WO2023244059A1 - Methods of treating cancer using dual androgen receptor and pde5 inhibitor compounds - Google Patents

Methods of treating cancer using dual androgen receptor and pde5 inhibitor compounds Download PDF

Info

Publication number
WO2023244059A1
WO2023244059A1 PCT/KR2023/008331 KR2023008331W WO2023244059A1 WO 2023244059 A1 WO2023244059 A1 WO 2023244059A1 KR 2023008331 W KR2023008331 W KR 2023008331W WO 2023244059 A1 WO2023244059 A1 WO 2023244059A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
compound
formula
cancer
methyl
Prior art date
Application number
PCT/KR2023/008331
Other languages
French (fr)
Inventor
Yoonsuk LEE
Kyung Sun Kim
Jeong-Ah Kim
Anna Moon
Dongkeun SONG
Juyoung JUNG
Jun-Su BAN
Soo-Jin Lee
Original Assignee
Ildong Pharmaceutical Co., Ltd.
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 Ildong Pharmaceutical Co., Ltd. filed Critical Ildong Pharmaceutical Co., Ltd.
Publication of WO2023244059A1 publication Critical patent/WO2023244059A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present disclosure provides a method of treating cancer using an androgen receptor (AR) inhibiting and phosphodiesterase 5 (PDE5) inhibiting compound, or a composition including the same.
  • AR androgen receptor
  • PDE5 phosphodiesterase 5
  • the androgen receptor (AR) is a member of the steroid-hormone family involved in the regulation of normal growth and development within a broad array of target organs. AR inhibitors and antagonists find use in various therapeutic applications. Enzalutamide and apalutamide are AR antagonist compounds that find use in treating cancer.
  • Phosphodiesterases (PDEs) encompass a large family of metallo phosphohydrolases involved in regulation of cellular cAMP and/or cyclic GMP (cGMP) levels by many stimuli. Compounds that selectively inhibit the catalytic activities of PDEs (e.g., PDE5) have been developed for the treatment of a variety of diseases.
  • PDE5 is a cGMP binding enzyme that specifically hydrolyzes cGMP to 5'-GMP. PDE5 inhibitors increase cGMP levels.
  • Cancer remains one of the most deadly threats to human health. Cancers, or malignant tumors, metastasize and grow rapidly in an uncontrolled manner, making timely detection and treatment extremely difficult.
  • Breast cancer is the most common cancer among women. Approximately 10-15% of breast cancers are triple-negative for expression of estrogen receptor (ER), progesterone receptor (PR), and hormone epidermal growth factor receptor 2 (HER2), also referred to as triple-negative breast cancer (TNBC).
  • ER estrogen receptor
  • PR progesterone receptor
  • HER2 hormone epidermal growth factor receptor 2
  • TNBC triple-negative breast cancer
  • TNBC is an aggressive type of cancer that is associated with a poor prognosis. Since the tumor cells lack the necessary receptors, common treatments like hormone therapy and drugs that target estrogen, progesterone, and HER-2 are ineffective.
  • the present disclosure provides a method of treating cancer using an androgen receptor (AR) inhibiting and phosphodiesterase 5 (PDE5) inhibiting compound, or a composition including the same.
  • AR androgen receptor
  • PDE5 phosphodiesterase 5
  • the AR- and PDE5-inhibiting compounds of this disclosure are further demonstrated to provide anticancer activity in vivo in various cancer models (e.g., as described herein). These results indicate that the AR- and PDE5-inhibiting compounds will be effective in treating cancer in a human patient.
  • the cancer is androgen positive triple-negative breast cancer (AR+ TNBC).
  • the present disclosure provides methods of treating a subject with a cancer including the step of administering to the subject an effective amount of an AR- and PDE5-inhibiting compound.
  • the AR- and PDE5-inhibiting compound is of formula (I):
  • L is a linking moiety
  • R 1 and R 2 are independently selected from -H, optionally substituted (C 1 -C 6 )alkyl, optionally substituted (C 3 -C 6 )cycloalkyl, optionally substituted (C 1 -C 6 )alkoxy, and optionally substituted (C 2 -C 4 )alkenyl;
  • each R 13 is selected from -H, optionally substituted (C 1 -C 6 )alkyl, and optionally substituted (C 1 -C 6 )alkoxy;
  • each R 14 is selected from -H, -CN, -OH, -NH 2 , -NO 2 , halogen, optionally substituted (C 1 -C 5 )alkyl, optionally substituted (C 1 -C 5 )haloalkyl, optionally substituted (C 1 -C 5 )alkoxy, optionally substituted (C 3 -C 6 )cycloalkyl, and optionally substituted (C 2 -C 4 )alkenyl;
  • X 1 is N or CR 14 ;
  • X 2 and X 3 are independently selected from N and CR 13 ;
  • Y 1 and Y 2 are independently selected from N and C, wherein one of Y 1 and Y 2 is N;
  • n 0 to 2;
  • n 1 to 4.
  • the compound of formula (I) is:
  • the compound of formula (I) is:
  • the compound of formula (I) is:
  • the compound of formula (I) is:
  • the compound of formula (I) is:
  • the subject has a cancer selected from breast cancer, gastric cancer gastrointestinal cancer, a gynecological cancer, kidney cancer, glioblastoma, high-grade-Glioma, non-small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, head and neck cancer, prostate cancer, metastatic castration-resistant prostate cancer (mCRPC), glioma, brain neoplasms and brain metastasis.
  • a cancer selected from breast cancer, gastric cancer gastrointestinal cancer, a gynecological cancer, kidney cancer, glioblastoma, high-grade-Glioma, non-small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, head and neck cancer, prostate cancer, metastatic castration-resistant prostate cancer (mCRPC), glioma, brain neoplasms and brain metastasis.
  • mCRPC metastatic castration-resistant prostate cancer
  • the subject has breast cancer.
  • the breast cancer is a triple-negative breast cancer (TNBC).
  • TNBC is a subtype selected from basal-like type 1 (BL1), basal-like type 2 (BL2), immunomodulatory (IM), mesenchymal (M), mesenchymal stem-like (MSL), and luminal androgen receptor (LAR) subtypes.
  • the TNBC is androgen receptor (AR)-positive triple-negative breast cancer (AR+ TNBC).
  • the TNBC comprises a BRCA1 mutation.
  • the subject has gastric cancer.
  • the present disclosure provides a pharmaceutical composition for use in inhibiting AR- and/or PDE5, or treating a cancer, comprising a therapeutically effective amount of an AR- and/or PDE5-inhibiting compound, or a pharmaceutically acceptable salt, a solvate, a hydrate, a prodrug, or a stereoisomer thereof.
  • the present disclosure provides a use of an AR- and/or PDE5-inhibiting compound, or a pharmaceutically acceptable salt, a solvate, a hydrate, a prodrug, or a stereoisomer thereof for inhibiting AR- and/or PDE5, or for treating or ameliorating of cancer.
  • the AR- and PDE5-inhibiting compound is of formula (I):
  • L is a linking moiety
  • each R 13 is selected from -H, optionally substituted (C 1 -C 6 )alkyl, and optionally substituted (C 1 -C 6 )alkoxy;
  • each R 14 is selected from -H, -CN, -OH, -NH 2 , -NO 2 , halogen, optionally substituted (C 1 -C 5 )alkyl, optionally substituted (C 1 -C 5 )haloalkyl, optionally substituted (C 1 -C 5 )alkoxy, optionally substituted (C 3 -C 6 )cycloalkyl, and optionally substituted (C 2 -C 4 )alkenyl;
  • X 1 is N or CR 14 ;
  • X 2 and X 3 are independently selected from N and CR 13 ;
  • n 0 to 2;
  • n 1 to 4.
  • aspects of the present disclosure include methods of treating cancer including administering to a subject having cancer a compound which exhibits dual functionality by: i) modulating the androgen receptor, and ii) modulating PDE5.
  • the inventors have demonstrated that compounds of this disclosure which have dual action as androgen receptor and PDE5 inhibiting compounds can provide potent anticancer activity in vivo in various therapeutic models of cancer (e.g., as described herein). Based on the results presented herein, the compounds of this disclosure would be effective in treating cancer, in particular AR-positive cancers associated with overexpression of PDE5.
  • AR and PDE5 inhibitor refers to compounds which exhibit the dual functionality or activity described herein.
  • the treatment goals for the methods of this disclosure include: 1) the prevention of cancer progression; 2) the relief of symptoms; 3) improvement in exercise tolerance; 3) improvement in health status; 4) the prevention and treatment of cancer exacerbations; 5) the prevention and treatment of cancer complications; 5) a reduction in mortality; and/or 6) minimization of side-effects from cancer treatment.
  • treating refers to obtaining a desired pharmacologic and/or physiologic effect, such as reduction or alleviation of one or more symptoms of the disease or disorder (i.e., cancer).
  • the effect may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease.
  • amelioration or any grammatical variation thereof (e.g., ameliorate, ameliorating, and amelioration etc.), includes, but is not limited to, delaying the onset, or reducing the severity of a disease or condition (e.g., cancer). Amelioration, as used herein, does not require the complete absence of symptoms. In some embodiments of the methods of this disclosure, administration of a AR- and PDE5-inhibiting compound to a subject having cancer at least ameliorates one or more symptoms of the cancer.
  • the AR and PDE5 inhibitors of the present disclosure are used for treating an AR positive cancer. In some embodiments, the AR- and PDE5 inhibitors of the present disclosure are used for treating a PDE5 expressing cancer. In some embodiments of the method, the subject has AR positive cancer cells and/or PDE5 expressing (e.g., PDE5 overexpressing) cancer cells.
  • the cancer is selected from breast cancer, gastric cancer gastrointestinal cancer, a gynecological cancer, kidney cancer, glioblastoma, high-grade-Glioma, non-small cell lung cancer, head and neck squamous cell carcinoma, ovarian cancer, uterine cancer, pancreatic cancer, head and neck cancer, prostate cancer, metastatic castration-resistant prostate cancer (mCRPC), glioma, brain neoplasms and brain metastasis.
  • mCRPC metastatic castration-resistant prostate cancer
  • breast cancers are ER+ (i.e., 1% or more of the cells tested express ER detectable by immunocytochemistry).
  • breast cancers contain cells that demonstrate estradiol-mediated growth.
  • patients to be treated have no detectable circulating levels of estradiol.
  • patients to be treated have circulating levels of estradiol greater than 10 pmol/L.
  • patients to be treated have circulating levels of estradiol less than 10 pmol/L.
  • a triple-negative breast cancer is AR+ (e.g., AR+ TNBC); i.e., it contains cells that express detectable androgen receptors as detected by immunohistochemistry, ligand binding, or other methods known in the art.
  • a triple negative breast cancer is AR-.
  • the breast cancer is ER+, i.e., it contains detectable levels of estrogen receptor, measured as described above, but is resistant to endocrine therapy.
  • Endocrine therapy includes administration of one or more aromatase inhibitors (e.g., anastrozole, exemestane, letrozole) and/or administration of one or more estrogen receptor modulators (e.g., tamoxifen, raloxifen, fulvestrant).
  • the breast cancer is ER+/AR+. In some embodiments the breast cancer is ER+/AR-. In some embodiments the breast cancer contains cells that are progesterone receptor positive (PR+) as detected by immunohistochemistry or ligand binding assays or any other method of detection. In some embodiments the breast cancer contains no detectable cells with progesterone receptors; e.g., the breast cancer is progesterone receptor negative (PR-). In some embodiments a breast cancer contains cells that are Her2 positive (Her2+) as detected by observable Her2 gene amplification after in situ hybridization.
  • PR+ progesterone receptor positive
  • PR- progesterone receptor negative
  • a breast cancer contains no detectable cells with amplification or expression or overexpression of Her2; e.g., the breast cancer is Her2 negative (Her2-).
  • Her2 Her2 negative
  • the progesterone receptors and Her2 can be present on the same or different populations of cells, which may be the same or different as the populations of cells expressing ER and/or AR.
  • a breast cancer is identified as AR+, ER+, and Her2+. In some embodiments a breast cancer is identified as AR+, ER+, and PR+. In some embodiments a breast cancer is identified as AR+, ER+, Her2+, and PR+. In some embodiments a breast cancer is identified as AR-, ER+, and Her2+. In some embodiments a breast cancer is identified as AR-, ER+, and PR+. In some embodiments a breast cancer is identified as AR-, ER+, Her2+, and PR+. In some embodiments, a breast cancer is identified as AR+, ER-, HER2+, PR-.
  • the AR and PDE5 inhibitors of the present disclosure are useful for treating prostate cancer, decreasing the incidence of, or halting or causing a regression of prostate cancer.
  • the AR and PDE5 inhibitors of the present disclosure are useful for treating various forms of prostate cancer.
  • the prostate cancer is associated with overexpression of AR.
  • the prostate cancer is hormone refractory prostate cancer (HRPC).
  • the prostate cancer is hormone sensitive prostate cancer (HSPC).
  • the prostate cancer is metastatic castration-resistant prostate cancer (mCRPC).
  • the AR and PDE5 inhibitors of the present disclosure are useful for treating uterine cancer.
  • the AR and PDE5 inhibitors of the present disclosure are useful for treating ovarian cancer.
  • the AR and PDE5 inhibitors of the present disclosure are useful for treating a gastric cancer. In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating a gastrointestinal cancer.
  • the AR and PDE5 inhibitors of the present disclosure are useful for treating a gynecological cancer.
  • the AR and PDE5 inhibitors of the present disclosure are useful for treating kidney cancer.
  • the AR and PDE5 inhibitors of the present disclosure are useful for treating glioma. In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating glioblastoma. In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating high-grade-glioma. In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating brain neoplasms. In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating brain metastasis.
  • the AR and PDE5 inhibitors of the present disclosure are useful for treating non-small cell lung cancer.
  • the AR and PDE5 inhibitors of the present disclosure are useful for treating head and neck cancer. In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating head and neck squamous cell carcinoma.
  • the AR and PDE5 inhibitors of the present disclosure are useful for treating pancreatic cancer.
  • one or more symptoms of the cancer of interest are reduced or alleviated in the subject after administration of the AR and PDE5 inhibitor as described herein.
  • the compounds can be administered via any convenient administration methods according to the methods of this disclosure.
  • the compound is administered orally, intravenously, subcutaneously, transdermally, intraperitoneally, or by inhalation.
  • a compound can be administered using pharmaceutical compositions comprising a therapeutically effective amount of the compound of formula (I) and a pharmaceutically acceptable carrier or diluent, in a variety of forms adapted to the chosen route of administration, for example, orally, nasally, intraperitoneally, or parenterally, by intravenous, intramuscular, topical or subcutaneous routes, or by injection into tissue.
  • the compound can also be administered intravenously or intraperitoneally by infusion or injection. Solutions of a compound can be prepared.
  • the pharmaceutical composition comprises dry powder.
  • the pharmaceutical composition is systematically administered.
  • the compound of formula (I) is administered intravenously.
  • the compound of formula (I) is administered enterically.
  • the compound of formula (I) is administered by mouth (p.o.).
  • the compound is administered by intranasal administration.
  • the compound of formula (I) is administered via an enteral feeding tube.
  • the enteral feeding tube is a nasogastric tube.
  • the effective amount of the compound of formula (I) is between 0.01 mg and 1,000 mg.
  • the dose is administered as a single daily dose.
  • the dose is administered as a plurality of equally divided sub-doses.
  • the compound of formula (I) is administered once a day, twice a day, or three times a day.
  • the compound of formula (I) is administered every two days, every three days, every four days, every five days, or every six days.
  • the compound of formula (I) is administered once a week, once every two weeks, or once every three weeks.
  • the present disclosure provides a method of treating a subject with a cancer.
  • the subject has a breast cancer. In some embodiments, the subject has a triple-negative breast cancer (TNBC). In some embodiments, the subject has a basal-like type 1 (BL1) subtype of TNBC. In some embodiments, the subject has a basal-like type 2 (BL2) subtype of TNBC. In some embodiments, the subject has an immunomodulatory (IM) subtype of TNBC. In some embodiments, the subject has a mesenchymal (M) subtype of TNBC. In some embodiments, the subject has a mesenchymal stem-like (MSL) subtype of TNBC. In some embodiments, the subject has a luminal androgen receptor (LAR) subtype of TNBC.
  • TNBC triple-negative breast cancer
  • BL1 basal-like type 1
  • BL2 basal-like type 2
  • IM immunomodulatory
  • M mesenchymal
  • MSL mesenchymal stem-like
  • LAR luminal androgen receptor
  • the subject has androgen receptor (AR)-positive triple-negative breast cancer (AR+ TNBC).
  • AR+ TNBC triple-negative breast cancer
  • the subject has a TNBC that comprises a BRCA1 mutation.
  • the subject has a tumor in a breast tissue before administration of the compound.
  • the subject has a refractory cancer.
  • the subject is not hospitalized. In another embodiment, the subject is hospitalized.
  • one or more additional active compounds are co-administered to the subject in conjunction with the AR- and PDE5- inhibiting compound.
  • “Co-administration" of compounds includes one or more compounds administered substantially simultaneously with the AR and PDE5 inhibitor of this disclosure, whether or not in the same pharmaceutical composition, or sequentially.
  • one or more of the compounds disclosed herein is administered or combined with other anticancer therapies, such as internal or external radiation, surgery, and chemotherapies.
  • Phosphodiesterase 5 is a phosphodiesterase. Inhibition of PDE5 suppresses the decomposition of cGMP, which can then lead to increased activity of PKG along with increasing the concentration of cGMP. Increasing the activity of PKG can then cause phosphorylation of numerous biologically important targets, relaxation of the smooth muscles, and increase in the flow of blood.
  • PDE5 inhibitors are a well characterized class of agent having a variety of activities.
  • a human phosphodiesterase5 (PDE5) inhibition assay in host cells can be used to assess the abilities of the compounds of the present disclosure to inhibit target PDE5.
  • the administration of the compounds of the present disclosure can cause significant changes in PDE5 activity as illustrated by Example 3.
  • the PDE5 inhibiting activity of the compounds can be assessed using in vitro enzyme assays.
  • Table 3 of Example 3 in the experimental section shows the IC 50 values for exemplary compounds in as in vitro PDE5 assay .
  • the AR and PDE5 inhibitors may inhibit the activity of AR and PDE5 in a sample, e.g., as assessed by a AR or PDE5 inhibition assay described in Examples 3 and 4.
  • AR and PDE5 inhibitors according to such methods may each have IC 50 values for AR and PDE5 inhibition, (e.g., as assessed by the assays of Example 3-4) of less than 5000 nM, such as 1000 nM or less, 200 nM or less, 100 nM or less, or 20 nM or less.
  • Biological systems may include subjects (e.g., human subjects).
  • the AR- and PDE5 inhibiting compounds include bicyclic core structures of substituted 1,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one ( ) or substituted imidazo[5,1-f][1,2,4]triazin-4(3H)-one ( ) covalently attached to various cyano-substituted aryl groups via i) a 1,3-phenylene, 2,4-pyridyl or 2,6-pyridyl, and ii) a variety of linking moieties such as substituted 2-thioxoimidazolidin-4-one ( ) groups, urea groups, or thiourea groups.
  • the AR- and PDE5-inhibiting compound can be a compound of formula (I):
  • L is a linking moiety
  • each R 14 is selected from -H, -CN, -OH, -NH 2 , -NO 2 , halogen, optionally substituted (C 1 -C 5 )alkyl, optionally substituted (C 1 -C 5 )haloalkyl, , optionally substituted (C 1 -C 5 )alkoxy, optionally substituted (C 3 -C 6 )cycloalkyl, and optionally substituted (C 2 -C 4 )alkenyl;
  • X 1 is N or CR 14 ;
  • Y 1 and Y 2 are independently selected from N and C, wherein one of Y 1 and Y 2 is N;
  • each R 13 is selected from -H, halogen and optionally substituted (C 1 -C 6 )alkoxy;
  • -A- is selected from a covalent bond, optionally substituted (C 6 -C 12 ) aryl or (C 3 -C 12 ) heteroaryl, optionally substituted-(C 3 -C 12 ) heteroaryl-(C 1 -C 5 )alkylene-, optionally substituted 3- to 6-membered heterocycle, -NHC(O)R 5 -, , and ; and
  • -B- is selected from a covalent bond, optionally substituted 3- to 6-membered heterocycle, -NHC(O)R 5 -, -O-, -S-, -NR 11 -, , , and ;
  • R 5 is selected from -OH, -(C 1 -C 5 )alkyl, -(C 1 -C 5 )haloalkyl an optionally substituted (C 1 -C 5 )alkylene;
  • R 6 and R 7 are each independently -H or optionally substituted (C 1 -C 3 )alkyl; or R 6 and R 7 together with the nitrogen atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered heterocycle;
  • Z 1 is selected from O and S;
  • At least one of -A- and -B- is not a covalent bond.
  • the compound is of formula (IIa) or (IIb):
  • R 3 is selected from -H, and optionally substituted (C 1 -C 6 )alkoxy;
  • each R 14 is independently -H or halogen. In some embodiments of formula (IIa) or (IIb), each R 14 is independently -H or -F. In some embodiments of formula (IIa) or (IIb), each R 14 is -H. In some embodiments of formula (IIa) or (IIb), each R 14 is -F. In some embodiments of formula (IIa) or (IIb), at least one R 14 is -F.
  • Z 1 is S. In some embodiments of formula (IIa) or (IIb), Z 1 is O.
  • R 6 and R 7 together with the nitrogen atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered heterocycle.
  • the compound is of formula (IIIa) or (IIIb):
  • R 8 and R 9 are independently selected from -H and optionally substituted (C 1 -C 3 )alkyl, or R 8 and R 9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered carbocycle or optionally substituted 3-to 6-membered heterocycle; and
  • Z 1 is O or S.
  • each R 14 is independently -H or halogen. In some embodiments of formula (IIIa) or (IIIb), each R 14 is independently -H or -F. In some embodiments of formula (IIIa) or (IIIb), each R 14 is -H. In some embodiments of formula (IIIa) or (IIIb), each R 14 is -F. In some embodiments of formula (IIIa) or (IIIb), at least one R 14 is -F.
  • Z 1 is S. In some embodiments of formula (IIIa) or (IIIb), Z 1 is O.
  • each R 14 is independently -H or halogen. In some embodiments of formula (IVa) or (IVb), each R 14 is independently -H or -F. In some embodiments of formula (IVa) or (IVb), each R 14 is -H. In some embodiments of formula (IVa) or (IVb), each R 14 is -F. In some embodiments of formula (IVa) or (IVb), at least one R 14 is -F.
  • Z 1 is S. In some embodiments of formula (IVa) or (IVb), Z 1 is O.
  • the compound is of formula (Va) or (Vb):
  • R 8 is -H.
  • R 9 is -H.
  • R 9 is an optionally substituted (C 1 -C 3 )alkyl. In another embodiment of formula (Va) or (Vb), R 9 is -CH 3 .
  • Z 1 is S. In some embodiments of formula (Va) or (Vb), Z 1 is O.
  • the optionally substituted 3- to 6-membered carbocycle or optionally substituted 3-to 6-membered heterocycle is selected from optionally substituted cyclobutyl, optionally substituted cyclopentyl, and optionally substituted tetrahydrofuran.
  • R 6 and R 7 are each independently -H or optionally substituted (C 1 -C 3 )alkyl. In some embodiments of formula (IIa) or (IIb), R 6 and R 7 are each -H.
  • Z 1 is S. In some embodiments of formula (VIa) or (VIb), Z 1 is O.
  • the compound is of formula (VIIa) or (VIIb):
  • -B- is wherein R 11 is -H or optionally substituted (C 1 -C 3 )alkyl.
  • the compound is of formula (VIIIa) or (VIIIb):
  • -A- is an optionally substituted 3- to 6-membered heterocycle.
  • -A- is an optionally substituted azetidine, optionally substituted pyrrolidine, optionally substituted piperidine, optionally substituted piperidin-2-one or optionally substituted pyrrolidin-2-one.
  • the -A- ring is connected to the adjacent 4-cyanophenyl or 2-cyanopyrid-5-yl ring via a N atom of the optionally substituted 3- to 6-membered heterocycle (e.g., optionally substituted azetidine, pyrrolidine, piperidine, piperidin-2-one or pyrrolidin-2-one).
  • a N atom of the optionally substituted 3- to 6-membered heterocycle e.g., optionally substituted azetidine, pyrrolidine, piperidine, piperidin-2-one or pyrrolidin-2-one.
  • r, s and t are independently is 0 or 1.
  • -A- is selected from:
  • each R 14 is independently -H or halogen. In some embodiments of formula (VIIIa) or (VIIIb), each R 14 is independently -H or -F. In some embodiments of formula (VIIIa) or (VIIIb), each R 14 is -H. In some embodiments of formula (VIIIa) or (VIIIb), each R 14 is -F. In some embodiments of formula (VIIIa) or (VIIIb), at least one R 14 is -F.
  • the compound is of formula (IXa) or (IXb):
  • -A- is an optionally substituted -(C 3 -C 12 )heteroaryl-(C 1 -C 5 )alkylene- (e.g., where the -(C 3 -C 12 )heteroaryl- and/or the -(C 1 -C 5 )alkylene- of -A- are each optionally substituted).
  • -L- is -A-B-, wherein -A- is an optionally substituted 3- to 6-membered heterocycle. In some embodiments, -A- is an optionally substituted pyrrolidin-2-one.
  • -A- is selected from
  • the compound is of formula (Xa) or (Xb):
  • R 3 is selected from -H, and optionally substituted (C 1 -C 6 )alkoxy;
  • R 12 is selected from -H and optionally substituted (C 1 -C 3 )alkyl.
  • R 12 is -H. In some embodiments of formula (Xa) or (Xb), R 12 is optionally substituted (C 1 -C 3 )alkyl. In some embodiments of formula (Xa) or (Xb), R 12 is ethyl.
  • -B- is selected from -O-, -S-, -NH-, -SO 2 -, and -NHSO 2 -.
  • -B- is -O-.
  • -B- is -S-.
  • -B- is -SO 2 -.
  • -B- is -NHSO 2 -.
  • -L- is -A-B-, wherein -A- is -NHC(O)R 5 -.
  • R 5 is .
  • the compound is of formula (XIa) or (XIb):
  • R 3 is selected from -H, and optionally substituted (C 1 -C 6 )alkoxy.
  • -B- is selected from -O-, -S-, -SO 2 - and -NHSO 2 -.
  • -B- is -O-.
  • -B- is -S-.
  • -B- is -SO 2 -.
  • -B- is -NHSO 2 -.
  • R 2 is an optionally substituted (C 1 -C 6 )alkyl. In any one of the embodiments of formula (I) to (XIb) described herein, R 2 is an optionally substituted (C 1 -C 3 )alkyl. In any one of the embodiments of formula (I) to (XIb) described herein, R 2 is n-propyl.
  • R 3 is optionally substituted (C 1 -C 3 )alkoxy. In any one of the embodiments of formula (I) to (XIb) described herein, R 3 is ethoxy.
  • X 3 is N. In another embodiment, X 1 and X 2 are each CH.
  • the compound is of formula (IVc) or (IVd):
  • X 1 is CH and R 14 is -F. In some embodiments of formula (IVc) or (IVd), X 1 is N and R 14 is -F. In some embodiments of formula (IVc) or (IVd), X 1 is CR 14 and each R 14 is -H. In some embodiments of formula (IVc) or (IVd), X 1 is N and R 14 is -H.
  • the compound is selected from:
  • R 8 and R 9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered carbocycle or optionally substituted 3- to 6-membere heterocycle (e.g., 4-membered or 5-membered carbocycle or heterocycle) that is selected from optionally substituted cyclobutyl, optionally substituted cyclopentyl, and optionally substituted tetrahydrofuran.
  • optionally substituted 3- to 6-membered carbocycle or optionally substituted 3- to 6-membere heterocycle e.g., 4-membered or 5-membered carbocycle or heterocycle
  • the compound is selected from:
  • the compound is of formula (IVc):
  • X 1 is CH or N
  • R 14 is -H or halogen
  • R 8 and R 9 are each independently H or (C 1 -C 3 )alkyl (e.g., R 8 and R 9 are each -CH 3 ), or R 8 and R 9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 5-membered carbocycle, or an optionally substituted 4-membered or 5-membered heterocycle (e.g., cyclopentane cyclobutane, cyclopentane, oxetane or tetrahydrofuran).
  • the compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoe)-2-aminoethyl
  • the compound is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoe)-2-aminoethyl
  • X 2 is N and X 3 is CH, or X 2 is CH and X 3 is N.
  • R 8 and R 9 are each optionally substituted (C 1 -C 3 )alkyl.
  • R 8 and R 9 are each independently -CH 3 .
  • R 8 and R 9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered carbocycle or optionally substituted 3- to 6-membered heterocycle (e.g., 4-membered or 5-membered carbocycle or heterocycle) that is selected from optionally substituted cyclobutyl, optionally substituted cyclopentyl, and optionally substituted tetrahydrofuran.
  • an optionally substituted 3- to 6-membered carbocycle or optionally substituted 3- to 6-membered heterocycle e.g., 4-membered or 5-membered carbocycle or heterocycle
  • the compound is of formula (Vc):
  • the compound is represented by the structure of one of the compounds in Table 1 , or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt form thereof.
  • the compounds disclosed in the present disclosure are isotopically-labeled compounds which are identical to those compounds as described herein, except that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature ("isotopologues").
  • the compounds of the present disclosure may also contain unnatural proportions of atomic isotopes at one or more atoms that constituted such compounds.
  • isotopes that can be incorporated into compounds described herein include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, such as 2 H ("D"), 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl, respectively.
  • a compound described herein can have one or more H atoms replaced with deuterium.
  • compounds described herein are intended to include compounds which differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13 C- or 14 C- enriched carbon are within the scope of the present disclosure.
  • certain isotopically-labeled compounds such as those labeled with 3 H and 14 C, can be useful in compound and/or substrate tissue distribution assays.
  • Tritiated ( 3 H) and carbon-14 ( 14 C) isotopes can be particularly preferred for their ease of preparation and detectability.
  • substitution with heavier isotopes such as deuterium can afford certain therapeutic advantages resulting from greater metabolic stability, such as increased in vivo half-life or reduced dosage requirements, and hence can be preferred in some circumstances.
  • Isotopically-labeled compounds can generally be prepared by following procedures analogous to those disclosed herein, for example, in the Examples section, by substituting an isotopically-labeled reagent for a non-isotopically-labeled reagent.
  • the compounds disclosed in the present disclosure are deuterated analogs of any of the compounds, or a salt thereof, as described herein.
  • a deuterated analog of a compound of any one of formulae (I)-(XIc) is a compound where one or more hydrogen atoms are substituted with a deuterium.
  • the deuterated analog is a compound of any one of formulae (I)-(XIc) that includes a deuterated R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , or R 14 group.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , and R 14 are independently selected from optionally substituted (C 1 -C 6 )alkyl, optionally substituted (C 1 -C 6 )alkoxy, optionally substituted (C 1 -C 6 )alkylene-heterocycloalkyl, optionally substituted monocyclic or bicyclic carbocycle, and optionally substituted monocyclic or bicyclic heterocycle including at least one deuterium atom.
  • Deuterium substituted compounds are synthesized using various methods such as described in: Dean, Dennis C.; Editor. Recent Advances in the Synthesis and Applications of Radiolabeled Compounds for Drug Discovery and Development. [In: Curr., Pharm. Des., 2000; 6(10)] 2000, 110 pp; George W.; Varma, Rajender S. The Synthesis of Radiolabeled Compounds via Organometallic Intermediates, Tetrahedron, 1989, 45(21), 6601-21; and Evans, E. Anthony. Synthesis of radiolabeled compounds, J. Radioanal. Chem., 1981, 64(1-2), 9-32.
  • the fluorinated analog is a compound of any one of formulae (I)-(XIc) that includes a fluorinated R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , or R 14 group.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , and R 14 group are independently selected from optionally substituted (C 1 -C 6 )alkyl, optionally substituted (C 1 -C 6 )alkoxy, optionally substituted (C 1 -C 6 )alkylene-heterocycloalkyl, optionally substituted monocyclic or bicyclic carbocycle, optionally substituted monocyclic or bicyclic heterocycle, optionally substituted aryl, and optionally substituted heteroaryl including at least one fluorine atom.
  • a fluorinated analog of a compound of any one of formulae (I)-(XIc) the hydrogen atom of an aliphatic or an aromatic C-H bond is replaced by a fluorine atom.
  • at least one hydrogen of an optionally substituted aryl or an optionally substituted heteroaryl is replaced by a fluorine atom.
  • a hydroxyl substituent (-OH) or an amino substituent (-NH 2 ) is replaced by a fluorine atom.
  • the compounds herein described may have asymmetric centers, geometric centers (e.g., double bond), or both. All chiral, diastereomeric, racemic forms and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomeric form is specifically indicated.
  • the compounds described herein have one or more chiral centers. It is understood that if an absolute stereochemistry is not expressly indicated, then each chiral center may independently be of the R-configuration or the S-configuration or a mixture thereof.
  • compounds described herein include enriched or resolved optical isomers at any or all asymmetric atoms as are apparent from the depictions.
  • Compounds of the present disclosure containing an asymmetrically substituted atom may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms, by synthesis from optically active starting materials, or through use of chiral auxiliaries.
  • Geometric isomers resulting from the arrangement of substituents around a carbon-carbon double bond or arrangement of substituents around a cycloalkyl or heterocyclic ring, can also exist in the compounds of the present disclosure.
  • cis and trans geometric isomers of the compounds of the present disclosure may also exist and may be isolated as a mixture of isomers or as separated isomeric forms.
  • Tautomeric forms result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton.
  • Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge.
  • Examples prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, amide - imidic acid pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, such as, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole.
  • Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
  • the compounds described herein are present in a salt form.
  • the compounds are provided in the form of pharmaceutically acceptable salts.
  • compositions that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids.
  • the acids that can be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds are those that form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions.
  • Salt forms of interest of the compounds described herein include, but not limited to, chloride, acetate, 2,2,2-trifluoroacetate (TFA), and formate salts.
  • Compounds containing an amine functional group or a nitrogen-containing heteroaryl group may be basic in nature and may react with a variety of inorganic and organic acids to form the corresponding pharmaceutically acceptable salts.
  • Inorganic acids commonly employed to form such salts include hydrochloric, and related inorganic acids.
  • Organic acids commonly employed to form such salts include formic acid, and related organic acids.
  • Such pharmaceutically acceptable salts thus include chloride, and related salts.
  • compositions that are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations.
  • examples of such salts include alkali metal or alkaline earth metal salts and, particularly, calcium, magnesium, sodium, lithium, zinc, potassium, and iron salts.
  • Compounds that include a basic or acidic moiety can also form pharmaceutically acceptable salts with various amino acids.
  • the compounds of the disclosure can contain both acidic and basic groups; for example, one amino and one carboxylic acid group.
  • the compound can exist as an acid addition salt, a zwitterion, or a base salt.
  • the compounds described herein can be present in various forms including crystalline, powder and amorphous forms of those compounds, pharmaceutically acceptable salts, including, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrates), conformational polymorphs, and amorphous forms of the compounds, as well as mixtures thereof.
  • the compounds described herein are present in a prodrug form.
  • Any convenient prodrug forms of the subject compounds can be prepared, for example, according to the strategies and methods described by Rautio et al. ("Prodrugs: design and clinical applications", Nature Reviews Drug Discovery 7, 255-270 (February 2008)).
  • Compounds of the present disclosure may be synthesized according to standard methods known in the art [see, e.g. Morrison and Boyd in "Organic Chemistry", 6 th edition, Prentice Hall (1992)]. Some compounds and/or intermediates of the present disclosure may be commercially available, known in the literature, or readily obtainable by those skilled in the art using standard procedures. Some compounds of the present disclosure may be synthesized using schemes, examples, or intermediates described herein.
  • Synthesized compounds may be validated for proper structure by methods known to those skilled in the art, for example by nuclear magnetic resonance (NMR) spectroscopy and/or mass spectrometry.
  • NMR nuclear magnetic resonance
  • the compound as described herein is represented by the structure of one of the compounds in Table 1.
  • the present disclosure is meant to encompass a compound of any one of Table 1, or a salt, a single stereoisomer, a mixture of stereoisomers and/or an isotopically labelled form thereof.
  • the compounds of the present disclosure that are administered to the subject may be composed in a pharmaceutical composition.
  • pharmaceutically acceptable excipient refers to any ingredient other than the inventive compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound, or any other convenient pharmaceutically acceptable carriers, excipients, diluent, adjuvant or additives) and having the properties of being substantially nontoxic and non-inflammatory in a patient.
  • pharmaceutically acceptable excipient includes both one and more than one such excipient, diluent, carrier, and/or adjuvant.
  • Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, dispensing, or dispersing agents, sweeteners, and waters of hydration.
  • the pharmaceutical composition comprises a compound as described herein, a pharmaceutically acceptable salt thereof, or a prodrug thereof in a therapeutically effective amount.
  • the pharmaceutical composition may be formulated according to any convenient methods, and may be prepared in various forms for oral administration such as tablets, pills, powders, capsules, syrups, emulsions and microemulsions, or in forms for non-oral administration such as eye drops or preparations for intramuscular, intravenous or subcutaneous administration.
  • the pharmaceutical composition may be administered through the eyes in the form of eyedrops.
  • the pharmaceutical composition may be an ophthalmic composition, such as an eye drop composition.
  • the pharmaceutical compositions are formulated for oral delivery.
  • additives or carriers which may be used include cellulose, calcium silicate, corn starch, lactose, sucrose, dextrose, calcium phosphate, magnesium stearate, stearic acid, stearate, talc, surfactant, suspending agent, emulsifier and diluent.
  • additives or carriers which may be used in a case wherein the pharmaceutical composition of the present disclosure is prepared as an injection include water, saline solution, glucose aqueous solution, pseudosugar solution, alcohol, glycol, ether (e.g., polyethylene glycol 400), oil, fatty acid, fatty acid ester, glyceride, surfactants, suspending agents and emulsifiers.
  • the pharmaceutical compositions are formulated for parenteral administration to a subject in need thereof. In some parenteral embodiments, the pharmaceutical compositions are formulated for intravenous administration to a subject in need thereof. In some parenteral embodiments, the pharmaceutical compositions are formulated for subcutaneous administration to a subject in need thereof.
  • C x -C y when used in conjunction with a chemical moiety, such as alkyl, alkenyl, or alkynyl is meant to include groups that contain from x to y carbons in the chain.
  • C 1 -C 6 alkyl refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups that contain from 1 to 6 carbons.
  • (C x -C y )alkylene refers to a substituted or unsubstituted alkylene chain with from x to y carbons in the alkylene chain.
  • (C x -C y )alkylene may be selected from methylene, ethylene, propylene, butylene, pentylene, and hexylene, any one of which is optionally substituted.
  • alkyl refers to an unbranched or branched saturated hydrocarbon chain.
  • alkyl as used herein has 1 to 20 carbon atoms ((C 1- C 20 )alkyl), 1 to 10 carbon atoms ((C 1- C 10 )alkyl), 1 to 8 carbon atoms ((C 1- C 8 )alkyl), 1 to 6 carbon atoms ((C 1- C 6 )alkyl), or 1 to 5 carbon atoms ((C 1- C 5 )alkyl).
  • Examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, 2-pentyl, isopentyl, neopentyl, n-hexyl, 2-hexyl, 3-hexyl, and 3-methyl pentyl.
  • alkyl residue having a specific number of carbons is named, all geometric isomers having that number of carbons may be encompassed.
  • butyl can include n-butyl, sec-butyl, isobutyl and t-butyl
  • ''propyl can include n-propyl and isopropyl.
  • an alkyl chain is optionally substituted by one or more substituents such as those substituents described herein.
  • alkylene refers to a straight divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation, and preferably having from 1 to 20 carbon atoms ((C 1- C 20 )alkylene), 1 to 10 carbon atoms ((C 1- C 10 )alkylene), 1 to 6 carbon atoms ((C 1- C 6 )alkylene), or 1 to 5 carbon atoms ((C 1- C 5 )alkylene). Examples include, but are not limited to, methylene, ethylene, propylene, butylene, and the like.
  • the alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond.
  • an alkylene chain is optionally substituted by one or more substituents such as those substituents described herein. Examples include, methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), propylene (-CH 2 CH 2 CH 2 -), 2-methylpropylene (-CH 2 -CH(CH 3 ) -CH 2 -), hexylene (-(CH 2 ) 6 -) and the like.
  • alkenyl refers to an aliphatic hydrocarbon group containing at least one carbon-carbon double bond including straight-chain, branched-chain and cyclic alkenyl groups. In some embodiments, the alkenyl group has 2-10 carbon atoms (a C 2 - 10 alkenyl). In another embodiment, the alkenyl group has 2-4 carbon atoms in the chain (a C 2 - 4 alkenyl).
  • alkenyl groups include, but are not limited to, ethenyl, propenyl, n-butenyl, i-butenyl, 3-methylbut-2-enyl, n-pentenyl, heptenyl, octenyl, cyclohexyl-butenyl and decenyl.
  • An alkylalkenyl is an alkyl group as defined herein bonded to an alkenyl group as defined herein.
  • the alkenyl group can be unsubstituted or substituted through available carbon atoms with one or more groups defined hereinabove for alkyl
  • alkynyl refers to straight or branched monovalent hydrocarbyl groups having from 2 to 6 carbon atoms and preferably 2 to 3 carbon atoms and having at least 1 and preferably from 1 to 2 sites of acetylenic (C ⁇ C-) unsaturation.
  • alkynyl groups include, but are not limited to, acetylenyl (C ⁇ CH), and propargyl (CH 2 C ⁇ CH).
  • aryl refers to a monocyclic or polycyclic group having at least one hydrocarbon aromatic ring, wherein all of the ring atoms of the at least one hydrocarbon aromatic ring is carbon.
  • Aryl may include groups with a single aromatic ring (e.g., phenyl) and multiple fused aromatic rings (e.g., naphthyl, anthryl).
  • Aryl may further include groups with one or more aromatic hydrocarbon rings fused to one or more non-aromatic hydrocarbon rings (e.g., fluorenyl; 2,3-dihydro-1H-indene; 1,2,3,4-tetrahydronaphthalene).
  • aryl includes groups with an aromatic hydrocarbon ring fused to a non-aromatic ring, wherein the non-aromatic ring comprises at least one ring heteroatom independently selected from the group consisting of N, O, and S.
  • aryl includes groups with a phenyl ring fused to a non-aromatic ring, wherein the non-aromatic ring comprises at least one ring heteroatom independently selected from the group consisting of N, O, and S (e.g., chromane; thiochromane; 2,3-dihydrobenzofuran; indoline).
  • aryl as used herein has from 6 to 14 carbon atoms ((C 6 -C 14 )aryl), or 6 to 10 carbon atoms ((C 6 -C 10 )aryl). Where the aryl includes fused rings, the aryl may connect to one or more substituents or moieties of the formulae described herein through any atom of the fused ring for which valency permits.
  • cycloalkyl refers to a monocyclic or polycyclic saturated hydrocarbon.
  • cycloalkyl has 3 to 20 carbon atoms ((C 3- C 20 )cycloalkyl), 3 to 8 carbon atoms ((C 3- C 8 )cycloalkyl), 3 to 6 carbon atoms ((C 3- C 6 )cycloalkyl), or 3 to 5 carbon atoms ((C 3- C 5 )cycloalkyl).
  • cycloalkyl has 3 to 8 carbon atoms having single or multiple cyclic rings including fused, bridged, and spiro ring systems.
  • Suitable cycloalkyl groups include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl, octahydropentalenyl, octahydro-1 H -indene, decahydronaphthalene, cubane, bicyclo[3.1.0]hexane, and bicyclo[1.1.1]pentane, and the like.
  • carbocycle refers to a saturated, unsaturated or aromatic ring in which each atom of the ring is carbon.
  • Carbocycle includes 3- to 10-membered monocyclic rings, 6- to 12-membered bicyclic rings, and 6- to 12-membered bridged rings.
  • Each ring of a bicyclic carbocycle may be selected from saturated, unsaturated, and aromatic rings.
  • an aromatic ring e.g., phenyl
  • a bicyclic carbocycle includes any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits.
  • a bicyclic carbocycle includes any combination of ring sizes such as 4-5 fused ring systems, 5-5 fused ring systems, 5-6 fused ring systems, 6-6 fused ring systems, 5-7 fused ring systems, 6-7 fused ring systems, 5-8 fused ring systems, and 6-8 fused ring systems.
  • Exemplary carbocycles include cyclopentyl, cyclohexyl, cyclohexenyl, adamantyl, phenyl, indanyl, and naphthyl.
  • heterocycle refers to a saturated, unsaturated or aromatic ring comprising one or more heteroatoms.
  • exemplary heteroatoms include N, O, Si, P, B, and S atoms.
  • Heterocycles include 3- to 10-membered monocyclic rings, 6- to 12-membered bicyclic rings, and 6- to 12-membered bridged rings.
  • a bicyclic heterocycle includes any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits.
  • an aromatic ring e.g., pyridyl
  • a saturated or unsaturated ring e.g., cyclohexane, cyclopentane, morpholine, piperidine or cyclohexene.
  • a bicyclic heterocycle includes any combination of ring sizes such as 4-5 fused ring systems, 5-5 fused ring systems, 5-6 fused ring systems, 6-6 fused ring systems, 5-7 fused ring systems, 6-7 fused ring systems, 5-8 fused ring systems, and 6-8 fused ring systems.
  • heteroaryl refers to an aromatic group of from 4 to 10 carbon atoms and 1 to 4 heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur within the ring.
  • Such heteroaryl groups can have a single ring (i.e., pyridinyl or furyl) or multiple condensed rings (i.e., indolizinyl or benzothienyl) wherein the condensed rings may or may not be aromatic and/or contain a heteroatom provided that the point of attachment is through an atom of the aromatic heteroaryl group.
  • the nitrogen and/or the sulfur ring atom(s) of the heteroaryl group are optionally oxidized to provide for the N oxide (N ⁇ O), sulfinyl, or sulfonyl moieties.
  • Preferred heteroaryls include 5 or 6 membered heteroaryls such as pyridinyl, pyrrolyl, indolyl, thiophenyl, and furanyl.
  • heteroalkyl refers to an alkyl substituent in which one or more of the carbon atoms and any attached hydrogen atoms are independently replaced with the same or different heteroatomic group. For example, 1, 2, or 3 carbon atoms may be independently replaced with the same or different heteroatomic substituent.
  • substituted refers to moieties having substituents replacing a hydrogen on one or more carbons or substitutable heteroatoms, e.g., an NH or NH2 of a compound. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound.
  • stable compounds include, but is not limited to, compounds which do not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc.
  • substituted refers to moieties having substituents replacing two hydrogen atoms on the same carbon atom, such as substituting the two hydrogen atoms on a single carbon with an oxo, imino or thioxo group.
  • substituted is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • substituents include alkyl, alkenyl, alkynyl, aryl, aralkyl, aralkenyl, aralkynyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl, and heteroarylalkyl, any of which may be optionally substituted by alkyl, alkenyl, alkynyl, halogen, haloalkyl, haloalkenyl, haloalkynyl, oxo, thioxo, cyano, nitro, imino, oximo, hydrazine, -R b OR a , -R b -OC(O)-R a , -R b -OC(O)-OR a , -R b -OC(O)-N(R a ) 2 , -R b -
  • isomers refers to two or more compounds comprising the same numbers and types of atoms, groups or components, but with different structural arrangement and connectivity of the atoms.
  • tautomer refers to one of two or more structural isomers which readily convert from one isomeric form to another and which exist in equilibrium.
  • stereoisomer refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable.
  • the present disclosure contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are non-superimposeable mirror images of one another.
  • Stereoselective syntheses a chemical or enzymatic reaction in which a single reactant forms an unequal mixture of stereoisomers during the creation of a new stereocenter or during the transformation of a pre-existing one, are well known in the art.
  • Stereoselective syntheses encompass both enantio- and diastereoselective transformations. See, for example, Carreira and Kvaerno, Classics in Stereoselective Synthesis, Wiley-VCH: Weinheim, 2009.
  • Geometric isomers resulting from the arrangement of substituents around a carbon-carbon double bond or arrangement of substituents around a cycloalkyl or heterocyclic ring, can also exist in the compounds of the present disclosure.
  • the symbol denotes a bond that may be a single, double or triple bond as described herein.
  • Substituents around a carbon-carbon double bond are designated as being in the "Z” or "E” configuration, where the terms “Z” and “E” are used in accordance with IUPAC standards. Unless otherwise specified, structures depicting double bonds encompass both the "E” and "Z” isomers.
  • the term “about” refers to a ⁇ 10% variation from the nominal value unless otherwise indicated or inferred. Where a percentage is provided with respect to an amount of a component or material in a composition, the percentage should be understood to be a percentage based on weight, unless otherwise stated or understood from the context.
  • a dash (“-") symbol that is not between two letters or symbols refers to a point of bonding or attachment for a substituent.
  • -NH 2 is attached through the nitrogen atom.
  • the terms " individual ,” “host ,” and “ subject ,” are used interchangeably, and refer to an animal, including, but not limited to, human and non-human primates, including simians and humans; rodents, including rats and mice; bovines; equines; ovines; felines; canines; and the like.
  • "Mammal” means a member or members of any mammalian species, and includes, by way of example, canines, felines, equines, bovines, ovines, rodentia, etc. and primates, i.e., non-human primates, and humans.
  • Non-human animal models, i.e. , mammals, non-human primates, murines, lagomorpha, etc. may be used for experimental investigations.
  • Patient refers to a human subject.
  • active agent drug
  • pharmacologically active agent active pharmaceutical ingredient
  • terapéuticaally effective amount refers to the amount of a compound that, when administered to a mammal or other subject for treating a disease, condition, or disorder, is sufficient to affect such treatment for the disease, condition, or disorder.
  • the “therapeutically effective amount” will vary depending on the compound, the disease and its severity and the age, weight, etc., of the subject to be treated.
  • references to or depiction of a certain element such as hydrogen or H is meant to include all isotopes of that element.
  • an R group is defined to include hydrogen or H, it also includes deuterium and tritium.
  • Compounds comprising radioisotopes such as tritium, 14 C, 32 P and 35 S are thus within the scope of the present technology. Procedures for inserting such labels into the compounds of the present technology will be readily apparent to those skilled in the art based on the disclosure herein.
  • NaHCO 3 sodium bicarbonate
  • HATU 1-[bis(dimethylamino)methylene]-1H-1,2,3-
  • LAH lithium aluminum hydride
  • EDCI 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide
  • Na 2 SO 4 sodium sulfate
  • UV ultraviolet
  • ⁇ M micromolar
  • HPLC-MS analysis was carried out with gradient elution.
  • Medium pressure liquid chromatography (MPLC) was performed with silica gel columns in both the normal phase and reverse phase.
  • Step 1
  • reaction mixture was poured into water (50 mL) and extracted with DCM (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na 2 SO 4 and concentrated under reduced pressure.
  • reaction mixture was poured into water (150 mL) and extracted with DCM (30 mLx3), the combined organic layers were washed with water (100 mL) and brine (100 mL), then dried over Na 2 SO 4 and concentrated under reduced pressure.
  • reaction mixture was poured into water (50 mL) and extracted with DCM (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na 2 SO 4 and concentrated under reduced pressure.
  • reaction mixture was poured into water (200 mL) and extracted with DCM (40 mLx3), the combined organic layers were washed with water (200 mL) and brine (200 mL), then dried over Na 2 SO 4 and concentrated under reduced pressure.
  • reaction mixture was poured into water (100 mL) and extracted with DCM (20 mLx3), the combined organic layers were washed with water (100 mL) and brine (100 mL), then dried over Na 2 SO 4 and concentrated under reduced pressure.
  • reaction mixture was poured into water (50 mL) and extracted with DCM (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na 2 SO 4 and concentrated under reduced pressure.
  • Step 2
  • reaction solution was cooled to room temperature and concentrated under reduced pressure, DMA (2 mL) and EtOH (20 mL) was added to the residue.
  • the mixture was heated to 70 °C and hydrogen chloride (2 M, 4 mL) was added, then stirred at this temperature for 2 h.
  • the resulting mixture was poured into water (200 mL) and extracted with EA (50mLx3), washed by brine (200 mL), dried over Na 2 SO 4 and concentrated.
  • reaction mixture was poured into water (80 mL) and extracted with DCM (20 mLx3), the combined organic layers were washed with water (100 mL) and brine (100 mL), then dried over Na 2 SO 4 and concentrated under reduced pressure.
  • reaction mixture was poured into water (80 mL) and extracted with DCM (20 mLx3), the combined organic layers were washed with water (80 mL) and brine (80 mL), then dried over Na 2 SO 4 and concentrated under reduced pressure.
  • reaction mixture was poured into water (80 mL) and extracted with DCM (20 mLx3), the combined organic layers were washed with water (80 mL) and brine (80 mL), then dried over Na 2 SO 4 and concentrated under reduced pressure.
  • This example illustrates the in vitro inhibition of human PDE5A1 by exemplary compounds of this disclosure (e.g., as described herein).
  • Sildenafil citrate (Catalog no. LKT-S3313, Axxora, San Diego, CA), Vardenafil hydrochloride trihydrate (Catalog no. SML2103, Sigma-Aldrich, St. Louis, MO), PDE Assay Buffer (Catalog no. 60393, BPS bioscience, San Diego, CA), PDE Binding Agent (Catalog no. 60390, BPS bioscience, San Diego, CA) and PDE Binding Agent Diluent (cGMP, Catalog no. 60392, BPS bioscience, San Diego, CA) were used for assays. Test compounds were supplied by Ildong Pharmaceuticals Co., Ltd.
  • Fluorescence intensity was measured at excitation of 485 nm and an emission of 528 nm using a Tecan Infinite M1000 microplate reader.
  • PDE activity assays were performed in duplicate at each concentration. Fluorescence intensity is converted to fluorescence polarization using the Tecan Magellan6 software. The fluorescence polarization (FP t ) in absence of the compound in each data set was defined as 100% activity. In the absence of PDE and the compound, the value fluorescent polarization (FP b ) in each data set was defined as 0% activity. The percent activity in the presence of compound was calculated according to Equation 1 :
  • Test compounds were received at 1000X (or greater) of the desired starting concentration in 100% DMSO. If compounds were supplied at greater that 1000X concentration, an initial dilution is made in 100% DMSO to bring the compounds to 1000X concentration. The 1000X test compounds were serially diluted (10 point
  • the Substrate Loading Solution consists of three Life Technologies reagents: Solution A (10 mM LiveBLAzerTM-FRET B/G Substrate), Solution B and Solution C.
  • AR-UAS-bla GripTiteTM 293 cells were thawed and resuspended in Assay Media (DMEM phenol red free, 2% CD-treated FBS, 0.1 mM NEAA, 1 mM Sodium Pyruvate, 100 U/mL/100 ⁇ g/mL Pen/Strep) to a concentration of 312,500 cells/mL.4 ⁇ L of a 10X serial dilution of Cyproterone Acetate (control antagonist starting concentration, 3,160 nM) or compounds are added to appropriate wells of a Poly-D-Lysine assay plate.
  • Assay Media DMEM phenol red free, 2% CD-treated FBS, 0.1 mM NEAA, 1 mM Sodium Pyruvate, 100 U/mL/100 ⁇ g/mL Pen/Strep
  • nM In Vitro Androgen Receptor (AR) Reporter Assay Cmpd No. Concentration Range (nM) A: IC 50 ⁇ 500 nM B: 500 nM ⁇ IC 50 ⁇ 1000 nM C: IC 50 > 1000 nM Cmpd No. Concentration Range(nM) A: IC 50 ⁇ 500 nM B: 500 nM ⁇ IC 50 ⁇ 1000 nM C: IC 50 > 1000 nM IC 50 IC 50 13 B 18 A 19 B 20 A 21 A 22 A 23 A 25 C 26 C 27 C 28 C 29 B 30 A 31 B 32 C 33 C 34 A 38 A 47 A 48 A 49 B 50 A
  • This example also illustrates the in vitro antagonistic activity toward androgen receptor (AR) exhibited by exemplary compounds of this disclosure (e.g., as described herein), and illustrates the binding affinity of the exemplary compounds.
  • AR androgen receptor
  • IC 50 values were determined by a non-linear, least squares regression analysis using MathIQTM (ID Business Solutions Ltd., UK).
  • IC 50 (nM) concentration ranges (A) refers to IC 50 ⁇ 50 nM; (B) refers to 50 nM ⁇ IC 50 ⁇ 200; and (C) refers to IC 50 > 200 nM.
  • exemplary compounds of this disclosure exhibit potent AR inhibitory activity and binding affinity.
  • Example 6 Antitumor effect of subject compounds in patient-derived xenograft ( PDX ) mouse model of triple negative breast cancer ( TNBC )
  • This example assesses the antitumor efficacy of the subject compounds in a TNB CTG-0670, with DHT tumor model.
  • test compounds 18 , 20 and 22 Three test compounds (subject compounds 18 , 20 and 22 ) and two positive controls (enzalutamide) were evaluated.
  • Polyethylene glycol (15)-hydroxystearate (Solutol, 30% in deionized water) and PEG-400 (20% in deionized water) were used as excipients to prepare test compounds 18 , 20 and 22 , and positive controls enzalutamide.
  • mice Female athymic nude mice were divided into 9 groups for dosing orally (p.o.) once daily over a 16 day period as set out in Table 6 below:
  • TNB CTG -0670 + DHT tumor model Study Design Study Groups Compound Dose Level (mg/kg/day) 1 Vehicle control 0 2 Vehicle control + DHT 0 3 Enzalutamide + DHT 25 4 Enzalutamide + DHT 50 5 Compound 18 + DHT 25 6 Compound 20 + DHT 25 7 Compound 20 + DHT 50 8 Compound 22 + DHT 25 9 Compound 22 + DHT 50
  • the mean tumor volume (MTV) was measured on day 0 and day 16 of treatment.
  • the mean tumor volume for each group and the maximum tumor growth inhibition (TGI) relative to the control group is set out in Table 7 below.
  • TGI was calculated using the formula below:
  • TGI [1-(final MTV - initial MTV of a treated group) / (final MTV - initial MTV of the control group)] x 100%.
  • TNB CTG -0670 + DHT tumor model Results Study Groups Compound Dose Level (mg/kg/day) Body Weight (g) Mean tumor volume (mm 3 ) Day 0 Mean tumor volume (mm 3 ) Day 16 TGI (%) 1 Vehicle control 0 24.1 190 1089 19 2 Vehicle control + DHT 0 25.9 196 1297 0 3 Enzalutamide + DHT 25 24.7 196 1243 5 4 Enzalutamide + DHT 50 25.1 196 1379 -7 5 Compound 18 + DHT 25 24.8 196 1119 17 6 Compound 20 + DHT 25 24.5 196 1015 26 7 Compound 20 + DHT 50 25.0 196 1135 15 8 Compound 22 + DHT 25 25.5 196 1139 15 9 Compound 22 + DHT 50 25.6 196 855 41
  • TNBC triple-negative breast cancer
  • Test compound is administered to ICR mice via IV (5 mg/kg and 5mL/kg) or PO (5 mg/kg and 5mL/kg).
  • ICR mice are administered via each route with the test compound dissolved in appropriate vehicles. Plasma samples are collected designated time and the samples are then stored at -70°C until analysis using LC-MS/MS system.
  • Plasma Protein precipitation is conducted on 20 ⁇ L of plasma samples with 180 ⁇ L of acetonitrile containing internal standard. After mixing and centrifugation, the supernatant(150 ⁇ L) is analyzed by LC-MS/MS system with MRM mode.
  • Brain Protein precipitation is conducted on 50 ⁇ L of brain samples with 200 ⁇ L of acetonitrile containing internal standard. After mixing and centrifugation, supernatant(150 ⁇ L) is analyzed by LC-MS/MS system with MRM mode.
  • LC/MS/MS data are analyzed by Analyst 1.7.2.
  • PK parameters are calculated by noncompartmental analysis using WinNonlin software.

Abstract

The present disclosure provides a method of treating a subject with cancer by using an androgen receptor (AR) inhibitor and phosphodiesterase 5 (PDE5) inhibitor compound, and compositions comprising the same. The compounds of this disclosure which have dual functionality for inhibiting or antagonizing the androgen receptor and for inhibiting PDE5 can provide activity in various models (e.g., as described herein) of cancer. The androgen receptor (AR) inhibitor and phosphodiesterase 5 (PDE5) inhibitor compounds can inhibit tumor growth in a triple-negative breast cancer model.

Description

METHODS OF TREATING CANCER USING DUAL ANDROGEN RECEPTOR AND PDE5 INHIBITOR COMPOUNDS
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application No. 63/352,583, filed June 15, 2022, which is hereby incorporated in its entirety by reference. The present disclosure provides a method of treating cancer using an androgen receptor (AR) inhibiting and phosphodiesterase 5 (PDE5) inhibiting compound, or a composition including the same.
BACKGROUND
[0001] The androgen receptor (AR) is a member of the steroid-hormone family involved in the regulation of normal growth and development within a broad array of target organs. AR inhibitors and antagonists find use in various therapeutic applications. Enzalutamide and apalutamide are AR antagonist compounds that find use in treating cancer. Phosphodiesterases (PDEs) encompass a large family of metallo phosphohydrolases involved in regulation of cellular cAMP and/or cyclic GMP (cGMP) levels by many stimuli. Compounds that selectively inhibit the catalytic activities of PDEs (e.g., PDE5) have been developed for the treatment of a variety of diseases. PDE5 is a cGMP binding enzyme that specifically hydrolyzes cGMP to 5'-GMP. PDE5 inhibitors increase cGMP levels.
[0002] Cancer remains one of the most deadly threats to human health. Cancers, or malignant tumors, metastasize and grow rapidly in an uncontrolled manner, making timely detection and treatment extremely difficult. Breast cancer is the most common cancer among women. Approximately 10-15% of breast cancers are triple-negative for expression of estrogen receptor (ER), progesterone receptor (PR), and hormone epidermal growth factor receptor 2 (HER2), also referred to as triple-negative breast cancer (TNBC). TNBC is an aggressive type of cancer that is associated with a poor prognosis. Since the tumor cells lack the necessary receptors, common treatments like hormone therapy and drugs that target estrogen, progesterone, and HER-2 are ineffective.
[0003] Accordingly, there is a need for treatment methods for cancers such as TNBC.
1. SUMMARY
[0004] The present disclosure provides a method of treating cancer using an androgen receptor (AR) inhibiting and phosphodiesterase 5 (PDE5) inhibiting compound, or a composition including the same. The inventors demonstrated that compounds of this disclosure have dual functionality and activity as AR inhibitors or antagonists, and PDE5 inhibitors, and are useful in treating therapeutic indications related to cancer.
[0005] The AR- and PDE5-inhibiting compounds of this disclosure are further demonstrated to provide anticancer activity in vivo in various cancer models (e.g., as described herein). These results indicate that the AR- and PDE5-inhibiting compounds will be effective in treating cancer in a human patient. In some embodiments, the cancer is androgen positive triple-negative breast cancer (AR+ TNBC).
[0006] Accordingly, the present disclosure provides methods of treating a subject with a cancer including the step of administering to the subject an effective amount of an AR- and PDE5-inhibiting compound. In some embodiments, the AR- and PDE5-inhibiting compound is of formula (I):
Figure PCTKR2023008331-appb-img-000001
(I)
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt thereof, wherein:
L is a linking moiety;
R1 and R2 are independently selected from -H, optionally substituted (C1-C6)alkyl, optionally substituted (C3-C6)cycloalkyl, optionally substituted (C1-C6)alkoxy, and optionally substituted (C2-C4)alkenyl;
each R13 is selected from -H, optionally substituted (C1-C6)alkyl, and optionally substituted (C1-C6)alkoxy;
each R14 is selected from -H, -CN, -OH, -NH2, -NO2, halogen, optionally substituted (C1-C5)alkyl, optionally substituted (C1-C5)haloalkyl, optionally substituted (C1-C5)alkoxy, optionally substituted (C3-C6)cycloalkyl, and optionally substituted (C2-C4)alkenyl;
X1 is N or CR14;
X2 and X3 are independently selected from N and CR13;
Y1 and Y2 are independently selected from N and C, wherein one of Y1 and Y2 is N;
m is 0 to 2; and
n is 1 to 4.
[0007] In some embodiments of the method, the compound of formula (I) is:
Figure PCTKR2023008331-appb-img-000002
20,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[0008] In some embodiments of the method, the compound of formula (I) is:
Figure PCTKR2023008331-appb-img-000003
22,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[0009] In some embodiments of the method, the compound of formula (I) is:
Figure PCTKR2023008331-appb-img-000004
18,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[0010] In some embodiments of the method, the compound of formula (I) is:
Figure PCTKR2023008331-appb-img-000005
38,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[0011] In some embodiments of the method, the compound of formula (I) is:
Figure PCTKR2023008331-appb-img-000006
47,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[0012] In some embodiments of the method, the compound of formula (I) is:
Figure PCTKR2023008331-appb-img-000007
48,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[0013] In some embodiments of the method, the compound of formula (I) is:
Figure PCTKR2023008331-appb-img-000008
49,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[0014] In some embodiments of the method, the compound of formula (I) is:
Figure PCTKR2023008331-appb-img-000009
50,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[0015] In some embodiments, the subject has a cancer selected from breast cancer, gastric cancer gastrointestinal cancer, a gynecological cancer, kidney cancer, glioblastoma, high-grade-Glioma, non-small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, head and neck cancer, prostate cancer, metastatic castration-resistant prostate cancer (mCRPC), glioma, brain neoplasms and brain metastasis.
[0016] In some embodiments, the subject has breast cancer. In some embodiments, the breast cancer is a triple-negative breast cancer (TNBC). In some embodiments, the TNBC is a subtype selected from basal-like type 1 (BL1), basal-like type 2 (BL2), immunomodulatory (IM), mesenchymal (M), mesenchymal stem-like (MSL), and luminal androgen receptor (LAR) subtypes. In some embodiments, the TNBC is androgen receptor (AR)-positive triple-negative breast cancer (AR+ TNBC). In some embodiments, the TNBC comprises a BRCA1 mutation.
[0017] In some embodiments, the subject has glioblastoma.
[0018] In some embodiments, the subject has gastric cancer.
[0019] In some embodiments, the subject has prostate cancer. In some embodiments, the subject has metastatic castration-resistant prostate cancer (mCRPC).
[0020] In some embodiments, the present disclosure provides a pharmaceutical composition for use in inhibiting AR- and/or PDE5, or treating a cancer, comprising a therapeutically effective amount of an AR- and/or PDE5-inhibiting compound, or a pharmaceutically acceptable salt, a solvate, a hydrate, a prodrug, or a stereoisomer thereof. In some embodiments, the present disclosure provides a use of an AR- and/or PDE5-inhibiting compound, or a pharmaceutically acceptable salt, a solvate, a hydrate, a prodrug, or a stereoisomer thereof for inhibiting AR- and/or PDE5, or for treating or ameliorating of cancer.
[0021] In some embodiments, the AR- and PDE5-inhibiting compound is of formula (I):
Figure PCTKR2023008331-appb-img-000010
(I)
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutically acceptable salt thereof, wherein:
L is a linking moiety;
R1 and R2 are independently selected from -H, optionally substituted (C1-C6)alkyl, optionally substituted (C3-C6)cycloalkyl, optionally substituted (C1-C6)alkoxy, and optionally substituted (C2-C4)alkenyl;
each R13 is selected from -H, optionally substituted (C1-C6)alkyl, and optionally substituted (C1-C6)alkoxy;
each R14 is selected from -H, -CN, -OH, -NH2, -NO2, halogen, optionally substituted (C1-C5)alkyl, optionally substituted (C1-C5)haloalkyl, optionally substituted (C1-C5)alkoxy, optionally substituted (C3-C6)cycloalkyl, and optionally substituted (C2-C4)alkenyl;
X1 is N or CR14;
X2 and X3 are independently selected from N and CR13;
Y1 and Y2 are independently selected from N and C, wherein one of Y1 and Y2 is N;
m is 0 to 2; and
n is 1 to 4.
2. DETAILED DESCRIPTION OF THE INVENTION
(i) 3.1 Methods of Treating Cancer
[0022] Aspects of the present disclosure include methods of treating cancer including administering to a subject having cancer a compound which exhibits dual functionality by: i) modulating the androgen receptor, and ii) modulating PDE5. The inventors have demonstrated that compounds of this disclosure which have dual action as androgen receptor and PDE5 inhibiting compounds can provide potent anticancer activity in vivo in various therapeutic models of cancer (e.g., as described herein). Based on the results presented herein, the compounds of this disclosure would be effective in treating cancer, in particular AR-positive cancers associated with overexpression of PDE5.
[0023] The terms "AR and PDE5 inhibitor", "AR- and PDE5-inhibiting compound", and "AR antagonist or inhibitor and PDE5 inhibitor compound" are used interchangeably herein to refer to compounds which exhibit the dual functionality or activity described herein.
[0024] In some embodiments, the treatment goals for the methods of this disclosure include: 1) the prevention of cancer progression; 2) the relief of symptoms; 3) improvement in exercise tolerance; 3) improvement in health status; 4) the prevention and treatment of cancer exacerbations; 5) the prevention and treatment of cancer complications; 5) a reduction in mortality; and/or 6) minimization of side-effects from cancer treatment.
[0025] The terms "treating," "treatment," and the like, refer to obtaining a desired pharmacologic and/or physiologic effect, such as reduction or alleviation of one or more symptoms of the disease or disorder (i.e., cancer). The effect may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease. "Treatment," as used herein, covers any treatment of a disease in a mammal, particularly in a human, and includes: (a) preventing the disease or a symptom of a disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it (i.e., including diseases that may be associated with or caused by a primary disease); (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease, i.e., causing regression of the disease (i.e., reduction in pain or other symptom).
[0026] The term "amelioration" or any grammatical variation thereof (e.g., ameliorate, ameliorating, and amelioration etc.), includes, but is not limited to, delaying the onset, or reducing the severity of a disease or condition (e.g., cancer). Amelioration, as used herein, does not require the complete absence of symptoms. In some embodiments of the methods of this disclosure, administration of a AR- and PDE5-inhibiting compound to a subject having cancer at least ameliorates one or more symptoms of the cancer.
[0027] In some embodiments, the AR and PDE5 inhibitors of the present disclosure are used for treating an AR positive cancer. In some embodiments, the AR- and PDE5 inhibitors of the present disclosure are used for treating a PDE5 expressing cancer. In some embodiments of the method, the subject has AR positive cancer cells and/or PDE5 expressing (e.g., PDE5 overexpressing) cancer cells.
[0028] In some embodiments of the method, the cancer is selected from breast cancer, gastric cancer gastrointestinal cancer, a gynecological cancer, kidney cancer, glioblastoma, high-grade-Glioma, non-small cell lung cancer, head and neck squamous cell carcinoma, ovarian cancer, uterine cancer, pancreatic cancer, head and neck cancer, prostate cancer, metastatic castration-resistant prostate cancer (mCRPC), glioma, brain neoplasms and brain metastasis.
(ii) 3.1.1 Breast Cancer
[0029] Encompassed within the scope of the present disclosure are methods for treating breast cancer, for delaying the progression of breast cancer, and for preventing and treating the recurrence of breast cancer and/or breast cancer metastasis, which comprise administering an AR- and PDE5 inhibiting compound (e.g., as described herein).
[0030] In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating various forms of breast cancer, whether or not the breast cancers express androgen receptors or estrogen receptors. Breast cancers that can be treated include, but are not limited to, basal-like breast cancer, BRCA1-related breast cancer, medullary breast cancer, metaplastic breast cancer, special histologic type of breast cancer, triple-negative breast cancer, and breast cancer resistant to endocrine therapy.
[0031] In some embodiments breast cancers are ER+ (i.e., 1% or more of the cells tested express ER detectable by immunocytochemistry). In some embodiments, breast cancers contain cells that demonstrate estradiol-mediated growth. In some embodiments patients to be treated have no detectable circulating levels of estradiol. In some embodiments patients to be treated have circulating levels of estradiol greater than 10 pmol/L. In some embodiments patients to be treated have circulating levels of estradiol less than 10 pmol/L.
[0032] In some of embodiments the breast cancer is a triple-negative breast cancer (TNBC) including, but not limited to, subtypes of triple-negative breast cancer such as of basal-like type 1 (BL1), basal-like type 2 (BL2), immunomodulatory (IM), mesenchymal (M), mesenchymal stem-like (MSL), and luminal androgen receptor (LAR) subtypes. "Triple-negative breast cancer" as used herein is characterized by lack of estrogen receptor (ER), progesterone receptor (PR), and lack of overexpression or amplification of Her2 gene. A tumor is negative for expression of ER or PR if fewer than 1% of the cells tested are positive for ER or PR, as measured by immunohistochemistry, and if the Her2 gene is not expressed (for example, amplification is not detected by FISH). Triple-negative breast cancer is clinically characterized as more aggressive and less responsive to standard treatment and is associated with poorer overall patient prognosis. It is diagnosed more frequently in younger women and in women with BRCA1 mutations.
[0033] In some embodiments a triple-negative breast cancer is AR+ (e.g., AR+ TNBC); i.e., it contains cells that express detectable androgen receptors as detected by immunohistochemistry, ligand binding, or other methods known in the art. In other embodiments a triple negative breast cancer is AR-.
[0034] Approximately 75% of breast cancers express the estrogen receptor (ER) and are candidates for endocrine therapy. The selective ER modulator tamoxifen is the most commonly prescribed endocrine therapy; however, approximately 30 percent of tumors that retain estrogen (ER) do not respond to estrogen/ER directed therapies such as tamoxifen or aromatase inhibitors (AI) and nearly all patients with metastatic disease develop resistance. In such patients, a compound can provide a therapeutic intervention.
[0035] In some embodiments the breast cancer is ER+, i.e., it contains detectable levels of estrogen receptor, measured as described above, but is resistant to endocrine therapy. "Endocrine therapy" as used herein includes administration of one or more aromatase inhibitors (e.g., anastrozole, exemestane, letrozole) and/or administration of one or more estrogen receptor modulators (e.g., tamoxifen, raloxifen, fulvestrant). "Resistant to endocrine therapy" as used herein means that the tumors (primary or metastases) do not respond to one or more of the above treatments by shrinking, but rather remains the same size or increases in size, or that recur in response to such treatment at any time in the patient's livespan.
[0036] In some embodiments the breast cancer is ER+/AR+. In some embodiments the breast cancer is ER+/AR-. In some embodiments the breast cancer contains cells that are progesterone receptor positive (PR+) as detected by immunohistochemistry or ligand binding assays or any other method of detection. In some embodiments the breast cancer contains no detectable cells with progesterone receptors; e.g., the breast cancer is progesterone receptor negative (PR-). In some embodiments a breast cancer contains cells that are Her2 positive (Her2+) as detected by observable Her2 gene amplification after in situ hybridization. In some embodiments a breast cancer contains no detectable cells with amplification or expression or overexpression of Her2; e.g., the breast cancer is Her2 negative (Her2-). The progesterone receptors and Her2 can be present on the same or different populations of cells, which may be the same or different as the populations of cells expressing ER and/or AR.
[0037] In some embodiments a breast cancer is identified as AR+, ER+, and Her2+. In some embodiments a breast cancer is identified as AR+, ER+, and PR+. In some embodiments a breast cancer is identified as AR+, ER+, Her2+, and PR+. In some embodiments a breast cancer is identified as AR-, ER+, and Her2+. In some embodiments a breast cancer is identified as AR-, ER+, and PR+. In some embodiments a breast cancer is identified as AR-, ER+, Her2+, and PR+. In some embodiments, a breast cancer is identified as AR+, ER-, HER2+, PR-.
(iii) 3.1.2 Prostate Cancer
In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating prostate cancer, decreasing the incidence of, or halting or causing a regression of prostate cancer. The AR and PDE5 inhibitors of the present disclosure are useful for treating various forms of prostate cancer. In certain embodiments, the prostate cancer is associated with overexpression of AR. In some embodiments, the prostate cancer is hormone refractory prostate cancer (HRPC). In some embodiments, the prostate cancer is hormone sensitive prostate cancer (HSPC). In some embodiments, the prostate cancer is metastatic castration-resistant prostate cancer (mCRPC).
(iv) 3.1.3 Other Cancers
[0038] In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating uterine cancer.
[0039] In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating ovarian cancer.
[0040] In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating a gastric cancer. In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating a gastrointestinal cancer.
[0041] In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating a gynecological cancer.
[0042] In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating kidney cancer.
[0043] In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating glioma. In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating glioblastoma. In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating high-grade-glioma. In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating brain neoplasms. In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating brain metastasis.
[0044] In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating non-small cell lung cancer.
[0045] In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating head and neck cancer. In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating head and neck squamous cell carcinoma.
[0046] In some embodiments, the AR and PDE5 inhibitors of the present disclosure are useful for treating pancreatic cancer.
[0047] In some embodiments, one or more symptoms of the cancer of interest are reduced or alleviated in the subject after administration of the AR and PDE5 inhibitor as described herein.
3.2 Administration Methods
[0048] The compounds can be administered via any convenient administration methods according to the methods of this disclosure. In some embodiments of the method of treating a cancer, the compound is administered orally, intravenously, subcutaneously, transdermally, intraperitoneally, or by inhalation. A compound can be administered using pharmaceutical compositions comprising a therapeutically effective amount of the compound of formula (I) and a pharmaceutically acceptable carrier or diluent, in a variety of forms adapted to the chosen route of administration, for example, orally, nasally, intraperitoneally, or parenterally, by intravenous, intramuscular, topical or subcutaneous routes, or by injection into tissue.
[0049] In some embodiments of the method of treating a cancer, the compound is delivered orally to the subject. A compound can be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier; or by inhalation or insufflation.
[0050] The compound can also be administered intravenously or intraperitoneally by infusion or injection. Solutions of a compound can be prepared.
[0051] In some embodiments, the pharmaceutical composition comprises dry powder.
[0052] In some embodiments, the pharmaceutical composition is systematically administered. In some embodiments of the method, the compound of formula (I) is administered intravenously. In another embodiment, the compound of formula (I) is administered enterically. In another embodiment, the compound of formula (I) is administered by mouth (p.o.). In another embodiment, the compound is administered by intranasal administration.
[0053] In some embodiments, the compound of formula (I) is administered via an enteral feeding tube. In another embodiment, the enteral feeding tube is a nasogastric tube.
(v) 3.2.1 Effective Dose
[0054] The terms "effective amount" or "pharmaceutically effective amount" or "therapeutically effective amount" of a compound or composition, is a quantity sufficient to achieve a desired therapeutic and/or prophylactic effect, e.g., an amount which results in the prevention of, or a decrease in, the symptoms associated with a disease (e.g., as described herein) that is being treated. The amount of a compound administered to the subject will depend on the type and severity of the disease and on the characteristics of the individual, such as general health, age, sex, body weight and tolerance to drugs. It will also depend on the degree, severity and type of disease. The skilled artisan will be able to determine appropriate dosages depending on these and other factors. The compounds of the present disclosure can also be administered in combination with one or more additional therapeutic compounds.
[0055] In some embodiments of the method, the effective amount of the compound of formula (I) is between 0.01 mg and 1,000 mg.
[0056] In some embodiments, the dose is administered as a single daily dose.
[0057] In some embodiments, the dose is administered as a plurality of equally divided sub-doses.
[0058] In some embodiments, the compound of formula (I) is administered once a day, twice a day, or three times a day.
[0059] In some embodiments, the compound of formula (I) is administered every two days, every three days, every four days, every five days, or every six days.
[0060] In some embodiments, the compound of formula (I) is administered once a week, once every two weeks, or once every three weeks.
[0061] In some embodiments, the compound of formula (I) is administered p.r.n.
[0062] In some embodiments of the method of treating breast cancer, the effective amount of the administered compound is an amount sufficient to decrease or inhibit tumor growth in breast tissue of the patient by at least 20% as compared to a baseline pre-treatment tumor size. In another embodiment, the effective amount is sufficient to decrease or inhibit tumor growth in breast tissue by at least 30% as compared to the pre-treatment tumor size. In another embodiment, the effective amount is sufficient to decrease or inhibit tumor growth in breast tissue by at least 40% as compared to the pre-treatment tumor size. In another embodiment, the effective amount is sufficient to decrease or inhibit tumor growth in breast tissue by at least 50% as compared to the pre-treatment tumor size. In another embodiment, the effective amount is sufficient to decrease or inhibit tumor growth in breast tissue by at least 60% as compared to the pre-treatment tumor size. In another embodiment, the effective amount is sufficient to decrease or inhibit tumor growth in breast tissue by at least 70% as compared to the pre-treatment tumor size. In another embodiment, the effective amount is sufficient to decrease or inhibit tumor growth in breast tissue by at least 80% as compared to the pre-treatment tumor size. In another embodiment, the effective amount is sufficient to decrease or inhibit tumor growth in breast tissue by at least 90% as compared to the pre-treatment tumor size.
(vi) 3.2.2 Patient Selection/Condition
[0063] The present disclosure provides a method of treating a subject with a cancer.
[0064] In some embodiments, the subject has a breast cancer. In some embodiments, the subject has a triple-negative breast cancer (TNBC). In some embodiments, the subject has a basal-like type 1 (BL1) subtype of TNBC. In some embodiments, the subject has a basal-like type 2 (BL2) subtype of TNBC. In some embodiments, the subject has an immunomodulatory (IM) subtype of TNBC. In some embodiments, the subject has a mesenchymal (M) subtype of TNBC. In some embodiments, the subject has a mesenchymal stem-like (MSL) subtype of TNBC. In some embodiments, the subject has a luminal androgen receptor (LAR) subtype of TNBC. In some embodiments, the subject has androgen receptor (AR)-positive triple-negative breast cancer (AR+ TNBC). In some embodiments, the subject has a TNBC that comprises a BRCA1 mutation. In some embodiments of the method, the subject has a tumor in a breast tissue before administration of the compound.
[0065] In some embodiment of the method of treating a cancer, patients to be treated are post-menopausal. In some embodiments patients to be treated are pre-menopausal. In some embodiments patients to be treated are peri-menopausal. In some embodiments patients to be treated are men.
[0066] In some embodiments, the subject has gastric cancer. In some embodiments, the subject has a gastrointestinal cancer. In some embodiments, the subject has a gynecological cancer. In some embodiments, the subject has kidney cancer. In some embodiments, the subject has glioma. In some embodiments, the subject has a brain neoplasm. In some embodiments, the subject has brain metastasis. In some embodiments, the subject has glioblastoma. In some embodiments, the subject has high-grade-Glioma. In some embodiments, the subject has non-small cell lung cancer (NSCLC). In some embodiments, the subject has head and neck squamous cell carcinoma. In some embodiments, the subject has pancreatic cancer. In some embodiments, the subject has head and neck cancer. In some embodiments, the subject has prostate cancer. In some embodiments, the subject has metastatic castration-resistant prostate cancer (mCRPC).
[0067] In some embodiments of the method, the subject has a refractory cancer.
[0068] In some embodiments, the subject is not hospitalized. In another embodiment, the subject is hospitalized.
(vii) 3.2.3 Combination therapies
[0069] In some embodiments of the methods of treating a cancer, one or more additional active compounds are co-administered to the subject in conjunction with the AR- and PDE5- inhibiting compound. "Co-administration" of compounds includes one or more compounds administered substantially simultaneously with the AR and PDE5 inhibitor of this disclosure, whether or not in the same pharmaceutical composition, or sequentially.
[0070] In some embodiments, one or more of the compounds disclosed herein is administered or combined with other anticancer therapies, such as internal or external radiation, surgery, and chemotherapies.
3.3 AR- and PDE5 -Inhibiting Compounds
[0071] As summarized above, the present disclosure provides use of compounds having dual activity, e.g., as androgen receptor (AR) and PDE5 inhibiting compounds, in treating cancer in a subject in need thereof.
[0072] The present disclosure also provides methods of treating cancer using compounds having inhibitory or antagonistic activity against the androgen receptor (AR). "Androgen receptor" or "nuclear receptor subfamily 3, group c, member 4" or "NR3C4" is a type of nuclear receptor that is activated by binding any of the androgenic hormones, including testosterone and dihydrotestosterone in the cytoplasm and then translocating into the nucleus. In some embodiments, the administration of the compounds of the present disclosure can cause significant changes in AR activity as illustrated by Examples 4 and 5.
[0073] The AR-inhibiting activity of the compounds can be assessed using cellular assays. For example, Table 4 of Example 4 in the experimental section shows the IC50 values for exemplary compounds in an in vitro AR reporter assay. As illustrated in Example 4, tested compounds exhibited superior antagonistic activity against AR with comparable or more potent IC50 values as compared to enzalutamide and apalutamide. Further, Table 5 of Example 5 in the experimental section shows the binding affinity for exemplary compounds in an in vitro radioligand binding assay. As illustrated in Example 5, exemplary compounds exhibited superior or comparable binding affinity and AR inhibition as compared to enzalutamide and apalutamide.
[0074] The present disclosure provides methods of treating a cancer using compounds having potent PDE5 inhibitory activity. Phosphodiesterase 5 (PDE5) is a phosphodiesterase. Inhibition of PDE5 suppresses the decomposition of cGMP, which can then lead to increased activity of PKG along with increasing the concentration of cGMP. Increasing the activity of PKG can then cause phosphorylation of numerous biologically important targets, relaxation of the smooth muscles, and increase in the flow of blood. PDE5 inhibitors are a well characterized class of agent having a variety of activities. A human phosphodiesterase5 (PDE5) inhibition assay in host cells can be used to assess the abilities of the compounds of the present disclosure to inhibit target PDE5. In some embodiments, the administration of the compounds of the present disclosure can cause significant changes in PDE5 activity as illustrated by Example 3.
[0075] The PDE5 inhibiting activity of the compounds can be assessed using in vitro enzyme assays. For example, Table 3 of Example 3 in the experimental section shows the IC50 values for exemplary compounds in as in vitro PDE5 assay. The AR and PDE5 inhibitors may inhibit the activity of AR and PDE5 in a sample, e.g., as assessed by a AR or PDE5 inhibition assay described in Examples 3 and 4. AR and PDE5 inhibitors according to such methods may each have IC50 values for AR and PDE5 inhibition, (e.g., as assessed by the assays of Example 3-4) of less than 5000 nM, such as 1000 nM or less, 200 nM or less, 100 nM or less, or 20 nM or less. Biological systems may include subjects (e.g., human subjects).
[0076] Any convenient compounds having dual AR-inhibiting and PDE5-inhibiting activity may be used in the methods of this disclosure. In general, the AR- and PDE5 inhibiting compounds include bicyclic core structures of substituted 1,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one (
Figure PCTKR2023008331-appb-img-000011
) or substituted imidazo[5,1-f][1,2,4]triazin-4(3H)-one (
Figure PCTKR2023008331-appb-img-000012
) covalently attached to various cyano-substituted aryl groups via i) a 1,3-phenylene, 2,4-pyridyl or 2,6-pyridyl, and ii) a variety of linking moieties such as substituted 2-thioxoimidazolidin-4-one (
Figure PCTKR2023008331-appb-img-000013
) groups, urea groups, or thiourea groups.
[0077] More specifically, the AR- and PDE5-inhibiting compound can be a compound of formula (I):
Figure PCTKR2023008331-appb-img-000014
(I)
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein:
L is a linking moiety;
R1 and R2 are independently selected from -H, optionally substituted (C1-C6)alkyl, optionally substituted (C3-C6)cycloalkyl, optionally substituted (C1-C6)alkoxy, and optionally substituted (C2-C4)alkenyl;
each R13 is selected from -H, optionally substituted (C1-C6)alkyl, and optionally substituted (C1-C6)alkoxy;
each R14 is selected from -H, -CN, -OH, -NH2, -NO2, halogen, optionally substituted (C1-C5)alkyl, optionally substituted (C1-C5)haloalkyl, , optionally substituted (C1-C5)alkoxy, optionally substituted (C3-C6)cycloalkyl, and optionally substituted (C2-C4)alkenyl;
X1 is N or CR14;
X2 and X3 are independently selected from N and CR13;
Y1 and Y2 are independently selected from N and C, wherein one of Y1 and Y2 is N;
m is 0 to 2; and
n is 1 to 4.
[0078] In some embodiments of formula (I), the compound is of formula (Ia):
Figure PCTKR2023008331-appb-img-000015
(Ia)
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof,
wherein:
each R13 is selected from -H, halogen and optionally substituted (C1-C6)alkoxy; and
R4 and each R14 is independently selected from -H, -CN, -OH, -NH2, -NO2, halogen, optionally substituted (C1-C5)alkyl, and optionally substituted (C1-C5)haloalkyl.
[0079] In some embodiments of formula (I)-(Ia), -L- is -A-B-, wherein:
-A- is selected from a covalent bond, optionally substituted (C6-C12) aryl or (C3-C12) heteroaryl, optionally substituted-(C3-C12) heteroaryl-(C1-C5)alkylene-, optionally substituted 3- to 6-membered heterocycle, -NHC(O)R5-,
Figure PCTKR2023008331-appb-img-000016
, and
Figure PCTKR2023008331-appb-img-000017
; and
-B- is selected from a covalent bond, optionally substituted 3- to 6-membered heterocycle, -NHC(O)R5-, -O-, -S-, -NR11-,
Figure PCTKR2023008331-appb-img-000018
,
Figure PCTKR2023008331-appb-img-000019
, and
Figure PCTKR2023008331-appb-img-000020
;
wherein:
R11 is H or optionally substituted (C1-C3)alkyl;
R5 is selected from -OH, -(C1-C5)alkyl, -(C1-C5)haloalkyl an optionally substituted (C1-C5)alkylene;
R6 and R7 are each independently -H or optionally substituted (C1-C3)alkyl; or R6 and R7 together with the nitrogen atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered heterocycle;
Z1 is selected from O and S; and
at least one of -A- and -B- is not a covalent bond.
[0080] In some embodiments of formula (Ia), -A- is
Figure PCTKR2023008331-appb-img-000021
.
[0081] In some embodiments of formula (I)-(Ia), the compound is of formula (IIa) or (IIb):
Figure PCTKR2023008331-appb-img-000022
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein:
R3 is selected from -H, and optionally substituted (C1-C6)alkoxy; and
-B- is selected from covalent bond and optionally substituted 3- to 6-membered heterocycle.
[0082] In some embodiments of formula (IIa) or (IIb), each R14 is independently -H or halogen. In some embodiments of formula (IIa) or (IIb), each R14 is independently -H or -F. In some embodiments of formula (IIa) or (IIb), each R14 is -H. In some embodiments of formula (IIa) or (IIb), each R14 is -F. In some embodiments of formula (IIa) or (IIb), at least one R14 is -F.
[0083] In some embodiments of formula (IIa) or (IIb), Z1 is S. In some embodiments of formula (IIa) or (IIb), Z1 is O.
[0084] In some embodiments of formula (IIa) or (IIb), R6 and R7 together with the nitrogen atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered heterocycle.
[0085] In some embodiments of formula (IIa) or (IIb), the compound is of formula (IIIa) or (IIIb):
Figure PCTKR2023008331-appb-img-000023
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein:
R8 and R9 are independently selected from -H and optionally substituted (C1-C3)alkyl, or R8 and R9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered carbocycle or optionally substituted 3-to 6-membered heterocycle; and
Z1 is O or S.
[0086] In some embodiments of formula (IIIa) or (IIIb), each R14 is independently -H or halogen. In some embodiments of formula (IIIa) or (IIIb), each R14 is independently -H or -F. In some embodiments of formula (IIIa) or (IIIb), each R14 is -H. In some embodiments of formula (IIIa) or (IIIb), each R14 is -F. In some embodiments of formula (IIIa) or (IIIb), at least one R14 is -F.
[0087] In some embodiments of formula (IIIa) or (IIIb), Z1 is S. In some embodiments of formula (IIIa) or (IIIb), Z1 is O.
[0088] In some embodiments of formula (IIIa) or (IIIb), -B- is a covalent bond and the compound is of formula (IVa) or (IVb):
Figure PCTKR2023008331-appb-img-000024
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[0089] In some embodiments of formula (IVa) or (IVb), each R14 is independently -H or halogen. In some embodiments of formula (IVa) or (IVb), each R14 is independently -H or -F. In some embodiments of formula (IVa) or (IVb), each R14 is -H. In some embodiments of formula (IVa) or (IVb), each R14 is -F. In some embodiments of formula (IVa) or (IVb), at least one R14 is -F.
[0090] In some embodiments of formula (IVa) or (IVb), Z1 is S. In some embodiments of formula (IVa) or (IVb), Z1 is O.
[0091] In some embodiments of formula (IIIa) or (IIIb), -B- is an optionally substituted 4- to 6-membered heterocycle.
[0092] In some embodiments of formula (IIIa) or (IIIb), the compound is of formula (Va) or (Vb):
Figure PCTKR2023008331-appb-img-000025
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein p and q are independently 1 or 2.
[0093] In some embodiments of formula (Va) or (Vb), -B- is selected from
Figure PCTKR2023008331-appb-img-000026
,
Figure PCTKR2023008331-appb-img-000027
, and
Figure PCTKR2023008331-appb-img-000028
.
[0094] In some embodiments of formula (Va) or (Vb), R8 is -H. In another embodiment of formula (Va) or (Vb), R9 is -H.
[0095] In some embodiments of formula (Va) or (Vb), R9 is an optionally substituted (C1-C3)alkyl. In another embodiment of formula (Va) or (Vb), R9 is -CH3.
[0096] In some embodiments of formula (Va) or (Vb), R8 and R9 are each independently optionally substituted (C1-C3)alkyl. In another embodiments of formula (Va) or (Vb), R8 and R9 are each -CH3.
[0097] In some embodiments of formula (Va) or (Vb), Z1 is S. In some embodiments of formula (Va) or (Vb), Z1 is O.
[0098] In some embodiments of formula (Va) or (Vb), -A- is selected from
Figure PCTKR2023008331-appb-img-000029
,
Figure PCTKR2023008331-appb-img-000030
, and
Figure PCTKR2023008331-appb-img-000031
.
[0099] In some embodiments of formula (Va) or (Vb), R8 and R9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered carbocycle or optionally substituted 3-to 6-membered heterocycle. In some embodiments of formula (Va) or (Vb), R8 and R9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 4-membered or 5-membered carbocycle or heterocycle. In another embodiment of formula (Va) or (Vb), the optionally substituted 3- to 6-membered carbocycle or optionally substituted 3-to 6-membered heterocycle is selected from optionally substituted cyclobutyl, optionally substituted cyclopentyl, and optionally substituted tetrahydrofuran.
[00100] In some embodiments of formula (Va) or (Vb), -A- is selected from
Figure PCTKR2023008331-appb-img-000032
,
Figure PCTKR2023008331-appb-img-000033
and
Figure PCTKR2023008331-appb-img-000034
.
[00101] In some embodiments of formula (Va) or (Vb), each R14 is independently -H or halogen. In some embodiments of formula (Va) or (Vb), each R14 is independently -H or -F. In some embodiments of formula (Va) or (Vb), each R14 is -H. In some embodiments of formula (Va) or (Vb), each R14 is -F. In some embodiments of formula (Va) or (Vb), at least one R14 is -F.
[00102] In some embodiments of formula (IIa) or (IIb), R6 and R7 are each independently -H or optionally substituted (C1-C3)alkyl. In some embodiments of formula (IIa) or (IIb), R6 and R7 are each -H.
[00103] In some embodiments of formula (IIa) or (IIb), the compound is of formula (VIa) or (VIb):
Figure PCTKR2023008331-appb-img-000035
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00104] In some embodiments of formula (VIa) or (VIb), Z1 is S. In some embodiments of formula (VIa) or (VIb), Z1 is O.
[00105] In some embodiments of formula (VIa) or (VIb), -B- is a covalent bond.
[00106] In some embodiments of formula (VIa) or (VIb), each R14 is independently -H or halogen. In some embodiments of formula (VIa) or (VIb), each R14 is independently -H or -F. In some embodiments of formula (VIa) or (VIb), each R14 is -H. In some embodiments of formula (VIa) or (VIb), each R14 is -F. In some embodiments of formula (VIa) or (VIb), at least one R14 is -F.
[00107] In some embodiments of formula (VIa) or (VIb), the compound is of formula (VIIa) or (VIIb):
Figure PCTKR2023008331-appb-img-000036
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00108] In some embodiments of formula (Ia), -B- is
Figure PCTKR2023008331-appb-img-000037
wherein R11 is -H or optionally substituted (C1-C3)alkyl.
[00109] In some embodiments of formula (Ia), the compound is of formula (VIIIa) or (VIIIb):
Figure PCTKR2023008331-appb-img-000038
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from -H and optionally substituted (C1-C5)alkoxy.
[00110] In some embodiments of formula (VIIIa) or (VIIIb), -A- is an optionally substituted 3- to 6-membered heterocycle. In some embodiments of formula (VIIIa) or (VIIIb), -A- is an optionally substituted azetidine, optionally substituted pyrrolidine, optionally substituted piperidine, optionally substituted piperidin-2-one or optionally substituted pyrrolidin-2-one. In some embodiments, the -A- ring is connected to the adjacent 4-cyanophenyl or 2-cyanopyrid-5-yl ring via a N atom of the optionally substituted 3- to 6-membered heterocycle (e.g., optionally substituted azetidine, pyrrolidine, piperidine, piperidin-2-one or pyrrolidin-2-one).
[00111] In some embodiments of formula (VIIIa) or (VIIIb), -A- is
Figure PCTKR2023008331-appb-img-000039
wherein:
R12 is selected from -H, -OH, optionally substituted (C1-C3)alkyl, and optionally substituted (C1-C5)haloalkyl; and
r, s and t are independently is 0 or 1.
[00112] In some embodiments of formula (VIIIa) or (VIIIb), -A- is selected from:
Figure PCTKR2023008331-appb-img-000040
.
[00113] In some embodiments of formula (VIIIa) or (VIIIb), -A- is a covalent bond.
[00114] In some embodiments of formula (VIIIa) or (VIIIb), each R14 is independently -H or halogen. In some embodiments of formula (VIIIa) or (VIIIb), each R14 is independently -H or -F. In some embodiments of formula (VIIIa) or (VIIIb), each R14 is -H. In some embodiments of formula (VIIIa) or (VIIIb), each R14 is -F. In some embodiments of formula (VIIIa) or (VIIIb), at least one R14 is -F.
[00115] In some embodiments of formula (VIIIa) or (VIIIb), the compound is of formula (IXa) or (IXb):
Figure PCTKR2023008331-appb-img-000041
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00116] In some embodiments of formula (VIIIa) or (VIIIb), -A- is an optionally substituted -(C3-C12)heteroaryl-(C1-C5)alkylene- (e.g., where the -(C3-C12)heteroaryl- and/or the -(C1-C5)alkylene- of -A- are each optionally substituted).
[00117] In some embodiments of formula (VIIIa) or (VIIIb), -A- is
Figure PCTKR2023008331-appb-img-000042
.
[00118] In some embodiments of formula (I)-(Ia), -L- is -A-B-, wherein -A- is an optionally substituted 3- to 6-membered heterocycle. In some embodiments, -A- is an optionally substituted pyrrolidin-2-one.
[00119] In some embodiments, -A- is selected from
Figure PCTKR2023008331-appb-img-000043
,
Figure PCTKR2023008331-appb-img-000044
, and
Figure PCTKR2023008331-appb-img-000045
.
[00120] In some embodiments of formula (Ia), the compound is of formula (Xa) or (Xb):
Figure PCTKR2023008331-appb-img-000046
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein:
B is as defined above;
R3 is selected from -H, and optionally substituted (C1-C6)alkoxy; and
R12 is selected from -H and optionally substituted (C1-C3)alkyl.
[00121] In some embodiments of formula (Xa) or (Xb), R12 is -H. In some embodiments of formula (Xa) or (Xb), R12 is optionally substituted (C1-C3)alkyl. In some embodiments of formula (Xa) or (Xb), R12 is ethyl.
[00122] In some embodiments of formula (Xa) or (Xb), -B- is selected from -O-, -S-, -NH-, -SO2-, and -NHSO2-. In some embodiments of formula (Xa) or (Xb), -B- is -O-. In some embodiments of formula (Xa) or (Xb), -B- is -S-. In some embodiments of formula (Xa) or (Xb), -B- is -SO2-. In some embodiments of formula (Xa) or (Xb), -B- is -NHSO2-.
[00123] In some embodiments of formula (Xa) or (Xb), each R14 is independently -H or halogen. In some embodiments of formula (Xa) or (Xb), each R14 is independently -H or -F. In some embodiments of formula (Xa) or (Xb), each R14 is -H. In some embodiments of formula (Xa) or (Xb), each R14 is -F. In some embodiments of formula (Xa) or (Xb), at least one R14 is -F.
[00124] In some embodiments of formula (I)-(Ia), -L- is -A-B-, wherein -A- is -NHC(O)R5-. In some embodiments of -A-, R5 is
Figure PCTKR2023008331-appb-img-000047
. In some embodiments, the compound is of formula (XIa) or (XIb):
Figure PCTKR2023008331-appb-img-000048
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from -H, and optionally substituted (C1-C6)alkoxy.
[00125] In some embodiments of formula (XIa) or (XIb), -B- is selected from -O-, -S-, -SO2- and -NHSO2-. In some embodiments of formula (XIa) or (XIb), -B- is -O-. In some embodiments of formula (XIa) or (XIb), -B- is -S-. In some embodiments of formula (XIa) or (XIb), -B- is -SO2-. In some embodiments of formula (XIa) or (XIb), -B- is -NHSO2-.
[00126] In some embodiments of formula (XIa) or (XIb), each R14 is independently -H or halogen. In some embodiments of formula (XIa) or (XIb), each R14 is independently -H or -F. In some embodiments of formula (XIa) or (XIb), each R14 is -H. In some embodiments of formula (XIa) or (XIb), each R14 is -F. In some embodiments of formula (XIa) or (XIb), at least one R14 is -F.
[00127] In any one of the embodiments of formula (I) to (XIb) described herein, R1 is optionally substituted (C1-C6)alkyl. In any one of the embodiments of formula (I) to (XIb) described herein, R1 is optionally substituted (C1-C3)alkyl. In any one of the embodiments of formula (I) to (XIb) described herein R1 is -CH3.
[00128] In any one of the embodiments of formula (I) to (XIb) described herein, R2 is an optionally substituted (C1-C6)alkyl. In any one of the embodiments of formula (I) to (XIb) described herein, R2 is an optionally substituted (C1-C3)alkyl. In any one of the embodiments of formula (I) to (XIb) described herein, R2 is n-propyl.
[00129] In any one of the embodiments of formula (I) to (XIb) described herein, R3 is optionally substituted (C1-C3)alkoxy. In any one of the embodiments of formula (I) to (XIb) described herein, R3 is ethoxy.
[00130] In various embodiments of the compound, each R14 and R4 is an optionally substituted (C1-C5)haloalkyl or halogen. In another embodiment, R14 and R4 are each -CF3, -F or -Cl.
[00131] In some embodiments of the compound, X1, X2, and X3 are each CH.
[00132] In some embodiments of the compound, X1 is N. In another embodiment, X2 and X3 are each CH.
[00133] In some embodiments of the compound, X2 is N. In another embodiment, X1 and X3 are each CH.
[00134] In some embodiments of the compound, X3 is N. In another embodiment, X1 and X2 are each CH.
[00135] In some embodiments, the compound is of formula (IVc) or (IVd):
Figure PCTKR2023008331-appb-img-000049
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein R14 is -H or halogen.
[00136] In some embodiments of formula (IVc) or (IVd), R14 is -H or -F. In some embodiments of formula (IVc) or (IVd), R14 is -H. In some embodiments of formula (IVc) or (IVd), R14 is -F.
[00137] In some embodiments of formula (IVc) or (IVd), X1 is CH and R14 is -F. In some embodiments of formula (IVc) or (IVd), X1 is N and R14 is -F. In some embodiments of formula (IVc) or (IVd), X1 is CR14 and each R14 is -H. In some embodiments of formula (IVc) or (IVd), X1 is N and R14 is -H.
[00138] In some embodiments of formula (IVc) or (IVd), R8 and R9 are each independently -H or optionally substituted (C1-C3)alkyl. In another embodiment, R8 and R9 are each independently -CH3.
[00139] In some embodiments of formula (IVc) or (IVd), the compound is selected from:
Figure PCTKR2023008331-appb-img-000050
and
Figure PCTKR2023008331-appb-img-000051
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00140] In some embodiments of formula (IVc) or (IVd),
Figure PCTKR2023008331-appb-img-000052
is
Figure PCTKR2023008331-appb-img-000053
.
[00141] In some embodiments, the compound is selected from
Figure PCTKR2023008331-appb-img-000054
Figure PCTKR2023008331-appb-img-000055
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00142] In some embodiments of formula (IVc) or (IVd), R8 and R9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered carbocycle or optionally substituted 3- to 6-membere heterocycle (e.g., 4-membered or 5-membered carbocycle or heterocycle) that is selected from optionally substituted cyclobutyl, optionally substituted cyclopentyl, and optionally substituted tetrahydrofuran.
[00143] In some embodiments of formula (IVc) or (IVd),
Figure PCTKR2023008331-appb-img-000056
is
Figure PCTKR2023008331-appb-img-000057
.
[00144] In some embodiments, the compound is selected from:
Figure PCTKR2023008331-appb-img-000058
,
Figure PCTKR2023008331-appb-img-000059
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00145] In some embodiments, the compound is of formula (IVa), or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein:
X1 is CH or N;
each R14 is independently -H or halogen; and
R8 and R9 are each independently H or (C1-C3)alkyl (e.g., R8 and R9 are each -CH3), or R8 and R9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 5-membered carbocycle, or an optionally substituted 4-membered or 5-membered heterocycle (e.g., cyclopentane cyclobutane, cyclopentane, oxetane or tetrahydrofuran).
[00146] In some embodiments, the compound is of formula (IVc):
Figure PCTKR2023008331-appb-img-000060
(IVc)
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein:
X1 is CH or N;
R14 is -H or halogen; and
R8 and R9 are each independently H or (C1-C3)alkyl (e.g., R8 and R9 are each -CH3), or R8 and R9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 5-membered carbocycle, or an optionally substituted 4-membered or 5-membered heterocycle (e.g., cyclopentane cyclobutane, cyclopentane, oxetane or tetrahydrofuran).
[00147] In some embodiments, the compound is selected from:
Figure PCTKR2023008331-appb-img-000061
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00148] In some embodiments, the compound is
Figure PCTKR2023008331-appb-img-000062
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00149] In some embodiments, the compound is
Figure PCTKR2023008331-appb-img-000063
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00150] In some embodiments, the compound is
Figure PCTKR2023008331-appb-img-000064
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00151] In some embodiments, the compound is
Figure PCTKR2023008331-appb-img-000065
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00152] In some embodiments, the compound is
Figure PCTKR2023008331-appb-img-000066
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00153] In some embodiments, the compound is
Figure PCTKR2023008331-appb-img-000067
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00154] In some embodiments of formula (IVc) or (IVd),
Figure PCTKR2023008331-appb-img-000068
is
Figure PCTKR2023008331-appb-img-000069
.
[00155] In some embodiments, the compound is selected from:
Figure PCTKR2023008331-appb-img-000070
and
Figure PCTKR2023008331-appb-img-000071
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00156] In some embodiments of formula (IVc) or (IVd),
Figure PCTKR2023008331-appb-img-000072
is
Figure PCTKR2023008331-appb-img-000073
.
[00157] In some embodiments, the compound is selected from:
Figure PCTKR2023008331-appb-img-000074
and
Figure PCTKR2023008331-appb-img-000075
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00158] In some embodiments of formula (IVc) or (IVd), either X2 is N and X3 is CH, or X2 is CH and X3 is N. In another embodiment, R8 and R9 are each optionally substituted (C1-C3)alkyl. In another embodiment, R8 and R9 are each independently -CH3.
[00159] In some embodiments, the compound is selected from:
Figure PCTKR2023008331-appb-img-000076
and
Figure PCTKR2023008331-appb-img-000077
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00160] In some embodiments of formula (IVc) or (IVd), R8 and R9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered carbocycle or optionally substituted 3- to 6-membered heterocycle (e.g., 4-membered or 5-membered carbocycle or heterocycle) that is selected from optionally substituted cyclobutyl, optionally substituted cyclopentyl, and optionally substituted tetrahydrofuran.
[00161] In some embodiments, the compound is selected from
Figure PCTKR2023008331-appb-img-000078
and
Figure PCTKR2023008331-appb-img-000079
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00162] In some embodiments, the compound is of formula (Vc):
Figure PCTKR2023008331-appb-img-000080
(Vc)
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00163] In some embodiments of formula (Vc), R8 and R9 are each independently optionally substituted (C1-C3)alkyl. In another embodiment of formula (Vc), R8 and R9 are each -CH3.
[00164] In some embodiments of formula (Vc), the compound is selected from:
Figure PCTKR2023008331-appb-img-000081
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00165] In some embodiments of formula (Vc), R8 and R9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered carbocycle or optionally substituted 3- to 6-membered heterocycle (e.g., 4-membered or 5-membered carbocycle or heterocycle) that is selected from optionally substituted cyclobutyl, optionally substituted cyclopentyl, and optionally substituted tetrahydrofuran.
[00166] In some embodiments, the compound is selected from:
Figure PCTKR2023008331-appb-img-000082
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00167] In some embodiments, the compound is of formula (VIIc):
Figure PCTKR2023008331-appb-img-000083
(VIIc)
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00168] In some embodiments, the compound is selected from:
Figure PCTKR2023008331-appb-img-000084
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00169] In some embodiments, the compound is of formula (VIIIc):
Figure PCTKR2023008331-appb-img-000085
(VIIIc)
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00170] In some embodiments of formula (VIIIc), -A- is
Figure PCTKR2023008331-appb-img-000086
wherein:
R12 is selected from -H, -OH, optionally substituted (C1-C3)alkyl, and optionally substituted (C1-C5)haloalkyl, ; and
r, s and t are independently is 0 or 1.
[00171] In some embodiments of formula (VIIIc), R4 is -CF3.
[00172] In some embodiments, the compound is selected from:
Figure PCTKR2023008331-appb-img-000087
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00173] In some embodiments, the compound is of formula (IXc) or (IXd):
Figure PCTKR2023008331-appb-img-000088
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00174] In some embodiments of formula (IXc) or (IXd), R4 is -CF3 or -Cl.
[00175] In some embodiments, the compound is selected from:
Figure PCTKR2023008331-appb-img-000089
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00176] In some embodiments, the compound is of formula (VIIId):
Figure PCTKR2023008331-appb-img-000090
(VIIId)
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00177] In some embodiments of formula (VIIId), R4 is -Cl.
[00178] In some embodiments, the compound is
Figure PCTKR2023008331-appb-img-000091
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00179] In some embodiments, the compound is of formula (XIc):
Figure PCTKR2023008331-appb-img-000092
(XIc)
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00180] In some embodiments, the compound is selected from:
Figure PCTKR2023008331-appb-img-000093
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00181] In some embodiments, the compound is of formula (Xc):
Figure PCTKR2023008331-appb-img-000094
(Xc)
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00182] In some embodiments of formula (Xc), -B- is selected from -NH-, -O-, -S-, and -SO2-.
[00183] In some embodiments, the compound is selected from:
Figure PCTKR2023008331-appb-img-000095
,
or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
[00184] In some embodiments, the compound is represented by the structure of one of the compounds in Table 1, or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt form thereof.
Exemplary AR Antagonist and/or PDE5 Inhibitor Compounds of Formula (I)-(XIc)
Cmpd No. Structure Name
1
Figure PCTKR2023008331-appb-img-000096
N-(4-cyano-3-(trifluoromethyl)phenyl)-4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)benzenesulfonamide
2
Figure PCTKR2023008331-appb-img-000097
(R)-N-(4-cyano-3-(trifluoromethyl)phenyl)-3-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)thio)-2-hydroxy-2-methylpropanamide
3
Figure PCTKR2023008331-appb-img-000098
(R)-N-(4-cyano-3-(trifluoromethyl)phenyl)-3-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)sulfonyl)-2-hydroxy-2-methylpropanamide
4
Figure PCTKR2023008331-appb-img-000099
(R)-N-(4-cyano-3-(trifluoromethyl)phenyl)-3-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)sulfonamido)-2-hydroxy-2-methylpropanamide
5
Figure PCTKR2023008331-appb-img-000100
N-(6-cyano-5-(trifluoromethyl)pyridin-3-yl)-4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)benzenesulfonamide
6
Figure PCTKR2023008331-appb-img-000101
N-(6-cyano-5-(trifluoromethyl)pyridin-3-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide
7
Figure PCTKR2023008331-appb-img-000102
N-(4-cyano-3-(trifluoromethyl)phenyl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide
8
Figure PCTKR2023008331-appb-img-000103
N-(3-chloro-4-cyanophenyl)-4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)benzenesulfonamide
9
Figure PCTKR2023008331-appb-img-000104
N-(3-chloro-4-cyanophenyl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide
10
Figure PCTKR2023008331-appb-img-000105
N-((3S,5R)-1-(4-cyano-3-(trifluoromethyl)phenyl)-5-(2,2,2-trifluoro-1-hydroxyethyl)pyrrolidin-3-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide
11
Figure PCTKR2023008331-appb-img-000106
(S)-N-(1-(3-(3-chloro-4-cyanophenyl)-1H-pyrazol-1-yl)propan-2-yl)-4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)benzenesulfonamide
12
Figure PCTKR2023008331-appb-img-000107
4-(3-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)amino)-2-ethyl-5-oxopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile
13
Figure PCTKR2023008331-appb-img-000108
4-(3-(4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile
14
Figure PCTKR2023008331-appb-img-000109
4-((2R,3R)-3-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)amino)-2-ethyl-5-oxopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile
15
Figure PCTKR2023008331-appb-img-000110
4-((2R,3S)-3-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)amino)-2-ethyl-5-oxopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile
16
Figure PCTKR2023008331-appb-img-000111
4-((2S,3R)-3-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)amino)-2-ethyl-5-oxopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile
17
Figure PCTKR2023008331-appb-img-000112
4-((2S,3S)-3-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)amino)-2-ethyl-5-oxopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile
18
Figure PCTKR2023008331-appb-img-000113
4-(3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile
19
Figure PCTKR2023008331-appb-img-000114
5-(3-(4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-3-(trifluoromethyl)picolinonitrile
20
Figure PCTKR2023008331-appb-img-000115
5-(3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-3-(trifluoromethyl)picolinonitrile
21
Figure PCTKR2023008331-appb-img-000116
4-(5-(4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7-yl)-2-(trifluoromethyl)benzonitrile
22
Figure PCTKR2023008331-appb-img-000117
4-(5-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7-yl)-2-(trifluoromethyl)benzonitrile
23
Figure PCTKR2023008331-appb-img-000118
5-(5-(4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7-yl)-3-(trifluoromethyl)picolinonitrile
24
Figure PCTKR2023008331-appb-img-000119
5-(5-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7-yl)-3-(trifluoromethyl)picolinonitrile
25
Figure PCTKR2023008331-appb-img-000120
4-(3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-5-oxo-2-thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile
26
Figure PCTKR2023008331-appb-img-000121
4-(3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4-methyl-5-oxo-2-thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile
27
Figure PCTKR2023008331-appb-img-000122
1-(4-cyano-3-(trifluoromethyl)phenyl)-3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)urea
28
Figure PCTKR2023008331-appb-img-000123
1-(4-cyano-3-(trifluoromethyl)phenyl)-3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo
[5,1-f][1,2,4]triazin-2-yl)phenyl)thiourea
29
Figure PCTKR2023008331-appb-img-000124
N-(1-(4-cyano-3-(trifluoromethyl)phenyl)azetidin-3-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide
30
Figure PCTKR2023008331-appb-img-000125
N-(1-(4-cyano-3-(trifluoromethyl)phenyl)piperidin-4-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide
31
Figure PCTKR2023008331-appb-img-000126
N-(1-(4-cyano-3-(trifluoromethyl)phenyl)pyrrolidin-3-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide
32
Figure PCTKR2023008331-appb-img-000127
N-(1-(4-cyano-3-(trifluoromethyl)phenyl)-2-oxopyrrolidin-3-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide
33
Figure PCTKR2023008331-appb-img-000128
N-(1-(4-cyano-3-(trifluoromethyl)phenyl)-5-oxopyrrolidin-3-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide
34
Figure PCTKR2023008331-appb-img-000129
4-(1-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4-oxo-2-thioxo-7-oxa-1,3-diazaspiro[4.4]nonan-3-yl)-2-(trifluoromethyl)benzonitrile
35
Figure PCTKR2023008331-appb-img-000130
4-(3-(4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenoxy)-2-ethyl-5-oxopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile
36
Figure PCTKR2023008331-appb-img-000131
4-(3-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)thio)-2-ethyl-5-oxopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile
37
Figure PCTKR2023008331-appb-img-000132
4-(3-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)sulfonyl)-2-ethyl-5-oxopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile
38
Figure PCTKR2023008331-appb-img-000133
4-(1-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4-oxo-2-thioxo-1,3-diazaspiro[4.4]nonan-3-yl)-2-(trifluoromethyl)benzonitrile
39
Figure PCTKR2023008331-appb-img-000134
4-(3-(5-ethoxy-6-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)pyridin-2-yl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile
40
Figure PCTKR2023008331-appb-img-000135
4-(3-(5-ethoxy-4-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)pyridin-2-yl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile
41
Figure PCTKR2023008331-appb-img-000136
4-(5-(5-ethoxy-6-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)pyridin-2-yl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7-yl)-2-(trifluoromethyl)benzonitrile
42
Figure PCTKR2023008331-appb-img-000137
4-(5-(5-ethoxy-4-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)pyridin-2-yl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7-yl)-2-(trifluoromethyl)benzonitrile
43
Figure PCTKR2023008331-appb-img-000138
4-(3-(1-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)azetidin-3-yl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile
44
Figure PCTKR2023008331-appb-img-000139
4-(3-(1-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)piperidin-4-yl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile
45
Figure PCTKR2023008331-appb-img-000140
4-(5-(1-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)azetidin-3-yl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7-yl)-2-(trifluoromethyl)benzonitrile
46
Figure PCTKR2023008331-appb-img-000141
4-(5-(1-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)piperidin-4-yl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7-yl)-2-(trifluoromethyl)benzonitrile
47
Figure PCTKR2023008331-appb-img-000142
4-(3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-3-fluoro-2-(trifluoromethyl)benzonitrile
48
Figure PCTKR2023008331-appb-img-000143
4-(5-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7-yl)-3-fluoro-2-(trifluoromethyl)benzonitrile
49
Figure PCTKR2023008331-appb-img-000144
4-(1-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4-oxo-2-thioxo-7-oxa-1,3-diazaspiro[4.4]nonan-3-yl)-3-fluoro-2-(trifluoromethyl)benzonitrile
50
Figure PCTKR2023008331-appb-img-000145
4-(1-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4-oxo-2-thioxo-1,3-diazaspiro[4.4]nonan-3-yl)-3-fluoro-2-(trifluoromethyl)benzonitrile
[00185] It is understood that all variations of salts, solvates, hydrates, prodrugs and/or stereoisomers of the compounds described herein (e.g., of Formula (I)-(XIc), such as a compound of Table 1) are meant to be encompassed by the present disclosure.
3.3.1 Isotopically Labelled Analogs
[00186] In some embodiments of the method of treating a cancer, the compounds disclosed in the present disclosure are isotopically-labeled compounds which are identical to those compounds as described herein, except that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature ("isotopologues"). The compounds of the present disclosure may also contain unnatural proportions of atomic isotopes at one or more atoms that constituted such compounds. Examples of isotopes that can be incorporated into compounds described herein include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, such as 2H ("D"), 3H, 13C, 14C, 15N, 18O, 17O, 31P, 32P, 35S, 18F, and 36Cl, respectively. For example, a compound described herein can have one or more H atoms replaced with deuterium.
[00187] Unless otherwise stated, compounds described herein are intended to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13C- or 14C- enriched carbon are within the scope of the present disclosure.
[00188] In some embodiments of the method, certain isotopically-labeled compounds, such as those labeled with 3H and 14C, can be useful in compound and/or substrate tissue distribution assays. Tritiated (3H) and carbon-14 (14C) isotopes can be particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium can afford certain therapeutic advantages resulting from greater metabolic stability, such as increased in vivo half-life or reduced dosage requirements, and hence can be preferred in some circumstances. Isotopically-labeled compounds can generally be prepared by following procedures analogous to those disclosed herein, for example, in the Examples section, by substituting an isotopically-labeled reagent for a non-isotopically-labeled reagent.
[00189] In some embodiments of the method, the compounds disclosed in the present disclosure are deuterated analogs of any of the compounds, or a salt thereof, as described herein. A deuterated analog of a compound of any one of formulae (I)-(XIc) is a compound where one or more hydrogen atoms are substituted with a deuterium. In some embodiments, the deuterated analog is a compound of any one of formulae (I)-(XIc) that includes a deuterated R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, or R14 group. In certain embodiments of a deuterated analog of a compound of any one of formulae (I)-(XIc), R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, and R14 are independently selected from optionally substituted (C1-C6)alkyl, optionally substituted (C1-C6)alkoxy, optionally substituted (C1-C6)alkylene-heterocycloalkyl, optionally substituted monocyclic or bicyclic carbocycle, and optionally substituted monocyclic or bicyclic heterocycle including at least one deuterium atom.
[00190] Deuterium substituted compounds are synthesized using various methods such as described in: Dean, Dennis C.; Editor. Recent Advances in the Synthesis and Applications of Radiolabeled Compounds for Drug Discovery and Development. [In: Curr., Pharm. Des., 2000; 6(10)] 2000, 110 pp; George W.; Varma, Rajender S. The Synthesis of Radiolabeled Compounds via Organometallic Intermediates, Tetrahedron, 1989, 45(21), 6601-21; and Evans, E. Anthony. Synthesis of radiolabeled compounds, J. Radioanal. Chem., 1981, 64(1-2), 9-32.
[00191] Deuterated starting materials are readily available and are subjected to the synthetic methods described herein to provide for the synthesis of deuterium-containing compounds. Large numbers of deuterium-containing reagents and building blocks are available commercially from chemical vendors, such as Aldrich Chemical Co.
3.3.2 Fluorinated Analogs
[00192] In some embodiments of the method of treating a cancer, the compounds disclosed in the present disclosure are fluorinated analogs of any of the compounds, or a salt thereof, as described herein. A fluorinated analog of a compound of any one of formulae (I)-(XIc) is a compound where one or more hydrogen atoms or substituents are substituted with a fluorine atom. In some embodiments, the fluorinated analog is a compound of any one of formulae (I)-(XIc) that includes a fluorinated R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, or R14 group. In some embodiments of a fluorinated analog of a compound of any one of formulae (I)-(XIc), R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, and R14 group are independently selected from optionally substituted (C1-C6)alkyl, optionally substituted (C1-C6)alkoxy, optionally substituted (C1-C6)alkylene-heterocycloalkyl, optionally substituted monocyclic or bicyclic carbocycle, optionally substituted monocyclic or bicyclic heterocycle, optionally substituted aryl, and optionally substituted heteroaryl including at least one fluorine atom. In some embodiments of a fluorinated analog of a compound of any one of formulae (I)-(XIc), the hydrogen atom of an aliphatic or an aromatic C-H bond is replaced by a fluorine atom. In some embodiments of a fluorinated analog of a compound of any one of formulae (I)-(XIc), at least one hydrogen of an optionally substituted aryl or an optionally substituted heteroaryl is replaced by a fluorine atom. In some embodiments of a fluorinated analog of a compound of formula (I), a hydroxyl substituent (-OH) or an amino substituent (-NH2) is replaced by a fluorine atom.
3.3.3 Isomers
[00193] The term "compound", as used herein, is meant to include all stereoisomers, geometric isomers, tautomers, and isotopes of the structures depicted.
[00194] The compounds herein described may have asymmetric centers, geometric centers (e.g., double bond), or both. All chiral, diastereomeric, racemic forms and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomeric form is specifically indicated. In some embodiments of the method of treating a cancer, the compounds described herein have one or more chiral centers. It is understood that if an absolute stereochemistry is not expressly indicated, then each chiral center may independently be of the R-configuration or the S-configuration or a mixture thereof. Thus, compounds described herein include enriched or resolved optical isomers at any or all asymmetric atoms as are apparent from the depictions. Racemic mixtures of R-enantiomer and S-enantiomer, and enantio-enriched stereoisomeric mixtures comprising of R- and S-enantiomers, as well as the individual optical isomers can be isolated or synthesized so as to be substantially free of their enantiomeric or diastereomeric partners, and these stereoisomers are all within the scope of the present technology.
[00195] Compounds of the present disclosure containing an asymmetrically substituted atom may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms, by synthesis from optically active starting materials, or through use of chiral auxiliaries.
[00196] Geometric isomers, resulting from the arrangement of substituents around a carbon-carbon double bond or arrangement of substituents around a cycloalkyl or heterocyclic ring, can also exist in the compounds of the present disclosure. Geometric isomers of olefins, C=N double bonds, or other types of double bonds may be present in the compounds described herein, and all such stable isomers are included in the present disclosure. Specifically, cis and trans geometric isomers of the compounds of the present disclosure may also exist and may be isolated as a mixture of isomers or as separated isomeric forms.
[00197] Compounds of the present disclosure also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Examples prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, amide - imidic acid pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, such as, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
3.3.4 Salts and other forms
[00198] In some embodiments of the method of treating a cancer, the compounds described herein are present in a salt form. In some embodiments, the compounds are provided in the form of pharmaceutically acceptable salts.
[00199] Compounds included in the present compositions that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids. The acids that can be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds are those that form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions. Salt forms of interest of the compounds described herein include, but not limited to, chloride, acetate, 2,2,2-trifluoroacetate (TFA), and formate salts.
[00200] Compounds containing an amine functional group or a nitrogen-containing heteroaryl group may be basic in nature and may react with a variety of inorganic and organic acids to form the corresponding pharmaceutically acceptable salts. Inorganic acids commonly employed to form such salts include hydrochloric, and related inorganic acids. Organic acids commonly employed to form such salts include formic acid, and related organic acids. Such pharmaceutically acceptable salts thus include chloride, and related salts.
[00201] Other examples of salts include anions of the compounds of the present disclosure compounded with a suitable cation such as N+, NH4 +, and NW4 + (where W can be a C1-C8 alkyl group), and the like. For therapeutic use, salts of the compounds of the present disclosure can be pharmaceutically acceptable. However, salts of acids and bases that are non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound.
[00202] Compounds included in the present compositions that are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations. Examples of such salts include alkali metal or alkaline earth metal salts and, particularly, calcium, magnesium, sodium, lithium, zinc, potassium, and iron salts.
[00203] Compounds that include a basic or acidic moiety can also form pharmaceutically acceptable salts with various amino acids. The compounds of the disclosure can contain both acidic and basic groups; for example, one amino and one carboxylic acid group. In such a case, the compound can exist as an acid addition salt, a zwitterion, or a base salt.
[00204] The compounds described herein can be present in various forms including crystalline, powder and amorphous forms of those compounds, pharmaceutically acceptable salts, including, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrates), conformational polymorphs, and amorphous forms of the compounds, as well as mixtures thereof.
[00205] The compounds described herein may exist as solvates, especially hydrates, and unless otherwise specified, all such solvates and hydrates are intended. Hydrates may form during manufacture of the compounds or compositions comprising the compounds, or hydrates may form over time due to the hygroscopic nature of the compounds. Compounds of the present technology may exist as organic solvates as well, including dimethylformamide (DMF), ether, and alcohol solvates, among others. The identification and preparation of any particular solvate is within the skill of the ordinary artisan of synthetic organic or medicinal chemistry.
[00206] In some embodiments, the compounds described herein are present in a solvate form. In some embodiments, the compounds described herein are present in a hydrate form when the solvent component of the solvate is water.
3.3.5 Prodrugs
[00207] In some embodiments of the method of treating a cancer, the compounds described herein are present in a prodrug form. Any convenient prodrug forms of the subject compounds can be prepared, for example, according to the strategies and methods described by Rautio et al. ("Prodrugs: design and clinical applications", Nature Reviews Drug Discovery 7, 255-270 (February 2008)).
3.3.6 Compound Synthesis
[00208] Compounds of the present disclosure may be synthesized according to standard methods known in the art [see, e.g. Morrison and Boyd in "Organic Chemistry", 6th edition, Prentice Hall (1992)]. Some compounds and/or intermediates of the present disclosure may be commercially available, known in the literature, or readily obtainable by those skilled in the art using standard procedures. Some compounds of the present disclosure may be synthesized using schemes, examples, or intermediates described herein. Where the synthesis of a compound, intermediate or variant thereof is not fully described, those skilled in the art can recognize that the reaction time, number of equivalents of reagents and/or temperature may be modified from reactions described herein to prepare compounds presented or intermediates or variants thereof and that different work-up and/or purification techniques may be necessary or desirable to prepare such compounds, intermediates, or variants.
[00209] Synthesized compounds may be validated for proper structure by methods known to those skilled in the art, for example by nuclear magnetic resonance (NMR) spectroscopy and/or mass spectrometry.
[00210] In various embodiments, the compound as described herein is represented by the structure of one of the compounds in Table 1. The present disclosure is meant to encompass a compound of any one of Table 1, or a salt, a single stereoisomer, a mixture of stereoisomers and/or an isotopically labelled form thereof.
3.4 Pharmaceutical Compositions
[00211] In some embodiments of the method of treating a cancer, the compounds of the present disclosure that are administered to the subject may be composed in a pharmaceutical composition.
[00212] Accordingly, the present disclosure provides a method of treating a cancer using a pharmaceutical composition comprising at least one compound described herein, or a pharmaceutically acceptable salt thereof, or a prodrug thereof, and at least one pharmaceutically acceptable excipient.
[00213] The term "pharmaceutical composition" is meant to encompass a composition suitable for administration to a subject, such as a mammal, especially a human. In general, a "pharmaceutical composition" is sterile, and preferably free of contaminants that are capable of eliciting an undesirable response within the subject (i.e., the compound(s) in the pharmaceutical composition is pharmaceutical grade). Pharmaceutical compositions can be designed for administration to subjects or patients in need thereof via a number of different routes of administration including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, intracheal, intramuscular, subcutaneous, and the like.
[00214] The terms "pharmaceutically acceptable excipient," "pharmaceutically acceptable diluent," "pharmaceutically acceptable carrier," and "pharmaceutically acceptable adjuvant" are used interchangeably and refer to any ingredient other than the inventive compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound, or any other convenient pharmaceutically acceptable carriers, excipients, diluent, adjuvant or additives) and having the properties of being substantially nontoxic and non-inflammatory in a patient. The phrase "pharmaceutically acceptable excipient" includes both one and more than one such excipient, diluent, carrier, and/or adjuvant. Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, dispensing, or dispersing agents, sweeteners, and waters of hydration. In some embodiments, the pharmaceutical composition comprises a compound as described herein, a pharmaceutically acceptable salt thereof, or a prodrug thereof in a therapeutically effective amount.
[00215] The pharmaceutical composition may be formulated according to any convenient methods, and may be prepared in various forms for oral administration such as tablets, pills, powders, capsules, syrups, emulsions and microemulsions, or in forms for non-oral administration such as eye drops or preparations for intramuscular, intravenous or subcutaneous administration. In one example, the pharmaceutical composition may be administered through the eyes in the form of eyedrops. In one example, the pharmaceutical composition may be an ophthalmic composition, such as an eye drop composition.
[00216] In some embodiments, the pharmaceutical compositions are formulated for oral delivery. In a case wherein the pharmaceutical composition is prepared in a form for oral administration, examples of additives or carriers which may be used include cellulose, calcium silicate, corn starch, lactose, sucrose, dextrose, calcium phosphate, magnesium stearate, stearic acid, stearate, talc, surfactant, suspending agent, emulsifier and diluent. Examples of additives or carriers which may be used in a case wherein the pharmaceutical composition of the present disclosure is prepared as an injection include water, saline solution, glucose aqueous solution, pseudosugar solution, alcohol, glycol, ether (e.g., polyethylene glycol 400), oil, fatty acid, fatty acid ester, glyceride, surfactants, suspending agents and emulsifiers.
[00217] In some embodiments, the pharmaceutical compositions are formulated for parenteral administration to a subject in need thereof. In some parenteral embodiments, the pharmaceutical compositions are formulated for intravenous administration to a subject in need thereof. In some parenteral embodiments, the pharmaceutical compositions are formulated for subcutaneous administration to a subject in need thereof.
3.5 Definitions
[00218] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure pertains.
[00219] It is understood that the definitions provided herein are not intended to be mutually exclusive. Accordingly, some chemical moieties may fall within the definition of more than one term.
[00220] As used herein, the symbol "
Figure PCTKR2023008331-appb-img-000146
" refers to a covalent bond comprising a single or a double bond.
[00221] The term "Cx-Cy" when used in conjunction with a chemical moiety, such as alkyl, alkenyl, or alkynyl is meant to include groups that contain from x to y carbons in the chain. For example, the term "C1-C6 alkyl" refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups that contain from 1 to 6 carbons. In some embodiments, the term "(Cx-Cy)alkylene" refers to a substituted or unsubstituted alkylene chain with from x to y carbons in the alkylene chain. For example "(Cx-Cy)alkylene may be selected from methylene, ethylene, propylene, butylene, pentylene, and hexylene, any one of which is optionally substituted.
[00222] The term "alkyl" refers to an unbranched or branched saturated hydrocarbon chain. In some embodiments, alkyl as used herein has 1 to 20 carbon atoms ((C1-C20)alkyl), 1 to 10 carbon atoms ((C1-C10)alkyl), 1 to 8 carbon atoms ((C1-C8)alkyl), 1 to 6 carbon atoms ((C1-C6)alkyl), or 1 to 5 carbon atoms ((C1-C5)alkyl). Examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, 2-pentyl, isopentyl, neopentyl, n-hexyl, 2-hexyl, 3-hexyl, and 3-methyl pentyl. When an alkyl residue having a specific number of carbons is named, all geometric isomers having that number of carbons may be encompassed. For example, "butyl" can include n-butyl, sec-butyl, isobutyl and t-butyl, and ''propyl" can include n-propyl and isopropyl. Unless stated otherwise specifically in the specification, an alkyl chain is optionally substituted by one or more substituents such as those substituents described herein.
[00223] The term "alkylene" refers to a straight divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation, and preferably having from 1 to 20 carbon atoms ((C1-C20)alkylene), 1 to 10 carbon atoms ((C1-C10)alkylene), 1 to 6 carbon atoms ((C1-C6)alkylene), or 1 to 5 carbon atoms ((C1-C5)alkylene). Examples include, but are not limited to, methylene, ethylene, propylene, butylene, and the like. The alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group are through the terminal carbons respectively. Unless stated otherwise specifically in the specification, an alkylene chain is optionally substituted by one or more substituents such as those substituents described herein. Examples include, methylene (-CH2-), ethylene (-CH2CH2-), propylene (-CH2CH2CH2-), 2-methylpropylene (-CH2-CH(CH3) -CH2-), hexylene (-(CH2)6-) and the like.
[00224] The term "alkenyl" refers to an aliphatic hydrocarbon group containing at least one carbon-carbon double bond including straight-chain, branched-chain and cyclic alkenyl groups. In some embodiments, the alkenyl group has 2-10 carbon atoms (a C2-10 alkenyl). In another embodiment, the alkenyl group has 2-4 carbon atoms in the chain (a C2-4 alkenyl). Exemplary alkenyl groups include, but are not limited to, ethenyl, propenyl, n-butenyl, i-butenyl, 3-methylbut-2-enyl, n-pentenyl, heptenyl, octenyl, cyclohexyl-butenyl and decenyl. An alkylalkenyl is an alkyl group as defined herein bonded to an alkenyl group as defined herein. The alkenyl group can be unsubstituted or substituted through available carbon atoms with one or more groups defined hereinabove for alkyl
[00225] The term "alkynyl" refers to straight or branched monovalent hydrocarbyl groups having from 2 to 6 carbon atoms and preferably 2 to 3 carbon atoms and having at least 1 and preferably from 1 to 2 sites of acetylenic (C≡C-) unsaturation. Examples of such alkynyl groups include, but are not limited to, acetylenyl (C≡CH), and propargyl (CH2C≡CH).
[00226] The term "aryl" refers to a monocyclic or polycyclic group having at least one hydrocarbon aromatic ring, wherein all of the ring atoms of the at least one hydrocarbon aromatic ring is carbon.  Aryl may include groups with a single aromatic ring (e.g., phenyl) and multiple fused aromatic rings (e.g., naphthyl, anthryl).  Aryl may further include groups with one or more aromatic hydrocarbon rings fused to one or more non-aromatic hydrocarbon rings (e.g., fluorenyl; 2,3-dihydro-1H-indene; 1,2,3,4-tetrahydronaphthalene). In certain embodiments, aryl includes groups with an aromatic hydrocarbon ring fused to a non-aromatic ring, wherein the non-aromatic ring comprises at least one ring heteroatom independently selected from the group consisting of N, O, and S.  For example, in some embodiments, aryl includes groups with a phenyl ring fused to a non-aromatic ring, wherein the non-aromatic ring comprises at least one ring heteroatom independently selected from the group consisting of N, O, and S (e.g., chromane; thiochromane; 2,3-dihydrobenzofuran; indoline).  In some embodiments, aryl as used herein has from 6 to 14 carbon atoms ((C6-C14)aryl), or 6 to 10 carbon atoms ((C6-C10)aryl).  Where the aryl includes fused rings, the aryl may connect to one or more substituents or moieties of the formulae described herein through any atom of the fused ring for which valency permits.
[00227] The term "cycloalkyl" refers to a monocyclic or polycyclic saturated hydrocarbon.  In some embodiments, cycloalkyl has 3 to 20 carbon atoms ((C3-C20)cycloalkyl), 3 to 8 carbon atoms ((C3-C8)cycloalkyl), 3 to 6 carbon atoms ((C3-C6)cycloalkyl), or 3 to 5 carbon atoms ((C3-C5)cycloalkyl). In some embodiments, cycloalkyl has 3 to 8 carbon atoms having single or multiple cyclic rings including fused, bridged, and spiro ring systems. Examples of suitable cycloalkyl groups include, but are not limited to, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl, octahydropentalenyl, octahydro-1H-indene, decahydronaphthalene, cubane, bicyclo[3.1.0]hexane, and bicyclo[1.1.1]pentane, and the like.
[00228] The term "carbocycle" refers to a saturated, unsaturated or aromatic ring in which each atom of the ring is carbon. Carbocycle includes 3- to 10-membered monocyclic rings, 6- to 12-membered bicyclic rings, and 6- to 12-membered bridged rings. Each ring of a bicyclic carbocycle may be selected from saturated, unsaturated, and aromatic rings. In an exemplary embodiment, an aromatic ring, e.g., phenyl, may be fused to a saturated or unsaturated ring, e.g., cyclohexane, cyclopentane, or cyclohexene. A bicyclic carbocycle includes any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits. A bicyclic carbocycle includes any combination of ring sizes such as 4-5 fused ring systems, 5-5 fused ring systems, 5-6 fused ring systems, 6-6 fused ring systems, 5-7 fused ring systems, 6-7 fused ring systems, 5-8 fused ring systems, and 6-8 fused ring systems. Exemplary carbocycles include cyclopentyl, cyclohexyl, cyclohexenyl, adamantyl, phenyl, indanyl, and naphthyl.
[00229] The term "heterocycle" refers to a saturated, unsaturated or aromatic ring comprising one or more heteroatoms. Exemplary heteroatoms include N, O, Si, P, B, and S atoms. Heterocycles include 3- to 10-membered monocyclic rings, 6- to 12-membered bicyclic rings, and 6- to 12-membered bridged rings. A bicyclic heterocycle includes any combination of saturated, unsaturated and aromatic bicyclic rings, as valence permits. In an exemplary embodiment, an aromatic ring, e.g., pyridyl, may be fused to a saturated or unsaturated ring, e.g., cyclohexane, cyclopentane, morpholine, piperidine or cyclohexene. A bicyclic heterocycle includes any combination of ring sizes such as 4-5 fused ring systems, 5-5 fused ring systems, 5-6 fused ring systems, 6-6 fused ring systems, 5-7 fused ring systems, 6-7 fused ring systems, 5-8 fused ring systems, and 6-8 fused ring systems.
[00230] The term "heteroaryl" refers to an aromatic group of from 4 to 10 carbon atoms and 1 to 4 heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur within the ring. Such heteroaryl groups can have a single ring (i.e., pyridinyl or furyl) or multiple condensed rings (i.e., indolizinyl or benzothienyl) wherein the condensed rings may or may not be aromatic and/or contain a heteroatom provided that the point of attachment is through an atom of the aromatic heteroaryl group. In one embodiment, the nitrogen and/or the sulfur ring atom(s) of the heteroaryl group are optionally oxidized to provide for the N oxide (N→O), sulfinyl, or sulfonyl moieties. Preferred heteroaryls include 5 or 6 membered heteroaryls such as pyridinyl, pyrrolyl, indolyl, thiophenyl, and furanyl.
[00231] The term "heteroalkyl" refers to an alkyl substituent in which one or more of the carbon atoms and any attached hydrogen atoms are independently replaced with the same or different heteroatomic group. For example, 1, 2, or 3 carbon atoms may be independently replaced with the same or different heteroatomic substituent.
[00232] The term "substituted" refers to moieties having substituents replacing a hydrogen on one or more carbons or substitutable heteroatoms, e.g., an NH or NH2 of a compound. It will be understood that "substitution" or "substituted with" includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound. For example, stable compounds include, but is not limited to, compounds which do not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. In certain embodiments, substituted refers to moieties having substituents replacing two hydrogen atoms on the same carbon atom, such as substituting the two hydrogen atoms on a single carbon with an oxo, imino or thioxo group. As used herein, the term "substituted" is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds.
[00233] It will be understood by those skilled in the art that substituents can themselves be substituted, if appropriate. Unless specifically stated as "unsubstituted," references to chemical moieties herein are understood to include substituted variants. For example, reference to a "heteroaryl" group or moiety implicitly includes both substituted and unsubstituted variants, unless specified otherwise.
[00234] The phrase "optionally substituted" refers to when a non-hydrogen substituent may or may not be present on a given atom, and, thus, the description includes structures wherein a non-hydrogen substituent is present and structures wherein a non-hydrogen substituent is not present.
[00235] In some embodiments, substituents may include any substituents described herein, for example: halogen, hydroxy, oxo (=O), thioxo (=S), cyano (-CN), nitro (-NO2), imino (=N-H), oximo (=N-OH), hydrazino (=N-NH2), -Rb-ORa, -Rb-OC(O)-Ra, -Rb-OC(O)-ORa, -Rb-OC(O)-N(Ra)2, -Rb-N(Ra)2, -Rb-C(O)Ra, -Rb-C(O)ORa, -Rb-C(O)N(Ra)2, -Rb-O-Rc-C(O)N(Ra)2, -Rb-N(Ra)C(O)ORa, -Rb-N(Ra)C(O)Ra, -RbN (Ra)S(O)tRa (where t is 1 or 2), -Rb-S(O)tRa  (where t is 1 or 2), -Rb-S(O)tORa (where t is 1 or 2), and -Rb-S(O)tN(Ra)2  (where t is 1 or 2). In another exemplary embodiment, substituents include alkyl, alkenyl, alkynyl, aryl, aralkyl, aralkenyl, aralkynyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl, and heteroarylalkyl, any of which may be optionally substituted by alkyl, alkenyl, alkynyl, halogen, haloalkyl, haloalkenyl, haloalkynyl, oxo, thioxo, cyano, nitro, imino, oximo, hydrazine, -RbORa, -Rb-OC(O)-Ra, -Rb-OC(O)-ORa, -Rb-OC(O)-N(Ra)2, -Rb-N(Ra)2, -Rb-C(O)Ra, -Rb-C(O)ORa, -Rb-C(O)N(Ra)2, -Rb-O-Rc-C(O)N(Ra)2, -Rb-N(Ra)C(O)ORa, -Rb-N(Ra)C(O)Ra, -Rb-N (Ra)S(O)tRa  (where t is 1 or 2), -Rb-S(O)tRa  (where t is 1 or 2), -Rb-S(O)tORa  (where t is 1 or 2) and -Rb-S(O)tN(Ra)2  (where t is 1 or 2); and wherein each Ra, Rb, and Rc  are independently selected from hydrogen, alkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl, and heteroarylalkyl; and wherein each Ra, Rb, and Rc, valence permitting, may be optionally substituted with alkyl, alkenyl, alkynyl, halogen, haloalkyl, haloalkenyl, haloalkynyl, oxo, thioxo, cyano, nitro, imino, oximo, hydrazine, -RbORa, -Rb-OC(O)-Ra, -Rb-OC(O)-ORa, -Rb-OC(O)-N(Ra)2, -Rb-N(Ra)2, -Rb-C(O)Ra, -Rb-C(O)ORa, -Rb-C(O)N(Ra)2, -Rb-O-Rc-C(O)N(Ra)2, -Rb-N(Ra)C(O)ORa, -Rb-N(Ra)C(O)Ra, -Rb-N (Ra)S(O)tRa  (where t is 1 or 2), -Rb-S(O)tRa  (where t is 1 or 2), -Rb-S(O)tORa  (where t is 1 or 2) and -Rb-S(O)tN(Ra)2  (where t is 1 or 2).
[00236] The term "isomers" refers to two or more compounds comprising the same numbers and types of atoms, groups or components, but with different structural arrangement and connectivity of the atoms.
[00237] The term "tautomer" refers to one of two or more structural isomers which readily convert from one isomeric form to another and which exist in equilibrium.
[00238] A "stereoisomer" refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable.  The present disclosure contemplates various stereoisomers and mixtures thereof and includes "enantiomers", which refers to two stereoisomers whose molecules are non-superimposeable mirror images of one another.
[00239] Individual enantiomers and diastereomers of compounds of the present disclosure can be prepared synthetically from commercially available starting materials that contain asymmetric or stereogenic centers, or by preparation of racemic mixtures followed by resolution methods well known to those of ordinary skill in the art. These methods of resolution are exemplified by (1) attachment of a mixture of enantiomers to a chiral auxiliary, separation of the resulting mixture of diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary, (2) salt formation employing an optically active resolving agent, (3) direct separation of the mixture of optical enantiomers on chiral liquid chromatographic columns, or (4) kinetic resolution using stereoselective chemical or enzymatic reagents. Racemic mixtures also can be resolved into their respective enantiomers by well-known methods, such as chiral-phase gas chromatography or crystallizing the compound in a chiral solvent. Stereoselective syntheses, a chemical or enzymatic reaction in which a single reactant forms an unequal mixture of stereoisomers during the creation of a new stereocenter or during the transformation of a pre-existing one, are well known in the art. Stereoselective syntheses encompass both enantio- and diastereoselective transformations. See, for example, Carreira and Kvaerno, Classics in Stereoselective Synthesis, Wiley-VCH: Weinheim, 2009.
[00240] Geometric isomers, resulting from the arrangement of substituents around a carbon-carbon double bond or arrangement of substituents around a cycloalkyl or heterocyclic ring, can also exist in the compounds of the present disclosure. The symbol = denotes a bond that may be a single, double or triple bond as described herein. Substituents around a carbon-carbon double bond are designated as being in the "Z" or "E" configuration, where the terms "Z" and "E" are used in accordance with IUPAC standards. Unless otherwise specified, structures depicting double bonds encompass both the "E" and "Z" isomers.
[00241] Substituents around a carbon-carbon double bond alternatively can be referred to as "cis" or "trans," where "cis" represents substituents on the same side of the double bond and "trans" represents substituent on opposite sides of the double bond. The arrangement of substituents around a carbocyclic ring can also be designated as "cis" or "trans." The term "cis" represents substituents on the same side of the plane of the ring and the term "trans" represents substituents on opposite sides of the plane of the ring. Mixtures of compound wherein the substituents are disposed on both the same and opposite sides of the plane of the ring are designated "cis/trans."
[00242] Singular articles such as "a," "an" and "the" and similar referents in the context of describing the elements are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, including the upper and lower bounds of the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (i.e., "such as") provided herein, is intended merely to better illuminate the embodiments and does not pose a limitation on the scope of the claims unless otherwise stated.
[00243] In some embodiments, where the use of the term "about" is before a quantitative value, the present disclosure also includes the specific quantitative value itself, unless specifically stated otherwise. As used herein, the term "about" refers to a ±10% variation from the nominal value unless otherwise indicated or inferred. Where a percentage is provided with respect to an amount of a component or material in a composition, the percentage should be understood to be a percentage based on weight, unless otherwise stated or understood from the context.
[00244] Where a molecular weight is provided and not an absolute value, for example, of a polymer, then the molecular weight should be understood to be an average molecule weight, unless otherwise stated or understood from the context.
[00245] It should be understood that the order of steps or order for performing certain actions is immaterial so long as the present disclosure remain operable. Moreover, two or more steps or actions can be conducted simultaneously.
[00246] A dash ("-") symbol that is not between two letters or symbols refers to a point of bonding or attachment for a substituent. For example, -NH2 is attached through the nitrogen atom.
[00247] The terms "individual," "host," and "subject," are used interchangeably, and refer to an animal, including, but not limited to, human and non-human primates, including simians and humans; rodents, including rats and mice; bovines; equines; ovines; felines; canines; and the like. "Mammal" means a member or members of any mammalian species, and includes, by way of example, canines, felines, equines, bovines, ovines, rodentia, etc. and primates, i.e., non-human primates, and humans. Non-human animal models, i.e., mammals, non-human primates, murines, lagomorpha, etc. may be used for experimental investigations.
[00248] "Patient" refers to a human subject.
[00249] The terms "active agent," "drug," "pharmacologically active agent," and "active pharmaceutical ingredient" are used interchangeably to refer to a compound or composition which, when administered to a subject, induces a desired pharmacologic or physiologic effect by local or systemic action or both.
[00250] The phrase "therapeutically effective amount" refers to the amount of a compound that, when administered to a mammal or other subject for treating a disease, condition, or disorder, is sufficient to affect such treatment for the disease, condition, or disorder. The "therapeutically effective amount" will vary depending on the compound, the disease and its severity and the age, weight, etc., of the subject to be treated.
[00251] Generally, reference to or depiction of a certain element such as hydrogen or H is meant to include all isotopes of that element. For example, if an R group is defined to include hydrogen or H, it also includes deuterium and tritium. Compounds comprising radioisotopes such as tritium, 14C, 32P and 35S are thus within the scope of the present technology. Procedures for inserting such labels into the compounds of the present technology will be readily apparent to those skilled in the art based on the disclosure herein.
3. EXAMPLES
[00252] The following examples are offered to illustrate the present disclosure and are not to be construed in any way as limiting the scope of the present technology. Any methods that are functionally equivalent are within the scope of the present technology. Various modifications of the present technology in addition to those described herein will become apparent to those skilled in the art from the foregoing description and accompanying figures. Such modifications fall within the scope of the appended claims.
[00253] Unless otherwise stated, all temperatures are in degrees Celsius. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperatures, etc.), but some experimental errors and deviation should be allowed for.
[00254] In the examples below, if an abbreviation is not defined, it has its generally accepted meaning.
aq. = aqueous
LC-MS = liquid chromatography-mass spectrometry
MS = mass spectrometry
THF = tetrahydrofuran
NaHCO3 = sodium bicarbonate
Cs2CO3 = cesium carbonate
NaH = sodium hydride
o/n = overnight
HATU = 1-[bis(dimethylamino)methylene]-1H-1,2,3-
triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate
r.t. = room temperature
LAH = lithium aluminum hydride
DCM = dichloromethane
DMF = dimethylformamide
DMSO = dimethyl sulfoxide
DIEA = diisopropylethylamine
equiv. = equivalent
EtOAc or EA = ethyl acetate
EtOH = ethanol
EDCI = 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide
g = gram
h = hours
HCl = hydrochloric acid
HPLC = high-performance liquid chromatography
HOAc = acetic acid
HOBT = hydroxybenzotriazole
M = molar
MeOH = methanol
mg = milligrams
mL = milliliters
mmol = millimols
mp = melting point
m/z = mass to charge ratio
NaCl = sodium chloride
Na2CO3 = sodium carbonate
NMR = nuclear magnetic resonance
NaOH = sodium hydroxide
Na2SO4 = sodium sulfate
ppm = parts per million
TFA = trifluoroacetic acid
TLC = thin layer chromatography
TsOH = p-Toluenesulfonic acid
UV = ultraviolet
wt % = weight percent
μM = micromolar
General Synthetic Methods
[00255] The compounds of this disclosure were prepared according to the methods summarized below, which are described in greater detail in International Application No. PCT/IB2021/000856, filed December 10, 2021, the disclosure of which is herein incorporated by reference.
[00256] Final compounds were confirmed by HPLC/MS analysis and determined to be >90% pure by weight. 1H and 13C NMR spectra were recorded in CDCl3 (residual internal standard CHCl3 = δ 7.26), DMSO-d6 (residual internal standard CD3SOCD2H = δ 2.50), methanol-d4 (residual internal standard CD2HOD = δ 3.31), or acetone-d6 (residual internal standard CD3COCD2H = δ 2.05). The chemical shifts (δ) reported are given in parts per million (ppm) and the coupling constants (J) are in Hertz (Hz). The spin multiplicities are reported as s = singlet, bs = broad singlet, bm = broad multiplet, d = doublet, t = triplet, q = quartet, p = pentuplet, dd = doublet of doublet, ddd = doublet of doublet of doublet, dt = doublet of triplet, td = triplet of doublet, tt = triplet of triplet, and m = multiplet.
[00257] HPLC-MS analysis was carried out with gradient elution. Medium pressure liquid chromatography (MPLC) was performed with silica gel columns in both the normal phase and reverse phase.
Example 1 - Synthesis of Intermediate Compounds
[00258] Described herein are details of the synthesis and characterization of several exemplary intermediate compounds or synthons that can be used to prepare a variety of final compounds of this disclosure. It is understood that the synthetic methods and intermediate compounds described, in combination with generally available starting materials, may readily be adapted to synthesize a variety of compounds of formula (I)-(XIc), including any of the compounds of Table 1.
Synthesis of Intermediate Compound 51
Figure PCTKR2023008331-appb-img-000147
Step 1:
[00259] To a solution of 5-(2-ethoxy-5-nitrophenyl)-1-methyl-3-propyl-1,6-dihydro-7H-pyrazolo[4,3-d] pyrimidin-7-one (500 mg, 1.40 mmol) in EtOH (10 mL) and H2O (2 mL) was added Fe (391 mg, 7.00 mmol) and NH4Cl (748 mg, 13.99 mmol), the reaction mixture was stirred at 60 °C for 1 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography to afford 5-(5-amino-2-ethoxyphenyl)-1-methyl-3-propyl-1,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one (430 mg, 93.88% yield) as a yellow solid. MS: m/z = 328.2 (M+1, ESI+).
Step 2:
[00260] To a solution of 5-(5-amino-2-ethoxyphenyl)-1-methyl-3-propyl-1,6-dihydro-7H-pyrazolo[4,3-d] pyrimidin-7-one (430 mg, 1.31 mmol) and 2-bromo-2-methylpropanoic acid (658 mg, 3.94 mmol) in i-PrOH (10 mL) was added TEA (399 mg, 3.94 mmol), the reaction mixture was stirred at 80 °C for 16 h. The reaction mixture was poured into water (100 mL) and extracted with EA (30 mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford 2-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)amino)-2-methylpropanoic acid (400 mg, 73.65% yield) as a white solid. MS: m/z = 414.3 (M+1, ESI+).
Step 3:
[00261] To a solution of 2-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin -5-yl)phenyl)amino)-2-methylpropanoic acid (370 mg, 894.87 umol) in MeOH (10 mL) was added SOCl2 (1 g, 8.41 mmol), the reaction mixture was stirred at 60 °C for 16 h. The reaction mixture was concentrated and the residue was poured into water (30 mL) and extracted with EA (10mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford intermediate compound 51, methyl 2-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)amino)-2-methylpropanoate (150 mg, 39.27% yield) as a white solid. MS: m/z = 428.4 (M+1, ESI+).
Synthesis of Intermediate Compound 52
Figure PCTKR2023008331-appb-img-000148
Step 1:
[00262] A mixture of 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl) benzenesulfonyl chloride (5 g, 12.17 mmol) in H2O (50 mL) was stirred at 70 °C for 3 h. The reaction mixture was filtered and the filter cake was dried under reduced pressure to afford 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonic acid (4.45 g, 93.18% yield) as a white solid. MS: m/z =393.1 (M+1, ESI+).
Step 2:
[00263] A mixture of 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl) benzenesulfonic acid (4.45 g, 11.34 mmol) was added to fuming nitric acid (20 mL) and DCM (20 mL) at -50 °C in portions, the reaction mixture was allowed to warm to room temperature and stirred for 2 h. The reaction mixture was poured into ice water (100 mL) and extracted with DCM (20 mLx3), the combined organic layers were washed with water (100 mL) and brine (100 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford 2-(2-ethoxy-5-nitrophenyl)-5-methyl-7-propylimidazo[5,1-f] [1,2,4] triazin -4(3H)-one (1.05 g, 25.91% yield) as a yellow solid. MS: m/z =358.2 (M+1, ESI+).
Step 3:
[00264] To a solution of 2-(2-ethoxy-5-nitrophenyl)-5-methyl-7-propylimidazo[5,1-f][1,2,4]triazin-4(3H)-one (1.10 g, 3.08 mmol) in EtOH (10 mL) and H2O (2 mL) was added Fe (516 mg, 9.23 mmol) and NH4Cl (540 mg, 9.23 mmol), the reaction mixture was stirred at 70 °C for 3 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography to afford 2-(5-amino-2-ethoxyphenyl)-5-methyl-7-propyl imidazo[5,1-f][1,2,4]triazin-4(3H)-one (980 mg, 97.25% yield) as a yellow solid. MS: m/z = 328.2 (M+1, ESI+).
Step 4:
[00265] To a solution of 2-(5-amino-2-ethoxyphenyl)-5-methyl-7-propylimidazo[5,1-f][1,2,4]triazin-4(3H)-one (980 mg, 2.99 mmol) and 2-bromo-2-methylpropanoic acid (1 g, 5.99 mmol) in i-PrOH (10 mL) was added TEA (909 mg, 8.98 mmol), the reaction mixture was stirred at 80 °C for 16 h. The reaction mixture was poured into water (100 mL) and extracted with EtOAc (30mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford 2-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino)-2-methylpropanoic acid (430 mg, 34.74% yield) as a yellow solid. MS: m/z = 414.2 (M+1, ESI+).
Step 5:
[00266] To a solution of 2-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino)-2-methylpropanoic acid (300 mg, 725.57 umol) in MeOH (10 mL) was added SOCl2 (259 mg, 2.18 mmol), the reaction mixture was stirred at 70 °C for 16 h. The reaction mixture was concentrated and the residue was poured into water (30 mL) and extracted with EA (10mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford intermediate compound 52, methyl 2-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino)-2-methyl propanoate (278 mg, 89.63% yield) as a yellow solid. MS: m/z = 428.3 (M+1, ESI+).
Synthesis of Intermediate Compound 53
Figure PCTKR2023008331-appb-img-000149
Step 1:
[00267] To a solution of 5-(5-amino-2-ethoxyphenyl)-1-methyl-3-propyl-1,6-dihydro-7H-pyrazolo[4,3-d] pyrimidin-7-one (2.5 g, 7.64 mmol) and 1-bromocyclobutane-1-carboxylic acid (2.05 g, 11.45 mmol) in i-PrOH (30 mL) was added TEA (2.32 g, 22.91 mmol), the reaction mixture was stirred at 80 °C for 16 h. The reaction mixture was concentrated under reduced pressure and the residue was purified by column chromatography to afford 1-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)amino)cyclobutane-1-carboxylic acid (1.5 g, 46.17% yield) as a yellow solid. MS: m/z = 426.2 (M+1, ESI+).
Step 2:
[00268] To a solution of 1-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin -5-yl)phenyl)amino)cyclobutane-1-carboxylic acid (1.5 g, 3.53 mmol) in MeOH (30 mL) was added SOCl2 (2.10 g, 17.63 mmol), the reaction mixture was stirred at 80 °C for 16 h. The reaction mixture was concentrated and the residue was poured into water (30 mL) and extracted with EA (10mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford intermediate compound 53 methyl 1-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)amino) cyclobutane-1-carboxylate (1 g, 64.51% yield) as a yellow solid. MS: m/z = 440.2 (M+1, ESI+).
Synthesis of Intermediate Compound 54
Figure PCTKR2023008331-appb-img-000150
Step 1:
[00269] To a solution of 2-(5-amino-2-ethoxyphenyl)-5-methyl-7-propylimidazo[5,1-f][1,2,4]triazin-4(3H)-one (3.2 g, 9.77 mmol) and 1-bromocyclobutane-1-carboxylic acid (3.5 g, 19.55 mmol) in i-PrOH (40 mL) was added TEA (2.97 g, 29.32 mmol), the reaction mixture was stirred at 90 °C for 16 h. The reaction mixture was concentrated under reduced pressure and the residue was purified by column chromatography to afford 1-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo [5,1-f][1,2,4]triazin-2-yl)phenyl)amino)cyclobutane-1-carboxylic acid (2 g, 48.09% yield) as a white solid. MS: m/z = 426.1 (M+1, ESI+).
Step 2:
[00270] To a solution of 1-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino)cyclobutane-1-carboxylic acid (2 g, 4.70 mmol) in MeOH (30 mL) was added SOCl2 (1.68 g, 14.10 mmol), the reaction mixture was stirred at 80 °C for 16 h. The reaction mixture was concentrated and the residue was poured into water (30 mL) and extracted with EA (10mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford intermediate compound 54, methyl 1-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino)cyclobutane-1-carboxylate (1 g, 48.40% yield) as a white solid. MS: m/z = 440.2(M+1, ESI+).
Synthesis of Intermediate Compound 55
Figure PCTKR2023008331-appb-img-000151
Step 1:
[00271] To a solution of 2-(5-amino-2-ethoxyphenyl)-5-methyl-7-propylimidazo[5,1-f][1,2,4]triazin-4(3H)-one (500 mg, 1.53 mmol) and 2-bromoacetic acid (255 mg, 1.83 mmol) in i-PrOH (10 mL) was added TEA (464 mg, 4.58 mmol), the reaction mixture was stirred at 80 °C for 16 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure, the residue was purified by column chromatography to afford (4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)glycine (400 mg, 67.95% yield) as a yellow solid. MS: m/z = 386.1 (M+1, ESI+).
Step 2:
[00272] To a solution of (4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)glycine (400 mg, 1.04 mmol) in MeOH (10 mL) was added SOCl2 (617.36 mg, 5.19 mmol), the reaction mixture was stirred at 80 °C for 16 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure, the residue was purified by column chromatography to afford intermediate compound 55, methyl (4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f] [1,2,4]triazin-2-yl)phenyl)glycinate (350 mg, 84.43% yield) as a yellow oil. MS: m/z = 400.1 (M+1, ESI+).
Synthesis of Intermdiate Compound 56
Figure PCTKR2023008331-appb-img-000152
Step 1:
[00273] To a solution of 2-(5-amino-2-ethoxyphenyl)-5-methyl-7-propylimidazo[5,1-f][1,2,4]triazin-4(3H)-one (500 mg, 1.53 mmol) and 2-bromopropanoic acid (280 mg, 1.83 mmol) in i-PrOH (10 mL) was added TEA (464 mg, 4.58 mmol), the reaction mixture was stirred at 80 °C for 16 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure, the residue was purified by column chromatography to afford (4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)alanine (360 mg, 59.01% yield) as a yellow solid. MS: m/z = 400.1 (M+1, ESI+).
Step 2:
[00274] To a solution of (4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)alanine (360 mg, 901 umol) in MeOH (8 mL) was added SOCl2 (536 mg, 4.51 mmol), the reaction mixture was stirred at 80 °C for 16 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure, the residue was purified by column chromatography to afford intermediate compound 56, methyl (4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f] [1,2,4]triazin-2-yl)phenyl)alaninate (310 mg, 83.19% yield) as a yellow oil. MS: m/z = 414.1 (M+1, ESI+).
Synthesis of Intermediate Compound 57
Figure PCTKR2023008331-appb-img-000153
[00275] To a solution of 2-(5-amino-2-ethoxyphenyl)-5-methyl-7-propylimidazo[5,1-f][1,2,4]triazin-4(3H)-one (1 g, 3.05 mmol) and dihydrofuran-3(2H)-one (525.93 mg, 6.11 mmol) in dioxane (30 mL) was added TMSCN (453.60 mg, 4.58 mmol) and ZnCl2 (83.08 mg, 610.91 umol), the reaction mixture was stirred at 50 °C for 16 h. The reaction mixture was cooled to room temperature and poured into water (80 mL), extracted with EA (30mLx3), washed by brine (80 mL), dried over Na2SO4 and concentrated. The residue was purified by column chromatography to afford intermediate compound 57, 3-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino) tetrahydrofuran-3-carbonitrile (1.09 g, 84.46% yield) as a yellow solid. MS: m/z =423.2 (M+1, ESI+).
Synthesis of Intermediate Compound 58
Figure PCTKR2023008331-appb-img-000154
Step 1:
[00276] A mixture of cyclopentanecarboxylic acid (10 g, 86.12 mmol) and red P (1.33 g, 43.06 mmol) at 0 °C was added Br2 (27.56 g, 172.24 mmol) in portions, after that, the reaction mixture was stirred at 60 °C for 6 h. The reaction mixture was diluted with EA (200 mL) and washed with brine (100 mLx2), the organic layer was dried over Na2SO4 and concentrated under reduced pressure, the residue was purified by column chromatography to afford 1-bromocyclopentane-1-carboxylic acid (12 g, crude) as a yellow oil. 1H NMR (400 MHz, DMSO-d6) δ 12.91 (s, 1H), 2.31-2.22 (m, 2H), 2.21-2.11 (m, 2H), 1.89-1.83 (m, 2H), 1.78-1.72 (m, 2H).
Step 2:
[00277] To a solution of 2-(5-amino-2-ethoxyphenyl)-5-methyl-7-propylimidazo[5,1-f][1,2,4]triazin-4(3H)-one (800 mg, 2.44 mmol) and 1-bromocyclopentane-1-carboxylic acid (943.43 mg, 4.89 mmol) in i-PrOH (15 mL) was added TEA (741.82 mg, 7.33 mmol), the reaction mixture was stirred at 80 °C for 16 h. The reaction mixture was cooled to room temperature and poured into water (100 mL) and extracted with EA (40mLx3), washed by brine (100 mL), dried over Na2SO4 and concentrated. The residue was purified by column chromatography to afford 1-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino)cyclopentane-1-carboxylic acid (336 mg, 31.29% yield) as a yellow solid. MS: m/z =440.1 (M+1, ESI+).
Step 3:
[00278] To a solution of 1-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino)cyclopentane-1-carboxylic acid (336 mg, 764.49 umol) in MeOH (8 mL) was added SOCl2 (2 mL) in portions, the reaction mixture was stirred at 70 °C for 16 h. The reaction mixture was concentrated and the residue was poured into water (30 mL) and extracted with EA (10mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford intermediate compound 58, methyl 1-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino) cyclopentane-1-carboxylate (140 mg, 40.38% yield) as a yellow solid. MS: m/z =454.2 (M+1, ESI+).
Synthesis of Intermediate Compound 59
Figure PCTKR2023008331-appb-img-000155
Step 1:
[00279] To a solution of 2-fluoro-3-(trifluoromethyl)aniline (25 g, 139.6 mmol) in DMF (200 mL) was added NBS (27.3 g, 153.5 mmol) in portions, the resulting mixture was stirred at 25°C for 16 h. The reaction mixture was poured into water (800 mL) and extracted with EA (200 mLx3), the combined organic layers were washed with water (800 mL) and brine (800 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford 4-bromo-2-fluoro-3-(trifluoromethyl)aniline (27.5 g, 76.6% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 7.32 (dd, 1H), 6.94 (t, 1H), 5.81 (br s, 2H).
Step 2:
[00280] To a solution of 4-bromo-2-fluoro-3-(trifluoromethyl)aniline (27.5 g, 107 mmol) in THF (200 mL) was added Ac2O (30 g, 294 mmol), the resulting mixture was stirred at 60 °C for 16 h. The reaction mixture was cooled to room temperature and concentrated under reduce pressure. The residue was purified by column chromatography to afford N-(4-bromo-2-fluoro-3-(trifluoromethyl)phenyl) acetamide (30 g, 93.4% yield) as a yellow solid. MS: m/z = 301.8 (M+1, ESI+).
Step 3:
[00281] To a solution of N-(4-bromo-2-fluoro-3-(trifluoromethyl)phenyl)acetamide (30 g, 100 mmol) in DMF (100 mL) was added CuCN (17.8 g, 200 mmol), the resulting mixture was stirred at 150 °C for 16 h. The reaction mixture cooled to room temperature and poured into water (800 mL) and extracted with EA (200 mLx3), the combined organic layers were washed with water (800 mL) and brine (800 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford N-(4-cyano-2-fluoro-3-(trifluoromethyl)phenyl)acetamide (19 g, 77% yield) as a yellow solid. MS: m/z = 246.9 (M+1, ESI+).
Step 4:
[00282] To a solution of N-(4-cyano-2-fluoro-3-(trifluoromethyl)phenyl)acetamide (19 g, 77 mmol) in EtOH (100 ml) was added 5 M HCl (100 mL), the resulting mixture was stirred at 80 °C for 2 h. The reaction mixture was cooled to room temperature and concentrated under reduce pressure. The residue was purified by column chromatography to afford 4-amino-3-fluoro-2-(trifluoromethyl) benzonitrile (12.8 g, 80% yield) as a yellow solid. MS: m/z = 205.1 (M+1, ESI+).
Step 5:
[00283] To a solution of triphosgene (20.3 g, 176.5 mmol) in H2O (200 mL) was added 4-amino-3-fluoro-2-(trifluoromethyl)benzonitrile (7.2 g, 35.3 mmol) in portions, the resulting mixture was stirred at 25 oC for 16 h. The reaction mixture was poured into ice water (200 mL) and extracted with DCM (100 mLx3), the combined organic layers were washed with water (200 mL) and brine (200 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford intermediate compound 59, 3-fluoro-4-isothiocyanato-2-(trifluoromethyl)benzonitrile (5.5 g, 62.8% yield) as a colorless oil.
Example 2 - Synthesis of Final Compounds
[00284] Described herein are details of the synthesis and characterization of several exemplary compounds of this disclosure. It is understood that the synthetic methods and materials described may readily be adapted to synthesize a variety of compounds of formula (I)-(XIc), including any of the compounds of Table 1.
Synthesis of Compound 1
Figure PCTKR2023008331-appb-img-000156
[00285] To a solution of 4-amino-2-(trifluoromethyl)benzonitrile (68 mg, 365 umol) in DMF (10 mL) at 0 °C was added NaH (29 mg, 730 umol, 60% purity), the reaction mixture was stirred at 0 °C for 0.5 h, then 4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl) benzenesulfonyl chloride (100 mg, 243 umol) was added to the above solution. The resulting mixture was stirred at 25 °C for 3 h. The reaction mixture was poured into water (100 mL) and extracted with EA (30 mLx3), the combined organic layers were washed with brine, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by Prep-HPLC to afford compound 1, N-(4-cyano-3-(trifluoromethyl)phenyl)-4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)benzenesulfonamide, (75 mg, 54.97% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 12.12 (s, 1H), 8.07-7.97 (m, 3H), 7.62-7.53 (m, 2H), 7.34 (d, 1H), 4.19-4.14 (m, 5H), 2.76 (t, 2H), 1.77-1.68 (m, 2H), 1.31 (t, 3H), 0.92 (t, 3H); MS: m/z = 561.4 (M+1, ESI+); HRMS: 561.1527.
Synthesis of Compound 5
Figure PCTKR2023008331-appb-img-000157
[00286] To a solution of 5-amino-3-(trifluoromethyl)picolinonitrile (170 mg, 0.9 mmol) in DMF (5 mL) at 0 °C was added NaH (55 mg, 1.3 mmol, 60% purity), the reaction mixture was stirred at 0 °C for 0.5 h, then 4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl) benzenesulfonyl chloride (300 mg, 0.9 mmol) was added to the above solution. The resulting mixture was stirred at 25 °C for 3 h. The reaction mixture was poured into water (100 mL) and extracted with EA (30 mLx3), the combined organic layers were washed with brine, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by Prep-HPLC to afford compound 5, N-(6-cyano-5-(trifluoromethyl)pyridin-3-yl)-4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)benzenesulfonamide (60 mg, 11.6% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 12.15 (s, 1H), 8.65 (s, 1H), 8.07-8.00 (m, 2H), 7.90 (s, 1H), 7.34 (d, 1H), 4.19-4.16 (m, 5H), 2.77 (t, 2H), 1.77-1.71 (m, 2H), 1.32 (t, 3H), 0.94 (t, 3H); MS: m/z = 562.0 (M+1, ESI+); HRMS: 562.1478.
Synthesis of Compound 6
Figure PCTKR2023008331-appb-img-000158
[00287] To a solution of 5-amino-3-(trifluoromethyl)picolinonitrile (170 mg, 0.9 mmol) in DMF (10 mL) at 0 °C was added NaH (55 mg, 1.3 mmol, 60% purity), the reaction mixture was stirred at 0 °C for 0.5 h, then 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl) benzenesulfonyl chloride (300 mg, 0.9 mmol) was added to the above solution. The resulting mixture was stirred at 25 °C for 3 h. The reaction mixture was poured into water (100 mL) and extracted with EA (30 mLx3), the combined organic layers were washed with brine, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by Prep-HPLC to afford compound 6, N-(6-cyano-5-(trifluoromethyl)pyridin-3-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide (50 mg, 10.0% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.68 (s, 1H), 8.68 (d, 1H), 8.08-8.05 (m, 2H), 7.90 (s, 1H), 7.37 (d, 1H), 4.19 (q, 2H), 2.83 (t, 2H), 2.49 (s, 2H), 1.77-1.71 (m, 2H), 1.31 (t, 3H), 0.93 (t, 3H); MS: m/z = 562.0 (M+1, ESI+); HRMS: 562.1478.
Synthesis of Compound 7
Figure PCTKR2023008331-appb-img-000159
[00288] To a solution of 4-amino-2-(trifluoromethyl)benzonitrile (68 mg, 365 umol) in DMF (10 mL) at 0 °C was added NaH (29 mg, 730 umol, 60% purity), the reaction mixture was stirred at 0 °C for 0.5 h, then 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl) benzenesulfonyl chloride (100 mg, 243 umol) was added to the above solution. The resulting mixture was stirred at 25 °C for 3 h. The reaction mixture was poured into water (100 mL) and extracted with EA (30 mLx3), the combined organic layers were washed with brine, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by Prep-HPLC to afford compound 7 N-(4-cyano-3-(trifluoromethyl)phenyl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide (60 mg, 43.98% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.77 (s, 1H), 8.10-8.06 (m, 3H), 7.71-7.63 (m, 2H), 7.41 (d, 1H), 4.21 (q, 2H), 2.87 (t, 2H), 2.52 (s, 3H), 1.80-1.74 (m, 2H), 1.33 (t, 3H), 0.95 (t, 3H); MS: m/z = 561.3 (M+1, ESI+); HRMS: 561.1525.
Synthesis of Compound 8
Figure PCTKR2023008331-appb-img-000160
[00289] To a solution of 4-amino-2-chlorobenzonitrile (84 mg, 548 umol) in DMF (10 mL) at 0 oC was added NaH (44 mg, 1.10 mmol, 60% purity), the reaction mixture was stirred at 0 °C for 0.5 h, then 4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl) benzenesulfonyl chloride (150 mg, 365 umol) was added to the above solution. The resulting mixture was stirred at 25 °C for 3 h. The reaction mixture was poured into water (100 mL) and extracted with EA (30 mLx3), the combined organic layers were washed with brine, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by Prep-HPLC to afford compound 8, N-(3-chloro-4-cyanophenyl)-4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo [4,3-d]pyrimidin-5-yl)benzenesulfonamide (90 mg, 46.78% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 12.14 (s, 1H), 11.33 (s, 1H), 8.04-7.97 (m, 2H), 7.85 (d, 1H), 7.37-7.33 (m, 2H), 7.26 (dd, 1H), 4.21-4.16 (m, 5H), 2.78 (t, 2H), 1.80-1.70 (m, 2H), 1.32 (t, 3H), 0.95 (t, 3H); MS: m/z = 527.3 (M+1, ESI+); HRMS: 527.1263.
Synthesis of Compound 9
Figure PCTKR2023008331-appb-img-000161
[00290] To a solution of 4-amino-2-chlorobenzonitrile (84 mg, 548 umol) in DMF (10 mL) at 0 oC was added NaH (44 mg, 1.10 mmol, 60% purity), the reaction mixture was stirred at 0 °C for 0.5 h, then 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl) benzenesulfonyl chloride (150 mg, 365 umol) was added to the above solution. The resulting mixture was stirred at 25 °C for 3 h. The reaction mixture was poured into water (100 mL) and extracted with EA (30 mLx3), the combined organic layers were washed with brine, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by Prep-HPLC to afford compound 9, N-(3-chloro-4-cyanophenyl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f] [1,2,4]triazin-2-yl)benzenesulfonamide (85 mg, 44.18% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.64 (s, 1H), 8.03-8.00 (m, 2H), 7.85 (d, 1H), 7.38-7.35 (m, 2H), 7.26 (d, 1H), 4.18 (q, 2H), 2.83 (t, 2H), 2.48 (s, 3H), 1.77-1.72 (m, 2H), 1.31 (t, 3H), 0.94 (t, 3H); MS: m/z = 527.3 (M+1, ESI+); HRMS: 527.1262.
Synthesis of Compound 10
Figure PCTKR2023008331-appb-img-000162
Step 1:
[00291] To a solution of 1-(tert-butyl) 2-methyl (2R,4R)-4-hydroxypyrrolidine-1,2-dicarboxylate (12.5 g, 50.96 mmol) in DCM (300 mL) was added Dess-martin (32.42 g, 76.45 mmol), the reaction mixture was stirred at 25 °C for 3 h. The reaction mixture was poured into water (500 mL) and extracted with DCM (100 mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford 1-(tert-butyl) 2-methyl (R)-4-oxopyrrolidine-1,2-dicarboxylate (10 g, 80.66% yield) as a white solid. MS: m/z =144.2 (M-100+1, ESI+).
Step 2:
[00292] To a solution of 1-(tert-butyl) 2-methyl (R)-4-oxopyrrolidine-1,2-dicarboxylate (10 g, 41.11 mmol) in DCM (300 mL) was added phenylmethanamine (5.29 g, 49.33 mmol), the reaction mixture was stirred at 25 °C for 0.5 h. Then NaBH(OAc)3 (13.07 g, 61.66 mmol) was added to the above reaction mixture, the resulting mixture was stirred at 25 °C for another 3 h. The reaction mixture was poured into water (500 mL) and extracted with DCM (100 mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford 1-(tert-butyl) 2-methyl (2R)-4-(benzylamino)pyrrolidine-1,2-dicarboxylate (10.5 g, 76.38% yield) as a yellow oil. MS: m/z =335.1 (M+1, ESI+).
Step 3:
[00293] To a solution of 1-(tert-butyl) 2-methyl (2R)-4-(benzylamino)pyrrolidine-1,2-dicarboxylate (7.5 g, 22.46 mmol) in MeOH (80 mL) was added Pd/C (0.8 g), the reaction mixture was stirred at 25 °C under H2 for 6 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to afford 1-(tert-butyl) 2-methyl (2R)-4-aminopyrrolidine-1,2-dicarboxylate (4.0 g, 76.38% yield) as a yellow oil. MS: m/z = 245.2 (M+1, ESI+).
Step 4:
[00294] To a solution of 1-(tert-butyl) 2-methyl (2R)-4-aminopyrrolidine-1,2-dicarboxylate (4 g, 16.37 mmol) and CbzCl (4.90 g, 19.65 mmol) in DCM (100 mL) was added TEA (6.35 g, 49.12 mmol), the reaction mixture was stirred at 25 °C for 5 h. The reaction mixture was poured into water (150 mL) and extracted with DCM (50 mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford 1-(tert-butyl) 2-methyl (2R)-4-(((benzyloxy)carbonyl)amino)pyrrolidine-1,2-dicarboxylate (5.4 g, 87.15% yield) as a yellow oil. MS: m/z = 279.1 (M-100+1, ESI+).
Step 5:
[00295] To a solution of 1-(tert-butyl) 2-methyl (2R)-4-(((benzyloxy)carbonyl)amino)pyrrolidine-1,2-dicarboxylate (5.4 g, 14.27 mmol) in THF (80 mL) at 0 °C was added LAH (813 mg, 21.40 mmol) in portions, then the reaction mixture was stirred at 25 °C for 3 h. The reaction mixture was poured into ice water (150 mL) slowly and extracted with EA (100 mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford tert-butyl (2R)-4-(((benzyloxy)carbonyl)amino)-2-(hydroxyl methyl)pyrrolidine-1-carboxylate (2.9 g, 58.00% yield) as a colorless oil. MS: m/z = 251.1 (M-100+1, ESI+).
Step 6:
[00296] To a solution of tert-butyl (2R)-4-(((benzyloxy)carbonyl)amino)-2-(hydroxymethyl)pyrrolidine-1-carboxylate (4.5 g, 12.84 mmol) in DCM (30 mL) was added Dess-martin (5.45 g, 12.84 mmol), the reaction mixture was stirred at 25 °C for 2 h. The reaction mixture was poured into water (100 mL) and extracted with DCM (30 mLx3). The combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford tert-butyl (2R)-4-(((benzyloxy)carbonyl)amino)-2-formylpyrrolidine-1-carboxylate (2.9 g, 64.82% yield) as a colorless oil. MS: m/z =249.2 (M-100+1, ESI+).
Step 7:
[00297] To a solution of tert-butyl (2R)-4-(((benzyloxy)carbonyl)amino)-2-formylpyrrolidine-1-carboxylate (2.9 g, 8.32 mmol) in THF (20 mL) was added TMSCF3 (1.54 g, 10.82 mmol) and CsF (127 mg, 832 umol), the reaction mixture was stirred at 25 °C for 2 h. The reaction mixture was poured into water (100 mL) and extracted with EA (30 mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford tert-butyl (2R)-4-(((benzyloxy)carbonyl)amino)-2-((S)-2,2,2-trifluoro-1-((trimethylsilyl)oxy)ethyl)pyrrolidine-1-carboxylate (2.35 g, 57.55% yield) as a yellow oil. MS: m/z = 391.2 (M-100+1, ESI+).
Step 8:
[00298] To a solution of tert-butyl (2R)-4-(((benzyloxy)carbonyl)amino)-2-((S)-2,2,2-trifluoro-1-((trimethylsilyl)oxy)ethyl)pyrrolidine-1-carboxylate (2.35 g, 4.79 mmol) in DCM (5 mL) was added 3M HCl in EA (5 mL), the reaction mixture was stirred at 25 °C for 2 h. The reaction mixture was poured into aq. NaHCO3 (40 mL) and extracted with EA (10 mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford benzyl ((5R)-5-((S)-2,2,2-trifluoro-1-hydroxyethyl)pyrrolidin-3-yl)carbamate (1.50 g, 4.71 mmol, 98.38% yield) as a brown oil. MS: m/z = 319.0 (M+1, ESI+).
Step 9:
[00299] To a solution of benzyl ((5R)-5-((S)-2,2,2-trifluoro-1-hydroxyethyl)pyrrolidin-3-yl)carbamate (1.50 g, 4.71 mmol) and 4-bromo-2-(trifluoromethyl)benzonitrile (1.77 g, 7.07 mmol) in toluene (25 mL) was added K2CO3 (1.95 g, 14.14 mmol) and RuPhosPd-G2 (183.02 mg, 235.63 umol), the reaction mixture was stirred at 110 °C under N2 for 16 h. The reaction mixture was cooled to room temperature and poured into water (100 mL) and extracted with EA (40 mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford benzyl ((5R)-1-(4-cyano-3-(trifluoromethyl) phenyl)-5-((S)-2,2,2-trifluoro-1-hydroxyethyl)pyrrolidin-3-yl)carbamate (869 mg, 37.83% yield) as a yellow oil. MS: m/z = 488.1 (M+1, ESI+).
Step 10:
[00300] To a solution of benzyl ((5R)-1-(4-cyano-3-(trifluoromethyl)phenyl)-5-((S)-2,2,2-trifluoro-1-hydroxyethyl)pyrrolidin-3-yl)carbamate (869 mg, 1.78 mmol) in MeCN (10 mL) was added TMSI (1.07 g, 5.35 mmol), the reaction mixture was stirred at 25 °C for 1 h. The reaction mixture was poured into water (100 mL) and extracted with EA (40 mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford 4-((2R)-4-amino-2-((S)-2,2,2-trifluoro-1-hydroxyethyl)pyrrolidin-1-yl)-2-(trifluoro methyl)benzonitrile hydrochloride (70 mg, 11.11% yield) as an off-white solid. 1H NMR (400 MHz, MeOD) δ 7.81 (d, 1H), 7.03 (d, 1H), 6.97 (dd, 1H), 4.59 (d, 1H), 4.38 (q, 1H), 4.12 (t, 1H), 3.88-3.83 (m, 1H), 3.72 (dd, 1H), 2.73-2.65 (m, 1H), 2.48 (d, 1H); MS: m/z = 354.0 (M+1, ESI+).
Step 11:
[00301] To a solution of 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonyl chloride (70 mg, 170 umol) in THF (5 mL) was added 4-((2R)-4-amino-2-((S)-2,2,2-trifluoro-1-hydroxyethyl)pyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile hydrochloride (60 mg, 170 umol) and TEA (51.50 mg, 508.91 umol), the reaction mixture was stirred at 25 °C for 5 h. The reaction mixture was poured into water (20 mL) and extracted with EA (10 mLx3), the combined organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 10, N-((5R)-1-(4-cyano-3-(trifluoromethyl)phenyl)-5-(2,2,2-trifluoro-1-hydroxyethyl)pyrrolidin-3-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydro imidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide (15 mg, 12.15% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 7.98-7.95 (m, 2H), 7.83 (d, 1H), 7.36 (d, 1H), 6.86-6.83 (m, 2H), 4.34-4.26 (m, 2H), 4.19 (q, 2H), 3.74-3.64 (m, 2H), 3.17 (t, 1H), 2.78 (t, 2H), 2.47 (s, 3H), 2.23-2.18 (m, 1H), 2.12-2.07 (m, 1H), 1.75-1.65 (m, 2H), 1.32 (t, 3H), 0.88 (t, 3H); MS: m/z = 728.3 (M+1, ESI+).
Synthesis of Compound 11
Figure PCTKR2023008331-appb-img-000163
[00302] To a solution of 4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)benzenesulfonyl chloride (250 mg,608.46 umol) and (S)-4-(1-(2-aminopropyl)-1H-pyrazol-3-yl)-2-chlorobenzonitrile (190 mg, 730.15 umol) in MeCN (10 mL) was added K2CO3 (168 mg,1.22 mmol), the reaction mixture was stirred at 25 oC for 16 h. Filtered and concentrated under reduced pressure, the residue was purified by prep-HPLC to afford compound 11, (S)-N-(1-(3-(3-chloro-4-cyanophenyl)-1H-pyrazol-1-yl)propan-2-yl)-4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d] pyrimidin-5-yl)benzenesulfonamide (102 mg, 26.35% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.84 (s, 1H), 7.94-7.90 (m, 3H), 7.82-7.76 (m, 2H), 7.68 (s, 2H), 7.11 (d, 1H), 6.76 (s, 1H), 4.17-4.06 (m, 7H), 3.71 (s, 1H), 2.78 (s, 2H), 1.76-1.75 (m, 2H), 1.34 (s, 3H), 1.07 (s, 3H), 0.95 (s, 3H); MS: m/z = 635.2 (M+1, ESI+); HRMS: 635.1952.
Synthesis of Compound 13
Figure PCTKR2023008331-appb-img-000164
Step 1:
[00303] To a solution of triphosgene (8.34 g, 72.53 mmol) in H2O (50 mL) was added 4-amino-2-(trifluoromethyl)benzonitrile (4.5 g, 24.18 mmol) in portions, the reaction mixture was stirred at 25 oC for 5 h. The reaction mixture was poured into ice water (100 mL) and extracted with DCM (40 mLx3), the combined organic layers were washed with water (100 mL) and brine (100 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford 4-isothiocyanato-2-(trifluoromethyl)benzonitrile (1.02 g, 18.49% yield) as a white solid.
Step 2:
[00304] To a solution of compound 51, methyl 2-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d] pyrimidin-5-yl)phenyl)amino)-2-methylpropanoate (150 mg, 350.88 umol) and 4-isothiocyanato-2-(trifluoromethyl)benzonitrile (160 mg, 702 umol) in DMSO (10 mL) was added isopropyl acetate (3 g, 29.37 mmol), the reaction mixture was stirred at 85 °C for 16 h. The reaction mixture was poured into water (150mL) and extracted with DCM (20 mLx3), the combined organic layers were washed with water (100 mL) and brine (100 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 13, 4-(3-(4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile (110 mg, 50.27% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 12.11 (s, 1H), 8.40 (d, 1H), 8.31 (s, 1H), 8.10 (dd, 1H), 7.62 (d, 1H), 7.50 (dd, 1H), 7.34 (d, 1H), 4.20 (q, 2H), 4.16 (s, 3H), 2.77 (t, 2H), 1.77-1.71 (m, 2H), 1.54 (s, 6H), 1.36 (t, 3H), 0.93 (t, 3H); MS: m/z = 624.2 (M+1, ESI+); HRMS: 624.1998.
Synthesis of Compound 18
Figure PCTKR2023008331-appb-img-000165
Step 1:
[00305] To a solution of triphosgene (8.34 g, 72.53 mmol) in H2O (50 mL) was added 4-amino-2-(trifluoromethyl)benzonitrile (4.5 g, 24.18 mmol) in portions, the reaction mixture was stirred at 25 oC for 5 h. The reaction mixture was poured into ice water (100 mL) and extracted with DCM (40 mLx3), the combined organic layers were washed with water (100 mL) and brine (100 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford 4-isothiocyanato-2-(trifluoromethyl)benzonitrile (1.02 g, 18.49% yield) as a white solid.
Step 2:
[00306] To a solution of compound 52, methyl 2-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4] triazin-2-yl)phenyl)amino)-2-methylpropanoate (278 mg, 650.30 umol) and 4-isothiocyanato-2-(trifluoromethyl)benzonitrile (178 mg, 780.36 umol) in DMSO (15 mL) was added isopropyl acetate (1.33 g, 13 mmol), the reaction mixture was stirred at 85 °C for 16 h. The reaction mixture was poured into water (150mL) and extracted with DCM (20 mLx3), the combined organic layers were washed with water (100 mL) and brine (100 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 18, 4-(3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile (100 mg, 24.66% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.63 (s, 1H), 8.39 (d, 1H), 8.30 (s, 1H), 8.08 (dd, 1H), 7.55-7.53 (m, 2H), 7.34 (d, 1H), 4.18 (q, 2H), 2.82 (t, 2H), 2.48 (s, 3H), 1.76-1.70 (m, 2H), 1.53 (s, 6H), 1.34 (t, 3H), 0.91 (t, 3H);MS: m/z = 624.1 (M+1, ESI+); HRMS: 624.1996.
Synthesis of Compound 19
Figure PCTKR2023008331-appb-img-000166
Step 1:
[00307] To a solution of triphosgene (6.14 g, 53.44 mmol) in H2O (15 mL) was added 5-amino-3-(trifluoromethyl)picolinonitrile (2.00 g, 10.69 mmol) in portions, the reaction mixture was stirred at 25 oC for 5 h. The reaction mixture was poured into ice water (100 mL) and extracted with DCM (40 mLx3), the combined organic layers were washed with water (100 mL) and brine (100 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford 5-isothiocyanato-3-(trifluoromethyl)picolinonitrile (1.0 g, 40.81% yield) as a yellow oil.
Step 2:
[00308] To a solution of compound 51, methyl 2-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4] triazin-2-yl)phenyl)amino)-2-methylpropanoate (350 mg, 818.72 umol) and 5-isothiocyanato-3-(trifluoromethyl)picolinonitrile (225 mg, 982.47 umol) in DMSO (5 mL) was added isopropyl acetate (1.67 g, 16.37 mmol), the reaction mixture was stirred at 85 °C for 3 h. The reaction mixture was poured into water (50 mL) and extracted with DCM (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 19, 5-(3-(4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-3-(trifluoromethyl)picolinonitrile (158 mg, 30.90% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 12.11 (s, 1H), 9.25 (d, 1H), 8.83 (d, 1H), 7.63 (d, 1H), 7.50 (dd, 1H), 7.35 (d, 1H), 4.20 (q, 2H), 4.16 (s, 3H), 2.77 (t, 2H), 1.77-1.72 (m, 2H), 1.56 (s, 6H), 1.37 (t, 3H), 0.93 (t, 3H); MS: m/z = 625.2 (M+1, ESI+); HRMS: 625.1951.
Synthesis of Compound 20
Figure PCTKR2023008331-appb-img-000167
[00309] To a solution of compound 52, methyl 2-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4] triazin-2-yl)phenyl)amino)-2-methylpropanoate (200 mg, 467.84 umol) and 5-isothiocyanato-3-(trifluoromethyl)picolinonitrile (161 mg, 702 umol) in NMP (10 mL) was added isopropyl acetate (5 mL), the reaction mixture was stirred at 115 °C for 16 h. The reaction mixture was poured into water (150 mL) and extracted with DCM (30 mLx3), the combined organic layers were washed with water (100 mL) and brine (100 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 20, 5-(3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-3-(trifluoromethyl)picolinonitrile (105 mg, 35.93% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.64 (s, 1H), 9.25 (s, 1H), 8.82 (s, 1H), 7.54-7.53 (m, 2H), 7.35 (d, 1H), 4.18 (q, 2H), 2.82 (t, 2H), 2.48 (s, 3H), 1.73 (q, 2H), 1.55 (s, 6H), 1.34 (t, 3H), 0.93-0.91 (t, 3H);MS: m/z = 625.2 (M+1, ESI+); HRMS: 625.1944.
Synthesis of Compound 21
Figure PCTKR2023008331-appb-img-000168
[00310] To a solution of compound 53, methyl 1-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d] pyrimidin-5-yl)phenyl)amino)cyclobutane-1-carboxylate (280 mg, 637 umol) and 4-isothiocyanato-2-(trifluoromethyl)benzonitrile (175 mg, 764 umol) in DMSO (5 mL) was added isopropyl acetate (1.30 g, 12.74 mmol) , the reaction mixture was stirred at 85 °C for 3 h. The reaction mixture was poured into water (50 mL) and extracted with DCM (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 21, 4-(5-(4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl)phenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7-yl)-2-(trifluoromethyl)benzonitrile (80 mg, 19.75% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 12.13 (s, 1H), 8.38 (d, 1H), 8.25 (s, 1H), 8.06 (dd, 1H), 7.65 (d, 1H), 7.52 (dd, 1H), 7.36 (d, 1H), 4.21 (q, 2H), 4.12 (s, 3H), 2.76 (t, 2H), 2.67-2.61 (m, 2H), 2.45-2.38 (m, 2H), 2.00-1.94 (m, 1H), 1.76-1.69 (m, 2H), 1.58-1.53 (m, 1H), 1.36 (t, 3H), 0.92 (t, 3H); MS: m/z = 636.2 (M+1, ESI+); HRMS: 636.1998.
Synthesis of Compound 22
Figure PCTKR2023008331-appb-img-000169
[00311] To a solution of compound 54, methyl 1-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino)cyclobutane-1-carboxylate (500 mg, 1.14 mmol) and 4-isothiocyanato-2-(trifluoromethyl)benzonitrile (260 mg, 1.14 mmol) in DMSO (20 mL) was added isopropyl acetate (10 mL), the reaction mixture was stirred at 85 °C for 16 h. The reaction mixture was poured into water (200 mL) and extracted with DCM (40 mLx3), the combined organic layers were washed with water (200 mL) and brine (200 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 22, 4-(5-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4] octan-7-yl)-2-(trifluoromethyl)benzonitrile (100 mg, 13.83% yield) as a white solid. 1H NMR (400 MHz, CDCl3) δ 9.90 (s, 1H), 8.09 (d, 1H), 8.00-7.98 (m, 2H), 7.88 (dd, 1H), 7.48 (dd, 1H), 7.27 (d, 1H), 4.36 (q, 2H), 2.97 (t, 2H), 2.76-2.71 (m, 2H), 2.64-2.54 (m, 5H), 2.31-2.24 (m, 1H), 1.90-1.81 (m, 2H), 1.78-1.70 (m, 1H), 1.62 (t, 3H), 0.99 (t, 3H); MS: m/z = 636.1 (M+1, ESI+); HRMS: 636.1992.
Synthesis of Compound 23
Figure PCTKR2023008331-appb-img-000170
[00312] To a solution of methyl 1-((4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d] pyrimidin-5-yl)phenyl)amino)cyclobutane-1-carboxylate (700 mg, 1.59 mmol) and 5-isothiocyanato-3-(trifluoromethyl)picolinonitrile (438 mg, 1.91 mmol) in DMSO (10 mL) was added isopropyl acetate (3.25 g, 31.85 mmol)), the reaction mixture was stirred at 85 °C for 3 h. The reaction mixture was poured into water (100 mL) and extracted with DCM (20 mLx3), the combined organic layers were washed with water (100 mL) and brine (100 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford 5-(5-(4-ethoxy-3-(1-methyl-7-oxo-3-propyl-6,7-dihydro-1H-pyrazolo[4,3-d]pyrimidin-5-yl) phenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7-yl)-3-(trifluoromethyl)picolinonitrile (200 mg, 19.72% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 12.15 (s, 1H), 9.22 (d, 1H), 8.76 (d, 1H), 7.67 (d, 1H), 7.53 (dd, 1H), 7.38 (d, 1H), 4.25-4.14 (m, 5H), 2.76 (t, 2H), 2.68-2.64 (m, 2H), 2.54-2.46 (m, 2H), 2.04-1.97 (m, 1H), 1.79-1.69 (m, 2H), 1.61-1.56 (m, 1H), 1.38 (t, 3H), 0.92 (t, 3H); MS: m/z = 637.0 (M+1, ESI+); HRMS: 637.1949.
Synthesis of Compound 24
Figure PCTKR2023008331-appb-img-000171
[00313] To a solution of compound 54, methyl 1-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino)cyclobutane-1-carboxylate (500 mg, 1.14 mmol) and 5-isothiocyanato-3-(trifluoromethyl)picolinonitrile (261 mg, 1.14 mmol) in DMSO (5 mL) was added isopropyl acetate (5 mL), the reaction mixture was stirred at 85 °C for 16 h. The reaction mixture was poured into water (50 mL) and extracted with DCM (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 24, 5-(5-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7-yl)-3-(trifluoromethyl)picolinonitrile (95 mg, 13.12% yield) as a white solid. 1H NMR (400 MHz, CDCl3) δ 9.89 (s, 1H), 9.13 (d, 1H), 8.41 (d, 1H), 8.07 (d, 1H), 7.48 (dd, 1H), 7.27 (s, 1H), 4.36 (dd, 2H), 2.96 (t, 2H), 2.79-2.73 (m, 2H), 2.64-2.57 (m, 5H), 2.33-2.25 (m, 1H), 1.90-1.72 (m, 3H), 1.62 (t, 3H), 0.99 (t, 3H); MS: m/z = 637.1 (M+1, ESI+); HRMS: 637.1945.
Synthesis of Compound 25
Figure PCTKR2023008331-appb-img-000172
[00314] To a solution of compound 55, methyl (4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4] triazin-2-yl)phenyl)glycinate (350 mg, 876.22 umol) and 4-isothiocyanato-2-(trifluoromethyl) benzonitrile (240 mg, 1.05 mmol) in toluene (10 mL) was added AcOH (526 mg, 8.76 mmol), the reaction mixture was stirred at 120 °C for 1 h. The reaction mixture was poured into water (50 mL) and extracted with DCM (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 25, 4-(3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-5-oxo-2-thioxoimidazolidin-1-yl)-2-(trifluoromethyl) benzonitrile (86 mg, 16.48% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.60 (s, 1H), 8.39 (d, 1H), 8.17 (s, 1H), 8.00 (dd, 1H), 7.88 (d, 1H), 7.82 (dd, 1H), 7.30 (d, 1H), 4.88 (s, 2H), 4.15 (q, 2H), 2.84 (t, 2H), 2.48 (s, 3H), 1.77-1.71 (m, 2H), 1.34 (t, 3H), 0.92 (t, 3H);MS: m/z = 596.3 (M+1, ESI+); HRMS: 596.1689.
Synthesis of Compound 26
Figure PCTKR2023008331-appb-img-000173
[00315] To a solution of compound 56 methyl (4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4] triazin-2-yl)phenyl)alaninate (310 mg, 749.75 umol) and 4-isothiocyanato-2-(trifluoromethyl) benzonitrile (205 mg, 900 umol) in toluene (10 mL) was added AcOH (450 mg, 7.50 mmol), the reaction mixture was stirred at 120 °C for 1 h. The reaction mixture was poured into water (50 mL) and extracted with DCM (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 26, 4-(3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4-methyl-5-oxo-2-thioxoimidazolidin-1-yl)-2-(trifluoromethyl)benzonitrile (80 mg, 17.50% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.63 (s, 1H), 8.39 (d, 1H), 8.23 (d, 1H), 8.04 (dd, 1H), 7.75-7.69 (m, 2H), 7.32 (d, 1H), 5.10 (q, 2H), 4.20-4.14 (m, 2H), 2.84 (t, 2H), 2.49 (s, 3H), 1.77-1.72 (m, 2H), 1.43 (d, 2H), 1.34 (t, 3H), 0.92 (t, 3H); MS: m/z = 610.2 (M+1, ESI+); HRMS: 610.1841.
Synthesis of Compound 27
Figure PCTKR2023008331-appb-img-000174
[00316] To a solution of 5-amino-2-(trifluoromethyl)benzonitrile (341.13 mg, 1.83 mmol) in DCM (15 mL) was added triphosgene (272 mg, 916 umol) at -10°C in portions and stirred at this temperature for 0.5 h, then TEA (232 mg, 2.29 mmol) was added stirred for another 15 min. After that, a solution of 2-(5-amino-2-ethoxyphenyl)-5-methyl-7-propylimidazo[5,1-f][1,2,4]triazin-4(3H)-one (500 mg, 1.53 mmol) in DCM (15 mL) was added to the above mixture and stirred at -10°C for 1 h. The resulting mixture was evaporated and the residue was purified by prep-HPLC to afford compound 27, 1-(4-cyano-3-(trifluoromethyl)phenyl)-3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo [5,1-f][1,2,4]triazin-2-yl)phenyl)urea (85 mg, 10.32% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.50 (s, 1H), 10.08 (s, 1H), 9.42 (s, 1H), 8.22 (d, 1H), 8.00 (d, 1H), 7.77 (dd, 1H), 7.65 (d, 1H), 7.60 (dd, 1H), 7.11 (d, 1H), 4.08 (dd, 2H), 2.83 (t, 2H), 2.48 (s, 3H), 1.77-1.71 (m, 2H), 1.29 (t, 3H), 0.92 (t, 3H);MS: m/z = 540.1 (M+1, ESI+); HRMS: 540.1967.
Synthesis of Compound 28
Figure PCTKR2023008331-appb-img-000175
[00317] A mixture of 2-(5-amino-2-ethoxyphenyl)-5-methyl-7-propylimidazo[5,1-f][1,2,4]triazin-4(3H)-one (350 mg, 1.07 mmol) and 4-isothiocyanato-2-(trifluoromethyl)benzonitrile (293 mg, 1.28 mmol) in NMP (6 mL) was stirred at 80°C for 3 h. The resulting mixture was cooled to room temperature and poured into water (60 mL) and extracted with DCM (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 28, 1-(4-cyano-3-(trifluoromethyl)phenyl)-3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4] triazin-2-yl)phenyl)thiourea (140 mg, 23.57% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.53 (s, 1H), 10.47 (s, 1H), 10.37 (s, 1H), 8.33 (d, 1H), 8.10-8.01 (m, 2H), 7.65-7.59 (m, 2H), 7.19 (d, 1H), 4.13 (dd, 2H), 2.81 (t, 2H), 2.48 (s, 3H), 1.75-1.69 (m, 2H), 1.33 (t, 3H), 0.90 (t, 3H); MS: m/z = 556.1 (M+1, ESI+); HRMS: 556.1739.
Synthesis of Compound 29
Figure PCTKR2023008331-appb-img-000176
Step 1:
[00318] To a solution of 4-iodo-2-(trifluoromethyl)benzonitrile (500 mg, 1.68 mmol) and tert-butyl azetidin-3-ylcarbamate (319 mg, 1.85 mmol) in toluene (10 mL) was added Pd2(dba)3 (154 mg, 168 umol), xantphos (49 mg, 84 umol) and Cs2CO3 (1.10 g, 3.37 mmol), the reaction mixture was stirred at 80°C for 1 h. The resulting mixture was cooled to room temperature and poured into water (60 mL) and extracted with EA (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford tert-butyl (1-(4-cyano-3-(trifluoromethyl)phenyl)azetidin-3-yl)carbamate (330 mg, 57.43% yield) as a yellow solid. MS: m/z = 342.1 (M+1, ESI+).
Step 2:
[00319] To a solution of tert-butyl (1-(4-cyano-3-(trifluoromethyl)phenyl)azetidin-3-yl)carbamate (330 mg, 967 umol) in DCM (6 mL) was added TFA (1.10 g, 9.67 mmol), the reaction mixture was stirred at 25°C for 16 h. The mixture was evaporated under reduced pressure to afford 4-(3-aminoazetidin-1-yl)-2-(trifluoromethyl)benzonitrile 2,2,2-trifluoroacetate salt (230 mg, 98.71% yield) as a yellow oil. MS: m/z = 242.2 (M+1, ESI+).
Step 3:
[00320] To a solution of 4-(3-aminoazetidin-1-yl)-2-(trifluoromethyl)benzonitrile 2,2,2-trifluoroacetate salt (230 mg, 954 umol) and 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl) benzenesulfonyl chloride (411 mg, 1.00 mmol) in MeCN (10 mL) was added K2CO3 (527 mg, 3.81 mmol), the reaction mixture was stirred at 25°C for 1 h. The mixture was filtered and the filtrate was evaporated under reduce pressure, the residue was purified by prep-HPLC to afford compound 29 N-(1-(4-cyano-3-(trifluoromethyl)phenyl)azetidin-3-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide (140 mg, 99.2% purity) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.73 (s, 1H), 8.56 (s, 1H), 7.96-7.94 (m, 2H), 7.78 (d, 1H), 7.38 (dd, 1H), 6.73 (d, 1H), 6.63 (dd, 1H), 4.30-4.18 (m, 5H), 3.74-3.70 (m, 2H), 2.82 (t, 2H), 2.48 (s, 3H), 1.75-1.69 (m, 2H), 1.34 (t, 3H), 0.89 (t, 3H); MS: m/z =616.1 (M+1, ESI+); HRMS: 616.1950.
Synthesis of Compound 30
Figure PCTKR2023008331-appb-img-000177
Step 1:
[00321] To a solution of 4-iodo-2-(trifluoromethyl)benzonitrile (500 mg, 1.68 mmol) and tert-butyl piperidin-4-ylcarbamate (405 mg, 2.02 mmol) in toluene (10 mL) was added Pd2(dba)3 (154 mg, 168 umol), xantphos (49 mg, 84 umol) and Cs2CO3 (1.10 g, 3.37 mmol), the reaction mixture was stirred at 80°C for 2 h. The resulting mixture was cooled to room temperature and poured into water (60 mL) and extracted with EA (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford tert-butyl (1-(4-cyano-3-(trifluoromethyl) phenyl)piperidin-4-yl)carbamate (390 mg, 62.72% yield) as a yellow solid. MS: m/z = 370.2 (M+1, ESI+).
Step 2:
[00322] To a solution of tert-butyl (1-(4-cyano-3-(trifluoromethyl)phenyl)piperidin-4-yl)carbamate (390 mg, 1.06 mmol) in DCM (8 mL) was added TFA (1.20 g, 10.56 mmol), the reaction mixture was stirred at 25°C for 16 h. The mixture was evaporated under reduced pressure to afford 4-(4-aminopiperidin-1-yl)-2-(trifluoromethyl)benzonitrile 2,2,2-trifluoroacetate salt (270 mg, 94.97% yield) as a yellow oil. MS: m/z = 270.2 (M+1, ESI+).
Step 3:
[00323] To a solution of 4-(4-aminopiperidin-1-yl)-2-(trifluoromethyl)benzonitrile 2,2,2-trifluoroacetate salt (270 mg, 1.00 mmol) and 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl) benzenesulfonyl chloride (411 mg, 1.00 mmol) in MeCN (10 mL) was added K2CO3 (554 mg, 4.01 mmol), the reaction mixture was stirred at 25°C for 1 h. The mixture was filtered and the filtrate was evaporated under reduce pressure, the residue was purified by prep-HPLC to afford compound 30, N-(1-(4-cyano-3-(trifluoromethyl)phenyl)piperidin-4-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide (185 mg, 28.58% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.70 (s, 1H), 7.99-7.96 (m, 2H), 7.86-7.78 (m, 2H), 7.36 (dd, 1H), 7.25 (d, 1H), 7.19 (dd, 1H), 4.20 (q, 2H), 3.89 (d, 2H), 3.36 (s, 1H), 3.06 (t, 2H), 2.83 (t, 2H), 2.49 (s, 3H), 1.76-1.68 (m, 4H), 1.44-1.32 (m, 5H), 0.90 (t, 3H); MS: m/z =644.1 (M+1, ESI+); HRMS: 644.2261.
Synthesis of Compound 31
Figure PCTKR2023008331-appb-img-000178
Step 1:
[00324] To a solution of 4-iodo-2-(trifluoromethyl)benzonitrile (500 mg, 1.68 mmol) and tert-butyl pyrrolidin-3-ylcarbamate (408 mg, 2.19 mmol) in toluene (20 mL) was added Pd2(dba)3 (77 mg, 84 umol), xantphos (146 mg, 253 umol) and Cs2CO3 (1.65 g, 5.05 mmol), the reaction mixture was stirred at 80°C for 3 h. The resulting mixture was cooled to room temperature and poured into water (60 mL) and extracted with EA (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford tert-butyl (1-(4-cyano-3-(trifluoromethyl)phenyl)pyrrolidin-3-yl)carbamate (531 mg, 88.76% yield) as a yellow solid. MS: m/z =356.1 (M+1, ESI+).
Step 2:
[00325] To a solution of tert-butyl (1-(4-cyano-3-(trifluoromethyl)phenyl)pyrrolidin-3-yl)carbamate (531 mg, 1.49 mmol) in DCM (8 mL) was added 3 M HCl in EA (4 mL), the reaction mixture was stirred at 25 °C for 2 h. The mixture was evaporated under reduced pressure to afford 4-(3-aminopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile hydrochloride (300 mg, 78.66% yield) as a yellow solid. MS: m/z = 256.1 (M+1, ESI+).
Step 3:
[00326] To a solution of 4-(3-aminopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile hydrochloride (100 mg, 391.79 umol) and 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl) benzenesulfonyl chloride (161 mg, 392 umol) in DCM (10 mL) was added TEA (119 mg, 1.18 mmol), the reaction mixture was stirred at 25°C for 3 h. The resulting mixture poured into water (60 mL) and extracted with DCM (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford N-(1-(4-cyano-3-(trifluoromethyl)phenyl) pyrrolidin-3-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl) benzenesulfonamide (130 mg, 52.70% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.69 (s, 1H), 8.13 (d, 1H), 7.99-7.97 (m, 2H), 7.76 (d, 1H), 7.36 (d, 1H), 6.80-6.75 (m, 2H), 4.21 (q, 2H), 3.94-3.90 (m, 1H), 3.54-3.46 (m, 2H), 3.39-3.33 (m, 1H), 3.25 (dd, 1H), 2.83 (t, 2H), 2.49 (s, 3H), 2.14-2.06 (m, 1H), 1.97-1.89 (m, 1H), 1.78-1.69 (m, 2H), 1.36 (t, 3H), 0.91 (t, 3H); MS: m/z = 630.1 (M+1, ESI+); HRMS: 630.2106.
Synthesis of Compound 32
Figure PCTKR2023008331-appb-img-000179
Step 1:
[00327] To a solution of 4-iodo-2-(trifluoromethyl)benzonitrile (300 mg, 1.01 mmol) and tert-butyl (2-oxopyrrolidin-3-yl)carbamate (263 mg, 1.31 mmol) in toluene (20 mL) was added Pd2(dba)3 (46 mg, 51 umol), xantphos (88 mg, 152 umol) and Cs2CO3 (987 mg, 3.03 mmol), the reaction mixture was stirred at 80°C for 3 h. The resulting mixture was cooled to room temperature and poured into water (60 mL) and extracted with EA (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford tert-butyl (1-(4-cyano-3-(trifluoromethyl)phenyl)-2-oxopyrrolidin-3-yl)carbamate (315 mg, 84.44% yield) as a yellow solid. MS: m/z =314.0 (M-56+1, ESI+).
Step 2:
[00328] To a solution of tert-butyl (1-(4-cyano-3-(trifluoromethyl)phenyl)-2-oxopyrrolidin-3-yl)carbamate (310 mg, 839 umol) in DCM (8 mL) was added 3 M HCl in EA (4 mL), the reaction mixture was stirred at 25 °C for 2 h. The mixture was evaporated under reduced pressure to afford 4-(3-amino-2-oxopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile hydrochloride (220 mg, 97.36% yield) as a yellow solid. MS: m/z = 270.0 (M+1, ESI+).
Step 3:
[00329] To a solution of 4-(3-amino-2-oxopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile hydrochloride(220 mg, 817 umol) and 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl) benzenesulfonyl chloride (336 mg, 817 umol) in DCM (10 mL) was added TEA (248 mg, 2.45 mmol), the reaction mixture was stirred at 25°C for 3 h. The resulting mixture poured into water (60 mL) and extracted with DCM (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 32, N-(1-(4-cyano-3-(trifluoromethyl)phenyl)-2-oxopyrrolidin-3-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)benzenesulfonamide (150 mg, 28.52% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.69 (s, 1H), 8.40-8.36 (m, 2H), 8.19 (d, 1H), 8.03-7.97 (m, 3H), 7.37 (d, 1H), 4.44-4.37 (m, 1H), 4.21 (q, 2H), 3.88 (t, 1H), 3.78-3.71 (m, 1H), 2.83 (t, 2H), 2.48 (s, 3H), 2.34-2.27 (m, 1H), 1.86-1.68 (m, 3H), 1.34 (t, 3H), 0.90 (t, 3H);MS: m/z = 644.1 (M+1, ESI+); HRMS: 644.1901.
Synthesis of Compound 33
Figure PCTKR2023008331-appb-img-000180
Step 1:
[00330] To a solution of 4-iodo-2-(trifluoromethyl)benzonitrile (500 mg, 1.68 mmol) and tert-butyl (5-oxopyrrolidin-3-yl)carbamate (438 mg, 2.19 mmol) in toluene (20 mL) was added Pd2(dba)3 (77 mg, 84 umol), xantphos (146 mg, 253 umol) and Cs2CO3 (1.65 g, 5.05 mmol), the reaction mixture was stirred at 80°C for 3 h. The resulting mixture was cooled to room temperature and poured into water (60 mL) and extracted with EA (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by column chromatography to afford tert-butyl (1-(4-cyano-3-(trifluoromethyl)phenyl)-5-oxopyrrolidin-3-yl)carbamate (522 mg, 83.96% yield) as a yellow solid. MS: m/z =370.1 (M+1, ESI+).
Step 2:
[00331] To a solution of tert-butyl (1-(4-cyano-3-(trifluoromethyl)phenyl)-5-oxopyrrolidin-3-yl)carbamate (522 mg, 1.41 mmol) in DCM (10 mL) was added 3 M HCl in EA (5 mL), the reaction mixture was stirred at 25 °C for 2 h. The mixture was evaporated under reduced pressure to afford 4-(4-amino-2-oxopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile hydrochloride (378 mg, 99.34% yield) as a yellow solid. MS: m/z = 270.1 (M+1, ESI+).
Step 3:
[00332] To a solution of 4-(4-amino-2-oxopyrrolidin-1-yl)-2-(trifluoromethyl)benzonitrile hydrochloride (378 mg, 1.40 mmol) and 4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl) benzenesulfonyl chloride (577 mg, 1.40 mmol) in DCM (20 mL) was added TEA (426 mg, 4.21 mmol), the reaction mixture was stirred at 25°C for 3 h. The resulting mixture poured into water (60 mL) and extracted with DCM (10 mLx3), the combined organic layers were washed with water (50 mL) and brine (50 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 33, N-(1-(4-cyano-3-(trifluoromethyl) phenyl)-5-oxopyrrolidin-3-yl)-4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f] [1,2,4]triazin-2-yl)benzenesulfonamide (170 mg, 18.81% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.70 (s, 1H), 8.37-8.31 (m, 2H), 8.15 (d, 1H), 7.98-7.96 (m, 2H), 7.89 (dd, 1H), 7.36 (dd, 1H), 4.23-4.05 (m, 4H), 3.77 (dd, 1H), 2.84-2.77 (m, 3H), 2.49 (s, 3H), 2.41 (dd, 1H), 1.77-1.68 (m, 2H), 1.34 (t, 3H), 0.91 (t, 3H);MS: m/z = 644.2 (M+1, ESI+); HRMS: 644.1899.
Synthesis of Compound 34
Figure PCTKR2023008331-appb-img-000181
[00333] To a solution of compound 57, 3-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino)tetrahydrofuran-3-carbonitrile (1.09 g, 2.58 mmol) and 4-amino-2-(trifluoromethyl)benzonitrile (576.27 mg, 3.10 mmol) in toluene (20 mL) was added TCDI (551.74 mg, 3.10 mmol), The reaction mixture was stirred at 105 °C for 22 h. The reaction solution was cooled to room temperature and concentrated under reduced pressure, DMA (2 mL) and EtOH (20 mL) was added to the residue. The mixture was heated to 70 °C and hydrogen chloride (2 M, 4 mL) was added, then stirred at this temperature for 2 h. The resulting mixture was poured into water (200 mL) and extracted with EA (50mLx3), washed by brine (200 mL), dried over Na2SO4 and concentrated. The residue was purified by Prep-HPLC to afford compound 34, 4-(1-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4-oxo-2-thioxo-7-oxa-1,3-diazaspiro[4.4]nonan-3-yl)-2-(trifluoromethyl)benzonitrile (60 mg, 3.57% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 11.67 (s, 1H), 8.39 (d, 1H), 8.26 (s, 1H), 8.07 (dd, 1H), 7.63-7.61 (m, 2H), 7.32 (d, 1H), 4.33 (d, 1H), 4.17 (q, 2H), 4.00 (d, 1H), 3.78 (dd, 1H), 3.54 (dd, 1H), 2.82 (t, 2H), 2.58 (t, 2H), 2.48 (s, 3H), 1.76-1.71 (m, 2H), 1.34 (t, 3H), 0.91 (t, 3H);MS: m/z = 652.4 (M+1, ESI+); HRMS: 652.1942.
Synthesis of Compound 38
Figure PCTKR2023008331-appb-img-000182
[00334] To a solution of compound 58, methyl 1-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4] triazin-2-yl)phenyl)amino)cyclopentane-1-carboxylate (140 mg, 309 umol) and 4-isothiocyanato-2-(trifluoromethyl)benzonitrile (85 mg, 370 umol) in NMP (8 mL) was added isopropyl acetate (4 mL), the reaction mixture was stirred at 85 °C for 16 h. The reaction mixture was poured into water (80 mL) and extracted with DCM (20 mLx3), the combined organic layers were washed with water (100 mL) and brine (100 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 38, 4-(1-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4-oxo-2-thioxo-1,3-diazaspiro[4.4]nonan-3-yl)-2-(trifluoromethyl)benzonitrile (130 mg, 64.82% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 12.15 (s, 1H), 8.38 (d, 1H), 8.29 (d, 1H), 8.08 (dd, 1H), 7.65-7.60 (m, 2H), 7.36 (d, 1H), 4.19 (q, 2H), 2.95 (t, 2H), 2.58 (s, 3H), 2.32-2.27 (m, 2H), 2.23-2.18 (m, 2H), 1.81-1.72 (m, 4H), 1.46-1.42 (m, 2H), 1.35 (t, 3H), 0.93 (t, 3H);MS: m/z = 650.4 (M+1, ESI+); HRMS: 650.2159.
Synthesis of Compound 47
Figure PCTKR2023008331-appb-img-000183
[00335] To a solution of compound 52, methyl 2-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4] triazin-2-yl)phenyl)amino)-2-methylpropanoate (540 mg, 1.26 mmol) and compound 59, 3-fluoro-4-isothiocyanato-2-(trifluoromethyl)benzonitrile (1.55 g, 6.3 mmol) in NMP (8 mL) was added isopropyl acetate (2 ml), the reaction mixture was stirred at 110 °C for 16 h. The reaction mixture was poured into water (80 mL) and extracted with DCM (20 mLx3), the combined organic layers were washed with water (80 mL) and brine (80 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 47, 4-(3-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4,4-dimethyl-5-oxo-2-thioxoimidazolidin-1-yl)-3-fluoro-2-(trifluoromethyl)benzonitrile (90 mg, 11% yield) as a pale yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 11.67 (s, 1H), 8.30-8.25 (m, 2H), 7.61-7.58 (m, 2H), 7.34 (d, 1H), 4.18 (q, 2H), 2.83 (t, 2H), 2.48 (s, 3H), 1.76-1.70 (m, 2H), 1.54 (dd, 6H), 1.33 (t, 3H), 0.91 (t, 3H); MS: m/z = 642.4 (M+1, ESI+); HRMS: 642.1904.
Synthesis of Compound 48
Figure PCTKR2023008331-appb-img-000184
[00336] To a solution of compound 54, methyl 1-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4] triazin-2-yl)phenyl)amino)cyclobutane-1-carboxylate (600 mg, 1.36 mmol) and compound 59, 3-fluoro-4-isothiocyanato-2-(trifluoromethyl)benzonitrile (1.67 g, 6.8 mmol) in NMP (8 mL) was added isopropyl acetate (2 ml), the reaction mixture was stirred at 110 °C for 16 h. The reaction mixture was poured into water (80 mL) and extracted with DCM (20 mLx3), the combined organic layers were washed with water (80 mL) and brine (80 mL), then dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by prep-HPLC to afford compound 48, 4-(5-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-8-oxo-6-thioxo-5,7-diazaspiro[3.4]octan-7-yl)-3-fluoro-2-(trifluoromethyl)benzonitrile (120 mg, 13.5% yield) as a pale yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 11.68 (s, 1H), 8.26-8.20 (m, 2H), 7.63-7.61 (m, 2H), 7.37 (dd, 1H), 4.19 (q, 2H), 2.82 (t, 2H), 2.68-2.62 (m, 1H), 2.58-2.52 (m, 2H), 2.49-2.44 (m, 4H), 2.02-1.95 (m, 1H), 1.78-1.69 (m, 2H), 1.60-1.56 (m, 1H), 1.35 (t, 3H), 0.91 (t, 3H); MS: m/z = 654.4 (M+1, ESI+); HRMS: 654.1901.
Synthesis of Compound 49
Figure PCTKR2023008331-appb-img-000185
[00337] To a solution of Compound 57, 3-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino)tetrahydrofuran-3-carbonitrile (900 mg, 2.13 mmol) in THF (50 mL) was added Compound 59, 3-fluoro-4-isothiocyanato-2-(trifluoromethyl)benzonitrile (1.57 g, 6.39 mmol), the reaction mixture was stirred at 70 °C for 16 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure, the residue was dissolved in EtOH (50 mL) and DMA (5 mL), then 2 N HCl (5 mL) was added to the above solution. The resulting mixture was stirred at 70 °C for 2 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure, the residue was poured into water (80 mL), extracted with EA (30mLx3), washed by brine (80 mL), dried over Na2SO4 and concentrated. The residue was purified by Prep-HPLC to afford Compound 49, 4-(1-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4-oxo-2-thioxo-7-oxa-1,3-diazaspiro[4.4]nonan-3-yl)-3-fluoro-2-(trifluoromethyl) benzonitrile (135 mg, 9.46% yield) as a pale yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 11.62 (s, 1H), 8.29-8.20 (m, 2H), 7.72-7.67 (m, 2H), 7.32 (d, 1H), 4.40 (dd, 1H), 4.18 (q, 2H), 4.03-3.96 (m, 1H), 3.80 (q, 1H), 3.61-3.51 (m, 1H), 2.83 (t, 2H), 2.72-2.52 (m, 2H), 2.48 (s, 3H), 1.78-1.69 (m, 2H), 1.34 (t, 3H), 0.92 (t, 3H); MS: m/z = 670.4 (M+1, ESI+); HRMS: 670.1849.
Synthesis of Compound 50
Figure PCTKR2023008331-appb-img-000186
Step 1:
[00338] To a solution of 2-(5-amino-2-ethoxyphenyl)-5-methyl-7-propylimidazo[5,1-f][1,2,4]triazin-4(3H)-one (1 g, 3.05 mmol) and cyclopentanone (514 mg, 6.11 mmol) in dioxane (30 mL) was added TMSCN (454 mg, 4.58 mmol) and ZnCl2 (83 mg, 610.91 umol), the reaction mixture was stirred at 50 °C for 16 h. The reaction mixture was cooled to room temperature and poured into water (80 mL), extracted with EA (30mLx3), washed by brine (80 mL), dried over Na2SO4 and concentrated. The residue was purified by column chromatography to afford 1-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino)cyclopentane-1-carbonitrile (1.15 g, 89.53% yield) as a yellow solid. MS: m/z =421.3 (M+1, ESI+).
Step 2:
[00339] To a solution of 1-((4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)amino)cyclopentane-1-carbonitrile (1 g, 2.38 mmol) in THF (50 mL) was added compound 59, 3-fluoro-4-isothiocyanato-2-(trifluoromethyl)benzonitrile (1.76 g, 7.13 mmol), the reaction mixture was stirred at 70 °C for 16 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure, the residue was dissolved in EtOH (50 mL) and DMA (5 mL), then 2 N HCl (5 mL) was added to the above solution. The resulting mixture was stirred at 70 °C for 2 h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure, the residue was poured into water (80 mL), extracted with EA (30mLx3), washed by brine (80 mL), dried over Na2SO4 and concentrated. The residue was purified by Prep-HPLC to afford compound 50, 4-(1-(4-ethoxy-3-(5-methyl-4-oxo-7-propyl-3,4-dihydroimidazo[5,1-f][1,2,4]triazin-2-yl)phenyl)-4-oxo-2-thioxo-1,3-diazaspiro[4.4]nonan-3-yl)-3-fluoro-2-(trifluoromethyl)benzonitrile (126 mg, 7.94% yield) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 11.67 (s, 1H), 8.26 (s, 2H), 7.65 (s, 2H), 7.32 (d, 1H), 4.18 (q, 2H), 2.82 (t, 2H), 2.48 (s, 3H), 2.32-2.20 (m, 4H), 1.76-1.71 (m, 4H), 1.50-1.43 (m, 2H), 1.34 (t, 3H), 0.92 (t, 3H); MS: m/z = 668.5 (M+1, ESI+); HRMS: 668.2057.
Example 3 - Human PDE5A1 Inhibition Assay
[00340] This example illustrates the in vitro inhibition of human PDE5A1 by exemplary compounds of this disclosure (e.g., as described herein).
Materials
[00341] Sildenafil citrate (Catalog no. LKT-S3313, Axxora, San Diego, CA), Vardenafil hydrochloride trihydrate (Catalog no. SML2103, Sigma-Aldrich, St. Louis, MO), PDE Assay Buffer (Catalog no. 60393, BPS bioscience, San Diego, CA), PDE Binding Agent (Catalog no. 60390, BPS bioscience, San Diego, CA) and PDE Binding Agent Diluent (cGMP, Catalog no. 60392, BPS bioscience, San Diego, CA) were used for assays. Test compounds were supplied by Ildong Pharmaceuticals Co., Ltd.
Experimental protocols
[00342] The enzymes and substrates used in this experiment are summarized in Table 2.
Enzymes and Substrates
Assay Catalog # Enzyme Lot# Enzyme Used (ng/ reaction) Substrate
PDE5A1 60050 181008-G 0.2 100 nM FAM-cGMP
[00343] The serial dilution of the compounds was first performed in 100% DMSO with the highest concentration at 1 mM and 0.1 mM. Each intermediate compound dilution (in 100% DMSO) will then get directly diluted 10x fold into assay buffer for 10% DMSO and 5 μL of the dilution was added to a 50 μL reaction so that the final concentration of DMSO is 1% in all reactions.
[00344] The enzymatic reactions were conducted at room temperature for 60 minutes in a 50 μL mixture containing PDE assay buffer, 100 nM FAM-cGMP, a PDE enzyme (Table 2) and the test compounds.
[00345] After enzymatic reaction, 100 μL of a binding solution (1:100 dilution of the binding agent with the binding agent diluent) was added to each reaction and the reaction was performed at room temperature for 60 minutes.
[00346] Fluorescence intensity was measured at excitation of 485 nm and an emission of 528 nm using a Tecan Infinite M1000 microplate reader.
Data Analysis
[00347] PDE activity assays were performed in duplicate at each concentration. Fluorescence intensity is converted to fluorescence polarization using the Tecan Magellan6 software. The fluorescence polarization (FPt) in absence of the compound in each data set was defined as 100% activity. In the absence of PDE and the compound, the value fluorescent polarization (FPb) in each data set was defined as 0% activity. The percent activity in the presence of compound was calculated according to Equation 1:
Figure PCTKR2023008331-appb-img-000187
(eqn . 1)
where FP = the fluorescence polarization in the presence of the compound.
[00348] The values of % activity versus a series of compound concentrations were then plotted using non-linear regression analysis of Sigmoidal dose-response curve generated with Equation 2:
Figure PCTKR2023008331-appb-img-000188
( eqn . 2)
where Y = percent activity, B = minimum percent activity, T = maximum percent activity, X = logarithm of compound, and Hill Slope = slope factor or Hill coefficient. The IC50 value was determined by the concentration causing a half-maximal percent activity.
Results
[00349] The results are tabulated in Table 3 with IC50 values shown as ranges.
In Vitro Inhibition of Human PDE5 Activities
Cmpd No. IC50 (nM)
A: IC50 ≤10 nM
B: 10 nM < IC50 ≤100 nM
C: IC50 > 100 nM
Cmpd No. IC50 (nM)
A: IC50 ≤10 nM
B: 10 nM < IC50 ≤100 nM
C: IC50 > 100 nM
PDE5 PDE5
1 A 2 B
3 B 4 B
5 A 6 A
7 A 8 A
9 A 10 A
11 C 13 C
18 A 19 C
20 A 21 C
22 A 23 C
24 A 25 A
26 A 27 A
28 A 29 A
30 A 31 A
32 A 33 A
34 A 38 A
47 A 48 A
49 A 50 A
[00350] As illustrated by this example, the tested compounds exhibited very good inhibitory activity against PDE5.
Example 4 - Androgen Receptor (AR) Reporter Assay
[00351] This example illustrates the in vitro antagonistic activity toward androgen receptor (AR) exhibited by exemplary compounds of this disclosure (e.g., as described herein). The protocols and results of this Example were carried out and obtained by Thermofisher Scientific.
Test Compounds
[00352] Test compounds were received at 1000X (or greater) of the desired starting concentration in 100% DMSO. If compounds were supplied at greater that 1000X concentration, an initial dilution is made in 100% DMSO to bring the compounds to 1000X concentration. The 1000X test compounds were serially diluted (10 point
½-log increments) in 100% DMSO.
Substrate Loading Solution
[00353] The Substrate Loading Solution consists of three Life Technologies reagents: Solution A (10 mM LiveBLAzer™-FRET B/G Substrate), Solution B and Solution C.
Androgen Receptor (AR) - Antagonist Screen, Activated by R1881
[00354] AR-UAS-bla GripTite™ 293 cells were thawed and resuspended in Assay Media (DMEM phenol red free, 2% CD-treated FBS, 0.1 mM NEAA, 1 mM Sodium Pyruvate, 100 U/mL/100 μg/mL Pen/Strep) to a concentration of 312,500 cells/mL.4 μL of a 10X serial dilution of Cyproterone Acetate (control antagonist starting concentration, 3,160 nM) or compounds are added to appropriate wells of a Poly-D-Lysine assay plate. 32 μL of cell suspension was added to the wells and pre-incubated at 37 ℃/5% CO2 in a humidified incubator with compounds and control antagonist titration for 30 minutes. 4 μL of 10X control agonist R1881 at the pre-determined EC80 concentration was added to wells containing the control antagonist or compounds. The plate was incubated for 16-24 hours at 37 ℃/5% CO2 in a humidified incubator. 8 μL of 1 μM Substrate Loading Solution is added to each well and the plate was incubated for 2 hours at room temperature. The plate was read on a fluorescence plate reader (Tecan Safire2).
Results
[00355] The results are tabulated in Table 4 with IC50 values shown as ranges.
In Vitro Androgen Receptor (AR) Reporter Assay
Cmpd No. Concentration Range (nM)
A: IC50 ≤ 500 nM
B: 500 nM < IC50 ≤1000 nM
C: IC50 > 1000 nM
Cmpd No. Concentration Range(nM)
A: IC50 ≤ 500 nM
B: 500 nM < IC50 ≤1000 nM
C: IC50 > 1000 nM
IC50 IC50
13 B 18 A
19 B 20 A
21 A 22 A
23 A 25 C
26 C 27 C
28 C 29 B
30 A 31 B
32 C 33 C
34 A 38 A
47 A 48 A
49 B 50 A
Example 5 - Androgen Receptor (AR) Radioligand Binding Assay
[00356] This example also illustrates the in vitro antagonistic activity toward androgen receptor (AR) exhibited by exemplary compounds of this disclosure (e.g., as described herein), and illustrates the binding affinity of the exemplary compounds.
Procedure
[00357] Methods employed in this study have been adapted from the following literature procedure.
[00358] Human androgen receptors obtained from human LNCaP cells are used in modified HEPES buffer pH 7.4. A 70 ㎍ (adjusted if necessary) aliquot is incubated with 0.5 nM [3H]Methyltrienolone for 20 hours at 4 oC. Non-specific binding is estimated in the presence of 1 μM testosterone. Receptors are filtered and washed, the filters are then counted to determine [3H]methyltrienolone specifically bound (Historic values: Kd = 0.71 nM: Specific binding = 75%; Bmax = 0.25 pmole/mg protein). (See, e.g., Traish, A. M et al., Binding of 7α, 17α-dimethyl-19-nortestosterone (Mibolerone) to androgen and progesterone receptors in human and animal tissues. Endocrinology. 118(4): 1327-1333, 1986).
[00359] Compounds are screened at 10 μM.
[00360] Where presented, IC50 values were determined by a non-linear, least squares regression analysis using MathIQTM (ID Business Solutions Ltd., UK).
Results
[00361] The results are tabulated in Table 5 with the following values shown as ranges: IC50 (nM) concentration ranges: (A) refers to IC50 ≤ 50 nM; (B) refers to 50 nM < IC50 ≤ 200; and (C) refers to IC50 > 200 nM.
In Vitro Androgen Receptor (AR) Binding Assay
Compound AR IC50 (nM)
13 C
18 A
19 C
20 B
21 C
22 B
23 C
24 B
26 C
29 B
30 A
31 B
32 C
33 C
34 B
38 A
47 A
48 A
49 A
50 A
[00362] As illustrated by this example, exemplary compounds of this disclosure exhibit potent AR inhibitory activity and binding affinity.
Example 6 - Antitumor effect of subject compounds in patient-derived xenograft ( PDX ) mouse model of triple negative breast cancer ( TNBC )
[00363] This example assesses the antitumor efficacy of the subject compounds in a TNB CTG-0670, with DHT tumor model.
Test Compounds
[00364] Three test compounds (subject compounds 18, 20 and 22) and two positive controls (enzalutamide) were evaluated.
Formulation Vehicles
Polyethylene glycol (15)-hydroxystearate (Solutol, 30% in deionized water) and PEG-400 (20% in deionized water) were used as excipients to prepare test compounds 18, 20 and 22, and positive controls enzalutamide.
Study Design
[00365] Female athymic nude mice were divided into 9 groups for dosing orally (p.o.) once daily over a 16 day period as set out in Table 6 below:
TNB CTG -0670 + DHT tumor model Study Design
Study Groups Compound Dose Level
(mg/kg/day)
1 Vehicle control 0
2 Vehicle control + DHT 0
3 Enzalutamide + DHT 25
4 Enzalutamide + DHT 50
5 Compound 18 + DHT 25
6 Compound 20 + DHT 25
7 Compound 20 + DHT 50
8 Compound 22 + DHT 25
9 Compound 22 + DHT 50
Results
[00366] The mean tumor volume (MTV) was measured on day 0 and day 16 of treatment. The mean tumor volume for each group and the maximum tumor growth inhibition (TGI) relative to the control group is set out in Table 7 below.
[00367] TGI was calculated using the formula below:
TGI = [1-(final MTV - initial MTV of a treated group) / (final MTV - initial MTV of the control group)] x 100%.
TNB CTG -0670 + DHT tumor model Results
Study Groups Compound Dose Level
(mg/kg/day)
Body Weight
(g)
Mean tumor volume (mm3) Day 0 Mean tumor volume (mm3) Day 16 TGI (%)
1 Vehicle control 0 24.1 190 1089 19
2 Vehicle control + DHT 0 25.9 196 1297 0
3 Enzalutamide + DHT 25 24.7 196 1243 5
4 Enzalutamide + DHT 50 25.1 196 1379 -7
5 Compound 18 + DHT 25 24.8 196 1119 17
6 Compound 20 + DHT 25 24.5 196 1015 26
7 Compound 20 + DHT 50 25.0 196 1135 15
8 Compound 22 + DHT 25 25.5 196 1139 15
9 Compound 22 + DHT 50 25.6 196 855 41
[00368] As seen above three subject compounds (18, 20 and 22) were studied in a TNB CTG-0670 + DHT tumor mouse model against enzalutamide, and vehicle as controls at two dose levels. Tumor growth inhibition was observed at both dose levels after treatment with compounds 20 and 22.
[00369] These results indicate that the subject compounds can be used to treat triple-negative breast cancer (TNBC).
Example 7 - Assessment of Pharmacokinetic Properties
[00370] The following methods can be adapted to assess the pharmacokinetic properties of exemplary compounds.
[00371] Animal Study:
[00372] Test compound is administered to ICR mice via IV (5 mg/kg and 5mL/kg) or PO (5 mg/kg and 5mL/kg).
[00373] ICR mice are administered via each route with the test compound dissolved in appropriate vehicles. Plasma samples are collected designated time and the samples are then stored at -70℃ until analysis using LC-MS/MS system.
[00374] Bioassays:
[00375] Plasma: Protein precipitation is conducted on 20 μL of plasma samples with 180 μL of acetonitrile containing internal standard. After mixing and centrifugation, the supernatant(150 μL) is analyzed by LC-MS/MS system with MRM mode.
[00376] Brain: Protein precipitation is conducted on 50 μL of brain samples with 200 μL of acetonitrile containing internal standard. After mixing and centrifugation, supernatant(150 μL) is analyzed by LC-MS/MS system with MRM mode.
[00377] Data analysis:
[00378] LC/MS/MS data are analyzed by Analyst 1.7.2. PK parameters are calculated by noncompartmental analysis using WinNonlin software.
EQUIVALENTS AND INCORPORATION BY REFERENCE
[00379] While the invention has been particularly shown and described with reference to a preferred embodiment and various alternate embodiments, it will be understood by persons skilled in the relevant art that various changes in form and details can be made therein without departing from the spirit and scope of the invention.
[00380] All references, issued patents and patent applications cited within the body of the instant specification are herein incorporated by reference in their entirety, for all purposes.

Claims (52)

  1. A method of treating a subject for cancer, comprising administering to a subject having cancer a therapeutically effective amount of an androgen receptor (AR)- and phosphodiesterase 5 (PDE5)-inhibiting compound.
  2. The method of claim 1, wherein the subject has a cancer selected from breast cancer, gastric cancer, gastrointestinal cancer, a gynecological cancer, kidney cancer, glioblastoma, high-grade-Glioma, non-small cell lung cancer, head and neck squamous cell carcinoma, pancreatic cancer, head and neck cancer, prostate cancer, metastatic castration-resistant prostate cancer (mCRPC), glioma, brain neoplasms and brain metastasis.
  3. The method of claim 2, wherein the subject has breast cancer.
  4. The method of claim 3, wherein the breast cancer is a triple-negative breast cancer (TNBC).
  5. The method of claim 4, wherein the TNBC is a subtype selected from basal-like type 1 (BL1), basal-like type 2 (BL2), immunomodulatory (IM), mesenchymal (M), mesenchymal stem-like (MSL), and luminal androgen receptor (LAR) subtypes.
  6. The method of claim 4, wherein the TNBC is androgen receptor (AR)-positive triple-negative breast cancer (AR+ TNBC).
  7. The method of any one of claims 4 to 6, wherein the TNBC comprises a BRCA1 mutation.
  8. The method of claim 2, wherein the subject has glioblastoma.
  9. The method of claim 2, wherein the subject has gastric cancer.
  10. The method of claim 2, wherein the subject has prostate cancer.
  11. The method of claim 2, therein the subject has metastatic castration-resistant prostate cancer (mCRPC).
  12. The method of any one of claims 1 to 11, wherein the AR- and PDE5-inhibiting compound is of formula (I):
    Figure PCTKR2023008331-appb-img-000189
    (I)
    or a pharmaceutically acceptable salt thereof, wherein:
    L is a linking moiety;
    R1 and R2 are independently selected from -H, optionally substituted (C1-C6)alkyl, optionally substituted (C3-C6)cycloalkyl, optionally substituted (C1-C6)alkoxy, and optionally substituted (C2-C4)alkenyl;
    each R13 is selected from -H, optionally substituted (C1-C6)alkyl, and optionally substituted (C1-C6)alkoxy;
    each R14 is independently selected from -H, -CN, -OH, -NH2, -NO2, halogen, optionally substituted (C1-C5)alkyl, optionally substituted (C1-C5)haloalkyl, optionally substituted (C1-C5)alkoxy, optionally substituted (C3-C6)cycloalkyl, and optionally substituted (C2-C4)alkenyl;
    X1 is N or CR14;
    X2 and X3 are independently selected from N and CR13;
    Y1 and Y2 are independently selected from N and C, wherein one of Y1 and Y2 is N;
    m is 0 to 2; and
    n is 1 to 4.
  13. The method of claim 12, wherein -L- is -A-B-,
    where:
    -A- is selected from a covalent bond, optionally substituted (C6-C12) aryl or (C3-C12) heteroaryl, optionally substituted-(C3-C12) heteroaryl-(C1-C5)alkylene-, optionally substituted 3- to 6-membered heterocycle, -NHC(O)R5-,
    Figure PCTKR2023008331-appb-img-000190
    , and
    Figure PCTKR2023008331-appb-img-000191
    ; and
    -B- is selected from a covalent bond, optionally substituted 3- to 6-membered heterocycle, -NHC(O)R5-, -O-, -S-, -NR11-,
    Figure PCTKR2023008331-appb-img-000192
    ,
    Figure PCTKR2023008331-appb-img-000193
    , and
    Figure PCTKR2023008331-appb-img-000194
    ;
    wherein:
    R11 is H or optionally substituted (C1-C3)alkyl;
    R5 is selected from -OH, -(C1-C5)alkyl, -(C1-C5)haloalkyl and optionally substituted (C1-C5)alkylene;
    R6 and R7 are each independently -H or optionally substituted (C1-C3)alkyl; or R6 and R7 together with the nitrogen atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered heterocycle;
    Z1 is selected from O and S; and
    at least one of -A- and -B- is not a covalent bond.
  14. The method of claim 12, wherein the compound is of formula (IIa) or (IIb):
    Figure PCTKR2023008331-appb-img-000195
    wherein:
    R3 is selected from -H, and optionally substituted (C1-C6)alkoxy; and
    -B- is selected from a covalent bond and optionally substituted 3- to 6-membered heterocycle.
  15. The method of claim 14, wherein the compound is of formula (IIIa) or (IIIb):
    Figure PCTKR2023008331-appb-img-000196
    wherein:
    R8 and R9 are independently selected from -H and optionally substituted (C1-C3)alkyl, or R8 and R9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered carbocycle or optionally substituted 3-to 6-membered heterocycle; and
    Z1 is O or S.
  16. The method of claim 15, wherein B is a covalent bond and the compound is of formula (IVa) or (IVb):
    Figure PCTKR2023008331-appb-img-000197
    .
  17. The method of claim 15, wherein the compound is of formula (Va) or (Vb):
    Figure PCTKR2023008331-appb-img-000198
    wherein p and q are independently 1 or 2.
  18. The method of claim 13, wherein R6 and R7 are each -H and the compound is of formula (VIa) or (VIb):
    Figure PCTKR2023008331-appb-img-000199
    .
  19. The method of claim 18, wherein -B- is a bond and the compound is of formula (VIIa) or (VIIb):
    Figure PCTKR2023008331-appb-img-000200
    .
  20. The method of claim 12, wherein the compound is of formula (VIIIa) or (VIIIb):
    Figure PCTKR2023008331-appb-img-000201
    wherein R3 is selected from -H, and optionally substituted (C1-C5)alkoxy.
  21. The method of claim 20, wherein -A- is a covalent bond and the compound is of formula (IXa) or (IXb):
    Figure PCTKR2023008331-appb-img-000202
    .
  22. The method of claim 12, wherein the compound is of formula (Xa) or (Xb);
    Figure PCTKR2023008331-appb-img-000203
    wherein:
    R3 is selected from -H, and optionally substituted (C1-C6)alkoxy;
    R12 is -H, or optionally substituted (C1-C3)alkyl; and
    -B- is -O-, -S-, -NH-, -SO2- , or -NHSO2-.
  23. The method of claim 12, wherein the compound is of formula (XIa) or (XIb):
    Figure PCTKR2023008331-appb-img-000204
    wherein:
    R3 is selected from -H, and optionally substituted (C1-C6)alkoxy; and
    -B- is selected from -O-, -S-, -SO2- and -NHSO2-.
  24. The method of any one of claims 1 to 23, wherein:
    R1 is optionally substituted (C1-C6)alkyl;
    R2 is optionally substituted (C1-C6)alkyl;
    each R13 and R3 is H or optionally substituted (C1-C3)alkoxy; and
    each R14 and R4 is selected from optionally substituted (C1-C5)haloalkyl, halogen and H.
  25. The method of any one of claims 1 to 24, wherein:
    X1, X2, and X3 are each CH;
    X1 is N, and X2 and X3 are each CH;
    X2 is N, and X1 and X3 are each CH; or
    X3 is N, and X1 and X2 are each CH.
  26. The method of claim 16, wherein the compound is of formula (IVc) or (IVd):
    Figure PCTKR2023008331-appb-img-000205
    wherein R14 is -H or halogen.
  27. The method of claim 26, wherein R8 and R9 are each independently H or optionally substituted (C1-C3)alkyl.
  28. The method of claim 27, wherein the compound is selected from
    Figure PCTKR2023008331-appb-img-000206
    .
  29. The method of claim 26, wherein R8 and R9 are each methyl.
  30. The method of claim 29, wherein the compound is selected from
    Figure PCTKR2023008331-appb-img-000207
    or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  31. The method of claim 26, wherein R8 and R9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered carbocycle or optionally substituted 3- to 6-membered heterocycle that is selected from optionally substituted cyclobutyl, optionally substituted cyclopentyl, and optionally substituted tetrahydrofuran.
  32. The method of claim 31, wherein the compound is selected from:
    Figure PCTKR2023008331-appb-img-000208
    ,
    or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  33. The method of claim 16, wherein the compound is of formula (IVc):
    Figure PCTKR2023008331-appb-img-000209
    (IVc)
    wherein:
    X1 is CH or N;
    R14 is -H or halogen; and
    R8 and R9 are each independently H or (C1-C3)alkyl (e.g., R8 and R9 are each -CH3), or R8 and R9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 5-membered carbocycle, or an optionally substituted 4-membered or 5-membered heterocycle (e.g., cyclopentane cyclobutane, cyclopentane, oxetane or tetrahydrofuran).
  34. The method of claim 33, wherein the compound is selected from:
    Figure PCTKR2023008331-appb-img-000210
    Figure PCTKR2023008331-appb-img-000211
    Figure PCTKR2023008331-appb-img-000212
    Figure PCTKR2023008331-appb-img-000213
    ,
    or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  35. The method of claim 16, wherein:
    either X2 is N and X3 is CH, or X2 is CH and X3 is N;
    R1 is -CH3;
    R1 is n-propyl;
    R1 is ethoxy; and
    R8 and R9 are each optionally substituted (C1-C3)alkyl, or R8 and R9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered carbocycle or optionally substituted 3- to 6-membered heterocycle that is selected from optionally substituted cyclobutyl, optionally substituted cyclopentyl, and optionally substituted tetrahydrofuran.
  36. The method of claim 35, wherein the compound is selected from:
    Figure PCTKR2023008331-appb-img-000214
    Figure PCTKR2023008331-appb-img-000215
    ,
    or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  37. The method of claim 17, wherein the compound is of formula (Vc):
    Figure PCTKR2023008331-appb-img-000216
    (Vc)
    wherein:
    R8 and R9 are each optionally substituted (C1-C3)alkyl, or R8 and R9 together with the carbon atom to which they are attached are cyclically linked to provide an optionally substituted 3- to 6-membered carbocycle or optionally substituted 3- to 6-membered heterocycle that is selected from optionally substituted cyclobutyl, optionally substituted cyclopentyl, and optionally substituted tetrahydrofuran.
  38. The method of claim 37, wherein the compound is selected from:
    Figure PCTKR2023008331-appb-img-000217
    ,
    or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  39. The method of claim 19, wherein the compound is of formula (VIIc):
    Figure PCTKR2023008331-appb-img-000218
    (VIIc).
  40. The method of claim 39, wherein the compound is selected from:
    Figure PCTKR2023008331-appb-img-000219
    ,
    or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  41. The method of claim 20, wherein the compound is of formula (VIIIc):
    Figure PCTKR2023008331-appb-img-000220
    (VIIIc).
  42. The method of claim 41, wherein -A- is
    Figure PCTKR2023008331-appb-img-000221
    wherein:
    R12 is selected from -H, -OH, optionally substituted (C1-C3)alkyl, and optionally substituted (C1-C5)haloalkyl; and
    r, s and t are independently is 0 or 1.
  43. The method of claim 42, wherein the compound is selected from:
    Figure PCTKR2023008331-appb-img-000222
    or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  44. The method of claim 21, wherein the compound is of formula (IXc) or (IXd):
    Figure PCTKR2023008331-appb-img-000223
    .
  45. The method of claim 44, wherein the compound is selected from:
    Figure PCTKR2023008331-appb-img-000224
    ,
    or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  46. The method of claim 20, wherein the compound is of formula (VIIId):
    Figure PCTKR2023008331-appb-img-000225
    (VIIId).
  47. The method of claim 46, wherein the compound is
    Figure PCTKR2023008331-appb-img-000226
    ,
    or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  48. The method of claim 23, wherein the compound is of formula (XIc):
    Figure PCTKR2023008331-appb-img-000227
    (XIc).
  49. The method of claim 48, wherein the compound is selected from:
    Figure PCTKR2023008331-appb-img-000228
    ,
    or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  50. The method of claim 22, wherein the compound is of formula (Xc):
    Figure PCTKR2023008331-appb-img-000229
    (Xc)
    wherein -B- is selected from -NH-, -O-, -S-, and -SO2-.
  51. The method of claim 50, wherein the compound is selected from:
    Figure PCTKR2023008331-appb-img-000230
    Figure PCTKR2023008331-appb-img-000231
    ,
    or a solvate, a hydrate, a prodrug, and/or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  52. The method of any one of claims 1 to 51, wherein the compound is in a pharmaceutical composition, comprising the compound and at least one pharmaceutically acceptable carrier or excipient.
PCT/KR2023/008331 2022-06-15 2023-06-15 Methods of treating cancer using dual androgen receptor and pde5 inhibitor compounds WO2023244059A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263352583P 2022-06-15 2022-06-15
US63/352,583 2022-06-15

Publications (1)

Publication Number Publication Date
WO2023244059A1 true WO2023244059A1 (en) 2023-12-21

Family

ID=89191673

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/008331 WO2023244059A1 (en) 2022-06-15 2023-06-15 Methods of treating cancer using dual androgen receptor and pde5 inhibitor compounds

Country Status (1)

Country Link
WO (1) WO2023244059A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6878708B2 (en) * 2000-03-02 2005-04-12 Bayer Aktiengesellschaft Imidazotriazinones and the use thereof
US9604916B2 (en) * 2012-07-13 2017-03-28 Gtx, Inc. Method of treating androgen receptor (AR)-positive breast cancers with selective androgen receptor modulator (SARMs)
WO2022123310A1 (en) * 2020-12-11 2022-06-16 Ildong Pharmaceutical Co., Ltd. Novel compounds as androgen receptor and phosphodiesterase dual inhibitor

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6878708B2 (en) * 2000-03-02 2005-04-12 Bayer Aktiengesellschaft Imidazotriazinones and the use thereof
US9604916B2 (en) * 2012-07-13 2017-03-28 Gtx, Inc. Method of treating androgen receptor (AR)-positive breast cancers with selective androgen receptor modulator (SARMs)
WO2022123310A1 (en) * 2020-12-11 2022-06-16 Ildong Pharmaceutical Co., Ltd. Novel compounds as androgen receptor and phosphodiesterase dual inhibitor

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LIM JIN, PIAZZA GARY A, PAMUKCU RIFAT, THOMPSON W. JOSEPH, WEINSTEIN I. BERNARD: "Exisulind and Related Compounds Inhibit Expression and Function of the Androgen Receptor in Human Prostate Cancer Cells", CLINICAL CANCER RESEARCH, ASSOCIATION FOR CANCER RESEARCH, US, vol. 9, no. 13, 15 October 2003 (2003-10-15), US, pages 4972 - 4982, XP093118334, ISSN: 1078-0432 *
PENG, T. ET AL.: "Inhibitors of phosphodiesterase as cancer therapeutics", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 150, 2018, pages 742 - 756, XP055875167, DOI: 10.1016/j.ejmech.2018.03.046 *

Similar Documents

Publication Publication Date Title
WO2019190259A1 (en) Novel sulfonamide derivative having inhibitory effect on epidermal growth factor receptor mutation
WO2017188694A1 (en) Heteroaryl compound comprising nitrogen, and use thereof
WO2013081400A2 (en) Novel benzamide derivative and use thereof
WO2020149723A1 (en) Pyrrolopyrimidine derivative, and pharmaceutical composition for preventing or treating protein kinase-related disease comprising same as active ingredient
WO2016032209A2 (en) Substituted n-(pyrrolidine-3-yl)-7h-pyrrolo[2,3-d]pyrimidine-4-amine as janus kinase inhibitor
WO2020096372A1 (en) Novel piperidine-2,6-dione derivative and use of same
WO2022035303A1 (en) Novel dioxoloisoquinolinone derivatives and use thereof
WO2023018155A1 (en) Compound having shp2 protein degrading activity, and medical uses thereof
AU2021226297B2 (en) 1,3,4-oxadiazole derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
AU2021225683B2 (en) 1,3,4-oxadiazole derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
WO2012030170A2 (en) Novel compound acting as a cannabinoid receptor-1 inhibitor
WO2018151562A2 (en) Novel benzimidazole derivative having jnk inhibitory activity and use thereof
WO2016006974A2 (en) Novel triazolopyrimidinone or triazolopyridinone derivatives, and use thereof
WO2020022787A1 (en) Novel imidazole derivative having jnk inhibitory activity, and pharmaceutical composition comprising same
WO2023244059A1 (en) Methods of treating cancer using dual androgen receptor and pde5 inhibitor compounds
WO2022139304A1 (en) Novel quinazoline derivative compound as sos1 inhibitor, and use thereof
AU2019344240B2 (en) Novel thiazole derivatives and pharmaceutically acceptable salts thereof
WO2022019597A1 (en) Compound for androgen receptor degradation, and pharmaceutical use thereof
WO2020263058A1 (en) Aminocyanopyridine derivative and use thereof
WO2021040422A1 (en) Novel pyrimido[4,5-d]pyrimidine-2-one derivative having protein kinase inhibitory activity
EP3166945A2 (en) Novel triazolopyrimidinone or triazolopyridinone derivatives, and use thereof
WO2023191536A1 (en) Thiobenzimidazole derivative or pharmaceutically acceptable salt thereof, and use thereof
WO2023211256A1 (en) Novel pim kinase inhibitors and uses thereof
WO2022231377A1 (en) Novel compound and pharmaceutical composition comprising same for prevention or treatment of cancer, and use thereof
WO2023153748A1 (en) Sos1 inhibitor and derivative thereof

Legal Events

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

Ref document number: 23824272

Country of ref document: EP

Kind code of ref document: A1