WO2023060431A1 - Processes of making 3-fluoro-5- ( ( (1s, 2ar) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1h-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile and polymorphs thereof - Google Patents

Processes of making 3-fluoro-5- ( ( (1s, 2ar) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1h-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile and polymorphs thereof Download PDF

Info

Publication number
WO2023060431A1
WO2023060431A1 PCT/CN2021/123248 CN2021123248W WO2023060431A1 WO 2023060431 A1 WO2023060431 A1 WO 2023060431A1 CN 2021123248 W CN2021123248 W CN 2021123248W WO 2023060431 A1 WO2023060431 A1 WO 2023060431A1
Authority
WO
WIPO (PCT)
Prior art keywords
amine
inden
cyclopenta
tetrahydro
oxy
Prior art date
Application number
PCT/CN2021/123248
Other languages
French (fr)
Inventor
Rongzhen Chen
Yan Dong
Charles M. Cook
Jiping Fu
Yigang He
Yan Lou
Original Assignee
Nikang Therapeutics, 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 Nikang Therapeutics, Inc. filed Critical Nikang Therapeutics, Inc.
Priority to PCT/CN2021/123248 priority Critical patent/WO2023060431A1/en
Priority to PCT/CN2021/123407 priority patent/WO2023060458A1/en
Priority to PCT/US2022/046323 priority patent/WO2023064305A1/en
Priority to AU2022362137A priority patent/AU2022362137A1/en
Priority to CA3234417A priority patent/CA3234417A1/en
Priority to US17/963,926 priority patent/US20230115881A1/en
Priority to IL311995A priority patent/IL311995A/en
Priority to CN202280004697.2A priority patent/CN116457335A/en
Priority to TW111138511A priority patent/TW202317074A/en
Publication of WO2023060431A1 publication Critical patent/WO2023060431A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C211/00Compounds containing amino groups bound to a carbon skeleton
    • C07C211/01Compounds containing amino groups bound to a carbon skeleton having amino groups bound to acyclic carbon atoms
    • C07C211/02Compounds containing amino groups bound to a carbon skeleton having amino groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton
    • C07C211/03Monoamines
    • C07C211/05Mono-, di- or tri-ethylamine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C253/00Preparation of carboxylic acid nitriles
    • C07C253/32Separation; Purification; Stabilisation; Use of additives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • the present disclosure provides certain processes of making 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile according to the following structure:
  • compositions comprising a crystalline polymorph form of Compound 1 and processes for preparing such polymorph forms.
  • Compound 1 is a hypoxia inducible factor-2 ⁇ (HIF-2 ⁇ ) inhibitor and is being developed for treating various diseases, including cancer, such as renal cancer, glioblastoma, neuroblastoma, pheochromocytomas, paragangliomas, somatostatinomas, hemangioblastomas, gastrointestinal stromal tumors (GIST) , pituitary tumors, leiomyomas, leiomyosarcomas, polycythaemia, and retinal tumors, and non-cancer diseases such as pulmonary artery hypertension (PAH) , reflux esophagitis, hepatic steatosis, NASH, inflammatory disease (such as inflammatory bowel disease) , autoimmune disease (such as Graft-versus-Host-Disease) , and iron overload.
  • cancer such as renal cancer, glioblastoma, neuroblastoma, pheochromocytomas, paragangliomas, somatos
  • Compound 1 is disclosed in Table 1, as compound No. 5, of PCT Application No. Publication No. WO 2020/214853.
  • the published processes for making Compound 1 generates impurities, requiring purification of the Compound 1 by column chromatography. Therefore, there is a need to identify a scalable process that can produce Compound 1 that meets regulatory and other purity requirements and is cost effective on a commercial scale.
  • the polymorphic behavior of small molecule drugs can be critical in pharmacology since the same small molecule can have different physical properties as a result of the arrangement of molecules in the crystal lattice. These different properties can affect pharmaceutical parameters such as storage stability, compressibility, density, hygroscopy, dissolution rates, and bioavailability. It is known that one polymorph can convert into another polymorph, and in some cases this happens spontaneously. Accordingly, there is a need to find a thermodynamically stable polymorph of a small molecule drug. The present disclosure fulfills these and related needs.
  • a crystalline form of Compound 1, designated as Form A polymorph having an X-ray powder diffraction pattern comprising peaks at angular positions 15.8 and 18.6, wherein the angular positions may vary by ⁇ 0.2°2 ⁇ as measured by X-ray powder diffraction at ambient temperature using (Cu K ⁇ ) an X-ray wavelength of
  • the X-ray powder diffraction is determined at about 23 to about 25°C.
  • R 1 is hydrogen and R 2 is C 2 -C 12 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 7 cycloalkyl, or C 3 -C 7 cycloalkyl-C 1-6 alkyl; (2) R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 7 cycloalkyl, or C 3 -C 7 cycloalkyl-C 1-6 alkyl and R 2 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 7 cycloalkyl, or C 3 -C 7 cycloalkyl-C 1-6 alkyl; or (3) R 1 and R 2 together with the nitrogen atom to which they are attached form cyclylamine; or
  • the X-ray powder diffraction is determined at about 23 to about 25°C.
  • R 1 is hydrogen and R 2 is C 2 -C 12 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 7 cycloalkyl, or C 3 -C 7 cycloalkyl-C 1-6 alkyl; (2) R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 7 cycloalkyl, or C 3 -C 7 cycloalkyl-C 1-6 alkyl and R 2 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 7 cycloalkyl, or C 3 -C 7 cycloalkyl-C 1-6 alkyl; or (3) R 1 and R 2 together with the nitrogen atom to which they are attached form cyclylamine; or
  • Step (b1) optionally adding (i) one or more anti-solvents and/or (ii) a solid crystalline seed of Compound 1, or a solid crystalline seed of the amine solvate of Compound 1, or a combination thereof, to the mixture of Step (a1) to precipitate the amine solvate of Compound 1;
  • Step (c1) isolating the solid of Step (b1) to obtain a solid amine solvate of Compound 1;
  • Step (b) purifying the mixture from Step (a) to obtain 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile.
  • Step (b2) optionally adding (i) one or more anti-solvents and/or (ii) a crystalline seed of Compound 1, or a solid crystalline seed of the amine solvate, or a combination thereof, to the mixture of Step (a2) to precipitate crystalline Form B polymorph of diethylamine solvate of Compound 1; and
  • a process of preparing crystalline Form A polymorph of Compound 1 (as defined in Embodiment A1 to A11) , from diethylamine solvate of Compound 1, comprising:
  • a process of making crystalline Form A polymorph of Compound 1, comprising:
  • a method of treating a disease treatable by inhibition of HIF-2 ⁇ in a patient preferably the patient is in need of such treatment, which method comprises administering to the patient, preferably a patient in need of such treatment, a therapeutically effective amount of crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof disclosed herein in a pharmaceutical composition comprising a pharmaceutically acceptable excipient.
  • the disease is cancer, such as renal cancer, clear cell renal cell carcinoma, liver cancer, hepatocellular carcinoma (HCC) , pancreatic cancer, pancreatic neuroendocrine tumor, gastric cancer, ovarian cancer, non-small cell lung cancer, colorectal cancer (CRC) , pancreatic ductal adenocarcinoma (PDAC) , biliary tract cancer (BTC) , glioblastoma (see PNAS 2017, 114, E6137-E6146) , neuroblastoma, pheochromocytomas and paragangliomas (see European Journal of Cancer 2017, 86, 1-4) , somatostatinomas, hemangioblastomas, gastrointestinal stromal tumors (GIST) , pituitary tumors, leiomyomas, leiomyosarcomas, polycythaemia, retinal tumors, cancers with one or more EPAS1/HIF2A mutations such as
  • a pharmaceutical composition comprising crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof disclosed herein; and a pharmaceutically acceptable excipient.
  • a pharmaceutical composition prepared with crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof disclosed herein; and a pharmaceutically acceptable excipient.
  • a method of inhibiting HIF2 ⁇ which method comprises contacting HIF2 ⁇ with crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof disclosed herein; or contacting HIF2 ⁇ with a pharmaceutical composition comprising crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof disclosed herein, and a pharmaceutically acceptable excipient.
  • the disease are those disclosed herein including those disclosed in the first embodiment of the eighth aspect.
  • any of the aforementioned aspects involving the treatment of cancer provided are further embodiments comprising administering crystalline Form A polymorph of Compound 5 as defined in the first aspect above or any one of embodiments thereof, in combination with at least one additional anticancer agent such as an EGFR inhibitor gefitinib, erlotinib, afatinib, icotinib, neratnib, rociletinib, cetuximab, panitumumab, zalutumumab, nimotuzumab, or matuzumab.
  • additional anticancer agent such as an EGFR inhibitor gefitinib, erlotinib, afatinib, icotinib, neratnib, rociletinib, cetuximab, panitumumab, zalutumumab, nimotuzumab, or matuzumab.
  • the crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof is administered in combination with a HER2/neu inhibitor including lapatinib, trastuzumab, and pertuzumab.
  • the crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof is administered in combination with a PI3k/mTOR inhibitor including idelalisib, buparlisib, BYL719, and LY3023414.
  • the crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof is administered in combination with a VEGF inhibitor such as bevacizumab, and/or a multi-tyrosine kinase inhibitors such as sorafenib, sunitinib, pazopanib, and cabozantinib.
  • a VEGF inhibitor such as bevacizumab
  • a multi-tyrosine kinase inhibitors such as sorafenib, sunitinib, pazopanib, and cabozantinib.
  • the crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof is administered in combination with an immunotherapeutic agents such as PD-1 and PD-L1 inhibitors, CTLA4 inhibitors, IDO inhibitors, TDO inhibitors, A2A agonists, A2B agonists, STING agonists, RIG-1 agonists, Tyro/Axl/Mer inhibitors, glutaminase inhibitors, arginase inhibitors, CD73 inhibitors, CD39 inhibitors, TGF- ⁇ inhibitors, IL-2, interferon, PI3K- ⁇ inhibitors, CSF-1R inhibitors, GITR agonists, OX40 agonists, TIM-3 antagonists, LAG-3 antagonists, CAR-T therapies, and therapeutic vaccines.
  • an immunotherapeutic agents such as PD-1 and PD-L1 inhibitors, CTLA4 inhibitors, IDO inhibitors, TDO inhibitors, A2A agonists, A2B agonists, STING agonist
  • Fig. 1 depicts a representative XRPD diffractogram of crystalline Form B polymorph of diethylamine (DEA) solvate of Compound 1, prepared according to the method described in Example 4.
  • DEA diethylamine
  • Fig 2 depicts a representative 1 HNMR spectrum of diethylamine solvate of Compound 1, prepared according to the method described in Example 4.
  • Fig. 3 depicts a representative XRPD diffractogram of crystalline Form A polymorph of Compound 1, prepared according to the method described in Example 3.
  • Fig. 4 depicts a comparative XRPD diffractograms of crystalline Form A polymorph of Compound 1, prepared according to the method described in Example 4 and Form B polymorph of diethylamine solvate of Compound 1, prepared according to method described in Example 4.
  • Alkyl means a linear or branched saturated monovalent hydrocarbon radical of one to six carbon atoms, unless stated otherwise e.g., C 2 -C 12 alkyl means an alkyl radical as defined above that contains 2 to 12 carbon atoms. Examples include, but are not limited to, methyl, ethyl, propyl, 2-propyl, butyl, pentyl, and the like.
  • C 1 -C 6 alkylene means a linear or branched saturated divalent hydrocarbon radical of one to six carbon atoms. Examples include, but are not limited to, methylene, -CH 2 CH 2 -, -CH 2 CH 2 CH 2 -, -CH 2 CH (CH 3 ) -, -CH 2 CH 2 CH 2 CH 2 - (and isomers thereof) , -CH 2 CH 2 CH 2 CH 2 CH 2 - (and isomers thereof) , and the like.
  • C 2 -C 6 alkenyl means a linear or branched monovalent hydrocarbon radical of two to six carbon atoms containing a single double bond. Examples include, but are not limited to, vinyl, allyl, and the like.
  • C 2 -C 6 alkynyl means a linear or branched monovalent hydrocarbon radical of two to six carbon atoms containing a single triple bond. Examples include, but are not limited to, ethynyl, propargyl, and the like.
  • Anti-solvent as used herein means a solvent in which Compound 1 is less soluble. In one embodiment, an antisolvent is a solvent in which Compound 1 has a solubility of less than about 50 mg/mL. In another embodiment, an antisolvent is a solvent in which Compound 1 has a solubility of less than about 25 mg/mL.
  • C 3 -C 7 cycloalkyl means a monocyclic saturated monovalent hydrocarbon radical of three to seven carbon atoms optionally substituted with one or two alkyl. Examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
  • C 3 -C 7 cycloalkyl-C 1-6 alkyl means a-C 1-6 alkylene-R group where C 1-6 alkylene is as defined above and R is C 3 -C 7 cycloalkyl as defined above. Examples include, but are not limited to, cyclopropylmethyl, cyclobutylmethyl, cyclopentylethyl, cyclohexylmethyl, and the like.
  • Cyclylamine means a saturated monocyclic ring of 4 to 8 ring atoms in which one ring atom is nitrogen and an additional ring atom can be a heteroatom independently selected from N, O, and S (O) n , where n is an integer selected from 0 to 2, the remaining ring atoms being C.
  • the cyclylamine may be substituted with one or two alkyl as defined herein. Representative examples include, but is not limited to, pyrrolidine, piperidine, homopiperidine, morpholine, piperazine, thiomorpholine, and the like.
  • solvate refers to forms of a compound where the compound is associated by a non-covalent bond to a solvent molecule. This physical association may include hydrogen bonding.
  • an amine solvate of Compound 1 refers to forms of Compound 1 that are associated with an amine of formula NHR 1 R 2 or R 3 R 4 N- (CH 2 ) n -NR 5 R 6 , each as defined herein and includes both stoichiometric solvates and non-stoichiometric solvates.
  • the amine solvate of Compound 1 are isolable, for example, when an amine moleculeis incorporated in the crystal lattice of a crystalline solid of Compound 1.
  • the amine solvate of Compound 1 are present in situ.
  • An amine solvate of Compound 1 thus includes both isolable and in situ solvates.
  • An amine solvate of Compound 1 present in situ are also referred to herein as adducts.
  • the present disclosure includes a polymorphic form of Compound 1.
  • Polymorphs are different crystalline forms of a compound that differ in arrangements of the molecules of that compound in a crystal lattice. Therefore, a single compound may give rise to a variety of polymorphic forms.
  • the polymorphs of a compound usually have different melting points, solubilities, densities, and optical properties.
  • Polymorphic forms of a compound can be distinguished by a number of techniques well known in the art such as X-ray diffractometry, IR or Raman spectroscopy.
  • XRPD means X-ray powder diffraction, an analytical technique which measures the diffraction of X-rays in the presence of a solid component. Materials which are crystalline and have regular repeating arrays of atoms generate a distinctive powder pattern.
  • “Substantially free” as used herein refers to crystalline Form A polymorph of Compound 1 (i.e. 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile) that has less than about 10%by weight of 3-fluoro-5- ( ( (1R, 2aS) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile enantiomer of Compound 1 (hereinafter the “ (1R, 2aS) enantiomer” ) .
  • crystalline Form A polymorph of Compound 1 has less than about 8%by weight the (1R, 2aS) enantiomer. In another embodiment, crystalline Form A polymorph of Compound 1 has less than about 7%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 6%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 5%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 4%by weight the (1R, 2aS) enantiomer.
  • crystalline Form A polymorph of Compound 1 has less than about 3%by weight the (1R, 2aS) enantiomer. In yet another crystalline Form A polymorph of Compound 1 has less than about 2%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 1%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.8%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.7%by weight the (1R, 2aS) enantiomer.
  • crystalline Form A polymorph of Compound 1 has less than about 0.6%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.5%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.4%by weight the(1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.3%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.2%by weight the (1R, 2aS) enantiomer.
  • crystalline Form A polymorph of Compound 1 has less than about 0.1%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.05%by weight the (1R, 2aS) enantiomer.
  • substantially pure refers to a solid-state form of Compound 1 that contain less than about 5%by weight total impurities.
  • the phrase “crystalline Form A polymorph is substantially pure” means that Form A polymorph of Compound 1 that contain less than about 5%by weight total impurities. In one embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 4%by weight total impurities. In another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 3%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 2%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 1%by weight total impurities.
  • crystalline Form A polymorph of Compound 1 that contain less than about 0.9%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.8%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.7%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.6%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.5%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.4%by weight total impurities.
  • crystalline Form A polymorph of Compound 1 that contain less than about 0.3%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.2%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.1%by weight total impurities.
  • Impurities include, but are not limited to, synthesis by-products, residual starting materials, reagents, residual organic solvent, and the like.
  • substantially identical refers to measured physical characteristics that are comparable in value or data traces that are comparable in peak position and amplitude or intensity within the scope of variations that are typically associated with sample positioning or handling or the identity of the instrument employed to acquire the traces or physical characteristics or due to other variations or fluctuations normally encountered within or between laboratory environments or analytical instrumentation.
  • reacting As used herein, the term “reacting” , “treating” or “contacting” when describing a certain process is used as known in the art and generally refers to the bringing together of chemical reagents in such a manner so as to allow their interaction at the molecular level to achieve a chemical or physical transformation.
  • the reacting steps of the processes described herein can be conducted for a time and under conditions suitable for preparing the identified product.
  • Suitable organic solvent or “first suitable organic solvent” refers to an organic solvent which, under the reaction conditions of the processes disclosed herein, does not enter into any appreciable reaction with either the reactants, intermediates and/or the products at the temperatures at which the reactions are carried out.
  • a given reaction disclosed herein can be carried out in one organic solvent or a mixture of two or more organic solvents.
  • suitable organic solvents that can be used in the reactions described herein include polar (protic and/or aprotic) and nonpolar organic solvents e.g., halogenated alkanes such as carbon tetrachloride, chloroform, dichloromethane, 1, 2-dichloroethane, and the like; ethers such as tetrahydrofuran, 2-methyltetrahydrofuran, 1, 3-dioxane, 1, 4-dioxane, diethyl ether, diisopropyl ether, methyl t-butyl ether, and the like; alcohols such as methanol, ethanol, 1-propanol, 2-propanol, 1-butanol, 2-butanol, tert-butyl alcohol, 1-, 2-, or 3-pentanol, neo-pentyl alcohol, and the like; esters such as ethyl acetate, isopropyl acetate, and the like;
  • Additional organic solvents that can be used in the reactions described herein include polar organic solvents including, but not limited to, acetonitrile, dimethylformamide, dimethylacetamide, dimethyl sulfoxide, ethyl acetate, alcohols, and the like.
  • polar organic solvents e.g., alcohols, acetonitrile, DMF, N-methylpyrrolidinone, nitromethane
  • solvents that are suitable for the particular reaction step can be readily selected by a person skilled in the art.
  • the processes described herein can be monitored according to any suitable method known in the art.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C) , infrared spectroscopy, spectrophotometry, or mass spectrometry; or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • HPLC high performance liquid chromatography
  • the compounds obtained by the reactions can be purified by any suitable method known in the art. For example, chromatography (medium pressure) on a suitable adsorbent (e.g., silica gel, alumina and the like) , HPLC, or preparative thin layer chromatography; distillation; sublimation, trituration, or recrystallization.
  • the purity of the compounds in general, are determined by physical methods such as measuring the melting point (in case of a solid) , obtaining an NMR spectrum, or performing a HPLC separation.
  • Alcohol refers to an aliphatic hydrocarbon compound that carries one or more such as 1 or 2 hydroxy group. Representative examples include, but are not limited to, methanol, ethanol, propanol, butanol, 1, 2-propanediol, and the like.
  • a “pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound.
  • Such salts include:
  • acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as formic acid, acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3- (4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1, 2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic
  • a metal ion e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion
  • organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
  • a “pharmaceutically acceptable carrier or excipient” means a carrier or an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes a carrier or an excipient that is acceptable for veterinary use as well as human pharmaceutical use. “A pharmaceutically acceptable carrier/excipient” as used in the specification and claims includes both one and more than one such excipient.
  • disease as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disorder, ” “syndrome, ” and “condition” (as in medical condition) , in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms, and causes the human or animal to have a reduced duration or quality of life.
  • combination therapy means the administration of two or more therapeutic agents to treat a disease or disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.
  • patient is generally synonymous with the term “subject” and includes all mammals including humans. Examples of patients include humans, livestock such as cows, goats, sheep, pigs, and rabbits, and companion animals such as dogs, cats, rabbits, and horses. Preferably, the patient is a human.
  • Treating” or “treatment” of a disease includes:
  • inhibiting the disease i.e., arresting (i.e., stabilizing) or reducing the development of the disease or its clinical symptoms;
  • a “therapeutically effective amount” means the amount of a compound of the present disclosure or a pharmaceutically acceptable salt thereof that, when administered to a patient for treating a disease, is sufficient to affect such treatment for the disease.
  • the “therapeutically effective amount” will vary depending on the compound, the disease and its severity and the age, weight, etc., of the mammal to be treated.
  • the present disclosure includes:
  • Embodiment A is a diagrammatic representation of Embodiment A.
  • embodiment A1 provided is a crystalline Form A polymorph of Compound 1 having an X-ray powder diffraction pattern as described in the first aspect in the Summary.
  • the crystalline Form A polymorph of embodiment A1 is wherein the Form A X-ray powder diffraction pattern further comprises a peak at angular position 20.1, wherein the angular positions may vary by ⁇ 0.2°2 ⁇ .
  • the crystalline Form A polymorph of embodiment A1 is wherein the Form A X-ray powder diffraction pattern further comprises peaks at angular positions 12.9 and 20.1, wherein the angular positions may vary by ⁇ 0.2°2 ⁇ .
  • the crystalline Form A polymorph of embodiment A1 is wherein the Form A X-ray powder diffraction pattern further comprises peaks at angular positions 11.4, 12.9, and 20.1, wherein the angular positions may vary by ⁇ 0.2°2 ⁇ .
  • the crystalline Form A polymorph of embodiment A1 is wherein the Form A X-ray powder diffraction pattern further comprises peaks at angular positions 10.1, 11.4, 12.9, and 20.1, wherein the angular positions may vary by ⁇ 0.2°2 ⁇ .
  • a crystalline Form A polymorph of Compound 1 having an X-ray powder diffraction pattern comprising at least two, at least three, at least four, at least five, at least six peaks, at least seven peaks, or at least eight peaks at angular positions selected from Table 1 below, wherein the angular positions may vary by ⁇ 0.2°2 ⁇ .
  • the crystalline Form A polymorph of embodiment A6 is wherein the at least two, at least three, at least four, at least five, or at least six peaks are selected from 10.1, 11.4, 12.9, 13.7, 15.8, 18.0, 19.6, 20.1, 21.4, 21.7, 25.0, and 26.0, wherein the angular positions may vary by ⁇ 0.2°2 ⁇ .
  • embodiment A8 provided is a crystalline Form A polymorph of Compound 1 having an X-ray powder diffraction pattern substantially identical as shown in Fig 1.
  • the crystalline Form A polymorph of any one of A1 to A8, are wherein the angular positions of the X-ray diffraction peaks denoted therein may vary by ⁇ 0.1°2 ⁇ .
  • the crystalline Form A polymorph of any one of A1 to A9 is substantially pure.
  • the crystalline Form A polymorph of any one of A1 to A10 is substantially free of 3-fluoro-5- ( ( (1R, 2aS) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile enantiomer of Compound 1.
  • Embodiment B is a diagrammatic representation of Embodiment B.
  • the amine solvate of embodiment B1 is wherein the amine is NHR 1 R 2 .
  • the amine solvate of embodiment B1 is wherein the amine is R 3 R 4 -N- (CH 2 ) n -NR 5 R 6 .
  • the amine solvate of embodiment B1 or B2 is wherein R 1 is hydrogen and R 2 is C 2 -C 12 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 7 cycloalkyl, or C 3 -C 7 cycloalkyl-C 1-6 alkyl.
  • the amine solvate of embodiment B1, B2, or B4 is wherein R 1 is hydrogen and R 2 is C 2 -C 12 alkyl.
  • the amine solvate of embodiment B1, B2, or B4 is wherein R 1 is hydrogen and R 2 is C 3 -C 7 cycloalkyl.
  • the amine solvate of embodiment B1, B2, or B4 is wherein R 1 is hydrogen and R 2 is C 3 -C 7 cycloalkyl-C 1-6 alkyl.
  • the amine solvate of embodiment B1 or B2 is wherein R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, or C 2 -C 6 alkynyl and R 2 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 7 cycloalkyl, or C 3 -C 7 cycloalkyl-C 1-6 alkyl.
  • the amine solvate of embodiment B1, B2, or B7 is wherein R 1 is C 1 -C 6 alkyl and R 2 is C 1 -C 6 alkyl.
  • the amine solvate of embodiment B1, B2, or B7 is wherein R 1 is C 1 -C 6 alkyl and R 2 is C 3 -C 7 cycloalkyl.
  • the amine solvate of embodiment B1, B2, or B7 is wherein R 1 is C 1 -C 6 alkyl and R 2 is C 3 -C 7 cycloalkyl-C 1-6 alkyl.
  • the amine solvate of embodiment B1 or B2 is wherein R 1 and R 2 are independently C 3 -C 7 cycloalkyl-C 1-6 alkyl.
  • the amine solvate of embodiment B1 or B2 is wherein R 1 and R 2 together with the nitrogen atom to which they are attached form cyclylamine.
  • n is selected from 2 to 5. In a first embodiment of B12, n is 3 to 5. In a second embodiment of B12, n is 4 or 5.
  • n is selected from 2 to 4. In a first embodiment of B13, n is 3 or4. In a second embodiment of B13, n is 3.
  • the amine solvate of any one of embodiments B1, B3, and B12 to B21 and subembodiments contained therein, is wherein R 5 is C 3 -C 7 cycloalkyl.
  • the amine solvate of any one of embodiments B1, B2, and B4, to B6A is wherein the amine is ethylamine, n-propylamine, isopropylamine, n-butylamine, sec-butylamine, tert-butylamine, n-pentylamine, sec-pentylamine, pentyl-3-amine, neopentylamine, n-hexylamine, 2-hexylamine, 3-hexylamine, isohexylamine, 1-methylpentylamine, 2-ethylbutylamine, 2-methylpentylamine, 1, 1-dimethylbutylamine, 1, 3-dimethylbutylamine, 3, 3-dimethylbutylamine, 2-methyl-3-pentylamine, 3-methylpentylamine, 3-methyl-3-pentylamine, 3-methyl-2-pentylamine, 2-methylbutylamine, 1, 2, 2-trimethyl
  • the amine solvate of any one of embodiments B1, B2, B4, and B5 is wherein the amine is ethylamine, n-propylamine, isopropylamine, n-butylamine, sec-butylamine, tert-butylamine, n-pentylamine, sec-pentylamine, pentyl-3-amine, neopentylamine, n-hexylamine, 2-hexylamine, 3-hexylamine, isohexylamine, 1-methylpentylamine, 2-ethylbutylamine, 2-butylamine, 2-methylpentylamine, 1, 1-dimethylbutylamine, 1, 3-dimethylbutylamine, 3, 3-dimethylbutylamine, 2-methyl-3-pentylamine, 3-methylpentylamine, 3-methyl-3-pentylamine, 3-methyl-2-pentylamine, 2-methylbutyl-1-amine
  • the amine solvate of any one of embodiments B1, B2, B4, and B6 is wherein the amine is cyclopropylamine, cyclobutylamine, cyclopentylamine, cyclohexylamine, 2-methylcyclopropylamine, 2-methylcyclopentylamine, 3-methyl-cyclopentylamine, or 2-ethylcyclopropylamine.
  • the amine solvate of any one of embodiments B1, B2, and B7 to B10 is wherein the amine is N-methylethylamine, N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methylisopropylamine, N-allylmethylamine, N-methylpropargylamine, N-ethyl-n-propylamine, N-ethylisopropylamine, N-methylbutylamine, N-methyl-2-butylamine, N-tert-butyl-methyl-amine, N-methyl-cyclobutylamine, N-ethylcyclopropylamine, 1-cyclopropyl-N-methylmethanamine, N-ethyl-prop-2-yn-1-amine, N-ethylallylamine, N-methylpentylamine, N-methyl-2-pentylamine, N-methyl-3-pentylamine, N, 3-d
  • the amine solvate of any one of embodiments B1, B2, B7, and B8 is wherein the amine is N-methylethylamine, N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methyl-isopropylamine, N-ethyl-n-propylamine, N-ethyl-isopropylamine, N-methylbutylamine, N-methyl-2-butylamine, N-tert-butyl-methyl-amine, N-methylpentyl-1-amine, N-methyl-2-pentylamine, N-methyl-3-pentylamine, N, 3-dimethylbutane-2-amine, N, 2-dimethylbutane-2-amine, N, 3-dimethylbutane-2-amine, N-ethyl-1-butylamine, N-ethyl-2- butylamine, N-ethyl-2-methyl-2-propylamine
  • the amine solvate of any one of embodiments B1, B2, B7, and B9 is wherein the amine is N-methylcyclobutylamine, N-ethylcyclopropylamine, N-methylcyclopentylamine, or N-ethylcyclobutylamine.
  • the amine solvate of any one of embodiments B1, B2, and B11 is wherein the amine is azetidine, pyrrolidine, piperidine, piperazine, morpholine, azepane, or azocane.
  • the amine solvate of any one of embodiments B1, B3, and B12 to B16, B19, B20, B22, B23, and B25 is wherein the amine is ethylenendiamine, propane-1, 3-diamine, butane-1, 4-diamine, pentane-1, 5-diamine, hexane-1, 6-diamine, N 1 , N 2 -dimethylethane-1, 2-diamine, or N 1 , N 2 -diethylethane-1, 2-diamine.
  • the amine solvate of embodiment B1 is wherein the amine is N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methyl-isopropylamine, N-ethyl-n-propylamine, N-ethyl-isopropylamine, azetidine, pyrrolidine, piperidine, or morpholine.
  • the amine solvate of embodiment B1 or B36 is wherein the amine is diethylamine.
  • the amine solvate of embodiment B37 is wherein the stoichiometric ratio of diethylamine to Compound 1 in the diethylamine solvate of Compound 1 is 1: 1.
  • the amine solvate of any one of embodiments B1 to B38 is wherein the amine solvate of Compound 1 is a solid.
  • the amine solvate of embodiment B37 or B38 is wherein the amine solvate of Compound 1 is a crystalline solid.
  • the amine solvate of any one of embodiments B1 to B38 is wherein the amine solvate of Compound 1 is present in situ.
  • the amine solvate of embodiment B40 is wherein the amine solvate of Compound 1 is present in a mixture comprising, one or more suitable organic solvents, Compound 1, and the amine.
  • the amine solvate of embodiment B37 is wherein the diethylamine solvate of Compound 1 is present in a mixture comprising, one or more suitable organic solvent, Compound 1, and diethylamine.
  • the amine solvate of any one of embodiments B40 to B42 is wherein the one or more suitable organic solvent is a polar organic solvent independently selected from ethers, alcohols, esters, halogenated alkanes, ketones, dimethylformamide, dimethylacetamide, acetonitrile, nitromethane, n-methyl pyrrolidinone, toluene, and xylene.
  • a polar organic solvent independently selected from ethers, alcohols, esters, halogenated alkanes, ketones, dimethylformamide, dimethylacetamide, acetonitrile, nitromethane, n-methyl pyrrolidinone, toluene, and xylene.
  • the amine solvate of embodiment B43 is wherein the one or more suitable organic solvent is independently selected from n-pentanol, methanol (MeOH) , ethanol, n-propanol, isopropanol (IPA) , n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAC) , tetra
  • the amine solvate of embodiment B43 is wherein the suitable organic solvent is methyl tert-butyl ether.
  • Embodiment C is a diagrammatic representation of Embodiment C.
  • embodiment C1 provided is a crystalline Form B polymorph of diethylamine solvate of Compound 1 having an X-ray powder diffraction pattern as described in the third aspect in the Summary.
  • the crystalline Form B of embodiment C1 is wherein the Form B X-ray powder diffraction pattern further comprises a peak at angular position 8.4, wherein the angular positions may vary by ⁇ 0.2°2 ⁇ .
  • the crystalline Form B of embodiment C1 is wherein the Form B X-ray powder diffraction pattern further comprises peaks at angular positions 8.4 and 23.5, wherein the angular positions may vary by ⁇ 0.2°2 ⁇ .
  • the crystalline Form B of embodiment C1 is wherein the Form B X-ray powder diffraction pattern further comprises peaks at angular positions 8.4, 23.5, and 11.7, wherein the angular positions may vary by ⁇ 0.2°2 ⁇ .
  • the crystalline Form B of embodiment C1 is wherein the Form B X-ray powder diffraction pattern further comprises peaks at angular positions 8.4, 23.5, 11.7, and 9.5, wherein the angular positions may vary by ⁇ 0.2°2 ⁇ .
  • the crystalline Form B of embodiment C6 is wherein the at least two, at least three, at least four, at least five, or six peaks are selected from 8.4, 9.5, 11.7, 13.8, 21.3, and 23.5 wherein the angular positions may vary by ⁇ 0.2°2 ⁇ .
  • the crystalline Form B of any one of C1 to C8, are wherein the peaks denoted therein have margin of error of ⁇ 0.1°2 ⁇ .
  • Embodiment D is a diagrammatic representation of Embodiment D.
  • embodiment D1 provided is a process of preparing an amine solvate of Compound 1 as described in the fourth aspect of the Summary.
  • the process of embodiment D1 is wherein Compound 1 is contacted with the amine by adding Compound 1 to the amine or by adding the amine to a mixture of Compound 1 in one or more suitable organic solvents in Step (a1) .
  • embodiment D3 the process of embodiment D1 or D2, is wherein the amine is added to a mixture of Compound 1 in one or more suitable organic solvents in Step (a1) .
  • the process of any one of embodiments D1 to D3, is wherein the amine is R 3 R 4 -N- (CH 2 ) n -NR 5 R 6 .
  • D5A the process of any one of embodiments D1 to D4, is wherein R 1 is hydrogen and R 2 is C 2 -C 12 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 7 cycloalkyl, or C 3 -C 7 cycloalkyl-C 1-6 alkyl.
  • D7A the process of any one of embodiments D1 to D4 and D5A, is wherein R 1 is hydrogen and R 2 is C 3 -C 7 cycloalkyl-C 1-6 alkyl.
  • any one of embodiments D1 to D4 is wherein R 1 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, or C 2 -C 6 alkynyl and R 2 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 7 cycloalkyl, or C 3 -C 7 cycloalkyl-C 1-6 alkyl.
  • process of any one of embodiments D1 to D4, and D7B is wherein R 1 is C 1 -C 6 alkyl and R 2 is C 3 -C 7 cycloalkyl-C 1-6 alkyl.
  • D10A the process of any one of embodiments D1 to D4, is wherein R 1 and R 2 are independently C 3 -C 7 cycloalkyl-C 1-6 alkyl.
  • n is selected from 2 to 5. In a first embodiment of D12, n is 3 to 5. In a second embodiment of D12, n is 4 or 5.
  • n is selected from 2 to 4. In a first embodiment of D13, n is 3 or4. In a second embodiment of D13, n is 3.
  • the process of any one of embodiments D1 to D4 and D5A to D7 is wherein the amine is ethylamine, n-propylamine, isopropylamine, n-butylamine, sec-butylamine, tert-butylamine, n-pentylamine, sec-pentylamine, pentyl-3-amine, neopentylamine, n-hexylamine, 2-hexylamine, 3-hexylamine, isohexylamine, 1-methylpentylamine, 2-ethylbutylamine, 2-methylpentylamine, 1, 1-dimethylbutylamine, 1, 3-dimethylbutylamine, 3, 3-dimethylbutylamine, 2-methyl-3-pentylamine, 3-methylpentylamine, 3-methyl-3-pentylamine, 3-methyl-2-pentylamine, 2-methylbutylamine, 1, 2, 2-trimethylpropylamine
  • the process of any one of embodiments D1 to D4, D5A and D6 is wherein the amine is ethylamine, n-propylamine, isopropylamine, n-butylamine, sec-butylamine, tert-butylamine, n-pentylamine, sec-pentylamine, pentyl-3-amine, neopentylamine, n-hexylamine, 2-hexylamine, 3-hexylamine, isohexylamine, 1-methylpentylamine, 2-ethylbutyl-amine, 2-butylamine, 2-methylpentylamine, 1, 1-dimethylbutylamine, 1, 3-dimethylbutylamine, 3, 3-dimethylbutylamine, 2-methyl-3-pentylamine, 3-methylpentylamine, 3-methyl-3-pentylamine, 3-methyl-2-pentylamine, 2-methylbutyl-1-amine,
  • the process of any one of embodiments D1 to D4, D5A and D7 is wherein the amine is cyclopropylamine, cyclobutylamine, cyclopentylamine, cyclohexylamine, 2-methylcyclopropylamine, 2-methylcyclopentylamine, 3-methylcyclopentyl-amine, or 2-ethylcyclopropylamine.
  • the process of any one of embodiments D1 to D4, and D7B to D9 is wherein the amine is N-methylethylamine, N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methylisopropylamine, N-allylmethylamine, N-methylpropargylamine, N-ethyl-n-propylamine, N-ethylisopropylamine, N-methylbutylamine, N-methyl-2-butylamine, N-tert-butyl-methyl-amine, N-methyl-cyclobutylamine, N-ethylcyclopropylamine, 1-cyclopropyl-N-methylmethanamine, N-ethyl-prop-2-yn-1-amine, N-ethylallylamine, N-methylpentylamine, N-methyl-2-pentylamine, N-methyl-3-pentylamine, N, 3-dimethyl
  • the process of any one of embodiments D1 to D4, D7B and D8 is wherein the amine is N-methylethylamine, N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methyl-isopropylamine, N-ethyl-n-propylamine, N-ethyl-isopropylamine, N-methylbutylamine, N-methyl-2-butylamine, N-tert-butyl-methyl-amine, N-methylpentyl-1-amine, N-methyl-2-pentylamine, N-methyl-3-pentylamine, N, 3-dimethylbutane-2-amine, N, 2-dimethylbutane-2-amine, N, 3-dimethylbutane-2-amine, N-ethyl-1-butylamine, N-ethyl-2-butylamine, N-ethyl-2-methyl-2-propylamine, N-N-
  • the process of any one of embodiments D1 to D4, and D7B, and D9 is wherein the amine is N-methylcyclobutylamine, N-ethylcyclopropylamine, N-methylcyclopentylamine, or N-ethylcyclobutylamine.
  • the process of any one of embodiments D1 to D4, and D11 is wherein the amine is azetidine, pyrrolidine, piperidine, piperazine, morpholine, azepane, or azocane.
  • the process of any one of embodiments D1 to D3, D5, and D12 to D16, D19, D20, D22, D23, and D25 is wherein the amine is ethylenendiamine, propane-1, 3-diamine, butane-1, 4-diamine, pentane-1, 5-diamine, hexane-1, 6-diamine, N 1 , N 2 -dimethyl-ethane-1, 2-diamine, or N 1 , N 2 -diethylethane-1, 2-diamine.
  • the process of embodiment D1 to D3 is wherein the amine is N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methyl-isopropylamine, N-ethyl-n-propylamine, N-ethyl-isopropylamine, azetidine, pyrrolidine, piperidine, or morpholine.
  • any one of embodiments D1 to D37 is wherein one or more anti-solvents are added to the mixture of Compound 1 of Step (a1) to obtain a solid amine solvate of Compound 1.
  • any one of embodiments D1 to D37 is wherein one or more anti-solvents and a solid crystalline seed of Compound 1 and/or a solid crystalline seed of the amine solvate of Compound 1 are added to the mixture of Step (a1) to obtain a solid amine solid of Compound 1.
  • the process of embodiment D38 or D39A is wherein the one or more anti-solvents in Step (b1) are independently selected from alkanes and water.
  • the one or more anti-solvents are independently selected from the group consisting of water, n-heptane, n-hexane, isooctane, pentane, cyclohexane, and cyclopentane.
  • the process of any one of embodiments D2 to D41 is wherein the one or more suitable organic solvents of Step (a1) are polar organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylformamide, and dimethylacetamide.
  • the one or more suitable organic solvents of Step (a1) are polar organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylformamide, and dimethylacetamide.
  • the process of embodiment any one of embodiments D2 to D41 is wherein the one or more suitable organic solvents of Step (a1) are independently selected from the group consisting of methanol, ethanol, n-propanol, isopropanol, n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAc) , tetrahydrofur
  • embodiment D46 the process of embodiment D45, is wherein the diethylamine solvate of Compound 1 is crystalline Form B polymorph having an X-ray powder diffraction pattern as described in the Summary.
  • embodiments D47-D54 the process of embodiment D46, is wherein the crystalline Form B has an X-ray powder diffraction pattern as described in above embodiments C2 to C9, respectively.
  • any one of embodiments D1 to D55 is wherein the amine solvate of Compound 1 is converted to Compound 1 by heating the amine solvate to remove the amine from the amine solvate.
  • any one of embodiments D1 to D55 is wherein the amine solvent of Compound 1 is converted to Compound 1 by partitioning the amine solvate between one or more suitable organic solvents and an aqueous acidic solution.
  • embodiment D58 the process of embodiment D57, is wherein the one or more suitable organic solvents containing Compound 1 is isolated and concentrated to provide a solid form of Compound 1.
  • embodiment D59 the process of embodiment D56 or D58, is wherein the crystalline Form A polymorph Compound 1 has an X-ray powder diffraction pattern as described in the Summary is produced.
  • embodiment D60-D69 the process of embodiment D59, is wherein crystalline Form A polymorph of Compound 1 has an X-ray powder diffraction pattern as described in above embodiments A2 to A11, respectively, is produced.
  • step (a1) the process of any one of embodiments D1 to D69, further comprises preparing the Compound 1 of Step (a1) , comprising:
  • embodiment D71 the process of embodiment D70, is wherein the deoxyfluorinating agent is pyridine-2-sulfonyl fluoride and the base is 1, 8-diazabicyclo- [5.4.0] undec-7-ene or 7-methyl-1, 5, 7-triaza-bicyclo- [4.4.0] dec-1-ene.
  • the deoxyfluorinating agent is pyridine-2-sulfonyl fluoride and the base is 1, 8-diazabicyclo- [5.4.0] undec-7-ene or 7-methyl-1, 5, 7-triaza-bicyclo- [4.4.0] dec-1-ene.
  • embodiment D72 the process of embodiment D70 or D71, is wherein the suitable organic solvent is tetrahydrofuran.
  • Embodiment E is a diagrammatic representation of Embodiment E.
  • embodiment E1 provided is a process of preparing crystalline Form B polymorph of diethylamine solvate of Compound 1 having an X-ray powder diffraction pattern as described in the fifth aspect of the Summary.
  • the process of embodiment E1 is wherein Compound 1 is contacted with diethylamine by adding Compound 1 to the diethylamine or by adding the diethylamine to a mixture of Compound 1 in one or more suitable organic solvents in Step (a2) .
  • embodiment E3 the process of embodiment E1, is wherein the diethylamine is added to the mixture of Compound 1 in one or more suitable organic solvents in Step (a2) .
  • any one of embodiments E1 to E3, is wherein one or more anti-solvents are added to the mixture of Step (a2) to obtain crystalline Form B polymorph of the diethylamine solvate of Compound 1.
  • any one of embodiments E1 to E3 is wherein one or more anti-solvents and a solid crystalline seed of Compound 1 and/or a solid crystalline seed of the diethylamine solvate of Compound 1 are added to the mixture of Step (a2) to obtain crystalline Form B polymorph of the diethylamine solvate of Compound 1.
  • the process of any one of embodiments E1 to E5A is wherein the one or more anti-solvents in Step (b2) are independently selected from alkanes and water.
  • the one or more anti-solvents are independently selected from the group consisting of water, n-heptane, n-hexane, isooctane, pentane, cyclohexane, and cyclopentane.
  • the process of any one of embodiments E1 to E7, is wherein the solution of Compound 1 in Step (b2) is in one or more suitable organic solvents are polar organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylacetamide, and dimethylformamide.
  • suitable organic solvents are polar organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylacetamide, and dimethylformamide.
  • the process of embodiment any one of embodiments E1 to E7 is wherein the one or more suitable organic solvents are independently selected from the group consisting of methanol, ethanol, n-propanol, isopropanol, n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAc) , etrahydrofuran (THF) ,
  • the process of any one of embodiments E1 to E10 is wherein the diethylamine of solvate Compound 1 is optionally converted to Compound 1 by heating the diethylamine solvate of Compound 1 to remove the diethylamine from the diethylamine solvate of Compound 1.
  • the process of any one of embodiments E1 to E10 is wherein the diethylamine of solvate Compound 1 is optionally converted to Compound 1 by partitioning the diethylamine solvate of Compound 1 between one or more suitable organic solvent and an aqueous acidic solution.
  • the acid is hydrochloric acid.
  • embodiment E13 the process of embodiment E12, is wherein the one or more organic solvents containing Compound 1 is isolated and concentrated to provide a solid form of Compound 1.
  • Embodiment F is a diagrammatic representation of Embodiment F.
  • embodiment F1 provided is a process of preparing crystalline Form A polymorph of Compound 1 from diethylamine solvate of Compound 1 as described in the sixth aspect of the Summary.
  • embodiments F2 to F10 the process of embodiment F1, is wherein the diethylamine solvate of Compound 1 is crystalline Form B polymorph having an X-ray powder diffraction pattern as described in above embodiments C1 to C9, respectively.
  • the process of any one of embodiments F1 to F10 is wherein the diethylamine is removed from the diethylamine solvate of Compound 1 by heating the diethylamine solvate of Compound 1.
  • the process of any one of embodiments F1 to F11 is wherein the diethylamine is removed from the diethylamine solvate of Compound 1 by partitioning the diethylamine solvate of Compound 1 between one or more suitable organic solvent and an aqueous acidic solution.
  • the acid is hydrochloric acid.
  • embodiment F13 the process of embodiment F12, is wherein the one or more suitable organic solvents is isolated and concentrated to provide crystalline Form A polymorph of Compound 1 having an X-ray powder diffraction pattern as described in any one of embodiments A1 to A11 above.
  • embodiment F15 the process of embodiment F14, is wherein the crystalline Form A polymorph of Compound 1 is dissolved in one or more suitable organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylacetamide and dimethylformamide.
  • suitable organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylacetamide and dimethylformamide.
  • the process of embodiment F15 is wherein the one or more suitable organic solvents are selected from the group consisting methanol, ethanol, n-propanol, isopropanol, n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAc) , tetrahydrofuran (THF) , 2-methyl THF (2-Me
  • embodiment F17 the process of embodiment F15 or F16, is wherein one or more anti-solvents and/or solid crystalline seed of Compound 1 are added.
  • embodiment F19 the process of embodiment F18, is wherein the one or more anti-solvents are independently selected from the group consisting of water, n-heptane, n-hexane, isooctane, pentane, cyclohexane, and cyclopentane.
  • embodiment F20 the process of embodiment F19, is wherein the anti-solvent is n-heptane.
  • Embodiment G is a diagrammatic representation of Embodiment G.
  • embodiment G1 provided is a process of making crystalline Form A polymorph of Compound 1 having an X-ray powder diffraction pattern as described in the seventh aspect of the Summary.
  • the process of embodiment G1 is wherein one or more anti-solvents are added to a solution of Compound 1 in one or more suitable organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylacetamide and dimethylformamide.
  • suitable organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylacetamide and dimethylformamide.
  • the process of embodiment G2 is wherein the one or more suitable organic solvents are selected from the group consisting methanol, ethanol, n-propanol, isopropanol, n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAc) , tetrahydrofuran (THF) , 2-methyl THF (2-MeTHF)
  • embodiment G4 the process of embodiment G2 or G3, is wherein one or more anti-solvents is added.
  • the process of embodiment G4, is wherein the one or more anti-solvents is independently selected from alkanes and water.
  • the process of embodiment G5 is wherein the one or more anti-solvents is independently selected from the group consisting of water, n-heptane, n-hexane, isooctane, pentane, cyclohexane, and cyclopentane.
  • embodiment G7 the process of embodiment G6, is wherein the anti-solvent is n-heptane.
  • the process of any one of embodiments G1 to G7, is wherein the process is carried out at about 0°C to about 70°C.
  • Compound 1 is useful for the treatment of HIF-2 ⁇ mediated diseases, which include but are not limited to, various types of cancer, liver disease such as nonalcoholic steatohepatitis (NASH) , inflammatory disease such as inflammatory bowel disease (IBD) , pulmonary diseases such as pulmonary arterial hypertension (PAH) , and iron load disorders.
  • liver disease such as nonalcoholic steatohepatitis (NASH)
  • IBD inflammatory bowel disease
  • pulmonary diseases such as pulmonary arterial hypertension (PAH)
  • iron load disorders iron load disorders.
  • HIF-2 ⁇ plays an important role in the initiation and progression of many human cancers. Many extensive studies have demonstrated the critical role of increased HIF-2 ⁇ activity in driving clear cell renal cell carcinoma (ccRCC) (see review by Shen and Kaelin, Seminars in Cancer Biology 23: 18-25, 2013) . Abnormal HIF-2 ⁇ activity is largely due to loss of function of a tumor suppressor, VHL. It is known that over eighty percent of ccRCC have defective VHL either through deletion, mutation or disturbed post-translational modification. Defective VHL leads to constitutively active HIF- ⁇ proteins regardless of oxygen level.
  • HIF-2 ⁇ is the key oncogenic substrate of VHL (see Kondo, et al. Cancer Cell 1: 237-246, 2002; Kondo, et al. PLoS Biology 1: 439-444, 2002; Maranchi, et al. Cancer Cell 1: 247-255, 2002; Zimmer, et al. Mol. Cancer Res 2: 89-95, 2004) .
  • knockdown of HIF-2 ⁇ in VHL-null tumors inhibited tumor formation; while reintroduction of VHL and overexpression of HIF-2 ⁇ overcame the tumor suppressive role of VHL.
  • single nucleotide polymorphism in HIF-2 ⁇ is associated with resistant to PHD-mediated degradation, has been linked to an increased risk of developing RCC.
  • the VHL-HIF-2 ⁇ axis has also been implicated in ccRCC tumor metastasis through its downstream CXCR4 and CYTIP (see Vanharanta et al. Nature Medicine 19: 50-59, 2013; Peter Staller et al. Nature. 2003 Sep 18; 425 (6955) : 307-11) .
  • these studies strongly support the potential therapeutic utility of HIF-2 ⁇ targeted agents for the treatment of ccRCC.
  • VHL not only predisposes patients to kidney cancer (with a70%lifetime risk) , but also to hemangioblastomas, pheochromocytoma, endolymphatic sac tumors, and pancreatic neuroendocrine tumors. Tumors derived from defective VHL are frequently driven by the constitutively active downstream HIF- ⁇ proteins, with the majority of these dependent on HIF-2 ⁇ activity (see Maher, et al. Eur. J. Hum. Genet. 19: 617-623, 2011) . Both genetic and epigenetic mechanisms can lead to the loss of function in VHL.
  • HIF-2 ⁇ has been linked to cancers of the retina, adrenal gland and pancreas through both loss of function in VHL and activating mutations in HIF-2 ⁇ .
  • HIF-2 ⁇ target gene products e.g., VEGF, PDGF, and cyclin Dl
  • HIF-2 ⁇ targeted therapy could be beneficial for the above cancers when driven by these signaling events downstream of abnormal HIF-2 ⁇ pathway activation.
  • HIF- ⁇ proteins are also frequently upregulated in the intratumor environment of rapidly growing tumors, due to the hypoxic condition resulting from poor vascularization in large tumors.
  • the activated HIF- ⁇ pathways in turn, further promotes tumor cell survival and proliferation by transcriptionally upregulating various essential factors.
  • HIF-2 ⁇ overexpression and poor prognosis have demonstrated a correlation between HIF-2 ⁇ overexpression and poor prognosis in various cancers including cancers of astrocytoma, breast, cervical, colorectal, glioblastoma, glioma, head and neck, liver, non-small cell lung, melanoma, neuroblastoma, ovarian, and prostate, thereby supporting the pursuit of HIF-2 ⁇ as a therapeutic target in treating these cancers (see reviewed in Keith, et al. Nature Rev. Cancer 12: 9-22, 2012) .
  • HIF-2 ⁇ has been demonstrated to augment the growth of APC mutant colorectal cancer through its regulation of genes involved in proliferation, iron utilization and inflammation (see Xue, et al.
  • HCC hepatocellular carcinoma
  • HIF-1 ⁇ does not correlate with reduced overall survival in lung cancer patients even though its expression is also often increased (see Giatromanolaki, et al. Br. J. Cancer 85: 881-890, 2001) .
  • Extensive studies in mice engineered with both non-degradable HIF-2 ⁇ and mutant KRAS tumors have demonstrated an increased tumor burden and a decreased survival when compared to mice with only mutant KRAS expression (see Kim, et al. J. Clin. Invest. 119: 2160-2170, 2009) .
  • HIF-2 ⁇ promotes tumor growth and progression in lung cancer and also negatively correlates with clinical prognosis.
  • HIF-2 ⁇ s activity has been linked to the progression of chronic obstructive pulmonary disease (COPD) , in addition to lung cancer, in mouse models (see Karoor, et al. Cancer Prev. Res. 5: 1061-1071, 2012) .
  • COPD chronic obstructive pulmonary disease
  • HIF-2 ⁇ activity has also been demonstrated to be important in cancers of the central nervous system (see Holmquist-Mengelbier, et al. Cancer Cell 10: 413-423, 2006 and Li, et al. Cancer Cell 15: 501-513, 2009) .
  • HIF-2 ⁇ knockdown reduced tumor growth in preclinical animal models of neuroblastoma
  • increased level of HIF-2 ⁇ correlated with advanced disease, poor prognosis and higher VEGF levels, which likely contribute to the poor clinical outcome.
  • higher HIF-2 ⁇ expression has been correlated with a poor survival in glioma.
  • inhibition of HIF-2 ⁇ in glioma stem cells reduced cell proliferation and survival in vitro and tumor initiation in vivo.
  • HIF-l ⁇ is expressed in both neural progenitors and brain tumor stem cells
  • HIF-2 ⁇ is found exclusively in the latter.
  • survival of glioma patients correlates to with HIF-2 ⁇ , but not HIF-1 ⁇ level.
  • HIF-2 ⁇ effector is cyclin D, an essential partner for the activation of CDK4 and CDK6. Therefore, administration of a HIF-2 ⁇ inhibitor with CDK4/6 inhibitors, including abemaciclib palbociclib andribociclib shouldresult in downregulation of cyclin D, thereby increasing antiproliferative effects of CDK4/6 inhibitors.
  • a recent study (Nicholson et al Sci Signal. 2019 Oct 1; 12 (601) ) suggests that the antiproliferative effects of CDK4/6 inhibition were synergistic with HIF-2 ⁇ inhibition in HIF-2 ⁇ -dependent VHL-/-ccRCC cells.
  • HIF-2 ⁇ targeted therapy could improve the response to radiation therapy in various cancers.
  • Somatostatinomas are somatostatin-producing neuroendocrine tumors that are rare, but often malignant. It has been found that HIF-2 ⁇ mutations lead to the disruption of the prolyl hydroxylation domain (PHD) of HIF-2 ⁇ , thus abolish the modification by PHDs, and subsequently reduce HIF-2 ⁇ degradation mediated by VHL (see Yang, et al. Blood. 121: 2563–2566, 2013) . The stabilized HIF-2 ⁇ can then translocate to the nucleus, driving increased expression of hypoxia-related genes to contribute to somatostatinoma. Thus, a HIF-2 ⁇ inhibitor will provide an alternative approach in treating somatostatinoma.
  • PHD prolyl hydroxylation domain
  • Polycythaemia is a hematologic disorder characterized by elevated hematocrit (the volume percentage of red blood cells in the blood) , also known as erythrocytosis.
  • Gain-of-function mutations in HIF-2 ⁇ are associated with autosomal dominant erythrocytosis (see Percy, et al. N. Engl. J. Med. 358: 162–8, 2008 and Wilson et al. Case Rep Hematol. 6373706, 2016) .
  • mutations in PHD of HIF-2 ⁇ which is responsible in signaling HIF-2 ⁇ for ubiquitination and degradation by VHL, have also been found to drive polycythaemia.
  • HIF-2 ⁇ inhibitor should be able to suppress HIF-2 ⁇ downstream genes, such as EPO and thereby reducing hematocrit of polycythaemia.
  • Pheochromocytomas and paragangliomas are rare neuroendocrine tumors that often develop on a background of predisposing genetic mutations, including loss of function in VHL or PHD2 or activating mutations of HIF-2 ⁇ , all of which result in highly expressed HIF-2 ⁇ protein and subsequently downstream genes to promote oncogenic progression (see Dahia, Nat Rev Cancer. 14: 108-19, 2014) .
  • germline heterozygous mutations in genes encoding succinate dehydrogenase (SDH) subunits and the SDH complex assembly factor 2 protein (SDHAF2) have been described in patients with hereditary phaeochromocytoma and paraganglioma (PPGL) .
  • Retinal capillary hemangioblastomas can be the ocular manifestations of VHL diseases, which are caused by loss of tumor suppressor VHL. Upregulation of HIF-2 ⁇ upon loss of VHL has been detected in retinal hemangioblastoma patients and is indicated to contribute to the aggressive course of retinal hemangioblastomas, resulting in the resistance to multiple anti-VEGF and radiation therapies (see Wang, et al. Graefes Arch. Clin. Exp. Ophthalmol. 252: 1319–1327, 2014) .
  • uncontrolled blood vessel growth is a central pathological component of many human blindness disorders, including diabetic retinopathy, age-related macular degeneration, glaucoma, and retinopathy of prematurity.
  • Neuronal cell death and vision loss observed in these diseases are often caused by aberrant, leaky vessels, results of pathological neovascularization (see Krock, et al. Genes Cancer. 2: 1117–1133, 2011) .
  • inhibitor of HIF-2 ⁇ may have potential utility in treating various diseases of blindness.
  • HIF-2 ⁇ In addition to a direct role in promoting the initiation, progression and metastasis of tumor cells (e.g. ccRCC) , HIF-2 ⁇ also indirectly contributes to tumorigenesis through augmenting the immunosuppressive effect of hypoxia within the tumor microenvironment. Expression of HIF-2 ⁇ has been detected in cells of the myeloid lineage (see Talks KL, et dal. Am J Pathol. 2000; 157 (2) : 411–421) . For example, HIF-2 ⁇ is shown to favor the polarization of macrophages to the immunosuppressive M2 phenotype and enhances migration and invasion of tumor-associated macrophages (see Imtiyaz HZ et al. J Clin Invest.
  • HIF-2 ⁇ in tumor-associated macrophages TAMs
  • TAMs tumor-associated macrophages
  • additional immunosuppressive pathways e.g. adenosine and arginase etc.
  • key signaling regulators such as adenosine A2B/A2A receptors and arginase.
  • HIF-2 ⁇ proteins Due to the key roles of HIF-2 ⁇ proteins in regulating physiological response to the fluctuation of oxygen levels, they have been causally associated with many hypoxia-related pathological processes in addition to cancer.
  • PAH a debilitating and life-threatening disease with very poor prognosis.
  • Recent studies demonstrated that HIF-2 ⁇ contributes to the process of hypoxic pulmonary vascular remodeling, reduced plasticity of the vascular bed, and ultimately, debilitating PAH see Andrew S., et al. Proc Natl Acad Sci U S A. 2016 Aug 2; 113 (31) : 8801–8806, Tang H, et al. Am J Physiol Lung Cell Mol Physiol. 2018 Feb 1; 314 (2) : L256-L275.
  • IBD hypoxia-related pathological processes
  • IBD a chronic relapsing inflammatory disease of the intestine. It is found that intestinal inflammation and subsequently IBD arose when a dysregulated epithelial oxygen tension occurs and intensifies across epithelial villi in the intestine (see Shah Y. M., Molecular and Cellular Pediatrics, 2016 Dec; 3 (1) : 1) .
  • HIF-2 ⁇ activation contributes to IBD, while HIF-1 ⁇ in intestinal epithelial cells is considered as a major protective factor in IBD (see Karhausen J, et al. J Clin Invest. 2004; 114 (8) : 1098–1106; Furuta GT, et al. J Exp Med. 2001; 193 (9) : 1027–1034) .
  • HIF-2 ⁇ activation not only leads to the upregulation of pro-inflammatory cytokines which promotes IBD directly, but also results in loss of intestine barrier integrity, thus indirectly contributes to the manifestation of IBD. (see Xue X, et al. Gastroenterology.
  • an HIF-2 ⁇ inhibitor holds the promise of reverting the pro-inflammatory condition and increasing the intestinal barrier integrity, thus alleviate the symptoms of IBD.
  • HIF-2 ⁇ inhibitor also represents a novel therapeutic approach in NASH, for which limited therapeutic options are available.
  • a recent study showed that an intestine-specific disruption of HIF-2 ⁇ led to a significant reduction of hepatic steatosis and obesity induced by high-fat-diet.
  • HIF-2 ⁇ HIF-1 ⁇
  • HIF-2 ⁇ HIF-1 ⁇
  • Mastrogiannaki M, et al. J Clin Invest. 2009; 119 (5) : 1159–1166 a small molecule inhibitor that targets HIF-2 ⁇ holds promise of improving iron homeostasis in patients with iron disorders.
  • the present disclosure provides a method for treating or lessening the severity of a disease, condition, or disorder where activation or over activation of HIF-2 ⁇ is implicated in the disease state.
  • the present disclosure provides a method of treating renal cell carcinoma of a subject with Compound 1.
  • HIF-2 ⁇ inhibitors also have therapeutic potentials for a broad range of non-cancer indications including but not limited to NASH, IBD, PAH, and iron overload.
  • Form A polymorph of Compound 1 will be administered in a therapeutically effective amount by any of the accepted modes of administration for agents that serve similar utilities.
  • Therapeutically effective amounts of Form A polymorph of Compound 1 may range from about 200 mg/per day to about 1000 mg/per day, which can be administered in single or multiple doses.
  • the compositions can be provided in the form of tablets containing about 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 750, 800, 900, and 1000 milligrams of the active ingredient.
  • the actual amount of Form A polymorph of Compound 1 will depend upon numerous factors such as the severity of the disease to be treated, the age and relative health of the patient, the potency of the compound being utilized, the route and form of administration, and other factors.
  • Form A polymorph of Compound 1 will be administered as pharmaceutical compositions by any one of the following routes: oral, systemic (e.g., transdermal, intranasal or by suppository) , or parenteral (e.g., intramuscular, intravenous or subcutaneous) administration.
  • routes oral, systemic (e.g., transdermal, intranasal or by suppository) , or parenteral (e.g., intramuscular, intravenous or subcutaneous) administration.
  • parenteral e.g., intramuscular, intravenous or subcutaneous
  • the preferred manner of administration is oral using a convenient daily dosage regimen, which can be adjusted according to the degree of affliction.
  • Compositions can take the form of tablets, pills, capsules, semisolids, powders, sustained release formulations, solutions, suspensions, elixirs, aerosols, or any other appropriate compositions.
  • formulations in the form of tablets, pills or capsules, including enteric coated or delayed release tablets, pills or capsules are preferred.
  • compositions are comprised of in general, Form A polymorph of Compound 1 in combination with at least one pharmaceutically acceptable excipient.
  • Acceptable excipients are non-toxic, aid administration, and do not adversely affect the therapeutic benefit of Form A polymorph of Compound 1.
  • excipient may be any solid, liquid, semi-solid or, in the case of an aerosol composition, gaseous excipient that is generally available to one of skill in the art.
  • Solid pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk and the like.
  • Liquid and semisolid excipients may be independently selected from glycerol, propylene glycol, water, ethanol and various oils, including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, mineral oil, sesame oil, etc.
  • Preferred liquid carriers, particularly for injectable solutions include water, saline, aqueous dextrose, and glycols.
  • Form A polymorph of Compound 1 may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in powder form or in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or sterile pyrogen-free water, immediately prior to use.
  • sterile liquid carrier for example, saline or sterile pyrogen-free water
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • Formulations for parenteral administration include aqueous and non-aqueous (oily) sterile injection solutions of Form A polymorph of Compound 1 which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of Form A polymorph of Compound 1 to allow for the preparation of highly concentrated solutions.
  • Form A polymorph of Compound 1 may also be formulated as a depot preparation. Such long-acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • Form A polymorph of Compound 1 may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • compositions may take the form of tablets, lozenges, pastilles, or gels formulated in conventional manner.
  • Such compositions may comprise Form A polymorph of Compound 1 in a flavored basis such as sucrose and acacia or tragacanth.
  • the level of Form A polymorph of Compound 1 in a formulation can vary within the full range employed by those skilled in the art. Typically, the formulation will contain, on a weight percent (wt. %) basis, from about 0.01-99.99 wt. %of Form A polymorph of Compound 1 based on the total formulation, with the balance being one or more suitable pharmaceutical excipients. For example, the compound is present at a level of about 1-80 wt. %.
  • Form A polymorph of Compound 1 may be used in combination with one or more other drugs in the treatment of diseases or conditions for which Form A polymorph of Compound 1 or the other drugs may have utility. Such other drug (s) may be administered, by a route and in an amount commonly used therefore, contemporaneously or sequentially with Form A polymorph of Compound 1.
  • a pharmaceutical composition in unit dosage form containing such other drugs and Form A polymorph of Compound 1 is preferred.
  • the combination therapy may also include therapies in which Form A polymorph of Compound 1 and one or more other drugs are administered on different overlapping schedules. It is also contemplated that when used in combination with one or more other active ingredients, Form A polymorph of Compound 1 and the other active ingredients may be used in lower doses than when each is used singly.
  • compositions of the present disclosure also include those that contain one or more other drugs, in addition to Form A polymorph of Compound 1.
  • the above combinations include combinations of Form A polymorph of Compound 1 not only with one other drug, but also with two or more other active drugs.
  • Form A polymorph of Compound 1 may be used in combination with other drugs that are used in the prevention, treatment, control, amelioration, or reduction of risk of the diseases or conditions for which Form A polymorph of Compound 1 is useful.
  • Such other drugs may be administered, by a route and in an amount commonly used therefore, contemporaneously or sequentially with Form A polymorph of Compound 1.
  • a pharmaceutical composition containing such other drugs in addition to Form A polymorph of Compound 1 can be used.
  • compositions of the present disclosure also include those that also contain one or more other active ingredients, in addition to Form A polymorph of Compound 1.
  • the weight ratio of Form A polymorph of Compound 1 to the second active ingredient may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used.
  • the subject in need is suffering from or at risk of suffering from cancer
  • the subject can be treated with Form A polymorph of Compound 1 in any combination with one or more other anti-cancer agents.
  • one or more of the anti-cancer agents are proapoptotic agents.
  • anti-cancer agents include, but are not limited to, any of the following: gossyphol, genasense, polyphenol E, Chlorofusin, all trans-retinoic acid (ATRA) , bryostatin, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) , 5-aza-2’-deoxycytidine, all trans retinoic acid, doxorubicin, vincristine, etoposide, gemcitabine, imatinib (Gleevec TM ) , geldanamycin, 17-N-Allylamino-17-Demethoxygeldanamycin (17-AAG) , flavopiridol, LY294002, bortezomib, trastuzumab, BAY 11-7082, PKC412, or PD184352, Taxol TM , also referred to as “paclitaxel” , which is a well-known anti-cancer
  • Suitable anti-cancer agents also include inhibitors of kinases associated cell proliferative disorder.
  • kinases include but not limited to Aurora-A, BTK, CDK1, CDK2, CDK3, CDK4, CDK6, CDK5, CDK7, CDK8, CDK9, ephrin receptor kinases, CHK1, CHK2, SRC, Yes, Fyn, Lck, Fer, Fes, Syk, Itk, Bmx, GSK3, JNK, MEK, PAK1, PAK2, PAK3, PAK4, PDK1, PKA, PKC, RAF, Rsk and SGK.
  • inhibitors of CDK4/6 including abemaciclib (Verzenio) , palbociclib (Ibrance) and ribociclib (Kisqali) , have the potential to be synergistic with HIF-2 ⁇ inhibitors and reverse the resistance to HIF-2 ⁇ inhibition; mitogen-activated protein kinase signaling, e.g., U0126, PD98059, PD184352, PD0325901, ARRY-142886, SB239063, SP600125, BAY 43-9006, wortmannin, or LY294002; Syk inhibitors; antibodies (e.g., rituxan) ; MET inhibitor such as foretinib, carbozantinib, or crizotinib; VEGFR inhibitor such as sunitinib, sorafenib, regorafinib, lenvatinib, vandetanib, carbozantinib, axitinib;
  • BEX235 (dactolisib) , CAL101 (idelalisib) , GSK2636771, TG100-115; MTOR inhibitor such as rapamycin (sirolimus) , temsirolimus, everolimus, XL388, XL765, AZD2013, PF04691502, PKI-587, BEZ235, GDC0349; MEK inhibitor such as AZD6244, trametinib, PD184352, pimasertinib, GDC-0973, AZD8330; CSF1R inhibitors (PLX3397, LY3022855, etc. ) and CSF1R antibodies (IMC-054, RG7155, etc) ; TGF beta receptor kinase inhibitor such as LY2157299; BTK inhibitor such as ibrutinib.
  • MTOR inhibitor such as rapamycin (sirolimus) , temsirolimus, everolimus,
  • anti-cancer agents include proteasome inhibitor such as carfilzomib, MLN9708, delanzomib, or bortezomib; BET inhibitors such as INCB054329, OTX015, CPI-0610; LSD1 inhibitors such as GSK2979552, INCB059872; HDAC inhibitors such as panobinostat, vorinostat; DNA methyl transferase inhibitors such as azacytidine, decitabine) , and other epigenetic modulator; SHP-2 inhibitor such as TNO155; Bcl2 inhibitor ABT-199, and other Bcl-2 family protein inhibitors; HIF-2 ⁇ inhibitors such as PT2977 and PT2385; Beta catenin pathway inhibitors, notch pathway inhibitors and hedgehog pathway inhibitors; Antibodies or other therapeutic proteins against VEGF include bevacizumab and aflibercept.
  • BET inhibitors such as INCB054329, OTX015, CPI-0610
  • LXR liver X receptor
  • AhR aryl hydrocarbon receptor
  • anti-cancer agents that can be employed in combination with Form A polymorph of Compound 1 include Adriamycin, Dactinomycin, Bleomycin, Vinblastine, Cisplatin, acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; aminoglutethimide; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carboplatin;
  • anti-cancer agents that can be employed in combination with Form A polymorph of Compound 1 include: 20-epi-1, 25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing morphogenetic protein-1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara
  • retinamide sub. 11 retinamide; rogletimide; rohitukine; romurtide; roquinimex; rubiginone B1; ruboxyl; safingol; saintopin; SarCNU; sarcophytol A; sargramostim; Sdi 1 mimetics; semustine; senescence derived 1; sense oligonucleotides; signal transduction inhibitors; signal transduction modulators; single chain antigen-binding protein; sizofuran; sobuzoxane; sodium borocaptate; sodium phenylacetate; solverol; somatomedin binding protein; sonermin; sparfosic acid; spicamycin D; spiromustine; splenopentin; spongistatin 1; squalamine; stem cell inhibitor; stem-cell division inhibitors; stipiamide; stromelysin inhibitors; sulfinosine; superactive vasoactive intestinal
  • anticancer agents that can be employed in combination with Form A polymorph of Compound 1 include alkylating agents, antimetabolites, natural products, or hormones, e.g., nitrogen mustards (e.g., mechloroethamine, cyclophosphamide, chlorambucil, etc. ) , alkyl sulfonates (e.g., busulfan) , nitrosoureas (e.g., carmustine, lomusitne, etc. ) , or triazenes (decarbazine, etc. ) .
  • nitrogen mustards e.g., mechloroethamine, cyclophosphamide, chlorambucil, etc.
  • alkyl sulfonates e.g., busulfan
  • nitrosoureas e.g., carmustine, lomusitne, etc.
  • triazenes decarbazine, etc.
  • antimetabolites include but are not limited to folic acid analog (e.g., methotrexate) , or pyrimidine analogs (e.g., cytarabine) , purine analogs (e.g., mercaptopurine, thioguanine, pentostatin) .
  • folic acid analog e.g., methotrexate
  • pyrimidine analogs e.g., cytarabine
  • purine analogs e.g., mercaptopurine, thioguanine, pentostatin
  • Examples of natural products useful in combination with Form A polymorph of Compound 1 include but are not limited to vinca alkaloids (e.g., vincristine) , epipodophyllotoxins (e.g., etoposide) , antibiotics (e.g., daunorubicin, doxorubicin, bleomycin) , enzymes (e.g., L-asparaginase) , or biological response modifiers (e.g., interferon alpha) .
  • vinca alkaloids e.g., vincristine
  • epipodophyllotoxins e.g., etoposide
  • antibiotics e.g., daunorubicin, doxorubicin, bleomycin
  • enzymes e.g., L-asparaginase
  • biological response modifiers e.g., interferon alpha
  • alkylating agents that can be employed in combination with Form A polymorph of Compound 1 include, but are not limited to, nitrogen mustards (e.g., mechloroethamine, cyclophosphamide, chlorambucil, melphalan, etc. ) , ethylenimine and methylmelamines (e.g., hexamethlymelamine, thiotepa) , alkyl sulfonates (e.g., busulfan) , nitrosoureas (e.g., carmustine, lomusitne, semustine, streptozocin, etc. ) , or triazenes (decarbazine, etc. ) .
  • nitrogen mustards e.g., mechloroethamine, cyclophosphamide, chlorambucil, melphalan, etc.
  • ethylenimine and methylmelamines e.g., hexame
  • antimetabolites include, but are not limited to folic acid analog (e.g., methotrexate) , or pyrimidine analogs (e.g., fluorouracil, floxuridine, cytarabine) , purine analogs (e.g., mercaptopurine, thioguanine, pentostatin.
  • folic acid analog e.g., methotrexate
  • pyrimidine analogs e.g., fluorouracil, floxuridine, cytarabine
  • purine analogs e.g., mercaptopurine, thioguanine, pentostatin.
  • hormones and antagonists useful in combination with Form A polymorph of Compound 1 include, but are not limited to, adrenocorticosteroids (e.g., prednisone) , progestins (e.g., hydroxyprogesterone caproate, megestrol acetate, medroxyprogesterone acetate) , estrogens (e.g., diethylstilbestrol, ethinyl estradiol) , antiestrogen (e.g., tamoxifen) , androgens (e.g., testosterone propionate, fluoxymesterone) , antiandrogen (e.g., flutamide) , gonadotropin releasing hormone analog (e.g., leuprolide) .
  • adrenocorticosteroids e.g., prednisone
  • progestins e.g., hydroxyprogesterone caproate, megestrol acetate,
  • platinum coordination complexes e.g., cisplatin, carboblatin
  • anthracenedione e.g., mitoxantrone
  • substituted urea e.g., hydroxyurea
  • methyl hydrazine derivative e.g., procarbazine
  • adrenocortical suppressant e.g., mitotane, aminoglutethimide
  • anti-cancer agents that can be employed in combination with Form A polymorph of Compound 1 include: anti-cancer agents which act by arresting cells in the G2-M phases due to stabilized microtubules and include Erbulozole (also known as R-55104) , Dolastatin 10 (also known as DLS-10 and NSC-376128) , Mivobulin isethionate (also known as CI-980) , Vincristine, NSC-639829, Discodermolide (also known as NVP-XX-A-296) , ABT-751 (Abbott, also known as E-7010) , Altorhyrtins (such as Altorhyrtin A and Altorhyrtin C) , Spongistatins (such as Spongistatin 1, Spongistatin 2, Spongistatin 3, Spongistatin 4, Spongistatin 5, Spongistatin 6, Spongistatin 7, Spongistatin 8, and Spongistatin 9)
  • HCl AC-7700 (Ajinomoto, also known as AVE-8062, AVE-8062A, CS-39-L-Ser. HCl, and RPR-258062A) , Vitilevuamide, Tubulysin A, Canadensol, Centaureidin (also known as NSC- 106969) , T-138067 (Tularik, also known as T-67, TL-138067 and TI-138067) , COBRA-1 (Parker Hughes Institute, also known as DDE-261 and WHI-261) , H10 (Kansas State University) , H16 (Kansas State University) , Oncocidin A1 (also known as BTO-956 and DIME) , DDE-313 (Parker Hughes Institute) , Fijianolide B.
  • AC-7700 Ajinomoto, also known as AVE-8062, AVE-8062A, CS-39-L-Ser. HCl, and RPR-258062
  • immune checkpoint inhibitors can be used in combination with Form A polymorph of Compound 1 for treatment of HIF-2 ⁇ -associated diseases, disorders or conditions.
  • exemplary immune checkpoint inhibitors include inhibitors (smack molecules or biologics) against immune checkpoint molecules such as CD27, CD28, CD40, CD122, CD96, CD73, CD39, CD47, OX40, GITR, CSF1R, JAK, PI3K delta, PI3K gamma, TAM, arginase, CD137 (also known as 4-1BB) , ICOS, A2AR, A2BR, SHP-2, B7-H3, B7-H4, BTLA, CTLA-4, LAG3, TIM3, VISTA, CD96, TIGIT, PD-1, PD-L1 and PD-L2.
  • inhibitors smack molecules or biologics
  • immune checkpoint molecules such as CD27, CD28, CD40, CD122, CD96, CD73, CD39, CD47, OX40, GITR, C
  • the immune checkpoint molecule is a stimulatory checkpoint molecule selected from CD27, CD28, CD40, ICOS, OX40, GITR, CD137 and STING.
  • the immune checkpoint molecule is an inhibitory checkpoint molecule selected from B7-H3, B7-H4, BTLA, CTLA-4, IDO, TDO, Arginase, KIR, LAG3, PD-1, TIM3, CD96, TIGIT and VISTA.
  • the compounds provided herein can be used in combination with one or more agents selected from KIR inhibitors, TIGIT inhibitors, LAIR1 inhibitors, CD160 inhibitors, 2B4 inhibitors and TGFR beta inhibitors.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-1, e.g., an anti-PD-1 monoclonal antibody.
  • the anti-PD-1 monoclonal antibody is nivolumab, pembrolizumab (also known as MK-3475) , pidilizumab, SHR-1210, PDR001, or AMP-224.
  • the anti-PD-1 monoclonal antibody is nivolumab, or pembrolizumab or PDR001.
  • the anti-PD1 antibody is pembrolizumab.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-L1, e.g., an anti-PD-L1 monoclonal antibody.
  • the anti-PD-L1 monoclonal antibody is BMS-935559, MEDI4736, MPDL3280A (also known as RG7446) , or MSB0010718C.
  • the anti-PD-L1 monoclonal antibody is MPDL3280A (atezolizumab) or MEDI4736 (durvalumab) .
  • the inhibitor of an immune checkpoint molecule is an inhibitor of CTLA-4, e.g., an anti-CTLA-4 antibody.
  • the anti-CTLA-4 antibody is ipilimumab or tremelimumab.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of LAG3, e.g., an anti-LAG3 antibody.
  • the anti-LAG3 antibody is BMS-986016 or LAG525.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of GITR, e.g., an anti-GITR antibody.
  • the anti-GITR antibody is TRX518 or, MK-4166, INCAGN01876 or MK-1248.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of OX40, e.g., an anti-OX40 antibody or OX40L fusion protein.
  • the anti-OX40 antibody is MEDI0562 or, INCAGN01949, GSK2831781, GSK-3174998, MOXR-0916, PF-04518600 or LAG525.
  • the OX40L fusion protein is MEDI6383.
  • Form A polymorph of Compound 1 can also be used to increase or enhance an immune response, including increasing the immune response to an antigen; to improve immunization, including increasing vaccine efficacy; and to increase inflammation.
  • the compounds of the invention can be sued to enhance the immune response to vaccines including, but not limited, Listeria vaccines, oncolytic viarl vaccines, and cancer vaccines such as (granulocyte-macrophage colony-stimulating factor (GM-CF) gene-transfected tumor cell vaccine) .
  • Anti-cancer vaccines include dendritic cells, synthetic peptides, DNA vaccines and recombinant viruses.
  • Other immune-modulatory agents also include those that block immune cell migration such as antagonists to chemokine receptors, including CCR2 and CCR4; Sting agonists and Toll receptor agonists.
  • anti-cancer agents also include those that augment the immune system such as adjuvants or adoptive T cell transfer.
  • Form A polymorph of Compound 1 may be effective in combination with CAR (Chimeric antigen receptor) T cell treatment as a booster for T cell activation.
  • the desired fractions were concentrated and the solvent was swapped with MTBE to obtain a MTBE solution (about 350 mL) .
  • n-Heptane 150 mL was added slowly over4 h at 20–30°C, followed by additional n-heptane (950 mL) over 8 h. After further stirring at 20–30°C for 3 h, the slurry was cooled slowly to-10–0°C over4 h, and then stirred at this temperature for 6 h. The mixture was filtered and the solid cake was rinsed with cold n-heptane. The solid was vacuum dried at 45-55°C for 12 h to obtain the title product (46.73g, 93.5%yield) .
  • the HPLC purity was 99.9 A%and chiral purity was 100.0%ee.
  • Compound 1 was dissolved in solvent, and anti-solvent was added with stirring at room temperature, 50 °C, or ice bath as shown in Table 4 below.
  • the crystalline solid was filtered and characterized by XRPD.
  • XRPD patterns were obtained with an X-ray diffractometer (Bruker D8 advance) , at ambient temperature (about 23-25°C) using an incident beam of Cu K ⁇ from a generator operating at 40kV and40 mA. The system was equipped with LynxEye detector. Samples of diethylamine solvate of 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile were scanned from 3 to 40°2 ⁇ , at a step size 0.02°2 ⁇ . Data was analyzed using DIFFRAC plus Evaluation Package Release 2010. The XRPD spectrum of crystalline Form B polymorph is shown in Figure 1. The peak listing for XRPD spectrum, diffraction angles (2-Theta) reported in degrees was given in Table 2 below.
  • a pharmaceutical composition for inhalation delivery 20 mg of a compound disclosed herein is mixed with 50 mg of anhydrous citric acid and 100 mL of0.9%sodium chloride solution. The mixture is incorporated into an inhalation delivery unit, such as a nebulizer, which is suitable for inhalation administration.
  • an inhalation delivery unit such as a nebulizer
  • a pharmaceutical topical gel composition 100 mg of a compound disclosed herein is mixed with 1.75 g of hydroxypropyl cellulose, 10 mL of propylene glycol, 10 mL of isopropyl myristate and 100 mL of purified alcohol USP. The resulting gel mixture is then incorporated into containers, such as tubes, which are suitable for topical administration.
  • a pharmaceutical ophthalmic solution composition 100 mg of a compound disclosed herein is mixed with 0.9 g of NaCl in 100 mL of purified water and filtered using a 0.2 micron filter. The resulting isotonic solution is then incorporated into ophthalmic delivery units, such as eye drop containers, which are suitable for ophthalmic administration.
  • a pharmaceutical nasal spray solution 10 g of a compound disclosed herein is mixed with 30 mL of a 0.05M phosphate buffer solution (pH 4.4) .
  • the solution is placed in a nasal administrator designed to deliver 100 ul of spray for each application.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

Disclosed are certain processes of making 3-fluoro-5-(((1S,2aR)-1,3,3,4,4-pentafluoro-2a-hydroxy-2,2a,3,4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy)-benzonitrile (Compound 1) and certain polymorphs thereof. Also disclosed are pharmaceutical compositions comprising a crystalline polymorph form of Compound 1 and processes for preparing such polymorph forms.

Description

PROCESSES OF MAKING 3-FLUORO-5- ( ( (1S, 2AR) -1, 3, 3, 4, 4-PENTAFLUORO-2A-HYDROXY-2, 2A, 3, 4-TETRAHYDRO-1H-CYCLOPENTA [CD] INDEN-7-YL) OXY) -BENZONITRILE AND POLYMORPHS THEREOF
Field of the disclosure
The present disclosure provides certain processes of making 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile according to the following structure:
Figure PCTCN2021123248-appb-000001
and certain polymorphs thereof. Also provided are pharmaceutical compositions comprising a crystalline polymorph form of Compound 1 and processes for preparing such polymorph forms.
Background
Compound 1 is a hypoxia inducible factor-2α (HIF-2α) inhibitor and is being developed for treating various diseases, including cancer, such as renal cancer, glioblastoma, neuroblastoma, pheochromocytomas, paragangliomas, somatostatinomas, hemangioblastomas, gastrointestinal stromal tumors (GIST) , pituitary tumors, leiomyomas, leiomyosarcomas, polycythaemia, and retinal tumors, and non-cancer diseases such as pulmonary artery hypertension (PAH) , reflux esophagitis, hepatic steatosis, NASH, inflammatory disease (such as inflammatory bowel disease) , autoimmune disease (such as Graft-versus-Host-Disease) , and iron overload.
Compound 1 is disclosed in Table 1, as compound No. 5, of PCT Application No. Publication No. WO 2020/214853. The published processes for making Compound 1 generates impurities, requiring purification of the Compound 1 by column chromatography. Therefore, there is a need to identify a scalable process that can produce Compound 1 that meets regulatory and other purity requirements and is cost effective on a commercial scale.
The polymorphic behavior of small molecule drugs can be critical in pharmacology since the same small molecule can have different physical properties as a result of the arrangement of molecules in the crystal lattice. These different properties can affect pharmaceutical parameters  such as storage stability, compressibility, density, hygroscopy, dissolution rates, and bioavailability. It is known that one polymorph can convert into another polymorph, and in some cases this happens spontaneously. Accordingly, there is a need to find a thermodynamically stable polymorph of a small molecule drug. The present disclosure fulfills these and related needs.
Summary
In a first aspect, provided is a crystalline form of Compound 1, designated as Form A polymorph, having an X-ray powder diffraction pattern comprising peaks at angular positions 15.8 and 18.6, wherein the angular positions may vary by±0.2°2θ as measured by X-ray powder diffraction at ambient temperature using (Cu Kα) an X-ray wavelength of
Figure PCTCN2021123248-appb-000002
In an embodiment of the first aspect, the X-ray powder diffraction is determined at about 23 to about 25℃.
In a second aspect, provided is an amine solvate of Compound 1, wherein the amine is:
(i) NHR 1R 2 where (1) R 1 is hydrogen and R 2 is C 2-C 12 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl; (2) R 1 is C 1-C 6 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl and R 2 is C 1-C 6 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl; or (3) R 1 and R 2 together with the nitrogen atom to which they are attached form cyclylamine; or
(ii) R 3R 4N- (CH 2n-NR 5R 6 where n is an integer selected from 1 to 6 and R 3, R 4, R 5, and R 6are independently H, C 1-C 6 alkyl, or C 3-C 7 cycloalkyl.
In a third aspect, provided is a crystalline form of diethylamine solvate of Compound 1, designated as Form B polymorph, having an X-ray powder diffraction pattern comprising peaks at angular positions 13.8 and21.3, wherein the angular positions may vary by+0.2°2θ as measured by X-ray powder diffraction at ambient temperature using (Cu Kα) an X-ray wavelength of
Figure PCTCN2021123248-appb-000003
Figure PCTCN2021123248-appb-000004
In an embodiment of the third aspect, the X-ray powder diffraction is determined at about 23 to about 25℃.
In a fourth aspect, provided is a process of preparing an amine solvate of Compound 1, wherein the amine is:
(i) NHR 1R 2 where (1) R 1 is hydrogen and R 2 is C 2-C 12 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl; (2) R 1 is C 1-C 6 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl and R 2 is C 1-C 6 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl; or (3) R 1 and R 2 together with the nitrogen atom to which they are attached form cyclylamine; or
(ii) R 3R 4N- (CH 2n-NR 5R 6 where n is an integer selected from 1 to 6 and R 3, R 4, R 5, and R 6are independently H, C 1-C 6 alkyl or C 3-C 7 cycloalkyl, comprising:
(a1) contacting Compound 1 with the amine NHR 1R 2 or R 3R 4-N- (CH 2n-R 5R 6 where R 1, R 2, R 3, R 4, R 5, and R 6, and n are as defined in (i) and (ii) , respectively, in the presence or absence of one or more suitable organic solvents;
(b1) optionally adding (i) one or more anti-solvents and/or (ii) a solid crystalline seed of Compound 1, or a solid crystalline seed of the amine solvate of Compound 1, or a combination thereof, to the mixture of Step (a1) to precipitate the amine solvate of Compound 1;
(c1) isolating the solid of Step (b1) to obtain a solid amine solvate of Compound 1; and
(d1) optionally converting the amine solvate of Compound 1 from any one of Steps (a1) to (c1) to Compound 1.
The process of the fourth aspect, further comprising preparing the Compound 1 of Step (a1) , comprising:
(a) treating 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile with a deoxyfluorinating agent in the presence of a base in a suitable organic solvent; and
(b) purifying the mixture from Step (a) to obtain 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile.
In a fifth aspect, provided is a process of preparing crystalline Form B polymorph of diethylamine solvate of Compound 1 as described in Embodiments C1 to C9, comprising:
(a2) contacting Compound 1 with diethylamine, in the presence or absence of one or more suitable organic solvents;
(b2) optionally adding (i) one or more anti-solvents and/or (ii) a crystalline seed of Compound 1, or a solid crystalline seed of the amine solvate, or a combination thereof, to the mixture of Step (a2) to precipitate crystalline Form B polymorph of diethylamine solvate of Compound 1; and
(c2) isolating the crystalline Form B polymorph of the diethylamine solvate of Compound 1; and
(d2) optionally converting the crystalline Form B polymorph of diethylamine solvate of Compound 1 from Step (c2) to Compound 1.
In a sixth aspect, provided is a process of preparing crystalline Form A polymorph of Compound 1 (as defined in Embodiment A1 to A11) , from diethylamine solvate of Compound 1, comprising:
(a3) removing of the diethylamine from the diethylamine solvate of Compound 1; and
(b3) optionally recrystallizing crystalline Form A polymorph of Compound 1 obtained from Step (a3) .
In a seventh aspect, provided is a process of making crystalline Form A polymorph of Compound 1, comprising:
a4) contacting a solution of Compound 1 in one or more suitable organic solvents with one or more anti-solvents;
b4) optionally adding a solid crystalline seed of Compound 1; and
c4) isolating the crystalline Form A polymorph of Compound 1 from the mixture.
In an eighth aspect, provided is a method of treating a disease treatable by inhibition of HIF-2αin a patient, preferably the patient is in need of such treatment, which method comprises administering to the patient, preferably a patient in need of such treatment, a therapeutically effective amount of crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof disclosed herein in a pharmaceutical composition comprising a pharmaceutically acceptable excipient.
In one embodiment of the eighth aspect, the disease is cancer, such as renal cancer, clear cell renal cell carcinoma, liver cancer, hepatocellular carcinoma (HCC) , pancreatic cancer, pancreatic neuroendocrine tumor, gastric cancer, ovarian cancer, non-small cell lung cancer, colorectal cancer (CRC) , pancreatic ductal adenocarcinoma (PDAC) , biliary tract cancer (BTC) , glioblastoma (see PNAS 2017, 114, E6137-E6146) , neuroblastoma, pheochromocytomas and paragangliomas (see European Journal of Cancer 2017, 86, 1-4) , somatostatinomas, hemangioblastomas, gastrointestinal stromal tumors (GIST) , pituitary tumors, leiomyomas, leiomyosarcomas, polycythaemia, retinal tumors, cancers with one or more EPAS1/HIF2A mutations such as uterine cancer, pheochromocytoma/paraganglioma, melanoma, stomach cancer, lung adenocarcinoma, esophagus cancer, lung squamous cancer, ovarian cancer, cervical cancer, head&neck cancer, liver cancer, colorectal cancer, breast cancer, renal cell carcinoma, diffuse large B-cell lymphoma, glioblastoma, thymoma, prostate cancer, pancreatic cancer, sarcoma, and low-grade gliomas, and cancers with one or more ELOC/TCEB1 mutations such as prostate cancer, uterine cancer, liver cancer, breast cancer, pancreatic cancer, ovarian cancer, lung adenocarcinoma, head and neck cancer, stomach cancer, sarcoma cancer, colorectal cancer, lung squamous cancer, esophagus cancer, diffuse large B-cell lymphoma, melanoma, renal cell carcinoma, cervical cancer, pheochromocytoma/paraganglioma, adenoid cystic carcinoma, acute myeloid leukemia, glioblastoma, and low-grade gliomas.
In another embodiment, non-cancer diseases that could benefit from Hif-2αinhibition include VHL (von Hippel-Lindau) disease (see Oncotarget, 2015, 6, 23036-23037) , PAH (pulmonary artery hypertension) (see Mol. Cell. Biol. 2016, 36, 1584-1594) , esophagitis, reflux esophagitis (see Current Opinion in Pharmacology 2017, 37: 93-99) , hepatic steatosis (see Nature Medicine 2017, 23, 1298-1308) , NASH, inflammatory disease such as inflammatory bowel disease (see Nature Reviews gastroenterology&Hepatology 2017, 14, 596) , autoimmune disease such as Graft-versus-Host-Disease (see Blood, 2015, 126, 1865) , and iron overload.
In a ninth aspect, provided is a pharmaceutical composition comprising crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof disclosed herein; and a pharmaceutically acceptable excipient.
In a tenth aspect, provided is a pharmaceutical composition prepared with crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof disclosed herein; and a pharmaceutically acceptable excipient.
In an eleventh aspect. provided is a method of inhibiting HIF2αwhich method comprises contacting HIF2αwith crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof disclosed herein; or contacting HIF2αwith a pharmaceutical composition comprising crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof disclosed herein, and a pharmaceutically acceptable excipient.
In a twelfth aspect, provided is crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof disclosed herein for use in the treatment of a disease mediated by HIF-2a. In an embodiment of the twelfth aspect, the disease are those disclosed herein including those disclosed in the first embodiment of the eighth aspect.
In any of the aforementioned aspects involving the treatment of cancer, provided are further embodiments comprising administering crystalline Form A polymorph of Compound 5 as defined in the first aspect above or any one of embodiments thereof, in combination with at least one additional anticancer agent such as an EGFR inhibitor gefitinib, erlotinib, afatinib, icotinib, neratnib, rociletinib, cetuximab, panitumumab, zalutumumab, nimotuzumab, or matuzumab. In another embodiment, the crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof, is administered in combination with a HER2/neu inhibitor including lapatinib, trastuzumab, and pertuzumab. In another embodiment, the crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof, is administered in combination with a PI3k/mTOR inhibitor including  idelalisib, buparlisib, BYL719, and LY3023414. In another embodiment, the crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof, is administered in combination with a VEGF inhibitor such as bevacizumab, and/or a multi-tyrosine kinase inhibitors such as sorafenib, sunitinib, pazopanib, and cabozantinib. In another embodiment, the crystalline Form A polymorph of Compound 1 as defined in the first aspect above or any one of embodiments thereof, is administered in combination with an immunotherapeutic agents such as PD-1 and PD-L1 inhibitors, CTLA4 inhibitors, IDO inhibitors, TDO inhibitors, A2A agonists, A2B agonists, STING agonists, RIG-1 agonists, Tyro/Axl/Mer inhibitors, glutaminase inhibitors, arginase inhibitors, CD73 inhibitors, CD39 inhibitors, TGF-βinhibitors, IL-2, interferon, PI3K-γ inhibitors, CSF-1R inhibitors, GITR agonists, OX40 agonists, TIM-3 antagonists, LAG-3 antagonists, CAR-T therapies, and therapeutic vaccines. When combination therapy is used, the agents can be administered simultaneously or sequentially.
Brief Description of the Drawings
Fig. 1 depicts a representative XRPD diffractogram of crystalline Form B polymorph of diethylamine (DEA) solvate of Compound 1, prepared according to the method described in Example 4.
Fig 2 depicts a representative  1HNMR spectrum of diethylamine solvate of Compound 1, prepared according to the method described in Example 4.
Fig. 3 depicts a representative XRPD diffractogram of crystalline Form A polymorph of Compound 1, prepared according to the method described in Example 3.
Fig. 4 depicts a comparative XRPD diffractograms of crystalline Form A polymorph of Compound 1, prepared according to the method described in Example 4 and Form B polymorph of diethylamine solvate of Compound 1, prepared according to method described in Example 4.
Detailed Description
Definitions:
Unless otherwise stated, the following terms used in the specification and claims are defined for the purposes of this Application and have the following meaning:
“Alkyl” means a linear or branched saturated monovalent hydrocarbon radical of one to six carbon atoms, unless stated otherwise e.g., C 2-C 12 alkyl means an alkyl radical as defined above that contains 2 to 12 carbon atoms. Examples include, but are not limited to, methyl, ethyl, propyl, 2-propyl, butyl, pentyl, and the like.
“C 1-C 6 alkylene” means a linear or branched saturated divalent hydrocarbon radical of one to six carbon atoms. Examples include, but are not limited to, methylene, -CH 2CH 2-, -CH 2CH 2CH 2-, -CH 2CH (CH 3) -, -CH 2CH 2CH 2CH 2- (and isomers thereof) , -CH 2CH 2CH 2CH 2CH 2- (and isomers thereof) , and the like.
“C 2-C 6alkenyl” means a linear or branched monovalent hydrocarbon radical of two to six carbon atoms containing a single double bond. Examples include, but are not limited to, vinyl, allyl, and the like.
“C 2-C 6alkynyl” means a linear or branched monovalent hydrocarbon radical of two to six carbon atoms containing a single triple bond. Examples include, but are not limited to, ethynyl, propargyl, and the like.
“Anti-solvent” as used herein means a solvent in which Compound 1 is less soluble. In one embodiment, an antisolvent is a solvent in which Compound 1 has a solubility of less than about 50 mg/mL. In another embodiment, an antisolvent is a solvent in which Compound 1 has a solubility of less than about 25 mg/mL.
“C 3-C 7 cycloalkyl” means a monocyclic saturated monovalent hydrocarbon radical of three to seven carbon atoms optionally substituted with one or two alkyl. Examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
“C 3-C 7 cycloalkyl-C 1-6 alkyl” means a-C 1-6 alkylene-R group where C 1-6 alkylene is as defined above and R is C 3-C 7 cycloalkyl as defined above. Examples include, but are not limited to, cyclopropylmethyl, cyclobutylmethyl, cyclopentylethyl, cyclohexylmethyl, and the like.
“Cyclylamine” means a saturated monocyclic ring of 4 to 8 ring atoms in which one ring atom is nitrogen and an additional ring atom can be a heteroatom independently selected from N, O, and S (O)  n, where n is an integer selected from 0 to 2, the remaining ring atoms being C. The cyclylamine may be substituted with one or two alkyl as defined herein. Representative examples include, but is not limited to, pyrrolidine, piperidine, homopiperidine, morpholine, piperazine, thiomorpholine, and the like.
The term “solvate” refers to forms of a compound where the compound is associated by a non-covalent bond to a solvent molecule. This physical association may include hydrogen bonding. As used herein, an amine solvate of Compound 1 refers to forms of Compound 1 that are associated with an amine of formula NHR 1R 2 or R 3R 4N- (CH 2n-NR 5R 6, each as defined herein and includes both stoichiometric solvates and non-stoichiometric solvates. In certain instances, the amine solvate of Compound 1 are isolable, for example, when an amine moleculeis incorporated in the crystal lattice of a crystalline solid of Compound 1. In certain instances, the amine  solvate of Compound 1 are present in situ. An amine solvate of Compound 1 thus includes both isolable and in situ solvates. An amine solvate of Compound 1 present in situ are also referred to herein as adducts.
The present disclosure includes a polymorphic form of Compound 1. Polymorphs are different crystalline forms of a compound that differ in arrangements of the molecules of that compound in a crystal lattice. Therefore, a single compound may give rise to a variety of polymorphic forms. The polymorphs of a compound usually have different melting points, solubilities, densities, and optical properties. Polymorphic forms of a compound can be distinguished by a number of techniques well known in the art such as X-ray diffractometry, IR or Raman spectroscopy.
“XRPD” means X-ray powder diffraction, an analytical technique which measures the diffraction of X-rays in the presence of a solid component. Materials which are crystalline and have regular repeating arrays of atoms generate a distinctive powder pattern.
“Substantially free” as used herein refers to crystalline Form A polymorph of Compound 1 (i.e. 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile) that has less than about 10%by weight of 3-fluoro-5- ( ( (1R, 2aS) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile enantiomer of Compound 1 (hereinafter the “ (1R, 2aS) enantiomer” ) . In one embodiment, crystalline Form A polymorph of Compound 1 has less than about 8%by weight the (1R, 2aS) enantiomer. In another embodiment, crystalline Form A polymorph of Compound 1 has less than about 7%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 6%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 5%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 4%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 3%by weight the (1R, 2aS) enantiomer. In yet another crystalline Form A polymorph of Compound 1 has less than about 2%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 1%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.8%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.7%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.6%by weight  the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.5%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.4%by weight the(1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.3%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.2%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.1%by weight the (1R, 2aS) enantiomer. In yet another embodiment, crystalline Form A polymorph of Compound 1 has less than about 0.05%by weight the (1R, 2aS) enantiomer.
“Substantially pure” as used herein refers to a solid-state form of Compound 1 that contain less than about 5%by weight total impurities. The phrase “crystalline Form A polymorph is substantially pure” means that Form A polymorph of Compound 1 that contain less than about 5%by weight total impurities. In one embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 4%by weight total impurities. In another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 3%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 2%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 1%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.9%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.8%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.7%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.6%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.5%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.4%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.3%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.2%by weight total impurities. In yet another embodiment, crystalline Form A polymorph of Compound 1 that contain less than about 0.1%by weight total impurities. Impurities include, but are not  limited to, synthesis by-products, residual starting materials, reagents, residual organic solvent, and the like.
“Substantially identical” as used herein refers to measured physical characteristics that are comparable in value or data traces that are comparable in peak position and amplitude or intensity within the scope of variations that are typically associated with sample positioning or handling or the identity of the instrument employed to acquire the traces or physical characteristics or due to other variations or fluctuations normally encountered within or between laboratory environments or analytical instrumentation.
As used herein, the term “reacting” , “treating” or “contacting” when describing a certain process is used as known in the art and generally refers to the bringing together of chemical reagents in such a manner so as to allow their interaction at the molecular level to achieve a chemical or physical transformation. The reacting steps of the processes described herein can be conducted for a time and under conditions suitable for preparing the identified product.
“Suitable organic solvent” or “first suitable organic solvent” refers to an organic solvent which, under the reaction conditions of the processes disclosed herein, does not enter into any appreciable reaction with either the reactants, intermediates and/or the products at the temperatures at which the reactions are carried out. A given reaction disclosed herein can be carried out in one organic solvent or a mixture of two or more organic solvents. Examples of suitable organic solvents that can be used in the reactions described herein include polar (protic and/or aprotic) and nonpolar organic solvents e.g., halogenated alkanes such as carbon tetrachloride, chloroform, dichloromethane, 1, 2-dichloroethane, and the like; ethers such as tetrahydrofuran, 2-methyltetrahydrofuran, 1, 3-dioxane, 1, 4-dioxane, diethyl ether, diisopropyl ether, methyl t-butyl ether, and the like; alcohols such as methanol, ethanol, 1-propanol, 2-propanol, 1-butanol, 2-butanol, tert-butyl alcohol, 1-, 2-, or 3-pentanol, neo-pentyl alcohol, and the like; esters such as ethyl acetate, isopropyl acetate, and the like; ketones such as acetone and the like; aromatic hydrocarbons such as benzene, toluene, xylene, or alkanes such as cyclohexane, pentane, hexane, heptane, and the like. Additional organic solvents that can be used in the reactions described herein include polar organic solvents including, but not limited to, acetonitrile, dimethylformamide, dimethylacetamide, dimethyl sulfoxide, ethyl acetate, alcohols, and the like. When polar organic solvents (e.g., alcohols, acetonitrile, DMF, N-methylpyrrolidinone, nitromethane) contain water they are referred to herein as aqueous organic solvent. Depending on the reaction step, solvents that are suitable for the particular reaction step can be readily selected by a person skilled in the art.
The processes described herein can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g.,  1H or  13C) , infrared spectroscopy, spectrophotometry, or mass spectrometry; or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography. The compounds obtained by the reactions can be purified by any suitable method known in the art. For example, chromatography (medium pressure) on a suitable adsorbent (e.g., silica gel, alumina and the like) , HPLC, or preparative thin layer chromatography; distillation; sublimation, trituration, or recrystallization. The purity of the compounds, in general, are determined by physical methods such as measuring the melting point (in case of a solid) , obtaining an NMR spectrum, or performing a HPLC separation.
“Alcohol” refers to an aliphatic hydrocarbon compound that carries one or more such as 1 or 2 hydroxy group. Representative examples include, but are not limited to, methanol, ethanol, propanol, butanol, 1, 2-propanediol, and the like.
A “pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. Such salts include:
acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as formic acid, acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3- (4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1, 2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, glucoheptonic acid, 4, 4’ -methylenebis- (3-hydroxy-2-ene-1-carboxylic acid) , 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or
salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like. It is understood that the pharmaceutically acceptable salts are non-toxic. Additional information on suitable pharmaceutically acceptable salts can be found in  Remington’s Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA, 1985, which is incorporated herein by reference in its entirety.
A “pharmaceutically acceptable carrier or excipient” means a carrier or an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes a carrier or an excipient that is acceptable for veterinary use as well as human pharmaceutical use. “A pharmaceutically acceptable carrier/excipient” as used in the specification and claims includes both one and more than one such excipient.
The term “about, ” as used herein, is intended to qualify the numerical values which it modifies, denoting such a value as variable within a margin of error. When no particular margin of error, such as a standard deviation to a mean value given in a chart or table of data, is recited, the term “about” should be understood to mean that range which would encompass±10%, preferably±5%, the recited value and the range is included.
The term “disease” as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disorder, ” “syndrome, ” and “condition” (as in medical condition) , in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms, and causes the human or animal to have a reduced duration or quality of life.
The term “combination therapy” means the administration of two or more therapeutic agents to treat a disease or disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.
The term “patient” is generally synonymous with the term “subject” and includes all mammals including humans. Examples of patients include humans, livestock such as cows, goats, sheep, pigs, and rabbits, and companion animals such as dogs, cats, rabbits, and horses. Preferably, the patient is a human.
“Treating” or “treatment” of a disease includes:
(1) inhibiting the disease, i.e., arresting (i.e., stabilizing) or reducing the development of the disease or its clinical symptoms; or
(3) relieving the disease, i.e., causing regression of the disease or its clinical symptoms.
A “therapeutically effective amount” means the amount of a compound of the present disclosure or a pharmaceutically acceptable salt thereof that, when administered to a patient for treating a disease, is sufficient to affect such treatment for the disease. The “therapeutically effective amount” will vary depending on the compound, the disease and its severity and the age, weight, etc., of the mammal to be treated.
Embodiments:
In further embodiments, the present disclosure includes:
Embodiment A:
A1. In embodiment A1, provided is a crystalline Form A polymorph of Compound 1 having an X-ray powder diffraction pattern as described in the first aspect in the Summary.
A2. In embodiment A2, the crystalline Form A polymorph of embodiment A1, is wherein the Form A X-ray powder diffraction pattern further comprises a peak at angular position 20.1, wherein the angular positions may vary by±0.2°2θ.
A3. In embodiment A3, the crystalline Form A polymorph of embodiment A1, is wherein the Form A X-ray powder diffraction pattern further comprises peaks at angular positions 12.9 and 20.1, wherein the angular positions may vary by±0.2°2θ.
A4. In embodiment A4, the crystalline Form A polymorph of embodiment A1, is wherein the Form A X-ray powder diffraction pattern further comprises peaks at angular positions 11.4, 12.9, and 20.1, wherein the angular positions may vary by±0.2°2θ.
A5. In embodiment A5, the crystalline Form A polymorph of embodiment A1, is wherein the Form A X-ray powder diffraction pattern further comprises peaks at angular positions 10.1, 11.4, 12.9, and 20.1, wherein the angular positions may vary by±0.2°2θ.
A6. In embodiment A6, provided is a crystalline Form A polymorph of Compound 1 having an X-ray powder diffraction pattern comprising at least two, at least three, at least four, at least five, at least six peaks, at least seven peaks, or at least eight peaks at angular positions selected from Table 1 below, wherein the angular positions may vary by±0.2°2θ.
Table 1
Figure PCTCN2021123248-appb-000005
Figure PCTCN2021123248-appb-000006
Figure PCTCN2021123248-appb-000007
A7. In embodiment A7, the crystalline Form A polymorph of embodiment A6, is wherein the at least two, at least three, at least four, at least five, or at least six peaks are selected from 10.1, 11.4, 12.9, 13.7, 15.8, 18.0, 19.6, 20.1, 21.4, 21.7, 25.0, and 26.0, wherein the angular positions may vary by±0.2°2θ.
A8. In embodiment A8, provided is a crystalline Form A polymorph of Compound 1 having an X-ray powder diffraction pattern substantially identical as shown in Fig 1.
A9. In embodiment A9, the crystalline Form A polymorph of any one of A1 to A8, are wherein the angular positions of the X-ray diffraction peaks denoted therein may vary by±0.1°2θ.
A10. In embodiment A10, the crystalline Form A polymorph of any one of A1 to A9, is substantially pure.
A11. In embodiment A10, the crystalline Form A polymorph of any one of A1 to A10, is substantially free of 3-fluoro-5- ( ( (1R, 2aS) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile enantiomer of Compound 1.
Embodiment B:
B1. In embodiment B1, provided is an amine solvate of Compound 1 as defined in the second aspect of the Summary.
B2. In embodiment B2, the amine solvate of embodiment B1, is wherein the amine is NHR 1R 2.
B3. In embodiment B3, the amine solvate of embodiment B1, is wherein the amine is R 3R 4-N- (CH 2n-NR 5R 6.
B4. In embodiment B4, the amine solvate of embodiment B1 or B2, is wherein R 1 is hydrogen and R 2 is C 2-C 12 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl.
B5. In embodiment B5, the amine solvate of embodiment B1, B2, or B4, is wherein R 1 is hydrogen and R 2 is C 2-C 12 alkyl.
B6. In embodiment B6, the amine solvate of embodiment B1, B2, or B4, is wherein R 1 is hydrogen and R 2 is C 3-C 7 cycloalkyl.
B6A. In embodiment B6A, the amine solvate of embodiment B1, B2, or B4, is wherein R 1 is hydrogen and R 2 is C 3-C 7 cycloalkyl-C 1-6 alkyl.
B7. In embodiment B7, the amine solvate of embodiment B1 or B2, is wherein R 1 is C 1-C 6 alkyl, C 2-C 6 alkenyl, or C 2-C 6 alkynyl and R 2 is C 1-C 6 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl.
B8. In embodiment B8, the amine solvate of embodiment B1, B2, or B7, is wherein R 1 is C 1-C 6alkyl and R 2 is C 1-C 6 alkyl.
B9. In embodiment B9, the amine solvate of embodiment B1, B2, or B7, is wherein R 1 is C 1-C 6alkyl and R 2 is C 3-C 7 cycloalkyl.
B10. In embodiment B10, the amine solvate of embodiment B1, B2, or B7, is wherein R 1 is C 1-C 6alkyl and R 2 is C 3-C 7 cycloalkyl-C 1-6 alkyl.
B10A. In embodiment B10A, the amine solvate of embodiment B1 or B2, is wherein R 1 and R 2 are independently C 3-C 7 cycloalkyl-C 1-6 alkyl.
B11. In embodiment B11, the amine solvate of embodiment B1 or B2, is wherein R 1 and R 2together with the nitrogen atom to which they are attached form cyclylamine.
B12. In embodiment B12, the amine solvate of embodiment B1 or B3, is wherein n is selected from 2 to 5. In a first embodiment of B12, n is 3 to 5. In a second embodiment of B12, n is 4 or 5.
B13. In embodiment B13, the amine solvate of embodiment B1 or B3, is wherein n is selected from 2 to 4. In a first embodiment of B13, n is 3 or4. In a second embodiment of B13, n is 3.
B14. In embodiment B14, the amine solvate of embodiment B1 or B3, is wherein n is 2 or 3.
B15. In embodiment B15, the amine solvate of embodiment B1 or B3, is wherein n is 2.
B16. In embodiment B16, the amine solvate of any one of embodiments B1, B3, and B12 to B15 and subembodiments contained therein, is wherein R 3 is hydrogen.
B17. In embodiment B17, the amine solvate of any one of embodiments B1, B3, and B12 to B15 and subembodiments contained therein, is wherein R 3 is C 1-C 6 alkyl.
B18. In embodiment B18, the amine solvate of any one of embodiments B1, B3, and B12 to B15 and subembodiments contained therein, is wherein R 3 is C 3-C 7 cycloalkyl.
B19. In embodiment B19, the amine solvate of any one of embodiments B1, B3, and B12 to B18 and subembodiments contained therein, is wherein R 4 is hydrogen.
B20. In embodiment B20, the amine solvate of any one of embodiments B1, B3, and B12 to B18 and subembodiments contained therein, is wherein R 4 is C 1-C 6 alkyl.
B21. In embodiment B21, the amine solvate of any one of embodiments B1, B3, and B12 to B18 and subembodiments contained therein, is wherein R 4 is C 3-C 7 cycloalkyl.
B22. In embodiment B22, the amine solvate of any one of embodiments B1, B3, and B12 to B21 and subembodiments contained therein, is wherein R 5 is hydrogen.
B23. In embodiment B23, the amine solvate of any one of embodiments B1, B3, and B12 to B21 and subembodiments contained therein, is wherein R 5 is C 1-C 6 alkyl.
B24. In embodiment B24, the amine solvate of any one of embodiments B1, B3, and B12 to B21 and subembodiments contained therein, is wherein R 5 is C 3-C 7 cycloalkyl.
B25. In embodiment B25, the amine solvate of any one of embodiments B1, B3, and B12 to B24 and subembodiments contained therein, is wherein R 6 is hydrogen.
B26. In embodiment B26, the amine solvate of any one of embodiments B1, B3, and B12 to B24 and subembodiments contained therein, is wherein R 6 is C 1-C 6 alkyl.
B27. In embodiment B27, the amine solvate of any one of embodiments B1, B3, and B12 to B24 and subembodiments contained therein, is wherein R 6 is C 3-C 7 cycloalkyl.
B28. In embodiment B28, the amine solvate of any one of embodiments B1, B2, and B4, to B6A is wherein the amine is ethylamine, n-propylamine, isopropylamine, n-butylamine, sec-butylamine, tert-butylamine, n-pentylamine, sec-pentylamine, pentyl-3-amine, neopentylamine, n-hexylamine, 2-hexylamine, 3-hexylamine, isohexylamine, 1-methylpentylamine, 2-ethylbutylamine, 2-methylpentylamine, 1, 1-dimethylbutylamine, 1, 3-dimethylbutylamine, 3, 3-dimethylbutylamine, 2-methyl-3-pentylamine, 3-methylpentylamine, 3-methyl-3-pentylamine, 3-methyl-2-pentylamine, 2-methylbutylamine, 1, 2, 2-trimethylpropylamine, cyclopropylamine, cyclobutylamine, cyclopentylamine, cyclohexylamine, cyclobutylmethylamine, cyclopropylmethylamine, 2-methylcyclopropylamine, 2-cyclopropylethylamine, 2-methylcyclopentylamine, 3-methylcyclopentylamine,  cyclopentylmethylamine, 2-cyclobutylethylamine, 3-cyclopropylpropylamine, 2-ethylcyclopropylamine, allyamine or propargylamine.
B29. In embodiment B29, the amine solvate of any one of embodiments B1, B2, B4, and B5 is wherein the amine is ethylamine, n-propylamine, isopropylamine, n-butylamine, sec-butylamine, tert-butylamine, n-pentylamine, sec-pentylamine, pentyl-3-amine, neopentylamine, n-hexylamine, 2-hexylamine, 3-hexylamine, isohexylamine, 1-methylpentylamine, 2-ethylbutylamine, 2-butylamine, 2-methylpentylamine, 1, 1-dimethylbutylamine, 1, 3-dimethylbutylamine, 3, 3-dimethylbutylamine, 2-methyl-3-pentylamine, 3-methylpentylamine, 3-methyl-3-pentylamine, 3-methyl-2-pentylamine, 2-methylbutyl-1-amine, or 1, 2, 2-trimethyl-propylamine.
B30. In embodiment B30, the amine solvate of any one of embodiments B1, B2, B4, and B6 is wherein the amine is cyclopropylamine, cyclobutylamine, cyclopentylamine, cyclohexylamine, 2-methylcyclopropylamine, 2-methylcyclopentylamine, 3-methyl-cyclopentylamine, or 2-ethylcyclopropylamine.
B31. In embodiment B31, the amine solvate of any one of embodiments B1, B2, and B7 to B10 is wherein the amine is N-methylethylamine, N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methylisopropylamine, N-allylmethylamine, N-methylpropargylamine, N-ethyl-n-propylamine, N-ethylisopropylamine, N-methylbutylamine, N-methyl-2-butylamine, N-tert-butyl-methyl-amine, N-methyl-cyclobutylamine, N-ethylcyclopropylamine, 1-cyclopropyl-N-methylmethanamine, N-ethyl-prop-2-yn-1-amine, N-ethylallylamine, N-methylpentylamine, N-methyl-2-pentylamine, N-methyl-3-pentylamine, N, 3-dimethylbutane-2-amine, N, 2-dimethyl-butane-2-amine, N, 3-dimethylbutane-2-amine, N-methylcyclopentylamine, N-ethyl-1-butylamine, N-ethyl-2-butylamine, N-ethyl-2-methyl-2-propylamine, N-ethyl-2-methylpropylamine, N-ethylcyclobutylamine, di-n-propylamine, di-isopropylamine, N-isopropylpropylamine, diallylamine, dipropargylamine, or allylpropargylamine.
B32. In embodiment B32, the amine solvate of any one of embodiments B1, B2, B7, and B8 is wherein the amine is N-methylethylamine, N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methyl-isopropylamine, N-ethyl-n-propylamine, N-ethyl-isopropylamine, N-methylbutylamine, N-methyl-2-butylamine, N-tert-butyl-methyl-amine, N-methylpentyl-1-amine, N-methyl-2-pentylamine, N-methyl-3-pentylamine, N, 3-dimethylbutane-2-amine, N, 2-dimethylbutane-2-amine, N, 3-dimethylbutane-2-amine, N-ethyl-1-butylamine, N-ethyl-2- butylamine, N-ethyl-2-methyl-2-propylamine, N-ethyl-2-methylpropylamine, di-n-propylamine, di-isopropylamine, or N-isopropylpropylamine.
B33. In embodiment B33, the amine solvate of any one of embodiments B1, B2, B7, and B9 is wherein the amine is N-methylcyclobutylamine, N-ethylcyclopropylamine, N-methylcyclopentylamine, or N-ethylcyclobutylamine.
B34. In embodiment B34, the amine solvate of any one of embodiments B1, B2, and B11 is wherein the amine is azetidine, pyrrolidine, piperidine, piperazine, morpholine, azepane, or azocane.
B35. In embodiment B35, the amine solvate of any one of embodiments B1, B3, and B12 to B16, B19, B20, B22, B23, and B25, is wherein the amine is ethylenendiamine, propane-1, 3-diamine, butane-1, 4-diamine, pentane-1, 5-diamine, hexane-1, 6-diamine, N 1, N 2-dimethylethane-1, 2-diamine, or N 1, N 2-diethylethane-1, 2-diamine.
B36. In embodiment B36, the amine solvate of embodiment B1 is wherein the amine is N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methyl-isopropylamine, N-ethyl-n-propylamine, N-ethyl-isopropylamine, azetidine, pyrrolidine, piperidine, or morpholine.
B37. In embodiment B37, the amine solvate of embodiment B1 or B36, is wherein the amine is diethylamine.
B38. In embodiment B38, the amine solvate of embodiment B37, is wherein the stoichiometric ratio of diethylamine to Compound 1 in the diethylamine solvate of Compound 1 is 1: 1.
B39. In embodiment B39, the amine solvate of any one of embodiments B1 to B38, is wherein the amine solvate of Compound 1 is a solid.
B39A. In embodiment B39A, the amine solvate of embodiment B37 or B38, is wherein the amine solvate of Compound 1 is a crystalline solid.
B40. In embodiment B40, the amine solvate of any one of embodiments B1 to B38, is wherein the amine solvate of Compound 1 is present in situ.
B41. In embodiment B41, the amine solvate of embodiment B40, is wherein the amine solvate of Compound 1 is present in a mixture comprising, one or more suitable organic solvents, Compound 1, and the amine.
B42. In embodiment B42, the amine solvate of embodiment B37, is wherein the diethylamine solvate of Compound 1 is present in a mixture comprising, one or more suitable organic solvent, Compound 1, and diethylamine.
B43. In embodiment B43, the amine solvate of any one of embodiments B40 to B42, is wherein the one or more suitable organic solvent is a polar organic solvent independently selected from ethers, alcohols, esters, halogenated alkanes, ketones, dimethylformamide, dimethylacetamide, acetonitrile, nitromethane, n-methyl pyrrolidinone, toluene, and xylene.
B44. In embodiment B44, the amine solvate of embodiment B43, is wherein the one or more suitable organic solvent is independently selected from n-pentanol, methanol (MeOH) , ethanol, n-propanol, isopropanol (IPA) , n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAC) , tetrahydrofuran (THF) , 2-methyl THF (2-MeTHF) , dimethoxyethane (DME) , toluene, acetone, methyl ethyl ketone (MEK) , nitromethane, acetonitrile (ACN) , and 1, 4-dioxane.
B45. In embodiment B45, the amine solvate of embodiment B43, is wherein the suitable organic solvent is methyl tert-butyl ether.
Embodiment C:
C1. In embodiment C1, provided is a crystalline Form B polymorph of diethylamine solvate of Compound 1 having an X-ray powder diffraction pattern as described in the third aspect in the Summary.
C2. In embodiment C2, the crystalline Form B of embodiment C1, is wherein the Form B X-ray powder diffraction pattern further comprises a peak at angular position 8.4, wherein the angular positions may vary by±0.2°2θ.
C3. In embodiment C3, the crystalline Form B of embodiment C1, is wherein the Form B X-ray powder diffraction pattern further comprises peaks at angular positions 8.4 and 23.5, wherein the angular positions may vary by±0.2°2θ.
C4. In embodiment C4, the crystalline Form B of embodiment C1, is wherein the Form B X-ray powder diffraction pattern further comprises peaks at angular positions 8.4, 23.5, and 11.7, wherein the angular positions may vary by±0.2°2θ.
C5. In embodiment C5, the crystalline Form B of embodiment C1, is wherein the Form B X-ray powder diffraction pattern further comprises peaks at angular positions 8.4, 23.5, 11.7, and 9.5, wherein the angular positions may vary by±0.2°2θ.
C6. In embodiment C6, provided is a crystalline Form B polymorph of diethylamine solvate of Compound 1 having an X-ray powder diffraction pattern comprising at least two, at least three, at least four, at least five, at least six, at least seven, or at least eight peaks at angular positions selected from in Table 2 below, wherein the angular positions may vary by±0.2°2θ.
Table 2
Figure PCTCN2021123248-appb-000008
Figure PCTCN2021123248-appb-000009
Figure PCTCN2021123248-appb-000010
C7. In embodiment C7, the crystalline Form B of embodiment C6, is wherein the at least two, at least three, at least four, at least five, or six peaks are selected from 8.4, 9.5, 11.7, 13.8, 21.3, and 23.5 wherein the angular positions may vary by±0.2°2θ.
C8. In embodiment C8, provided is a crystalline Form B of polymorph of Compound 1 having an X-ray powder diffraction pattern substantially identical as shown in Fig 2.
C9. In embodiment C9, the crystalline Form B of any one of C1 to C8, are wherein the peaks denoted therein have margin of error of±0.1°2θ.
Embodiment D:
D1. In embodiment D1, provided is a process of preparing an amine solvate of Compound 1 as described in the fourth aspect of the Summary.
D2. In embodiment D2, the process of embodiment D1, is wherein Compound 1 is contacted with the amine by adding Compound 1 to the amine or by adding the amine to a mixture of Compound 1 in one or more suitable organic solvents in Step (a1) .
D3. In embodiment D3, the process of embodiment D1 or D2, is wherein the amine is added to a mixture of Compound 1 in one or more suitable organic solvents in Step (a1) .
D4. In embodiment D4, the process of any one of embodiments D1 to D3, is wherein the amine is NHR 1R 2.
D5. In embodiment D5, the process of any one of embodiments D1 to D3, is wherein the amine is R 3R 4-N- (CH 2n-NR 5R 6.
D5A. In embodiment D5A, the process of any one of embodiments D1 to D4, is wherein R 1 is hydrogen and R 2 is C 2-C 12 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl.
D6. In embodiment D6, the process of any one of embodiments D1 to D4 and D5A, is wherein R 1 is hydrogen and R 2 is C 2-C 12 alkyl.
D7. In embodiment D7, the process of any one of embodiments D1 to D4 and D5A, is wherein R 1 is hydrogen and R 2 is C 3-C 7 cycloalkyl.
D7A. In embodiment D7A, the process of any one of embodiments D1 to D4 and D5A, is wherein R 1 is hydrogen and R 2 is C 3-C 7 cycloalkyl-C 1-6 alkyl.
D7B. In embodiment D7B, the process of any one of embodiments D1 to D4, is wherein R 1 is C 1-C 6 alkyl, C 2-C 6 alkenyl, or C 2-C 6 alkynyl and R 2 is C 1-C 6 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl.
D8. In embodiment D8, the process of any one of embodiments D1 to D4, and D7B, is wherein R 1 is C 1-C 6alkyl and R 2 is C 1-C 6 alkyl.
D9. In embodiment D9, the process of any one of embodiments D1 to D4, and D7B, is wherein R 1 is C 1-C 6alkyl and R 2 is C 3-C 7 cycloalkyl.
D10. In embodiment D10, process of any one of embodiments D1 to D4, and D7B, is wherein R 1 is C 1-C 6alkyl and R 2 is C 3-C 7 cycloalkyl-C 1-6 alkyl.
D10A. In embodiment D10A, the process of any one of embodiments D1 to D4, is wherein R 1 and R 2 are independently C 3-C 7 cycloalkyl-C 1-6 alkyl.
D11. In embodiment D11, the process of any one of embodiments D1 to D4, is wherein R 1 and R 2together with the nitrogen atom to which they are attached form cyclylamine.
D12. In embodiment D12, the process of any one of embodiments D1 to D3 and D5, is wherein n is selected from 2 to 5. In a first embodiment of D12, n is 3 to 5. In a second embodiment of D12, n is 4 or 5.
D13. In embodiment D13, the process of any one of embodiments D1 to D3 and D5, is wherein n is selected from 2 to 4. In a first embodiment of D13, n is 3 or4. In a second embodiment of D13, n is 3.
D14. In embodiment D14, the process of any one of embodiments D1 to D3 and D5, is wherein n is 2 or 3.
D15. In embodiment D15, the process of any one of embodiments D1 to D3 and D5, is wherein n is 2.
D16. In embodiment D16, the process of any one of embodiments D1 to D3 and D12 to D15 and subembodiments contained therein, is wherein R 3 is hydrogen.
D17. In embodiment D17, the process of any one of embodiments D1 to D3 and D12 to D15 and subembodiments contained therein, is wherein R 3 is C 1-C 6 alkyl.
D18. In embodiment D18, the process of any one of embodiments D1 to D3 and D12 to D15 and subembodiments contained therein, is wherein R 3 is C 3-C 7 cycloalkyl.
D19. In embodiment D19, the process of any one of embodiments D1 to D3 and D12 to D18 and subembodiments contained therein, is wherein R 4 is hydrogen.
D20. In embodiment D20, the process of any one of embodiments D1 to D3 and D12 to D18 and subembodiments contained therein, is wherein R 4 is C 1-C 6 alkyl.
D21. In embodiment D21, the process of any one of embodiments D1 to D3 and D12 to D18 and subembodiments contained therein, is wherein R 4 is C 3-C 7 cycloalkyl.
D22. In embodiment D22, the process of any one of embodiments D1 to D3 and D12 to D21 and subembodiments contained therein, is wherein R 5 is hydrogen.
D23. In embodiment D23, the process of any one of embodiments D1 to D3 and D12 to D21 and subembodiments contained therein, is wherein R 5 is C 1-C 6 alkyl.
D24. In embodiment D24, the process of any one of embodiments D1 and D3 and D12 to D21 and subembodiments contained therein, is wherein R 5 is C 3-C 7 cycloalkyl.
D25. In embodiment D25, the process of any one of embodiments D1 to D3 and D12 to D24 and subembodiments contained therein, is wherein R 6 is hydrogen.
D26. In embodiment D26, the process of any one of embodiments D1 to D3 and D12 to D24 and subembodiments contained therein, is wherein R 6 is C 1-C 6 alkyl.
D27. In embodiment D27, the process of any one of embodiments D1 to D3 and D12 to D24 and subembodiments contained therein, is wherein R 6 is C 3-C 7 cycloalkyl.
D28. In embodiment D28, the process of any one of embodiments D1 to D4 and D5A to D7 is wherein the amine is ethylamine, n-propylamine, isopropylamine, n-butylamine, sec-butylamine, tert-butylamine, n-pentylamine, sec-pentylamine, pentyl-3-amine, neopentylamine, n-hexylamine, 2-hexylamine, 3-hexylamine, isohexylamine, 1-methylpentylamine, 2-ethylbutylamine, 2-methylpentylamine, 1, 1-dimethylbutylamine, 1, 3-dimethylbutylamine, 3, 3-dimethylbutylamine, 2-methyl-3-pentylamine, 3-methylpentylamine, 3-methyl-3-pentylamine, 3-methyl-2-pentylamine, 2-methylbutylamine, 1, 2, 2-trimethylpropylamine, cyclopropylamine, cyclobutylamine, cyclopentylamine, cyclohexylamine, cyclobutylmethylamine, cyclopropylmethylamine, 2-methylcyclopropylamine, 2-cyclopropylethylamine, 2-methyl-cyclopentylamine, 3-methylcyclopentylamine, cyclopentylmethylamine, 2-cyclobutylethylamine, 3-cyclopropylpropyl-amine, 2-ethylcyclopropylamine, allyamine or propargylamine.
D29. In embodiment D29, the process of any one of embodiments D1 to D4, D5A and D6 is wherein the amine is ethylamine, n-propylamine, isopropylamine, n-butylamine, sec-butylamine, tert-butylamine, n-pentylamine, sec-pentylamine, pentyl-3-amine, neopentylamine, n-hexylamine, 2-hexylamine, 3-hexylamine, isohexylamine, 1-methylpentylamine, 2-ethylbutyl-amine, 2-butylamine, 2-methylpentylamine, 1, 1-dimethylbutylamine, 1, 3-dimethylbutylamine, 3, 3-dimethylbutylamine, 2-methyl-3-pentylamine, 3-methylpentylamine, 3-methyl-3-pentylamine, 3-methyl-2-pentylamine, 2-methylbutyl-1-amine, or 1, 2, 2-trimethyl-propylamine.
D30. In embodiment D30, the process of any one of embodiments D1 to D4, D5A and D7 is wherein the amine is cyclopropylamine, cyclobutylamine, cyclopentylamine, cyclohexylamine, 2-methylcyclopropylamine, 2-methylcyclopentylamine, 3-methylcyclopentyl-amine, or 2-ethylcyclopropylamine.
D31. In embodiment D31, the process of any one of embodiments D1 to D4, and D7B to D9 is wherein the amine is N-methylethylamine, N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methylisopropylamine, N-allylmethylamine, N-methylpropargylamine, N-ethyl-n-propylamine, N-ethylisopropylamine, N-methylbutylamine, N-methyl-2-butylamine, N-tert-butyl-methyl-amine, N-methyl-cyclobutylamine, N-ethylcyclopropylamine, 1-cyclopropyl-N-methylmethanamine, N-ethyl-prop-2-yn-1-amine, N-ethylallylamine, N-methylpentylamine, N-methyl-2-pentylamine, N-methyl-3-pentylamine, N, 3-dimethylbutane-2-amine, N, 2-dimethylbutane-2-amine, N, 3-dimethylbutane-2-amine, N-methylcyclopentylamine, N-ethyl-1-butylamine, N-ethyl-2-butylamine, N-ethyl-2-methyl-2-propylamine, N-ethyl-2-methylpropyl-amine, N-ethylcyclobutylamine, di-n-propylamine, di-isopropylamine, N-isopropylpropylamine, diallylamine, dipropargylamine, or allylpropargylamine.
D32. In embodiment D32, the process of any one of embodiments D1 to D4, D7B and D8 is wherein the amine is N-methylethylamine, N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methyl-isopropylamine, N-ethyl-n-propylamine, N-ethyl-isopropylamine, N-methylbutylamine, N-methyl-2-butylamine, N-tert-butyl-methyl-amine, N-methylpentyl-1-amine, N-methyl-2-pentylamine, N-methyl-3-pentylamine, N, 3-dimethylbutane-2-amine, N, 2-dimethylbutane-2-amine, N, 3-dimethylbutane-2-amine, N-ethyl-1-butylamine, N-ethyl-2-butylamine, N-ethyl-2-methyl-2-propylamine, N-ethyl-2-methylpropylamine, di-n-propylamine, di-isopropylamine, or N-isopropylpropylamine.
D33. In embodiment D33, the process of any one of embodiments D1 to D4, and D7B, and D9 is wherein the amine is N-methylcyclobutylamine, N-ethylcyclopropylamine, N-methylcyclopentylamine, or N-ethylcyclobutylamine.
D34. In embodiment D34, the process of any one of embodiments D1 to D4, and D11 is wherein the amine is azetidine, pyrrolidine, piperidine, piperazine, morpholine, azepane, or azocane.
D35. In embodiment D35, the process of any one of embodiments D1 to D3, D5, and D12 to D16, D19, D20, D22, D23, and D25, is wherein the amine is ethylenendiamine, propane-1, 3-diamine, butane-1, 4-diamine, pentane-1, 5-diamine, hexane-1, 6-diamine, N 1, N 2-dimethyl-ethane-1, 2-diamine, or N 1, N 2-diethylethane-1, 2-diamine.
D36. In embodiment D36, the process of embodiment D1 to D3 is wherein the amine is N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methyl-isopropylamine, N-ethyl-n-propylamine, N-ethyl-isopropylamine, azetidine, pyrrolidine, piperidine, or morpholine.
D37. In embodiment D37, the process of embodiment D1 to D3, or D36, is wherein the amine is diethylamine.
D38. In embodiment D38, the process of any one of embodiments D1 to D37, is wherein one or more anti-solvents are added to the mixture of Compound 1 of Step (a1) to obtain a solid amine solvate of Compound 1.
D39. In embodiment D39, the process of any one of embodiments D1 to D37, is wherein one or more anti-solvents and a solid crystalline seed of Compound 1 and/or a solid crystalline seed of the amine solvate of Compound 1 are added to the mixture of Step (a1) to obtain a solid amine solid of Compound 1.
D39A. In embodiment D39A, the process of any one of embodiments D1 to D39, is wherein the mixture of Step (a1) is a solution.
D40. In embodiment D40, the process of embodiment D38 or D39A is wherein the one or more anti-solvents in Step (b1) are independently selected from alkanes and water. In a subembodiment of embodiment D40, the one or more anti-solvents are independently selected from the group consisting of water, n-heptane, n-hexane, isooctane, pentane, cyclohexane, and cyclopentane.
D41. In embodiment D41, the process of embodiment D38, D39, D39A, or D40, is wherein the anti-solvent in Step (b1) is n-heptane.
D42. In embodiment D42, the process of any one of embodiments D2 to D41, is wherein the one or more suitable organic solvents of Step (a1) are polar organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylformamide, and dimethylacetamide.
D43. In embodiment D43, the process of embodiment any one of embodiments D2 to D41, is wherein the one or more suitable organic solvents of Step (a1) are independently selected from the group consisting of methanol, ethanol, n-propanol, isopropanol, n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAc) , tetrahydrofuran (THF) , 2-methyl THF (2-MeTHF) , dimethoxyethane (DME) , toluene, acetone, methyl ethyl ketone (MEK) , nitromethane, acetonitrile (ACN) , and 1, 4-dioxane.
D44. In embodiment D44, the process of embodiments D42, is wherein the suitable organic solvent of Step (a1) is MTBE.
D45. In embodiment D45, the process of any one of embodiments D37 to D44, is wherein the diethylamine solvate of Compound 1 is a crystalline solid.
D46. In embodiment D46, the process of embodiment D45, is wherein the diethylamine solvate of Compound 1 is crystalline Form B polymorph having an X-ray powder diffraction pattern as described in the Summary.
D47-D54. In embodiments D47 to D54, the process of embodiment D46, is wherein the crystalline Form B has an X-ray powder diffraction pattern as described in above embodiments C2 to C9, respectively.
D55. In embodiment D55, the process of any one of embodiments D1 to D54, is wherein the amine solvate of Compound 1 is converted to Compound 1.
D56. In embodiment D56, the process of any one of embodiments D1 to D55, is wherein the amine solvate of Compound 1 is converted to Compound 1 by heating the amine solvate to remove the amine from the amine solvate.
D57. In embodiment D57, the process of any one of embodiments D1 to D55, is wherein the amine solvent of Compound 1 is converted to Compound 1 by partitioning the amine solvate between one or more suitable organic solvents and an aqueous acidic solution.
D58. In embodiment D58, the process of embodiment D57, is wherein the one or more suitable organic solvents containing Compound 1 is isolated and concentrated to provide a solid form of Compound 1.
D59. In embodiment D59, the process of embodiment D56 or D58, is wherein the crystalline Form A polymorph Compound 1 has an X-ray powder diffraction pattern as described in the Summary is produced.
D60-D69. In embodiment D60 to D69, the process of embodiment D59, is wherein crystalline Form A polymorph of Compound 1 has an X-ray powder diffraction pattern as described in above embodiments A2 to A11, respectively, is produced.
D70. In embodiment D70, the process of any one of embodiments D1 to D69, further comprises preparing the Compound 1 of Step (a1) , comprising:
(a) treating 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile with a deoxyfluorinating agent in the presence of a base in a suitable organic solvent;
(b) purifying the mixture from Step (a) to obtain Compound 1.
D71. In embodiment D71, the process of embodiment D70, is wherein the deoxyfluorinating agent is pyridine-2-sulfonyl fluoride and the base is 1, 8-diazabicyclo- [5.4.0] undec-7-ene or 7-methyl-1, 5, 7-triaza-bicyclo- [4.4.0] dec-1-ene.
D72. In embodiment D72, the process of embodiment D70 or D71, is wherein the suitable organic solvent is tetrahydrofuran.
Embodiment E:
E1. In embodiment E1, provided is a process of preparing crystalline Form B polymorph of diethylamine solvate of Compound 1 having an X-ray powder diffraction pattern as described in the fifth aspect of the Summary.
E2. In embodiment E2, the process of embodiment E1, is wherein Compound 1 is contacted with diethylamine by adding Compound 1 to the diethylamine or by adding the diethylamine to a mixture of Compound 1 in one or more suitable organic solvents in Step (a2) .
E3. In embodiment E3, the process of embodiment E1, is wherein the diethylamine is added to the mixture of Compound 1 in one or more suitable organic solvents in Step (a2) .
E4. In embodiment E4, the process of any one of embodiments E1 to E3, is wherein one or more anti-solvents are added to the mixture of Step (a2) to obtain crystalline Form B polymorph of the diethylamine solvate of Compound 1.
E5. In embodiment E5, the process of any one of embodiments E1 to E3, is wherein one or more anti-solvents and a solid crystalline seed of Compound 1 and/or a solid crystalline seed of the diethylamine solvate of Compound 1 are added to the mixture of Step (a2) to obtain crystalline Form B polymorph of the diethylamine solvate of Compound 1.
E5A. In embodiment E5A, the process of any one of embodiments E1 to E5, is wherein the mixture of Step (a2) is a solution.
E6. In embodiment E6, the process of any one of embodiments E1 to E5A, is wherein the one or more anti-solvents in Step (b2) are independently selected from alkanes and water. In a subembodiment of embodiment E6, the one or more anti-solvents are independently selected from the group consisting of water, n-heptane, n-hexane, isooctane, pentane, cyclohexane, and cyclopentane.
E7. In embodiment E7, the process of any one of embodiments E1 to E6, is wherein the anti-solvent in Step (b2) is n-heptane.
E8. In embodiment E8, the process of any one of embodiments E1 to E7, is wherein the solution of Compound 1 in Step (b2) is in one or more suitable organic solvents are polar organic  solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylacetamide, and dimethylformamide.
E9. In embodiment E9, the process of embodiment any one of embodiments E1 to E7, is wherein the one or more suitable organic solvents are independently selected from the group consisting of methanol, ethanol, n-propanol, isopropanol, n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAc) , etrahydrofuran (THF) , 2-methyl THF (2-MeTHF) , dimethoxyethane (DME) , toluene, acetone, methyl ethyl ketone (MEK) , nitromethane, acetonitrile (ACN) , and 1, 4-dioxane.
E10. In embodiment D10, the process of embodiment E9, is wherein the suitable organic solvent is MTBE.
E11. In embodiment E11, the process of any one of embodiments E1 to E10, is wherein the diethylamine of solvate Compound 1 is optionally converted to Compound 1 by heating the diethylamine solvate of Compound 1 to remove the diethylamine from the diethylamine solvate of Compound 1.
E12. In embodiment E12, the process of any one of embodiments E1 to E10, is wherein the diethylamine of solvate Compound 1 is optionally converted to Compound 1 by partitioning the diethylamine solvate of Compound 1 between one or more suitable organic solvent and an aqueous acidic solution. In one embodiment, the acid is hydrochloric acid.
E13. In embodiment E13, the process of embodiment E12, is wherein the one or more organic solvents containing Compound 1 is isolated and concentrated to provide a solid form of Compound 1.
E14. In embodiment E14 the process of any one of embodiments E11 to E13, wherein is crystalline Form A polymorph of Compound 1 has an X-ray powder diffraction pattern as defined in any one of Embodiments A1 to A11, is produced.
Embodiment F:
F1. In embodiment F1, provided is a process of preparing crystalline Form A polymorph of Compound 1 from diethylamine solvate of Compound 1 as described in the sixth aspect of the Summary.
F2 to F10. In embodiments F2 to F10, the process of embodiment F1, is wherein the diethylamine solvate of Compound 1 is crystalline Form B polymorph having an X-ray powder diffraction pattern as described in above embodiments C1 to C9, respectively.
F11. In embodiment F11, the process of any one of embodiments F1 to F10, is wherein the diethylamine is removed from the diethylamine solvate of Compound 1 by heating the diethylamine solvate of Compound 1.
F12. In embodiment F12, the process of any one of embodiments F1 to F11, is wherein the diethylamine is removed from the diethylamine solvate of Compound 1 by partitioning the diethylamine solvate of Compound 1 between one or more suitable organic solvent and an aqueous acidic solution. In one embodiment the acid is hydrochloric acid.
F13. In embodiment F13, the process of embodiment F12, is wherein the one or more suitable organic solvents is isolated and concentrated to provide crystalline Form A polymorph of Compound 1 having an X-ray powder diffraction pattern as described in any one of embodiments A1 to A11 above.
F14. In embodiment F14, the process of any one of embodiments F1 to F11, is wherein the crystalline Form A polymorph of Compound 1 is recrystallized from one or more suitable organic solvents.
F15. In embodiment F15, the process of embodiment F14, is wherein the crystalline Form A polymorph of Compound 1 is dissolved in one or more suitable organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylacetamide and dimethylformamide.
F16. In embodiment F16, the process of embodiment F15, is wherein the one or more suitable organic solvents are selected from the group consisting methanol, ethanol, n-propanol, isopropanol, n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAc) , tetrahydrofuran (THF) , 2-methyl THF (2-MeTHF) , dimethoxyethane (DME) , toluene, acetone, methyl ethyl ketone (MEK) , nitromethane, acetonitrile (ACN) , and 1, 4-dioxane. In an embodiment the suitable organic solvent is isopropyl acetate.
F17. In embodiment F17, the process of embodiment F15 or F16, is wherein one or more anti-solvents and/or solid crystalline seed of Compound 1 are added.
F18. In embodiment F18, the process of embodiment 17, is wherein the one or more anti-solvents are independently selected from alkanes and water.
F19. In embodiment F19, the process of embodiment F18, is wherein the one or more anti-solvents are independently selected from the group consisting of water, n-heptane, n-hexane, isooctane, pentane, cyclohexane, and cyclopentane.
F20. In embodiment F20, the process of embodiment F19, is wherein the anti-solvent is n-heptane.
Embodiment G:
G1. In embodiment G1, provided is a process of making crystalline Form A polymorph of Compound 1 having an X-ray powder diffraction pattern as described in the seventh aspect of the Summary.
G2. In embodiment G2, the process of embodiment G1, is wherein one or more anti-solvents are added to a solution of Compound 1 in one or more suitable organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylacetamide and dimethylformamide.
G3. In embodiment G3, the process of embodiment G2 is wherein the one or more suitable organic solvents are selected from the group consisting methanol, ethanol, n-propanol, isopropanol, n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAc) , tetrahydrofuran (THF) , 2-methyl THF (2-MeTHF) , dimethoxyethane (DME) , toluene, acetone, methyl ethyl ketone (MEK) , nitromethane, acetonitrile (ACN) , and 1, 4-dioxane. In an embodiment the suitable organic solvent is isopropyl acetate.
G4. In embodiment G4, the process of embodiment G2 or G3, is wherein one or more anti-solvents is added.
G5. In embodiment G5, the process of embodiment G4, is wherein the one or more anti-solvents is independently selected from alkanes and water.
G6. In embodiment G6, the process of embodiment G5, is wherein the one or more anti-solvents is independently selected from the group consisting of water, n-heptane, n-hexane, isooctane, pentane, cyclohexane, and cyclopentane.
G7. In embodiment G7, the process of embodiment G6, is wherein the anti-solvent is n-heptane.
G8. In embodiment G8, the process of any one of embodiments G1 to G7, is wherein the process is carried out at about 0℃ to about 70℃.
Utility
Compound 1 is useful for the treatment of HIF-2αmediated diseases, which include but are not limited to, various types of cancer, liver disease such as nonalcoholic steatohepatitis (NASH) , inflammatory disease such as inflammatory bowel disease (IBD) , pulmonary diseases such as pulmonary arterial hypertension (PAH) , and iron load disorders.
HIF-2αplays an important role in the initiation and progression of many human cancers. Many extensive studies have demonstrated the critical role of increased HIF-2αactivity in driving clear cell renal cell carcinoma (ccRCC) (see review by Shen and Kaelin, Seminars in Cancer Biology 23: 18-25, 2013) . Abnormal HIF-2αactivity is largely due to loss of function of a tumor suppressor, VHL. It is known that over eighty percent of ccRCC have defective VHL either through deletion, mutation or disturbed post-translational modification. Defective VHL leads to constitutively active HIF-αproteins regardless of oxygen level. Various studies employing gain-of-function and loss-of-function approaches in mouse models have demonstrated that HIF-2αis the key oncogenic substrate of VHL (see Kondo, et al. Cancer Cell 1: 237-246, 2002; Kondo, et al. PLoS Biology 1: 439-444, 2002; Maranchi, et al. Cancer Cell 1: 247-255, 2002; Zimmer, et al. Mol. Cancer Res 2: 89-95, 2004) . For example, knockdown of HIF-2αin VHL-null tumors inhibited tumor formation; while reintroduction of VHL and overexpression of HIF-2αovercame the tumor suppressive role of VHL. Moreover, single nucleotide polymorphism in HIF-2α, is associated with resistant to PHD-mediated degradation, has been linked to an increased risk of developing RCC. In addition to serving as an archetypical tumor-initiating event in ccRCC, the VHL-HIF-2αaxis has also been implicated in ccRCC tumor metastasis through its downstream CXCR4 and CYTIP (see Vanharanta et al. Nature Medicine 19: 50-59, 2013; Peter Staller et al. Nature. 2003 Sep 18; 425 (6955) : 307-11) . Taken together, these studies strongly support the potential therapeutic utility of HIF-2αtargeted agents for the treatment of ccRCC.
Defective VHL not only predisposes patients to kidney cancer (with a70%lifetime risk) , but also to hemangioblastomas, pheochromocytoma, endolymphatic sac tumors, and pancreatic neuroendocrine tumors. Tumors derived from defective VHL are frequently driven by the constitutively active downstream HIF-αproteins, with the majority of these dependent on HIF-2α activity (see Maher, et al. Eur. J. Hum. Genet. 19: 617-623, 2011) . Both genetic and epigenetic mechanisms can lead to the loss of function in VHL. Epigenetic inactivation of VHL expression and thus constitutive activation of HIF-αproteins has been found in many cancers including RCC, multiple myeloma, retinoblastoma, NSCLC, pancreatic endocrine tumors, squamous cell carcinoma, acute myeloid leukemia, myelodysplastic syndrome, and esophageal squamous cell carcinoma (see reviewed in Nguyen, et al. Arch. Phann. Res 36: 252-263, 2013) . HIF-2αhas also been linked to cancers of the retina, adrenal gland and pancreas through both loss of function in VHL and activating mutations in HIF-2α. Recently, gain-of-function HIF-2αmutations have been identified in erythrocytosis and paraganglioma with polycythemia (see Zhuang, et al. NEJM 367: 922-930, 2012; Percy, et al. NEJM 358: 162-168, 2008; and Percy, et al. Am. J. Hematol. 87: 439-442, 2012) . Notably, many of the known HIF-2αtarget gene products (e.g., VEGF, PDGF, and cyclin Dl) have been demonstrated to play pivotal roles in cancers derived from kidney, liver, colon, lung, and brain. Thus, a HIF-2αtargeted therapy could be beneficial for the above cancers when driven by these signaling events downstream of abnormal HIF-2αpathway activation. In addition to loss of function in VHL and activating mutation of HIF-2α, HIF-αproteins are also frequently upregulated in the intratumor environment of rapidly growing tumors, due to the hypoxic condition resulting from poor vascularization in large tumors. The activated HIF-αpathways, in turn, further promotes tumor cell survival and proliferation by transcriptionally upregulating various essential factors.
A large body of studies have demonstrated a correlation between HIF-2αoverexpression and poor prognosis in various cancers including cancers of astrocytoma, breast, cervical, colorectal, glioblastoma, glioma, head and neck, liver, non-small cell lung, melanoma, neuroblastoma, ovarian, and prostate, thereby supporting the pursuit of HIF-2αas a therapeutic target in treating these cancers (see reviewed in Keith, et al. Nature Rev. Cancer 12: 9-22, 2012) . HIF-2αhas been demonstrated to augment the growth of APC mutant colorectal cancer through its regulation of genes involved in proliferation, iron utilization and inflammation (see Xue, et al. Cancer Res 72: 2285-2293, 2012; and Xue and Shah, Carcinogenesis 32: 163-169, 2013) . In hepatocellular carcinoma (HCC) , knock-down of HIF-2αin preclinical models led to the inhibition of cell proliferation in vitro and tumor growth in vivo through the downregulation of VEGF and cyclin D 1 (see He, et al. Cancer Sci. 103: 528-534, 2012) . In NSCLC, around 50%of patients exhibited overexpression of HIF-2αprotein, which strongly correlates with higher VEGF expression and more importantly, reduced overall survival. Interestingly, HIF-1αdoes not correlate with reduced overall survival in lung cancer patients even though its expression is also  often increased (see Giatromanolaki, et al. Br. J. Cancer 85: 881-890, 2001) . Extensive studies in mice engineered with both non-degradable HIF-2αand mutant KRAS tumors have demonstrated an increased tumor burden and a decreased survival when compared to mice with only mutant KRAS expression (see Kim, et al. J. Clin. Invest. 119: 2160-2170, 2009) . These studies demonstrate that HIF-2αpromotes tumor growth and progression in lung cancer, and also negatively correlates with clinical prognosis.
HIF-2αs activity has been linked to the progression of chronic obstructive pulmonary disease (COPD) , in addition to lung cancer, in mouse models (see Karoor, et al. Cancer Prev. Res. 5: 1061-1071, 2012) . HIF-2αactivity has also been demonstrated to be important in cancers of the central nervous system (see Holmquist-Mengelbier, et al. Cancer Cell 10: 413-423, 2006 and Li, et al. Cancer Cell 15: 501-513, 2009) . HIF-2αknockdown reduced tumor growth in preclinical animal models of neuroblastoma, Conversely, increased level of HIF-2αcorrelated with advanced disease, poor prognosis and higher VEGF levels, which likely contribute to the poor clinical outcome. Similarly, higher HIF-2αexpression has been correlated with a poor survival in glioma. Experimentally, inhibition of HIF-2αin glioma stem cells reduced cell proliferation and survival in vitro and tumor initiation in vivo. While HIF-lαis expressed in both neural progenitors and brain tumor stem cells, HIF-2αis found exclusively in the latter. Moreover, survival of glioma patients correlates to with HIF-2α, but not HIF-1αlevel.
One of downstream HIF-2αeffector is cyclin D, an essential partner for the activation of CDK4 and CDK6. Therefore, administration of a HIF-2αinhibitor with CDK4/6 inhibitors, including abemaciclib
Figure PCTCN2021123248-appb-000011
palbociclib
Figure PCTCN2021123248-appb-000012
andribociclib
Figure PCTCN2021123248-appb-000013
shouldresult in downregulation of cyclin D, thereby increasing antiproliferative effects of CDK4/6 inhibitors. A recent study (Nicholson et al Sci Signal. 2019 Oct 1; 12 (601) ) suggests that the antiproliferative effects of CDK4/6 inhibition were synergistic with HIF-2αinhibition in HIF-2α-dependent VHL-/-ccRCC cells.
Radiation therapy is frequently used for approximately 50%of cancer patients, either alone or in combination with other therapies. However, the hypoxia microenvironment within the tumor has long been associated with resistance to radiation therapy. Bhatt and co-workers found that decreased level of HIF-2αleads to increased sensitivity to ionizing radiation in renal cell carcinoma cell lines (see Bhatt, et al. BJU Int. 102: 358-363, 2008) . Furthermore, mechanistic studies from Bertout et. al, have demonstrated that HIF-2αinhibition enhances the effectiveness of radiation through increased p53-dependent apoptosis (see Bertout, et al. PNAS 106: 14391-14396,  2009) . Thus, HIF-2αtargeted therapy could improve the response to radiation therapy in various cancers.
Somatostatinomas are somatostatin-producing neuroendocrine tumors that are rare, but often malignant. It has been found that HIF-2αmutations lead to the disruption of the prolyl hydroxylation domain (PHD) of HIF-2α, thus abolish the modification by PHDs, and subsequently reduce HIF-2αdegradation mediated by VHL (see Yang, et al. Blood. 121: 2563–2566, 2013) . The stabilized HIF-2αcan then translocate to the nucleus, driving increased expression of hypoxia-related genes to contribute to somatostatinoma. Thus, a HIF-2αinhibitor will provide an alternative approach in treating somatostatinoma.
Polycythaemia is a hematologic disorder characterized by elevated hematocrit (the volume percentage of red blood cells in the blood) , also known as erythrocytosis. Gain-of-function mutations in HIF-2αare associated with autosomal dominant erythrocytosis (see Percy, et al. N. Engl. J. Med. 358: 162–8, 2008 and Wilson et al. Case Rep Hematol. 6373706, 2016) . In addition, mutations in PHD of HIF-2α, which is responsible in signaling HIF-2αfor ubiquitination and degradation by VHL, have also been found to drive polycythaemia. Thus, inhibiting HIF-2αn, which is stabilized either by gain of function HIF-2αmutations or by loss of function mutations in PHD, VHL, by an HIF-2αinhibitor should be able to suppress HIF-2αdownstream genes, such as EPO and thereby reducing hematocrit of polycythaemia.
Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors that often develop on a background of predisposing genetic mutations, including loss of function in VHL or PHD2 or activating mutations of HIF-2α, all of which result in highly expressed HIF-2αprotein and subsequently downstream genes to promote oncogenic progression (see Dahia, Nat Rev Cancer. 14: 108-19, 2014) . Furthermore, germline heterozygous mutations in genes encoding succinate dehydrogenase (SDH) subunits and the SDH complex assembly factor 2 protein (SDHAF2) have been described in patients with hereditary phaeochromocytoma and paraganglioma (PPGL) . These mutations can lead to the accumulation of succinate, which in turn causes an inhibition of prolyl-hydroxylases that is essential in mediating ubiquitination/degradation of HIF proteins by VHL complex. Pituitary adenoma has been frequently found to be co-existing with PPGLs. Thus, inhibiting HIF-2αshould be useful for treating both PPGLs and pituitary tumors. Succinate dehydrogenase subunits mutations have also been associated with gastrointestinal stromal tumors (GIST) , thus supporting exploration of HIF-2αinhibitor for the treatment of GIST (see Janeway, et al. Proc. Natl Acad. Sci. USA 108: 314–318, 2011) .
Loss-of-function mutations of fumarate hydratase (FH) predispose patients to the autosomal dominant syndrome of both cutaneous and uterine leiomyomatosis. It has been suggested that activation of HIF proteins contributes to FH-associated tumor development by activation of hypoxia pathways. (see O'Flaherty, et al. Hum Mol Genet. 19: 3844–3851, 2010 and Wei, et al. J Med Genet. 43: 18-27, 2006) . Furthermore, high expression of HIF-2αis found in leiomyosarcomas, a rare neoplasm of smooth-muscle origin (see Mayer, et al. Cancer Res. 68: 4719, 2008) Thus, inhibition of HIF-2αcould be beneficial in treating both leiomyomas and leiomyosarcomas.
Retinal capillary hemangioblastomas can be the ocular manifestations of VHL diseases, which are caused by loss of tumor suppressor VHL. Upregulation of HIF-2αupon loss of VHL has been detected in retinal hemangioblastoma patients and is indicated to contribute to the aggressive course of retinal hemangioblastomas, resulting in the resistance to multiple anti-VEGF and radiation therapies (see Wang, et al. Graefes Arch. Clin. Exp. Ophthalmol. 252: 1319–1327, 2014) . Moreover, uncontrolled blood vessel growth is a central pathological component of many human blindness disorders, including diabetic retinopathy, age-related macular degeneration, glaucoma, and retinopathy of prematurity. Neuronal cell death and vision loss observed in these diseases are often caused by aberrant, leaky vessels, results of pathological neovascularization (see Krock, et al. Genes Cancer. 2: 1117–1133, 2011) . Given the causal role of HIFs in neovascularization, inhibitor of HIF-2αmay have potential utility in treating various diseases of blindness. In fact, systemic reduction of HIF-2αexpression with a hypomorphic Hif-2αallele caused marked decreases in retinal neovascularization that was accompanied by defects in EPO expression (see Morita, et al. EMBO J. 22: 1134-46, 2003) .
In addition to a direct role in promoting the initiation, progression and metastasis of tumor cells (e.g. ccRCC) , HIF-2αalso indirectly contributes to tumorigenesis through augmenting the immunosuppressive effect of hypoxia within the tumor microenvironment. Expression of HIF-2αhas been detected in cells of the myeloid lineage (see Talks KL, et dal. Am J Pathol. 2000; 157 (2) : 411–421) . For example, HIF-2αis shown to favor the polarization of macrophages to the immunosuppressive M2 phenotype and enhances migration and invasion of tumor-associated macrophages (see Imtiyaz HZ et al. J Clin Invest. 2010; 120 (8) : 2699–2714) . Thus, increased level of HIF-2αin tumor-associated macrophages (TAMs) is associated with high-grade human tumors and correlates with poor prognosis. Furthermore, HIF-2αcan indirectly promote additional immunosuppressive pathways (e.g. adenosine and arginase etc. ) by modulating the expression of key signaling regulators such as adenosine A2B/A2A receptors and arginase. These data support  that HIF-2αis a potential therapeutic target for treating a broader range of inflammatory disorders and cancer either as a single agent or in combination with other therapeutic agents e.g., immunotherapies.
Due to the key roles of HIF-2αproteins in regulating physiological response to the fluctuation of oxygen levels, they have been causally associated with many hypoxia-related pathological processes in addition to cancer. One such disease is PAH, a debilitating and life-threatening disease with very poor prognosis. Recent studies demonstrated that HIF-2αcontributes to the process of hypoxic pulmonary vascular remodeling, reduced plasticity of the vascular bed, and ultimately, debilitating PAH (see Andrew S., et al. Proc Natl Acad Sci U S A. 2016 Aug 2; 113 (31) : 8801–8806, Tang H, et al. Am J Physiol Lung Cell Mol Physiol. 2018 Feb 1; 314 (2) : L256-L275. ) . These studies offered new understanding in the role of pulmonary endothelial HIF-2αin regulating the pulmonary vascular response to hypoxia, and offer an much needed new therapeutic strategy by targeting HIF-2α. Another example of hypoxia-related pathological processes is IBD, a chronic relapsing inflammatory disease of the intestine. It is found that intestinal inflammation and subsequently IBD arose when a dysregulated epithelial oxygen tension occurs and intensifies across epithelial villi in the intestine (see Shah Y. M., Molecular and Cellular Pediatrics, 2016 Dec; 3 (1) : 1) . HIF-2αactivation contributes to IBD, while HIF-1αin intestinal epithelial cells is considered as a major protective factor in IBD (see Karhausen J, et al. J Clin Invest. 2004; 114 (8) : 1098–1106; Furuta GT, et al. J Exp Med. 2001; 193 (9) : 1027–1034) . Mechanistically, HIF-2αactivation not only leads to the upregulation of pro-inflammatory cytokines which promotes IBD directly, but also results in loss of intestine barrier integrity, thus indirectly contributes to the manifestation of IBD. (see Xue X, et al. Gastroenterology. 2013; 145 (4) : 831–841; Glover LE, et al. Proc Natl Acad Sci U S A. 2013; 110 (49) : 19820–19825) . Therefore, an HIF-2αinhibitor holds the promise of reverting the pro-inflammatory condition and increasing the intestinal barrier integrity, thus alleviate the symptoms of IBD.
HIF-2αinhibitor also represents a novel therapeutic approach in NASH, for which limited therapeutic options are available. A recent study showed that an intestine-specific disruption of HIF-2αled to a significant reduction of hepatic steatosis and obesity induced by high-fat-diet. Mechanistically, intestine HIF-2αpositively regulates the gene encoding neuraminidase 3, thus regulates ceramide metabolism which contributes to the development of NASH (see Xie C, et al. Nat Med. 2017 Nov; 23 (11) : 1298-1308. ) . Therefore, a HIF-2αinhibitor should have preventive and therapeutic effects on metabolic disorders, such as NASH.
Several connections between the level of HIF-2αand iron homeostasis have been identified (see Peyssonnaux C et al, Cell Cycle. 2008; 7 (1) : 28–32) . Multiple studies have demonstrated the important role of HIF-2αin iron load disorders. HIF-2α, not HIF-1α, has emerged as an important “local” regulator of intestinal iron status through its regulation of various genes essential in iron transport and absorption (see Mastrogiannaki M, et al. J Clin Invest. 2009; 119 (5) : 1159–1166) . Therefore, a small molecule inhibitor that targets HIF-2αholds promise of improving iron homeostasis in patients with iron disorders.
Accordingly, the present disclosure provides a method for treating or lessening the severity of a disease, condition, or disorder where activation or over activation of HIF-2αis implicated in the disease state. In another aspect, the present disclosure provides a method of treating renal cell carcinoma of a subject with Compound 1.
HIF-2αinhibitors also have therapeutic potentials for a broad range of non-cancer indications including but not limited to NASH, IBD, PAH, and iron overload.
Pharmaceutical Compositions
In general, Form A polymorph of Compound 1 will be administered in a therapeutically effective amount by any of the accepted modes of administration for agents that serve similar utilities. Therapeutically effective amounts of Form A polymorph of Compound 1 may range from about 200 mg/per day to about 1000 mg/per day, which can be administered in single or multiple doses. For oral administration, the compositions can be provided in the form of tablets containing about 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 750, 800, 900, and 1000 milligrams of the active ingredient. The actual amount of Form A polymorph of Compound 1 will depend upon numerous factors such as the severity of the disease to be treated, the age and relative health of the patient, the potency of the compound being utilized, the route and form of administration, and other factors.
In general, Form A polymorph of Compound 1 will be administered as pharmaceutical compositions by any one of the following routes: oral, systemic (e.g., transdermal, intranasal or by suppository) , or parenteral (e.g., intramuscular, intravenous or subcutaneous) administration. The preferred manner of administration is oral using a convenient daily dosage regimen, which can be adjusted according to the degree of affliction. Compositions can take the form of tablets, pills, capsules, semisolids, powders, sustained release formulations, solutions, suspensions, elixirs, aerosols, or any other appropriate compositions.
The choice of formulation depends on various factors such as the mode of drug administration (e.g., for oral administration, formulations in the form of tablets, pills or capsules, including enteric coated or delayed release tablets, pills or capsules are preferred) and the bioavailability of the drug substance.
The compositions are comprised of in general, Form A polymorph of Compound 1 in combination with at least one pharmaceutically acceptable excipient. Acceptable excipients are non-toxic, aid administration, and do not adversely affect the therapeutic benefit of Form A polymorph of Compound 1. Such excipient may be any solid, liquid, semi-solid or, in the case of an aerosol composition, gaseous excipient that is generally available to one of skill in the art.
Solid pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice flour, chalk, silica gel, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk and the like. Liquid and semisolid excipients may be independently selected from glycerol, propylene glycol, water, ethanol and various oils, including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, mineral oil, sesame oil, etc. Preferred liquid carriers, particularly for injectable solutions, include water, saline, aqueous dextrose, and glycols.
Form A polymorph of Compound 1 may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in powder form or in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or sterile pyrogen-free water, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
Formulations for parenteral administration include aqueous and non-aqueous (oily) sterile injection solutions of Form A polymorph of Compound 1 which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection  suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of Form A polymorph of Compound 1 to allow for the preparation of highly concentrated solutions.
In addition to the formulations described previously, Form A polymorph of Compound 1 may also be formulated as a depot preparation. Such long-acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, Form A polymorph of Compound 1 may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
For buccal or sublingual administration, the compositions may take the form of tablets, lozenges, pastilles, or gels formulated in conventional manner. Such compositions may comprise Form A polymorph of Compound 1 in a flavored basis such as sucrose and acacia or tragacanth.
Other suitable pharmaceutical excipients and their formulations are described in Remington’s Pharmaceutical Sciences, edited by E. W. Martin (Mack Publishing Company, 20th ed., 2000) .
The level of Form A polymorph of Compound 1 in a formulation can vary within the full range employed by those skilled in the art. Typically, the formulation will contain, on a weight percent (wt. %) basis, from about 0.01-99.99 wt. %of Form A polymorph of Compound 1 based on the total formulation, with the balance being one or more suitable pharmaceutical excipients. For example, the compound is present at a level of about 1-80 wt. %.
Combinations and Combination Therapies
Form A polymorph of Compound 1 may be used in combination with one or more other drugs in the treatment of diseases or conditions for which Form A polymorph of Compound 1 or the other drugs may have utility. Such other drug (s) may be administered, by a route and in an amount commonly used therefore, contemporaneously or sequentially with Form A polymorph of Compound 1. When Form A polymorph of Compound 1 is used contemporaneously with one or more other drugs, a pharmaceutical composition in unit dosage form containing such other drugs and Form A polymorph of Compound 1 is preferred. However, the combination therapy may also include therapies in which Form A polymorph of Compound 1 and one or more other drugs are administered on different overlapping schedules. It is also contemplated that when used in  combination with one or more other active ingredients, Form A polymorph of Compound 1 and the other active ingredients may be used in lower doses than when each is used singly.
Accordingly, the pharmaceutical compositions of the present disclosure also include those that contain one or more other drugs, in addition to Form A polymorph of Compound 1.
The above combinations include combinations of Form A polymorph of Compound 1 not only with one other drug, but also with two or more other active drugs. Likewise, Form A polymorph of Compound 1 may be used in combination with other drugs that are used in the prevention, treatment, control, amelioration, or reduction of risk of the diseases or conditions for which Form A polymorph of Compound 1 is useful. Such other drugs may be administered, by a route and in an amount commonly used therefore, contemporaneously or sequentially with Form A polymorph of Compound 1. When Form A polymorph of Compound 1 is used contemporaneously with one or more other drugs, a pharmaceutical composition containing such other drugs in addition to Form A polymorph of Compound 1 can be used. Accordingly, the pharmaceutical compositions of the present disclosure also include those that also contain one or more other active ingredients, in addition to Form A polymorph of Compound 1. The weight ratio of Form A polymorph of Compound 1 to the second active ingredient may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used.
Where the subject in need is suffering from or at risk of suffering from cancer, the subject can be treated with Form A polymorph of Compound 1 in any combination with one or more other anti-cancer agents. In some embodiments, one or more of the anti-cancer agents are proapoptotic agents. Examples of anti-cancer agents include, but are not limited to, any of the following: gossyphol, genasense, polyphenol E, Chlorofusin, all trans-retinoic acid (ATRA) , bryostatin, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) , 5-aza-2’-deoxycytidine, all trans retinoic acid, doxorubicin, vincristine, etoposide, gemcitabine, imatinib (Gleevec TM) , geldanamycin, 17-N-Allylamino-17-Demethoxygeldanamycin (17-AAG) , flavopiridol, LY294002, bortezomib, trastuzumab, BAY 11-7082, PKC412, or PD184352, Taxol TM, also referred to as “paclitaxel” , which is a well-known anti-cancer drug which acts by enhancing and stabilizing microtubule formation, and analogs of Taxol TM., such as Taxotere TM. Compounds that have the basic taxane skeleton as a common structure feature, have also been shown to have the ability to arrest cells in the G2-M phases due to stabilized microtubules and may be useful for treating cancer in combination with the compounds described herein. Suitable anti-cancer agents also include inhibitors of kinases associated cell proliferative disorder. These kinases include but not limited to Aurora-A, BTK, CDK1, CDK2, CDK3, CDK4, CDK6, CDK5, CDK7, CDK8, CDK9, ephrin receptor kinases, CHK1, CHK2, SRC, Yes, Fyn, Lck, Fer, Fes, Syk, Itk, Bmx, GSK3, JNK, MEK, PAK1, PAK2, PAK3, PAK4, PDK1, PKA, PKC, RAF, Rsk and SGK. In particular, inhibitors of CDK4/6, including abemaciclib (Verzenio) , palbociclib (Ibrance) and ribociclib (Kisqali) , have the potential to be synergistic with HIF-2αinhibitors and reverse the resistance to HIF-2αinhibition; mitogen-activated protein kinase signaling, e.g., U0126, PD98059, PD184352, PD0325901, ARRY-142886, SB239063, SP600125, BAY 43-9006, wortmannin, or LY294002; Syk inhibitors; antibodies (e.g., rituxan) ; MET inhibitor such as foretinib, carbozantinib, or crizotinib; VEGFR inhibitor such as sunitinib, sorafenib, regorafinib, lenvatinib, vandetanib, carbozantinib, axitinib; EGFR inhibitor such as afatinib, brivanib, carbozatinib, erlotinib, gefitinib, neratinib, lapatinib; PI3K inhibitor such as XL147, XL765, BKM120 (buparlisib) , GDC-0941, BYL719, IPI145, BAY80-6946. BEX235 (dactolisib) , CAL101 (idelalisib) , GSK2636771, TG100-115; MTOR inhibitor such as rapamycin (sirolimus) , temsirolimus, everolimus, XL388, XL765, AZD2013, PF04691502, PKI-587, BEZ235, GDC0349; MEK inhibitor such as AZD6244, trametinib, PD184352, pimasertinib, GDC-0973, AZD8330; CSF1R inhibitors (PLX3397, LY3022855, etc. ) and CSF1R antibodies (IMC-054, RG7155, etc) ; TGF beta receptor kinase inhibitor such as LY2157299; BTK inhibitor such as ibrutinib.
Other anti-cancer agents include proteasome inhibitor such as carfilzomib, MLN9708, delanzomib, or bortezomib; BET inhibitors such as INCB054329, OTX015, CPI-0610; LSD1 inhibitors such as GSK2979552, INCB059872; HDAC inhibitors such as panobinostat, vorinostat; DNA methyl transferase inhibitors such as azacytidine, decitabine) , and other epigenetic modulator; SHP-2 inhibitor such as TNO155; Bcl2 inhibitor ABT-199, and other Bcl-2 family protein inhibitors; HIF-2αinhibitors such as PT2977 and PT2385; Beta catenin pathway inhibitors, notch pathway inhibitors and hedgehog pathway inhibitors; Antibodies or other therapeutic proteins against VEGF include bevacizumab and aflibercept.
Other anti-cancer agents/drugs that can be used in combination with the compounds of the invention include, but are not limited to, liver X receptor (LXR) modulators, including LXR agonists and LXR beta-selective agonists; aryl hydrocarbon receptor (AhR) inhibitors.
Other anti-cancer agents that can be employed in combination with Form A polymorph of Compound 1 include Adriamycin, Dactinomycin, Bleomycin, Vinblastine, Cisplatin, acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; aminoglutethimide; amsacrine; anastrozole; anthramycin; asparaginase;  asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carboplatin; carmustine; carubicin hydrochloride; carzelesin; cedefingol; chlorambucil; cirolemycin; cladribine; crisnatol mesylate; cyclophosphamide; cytarabine; dacarbazine; daunorubicin hydrochloride; decitabine; dexormaplatin; dezaguanine; dezaguanine mesylate; diaziquone; doxorubicin; doxorubicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflornithine hydrochloride; elsamitrucin; enloplatin; enpromate; epipropidine; epirubicin hydrochloride; erbulozole; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; fadrozole hydrochloride; fazarabine; fenretinide; floxuridine; fludarabine phosphate; fluorouracil; flurocitabine; fosquidone; fostriecin sodium; gemcitabine; gemcitabine hydrochloride; hydroxyurea; idarubicin hydrochloride; ifosfamide; ilmofosine; interleukin II (including recombinant interleukin II, or Ril2) , interferon alfa-2a; interferon alfa-2b; interferon alfa-n1; interferon alfa-n3; interferon beta-1a;interferon gamma-1 b; iproplatin; irinotecan hydrochloride; lanreotide acetate; letrozole; leuprolide acetate; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine; mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate; melphalan; menogaril; mercaptopurine; methotrexate; methotrexate sodium; metoprine; meturedepa; mitindomide; mitocarcin; mitocromin; mitogillin; mitomalcin; mitomycin; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nocodazole; nogalamycin; ormaplatin; oxisuran; pegaspargase; peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; plicamycin; plomestane; porfimer sodium; porfiromycin; prednimustine; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazofurin; riboprine; rogletimide; safingol; safingol hydrochloride; semustine; simtrazene; sparfosate sodium; sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatin; streptonigrin; streptozocin; sulofenur; talisomycin; tecogalan sodium; tegafur; teloxantrone hydrochloride; temoporfin; teniposide; teroxirone; testolactone; thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; toremifene citrate; trestolone acetate; triciribine phosphate; trimetrexate; trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; uracil mustard; uredepa; vapreotide; verteporfin; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorozole; zeniplatin; zinostatin; zorubicin hydrochloride.
Other anti-cancer agents that can be employed in combination with Form A polymorph of Compound 1 include: 20-epi-1, 25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing morphogenetic protein-1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-DL-PTBA; arginine deaminase; asulacrine; atamestane; atrimustine; axinastatin 1; axinastatin 2; axinastatin 3; azasetron; azatoxin; azatyrosine; baccatin III derivatives; balanol; batimastat; BCR/ABL antagonists; benzochlorins; benzoylstaurosporine; beta lactam derivatives; beta-alethine; betaclamycin B; betulinic acid; Bfgf inhibitor; bicalutamide; bisantrene; bisaziridinylspermine; bisnafide; bistratene A; bizelesin; breflate; bropirimine; budotitane; buthionine sulfoximine; calcipotriol; calphostin C; camptothecin derivatives; canarypox IL-2; capecitabine; carboxamide-amino-triazole; carboxyamidotriazole; CaRest M3; CARN 700; cartilage derived inhibitor; carzelesin; casein kinase inhibitors (ICOS) ; castanospermine; cecropin B;cetrorelix; chlorlns; chloroquinoxaline sulfonamide; cicaprost; cis-porphyrin; cladribine; clomifene analogues; clotrimazole; collismycin A; collismycin B; combretastatin A4; combretastatin analogue; conagenin; crambescidin 816; crisnatol; cryptophycin 8; cryptophycin A derivatives; curacin A; cyclopentanthraquinones; cycloplatam; cypemycin; cytarabine ocfosfate; cytolytic factor; cytostatin; dacliximab; decitabine; dehydrodidemnin B; deslorelin; dexamethasone; dexifosfamide; dexrazoxane; dexverapamil; diaziquone; didemnin B; didox; diethylnorspermine; dihydro-5-azacytidine; 9-dioxamycin; diphenyl spiromustine; docosanol; dolasetron; doxifluridine; droloxifene; dronabinol; duocarmycin SA; ebselen; ecomustine; edelfosine; edrecolomab; eflomithine; elemene; emitefur; epirubicin; epristeride; estramustine analogue; estrogen agonists; estrogen antagonists; etanidazole; etoposide phosphate; exemestane; fadrozole; fazarabine; fenretinide; filgrastim; fmasteride; flavopiridol; flezelastine; fluasterone; fludarabine; fluorodaunorunicin hydrochloride; forfenimex; formestane; fostriecin; fotemustine; gadolinium texaphyrin; gallium nitrate; galocitabine; ganirelix; gelatinase inhibitors; gemcitabine; glutathione inhibitors; hepsulfam; heregulin; hexamethylene bisacetamide; hypericin; ibandronic acid; idarubicin; idoxifene; idramantone; ilmofosine; ilomastat; imidazoacridones; imiquimod; immunostimulant peptides; insulin-like growth factor-1 receptor inhibitor; interferon agonists; interferons; interleukins; iobenguane; iododoxorubicin; ipomeanol, iroplact; irsogladine; isobengazole; isohomohalicondrin B; itasetron; jasplakinolide; kahalalide F; lamellarin-N  triacetate; lanreotide; leinamycin; lenograstim; lentinan sulfate; leptolstatin; letrozole; leukemia inhibiting factor; leukocyte alpha interferon; leuprolide+estrogen+progesterone; leuprorelin; levamisole; liarozole; linear polyamine analogue; lipophilic disaccharide peptide; lipophilic platinum compounds; lissoclinamide 7; lobaplatin; lombricine; lometrexol; lonidamine; losoxantrone; lovastatin; loxoribine; lurtotecan; lutetium texaphyrin; lysofylline; lytic peptides; maitansine; mannostatin A; marimastat; masoprocol; maspin; matrilysin inhibitors; matrix metalloproteinase inhibitors; menogaril; merbarone; meterelin; methioninase; metoclopramide; MIF inhibitor; mifepristone; miltefosine; mirimostim; mismatched double stranded RNA; mitoguazone; mitolactol; mitomycin analogues; mitonafide; mitotoxin fibroblast growth factor-saporin; mitoxantrone; mofarotene; molgramostim; monoclonal antibody, human chorionic gonadotrophin; mopidamol; multiple drug resistance gene inhibitor; multiple tumor suppressor 1-based therapy; mustard anticancer agent; mycaperoxide B; mycobacterial cell wall extract; myriaporone; N-acetyldinaline; N-substituted benzamides; nafarelin; nagrestip; naloxone+pentazocine; napavin; naphterpin; nartograstim; nedaplatin; nemorubicin; neridronic acid; neutral endopeptidase; nilutamide; nisamycin; nitric oxide modulators; nitroxide antioxidant; nitrullyn; O6-benzylguanine; octreotide; okicenone; oligonucleotides; onapristone; ondansetron; ondansetron; oracin; oral cytokine inducer; ormaplatin; osaterone; oxaliplatin; oxaunomycin; palauamine; palmitoylrhizoxin; pamidronic acid; panaxytriol; panomifene; parabactin; pazelliptine; pegaspargase; peldesine; pentosan polysulfate sodium; pentostatin; pentrozole; perflubron; perfosfamide; perillyl alcohol; phenazinomycin; phenylacetate; phosphatase inhibitors; picibanil; pilocarpine hydrochloride; pirarubicin; piritrexim; placetin A; placetin B; plasminogen activator inhibitor; platinum complex; platinum compounds; platinum-triamine complex; porfimer sodium; porfiromycin; prednisone; propyl bis-acridone; prostaglandin J2; proteasome inhibitors; protein A-based immune modulator; protein kinase C inhibitors, microalgal; protein tyrosine phosphatase inhibitors; purine nucleoside phosphorylase inhibitors; purpurins; pyrazoloacridine; pyridoxylated hemoglobin polyoxyethylerie conjugate; raf antagonists; raltitrexed; ramosetron; ras farnesyl protein transferase inhibitors; ras inhibitors; ras-GAP inhibitor; retelliptine demethylated; rhenium Re 186 etidronate; rhizoxin; ribozymes; R. sub. 11 retinamide; rogletimide; rohitukine; romurtide; roquinimex; rubiginone B1; ruboxyl; safingol; saintopin; SarCNU; sarcophytol A; sargramostim; Sdi 1 mimetics; semustine; senescence derived 1; sense oligonucleotides; signal transduction inhibitors; signal transduction modulators; single chain antigen-binding protein; sizofuran; sobuzoxane; sodium borocaptate; sodium phenylacetate; solverol; somatomedin binding protein; sonermin; sparfosic acid; spicamycin D; spiromustine; splenopentin; spongistatin 1; squalamine;  stem cell inhibitor; stem-cell division inhibitors; stipiamide; stromelysin inhibitors; sulfinosine; superactive vasoactive intestinal peptide antagonist; suradista; suramin; swainsonine; synthetic glycosaminoglycans; tallimustine; tamoxifen methiodide; tauromustine; tazarotene; tecogalan sodium; tegafur; tellurapyrylium; telomerase inhibitors; temoporfin; temozolomide; teniposide; tetrachlorodecaoxide; tetrazomine; thaliblastine; thiocoraline; thrombopoietin; thrombopoietin mimetic; thymalfasin; thymopoietin receptor agonist; thymotrinan; thyroid stimulating hormone; tin ethyl etiopurpurin; tirapazamine; titanocene bichloride; topsentin; toremifene; totipotent stem cell factor; translation inhibitors; tretinoin; triacetyluridine; triciribine; trimetrexate; triptorelin; tropisetron; turosteride; tyrosine kinase inhibitors; tyrphostins; UBC inhibitors; ubenimex; urogenital sinus-derived growth inhibitory factor; urokinase receptor antagonists; vapreotide; variolin B; vector system, erythrocyte gene therapy; velaresol; veramine; verdins; verteporfin; vinorelbine; vinxaltine; vitaxin; vorozole; zanoterone; zeniplatin; zilascorb; and zinostatin stimalamer.
Yet other anticancer agents that can be employed in combination with Form A polymorph of Compound 1 include alkylating agents, antimetabolites, natural products, or hormones, e.g., nitrogen mustards (e.g., mechloroethamine, cyclophosphamide, chlorambucil, etc. ) , alkyl sulfonates (e.g., busulfan) , nitrosoureas (e.g., carmustine, lomusitne, etc. ) , or triazenes (decarbazine, etc. ) . Examples of antimetabolites include but are not limited to folic acid analog (e.g., methotrexate) , or pyrimidine analogs (e.g., cytarabine) , purine analogs (e.g., mercaptopurine, thioguanine, pentostatin) .
Examples of natural products useful in combination with Form A polymorph of Compound 1 include but are not limited to vinca alkaloids (e.g., vincristine) , epipodophyllotoxins (e.g., etoposide) , antibiotics (e.g., daunorubicin, doxorubicin, bleomycin) , enzymes (e.g., L-asparaginase) , or biological response modifiers (e.g., interferon alpha) .
Examples of alkylating agents that can be employed in combination with Form A polymorph of Compound 1 include, but are not limited to, nitrogen mustards (e.g., mechloroethamine, cyclophosphamide, chlorambucil, melphalan, etc. ) , ethylenimine and methylmelamines (e.g., hexamethlymelamine, thiotepa) , alkyl sulfonates (e.g., busulfan) , nitrosoureas (e.g., carmustine, lomusitne, semustine, streptozocin, etc. ) , or triazenes (decarbazine, etc. ) . Examples of antimetabolites include, but are not limited to folic acid analog (e.g., methotrexate) , or pyrimidine analogs (e.g., fluorouracil, floxuridine, cytarabine) , purine analogs (e.g., mercaptopurine, thioguanine, pentostatin.
Examples of hormones and antagonists useful in combination with Form A polymorph of Compound 1 include, but are not limited to, adrenocorticosteroids (e.g., prednisone) , progestins (e.g., hydroxyprogesterone caproate, megestrol acetate, medroxyprogesterone acetate) , estrogens (e.g., diethylstilbestrol, ethinyl estradiol) , antiestrogen (e.g., tamoxifen) , androgens (e.g., testosterone propionate, fluoxymesterone) , antiandrogen (e.g., flutamide) , gonadotropin releasing hormone analog (e.g., leuprolide) . Other agents that can be used in the methods and compositions described herein for the treatment or prevention of cancer include platinum coordination complexes (e.g., cisplatin, carboblatin) , anthracenedione (e.g., mitoxantrone) , substituted urea (e.g., hydroxyurea) , methyl hydrazine derivative (e.g., procarbazine) , adrenocortical suppressant (e.g., mitotane, aminoglutethimide) .
Other anti-cancer agents that can be employed in combination with Form A polymorph of Compound 1 include: anti-cancer agents which act by arresting cells in the G2-M phases due to stabilized microtubules and include Erbulozole (also known as R-55104) , Dolastatin 10 (also known as DLS-10 and NSC-376128) , Mivobulin isethionate (also known as CI-980) , Vincristine, NSC-639829, Discodermolide (also known as NVP-XX-A-296) , ABT-751 (Abbott, also known as E-7010) , Altorhyrtins (such as Altorhyrtin A and Altorhyrtin C) , Spongistatins (such as Spongistatin 1, Spongistatin 2, Spongistatin 3, Spongistatin 4, Spongistatin 5, Spongistatin 6, Spongistatin 7, Spongistatin 8, and Spongistatin 9) , Cemadotin hydrochloride (also known as LU-103793 and NSC-D-669356) , Epothilones (such as Epothilone A, Epothilone B, Epothilone C (also known as desoxyepothilone A or dEpoA) , Epothilone D (also referred to as KOS-862, dEpoB, and desoxyepothilone B) , Epothilone E, Epothilone F, Epothilone B N-oxide, Epothilone A N-oxide, 16-aza-epothilone B, 21-aminoepothilone B (also known as BMS-310705) , 21-hydroxyepothilone D (also known as Desoxyepothilone F and dEpoF) , 26-fluoroepothilone) , Auristatin PE (also known as NSC-654663) , Soblidotin (also known as TZT-1027) , LS-4559-P (Pharmacia, also known as LS-4577) , LS-4578 (Pharmacia, also known as LS-477-P) , LS-4477 (Pharmacia) , LS-4559 (Pharmacia) , RPR-112378 (Aventis) , Vincristine sulfate, DZ-3358 (Daiichi) , FR-182877 (Fujisawa, also known as WS-9885B) , GS-164 (Takeda) , GS-198 (Takeda) , KAR-2 (Hungarian Academy of Sciences) , BSF-223651 (BASF, also known as ILX-651 and LU-223651) , SAH-49960 (Lilly/Novartis) , SDZ-268970 (Lilly/Novartis) , AM-97 (Armad/Kyowa Hakko) , AM-132 (Armad) , AM-138 (Armad/Kyowa Hakko) , IDN-5005 (Indena) , Cryptophycin 52 (also known as LY-355703) , AC-7739 (Ajinomoto, also known as AVE-8063A and CS-39. HCl) , AC-7700 (Ajinomoto, also known as AVE-8062, AVE-8062A, CS-39-L-Ser. HCl, and RPR-258062A) , Vitilevuamide, Tubulysin A, Canadensol, Centaureidin (also known as NSC- 106969) , T-138067 (Tularik, also known as T-67, TL-138067 and TI-138067) , COBRA-1 (Parker Hughes Institute, also known as DDE-261 and WHI-261) , H10 (Kansas State University) , H16 (Kansas State University) , Oncocidin A1 (also known as BTO-956 and DIME) , DDE-313 (Parker Hughes Institute) , Fijianolide B. Laulimalide, SPA-2 (Parker Hughes Institute) , SPA-1 (Parker Hughes Institute, also known as SPIKET-P) , 3-IAABU (Cytoskeleton/Mt. Sinai School of Medicine, also known as MF-569) , Narcosine (also known as NSC-5366) , Nascapine, D-24851 (Asta Medica) , A-105972 (Abbott) , Hemiasterlin, 3-BAABU (Cytoskeleton/Mt. Sinai School of Medicine, also known as MF-191) , TMPN (Arizona State University) , Vanadocene acetylacetonate, T-138026 (Tularik) , Monsatrol, Inanocine (also known as NSC-698666) , 3-1AABE (Cytoskeleton/Mt. Sinai School of Medicine) , A-204197 (Abbott) , T-607 (Tuiarik, also known as T-900607) , RPR-115781 (Aventis) , Eleutherobins (such as Desmethyleleutherobin, Desaetyleleutherobin, Isoeleutherobin A, and Z-Eleutherobin) , Caribaeoside, Caribaeolin, Halichondrin B, D-64131 (Asta Medica) , D-68144 (Asta Medica) , Diazonamide A, A-293620 (Abbott) , NPI-2350 (Nereus) , Taccalonolide A, TUB-245 (Aventis) , A-259754 (Abbott) , Diozostatin, (-) -Phenylahistin (also known as NSCL-96F037) , D-68838 (Asta Medica) , D-68836 (Asta Medica) , Myoseverin B, D-43411 (Zentaris, also known as D-81862) , A-289099 (Abbott) , A-318315 (Abbott) , HTI-286 (also known as SPA-110, trifluoroacetate salt) (Wyeth) , D-82317 (Zentaris) , D-82318 (Zentaris) , SC-12983 (NCI) , Resverastatin phosphate sodium, BPR-OY-007 (National Health Research Institutes) , and SSR-250411 (Sanofi) .
One or more additional immune checkpoint inhibitors can be used in combination with Form A polymorph of Compound 1 for treatment of HIF-2α-associated diseases, disorders or conditions. Exemplary immune checkpoint inhibitors include inhibitors (smack molecules or biologics) against immune checkpoint molecules such as CD27, CD28, CD40, CD122, CD96, CD73, CD39, CD47, OX40, GITR, CSF1R, JAK, PI3K delta, PI3K gamma, TAM, arginase, CD137 (also known as 4-1BB) , ICOS, A2AR, A2BR, SHP-2, B7-H3, B7-H4, BTLA, CTLA-4, LAG3, TIM3, VISTA, CD96, TIGIT, PD-1, PD-L1 and PD-L2. In some embodiments, the immune checkpoint molecule is a stimulatory checkpoint molecule selected from CD27, CD28, CD40, ICOS, OX40, GITR, CD137 and STING. In some embodiments, the immune checkpoint molecule is an inhibitory checkpoint molecule selected from B7-H3, B7-H4, BTLA, CTLA-4, IDO, TDO, Arginase, KIR, LAG3, PD-1, TIM3, CD96, TIGIT and VISTA. In some embodiments, the compounds provided herein can be used in combination with one or more agents selected from KIR inhibitors, TIGIT inhibitors, LAIR1 inhibitors, CD160 inhibitors, 2B4 inhibitors and TGFR beta inhibitors.
In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of PD-1, e.g., an anti-PD-1 monoclonal antibody. In some embodiments, the anti-PD-1 monoclonal antibody is nivolumab, pembrolizumab (also known as MK-3475) , pidilizumab, SHR-1210, PDR001, or AMP-224. In some embodiments, the anti-PD-1 monoclonal antibody is nivolumab, or pembrolizumab or PDR001. In some embodiments, the anti-PD1 antibody is pembrolizumab.
In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of PD-L1, e.g., an anti-PD-L1 monoclonal antibody. In some embodiments, the anti-PD-L1 monoclonal antibody is BMS-935559, MEDI4736, MPDL3280A (also known as RG7446) , or MSB0010718C. In some embodiments, the anti-PD-L1 monoclonal antibody is MPDL3280A (atezolizumab) or MEDI4736 (durvalumab) .
In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of CTLA-4, e.g., an anti-CTLA-4 antibody. In some embodiments, the anti-CTLA-4 antibody is ipilimumab or tremelimumab. In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of LAG3, e.g., an anti-LAG3 antibody. In some embodiments, the anti-LAG3 antibody is BMS-986016 or LAG525. In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of GITR, e.g., an anti-GITR antibody. In some embodiments, the anti-GITR antibody is TRX518 or, MK-4166, INCAGN01876 or MK-1248. In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of OX40, e.g., an anti-OX40 antibody or OX40L fusion protein. In some embodiments, the anti-OX40 antibody is MEDI0562 or, INCAGN01949, GSK2831781, GSK-3174998, MOXR-0916, PF-04518600 or LAG525. In some embodiments, the OX40L fusion protein is MEDI6383.
Form A polymorph of Compound 1 can also be used to increase or enhance an immune response, including increasing the immune response to an antigen; to improve immunization, including increasing vaccine efficacy; and to increase inflammation. In some embodiments, the compounds of the invention can be sued to enhance the immune response to vaccines including, but not limited, Listeria vaccines, oncolytic viarl vaccines, and cancer vaccines such as
Figure PCTCN2021123248-appb-000014
 (granulocyte-macrophage colony-stimulating factor (GM-CF) gene-transfected tumor cell vaccine) . Anti-cancer vaccines include dendritic cells, synthetic peptides, DNA vaccines and recombinant viruses. Other immune-modulatory agents also include those that block immune cell migration such as antagonists to chemokine receptors, including CCR2 and CCR4; Sting agonists and Toll receptor agonists.
Other anti-cancer agents also include those that augment the immune system such as adjuvants or adoptive T cell transfer. Form A polymorph of Compound 1 may be effective in  combination with CAR (Chimeric antigen receptor) T cell treatment as a booster for T cell activation.
Examples
The following preparations of Compound 1 are given to enable those skilled in the art to more clearly understand and to practice the present disclosure. They should not be considered as limiting the scope of the disclosure, but merely as being illustrative and representative thereof.
Example 1
Synthesis and Polymorph characterization of diethylamine solvate of 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile
Figure PCTCN2021123248-appb-000015
To a stirred solution of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile (100.00 g, 260.91 mmol, 1.00 eq. ) and DBU (79.44 g, 521.81 mmol, 2.00 eq. ) in THF (2.2 L) was added a solution of pyridine-2-sulfonyl fluoride (50.46 g, 313.12 mmol, 1.20 eq. ) in THF (400 mL) slowly at 20-25℃ over 2 h under nitrogen atmosphere. The resulting mixture was stirred further for 16 h at 20-25℃, and then quenched with 0.78 N aqueous NaOH (1.0 L) . After stirring at 20-30 ℃ for 30 min, the layers were separated. The aqueous layer was extracted with MTBE. The combined organic layer was washed with 10%brine, and then concentrated to about a 350 mL solution. This solution was diluted with MTBE, washed with 0.5 N aqueous HCl, and then water. The organic layer was concentrated to about 200 mL (HPLC purity of 94.1 A%, and chiral purity was 97.2%ee) , diluted with EtOAc, and then loaded onto a silica gel pad and the pad was rinsed with EtOAc/n-heptane =1/5 until no product eluted. The desired fractions were concentrated and the solvent was swapped with MTBE to obtain a MTBE solution (about 350 mL) .
Diethylamine (600 mL) was added and the resulting solution was heated to 35-45℃, and n-heptane (400 mL) was added slowly at this temperature over 2 h. The resulting slurry was stirred at 35-45℃ for 1 h, and then solid crystalline seed of 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile Form A polymorph (0.5 g) was added. After further stirring at 35-45℃ for 1 h, the slurry was cooled  slowly to-5 to 5℃, and then n-heptane (2.0 L) was added slowly over 5 h. After further stirring at-5 to 5℃ for 6 h, the mixture was filtered and the solid cake was washed with cold DEA/n-heptane (1: 4) to give the title compound.
Example 2
Synthesis of crystalline Form A of 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile
Figure PCTCN2021123248-appb-000016
3-Fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile: DEA solvate was vacuum dried at 65-70℃ for 20 h to obtain 53.2 g of the title compound. The HPLC purity of the title compound was 99.8 A%and chiral purity was 100.0%ee.. ) .  1H NMR (400MHz, CDCl 3) δ=7.71-7.67 (m, 1H) , 7.29-7.26 (m, 2H) , 7.25-7.09 (m, 2H) , 6.60-5.80 (ddd, 1H) , 2.87 (s, 1H) , 2.91-2.57 (m, 2H) .
To a stirred clear solution of 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile (50.0 g) in IPA (150 mL) was added slowly n-heptane (100 mL) at 20–30℃ over 1 h, followed by seed crystal of the title compound (0.25 g of Form A polymorph of Compound 1) , and the resulting slurry was stirred at 20-30℃ for 3 h. n-Heptane (150 mL) was added slowly over4 h at 20–30℃, followed by additional n-heptane (950 mL) over 8 h. After further stirring at 20–30℃ for 3 h, the slurry was cooled slowly to-10–0℃ over4 h, and then stirred at this temperature for 6 h. The mixture was filtered and the solid cake was rinsed with cold n-heptane. The solid was vacuum dried at 45-55℃ for 12 h to obtain the title product (46.73g, 93.5%yield) . The HPLC purity was 99.9 A%and chiral purity was 100.0%ee.
Alternate Method for Preparing Compound 1 from its DEA solvate:
To a stirred mixture of Compound 1/DEA solvate (10.0 g) in EtOAc (150 mL) is added 0.5 M aqueous HCl (150 mL) at 20-30℃. The resulting mixture is stirred at this temperature for 30 min. The organic layer is separated, and then washed with water. The organic layer is concentrated and the residue is recrystallized from IPA/n-heptane to obtain the title Compound 1.
Example 3
Alternative synthesis, purification, and Polymorph Characterization of Compound 1
Figure PCTCN2021123248-appb-000017
Into a 3000 mL 3-necked round-bottom flask were added 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile (350 g, 0.762 mol, 1.0 equiv. ) and DBU (232.1 g, 1.53 mol, 2.0 equiv. ) in THF (3.5 L) at room temperature. To this solution was added dropwise a solution of pyridine-2-sulfonyl fluoride (159.7 g,0.991 mmol, 1.3 equiv. ) in THF (700 mL) at 10~15℃. The resulting mixture was stirred for 12 h at room temperature then diluted with water and EtOAc. The suspension was filtered through silica gel and the phases were separated. The aqueous layer was extracted with EtOAc and the combined organic layers were washed with water and brine, dried over anhydrous Na 2SO 4 and concentrated. The residue was purified by silica gel column chromatography (Petroleum ether/ethyl acetate=20/1 to 7/1) to give crude product (400 g) as off-white solid. The crude product was separated by SFC (column: DAICEL CHIRALPAK AS (250mm*50mm, 10um) to give 250 g of crude product. To the crude product was added heptane (1.0 L) and the mixture was stirred at 40℃ for 2 hours. The mixture was filtered to give pure title product (230 g) The HPLC purity of the tile compound was 98.7%and chiral purity was 99.1%ee. SFC condition: Column: Chiralpak AD-350×4.6mm I. D., 3μm Mobile phase: Phase A, CO 2; Phase B, EtOH (0.05%DEA) Gradient elution: B in A from 5%to 40%; Flow rate: 3mL/min Column Temp: 35 ℃; Back Pressure: 100 Bar; t R: 0.904 min.
Polymorph characterization:
Compound 1 (15.5 mg) obtained from Example 3, was added into 0.4 mL of a mixture of n-pentanol/heptane (9: 1) at 50℃. After stirring the mixture for 3 days, the solid was collected by filtration to give crystalline Form A polymorph. XRPD patterns were obtained with an X-ray diffractometer (Bruker D8 advance) , at ambient temperature (23-25℃) using an incident beam of Cu Kα
Figure PCTCN2021123248-appb-000018
from a generator operating at 40kV and40 mA. The system was equipped with LynxEye detector. Samples of Compound 5 were scanned from 3 to 40° 2θ, at a step size 0.02°2θ.  Data was analyzed using DIFFRAC plus Evaluation Package Release 2010. The XRPD spectrum of crystalline Form A polymorph is shown in Figure 3. The peak listing for XRPD spectrum, diffraction angles (2-Theta) reported in degrees was given in Table 1 below.
Table 1. Peak list of crystalline Compound 1 obtained from n-pentanol/heptane
Figure PCTCN2021123248-appb-000019
Figure PCTCN2021123248-appb-000020
Crystallization of Compound 1 under following conditions also gave Form A polymorph.
Method A
Compound 1 (15 mg/ml) was dissolved in MeOH, IPA, MTBE, EtOAc, IPAc, or toluene and the solutions were allowed to evaporate at room temperature to give Compound 1 as a crystalline solid. XRPD analysis showed the crystalline solid was Form A polymorph.
Method B
Compound 1 (~10 mg) was added into 0.5 mL of anti-solvent at 50 ℃ and then solvent was added to dissolve solid as shown in Table 3 below. The mixture was filtered and the solution was slowly cooled to room temperature and stirred at room temperature for one day. The crystals were filtered and XRPD analysis was performed.
Table 3.
Anti-solvent Solvent Crystal form
water EtOH Form A
water IPA Form A
Heptane EtOH Form A
Heptane IPA Form A
Heptane toluene Form A
Method C
Compound 1 was dissolved in solvent, and anti-solvent was added with stirring at room temperature, 50 ℃, or ice bath as shown in Table 4 below. The crystalline solid was filtered and characterized by XRPD.
Table 4.
Figure PCTCN2021123248-appb-000021
Method D
Compound 1 (~10 mg) was dissolved in solvent and then the resulting solution was added into 0.5 mL of anti-solvent. The mixture was kept stirring at room temperature for one day and the crystalline solid was analyzed by XRPD. The results were shown in Table 5 below.
Table 5.
Figure PCTCN2021123248-appb-000022
Example 4
Preparation of DEA solvate of Compound 1 and Compound 1
Polymorph characterization of DEA solvate of Compound 1:
To a stirred solution of crude 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile (25.1 g) in MTBE (75 mL) was added DEA (150 mL) at 35-45℃, followed by n-heptane (100 mL) slowly, and solid crystalline seed of 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile Form A polymorph was added. The resulting slurry was stirred at 35-45℃ for 1 h, cooled slowly to-5 to 5℃, and then stirred at this temperature for 6 h. n-Heptane (500 mL) was added slowly over 4 h at-5 to 5℃. After stirring at-5 to 5℃ for 4 h, the slurry was filtered, washed with cold solution of DEA/Heptane=1V/4V and XPRD and NMR was done on the DEA solvate of Compound 1.
XRPD patterns were obtained with an X-ray diffractometer (Bruker D8 advance) , at ambient temperature (about 23-25℃) using an incident beam of Cu Kα
Figure PCTCN2021123248-appb-000023
from a generator operating at 40kV and40 mA. The system was equipped with LynxEye detector. Samples of diethylamine solvate of 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile were scanned from 3 to 40°2θ, at a step size 0.02°2θ. Data was analyzed using DIFFRAC plus Evaluation Package Release 2010. The XRPD spectrum of crystalline Form B polymorph is shown in Figure 1. The peak listing for XRPD spectrum, diffraction angles (2-Theta) reported in degrees was given in Table 2 below.
Table 2
Figure PCTCN2021123248-appb-000024
Figure PCTCN2021123248-appb-000025
Figure PCTCN2021123248-appb-000026
Figure PCTCN2021123248-appb-000027
Compound 1:
A solution of crude 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile (26.3 g) in DCM (50 mL) was concentrated to dryness. To the residue was charged DCM (38 mL) and the resulting solution was heated to 35-40℃, followed by the addition of DEA (114 mL) . After stirring at 35-40℃ for 10 min, n-heptane (190 mL) was added slowly. The resulting mixture was stirred at 35-40℃ for 30 min, and solid crystalline seed of 3-fluoro-5- ( ( (1S, 2aR) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile Form A polymorph was added. The slurry was stirred at 35-40℃ for 3 h, n-heptane (266 mL) then was added slowly over 8 h. The slurry was cooled slowly to 0℃, and then stirred at this temperature for 5.5 h. The slurry was filtered, washed with cold solution of DEA/Heptane=1V/4V. The solid was dried at 65℃ under vacuum to obtain the title compound (14.7 g) . XRPD measurement was taken under conditions described in Example 4.
Formulation Examples
The following are representative pharmaceutical formulations containing a compound of the present disclosure.
Tablet Formulation
The following ingredients are mixed intimately and pressed into single scored tablets.
Figure PCTCN2021123248-appb-000028
Capsule Formulation
The following ingredients are mixed intimately and loaded into a hard-shell gelatin capsule.
Figure PCTCN2021123248-appb-000029
Injectable Formulation
Compound of the disclosure (e.g., a compound of Formula I) in 2%HPMC, 1%Tween 80 in DI water, q.s. to at least 20 mg/mL
Inhalation Composition
To prepare a pharmaceutical composition for inhalation delivery, 20 mg of a compound disclosed herein is mixed with 50 mg of anhydrous citric acid and 100 mL of0.9%sodium chloride solution. The mixture is incorporated into an inhalation delivery unit, such as a nebulizer, which is suitable for inhalation administration.
Topical Gel Composition
To prepare a pharmaceutical topical gel composition, 100 mg of a compound disclosed herein is mixed with 1.75 g of hydroxypropyl cellulose, 10 mL of propylene glycol, 10 mL of isopropyl myristate and 100 mL of purified alcohol USP. The resulting gel mixture is then incorporated into containers, such as tubes, which are suitable for topical administration.
Ophthalmic Solution Composition
To prepare a pharmaceutical ophthalmic solution composition, 100 mg of a compound disclosed herein is mixed with 0.9 g of NaCl in 100 mL of purified water and filtered using a 0.2 micron filter. The resulting isotonic solution is then incorporated into ophthalmic delivery units, such as eye drop containers, which are suitable for ophthalmic administration.
Nasal spray solution
To prepare a pharmaceutical nasal spray solution, 10 g of a compound disclosed herein is mixed with 30 mL of a 0.05M phosphate buffer solution (pH 4.4) . The solution is placed in a nasal administrator designed to deliver 100 ul of spray for each application.

Claims (129)

  1. A crystalline form of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile, designated as Form A polymorph, having an X-ray powder diffraction pattern comprising peaks at angular positions 15.8 and 18.6, wherein the angular positions may vary by ± 0.2°2θ as measured by X-ray powder diffraction at ambient temperature using (Cu Kα) an X-ray wavelength of
    Figure PCTCN2021123248-appb-100001
  2. The crystalline Form A polymorph of claim 1, wherein the Form A X-ray powder diffraction pattern further comprises a peak at angular position 20.1, wherein the angular positions may vary by ± 0.2°2θ.
  3. The crystalline Form A polymorph of claim 1, wherein the Form A X-ray powder diffraction pattern further comprises peaks at angular positions 12.9 and 20.1, wherein the angular positions may vary by ± 0.2°2θ.
  4. The crystalline Form A polymorph of claim 1, wherein the Form A X-ray powder diffraction pattern further comprises peaks at angular positions 11.4, 12.9, and 20.1, wherein the angular positions may vary by ± 0.2°2θ.
  5. The crystalline Form A polymorph of claim 1, wherein the Form A X-ray powder diffraction pattern further comprises peaks at angular positions 10.1, 11.4, 12.9, and 20.1, wherein the angular positions may vary by ± 0.2°2θ.
  6. A crystalline form of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile, designated as Form A polymorph, having an X-ray powder diffraction pattern comprising at least two, at least three, at least four, at least five, at least six peaks, at least seven peaks, or at least eight peaks at angular positions selected from Table 1 below, wherein the angular positions may vary by ± 0.2°2θ:
    Table 1
    Figure PCTCN2021123248-appb-100002
    Figure PCTCN2021123248-appb-100003
    Figure PCTCN2021123248-appb-100004
  7. The crystalline Form A polymorph of claim 6, wherein the at least two, at least three, at least four, at least five, or at least six peaks are selected from 10.1, 11.4, 12.9, 13.7, 15.8, 18.0, 19.6, 20.1, 21.4, 21.7, 25.0, and 26.0, wherein the angular positions may vary by ± 0.2°2θ.
  8. The crystalline Form A polymorph of any one of claims 1 to 7, wherein the angular positions of the X-ray diffraction peaks denoted therein may vary by ± 0.1°2θ.
  9. A crystalline form of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile, designated as Form A polymorph, having an X-ray powder diffraction pattern substantially identical as shown in Fig 1.
  10. The crystalline Form A polymorph of any one of claims 1 to 9, wherein the crystalline Form A polymorph is substantially pure.
  11. The crystalline Form A polymorph of any one of claims 1 to 10, wherein the crystalline Form A polymorph is substantially free of 3-fluoro-5- ( ( (1R, 2aS) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile enantiomer.
  12. An amine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile, wherein the amine is:
    (i) NHR 1R 2 where (1) R 1 is hydrogen and R 2 is C 2-C 12 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl; (2) R 1 is C 1-C 6 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl and R 2 is C 1-C 6 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl; or (3) R 1 and R 2 together with the nitrogen atom to which they are attached form cyclylamine; or
    (ii) R 3R 4N- (CH 2n-NR 5R 6 where n is an integer selected from 1 to 6 and R 3, R 4, R 5, and R 6 are independently H, C 1-C 6 alkyl, or C 3-C 7 cycloalkyl.
  13. The amine solvate of claim 12, wherein the amine is NHR 1R 2.
  14. the amine solvate of claim 12, wherein the amine is R 3R 4-N- (CH 2n-NR 5R 6.
  15. The amine solvate of claim 12 or 13, wherein R 1 is hydrogen and R 2 is C 2-C 12 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl.
  16. The amine solvate of claim 12, 13, or 15, wherein R 1 is hydrogen and R 2 is C 2-C 12 alkyl.
  17. The amine solvate of claim 12, 13, or 15, wherein R 1 is hydrogen and R 2 is C 3-C 7 cycloalkyl.
  18. The amine solvate of claim 12, 13, or 15, wherein R 1 is hydrogen and R 2 is C 3-C 7 cycloalkyl-C 1-6 alkyl.
  19. The amine solvate of claim 12 or 13, wherein R 1 is C 1-C 6 alkyl, C 2-C 6 alkenyl, or C 2-C 6 alkynyl and R 2 is C 1-C 6 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl.
  20. The amine solvate of claim 12, 13, or 19, wherein R 1 is C 1-C 6 alkyl and R 2 is C 1-C 6 alkyl.
  21. The amine solvate of claim 12, 13, or 19, wherein R 1 is C 1-C 6 alkyl and R 2 is C 3-C 7 cycloalkyl.
  22. The amine solvate of claim 12, 13, or 19, wherein R 1 is C 1-C 6 alkyl and R 2 is C 3-C 7 cycloalkyl-C 1-6 alkyl.
  23. The amine solvate of claim 12 or 13, wherein R 1 and R 2 together with the nitrogen atom to which they are attached form cyclylamine.
  24. The amine solvate of any one of claims 12, 13, and 15-18 wherein the amine is ethylamine, n-propylamine, isopropylamine, n-butylamine, sec-butylamine, tert-butylamine, n-pentylamine, sec-pentylamine, pentyl-3-amine, neopentylamine, n-hexylamine, 2-hexylamine, 3-hexylamine, isohexylamine, 1-methylpentylamine, 2-ethylbutylamine, 2-methylpentylamine, 1, 1-dimethylbutylamine, 1, 3-dimethylbutylamine, 3, 3-dimethylbutylamine, 2-methyl-3-pentylamine, 3-methylpentylamine, 3-methyl-3-pentylamine, 3-methyl-2-pentylamine, 2-methylbutylamine, 1, 2, 2-trimethylpropylamine, cyclopropylamine, cyclobutylamine, cyclopentylamine, cyclohexylamine, cyclobutylmethylamine, cyclopropylmethylamine, 2-methylcyclopropylamine, 2-cyclopropylethylamine, 2-methylcyclopentylamine, 3-methylcyclopentylamine, cyclopentylmethylamine, 2-cyclobutylethylamine, 3-cyclopropylpropylamine, 2-ethylcyclopropylamine, allyamine or propargylamine.
  25. The amine solvate of any one of claims 12, 13, 15, and 16, wherein the amine is ethylamine, n-propylamine, isopropylamine, n-butylamine, sec-butylamine, tert-butylamine, n-pentylamine, sec-pentylamine, pentyl-3-amine, neopentylamine, n-hexylamine, 2-hexylamine, 3-hexylamine, isohexylamine, 1-methylpentylamine, 2-ethylbutylamine, 2-butylamine, 2-methylpentylamine, 1, 1-dimethylbutylamine, 1, 3-dimethylbutylamine, 3, 3-dimethylbutylamine, 2-methyl-3-pentylamine, 3-methylpentylamine, 3-methyl-3-pentylamine, 3-methyl-2-pentylamine, 2-methylbutyl-1-amine, or 1, 2, 2-trimethyl-propylamine.
  26. The amine solvate of any one of claims 12, 13, and 19 to 22, wherein the amine is N-methylethylamine, N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methylisopropylamine, N-allylmethylamine, N-methylpropargylamine, N-ethyl-n-propylamine, N-ethylisopropylamine, N-methylbutylamine, N-methyl-2-butylamine, N-tert-butyl-methyl-amine, N-methyl-cyclobutylamine, N-ethylcyclopropylamine, 1-cyclopropyl-N-methylmethanamine, N-ethyl-prop-2-yn-1-amine, N-ethylallylamine, N-methylpentylamine, N-methyl-2-pentylamine, N-methyl-3-pentylamine, N, 3-dimethylbutane-2-amine, N, 2-dimethyl-butane-2-amine, N, 3-dimethylbutane-2-amine, N-methylcyclopentylamine, N-ethyl-1-butylamine, N-ethyl-2-butylamine, N-ethyl-2-methyl-2-propylamine, N-ethyl-2-methylpropylamine, N-ethylcyclobutylamine, di-n-propylamine, di-isopropylamine, N-isopropylpropylamine, diallylamine, dipropargylamine, or allylpropargylamine.
  27. The amine solvate of any one of claims 12, 13, 19, and 20, wherein the amine is N-methylethylamine, N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methyl-isopropylamine, N-ethyl-n-propylamine, N-ethyl-isopropylamine, N-methylbutylamine, N-methyl-2-butylamine, N-tert-butyl-methyl-amine, N-methylpentyl-1-amine, N-methyl-2-pentylamine, N-methyl-3-pentylamine, N, 3-dimethylbutane-2-amine, N, 2-dimethylbutane-2-amine, N, 3-dimethylbutane-2-amine, N-ethyl-1-butylamine, N-ethyl-2-butylamine, N-ethyl-2-methyl-2-propylamine, N-ethyl-2-methylpropylamine, di-n-propylamine, di-isopropylamine, or N-isopropylpropylamine.
  28. The amine solvate of any one of claims 12, 13, and 23 wherein the amine is azetidine, pyrrolidine, piperidine, piperazine, morpholine, azepane, or azocane.
  29. The amine solvate of claim 12, wherein the amine is diethylamine.
  30. The amine solvate of claim 29, wherein the stoichiometric ratio of diethylamine to 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile in the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is 1: 1.
  31. The amine solvate of any one of claims 12 to 30, wherein the amine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is a solid.
  32. The amine solvate of claim 29 or 30, wherein the amine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is a crystalline solid.
  33. The amine solvate of any one of claims 12 to 30, wherein the amine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is present in situ.
  34. The amine solvate of claim 33, wherein the amine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is present in a mixture comprising, one or more suitable organic solvents, 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile, and the amine.
  35. The amine solvate of claim 29, wherein the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is present in a mixture comprising, one or more suitable organic solvent, 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile, and diethylamine.
  36. The amine solvate of any one of claims 33 to 35, wherein the one or more suitable organic solvent is a polar organic solvent independently selected from ethers, alcohols, esters, halogenated alkanes, ketones, dimethylformamide, dimethylacetamide, acetonitrile, nitromethane, n-methyl pyrrolidinone, toluene, and xylene.
  37. The amine solvate of claim 36, wherein the one or more suitable organic solvent is independently selected from n-pentanol, methanol (MeOH) , ethanol, n-propanol, isopropanol (IPA) , n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAC) , tetrahydrofuran (THF) , 2-methyl THF (2-MeTHF) , dimethoxyethane (DME) , toluene, acetone, methyl ethyl ketone (MEK) , nitromethane, acetonitrile (ACN) , and 1, 4-dioxane.
  38. The amine solvate of claim 36, wherein the suitable organic solvent is methyl tert-butyl ether.
  39. A crystalline form of diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile, designated as Form B polymorph, having an X-ray powder diffraction pattern comprising peaks at angular positions 13.8 and 21.3, wherein the angular positions may vary by ± 0.2°2θ as measured by X-ray powder diffraction at ambient temperature using (Cu Kα) an X-ray wavelength of
    Figure PCTCN2021123248-appb-100005
    Figure PCTCN2021123248-appb-100006
  40. The crystalline Form B of claim 39, wherein the Form B X-ray powder diffraction pattern further comprises a peak at angular position 8.4, wherein the angular positions may vary by ± 0.2°2θ.
  41. The crystalline Form B of claim 39, wherein the Form B X-ray powder diffraction pattern further comprises peaks at angular positions 8.4 and 23.5, wherein the angular positions may vary by ± 0.2°2θ.
  42. The crystalline Form B of claim 39, wherein the Form B X-ray powder diffraction pattern further comprises peaks at angular positions 8.4, 23.5, and 11.7, wherein the angular positions may vary by ± 0.2°2θ.
  43. The crystalline Form B of claim 39, wherein the Form B X-ray powder diffraction pattern further comprises peaks at angular positions 8.4, 23.5, 11.7, and 9.5, wherein the angular positions may vary by ± 0.2°2θ.
  44. A crystalline Form B polymorph of diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile having an X-ray powder diffraction pattern comprising at least two, at least three, at least four, at least five, at least six, at least seven, or at least eight peaks at angular positions selected from in Table 2 below, wherein the angular positions may vary by ± 0.2°2θ.
    Table 2
    Figure PCTCN2021123248-appb-100007
    Figure PCTCN2021123248-appb-100008
    Figure PCTCN2021123248-appb-100009
    Figure PCTCN2021123248-appb-100010
  45. The crystalline Form B of claim 44, wherein the at least two, at least three, at least four, at least five, or six peaks are selected from 8.4, 9.5, 11.7, 13.8, 21.3, and23.5 wherein the angular positions may vary by ± 0.2°2θ.
  46. The crystalline Form B of any one of 39 to 45, are wherein the peaks denoted therein have margin of error of ± 0.1°2θ.
  47. A crystalline Form B of polymorph of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile having an X-ray powder diffraction pattern substantially identical as shown in Fig 2.
  48. A process of preparing an amine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile, wherein the amine is:
    (i) NHR 1R 2 where (1) R 1 is hydrogen and R 2 is C 2-C 12 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl; (2) R 1 is C 1-C 6 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl and R 2 is C 1-C 6 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl; or (3) R 1 and R 2 together with the nitrogen atom to which they are attached form cyclylamine; or
    (ii) R 3R 4N- (CH 2n-NR 5R 6 where n is an integer selected from 1 to 6 and R 3, R 4, R 5, and R 6 are independently H, C 1-C 6 alkyl or C 3-C 7 cycloalkyl, comprising:
    (a1) contacting Compound 1 with the amine NHR 1R 2 or R 3R 4-N- (CH 2n-R 5R 6 where R 1, R 2 ,R 3, R 4, R 5, and R 6, and n are as defined in (i) and (ii) , respectively, in the presence or absence of one or more suitable organic solvents;
    (b1) optionally adding (i) one or more anti-solvent and/or (ii) a solid crystalline seed of Compound 1, or a solid crystalline seed of the amine solvate of Compound 1, or a combination thereof, to the mixture of Step (a1) to precipitate the amine solvate of Compound 1;
    (c1) isolating the solid of Step (b1) to obtain a solid amine solvate of Compound 1; and
    (d1) optionally converting the amine solvate of Compound 1 from any one of Steps (a1) to (c1) to Compound 1.
  49. The process of claim48, wherein 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is contacted with the amine by adding 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile to the amine or by adding the amine to a mixture of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile in one or more suitable organic solvents in Step (a1) .
  50. The process of claim 48 or 49, wherein the amine is added to a mixture of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile in one or more suitable organic solvents in Step (a1) .
  51. The process of any one of claims 48 to 50, wherein the amine is NHR 1R 2.
  52. The process of any one of claims 48 to 50, wherein the amine is R 3R 4-N- (CH 2n-NR 5R 6.
  53. The process of any one of claims 48 to 51, wherein R 1 is hydrogen and R 2 is C 2-C 12 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl.
  54. The process of any one of claims 48 to 51 and 53, wherein R 1 is hydrogen and R 2 is C 2-C 12 alkyl.
  55. The process of any one of claims 48 to 51 and 53, wherein R 1 is hydrogen and R 2 is C 3-C 7 cycloalkyl.
  56. The process of any one of claims 48 to 51 and 53, wherein R 1 is hydrogen and R 2 is C 3-C 7 cycloalkyl-C 1-6 alkyl.
  57. The process of any one of claims 48 to 51, wherein R 1 is C 1-C 6 alkyl, C 2-C 6 alkenyl, or C 2-C 6 alkynyl and R 2 is C 1-C 6 alkyl, C 2-C 6 alkenyl, C 2-C 6 alkynyl, C 3-C 7 cycloalkyl, or C 3-C 7 cycloalkyl-C 1-6 alkyl.
  58. The process of any one of claims 48 to 51 and 57, wherein R 1 is C 1-C 6 alkyl and R 2 is C 1-C 6 alkyl.
  59. The process of any one of claims 48 to 51 and 57, wherein R 1 is C 1-C 6 alkyl and R 2 is C 3-C 7 cycloalkyl.
  60. The process of any one of claims 48 to 51 and 57, wherein R 1 is C 1-C 6 alkyl and R 2 is C 3-C 7 cycloalkyl-C 1-6 alkyl.
  61. The process of any one of claims 48 to 51, wherein R 1 and R 2 together with the nitrogen atom to which they are attached form cyclylamine.
  62. The process of any one of claims 48 to 51 and 53 to 55 wherein the amine is ethylamine, n-propylamine, isopropylamine, n-butylamine, sec-butylamine, tert-butylamine, n-pentylamine, sec-pentylamine, pentyl-3-amine, neopentylamine, n-hexylamine, 2-hexylamine, 3-hexylamine, isohexylamine, 1-methylpentylamine, 2-ethylbutylamine, 2-methylpentylamine, 1, 1-dimethylbutylamine, 1, 3-dimethylbutylamine, 3, 3-dimethylbutylamine, 2-methyl-3-pentylamine, 3-methylpentylamine, 3-methyl-3-pentylamine, 3-methyl-2-pentylamine, 2-methylbutylamine, 1, 2, 2-trimethylpropylamine, cyclopropylamine, cyclobutylamine, cyclopentylamine, cyclohexylamine, cyclobutylmethylamine, cyclopropylmethylamine, 2-methylcyclopropylamine, 2-cyclopropylethylamine, 2-methyl-cyclopentylamine, 3-methylcyclopentylamine, cyclopentylmethylamine, 2-cyclobutylethylamine, 3-cyclopropylpropyl-amine, 2-ethylcyclopropylamine, allyamine or propargylamine.
  63. The process of any one of claims 48 to 51, 53 and 54 wherein the amine is ethylamine, n-propylamine, isopropylamine, n-butylamine, sec-butylamine, tert-butylamine, n-pentylamine, sec-pentylamine, pentyl-3-amine, neopentylamine, n-hexylamine, 2-hexylamine, 3-hexylamine, isohexylamine, 1-methylpentylamine, 2-ethylbutyl-amine, 2-butylamine, 2-methylpentylamine, 1, 1-dimethylbutylamine, 1, 3-dimethylbutylamine, 3, 3-dimethylbutylamine, 2-methyl-3-pentylamine, 3-methylpentylamine, 3-methyl-3-pentylamine, 3-methyl-2-pentylamine, 2-methylbutyl-1-amine, or 1, 2, 2-trimethyl-propylamine.
  64. The process of any one of claims 48 to 51, and 57 to 59 wherein the amine is N-methylethylamine, N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methylisopropylamine, N-allylmethylamine, N-methylpropargylamine, N-ethyl-n-propylamine, N-ethylisopropylamine, N-methylbutylamine, N-methyl-2-butylamine, N-tert-butyl-methyl-amine, N-methyl-cyclobutylamine, N-ethylcyclopropylamine, 1-cyclopropyl-N-methylmethanamine, N-ethyl-prop-2-yn-1-amine, N-ethylallylamine, N-methylpentylamine, N-methyl-2-pentylamine, N-methyl-3-pentylamine, N, 3-dimethylbutane-2-amine, N, 2-dimethylbutane-2-amine, N, 3-dimethylbutane-2-amine, N-methylcyclopentylamine, N-ethyl-1-butylamine, N-ethyl-2-butylamine, N-ethyl-2-methyl-2-propylamine, N-ethyl-2-methylpropyl-amine, N-ethylcyclobutylamine, di-n-propylamine, di-isopropylamine, N-isopropylpropylamine, diallylamine, dipropargylamine, or allylpropargylamine.
  65. The process of any one of claims 48 to 51, and 57 and 58 wherein the amine is N-methylethylamine, N-methylethylamine, diethylamine, N-methyl-n-propylamine, N-methyl-isopropylamine, N-ethyl-n-propylamine, N-ethyl-isopropylamine, N-methylbutylamine, N-methyl-2-butylamine, N-tert-butyl-methyl-amine, N-methylpentyl-1-amine, N-methyl-2-pentylamine, N-methyl-3-pentylamine, N, 3-dimethylbutane-2-amine, N, 2-dimethylbutane-2-amine, N, 3-dimethylbutane-2-amine, N-ethyl-1-butylamine, N-ethyl-2-butylamine, N-ethyl-2-methyl-2-propylamine, N-ethyl-2-methylpropylamine, di-n-propylamine, di-isopropylamine, or N-isopropylpropylamine.
  66. The process of any one of claims 48 to 51, and 61 wherein the amine is azetidine, pyrrolidine, piperidine, piperazine, morpholine, azepane, or azocane.
  67. The process of any one of claims 48-50, wherein the amine is diethylamine.
  68. The process of any one of claims 48 to 67, wherein one or more anti-solvents are added to the mixture of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile of Step (a1) to obtain a solid amine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile.
  69. The process of any one of claims 48 to 67, wherein one or more anti-solvents and a solid crystalline seed of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile and/or a solid crystalline seed of the amine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile are added to the solution of Step (a1) to obtain a solid amine solid of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile.
  70. The process of claim 68 or 69 wherein the one or more anti-solvents in Step (b1) are independently selected from alkanes and water.
  71. The process of claim of claim 70, wherein the one or more anti-solvents are independently selected from the group consisting of water, n-heptane, n-hexane, isooctane, pentane, cyclohexane, and cyclopentane.
  72. The process of claim 68, 69, or 70, wherein the anti-solvent in Step (b1) is n-heptane.
  73. The process of any one of claims 49 to 72, wherein the one or more suitable organic solvents of Step (a1) are polar organic solvents independently selected from the group  consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylformamide, and dimethylacetamide.
  74. The process of any one of claims 49 to 72, wherein the one or more suitable organic solvents of Step (a1) are independently selected from the group consisting of methanol, ethanol, n-propanol, isopropanol, n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAc) , tetrahydrofuran (THF) , 2-methyl THF (2-MeTHF) , dimethoxyethane (DME) , toluene, acetone, methyl ethyl ketone (MEK) , nitromethane, acetonitrile (ACN) , and 1, 4-dioxane.
  75. The process of claims 73, wherein the suitable organic solvent of Step (a1) is MTBE.
  76. The process of any one of claims 67 to 75, wherein the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is a crystalline solid.
  77. The process of claim 76, wherein the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is crystalline Form B polymorph having an X-ray powder diffraction pattern as described in the Summary.
    78-85. The process of claim 77, wherein the crystalline Form B has an X-ray powder diffraction pattern as described in above claims 40 to 47, respectively.
  78. The process of any one of claims 48 to 85, wherein the amine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is converted to 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile.
  79. The process of any one of claims 48 to 86, wherein the amine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is converted to 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile by heating the amine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile to remove the amine from the amine solvate.
  80. The process of any one of claims 48 to 86, wherein the amine solvent of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is converted to 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile by partitioning the amine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile between one or more suitable organic solvents and an aqueous acidic solution.
  81. The process of claim 88, wherein the one or more suitable organic solvents containing 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is isolated and concentrated to provide a solid form of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile.
  82. The process of claim 87 or 89, wherein the crystalline Form A polymorph 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile has an X-ray powder diffraction pattern as described in the Summary is produced.
    91-100. The process of claim 90, wherein crystalline Form A polymorph of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile has an X-ray powder diffraction pattern as described in above claims 2 to 11, respectively, is produced.
  83. The process of any one of claims 49 to 100, further comprises preparing the 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile of Step (a1) , comprising:
    (a) treating 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) oxy) benzonitrile with a deoxyfluorinating agent in the presence of a base in a suitable organic solvent;
    (b) purifying the mixture from Step (a) to obtain 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile.
  84. the process of claim 101, wherein the deoxyfluorinating agent is pyridine-2-sulfonyl fluoride and the base is 1, 8-diazabicyclo- [5.4.0] undec-7-ene or 7-methyl-1, 5, 7-triaza-bicyclo- [4.4.0] dec-1-ene.
  85. The process of claim 101 or 102, wherein the suitable organic solvent is tetrahydrofuran.
  86. A process of preparing crystalline Form B polymorph of diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile as described in any one of Claims 39 to 47, comprising:
    (a2) contacting 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile with diethylamine, in the presence or absence of one or more suitable organic solvents;
    (b2) optionally adding (i) one or more anti-solvent and/or (ii) a crystalline seed of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile, or a solid crystalline seed of the amine solvate, or a combination thereof, to the mixture of Step (a2) to precipitate crystalline Form B polymorph of diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile; and
    (c2) isolating the crystalline Form B polymorph of the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile; and
    (d2) optionally converting the crystalline Form B polymorph of diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile from Step (c2) to 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile.
  87. The process of claim 104, wherein 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is contacted with diethylamine by adding 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile to the diethylamine or by adding the diethylamine to a mixture of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile in one or more suitable organic solvents in Step (a2) .
  88. The process of claim 104, wherein the diethylamine is added to the mixture of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile in one or more suitable organic solvents in Step (a2) .
  89. The process of any one of claims 104 to 106, wherein one or more anti-solvents are added to the mixture of Step (a2) to obtain crystalline Form B polymorph of the diethylamine  solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile.
  90. The process of any one of claims 104 to 106, wherein one or more anti-solvents and a solid crystalline seed of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile and/or a solid crystalline seed of the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile are added to the solution of Step (a2) ) to obtain crystalline Form B polymorph of the diethylamine solvate of Compound 1.
  91. The process of any one of claims 104 to 108, wherein the one or more anti-solvents in Step (b2) are independently selected from alkanes and water, preferably wherein the one or more anti-solvents are independently selected from the group consisting of water, n-heptane, n-hexane, isooctane, pentane, cyclohexane, and cyclopentane.
  92. The process of any one of claims 104 to 109, wherein the anti-solvent in Step (b2) is n-heptane.
  93. The process of any one of claims 104 to 110, wherein the solution of Compound 1 in Step (b2) is in one or more suitable organic solvents are polar organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylacetamide, and dimethylformamide.
  94. The process of claim any one of claims 104 to 110, wherein the one or more suitable organic solvents are independently selected from the group consisting of methanol, ethanol, n-propanol, isopropanol, n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAc) , etrahydrofuran (THF) , 2-methyl THF (2-MeTHF) , dimethoxyethane (DME) , toluene, acetone, methyl ethyl ketone (MEK) , nitromethane, acetonitrile (ACN) , and 1, 4-dioxane.
  95. The process of claim 112, wherein the suitable organic solvent is MTBE.
  96. The process of any one of claims 104 to 113, wherein the diethylamine of solvate 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is optionally converted to 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) - oxy) benzonitrile by heating the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile to remove the diethylamine from the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile.
  97. The process of any one of claims 104 to 113, wherein the diethylamine of solvate 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is optionally converted to 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile by partitioning the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile between one or more suitable organic solvent and an aqueous acidic solution.
  98. The process of claim 115, the acid is hydrochloric acid.
  99. The process of claim 115 or 116, wherein the one or more organic solvents containing 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is isolated and concentrated to provide a solid form of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile.
  100. The process of any one of claims 114 to 117, wherein is crystalline Form A polymorph of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile has an X-ray powder diffraction pattern as defined in any one of Claims 1 to 11, is produced.
  101. A process of preparing crystalline Form A polymorph of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrileas defined in any one of Claims 1 to 11, from diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile, comprising:
    (a3) removing of the diethylamine from the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile; and
    (b3) optionally recrystallizing crystalline Form A polymorph of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile obtained from Step (a3) .
    120 to 128. The process of claim 119, wherein the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is crystalline Form B polymorph having an X-ray powder diffraction pattern as described in above claims 39 to 47, respectively.
  102. The process of any one of claims 119 to 128, wherein the diethylamine is removed from the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile by heating the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile.
  103. The process of any one of claims 119 to 129, wherein the diethylamine is removed from the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile by partitioning the diethylamine solvate of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile between one or more suitable organic solvent and an aqueous acidic solution.
  104. The process of claim 130, wherein the acid is hydrochloric acid.
  105. The process of claim 130 or 131, wherein the one or more suitable organic solvents is isolated and concentrated to provide crystalline Form A polymorph of Compound 1 having an X-ray powder diffraction pattern as described in any one of claims 1 to 11.
  106. The process of any one of claims 119 to 129, wherein the crystalline Form A polymorph of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is recrystallized from one or more suitable organic solvents.
  107. The process of claim 133, wherein the crystalline Form A polymorph of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile is dissolved in one or more suitable organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylacetamide and dimethylformamide.
  108. The process of claim 134, wherein the one or more suitable organic solvents are selected from the group consisting methanol, ethanol, n-propanol, isopropanol, n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone  (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAc) , tetrahydrofuran (THF) , 2-methyl THF (2-MeTHF) , dimethoxyethane (DME) , toluene, acetone, methyl ethyl ketone (MEK) , nitromethane, acetonitrile (ACN) , and 1, 4-dioxane.
  109. The process of claim 134, wherein the suitable organic solvent is isopropyl acetate.
  110. The process of any one of claims 134 to 136, wherein one or more anti-solvents and/or solid crystalline seed of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile are added.
  111. The process of claim 137, wherein the one or more anti-solvents are independently selected from alkanes and water.
  112. The process of claim 138, wherein the one or more anti-solvents are independently selected from the group consisting of water, n-heptane, n-hexane, isooctane, pentane, cyclohexane, and cyclopentane.
  113. The process of claim 139, wherein the anti-solvent is n-heptane.
  114. A process of making crystalline Form A polymorph of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile, comprising:
    a4) contacting a solution of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile in one or more suitable organic solvents with one or more anti-solvents;
    b4) optionally adding a solid crystalline seed of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile; and
    c4) isolating the crystalline Form A polymorph of Compound 1 from the mixture.
  115. The process of claim 141, wherein one or more anti-solvents are added to a solution of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile in one or more suitable organic solvents independently selected from the group consisting of alcohols, ethers, toluene, ketones, esters, halogenated alkanes, nitromethane, N-methylpyrrolidinone, acetonitrile, dimethylacetamide and dimethylformamide.
  116. The process of claim 142 wherein the one or more suitable organic solvents are selected from the group consisting methanol, ethanol, n-propanol, isopropanol, n-butanol, 1, 2-propanediol, methylene dichloride, chloroform, 1, 2-dichloroethane, diethyl ether, diisopropyl ether, methyl tert-butyl ether (MTBE) , methyl tert-amyl ether, methyl acetate (MeOAc) , ethyl  acetate (EtOAc) , n-propylacetate, isopropylacetate (IPAc) , butyl acetate, n-methyl pyrrolidinone (NMP) , dimethyl formamide (DMF) , dimethylacetamide (DMAc) , tetrahydrofuran (THF) , 2-methyl THF (2-MeTHF) , dimethoxyethane (DME) , toluene, acetone, methyl ethyl ketone (MEK) , nitromethane, acetonitrile (ACN) , and 1, 4-dioxane.
  117. The process of claim 141 or 142, wherein the suitable organic solvent is isopropyl acetate.
  118. The process of claim 142, 143, or 144, wherein one or more anti-solvents is added.
  119. The process of claim 145, wherein the one or more anti-solvents is independently selected from alkanes and water.
  120. The process of claim 146, wherein the one or more anti-solvents is independently selected from the group consisting of water, n-heptane, n-hexane, isooctane, pentane, cyclohexane, and cyclopentane.
  121. The process of claim 147 wherein the anti-solvent is n-heptane.
  122. The process of any one of claims 141 to 148, wherein the process is carried out at about 0℃ to about 70℃.
  123. A pharmaceutical composition comprising crystalline Form A polymorph of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile of any one of claims 1 to 11; and a pharmaceutically acceptable excipient.
  124. A pharmaceutical composition prepared with crystalline Form A polymorph of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile of any one of claims 1 to 11; and a pharmaceutically acceptable excipient.
  125. A method of treating a disease treatable by inhibition of HIF-2αin a patient, preferably the patient is in need of such treatment, which method comprises administering to the patient, preferably a patient in need of such treatment, a therapeutically effective amount of crystalline Form A polymorph of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile of any one of claims 1 to 11 optionally in a pharmaceutical composition comprising a pharmaceutically acceptable excipient.
  126. The method of claim 152, wherein, the disease is cancer.
  127. The method of claim 153, wherein the cancer is selected from the group consisting of renal cancer, clear cell renal cell carcinoma, liver cancer, hepatocellular carcinoma (HCC) , pancreatic cancer, pancreatic neuroendocrine tumor, non-small cell lung cancer, colorectal cancer  (CRC) , pancreatic ductal adenocarcinoma (PDAC) , biliary tract cancer (BTC) , glioblastoma, neuroblastoma, pheochromocytomas and paragangliomas, somatostatinomas, hemangioblastomas, gastrointestinal stromal tumors (GIST) , pituitary tumors, leiomyomas, leiomyosarcomas, polycythaemia, and retinal tumors.
  128. The method of any one of claims 152 to 154, further comprising administering crystalline Form A polymorph of 3-fluoro-5- ( ( (1R, 2aR) -3, 3, 4, 4-tetrafluoro-1, 2a-dihydroxy-2, 2a, 3, 4-tetrahydro-1H-cyclopenta [cd] inden-7-yl) -oxy) benzonitrile with one or more other anti-cancer agents.
  129. The method of claim 155, wherein the one or more other anticancer agent is selected from the group consisting of an EGFR inhibitor, a HER2/neu inhibitor, a PI3k/mTOR inhibitor, a VEGF inhibitor, a multi-tyrosine kinase inhibitor, an immunotherapeutic agent, CAR-T therapies, and therapeutic vaccines.
PCT/CN2021/123248 2021-10-12 2021-10-12 Processes of making 3-fluoro-5- ( ( (1s, 2ar) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1h-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile and polymorphs thereof WO2023060431A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
PCT/CN2021/123248 WO2023060431A1 (en) 2021-10-12 2021-10-12 Processes of making 3-fluoro-5- ( ( (1s, 2ar) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1h-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile and polymorphs thereof
PCT/CN2021/123407 WO2023060458A1 (en) 2021-10-12 2021-10-13 Processes of making 3-fluoro-5- ( ( (1s, 2ar) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1h-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile and polymorphs thereof
PCT/US2022/046323 WO2023064305A1 (en) 2021-10-12 2022-10-11 Processes of making 3-fluoro-5-(((1s,2ar)-1,3,3,4,4-pentafluoro-2a- hydroxy-2,2a,3,4-tetrahydro-1h-cyclopenta[cd]inden-7-yl)oxy)- benzonitrile and polymorphs thereof
AU2022362137A AU2022362137A1 (en) 2021-10-12 2022-10-11 Processes of making 3-fluoro-5-(((1s,2ar)-1,3,3,4,4-pentafluoro-2a- hydroxy-2,2a,3,4-tetrahydro-1h-cyclopenta[cd]inden-7-yl)oxy)- benzonitrile and polymorphs thereof
CA3234417A CA3234417A1 (en) 2021-10-12 2022-10-11 Processes of making 3-fluoro-5-(((1s,2ar)-1,3,3,4,4-pentafluoro-2a- hydroxy-2,2a,3,4-tetrahydro-1h-cyclopenta[cd]inden-7-yl)oxy)- benzonitrile and polymorphs thereof
US17/963,926 US20230115881A1 (en) 2021-10-12 2022-10-11 PROCESSES OF MAKING 3-FLUORO-5-(((1S,2aR)-1,3,3,4,4-PENTAFLUORO-2a-HYDROXY-2,2a,3,4-TETRAHYDRO-1H-CYCLOPENTA[CD]INDEN-7-YL)OXY)-BENZONITRILE AND POLYMORPHS THEREOF
IL311995A IL311995A (en) 2021-10-12 2022-10-11 Processes of making 3-fluoro-5-(((1s,2ar)-1,3,3,4,4-pentafluoro-2a- hydroxy-2,2a,3,4-tetrahydro-1h-cyclopenta[cd]inden-7-yl)oxy)- benzonitrile and polymorphs thereof
CN202280004697.2A CN116457335A (en) 2021-10-12 2022-10-11 Process for preparing 3-fluoro-5- (((1S, 2 aR) -1,3, 4-pentafluoro-2 a-hydroxy-2, 2a,3, 4-tetrahydro-1H-cyclopenta [ cd ] inden-7-yl) oxy) benzonitrile and polymorphs thereof
TW111138511A TW202317074A (en) 2021-10-12 2022-10-11 PROCESSES OF MAKING 3-FLUORO-5-(((1S,2aR)-1,3,3,4,4-PENTAFLUORO-2a-HYDROXY-2,2a,3,4-TETRAHYDRO-1H-CYCLOPENTA[CD]INDEN-7-YL)OXY)-BENZONITRILE AND POLYMORPHS THEREOF

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2021/123248 WO2023060431A1 (en) 2021-10-12 2021-10-12 Processes of making 3-fluoro-5- ( ( (1s, 2ar) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1h-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile and polymorphs thereof

Publications (1)

Publication Number Publication Date
WO2023060431A1 true WO2023060431A1 (en) 2023-04-20

Family

ID=78401982

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2021/123248 WO2023060431A1 (en) 2021-10-12 2021-10-12 Processes of making 3-fluoro-5- ( ( (1s, 2ar) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1h-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile and polymorphs thereof
PCT/CN2021/123407 WO2023060458A1 (en) 2021-10-12 2021-10-13 Processes of making 3-fluoro-5- ( ( (1s, 2ar) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1h-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile and polymorphs thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/123407 WO2023060458A1 (en) 2021-10-12 2021-10-13 Processes of making 3-fluoro-5- ( ( (1s, 2ar) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1h-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile and polymorphs thereof

Country Status (1)

Country Link
WO (2) WO2023060431A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020214853A1 (en) * 2019-04-18 2020-10-22 Nikang Therapeutics, Inc. Tetrahydro-1h-cyclopenta[cd]indene derivatives as hypoxia inducible factor-2(alpha) inhibitors
WO2021016280A1 (en) * 2019-07-22 2021-01-28 Nikang Therapeutics, Inc. Tricyclic derivatives as hypoxia inducible factor-2(alpha) inhibitors
WO2022082329A1 (en) * 2020-10-19 2022-04-28 Nikang Therapeutics, Inc. Processes of preparing 3-fluoro-5- ( ( (1s, 2ar) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1h-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020214853A1 (en) * 2019-04-18 2020-10-22 Nikang Therapeutics, Inc. Tetrahydro-1h-cyclopenta[cd]indene derivatives as hypoxia inducible factor-2(alpha) inhibitors
WO2021016280A1 (en) * 2019-07-22 2021-01-28 Nikang Therapeutics, Inc. Tricyclic derivatives as hypoxia inducible factor-2(alpha) inhibitors
WO2022082329A1 (en) * 2020-10-19 2022-04-28 Nikang Therapeutics, Inc. Processes of preparing 3-fluoro-5- ( ( (1s, 2ar) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1h-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile

Also Published As

Publication number Publication date
WO2023060458A1 (en) 2023-04-20

Similar Documents

Publication Publication Date Title
US11753366B2 (en) Tetrahydro-1H-cyclopenta[cd]indene derivatives as hypoxia inducible factor-2(alpha) inhibitors
EP3866927A1 (en) Indane derivatives as hypoxia inducible factor-2(alpha) inhibitors
WO2021016280A1 (en) Tricyclic derivatives as hypoxia inducible factor-2(alpha) inhibitors
CA3179508A1 (en) Hypoxia inducible factor-2(alpha) inhibitors and their use in the treatment of diseases
WO2020140001A1 (en) Quinazoline derivatives as ectonucleotide pyrophosphatase phosphodiesterase 1 inhibitors
EP3952995B1 (en) Bicyclic heteroaryl derivatives as ectonucleotide pyrophosphatase phosphodiesterase 1 inhibitors
EP3849541A1 (en) 2,3-dihydrobenzo[b]thiophene derivatives as hypoxia inducible factor-2(alpha) inhibitors
TW201804998A (en) Histone methyltransferase inhibitors
WO2023060431A1 (en) Processes of making 3-fluoro-5- ( ( (1s, 2ar) -1, 3, 3, 4, 4-pentafluoro-2a-hydroxy-2, 2a, 3, 4-tetrahydro-1h-cyclopenta [cd] inden-7-yl) oxy) -benzonitrile and polymorphs thereof
US20230115881A1 (en) PROCESSES OF MAKING 3-FLUORO-5-(((1S,2aR)-1,3,3,4,4-PENTAFLUORO-2a-HYDROXY-2,2a,3,4-TETRAHYDRO-1H-CYCLOPENTA[CD]INDEN-7-YL)OXY)-BENZONITRILE AND POLYMORPHS THEREOF
WO2023070483A1 (en) PROCESSES OF MAKING 3-FLUORO-5- ( ( (1R, 2S, 2aS) -1, 2, 3, 3, 4, 4-HEXAFLUORO-2A-HYDROXY-2, 2A, 3, 4-TETRAHYDRO-1H-CYCLOPENTA [CD] INDEN-7-YL) -OXY) -BENZONITRILE AND POLYMORPHS THEREOF
US11420936B2 (en) Hypoxia inducible factor-2(alpha) inhibitors and their use in the treatment of diseases
KR20240090359A (en) 3-fluoro-5-(((1S,2aR)-1,3,3,4,4-pentafluoro-2a-hydroxy-2,2a,3,4-tetrahydro-1H-cyclopenta[ CD] Process for preparing inden-7-yl)oxy)-benzonitrile and its polymorphs
CA3235013A1 (en) Hypoxia inducible factor-2(alpha) inhibitors for the treatment of bladder cancer
WO2022197734A1 (en) Bicyclic heteroaryl boronate derivatives as ectonucleotide pyrophosphatase phosphodiesterase 1 inhibitors
WO2022086882A1 (en) Diagnostic and therapeutic methods for treatment of cancer with a hif-2(alpha) inhibitor
US20240209005A1 (en) Bicyclic heteroaryl boronate derivatives as ectonucleotide pyrophosphatase phosphodiesterase 1 inhibitors
WO2022212488A1 (en) Bicyclic heteroaryl phosphonate derivatives as ectonucleotide pyrophosphatase phosphodiesterase 1 inhibitors

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: 21960180

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE