WO2018169742A1 - Apoptosis signal-regulating kinase inhibtor - Google Patents

Apoptosis signal-regulating kinase inhibtor Download PDF

Info

Publication number
WO2018169742A1
WO2018169742A1 PCT/US2018/021334 US2018021334W WO2018169742A1 WO 2018169742 A1 WO2018169742 A1 WO 2018169742A1 US 2018021334 W US2018021334 W US 2018021334W WO 2018169742 A1 WO2018169742 A1 WO 2018169742A1
Authority
WO
WIPO (PCT)
Prior art keywords
fluoro
cyclopropyl
pyridin
triazol
imidazol
Prior art date
Application number
PCT/US2018/021334
Other languages
French (fr)
Inventor
Jennifer Lynne ALLEVA
Kassibla Elodie DEMPAH
Julie Farand
Gregory Notte
Original Assignee
Gilead Sciences, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gilead Sciences, Inc. filed Critical Gilead Sciences, Inc.
Publication of WO2018169742A1 publication Critical patent/WO2018169742A1/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
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics

Definitions

  • the present disclosure relates to novel compounds that are inhibitors of ASK 1 and that can be used in the treatment of ASK1 -mediated diseases.
  • the disclosure also relates to methods of preparing the compounds of Formula (I) and methods of treating disease by administering compounds of Formula (I), a salt, co-crystal, solvate, or hydrate thereof.
  • Apoptosis signal-regulating kinase 1 is a member of the mitogen- activated protein kinase ("MAP3K”) family that activates the c-Jun N-terminal protein kinase (“JNK”) and p38 MAP kinase (Ichijo, H., Nishida, E., Irie, K., Dijke, P. T.,
  • ASK1 is activated by a variety of stimuli including oxidative stress, reactive oxygen species (ROS), LPS, TNF-a, FasL, ER stress, and increased intracellular calcium concentrations (Hattori, K., Naguro, I., Runchel, C, and Ichijo, H. (2009) Cell Comm. Signal. 7:1-10; Takeda, K., Noguchi, T., Naguro, I., and Ichijo, H. (2007) Annu. Rev. Pharmacol. Toxicol.
  • ROS reactive oxygen species
  • ASK1 activation and signaling have been reported to play an important role in a broad range of diseases including liver diseases (such as non-alcoholic steatohepatitis, non-alcoholic fatty liver disease, alcoholic hepatitis), cardiovascular diseases (such as pulmonary arterial hypertension and heart failure), kidney disease (such as chronic kidney disease) and also neurodegenerative, inflammatory, autoimmune, and metabolic disorders.
  • liver diseases such as non-alcoholic steatohepatitis, non-alcoholic fatty liver disease, alcoholic hepatitis
  • cardiovascular diseases such as pulmonary arterial hypertension and heart failure
  • kidney disease such as chronic kidney disease
  • neurodegenerative, inflammatory, autoimmune, and metabolic disorders such as chronic kidney disease
  • U.S. Patent No. 8,742, 126, U.S. Patent No. 9,067,933, U.S. Patent No. 9,254,284, U.S. Patent No. 9,333,197, U.S. Patent No. 9,586,932, U.S. Patent Publication No. 2015/0342943, and U.S. Patent Publication No. 2016/0166556 disclose compounds useful as ASK1 inhibitors and methods of treating disease with an ASK1 inhibitor. Surprisingly, applicants have discovered a novel compound exhibiting good potency and pharmacokinetic and/or pharmacodynamic parameters.
  • compositions useful as an inhibitor of ASK1.
  • the compound disclosed herein may find use in pharmaceutical compositions, together with at least one pharmaceutically acceptable excipient, for treating a subject in need thereof.
  • the compound of the present disclosure has been found to inhibit ASK1.
  • compositions including pharmaceutical compositions, kits that include the compound of Formula (I), and methods of making and using compounds of Formula (I). Also provided are salts, co- crystals, hydrates and solvates of the compounds of Formula (I).
  • the compound of Formula (I) is a freebase. In another embodiment of the disclosure, the compound of Formula (I) is a salt. In one embodiment, the compound of Formula (I) is a salt selected from succinate, hydrochloride, phosphate, malate, sulfate, citrate, glutarate, and methane sulfonate.
  • the compound of Formula (I) or a pharmaceutically acceptable salt thereof is amorphous.
  • the compound of Formula (I) freebase is crystalline Form I.
  • the compound of Formula (I) freebase Form I is characterized by an X-ray powder diffractogram having peaks at 8.7, 10.2, and 18.1 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) freebase Form I is characterized by an X-ray powder diffractogram having peaks at 15.8, 17.3, and 23.5 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) freebase Form I is characterized by an X-ray powder diffractogram having peaks at 15.2, 18.5, and 24.9 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • DSC differential scanning calorimetry
  • the disclosure provides a compound of Formula (I) freebase Form II.
  • the compound is characterized by an X-ray powder diffractogram having peaks at 8.7, 10.0, and 17.0 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound is characterized by an X-ray powder diffractogram having peaks at 13.9, 21.3, and 22.8 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound is characterized by an X-ray powder diffractogram having peaks at 24.0, 28.0, and 29.1 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • a compound of Formula (I) freebase Form II is characterized by an X-ray powder diffractogram substantially as shown in FIG. 4.
  • a compound of Formula (I) freebase Form II is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 5.
  • the disclosure provides a crystalline succinate Form I of the compound of Formula (I).
  • the succinate Form I of the compound of Formula (I) is characterized by an X-ray powder diffractogram having peaks at 7.5, 20.9, and 24.9 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the succinate Form I is characterized by an X-ray powder diffractogram having peaks at 15.4, 22.4, and 31.1 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the succinate Form I is characterized by an X- ray powder diffractogram having peaks at 14.9, 23.2, and 25.4 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • crystalline compound of Formula (I) succinate Form I is characterized by an X-ray powder diffractogram substantially as shown in FIG. 7.
  • crystalline compound of Formula (I) succinate Form I is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 8.
  • the disclosure provides a crystalline compound of Formula (I) succinate Form II.
  • the crystalline compound of Formula (I) succinate Form II is characterized by an X-ray powder diffractogram having peaks at 9.8, 11.7, and 23.3 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) succinate Form II is characterized by an X-ray powder diffractogram having peaks at 15.9, 21.5 and 32.0 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) succinate Form II is characterized by an X-ray powder diffractogram having peaks at 16.3, 20.8, and 24.8 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • crystalline compound of Formula (I) succinate Form II is characterized by an X-ray powder diffractogram substantially as shown in FIG. 10.
  • crystalline compound of Formula (I) succinate Form II is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 11.
  • the disclosure provides a crystalline compound of
  • Formula (I) hydrochloride Form I the crystalline compound of Formula (I) hydrochloride Form I is characterized by an X-ray powder diffractogram having peaks at 9.3, 21.2, and 26.2 °2 ⁇ ⁇ 0.2 °2 ⁇ . In one embodiment, the crystalline compound of Formula (I) hydrochloride Form I is characterized by an X-ray powder diffractogram having peaks at 20.9, 24.7, and 27.6 °2 ⁇ ⁇ 0.2 °2 ⁇ . In one embodiment, the crystalline compound of Formula (I) hydrochloride Form I X-ray powder
  • crystalline compound of Formula (I) hydrochloride Form I is characterized by an X-ray powder diffractogram substantially as shown in FIG. 13.
  • crystalline compound of Formula (I) hydrochloride Form I is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 14.
  • the disclosure provides a crystalline compound of Formula (I) hydrochloride Form II.
  • the crystalline compound of Formula (I) hydrochloride Form II is characterized by an X-ray powder diffractogram having peaks at 8.3, 12.0, and 26.0 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the crystalline compound of Formula (I) hydrochloride Form II is characterized by an X-ray powder diffractogram having peaks at 19.3, 21.7, and 24.0 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the crystalline compound of Formula (I) hydrochloride Form II X-ray powder diffractogram having peaks at 9.8, 15.4, and 28.9 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • crystalline compound of Formula (I) hydrochloride Form II is characterized by an X-ray powder diffractogram substantially as shown in FIG. 16.
  • crystalline compound of Formula (I) hydrochloride Form II is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 33.
  • the disclosure provides a crystalline compound of
  • the crystalline compound of Formula (I) hydrochloride Form III is characterized by an X-ray powder diffractogram having peaks at 6.6, 12.9, and 19.7 °2 ⁇ ⁇ 0.2 °2 ⁇ . In one embodiment, the crystalline compound of Formula (I) hydrochloride Form III is characterized by an X-ray powder diffractogram having peaks at 20.4, 23.4, and 24.8 °2 ⁇ ⁇ 0.2 °2 ⁇ . In one embodiment, the crystalline compound of Formula (I) hydrochloride Form III X-ray powder diffractogram having peaks 18.1, 25.4, and 26.7 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • crystalline compound of Formula (I) hydrochloride Form III is characterized by an X-ray powder diffractogram substantially as shown in FIG. 17.
  • crystalline compound of Formula (I) hydrochloride Form III is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 18.
  • the disclosure provides a crystalline compound of Formula (I) L-malate.
  • the crystalline compound of Formula (I) L-malate is characterized by an X-ray powder diffractogram having peaks at 7.2, 21.9, and 24.5 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) L-malate is characterized by an X-ray powder diffractogram having peaks at 10.9, 16.6, and 19.7 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) L-malate is characterized by an X-ray powder diffractogram having peaks at 8.2, 22.8, and 27.3 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • crystalline compound of Formula (I) L- malate is characterized by an X-ray powder diffractogram substantially as shown in FIG. 19.
  • crystalline compound of Formula (I) L- malate is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 20.
  • the disclosure provides a crystalline compound of Formula (I) phosphate Material A.
  • the crystalline compound of Formula (I) phosphate Material A is characterized by an X-ray powder diffractogram having peaks at 6.9, 8.0, and 21.2 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) phosphate Material A is characterized by an X-ray powder diffractogram having peaks at 22.8, 25.3, and 26.0 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) phosphate Material A is characterized by an X-ray powder diffractogram having peaks at 19.8, 20.6, 23.2 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • crystalline compound of Formula (I) phosphate Material A is characterized by an X-ray powder diffractogram substantially as shown in FIG. 22.
  • the disclosure provides a crystalline compound of Formula (I) phosphate Material B.
  • the crystalline compound of Formula (I) phosphate Material B is characterized by an X-ray powder diffractogram having peaks at 13.4, 23.5, and 24.3 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) phosphate Material B is characterized by an X-ray powder diffractogram having peaks at 21.7, 25.1, and 25.8 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) phosphate Material B is characterized by an X-ray powder diffractogram having peaks at 17.0, 18.6, and 22.5 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • crystalline compound of Formula (I) phosphate Material B is characterized by an X-ray powder diffractogram substantially as shown in FIG. 23.
  • the disclosure provides a crystalline compound of Formula (I) sulfate.
  • the crystalline compound of Formula (I) sulfate is characterized by an X-ray powder diffractogram having peaks at 7.2, 13.5, and 21.5 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) sulfate is characterized by an X-ray powder diffractogram having peaks at 16.9, 20.5, and 25.3 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) sulfate is characterized by an X-ray powder diffractogram having peaks at 18.2, 20.6, and 27.2 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • crystalline compound of Formula (I) sulfate is characterized by an X-ray powder diffractogram substantially as shown in FIG. 24.
  • crystalline compound of Formula (I) sulfate is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 25.
  • the disclosure provides a crystalline compound of Formula (I) citrate Material A.
  • the crystalline compound of Formula (I) citrate Material A is characterized by an X-ray powder diffractogram having peaks at 5.6, 9.1, 16.7 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) citrate Material A is characterized by an X-ray powder diffractogram having peaks at 12.2, 21.0, 23.0 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) citrate Material A is characterized by an X-ray powder diffractogram having peaks at 18.7, 24.9, 25.8 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • crystalline compound of Formula (I) citrate Material A is characterized by an X-ray powder diffractogram substantially as shown in FIG. 27.
  • the disclosure provides a crystalline compound of Formula
  • the crystalline compound of Formula (I) citrate Material B is characterized by an X-ray powder diffractogram having peaks at 5.6, 8.6, and 9.1 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) citrate Material B is characterized by an X-ray powder diffractogram having peaks at 12.2, 23.2, and 23.9 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • Material B is characterized by an X-ray powder diffractogram having peaks at 16.8, 18.5, and 21.1 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • crystalline compound of Formula (I) citrate Material B is characterized by an X-ray powder diffractogram substantially as shown in FIG. 28. In one embodiment of the disclosure crystalline compound of Formula (I) citrate Material B is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 29.
  • DSC differential scanning calorimetry
  • the disclosure provides a crystalline compound of Formula (I) glutarate.
  • the crystalline compound of Formula (I) glutarate is characterized by an X-ray powder diffractogram having peaks at 6.7, 8.0, and 12.1 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) glutarate is characterized by an X-ray powder diffractogram having peaks at 14.6, 20.9, and 21.8 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • the compound of Formula (I) glutarate is characterized by an X-ray powder diffractogram having peaks at 17.9, 19.3, and 26.2 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • crystalline compound of Formula (I) glutarate is characterized by an X-ray powder diffractogram substantially as shown in FIG. 30.
  • crystalline compound of Formula (I) glutarate is characterized by a differential scanning calorimetry (DSC) curve
  • the disclosure provides a crystalline compound of Formula (I) methanesulfonate.
  • the crystalline compound of Formula (I) methanesulfonate is characterized by an X-ray powder diffractogram having peaks at 8.4,
  • the compound of Formula (I) methanesulfonate is characterized by an X-ray powder diffractogram having peaks at
  • the compound of Formula (I) methanesulfonate is characterized by an X-ray powder diffractogram having peaks at 12.8, 21.8, and 25.2 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • crystalline compound of Formula (I) methanesulfonate is characterized by an X-ray powder diffractogram substantially as shown in FIG. 32.
  • the disclosure provides a therapeutically effective amount of a compound of Formula (I) a salt, co-crystal, solvate, or hydrate thereof with a pharmaceutically acceptable excipient.
  • the disclosure provides a composition comprising a compound of Formula (I) a salt, co-crystal, solvate, or hydrate thereof with a pharmaceutically acceptable excipient and one, two, or three additional agents.
  • the disclosure provides a composition comprising a compound of Formula (I), a salt, co-crystal, solvate, or hydrate thereof with a pharmaceutically acceptable excipient and one additional agent that is an FXR agonist.
  • the FXR agonist may be a compound of Formula (III) or a compound of Formula (IV) as defined here.
  • the disclosure provides a composition
  • a composition comprising a compound of Formula (I) a salt, co-crystal, solvate, or hydrate thereof with a pharmaceutically acceptable excipient and one additional agent that is an ACC inhibitor.
  • the ACC inhibitor may be a compound of Formula (V) as defined herein.
  • the disclosure provides methods for inhibiting ASK1 with a compound of
  • the disclosure provides a method of treating a condition selected from non-alcoholic steatohepatitis, alcoholic hepatitis, pulmonary arterial hypertension, heart failure with preserved ejection fraction, and diabetic kidney disease by administering a therapeutically effective amount of a compound of Formula (I) or a salt, co-crystal, solvate, or hydrate thereof.
  • the disclosure provides a method of treating fibrosis with a therapeutically effective amount of a compound of Formula (I) or a salt, co-solvate, or hydrate thereof.
  • the disclosure provides a compound of Formula (la):
  • FIG. 1 is a XRPD pattern for a compound of Formula (I) freebase Form I.
  • FIG. 2 is a DSC thermogram for a compound of Formula (I) freebase Form I.
  • FIG. 3 is a TGA thermogram for a compound of Formula (I) freebase Form I.
  • FIG. 4 is a XRPD pattern for a compound of Formula (I) freebase Form II.
  • FIG. 5 is a DSC thermogram for a compound of Formula (I) freebase Form II.
  • FIG. 6 is a TGA thermogram for a compound of Formula (I) freebase Form II.
  • FIG. 7 is a XRPD pattern for a compound of Formula (I) succinate Form I.
  • FIG. 8 is a DSC thermogram for a compound of Formula (I) succinate Form I.
  • FIG. 9 is a TGA thermogram for a compound of Formula (I) succinate Form I.
  • FIG. 10 is a XRPD pattern for a compound of Formula (I) succinate Form II.
  • FIG. 11 is a DSC thermogram for a compound of Formula (I) succinate form II.
  • FIG. 12 is a TGA thermogram for a compound of Formula (I) succinate Form II.
  • FIG. 13 is a XRPD pattern for a compound of Formula (I) HC1 Form I.
  • FIG. 14 is a DSC thermogram for a compound of Formula (I) HC1 Form I.
  • FIG. 15 is a TGA thermogram for a compound of Formula (I) HC1 Form I.
  • FIG. 16 is a XRPD pattern for a compound of Formula (I) HC1 Form II.
  • FIG. 17 is a XRPD pattern for a compound of Formula (I) HC1 Form III.
  • FIG. 18 is a DSC thermogram for a compound of Formula (I) HC1 Form III.
  • FIG. 19 is a XRPD pattern for a compound of Formula (I) L-malate.
  • FIG. 20 is a DSC thermogram for a compound of Formula (I) L-malate.
  • FIG. 21 is a TGA thermogram for a compound of Formula (I) L-malate.
  • FIG. 22 is a XRPD pattern for a compound of Formula (I) phosphate Material A
  • FIG. 23 is a XRPD pattern for a compound of Formula (I) phosphate Material B.
  • FIG. 24 is a XRPD pattern for a compound of Formula (I) sulfate.
  • FIG. 25 is a DSC thermogram for a compound of Formula (I) sulfate.
  • FIG. 26 is a TGA thermogram for a compound of Formula (I) sulfate.
  • FIG. 27 is a XRPD pattern for a compound of Formula (I) citrate Material A.
  • FIG. 28 is a XRPD pattern for a compound of Formula (I) citrate Material B.
  • FIG. 29 is a DSC thermogram for a compound of Formula (I) citrate Material B.
  • FIG. 30 is a XRPD pattern for a compound of Formula (I) glutarate.
  • FIG. 31 is a DSC thermogram for a compound of Formula (I) glutarate.
  • FIG. 32 is a XRPD pattern for a compound of Formula (I) methanesulfonate.
  • FIG. 33 is a DSC thermogram for a compound of Formula (I) HC1 Form II.
  • a dash (“-") that is not between two letters or symbols is used to indicate a point of attachment for a substituent.
  • -C(0)NH 2 is attached through the carbon atom.
  • a dash at the front or end of a chemical group is a matter of convenience;
  • chemical groups may be depicted with or without one or more dashes without losing their ordinary meaning.
  • a wavy line drawn through a line in a structure indicates a point of attachment of a group. Unless chemically or structurally required, no directionality is indicated or implied by the order in which a chemical group is written or named.
  • references to "about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se.
  • the term “about” includes the indicated amount + 10%.
  • the term “about” includes the indicated amount + 5%.
  • the term “about” includes the indicated amount ⁇ 1%.
  • to the term “about X” includes description of "X”.
  • the singular forms "a” and “the” include plural references unless the context clearly dictates otherwise.
  • reference to “the compound” includes a plurality of such compounds and reference to “the assay” includes reference to one or more assays and equivalents thereof known to those skilled in the art.
  • isotopes that can be incorporated into compounds of the disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as, but not limited to 2 H (deuterium, D), 3 H (tritium), n C, 13 C, 14 C, 15 N,
  • isotopically labeled compounds of the present disclosure for example those into which radioactive isotopes such as 3 H, 13 C and 14 C are incorporated.
  • isotopically labelled compounds may be useful in metabolic studies, reaction kinetic studies, detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • the disclosure also includes "deuterated analogues" of compounds of Formula (I) in which from 1 to n hydrogens attached to a carbon atom is/are replaced by deuterium, in which n is the number of hydrogens in the molecule.
  • deuterated analogues of compounds of Formula (I) in which from 1 to n hydrogens attached to a carbon atom is/are replaced by deuterium, in which n is the number of hydrogens in the molecule.
  • Such compounds exhibit increased resistance to metabolism and are thus useful for increasing the half-life of any compound of Formula (I) when administered to a mammal, particularly a human. See, for example, Foster, “Deuterium Isotope Effects in Studies of Drug Metabolism," Trends Pharmacol. Sci. 5(12):524-527 (1984).
  • Such compounds are synthesized by means well known in the art, for example by employing starting materials in which one or more hydrogens have been replaced by deuterium.
  • Deuterium labelled or substituted therapeutic compounds of the disclosure may have improved DMPK (drug metabolism and pharmacokinetics) properties, relating to distribution, metabolism and excretion (ADME). Substitution with heavier isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life, reduced dosage requirements and/or an improvement in therapeutic index.
  • An 18 F labeled compound may be useful for PET or SPECT studies.
  • Isotopically labeled compounds of this disclosure and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • deuterium in this context is regarded as a substituent in the compound of Formula (I).
  • concentration of such a heavier isotope, specifically deuterium may be defined by an isotopic enrichment factor.
  • any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom.
  • a position is designated specifically as “H” or “hydrogen”
  • the position is understood to have hydrogen at its natural abundance isotopic composition.
  • any atom specifically designated as a deuterium (D) is meant to represent deuterium.
  • the compound of Formula (I) is capable of forming salts as described herein.
  • pharmaceutically acceptable salts, hydrates, solvates, tautomeric forms, polymorphs, and prodrugs of the compounds described herein are pharmaceutically acceptable salts, hydrates, solvates, tautomeric forms, polymorphs, and prodrugs of the compounds described herein.
  • “Pharmaceutically acceptable” or “physiologically acceptable” refer to compounds, salts, compositions, dosage forms and other materials which are useful in preparing a pharmaceutical composition that is suitable for veterinary or human pharmaceutical use.
  • the salt may be any acid or base capable of forming a salt with the compound of Formula (I). It may be, for example, selected from a pharmaceutically acceptable salt or synthetically useful salt.
  • pharmaceutically acceptable salt of a given compound refers to salts that retain the biological effectiveness and properties of the given compound, and which are not biologically or otherwise undesirable.
  • “Pharmaceutically acceptable salts” or “physiologically acceptable salts” include, for example, salts with inorganic acids and salts with organic acids.
  • the free base can be obtained by basifying a solution of the acid salt.
  • an addition salt, particularly a pharmaceutically acceptable addition salt may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds.
  • Pharmaceutically acceptable acid addition salts may be prepared from inorganic and organic acids.
  • Salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Salts derived from organic acids include acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene-sulfonic acid, salicylic acid, and the like.
  • pharmaceutically acceptable base addition salts can be prepared from inorganic and organic bases.
  • Salts derived from inorganic bases include, by way of example only, sodium, potassium, lithium, ammonium, calcium and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary and tertiary amines, such as alkyl amines (i.e., NH2(alkyl)), dialkyl amines (i.e., HN(alkyl)2), trialkyl amines (i.e., N(alkyl) 3 ), substituted alkyl amines (i.e., NH2(substituted alkyl)), di(substituted alkyl) amines (i.e., HN(substituted alkyl)2), tri(substituted alkyl) amines (i.e., N(substituted alkyl) 3 ), alkenyl amines (i.e., NH2(alkyl)), dialkyl amine
  • dialkenyl amines i.e., HN(alkenyl)2
  • trialkenyl amines i.e.,
  • N(alkenyl)s substituted alkenyl amines (i.e., NH2(substituted alkenyl)), di(substituted alkenyl) amines (i.e., HN(substituted alkenyl ⁇ ), tri(substituted alkenyl) amines (i.e., N(substituted alkenyl) 3 , mono-, di- or tri- cycloalkyl amines (i.e., NH 2 (cycloalkyl), HN(cycloalkyl)2, N(cycloalkyl) 3 ), mono-, di- or tri- arylamines (i.e., NH2(aryl), HN(aryl)2, N(aryl)s), or mixed amines, etc.
  • substituted alkenyl amines i.e., NH2(substituted alkenyl)
  • Suitable amines include, by way of example only, isopropylamine, trimethyl amine, diethyl amine, tri(iso-propyl) amine, tri(n-propyl) amine, ethanolamine, 2-dimethylaminoethanol, piperazine, piperidine, morpholine, N-ethylpiperidine, and the like.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
  • the use of such media and agents for pharmaceutically active substances are known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Additional active ingredients can also be incorporated into the compositions.
  • therapeutically effective amount refers to an amount of the compound of Formula (I) that is sufficient to effect treatment as defined below, when administered to a patient (particularly a human) in need of such treatment in one or more doses.
  • the therapeutically effective amount will vary, depending upon the patient, the disease being treated, the weight and/or age of the patient, the severity of the disease, or the manner of administration as determined by a qualified prescriber or care giver.
  • treatment means administering a compound or pharmaceutically acceptable salt of Formula (I) for the purpose of: (i) delaying the onset of a disease, that is, causing the clinical symptoms of the disease not to develop or delaying the development thereof; (ii) inhibiting the disease, that is, arresting the development of clinical symptoms; and/or (iii) relieving the disease, that is, causing the regression of clinical symptoms or the severity thereof.
  • a method of treating and/or preventing liver disease in a patient in need thereof comprising administering to the patient a therapeutically effective amount of a compound of Formula (I).
  • the presence of active liver disease can be detected by the existence of elevated enzyme levels in the blood.
  • blood levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) above clinically accepted normal ranges are known to be indicative of on-going liver damage.
  • Routine monitoring of liver disease patients for blood levels of ALT and AST is used clinically to measure progress of the liver disease while on medical treatment. Reduction of elevated ALT and AST to within the accepted normal range is taken as clinical evidence reflecting a reduction in the severity of the patient's on- going liver damage.
  • the liver disease is a chronic liver disease.
  • Chronic liver diseases involve the progressive destruction and regeneration of the liver parenchyma, leading to fibrosis and cirrhosis.
  • chronic liver diseases can be caused by viruses (such as hepatitis B, hepatitis C, cytomegalovirus (CMV), or Epstein Barr Virus (EBV)), toxic agents or drugs (such as alcohol, methotrexate, or nitrofurantoin), a metabolic disease (such as non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), haemochromatosis, or Wilson's Disease), an autoimmune disease (such as Autoimmune Chronic Hepatitis, Primary Biliary Cholangitis (formerly known as Primary Biliary Cirrhosis), or Primary Sclerosing Cholangitis), or other causes (such as right heart failure).
  • viruses such as hepatitis B, hepatitis C, cytomegalovirus (CMV), or Epstein
  • cirrhosis is characterized pathologically by loss of the normal microscopic lobular architecture, with fibrosis and nodular regeneration.
  • the level of cirrhosis is reduced by about 5% to about 100%. In one embodiment, the level of cirrhosis is reduced by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100% in the subject.
  • the liver disease is a metabolic liver disease.
  • the liver disease is non-alcoholic fatty liver disease (NAFLD).
  • NAFLD is associated with insulin resistance and metabolic syndrome (obesity, combined hyperlipidemia, diabetes mellitus (type II) and high blood pressure). NAFLD is considered to cover a spectrum of disease activity, and begins as fatty accumulation in the liver (hepatic steatosis).
  • NAFLD has several other known causes.
  • NAFLD can be caused by certain medications, such as amiodarone, antiviral drugs (e.g., nucleoside analogues), aspirin (rarely as part of Reye's syndrome in children), corticosteroids, methotrexate, tamoxifen, or tetracycline.
  • NAFLD has also been linked to the consumption of soft drinks through the presence of high fructose corn syrup which may cause increased deposition of fat in the abdomen, although the consumption of sucrose shows a similar effect (likely due to its breakdown into fructose). Genetics has also been known to play a role, as two genetic mutations for this susceptibility have been identified.
  • NAFLD non-alcoholic steatohepatitis
  • NASH non-alcoholic steatohepatitis
  • liver fibrosis is the excessive accumulation of extracellular matrix proteins including collagen that occurs in most types of chronic liver diseases.
  • advanced liver fibrosis results in cirrhosis and liver failure.
  • Methods for measuring liver histologies such as changes in the extent of fibrosis, lobular hepatitis, and periportal bridging necrosis, are well known in the art.
  • treatment as described herein may improve a patient' s fibrosis from baseline, for example, improving from F4 to F3, F3 to F2, or F2 to Fl.
  • a patient's fibrosis score is improved by one or more following 24 weeks of daily treatment.
  • Liver disease can be classified into 4 stages: F0 indicates no fibrosis; Fl indicates mild fibrosis; F2 indicates moderate fibrosis; F3 indicates severe fibrosis; and F4 indicates cirrhosis.
  • F0 indicates no fibrosis
  • Fl indicates mild fibrosis
  • F2 indicates moderate fibrosis
  • F3 indicates severe fibrosis
  • F4 indicates cirrhosis.
  • “Fibrosis Score” refers to a scoring system for fibrosis as described by Kleiner et al. (Hepatology, Design and validation of a histological scoring system for nonalcoholic fatty liver disease (2005), 41: 1313-1321).
  • the level of liver fibrosis which is the formation of fibrous tissue, fibroid or fibrous degeneration, is reduced by more than about 90%. In one embodiment, the level of fibrosis, which is the formation of fibrous tissue, fibroid or fibrous degeneration, is reduced by at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30%, at least about 20%, at least about 10%, at least about 5% or at least about 2%.
  • the compounds provided herein reduce the level of fibrogenesis in the liver.
  • Liver fibrogenesis is the process leading to the deposition of an excess of extracellular matrix components in the liver known as fibrosis. It is observed in a number of conditions such as chronic viral hepatitis B and C, alcoholic liver disease, drug-induced liver disease, hemochromatosis, auto-immune hepatitis, Wilson disease, Primary Biliary Cholangitis (formerly known as Primary Biliary Cirrhosis), sclerosing cholangitis, liver schistosomiasis and others.
  • the level of fibrogenesis is reduced by more than about 90%.
  • the level of fibrogenesis is reduced by at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least 40%, at least about 30%, at least about 20%, at least about 10%, at least about 5% or at least about 2%.
  • provided herein is a method of treating and/or preventing primary sclerosing cholangitis (PSC) in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of Formula (I).
  • PSC primary sclerosing cholangitis
  • Cardiovascular diseases refer to any one or more than one of, for example, heart failure (including congestive heart failure, diastolic heart failure, systolic heart failure, heart failure with preserved ejection fraction), acute heart failure, ischemia, recurrent ischemia, myocardial infarction, arrhythmias, angina (including exercise-induced angina, variant angina, stable angina, unstable angina), acute coronary syndrome, diabetes, intermittent claudication, and idiopathic pulmonary fibrosis.
  • heart failure including congestive heart failure, diastolic heart failure, systolic heart failure, heart failure with preserved ejection fraction
  • acute heart failure ischemia, recurrent ischemia, myocardial infarction, arrhythmias
  • angina including exercise-induced angina, variant angina, stable angina, unstable angina
  • acute coronary syndrome diabetes, intermittent claudication, and idiopathic pulmonary fibrosis.
  • Also provided herein is a method of treating and/or preventing pulmonary vascular disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of Formula (I) or a salt, co-crystal, solvate, or hydrate thereof.
  • the pulmonary vascular disease is a pulmonary arterial hypertension (PAH).
  • PAH pulmonary arterial hypertension
  • the patient is diagnosed with Group 1, ⁇ , 1", 2, 3, 4, or 5 pulmonary hypertension.
  • a method of treating and/or preventing right ventricle dysfunction in a patient in need thereof comprising administering to the patient a therapeutically effective amount of an ASKl inhibitor.
  • Also provided herein is a method of treating, preventing, and/or reversing the narrowing or restricting of pulmonary arteries in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of an ASKl inhibitor. Additionally, provided herein is a method of reducing or normalizing high mean pulmonary arterial pressure (mPAP) and/or high pulmonary vascular resistance in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of an ASKl inhibitor. In one embodiment, the high mPAP > 25 mmHg at rest may be reduced to levels within the normal range at rest by the methods described herein.
  • mPAP mean pulmonary arterial pressure
  • the high mPAP > 25 mmHg at rest may be reduced to levels within the normal range at rest by the methods described herein.
  • the high mPAP > 25 mmHg at rest may be reduced to about 22 mmHg, 20 mmHg, 18 mmHg, 16 mmHg, or 14 mmHg at rest by the methods described herein.
  • mPAP is determined by right heart catheterization (RHC).
  • PAH symptoms include and are not limited to breathlessness or shortness of breath (dyspnea), fatigue, dizziness, fainting (syncope), swollen ankles and legs (edema), chest pain, right heart failure and/or dysfunction.
  • the improvement may be determined by a change from baseline in pulmonary vascular resistance (PVR), a change from baseline in cardiac index (CI) such as mean pulmonary artery pressure (mPAP), mean right atrial pressure (mRAP), mixed venous oxygen saturation (Sv02), and right ventricular cardiac power, a change from baseline in clinical measures of symptoms and function, including but not limited to submaximal exercise (6-minute walk test (6MWT)), heart rate recovery (HRR) after the 6MWT, the Borg dyspnea index, WHO Functional Class, N-terminal pro-brain natriuretic peptide, and/or quality of life by the SF-36 ® Health Survey.
  • PVR is determined by right heart catheterization.
  • cardiac function is determined by echocardiography or cardiac hemodynamic data. Additionally provided herein is a method of reducing the remodeling of pulmonary vasculature or arteries in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of an ASK1 inhibitor.
  • the right ventricle failure or dysfunction may be detected or monitored by cardiac imaging such as echocardiography and cardiac MRI.
  • hypertrophy, and/or RV function in a patient in need thereof comprising administering to the patient a therapeutically effective amount of an ASK1 inhibitor.
  • the present application provides a therapy or treatment to a patient in need, wherein the patient has or is suspected to have pulmonary vascular disease such as pulmonary hypertension or pulmonary arterial hypertension.
  • the patients experience one or more symptoms selected from breathlessness or shortness of breath (dyspnea), fatigue, dizziness, fainting (syncope), swollen ankles and legs (edema), or chest pain (e.g. angina).
  • the patients may be at various clinical or treatment stages, including patients who have not received any prior treatment to pulmonary hypertension or pulmonary arterial hypertension, patient who have received prior therapies or drugs for pulmonary hypertension or pulmonary arterial hypertension and remains
  • the patient may have received the therapeutics of the present application (e.g. the ASK1 inhibitor or a pharmaceutical composition thereof) and other PAH drugs concurrently.
  • the therapeutics of the present application e.g. the ASK1 inhibitor or a pharmaceutical composition thereof
  • other PAH drugs concurrently.
  • the treatment, prevention, reduction, reversion, and/or improvement by the method described herein may be determined by a change from baseline in pulmonary vascular resistance (PVR), a change from baseline in cardiac index (CI) such as mean pulmonary artery pressure (mPAP), mean right atrial pressure (mRAP), mixed venous oxygen saturation (Sv02), and right ventricular cardiac power, a change from baseline in clinical measures of symptoms and function, including but not limited to submaximal exercise (6-minute walk test (6MWT)), heart rate recovery (HRR) after the 6MWT, the Borg dyspnea index, WHO Functional Class, N-terminal pro-brain natriuretic peptide, an/or quality of life by the SF-36 ® Health Survey.
  • PVR pulmonary vascular resistance
  • CI cardiac index
  • mPAP mean pulmonary artery pressure
  • mRAP mean right atrial pressure
  • Sv02 mixed venous oxygen saturation
  • RV right ventricular cardiac power
  • PVR is determined by right heart catheterization.
  • cardiac function is determined by echocardiography or cardiac hemodynamic data.
  • the baseline refers to a value, number, or reading that is determined or measured from the subject prior to any treatment.
  • the baseline is a value, number, or reading from a patient prior to being treated with the methods described herein, from a healthy individual, from a group of subjects, or from suitable guidelines.
  • the baseline is a value, number, or reading from a patient prior to being treated with the methods described herein.
  • the baseline value or number may be determined or measured by any suitable methods.
  • RV dysfunction refers to the failure of right ventricle or right heart is unable to carry out the normal function (e.g. pumping blood out of the heart into the lungs to be replenished with oxygen, and/or maintaining sufficient blood flow to meet the needs of the body).
  • RV dysfunction may be determined or detected by cardiac imaging including echocardiography and cardiac MRI which characterizes structural changes (myocardial hypertrophy followed by progressive contractile dysfunction and chamber dilation) and/or functional changes (reduced fractional shortening, increased filling pressures, reduced right ventricular ejection fraction and decreased cardiac output). Other commonly used methods may also be used to determine or detect RV dysfunction.
  • promoting or “stimulating” refer to one or more factor that may cause or contribute to progressing of activity, disease, disorder, or condition.
  • promoting or contributing to PAH is used to describe one or more factor that may cause or contribute to progressing or developing of PAH.
  • kidney disease refers to progressive loss of kidney function over time typically months or even years.
  • Chronic kidney disease CKD
  • Chronic kidney disease includes by implication kidney disease.
  • diabetic kidney disease refers to kidney disease caused by diabetes, exacerbated by diabetes, or co- presenting with diabetes. It is a form of chronic kidney disease occurring in approximately 30% of patients with diabetes. It is defined as diabetes with the presence of albuminuria and/or impaired renal function (i.e. decreased glomerular filtration rate (See de B, I, et al. Temporal trends in the prevalence of diabetic kidney disease in the United States. JAMA 2011 Jun. 22; 305(24):2532-2539).
  • a compound of Formula (I), or salt thereof can be used to prevent or decrease the likelihood of transplant rejection.
  • an inflammatory disease such as Rheumatoid Arthritis is treated by administering a compound of Formula (I), or salt thereof.
  • systemic lupus erythematosus is treated by administering a compound of Formula (I), or salt thereof.
  • neuropathy such as HIV-associated neuropathy is treated by administering a compound of Formula (I), or salt thereof.
  • Also provided herein is a method of improving pathological consequence or outcome associated with oxidative stress in a patient in need thereof comprising administering to the patient a therapeutically effective amount of ASK1 inhibitor.
  • the compound of Formula (I) is combined with an additional active agent selected from an ACE inhibitor, Acetyl CoA carboxylase inhibitor, Adenosine A3 receptor agonist, Adiponectin receptor agonist, AKT protein kinase inhibitor AMP-activated protein kinase (AMPK) inhibitor, Amylin receptor agonist,
  • an additional active agent selected from an ACE inhibitor, Acetyl CoA carboxylase inhibitor, Adenosine A3 receptor agonist, Adiponectin receptor agonist, AKT protein kinase inhibitor AMP-activated protein kinase (AMPK) inhibitor, Amylin receptor agonist,
  • an additional active agent selected from an ACE inhibitor, Acetyl CoA carboxylase inhibitor, Adenosine A3 receptor agonist, Adiponectin receptor agonist, AKT protein kinase inhibitor AMP-activated protein kinase (AMPK) inhibitor, Amylin receptor agonist,
  • AMPK AMP-activated protein kinas
  • Angiotensin II AT-1 receptor antagonist Autotaxin inhibitors, Bioactive lipid, Calcitonin agonist, Caspase inhibitor, Caspase-3 stimulator, Cathepsin B inhibitor, Cathepsin K inhibitor, Cathepsin S inhibitor, Caveolin 1 inhibitor, CCR2 chemokine antagonist, CCR3 chemokine antagonist, CCR5 chemokine antagonist, Chloride channel stimulator, CNR1 inhibitor, Cyclin Dl inhibitor, Cytochrome P450 7A1 inhibitor, DGAT2 gene inhibitor, Dipeptidyl peptidase IV inhibitor, Eotaxin ligand inhibitor, Extracellular matrix protein modulator, Farnesoid X receptor agonist, FGF1 receptor agonist, FGF-19 ligand, Galectin-3 inhibitor, Glucagon receptor agonist, Glucagon-like peptide 1 agonist, G-protein coupled bile acid receptor 1 agonist, Hedgehog (Hh) modulators, Hepatitis C virus NS3 protease inhibitor, H
  • Phospholipase C inhibitor Phospholipase C inhibitor, PPAR alpha agonist, PPAR delta agonist, PPAR gamma agonist, PPAR gamma modulator, Protease- activated receptor-2 antagonist, Protein kinase modulator, Rho associated protein kinase 2 inhibitor, Sodium glucose transporter- 2 inhibitor, SREBP transcription factor inhibitor, STAT-1 inhibitor, Stearoyl CoA desaturase-1 inhibitor, Suppressor of cytokine signalling- 1 stimulator, Suppressor of cytokine signalling-3 stimulator, Transforming growth factor ⁇ (TGF- ⁇ ), Thyroid hormone receptor beta agonist, TLR-4 antagonist, Transglutaminase inhibitor, Tyrosine kinase receptor modulator, Unspecified GPCR modulator, Unspecified nuclear hormone receptor modulator, WNT modulators, YAP/TAZ modulators.
  • TGF- ⁇ TGF- ⁇
  • Thyroid hormone receptor beta agonist TLR-4 antagonist
  • Specific examples include A-4250, AC-3174, acetylsalicylic acid, AK-20, alipogene tiparvovec, aramchol, ARI-3037MO, atorvastatin, AZ compound (NASH/NAFLD), bertilimumab, Betaine anhydrous, BI-1467335, BMS-986036, BMT-053011, BOT-191, CAT-2003, cenicriviroc, CER-209, CF-102, CNX-014, CNX-023, CNX-024, CNX-025, cobiprostone, colesevelam, dapagliflozin, deuterated pioglitazone R-enantiomer 2,4- dinitrophenol, DRX-065, DS-102, dual acting glucagon-like peptide 1 receptor/glucagon receptor co-agonist, DUR-928, EDP-305, elafibranor, emricasan, enalapri
  • norursodeoxycholic acid O-304, obeticholic acid, olesoxime, PAT-505, PAT-048, peg- ilodecakin, pioglitazone, Px-102, PXS-4728A, PZ-235, RDX-009 program, remogliflozin etabonate, RG-125, saroglitazar, semaglutide, pumpuzumab, solithromycin, sotagliflozin, TCM-606F, TEV-45478, tipelukast (MN-001), TRX-318, UD-009, ursodeoxycholic acid, VBY-376, VBY-825, VK-2809, volixibat potassium ethanolate hydrate (SHP-626), VVP-100X, WAV-301, and ZGN-83.
  • an FXR agonist is administered or formulated with the compound of Formula (I) or salts and/or forms thereof.
  • the FXR agonist is a compound having the structure of Formula (III):
  • the FXR agonist is a compound having the structure of Formula (IV):
  • the compounds of Formula (III) and Formula (IV) may be synthesized and characterized using methods known to those of skill in the art, such as those described in U.S. Publication No. 9,139,539.
  • the compound of Formula (I) or a salt, co-crystal, solvate, or hydrate thereof is combined with an ACC inhibitor.
  • the ACC inhibitor is a compound Formula (V):
  • Cardiovascular related diseases or conditions that can benefit from a combination treatment of ASK1 inhibitors with other therapeutic agents include, without limitation, angina, including stable angina, unstable angina (UA), exercised-induced angina, variant angina, arrhythmias, intermittent claudication, myocardial infarction including non-STE myocardial infarction (NSTEMI), heart failure including congestive (or chronic) heart failure, acute heart failure, or recurrent ischemia.
  • angina including stable angina, unstable angina (UA), exercised-induced angina, variant angina, arrhythmias, intermittent claudication, myocardial infarction including non-STE myocardial infarction (NSTEMI), heart failure including congestive (or chronic) heart failure, acute heart failure, or recurrent ischemia.
  • Therapeutic agents suitable for treating cardiovascular related diseases or conditions include anti-anginals, heart failure agents, antithrombotic agents, antiarrhythmic agents, antihypertensive agents, and lipid lowering agents.
  • ASK1 inhibitors with therapeutic agents suitable for treating cardiovascular related conditions allows enhancement in the standard of care therapy the patient is currently receiving.
  • Anti-anginals include beta-blockers, calcium channel blockers, and nitrates. Beta blockers reduce the heart's need for oxygen by reducing its workload resulting in a decreased heart rate and less vigorous heart contraction.
  • beta-blockers include acebutolol (Sectral), atenolol (Tenormin), betaxolol (Kerlone),
  • bisoprolol/hydrochlorothiazide Ziac
  • bisoprolol Zebeta
  • carteolol Cartrol
  • esmolol Brevibloc
  • labetalol Normalne, Trandate
  • metoprolol Lipressor
  • Toprol XL nadolol
  • nadolol Corgard
  • propranolol Inderal
  • sotalol Badore
  • timolol Blocadren
  • Nitrates dilate the arteries and veins thereby increasing coronary blood flow and decreasing blood pressure.
  • examples of nitrates include nitroglycerin, nitrate patches, isosorbide dinitrate, and isosorbide-5 -mononitrate.
  • Calcium channel blockers prevent the normal flow of calcium into the cells of the heart and blood vessels causing the blood vessels to relax thereby increasing the supply of blood and oxygen to the heart.
  • Examples of calcium channel blockers include amlodipine (Norvasc, Lotrel), bepridil (Vascor), diltiazem (Cardizem, Tiazac), felodipine (Plendil), nifedipine (Adalat, Procardia), nimodipine (Nimotop), nisoldipine (Sular), verapamil (Calan, Isoptin, Verelan), and nicardipine.
  • Diuretics eliminate excess fluids in the tissues and circulation thereby relieving many of the symptoms of heart failure.
  • diuretics include hydrochlorothiazide, metolazone (Zaroxolyn), furosemide (Lasix), bumetanide (Bumex), spironolactone (Aldactone), and eplerenone (Inspra).
  • Angiotensin converting enzyme (ACE) inhibitors reduce the workload on the heart by expanding the blood vessels and decreasing resistance to blood flow.
  • ACE inhibitors include benazepril (Lotensin), captopril (Capoten), enalapril
  • Vasodilators reduce pressure on the blood vessels by making them relax and expand.
  • vasodilators include hydralazine, diazoxide, prazosin, clonidine, and methyldopa.
  • ACE inhibitors, nitrates, potassium channel activators, and calcium channel blockers also act as vasodilators.
  • Cardiac glycosides are compounds that increase the force of the heart's contractions. These compounds strengthen the pumping capacity of the heart and improve irregular heartbeat activity. Examples of cardiac glycosides include digitalis, digoxin, and digitoxin.
  • Antithrombotics inhibit the clotting ability of the blood.
  • platelet inhibitors inhibit the clotting activity of platelets, thereby reducing clotting in the arteries.
  • platelet inhibitors include acetyls alicylic acid (aspirin), ticlopidine, clopidogrel (plavix), dipyridamole, cilostazol, persantine sulfinpyrazone, dipyridamole, indomethacin, and glycoprotein llb/llla inhibitors, such as abciximab, tirofiban, and eptifibatide (Integrelin).
  • Beta blockers and calcium channel blockers also have a platelet- inhibiting effect.
  • Anticoagulants prevent blood clots from growing larger and prevent the formation of new clots.
  • anticoagulants include bivalirudin (Angiomax), warfarin (Coumadin), unfractionated heparin, low molecular weight heparin, danaparoid, lepirudin, and argatroban.
  • Thrombolytic agents act to break down an existing blood clot.
  • examples of thrombolytic agents include streptokinase, urokinase, and tenecteplase (TNK), and tissue plasminogen activator (t-PA).
  • Antiarrhythmic agents are used to treat disorders of the heart rate and rhythm.
  • antiarrhythmic agents include amiodarone, quinidine, procainamide, lidocaine, and propafenone.
  • Cardiac glycosides and beta blockers are also used as antiarrhythmic agents.
  • Antihypertensive agents are used to treat hypertension, a condition in which the blood pressure is consistently higher than normal. Hypertension is associated with many aspects of cardiovascular disease, including congestive heart failure, atherosclerosis, and clot formation.
  • antihypertensive agents include alpha- 1 -adrenergic antagonists, such as prazosin (Minipress), doxazosin mesylate (Cardura), prazosin hydrochloride (Minipress), prazosin, polythiazide (Minizide), and terazosin hydrochloride (Hytrin); beta-adrenergic antagonists, such as propranolol (Inderal), nadolol (Corgard), timolol (Blocadren), metoprolol (Lopressor), and pindolol (Visken); central alpha-adrenoceptor agonists, such as clonidine hydrochloride (Catapres), clonidine hydrochloride and chlorthalidone (Clorpres, Combipres), guanabenz Acetate (Wytensin), guanfacine hydrochloride (Tenex), methyldopa (Aldomet), a
  • alpha/be ta- adrenergic antagonists such as labetalol (Normodyne, Trandate), Carvedilol (Coreg); adrenergic neuron blocking agents, such as guanethidine (Ismelin), reserpine (Serpasil); central nervous system-acting antihypertensives, such as clonidine (Catapres), methyldopa (Aldomet), guanabenz (Wytensin); anti- angiotensin II agents; ACE inhibitors, such as perindopril (Aceon) captopril (Capoten), enalapril (Vasotec), lisinopril (Prinivil, Zestril); angiotensin- II receptor antagonists, such as Candesartan (Atacand), Eprosartan
  • hydralazine Presoline
  • minoxidil Lithiten
  • verapamil hydralazine
  • potassium channel activators such as aprikalim, bimakalim, cromakalim, emakalim, nicorandil, and pinacidil.
  • Lipid lowering agents are used to lower the amounts of cholesterol or fatty sugars present in the blood.
  • lipid lowering agents include bezafibrate (Bezalip), ciprofibrate (Modalim), and statins, such as atorvastatin (Lipitor), fluvastatin (Lescol), lovastatin (Mevacor, Altocor), mevastatin, pitavastatin (Livalo, Pitava) pravastatin (Lipostat), rosuvastatin (Crestor), and simvastatin (Zocor).
  • statins such as atorvastatin (Lipitor), fluvastatin (Lescol), lovastatin (Mevacor, Altocor), mevastatin, pitavastatin (Livalo, Pitava) pravastatin (Lipostat), rosuvastatin (Crestor), and simvastatin (Zocor).
  • a subject in need of the ASK1 inhibitor often suffers from secondary medical conditions such as one or more of a metabolic disorder, a pulmonary disorder, a peripheral vascular disorder, or a gastrointestinal disorder.
  • secondary medical conditions such as one or more of a metabolic disorder, a pulmonary disorder, a peripheral vascular disorder, or a gastrointestinal disorder.
  • Such patients can benefit from treatment of a combination therapy comprising administering to the patient the compounds of the invention in combination with at least one therapeutic agent.
  • Pulmonary disorder refers to any disease or condition related to the lungs.
  • pulmonary disorders include, without limitation, asthma, chronic obstructive pulmonary disease (COPD), bronchitis, and emphysema.
  • COPD chronic obstructive pulmonary disease
  • bronchitis bronchitis
  • emphysema emphysema
  • therapeutics agents used to treat pulmonary disorders include bronchodilators including beta2 agonists and anticholinergics, corticosteroids, and electrolyte supplements.
  • Specific examples of therapeutic agents used to treat pulmonary disorders include epinephrine, terbutaline (Brethaire, Bricanyl), albuterol (Proventil), salmeterol (Serevent, Serevent Diskus), theophylline, ipratropium bromide (Atrovent), tiotropium (Spiriva), methylprednisolone (Solu-Medrol, Medrol), magnesium, and potassium.
  • metabolic disorders include, without limitation, diabetes, including type I and type II diabetes, metabolic syndrome, dyslipidemia, obesity, glucose intolerance, hypertension, elevated serum cholesterol, and elevated triglycerides.
  • therapeutic agents used to treat metabolic disorders include antihypertensive agents and lipid lowering agents.
  • Additional therapeutic agents used to treat metabolic disorders include insulin, sulfonylureas, biguanides, alpha-glucosidase inhibitors, and incretin mimetics.
  • Peripheral vascular disorders are disorders related to the blood vessels (arteries and veins) located outside the heart and brain, including, for example peripheral arterial disease (PAD), a condition that develops when the arteries that supply blood to the internal organs, arms, and legs become completely or partially blocked as a result of atherosclerosis.
  • PAD peripheral arterial disease
  • Gastrointestinal disorders refer to diseases and conditions associated with the gastrointestinal tract. Examples of gastrointestinal disorders include gastroesophageal reflux disease (GERD), inflammatory bowel disease (IBD), gastroenteritis, gastritis and peptic ulcer disease, and pancreatitis.
  • GFD gastroesophageal reflux disease
  • IBD inflammatory bowel disease
  • pancreatitis pancreatitis
  • therapeutic agents used to treat gastrointestinal disorders include proton pump inhibitors, such as pantoprazole (Protonix), lansoprazole (Prevacid), esomeprazole (Nexium), omeprazole (Prilosec), rabeprazole; H2 blockers, such as cimetidine (Tagamet), ranitidine (Zantac), famotidine (Pepcid), nizatidine (Axid);
  • proton pump inhibitors such as pantoprazole (Protonix), lansoprazole (Prevacid), esomeprazole (Nexium), omeprazole (Prilosec), rabeprazole
  • H2 blockers such as cimetidine (Tagamet), ranitidine (Zantac), famotidine (Pepcid), nizatidine (Axid);
  • prostaglandins such as misoprostol (Cytotec); sucralfate; and antacids.
  • a compound of Formula (I) or a salt, co-crystal, solvate, or hydrate thereof as disclosed herein may be used in combination with one or more additional therapeutic agent that are being used and/or developed to treat gastrointestinal disorders.
  • a compound of Formula (I) or a salt, co-crystal, solvate, or hydrate thereof disclosed herein may be used in combination with one or more additional therapeutic agent that are being used and/or developed to treat inflammatory disorders (e.g., IBD).
  • the one or more additional therapeutic agent may be a ⁇ 4 ⁇ 7 inhibitor, a steroid, a MMP-9 antibody, a S1P1 agonist, a TNF biologic, or any combination thereof.
  • the one or more additional therapeutic agent may be a ⁇ 4 ⁇ 7 integrin inhibitor, or an agent that inhibits the expression and/or activity of ⁇ 4 ⁇ 7 integrin.
  • the inhibitor can be small molecule or biologic.
  • the ⁇ 4 ⁇ 7 integrin inhibitor can be natalizumab or vedolizumab.
  • the one or more additional therapeutic agent may be a steroid, including but not limited to, corticosteroids.
  • Corticosteroids may be administered by various routes, including intravenously (i.e., methylprednisolone, hydrocortisone), orally (i.e., prednisone, prednisolone, budesonide, dexamethasone), or topically (i.e., enema, suppository, or foam preparations).
  • the one or more additional therapeutic agent may be an MMP9 inhibitor, or an agent that inhibits the expression and/or activity of MMP9.
  • a representative protein sequence for MMP9 is GenBank Accession No. NP_004985.
  • the inhibitor can be small molecule or biologic.
  • SB-3CT CAS 292605-14-2
  • siRNA, antisense RNA and antibodies have also been demonstrated to inhibit the expression or activity of MMP9 and are within the scope of the present disclosure.
  • an MMP9 inhibitor is a monoclonal anti- MMP9 antibody.
  • the one or more additional therapeutic agent includes an MMP9 inhibitor and a nucleoside analog such as gemcitabine.
  • the one or more additional therapeutic agent may be a
  • S1P1 Sphingosine 1 -Phosphate Receptor
  • the inhibitor can be small molecule or biologic.
  • the S1P1 inhibitor can be RPC1063.
  • the one or more additional therapeutic agent may be a TNF inhibitor, or an agent that inhibits the expression and/or activity of TNF.
  • the inhibitor can be small molecule or biologic.
  • the TNF inhibitor can be golimumab.
  • the one or more additional therapeutic agent is being used and/or developed to treat ulcerative colitis (UC) and/or Crohn disease (CD).
  • the agent can be a biologic or small molecule.
  • the agent is a modulator
  • the JAK inhibitor is filgotinib.
  • Non- limiting examples of agents being used and/or developed to treat ulcerative colitis include GSK3050002 (CCL20 modulator, by GSK), GS-5745 (MMP9 modulator, by Gilead), AVX-470 (TNF modulator, by Avaxia), Bertilimumab (Eotaxin modulator, by Immune Pharma), Simponi (TNF modulator, by Johnson & Johnson and Merck), RX-10001 (by Resolvyx), IBD-98 (5-ASA modulator, by Holy Stone), SP-333 (GC modulator, by Synergy), KAG-308 (EP4 modulator, by Kaken), SB012 (GATA-3 modulator, by Sterna), AJM300 ( ⁇ x4 modulator, by Ajinomoto), BL-7040 (TLR9 modulator, by BiolineRx), TAK-114 (SAT3 modulator, by Takeda), CyCol (by Sigmoid), GWP-42003 (CB modulator, by GW Pharma), ASP
  • ASA modulators such as Delzicol (by Actavis), Canasa (by Aptalis), Asacol (by Actavis), Pentasa (by Shire/Ferring), Lialda (by Shire), Mezavant (by Shire), Apriso (by Salix), Colazal (by Salix), Giazo (by Salix), and Salofalk (by Dr Falk).
  • agents being used and/or developed to treat Crohn disease (CD) include
  • FFP102 CD40 modulator, by Fast Forward
  • E6011 CX3CL1 modulator, by Eisai
  • PF- 06480605 by Pfizer
  • QBECO SSI Immunomod modulator, by Qu Biologies
  • PDA- 001 by Celgene
  • BI 655066 IL-12/IL-23 modulator, by Boehringer
  • TNFa kinoid TNF modulator, by Neovacs
  • AMG 139/MEDI-2070 IL-12/IL-23 modulator, by AstraZeneca
  • PF-04236921 IL-6 modulator, by Pfizer
  • Tysabri ⁇ 7 modulator, marketed by Biogen pou in the U.S.
  • Cimzia marketed by UCB in the U.S.
  • JNJ- 40346527 FMS modulator, by J&J
  • SGX-203 StepX-203
  • StepX-203 StepX-203
  • StepX-203 StepX-203
  • CyCron by
  • Non-limiting examples of agents being used and/or developed to treat ulcerative colitis (UC) and Crohn disease (CD) include PF-06410293 (by Pfizer), SAN-300 (VLA- 1 modulator, by Salix), SAR252067 (LIGHT modulator, by Sanofi), PF-00547659
  • the one or more additional therapeutic agent may be a JAK inhibitor, particularly a JAK- 1 selective inhibitor.
  • the inhibitor can be small molecule or biologic.
  • the JAK inhibitor can be Filgotinib, GLPG0634 (JAK modulator, by Galapagos).
  • Patients presenting with an acute coronary disease event may exhibit conditions that benefit from administration of therapeutic agent or agents that are antibiotics, analgesics, antidepressant and anti-anxiety agents in combination with ranolazine.
  • Antibiotics are therapeutic agents that kill, or stop the growth of,
  • antibiotic agents include ⁇ -Lactam antibiotics, including penicillins (amoxicillin), cephalosporins, such as cefazolin, cefuroxime, cefadroxil (Duricef), cephalexin (Keflex), cephradine (Velosef), cefaclor (Ceclor), cefuroxime axtel (Ceftin), cefprozil (Cefzil), loracarbef (Lorabid), cefixime (Suprax), cefpodoxime proxetil (Vantin), ceftibuten (Cedax), cefdinir
  • penicillins amoxicillin
  • cephalosporins such as cefazolin, cefuroxime, cefadroxil (Duricef), cephalexin (Keflex), cephradine (Velosef), cefaclor (Ceclor), cefuroxime axtel (Ceftin), cefprozil
  • tetracyclines such as tetracycline
  • macrolide antibiotics such as erythromycin
  • aminoglycosides such as gentamicin, tobramycin, amikacin
  • quinolones such as ciprofloxacin
  • cyclic peptides such as vancomycin, streptogramins, polymyxins
  • lincosamides such as clindamycin
  • oxazolidinoes such as linezolid
  • sulfa antibiotics such as sulfisoxazole.
  • Analgesics are therapeutic agents that are used to relieve pain.
  • Examples of analgesics include opiates and morphinomimetics, such as fentanyl and morphine; paracetamol; NSAIDs, and COX-2 inhibitors.
  • Antidepressant and anti-anxiety agents include those agents used to treat anxiety disorders, depression, and those used as sedatives and tranquilzers.
  • Examples of antidepressant and anti-anxiety agents include benzodiazepines, such as diazepam, lorazepam, and midazolam; enzodiazepines; barbiturates; glutethimide; chloral hydrate; meprobamate; sertraline (Zoloft, Lustral, Apo-Sertral, Asentra, Gladem, Serlift,
  • escitalopram (Lexapro, Cipralex); fluoxetine (Prozac, Sarafem, Fluctin, Fontex, Prodep, Fludep, Lovan); venlafaxine (Effexor XR, Efexor); citalopram (Celexa, Cipramil, Talohexane); paroxetine (Paxil, Seroxat, Aropax); trazodone (Desyrel);
  • amitriptyline (Elavil); and bupropion (Wellbutrin, Zyban).
  • ACE angiotensin converting enzyme
  • ARB angiontesin II receptor blockers
  • antihypertensive agents such as amlodipine, nifedipine, and felodipine.
  • the benefit of combination may be increased efficacy and/or reduced side effects for a component as the dose of that component may be adjusted down to reduce its side effects while benefiting from its efficacy augmented by the efficacy of Formula (I) or a salt, co- crystal, solvate, or hydrate thereof.
  • Patients presenting with chronic kidney disease treatable with ASKl inhibitors such as a compound of Formula (I) may also exhibit conditions that benefit from co- administration (as directed by a qualified caregiver) of a therapeutic agent or agents that are antibiotic, analgesic, antidepressant and/or anti-anxiety agents in combination with a compound of Formula (I).
  • Combination treatments may be administered simultaneously or one after the other within intervals as directed by a qualified caregiver or via a fixed dose (all active ingredients are combined into a single dosage form e.g. tablet) presentation of two or more active agents.
  • Coronary patients being treated for an acute cardiovascular disease event by administration of ASKl inhibitors often exhibit diseases or conditions that benefit from treatment with other therapeutic agents. These diseases or conditions can be of the cardiovascular nature or can be related to pulmonary disorders, metabolic disorders, gastrointestinal disorders and the like. Additionally, some coronary patients being treated for an acute cardiovascular disease event by administration of an ASKl inhibitor exhibit conditions that can benefit from treatment with therapeutic agents that are antibiotics, analgesics, and/or antidepressants and antianxiety agents.
  • compositions While it is possible for the active ingredients to be administered alone it may be preferable to present them as pharmaceutical formulations (compositions).
  • compositions both for veterinary and for human use, of the invention comprise at least one active ingredient, as above defined, together with one or more acceptable carriers therefor and optionally other therapeutic ingredients.
  • the carrier(s) must be
  • the formulations include those suitable for the foregoing administration routes.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, PA). Such methods include the step of bringing into association the active ingredient with inactive ingredients (e.g., a carrier, pharmaceutical excipient, etc.) which constitutes one or more accessory ingredients.
  • inactive ingredients e.g., a carrier, pharmaceutical excipient, etc.
  • the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • formulations suitable for oral administration are presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient.
  • the pharmaceutical formulations include one or more compounds of the invention together with one or more pharmaceutically acceptable carriers or excipients and optionally other therapeutic agents.
  • Pharmaceutical formulations containing the active ingredient may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation.
  • Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable.
  • excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as cellulose, microcrystalline cellulose, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc.
  • inert diluents such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate
  • granulating and disintegrating agents such as maize starch, or alginic acid
  • binding agents such as cellulose, microcrystalline cellulose, starch,
  • Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
  • a dosage form for oral administration to humans contains approximately 1 to 1000 mg of active material formulated with an appropriate and convenient amount of carrier material (e.g., inactive ingredient or excipient material).
  • carrier material e.g., inactive ingredient or excipient material.
  • the carrier material varies from about 5 to about 95% of the total compositions (weight: weight).
  • the pharmaceutical compositions described herein contain about 1 to about 800 mg, about 1 to 600 mg, about 1 to 400 mg, about 1 to 200 mg, about 1 to about 100 mg or about 1 to about 50 mg of the compound of Formula (I), or a pharmaceutically acceptable salt, co-crystal, solvate, or hydrate thereof. In some embodiments, the pharmaceutical compositions described herein contain no more than about 400 mg of the compound of Formula (I). In some embodiments, the pharmaceutical compositions described herein contain about 100 mg of the compound of Formula (I), or a salt, co- crystal, solvate, or hydrate thereof.
  • formulations disclosed herein may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
  • compositions comprising at least one active ingredient as above defined together with a veterinary carrier are further provided.
  • Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered orally, parenterally or by any other desired route.
  • Effective dose of active ingredient depends at least on the nature of the condition being treated, toxicity, whether the compound is being used prophylactically (lower doses), the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies.
  • One or more compounds of Formula (I) are administered by any route appropriate to the condition to be treated. Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), and the like. It will be appreciated that the preferred route may vary with for example the condition of the recipient.
  • compositions described herein are oral dosage forms.
  • the pharmaceutical compositions described herein are oral solid dosage forms.
  • the oral daily dosage of Formula (I) (as a free base) is between about 0.5 mg and about 100 mg as a daily dose.
  • the dose is about 1 mg daily, about 2 mg daily, about 3 mg daily, about 4 mg daily, about 5 mg daily, about 6 mg daily, about 7 mg daily, about 8 mg daily, about 9 mg daily, about 10 mg daily, about 11 mg daily, about 12 mg daily, about 13 mg daily, about 14 mg daily, about 15 mg daily, about 16 mg daily, about 17 mg daily, about 18 mg daily, about 19 mg daily, about 20 mg daily, about 25 mg daily, about 30 mg daily, or about 35 mg daily.
  • the daily dose of the free base of Formula (I) is less than about 18 mg daily.
  • DSC thermograms were collected on a TA Instruments Q2000 system equipped with a 50 position auto-sampler. The calibration for energy and temperature was carried out using certified indium. Typically 1 - 5 mg of each sample, in a pin-holed aluminium pan, was heated at 10 °C/min from 25 °C to 250 °C or above.
  • TGA thermograms were collected on a TA Instruments Q5000 or Q500. Typically 1 - 5 mg of each sample in, an open aluminium pan, was heated at 10 °C/min from 25 °C to 250 °C or above.
  • the DSC values may vary by up to about 3%, or about 2% or about 1%.
  • MeMgX Methylmagnesium halide (Grignard reagent), where X is Fluoro, Chloro, Bromo or lodo
  • a compound of Formula (I) may be prepared by reacting compound (6) with a suitable chlorinating reagent, including but not limited to, oxalyl chloride or thionyl chloride, to produce the corresponding acid chloride.
  • a suitable chlorinating reagent including but not limited to, oxalyl chloride or thionyl chloride.
  • Suitable reagents are known to those of ordinary skill in the art.
  • Subsequent amide bond formation between the acid chloride and compound (12) gives a compound of Formula (I).
  • Suitable reagents to facilitate amide bond formation are known to those of skill in the art.
  • the reaction was stirred for about 30 minutes at a temperature of about 45 °C, then cooled to room temperature, and a solution of 20% wt/wt sodium sulfite in water (6 mL) was slowly added. The mixture was stirred for 30 minutes and then solvents were removed under reduced pressure. The residue was suspended in 250 mL of water and 4N aqueous ammonium hydroxide was added to bring the pH to -10. The mixture was extracted with dichloromethane (3 x 200 mL), the organics combined, dried over magnesium sulfate, and the solvent was removed under reduced pressure.
  • reaction mixture was cooled to about 0 °C and then N,N-diisopropylethylamine (0.6 equiv) was charged followed by (S)-6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-amine (12) (1.1 equiv) which was charged slowly.
  • the reaction mixture was warmed to about 22 °C and maintained until the reaction was complete.
  • the reaction was concentrated to 5 L/kg and to this was then charged l-methyl-2-pyrrolidinone (10 L/kg).
  • the reaction was concentrated to 10 L/kg followed by a temperature adjustment to about 65 °C.
  • a solution of citric acid (0.5 equiv) and water (10 L/kg) was charged maintaining a temperature above about 55 °C.
  • the pH was adjusted to about 6 using 50 wt% aqueous sodium hydroxide.
  • the contents were heated at about 65 °C for about 1 h and then the reaction mixture was cooled to about 20 °C.
  • the product was isolated by filtration, washed with water (2 x 5 L/kg), and dried to afford the compound of Formula (I).
  • Formula (I) freebase Form I was crystallized from the reaction mixture using the following procedure: 1.0 equivalent of oxalyl chloride was added to compound (6) and the mixture was agitated at about 35 °C. The solution was then distilled to dryness, and 15 volumes of DCM were added. 0.7 equivalents of DIPEA followed by 1.1 equivalents of compound (12) were then added. The mixture was agitated at about 22 °C. 10 volumes of NMP were then charged to the reactor and the DCM was distilled down to a 15 wt% level. One equivalent of sodium citrate dissolved in 2 volumes of water was then added to the reactor, at about 22 °C. Three volumes of a IN NaOH solution and 5 volumes of water were then added. The solution was then aged overnight.
  • FIG. 1 is the DSC thermogram of Formula (I) freebase Form I. There is a melting transition with an onset of about 223 °C.
  • FIG. 3 is the TGA thermogram of the compound of Formula (I) freebase Form I. There is a 0.9 % weight loss between 25 and 220 °C.
  • Formula (I) freebase Form II was crystallized from the reaction mixture using the following procedure: 1.0 equivalent of oxalyl chloride was added to compound (6) and the mixture was agitated at about 35 °C. The solution was then distilled to 5 volumes and 10 volumes of DCM were added. 0.6 equivalents of DIPEA were added, followed by 1.1 equivalents of compound (12). The mixture was agitated at about 22 °C and the DCM was distilled to dryness. 10 volumes of NMP were then added. The content of the reactor was then heated to about 70 °C and a solution of 0.5 equiv. sodium citrate, 1.5 equivalents of NaOH, and water were added at a rate allowing the internal temperature of the solution to remain above about 58 °C. The mixture was then held at about 65 °C for about 1 h, and cooled to about 20 °C over 3 h. The resulting slurry was then filtered and rinsed with two times 5 volumes of water. The solid was then dried.
  • Formula (I) freebase Form II can also be obtained by slurrying Formula (I) freebase Form I in a variety of solvents, including ethanol.
  • FIG. 4 shows the XRPD pattern of Formula (I) freebase Form II. It contains the following characteristic peaks: (1) 8.7, 10.0, and 17.0 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (2) 13.9, 21.3, and 22.8 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (3) 24.0, 28.0, and 29.1 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • FIG. 5 is the DSC thermogram of Formula (I) freebase Form II. It shows a melting endotherm that has an onset at about 208 °C.
  • FIG. 6 is the TGA thermogram of Formula (I) freebase Form II. The solid loses about 1% weight upon heating to 200 °C.
  • FIG. 8 shows the DSC thermogram of Formula (I) succinate Form I.
  • the solid has a melting onset at approximately 159 °C.
  • FIG. 9 shows the TGA thermogram of Formula (I) succinate Form I. The solid loses about 0.6% weight between ambient and approximately 160 °C.
  • Formula (I) succinate Form II is a methanol solvate. It was first obtained by slurrying Formula (I) succinate Form I in pure methanol at about 22 °C for
  • FIG. 10 shows the XRPD pattern of Formula (I) succinate Form II. The following peaks are characteristic of this pattern: (1) 9.8, 11.7, 23.3 (2) 15.9, 21.5, 32.0 (3) 16.3, 20.8, 24.8 °2 ⁇ .
  • FIG. 11 shows the DSC thermogram of Formula (I) succinate Form II. It shows one endothermic event with an onset at about 100 °C, followed by two consecutive endothermic events with onsets at about 159, and about 162 °C.
  • FIG. 12 is the TGA thermogram of Formula (I) succinate Form II. There are two weight losses of 4.6 and 9.2 % weight at about 100 and 200 °C, respectively.
  • Formula (I) HCl Form I was first prepared by adding one equivalent of concentrated HCl to 47 mg of Formula (I) freebase Form II suspended in acetonitrile. The resulting solution was stirred at approximately 22°C for about 16 h.
  • FIG. 13 is the DSC thermogram of Formula (I) HCl Form I. The melt/dehydration occurs around 152 °C.
  • FIG. 15 is the TGA thermogram of Formula (I) HCl Form I. There are two weight losses , the first one is about 4.2 % weight, and occurs between about 30 °C and 140 °C. The second one is about 1.7 % weight and takes place between about 140 and 200 °C.
  • Formula (I) HCl Form II was first prepared by slurrying a mixture of Formula (I) HCl Form I and Material A (about 1:1 w/w) in 10:90 water: acetonitrile (v/v) for 16 h at approximately 22 °C. After 16h the XRPD of the centrifuged and air-dried solid was collected.
  • Formula (I) HCl Form II was also prepared by slurrying Formula (I) HCl Form I in 10:90 water: acetonitrile (v/v), centrifuging it and drying it in the vacuum oven at about 50 °C.
  • FIG. 16 shows the XRPD pattern of Formula (I) HCl Form II. It contains the following characteristic peaks: (1) 8.3, 12.0, and 26.0 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (2) 19.3, 21.7, and 24.0 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (3) 9.8, 15.4, and 28.9 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • FIG. 33 is the DSC thermogram of Formula (I) HCl Form II, in which there are two endothermic events with onsets at about 135 and about 205 °C.
  • Formula (I) HC1 Material A was first prepared by adding one equivalent of 1M HCl/dioxane to Formula (I) freebase Form II suspended in acetonitrile. The resulting solution was stirred at approximately 22°C for about 16 h and XRPD of the centrifuged and dried solid was collected.
  • FIG. 17 shows the XRPD pattern of Formula (I) HC1 Form III. It contains the following characteristic peaks: (1) 6.6, 12.9, and 19.7 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (2) 20.4, 23.4, and 24.8 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (3) 18.1, 25.4, and 26.7
  • FIG. 18 is the DSC thermogram of Formula (I)-HC1 Form III. No clear thermal events can be seen.
  • Formula (I) L-malate was first obtained by slurrying about 40 mg of Formula (I) freebase Form II in 0.2 mL of acetonitrile with one equivalent of L-malic acid, about 12 mg, for approximately 16 h at about 22 °C. The slurry was then centrifuged and an XRPD pattern of the dry solid was collected.
  • FIG. 19 shows the XRPD pattern of Formula (I) L-malate. It contains the following characteristic peaks: (1) 7.2, 21.9, and 24.5 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (2) 10.9, 16.6, and 19.7 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (3) 8.2, 22.8, and 27.3 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • FIG. 20 shows the DSC thermogram of Formula (I) L-malate. There are two endothermic events, the first one around 23 °C, and the second one, the melting endotherm, around 110 °C.
  • FIG. 21 shows the TGA thermogram of Formula (I) L-malate. The solid loses about 2.5 % weight when heated to about 150°C
  • Table 8 Formula (I) L-malate XRPD peak table
  • Formula (I) phosphate Material A was obtained by slurrying about 37 mg of Formula (I) freebase Form II in 0.2 mL of acetonitrile and adding one equivalent of 14.8 M phosphoric acid. After stirring for 16 h at approximately 22 °C, the solution was rotavaped and 0.2 mL of IPA was added. This solution was stirred for 10 days. A slurry was obtained and the XRPD of the centrifuged and dried solid was collected.
  • the XRPD pattern of Formula (I) phosphate Material A is shown in FIG. 22, it has the following characteristic peaks: (1) 6.9, 8.0, and 21.2 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (2) 22.8, 25.3, and 26.0 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (3) 19.8, 20.6, 23.2 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • Formula (I) phosphate Material B was obtained by placing about 1 mg of Formula (I) phosphate Material A on an aluminum pan and submitting it to the following relative humidity conditions inside the Dynamic Vapor Sorption (DVS) instrument: equilibrate at 25 °C and 10 % RH for 20 min, ramp to 90% RH and hold for 30 min.
  • the XRPD pattern of the solid was collected after the DVS cycle.
  • FIG. 23 shows the XRPD pattern of Formula (I) phosphate Material B.
  • Formula (I) sulfate was first obtained by adding 1 equivalent of 98 % sulfuric acid to 38 mg of Formula (I) freebase Form II in 0.2 mL of acetonitrile. The solution was stirred for about 16 h at approximately 22 °C, then rotavaped. 0.2 mL of IPA was added and the solution was stirred for about 16 h. The slurry was then centrifuged and XRPD of the solid dried in the vacuum oven at about 50 °C was obtained.
  • FIG. 24 The XRPD pattern of Formula (I) sulfate is presented in FIG. 24. It has the following characteristic peaks: (1) 7.2, 13.5, and 21.5 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (2) 16.9, 20.5, and 25.3 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (3) 18.2, 20.6, and 27.2 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • FIG. 25 shows the DSC thermogram of Formula (I) sulfate. The solid melts at approximately 210 °C.
  • FIG. 26 is the TGA thermogram of Formula (I) sulfate. It shows a weight loss of about 1.8 % around 225 °C.
  • Formula (I) citrate was obtained by slurrying about 42 mg of Formula (I) freebase Form II and one equivalent of citric acid, about 18 mg, in 0.2 mL of acetonitrile at about 22 °C for approximately 16 h.
  • the XRPD pattern of the centrifuged wet solid was obtained.
  • FIG. 27 shows the XRPD pattern of Formula (I) citrate Material A.
  • the pattern has the following characteristic peaks: (1) 5.6, 9.1, 16.7 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (2)12.2, 21.0, 23.0 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (2) 18.7, 24.9, 25.8 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • Formula (I) citrate Material ⁇ was obtained by drying Formula (I) citrate Material A in the vacuum oven at about 50 °C.
  • the XRPD pattern of Formula (I) citrate Material B is shown in FIG. 28.
  • FIG. 29 shows the DSC
  • thermogram of Formula (I) citrate Material B There are two thermal events, with onsets at about 54 and about 122 °C.
  • TczWe 12b Formula (I) Citrate Material B XRPD Peak Table No. Pos. [°2Th.] Rel. Int. [%]
  • Formula (I) glutarate was first prepared by stirring together 55.1 mg of Formula (I) freebase Form II and one equivalent of glutaric acid, approximately 17 mg, in 0.2 mL of acetonitrile for about 16 h at approximately 22 °C.
  • FIG. 30 is the XRPD pattern of Formula (I) glutarate. It contains the following characteristic peaks: (1) 6.7, 8.0, and 12.1 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (2) 14.6, 20.9, and 21.8 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (3) 17.9, 19.3, and 26.2 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • FIG. 31 is the DSC thermogram of Formula (I) glutarate. The onset of melting is about 150 °C.
  • Formula (I) methanesulfonate was first prepared by adding one equivalent of methanesulfonic acid to 53.5 mg of Formula (I) freebase Form II slurried in 0.2 mL of acetone at about 22 °C. The slurry was centrifuged and the solid was dried in the vacuum oven at about 50 °C.
  • FIG. 32 is the XRPD pattern of Formula (I) methanesulfonate. It contains the following characteristic peaks: (1) 8.4, 19.1, and 21.2 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (2) 17.2, 19.7, and 20.2 °2 ⁇ ⁇ 0.2 °2 ⁇ ; (3) 12.8, 21.8, and 25.2 °2 ⁇ ⁇ 0.2 °2 ⁇ .
  • Example 1 ASKl (Apoptosis Signal-Regulating Kinase 1) TR-FRET Kinase Assay (Biochemical IC50)
  • HTRF detection was used to measure the phosphorylation level of a biotinylated peptide substrate by the ASKl kinase.
  • This is a competitive, time-resolved fluorescence resonance energy transfer (TR-FRET) immunoassay, based on HTRF® KinEASETM- STK manual from Cisbio.
  • TR-FRET time-resolved fluorescence resonance energy transfer
  • the fluorescence was measured at 615 nm (Cryptate) and 665 nm (XL665) and a ratio of 665 nm/615 nm is calculated for each well.
  • the resulting TR-FRET level (a ratio of 665 nm/615 nm) is proportional to the phosphorylation level. Under these assay conditions, the degree of phosphorylation of peptide substrate was linear with time and
  • ASKl is bound and repressed by the reduced form of the antioxidant protein thioredoxin.
  • ASKl autophosphorylation and activation.
  • Activated ASKl phosphorylates and activates downstream MAPKKs, including MAPKK3, 4, 6, and 7, which in turn phosphorylate p38 and JNK. This is followed by increased phosphorylation of the p38 and JNK substrates ATF2 and c-Jun, respectively; and increased expression of a variety of known ATF2/cJun/APl target genes including the chemokines CXCL1/KC. All of these auranofin-induced signaling events are blocked by ASKl inhibitors, demonstrating that ASKl drives the chain of signaling events following auranofin stimulation.
  • EC5 0 of the compound of Formula (I) was determined in HK-2 as 9.2 nM.
  • the compound of Formula (I) inhibited auranofin- induced phosphorylated p38 with the mean EC 50 of 6.4 nM.
  • the CXCL1 assay is conducted in human whole blood and measures the ability of a test compound to inhibit production of the CXCL1 cytokine after auranofin stimulation of the ASK1 pathway.
  • the compound of Formula (I) inhibited auranofin- induced CXCL1 production in whole blood with an average EC5 0 of 80.8 nM.
  • the compound of Formula (I) inhibits ASK1 activity in multiple cellular assays with or without human whole blood in the medium.
  • the compound of Formula (I) inhibits endogenous ASK1 downstream targets such as phos-p38 and the production of CXCL-1.
  • kinase-tagged T7 phage strains were prepared in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage and incubated with shaking at 32°C until lysis. The lysates were centrifuged and filtered to remove cell debris. The remaining kinases were produced in HEK-293 cells and subsequently tagged with DNA for qPCR detection. Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for about 30 minutes at room temperature to generate affinity resins for kinase assays.
  • the liganded beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1% (bovine serum albumin), 0.05% Tween 20, 1 mM
  • Binding reactions were assembled by combining kinases, liganded affinity beads, and test compounds in lx binding buffer (20% SeaBlock, 0.17x PBS, 0.05% Tween 20, 6 mM DTT). All reactions were performed in polystyrene 96-well plates in a final volume of 0.135 mL. The assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (lx PBS, 0.05% Tween 20).
  • elution buffer lx PBS, 0.05% Tween 20, 0.5 ⁇ non-biotinylated affinity ligand
  • the compound of Formula (I) exhibited a 3 ⁇ 4 of 0.17 nM. This data suggests that the compound of Formula (I) binds potently to ASK1 receptor in the absence of ATP.
  • the compound of Formula (I) was formulated in 5% dextrose at 0.50 mg/mL for
  • Each dosing group consisted of 3 male, cynomolgus monkeys. At dosing, the animals weighed -4.4 kg. The animals were fasted overnight prior to dose administration and up to 4 hr after dosing.
  • the test article was administered by intravenous infusion over 30 min. The rate of infusion was adjusted according to the body weight of each animal to deliver a dose of 1 mg/kg.
  • Serial venous blood samples (approximately 0.3 mL each) were taken at specified time points after dosing from each animal (see Appendix I for the time points).
  • the blood samples were collected into VacutainerTM tubes containing EDTA-K2 as the anti-coagulant and were immediately placed on wet ice pending centrifugation for plasma.
  • An LC/MS/MS method was used to measure the concentration of the compound of Formula (I) in plasma.
  • the terminal ti/2 was -9.8 h and the mean residence time (MRT) was -9.4 h.
  • the terminal t ⁇ for 5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-isopropyl-4H- l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide is -3.16 h.
  • the required amount of solid compound of Formula (I) (free base or salt form) was added to a reaction vessel equipped with a stir bar.
  • the buffer solution was then added to the stirring vessel and the reaction timer was initiated.
  • Samples of the solution were taken at discreet time points (5, 10, 20, 30, 60, and 120 minutes) and filtered through a GHP acrodisc filter (0.45 um).
  • the resulting filtrate was diluted 1: 1 with 65/35 v/v 3 ⁇ 40 0.1% TFA/ACN diluent and the amount of the compound of Formula (I) was quantified by UPLC with comparison to a reference standard.
  • a final sample was taken to analyze the equilibration concentration of each salt form.
  • the remaining solids from the reaction vessel were collected and analyzed by X- ray powder diffraction to determine the final solid form.
  • FIG. 34 and FIG. 35 show that the salts of the compound of Formula (I) have improved kinetic solubility over the freebase.
  • the crystalline succinate Form I was also stable under mechanical stress.
  • the crystal has plate morphology and thus poses less risk to flow compared to needles.
  • the powder was then subjected to grinding for one minute and five minutes with a mortar and pestle and analyzed by X-ray powder diffraction (XRPD). Compression was performed at 2000 psi on a Carver press. The morphology was then characterized by light microscopy (Table 16).
  • Table 17 compares Formula (I) formulated as free base, Form II, in a tablet, and Formula (I) salts formulated as a powder in a capsule (PIC) in a 1:2 ratio of Formula (I):pre-gelatinized starch (PGS).
  • PIC a capsule
  • PPS pre-gelatinized starch
  • Formula (I) free base shows a ⁇ 12x decrease in plasma exposure when dosed in famotidine pre-treated dogs suggesting that Formula (I) free base will have a significant drug-drug interaction when used to treat patients who use acid-suppressive agents such as famotidine or omeprazole.
  • salts of Formula (I) abrogate this effect and show ⁇ 2x difference between pentagastrin and famotidine pre-treatment.

Abstract

Provided is a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in treatment of non-alcoholic steatohepatitis, alcoholic hepatitis, pulmonary arterial hypertension, heart failure with preserved ejection fraction, and diabetic kidney disease. Also provided are amorphous and crystalline forms of the compound of Formula (I) and salts, co-crystals, solvates, and hydrates thereof.

Description

APOPTOSIS SIGNAL-REGULATING KINASE INHIBITOR
CROSS-REFERENCE TO RELATED APPLICATION
This application claims the benefit under 35 U.S.C. § 119(e) of United States Provisional Application No. 62/471,300, filed March 14, 2017, which is hereby incorporated by reference in its entirety.
FIELD
The present disclosure relates to novel compounds that are inhibitors of ASK 1 and that can be used in the treatment of ASK1 -mediated diseases. The disclosure also relates to methods of preparing the compounds of Formula (I) and methods of treating disease by administering compounds of Formula (I), a salt, co-crystal, solvate, or hydrate thereof.
BACKGROUND
Apoptosis signal-regulating kinase 1 (ASK1) is a member of the mitogen- activated protein kinase ("MAP3K") family that activates the c-Jun N-terminal protein kinase ("JNK") and p38 MAP kinase (Ichijo, H., Nishida, E., Irie, K., Dijke, P. T.,
Saitoh, M., Moriguchi, T., Matsumoto, K., Miyazono, K., and Gotoh, Y. (1997) Science, 275, 90-94). ASK1 is activated by a variety of stimuli including oxidative stress, reactive oxygen species (ROS), LPS, TNF-a, FasL, ER stress, and increased intracellular calcium concentrations (Hattori, K., Naguro, I., Runchel, C, and Ichijo, H. (2009) Cell Comm. Signal. 7:1-10; Takeda, K., Noguchi, T., Naguro, I., and Ichijo, H. (2007) Annu. Rev. Pharmacol. Toxicol. 48: 1-8.27; Nagai, H., Noguchi, T., Takeda, K., and Ichijo, I. (2007) /. Biochem. Mol. Biol. 40: 1-6). ASK1 activation and signaling have been reported to play an important role in a broad range of diseases including liver diseases (such as non-alcoholic steatohepatitis, non-alcoholic fatty liver disease, alcoholic hepatitis), cardiovascular diseases (such as pulmonary arterial hypertension and heart failure), kidney disease (such as chronic kidney disease) and also neurodegenerative, inflammatory, autoimmune, and metabolic disorders. Thus, therapeutic agents that function as inhibitors of ASK1 signaling have the potential to remedy or improve the lives of patients in need of treatment for a variety of diseases.
U.S. Patent No. 8,378, 108, U.S. Patent No. 8,552,196, U.S. Patent No. 8,598,360,
U.S. Patent No. 8,742, 126, U.S. Patent No. 9,067,933, U.S. Patent No. 9,254,284, U.S. Patent No. 9,333,197, U.S. Patent No. 9,586,932, U.S. Patent Publication No. 2015/0342943, and U.S. Patent Publication No. 2016/0166556 disclose compounds useful as ASK1 inhibitors and methods of treating disease with an ASK1 inhibitor. Surprisingly, applicants have discovered a novel compound exhibiting good potency and pharmacokinetic and/or pharmacodynamic parameters.
SUMMARY
Provided herein is a compound and pharmaceutical compositions useful as an inhibitor of ASK1. The compound disclosed herein may find use in pharmaceutical compositions, together with at least one pharmaceutically acceptable excipient, for treating a subject in need thereof. The compound of the present disclosure has been found to inhibit ASK1. The disclosure also provides compositions, including pharmaceutical compositions, kits that include the compound of Formula (I), and methods of making and using compounds of Formula (I). Also provided are salts, co- crystals, hydrates and solvates of the compounds of Formula (I).
In one embodiment of the disclosure, is provided a compound of Formula (I):
Figure imgf000003_0001
(I) or a pharmaceutically acceptable salt, co- crystal, solvate or hydrate thereof.
In one embodiment of the disclosure, the compound of Formula (I) is a freebase. In another embodiment of the disclosure, the compound of Formula (I) is a salt. In one embodiment, the compound of Formula (I) is a salt selected from succinate, hydrochloride, phosphate, malate, sulfate, citrate, glutarate, and methane sulfonate.
In one embodiment of the disclosure, the compound of Formula (I) or a pharmaceutically acceptable salt thereof is amorphous.
In one embodiment of the disclosure, the compound of Formula (I) freebase is crystalline Form I. In one embodiment, the compound of Formula (I) freebase Form I is characterized by an X-ray powder diffractogram having peaks at 8.7, 10.2, and 18.1 °2Θ ±0.2 °2Θ. In one embodiment, the compound of Formula (I) freebase Form I is characterized by an X-ray powder diffractogram having peaks at 15.8, 17.3, and 23.5 °2Θ ±0.2 °2Θ. In one embodiment, the compound of Formula (I) freebase Form I is characterized by an X-ray powder diffractogram having peaks at 15.2, 18.5, and 24.9 °2 Θ ±0.2 °2Θ.
In one embodiment, the compound of Formula (I) freebase Form I is
characterized by an X-ray powder diffractogram that is substantially as shown in FIG. 1.
In one embodiment, the compound of Formula (I) freebase Form I is
characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 2.
In another embodiment, the disclosure provides a compound of Formula (I) freebase Form II. In one embodiment, the compound is characterized by an X-ray powder diffractogram having peaks at 8.7, 10.0, and 17.0 °2 Θ ±0.2 °2Θ. In another embodiment, the compound is characterized by an X-ray powder diffractogram having peaks at 13.9, 21.3, and 22.8 °2 Θ ±0.2 °2Θ. In one embodiment, the compound is characterized by an X-ray powder diffractogram having peaks at 24.0, 28.0, and 29.1 °2 Θ ±0.2 °2Θ.
In one embodiment of the disclosure a compound of Formula (I) freebase Form II is characterized by an X-ray powder diffractogram substantially as shown in FIG. 4.
In one embodiment of the disclosure a compound of Formula (I) freebase Form II is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 5.
In another embodiment, the disclosure provides a crystalline succinate Form I of the compound of Formula (I). In one embodiment, the succinate Form I of the compound of Formula (I) is characterized by an X-ray powder diffractogram having peaks at 7.5, 20.9, and 24.9 °2 Θ ±0.2 °2Θ. In one embodiment, the succinate Form I is characterized by an X-ray powder diffractogram having peaks at 15.4, 22.4, and 31.1 °2 Θ ±0.2 °2Θ. In still another embodiment, the succinate Form I is characterized by an X- ray powder diffractogram having peaks at 14.9, 23.2, and 25.4 °2 Θ ±0.2 °2Θ.
In one embodiment of the disclosure, crystalline compound of Formula (I) succinate Form I is characterized by an X-ray powder diffractogram substantially as shown in FIG. 7.
In one embodiment of the disclosure, crystalline compound of Formula (I) succinate Form I is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 8. In one embodiment, the disclosure provides a crystalline compound of Formula (I) succinate Form II. In one embodiment, the crystalline compound of Formula (I) succinate Form II is characterized by an X-ray powder diffractogram having peaks at 9.8, 11.7, and 23.3 °2 Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) succinate Form II is characterized by an X-ray powder diffractogram having peaks at 15.9, 21.5 and 32.0 °2 Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) succinate Form II is characterized by an X-ray powder diffractogram having peaks at 16.3, 20.8, and 24.8 °2 Θ ±0.2 °2Θ.
In one embodiment of the disclosure crystalline compound of Formula (I) succinate Form II is characterized by an X-ray powder diffractogram substantially as shown in FIG. 10.
In one embodiment of the disclosure crystalline compound of Formula (I) succinate Form II is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 11.
In another embodiment, the disclosure provides a crystalline compound of
Formula (I) hydrochloride Form I. In one embodiment, the crystalline compound of Formula (I) hydrochloride Form I is characterized by an X-ray powder diffractogram having peaks at 9.3, 21.2, and 26.2 °2 Θ ±0.2 °2Θ. In one embodiment, the crystalline compound of Formula (I) hydrochloride Form I is characterized by an X-ray powder diffractogram having peaks at 20.9, 24.7, and 27.6 °2 Θ ±0.2 °2Θ. In one embodiment, the crystalline compound of Formula (I) hydrochloride Form I X-ray powder
diffractogram having peaks at 23.8, 24.3, and 26.6 °2 Θ ±0.2 °2Θ.
In one embodiment of the disclosure crystalline compound of Formula (I) hydrochloride Form I is characterized by an X-ray powder diffractogram substantially as shown in FIG. 13.
In one embodiment of the disclosure crystalline compound of Formula (I) hydrochloride Form I is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 14.
In another embodiment, the disclosure provides a crystalline compound of Formula (I) hydrochloride Form II. In one embodiment, the crystalline compound of Formula (I) hydrochloride Form II is characterized by an X-ray powder diffractogram having peaks at 8.3, 12.0, and 26.0 °2 Θ ±0.2 °2Θ. In one embodiment, the crystalline compound of Formula (I) hydrochloride Form II is characterized by an X-ray powder diffractogram having peaks at 19.3, 21.7, and 24.0 °2 Θ ±0.2 °2Θ. In one embodiment, the crystalline compound of Formula (I) hydrochloride Form II X-ray powder diffractogram having peaks at 9.8, 15.4, and 28.9 °2 Θ ±0.2 °2Θ.
In one embodiment of the disclosure, crystalline compound of Formula (I) hydrochloride Form II is characterized by an X-ray powder diffractogram substantially as shown in FIG. 16.
In one embodiment of the disclosure crystalline compound of Formula (I) hydrochloride Form II is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 33.
In another embodiment, the disclosure provides a crystalline compound of
Formula (I) hydrochloride Form III. In one embodiment, the crystalline compound of Formula (I) hydrochloride Form III is characterized by an X-ray powder diffractogram having peaks at 6.6, 12.9, and 19.7 °2 Θ ±0.2 °2Θ. In one embodiment, the crystalline compound of Formula (I) hydrochloride Form III is characterized by an X-ray powder diffractogram having peaks at 20.4, 23.4, and 24.8 °2 Θ ±0.2 °2Θ. In one embodiment, the crystalline compound of Formula (I) hydrochloride Form III X-ray powder diffractogram having peaks 18.1, 25.4, and 26.7 °2 Θ ±0.2 °2Θ.
In one embodiment of the disclosure, crystalline compound of Formula (I) hydrochloride Form III is characterized by an X-ray powder diffractogram substantially as shown in FIG. 17.
In one embodiment of the disclosure crystalline compound of Formula (I) hydrochloride Form III is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 18.
In one embodiment, the disclosure provides a crystalline compound of Formula (I) L-malate. In one embodiment, the crystalline compound of Formula (I) L-malate is characterized by an X-ray powder diffractogram having peaks at 7.2, 21.9, and 24.5 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) L-malate is characterized by an X-ray powder diffractogram having peaks at 10.9, 16.6, and 19.7 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) L-malate is characterized by an X-ray powder diffractogram having peaks at 8.2, 22.8, and 27.3 °2Θ ±0.2 °2Θ. In one embodiment of the disclosure crystalline compound of Formula (I) L- malate is characterized by an X-ray powder diffractogram substantially as shown in FIG. 19.
In one embodiment of the disclosure crystalline compound of Formula (I) L- malate is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 20.
In one embodiment, the disclosure provides a crystalline compound of Formula (I) phosphate Material A. In one embodiment, the crystalline compound of Formula (I) phosphate Material A is characterized by an X-ray powder diffractogram having peaks at 6.9, 8.0, and 21.2 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) phosphate Material A is characterized by an X-ray powder diffractogram having peaks at 22.8, 25.3, and 26.0 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) phosphate Material A is characterized by an X-ray powder diffractogram having peaks at 19.8, 20.6, 23.2 °2Θ ±0.2 °2Θ.
In one embodiment of the disclosure crystalline compound of Formula (I) phosphate Material A is characterized by an X-ray powder diffractogram substantially as shown in FIG. 22.
In one embodiment, the disclosure provides a crystalline compound of Formula (I) phosphate Material B. In one embodiment, the crystalline compound of Formula (I) phosphate Material B is characterized by an X-ray powder diffractogram having peaks at 13.4, 23.5, and 24.3 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) phosphate Material B is characterized by an X-ray powder diffractogram having peaks at 21.7, 25.1, and 25.8 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) phosphate Material B is characterized by an X-ray powder diffractogram having peaks at 17.0, 18.6, and 22.5 °2Θ ±0.2 °2Θ.
In one embodiment of the disclosure crystalline compound of Formula (I) phosphate Material B is characterized by an X-ray powder diffractogram substantially as shown in FIG. 23.
In one embodiment, the disclosure provides a crystalline compound of Formula (I) sulfate. In one embodiment, the crystalline compound of Formula (I) sulfate is characterized by an X-ray powder diffractogram having peaks at 7.2, 13.5, and 21.5 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) sulfate is characterized by an X-ray powder diffractogram having peaks at 16.9, 20.5, and 25.3 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) sulfate is characterized by an X-ray powder diffractogram having peaks at 18.2, 20.6, and 27.2 °2Θ ±0.2 °2Θ.
In one embodiment of the disclosure crystalline compound of Formula (I) sulfate is characterized by an X-ray powder diffractogram substantially as shown in FIG. 24.
In one embodiment of the disclosure crystalline compound of Formula (I) sulfate is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 25.
In one embodiment, the disclosure provides a crystalline compound of Formula (I) citrate Material A. In one embodiment, the crystalline compound of Formula (I) citrate Material A is characterized by an X-ray powder diffractogram having peaks at 5.6, 9.1, 16.7 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) citrate Material A is characterized by an X-ray powder diffractogram having peaks at 12.2, 21.0, 23.0 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) citrate Material A is characterized by an X-ray powder diffractogram having peaks at 18.7, 24.9, 25.8 °2Θ ±0.2 °2Θ.
In one embodiment of the disclosure crystalline compound of Formula (I) citrate Material A is characterized by an X-ray powder diffractogram substantially as shown in FIG. 27.
In one embodiment, the disclosure provides a crystalline compound of Formula
(I) citrate Material B. In one embodiment, the crystalline compound of Formula (I) citrate Material B is characterized by an X-ray powder diffractogram having peaks at 5.6, 8.6, and 9.1 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) citrate Material B is characterized by an X-ray powder diffractogram having peaks at 12.2, 23.2, and 23.9 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) citrate
Material B is characterized by an X-ray powder diffractogram having peaks at 16.8, 18.5, and 21.1 °2Θ ±0.2 °2Θ.
In one embodiment of the disclosure crystalline compound of Formula (I) citrate Material B is characterized by an X-ray powder diffractogram substantially as shown in FIG. 28. In one embodiment of the disclosure crystalline compound of Formula (I) citrate Material B is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 29.
In one embodiment, the disclosure provides a crystalline compound of Formula (I) glutarate. In one embodiment, the crystalline compound of Formula (I) glutarate is characterized by an X-ray powder diffractogram having peaks at 6.7, 8.0, and 12.1 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) glutarate is characterized by an X-ray powder diffractogram having peaks at 14.6, 20.9, and 21.8 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) glutarate is characterized by an X-ray powder diffractogram having peaks at 17.9, 19.3, and 26.2 °2Θ ±0.2 °2Θ.
In one embodiment of the disclosure crystalline compound of Formula (I) glutarate is characterized by an X-ray powder diffractogram substantially as shown in FIG. 30.
In one embodiment of the disclosure crystalline compound of Formula (I) glutarate is characterized by a differential scanning calorimetry (DSC) curve
substantially as shown in FIG. 31.
In one embodiment, the disclosure provides a crystalline compound of Formula (I) methanesulfonate. In one embodiment, the crystalline compound of Formula (I) methanesulfonate is characterized by an X-ray powder diffractogram having peaks at 8.4,
19.1, and 21.2 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) methanesulfonate is characterized by an X-ray powder diffractogram having peaks at
17.2, 19.7, and 20.2 °2Θ ±0.2 °2Θ. In another embodiment, the compound of Formula (I) methanesulfonate is characterized by an X-ray powder diffractogram having peaks at 12.8, 21.8, and 25.2 °2Θ ±0.2 °2Θ.
In one embodiment of the disclosure crystalline compound of Formula (I) methanesulfonate is characterized by an X-ray powder diffractogram substantially as shown in FIG. 32.
In one embodiment, the disclosure provides a therapeutically effective amount of a compound of Formula (I) a salt, co-crystal, solvate, or hydrate thereof with a pharmaceutically acceptable excipient. In one embodiment, the disclosure provides a composition comprising a compound of Formula (I) a salt, co-crystal, solvate, or hydrate thereof with a pharmaceutically acceptable excipient and one, two, or three additional agents.
In one embodiment, the disclosure provides a composition comprising a compound of Formula (I), a salt, co-crystal, solvate, or hydrate thereof with a pharmaceutically acceptable excipient and one additional agent that is an FXR agonist. The FXR agonist may be a compound of Formula (III) or a compound of Formula (IV) as defined here.
In one embodiment, the disclosure provides a composition comprising a compound of Formula (I) a salt, co-crystal, solvate, or hydrate thereof with a pharmaceutically acceptable excipient and one additional agent that is an ACC inhibitor.
The ACC inhibitor may be a compound of Formula (V) as defined herein.
The disclosure provides methods for inhibiting ASK1 with a compound of
Formula (I) or a salt, co-crystal, solvate, or hydrate thereof.
In another embodiment, the disclosure provides a method of treating a condition selected from non-alcoholic steatohepatitis, alcoholic hepatitis, pulmonary arterial hypertension, heart failure with preserved ejection fraction, and diabetic kidney disease by administering a therapeutically effective amount of a compound of Formula (I) or a salt, co-crystal, solvate, or hydrate thereof.
In one embodiment, the disclosure provides a method of treating fibrosis with a therapeutically effective amount of a compound of Formula (I) or a salt, co-solvate, or hydrate thereof.
In one embodiment, the disclosure provides a compound of Formula (la):
Figure imgf000010_0001
DESCRIPTION OF THE DRAWINGS FIG. 1 is a XRPD pattern for a compound of Formula (I) freebase Form I.
FIG. 2 is a DSC thermogram for a compound of Formula (I) freebase Form I. FIG. 3 is a TGA thermogram for a compound of Formula (I) freebase Form I.
FIG. 4 is a XRPD pattern for a compound of Formula (I) freebase Form II.
FIG. 5 is a DSC thermogram for a compound of Formula (I) freebase Form II.
FIG. 6 is a TGA thermogram for a compound of Formula (I) freebase Form II. FIG. 7 is a XRPD pattern for a compound of Formula (I) succinate Form I.
FIG. 8 is a DSC thermogram for a compound of Formula (I) succinate Form I.
FIG. 9 is a TGA thermogram for a compound of Formula (I) succinate Form I.
FIG. 10 is a XRPD pattern for a compound of Formula (I) succinate Form II.
FIG. 11 is a DSC thermogram for a compound of Formula (I) succinate form II. FIG. 12 is a TGA thermogram for a compound of Formula (I) succinate Form II.
FIG. 13 is a XRPD pattern for a compound of Formula (I) HC1 Form I.
FIG. 14 is a DSC thermogram for a compound of Formula (I) HC1 Form I.
FIG. 15 is a TGA thermogram for a compound of Formula (I) HC1 Form I.
FIG. 16 is a XRPD pattern for a compound of Formula (I) HC1 Form II.
FIG. 17 is a XRPD pattern for a compound of Formula (I) HC1 Form III.
FIG. 18 is a DSC thermogram for a compound of Formula (I) HC1 Form III.
FIG. 19 is a XRPD pattern for a compound of Formula (I) L-malate.
FIG. 20 is a DSC thermogram for a compound of Formula (I) L-malate.
FIG. 21 is a TGA thermogram for a compound of Formula (I) L-malate.
FIG. 22 is a XRPD pattern for a compound of Formula (I) phosphate Material A
FIG. 23 is a XRPD pattern for a compound of Formula (I) phosphate Material B.
FIG. 24 is a XRPD pattern for a compound of Formula (I) sulfate.
FIG. 25 is a DSC thermogram for a compound of Formula (I) sulfate.
FIG. 26 is a TGA thermogram for a compound of Formula (I) sulfate.
FIG. 27 is a XRPD pattern for a compound of Formula (I) citrate Material A.
FIG. 28 is a XRPD pattern for a compound of Formula (I) citrate Material B. FIG. 29 is a DSC thermogram for a compound of Formula (I) citrate Material B.
FIG. 30 is a XRPD pattern for a compound of Formula (I) glutarate.
FIG. 31 is a DSC thermogram for a compound of Formula (I) glutarate.
FIG. 32 is a XRPD pattern for a compound of Formula (I) methanesulfonate. FIG. 33 is a DSC thermogram for a compound of Formula (I) HC1 Form II.
FIGS. 34 and 35 show kinetic solubility profiles of various salts of the compound of Formula (I) (50 mM Acetate, pH=5.5, 1=150 mM, FaSSIF).
DETAILED DESCRIPTION
Definitions
The following description sets forth exemplary methods, parameters and the like.
It should be recognized, however, that such description is not intended as a limitation on the scope of the present disclosure but is instead provided as a description of exemplary embodiments.
A dash ("-") that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, -C(0)NH2 is attached through the carbon atom. A dash at the front or end of a chemical group is a matter of convenience;
chemical groups may be depicted with or without one or more dashes without losing their ordinary meaning. A wavy line drawn through a line in a structure indicates a point of attachment of a group. Unless chemically or structurally required, no directionality is indicated or implied by the order in which a chemical group is written or named.
Reference to "about" a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. In certain embodiments, the term "about" includes the indicated amount + 10%. In other embodiments, the term "about" includes the indicated amount + 5%. In certain other embodiments, the term "about" includes the indicated amount ± 1%. Also, to the term "about X" includes description of "X". Also, the singular forms "a" and "the" include plural references unless the context clearly dictates otherwise. Thus, e.g., reference to "the compound" includes a plurality of such compounds and reference to "the assay" includes reference to one or more assays and equivalents thereof known to those skilled in the art. Any formula or structure given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds. Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass
number. Examples of isotopes that can be incorporated into compounds of the disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as, but not limited to 2H (deuterium, D), 3H (tritium), nC, 13C, 14C, 15N,
18 31 32 35 36 125
F, P, P, S, Cl and I. Various isotopically labeled compounds of the present disclosure, for example those into which radioactive isotopes such as 3H, 13C and 14C are incorporated. Such isotopically labelled compounds may be useful in metabolic studies, reaction kinetic studies, detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays or in radioactive treatment of patients.
The disclosure also includes "deuterated analogues" of compounds of Formula (I) in which from 1 to n hydrogens attached to a carbon atom is/are replaced by deuterium, in which n is the number of hydrogens in the molecule. Such compounds exhibit increased resistance to metabolism and are thus useful for increasing the half-life of any compound of Formula (I) when administered to a mammal, particularly a human. See, for example, Foster, "Deuterium Isotope Effects in Studies of Drug Metabolism," Trends Pharmacol. Sci. 5(12):524-527 (1984). Such compounds are synthesized by means well known in the art, for example by employing starting materials in which one or more hydrogens have been replaced by deuterium.
Deuterium labelled or substituted therapeutic compounds of the disclosure may have improved DMPK (drug metabolism and pharmacokinetics) properties, relating to distribution, metabolism and excretion (ADME). Substitution with heavier isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life, reduced dosage requirements and/or an improvement in therapeutic index. An 18F labeled compound may be useful for PET or SPECT studies. Isotopically labeled compounds of this disclosure and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent. It is understood that deuterium in this context is regarded as a substituent in the compound of Formula (I). The concentration of such a heavier isotope, specifically deuterium, may be defined by an isotopic enrichment factor. In the compounds of this disclosure any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom. Unless otherwise stated, when a position is designated specifically as "H" or "hydrogen", the position is understood to have hydrogen at its natural abundance isotopic composition. Accordingly, in the compounds of this disclosure any atom specifically designated as a deuterium (D) is meant to represent deuterium.
The compound of Formula (I) is capable of forming salts as described herein. Provided are pharmaceutically acceptable salts, hydrates, solvates, tautomeric forms, polymorphs, and prodrugs of the compounds described herein. "Pharmaceutically acceptable" or "physiologically acceptable" refer to compounds, salts, compositions, dosage forms and other materials which are useful in preparing a pharmaceutical composition that is suitable for veterinary or human pharmaceutical use.
The salt may be any acid or base capable of forming a salt with the compound of Formula (I). It may be, for example, selected from a pharmaceutically acceptable salt or synthetically useful salt. The term "pharmaceutically acceptable salt" of a given compound refers to salts that retain the biological effectiveness and properties of the given compound, and which are not biologically or otherwise undesirable.
"Pharmaceutically acceptable salts" or "physiologically acceptable salts" include, for example, salts with inorganic acids and salts with organic acids. In addition, if the compounds described herein are obtained as an acid addition salt, the free base can be obtained by basifying a solution of the acid salt. Conversely, if the product is a free base, an addition salt, particularly a pharmaceutically acceptable addition salt, may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds. Those skilled in the art will recognize various synthetic methodologies that may be used to prepare nontoxic pharmaceutically acceptable addition salts. Pharmaceutically acceptable acid addition salts may be prepared from inorganic and organic acids. Salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Salts derived from organic acids include acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene-sulfonic acid, salicylic acid, and the like. Likewise, pharmaceutically acceptable base addition salts can be prepared from inorganic and organic bases. Salts derived from inorganic bases include, by way of example only, sodium, potassium, lithium, ammonium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary and tertiary amines, such as alkyl amines (i.e., NH2(alkyl)), dialkyl amines (i.e., HN(alkyl)2), trialkyl amines (i.e., N(alkyl)3), substituted alkyl amines (i.e., NH2(substituted alkyl)), di(substituted alkyl) amines (i.e., HN(substituted alkyl)2), tri(substituted alkyl) amines (i.e., N(substituted alkyl)3), alkenyl amines (i.e.,
N]¾(alkenyl)), dialkenyl amines (i.e., HN(alkenyl)2), trialkenyl amines (i.e.,
N(alkenyl)s), substituted alkenyl amines (i.e., NH2(substituted alkenyl)), di(substituted alkenyl) amines (i.e., HN(substituted alkenyl^), tri(substituted alkenyl) amines (i.e., N(substituted alkenyl)3, mono-, di- or tri- cycloalkyl amines (i.e., NH2(cycloalkyl), HN(cycloalkyl)2, N(cycloalkyl)3), mono-, di- or tri- arylamines (i.e., NH2(aryl), HN(aryl)2, N(aryl)s), or mixed amines, etc. Specific examples of suitable amines include, by way of example only, isopropylamine, trimethyl amine, diethyl amine, tri(iso-propyl) amine, tri(n-propyl) amine, ethanolamine, 2-dimethylaminoethanol, piperazine, piperidine, morpholine, N-ethylpiperidine, and the like.
As used herein, "pharmaceutically acceptable carrier" or "pharmaceutically acceptable excipient" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances are known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Additional active ingredients can also be incorporated into the compositions.
The term "therapeutically effective amount" refers to an amount of the compound of Formula (I) that is sufficient to effect treatment as defined below, when administered to a patient (particularly a human) in need of such treatment in one or more doses. The therapeutically effective amount will vary, depending upon the patient, the disease being treated, the weight and/or age of the patient, the severity of the disease, or the manner of administration as determined by a qualified prescriber or care giver.
The term "treatment" or "treating" means administering a compound or pharmaceutically acceptable salt of Formula (I) for the purpose of: (i) delaying the onset of a disease, that is, causing the clinical symptoms of the disease not to develop or delaying the development thereof; (ii) inhibiting the disease, that is, arresting the development of clinical symptoms; and/or (iii) relieving the disease, that is, causing the regression of clinical symptoms or the severity thereof.
Methods
Disclosed herein is a method of treating and/or preventing liver disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of Formula (I). The presence of active liver disease can be detected by the existence of elevated enzyme levels in the blood. Specifically, blood levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) above clinically accepted normal ranges are known to be indicative of on-going liver damage. Routine monitoring of liver disease patients for blood levels of ALT and AST is used clinically to measure progress of the liver disease while on medical treatment. Reduction of elevated ALT and AST to within the accepted normal range is taken as clinical evidence reflecting a reduction in the severity of the patient's on- going liver damage.
In certain embodiments, the liver disease is a chronic liver disease. Chronic liver diseases involve the progressive destruction and regeneration of the liver parenchyma, leading to fibrosis and cirrhosis. In general, chronic liver diseases can be caused by viruses (such as hepatitis B, hepatitis C, cytomegalovirus (CMV), or Epstein Barr Virus (EBV)), toxic agents or drugs (such as alcohol, methotrexate, or nitrofurantoin), a metabolic disease (such as non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), haemochromatosis, or Wilson's Disease), an autoimmune disease (such as Autoimmune Chronic Hepatitis, Primary Biliary Cholangitis (formerly known as Primary Biliary Cirrhosis), or Primary Sclerosing Cholangitis), or other causes (such as right heart failure).
In one embodiment, provided herein is a method for reducing the level of cirrhosis. In one embodiment, cirrhosis is characterized pathologically by loss of the normal microscopic lobular architecture, with fibrosis and nodular regeneration.
Methods for measuring the extent of cirrhosis are well known in the art. In one embodiment, the level of cirrhosis is reduced by about 5% to about 100%. In one embodiment, the level of cirrhosis is reduced by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100% in the subject.
In certain embodiments, the liver disease is a metabolic liver disease. In one embodiment, the liver disease is non-alcoholic fatty liver disease (NAFLD). NAFLD is associated with insulin resistance and metabolic syndrome (obesity, combined hyperlipidemia, diabetes mellitus (type II) and high blood pressure). NAFLD is considered to cover a spectrum of disease activity, and begins as fatty accumulation in the liver (hepatic steatosis).
It has been shown that both obesity and insulin resistance probably play a strong role in the disease process of NAFLD. In addition to a poor diet, NAFLD has several other known causes. For example, NAFLD can be caused by certain medications, such as amiodarone, antiviral drugs (e.g., nucleoside analogues), aspirin (rarely as part of Reye's syndrome in children), corticosteroids, methotrexate, tamoxifen, or tetracycline. NAFLD has also been linked to the consumption of soft drinks through the presence of high fructose corn syrup which may cause increased deposition of fat in the abdomen, although the consumption of sucrose shows a similar effect (likely due to its breakdown into fructose). Genetics has also been known to play a role, as two genetic mutations for this susceptibility have been identified.
If left untreated, NAFLD can develop into non-alcoholic steatohepatitis (NASH), which is the most extreme form of NAFLD, a state in which steatosis is combined with inflammation and fibrosis. NASH is regarded as a major cause of cirrhosis of the liver. Accordingly, provided herein is a method of treating and/or preventing nonalcoholic steatohepatitis (NASH) in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of Formula (I).
Also provided herein is a method of treating and/or preventing liver fibrosis in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of Formula (I). Liver fibrosis is the excessive accumulation of extracellular matrix proteins including collagen that occurs in most types of chronic liver diseases. In certain embodiments, advanced liver fibrosis results in cirrhosis and liver failure. Methods for measuring liver histologies, such as changes in the extent of fibrosis, lobular hepatitis, and periportal bridging necrosis, are well known in the art. In one embodiment, treatment as described herein may improve a patient' s fibrosis from baseline, for example, improving from F4 to F3, F3 to F2, or F2 to Fl. In one embodiment, a patient's fibrosis score is improved by one or more following 24 weeks of daily treatment. In one embodiment, provided is a method for treating liver fibrosis in a patient in need thereof, wherein the liver fibrosis stage of the patient is F3, comprising administering to the patient a therapeutically effective amount of an ASKl inhibitor. In one embodiment, provided is a method for treating liver fibrosis in a patient in need thereof, wherein the liver fibrosis stage of the patient is F3, comprising administering to the patient a therapeutically effective amount of an ASKl inhibitor in combination with a therapeutically effective amount of an ACC inhibitor. In one embodiment, provided is a method for treating liver fibrosis in a patient in need thereof, wherein the liver fibrosis stage of the patient is F4, comprising administering to the patient a therapeutically effective amount of an ASKl inhibitor. In another embodiment, provided is a method for treating liver fibrosis in a patient in need thereof, wherein the liver fibrosis stage of the patient is F4, comprising administering to the patient a therapeutically effective amount of an ASKl inhibitor in combination with a
therapeutically effective amount of an ACC inhibitor.
In still other embodiments, provided is a method for treating NASH in a patient in need thereof, wherein the liver fibrosis stage of the patient is F3, comprising administering to the patient a therapeutically effective amount of an ASKl inhibitor.. In still other embodiments, provided is a method for treating NASH in a patient in need thereof, wherein the liver fibrosis stage of the patient is F4, comprising administering to the patient a therapeutically effective amount of an ASKl inhibitor.
Liver disease can be classified into 4 stages: F0 indicates no fibrosis; Fl indicates mild fibrosis; F2 indicates moderate fibrosis; F3 indicates severe fibrosis; and F4 indicates cirrhosis. As used herein, "Fibrosis Score" refers to a scoring system for fibrosis as described by Kleiner et al. (Hepatology, Design and validation of a histological scoring system for nonalcoholic fatty liver disease (2005), 41: 1313-1321).
In one embodiment, the level of liver fibrosis, which is the formation of fibrous tissue, fibroid or fibrous degeneration, is reduced by more than about 90%. In one embodiment, the level of fibrosis, which is the formation of fibrous tissue, fibroid or fibrous degeneration, is reduced by at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30%, at least about 20%, at least about 10%, at least about 5% or at least about 2%.
In one embodiment, the compounds provided herein reduce the level of fibrogenesis in the liver. Liver fibrogenesis is the process leading to the deposition of an excess of extracellular matrix components in the liver known as fibrosis. It is observed in a number of conditions such as chronic viral hepatitis B and C, alcoholic liver disease, drug-induced liver disease, hemochromatosis, auto-immune hepatitis, Wilson disease, Primary Biliary Cholangitis (formerly known as Primary Biliary Cirrhosis), sclerosing cholangitis, liver schistosomiasis and others. In one embodiment, the level of fibrogenesis is reduced by more than about 90%. In one embodiment, the level of fibrogenesis is reduced by at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least 40%, at least about 30%, at least about 20%, at least about 10%, at least about 5% or at least about 2%.
In still other embodiments, provided herein is a method of treating and/or preventing primary sclerosing cholangitis (PSC) in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of Formula (I).
Also disclosed herein is a method of treating or preventing cardiovascular disorder a patient in need of such treatment comprising administering a therapeutically effective amount of a compound of Formula (I). Cardiovascular diseases refer to any one or more than one of, for example, heart failure (including congestive heart failure, diastolic heart failure, systolic heart failure, heart failure with preserved ejection fraction), acute heart failure, ischemia, recurrent ischemia, myocardial infarction, arrhythmias, angina (including exercise-induced angina, variant angina, stable angina, unstable angina), acute coronary syndrome, diabetes, intermittent claudication, and idiopathic pulmonary fibrosis.
Also provided herein is a method of treating and/or preventing pulmonary vascular disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of Formula (I) or a salt, co-crystal, solvate, or hydrate thereof. In certain embodiments, the pulmonary vascular disease is a pulmonary arterial hypertension (PAH). In some embodiments, the patient is diagnosed with Group 1, Γ, 1", 2, 3, 4, or 5 pulmonary hypertension. Provided herein is a method of treating and/or preventing right ventricle dysfunction in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of an ASKl inhibitor.
Also provided herein is a method of treating, preventing, and/or reversing the narrowing or restricting of pulmonary arteries in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of an ASKl inhibitor. Additionally, provided herein is a method of reducing or normalizing high mean pulmonary arterial pressure (mPAP) and/or high pulmonary vascular resistance in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of an ASKl inhibitor. In one embodiment, the high mPAP > 25 mmHg at rest may be reduced to levels within the normal range at rest by the methods described herein. In some embodiment, the high mPAP > 25 mmHg at rest may be reduced to about 22 mmHg, 20 mmHg, 18 mmHg, 16 mmHg, or 14 mmHg at rest by the methods described herein. In certain embodiment, mPAP is determined by right heart catheterization (RHC).
Provided herein is a method of improving or reducing PAH symptoms in a patient in need thereof, comprising administering a therapeutically effective amount of ASKl inhibitor. In some embodiments, PAH symptoms include and are not limited to breathlessness or shortness of breath (dyspnea), fatigue, dizziness, fainting (syncope), swollen ankles and legs (edema), chest pain, right heart failure and/or dysfunction. In certain embodiments, the improvement may be determined by a change from baseline in pulmonary vascular resistance (PVR), a change from baseline in cardiac index (CI) such as mean pulmonary artery pressure (mPAP), mean right atrial pressure (mRAP), mixed venous oxygen saturation (Sv02), and right ventricular cardiac power, a change from baseline in clinical measures of symptoms and function, including but not limited to submaximal exercise (6-minute walk test (6MWT)), heart rate recovery (HRR) after the 6MWT, the Borg dyspnea index, WHO Functional Class, N-terminal pro-brain natriuretic peptide, and/or quality of life by the SF-36® Health Survey. In other embodiments, PVR is determined by right heart catheterization. In additional embodiments, cardiac function is determined by echocardiography or cardiac hemodynamic data. Additionally provided herein is a method of reducing the remodeling of pulmonary vasculature or arteries in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of an ASK1 inhibitor.
Further provided herein is a method of treating and/or preventing right ventricle failure or right ventricle dysfunction in a patient in need thereof comprising
administering to the patient a therapeutically effective amount of an ASK1 inhibitor. In one embodiment, the right ventricle failure or dysfunction may be detected or monitored by cardiac imaging such as echocardiography and cardiac MRI.
Provided herein is a method of improving and/or reducing PVR, pulmonary pressure, pulmonary vascular remodeling, vascular function, maladaptive RV
hypertrophy, and/or RV function in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of an ASK1 inhibitor.
The present application provides a therapy or treatment to a patient in need, wherein the patient has or is suspected to have pulmonary vascular disease such as pulmonary hypertension or pulmonary arterial hypertension. In one embodiment, the patients experience one or more symptoms selected from breathlessness or shortness of breath (dyspnea), fatigue, dizziness, fainting (syncope), swollen ankles and legs (edema), or chest pain (e.g. angina). The patients may be at various clinical or treatment stages, including patients who have not received any prior treatment to pulmonary hypertension or pulmonary arterial hypertension, patient who have received prior therapies or drugs for pulmonary hypertension or pulmonary arterial hypertension and remains
symptomatic, and patients who currently receive other therapies or drugs for pulmonary hypertension or pulmonary arterial hypertension. For example, the patient may have received the therapeutics of the present application (e.g. the ASK1 inhibitor or a pharmaceutical composition thereof) and other PAH drugs concurrently.
In any of the foregoing, the treatment, prevention, reduction, reversion, and/or improvement by the method described herein may be determined by a change from baseline in pulmonary vascular resistance (PVR), a change from baseline in cardiac index (CI) such as mean pulmonary artery pressure (mPAP), mean right atrial pressure (mRAP), mixed venous oxygen saturation (Sv02), and right ventricular cardiac power, a change from baseline in clinical measures of symptoms and function, including but not limited to submaximal exercise (6-minute walk test (6MWT)), heart rate recovery (HRR) after the 6MWT, the Borg dyspnea index, WHO Functional Class, N-terminal pro-brain natriuretic peptide, an/or quality of life by the SF-36® Health Survey. In other embodiments, PVR is determined by right heart catheterization. In additional embodiments, cardiac function is determined by echocardiography or cardiac hemodynamic data. The baseline refers to a value, number, or reading that is determined or measured from the subject prior to any treatment. By way of example, the baseline is a value, number, or reading from a patient prior to being treated with the methods described herein, from a healthy individual, from a group of subjects, or from suitable guidelines. In one embodiment, the baseline is a value, number, or reading from a patient prior to being treated with the methods described herein. The baseline value or number may be determined or measured by any suitable methods.
As used herein, the terms "right ventricle (RV) dysfunction," "right ventricular dysfunction," "right heart failure," or variants thereof refer to the failure of right ventricle or right heart is unable to carry out the normal function (e.g. pumping blood out of the heart into the lungs to be replenished with oxygen, and/or maintaining sufficient blood flow to meet the needs of the body). RV dysfunction may be determined or detected by cardiac imaging including echocardiography and cardiac MRI which characterizes structural changes (myocardial hypertrophy followed by progressive contractile dysfunction and chamber dilation) and/or functional changes (reduced fractional shortening, increased filling pressures, reduced right ventricular ejection fraction and decreased cardiac output). Other commonly used methods may also be used to determine or detect RV dysfunction. Also, "promoting" or "stimulating" refer to one or more factor that may cause or contribute to progressing of activity, disease, disorder, or condition. For example, promoting or contributing to PAH is used to describe one or more factor that may cause or contribute to progressing or developing of PAH.
Also disclosed herein a method of treating or preventing kidney disorders such as chronic kidney disease or diabetic kidney disease. The term "chronic kidney disease" as used herein refers to progressive loss of kidney function over time typically months or even years. Chronic kidney disease (CKD) is diagnosed by a competent care giver using appropriate information, tests or markers known to one of skill in the art. Chronic kidney disease includes by implication kidney disease. The term "diabetic kidney disease" as used herein refers to kidney disease caused by diabetes, exacerbated by diabetes, or co- presenting with diabetes. It is a form of chronic kidney disease occurring in approximately 30% of patients with diabetes. It is defined as diabetes with the presence of albuminuria and/or impaired renal function (i.e. decreased glomerular filtration rate (See de B, I, et al. Temporal trends in the prevalence of diabetic kidney disease in the United States. JAMA 2011 Jun. 22; 305(24):2532-2539).
In alternative embodiments, a compound of Formula (I), or salt thereof, can be used to prevent or decrease the likelihood of transplant rejection.
In still other embodiments, an inflammatory disease, such as Rheumatoid Arthritis is treated by administering a compound of Formula (I), or salt thereof.
In certain embodiments, systemic lupus erythematosus is treated by administering a compound of Formula (I), or salt thereof.
In still other embodiments, neuropathy, such as HIV-associated neuropathy is treated by administering a compound of Formula (I), or salt thereof.
Also provided herein is a method of improving pathological consequence or outcome associated with oxidative stress in a patient in need thereof comprising administering to the patient a therapeutically effective amount of ASK1 inhibitor.
Combinations
In one embodiment, the compound of Formula (I) is combined with an additional active agent selected from an ACE inhibitor, Acetyl CoA carboxylase inhibitor, Adenosine A3 receptor agonist, Adiponectin receptor agonist, AKT protein kinase inhibitor AMP-activated protein kinase (AMPK) inhibitor, Amylin receptor agonist,
Angiotensin II AT-1 receptor antagonist, Autotaxin inhibitors, Bioactive lipid, Calcitonin agonist, Caspase inhibitor, Caspase-3 stimulator, Cathepsin B inhibitor, Cathepsin K inhibitor, Cathepsin S inhibitor, Caveolin 1 inhibitor, CCR2 chemokine antagonist, CCR3 chemokine antagonist, CCR5 chemokine antagonist, Chloride channel stimulator, CNR1 inhibitor, Cyclin Dl inhibitor, Cytochrome P450 7A1 inhibitor, DGAT2 gene inhibitor, Dipeptidyl peptidase IV inhibitor, Eotaxin ligand inhibitor, Extracellular matrix protein modulator, Farnesoid X receptor agonist, FGF1 receptor agonist, FGF-19 ligand, Galectin-3 inhibitor, Glucagon receptor agonist, Glucagon-like peptide 1 agonist, G-protein coupled bile acid receptor 1 agonist, Hedgehog (Hh) modulators, Hepatitis C virus NS3 protease inhibitor, HMG CoA reductase inhibitor, IL-10 agonist, IL-17 antagonist, Ileal sodium bile acid cotransporter inhibitor, Insulin sensitizer, β3 or βΐ Integrins , Interleukin- 1 receptor-associated kinase 4 (IRAK4), Jak2 tyrosine kinase inhibitor, Klotho beta stimulator, 5 -Lipoxygenase inhibitor, Lipoprotein lipase inhibitor, LPL gene stimulator, Lysophosphatidate- 1 receptor antagonist, Lysyl oxidase homolog 2 inhibitor, Matrix metalloproteinases (MMPs) MEKK-5 protein kinase inhibitor, Membrane copper amine oxidase (VAP-1) inhibitor, Methionine aminopeptidase-2 inhibitor, Methyl CpG binding protein 2 modulator, Mitochondrial uncouplers, Myelin basic protein stimulator, NACHT LRR PYD domain protein 3 (NLRP3) inhibitor NAD- dependent deacetylase sirtuin stimulator, NADPH oxidase 1 inhibitor, NADPH oxidase 4 inhibitor, Nicotinic acid receptor 1 agonist, P2Y13 purinoceptor stimulator, PDE 3 inhibitor, PDE 4 inhibitor, PDE 5 inhibitor, PDGF receptor beta modulator,
Phospholipase C inhibitor, PPAR alpha agonist, PPAR delta agonist, PPAR gamma agonist, PPAR gamma modulator, Protease- activated receptor-2 antagonist, Protein kinase modulator, Rho associated protein kinase 2 inhibitor, Sodium glucose transporter- 2 inhibitor, SREBP transcription factor inhibitor, STAT-1 inhibitor, Stearoyl CoA desaturase-1 inhibitor, Suppressor of cytokine signalling- 1 stimulator, Suppressor of cytokine signalling-3 stimulator, Transforming growth factor βΐ (TGF-βΙ), Thyroid hormone receptor beta agonist, TLR-4 antagonist, Transglutaminase inhibitor, Tyrosine kinase receptor modulator, Unspecified GPCR modulator, Unspecified nuclear hormone receptor modulator, WNT modulators, YAP/TAZ modulators. Specific examples include A-4250, AC-3174, acetylsalicylic acid, AK-20, alipogene tiparvovec, aramchol, ARI-3037MO, atorvastatin, AZ compound (NASH/NAFLD), bertilimumab, Betaine anhydrous, BI-1467335, BMS-986036, BMT-053011, BOT-191, CAT-2003, cenicriviroc, CER-209, CF-102, CNX-014, CNX-023, CNX-024, CNX-025, cobiprostone, colesevelam, dapagliflozin, deuterated pioglitazone R-enantiomer 2,4- dinitrophenol, DRX-065, DS-102, dual acting glucagon-like peptide 1 receptor/glucagon receptor co-agonist, DUR-928, EDP-305, elafibranor, emricasan, enalapril, ertugliflozin, GKT-831, GNF-5120, GR-MD-02, GS-4997, GS-9674, hydrochlorothiazide, icosapent ethyl ester, IMM-124-E, INT-767, IONIS-DGAT2Rx, ipragliflozin, Irbesarta + propagermanium, IVA-337, JKB-121, KB-GE-001, KBP-042, KD-025, leucine + metformin + sildenafil, linagliptin, liraglutide, LJN-452, MBX-8025, MDV-4463, mercaptamine, MGL-3196, MGL-3745, MSDC-0602K, namacizumab, NC-101, NDI- 010976, ND-L02-s0201, NGM-282, NGM-313, NGM-386, NGM-395,
norursodeoxycholic acid, O-304, obeticholic acid, olesoxime, PAT-505, PAT-048, peg- ilodecakin, pioglitazone, Px-102, PXS-4728A, PZ-235, RDX-009 program, remogliflozin etabonate, RG-125, saroglitazar, semaglutide, simtuzumab, solithromycin, sotagliflozin, TCM-606F, TEV-45478, tipelukast (MN-001), TRX-318, UD-009, ursodeoxycholic acid, VBY-376, VBY-825, VK-2809, volixibat potassium ethanolate hydrate (SHP-626), VVP-100X, WAV-301, and ZGN-83.
In certain embodiments of the methods and pharmaceutical compositions disclosed herein, an FXR agonist is administered or formulated with the compound of Formula (I) or salts and/or forms thereof. In one embodiment, the FXR agonist is a compound having the structure of Formula (III):
Figure imgf000025_0001
, (III) or a pharmaceutically acceptable salt, co-crystal, solvate, or hydrate thereof.
In other embodiments, the FXR agonist is a compound having the structure of Formula (IV):
Figure imgf000025_0002
, (IV) or a pharmaceutically acceptable salt, co-crystal, solvate, or hydrate thereof.
The compounds of Formula (III) and Formula (IV) may be synthesized and characterized using methods known to those of skill in the art, such as those described in U.S. Publication No. 9,139,539.
In another embodiment, the compound of Formula (I) or a salt, co-crystal, solvate, or hydrate thereof is combined with an ACC inhibitor. In one embodiment, the ACC inhibitor is a compound Formula (V):
Figure imgf000026_0001
(V), or a pharmaceutically acceptable salt, co-crystal, solvate, or hydrate thereof.
Cardiovascular related diseases or conditions that can benefit from a combination treatment of ASK1 inhibitors with other therapeutic agents include, without limitation, angina, including stable angina, unstable angina (UA), exercised-induced angina, variant angina, arrhythmias, intermittent claudication, myocardial infarction including non-STE myocardial infarction (NSTEMI), heart failure including congestive (or chronic) heart failure, acute heart failure, or recurrent ischemia.
Therapeutic agents suitable for treating cardiovascular related diseases or conditions include anti-anginals, heart failure agents, antithrombotic agents, antiarrhythmic agents, antihypertensive agents, and lipid lowering agents.
The co-administration of ASK1 inhibitors with therapeutic agents suitable for treating cardiovascular related conditions allows enhancement in the standard of care therapy the patient is currently receiving.
Anti-anginals include beta-blockers, calcium channel blockers, and nitrates. Beta blockers reduce the heart's need for oxygen by reducing its workload resulting in a decreased heart rate and less vigorous heart contraction. Examples of beta-blockers include acebutolol (Sectral), atenolol (Tenormin), betaxolol (Kerlone),
bisoprolol/hydrochlorothiazide (Ziac), bisoprolol (Zebeta), carteolol (Cartrol), esmolol (Brevibloc), labetalol (Normodyne, Trandate), metoprolol (Lopressor, Toprol XL), nadolol (Corgard), propranolol (Inderal), sotalol (Betapace), and timolol (Blocadren).
Nitrates dilate the arteries and veins thereby increasing coronary blood flow and decreasing blood pressure. Examples of nitrates include nitroglycerin, nitrate patches, isosorbide dinitrate, and isosorbide-5 -mononitrate.
Calcium channel blockers prevent the normal flow of calcium into the cells of the heart and blood vessels causing the blood vessels to relax thereby increasing the supply of blood and oxygen to the heart. Examples of calcium channel blockers include amlodipine (Norvasc, Lotrel), bepridil (Vascor), diltiazem (Cardizem, Tiazac), felodipine (Plendil), nifedipine (Adalat, Procardia), nimodipine (Nimotop), nisoldipine (Sular), verapamil (Calan, Isoptin, Verelan), and nicardipine.
Agents used to treat heart failure include diuretics, ACE inhibitors, vasodilators, and cardiac glycosides. Diuretics eliminate excess fluids in the tissues and circulation thereby relieving many of the symptoms of heart failure. Examples of diuretics include hydrochlorothiazide, metolazone (Zaroxolyn), furosemide (Lasix), bumetanide (Bumex), spironolactone (Aldactone), and eplerenone (Inspra).
Angiotensin converting enzyme (ACE) inhibitors reduce the workload on the heart by expanding the blood vessels and decreasing resistance to blood flow. Examples of ACE inhibitors include benazepril (Lotensin), captopril (Capoten), enalapril
(Vasotec), fosinopril (Monopril), lisinopril (Prinivil, Zestril), moexipril (Univasc), perindopril (Aceon), quinapril (Accupril), ramipril (Altace), and trandolapril (Mavik).
Vasodilators reduce pressure on the blood vessels by making them relax and expand. Examples of vasodilators include hydralazine, diazoxide, prazosin, clonidine, and methyldopa. ACE inhibitors, nitrates, potassium channel activators, and calcium channel blockers also act as vasodilators.
Cardiac glycosides are compounds that increase the force of the heart's contractions. These compounds strengthen the pumping capacity of the heart and improve irregular heartbeat activity. Examples of cardiac glycosides include digitalis, digoxin, and digitoxin.
Antithrombotics inhibit the clotting ability of the blood. There are three main types of antithrombotics— platelet inhibitors, anticoagulants, and thrombolytic agents. Platelet inhibitors inhibit the clotting activity of platelets, thereby reducing clotting in the arteries. Examples of platelet inhibitors include acetyls alicylic acid (aspirin), ticlopidine, clopidogrel (plavix), dipyridamole, cilostazol, persantine sulfinpyrazone, dipyridamole, indomethacin, and glycoprotein llb/llla inhibitors, such as abciximab, tirofiban, and eptifibatide (Integrelin). Beta blockers and calcium channel blockers also have a platelet- inhibiting effect.
Anticoagulants prevent blood clots from growing larger and prevent the formation of new clots. Examples of anticoagulants include bivalirudin (Angiomax), warfarin (Coumadin), unfractionated heparin, low molecular weight heparin, danaparoid, lepirudin, and argatroban.
Thrombolytic agents act to break down an existing blood clot. Examples of thrombolytic agents include streptokinase, urokinase, and tenecteplase (TNK), and tissue plasminogen activator (t-PA).
Antiarrhythmic agents are used to treat disorders of the heart rate and rhythm. Examples of antiarrhythmic agents include amiodarone, quinidine, procainamide, lidocaine, and propafenone. Cardiac glycosides and beta blockers are also used as antiarrhythmic agents.
Antihypertensive agents are used to treat hypertension, a condition in which the blood pressure is consistently higher than normal. Hypertension is associated with many aspects of cardiovascular disease, including congestive heart failure, atherosclerosis, and clot formation.
Examples of antihypertensive agents include alpha- 1 -adrenergic antagonists, such as prazosin (Minipress), doxazosin mesylate (Cardura), prazosin hydrochloride (Minipress), prazosin, polythiazide (Minizide), and terazosin hydrochloride (Hytrin); beta-adrenergic antagonists, such as propranolol (Inderal), nadolol (Corgard), timolol (Blocadren), metoprolol (Lopressor), and pindolol (Visken); central alpha-adrenoceptor agonists, such as clonidine hydrochloride (Catapres), clonidine hydrochloride and chlorthalidone (Clorpres, Combipres), guanabenz Acetate (Wytensin), guanfacine hydrochloride (Tenex), methyldopa (Aldomet), methyldopa and chlorothiazide
(Aldoclor), methyldopa and hydrochlorothiazide (Aldoril); combined alpha/be ta- adrenergic antagonists, such as labetalol (Normodyne, Trandate), Carvedilol (Coreg); adrenergic neuron blocking agents, such as guanethidine (Ismelin), reserpine (Serpasil); central nervous system-acting antihypertensives, such as clonidine (Catapres), methyldopa (Aldomet), guanabenz (Wytensin); anti- angiotensin II agents; ACE inhibitors, such as perindopril (Aceon) captopril (Capoten), enalapril (Vasotec), lisinopril (Prinivil, Zestril); angiotensin- II receptor antagonists, such as Candesartan (Atacand), Eprosartan (Teveten), Irbesartan (Avapro), Losartan (Cozaar), Telmisartan (Micardis), Valsartan (Diovan); calcium channel blockers, such as verapamil (Calan, Isoptin), diltiazem (Cardizem), nifedipine (Adalat, Procardia); diuretics; direct vasodilators, such as nitroprusside (Nipride), diazoxide. (Hyperstat IV), hydralazine (Apresoline), minoxidil (Loniten), verapamil; and potassium channel activators, such as aprikalim, bimakalim, cromakalim, emakalim, nicorandil, and pinacidil.
Lipid lowering agents are used to lower the amounts of cholesterol or fatty sugars present in the blood. Examples of lipid lowering agents include bezafibrate (Bezalip), ciprofibrate (Modalim), and statins, such as atorvastatin (Lipitor), fluvastatin (Lescol), lovastatin (Mevacor, Altocor), mevastatin, pitavastatin (Livalo, Pitava) pravastatin (Lipostat), rosuvastatin (Crestor), and simvastatin (Zocor).
In this disclosure, a subject in need of the ASK1 inhibitor often suffers from secondary medical conditions such as one or more of a metabolic disorder, a pulmonary disorder, a peripheral vascular disorder, or a gastrointestinal disorder. Such patients can benefit from treatment of a combination therapy comprising administering to the patient the compounds of the invention in combination with at least one therapeutic agent.
Pulmonary disorder refers to any disease or condition related to the lungs.
Examples of pulmonary disorders include, without limitation, asthma, chronic obstructive pulmonary disease (COPD), bronchitis, and emphysema.
Examples of therapeutics agents used to treat pulmonary disorders include bronchodilators including beta2 agonists and anticholinergics, corticosteroids, and electrolyte supplements. Specific examples of therapeutic agents used to treat pulmonary disorders include epinephrine, terbutaline (Brethaire, Bricanyl), albuterol (Proventil), salmeterol (Serevent, Serevent Diskus), theophylline, ipratropium bromide (Atrovent), tiotropium (Spiriva), methylprednisolone (Solu-Medrol, Medrol), magnesium, and potassium.
Examples of metabolic disorders include, without limitation, diabetes, including type I and type II diabetes, metabolic syndrome, dyslipidemia, obesity, glucose intolerance, hypertension, elevated serum cholesterol, and elevated triglycerides.
Examples of therapeutic agents used to treat metabolic disorders include antihypertensive agents and lipid lowering agents. Additional therapeutic agents used to treat metabolic disorders include insulin, sulfonylureas, biguanides, alpha-glucosidase inhibitors, and incretin mimetics.
Peripheral vascular disorders are disorders related to the blood vessels (arteries and veins) located outside the heart and brain, including, for example peripheral arterial disease (PAD), a condition that develops when the arteries that supply blood to the internal organs, arms, and legs become completely or partially blocked as a result of atherosclerosis.
Gastrointestinal disorders refer to diseases and conditions associated with the gastrointestinal tract. Examples of gastrointestinal disorders include gastroesophageal reflux disease (GERD), inflammatory bowel disease (IBD), gastroenteritis, gastritis and peptic ulcer disease, and pancreatitis.
Examples of therapeutic agents used to treat gastrointestinal disorders include proton pump inhibitors, such as pantoprazole (Protonix), lansoprazole (Prevacid), esomeprazole (Nexium), omeprazole (Prilosec), rabeprazole; H2 blockers, such as cimetidine (Tagamet), ranitidine (Zantac), famotidine (Pepcid), nizatidine (Axid);
prostaglandins, such as misoprostol (Cytotec); sucralfate; and antacids.
In one embodiment, a compound of Formula (I) or a salt, co-crystal, solvate, or hydrate thereof as disclosed herein may be used in combination with one or more additional therapeutic agent that are being used and/or developed to treat gastrointestinal disorders. In one embodiment, a compound of Formula (I) or a salt, co-crystal, solvate, or hydrate thereof disclosed herein may be used in combination with one or more additional therapeutic agent that are being used and/or developed to treat inflammatory disorders (e.g., IBD). In such embodiments, the one or more additional therapeutic agent may be a α4β7 inhibitor, a steroid, a MMP-9 antibody, a S1P1 agonist, a TNF biologic, or any combination thereof.
In some embodiments, the one or more additional therapeutic agent may be a α4β7 integrin inhibitor, or an agent that inhibits the expression and/or activity of α4β7 integrin. The inhibitor can be small molecule or biologic. For example, the α4β7 integrin inhibitor can be natalizumab or vedolizumab.
In some embodiments, the one or more additional therapeutic agent may be a steroid, including but not limited to, corticosteroids. Corticosteroids may be administered by various routes, including intravenously (i.e., methylprednisolone, hydrocortisone), orally (i.e., prednisone, prednisolone, budesonide, dexamethasone), or topically (i.e., enema, suppository, or foam preparations).
In some embodiments, the one or more additional therapeutic agent may be an MMP9 inhibitor, or an agent that inhibits the expression and/or activity of MMP9. A representative protein sequence for MMP9 is GenBank Accession No. NP_004985. The inhibitor can be small molecule or biologic. For instance, Gu et al., The Journal of Neuroscience, 25(27): 6401-6408 (2005) discloses a specific MMP9 inhibitor, SB-3CT (CAS 292605-14-2). Further, siRNA, antisense RNA and antibodies have also been demonstrated to inhibit the expression or activity of MMP9 and are within the scope of the present disclosure. In one embodiment, an MMP9 inhibitor is a monoclonal anti- MMP9 antibody. In some embodiment, the one or more additional therapeutic agent includes an MMP9 inhibitor and a nucleoside analog such as gemcitabine.
In some embodiments, the one or more additional therapeutic agent may be a
Sphingosine 1 -Phosphate Receptor (S1P1) inhibitor, or an agent that inhibits the expression and/or activity of S1P1. The inhibitor can be small molecule or biologic. For example, the S1P1 inhibitor can be RPC1063.
In some embodiments, the one or more additional therapeutic agent may be a TNF inhibitor, or an agent that inhibits the expression and/or activity of TNF. The inhibitor can be small molecule or biologic. For example, the TNF inhibitor can be golimumab.
In some embodiments, the one or more additional therapeutic agent is being used and/or developed to treat ulcerative colitis (UC) and/or Crohn disease (CD). The agent can be a biologic or small molecule. In some embodiments, the agent is a modulator
(e.g., agonist or antagonist) of S1P1, IL-6, CX3CL1 , DHODH, <χ4, β7, JAK, TNF, CB, IL-12/IL-23, CCL20, TLR9, MAdCAM, CCR9, CXCLIO, Smad7, PDE4, MC, VLA-1, GC, GATA-3, Eotaxin, FFA2, LIGHT, FMS, MMP9, CD40, Steroid, 5-ASA,
Immunomod, STAT3, and/or EP4. In some embodiments, the JAK inhibitor is filgotinib.
Non- limiting examples of agents being used and/or developed to treat ulcerative colitis (UC) include GSK3050002 (CCL20 modulator, by GSK), GS-5745 (MMP9 modulator, by Gilead), AVX-470 (TNF modulator, by Avaxia), Bertilimumab (Eotaxin modulator, by Immune Pharma), Simponi (TNF modulator, by Johnson & Johnson and Merck), RX-10001 (by Resolvyx), IBD-98 (5-ASA modulator, by Holy Stone), SP-333 (GC modulator, by Synergy), KAG-308 (EP4 modulator, by Kaken), SB012 (GATA-3 modulator, by Sterna), AJM300 (<x4 modulator, by Ajinomoto), BL-7040 (TLR9 modulator, by BiolineRx), TAK-114 (SAT3 modulator, by Takeda), CyCol (by Sigmoid), GWP-42003 (CB modulator, by GW Pharma), ASP3291 (MC modulator, by Drais), GLPG0974 (FFA2 modulator, by Galapagos), Ozanimod (S1P1 modulator, by Receptos), ASP015K (JAK modulator, by Astellas), Apremilast (PDE4 modulator, by Celgene), Zoenasa (by Altheus), Kappaproct (TLR9 modulator, by InDex),
Phosphatidylcholine (by Dr Falk/Lipid Tx), Tofacitinib (JAk modulator, by Pfizer), Cortment (Steroid modulator, by Ferring), Uceris (Steroid modulator, by Salix), and 5- ASA modulators such as Delzicol (by Actavis), Canasa (by Aptalis), Asacol (by Actavis), Pentasa (by Shire/Ferring), Lialda (by Shire), Mezavant (by Shire), Apriso (by Salix), Colazal (by Salix), Giazo (by Salix), and Salofalk (by Dr Falk). Non-limiting examples of agents being used and/or developed to treat Crohn disease (CD) include
FFP102 (CD40 modulator, by Fast Forward), E6011 (CX3CL1 modulator, by Eisai), PF- 06480605 (by Pfizer), QBECO SSI (Immunomod modulator, by Qu Biologies), PDA- 001 (by Celgene), BI 655066 (IL-12/IL-23 modulator, by Boehringer), TNFa kinoid (TNF modulator, by Neovacs), AMG 139/MEDI-2070 (IL-12/IL-23 modulator, by AstraZeneca), PF-04236921 (IL-6 modulator, by Pfizer), Tysabri (β7 modulator, marketed by Biogen Idee in the U.S.), Cimzia (marketed by UCB in the U.S.), JNJ- 40346527 (FMS modulator, by J&J), SGX-203 (Steroid modulator, by Solgenix), CyCron (by Sigmoid), CCX507 (CCR9 modulator, by ChemoCentryx), MT1303 (S1P1 modulator, by Mitsubishi), 6-MP (by Teva), ABT-494 (JAk modulator, by Abbvie), Tofacitinib (JAk modulator, by Pfizer), TRK-170 (β7 modulator, by Toray), Mongersen (Smad7 modulator, by Celgene), RHB-104 (by Redhill), Rifaxmin EIR (by Salix), Budenofalk (by Dr Falk), and Entocort (by AstraZeneca).
Non-limiting examples of agents being used and/or developed to treat ulcerative colitis (UC) and Crohn disease (CD) include PF-06410293 (by Pfizer), SAN-300 (VLA- 1 modulator, by Salix), SAR252067 (LIGHT modulator, by Sanofi), PF-00547659
(MAdCAM modulator, by Pfizer), Eldelumab (Smad7 modulator, by BMS), AMG 181/ MEDI-7183 (β7 modulator, by Amgen/AstraZeneca), Etrolizumab (β7 modulator, by Roche), Ustekinumab (IL-12/IL-23 modulator, by J&J), Remicade (TNF modulator, by J&J and Merck), Entyvio (β7 modulator, by Takeda), Humira (TNF modulator, by Abbvie), Infliximab (by Celtrion), PF-06651600 (by Pfizer), GSK2982772 (by GSK), GLPG1205 (FFA2 modulator, by Galapagos), AG014 (by Intrexon) and Vidofludimus (DHODH modulator, by 4SC).
In some embodiments, the one or more additional therapeutic agent may be a JAK inhibitor, particularly a JAK- 1 selective inhibitor. The inhibitor can be small molecule or biologic. For example, the JAK inhibitor can be Filgotinib, GLPG0634 (JAK modulator, by Galapagos).
Patients presenting with an acute coronary disease event may exhibit conditions that benefit from administration of therapeutic agent or agents that are antibiotics, analgesics, antidepressant and anti-anxiety agents in combination with ranolazine.
Antibiotics are therapeutic agents that kill, or stop the growth of,
microorganisms, including both bacteria and fungi. Example of antibiotic agents include β-Lactam antibiotics, including penicillins (amoxicillin), cephalosporins, such as cefazolin, cefuroxime, cefadroxil (Duricef), cephalexin (Keflex), cephradine (Velosef), cefaclor (Ceclor), cefuroxime axtel (Ceftin), cefprozil (Cefzil), loracarbef (Lorabid), cefixime (Suprax), cefpodoxime proxetil (Vantin), ceftibuten (Cedax), cefdinir
(Omnicef), ceftriaxone (Rocephin), carbapenems, and monobactams; tetracyclines, such as tetracycline; macrolide antibiotics, such as erythromycin; aminoglycosides, such as gentamicin, tobramycin, amikacin; quinolones such as ciprofloxacin; cyclic peptides, such as vancomycin, streptogramins, polymyxins; lincosamides, such as clindamycin; oxazolidinoes, such as linezolid; and sulfa antibiotics, such as sulfisoxazole.
Analgesics are therapeutic agents that are used to relieve pain. Examples of analgesics include opiates and morphinomimetics, such as fentanyl and morphine; paracetamol; NSAIDs, and COX-2 inhibitors.
Antidepressant and anti-anxiety agents include those agents used to treat anxiety disorders, depression, and those used as sedatives and tranquilzers. Examples of antidepressant and anti-anxiety agents include benzodiazepines, such as diazepam, lorazepam, and midazolam; enzodiazepines; barbiturates; glutethimide; chloral hydrate; meprobamate; sertraline (Zoloft, Lustral, Apo-Sertral, Asentra, Gladem, Serlift,
Stimuloton); escitalopram (Lexapro, Cipralex); fluoxetine (Prozac, Sarafem, Fluctin, Fontex, Prodep, Fludep, Lovan); venlafaxine (Effexor XR, Efexor); citalopram (Celexa, Cipramil, Talohexane); paroxetine (Paxil, Seroxat, Aropax); trazodone (Desyrel);
amitriptyline (Elavil); and bupropion (Wellbutrin, Zyban).
Patients being treated for cardio-renal diseases such as chronic kidney disease may benefit from combination drug treatment. For example the compound of the present invention may be combined with one or more of angiotensin converting enzyme (ACE) inhibitors such as enalapril, captopril, ramipril, lisinopril, and quinapril; or angiontesin II receptor blockers (ARBs) such as losartan, olmesartan, and irbesartan; or
antihypertensive agents such as amlodipine, nifedipine, and felodipine. The benefit of combination may be increased efficacy and/or reduced side effects for a component as the dose of that component may be adjusted down to reduce its side effects while benefiting from its efficacy augmented by the efficacy of Formula (I) or a salt, co- crystal, solvate, or hydrate thereof.
Patients presenting with chronic kidney disease treatable with ASKl inhibitors such as a compound of Formula (I) may also exhibit conditions that benefit from co- administration (as directed by a qualified caregiver) of a therapeutic agent or agents that are antibiotic, analgesic, antidepressant and/or anti-anxiety agents in combination with a compound of Formula (I). Combination treatments may be administered simultaneously or one after the other within intervals as directed by a qualified caregiver or via a fixed dose (all active ingredients are combined into a single dosage form e.g. tablet) presentation of two or more active agents.
Coronary patients being treated for an acute cardiovascular disease event by administration of ASKl inhibitors often exhibit diseases or conditions that benefit from treatment with other therapeutic agents. These diseases or conditions can be of the cardiovascular nature or can be related to pulmonary disorders, metabolic disorders, gastrointestinal disorders and the like. Additionally, some coronary patients being treated for an acute cardiovascular disease event by administration of an ASKl inhibitor exhibit conditions that can benefit from treatment with therapeutic agents that are antibiotics, analgesics, and/or antidepressants and antianxiety agents.
Pharmaceutical Compositions
While it is possible for the active ingredients to be administered alone it may be preferable to present them as pharmaceutical formulations (compositions). The formulations, both for veterinary and for human use, of the invention comprise at least one active ingredient, as above defined, together with one or more acceptable carriers therefor and optionally other therapeutic ingredients. The carrier(s) must be
"acceptable" in the sense of being compatible with the other ingredients of the formulation and physiologically innocuous to the recipient thereof.
The formulations include those suitable for the foregoing administration routes. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, PA). Such methods include the step of bringing into association the active ingredient with inactive ingredients (e.g., a carrier, pharmaceutical excipient, etc.) which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
In certain embodiments, formulations suitable for oral administration are presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient.
In certain embodiments, the pharmaceutical formulations include one or more compounds of the invention together with one or more pharmaceutically acceptable carriers or excipients and optionally other therapeutic agents. Pharmaceutical formulations containing the active ingredient may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable. These excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as cellulose, microcrystalline cellulose, starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
The amount of active ingredient that is combined with the inactive ingredients to produce a dosage form will vary depending upon the host treated and the particular mode of administration. For example, in some embodiments, a dosage form for oral administration to humans contains approximately 1 to 1000 mg of active material formulated with an appropriate and convenient amount of carrier material (e.g., inactive ingredient or excipient material). In certain embodiments, the carrier material varies from about 5 to about 95% of the total compositions (weight: weight). In some embodiments, the pharmaceutical compositions described herein contain about 1 to about 800 mg, about 1 to 600 mg, about 1 to 400 mg, about 1 to 200 mg, about 1 to about 100 mg or about 1 to about 50 mg of the compound of Formula (I), or a pharmaceutically acceptable salt, co-crystal, solvate, or hydrate thereof. In some embodiments, the pharmaceutical compositions described herein contain no more than about 400 mg of the compound of Formula (I). In some embodiments, the pharmaceutical compositions described herein contain about 100 mg of the compound of Formula (I), or a salt, co- crystal, solvate, or hydrate thereof.
It should be understood that in addition to the ingredients particularly mentioned above the formulations disclosed herein may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
Veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier are further provided.
Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered orally, parenterally or by any other desired route.
Effective dose of active ingredient depends at least on the nature of the condition being treated, toxicity, whether the compound is being used prophylactically (lower doses), the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies. Routes of Administration
One or more compounds of Formula (I) (herein referred to as the active ingredients), or a pharmaceutically acceptable salt, co-crystal, solvate, or hydrate thereof, are administered by any route appropriate to the condition to be treated. Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), and the like. It will be appreciated that the preferred route may vary with for example the condition of the recipient.
An advantage of the compounds of this invention is that they are orally bioavailable and can be dosed orally. Accordingly, in one embodiment, the
pharmaceutical compositions described herein are oral dosage forms. In certain embodiments, the pharmaceutical compositions described herein are oral solid dosage forms.
In one embodiment, the oral daily dosage of Formula (I) (as a free base) is between about 0.5 mg and about 100 mg as a daily dose. In preferred embodiments, the dose is about 1 mg daily, about 2 mg daily, about 3 mg daily, about 4 mg daily, about 5 mg daily, about 6 mg daily, about 7 mg daily, about 8 mg daily, about 9 mg daily, about 10 mg daily, about 11 mg daily, about 12 mg daily, about 13 mg daily, about 14 mg daily, about 15 mg daily, about 16 mg daily, about 17 mg daily, about 18 mg daily, about 19 mg daily, about 20 mg daily, about 25 mg daily, about 30 mg daily, or about 35 mg daily. In another embodiment, the daily dose of the free base of Formula (I) is less than about 18 mg daily.
EXAMPLES
The following examples are included to demonstrate specific embodiments of the disclosure. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques to function well in the practice of the disclosure, and thus can be considered to constitute specific modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the disclosure. Crystalline forms and salts thereof were analyzed by XRPD, DSC, and TGA. XRPD patterns were collected with a PANalytical X'Pert RO MPD diffractometer generally on the following settings: 5kB, 40mA, Kal=1.5406 A, scan range 2-40 °2 Θ, step size 0.0167 °2 . All °2 Θ values are ±0.2 °2 Θ. DSC thermograms were collected on a TA Instruments Q2000 system equipped with a 50 position auto-sampler. The calibration for energy and temperature was carried out using certified indium. Typically 1 - 5 mg of each sample, in a pin-holed aluminium pan, was heated at 10 °C/min from 25 °C to 250 °C or above. A purge of dry nitrogen at 50 mL/min was maintained over the sample throughout the measurement. TGA thermograms were collected on a TA Instruments Q5000 or Q500. Typically 1 - 5 mg of each sample in, an open aluminium pan, was heated at 10 °C/min from 25 °C to 250 °C or above. The DSC values may vary by up to about 3%, or about 2% or about 1%.
List of Abbreviations and Acronyms
Abbreviation Meaning
°C Degree Celsius
Ac Acetyl
aq. Aqueous
ATP Adenosine triphosphate
br Broad
BSA Bovine serum albumin
d Doublet
DCM Dichloromethane
dd Doublet of doublets
ddd Doublet of doublet of doublets
DMA Dimethylacetamide
DME 1 ,2-Dimethoxy ethane
DMF Dimethylformamide
DMSO Dimethylsulfoxide
dt Doublet-triplet
DTT Dithiothreitol (Cleland's reagent)
EDTA Ethylenediaminetetraacetic acid
Equiv. Equivalents
ES/MS Electrospray mass spectrometry Et Ethyl
EtOAc Ethyl acetate
EtOH Ethanol (Ethyl alcohol)
FBS Fetal bovine serum
g Grams
HEPES 2- [4-(2-hydroxyethyl)piperazin- 1 - yljethanesulfonic acid
HC1 Hydrochloric acid
HPLC High pressure liquid chromatography h Hours
Hz Hertz
IBD Inflammatory bowel disease
IC50 Half-maximal inhibitory concentration z'-pr Isopropyl
/ Coupling constant (Hz)
LPS Lipopolysaccharide
M Molar
m Multiplet
M+ Mass peak
M+H+ Mass peak plus hydrogen
Me Methyl
MeCN Acetonitrile
MeOH Methanol (Methyl alcohol)
MeLi Methyllithium
MeMgX Methylmagnesium halide (Grignard reagent), where X is Fluoro, Chloro, Bromo or lodo
Me6Sn2 Hexamethyldistannane (hexamethylditin) mg Milligram
MgS04 Magnesium sulfate
MHz Megahertz
min Minute
ml/mL Milliliter
mM Millimolar mmol Millimole
NBS N-Bromosuccinimide
n- Normal
nBu/Bu n-Butyl (normal Butyl)
n-BuLi n-Butyl Lithium
NaH Sodium hydride
NaHC03 Sodium bicarbonate
nL Nanoliter
NMR Nuclear magnetic resonance
NP-40 Nonyl phenoxypolyethoxylethanol
Ph Phenyl
q Quartet
q.s. Quantity sufficient to achieve a stated function
RP Reverse phase
RRT Relative Retention Time
S Singlet
Sat. Saturated
t Triplet
THF Tetrahydrofuran
TFA Trifluoroacetic acid
General Scheme 1:
Figure imgf000041_0001
Figure imgf000041_0002
12
suitable reagent
Figure imgf000041_0003
A compound of Formula (I) may be prepared by reacting compound (6) with a suitable chlorinating reagent, including but not limited to, oxalyl chloride or thionyl chloride, to produce the corresponding acid chloride. Suitable reagents are known to those of ordinary skill in the art. Subsequent amide bond formation between the acid chloride and compound (12) gives a compound of Formula (I). Suitable reagents to facilitate amide bond formation are known to those of skill in the art.
Reaction Scheme I: Preparation of 5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-4- methylbenzoic acid hydrochloride (6)
Figure imgf000041_0004
Figure imgf000041_0005
Step 1 - Preparation of 5-amino-2-fluoro-4-methylbenzonitrile - Compound (2)
5-bromo-4-fluoro-2-methylaniline (1) (20g, 98 mmol) was dissolved in anhydrous l-methyl-2-pyrrolidinone (10 mL), and copper (I) cyanide (17.6g, 196 mmol) was added. The reaction was heated to about 180 °C for 3 about 3 hours, cooled to room temperature, and water (300 mL) and concentrated ammonium hydroxide (300 mL) added. The mixture was stirred for about 30 minutes, then extracted with ethyl acetate (3 x 200 mL). The combined extracts were dried over magnesium sulfate, and the solvent was removed under reduced pressure. The oily residue was washed with hexanes (2 x 100 mL), and the solid dissolved in dichloromethane and loaded onto a silica gel column. Eluting with 0 to 25% ethyl acetate in hexanes gradient provided 5-amino-2-fluoro-4- methylbenzonitrile. LC/MS (m/z:151 M+1).
Step 2 - Preparation of 5-(2-cyclopropyl-2-oxoethylamino)-2-fluoro-4- methylbenzonitrile - Compound (3)
5-Amino-2-fluoro-4-methylbenzonitrile (80mmol) was dissolved in anhydrous N,N-dimethylformamide (160 mL) under nitrogen, and potassium carbonate (96 mmol) and potassium iodide (88 mmol) were added as solids with stirring. The reaction was stirred for 5 minutes at room temperature and then bromomethyl cyclopropylketone (180 mmol) was added. The reaction mixture was heated to 60 °C for 3 hours, and then the solvents removed under reduced pressure. The residue was dissolved in ethyl acetate (400 mL) and washed with 400 mL of water. The organic layer was dried over magnesium sulfate, and solvent was removed under reduced pressure. The residue was re-dissolved in a minimum amount of ethyl acetate, and hexanes were added to bring the solution to 3:1 hexanes: ethyl acetate by volume. The product precipitated out of solution and was collected by filtration, to provide 5-(2-cyclopropyl-2-oxoethylamino)-2-fluoro- 4-methylbenzonitrile. LC/MS (m z : 233, M+1)
Step 3 - Preparation of 5-(4-cyclopropyl-2-mercapto-lH-imidazol-l-yl)-2-fluoro-4- methylbenzonitrile - Compound (4)
5-(2-Cyclopropyl-2-oxoethylamino)-2-fluoro-4-methylbenzonitrile (14.19 g, 61.2 mmol) was dissolved in glacial acetic acid (300 mL). Potassium thiocyanate (11.9 g, 122.4 mmol) was then added as a solid with stirring, and the reaction heated to about 110 °C for about 4 hours, at which time the solvent was removed under reduced pressure. The residue was taken up in dichloromethane (200 mL) and washed with 200 mL water. The aqueous extract was extracted with (2 x 200 mL) additional dichloromethane, the organic extracts combined and dried over magnesium sulfate. The solvent was removed under reduced pressure, the oily residue re-dissolved in ethyl acetate (50 ml), and 150 mL hexanes was added. A dark layer formed and a stir bar was added to the flask. Vigorous stirring caused the product to crash out as a peach colored solid. The product was collected by filtration, to yield 5-(4-cyclopropyl-2-mercapto-lH-imidazol-l-yl)-2- fluoro-4-methylbenzonitrile. Anal. LC/MS (m/z : 274, M+1).
Step 4 - Preparation of 5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-4- methylbenzonitrile - Compound (5)
In a 500 mL three neck round bottom flask was placed acetic acid (96 mL), water
(19 mL) and then hydrogen peroxide (30%, 7.47 mL, 65.88 mmol). The mixture was heated to about 45 °C with stirring under nitrogen while monitoring the internal temperature. 5-(4-Cyclopropyl-2-mercapto- lH-imidazol- 1 -yl)-2-fluoro-4- methylbenzonitrile (6.00 g, 21.96 mmol) was then added as a solid in small portions over about 30 minutes while maintaining an internal temperature below about 55 °C. When addition of the thioimidazole was complete the reaction was stirred for about 30 minutes at a temperature of about 45 °C, then cooled to room temperature, and a solution of 20% wt/wt sodium sulfite in water (6 mL) was slowly added. The mixture was stirred for 30 minutes and then solvents were removed under reduced pressure. The residue was suspended in 250 mL of water and 4N aqueous ammonium hydroxide was added to bring the pH to -10. The mixture was extracted with dichloromethane (3 x 200 mL), the organics combined, dried over magnesium sulfate, and the solvent was removed under reduced pressure. The residue was dissolved in 20 mL ethyl acetate, and 80 mL of hexanes were added with stirring. The solvents were decanted off and an oily residue was left behind. This process was repeated and the product, 5-(4-cyclopropyl-lH- imidazol-l-yl)-2-fluoro-4-methylbenzonitrile was obtained as a viscous oil. Anal.
LC/MS (m z : 242, M+1).
Step 5 - Preparation of 5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-4- methylbenzoic acid hydrochloride (6)
5-(4-Cyclopropyl-lH-imidazol-l-yl)-2-fluoro-4-methylbenzonitrile (11.21 g,
46.50 mmol) was placed in a round bottom flask fitted with a reflux condenser, and suspended in 38% hydrochloric acid (200 mL). The mixture was heated to about 100 °C for about 4.5 hours, and then cooled to room temperature. Solvent was removed under reduced pressure to give a pink solid, to which was added 100 mL of ethyl acetate. The solid product was collected by filtration and washed with 3 xlOO mL ethyl acetate. To the solid product was added 100 mL 10% methanol in dichloromethane, the mixture stirred, and the filtrate collected. This was repeated with 2 more 100 mL portions of 10% methanol in dichloromethane. The filtrates were combined and solvent was removed under reduced pressure, to provide crude 5-(4-cyclopropyl-lH-imidazol-l-yl)- 2-fluoro-4-methylbenzoic acid hydrochloride. No further purification was carried out. Anal. LC/MS (m/z : 261, M+1).
Reaction Scheme II:
Figure imgf000044_0001
Step 1 - Preparation of methyl 6-(((benzyloxy)carbonyl)amino)picolinate (8)
A solution of methyl 6-aminopicolinate (70 g, 460.53 mmol, 1.0 equiv.) and NaHC03 (96.71 g, 1151.32 mmol, 2.5 equiv.) in acetone/H20 (500ml/250ml) was added CbzCl (98.54 mL, 690.79 mmol, 1.5 equiv.) dropwise at about 0 °C. The mixture was allowed to warm to room temperature and stir at room temperature overnight. TLC showed the reaction was complete. Water (1500ml) was added and the mixture was stirred for about 20 mins. Then it was filtered to afford the desired product. 1H NMR (400 MHz, CDC13) δ 8.20-8.22 (1H, m), 7.75-7.84 (3H, m), 7.34-7.39 (5H, m), 5.23 (2H, s), 3.98 (3H, s).
Step 2 - Preparation of benzyl (6-(hydrazinecarbonyl)pyridin-2-yl)carbamate (9)
A solution of benzyl (6-(hydrazinecarbonyl)pyridin-2-yl)carbamate (121 g, 423.08 mmol, 1.0 equiv.) in MeOH (500ml) was added Η2ΝΝΗ2Ή20 (31.73 g, 634.62 mmol, 1.5 equiv.). The mixture was refluxed overnight. TLC showed the reaction was complete. After cooling to room temperature, the solid was precipitated. The mixture was stirred for about 0.5 h at ice-water and filtered to afford the desired product as a white solid. H NMR (400 MHz, CDC13) δ 8.66 (1H, s), 8.13-8.16 (1H, m), 7.84-7.86 (2H, m), 7.36-7.43 (6H, m), 5.24 (2H, s), 4.04 (2H, d, J = 4.4 Hz).
Step 3 - Preparation of benzyl (E)-(6-(2-((dimethylamino)methylene)hydrazine-l- carbonyl)pyridin-2-yl)carbamate (10)
A mixture of benzyl (6-(hydrazinecarbonyl)pyridin-2-yl)carbamate (170.2 g,
595.10 mmol, 1.0 equiv.) in DMF-DMA (350ml) was refluxed under Argon for 45 mins. TLC showed the reaction was complete. The mixture was cooled to room temperature and concentrated to get a solid. It was washed with ethyl acetate to get 184.2 g of the desired compound as a light yellow solid. [M+H]=342.
Step 4 - Preparation of benzyl (S)-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3- yl)pyridin-2-yl)carbamate (11)
To a solution of benzyl (E)-(6-(2-((dimethylamino)methylene)hydrazine-l- carbonyl)pyridin-2-yl)carbamate (160 g, 469.21 mmol, 1.0 equiv.) in MeCN/AcOH (800ml/200ml) was added (S)-l-fluoropropan-2-amine hydrochloride (106.04 g, 938.42 mmol, 2.0 equiv.). The mixture was refluxed under argon overnight. TLC and LCMS showed the reaction was complete. The mixture was concentrated and adjusted to pH=8 by adding NaHC03 solution. The mixture was filtered to afford the crude product as a solid. Ethyl acetate was added (400ml) and stirred under reflux for 1 h. After cooling to room temperature, it was filtered to afford the pure product. The filtrate was then concentrated to the crude product. Ethyl acetate was added (100ml) and stirred at room temperature for 10 mins. The solid was precipitated and refluxed for about 0.5 h. The mixture was filtered to afford the product. [M+H]=356.
Step 5 - Preparation of (S)-6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin- 2-amine (12)
A solution of benzyl (S)-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3- yl)pyridin-2-yl)carbamate (46.2 g, 130.14 mmol, 1.0 equiv.) in MeOH (450ml) was added Pd/C (3.17 g, 10%). The mixture was stirred at room temperature under ¾ overnight. TLC showed the reaction was complete, and it was filtered. The filtrate was concentrated and purified by column by column (MeOH: DCM=1:100-1:70) to get a white solid. The solid was washed by PE and EA (EA:PE=1:50) to afford of the desired product. [M+H]=222, H NMR (400 MHz, CDC13) δ 8.39 (1H, s), 7.66 (1H, d, J = 7.6 Hz), 7.59 (IH, t, / = 7.8 Hz), 6.57 (IH, d, / = 8.0 Hz), 5.83-5.91 (IH, m), 4.74 (IH, d, / = 3.8 Hz), 4.63 (IH, d, / = 3 Hz), 4.46 (IH, s), 1.64 (3H, d, J = 7.2 Hz).
Reaction Scheme III:
Figure imgf000047_0001
Preparation of (S)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide - Formula (I)
5-(4-Cyclopropyl-lH-imidazol-l-yl)-2-fluoro-4-methylbenzoic acid
hydrochloride (1.0 eq) was suspended in dichloromethane at room temperature and DMF (0.1 eq) was added. Oxalyl chloride (1.1 eq) was then slowly added with stirring under nitrogen. The mixture was stirred for about 5 hrs. at room temperature, and the solvent was concentrated under reduced pressure to approximately half the volume, and dichloromethane (2090 mL) was added. The solution was cooled to about 5 °C and diisopropylethyl amine was added slowly. The mixture was warmed to 15 °C and (S)-6- (4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-amine (1.1 eq) was added. The mixture was stirred at ambient temperature for 17 hours and a solution of sodium citrate tribasic dehydrate (leq) was added. IN NaOH was added and the mixture was stirred for about 10 min. The layers were separated and the organic layer was washed with water and concentrated under reduced pressure. The solids were suspended in in EtOH (1000 mL) and heated to reflux. The mixture was cooled, stirred at ambient temperature for about 3.5 hr, and filtered. The solids were rinsed with EtOH to afford (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol- 3-yl)pyridin-2-yl)-4-methylbenzamide. H NMR (400 MHz, DMSO-d6) δ 10.93 (s, 1H), 8.88 (s, 1H), 8.17 (dd, / = 8.3, 0.9 Hz, 1H), 8.04 (t, / = 8.0 Hz, 1H), 7.93 (dd, / = 7.6, 0.9 Hz, 1H), 7.70 (d, / = 1.4 Hz, 1H), 7.65 (d, / = 6.5 Hz, 1H), 7.50 (d, / = 10.7 Hz, 1H), 7.18 (d, / = 1.4 Hz, 1H), 6.04-5.89 (m, 1H), 4.78-4.71 (m, 1H), 4.67-4.59 (m, 1H), 2.25 (s, 3H), 1.88-1.81 (m, 1H), 1.51 (d, / = 6.9 Hz, 3H), 0.82-0.78 (m, 2H), 0.71-0.68 (m, 2H). LCMS-ES (m/z): [M+H]+ calcd 464.20; found 464.21.
Preparation of (S)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide - Formula (I) 5-(4-Cyclopropyl-lH-imidazol-l-yl)-2-fluoro-4-methylbenzoic acid
hydrochloride (1.0 equiv) was combined with dichloromethane (10 L/kg) and N,N- dimethylformamide (0.1 equiv). To the resulting mixture was charged oxalyl chloride (1.1 equiv) at such a rate to control the gas evolution. The reaction mixture was heated at about 35 °C until the reaction was complete. The reaction mixture was concentrated to 5 L/kg and then dichloromethane (10 L/kg) was charged. The reaction mixture was cooled to about 0 °C and then N,N-diisopropylethylamine (0.6 equiv) was charged followed by (S)-6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-amine (12) (1.1 equiv) which was charged slowly. The reaction mixture was warmed to about 22 °C and maintained until the reaction was complete. The reaction was concentrated to 5 L/kg and to this was then charged l-methyl-2-pyrrolidinone (10 L/kg). The reaction was concentrated to 10 L/kg followed by a temperature adjustment to about 65 °C. A solution of citric acid (0.5 equiv) and water (10 L/kg) was charged maintaining a temperature above about 55 °C. The pH was adjusted to about 6 using 50 wt% aqueous sodium hydroxide. The contents were heated at about 65 °C for about 1 h and then the reaction mixture was cooled to about 20 °C. The product was isolated by filtration, washed with water (2 x 5 L/kg), and dried to afford the compound of Formula (I).
Preparation 2 of (S)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide - Formula (I)
A mixture of 5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-4-methylbenzoic acid hydrochloride (1.0 equiv) in l-methyl-2-pyrrolidinone (10 L/kg) was cooled to about 5 °C. To this was charged thionyl chloride (1.1 equiv) followed by (S)-6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-amine (12) (1.0 equiv). This reaction mixture was maintained at about 25 °C until the reaction was complete and then it was heated to about 70 °C. In a separate reaction vessel sodium hydroxide (3.5 equiv), sodium citrate (0.5 equiv) and water (10 L/kg) were combined. The resulting solution was then charged to the reaction mixture. The pH was adjusted to about 6 using 50 wt% aqueous sodium hydroxide. The resulting mixture was heated at about 70 °C and then cooled to about 5 °C. The product was isolated by filtration, washed with water (2 x 5 L/kgkg), and dried to afford the compound of Formula (I). Preparation of the compound of Formula (I) Freebase Form I
Formula (I) freebase Form I was crystallized from the reaction mixture using the following procedure: 1.0 equivalent of oxalyl chloride was added to compound (6) and the mixture was agitated at about 35 °C. The solution was then distilled to dryness, and 15 volumes of DCM were added. 0.7 equivalents of DIPEA followed by 1.1 equivalents of compound (12) were then added. The mixture was agitated at about 22 °C. 10 volumes of NMP were then charged to the reactor and the DCM was distilled down to a 15 wt% level. One equivalent of sodium citrate dissolved in 2 volumes of water was then added to the reactor, at about 22 °C. Three volumes of a IN NaOH solution and 5 volumes of water were then added. The solution was then aged overnight. The resulting slurry was then filtered, and rinsed with 5 volumes of water and 2 volumes of methanol. The resulting solid was then dried at about 50 °C. The XRPD pattern of Formula (I) freebase Form I is presented in FIG. 1. The pattern contains the following characteristic peaks: (1) 8.7, 10.2, and 18.1 °2Θ ±0.2 °2Θ; (2) 15.8, 17.3, and 23.5 °2Θ ±0.2 °2Θ; (3) 15.2, 18.5, and 24.9 °2Θ ±0.2 °2Θ. FIG. 2 is the DSC thermogram of Formula (I) freebase Form I. There is a melting transition with an onset of about 223 °C. FIG. 3 is the TGA thermogram of the compound of Formula (I) freebase Form I. There is a 0.9 % weight loss between 25 and 220 °C.
Table 1: Formula (I) Freebase Form I XRPD Peak Table
Figure imgf000049_0001
No. Pos. [°2Th.] Rel. Int. [%]
10 23.0 11
11 23.2 15
12 23.5 16
13 23.9 4
14 24.9 13
15 26.0 9
16 26.1 8
17 26.6 5
18 26.9 4
19 27.8 3
20 28.3 7
21 28.6 11
22 29.2 5
23 31.4 3
24 31.9 2
25 37.2 4
Preparation of the compound of Formula (I) Freebase Form II
Formula (I) freebase Form II was crystallized from the reaction mixture using the following procedure: 1.0 equivalent of oxalyl chloride was added to compound (6) and the mixture was agitated at about 35 °C. The solution was then distilled to 5 volumes and 10 volumes of DCM were added. 0.6 equivalents of DIPEA were added, followed by 1.1 equivalents of compound (12). The mixture was agitated at about 22 °C and the DCM was distilled to dryness. 10 volumes of NMP were then added. The content of the reactor was then heated to about 70 °C and a solution of 0.5 equiv. sodium citrate, 1.5 equivalents of NaOH, and water were added at a rate allowing the internal temperature of the solution to remain above about 58 °C. The mixture was then held at about 65 °C for about 1 h, and cooled to about 20 °C over 3 h. The resulting slurry was then filtered and rinsed with two times 5 volumes of water. The solid was then dried.
Formula (I) freebase Form II can also be obtained by slurrying Formula (I) freebase Form I in a variety of solvents, including ethanol.
FIG. 4 shows the XRPD pattern of Formula (I) freebase Form II. It contains the following characteristic peaks: (1) 8.7, 10.0, and 17.0 °2Θ ±0.2 °2Θ; (2) 13.9, 21.3, and 22.8 °2Θ ±0.2 °2Θ; (3) 24.0, 28.0, and 29.1 °2Θ ±0.2 °2Θ. FIG. 5 is the DSC thermogram of Formula (I) freebase Form II. It shows a melting endotherm that has an onset at about 208 °C. FIG. 6 is the TGA thermogram of Formula (I) freebase Form II. The solid loses about 1% weight upon heating to 200 °C.
Table 2: Formula I freebase Form II XRPD Peak Table
Figure imgf000051_0001
No. Pos. [°2Th.] Rel. Int. [%]
16 20.5 11
17 21.3 72
18 22.1 9
19 22.5 27
20 22.8 100
21 23.1 14
22 23.5 5
23 24.0 28
24 25.1 6
25 25.5 10
26 26.1 8
27 26.5 11
28 27.2 6
29 28.0 31
30 28.6 7
31 29.1 49
32 29.5 17
33 31.4 6
34 32.2 5
Reaction Scheme IV:
Figure imgf000052_0001
Preparation of (S)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide succinate
A solution of succinic acid (1.0 equiv), methanol (1.1 L/kg) and acetonitrile (9.9 L/kg) was combined with (S)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide (1.0 equiv). The resulting mixture was heated to about 80 °C and then cooled to about 5 °C. The product was isolated by filtration, washed with acetonitrile (2 x 3 L/kg), and dried to afford the compound of Formula (I) as the succinate.
Preparation of Formula (I) Succinate Form I
Formula (I) succinate Form I was first obtained by slurrying about 40 mg of
Formula (I) Freebase Form II in 0.2 mL of acetonitrile with one equivalent of succinic acid, which is about 11 mg. The slurry was stirred with a stir bar at about 22 °C for approximately 16 hours. A small amount of slurry was then centrifuged and the XRPD pattern of the solid was obtained after it was dried in the vacuum oven at about 50 °C. The XRPD pattern of succinate Form I is presented in FIG. 7. The pattern contains the following characteristic peaks: (1) 7.5, 20.9, and 24.9 °2Θ ±0.2 °2Θ; (2) 15.4, 22.4, and 31.1 °2Θ ±0.2 °2Θ; (3) 14.9, 23.2, and 25.4 °2Θ ±0.2 °2Θ. FIG. 8 shows the DSC thermogram of Formula (I) succinate Form I. The solid has a melting onset at approximately 159 °C. FIG. 9 shows the TGA thermogram of Formula (I) succinate Form I. The solid loses about 0.6% weight between ambient and approximately 160 °C.
Table 3: Formula (I) Succinate Form I XRPD Peak Table
Figure imgf000053_0001
. [°2Th.] Rel. Int. [%]
18.9 3
19.3 15
19.9 5
20.2 8
20.9 100
21.8 3
22.4 17
23.2 11
23.4 9
24.3 3
24.9 79
25.4 10
25.7 5
25.9 3
26.4 4
27.0 4
27.7 2
28.2 2
28.9 9
29.2 3
30.0 9
30.3 4
31.1 18
32.2 2 No. Pos. [°2Th.] Rel. Int. [%]
32 32.7 3
33 33.9 2
34 35.9 3
35 37.9 4
Preparation of Formula (I) succinate Form II
Formula (I) succinate Form II is a methanol solvate. It was first obtained by slurrying Formula (I) succinate Form I in pure methanol at about 22 °C for
approximately 16 hours. The slurry was then centrifuged and the XRPD pattern of the solid dried at about 50 °C was obtained. Formula (I) succinate Form II is a methanol solvate
It was also obtained by slurrying Formula (I) succinate Form I in a solvent mixture composed of 4.7% water and 95.3 % methanol, as well as by slurrying Formula (I) succinate Form I in a solvent mixture composed of 20% acetonitrile, and 80 % methanol.
FIG. 10 shows the XRPD pattern of Formula (I) succinate Form II. The following peaks are characteristic of this pattern: (1) 9.8, 11.7, 23.3 (2) 15.9, 21.5, 32.0 (3) 16.3, 20.8, 24.8 °2 Θ. FIG. 11 shows the DSC thermogram of Formula (I) succinate Form II. It shows one endothermic event with an onset at about 100 °C, followed by two consecutive endothermic events with onsets at about 159, and about 162 °C. FIG. 12 is the TGA thermogram of Formula (I) succinate Form II. There are two weight losses of 4.6 and 9.2 % weight at about 100 and 200 °C, respectively.
Table 4: Formula (I) succinate Form II XRPD Peak Table
Figure imgf000055_0001
. [°2Th.] Rel. Int. [%]
11.7 26
12.3 3
13.1 3
13.7 9
14.9 9
15.4 8
15.9 24
16.1 14
16.3 22
16.6 9
16.9 4
17.3 8
18.6 5
18.8 9
19.2 3
19.6 5
20.2 8
20.8 15
21.5 12
22.8 5
23.3 100
23.4 68
23.9 12
24.2 28 No. Pos. [°2Th.] Rel. Int. [%]
28 24.5 10
29 24.8 23
30 25.7 7
31 26.1 4
32 26.5 6
33 27.0 12
34 27.6 5
35 27.9 6
36 28.4 4
37 29.0 4
38 29.8 5
39 30.1 7
40 31.5 6
41 32.0 26
42 32.1 22
43 32.5 4
44 33.1 4
45 34.0 2
46 34.3 3
47 34.9 4
48 35.4 3
49 36.5 3
Preparation of Formula (I) HCl Form I Formula (I) HCl Form I was first prepared by adding one equivalent of concentrated HCl to 47 mg of Formula (I) freebase Form II suspended in acetonitrile. The resulting solution was stirred at approximately 22°C for about 16 h.
The XRPD pattern of Formula (I) HCl Form I is presented in FIG. 13. It contains the following representative peaks (1) 9.3, 21.2, 26.2 (2) 20.9, 24.7, 27.6 (3) 23.8, 24.3, 26.6 °2 Θ. FIG. 14 is the DSC thermogram of Formula (I) HCl Form I. The melt/dehydration occurs around 152 °C. FIG. 15 is the TGA thermogram of Formula (I) HCl Form I. There are two weight losses , the first one is about 4.2 % weight, and occurs between about 30 °C and 140 °C. The second one is about 1.7 % weight and takes place between about 140 and 200 °C.
Table 5: Formula (I) HCl Form I XRPD Peak Table
Figure imgf000058_0001
No. Pos. [°2Th.] Rel. Int. [%]
16 24.3 14
17 24.7 16
18 25.7 5
19 26.2 28
20 26.6 14
21 27.6 16
22 27.8 8
23 28.0 6
24 28.8 10
25 29.1 10
26 29.4 6
27 29.8 7
28 30.2 4
29 31.1 2
30 33.1 2
31 35.9 2
Preparation of Formula (I) HCl Form II
Formula (I) HCl Form II was first prepared by slurrying a mixture of Formula (I) HCl Form I and Material A (about 1:1 w/w) in 10:90 water: acetonitrile (v/v) for 16 h at approximately 22 °C. After 16h the XRPD of the centrifuged and air-dried solid was collected.
Formula (I) HCl Form II was also prepared by slurrying Formula (I) HCl Form I in 10:90 water: acetonitrile (v/v), centrifuging it and drying it in the vacuum oven at about 50 °C. FIG. 16 shows the XRPD pattern of Formula (I) HCl Form II. It contains the following characteristic peaks: (1) 8.3, 12.0, and 26.0 °2Θ ±0.2 °2Θ; (2) 19.3, 21.7, and 24.0 °2Θ ±0.2 °2Θ; (3) 9.8, 15.4, and 28.9 °2Θ ±0.2 °2Θ. FIG. 33 is the DSC thermogram of Formula (I) HCl Form II, in which there are two endothermic events with onsets at about 135 and about 205 °C.
Table 6: Formula (I) HCl Form II XRPD Peak Table
Figure imgf000060_0001
No. Pos. [°2Th.] Rel. Int. [%]
22 26.6 12
23 27.6 9
24 28.4 8
25 28.9 19
26 29.6 6
27 30.0 8
28 30.7 8
29 30.8 9
30 32.0 5
31 33.5 12
Preparation of Formula (I) HC1 Form III
Formula (I) HC1 Material A was first prepared by adding one equivalent of 1M HCl/dioxane to Formula (I) freebase Form II suspended in acetonitrile. The resulting solution was stirred at approximately 22°C for about 16 h and XRPD of the centrifuged and dried solid was collected. FIG. 17 shows the XRPD pattern of Formula (I) HC1 Form III. It contains the following characteristic peaks: (1) 6.6, 12.9, and 19.7 °2Θ ±0.2 °2Θ; (2) 20.4, 23.4, and 24.8 °2Θ ±0.2 °2Θ; (3) 18.1, 25.4, and 26.7
°2Θ ±0.2 °2Θ. FIG. 18 is the DSC thermogram of Formula (I)-HC1 Form III. No clear thermal events can be seen.
Table 7: Formula (I) HCl Form III XRPD Peak Table
Figure imgf000061_0001
No. Pos. [°2Th.] Rel. Int. [%]
5 16.1 11
6 18.1 37
7 19.0 28
8 19.7 59
9 20.4 100
10 21.7 33
11 23.4 99
12 24.8 49
13 25.4 49
14 25.9 40
15 26.7 55
16 27.5 34
17 29.4 33
18 30.9 13
19 33.4 10
20 34.0 9
Preparation of Formula (I) L-malate
Formula (I) L-malate was first obtained by slurrying about 40 mg of Formula (I) freebase Form II in 0.2 mL of acetonitrile with one equivalent of L-malic acid, about 12 mg, for approximately 16 h at about 22 °C. The slurry was then centrifuged and an XRPD pattern of the dry solid was collected.
FIG. 19 shows the XRPD pattern of Formula (I) L-malate. It contains the following characteristic peaks: (1) 7.2, 21.9, and 24.5 °2Θ ±0.2 °2Θ; (2) 10.9, 16.6, and 19.7 °2Θ ±0.2 °2Θ; (3) 8.2, 22.8, and 27.3 °2Θ ±0.2 °2Θ. FIG. 20 shows the DSC thermogram of Formula (I) L-malate. There are two endothermic events, the first one around 23 °C, and the second one, the melting endotherm, around 110 °C. FIG. 21 shows the TGA thermogram of Formula (I) L-malate. The solid loses about 2.5 % weight when heated to about 150°C
Table 8: Formula (I) L-malate XRPD peak table
Figure imgf000063_0001
No. Pos. [°2Th.] Rel. Int. [%]
22 30.3 7
23 32.1 3
24 33.6 7
Preparation of Formula (I) Phosphate Material A
Formula (I) phosphate Material A was obtained by slurrying about 37 mg of Formula (I) freebase Form II in 0.2 mL of acetonitrile and adding one equivalent of 14.8 M phosphoric acid. After stirring for 16 h at approximately 22 °C, the solution was rotavaped and 0.2 mL of IPA was added. This solution was stirred for 10 days. A slurry was obtained and the XRPD of the centrifuged and dried solid was collected.
The XRPD pattern of Formula (I) phosphate Material A is shown in FIG. 22, it has the following characteristic peaks: (1) 6.9, 8.0, and 21.2 °2Θ ±0.2 °2Θ; (2) 22.8, 25.3, and 26.0 °2Θ ±0.2 °2Θ; (3) 19.8, 20.6, 23.2 °2Θ ±0.2 °2Θ.
Table 9: Formula (I) Phosphate Material A XRPD Peak Table
Figure imgf000064_0001
. [°2Th.] Rel. Int. [%]
15.7 42
16.4 7
16.9 27
17.5 17
17.9 15
18.6 31
18.8 25
19.1 43
19.8 46
20.6 45
21.2 47
21.4 40
21.8 26
22.1 23
22.8 63
23.2 45
23.7 33
24.3 31
25.3 100
25.8 42
26.0 46
26.4 29
26.9 28
27.7 26 No. Pos. [°2Th.] Rel. Int. [%]
36 28.0 15
37 28.7 11
38 29.1 24
39 30.6 5
40 31.7 5
41 32.5 6
42 35.0 3
43 38.4 3
Formula (I) phosphate Material B was obtained by placing about 1 mg of Formula (I) phosphate Material A on an aluminum pan and submitting it to the following relative humidity conditions inside the Dynamic Vapor Sorption (DVS) instrument: equilibrate at 25 °C and 10 % RH for 20 min, ramp to 90% RH and hold for 30 min. The XRPD pattern of the solid was collected after the DVS cycle. FIG. 23 shows the XRPD pattern of Formula (I) phosphate Material B. It contains the following characteristic peaks: (1) 13.4, 23.5, and 24.3 °2Θ ±0.2 °2Θ; (2) 21.7, 25.1, and 25.8 °2Θ ±0.2 °2Θ; (3) 17.0, 18.6, and 22.5 °2Θ ±0.2 °2Θ.
Table 10: Formula (I) Phosphate Material B XRPD Peak Table
Figure imgf000066_0001
. [°2Th.] Rel. Int. [%]
13.4 84
15.0 17
15.3 16
15.9 8
17.0 49
17.3 52
18.0 25
18.6 40
19.7 15
20.2 26
20.8 33
21.4 38
21.7 55
22.0 26
22.5 53
23.5 84
24.3 100
24.7 26
25.1 61
25.8 66
26.3 25
27.5 24
28.0 17
28.5 10 No. Pos. [°2Th.] Rel. Int. [%]
32 31.4 10
33 35.9 5
Preparation of Formula (I) sulfate
Formula (I) sulfate was first obtained by adding 1 equivalent of 98 % sulfuric acid to 38 mg of Formula (I) freebase Form II in 0.2 mL of acetonitrile. The solution was stirred for about 16 h at approximately 22 °C, then rotavaped. 0.2 mL of IPA was added and the solution was stirred for about 16 h. The slurry was then centrifuged and XRPD of the solid dried in the vacuum oven at about 50 °C was obtained.
The XRPD pattern of Formula (I) sulfate is presented in FIG. 24. It has the following characteristic peaks: (1) 7.2, 13.5, and 21.5 °2Θ ±0.2 °2Θ; (2) 16.9, 20.5, and 25.3 °2Θ ±0.2 °2Θ; (3) 18.2, 20.6, and 27.2 °2Θ ±0.2 °2Θ. FIG. 25 shows the DSC thermogram of Formula (I) sulfate. The solid melts at approximately 210 °C. FIG. 26 is the TGA thermogram of Formula (I) sulfate. It shows a weight loss of about 1.8 % around 225 °C.
Table 11: Formula (I) Sulfate Peak Table
Figure imgf000068_0001
. [°2Th.] Rel. Int. [%]
18.2 38
18.5 16
18.9 17
19.5 19
20.1 31
20.5 54
20.6 41
21.2 18
21.5 69
22.7 8
23.2 17
23.9 6
24.4 22
25.3 86
26.0 26
26.3 22
26.6 19
27.2 53
27.8 15
28.7 11
29.0 14
30.5 9
31.5 7
32.5 7 No. Pos. [°2Th.] Rel. Int. [%]
34 35.5 5
Preparation of Formula (I) Citrate Material A and Material B
Formula (I) citrate was obtained by slurrying about 42 mg of Formula (I) freebase Form II and one equivalent of citric acid, about 18 mg, in 0.2 mL of acetonitrile at about 22 °C for approximately 16 h. The XRPD pattern of the centrifuged wet solid was obtained.
FIG. 27 shows the XRPD pattern of Formula (I) citrate Material A. The pattern has the following characteristic peaks: (1) 5.6, 9.1, 16.7 °2Θ ±0.2 °2Θ; (2)12.2, 21.0, 23.0 °2Θ ±0.2 °2Θ; (2) 18.7, 24.9, 25.8 °2Θ ±0.2 °2Θ. Formula (I) citrate Material Β was obtained by drying Formula (I) citrate Material A in the vacuum oven at about 50 °C. The XRPD pattern of Formula (I) citrate Material B is shown in FIG. 28. It has the following characteristic peaks: (1) 5.6, 8.6, and 9.1 °2Θ ±0.2 °2Θ; (2) 12.2, 23.2, and 23.9 °2Θ ±0.2 °2Θ; (3) 16.8, 18.5, and 21.1 °2Θ ±0.2 °2Θ. FIG. 29 shows the DSC
thermogram of Formula (I) citrate Material B. There are two thermal events, with onsets at about 54 and about 122 °C.
Table 12a: Formula (I) Citrate Material A Peak Table
Figure imgf000070_0001
Pos. [°2Th.] Rel. Int. [%]
15.9 16
16.4 13
16.7 65
17.1 10
18.3 28
18.7 35
19.5 13
20.1 23
20.3 27
21.0 81
21.4 30
21.6 34
22.4 16
23.0 100
23.7 31
24.9 52
25.5 17
25.8 37
26.4 18
26.9 21
27.3 17
27.9 21
28.9 22
29.9 18 No. Pos. [°2Th.] Rel. Int. [%]
34 31.3 16
35 31.9 10
36 32.5 11
37 34.4 6
38 34.9 5
39 35.9 6
40 37.2 4
41 38.0 7
42 38.7 9
43 39.3 5
TczWe 12b: Formula (I) Citrate Material B XRPD Peak Table
Figure imgf000072_0001
No. Pos. [°2Th.] Rel. Int. [%]
12 16.8 48
13 18.3 57
14 18.5 59
15 20.3 40
16 21.1 76
17 21.6 63
18 23.2 96
19 23.9 100
20 25.1 45
21 26.1 43
22 27.2 31
23 28.2 21
24 28.9 20
25 29.9 17
26 32.3 2
Preparation of Formula (I) Glutarate
Formula (I) glutarate was first prepared by stirring together 55.1 mg of Formula (I) freebase Form II and one equivalent of glutaric acid, approximately 17 mg, in 0.2 mL of acetonitrile for about 16 h at approximately 22 °C.
FIG. 30 is the XRPD pattern of Formula (I) glutarate. It contains the following characteristic peaks: (1) 6.7, 8.0, and 12.1 °2Θ ±0.2 °2Θ; (2) 14.6, 20.9, and 21.8 °2Θ ±0.2 °2Θ; (3) 17.9, 19.3, and 26.2 °2Θ ±0.2 °2Θ. FIG. 31 is the DSC thermogram of Formula (I) glutarate. The onset of melting is about 150 °C.
Table 13: Formula (I) Glutarate Peak Table . [°2Th.] Rel. Int. [%]
6.7 74
8.0 42
9.8 3
12.1 100
12.8 3
13.3 3
14.6 58
14.9 5
15.2 11
16.0 4
16.6 6
17.2 4
17.9 62
18.7 8
19.3 57
19.7 14
20.1 16
20.9 65
21.8 30
22.1 10
22.5 15
22.9 6
23.9 11
24.2 27 . [°2Th.] Rel. Int. [%]
24.4 29
24.6 52
24.7 81
24.9 55
25.1 44
25.7 5
26.2 22
26.6 8
26.8 15
27.3 16
27.6 14
28.4 5
28.9 7
29.8 13
30.1 22
30.8 6
31.6 10
32.4 6
33.1 6
34.4 5
34.8 4
35.5 2
36.3 13
37.0 4 No. Pos. [°2Th.] Rel. Int. [%]
49 37.7 4
50 39.2 5
Preparation of Form (I) Methane Sulfonate
Formula (I) methanesulfonate was first prepared by adding one equivalent of methanesulfonic acid to 53.5 mg of Formula (I) freebase Form II slurried in 0.2 mL of acetone at about 22 °C. The slurry was centrifuged and the solid was dried in the vacuum oven at about 50 °C.
FIG. 32 is the XRPD pattern of Formula (I) methanesulfonate. It contains the following characteristic peaks: (1) 8.4, 19.1, and 21.2 °2Θ ±0.2 °2Θ; (2) 17.2, 19.7, and 20.2 °2Θ ±0.2 °2Θ; (3) 12.8, 21.8, and 25.2 °2Θ ±0.2 °2Θ.
Table 14: Peak Table: Form (I) Methane Sulfonate
Figure imgf000076_0001
No. Pos. [°2Th.] Rel. Int. [%]
13 19.1 64
14 19.5 53
15 19.7 57
16 20.2 78
17 21.2 100
18 21.8 49
19 22.6 49
20 23.7 30
21 24.0 42
22 24.3 58
23 25.2 48
24 26.4 38
25 26.9 28
26 28.0 19
27 28.4 20
28 29.0 20
29 29.5 17
30 34.4 11
31 38.3 10
Example 1: ASKl (Apoptosis Signal-Regulating Kinase 1) TR-FRET Kinase Assay (Biochemical IC50)
HTRF detection was used to measure the phosphorylation level of a biotinylated peptide substrate by the ASKl kinase. This is a competitive, time-resolved fluorescence resonance energy transfer (TR-FRET) immunoassay, based on HTRF® KinEASE™- STK manual from Cisbio. The compound of Formula (I), 1 μΜ STK3 peptide substrate, 4 nM of ASKl kinase are incubated with 10 mM MOP buffer, pH. 7.0 containing 10 mM Mg-acetate, 0.025 % NP-40, 1 mM DTT, 0.05% BSA and 1.5% glycerol for about 30 minutes then 100 μΜ ATP were added to start the kinase reaction and incubated for 3 hr. Peptide antibody labeled with IX Eu3+ Cryptate buffer containing 10 mM EDTA and 125 nM Streptavidin XL665 were added to stop the reaction and phosphorylated peptide substrate is detected using Envision 2103 Multilabeled reader from PerkinElmer. The fluorescence was measured at 615 nm (Cryptate) and 665 nm (XL665) and a ratio of 665 nm/615 nm is calculated for each well. The resulting TR-FRET level (a ratio of 665 nm/615 nm) is proportional to the phosphorylation level. Under these assay conditions, the degree of phosphorylation of peptide substrate was linear with time and
concentration for the enzyme. The assay system yielded consistent results with regard to Km and specific activities for the enzyme. For inhibition experiments (IC50 values), activities were performed with constant concentrations of ATP, peptide and several fixed concentrations of inhibitors. Staurosporine, the nonselective kinase inhibitor, was used as the positive control. All enzyme activity data is reported as an average of
quadruplicate determination. The compound of Formula (I) inhibited ASKl with an IC50 of 2.4 nM.
Example 2: ASKl Cellular and Whole Blood Potency
To further characterize cellular potency of the compound of Formula (I), auranofin-induced ASKl activation (Thr845 autophosphorylation) and downstream activation of p38 and JNK MAP kinases were evaluated in HK-2 human renal proximal tubular epithelial cells, and Neonatal Rat Ventricular Myocytes (NRVMs). In resting cells, ASKl is bound and repressed by the reduced form of the antioxidant protein thioredoxin. Auranofin agonizes the ASKl pathway by causing the oxidation of thioredoxin, dissociation of the thioredoxin/ASKl complex, and ASKl
autophosphorylation and activation. Activated ASKl phosphorylates and activates downstream MAPKKs, including MAPKK3, 4, 6, and 7, which in turn phosphorylate p38 and JNK. This is followed by increased phosphorylation of the p38 and JNK substrates ATF2 and c-Jun, respectively; and increased expression of a variety of known ATF2/cJun/APl target genes including the chemokines CXCL1/KC. All of these auranofin-induced signaling events are blocked by ASKl inhibitors, demonstrating that ASKl drives the chain of signaling events following auranofin stimulation. Using phosphorylated p38 as a surrogate marker for ASKl activity in serum free environment, EC50 of the compound of Formula (I) was determined in HK-2 as 9.2 nM. In NRVMs, the compound of Formula (I) inhibited auranofin- induced phosphorylated p38 with the mean EC50 of 6.4 nM.
The CXCL1 assay is conducted in human whole blood and measures the ability of a test compound to inhibit production of the CXCL1 cytokine after auranofin stimulation of the ASK1 pathway. The compound of Formula (I) inhibited auranofin- induced CXCL1 production in whole blood with an average EC50 of 80.8 nM.
In conclusion, the compound of Formula (I) inhibits ASK1 activity in multiple cellular assays with or without human whole blood in the medium. The compound of Formula (I) inhibits endogenous ASK1 downstream targets such as phos-p38 and the production of CXCL-1.
Example 3: Determination of Kd
Kinase assays
Kinase-tagged T7 phage strains were prepared in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage and incubated with shaking at 32°C until lysis. The lysates were centrifuged and filtered to remove cell debris. The remaining kinases were produced in HEK-293 cells and subsequently tagged with DNA for qPCR detection. Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for about 30 minutes at room temperature to generate affinity resins for kinase assays.
The liganded beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1% (bovine serum albumin), 0.05% Tween 20, 1 mM
DTT(dithiothreitol)) to remove unbound ligand and to reduce non-specific binding. Binding reactions were assembled by combining kinases, liganded affinity beads, and test compounds in lx binding buffer (20% SeaBlock, 0.17x PBS, 0.05% Tween 20, 6 mM DTT). All reactions were performed in polystyrene 96-well plates in a final volume of 0.135 mL. The assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (lx PBS, 0.05% Tween 20). The beads were then re-suspended in elution buffer (lx PBS, 0.05% Tween 20, 0.5 μΜ non-biotinylated affinity ligand) and incubated at room temperature with shaking for about 30 minutes. The kinase concentration in the eluates was measured by qPCR. Binding constants (Kds) were calculated with a standard dose-response curve using the Hill equation.
Result
The compound of Formula (I) exhibited a ¾ of 0.17 nM. This data suggests that the compound of Formula (I) binds potently to ASK1 receptor in the absence of ATP.
Example 4: CYP-Mediated Metabolism in Cryo-preserved Human Microsomes
To study the mechanism of CYP-mediated metabolism, compounds (1 μΜ) were incubated with human liver microsomes at 2, 12, 25, 45, and 65 minutes time points with NADPH co-factors. Samples were collected at the various time points and the reactions stopped by adding 10 % MeOH, 90 % acetonitrile, 0.1% Formic Acid, and 50 nM internal standard. The concentration of the compound in each sample was analyzed using LC/MS/MS. The disappearance half-life of the compound in hepatocyte suspension was determined by fitting the concentration-time data with a monophasic exponential equation. The data was also scaled up to represent intrinsic hepatic clearance and/or total hepatic clearance.
Appreciable metabolism of the compound of Formula (I) was not observed, whereas considerable loss of parent via N-dealkylation was observed for N-(6-(4-(tert- butyl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro- 4-methylbenzamide; 5-(4-cyclopropyl- lH-imidazol- l-yl)-N-(6-(4-(cyclopropylmethyl)- 4H-l,2,4-triazol-3-yl)pyridin-2-yl)-2-fluoro-4-methylbenzamide; and N-(6-(4-benzyl- 4H-l,2,4-triazol-3-yl)pyridin-2-yl)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-4- methylbenzamide.
Example 5: Pharmacokinetics of Formula (I) in Cynomolgus Monkey
Cynomolgus Monkeys
The compound of Formula (I) was formulated in 5% dextrose at 0.50 mg/mL for
IV infusion. Each dosing group consisted of 3 male, cynomolgus monkeys. At dosing, the animals weighed -4.4 kg. The animals were fasted overnight prior to dose administration and up to 4 hr after dosing. For the IV infusion group, the test article was administered by intravenous infusion over 30 min. The rate of infusion was adjusted according to the body weight of each animal to deliver a dose of 1 mg/kg. Serial venous blood samples (approximately 0.3 mL each) were taken at specified time points after dosing from each animal (see Appendix I for the time points). The blood samples were collected into Vacutainer™ tubes containing EDTA-K2 as the anti-coagulant and were immediately placed on wet ice pending centrifugation for plasma. An LC/MS/MS method was used to measure the concentration of the compound of Formula (I) in plasma. The terminal ti/2 was -9.8 h and the mean residence time (MRT) was -9.4 h. The terminal t^ for 5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-isopropyl-4H- l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide is -3.16 h.
Example 6: Kinetic Solubility Studies
The required amount of solid compound of Formula (I) (free base or salt form) was added to a reaction vessel equipped with a stir bar. The buffer solution was then added to the stirring vessel and the reaction timer was initiated. Samples of the solution were taken at discreet time points (5, 10, 20, 30, 60, and 120 minutes) and filtered through a GHP acrodisc filter (0.45 um). The resulting filtrate was diluted 1: 1 with 65/35 v/v ¾0 0.1% TFA/ACN diluent and the amount of the compound of Formula (I) was quantified by UPLC with comparison to a reference standard. After 20 hours of stirring, a final sample was taken to analyze the equilibration concentration of each salt form. The remaining solids from the reaction vessel were collected and analyzed by X- ray powder diffraction to determine the final solid form.
The data in FIG. 34 and FIG. 35 show that the salts of the compound of Formula (I) have improved kinetic solubility over the freebase.
Example 7: Stability Studies
Crystalline lots were prepared of three salts of Formula (I) (succinate Form I, HC1 Form I and malate Form I) and each was placed in a 40°C/75% RH stability chamber for one month in an open container. After one month, samples were tested for appearance, assay and impurities by HPLC, water content, and by X-ray powder diffraction (XRPD) (Table 15). The HC1 Form I was chemically and physically stable however, it sorbs a lot of water. The malate salt was less stable and dissociated back to the free base after one month of storage at 40°C/75% RH. The succinate Form I was physically and chemically stable under open conditions and was non-hygroscopic. Table 15.
Figure imgf000081_0001
Initial 1 Month Initial 1 Month Initial 1 Month
Off-White
White Off-White Off-White Off-White White
Appearance Powder
Powder Powder Powder Powder Powder
(sticky)
Assay 100.6 100.3 96.4 98.0 100.4 102.1
Water 0.0 0.1 2.7 3.8 1.3 5.8
Total
0.05 0.06 0.05 0.06 0.12 0.22 Impurities*
RRT 0.55 ND ND 0.03 0.04 ND 0.06
RRT 0.67 ND ND 0.03 0.03 ND ND
RRT 0.79 ND ND ND ND ND 0.10
RRT 0.82 ND ND ND ND 0.03 ND
RRT 0.85 ND ND 0.03 0.03 ND ND
RRT 0.97 0.05 0.06 0.05 0.06 0.06 0.06
RRT 0.98 0.04 0.04 0.04 0.03 0.04 0.03
RRT 1.21 ND ND ND ND 0.06 ND
No change No change in
Freebase
XRPD Crystalline in XRD Crystalline XRD from Crystalline
Form II from initial initial
* Includes individual impurities > 0.05%
The crystalline succinate Form I was also stable under mechanical stress. The crystal has plate morphology and thus poses less risk to flow compared to needles. The powder was then subjected to grinding for one minute and five minutes with a mortar and pestle and analyzed by X-ray powder diffraction (XRPD). Compression was performed at 2000 psi on a Carver press. The morphology was then characterized by light microscopy (Table 16).
Table 16.
Figure imgf000082_0001
Formula (I) Succinate
159 No change in XRPD pattern Plate-like Form I
Formula (I) HC1 Form I 136 No change in XRPD pattern Needles
Example 8: Drug-drug Interaction
The following data shows that salts of Formula (I) reduce the acid-suppressive effect that is observed with free base. Table 17 compares Formula (I) formulated as free base, Form II, in a tablet, and Formula (I) salts formulated as a powder in a capsule (PIC) in a 1:2 ratio of Formula (I):pre-gelatinized starch (PGS). The tablets were administered as a fixed dose of 6 mg into either pentagastrin or famotidine pre-treated male beagle dogs. Formula (I) free base shows a ~12x decrease in plasma exposure when dosed in famotidine pre-treated dogs suggesting that Formula (I) free base will have a significant drug-drug interaction when used to treat patients who use acid-suppressive agents such as famotidine or omeprazole. However, salts of Formula (I) abrogate this effect and show ~2x difference between pentagastrin and famotidine pre-treatment.
Table 17.
Figure imgf000083_0001

Claims

1. A compound of Formula (I):
Figure imgf000084_0001
or a pharmaceutically acceptable salt, co-crystal, solvate, or hydrate thereof.
2. The compound of claim 1, wherein the compound is a freebase.
3. The compound of claim 1, wherein the compound is a salt selected from succinate, hydrochloric acid, phosphate, malate, sulfate, citrate, glutarate, and methane sulfonate.
4. Amorphous (S)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide.
5. Crystalline Form I of (S)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide.
6. The crystalline Form I of claim 5, wherein the compound is characterized by an X- ray powder diffractogram having peaks at 8.7, 10.2, and 18.1 °2Θ ±0.2 °2Θ.
7. The crystalline Form I of either claim 5 or 6, wherein the (S)-5-(4-cyclopropyl-lH- imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin- 2-yl)-4-methylbenzamide is characterized by an X-ray powder diffractogram having peaks at 15.8, 17.3, and 23.5 °2Θ ±0.2 °2Θ.
8. The crystalline Form I of any one of claims 5 to 7, wherein the (S)-5-(4-cyclopropyl- lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3- yl)pyridin-2-yl)-4-methylbenzamide is characterized by an X-ray powder diffractogram having peaks at 15.2, 18.5, and 24.9 °2Θ ±0.2 °2Θ.
9. The crystalline Form I of any one of claims 5 to 8, wherein the (S)-5-(4-cyclopropyl- lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3- yl)pyridin-2-yl)-4-methylbenzamide is characterized by an X-ray powder diffractogram that is substantially as shown in FIG. 1.
10. The crystalline Form I of any one of claims 5 to 9, wherein the (S)-5-(4-cyclopropyl- lH midazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3- yl)pyridin-2-yl)-4-methylbenzamide is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 2.
11. Crystalline Form II of (S)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide.
12. The crystalline Form II of claim 11, wherein the (S)-5-(4-cyclopropyl-lH-imidazol- l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4- methylbenzamide is characterized by an X-ray powder diffractogram having peaks at 8.7, 10.0, and 17.0 °2Θ ±0.2 °2Θ.
13. The crystalline Form II of either claims 11 to 12, wherein the (S)-5-(4-cyclopropyl- lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3- yl)pyridin-2-yl)-4-methylbenzamide is characterized by an X-ray powder diffractogram having peaks at 13.9, 21.3, and 22.8 °2Θ ±0.2 °2Θ.
14. The crystalline Form II of any one of claims 11 to 13, wherein the (S)-5-(4- cyclopropyl- lH-imidazol- l-yl)-2-fluoro-N-(6-(4-( 1 -fluoropropan-2-yl)-4H- 1 ,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide is characterized by an X-ray powder diffractogram having peaks at 24.0, 28.0, and 29.1 °2Θ ±0.2 °2Θ.
15. The crystalline Form II of any one of claims 11 to 14, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide is characterized by an X-ray powder diffractogram substantially as shown in FIG. 4.
16. The crystalline Form II of any one of claims 11 to 15, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 5.
17. Crystalline (S)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide succinate Form I.
18. The crystalline succinate Form I of claim 17, wherein the (S)-5-(4-cyclopropyl-lH- imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin- 2-yl)-4-methylbenzamide succinate is characterized by an X-ray powder
diffractogram having peaks at 7.5, 20.9, and 24.9 °2Θ ±0.2 °2Θ.
19. The crystalline succinate Form I of either claim 17 or 18, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide succinate is characterized by an X-ray powder diffractogram having peaks at 15.4, 22.4, and 31.1 °2Θ ±0.2 °2Θ.
20. The crystalline succinate Form I of any one of claims 17 to 19, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide succinate is characterized by an X-ray powder diffractogram having peaks at 14.9, 23.2, and 25.4 °2Θ ±0.2 °2Θ.
21. The crystalline succinate Form I of any one of claims 17 to 20, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide succinate is characterized by an X-ray powder diffractogram substantially as shown in FIG. 7.
22. The crystalline succinate Form I of any one of claims 17 to 21, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide succinate is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 8.
23. Crystalline (S)-5-(4-cyclopropyl- lH-imidazol- 1 -yl)-2-fluoro-N-(6-(4-( 1 - fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide succinate
Form II.
24. The crystalline succinate Form II of claim 23, wherein the (S)-5-(4-cyclopropyl-lH- imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin- 2-yl)-4-methylbenzamide succinate is characterized by an X-ray powder
diffractogram having peaks at 9.8, 11.7, and 23.3 °2Θ ±0.2 °2Θ.
25. The crystalline succinate Form II of either claim 23 or 24, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide succinate is characterized by an X-ray powder diffractogram having peaks at 15.9, 21.5, and 32.0 °2Θ ±0.2 °2Θ.
26. The crystalline succinate Form II of any one of claims 23 to 25, wherein the (S)-5-(4- cyclopropyl- lH-imidazol- l-yl)-2-fluoro-N-(6-(4-( 1 -fluoropropan-2-yl)-4H- 1 ,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide succinate is characterized by an X-ray powder diffractogram having peaks at 16.3, 20.8, and 24.8 °2Θ ±0.2 °2Θ.
27. The crystalline succinate Form II of any one of claims 23 to 26, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide succinate is characterized by an X-ray powder diffractogram substantially as shown in FIG. 10.
28. The crystalline succinate Form II of any one of claims 23 to 27, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide succinate is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 11.
29. Crystalline (S)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HC1 Form I.
30. The crystalline HC1 Form I of claim 29, wherein the (S)-5-(4-cyclopropyl-lH- imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin- 2-yl)-4-methylbenzamide HC1 is characterized by an X-ray powder diffractogram having peaks at 9.3, 21.2, and 26.2 °2Θ ±0.2 °2Θ.
31. The crystalline HC1 Form I of either claim 29 or 30, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HC1 is characterized by an X-ray powder diffractogram having peaks at 20.9, 24.7, and 27.6 °2Θ ±0.2 °2Θ.
32. The crystalline HC1 Form I of any one of claims 29 to 31, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HC1 is characterized by an X-ray powder diffractogram having peaks at 23.8, 24.3, and 26.6 °2Θ ±0.2 °2Θ.
33. The crystalline HC1 Form I of any one of claims 29 to 32, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HC1 is characterized by an X-ray powder diffractogram substantially as shown in FIG. 13.
34. The crystalline HC1 Form I of any one of claims 29 to 33, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HCl is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 14.
35. Crystalline (S)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HCl Form II.
36. The crystalline HCl Form II of claim 35, wherein the (S)-5-(4-cyclopropyl-lH- imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin- 2-yl)-4-methylbenzamide HCl is characterized by an X-ray powder diffractogram having peaks at 8.3, 12.0, and 26.0 °2Θ ±0.2 °2Θ.
37. The crystalline HCl Form II of either claim 35 or 36, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HCl is characterized by an X-ray powder diffractogram having peaks at 19.3, 21.7, and 24.0 °2Θ ±0.2 °2Θ.
38. The crystalline HCl Form II of any one of claims 35 to 37, wherein the (S)-5-(4- cyclopropyl- 1 H-imidazol- 1 -yl)-2-fluoro-N-(6-(4-( 1 -fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HCl is characterized by an X-ray powder diffractogram having peaks at 9.8, 15.4, and 28.9 °2Θ ±0.2 °2Θ.
39. The crystalline HCl Form II of any one of claims 35 to 38, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HCl is characterized by an X-ray powder diffractogram substantially as shown in FIG. 16.
40. The crystalline HCl Form II of any one of claims 35 to 39, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HCl is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 33.
41. Crystalline (S)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HCl Form III.
42. The crystalline HCl Form III of claim 41, wherein the (S)-5-(4-cyclopropyl-lH- imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin- 2-yl)-4-methylbenzamide HCl is characterized by an X-ray powder diffractogram having peaks at 6.6, 12.9, and 19.7 °2Θ ±0.2 °2Θ.
43. The crystalline HCl Form III of either claim 41 or 42, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HCl is characterized by an X-ray powder diffractogram having peaks at 20.4, 23.4, and 24.8 °2Θ ±0.2 °2Θ.
44. The crystalline HCl Form III of any one of claims 41 to 43, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HCl is characterized by an X-ray powder diffractogram having peaks at 18.1, 25.4, and 26.7 °2Θ ±0.2 °2Θ.
45. The crystalline HCl Form III of any one of claims 41 to 44, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HCl is characterized by an X-ray powder diffractogram substantially as shown in FIG. 17.
46. The crystalline HCl Form III of any one of claims 41 to 45, wherein the (S)-5-(4- cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l-fluoropropan-2-yl)-4H- 1,2,4- triazol-3-yl)pyridin-2-yl)-4-methylbenzamide HCl is characterized by a differential scanning calorimetry (DSC) curve substantially as shown in FIG. 18.
47. A pharmaceutical composition comprising a therapeutically effective amount of the crystalline (S)-5-(4-cyclopropyl-lH-imidazol-l-yl)-2-fluoro-N-(6-(4-(l- fluoropropan-2-yl)-4H-l,2,4-triazol-3-yl)pyridin-2-yl)-4-methylbenzamide of any one of claims 1 to 45 and a pharmaceutically acceptable excipient.
48. The pharmaceutical composition of claim 47, further comprising one, two, or three additional therapeutic agents.
49. The pharmaceutical composition of claim 48, wherein the pharmaceutical
composition comprises one additional therapeutic agent that is an FXR agonist.
50. The pharmaceutical composition of claim 49, wherein the FXR agonist is a
compound of Formula (III):
Figure imgf000090_0001
(III), or a pharmaceutical salt, co- crystal, solvate, or hydrate thereof.
51. The pharmaceutical composition of claim 49, wherein the FXR agonist is a
compound of Formula (IV):
Figure imgf000090_0002
(IV), or a pharmaceutically acceptable salt, co-crystal, solvate, or hydrate thereof.
52. The pharmaceutical composition of claim 49, wherein the pharmaceutical
composition comprises one additional therapeutic agent that is an inhibitor of ACC.
53. The composition of claim 52, wherein the ACC inhibitor is a compound of Formula (V):
Figure imgf000090_0003
or a pharmaceutically acceptable salt, co-crystal, solvate, or hydrate thereof.
54. A method of inhibiting ASK1 with a compound or pharmaceutical composition of any one of claims 1 to 53.
55. A method of treating a condition mediated by ASK1, the method comprising administering a compound or pharmaceutical composition of any one of claims 1 to 53 to a patient in need thereof.
56. A method of treating a condition selected from non-alcoholic steatohepatitis,
alcoholic hepatitis, pulmonary arterial hypertension, heart failure with preserved ejection fraction, and diabetic kidney disease, the method comprising administering a therapeutically effective amount of a compound or pharmaceutical composition of any one of claims 1 to 53 to a patient in need thereof.
57. A method of treating non-alcoholic steatohepatitis, the method comprising
administering therapeutically effective amount of a compound or pharmaceutical composition of any one of claims 1 to 53 to a patient in need thereof.
58. A method of treating fibrosis, the method comprising administering therapeutically effective amount of a compound or composition of any one of claims 1 to 53 to a patient in need thereof.
59. The compound of claim 1, wherein the compound is Formula (la):
Figure imgf000091_0001
PCT/US2018/021334 2017-03-14 2018-03-07 Apoptosis signal-regulating kinase inhibtor WO2018169742A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762471300P 2017-03-14 2017-03-14
US62/471,300 2017-03-14

Publications (1)

Publication Number Publication Date
WO2018169742A1 true WO2018169742A1 (en) 2018-09-20

Family

ID=61750538

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/021334 WO2018169742A1 (en) 2017-03-14 2018-03-07 Apoptosis signal-regulating kinase inhibtor

Country Status (6)

Country Link
AR (1) AR111178A1 (en)
BR (1) BR102018004862A2 (en)
JO (1) JOP20180017A1 (en)
TW (1) TW201840551A (en)
UY (1) UY37631A (en)
WO (1) WO2018169742A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019213244A1 (en) 2018-05-02 2019-11-07 Enanta Pharmaceuticals, Inc. Tetrazole containing apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US10597382B2 (en) 2017-08-28 2020-03-24 Enanta Pharmaceuticals, Inc. Tetrazole containing apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US10683289B2 (en) 2018-05-02 2020-06-16 Enanta Pharmaceuticals, Inc. Apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US10683279B2 (en) * 2017-05-12 2020-06-16 Enanta Pharmaceuticals, Inc. Apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US10968199B2 (en) 2018-08-22 2021-04-06 Enanta Pharmaceuticals, Inc. Cycloalkyl-containing apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
USRE48711E1 (en) 2009-07-13 2021-08-31 Gilead Sciences, Inc. Apoptosis signal-regulating kinase inhibitors
US11345699B2 (en) 2018-11-19 2022-05-31 Enanta Pharmaceuticals, Inc. Apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US11466033B2 (en) 2019-03-25 2022-10-11 Enanta Pharmaceuticals, Inc. Substituted pyridines as apoptosis signal-regulating kinase 1 inhibitors
WO2023125904A1 (en) * 2021-12-30 2023-07-06 苏州晶云药物科技股份有限公司 Crystal form of azacyclobutyl nicotinic acid compound and preparation method therefor
WO2024027593A1 (en) * 2022-07-30 2024-02-08 中国医学科学院阜外医院 Use of jun inhibitor in preparing medicament for treating heart failure with preserved ejection fraction

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110818683B (en) * 2018-08-10 2023-04-14 中国科学院上海药物研究所 2-pyridine substituted urea structure small molecule compound and synthesis and application thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8378108B2 (en) 2009-07-13 2013-02-19 Gilead Sciences, Inc. Apoptosis signal-regulating kinase inhibitors
US8742126B2 (en) 2012-01-27 2014-06-03 Gilead Sciences, Inc. Apoptosis signal-regulating kinase inhibitor
US9139539B2 (en) 2011-07-13 2015-09-22 Gilead Sciences, Inc. FXR (NR1H4) binding and activity modulating compounds
US20150342943A1 (en) 2014-06-03 2015-12-03 Gilead Sciences, Inc. Methods of treating liver disease
US20160166556A1 (en) 2014-08-13 2016-06-16 Gilead Sciences, Inc. Methods of treating pulmonary hypertension
WO2016105453A1 (en) * 2014-12-23 2016-06-30 Gilead Sciences, Inc. Solid forms of an ask1 inhibitor

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8378108B2 (en) 2009-07-13 2013-02-19 Gilead Sciences, Inc. Apoptosis signal-regulating kinase inhibitors
US8552196B2 (en) 2009-07-13 2013-10-08 Gilead Sciences, Inc. Apoptosis signal-regulating kinase inhibitors
US8598360B2 (en) 2009-07-13 2013-12-03 Gilead Sciences, Inc. Apoptosis signal-regulating kinase inhibitors
US9067933B2 (en) 2009-07-13 2015-06-30 Gilead Sciences, Inc. Apoptosis signal-regulating kinase inhibitors
US9586932B2 (en) 2009-07-13 2017-03-07 Gilead Sciences, Inc. Apoptosis signal-regulating kinase inhibitors
US9139539B2 (en) 2011-07-13 2015-09-22 Gilead Sciences, Inc. FXR (NR1H4) binding and activity modulating compounds
US8742126B2 (en) 2012-01-27 2014-06-03 Gilead Sciences, Inc. Apoptosis signal-regulating kinase inhibitor
US9254284B2 (en) 2012-01-27 2016-02-09 Gilead Sciences, Inc. Apoptosis signal-regulating kinase inhibitor
US9333197B2 (en) 2012-01-27 2016-05-10 Gilead Sciences, Inc. Apoptosis signal-regulating kinase inhibitor
US20150342943A1 (en) 2014-06-03 2015-12-03 Gilead Sciences, Inc. Methods of treating liver disease
US20160166556A1 (en) 2014-08-13 2016-06-16 Gilead Sciences, Inc. Methods of treating pulmonary hypertension
WO2016105453A1 (en) * 2014-12-23 2016-06-30 Gilead Sciences, Inc. Solid forms of an ask1 inhibitor

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", MACK PUBLISHING CO.
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 292605-14-2
DE B, I ET AL.: "Temporal trends in the prevalence of diabetic kidney disease in the United States", JAMA, vol. 305, no. 24, 22 June 2011 (2011-06-22), pages 2532 - 2539
FOSTER: "Deuterium Isotope Effects in Studies of Drug Metabolism", TRENDS PHARMACOL. SCI., vol. 5, no. 12, 1984, pages 524 - 527, XP025943358, DOI: doi:10.1016/0165-6147(84)90534-0
GU ET AL., THE JOURNAL OF NEUROSCIENCE, vol. 25, no. 27, 2005, pages 6401 - 6408
HATTORI, K.; NAGURO, I.; RUNCHEL, C.; ICHIJO, H., CELL COMM. SIGNAL., vol. 7, 2009, pages 1 - 10
ICHIJO, H.; NISHIDA, E.; IRIE, K.; DIJKE, P. T.; SAITOH, M.; MORIGUCHI, T.; MATSUMOTO, K.; MIYAZONO, K.; GOTOH, Y., SCIENCE, vol. 275, 1997, pages 90 - 94
KLEINER ET AL.: "Design and validation of a histological scoring system for nonalcoholic fatty liver disease", HEPATOLOGY, vol. 41, 2005, pages 1313 - 1321, XP055123202, DOI: doi:10.1002/hep.20701
NAGAI, H.; NOGUCHI, T.; TAKEDA, K.; ICHIJO, I., J. BIOCHEM. MOL. BIOL., vol. 40, 2007, pages 1 - 6
TAKEDA, K.; NOGUCHI, T.; NAGURO, I.; ICHIJO, H., ANNU. REV. PHARMACOL. TOXICOL., vol. 48, 2007, pages 1 - 8.27

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE48711E1 (en) 2009-07-13 2021-08-31 Gilead Sciences, Inc. Apoptosis signal-regulating kinase inhibitors
US10988458B2 (en) 2017-05-12 2021-04-27 Enanta Pharmaceuticals, Inc. Apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US11560368B2 (en) 2017-05-12 2023-01-24 Enanta Pharmaceuticals, Inc. Apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US10683279B2 (en) * 2017-05-12 2020-06-16 Enanta Pharmaceuticals, Inc. Apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US10597382B2 (en) 2017-08-28 2020-03-24 Enanta Pharmaceuticals, Inc. Tetrazole containing apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US11008304B2 (en) 2018-05-02 2021-05-18 Enanta Pharmaceuticals, Inc. Tetrazole containing apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
WO2019213244A1 (en) 2018-05-02 2019-11-07 Enanta Pharmaceuticals, Inc. Tetrazole containing apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US10683289B2 (en) 2018-05-02 2020-06-16 Enanta Pharmaceuticals, Inc. Apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US11834436B2 (en) 2018-05-02 2023-12-05 Enanta Pharmaceuticals, Inc. Tetrazole containing apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US10968199B2 (en) 2018-08-22 2021-04-06 Enanta Pharmaceuticals, Inc. Cycloalkyl-containing apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US11345699B2 (en) 2018-11-19 2022-05-31 Enanta Pharmaceuticals, Inc. Apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
US11466033B2 (en) 2019-03-25 2022-10-11 Enanta Pharmaceuticals, Inc. Substituted pyridines as apoptosis signal-regulating kinase 1 inhibitors
WO2023125904A1 (en) * 2021-12-30 2023-07-06 苏州晶云药物科技股份有限公司 Crystal form of azacyclobutyl nicotinic acid compound and preparation method therefor
WO2024027593A1 (en) * 2022-07-30 2024-02-08 中国医学科学院阜外医院 Use of jun inhibitor in preparing medicament for treating heart failure with preserved ejection fraction

Also Published As

Publication number Publication date
TW201840551A (en) 2018-11-16
UY37631A (en) 2018-09-28
BR102018004862A2 (en) 2019-01-02
AR111178A1 (en) 2019-06-12
JOP20180017A1 (en) 2019-01-30

Similar Documents

Publication Publication Date Title
WO2018169742A1 (en) Apoptosis signal-regulating kinase inhibtor
US10946004B2 (en) Solid forms of an ASK1 inhibitor
USRE48150E1 (en) Apoptosis signal-regulating kinase inhibitors
JP5745721B2 (en) Apoptosis signal-regulating kinase inhibitor
US8912190B2 (en) Substituted heterocyclic compounds
EP3083608B1 (en) Fused heterocyclic compounds as ion channel modulators

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: 18712766

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18712766

Country of ref document: EP

Kind code of ref document: A1