)quinoline-6-carbalde.hyde (13-6). To a suspension of 13-5 (500 mg, 1.26 mmol) in DMF (8.0 mL) was added. MnO2 (3.05 g, 31.6 mmol) and the resulting mixture was stirred at 23 °C for 40 h. The crude reaction was filtered through Celite, washing with DMF (10 mL). The filtrate was concentrated under reduced pressure and the residue was purified via flash chromatography (0-10% MeOH/DCM) to afford 13-6 (168 mg) as a white solid. [M+H]+ calcd for C19H14N4O 315.1, found 315.
[0308] Step F: Preparation of methyl 2-(4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinolin-6-yl)oxazole- 4-carboxylate (13-8). To a suspension of 1-serine methyl ester (13-7, 26.7 mg, 0.172 mmol) and K2CO3 (47.5 mg, 0.344 mmol) in DMF (200 p.L) was added a solution of 13-6 (54 mg, 0.172. mmol) in DMA (277 pl.,) and the mixture was stirred at 23 °C for 15 h. The reaction mixture was cooled to 0 °C and treated with bromotrichloromethane (50.8 pL, 0.515 mmol) followed by DBU (78 μL, 0.515 mmol) and the reaction was slowly warmed to 23 °C. The reaction was diluted with 'water (0.5 mL) and extracted with EtOAc (2 x 4 mL). The combined organics were dried over Na2SO4, filtered, and concentrated under reduced pressure. The crude residue was purified via flash chromatography (0- 10% MeOH/DCM) to afford 13-8 (7.0 mg) as a yellow solid. [M+H]+ calcd for C23H17N5O3412.1, found 412.
[0309] Step G: Preparation of 2-(4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinolin-6-yl)oxazole-4- carboxylic acid (13-9). To a suspension of 13-8 (15.8 mg, 0.038 mmol) in THF (96 μL) / MeOH (32.0 μL) / water (32.0 μL) was added LiOH (1.6 mg, 0.067 mmol) at 0 °C. The reaction was warmed to 23 °C and was stirred, for 1 h. The solvents were removed under reduced pressure and. the residue was purified by preparative HPLC chromatography to obtain the bis-TFA salt of 13-9 as a white solid (14.1 mg). [M+H]+ calcd for C22H15N5O3 398.1, found 398.2.
[0310] Step H: Preparation of (4-isopropylpiperazin-1-yl)(2-(4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4- yl)quinolin-6-yl)oxazol-4-yl)methanone (441). To a solution of (13-9) (8.0 mg, 0.013 mmol) in DMF (128 μL) stirring at 23 °C was added HATH (5.59 mg, 0.015 mmol) followed by iPr2EtN (11.2 μL, 0.064 mmol) and the resulting mixture was stirred for 10 min. 13-10 (2.29 pL, 0.016 mmol) was added and the resulting mixture was stirred at 23 °C for 16 h. The solvent was removed under reduced pressure and the residue was purified by preparative HPLC chromatography to obtain the bis-TFA salt of 441 (0.6 mg). [M+H]+ calcd for C29H29N7O2508.24, found 508.
[0311] Step I: Preparation of 4-((4-isopropylpiperazin-l-yl)methyl)-2-(4-(5-(6-methylpyridin-2-yl)-1H- pyrazol-4-yl)quinolin-6-yl)oxazole (60). To a solution of 441 (2.0 mg, 2.72 μmol) in THE (25 μL) stirring at 23 °C under N2 was added Li AIH4 (10 pL, 10.0 pmol, 1.0 M in THE) and the resulting mixture was stirred for 2 h. The reaction was quenched with a solution of AcOH/H2O (1:1, 100 pL) and the resulting suspension was filtered. The filtrate was purified by preparative HPLC chromatography to obtain the bis-TFA salt of the title compound (0.6 mg). [M+H]+ calcd for C29H31N7O 494.3, found 494.0.
[0312] Example 14: Synthesis of 4-(5-(6-methyipyridin-2-yi)-1H-pyrazol-4-yl)-7-(2-azaspiro[3.5]non-6-en- 7-yl)quinoline (551).
Figure imgf000240_0001
[0313] Step A: Preparation of (Z)-2-(7-bromoquinolin-4-yl)-3-(dimethylamino)-1-(6-methylpyridin-2- yl)prop-2-en-1-one (13-2). A solution of 13-1 (32.0 g, 93.8 mmol) in N,N-dimethylformaniide dimethyl acetal (250 mL) was stirred at 80 °C for 6 h. The reaction was concentrated under reduced pressure to afford crude 13-2 as a red oil (32.0 g) which was used directly in the next step. [M+H]+ calcd for C2oHisBrN30 396.0/398.0, found 397.8.
[0314] Step B: Preparation of 7-bromo-4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)quinoline (13-3). A solution of 13-2 (32.0 g, 80.8 mmol) and NH2NH2H2O (20.0 g, 404 mmol) in EtOH (300 mL) was stirred at 80 °C for 4 b. The solution was concentrated under reduced pressure and the residue was purified by flash chromatography (0-40% EtOAc/petroIeum ether) and recrystallization (50 mL, of EtOAc) to obtain 13-3 (5.70 g) as a white solid. [M+H]+ calcd for C18H13BrN4 365.0/367.0, found 364.9.
[0315] Step C: Preparation of 4-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)-7-(2-azaspiro[3.5]non-6-en-7- yl)quinoline (551). To a suspension of 13-3 (0.018 g, 0.05 mmol), 13-4 (21.0 mg, 0.060 mmol), and Pd(dppf)Cl2 (1.83 mg, 2.500 μmol) in 1,4-dioxane (0.25 mL) was added a solution of Na2CO3 (0.150 mL, 1.0 M, 0.150 mmol). The mixture was degassed via vacuum/N) backfill (5x) and the vial was sealed and heated to 100 °C for 20 h. The reaction was concentrated under reduced pressure and the residue was dissolved in TFA (1.00 mL) and stirred at 23 °C for 2 h. The solvent was removed under reduced pressure and the residue was purified by preparative HPLC chromatography to obtain the bis-TFA salt of the title compound (9.7 mg). [M+H]+ calcd for C26H25N5 408.2, found 408.3.
[0316] Example 15: Synthesis of 7-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)-4-(4-(piperidin-3- yl)phenyl)isoquinoline (557).
Figure imgf000241_0001
[0317] Step A: Preparation of 4,7- dibromoisoquinoline (15-2). To a stirred solution of 15-1 (3.0 g, 14.42 mmol) in acetic acid (30.0 ml) was added NBS (3.08 g, 17.30 mmol) at 0 °C. The reaction mass was allowed to warm to RT and then stirred for 6 h at 110 °C. The reaction was cooled back to RT and concentrated under reduced pressure to get the crude compound. The crude compound was quenched using saturated NaHCCh solution and extracted with EtOAc. The organic layer was dried over NasSOa, concentrated under vacuum, and purified by silica column chromatography (0-5% EtOAc in heptane), yielding the title compound (2.2 g). [M+H]+ calcd for C9H3Br2N 284.88, found 285.85.
[0318] Step B: Preparation of 4-bromo-7-(5-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)isoquinoline (15-4). To a stirred solution of 15-2 (3.0 g, 10.45 mmol) in 1,4-dioxane (30 ml.,) and H2O (6 mL) was added 15-3 (4.62 g, 12.54 mmol) and Na2CO3 (2.21 g, 20.9 mmol). The reaction mixture was purged with argon for 5 minutes, then Pd(dppf)Cl2-DCM added (852 mg, 1.045 mmol). The reaction was purged with argon for 2 minutes. Finally, the reaction was allowed to stir at 95 °C for 4 h. The reaction mixture was filtered through Cehte and the filtrate partitioned between EtOAc and water. The organic layer was separated, dried over sodium sulfate and concentrated under reduced pressure to afford the crude material, which was purified by silica column chromatography (0-30% EtOAc in hexanes) and further purified by reverse phase HPLC (30-55% acetonitrile in a 5mM solution of ammonium bicarbonate in water) to separate regioisomers, yielding the title compound (1.4 g) as an off-white solid. [M+H]+ calcd for C23H21BrN4O 448.09, found. 449.14.
[0319] Step C: Preparation of 7-(5-(6-methyIpyridin-2-yl)-1H-pyrazoI-4-yl)-4-(4-(piperidin-3- yl)phenyl)isoquinoline (557). A vial of 15-4 (20 mg, 0.045 mmol), 15-5 (16.30 mg, 0.053 mmol), 2M sodium carbonate in water (66.8 μL), and Pd(dppf)Cl2 (3.26 mg, 4.45 μmol) in degassed 1,4-dioxane (0.5 mL) was heated at 105 °C for 16 h. The reaction mixture was concentrated in vacuum. TFA (0.5 mL) was added to the residue and stirred at 50 °C for 1 h. TFA was removed in vacuum and the residue was petrified by preparative HPLC chromatography using a gradient (5 to 35%) of acetonitrile in water with 0.05% trifluoroacetic acid to yield a TFA salt of the title compound (19.3 mg). [M+H]+ calcd for C29H27N5445.23, found 446.2.
[0320] Example 16: Synthesis of 2-(4-(2-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)thieno['3,2-c]pyridin-7- yl) - 1 H-pyrazol - 1 -ylfethan- 1 -amine (294).
Figure imgf000242_0001
[0321] Step A: Preparation of 7-bromothieno[3,2-c]pyridine (16-2). To a stirred solution of 16-1 (4.00 g,
15.5 mmol) in DMSO (25.0 ml) was added Ph2O (25.0 mL) and the resulting mixture stirred at 230 °C for 8 h. Water was added and the reaction extracted using EtOAc thrice followed by washing with brine. The combined organics were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to get the crude compound which was purified by silica gel column chromatography using 0-30% EtOAc/hexanes as an eluent to afford the title compound as brown colored semisolid compound (1.60 g). [M+H]+ calcd for C7H4BrNS 41212.92, found 213.92.
[0322] Step B: Preparation of 7-bronio-2-(trimethyistannyl)thieno[3,2-c]pyridine (16-3). To a stirred solution of 16-2 (1.50 g, 7.01 mmol) in anhydrous THF (20.0 mL) was added EDA (5.26 mb, 10.5 mmol) at -78 °C under argon atmosphere. The reaction was stirred at -78 °C for 1 h followed by addition of MesSnCl (2.09 g,
10.5 mmol) dissolved in anhydrous THF (10.0 mb) dropwise at -78 °C. The reaction mixture was allowed to stir at -78 °C for 10 minutes and then stirred, at rt for 2 h. The reaction was quenched with saturated NH4CI solution and extracted using EtOAc twice. The organic layer was dried over anhydrous Na^SOr, filtered and concentrated under reduced pressure to get the crude material which was purified by column chromatography in neutral alumina using 5-15% EtOAc/heptane as an eluent to afford the title compound as a dark brown viscous compound (950 mg). [M+H]+ calcd for C10H18BrNSSn 376.89, found 377.85.
[0323] Step C: Preparation of 7-bromo-2-(5-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)thieno[3,2-c]pyridine (16-5). A stirred solution of 16-4 (686 mg, 1.86 mmol) in toluene (12.0 mL) was purged with argon for three minutes followed by addition of 16-3 (700 mg, 1.86 mmol) and tetralds(triphenylphosphine)palladium(0) (215 mg, 0.186 mmol). The reaction mixture was allowed to stir at 80 °C for 5 h, then concentrated and purified by silica gel column chromatography using 45-50% EtOAc- heptane as an eluent. The product was then further purified using reverse phase C-18 column chromatography using 35% ACN:(5 mM ammonium acetate buffer) water eluent to afford the title compound (530 mg). [M+H]+ caicd for C21H19BrN4OS 454.05, found 455.19.
[0324] Step D: Preparation of 2-(4-(2-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)thieno[3,2-c]pyridin-7-yl)- 1 H-pyrazol-l-yl)ethan-1 -amine (294). A vial of 16-5 (20 mg, 0.044 mmol), 16-6 (17.77 mg, 0.053 mmol), 2M sodium carbonate in water (65.9 μL), and Pd(dppf)Cl2 (3.21 mg, 4.36 μmol) in degassed 1,4-dioxane (0.5 mL) was heated at 105 °C for 16 h. The reaction mixture was concentrated in vacuum. TFA (0.5 mL) was added to the residue and stirred at 50 °C for 1 h. TFA was removed in vacuum and the residue was purified by preparative HPLC chromatography using a gradient (10 to 20%) of acetonitrile in water with 0.05% trifluoroacetic acid to yield a TEA salt of the title compound (7.0 mg). [M+H1+ caicd for C21H19N7S 401.14, found 402.1.
[0325] Example 17: Synthesis of 2-(5-(6-methylpyridin-2-yl)-l-(tetrahydro-2H-pyran-2-yi)-1H-pyrazol-4 yl)-N-(4-(piperazin-1 -yl)phenyl)tbieno[3,2-c]pyridin-6-amine (664).
Figure imgf000243_0001
[0326] Step A: Preparation of 6 -chloro -2-(trimethylstannyl)thieno[3,2-c]pyridine (17-2). To 17-1 (750 nig, 4.42 mmol) in THF (10 ml) was added LDA (3.32 mL, 6.63 mmol) at -78 °C. The reaction mixture was stirred at -78 °C for 30 min, then trimethyltin chloride ( 1.33 g, 6.63 mmol) added dropwise. The reaction mixture was allowed to reach room temperature within 15 min, then stirred for 2h at rt. The reaction mixture was diluted with EtOAc and saturated NH4CI solution and the organics were separated, dried over Na2SO4 and concentrated under vacuo to give the crude product, used as such without further purification.
[0327] Step B: Preparation of 6-chloro-2-(5-(6-methylpyridin-2-yl)-l-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)thieno[3,2-c]pyridine (17-3). A stirred solution of 17-2 (1.00 g, 3.01 mmol) and 16-4 (777 mg, 2.11 mmol) in toluene (10 ml,) was purged with N2 for 10 min. To it, Pd(PPh3)4 (348 mg, 0.301 mmol) was added, and the reaction mixture was heated at 100 °C for 6 h. The reaction mixture was passed through celite and washed with EtOAc (2-3 times). The combined organics were concentrated under vacuo to give the crude product. The crude material was purified by reverse phase HPLC (10-55% acetonitrile in 0.1% TFA in water) to give the title product as an off-white solid (408 mg). [M+H]+ calcd for C21H19CIN4OS 410.10, found 411.26.
[0328] Step C: Preparation of 2-(5-(6-niethyIpyridin-2-yl)- §-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4-yl)-N- (4-(piperazin-1-yl)phenyI)thieno[3,2-c]pyridin-6-amine (664). A vial of 17-3 (25 mg, 0.061 mmol), 17-4 (20.25 mg, 0.073 mmol), cesium carbonate (59.5 mg, 0.183 mmol), BrettPhos (3.27 mg, 6.08 μmol), and BrettPhos Pd G4 (5.60 mg, 6.08 μmol) in degassed 1,4-dioxane (1 mL) was heated at 105 °C for 16 h. The reaction mixture was concentrated in vacuum. TFA (0.5 mL) was added to the residue and stirred at 50 °C for
1 h. TFA was removed in vacuum and the residue was purified by preparative HPLC chromatography using a gradient (5 to 35%) of acetonitrile in water with 0.05% trifluoroacetic acid to yield a TFA salt of the title compound (12.6 mg). [M+H]+ calcd for C26H25N7S 467.19, found 468.1.
[0329] Example 18: Synthesis of 2-(4-(2-(5 -(6-methylpyridin-2-yl)-1 H-pyrazol -4-yi)thieno[3,2-c]pyridin-7- yl)-1H-pyrazol-1-yl)ethan-1 -amine (603).
Figure imgf000244_0001
[0330] Step A: Preparation of 4-bromothieno[2,3-c]pyridine (18-2). To 18-1 (3.0 g, 11.66 mmol), DMSO:diphenyl ether (20 rnL:20 mL) was added and heated at 230 °C for 4h. The reaction mixture was partitioned between EtOAc and water and the organics were separated, dried over Na2SOr and concentrated under vacuum to give a crude residue. The residue was purified by silica column chromatography (0-100% EtOAc :hexanes) to yield the title compound (1.3 g) [M+H]+ calcd for C7H4BrNS 212.92, found 213.9.
[0331] Step B: Preparation of 4-bromo-2-(trimethylstannyl)thieno[2,3-c]pyridine (18-3). To 18-2 (980 mg., 4.60 mmol) in dry THF (10 ml) was added LDA (6.9 ml, 13.80 mmol) at -78 °C under nitrogen atmosphere. The reaction mixture was stirred at -78 °C for 30 min. To it, MesSnCl (1 .37 g, 6.90 mmol in 5 ml THF) was added dropwise and the resulting mixture allowed to reach room temperature within 15 min, then stirred for 1 h. The reaction mixture was diluted with EtOAc and NH4Cl solution and the organics were separated, dried over Na2SO4 and concentrated under vacuum to give crude title compound as a brown viscous liquid (2.4 g), used in the next step without further purification. [M+H]+ calcd for C10H12BrNSSn 376.89, found 377.83.
[0332] Step C: Preparation of 4-bromo-2-(5-(6-methylpyridin-2-yl)-l-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol -4-yi)thieno[2,3-c]pyridine (18-4). To etude 18-3 (2.4 g, 3.18 mmol) in toluene (10 mi) was added 16- 4 (1.17 g, 3.18 mmol) at rt. The reaction mixture was purged with N2 for 10 min. To this, Pd(PPh3)4 (367 mg, 0.318 mmol) was added and stirred at 80 °C for 4 h. The reaction mass was filtered through celite, the residue washed with EtOAc and the filtrate concentrated and purified by reverse phase HPLC (10-55% acetonitrile / 0.1% TFA in water), yielding the title compound as a reddish-brown solid. (760 mg). [M+H] + calcd for C21H19Br N4OS 454.05, found 455.08.
[0333] Step D: Preparation of 2-(4-(2-(5-(6-methylpyridin-2-yl)-1H-pyrazol-4-yl)thieno[3,2-c]pyridin-7-yl)- 1H-pyrazol-1-yl)ethan-1-amine (603). A vial of 18-4 (20 mg, 0.044 mmol), 16-6 (17.77 mg, 0.053 mmol), 2M sodium carbonate in water (65.9 μL), Pd(dppf)Cl2 (3.21 mg, 4.36 μmol) in degassed 1,4-dioxane (0.5 mL) was heated at 1.05 °C for 1.6 h. The reaction mixture was concentrated in vacuum. TFA (0.5 mL) was added to the residue and stirred at 50 °C for 1 h. TFA was removed in vacuum and the residue was purified by preparative HPLC chromatography using a gradient (2 to 20%) of acetonitrile in water with 0.05% trifluoroacetic acid to yield a TFA salt of the title compound (9.3 mg). [M+H]+ calcd for C21H19N7S 401.14, found 402.1.
[0334] Example 19: Synthesis of 6-(5-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-4- yl)-N-(4-(piperazin-1 -yl)phenyl)tbieno[2,3-d]pyrimidin-2-amine (667).
Figure imgf000245_0001
[0335] Step A: Preparation of 6-bromo-2-chlorothieno[2,3-d]pyriinidine (19-2). To a stirred solution of 19-1 (1.0 g, 5.86 mmol) in THF (10 mL) was added HBr (6 mL , 48% aq.) and the resulting mixture stirred for 5 min before adding H2O2 (1-5 mL, 30% aq.) slowly. The resulting mixture was stirred for 16 h. Sodium bicarbonate solution (20 mL) was added and the mixture was stirred for 5 min, then diluted with EtOAc. The organic layer was separated, dried over sodium sulfate, and. concentrated under vacuum to get the crude compound. The crude compound was purified by silica column chromatography (5% EtOAc/hexanes), yielding the title compound as an off-white solid (1.2 g). [M+H]+ calcd for C6H2BrClN2S 247.88, found 249. [0336] Step B: Preparation of 2-chloro-6-(5-(6-methylpyridin-2-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H- pyrazol-4-yl)thieno[2,3-d]pyrimidine (19-4). To a stirred solution of 19-2 (250 mg, 1.00 mmol) in dioxane:THF (10 mL) was added 19-3 (446 mg, 1.20 mmol) and CS2CO3 (986 mg, 3.02 mmol) in H2O (2.5 ml.,). The reaction mixture was purged with argon for 10 minutes, then PdCl2(PPh3) (70 mg, 0.10 mmol) added and the mixture again purged with argon for 2 minutes. The reaction was allowed to stir overnight at 110 °C. After 16 h, the reaction mixture was diluted with EtOAc and water. The organic layer was separated, dried over sodium sulfate, and concentrated under vacuum to get the crude product, which was purified by reverse phase HPLC (10-55% acetonitrile in 5mM ammonium acetate in water) to yield the title compound as a light yellow solid (970 mg). [M+H]+ calcd for C20H18ClNOS 411.09, found 412.24.
[0337] Step C: Preparation of 6-(5-(6-methylpyridin-2-yl)- 1- (tetrahydro -2H-pyran-2-yl)-1 H-pyrazol -4-yl)-N- (4-(piperazin-1-yl)phenyl)thieno[2,3-d]pyrimidin-2-amine (667). A vial of 19-4 (25 mg, 0.061 mmol), 17-4 (20.20 mg, 0.073 mmol), cesium carbonate (59.3 mg, 0.182 mmol), BrettPbos (3.26 mg, 6.07 μmol), and BrettPhos Pd G4 (5.59 mg, 6.07 jrmol) in degassed 1 ,4-dioxane (1 mL) was heated at 105 °C for 16 h. The reaction mixture was concentrated in vacuum. TFA (0.5 mL) was added to the residue and stirred at 50 °C for 1 h. TFA was removed in vacuum and the residue was purified by preparative HPLC chromatography using a gradient (7 to 42%) of acetonitrile in water with 0.05% trifluoroacetic acid to yield a TFA salt of the title compound (18.8 mg). [M+H]+ calcd for C25H24N8S 468.18, found 469.1.
[0338] Example 20: Synthesis of 1-[6-[4-(4-methylthiazol-2-yl)-1H-imidazol-5-yl]-1,3-benzothiazol-2-yl]- 3-(3-pyridyl)urea (449).
Figure imgf000246_0001
[0339] Step A: Preparation of tert-butyl (6-bromobenzo[d]tliiazol-2-yl)(tert-butoxycarbonyl)carbamate (20- 2). To a solution of 20-1 (19.0 g, 82.9 mmol) in THF (300 mL) was added DIPEA (21.4 g, 166 mmol) and DMAP (2.0 g, 16.6 mmol), then Boc2O (145 g, 663 mmol) was added. The reaction mixture was stirred for 2 h at 70 °C. The mixture was concentrated in vacuum to give the residue. The residue was purified by trituration (PE, 100 mL) to afford 20-2 (28.0 g) as a yellow solid. [M+HF calcd for C17H21BrN2O4S 429.04, found 429.1.
[0340] Step B: Preparation of tert-butyl (6-((4-methylthiazol-2-yl)ethynyl)benzo[d]thiazol-2-yl)carbamate (20-4). A mixture of 20-3 (6.0 g, 48.7 mmol), 20-2 (20.9 g, 48.7 mmol), XPhos-Pd-G2 (1.9 g, 2.44 mmol), and CyrNMe (28.5 g, 146 mmol) in MeCN (100 mL) was stirred at 90 °C for 12 h trader Nr. The mixture was concentrated in vacuum to give the residue. The residue was purified by column chromatography (PE/EA = 10/1-3/1) to afford 20-4 (6.0 g) as a yellow solid. [M+H]+ calcd for C18H17N3O2S2372.08, found 372.2.
[0341] Step C: Preparation of tert-butyl (6-(2-(4-methyithiazol -2-yl)-2-oxoacetyl)benzo[d]thiazol-2- ylicarbamate (20-5). In three separate batches: to a solution of 20-4 (2.0 g, 5.38 mmol) in acetone (50 mL) was added a solution of NaHCO3 (271 mg, 3.23 mmol) and MgSOr (2.7 g, 22.1 mmol) in H2O (30 mL). KMnO.4 (3.3 g, 21.0 mmol) was added to the reaction mixture and the mixture was stirred at 20 °C for 1.5 h. NaNOr (1.4 g, 19.9 mmol) and 10 % H2SO4 (24 mL) were added to the reaction mixture at 0 °C. The reaction mixture was stirred at 0 °C for 30 min. The reaction mixture was diluted with water (200 mL), extracted with EtOAc (400 mL x 3), dried over Na2SO4, filtered and the organic layer was concentrated in vacuum to afford 20-5 (5.0 g, 50% purity) as a yellow solid. The crude product was used directly without further purification. [M+HF calcd for C18H18N3O4S2404.07, found 404.2.
[0342] Step D: Preparation of 6-(4-(4-methylthiazol-2-yl)- 1H-imidazol-5-yI)benzo[d]tliiazol-2 -amine (20-7). In two separate batches: a solution of 20-5 (2.5 g, 3.10 mmol), 20-6 (1.3 g, 9.30 mmol) and NH4OAc (1.4 g, 18.6 mmol) in AcOH (40 mL) was stirred for 2.5 h at 100 °C. The reaction mixture was concentrated in vacuum and basified with sat. NaHCO3 (200 mL) to pH=9. The mixture was extracted with EtOAc (400ml, x 3). The combined organic phase was washed with brine (50 mL x 2), dried over Na2SO4 filtered, and concentrated in vacuum to give a residue. The residue was purified by column chromatography (EA/MeOH - 1/0-10/1) and Prep-HPLC (1% to 21% MeCN in H2O (0.05%HCl)) to afford 20-7 (920 mg) as a yellow solid. [M+HF calcd for CuHnNsSr 314.05, found 314.2.
[0343] Step E: Preparation of 1-[6-[4-(4-methylthiazol-2-yl)-1H-imidazol-5-yl]-1,3-benzothiazol-2-yl]-3-(3- pyridyl)urea (449). To a mixture of 20-7 (30 mg, 0.086 mmol) and. DIPEA (44.9 ,μL, 0.257 mmol) in DMF (857 μL) was added 20-8 (20.6 mg, 0.171 mmol). The reaction mixture was allowed to stir for 1 h at 90 °C. The reaction mixture was concentrated under vacuum. The residue was purified by preparative HPLC chromatography using a gradient (14 to 29%) of acetonitrile in water with 0.05% trifluoroacetic acid to yield a TEA salt of the title compound (3.9 mg). [M+H]+ calcd for C20H15N7OS2434.08, found 434.0.
[0344] Example 21: Biochemical ALK5 (TGF-βR1) Assay to Measure pKi.
[0345] Apparent pKi values for compounds of the present disclosure were determined using a recombinant human ALK5 (TGF-βRl) protein (Product No. PR9075A or equivalent, Life Technologies) and a commercially -available kinase assay (LANCE®1 (lanthanide chelate excite) Ultra ULight™ kinase assay. Product Nos. TRF0130-M and TRF02108-M, Perkin Elmer) as described below. [0346] The assays were performed in a 384-well piate (24 columns x 16 wells/rows). An Echo®5550 Liquid Handler (Labcyte) was used to prepare various intermediate concentrations of compounds of the present disclosure in 100% DMSO. From the intermediate concentrations, a range of concentrations (from 10 pM to 25 pM corresponding to volumes up to 105 nL) were prepared and ejected into a final assay plate to be used to create individual dose response curves for each of the subject compounds. To a separate column within the assay plate, 105 nL of DMSO in each well was used to establish a maximum assay signal. Additionally, 105 nL of 100 pM SD-208, a selective TGF-βR1 inhibitor (Catalog #87624, Selleck Chemicals), was used in another column of wells to establish a minimal assay signal.
[0347] With a multidrop dispenser, 8 pL of enzyme mixture (1.25x final) was added to each well. The enzyme mixture consisted of 250 pM ALK5 enzyme and 62.5 nM peptide substrate (LANCE®5 (lanthanide chelate excite) Ultra ULight™-DNA Topoisomerase 2-alpha (Th1l 342)) prepared in assay buffer (50 fflM HEPES, 10 mM MgCl2, 1 mM EGTA, 0.01% Tween-20, pH 7.5 at room temperature) with 2 mM DTT added prior to use. The plate was then sealed with an adhesive seal and. allowed, to equilibrate for 60 minutes at room temperature.
[0348] Next, 2 μL of 125 μM ATP (5x final, 125 pM ATP prepared in assay buffer with 2 mM DTT) was added to the incubated mixtures, covered with a MicroClime® Environmental Lid (Product No. LLS-0310, Labcyte) and. immediately transferred to 37 °C. The reactions were allowed to proceed, at 37 °C for 60 minettes before terminating with the addition of 1.0 pL of detection antibody (LANCE®5 (lanthanide chelate excite) Ultra Europium-anti -phospo-DNA Topoisomerase 2-alpha (Thr1342)) in detection mixture (12 mM EDTA, 4 nM detection antibody prepared in detection buffer (50 mM Tris-HCl, 150 mM NaCl, 0.5% BSA (Fraction V), pH 7.0)) at room temperature. The plate was then read, on a Perkin Elmer EnVision Plate Reader using europium specific reader settings with excitation and emission wavelengths set to 320 or 340 nm and 665 nm, respectively. These data were used to calculate percent enzyme inhibition values based on DMSO and SD-208 background controls.
[0349] For dose -response analyses, percent inhibition versus compound concentrations were plotted, and pIC50 values were determined from a 4-parameter robust fit model with GraphPad Prism V5 Software (GraphPad Software, Inc., La Jolla, CA). This model obtains pIC50 values by fitting the sigmoidal dose- response (variable slope) equation to the data. Results were expressed as pIC50 (negative logarithm, of IC50) and subsequently converted to pKi (negative logarithm of dissociate constant, Ki) using the Cheng-Prusoff equation. The higher the value of pKi (lower value of Ki), the greater the inhibition of ALK5 activity. Certain compounds disclosed herein exhibited pKi values of greater than 8 or greater than 9 when tested in the biochemical ALK5 assay.
[0350] Table 2 shows biological activities of selected compounds in a biochemical ALK5 assay. Compound numbers correspond to the numbers and structures provided in Table 1 and Examples 1-20. Table 2
Figure imgf000249_0001
[0351] Example 22: Cellular ALK5 Potency Assay to Measure pIC50, Inhibition of TGF-β Stimulated pSMAD3 Formation in BEAS-2B Cells.
[0352] The potency of compounds of the present disclosure for inhibition of TGF-β-stimulated SM AD3 phosphorylation was measured in BEAS-2B cells, a human lung epithelial cell line. TGF-β signals through activin receptor-like kinase 5 (ALK5) immediately prior to SMAD3 phosphorylation. As the AlphaLISA
SureFire Ultra kit (Perkin Elmer) quantitatively measures pSMAD3 levels in lysate, the assay demonstrates the ALK5 cellular potency of a test compound.
[0353] BEAS-2.B cells were grown using 50% DMEM (Life Technologies) and 50% F-12. (Life
Technologies) media, supplemented with 10% Fetal Bovine Serum (ATCC), 25 mM HEPES (Life
Technologies), and lx Pen-Strep (Life Technologies). Cells were cultured in a humidified incubator set at 37
°C, 5% CO2, and trypsonized using 0.25% Trypsin with 0.5% polyvinylpyrrolidone (PVP).
[0354] For the assay, BEAS-2B cells were seeded at 7,500 cells/well (25 μL/well) in a 384-well plate and cultured overnight. Before dosing, growth media was aspirated and the wells were rinsed with HBSS Buffer
(HBSS with Calcium and Magnesium, Life Technologies) supplemented with 25 mM HEPES (Life
Technologies) and 1% Bovine Serum Albumin (Roche). Compounds were serially diluted in DMSO, then further diluted with supplemented HBSS Buffer (50 pL/well) to create a compound plate 3x of the final assay concentration, at 0.3% DMSO. The diluted compounds were then added to the cells (8 μL/well) and incubated at 37 °C, 5% CO2 for 1 hour. After the compound incubation, TGF-β (R&D Systems) reconstituted in supplemented HBSS Buffer was added to the cells (12 pL/well, final concentration 10 ng/mL) and incubated for a further 30 minutes, after which the cells were immediately lysed with AlphaLISA lysis buffer
(PerkinElmer). AlphaLISA Acceptor and Detector beads (PerkinElmer) were added 2 hours apart, then incubated overnight to be read the next day. The potency of the compound was determined through analysis of dose-dependent quantified changes in pSMAD3 signal from baseline ( non-compound treated TGF-β stimulated cells). Data are expressed as pIC50 (negative decadic logarithm IC50) values. Certain compounds disclosed herein exhibited pIC50 values of greater than 6 or greater than 7 when tested in BEAS-2B cells.
[0355] Table 3 shows biological activities of selected compounds in a cellular ALK5 potency assay.
Compound numbers correspond to the numbers and structures provided in Table 1 and Examples 1-20.
Table 3
Figure imgf000250_0001
Figure imgf000251_0001
[0356] Example 23: Cytotoxicity Measured by Premature Chromosome Condensation [15] (pCCis).
[0357] The impact of a compound of the present disclosure on cellular adenosine triphosphate (ATP) levels was measured in Beas2B cells, a human lung epithelial cell line. Levels of ATP are correlated with the viability of cells and are often measured to determine tire potential cytotoxicity of compounds. CellTiter-GIo, which lyses the cells and produces a luminescent signal proportional to the amount of ATP present, was used to determine the effect of test compound on cell viability. [0358] Beas2B cells were grown in 50% DMEM (Life Technologies) and 50% F-12 (Life Technologies) media, supplemented with 10% Fetal Bovine Serum ( ATCC), 25 mM HEPES (Life Technologies), and I x
Pen-Strep (Life Technologies). Cells were cultured in a humidified incubator set at 37 °C, 5% CO2, and trypsinized using 0.25% Trypsin with 0.5% polyvinylpyrrolidone (PVP).
[0359] For the assay, Beas2B cells were seeded at 500 cells/well (25 μL/well) in a 384- well plate and cultured overnight. Compounds were serially diluted in DMSO, then further diluted with growth media (40 μL/well) to create a compound plate 6x of the final assay concentration, at 0.6% DMSO. The diluted compounds were then added to the cells (5 μL/well) and incubated at 37 °C, 5% CO2 for 48 hours. After the compound incubation, CeilTiter-Glo (Promega) was added directly to the cells (30 μL/mL). The assay plate was sealed and shaken at 700 rpm for 15 minutes in a darkened environment, then centrifuged for 2 minutes at 1500 rpm to settle the lysate at the bottom of the well. The effect of the compound on cell viability was determined through analysis of dose- dependent quantified changes in ATP from baseline (non -compound treated cells) and wells treated with 60 μM AT9283, a well -characterized cytotoxic compound. Data are expressed as pCC15 (negative decadic logarithm CC15) values. Certain compounds disclosed herein exhibited pCC15 values of less than 6 or less than 5.5 when tested in Beas2B cells.
[0360] Table 4 shows cytotoxicities of selected compounds in a premature chromosome condensation assay.
Compound numbers correspond to the numbers and structures provided in Table 1 and. Examples 1-20.
Table 4
Figure imgf000252_0001
Figure imgf000253_0001
[0361] Example 24: In Vitro Human Liver Microsome Intrinsic Clearance (HLM Clint).
[0362] Liver microsomes were used for in vitro determination of hepatic clearance of compounds of the present disclosure. A microsomal incubation cofactor solution was prepared with 100 mM potassium phosphate buffered to pH 7.4 (BD Biosciences, Woburn, MA) supplemented with 2 mM NADPH (Sigma-
Aidrich, St. Louis, MO). 10 mM DMSO stocks of test compound were diluted and spiked into the cofactor solution to yield a 0.2 μM concentration (0.02% v/v DMSO). Aliquots of frozen human liver microsomes
(Bioreclamation IVT, Baltimore MD) were thawed and diluted, into 100 mM potassium phosphate buffer to yield microsomal protein concentrations of 0.2 mg/mL. Cofactor/drug and microsomal solutions were pre-warmed separately for 4 minutes in a water bath held at 37°C. Incubations (n=1) were started by the combination of equal volumes of cofactor/drug solution with microsomal solution. The final concentration of test compound was 0.1 μM with a final protein concentration of 0.1 mg/mL and final NADPH concentration of 1mM. Samples were collected at times 0, 3, 8, 15, 30, and 45 minutes to monitor the disappearance of test compound. At each time point, 50 μL of incubation sample was removed and spiked into 25 μL of water plus
3% formic acid plus Internal Standard for reaction termination. Samples were then injected onto an AB Sciex
API 4000 triple quadrupole mass spectrometer for quantitation by LC-MS/MS. Mobile Phase A consisted of
HPLC grade water with 0.2% formic acid and Mobile Phase B consisted of HPLC grade acetonitrile with
0.2% formic acid with all samples run through a Thermo HyPURITY C1850 x 2.1 mm column (Waltham,
MA). HLM Clint data was reported in units of pL/min/mg. See Riley, R.J., et al., Drug Metab. Dispos., 2005, September, 33(9), pp. 1304-1311. Certain compounds disclosed herein exhibited HLM Clint of greater than 50 μL/min/mg or greater than 100 pL/min/mg.
[0363] Example 25: Lung PK/PD.
[0364] In-Life Portion
[0365] C57bl/6n mice were acclimated for at least 3 days before use. On the day of the experiment, animals were grouped into sample sizes of 5 (n = 10 for the TGF-β stimulated group). Compounds of the present disclosure (formulated in 3% glycerol in PBS; pH = 4) were pre-treated via oral aspiration (OA; animals are forced to aspirate solution into the lungs by covering their nose). All oral aspirations were performed using a 50 pL dosing volume and accompanied by the appropriate vehicle control groups. Following compound OA treatment, the animals were returned to their home cages and monitored. Compound pre-treatment occurred 4 hours prior to harvest for screening and dose-response studies; duration studies had variable compound pre- treatment times. One hour prior to harvest, animals were challenged via oral aspiration a second time with PBS vehicle or recombinant human TGF-β1 protein (0.01 μg per animal dissolved in 1 part 4 mM HCl and 2 parts 3% glycerol in PBS). Five minutes prior to harvest, animals were deeply anesthetized under isoflurane and euthanized via cervical dislocation. Bronchoalveolar lavage fluid (BALF), plasma and left lung lobes were collected during harvest.
[0366] Sample Collection and Processing
[0367] Blood plasma was collected via open cardiac puncture. After whole blood collection, the samples were placed in EDTA-coated tubes to prevent coagulation. Blood samples were spun at L5300xg’s for 4 minutes at 4 °C to separate the plasma. Plasma was immediately isolated, frozen and submitted for bioanalytical (BA) analysis.
[0368] In order to collect BALF, the lungs were flushed via the trachea with 0.7 mL of PBS 3 times. The BALF, which consists almost entirely of tissue-derived macrophages, was immediately centrifuged at 700xg’s for 15 minutes. After centrifugation, the supernatant was removed, the BALF was re-suspended in IX cell lysis buffer, and immediately frozen. Prior to BA submission, the BALF was dethawed and sonicated for 30 minutes on cold water to lyse open the cells
[0369] Left lung lobes were harvested immediately after BALF collection. Lung samples were homogenized in 500 jiL of lx cell lysis buffer. After homogenization, the samples were split: half of the sample was immediately placed on a rotisserie for 10 minutes while the other half was immediately frozen for BA analysis. The samples placed on the rotisserie were then centrifuged at 10,000xg’s for 10 minutes in order to separate the protein in the supernatant from pelleted debris. Following collection of the supernatant, a total protein quantification assay (Bradford) was performed to normalize the concentrations of all samples. Using the Hamilton star liquid handling system, each sample was diluted in lx cell lysis buffer to 2 mg/mL of protein. Samples were stored at -80 °C or immediately processed using the Meso-scale Discovery system. [0370] Phospho-SMAD3 (pSMAD3) and Total- SMAD3 (tSMAD3) Quantification using Meso-scale Discovery
[0371] Meso-scale Discovery (MSD) is an electrochemical protein quantification assay that requires specialized microplates with carbon electrodes attached to the bottom. These carbon electrodes allow for greater attachment of biological reagent to microplates, thus allowing for a more sensitive read-out when compared to a traditional ELISA. Similar to a standard sandwich ELISA, MSD requires use of a coating antibody that binds the target protein(s) within the sample. After sample incubation, a primary antibody is used to bind the epitope of interest. Following addition of the primary antibody, a secondary -antibody with a SULFO-TAG detection is used to allow for quantification of the epitope of interest. Lastly, the microplate is read via an electric pulse that causes the SULFO-TAG to emit light, which serves as the final read-out of the assay.
[0372] The coating antibody (SMAD3, clone = 5G- 11) was incubated overnight in the specialized MSD microplates at 4 °C. The next day, the microplates were blocked in 3% BSA (bovine serum albumin) for 70 minutes to prevent non-specific protein binding to the bottom of the microplate. After a wash step, 50 pg of hing samples were loaded into the MSD-piate and incubated for 2 hours at room temperature. The plates were washed again to remove unbound sample; either phospo-SMAD3 (pSMAD3; clone = EP568Y) or total- SMAD3 (tSMAD3) primary antibody were incubated for 1 hour. Following a wash step, the anti-rabbit SULFO-tag detection antibody was incubated for 50 minutes. After a final wash step, MSD-read buffer was added to each sample. pSMAD3 and I.SMAD3 quantification was performed using an MSD-specific plate reader (Sector S 600).
[0373] Data Analysis
[0374] Samples were immediately analyzed using an outlier analysis (Grubbs test, a = 0.05). After outlier removal, the raw pSMAD3 were divided by the tSMAD3 luminescent readings. In screening and doseresponse studies, the pSMAD3/tSMAD3 ratio was normalized to the TGF-β induction group (set to 100%) in order to minimize the variability between stimulation. First, the 3% glycerol/PBS group was compared with the 3% glycerol/TGF-β with a student's t-test (cut-off: p=0.05) to ensure a pSMAD3 window was present. A one-way ANOVA (fisher’s uncorrected LSD) was used to compare all drug treated groups with the 3% glycerol/TGF-β group to determine if statistically significant differences are observed. Percent pSMAD3 inhibition was calculated using the vehicle pSMAD3 as a baseline value and displayed as the final readout. Dose-response curves were fitted with a 4-paramater non-linear regression algorithm; the minimum response was set to 0% pSMAD3 inhibition and the maximum response set to 100% pSMAD3 inhibition. Compound potencies were obtained from the regression and reported as ID50s.
[0375] PK Study
[0376] Plasma, lung and macrophage drug concentrations were quantified. Total macrophage concentration was normalized to the total macrophage cell volume over the total drug recovered in the BALF. The alveolar macrophage volume used in the calculation was based on a publication by Krombach et ah (Environmental Health Perspectives, September 1997, Vol. 105, Supplement 5, pp. 1261-1263) which estimated the rat alveolar macrophage volume to be approximately 1200 μm3 or 1.2e-9 mL. The assumption was made that the mouse alveolar macrophage volume is similar to that of the rat. Normalized total macrophage concentration recovered = (total drug recovered from BALE) / (total cell counts* 1.2e-9 mL).
[0377] Certain compounds disclosed herein exhibited (lung AUC0-t):(plasma AUC0-t) ratios of greater than 10, such as greater than 50, greater than 75, or greater than 100. A compound intended for local delivery to the lung with minimal systemic exposure, in some embodiments, exhibits a (lung AUC0-t): (plasma AUC0-t) ratio of greater than 50. Certain compounds provided in Table 2 having pKi values of greater than 9.5 exhibited a (lung AUC0-t):(plasma AUC0-t) ratio of greater than 75.
[0378] Example 26: Cardiac PK/PD.
[0379] In-Life Portion
[0380] C57bl/6n mice were acclimated for at least 3 days before use. On the day of the experiment, animals were grouped into sample sizes of 5-10. Test compounds were pre-treated via oral aspiration (OA; animals are forced to aspirate solution into the lungs by covering their nose). All oral aspirations were performed using a 50 pL dosing volume and accompanied by a vehicle control group (3% glycerol in PBS, pH = 4). Following compound OA treatment, the animals were returned to their home cages and monitored. Compound, pre- treatment occurred either 2 or 4 hours prior to harvest. One hour prior to harvest, animals were challenged via tail-vein intravenous injection with PBS vehicle or recombinant human TGF-βl protein (1 pg per animal dissolved in 1 part 4 mM HC1 and 2 parts 3% glycerol in PBS). Five minutes prior to harvest, animals were deeply anesthetized under isoflurane and euthanized via cervical dislocation. Plasma, left lung lobes and whole hearts were collected during harvest.
[0381] Sample Collection and Processing
[0382] Blood plasma was harvested as described above in the Lung PK/PD experiment. Whole hearts were processed in the same manners as left lung lobes in the Lung PK/PD experiment. Left lung lobes were homogenized in 500 μL of water and submitted for BA Analysis.
[0383] Phospho-SMAD3 (pSMAD3) and Total-SMAD3 (1SMAD3) Quantification using Meso-scale Discoven;
[0384] Heart samples were processed using MSD in the same manner as the left lung lobes above. Data analysis was performed in the same manners as the lung PK/PD experiment. Plasma, lung and heart drug concentrations were quantified.
[0385] There was minimal target engagement systemically following treatment with one or more compound disclosed herein, as measured by SMAD3 phosphorylation inhibition. In some examples, a compound disclosed, herein exhibited less than 10% target engagement systemically as measured by SMAD3 phosphorylation inhibition. [0386] Example 27: Efficacy study in syngeneic cancer model.
[0387] One or more compounds disclosed herein, e.g., a compound provided in Table 1 having an ALK5 pKi value of greater than 9.5, or greater than 10.5 (a measurement reflecting the ability of the compound to inhibit ALK5 activity, measured in accordance with Example 21), are expected to suppress tumor growth in syngeneic cancer models when administered alone or in combination with an immunotherapeutic agent. Six- to 8-week old BALB/c mice are used for In vivo efficacy studies in accordance with IACUC guidelines. Commercially available 4T1 cells (0.5-2.0 x 1.04 cells/mouse) are implanted subcutaneously into the right flanks of BALB/c mice. When the tumor reaches a palpable size of approximately 8-10 mm. in diameter, the primary tumors are surgically removed, and the mice are randomly assigned to vehicle control or compound treatment groups. Alternatively, CT26 cells (0.5-2.0 x 104 cells/mouse) are injected intravenously into BALB/c mice to generate the cancer model. Two days following the surgery, or 7 days following injection of CT26 cells, the mice are treated with either (1) vehicle control, (2) a compound of the present disclosure at an appropriate amount and frequency (formulated in 3% glycerol in PBS: pH 4) via oral aspiration or intranasaliy, (3) an immunotherapeutic agent (e.g., pembrolizumab or durvalumab) at an appropriate amount and frequency, or (4) a compound of the present disclosure and an immunotherapeutic agent, each at an appropriate amount and frequency.
[0388] Body weight is measured twice weekly. Following 2- to 4- weeks of treatment, the lung and liver of each animal is harvested, and the number of metastatic cells in each tissue sample determined using a clonogenic metastasis assay. Cells may be further subjected to one or more of FACS analysis, T-cell function assay, and RNA extraction. It is expected that the animal group treated with one or more of the ALK5 inhibitors disclosed herein exhibits reduction in lung tumor burden. Activation of an immune response by the ALK5 inhibitor may stimulate both local and systemic antitumor T-cell activation, thus a reduction in liver tumor burden may also be observed. When administered in combination with an immunotherapeutic agent, a compound of the present disclosure, such as a compound provided in Table 1, is expected to produce an increased reduction in lung tumor burden relative to the reduction in tumor burden observed in animals treated with either single agent alone. The compounds described herein are expected to interact synergistically with an immunotherapeutic agent to suppress tumor growth and increase survival.
[0389] Example 28: Prophylactic study in murine DSS-induced intestinal fibrosis model.
[0390] One or more compounds disclosed herein, e.g., a compound provided in Table 1 having an ALK5 pKi value of greater than 9.5, or greater than 10.5 (a measurement reflecting the ability of the compound to inhibit ALK5 activity, measured in accordance with Example 21), are expected to slow, halt or reverse the progression of intestinal fibrosis in a murine colitis model. Six to 8-week old male C57BL/6J mice are tagged and weighed. The drinking water of the animals is treated with 2.5% dextran sulfate sodium (DSS) for 7 days to induce acute colitis, followed by 2 days of normal drinking water. Three, 3 -week cycles of 2.5% DSS treatment (1 week of 2.5% DSS in water; 2 weeks of normal water) are then completed to induce intestinal fibrosis.
[0391] Starting on day one of DSS administration, mice are treated with either vehicle control or a compound of the present disclosure at an appropriate amount and frequency via oral gavage (e.g., once daily). The animals are sacrificed 9 weeks after the first DSS administration, then distal, mid and proximal sections of the colon harvested for histologic analysis, RNA extraction and cytokine measurement. A compound of the present disclosure, such as a compound provided in Table 1, is expected to decrease ALK5 activity in the colon and to slow or prevent intestinal fibrosis as evidenced by one or more of (1) reduction in the ratio of colon weight to colon length; (2) reduction in deposition of extracellular matrix as observed by histology; (3) reduction in expression of collagen 1 (Colla1) and connective tissue growth factor (Ctgf) in colon tissue; and (4) reduction in production of TGF-β1 and IL6 in the colon, relative to vehicle-treated controls.
[0392] Example 29: Efficacy study in murine DSS-induced intestinal fibrosis model.
[0393] One or more compounds disclosed herein, e.g., a compound provided in Table 1 having an ALK5 pK, value of greater than 9.5, or greater than 10.5 (a measurement reflecting the ability of the compound to inhibit ALK5 activity, measured in accordance with Example 21), are expected to slow, halt or reverse the progression of intestinal fibrosis in a murine colitis model. Six to 8-week old male C57BL/6J mice are tagged and weighed. The drinking water of the animals is treated with 2.5% dextran sulfate sodium (DSS) for 7 days to induce acute colitis, followed by 2 days of normal drinking water. Three, 3 -week cycles of 2.5% DSS treatment (1 week of 2.5% DSS in water; 2 weeks of normal water) are then completed to induce intestinal fibrosis.
[0394] Following the second of the 3 cycles of DSS administration, mice are treated with either vehicle control or a compound of the present disclosure at an appropriate amount and frequency via oral gavage (e.g., once daily). Animals are sacrificed at either 6, 9 or 12 weeks after the first DSS cycle, then distal, mid and proximal sections of the colon harvested for histologic analysis, RNA extraction and cytoki ne measurement. A compound of the present disclosure, such as a compound provided in Table 1, is expected to decrease ALK5 activity in the colon and to slow, halt or reverse intestinal fibrosis as evidenced by one or more of (1) reduction in the ratio of colon weight to colon length; (2) reduction in deposition of extracellular matrix as observed by histology; (3) reduction in expression of collagen 1 (Coll a1) and connective tissue growth factor (Cigf) in colon tissue; and (4) reduction in production of TGF-β1 and IL6 in the colon, relative to vehicle - treated controls.
[0395] Example 30: Efficacy study in adoptive T-cell transfer model of colitis.
[0396] One or more compounds disclosed herein, e.g., a compound provided in Table 1 having an ALK5 pKi value of greater than 9.5, or greater than 10.5 (a measurement reflecting the ability of the compound to inhibit ALK5 activity, measured in accordance with Example 21), are expected to slow, halt or reverse the progression of intestinal fibrosis in an adoptive T-cell transfer model of colitis. Six- to 8 -week old female CB17 SCID mice are tagged and weighed, then administered CD4* CD25" CD62L* naive T cells isolated from the spleens of Balb/C mice (IP; lx 106 cells) to induce colitis.
[0397] Once diarrhea and a 10% or greater decrease in body weight are observed (typically around week 2), mice are treated with either vehicle control or a compound of the present disclosure at an appropriate amount and frequency via oral gavage (e.g., once daily). Animals are sacrificed 45 days after induction of colitis, then distal, mid and proximal sections of the coion harvested for histologic analysis, RNA extraction and cytokine measurement. A compound of the present disclosure, such as a compound provided in Table 1, is expected to decrease ALK5 activity in the colon and to slow, halt or reverse intestinal fibrosis as evidenced by one or more of (1) reduction in the ratio of colon weight to colon length; (2) reduction in deposition of extracellular matrix as observed by histology; (3) reduction in expression of collagen 1 (Colla1) and connective tissue growth factor (Ctgf) in colon tissue; and (4) reduction in production of TGF-β1 and IL6 in the colon, relative to vehicle- treated controls.
[0398] Example 31: Efficacy study in monocrotaline model of severe pulmonary hypertension.
[0399] One or more compounds disclosed herein, e.g., a compound provided in Table 1 having an ALK5 pKi value of greater than 9.5, or greater than 10.5 (a measurement reflecting the ability of the compound to inhibit ALK5 activity, measured in accordance with Example 21), are expected to slow, halt or reverse the progression of pulmonary hypertension in a monocrotaline (MCT) model of severe pulmonary hypertension. Male Sprague-Dawley rats are tagged, weighed, and randomly divided into control and MCT-treated groups. The rats in the MCT-treated group are administered a single dose of MCT (60 mg/kg, s.c.), then treated with either (I) vehicle control; (2) sildenafil (30 mg/kg, p.o., b.i.d.); or (3) a compound of the present disclosure at an appropriate amount and frequency (formulated in 3% glycerol in PBS; pH = 4) via oral aspiration.
[0400] Following 2-weeks of treatment, the animals are anesthetized with ketamine/xylazine for terminal monitoring of pulmonary and systemic arterial pressures along with heart rate. The lungs of each animal are then harvested for histologic analysis. A compound of the present disclosure, such as a compound provided in Table 1, is expected to decrease ALK5 activity in the lung and slow, halt or reverse the progression of pulmonary hypertension as evidenced by one or more of (1) reduction in systolic pulmonary arterial pressure; (2) reduction in right ventricular (RV) systolic pressure; (3) reduction in RV diastolic pressure; (4) increase in cardiac output; (5) reduction in RV hypertrophy; (6) reduction in pSmad2 or pSmad3 staining within vascular and/or alveolar cells; (7) reduction in medial thickness; (8) reduction in vascular smooth muscle cell proliferation; (9) reduction in vascular smooth muscle hypertrophy; and (10) reduction in expression of matrix metalloproteinase (MMP)-2 and/or MMP-9.

Claims

CLAIMS WHAT IS CLAIMED IS:
1. A compound of Formula (I):
Figure imgf000260_0001
or a pharmaceutically acceptable salt thereof, wherein:
X and Y are each independently selected from CH and N;
A is a 9- or 10-membered bicyclic heteroaryl group selected from:
Figure imgf000260_0002
Figure imgf000260_0003
and
Figure imgf000260_0004
B is selected from phenyl, pyridyl, and thiazolyl;
RA is selected from:
-NO2, -CN, -SR1, -NR1R4, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)OR5, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2; C2-10 alkenyl and C2-10 alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2 -P(O)(R1)2, =O, =S, =N(R1), C3-12 carbocycle, and 3- to 12-membered heterocycle; C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, C3. 12 carbocycle, and 3- to 12-membered heterocycle;
C9-10 aryl, C3-12 cycloalkyl, and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, (O)N-O(RC1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
RB is independently selected at each occurrence from halogen, -CN, -NH2, -NHCH3, -NHCH2CH3, -C(O)CH3, -OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CH2F, -CHF2, -CF3, C3-4 carbocycle, and 3- to 4-membered heterocycle; n is an integer from 0 to 3;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle;
R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1;
R4 is independently selected at each occurrence from C1-3 alkyl, C2-6 alkenyl, C2-6 alkynyl, CH2-(C3-12 cycloalkyl), CH2-(C5-12 cycloalkenyl), C2-3 alkyl-(C3-12 carbocycle), and C1-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OCH2CH3, -CH(CH3)2, -C(CH3)3, C3. 12 cycloalkyl, and 3- to 6-membered heterocycle; and
R5 is independently selected at each occurrence from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6- membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH2OH,
-CH2CH2OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6- membered heterocycle.
2. A compound of Formula (I):
Figure imgf000262_0001
or a pharmaceutically acceptable salt thereof, wherein:
X and Y are each independently selected from CH and N;
A is a 9- or 10-membered bicyclic heteroaryl group selected from:
Figure imgf000262_0002
B is selected from phenyl, pyridyl, and thiazolyl;
RA is selected from:
-NO2, -CN, -SR1, -NR1R4, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)OR5, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2;
C2-10 alkenyl and C2-10 alkynyl, each of which is independently optionally substituted at each occurrence with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), C3-12 carbocycle, and 3- to 12-membered heterocycle; C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, C3- 12 carbocycle, and 3- to 12-membered heterocycle;
C9-10 aryl, C3-12 cycloalkyl, and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -OC(O)OR1, -OC(O)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -NO2, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -S(=O)R1, -S(=O)2R1, -S(=O)2N(R1)2, -S(=O)2NR2R3, -NR1S(=O)2R1, -NR1S(=O)2N(R1)2, -NR1S(=O)2NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -O(CO)OR1, -O(CO)N(R1)2, -OC(O)NR2R3, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, -P(O)(OR1)2, -P(O)(R1)2, =O, =S, =N(R1), R1, C1-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl;
RB is independently selected at each occurrence from halogen, -CN, -NH2, -NHCH3, -NHCH2CH3, -C(O)CH3, -OH, -OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CH2F, -CHF2, -CF3, C3-4 carbocycle, and 3- to 4-membered heterocycle; n is an integer from 0 to 3;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3,
-C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH,
-OCH3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, -CF3, C3-12 carbocycle, and 3- to 6-membered heterocycle;
R2 and R3 are taken together with the nitrogen atom to which they are attached to form a heterocycle, optionally substituted with one or more R1;
R4 is independently selected at each occurrence from C1-3 alkyl, C2-6 alkenyl, C2-6 alkynyl, CH2-(C3-12 cycloalkyl), CH2-(C5-12 cycloalkenyl), C2-3 alkyl-(C3-12 carbocycle), and C1-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)OCH3, -C(O)OCH2CH3, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OCH3, -OOCCH2CH3, -CH(CH3)2, -C(CH3)3, C3- 12 cycloalkyl, and 3- to 6-membered heterocycle; and
R5 is independently selected at each occurrence from C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, 1- to 6- membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -CN, -NO2, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -C(O)CH3, -C(O)OH, -C(O)NH2, =O, -OH, -CH2OH, -CH2CH2OH, -OHC3, -OCH2CH3, -CH3, -CH2CH3, -CH(CH3)2, -C(CH3)3, C3-12 carbocycle, and 3- to 6- membered heterocycle.
3. The compound or salt of any one of claims 1 or 2, wherein X is CH and Y is N.
4. The compound or salt of any one of claims 1 or 2, wherein X is N and Y is CH.
5. The compound or salt of any one of claims 1 or 2, wherein X and Y are each N.
6. The compound or salt of any one of claims 1 to 5, wherein B is selected from
Figure imgf000264_0001
and
Figure imgf000264_0002
7. The compound or salt of any one of claims 1 to 5, wherein B is
Figure imgf000264_0003
8. The compound or salt of any one of claims 1 to 5, wherein B is
Figure imgf000264_0004
9. The compound or salt of any one of claims 1 or 2, wherein the compound is selected from: and
Figure imgf000265_0001
Figure imgf000265_0002
10. The compound or salt of any one of claims 1 to 9, wherein A is selected from
Figure imgf000265_0003
Figure imgf000265_0004
and
Figure imgf000265_0005
11. The compound or salt of any one of claims 1 to 9, wherein A is selected from
Figure imgf000265_0006
, and
Figure imgf000265_0007
Figure imgf000265_0008
12. The compound or salt of any one of claims 1 to 9, wherein A is selected from
Figure imgf000265_0009
and
Figure imgf000265_0010
13. The compound or salt of any one of claims 1 to 9, wherein A is selected from
Figure imgf000265_0011
and
Figure imgf000265_0012
14. The compound or salt of any one of claims 1 to 9, wherein A is selected from
Figure imgf000265_0013
and
Figure imgf000265_0014
Figure imgf000265_0015
15. The compound or salt of any one of claims 1 to 9, wherein A is selected from
Figure imgf000266_0001
, and
Figure imgf000266_0002
Figure imgf000266_0003
16. The compound or salt of any one of claims 1 to 15, wherein RA is selected from:
-NR1R4, -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3; C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)R1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle;
C10 aryl, C3-12 cycloalkyl, and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -CN, -OR1, -SR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -C(O)R1, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)OR1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, -C(O)N(R1)2, -C(O)NR2R3, =O, R1, C1.6 alkyl, and C1-6 haloalkyl.
17. The compound or salt of claim 16, wherein RA is selected from:
-NR1R4, -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3; C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; C3-12 cycloalkyl and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
18. The compound or salt of claim 17, wherein RA is selected from 3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12 carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
19. The compound or salt of claim 17, wherein RA is selected from -C(O)OR5, -NR1C(O)R1, and -NR1C(O)N(R1)2.
20. The compound or salt of any one of claims 1 to 19, wherein R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl, 1- to 6-membered heteroalkyl, C0-3 alkyl-(C3-12 carbocycle), and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from halogen, -NH2, -N(CH3)2, -C(O)OCH2CH3, =O, and -CH3.
21. The compound or salt of claim 20, wherein R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by one or more substituents selected from -NH2 and -CH3.
22. The compound or salt of any one of claims 1 to 21, wherein R5 is independently selected at each occurrence from C1-6 alkyl and C0-3 alkyl-(3- to 12-membered heterocycle).
23. The compound or salt of any one of claims 1 to 15, wherein RA is selected from -NH-(3- to 12-membered heterocycle) and -NH-(C3-12 carbocycle), each of which is optionally substituted with C0-3 alkyl- (3- to 12-membered heterocycle), wherein the C0-3 alkyl-(3- to 12-membered heterocycle) is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -CH3, and -CF3.
24. The compound or salt of any one of claims 1 to 15, wherein: RA is selected from:
-C(O)OR5, -NHC(O)NHR1; C1-10 alkyl and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1 and -CH2NHR1;
C5-12 cycloalkenyl, optionally substituted with -NHR1; and
3- to 12-membered heterocycle, -NH-(3- to 12-membered heterocycle), and -NH-(C3- 12 carbocycle), each of which is independently optionally substituted with one or more substituents selected from halogen, -CH2NHR1, and R1;
R1 is independently selected at each occurrence from hydrogen; and C1-6 alkyl and C0-3 alkyl-(3- to 12-membered heterocycle), each of which is optionally substituted by -NH2; and
R5 is independently selected at each occurrence from C1-6 alkyl and C0-3 alkyl-(3- to 12-membered heterocycle).
25. The compound or salt of any one of claims 1 to 5, wherein:
A is selected from , and
Figure imgf000268_0001
Figure imgf000268_0002
B is selected from
Figure imgf000268_0003
and ; and
Figure imgf000268_0004
RA is selected from:
-NR1R4, -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3; C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; C3-12 cycloalkyl and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, (O)R-1O, -CNR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
26. The compound or salt of any one of claims 1 to 5, wherein:
A is
Figure imgf000269_0001
B is selected from
Figure imgf000269_0002
and
Figure imgf000269_0003
; and
RA is selected from -NH-(3- to 12-membered heterocycle) and -NH-(C3-12 carbocycle), each of which is optionally substituted with C0-3 alkyl-(3- to 12-membered heterocycle), wherein the C0-3 alkyl-(3- to 12- membered heterocycle) is optionally substituted by one or more substituents selected from halogen, -NH2, -NHCH3, -N(CH3)2, -NHCH2CH3, -CH2CH2N(CH3)2, -CH3, and -CF3.
27. The compound or salt of any one of claims 1 to 5, wherein:
A is selected from
Figure imgf000269_0005
and
Figure imgf000269_0004
B is selected from
Figure imgf000269_0006
and
Figure imgf000269_0007
and
RA is selected from:
-NR1R4, -NR2R3, -C(O)OR5, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3; C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1, -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
28. The compound or salt of any one of claims 1 to 5, wherein: A is selected from
Figure imgf000270_0001
and
Figure imgf000270_0002
B is selected from
Figure imgf000270_0003
and
Figure imgf000270_0004
and
RA is selected from:
-NR1R4, -NR2R3, -C(O)OR5, -OC(O)R1 , -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, C1-10 alkyl, -N(R1)-C2-10 alkyl, and phenyl, each of which is independently substituted at each occurrence with one or more substituents selected from -OR1, -CH2N(R1)2, -N(R1)2, -CH2NR2R3, -NR2R3, -C(O)OR1, -OC(O)R1 , -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, C3-12 carbocycle, and 3- to 12-membered heterocycle; C3-12 cycloalkyl and C5-12 cycloalkenyl, each of which is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, -C(O)OR1, -OC(O)R1 , -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl; and
3- to 12-membered heterocycle, -N(R1)-(3- to 12-membered heterocycle), and -N(R1)-(C3-12carbocycle), wherein each C3-12 carbocycle and 3- to 12-membered heterocycle in RA is independently optionally substituted with one or more substituents selected from halogen, -OR1, -CH2N(R1)2, -N(R1)2, -NR2R3, -CH2C(O)OR1, - C(O)OR1 , -OC(O)R1 , -NR1C(O)R1, -NR1C(O)N(R1)2, -NR1C(O)NR2R3, =O, R1, C1-6 alkyl, and C1-6 haloalkyl.
29. The compound or salt of any one of claims 25 to 28, wherein B is
Figure imgf000270_0005
30. A substantially pure stereoisomer of the compound or salt of any one of claims 1 to 29.
31. The compound or salt of claim 30, wherein the stereoisomer is provided in at least 90% enantiomeric excess.
32. A compound selected from Table 1, or a pharmaceutically acceptable salt thereof.
33. A compound selected from Table la, or a pharmaceutically acceptable salt thereof.
34. A conjugate of the formula:
Figure imgf000270_0006
wherein:
A' is an antibody construct or targeting moiety;
L1 is a linker;
D' is a compound or salt of any one of claims 1 to 32; and p is an integer from 1 to 20.
35. A pharmaceutical composition comprising the compound or salt of any one of claims 1 to 33 and a pharmaceutically acceptable carrier.
36. The pharmaceutical composition of claim 35, wherein the pharmaceutical composition is formulated for inhalation.
37. A method of inhibiting ALK5, comprising contacting ALK5 with an effective amount of the compound or salt of any one of claims 1 to 33.
38. A method of treating an ALK5-mediated disease or condition in a subject, comprising administering to the subject a therapeutically effective amount of the compound or salt of any one of claims 1 to 33.
39. The method of claim 38, wherein the disease or condition is selected from fibrosis, alopecia, and cancer.
40. The method of claim 38, wherein the disease or condition is fibrosis.
41. A method of treating fibrosis, comprising administering to a patient a therapeutically effective amount of the compound or salt of any one of claims 1 to 33.
42. The method of claim 40 or 41, where the fibrosis is selected from systemic sclerosis, nephrogenic systemic fibrosis, organ-specific fibrosis, fibrosis associated with cancer, cystic fibrosis, and fibrosis associated with an autoimmune disease.
43. The method of claim 42, where the organ-specific fibrosis is selected from cardiac fibrosis, kidney fibrosis, pulmonary fibrosis, liver fibrosis, portal vein fibrosis, skin fibrosis, bladder fibrosis, intestinal fibrosis, peritoneal fibrosis, myelofibrosis, oral submucous fibrosis, and retinal fibrosis.
44. The method of claim 43, where the pulmonary fibrosis is selected from idiopathic pulmonary fibrosis (IPF), familial pulmonary fibrosis (FPF), interstitial lung fibrosis, fibrosis associated with asthma, fibrosis associated with chronic obstructive pulmonary disease (COPD), silica-induced fibrosis, asbestos- induced fibrosis, and chemotherapy-induced lung fibrosis.
45. The method of claim 43, where the pulmonary fibrosis is idiopathic pulmonary fibrosis (IPF).
46. The method of claim 42, wherein the organ-specific fibrosis is intestinal fibrosis.
47. The method of claim 38, wherein the disease or condition is cancer.
48. The method of claim 47, wherein the cancer is selected from breast cancer, colon cancer, prostate cancer, lung cancer, hepatocellular carcinoma, glioblastoma, melanoma, and pancreatic cancer.
49. The method of claim 48, wherein the lung cancer is non-small cell lung cancer.
50. The method of any one of claims 37 to 49, comprising administering a second therapeutic agent.
51. The method of claim 50, wherein the second therapeutic agent is an immunotherapeutic agent.
52. The method of claim 51, wherein the immunotherapeutic agent is a PD-1 inhibitor or a CTLA-4 inhibitor.
53. The method of claim 51, wherein the immunotherapeutic agent is selected from pembrolizumab and durvalumab.
54. The method of any one of claims 37 to 53, further comprising administering an effective amount of radiation.
55. The method of any one of claims 37 to 54, wherein the compound or salt is administered by inhalation.
56. A compound or salt of any one of claims 1 to 33 for use in treating fibrosis.
57. The use of a compound or salt of any one of claims 1 to 33 for the manufacture of a medicament for treating fibrosis.
PCT/US2022/030925 2021-05-26 2022-05-25 Bicyclic inhibitors of alk5 and methods of use WO2022251359A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163202062P 2021-05-26 2021-05-26
US63/202,062 2021-05-26

Publications (1)

Publication Number Publication Date
WO2022251359A1 true WO2022251359A1 (en) 2022-12-01

Family

ID=82361407

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/030925 WO2022251359A1 (en) 2021-05-26 2022-05-25 Bicyclic inhibitors of alk5 and methods of use

Country Status (1)

Country Link
WO (1) WO2022251359A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115785071A (en) * 2023-01-09 2023-03-14 中国药科大学 3-ethynyl-5- (1H-1,2,3-triazole-4-yl) -1H-indazole compound and application thereof

Citations (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US156A (en) 1837-03-30 Improvement in machines for packing and pressing flour
US868A (en) 1838-08-01 Improvement in brewing beer and ale
US6207A (en) 1849-03-20 Improved spriing snap-hook
US7722A (en) 1850-10-15 Machine for folding paper
US5323907A (en) 1992-06-23 1994-06-28 Multi-Comp, Inc. Child resistant package assembly for dispensing pharmaceutical medications
US5811097A (en) 1995-07-25 1998-09-22 The Regents Of The University Of California Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
WO1998042752A1 (en) 1997-03-21 1998-10-01 Brigham And Women's Hospital Inc. Immunotherapeutic ctla-4 binding peptides
US5855887A (en) 1995-07-25 1999-01-05 The Regents Of The University Of California Blockade of lymphocyte down-regulation associated with CTLA-4 signaling
US5977318A (en) 1991-06-27 1999-11-02 Bristol Myers Squibb Company CTLA4 receptor and uses thereof
US6051227A (en) 1995-07-25 2000-04-18 The Regents Of The University Of California, Office Of Technology Transfer Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
WO2000037504A2 (en) 1998-12-23 2000-06-29 Pfizer Inc. Human monoclonal antibodies to ctla-4
WO2001014424A2 (en) 1999-08-24 2001-03-01 Medarex, Inc. Human ctla-4 antibodies and their uses
US20020039581A1 (en) 2000-01-27 2002-04-04 Carreno Beatriz M. Antibodies against CTLA4 and uses therefor
WO2002040476A1 (en) * 2000-11-16 2002-05-23 Smithkline Beecham Corporation Pyridyl-substituted triazoles as tgf inhibitors
US20020086014A1 (en) 1999-08-24 2002-07-04 Korman Alan J. Human CTLA-4 antibodies and their uses
WO2003042211A1 (en) * 2001-11-15 2003-05-22 Smithkline Beecham Corporation Phenyl substituted triazoles and their use as selective inhibors of akl5 kinase
US6682736B1 (en) 1998-12-23 2004-01-27 Abgenix, Inc. Human monoclonal antibodies to CTLA-4
WO2004035607A2 (en) 2002-10-17 2004-04-29 Genmab A/S Human monoclonal antibodies against cd20
WO2004056875A1 (en) 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
US7109003B2 (en) 1998-12-23 2006-09-19 Abgenix, Inc. Methods for expressing and recovering human monoclonal antibodies to CTLA-4
WO2006121168A1 (en) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
WO2007005874A2 (en) 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
WO2007070866A2 (en) * 2005-12-16 2007-06-21 Alcon, Inc. Control of intraocular pressure using alk5 modulation agents
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
WO2009014708A2 (en) 2007-07-23 2009-01-29 Cell Genesys, Inc. Pd-1 antibodies in combination with a cytokine-secreting cell and methods of use thereof
WO2009101611A1 (en) 2008-02-11 2009-08-20 Curetech Ltd. Monoclonal antibodies for tumor treatment
WO2009114335A2 (en) 2008-03-12 2009-09-17 Merck & Co., Inc. Pd-1 binding proteins
US7595048B2 (en) 2002-07-03 2009-09-29 Ono Pharmaceutical Co., Ltd. Method for treatment of cancer by inhibiting the immunosuppressive signal induced by PD-1
WO2010027827A2 (en) 2008-08-25 2010-03-11 Amplimmune, Inc. Targeted costimulatory polypeptides and methods of use to treat cancer
WO2010036959A2 (en) 2008-09-26 2010-04-01 Dana-Farber Cancer Institute Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
WO2010077634A1 (en) 2008-12-09 2010-07-08 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2010089411A2 (en) 2009-02-09 2010-08-12 Universite De La Mediterranee Pd-1 antibodies and pd-l1 antibodies and uses thereof
US7794710B2 (en) 2001-04-20 2010-09-14 Mayo Foundation For Medical Education And Research Methods of enhancing T cell responsiveness
WO2010126022A1 (en) * 2009-04-28 2010-11-04 大正製薬株式会社 Imidazole derivative
WO2011066342A2 (en) 2009-11-24 2011-06-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
WO2013019906A1 (en) 2011-08-01 2013-02-07 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors
US8435516B2 (en) 2009-10-12 2013-05-07 Pfizer Inc. Cancer treatment
WO2013181452A1 (en) 2012-05-31 2013-12-05 Genentech, Inc. Methods of treating cancer using pd-l1 axis binding antagonists and vegf antagonists
WO2014022758A1 (en) 2012-08-03 2014-02-06 Dana-Farber Cancer Institute, Inc. Single agent anti-pd-l1 and pd-l2 dual binding antibodies and methods of use
US8652465B2 (en) 2005-06-08 2014-02-18 Emory University Methods and compositions for the treatment of persistent infections
US8735553B1 (en) 2013-09-13 2014-05-27 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
WO2014100079A1 (en) 2012-12-21 2014-06-26 Merck Sharp & Dohme Corp. Antibodies that bind to human programmed death ligand 1 (pd-l1)
WO2014206107A1 (en) 2013-06-26 2014-12-31 上海君实生物医药科技有限公司 Anti-pd-1 antibody and use thereof
US20150071910A1 (en) 2013-03-15 2015-03-12 Genentech, Inc. Biomarkers and methods of treating pd-1 and pd-l1 related conditions
WO2015036927A1 (en) 2013-09-10 2015-03-19 Aurigene Discovery Technologies Limited Immunomodulating peptidomimetic derivatives
WO2015036394A1 (en) 2013-09-10 2015-03-19 Medimmune Limited Antibodies against pd-1 and uses thereof
WO2015044900A1 (en) 2013-09-27 2015-04-02 Aurigene Discovery Technologies Limited Therapeutic immunomodulating compounds
WO2015048520A1 (en) 2013-09-27 2015-04-02 Genentech, Inc. Anti-pdl1 antibody formulations
WO2015061668A1 (en) 2013-10-25 2015-04-30 Dana-Farber Cancer Institute, Inc. Anti-pd-l1 monoclonal antibodies and fragments thereof
WO2015085847A1 (en) 2013-12-12 2015-06-18 上海恒瑞医药有限公司 Pd-1 antibody, antigen-binding fragment thereof, and medical application thereof
WO2015109124A2 (en) 2014-01-15 2015-07-23 Kadmon Corporation, Llc Immunomodulatory agents
WO2015112800A1 (en) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Human antibodies to pd-1
WO2015112805A1 (en) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Human antibodies to pd-l1
WO2015112900A1 (en) 2014-01-24 2015-07-30 Dana-Farber Cancer Institue, Inc. Antibody molecules to pd-1 and uses thereof
WO2016140884A1 (en) * 2015-03-02 2016-09-09 Rigel Pharmaceuticals, Inc. TGF-β INHIBITORS
WO2020123453A2 (en) * 2018-12-11 2020-06-18 Theravance Biopharma R&D Ip, Llc Alk5 inhibitors

Patent Citations (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US868A (en) 1838-08-01 Improvement in brewing beer and ale
US6207A (en) 1849-03-20 Improved spriing snap-hook
US7722A (en) 1850-10-15 Machine for folding paper
US156A (en) 1837-03-30 Improvement in machines for packing and pressing flour
US5977318A (en) 1991-06-27 1999-11-02 Bristol Myers Squibb Company CTLA4 receptor and uses thereof
US5323907A (en) 1992-06-23 1994-06-28 Multi-Comp, Inc. Child resistant package assembly for dispensing pharmaceutical medications
US5855887A (en) 1995-07-25 1999-01-05 The Regents Of The University Of California Blockade of lymphocyte down-regulation associated with CTLA-4 signaling
US5811097A (en) 1995-07-25 1998-09-22 The Regents Of The University Of California Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
US6051227A (en) 1995-07-25 2000-04-18 The Regents Of The University Of California, Office Of Technology Transfer Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
WO1998042752A1 (en) 1997-03-21 1998-10-01 Brigham And Women's Hospital Inc. Immunotherapeutic ctla-4 binding peptides
WO2000037504A2 (en) 1998-12-23 2000-06-29 Pfizer Inc. Human monoclonal antibodies to ctla-4
US8143379B2 (en) 1998-12-23 2012-03-27 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
US8883984B2 (en) 1998-12-23 2014-11-11 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
US6682736B1 (en) 1998-12-23 2004-01-27 Abgenix, Inc. Human monoclonal antibodies to CTLA-4
US7132281B2 (en) 1998-12-23 2006-11-07 Amgen Fremont Inc. Methods and host cells for producing human monoclonal antibodies to CTLA-4
US7109003B2 (en) 1998-12-23 2006-09-19 Abgenix, Inc. Methods for expressing and recovering human monoclonal antibodies to CTLA-4
WO2001014424A2 (en) 1999-08-24 2001-03-01 Medarex, Inc. Human ctla-4 antibodies and their uses
US8318916B2 (en) 1999-08-24 2012-11-27 Medarex, Inc. Human CTLA-4 antibodies and their uses
US7605238B2 (en) 1999-08-24 2009-10-20 Medarex, Inc. Human CTLA-4 antibodies and their uses
US8784815B2 (en) 1999-08-24 2014-07-22 Medarex, L.L.C. Human CTLA-4 antibodies and their uses
US20050201994A1 (en) 1999-08-24 2005-09-15 Medarex, Inc. Human CTLA-4 antibodies and their uses
US6984720B1 (en) 1999-08-24 2006-01-10 Medarex, Inc. Human CTLA-4 antibodies
US20020086014A1 (en) 1999-08-24 2002-07-04 Korman Alan J. Human CTLA-4 antibodies and their uses
EP1212422A2 (en) 1999-08-24 2002-06-12 Medarex, Inc. Human ctla-4 antibodies and their uses
US20020039581A1 (en) 2000-01-27 2002-04-04 Carreno Beatriz M. Antibodies against CTLA4 and uses therefor
WO2002040476A1 (en) * 2000-11-16 2002-05-23 Smithkline Beecham Corporation Pyridyl-substituted triazoles as tgf inhibitors
US7794710B2 (en) 2001-04-20 2010-09-14 Mayo Foundation For Medical Education And Research Methods of enhancing T cell responsiveness
WO2003042211A1 (en) * 2001-11-15 2003-05-22 Smithkline Beecham Corporation Phenyl substituted triazoles and their use as selective inhibors of akl5 kinase
US7595048B2 (en) 2002-07-03 2009-09-29 Ono Pharmaceutical Co., Ltd. Method for treatment of cancer by inhibiting the immunosuppressive signal induced by PD-1
WO2004035607A2 (en) 2002-10-17 2004-04-29 Genmab A/S Human monoclonal antibodies against cd20
WO2004056875A1 (en) 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
US7521051B2 (en) 2002-12-23 2009-04-21 Medimmune Limited Methods of upmodulating adaptive immune response using anti-PD-1 antibodies
WO2006121168A1 (en) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US8652465B2 (en) 2005-06-08 2014-02-18 Emory University Methods and compositions for the treatment of persistent infections
US8383796B2 (en) 2005-07-01 2013-02-26 Medarex, Inc. Nucleic acids encoding monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2007005874A2 (en) 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
WO2007070866A2 (en) * 2005-12-16 2007-06-21 Alcon, Inc. Control of intraocular pressure using alk5 modulation agents
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
WO2009014708A2 (en) 2007-07-23 2009-01-29 Cell Genesys, Inc. Pd-1 antibodies in combination with a cytokine-secreting cell and methods of use thereof
WO2009101611A1 (en) 2008-02-11 2009-08-20 Curetech Ltd. Monoclonal antibodies for tumor treatment
WO2009114335A2 (en) 2008-03-12 2009-09-17 Merck & Co., Inc. Pd-1 binding proteins
WO2010027827A2 (en) 2008-08-25 2010-03-11 Amplimmune, Inc. Targeted costimulatory polypeptides and methods of use to treat cancer
WO2010036959A2 (en) 2008-09-26 2010-04-01 Dana-Farber Cancer Institute Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
WO2010077634A1 (en) 2008-12-09 2010-07-08 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2010089411A2 (en) 2009-02-09 2010-08-12 Universite De La Mediterranee Pd-1 antibodies and pd-l1 antibodies and uses thereof
WO2010126022A1 (en) * 2009-04-28 2010-11-04 大正製薬株式会社 Imidazole derivative
US8435516B2 (en) 2009-10-12 2013-05-07 Pfizer Inc. Cancer treatment
WO2011066342A2 (en) 2009-11-24 2011-06-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
WO2013019906A1 (en) 2011-08-01 2013-02-07 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors
WO2013181452A1 (en) 2012-05-31 2013-12-05 Genentech, Inc. Methods of treating cancer using pd-l1 axis binding antagonists and vegf antagonists
WO2014022758A1 (en) 2012-08-03 2014-02-06 Dana-Farber Cancer Institute, Inc. Single agent anti-pd-l1 and pd-l2 dual binding antibodies and methods of use
WO2014100079A1 (en) 2012-12-21 2014-06-26 Merck Sharp & Dohme Corp. Antibodies that bind to human programmed death ligand 1 (pd-l1)
US20150071910A1 (en) 2013-03-15 2015-03-12 Genentech, Inc. Biomarkers and methods of treating pd-1 and pd-l1 related conditions
WO2014206107A1 (en) 2013-06-26 2014-12-31 上海君实生物医药科技有限公司 Anti-pd-1 antibody and use thereof
WO2015036927A1 (en) 2013-09-10 2015-03-19 Aurigene Discovery Technologies Limited Immunomodulating peptidomimetic derivatives
WO2015036394A1 (en) 2013-09-10 2015-03-19 Medimmune Limited Antibodies against pd-1 and uses thereof
US8735553B1 (en) 2013-09-13 2014-05-27 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
WO2015035606A1 (en) 2013-09-13 2015-03-19 Beigene, Ltd. Anti-pd1 antibodies and their use as therapeutics and diagnostics
WO2015048520A1 (en) 2013-09-27 2015-04-02 Genentech, Inc. Anti-pdl1 antibody formulations
WO2015044900A1 (en) 2013-09-27 2015-04-02 Aurigene Discovery Technologies Limited Therapeutic immunomodulating compounds
WO2015061668A1 (en) 2013-10-25 2015-04-30 Dana-Farber Cancer Institute, Inc. Anti-pd-l1 monoclonal antibodies and fragments thereof
WO2015085847A1 (en) 2013-12-12 2015-06-18 上海恒瑞医药有限公司 Pd-1 antibody, antigen-binding fragment thereof, and medical application thereof
WO2015109124A2 (en) 2014-01-15 2015-07-23 Kadmon Corporation, Llc Immunomodulatory agents
WO2015112800A1 (en) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Human antibodies to pd-1
WO2015112805A1 (en) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Human antibodies to pd-l1
WO2015112900A1 (en) 2014-01-24 2015-07-30 Dana-Farber Cancer Institue, Inc. Antibody molecules to pd-1 and uses thereof
WO2016140884A1 (en) * 2015-03-02 2016-09-09 Rigel Pharmaceuticals, Inc. TGF-β INHIBITORS
WO2020123453A2 (en) * 2018-12-11 2020-06-18 Theravance Biopharma R&D Ip, Llc Alk5 inhibitors

Non-Patent Citations (34)

* Cited by examiner, † Cited by third party
Title
ARAUJO-JORGE, T.C. ET AL., CLIN. PHARMACOL. TITER., vol. 92, no. 5, 2012, pages 613 - 621
AYERS, N.B. ET AL., JOURNAL OF BIOMEDICAL RESEARCH, vol. 32, no. 1, 2018, pages 3 - 12
BUNDGARD, H.: "Design of Prodrugs", 1985, ELSEVIER, pages: 7 - 9
CAMACHO ET AL., J CLIN ONCOLOGY, vol. 22, 2004, pages 145
CHILOSI, M. ET AL., RESPIT-. RES., vol. 13, no. 1, 2012, pages 3
CURVO, E., MEM INST OSWALDO CRUZ, vol. 113, no. 4, 2018, pages e170440
FABREGAT, I. ET AL., THE FEBS J., vol. 283, no. 12, 2016, pages 2219 - 2232
H.C. ANSEL: "Pharmaceutical Dosage Forms and Drug Delivery Systems", 1999, LIPPINCOTT WILLIAMS & WHITE
HIGUCHI, T. ET AL.: "Pro-drugs as Novel Delivery Systems", vol. 14, 1987
HURWITZ ET AL., PNAS, vol. 95, no. 17, 1998, pages 10067 - 10071
J. DIEZ, J. CLIN. HYPERTENS., no. 7, 9 July 2007 (2007-07-09), pages 546 - 550
KARIYAWASAM, H.H., J ALLERGY CLIN IMMUNOL., vol. 4, no. 3, 12 September 2009 (2009-09-12), pages 454 - 462
KELLY, J. AM. ACAD. DERMATOL., vol. 58, no. 6, 2008, pages 1025 - 1030
KITAO, A. ET AL., AM. J. PATHOL., vol. 175, 2009, pages 616 - 626
KROMBACH ET AL., ENVIRONMENTAL HEALTH PERSPECTIVES, vol. 105, September 1997 (1997-09-01), pages 1261 - 1263
LAKES, G. ET AL., AM. J. PATHOL., vol. 165, no. 1, 2004, pages 203 - 217
LAPING, N.J., CURRENT OPINION IN PHARMACOLOGY, vol. 3, 2003, pages 204 - 208
LATELLA, G.RIEDER, F., CURR. OPIN. GASTROENTEROL, vol. 33, no. 4, 2017, pages 239 - 245
LOFFEK, S., J. ONCOLO., 2018, pages 1 - 9
MCCORMICK, L.L. ET AL., J. IMMUNOL, vol. 163, 1999, pages 5693 - 5699
MEHDIPOUR, M. ET AL., AGING, vol. 10, 2018, pages 5628 - 5645
MOKYR ET AL., CANCER RESEARCH, vol. 58, 1998, pages 5301 - 5304
NAKANUMA, Y., HEPATOL. RES., vol. 39, 2009, pages 1023 - 1031
PARDALI, E., INT../. MOL. SCI., vol. 18, no. 2157, pages 1 - 22
PENG, Z-W. ET AL., HEPATOLOGY, vol. 63, 2016, pages 217 - 232
REMINGTON: "The Science and Practice of Pharmacy", 2000, LIPPINCOTT WILLIAMS & WHITE
RILEY, R.J. ET AL., DRUG METAB. DISPOS., vol. 3, no. 9, 3 September 2005 (2005-09-03), pages 1304 - 1311
ROSENBLOOM, FIBROSIS: METHODS AND PROTOCOLS, METHODS IN MOLECULAR BIOLOGY, vol. 1627, 2017, pages 1 - 21
WYNN, T.A., J CLIN INVEST., vol. 117, 2007, pages 524 - 529
XIONG, Y. ET AL., EMERGING MICROBES & INFECTIONS, vol. 9, no. 1, 2020, pages 761 - 770
YANG, Y.C., ALLERGY, vol. 67, 2012, pages 1193 - 1202
YINGLING ET AL., NATURE REVIEWS: DRUG DISCOVERY, vol. 3, December 2004 (2004-12-01), pages 1011 - 1022
ZAIMAN, A. L. ET AL., AM. J. RESPIR. CRIT. CARE MED., vol. 177, 2008, pages 896 - 905
ZHAO, X., 1. BIOL. CHEM., vol. 283, no. 6, 2008, pages 3272 - 3280

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115785071A (en) * 2023-01-09 2023-03-14 中国药科大学 3-ethynyl-5- (1H-1,2,3-triazole-4-yl) -1H-indazole compound and application thereof
CN115785071B (en) * 2023-01-09 2024-01-26 中国药科大学 3-ethynyl-5- (1H-1, 2, 3-triazole-4-yl) -1H-indazole compound and application thereof

Similar Documents

Publication Publication Date Title
CA3159561A1 (en) Ras inhibitors
JP2022553859A (en) RAS inhibitor
AU2014364744B2 (en) Novel carboxamides, method for the production thereof, pharmaceutical preparations comprising them, and use thereof for producing medicaments
US11730720B2 (en) ALK5 inhibitors
AU2016226468A1 (en) TGF-beta inhibitors
AU2018255300A1 (en) Bicyclic compounds and their use in the treatment of cancer
US20230218597A1 (en) Substituted pyridines and methods of use
EP4334321A1 (en) Ras inhibitors
WO2022251359A1 (en) Bicyclic inhibitors of alk5 and methods of use
US20230399312A1 (en) Substituted pyrimidines and methods of use
US20210387983A1 (en) Crystalline alk5 inhibitors and uses thereof
JP2024517847A (en) RAS inhibitors
WO2024010762A1 (en) C-myc mrna translation modulators and uses thereof in the treatment of cancer
CN117337292A (en) Pyridine derivatives having N-linked cyclic substituents as cGAS inhibitors
KR20240035402A (en) Novel Selective Angiotensin II Compounds

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: 22736411

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE