[go: up one dir, main page]

WO2020047487A1 - Methods for treating cancer with rorgamma inhibitors and statins - Google Patents

Methods for treating cancer with rorgamma inhibitors and statins Download PDF

Info

Publication number
WO2020047487A1
WO2020047487A1 PCT/US2019/049178 US2019049178W WO2020047487A1 WO 2020047487 A1 WO2020047487 A1 WO 2020047487A1 US 2019049178 W US2019049178 W US 2019049178W WO 2020047487 A1 WO2020047487 A1 WO 2020047487A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
cancer
rory
combination
statin
Prior art date
Legal status (The legal status 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 status listed.)
Ceased
Application number
PCT/US2019/049178
Other languages
French (fr)
Inventor
Hongwu CHEN
Demin CAI
Yuqian Jiang
Junjian WANG
June X. Zou
Hongye ZOU
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of California Berkeley
University of California San Diego UCSD
Original Assignee
University of California Berkeley
University of California San Diego UCSD
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 University of California Berkeley, University of California San Diego UCSD filed Critical University of California Berkeley
Publication of WO2020047487A1 publication Critical patent/WO2020047487A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

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
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/366Lactones having six-membered rings, e.g. delta-lactones
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca

Definitions

  • the present invention provides a method for preventing or treating a cancer in a subject, the method comprising administering to the subject an effective amount of a RORy inhibitor and a statin.
  • the RORy inhibitor is a small- molecule compound, an anti-RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
  • the RORy inhibitor is selected from the group consisting of a compound according to Formula I:
  • GSK805, SR2211, a compound listed in Table 5, pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and solvates thereof, wherein X is C( 0) or S0 2; n is an integer selected from the group consisting of 0, 1, 2, or 3; Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF 3 )2, heterocyclyl, and cycloalkyl, wherein R 5 is selected from the group consisting of H, and C 1 -C 3 alkyl group; R 2 is selected from the group consisting of H, halogen, and alkyl; R 3 is selected from the group consisting of H and alkyl; R 4 is selected from the group consisting of C 0 -C 4 alkylene-R 6 , Co- C 4 alkylene-Rvcycl
  • the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP-43742, and a combination thereof.
  • the statin is selected form the group consisting of atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, pitavastatin, cerivastatin, pravastatin, and a combination thereof.
  • the cancer is resistant to an anticancer drug.
  • the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
  • the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
  • the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof.
  • the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
  • the cancer is selected from the group consisting of breast cancer, prostate cancer, lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, and glioma.
  • the breast cancer is a triple-negative breast cancer (TNBC), tamoxifen-resistant breast cancer, radiation-resistant breast cancer, HER2-positive breast cancer, or ER-positive breast cancer.
  • the prostate cancer is a castration-resistant prostate cancer.
  • the lung cancer is a non-small-cell lung cancer (NSCLC), K-Ras mutant lung cancer, BRAF mutant lung cancer, EGFR mutant lung cancer, tyrosine kinase inhibitor- resistant lung cancer, or small cell lung cancer (SCLC).
  • NSCLC non-small-cell lung cancer
  • K-Ras mutant lung cancer K-Ras mutant lung cancer
  • BRAF mutant lung cancer BRAF mutant lung cancer
  • EGFR mutant lung cancer tyrosine kinase inhibitor- resistant lung cancer
  • SCLC small cell lung cancer
  • the method further comprises administering the anti-cancer drug to the subject.
  • the subject is a human in need of cancer treatment.
  • administering the RORy inhibitor and the statin enhances the therapeutic effect of the anticancer drug.
  • administering the RORy inhibitor and the statin reverses or reduces cancer cell resistance to the anti cancer drug and/or sensitizes cancer cells to the anticancer drug.
  • administering the RORy inhibitor and the statin produces a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof.
  • enhancement of the anticancer drug therapeutic effect and/or the beneficial effect that is produced are greater when the RORy inhibitor and the statin are administered in combination compared to when the RORy inhibitor or the statin are administered alone.
  • administering the RORy inhibitor and the statin in combination produces a synergistic enhancement of the anticancer drug and/or produces a synergistic beneficial effect.
  • the cancer is sensitized to the statin. [0012] In some embodiments, the RORy inhibitor and the statin are administered concomitantly. In some embodiments, the RORy inhibitor and the statin are administered sequentially.
  • the present invention provides a composition comprising a RORy inhibitor and a statin.
  • the RORy inhibitor is a small-molecule compound, an anti-RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
  • the RORy inhibitor is selected from the group consisting of a compound according to Formula I:
  • GSK805, SR2211, a compound listed in Table 5, pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and solvates thereof, wherein X is C( 0) or S0 2; n is an integer selected from the group consisting of 0, 1, 2, or 3; Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF 3 )2, heterocyclyl, and cycloalkyl, wherein R5 is selected from the group consisting of H, and C1-C3 alkyl group; R 2 is selected from the group consisting of H, halogen, and alkyl; R 3 is selected from the group consisting of H and alkyl; R 4 is selected from the group consisting of C0-C4 alkylene-R 6 , Co- C 4 alkylene-Rvcycloalkyl
  • the RORy inhibitor selectively binds to RORy and inhibits RORy activity.
  • the compound of Formula I is represented by a compound according to any one of Formulas Ic to Ii:
  • the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP-43742, and a combination thereof.
  • the statin is selected form the group consisting of atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, pitavastatin, cerivastatin, pravastatin, and a combination thereof.
  • the composition comprises an effective amount of the RORy inhibitor and the statin. In some embodiments, the composition comprises an effective amount of the RORy inhibitor that is sufficient to sensitize a cancer to the statin. In some embodiments, the effective amount of the RORy inhibitor and the statin is sufficient to enhance an anticancer drug therapeutic effect and/or produce a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof.
  • enhancing the anticancer drug therapeutic effect comprises reversing or reducing cancer cell resistance to the anticancer drug and/or sensitizing cancer cells to the anticancer drug.
  • the enhancement of the anticancer drug therapeutic effect and/or the beneficial effect that is produced is greater when the RORy inhibitor and the statin are administered in combination compared to when the RORy inhibitor or the statin are administered alone.
  • the anticancer drug therapeutic effect is synergistically enhanced and/or the beneficial effect is synergistically produced when the RORy inhibitor and the statin are administered in combination.
  • the composition further comprises an anticancer drug.
  • the anticancer drug is selected from the group consisting of an anti androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
  • the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
  • the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof.
  • the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
  • the composition further comprises a pharmaceutically acceptable excipient or diluent.
  • the present invention provides a kit comprising a RORy inhibitor and a statin.
  • the RORy inhibitor is a small-molecule compound, an anti-RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
  • the RORy inhibitor is selected from the group consisting of a compound according to Formula I:
  • GSK805, SR2211, a compound listed in Table 5, pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and solvates thereof, wherein X is C( 0) or S0 2; n is an integer selected from the group consisting of 0, 1, 2, or 3; Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF 3 )2, heterocyclyl, and cycloalkyl, wherein R 5 is selected from the group consisting of H, and C 1 -C 3 alkyl group; R 2 is selected from the group consisting of H, halogen, and alkyl; R 3 is selected from the group consisting of H and alkyl; R 4 is selected from the group consisting of C 0 -C 4 alkylene-R 6 , Co- C 4 alkylene-Rvcycl
  • the RORy inhibitor selectively binds to RORy and inhibits RORy activity.
  • the compound of Formula I is represented by a compound according to any one of Formulas Ic to Ii:
  • the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP-43742, and a combination thereof.
  • the statin is selected form the group consisting of atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, pitavastatin, cerivastatin, pravastatin, and a combination thereof.
  • the kit comprises an effective amount of the RORy inhibitor and the statin. In some embodiments, the kit comprises an effective amount of the RORy inhibitor that is sufficient to sensitize the cancer to the statin. In some embodiments, the effective amount of the RORy inhibitor and the statin is sufficient to enhance an anticancer drug therapeutic effect and/or produce a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof. In some embodiments, enhancing the anticancer drug therapeutic effect comprises reversing or reducing cancer cell resistance to the anticancer drug and/or sensitizing cancer cells to the anticancer drug.
  • the enhancement of the anticancer drug therapeutic effect and/or the beneficial effect that is produced is greater when the RORy inhibitor and the statin are administered in combination compared to when the RORy inhibitor or the statin are administered alone.
  • the anticancer drug therapeutic effect is synergistically enhanced and/or the beneficial effect is synergistically produced when the RORy inhibitor and the statin are administered in combination.
  • the kit further comprises an anticancer drug.
  • the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
  • the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
  • the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof.
  • the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
  • the kit further comprises a label with instructions for administering the RORy inhibitor and the statin to the subject.
  • the subject is a human in need of cancer treatment.
  • the present invention provides a method for preventing or treating a cancer in a subject, the method comprising administering to the subject an effective amount of a RORy inhibitor, wherein the RORy inhibitor is a compound listed in Table 5, a pharmaceutically acceptable salt thereof, an isomer thereof, a racemate thereof, a prodrug thereof, a co-crystalline complex thereof, a hydrate thereof, or a solvate thereof.
  • the RORy inhibitor selectively binds to RORy and inhibits RORy activity.
  • the RORy inhibitor is VTP-43742.
  • the cancer is resistant to an anticancer drug.
  • the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
  • the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
  • the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof.
  • taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
  • the cancer is selected from the group consisting of prostate cancer, lung cancer, breast cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, and glioma.
  • the prostate cancer is a castration-resistant prostate cancer.
  • the lung cancer is a non-small-cell lung cancer (NSCLC), K-Ras mutant lung cancer, BRAF mutant lung cancer, EGFR mutant lung cancer, tyrosine kinase inhibitor-resistant lung cancer, or small cell lung cancer (SCLC).
  • the breast cancer is a triple-negative breast cancer (TNBC), tamoxifen-resistant breast cancer, radiation-resistant breast cancer, HER2-positive breast cancer, or ER-positive breast cancer.
  • the method further comprises administering the anti-cancer drug to the subject.
  • the subject is a human in need of cancer treatment.
  • administering the RORy inhibitor enhances the therapeutic effect of the anticancer drug.
  • administering the RORy inhibitor reverses or reduces cancer cell resistance to the anticancer drug and/or sensitizes cancer cells to the anticancer drug.
  • administering the RORy inhibitor produces a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof.
  • FIGS. 1A-1D show that RORy is required for cancer cell growth and survival of breast cancer.
  • FIG. 1B The TNBC cells were infected as in FIG. 1A. Three days later, cells were harvested for Western blotting analysis of indicated proteins with specific antibodies.
  • FIG. 1C The TNBC cells were transfected with control siRNA (siNeal) or two siRNAs against RORC.
  • FIG. 1D The HCC70 TNBC cells were transfected as in FIG. 1C. Three days later, cells were harvested for Western blotting analysis of indicated proteins with specific antibodies.
  • FIG. 2 shows that RORy is required for growth and survival of multiple different TNBC cell lines but not for non-malignant MCF10A cells.
  • FIG. 3 shows that RORy is required for growth and survival of mutant KRAS lung cancer cells.
  • FIGS. 4A-4C show that RORy inhibition suppresses the expression of genes controlling cholesterol synthesis.
  • FIG. 4A An abbreviated pathway of cholesterol synthesis showing statin and azole inhibitors and their corresponding target enzymes.
  • FIG. 4B Heat map display of fold changes (in log2) in gene expression of cholesterol biosynthesis pathway between cells treated by the indicated inhibitors and cells treated with vehicle control, as detected by RNA-seq analysis of HCC70 TNBC cells treated by vehicle, 2.5 mM RORy inhibitor XY018, or GSK805 for 48 hours.
  • FIG. 4C Western blotting analysis of proteins in cholesterol synthesis pathway with MDA-MB468 cells treated by indicated concentrations of RORy inhibitor XY018 and XY063.
  • FIG. 5 shows that RORy inhibition abolishes statin treatment-induced up-regulation of cholesterol synthesis and homeostasis genes.
  • FIGS. 6A-6D show that RORy inhibitors in combination with statins display strong synergy in the inhibition of TNBC cancer cell growth.
  • Breast cancer cells HCC70 FIG. 6A
  • MDA-MB468 cells FIGGS. 6B and 6D
  • SUM159 cells FIG. 6C
  • FIGS. 7A-7D show that RORy inhibitors in combination with statins display strong synergy in the killing of TNBC cancer cells.
  • FIGS. 8A and 8B show that RORy inhibitors in combination with statins display strong synergy in the inhibition of lung cancer cell growth.
  • A549 lung cancer cells were treated by indicated concentrations of fluvastatin (FIG. 8A) or lovastatin (FIG. 8B), either alone or in combination with indicated concentrations of RORy inhibitor XY018 or XY063 for 4 days.
  • FIGS. 9A-9D show that RORy inhibitors in combination with statins display strong synergy in the inhibition of CRPC prostate cancer cell growth.
  • CRPC prostate cancer cells 22Rvl and C4-2B cells were treated by indicated concentrations of atorvastatin (FIGS. 9B and 9D, respectively) or simvastatin (FIGS. 9 A and 9C, respectively), either alone or in combination with indicated concentrations of RORy inhibitor XY018 or XY063 for 4 days.
  • FIGS. 10A-10I show that RORy inhibitors in combination with statins display strong synergy in the inhibition of gastric cancer cell growth.
  • FIGS. 10 A, 10B, and 10C show cell viability assays of AGS, HGC-27, and MGC-803 gastric cancer cells, respectively, that were treated by indicated concentrations of RORy inhibitors GSK805, XY018, or SR2211 for 4 days. The cell viability was measured by CellTiter-Glo.
  • FIGS. 10D-10I show cell viability assays of gastric cancer cells that were treated by atorvastatin (AGS, HGC-27, and MGC-803 cells shown in FIGS.
  • FIGS. 11A-11D show that RORy inhibitor VTP -43742 in combination with statins displays strong synergy in the inhibition of breast cancer and prostate cancer cell growth.
  • FIGS. 11A and 11B cells of the HCC70 triple-negative breast cancer (TNBC) cell model were treated by the indicated statin (simvastatin (FIG. 11 A) or atorvastatin (FIG. 11B)) at increasing concentrations (from 0.625 pM to 10 pM) either alone (plus vehicle control) or with 2.5 pM RORy inhibitor/antagonist VTP -43742. Cells were treated for 4 days.
  • FIG. 11C and 11D cells of the C4-2B castration-resistant prostate cancer (CRPC) cell model were treated by the indicated statin (simvastatin (FIG. 11C) or atorvastatin (FIG. 11D)) at increasing concentrations (from 0.625 pM to 10 pM) either alone (plus vehicle control) or with 2.5 pM RORy inhibitor/antagonist VTP-43742. Cells were treated for 4 days. For data shown in all panels, cell viability was measured by CellTiter-GLO. The cell viability obtained from cells treated with vehicle only ( i.e without either the statin or the ROR-y inhibitor/antagonist) was set as 100. The experiments were repeated three times and in sexplicate.
  • statin statin
  • FIG. 11D atorvastatin
  • FIGS. 12A and 12B show that inhibition of RORy with small-interfering RNA (siRNA) strongly sensitizes prostate cancer and breast cancer cells to killing by statins.
  • Cells of the MDA-MB468 triple-negative breast cancer (TNBC) cell model or the C4-2B castration-resistant prostate cancer (CRPC) cell model were first transfected with 5 nM of control siRNA or two RORC gene siRNAs (si-RORC-l and siRORC-2) as previously reported in J. Wang et al, Nature Medicine , 22, 488-496 (2016).
  • FIGS. 13A-13D show that oral administration of RORy inhibitors in combination with statins suppresses breast cancer tumor growth more effectively than administration of either alone.
  • Mammary gland-orthotopic xenograft tumors were established by implanting breast cancer MDA-MB468 cells into mammary fat pads of SCID mice. Mice were treated 5 times per week with vehicle, RORy inhibitors alone (XY018 10 mg/kg p.o. or XY063 10 mg/kg p.o.), atorvastatin alone (20 mg/kg p.o.), or a combination of RORy inhibitors with atorvastatin (XY018 10 mg/kg p.o. + Atorva. 20 mg/kg p.o.
  • FIG. 13 A shows tumor volume.
  • FIG. 13B shows tumor weight.
  • FIG. 13C shows representative images or tumors.
  • FIG. 13D shows mouse body weight. Significance was calculated using Student’s / test. * P ⁇ 0.001, ** P ⁇ 0.0005, *** P ⁇ 0.0001.
  • FIGS. 14A-14D show that RORy inhibitors in combination with statins suppress lung tumor growth more effectively than administration of either alone.
  • A549 lung xenograft tumors were established by subcutaneous (s.c.) injection of A549 cells into NOD-SCID female mice. When tumors reached approximately 100 mm 3 , mice were randomized into groups for treatment by either vehicle, intraperitoneal (i.p.) injection of XY018 (5 mg/kg, five times per week), oral gavage of atorvastatin (20 mg/kg, five times per week), or a combination of XY018 and atorvastatin (five times per week).
  • FIG. 14A shows tumor volume.
  • FIG. 14B shows tumor weight.
  • FIG. 14C shows representative images or tumors.
  • FIG. 14D shows mouse body weight.
  • FIG. 15 shows the structures of various statins.
  • FIGS. 16A-16C show that inhibition of RORy with antagonists XY018 and XY063 strongly sensitizes lung cancer cells to killing by different statins.
  • FIG. 16A shows the combination index (Cl) and supporting data.
  • FIG. 16B shows viability of A549 cells using fluvastatin and XY018 or XY063.
  • FIG. 16C shows viability of A549 cells using lovastatin and XY018 or XY063. Data shown are mean ⁇ s.d. ** P ⁇ 0.002; Student’s t test.
  • FIGS. 17A-17C show that combined treatment with RORy inhibitors/antagonists and statins is synergistic in the inhibition of lung cancer tumor growth.
  • FIG. 17A shows tumor size data.
  • FIG. 17B shows tumor weight data.
  • FIG. 17C shows images of representative tumors. Significance was calculated using Student’s t test. * P ⁇ 0.05, *** P ⁇ 0 01
  • FIGS. 18A and 18B show that a combined treatment of cancer cells with RORy inhibitor and statin synergistically inhibits the oncogenic kinase signaling by receptor tyrosine kinase (RTK) and serine/threonine kinase.
  • FIG. 18A shows data from PC9 cells.
  • FIG. 18B shows data from A549 cells.
  • FIG. 19 shows biochemical analysis of serum samples for the assessment of significant alteration of key blood/serum biochemical parameters or toxicity indicators.
  • FIGS. 20A-20K shows that RORy inhibitors alone or synergize with statins strongly inhibit TNBC tumor growth in vivo.
  • FIG. 20A Mice bearing MDA-MB468 were treated, p.o., 5 times per week, with vehicle or indicated doses of RORy inhibitor XY018 for 52 days. Tumor volume (top), representative images (middle) and tumor weight (
  • FIG. 20E Mice bearing MCF-7 were treated, i.p.
  • mice bearing PDX-1079 or PDX- 1173 treated, 5 times per week, p.o., with vehicle, 20 mg/kg XY018 alone, 15 mg/kg ATV alone, or both XY018 + ATV for indicated days n 7 mice per group.
  • FIG. 20J: PDX-1079 were treated, 5 times per week, p.o., with vehicle, 20 mg/kg XY018 alone, l5mg/kg simvastatin alone (SIM) or both of XY018 + SIM for 47 days. Tumor weight is shown n 7.
  • FIGS. 21A-21G show that RORy inhibitors alone or with statins cause tumor regression and block metastasis.
  • MV A mevalonate pathway
  • SREBP1 and -2 transcription factors sterol regulatory element-binding protein 1 and 2
  • LXRs 2, 5
  • Statins are a class of cholesterol/lipid-lowering drugs used for treating patients with hypercholesterolemia to reduce the associated high risk of cardiovascular disease (CVD) (1).
  • Statin treatment strongly reduces cholesterol production initially, and, as an adaptive response, the reduced cellular cholesterol level triggers the SREBP-mediated activation of gene expression including that of LDLR in liver and other tissues, which leads to an increased uptake of LDL from the circulation, hence lowering blood cholesterol level.
  • Epidemiological studies comparing cancer patients receiving statins for their CVD risk to those not receiving statins suggest that the use of statins is associated with a statistically significant reduction of the risk of cancer relapse and cancer-related death.
  • the present invention is based, in part, on the discovery that RORy inhibitors effectively sensitize cancer cells to statin-induced cell death and growth inhibition, and that a combination treatment with RORy inhibitors and statins results in highly synergistic killing of cancer cells of multiple cancer types.
  • retinoic acid receptor-related orphan receptor y and“RORy” refers to either or both isoforms encoded by the RORC (RAR-related orphan receptor C; see, e.g., NCBI Gene ID 6097 for the human gene) gene, namely RORy (also referred to as RORyl or RORC1; see, e.g, NCBI Reference Sequence NP 005051.2 for the human sequence) and RORyt (also known as RORy 2 or RORC2; see, e.g., NCBI Reference Sequence NP_001001523.1 for the human sequence).
  • RORC RAR-related orphan receptor C
  • RORy also referred to as RORyl or RORC1
  • RORyt also known as RORy 2 or RORC2; see, e.g., NCBI Reference Sequence NP_001001523.1 for the human sequence
  • the terms“subject”,“patient” or“individual” are used herein interchangeably to include a human or animal.
  • the animal subject may be a mammal, a primate (e.g, a monkey), a livestock animal (e.g, a horse, a cow, a sheep, a pig, or a goat), a companion animal (e.g, a dog, a cat), a laboratory test animal (e.g, a mouse, a rat, a guinea pig, a bird), an animal of veterinary significance, or an animal of economic significance.
  • a primate e.g, a monkey
  • livestock animal e.g, a horse, a cow, a sheep, a pig, or a goat
  • a companion animal e.g, a dog, a cat
  • a laboratory test animal e.g, a mouse, a rat, a guinea pig, a bird
  • an animal of veterinary significance e.
  • an effective amount includes a dosage sufficient to produce a desired result with respect to the indicated disorder, condition, or mental state.
  • the desired result may comprise a subjective or objective improvement in the recipient of the dosage.
  • an effective amount of a RORy inhibitor and a statin includes an amount sufficient to alleviate the signs, symptoms, or causes of a cancer, e.g., a therapeutically or prophylactically effective amount.
  • an effective amount can be an amount that slows or reverses tumor growth (e.g, slows tumor size), increases mean time of survival, inhibits tumor progression or metastasis, or sensitizes a cancer cell to an anticancer drug to which it has become or is resistant.
  • an effective amount of a RORy inhibitor and a statin includes an amount sufficient to cause a substantial improvement in a subject having cancer when administered to the subject.
  • the amount will vary with the type of cancer being treated, the stage of advancement of the cancer, the type and concentration of composition applied, and the amount of an anticancer drug (e.g, anti-androgen drug) that is also administered to the subject.
  • an effective amount of a RORy inhibitor and a statin can include an amount that is effective in enhancing the therapeutic activity of anticancer drugs such as anti-androgen drugs (e.g, bicalutamide, enzalutamide, arbiraterone, etc.) and/or chemotherapeutic agents (e.g, tamoxifen and/or taxanes such as docetaxel), and/or sensitizing a cancer (e.g, a cancer cell) to an anticancer drug or other therapy.
  • an effective amount e.g. of a RORy inhibitor
  • an effective amount is an amount that is sufficient to sensitize a cancer to a statin.
  • small-molecule compound refers to a compound or drug that has a low molecular weight.
  • the small-molecule compound may be a small organic molecule, either naturally occurring or synthetic.
  • the small-molecule compound has a molecular weight of more than about 50 Daltons and less than about 1,500 Daltons, e.g., between about 100 Daltons and about 1,000 Daltons or between about 200 Daltons and about 600 Daltons.
  • interfering RNA or“RNAi” or“interfering RNA sequence” as used herein includes single-stranded RNA (e.g ., mature miRNA, ssRNAi oligonucleotides, ssDNAi oligonucleotides), double-stranded RNA (i.e., duplex RNA such as siRNA, Dicer- substrate dsRNA, shRNA, aiRNA, or pre-miRNA), a DNA-RNA hybrid (see, e.g., PCT Publication No. WO 2004/078941), or a DNA-DNA hybrid (see, e.g., PCT Publication No.
  • Interfering RNA thus refers to the single-stranded RNA that is complementary to a target mRNA sequence or to the double-stranded RNA formed by two complementary strands or by a single, self-complementary strand.
  • Interfering RNA may have substantial or complete identity to the target gene or sequence, or may comprise a region of mismatch (i.e., a mismatch motif).
  • the sequence of the interfering RNA can correspond to the full-length target gene, or a subsequence thereof.
  • the interfering RNA molecules are chemically synthesized.
  • Interfering RNA includes“small-interfering RNA” or“siRNA,” e.g., interfering RNA of about 15-60, 15-50, or 15-40 (duplex) nucleotides in length, more typically about 15- 30, 15-25, or 19-25 (duplex) nucleotides in length, and is preferably about 20-24, 21-22, or 21-23 (duplex) nucleotides in length (e.g, each complementary sequence of the double- stranded siRNA is 15-60, 15-50, 15-40, 15-30, 15-25, or 19-25 nucleotides in length, preferably about 20-24, 21-22, or 21-23 nucleotides in length, and the double-stranded siRNA is about 15-60, 15-50, 15-40, 15-30, 15-25, or 19-25 base pairs in length, preferably about 18-22, 19-20, or 19-21 base pairs in length).
  • siRNA e.g., interfering RNA of about 15-60, 15-50, or 15-40 (duplex) nucle
  • siRNA duplexes may comprise 3’ overhangs of about 1 to about 4 nucleotides or about 2 to about 3 nucleotides and 5’ phosphate termini.
  • siRNA includes RNA-RNA duplexes as well as DNA-RNA hybrids (see, e.g., PCT Publication No. WO 2004/078941).
  • siRNA can be chemically synthesized or can be generated by cleavage of longer dsRNA (e.g, dsRNA greater than about 25 nucleotides in length), e.g., with the E. coli RNase III or Dicer. These enzymes process the dsRNA into biologically active siRNA (see, e.g., Yang el al, Proc. Natl. Acad. Sci. USA, 99:9942-9947 (2002); Calegari el al, Proc. Natl. Acad. Sci.
  • dsRNA are at least 50 nucleotides to about 100, 200, 300, 400, or 500 nucleotides in length.
  • a dsRNA may be as long as 1000, 1500, 2000, 5000 nucleotides in length, or longer.
  • the dsRNA can encode an entire gene transcript or a partial gene transcript.
  • mismatch motif or“mismatch region” refers to a portion of an interfering RNA sequence that does not have 100% complementarity to its target sequence.
  • An interfering RNA may have at least one, two, three, four, five, six, or more mismatch regions.
  • the mismatch regions may be contiguous or may be separated by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more nucleotides.
  • the mismatch motifs or regions may comprise a single nucleotide or may comprise two, three, four, five, or more nucleotides.
  • the term“treating” includes, but is not limited to, methods and manipulations to produce beneficial changes in a recipient's health status, e.g. , a patient’s cancer status.
  • the changes can be either subjective or objective and can relate to features such as symptoms or signs of the cancer being treated. For example, if the patient notes decreased pain, then successful treatment of pain has occurred. For example, if a decrease in the amount of swelling has occurred, then a beneficial treatment of inflammation has occurred. Similarly, if the clinician notes objective changes, such as reducing the number of cancer cells, the growth of the cancer cells, the size of cancer tumors, or the resistance of the cancer cells to another cancer drug, then treatment of cancer has also been beneficial.
  • Treating also includes administering combination of a RORy inhibitor and a statin alone with or without an anticancer drug to a subject having cancer.
  • the cancer is breast cancer, prostate cancer, lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or glioblastoma multiforme.
  • administering includes activities associated with providing a patient an amount of a compound described herein, e.g, a RORy inhibitor and a statin.
  • Administering includes providing unit dosages of compositions set forth herein to a patient in need thereof.
  • Administering includes providing effective amounts of compounds for a specified period of time, e.g, for about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or more days, or in a specified sequence, e.g. , administration of a RORy inhibitor and a statin followed by the administration of one or more anticancer drugs, or vice versa.
  • the term“co-administering” includes sequential or simultaneous administration of two or more structurally different compounds.
  • two or more structurally different pharmaceutically active compounds can be co-administered by administering a pharmaceutical composition adapted for oral administration that contains two or more structurally different active pharmaceutically active compounds.
  • two or more structurally different compounds can be co-administered by administering one compound and then administering the other compound.
  • the co-administered compounds are administered by the same route.
  • the co-administered compounds are administered via different routes.
  • one compound can be administered orally, and the other compound can be administered, e.g, sequentially or simultaneously, via intravenous or intraperitoneal injection.
  • cancer refers to conditions including solid cancers, lymphomas, and leukemias.
  • types of cancers include, but are not limited to, prostate cancer, lung cancer (e.g, non-small cell lung cancer or NSCLC), ovarian cancer, colorectal cancer, liver cancer (i.e., hepatocarcinoma), renal cancer (i.e., renal cell carcinoma), bladder cancer, breast cancer, thyroid cancer, pleural cancer, pancreatic cancer, uterine cancer, cervical cancer, testicular cancer, anal cancer, bile duct cancer, gastrointestinal carcinoid tumors, esophageal cancer, gall bladder cancer, appendix cancer, small intestine cancer, stomach (gastric) cancer, cancer of the central nervous system, skin cancer, choriocarcinoma, head and neck cancer, blood cancer, endometrial cancer, osteogenic sarcoma, fibrosarcoma, neuroblastoma, glioma, mela
  • the cancer can be metastatic.
  • the cancer is prostate cancer, lung cancer, breast cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma such as glioblastoma multiforme.
  • the cancer can be resistant to an anticancer drug, e.g, an anti-androgen-resistant cancer, a taxane-resistant cancer (e.g, docetaxel- resistant cancer), a tamoxifen-resistant cancer, a radiation-resistant cancer, or a tyrosine kinase inhibitor-resistant cancer.
  • the terms“prostate cancer” or“prostate cancer cell” refer to a cancer cell or cells that reside in prostate tissue.
  • the prostate cancer can be benign, malignant, or metastatic.
  • the prostate cancer can be androgen-insensitive, hormone-resistant, or castrate-resistant.
  • the prostate cancer can be“advanced stage prostate cancer” or “advanced prostate cancer.”
  • Advanced stage prostate cancer includes a class of prostate cancers that has progressed beyond early stages of the disease. Typically, advanced stage prostate cancers are associated with a poor prognosis.
  • Types of advanced stage prostate cancers include, but are not limited to, metastatic prostate cancer, drug-resistant prostate cancer such as anti -androgen -resistant prostate cancer (e.g ., enzalutamide-resistant prostate cancer, abiraterone-resistant prostate cancer, bicalutamide-resistant prostate cancer, etc.), taxane-resistant prostate cancer (e.g., docetaxel-resistant prostate cancer) and the like, hormone refractory prostate cancer, castration-resistant prostate cancer (CRPC), metastatic castration-resistant prostate cancer, AR-V7-induced drug-resistant prostate cancer such as AR-V7-induced anti-androgen-resistant prostate cancer (e.g, AR-V7-induced enzalutamide- resistant prostate cancer), AKR1C3 -induced drug-resistant prostate cancer such as AKR1C3- induced anti -androgen-resistant prostate cancer (e.g, AKR1C3 -induced enzalutamide- resistant prostate cancer), and combinations thereof.
  • the advanced stage prostate cancers do not generally respond, or are resistant, to treatment with one or more of the following conventional prostate cancer therapies: enzalutamide, abiraterone, bicalutamide, and docetaxel.
  • Compounds, compositions, and methods of the present invention are provided for treating prostate cancer, such as advanced stage prostate cancer, including any one or more (e.g, two, three, four, five, six, seven, eight, nine, ten, or more) of the types of advanced stage prostate cancers disclosed herein.
  • the phrase“enhancing the therapeutic effects” includes any of a number of subjective or objective factors indicating a beneficial response or improvement of the condition being treated as discussed herein.
  • enhancing the therapeutic effects of an anticancer drug such as an anti -androgen drug (e.g, enzalutamide, abiraterone, or bicalutamide) or a chemotherapeutic agent such as tamoxifen or a taxane (e.g, docetaxel) includes reversing or reducing cancer cell resistance and/or sensitizing a drug-resistant cancer to the anticancer drug therapy.
  • enhancing the therapeutic effects of an anticancer drug includes altering drug-resistant cancer cells so that the cells are not resistant to the anticancer drug.
  • enhancing the therapeutic effects of an anticancer drug includes additively or synergistically improving or increasing the activity of the anticancer drug.
  • the enhancement includes a one-fold, two-fold, three-fold, five-fold, ten-fold, twenty-fold, fifty-fold, hundred-fold, or thousand-fold increase in the therapeutic activity of an anticancer drug used to treat cancer.
  • the phrase“reversing cancer cell resistance” includes altering or modifying a cancer cell that is resistant to anticancer drug therapy so that the cell is no longer resitant to anticancer drug therapy.
  • the phrase“reducing cancer cell resistance” includes increasing the therapeutic activity of an anticancer drug towards cancer cells that are, or previously were, resistant to anticancer drug therapy.
  • the phrase“sensitizing cancer cell resistance” includes inducing sensitization towards anticancer drug therapy in cancer cells that are resistant to anticancer drug therapy. Sensitization as used herein includes inducing or enhancing the ability of a cancer cell to be effectively treated with an anticancer drug. Sensitization also includes reducing the dosage required to achieve a beneficial effect with an anticancer drug.
  • anti-androgen drug includes anti-androgen compounds that alter the androgen pathway by blocking the androgen receptors, competing for binding sites on the cell’s surface, or affecting or mediating androgen production.
  • Anti-androgen drugs are useful for treating several diseases including, but not limited to, prostate cancer.
  • Anti-androgen drugs include, but are not limited to, enzalutamide, abiraterone, bicalutamide, flutamide, nilutamide, apalutamide, finasteride, dutasteride, alfatradiol, and combinations thereof.
  • AR androgen receptor
  • AREs Androgen Response Elements
  • AR variant includes a splice variant of full-length AR.
  • Various AR variants are known. See , Guo el al ., Cancer Res., 69(6):2305-l3 (2009).
  • Exemplary AR variants include, but are not limited to, variants lacking a functional ligand binding domain (LBD).
  • LBD functional ligand binding domain
  • An example of an AR variant that lacks an LBD is AR-V7.
  • AR- V7 includes androgen receptor splice variant 7, a contituitively active variant of an AR that lacks a functional ligand binding domain (LBD). See , e.g., Hu el al. , Cancer Research , 69(1): 16-22 (2009).
  • “Pharmaceutically acceptable” or“therapeutically acceptable” includes a substance which does not interfere with the effectiveness or the biological activity of the active ingredients and which is not toxic to the hosts in the amounts used, and which hosts may be either humans or animals to which it is to be administered.
  • Alkyl refers to a straight or branched, saturated, aliphatic radical having the number of carbon atoms indicated. Alkyl can include any number of carbons, such as Ci- 2 , Ci-3, Ci-4, Ci-5, Ci- 6 , Ci-7, Ci-8, Ci-9, Ci-io, C 2 - 3 , C 2-4 , C 2 -5, C 2 - 6 , C 3-4 , C 3-5 , C 3-6 , C 4-5 , C 4 - 6 and C5-6.
  • Ci -6 alkyl includes, but is not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, hexyl, etc.
  • Alkyl can also refer to alkyl groups having up to 20 carbons atoms, such as, but not limited to heptyl, octyl, nonyl, decyl, etc. Alkyl groups can be substituted or unsubstituted.
  • Alkylene refers to a straight or branched, saturated, aliphatic radical having the number of carbon atoms indicated, and linking at least two other groups, i.e., a divalent hydrocarbon radical.
  • the two moieties linked to the alkylene can be linked to the same atom or different atoms of the alkylene group.
  • a straight chain alkylene can be the bivalent radical of -(CH2) n - where n is 1, 2, 3, 4, 5 or 6.
  • Representative alkylene groups include, but are not limited to, methylene, ethylene, propylene, isopropylene, butylene, isobutylene, sec-butylene, pentylene and hexylene.
  • Alkylene groups can be substituted or unsubstituted.
  • Alkenyl refers to a straight chain or branched hydrocarbon having at least 2 carbon atoms and at least one double bond. Alkenyl can include any number of carbons, such as C 2 , C2-3, C2-4, C2-5, C2-6, C2-7, C2-8, C2-9, C2-10, C3, C3-4, C3-5, C3-6, C 4 , C4-5, C4-6, C5, C5-6, and C 6. Alkenyl groups can have any suitable number of double bonds, including, but not limited to, 1, 2, 3, 4, 5 or more.
  • alkenyl groups include, but are not limited to, vinyl (ethenyl), propenyl, isopropenyl, l-butenyl, 2-butenyl, isobutenyl, butadienyl, l-pentenyl, 2-pentenyl, isopentenyl, l,3-pentadienyl, l,4-pentadienyl, l-hexenyl, 2-hexenyl, 3-hexenyl, l,3-hexadienyl, l,4-hexadienyl, l,5-hexadienyl, 2,4-hexadienyl, or l,3,5-hexatrienyl.
  • Heteroalkyl refers to an alkyl group of any suitable length and having from 1 to 3 heteroatoms such as N, O and S. Additional heteroatoms can also be useful, including, but not limited to, B, Al, Si and P. The heteroatoms can also be oxidized, such as, but not limited to, -S(O)- and -S(0) 2 -.
  • heteroalkyl can include ethers, thioethers and alkyl-amines.
  • the heteroatom portion of the heteroalkyl can replace a hydrogen of the alkyl group to form a hydroxy, thio or amino group.
  • the heteroatom portion can be the connecting atom, or be inserted between two carbon atoms.
  • Haloalkyl refers to an alkyl group, where some or all of the hydrogen atoms are replaced with halogen atoms.
  • alkyl group haloalkyl groups can have any suitable number of carbon atoms, such as Ci- 6.
  • haloalkyl includes trifluoromethyl, fluoromethyl, etc.
  • perfluoro can be used to define a compound or radical where all the hydrogens are replaced with fluorine.
  • perfluoromethyl refers to 1,1,1 -trifluoromethyl.
  • Alkoxy refers to an alkyl group having an oxygen atom that connects the alkyl group to the point of attachment: alkyl-O-.
  • alkyl group alkoxy groups can have any suitable number of carbon atoms, such as Ci -6.
  • Alkoxy groups include, for example, methoxy, ethoxy, propoxy, iso-propoxy, butoxy, 2-butoxy, iso-butoxy, sec-butoxy, tert-butoxy, pentoxy, hexoxy, etc.
  • the alkoxy groups can be further substituted with a variety of substituents described within. Alkoxy groups can be substituted or unsubstituted.
  • Aryl refers to an aromatic ring system having any suitable number of ring atoms and any suitable number of rings.
  • Aryl groups can include any suitable number of ring atoms, such as, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or 16 ring atoms, as well as from 6 to 10, 6 to 12, or 6 to 14 ring members.
  • Aryl groups can be monocyclic, fused to form bicyclic or tricyclic groups, or linked by a bond to form a biaryl group.
  • Representative aryl groups include phenyl, naphthyl and biphenyl. Other aryl groups include benzyl, having a methylene linking group.
  • aryl groups have from 6 to 12 ring members, such as phenyl, naphthyl or biphenyl. Other aryl groups have from 6 to 10 ring members, such as phenyl or naphthyl. Some other aryl groups have 6 ring members, such as phenyl.
  • Aryl groups can be substituted or unsubstituted.
  • Heteroaryl refers to a monocyclic or fused bicyclic or tricyclic aromatic ring assembly containing 5 to 16 ring atoms, where from 1 to 5 of the ring atoms are a heteroatom such as N, O or S. Additional heteroatoms can also be useful, including, but not limited to, B, Al, Si and P. The heteroatoms can also be oxidized, such as, but not limited to, -S(O)- and -S(0) 2 -. Heteroaryl groups can include any number of ring atoms, such as, 3 to 6, 4 to 6, 5 to 6, 3 to 8, 4 to 8, 5 to 8, 6 to 8, 3 to 9, 3 to 10, 3 to 11, or 3 to 12 ring members.
  • heteroaryl groups can have from 5 to 8 ring members and from 1 to 4 heteroatoms, or from 5 to 8 ring members and from 1 to 3 heteroatoms, or from 5 to 6 ring members and from 1 to 4 heteroatoms, or from 5 to 6 ring members and from 1 to 3 heteroatoms.
  • the heteroaryl group can include groups such as pyrrole, pyridine, imidazole, pyrazole, triazole, tetrazole, pyrazine, pyrimidine, pyridazine, triazine (1,2,3-, 1,2,4- and 1,3, 5-isomers), thiophene, furan, thiazole, isothiazole, oxazole, and isoxazole.
  • heteroaryl groups can also be fused to aromatic ring systems, such as a phenyl ring, to form members including, but not limited to, benzopyrroles such as indole and isoindole, benzopyridines such as quinoline and isoquinoline, benzopyrazine (quinoxaline), benzopyrimidine (quinazoline), benzopyridazines such as phthalazine and cinnoline, benzothiophene, and benzofuran.
  • Other heteroaryl groups include heteroaryl rings linked by a bond, such as bipyridine. Heteroaryl groups can be substituted or unsubstituted.
  • Cycloalkyl refers to a saturated or partially unsaturated, monocyclic, fused bicyclic or bridged polycyclic ring assembly containing from 3 to 12 ring atoms, or the number of atoms indicated. Cycloalkyl can include any number of carbons, such as C 3-6 , C 4 - 6 , C5- 6 , C3-8, C4-8, C5- 8 , C 6 - 8 , C3- 9 , C 3 -1 0 , C 3 -11, and C 3 -12.
  • Saturated monocyclic cycloalkyl rings include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cyclooctyl.
  • Saturated bicyclic and polycyclic cycloalkyl rings include, for example, norbomane, [2.2.2] bicyclooctane, decahydronaphthalene and adamantane. Cycloalkyl groups can also be partially unsaturated, having one or more double or triple bonds in the ring.
  • Representative cycloalkyl groups that are partially unsaturated include, but are not limited to, cyclobutene, cyclopentene, cyclohexene, cyclohexadiene (1,3- and 1, 4-isomers), cycloheptene, cycloheptadiene, cyclooctene, cyclooctadiene (1,3-, 1,4- and 1, 5-isomers), norbomene, and norbomadiene.
  • exemplary groups include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
  • exemplary groups include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Cycloalkyl groups can be substituted or unsubstituted.
  • Heterocycloalkyl refers to a saturated or partially unsaturated ring system having from 3 to 13 ring members and from 1 to 4 heteroatoms of N, O and S. Heterocycloalkyl groups can include fused bi- or tri- cyclic systems, and can include one or more points of unsaturation. Additional heteroatoms can also be useful, including, but not limited to, B, Al, Si and P. The heteroatoms can also be oxidized, such as, but not limited to, -S(O)- and -S(0) 2 -.
  • Heterocycloalkyl groups can include any number of ring atoms, such as, 3 to 6, 4 to 6, 5 to 6, 3 to 8, 4 to 8, 5 to 8, 6 to 8, 3 to 9, 3 to 10, 3 to 11, 3 to 12 or 3 to 13 ring members. Any suitable number of heteroatoms can be included in the heterocycloalkyl groups, such as 1, 2, 3, or 4, or 1 to 2, 1 to 3, 1 to 4, 2 to 3, 2 to 4, or 3 to 4.
  • the heterocycloalkyl group can include groups such as aziridine, azetidine, pyrrolidine, piperidine, azepane, azocane, quinuclidine, pyrazolidine, imidazolidine, piperazine (1,2-, 1,3- and 1, 4-isomers), oxirane, oxetane, tetrahydrofuran, oxane (tetrahydropyran), oxepane, thiirane, thietane, thiolane (tetrahydrothiophene), thiane (tetrahydrothiopyran), oxazolidine, isoxazolidine, thiazolidine, isothiazolidine, dioxolane, dithiolane, or morpholine.
  • groups such as aziridine, azetidine, pyrrolidine, piperidine, azepane, azocane, quinuclidine, pyra
  • heterocycloalkyl groups can also be fused to aromatic or non-aromatic ring systems to form members including, but not limited to, indoline.
  • Heterocycloalkyl groups can be unsubstituted or substituted.
  • heterocycloalkyl groups can be linked via any position on the ring.
  • piperidine can be 1-, 2-, 3- or 4-piperidine
  • pyrazolidine can be 1-, 2-, 3-, or 4- pyrazolidine
  • imidazolidine can be 1-, 2-, 3- or 4-imidazolidine
  • piperazine can be 1-, 2-, 3- or 4-piperazine
  • morpholine can be 1-, 2-, 3- or 4-morpholine.
  • the term“synergy” or“synergistic effect” refers to an effect produced by two or more compounds (e.g a RORy inhibitor and a statin) that is greater than the effect produced by a sum of the effects of the individual compounds (i.e., an effect that is greater than an additive effect).
  • a combination of drugs produces a synergistic effect.
  • the Highest Single Agent approach simply reflects that the fact that the resulting effect of a combination of drugs (EAB) is greater than the effects of the individual drugs (EA and EB).
  • a combination index (Cl) can be calculated according to the formula:
  • the Bliss Independence model is based on the principle that drug effects are the outcomes of probabilistic processes, and makes the assumption that drugs act independently such that they do not interfere with each other (i.e., different sites of action). However, the model also assumes that each drug contributes to the production of a common result. According to this method, the observed combination effect is expressed as a probability (0 ⁇ EA B £ 1) and is compared to the expected additive effect expressed as:
  • EA + EB (1 -EA) EA + EB - EAEB, where 0 ⁇ EA £ 1 and 0 ⁇ E B ⁇ 1.
  • the Cl for this method is calculated using the formula:
  • compositions, methods, and kits comprising one or more RORy inhibitors (e.g., a small-molecule compound such as a compound of Formula I ( e.g. , XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statin drugs (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) that are useful for treating cancer, e.g.
  • RORy inhibitors e.g., a small-molecule compound such as a compound of Formula I ( e.g. , XY018, XY063), GSK805, SR2211, and/or a
  • compositions, methods and kits further comprise an anticancer drug such as an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen- specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, or a combination thereof.
  • an anticancer drug such as an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen- specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, or a combination thereof.
  • the compositions, methods, and kits comprise an effective amount of the RORy inhibitor, statin, and/or anticancer drug.
  • RORy inhibitors used in compositions, methods, and kits of the present invention specifically bind to RORy and inhibit RORy activity.
  • Receptor-related orphan receptor y (RORy) inhibitors include small-molecule compounds that inhibit retinoic acid receptor-related orphan receptor y (RORy) transcription, translation, stability, and/or activity.
  • a RORy inhibitor binds to RORy and inhibits the activity of the receptor.
  • a RORy inhibitor selectively binds to RORy.
  • a RORy inhibitor selectively binds to RORy relative to RORa and/or RORp.
  • a RORy inhibitor selectively binds to RORy and inhibits RORy activity.
  • a RORy inhibitor inhibits RORy activity relative to RORa and/or RORp.
  • inhibition of RORy activity includes inhibition of recruitment of coactivators such as SRC-l and/or SRC-3 to an androgen receptor (AR) ROR response element (RORE).
  • AR androgen receptor
  • inhibition of RORy activity includes inhibition of transcription of the AR gene and/or a variant thereof such as AR-V7.
  • the terms“RORy inhibitor,”“RORy antagonist,” and “RORy inverse agonist” are used interchangeably.
  • Non-limiting examples of small-molecule RORy inhibitors include compounds of Formula I (e.g ., those listed in Table 2, such as XY018 and XY063), GSK805, SR2211, and those described in Pandya et al, J Med. Chem. (2016) (DOI: l0. l02l/acs.jmedchem.8b00588), e.g., those listed in Table 5, such as VTP -43742. Any combination of RORy inhibitors may be used. In some embodiments, the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP-43742, and a combination thereof.
  • compounds of Formula I can be used in methods of the present invention as RORy inhibitors or RORy antagonists.
  • compounds of Formula I e.g., XY018 or XY063 include inverse agonists that bind to RORy and decrease its activity below a constitutive (e.g., intrinsic or basal) level of activity in the absence of any ligand.
  • compounds of Formula I include pharmaceutically acceptable salts, derivatives, analogs, isomers, racemates, prodrugs, co-crystalline complexes, hydrates, and solvates thereof.
  • the compound of Formula I has a half-maximal inhibitory concentration (IC50) value of from about 100 nM to about 100 mM, e.g, from about 100 nM to about 50 pM, from about 100 nM to about 25 pM, from about 100 nM to about 10 pM, from about 500 nM to about 100 pM, from about 500 nM to about 50 pM, from about 500 nM to about 25 pM, from about 500 nM to about 10 pM, from about 1 pM to about 100 pM, from about 1 pM to about 50 pM, from about 1 pM to about 25 pM, from about 1 pM to about 10 pM, or about 100 nM, 200 nM, 300 nM, 400 nM, 500 nM, 600 nM, 700 nM, 800 nM, 900 nM
  • the IC50 value for a specific compound of Formula I is measured using an in vitro assay in cancer cells that have been incubated with the compound.
  • the IC50 value can be determined based on the effect of the compound of Formula I in inhibiting the survival of cancer cells such as cells from a cancer cell line or primary tumor cells.
  • the compound of Formula I has an inhibitor constant (Kp that is essentially the same numerical value as the IC50 value, or is about one-half the value of the IC50 value.
  • the compound of Formula I has the following structure:
  • n is an integer selected from the group consisting of 0, 1, 2, or 3,
  • Ki is selected from the group consisting of FI, halo, alkyl, trifluoromethyl, cyano, -COQRs, -COR.5, -ORs, -CQH(CF 3 ) 2 , heterocydyi, and cycloalkyl,
  • R5 is selected from the group consisting of H, and Ci-C 3 alkyl group
  • R2 is selected from the group consisting of FI, halogen, and alkyl
  • R 3 is selected from the group consisting of H and alkyl
  • R 4 is selected from the group consisting of C 0 -C 4 alkylene-R 6 , C 0 -C 4 alkylene- R 7 -cycloa3ky3, and C0-C4 alkylene-R 7 -heterocyclyl,
  • Re is selected from the group consisting of -Rg, -ORg, -CQRg, -COOR8, -S(0) m R8, cycloalkyl, and heterocyclyl, m is 0 or 2, and R? is selected from the group consisting of -OR9, -C(0)33 ⁇ 4, -NR9, -SR9, ⁇ S(())R9, -S(0) 2 R9,
  • Rg is selected from the group consisting of H, and C 1 -C 3 alkyl group, and R9 is C1-C3 alkyl ene;
  • each cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, cafboxy, amino, -CONH2, -COOR10, -CQR10, -OR10, -NHCOR10, -NHCOOR10, and -COH(CF 3 )2,
  • each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono- , bi- or tri -cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, C 1 -C 4 alkyl, trifluoromethyl, cyano, carhoxy, nitro, amino, -CON!
  • Rio is independently selected from the group consisting of H, C I -C I alkyl, and phenyl, and
  • R 11 is independently selected from the group consisting of C 1 -C 4 alkyl, halogen, acetyl, methoxy, and ethoxy.
  • Ra is selected from the group consisting of H, methyl, ethyl, propyl, fluoro, chloro, bromo, trifluoromethyl, cyano, -COH(CF3)2, -COOR5, -COR5, -ORs, heterocyclyl, and cycloalkyl, wherein R 5 is selected from the group consisting of H, methyl, ethyl, and propyl.
  • R 2 is selected from the group consisting of H, methyl, ethyl, propyl, fluoro, chloro, and bromo.
  • R 3 is selected from the group consisting of H, methyl, ethyl, propyl, and isopropyl.
  • R 4 is selected from the group consisting of: (1) ( ' :. ⁇ ( ⁇ alkylene-Re, wherein R 6 is selected from the group consisting of -R 8 , -ORg, -CORg, -COORg, -S(0) m R8, cycloalkyl, and heterocyclyl, m is 0 or 2, and Rg is selected from the group consisting of H, methyl, ethyl, and propyl group;
  • said eycioalkyi group is selected from the group consisting of cyclobutane, cyclopentane, cyclohexane, cycloheptane, phenyl, and naphthyl, and said eycioalkyi group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, carboxy, amino, -COM !
  • said heterocyclyl group is selected from the group consisting of imidazoly!, triazolyl, pyrazolyl, thienyl, oxazoly!, isoxazoiyl, pyrazinyi, pyridazinyi, pyrimidinyl, pyrrol yl, piperazinyl, tetrahydro-pyrrolyl, piped dinyl, morpholinyl, 1,3- dioxolany!, isoquinolinyl, indoiine group, 1H- indazolyl, 1H- benzo [d] imidazolyl, 1 H- indolyl, benzo [d] [1,3] dioxoie, benzo [d] thiazolyi, and a member of the moieties shown in Table 1, and said heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, C1-C4 alkyl, trifluoromethyl,
  • R ⁇ is selected from the group consisting of -ORg, -C(0)R9, -NR 9 , -SR 9 , -S(0)R9, -S(0) 2 R9,
  • R9 is C1-C3 alkylene
  • Rio is independently selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, and phenyl,
  • Rn is independently selected from the group consisting of methyl, ethyl, propyl, isopropyl, fluoro, chloro, bromo, acetyl, methoxy, and ethoxy, and
  • R 12 is selected from the group consisting of H, methyl, ethyl , propyl, and isopropyl.
  • Table 1 Possible Heterocyclic Moieties
  • the RORy inhibitor compound of Formula I is represented by a compound according to Formula la:
  • Ri is selected from the group consisting of H, methyl, ethyl, propyl, fluoro, ch!oro, bromo, trifluoro ethyl, cyano, -COH(CF 3 )2, -COORs, -CORs, -OR5, heterocyclyl, and cycloalkyl, wherein R5 is selected from the group consisting of FI, and Ci-saikyl group;
  • R 2 is selected from the group consisting of H, fluoro, chloro, bromo;
  • R 3 is selected from the group consisting of H, methyl, ethyl, propyl, and isopropyl;
  • i is selected from the group consisting of
  • R & is selected from the group consisting of -Rs, -ORg, -CORs, -COORg, -S(0) m Rs, cycloalkyl, and heterocyclyl , m is 0 or 2, and Rg is selected from the group consisting of H, and Ci- 3 alkyl;
  • R ? is selected from the group consisting of -OR9, -C(0)R9, -NR9, -SR9, -S(0)Rg, -S(0) 2 R 9 , wherein
  • R9 is selected from the group consisting of C l-3 aikylene, heterocyclyl, and cycloalkyl,
  • each cycloalkyi group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyi group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethy!, cyano, carboxy, amino, -CONH 2 , -CQORio, -CORio, -OR l0 , -NHCORio, -NHCOORio, and -COI Kdrik
  • each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono- , bi- or tri -cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, Ci-C 4 alkyl, trifluoromethyi, cyano, carboxy, nitro, amino, -COM l ⁇ , -COORio, -COR l0, -OR l0 , -NHCORio, -NHCOORio, -COH(CF 3 ) 2 , -CcHsRn, morpholinyl, piperidinyl, tetrahydrofuranyl , substituted pyridyl group,
  • R l0 is independently selected from the group consisting of C 1 -C 4 alkyl, and phenyl, and
  • Rn is independently selected from the group consisting of C1-C4 alkyl, halogen, acetyl, methoxy, and ethoxy.
  • Ri is a cycloalkyl group having an un saturated ring structure of 6 carbon atoms (e.g, a phenyl group), wherein the cycloalkyl group is substituted with a fluoro group (e.g., at the 2' position) and a 1,1, 1,3,3, 3-hexafluoro-2-hydroxypropan-2- yl group (e.g, at the 4' position).
  • R 2 is H.
  • R 3 is H.
  • the RORy inhibitor compound of Formula I is represented by a compound according to Formula lb:
  • R is selected from the group consisting of H, methyl, ethyl, propyl, fluoro, chloro, bromo, trifluoromethyi, cyano, -COH(CF 3 ) 2 , -COQRs, -CORs, -OR 5 , heterocyclyl, and cycloalkyl, wherein
  • R 5 is selected from the group consisting of H, and Ci- 3 alkyl group, and said cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms,
  • said cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C -C 4 alkyl group. trifluoromethyl, cyano, carboxy, amino, -CONH 2 , -COOR10, -COR10, -OR l0 , -NHCORio,
  • heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono-, bi- or tri ⁇ cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, (), and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, C1-C4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -COM l ⁇ , -COOR10, -COR l0, -OR l0 , -NHCORio, -NHCOOR10, -COH(CF 3 ) 2 , -CeHjRn, morpholinyl, piperidinyl, tetrahydrofuranyl , substituted pyridyl group;
  • R 2 is selected from the group consisting of H, fluoro, chloro, bromo;
  • Ri is selected from the group consisting of H, methyl, ethyl, propyl, and isopropyl;
  • R 4 is selected from the group consisting of
  • C0-C4 alkylene-Re wherein R 6 is selected from the group consisting of -Rg, -ORg, and cycloalkyl, and Rg is selected from the group consisting of H, and Cusalkyl;
  • R? is selected from the group consisting of -OR9, -C(0)R 9 , -NR ⁇ , ⁇ . -SR 9 , -S(0)R 9 , -S(0) 2 R9, and R9 is selected from the group consisting of Ciualkyiene, heterocyclyl, and cycloalkyl,
  • heterocyclyl group is selected from the group consisting of imidazolyl, triazolyl, pyrazolyl, thienyl, oxazolyl, isoxazolyl, pyrazinyl, pyridazinyl, pyrimidinyl, pyrrolyl, piperazinyl, tetrahydro-pyrroly!
  • R l0 is independently selected from the group consisting of Ch-4 alkyl and
  • R J J is independently selected from the group consisting of methyl, ethyl, propyl, isopropyl, halogen, acetyl, methoxy, and ethoxy, and
  • R 12 is selected from the group consisting of H, methyl, ethyl , propyl, isopropyl.
  • the compound of Formula I is selected from the group consisting of l-ethyl -N- (2- fluoro-4- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) phenyl) -2- oxo-l, 2- dihydro-benzo [cd] indole-6-sulfonamide, N- (2- fluoro-4- (1, 1, 1,3,3, 3-hexafluoro- 2-hydroxy-2-yl) phenyl) -4,4-dimethyl-2-oxo- l,2,3,4-tetrahydro-quinolin-6-sulfonamide, N- (2- fluoro-4- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) phenyl) - 2-oxo
  • the RORy inhibitor compound of Formula I is a compound in Table 2.
  • the RORy inhibitor compound of Formula I is represented by a compound according to any one of Formulas Ic to Ii:
  • the compound of Formula Ic is also called XY018 or F18 and corresponds to Structure No. 37 in Table 2.
  • the compound of Formula Id is also called F17 and corresponds to Structure No. 36 in Table 2.
  • the compound of Formula Ie is also called F62 and corresponds to Structure No. 80 in Table 2.
  • the compound of Formula If is also called XY063 or F63 and corresponds to Structure No. 81 in Table 2.
  • the compound of Formula Ig is also called F64 and corresponds to Structure No. 82 in Table 2.
  • the compound of Formula Ih is also called F65 and corresponds to Structure No. 83 in Table 2.
  • the compound of Formula Ii is also called F68 and corresponds to Structure No. 86 in Table 2.
  • the RORy inhibitor compound of Formula I is a compound disclosed in Chinese Patent Application No. 201410344302.0, filed July 18, 2014, the disclosure of which is hereby incorporated by reference in its entirety for all purposes.
  • the RORy inhibitor compound of Formula I is a compound disclosed in Chinese Patent Publication No. CN 105272904, the disclosure of which is hereby incorporated by reference in its entirety for all purposes.
  • GSK805 refers to N-[2,6-dichloro-2'-(trifluoromethoxy)[l,r-biphenyl]- 4-yl]-4-(ethylsulfonyl)-benzeneacetamide, which has the following structure:
  • GSK805 is an orally bioavailable retinoic acid receptor-related orphan receptor g ⁇ (RORyt) inverse agonist that directly and reversibly interacts with the receptor's putative ligand binding domain to block its effects on transcription without exerting significant effects on DNA binding.
  • GSK805 inhibits the expression of IL-17 (e.g., at a concentration of 0.5 mM) in naive CD4 + T cells activated under Thl 7-cell-polarizing conditions and affects the broader RORyt-dependent gene network, inhibiting the development and pathogenic function of Thl7 cells.
  • SR2211 refers to 2-fluoro-4’-[[4-(4-pyridinylmethyl)-l- piperazinyl]methyl]-a,a-Z>A(trifluoromethyl)-[l,r-biphenyl]-4-methanol, which as the following structure: as well as pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and/or solvates thereof.
  • RORy inhibitors i.e., that can be used in methods of the present invention, e.g., either alone or with a statin as described herein
  • Suitable RORy inhibitors are described in Pandya et al., J Med. Chem. (2016) (DOI: l0. l02l/acs.jmedchem.8b00588; hereby incorporated by reference in its entirety for all purposes), some of which are discussed below.
  • the RORy inhibitor is a sulfonamide of a cyclic amine.
  • the sulfonamide of a cyclic amine has the following structure:
  • R 13 is H or F.
  • the sulfonamide of a cyclic amine is N-((R)-2-fluoro-3- hydroxy-3 -methylbutyl)-2-((S)- 1 -((4-fluorophenyl)sulfonyl)-6-( 1 , 1 , 1 ,3 ,3 ,3 -hexafluoro-2- hydroxypropan-2-yl)-l,2,3,4-tetrahydroquinolin-2-yl)acetamide (chemical formula: C25H26F8N2O5S, molecular weight: 618.54), which has the following structure:
  • the sulfonamide of a cyclic amine has the following structure:
  • R14 is CFh or O.
  • the sulfonamide of a cyclic amine is 2-chloro-6-fluoro-N-(2- ((3-(trifluoromethyl)phenyl)sulfonyl)-2,3,4,5-tetrahydro-lH-benzo[c]azepin-8-yl)benzamide (chemical formula: C 24 H 19 CIF 4 N 2 O 3 S, molecular weight: 526.93), which has the following structure:
  • the RORy inhibitor is an aryl sulfonyl compound.
  • a sulfonyl group attached to an aryl or heteroaryl ring can participate in hydrogen bond interactions (e.g with Arg367 and/or Leu287 of RORy) that contribute to RORy inhibitory potency.
  • the aryl sulfonyl compound is a cyclopropyl derivative, e.g., N-(3,5-dichloro-4-(l-(5-(4-fluorophenyl)-l,2,4-oxadiazol-3- yl)cyclopropyl)phenyl)-2-(4-(ethylsulfonyl)phenyl)acetamide (chemical formula: C 27 H 22 CI 2 FN 3 O 4 S, molecular weight: 574.45), which has the following structure:
  • the cyclopropyl ring (e.g, 1,1 -di substituted cyclopropyl ring) can, at least in some instances, project an attached aryl or heteroaryl group such that occupancy of the hydrophobic pocket of a RORy ligand binding domain is improved.
  • the aryl sulfonyl compound contains a hydroxymethyl group, e.g, (S)-7-ethyl-N-((R,4Z,6E)-6-(ethylsulfonyl)-l-hydroxyocta-4,6-dien-2-yl)-6-(((lr,4S)-4- (trifluoromethyl) cyclohexyl)methyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridine-3-carboxamide (chemical formula: C28H36F3N3O4S, molecular weight: 567.67), which has the following structure:
  • the aryl sulfonyl compound is (S)-N-((5- (ethylsulfonyl)pyridin-2-yl)methyl)-7-isopropyl-6-(((lr,4S)-4-(trifluoromethyl) cyclohexyl) methyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridine-3 -carboxamide (also known as VTP -43742) (chemical formula: C27H35F3N4O3S, molecular weight: 552.66), which has the following structure:
  • the aryl sulfonyl compound contains a terminal CF 3 group that interacts with His479 and/or Trp3 l7 of RORy, thus creating a steric clash that destabilizes helix 12 of RORy (i.e., in the agonist position).
  • the aryl sulfonyl compound inhibits RORy by trapping a water molecule, thus forming a hydrogen bond to Tyr502 of RORy.
  • connectivity of the carboxamide group to the central core of the aryl sulfonyl compound is inverted (e.g ., (S)-7-ethyl-N-((R,4Z,6E)-6- (ethylsulfonyl)-l-hydroxyocta-4,6-dien-2-yl)-6-(((lr,4S)-4-(trifluoromethyl) cyclohexyl) methyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridine-3 -carboxamide or (S)-N-((5-
  • the aryl sulfonyl compound is an isoindoline derivative, e.g., (R)-2-acetyl-N-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl)-5-(methylsulfonyl) isoindoline-l-carboxamide (chemical formula: C21H18F6N2O5S, molecular weight: 524.43), which has the following structure:
  • the aryl sulfonyl compound contains an N-acetyl group comprising an oxygen atom that hydrogen bonds with the RORy ligand binding domain. In some embodiments, the aryl sulfonyl compound contains a hexafluoroisopropanol moiety that interacts with helices 11 and/or 12 of RORy.
  • the RORy inhibitor is an amide/bis-amide derivative.
  • the amide/bis-amide derivative contains one or two amide groups that either directly or indirectly (e.g water-mediated) participate in hydrogen bond interactions with Phe 377, Glu379, and/or His479 of RORy.
  • the amide/bis-amide derivative is a piperazine derivative, e-g (S)-N-(8-((4-(cyclopentanecarbonyl)-3-methylpiperazin-l-yl)methyl)-7- methylimidazo[l,2-a]pyridin-6-yl)-2-methylpyrimidine-5-carboxamide (chemical formula: C26H33N7O2, molecular weight: 457.60), which has the following structure:
  • the amide/bis-amide derivative contains a pyrimidine ring comprising two nitrogen atoms that form water-mediated hydrogen bonds to residues Arg367, Leu287, and/or Glu379 of RORy. In some embodiments, the amide/bis-amide derivative contains an NH group that participates in a hydrogen bond to Phe377 of RORy.
  • the amide/bis-amide derivative is 2-((lS,3s)-3-((R)-5-((7- fluoro-l,l-dimethyl-2,3-dihydro-lH-inden-5-yl)carbamoyl)-2-methoxy-5,6,7,8-tetrahydro- l,6-naphthyridine-6-carbonyl)cyclobutyl)acetic acid (chemical formula: C28H32FN3O5, molecular weight: 509.58), which has the following structure:
  • the amide/bis-amide derivative contains an amide group that participates in a hydrogen bond interaction with Phe377 of RORy, an amide group that participates in a hydrogen bond interaction with Glu379 of RORy, a pyridine ring comprising a nitrogen atom that participates in a hydrogen bond interaction with Gln286 of RORy, and/or a methoxy group that participates in a hydrogen bond interaction with Arg364 of RORy.
  • the amide/bis-amide derivative is 3-cyano-N-(3-(l- (cyclopentanecarbonyl)piperidin-4-yl)-l-methyl-4-(trifluoromethyl)-lH-pyrrolo[2,3-b] pyridin-5-yl)benzamide (chemical formula: C28H28F3N5O2, molecular weight: 523.56), which has the following structure:
  • the RORy inhibitor is an indole derivative.
  • the indole derivative is (2,4-dichloro-3-((l,4-dimethyl-6-(trifluoromethyl)-lH- indol-2-yl)methyl)phenyl)(4-(oxetan-3-yl)piperazin-l-yl)methanone (chemical formula: C26H26CI2F3N3O2, molecular weight: 540.41), which has the following structure:
  • Statins are a group of compounds that are 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase inhibitors (also known as HMG-CoA inhibitors or HMGCR inhibitors) that function as lipid-lowering compounds.
  • HMG-CoA is an enzyme ⁇ i.e., NADH-dependent (EC 1.1.1.88), NADPH-dependent (EC 1.1.1.34)) that is the rate-limiting enzyme of the mevalonate pathway, i.e., the metabolic pathway that produces cholesterol and other isoprenoids.
  • HMG-CoA is normally suppressed by cholesterol that is derived from the internalization and degradation of low density lipoprotein (LDL) via the LDL receptor, as well as oxidized species of cholesterol.
  • LDL low density lipoprotein
  • statins suitable for use in compositions, methods, and kits of the present invention include atorvastatin, cerivastatin, fluvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin, and simvastatin, the chemical structures of which are shown in FIG. 15. Furthermore, it is understood that combinations of statins can be used in compositions, methods, and kits of the present invention.
  • the RORy inhibitor e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and the statin (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) can be used in combination with an anticancer drug to reduce or reverse cancer cell resistance to the anticancer drug, e.g, by sensitizing the cancer cell to the anticancer drug.
  • a compound of Formula I e.g, XY018, XY063
  • GSK805 e.g, SR2211
  • a compound listed in Table 5 such as VTP- 4374
  • Non-limiting examples of anticancer drugs include anti-androgen drugs, chemotherapeutic agents, radiotherapeutic agents, antigen-specific immunotherapeutic agents, endocrine therapies, tyrosine kinase inhibitors, and combinations thereof.
  • Anti-androgen drugs are compounds that inhibit the transcription, translation, stability, and/or activity of androgen receptors (AR) or variants thereof (e.g. AR-V7).
  • Inhibition of AR activity can include inhibition of recruitment of AR to Androgen Response Elements (AREs).
  • AREs Androgen Response Elements
  • inhibition of AR activity can include inhibition of recruitment of AR to the PSA promoter.
  • inhibition of AR activity can include inhibition of AR-induced activation of the PSA promoter.
  • inhibition of AR activity can include inhibition of AR-induced PSA production.
  • inhibition of AR can include inhibition of production of PSA in the absence of DHT.
  • Anti-androgen drugs include, but are not limited to, enzalutamide, abiraterone, bicalutamide, flutamide, nilutamide, apalutamide, finasteride, dutasteride, alfatradiol, and combinations thereof.
  • the present invention provides a composition comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more anti-androgen drugs.
  • the composition further comprises a pharmaceutically acceptable excipient or diluent.
  • the composition is formulated for oral or parenteral administration.
  • the present invention provides a method for treating cancer in a subject comprising administering to the subject an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more anti-androgen drugs.
  • RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such
  • the effective amount of the RORy inhibitor and the statin is an amount sufficient to sensitize an anti-androgen drug-resistant cancer such as anti -androgen drug-resistant prostate cancer (e.g, castration-resistant prostate cancer) to anti -androgen drug treatment.
  • an anti-androgen drug-resistant cancer such as anti -androgen drug-resistant prostate cancer (e.g, castration-resistant prostate cancer)
  • Combinations of RORy inhibitors and statins with anti-androgen drugs can be delivered to a subject via the same route of administration (e.g, orally or parenterally) or via different routes of administration (e.g, intravenously for RORy inhibitors and/or a statins and orally for anti -androgen drugs, or vice versa).
  • Chemotherapeutic agents are well known in the art and include, but are not limited to, anthracenediones (anthraquinones) such as anthracy clines (e.g, daunorubicin (daunomycin; rubidomycin), doxorubicin, epirubicin, idarubicin, and valrubicin), mitoxantrone, and pixantrone; platinum-based agents (e.g, cisplatin, carboplatin, oxaliplatin, satraplatin, picoplatin, nedaplatin, triplatin, and lipoplatin); tamoxifen and metabolites thereof such as 4-hydroxytamoxifen (afimoxifene) and N-desmethyl-4-hydroxytamoxifen (endoxifen); taxanes such as paclitaxel (taxol), docetaxel, cabazitaxel, hongdoushan A, hongdoushan
  • the present invention provides a composition comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more chemotherapeutic agents.
  • the composition further comprises a pharmaceutically acceptable excipient or diluent.
  • the composition is formulated for oral or parenteral administration.
  • the present invention provides a method for treating cancer in a subject comprising administering to the subject an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more chemotherapeutic agents.
  • RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such
  • the effective amount of the one or more RORy inhibitors and one or more statins is an amount sufficient to sensitize a chemotherapy drug-resistant cancer such as a tamoxifen- resistant cancer (e.g ., tamoxifen -resistant breast cancer) or a taxane-resistant cancer (e.g, docetaxel-resistant prostate cancer) to chemotherapy drug treatment.
  • a chemotherapy drug-resistant cancer such as a tamoxifen- resistant cancer (e.g ., tamoxifen -resistant breast cancer) or a taxane-resistant cancer (e.g, docetaxel-resistant prostate cancer) to chemotherapy drug treatment.
  • Combinations of RORy inhibitors, statins, and chemotherapeutic agents can be delivered to a subject via the same route of administration (e.g, orally or parenterally) or via different routes of administration (e.g, intravenously for RORy inhibitors and statins and orally for chemotherapeutic agents, or vice versa).
  • Radiotherapeutic agents are well known in the art and can comprise external-beam radiation therapy and/or internal radiation therapy.
  • External beam radiation therapy delivers radioactive beams of high energy X-rays and/or gamma rays to a patient’s tumor
  • internal radiation therapy delivers radioactive atoms to a patient’s tumor.
  • Both external beam radiation therapy and internal radiation therapy are used to suppress tumor growth or kill cancer cells by delivering a sufficient quantity of radioactivity to the target site.
  • the radiotherapeutic agent comprises a radioactive atom and is complexed with a biologic or synthetic agent to increase delivery to the target site.
  • biologic or synthetic agents are known in the art.
  • radioactive atoms for use with the RORy inhibitors include any of the radionuclides described herein, or any other isotope which emits enough energy to destroy a targeted tissue or cell.
  • radiotherapeutic agents may be coupled to targeting moieties, such as antibodies, to improve the localization of radiotherapeutic agents to cancerous cells.
  • radioactivity refers to the radiation, including alpha particles, beta particles, nucleons, electrons, positrons, neutrinos, and gamma rays, emitted by a radioactive substance.
  • radionuclides suitable for use in the present invention include, but are not limited to, fluorine 18 ( 18 F), fluorine 19 ( 19 F), phosphorus 32 ( 32 P), scandium 47 ( 47 Sc), cobalt 55 ( 55 Co), copper 60 ( 60 Cu), copper 61 ( 61 Cu), copper 62 ( 62 Cu), copper 64 ( 64 Cu), gallium 66 ( 66 Ga), copper 67 ( 67 Cu), gallium 67 ( 67 Ga), gallium 68 ( 68 Ga), rubidium 82 ( 82 Rb), yttrium 86 ( 86 Y), yttrium 87 ( 87 Y), strontium 89 ( 89 Sr), yttrium 90 ( 90 Y), rhodium 105 ( 105 Rh), silver 111 ( lu Ag), indium 111 ( U1 ln), iodine 124 ( 124 I), iodine 125 ( 125 I), iodine 131 ( 131 I), tin H
  • the“m” in 117m Sn and 99m Tc stands for the meta state.
  • naturally-occurring radioactive elements such as uranium, radium, and thorium, which typically represent mixtures of radioisotopes, are suitable examples of radionuclides.
  • 67 Cu, 131 I, 177 Lu, and 186 Re are beta- and gamma-emitting radionuclides.
  • 212 Bi is an alpha- and beta-emitting radionuclide.
  • 211 At is an alpha-emitting radionuclide.
  • 32 P, 47 Sc, 89 Sr, 90 Y, 105 Rh, lu Ag, 117m Sn, 149 Pm, 153 Sm, 166 HO, and 188 Re are examples of beta- emitting radionuclides.
  • 67 Ga, U1 ln, 99m Tc, and 201 Tl are examples of gamma-emitting radionuclides.
  • 55 Co, 60 Cu, 61 Cu, 62 Cu, 66 Ga, 68 Ga, 82 Rb, and 86 Y are examples of positron- emitting radionuclides.
  • 64 Cu is a beta- and positron-emitting radionuclide.
  • the present invention provides a composition comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more radiotherapeutic agents.
  • the composition further comprises a pharmaceutically acceptable excipient or diluent.
  • the composition is formulated for oral or parenteral administration.
  • the present invention provides a method for treating cancer in a subject comprising administering to the subject an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more radiotherapeutic agents.
  • RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as V
  • the effective amount of the one or more RORy inhibitors and one or more statins is an amount sufficient to sensitize a radiation-resistant cancer such as a radiation-resistant breast cancer to radiation treatment.
  • One or more RORy inhibitors, one or more statins, and radiotherapeutic agents can be delivered to a subject via the same route of administration (e.g, orally or parenterally) or via different routes of administration (e.g, intravenously for RORy inhibitors and/or statins and orally for radiotherapeutic agents, or vice versa).
  • Endocrine therapy is the manipulation of the endocrine system through the administration of specific hormones or drugs which inhibit or decrease the production or activity of targeted hormones or alter the gene expression pattern of targeted cells. Endocrine therapy is particularly useful in certain types of cancer, including breast cancer. Any known hormone antagonist or modulator may be used in the present invention.
  • Endocrine therapies useful in the present invention include, but are not limited to, aromatase inhibitors (e.g. letrozole), megestrol acetate, flutamide, tamoxifen, raloxifene, lasofoxifene, apeledoxifene, thaledoxifene/conjugated estrogens, and combinations thereof.
  • the present invention provides a composition comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more endocrine therapies.
  • the composition further comprises a pharmaceutically acceptable excipient or diluent.
  • the composition is formulated for oral or parenteral administration.
  • the present invention provides a method for treating cancer in a subject comprising administering to the subject an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more endocrine therapies.
  • RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as
  • the effective amount of the one or more RORy inhibitors and one or more statins is an amount sufficient to sensitize an endocrine therapy-resistant cancer such as a tamoxifen -resistant breast cancer to endocrine therapy.
  • RORy inhibitors, statins, and endocrine therapies can be delivered to a subject via the same route of administration (e.g ., orally or parenterally) or via different routes of administration (e.g., intravenously for RORy inhibitors and/or statins and orally for endocrine therapies, or vice versa).
  • Tyrosine kinase inhibitors are small-molecules that inhibit tyrosine kinase proteins.
  • Tyrosine kinases are enzymes that activate many proteins in cellular signal transduction cascades by addition of a phosphate group to the protein. High expression and aberrant activation, of tyrosine kinase proteins can cause undesirable“switching on” of cellular signaling pathways that can result in uncontrolled cellular proliferation associated with cancerous cellular phenotypes.
  • Various forms of cancer are currently treated by inhibiting or reducing the activity of poorly regulated tyrosine kinase proteins with tyrosine kinase inhibitors.
  • Treatment regimens with tyrosine kinase inhibitors can suppress, reduce the incidence, reduce the severity, or inhibit the progression of cancer.
  • tyrosine kinase inhibitors include, but are not limited to, gefitinib, erlotinib, sorafenib, sunitinib, dasatinib, lapatinib, nilotinib, bortezomib, salinomycin, and combinations thereof.
  • the present invention provides a composition comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more tyrosine kinase inhibitors.
  • the composition further comprises a pharmaceutically acceptable excipient or diluent.
  • the composition is formulated for oral or parenteral administration.
  • the present invention provides a method for treating cancer in a subject comprising administering to the subject an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more tyrosine kinase inhibitors.
  • RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a
  • the effective amount of the one or more RORy inhibitors and one or more statins is an amount sufficient to sensitize a tyrosine kinase inhibitor-resistant cancer such as a tyrosine kinase inhibitor-resistant non-small-cell lung cancer (NSCLC) to tyrosine kinase inhibitor therapy.
  • RORy inhibitors, statins, and tyrosine kinase inhibitors can be delivered to a subject via the same route of administration (e.g ., orally or parenterally) or via different routes of administration (e.g., intravenously for RORy inhibitors and/or statins and orally for tyrosine kinase inhibitors, or vice versa).
  • antigen-specific immunotherapeutic agents include compounds and compositions designed to stimulate the immune system to specifically recognize antigens expressed or overexpressed by cancerous cells.
  • antigen-specific immunotherapeutic agents include compounds and compositions that will specifically recognize antigens expressed or overexpressed by cancerous ceils.
  • tumor microenvironment-targeting agents are used that may include, for example, compounds, antibodies, and other compositions that specifically recognize immune- modulating cytokines and suppress their tumor-promoting activities.
  • Non-limiting examples of antigen-specific or tumor microenvironment-targeting immunotherapeutic agents include vaccines (e.g., peptide vaccines), antibodies, cytotoxic T cell lymphocytes (CTLs), chimeric antigen receptor T cells (CAR-T cells), immune checkpoints (e.g, CTLA-4, PD-l, and PD- Ll), antibodies against immune modulating cytokines (e.g, IL-6, IL-23 and IL-17), and combinations thereof.
  • the antigens presented by cancerous cells are highly specific to each cancer type, and the vaccines, antibodies, CTLs, and/or CAR-T cells used is dependent on the cancer type being treated.
  • a vaccine can stimulate the immune system to specifically recognize and attack antigens presented by cancerous cells.
  • Vaccines can comprise one or more peptides, peptide fragments, fusion peptides, DNA, RNA, other biologic or non-biologic material, or combinations thereof.
  • one or more peptides, peptide fragments, or fusion peptides may be used for a peptide vaccine.
  • the peptides may be harvested from an endogenous source or chemically synthesized.
  • the peptides chosen are specific for the type of cancer being treated. For example, when targeting cancer cells, some commonly targeted proteins include GM-CSF, IL-l3Ra2, EphA2, and Survivin; however, specific cancer types will have specifically preferred peptides used for targeting afflicted cells.
  • the one or more peptides in the peptide vaccine are free soluble peptides.
  • the one or more peptides in the peptide vaccine are tethered together using any means known in the art.
  • vaccines include cancer vaccines such as, e.g., tecemotide (L-BLP25), oncophage, sipuleucel-T, and combinations thereof.
  • Tecemotide (L-BLP25) is a liposomal antigen-specific cancer immunotherapy that contains 25 amino acids from the immunogenic tandem-repeat region of MUC1 (see, e.g, Mehta NR el al. , Clin. Cancer Res., 18:2861-2871 (2012)).
  • Antibodies can recognize antigens expressed or overexpressed by cancerous cells Antigens recognized by these antibodies can be proteins expressed, activated, or overexpressed on the cell surface or proteins secreted into the extracellular fluid. In some embodiments, antibodies can be used to target human effector ceils (e.g., macrophages) against the cancerous cells. In some embodiments, antibodies are used to inhibit the normal function of cell surface receptors. In some embodiments, antibodies bind to the ligands of eeli surface receptors to block the cellular signaling cascade.
  • Antibodies used as antigen- specific immunotherapeutic agents can be monoclonal or polyclonal antibodies as well as chimeric, humanized, or human antibodies, and can be previously isolated from the patient or produced from another biologic source. Methods of producing antibodies are well known in the art, and may be made by any known means. For example, antibodies described herein can be produced by conventional monoclonal antibody methodology e.g, the standard somatic cell hybridization technique of Kohler and Milstein, Nature 256: 495 (1975), the contents of which are herein incorporated by reference for all purposes.
  • antibodies useful in the treatment of cancer include immune checkpoint inhibitors.
  • antibodies useful in the treatment of cancer include, but are not limited to, alemtuzumab.
  • bevacizumab cetuximab, ipilimumab, nivolumab, ofatumumab, panitumumab, pembrolizumab, atezolizumab, rituximab, trastuzumab, and combinations thereof.
  • CTLs and CAR-T cells are used as antigen-specific immunotherapeutic agents.
  • adoptive T cell transfer therapy is a technique that can boost the natural immune system’s ability to combat cancer by enriching for and/or designing T cells that are able to effectively recognize, bind, and kill a diseased cell.
  • CTLs can recognize and bind cancerous cells using T-cell receptors (TCR).
  • TCRs contain a highly variable binding region that allows them to recognize a large range of antigens.
  • TCRs bind to the major histocompatibility complex I (MHC I) of cancerous cells presenting an appropriate antigen.
  • MHC I major histocompatibility complex I
  • TCR binding is highly specific, so only a small number of CTLs will be able to recognize a particular antigen. Once an antigen is recognized by CTLs binding to the MHC I complex of the cancerous cell, they activate to induce cellular death. Activated CTLs proliferate to fight the detected cancer.
  • CTLs administered in this therapy may be derived from the subject or may be derived from other biological sources.
  • Methods for producing CTLs directed to a particular antigen are well known in the art, and can be harvested from an individual possessing a CTL directed to a particular antigen or produced outside of the body (ex vivo). For example, when treating cancer, cytotoxic T cells from a subject’s tumor are isolated, the cytotoxic T cells with the greatest antitumor activity are identified, the identified cytotoxic T cells are cultured to produce large amounts of the most effective cells, and the cultured cytotoxic T cells are reintroduced into the subject to treat the cancer.
  • CTLs can also be produced in healthy individuals using ex vivo techniques described in U.S. Patent No. 5,962,318, and U.S. Patent Application Publication No. 2009/0324539, the contents of which are herein incorporated by reference for all purposes. The ex vivo methods described herein can be useful for individuals both before cancer onset or after cancer onset.
  • CAR-T cells are modified T cells which have been engineered to possess a cellular specificity domain that has not been produced naturally.
  • the natural specificity domain of T cells are T-cell receptors that recognize a particular antigen presented on MHC class I molecules.
  • CAR-T cells possess a T-cell receptor that has not been naturally produced in a subject’s body.
  • the cellular specificity domain is a monoclonal antibody that is specific for the targeted cells or tissue.
  • CAR-T cells can be produced using any means known in the art.
  • cytotoxic T cells are harvested from a subject’s blood, the cytotoxic T cells are genetically modified by inserting a gene that encodes for a receptor that recognizes an antigen specific to the cancer affecting the subject, the CAR-T cells are cultured and can be stored for later use or reintroduced into the subject’s body to treat the cancer.
  • the CAR-T cells are cultured and can be stored for later use or reintroduced into the subject’s body to treat the cancer.
  • the present invention provides a composition comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more antigen-specific immunotherapeutic agents.
  • the composition further comprises a pharmaceutically acceptable excipient or diluent.
  • the composition is formulated for oral or parenteral administration.
  • the present invention provides a method for treating cancer in a subject comprising administering to the subject an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more antigen-specific immunotherapeutic agents.
  • RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table
  • the effective amount of the one or more RORy inhibitors and one or more statins is an amount sufficient to sensitize a cancer that is resistant to treatment with antigen- specific immunotherapeutic agents to such treatment.
  • RORy inhibitors, statins, and antigen- specific immunotherapeutic agents can be delivered to a subject via the same route of administration (e.g, orally or parenterally) or via different routes of administration (e.g, intravenously for RORy inhibitors and/or statins and orally for antigen-specific immunotherapeutic agents, or vice versa).
  • a cancer can be treated or prevented by administering one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin).
  • RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.
  • the one or more RORy inhibitors and one or more statins are administered in combination with an anticancer drug.
  • Cancer generally includes any of various malignant neoplasms characterized by the proliferation of anaplastic cells that tend to invade surrounding tissue and metastasize to new body sites.
  • Non-limiting examples of different types of cancer suitable for treatment using the compositions of the present invention include breast cancer, prostate cancer (including drug- resistant prostate cancers, e.g., apalutamide-resistant prostate cancer), lung cancer, ovarian cancer, bladder cancer, thyroid cancer, liver cancer, pleural cancer, pancreatic cancer, cervical cancer, testicular cancer, colon cancer, anal cancer, bile duct cancer, gastrointestinal carcinoid tumors, esophageal cancer, gall bladder cancer, rectal cancer, appendix cancer, small intestine cancer, stomach (gastric) cancer, renal cancer ( i.e ., renal cell carcinoma), cancer of the central nervous system, skin cancer, choriocarcinomas, head and neck cancers, bone cancer, osteogenic sarcomas, fibrosarcoma, neuroblastoma, glioma, endometrial cancer, melanoma, leukemia (e.g., acute lymphocytic leukemia, chronic lymphocytic leukemia, acute my
  • the cancer is an epithelial cancer (e.g, prostate cancer, ovarian cancer, breast cancer, and the like), or a blood cancer (e.g, leukemia, lymphoma, multiple myeloma).
  • the cancer is a prostate cancer.
  • the prostate cancer is an advanced stage prostate cancer selected from one or more of metastatic prostate cancer, drug-resistant prostate cancer (e.g, anti -androgen- resistant prostate cancer such as enzalutamide-resistant prostate cancer, abiraterone-resistant prostate cancer, bicalutamide-resistant prostate cancer, etc.; taxane-resistant prostate cancer; docetaxel-resistant prostate cancer; and the like), hormone refractory prostate cancer, castration-resistant prostate cancer (CRPC), metastatic castration-resistant prostate cancer, AR-V7-induced drug-resistant prostate cancer such as AR-V7-induced anti-androgen- resistant prostate cancer (e.g, AR-V7-induced enzalutamide-resistant prostate cancer), AKR1C3 -induced drug-resistant prostate cancer such as AKR1C3 -induced anti-androgen- resistant prostate cancer (e.g, AKR1C3 -induced enzalutamide-resistant prostate cancer), and combinations thereof.
  • drug-resistant prostate cancer e.g, anti -
  • the prostate cancer is an advanced stage prostate cancer selected from drug-resistant tumors with neuroendocrine (NE) phenotypes or NEPC.
  • the cancer is a lung cancer, breast cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma.
  • the lung cancer is a non-small-cell lung cancer (NSCLC), K-Ras mutant lung cancer, BRAF mutant lung cancer, tyrosine kinase inhibitor-resistant lung cancer, small cell lung cancer (SCLC), adenocarcinoma (e.g ., adenocarcinoma in situ), squamous cell carcinoma, large cell carcinoma, bronchial carcinoid, or combinations thereof.
  • NSCLC non-small-cell lung cancer
  • K-Ras mutant lung cancer BRAF mutant lung cancer
  • tyrosine kinase inhibitor-resistant lung cancer small cell lung cancer (SCLC)
  • SCLC small cell lung cancer
  • adenocarcinoma e.g ., adenocarcinoma in situ
  • squamous cell carcinoma large cell carcinoma
  • bronchial carcinoid or combinations thereof.
  • the breast cancer is triple-negative breast cancer (TNBC), tamoxifen- resistant breast cancer, radiation-resistant breast cancer, ductal carcinoma in situ , invasive ductal carcinoma, HER2-positive breast cancer, ER-positive breast cancer, inflammatory breast cancer, metastatic breast cancer, medullary carcinoma, tubular carcinoma, mucinous carcinoma (colloid), or combinations thereof.
  • the liver cancer is a hepatocellular carcinoma (HCC), cholangiocarcinoma (bile duct cancer), angiosarcoma, hepatoblastoma, or combinations thereof.
  • the glioma is an ependymoma, astrocytoma (e.g., glioblastoma multiforme), oligodendroglioma, brainstem glioma, optic nerve glioma, or combinations thereof (e.g, mixed glioma).
  • the gastric (stomach) cancer is an adenocarcinoma of the distal esophagus, gastroesophageal junction and/or stomach, a gastrointestinal carcinoid tumor, a gastrointestinal stromal tumor, an associated lymphoma, a cancer linked to infection with H. pylori , or combinations thereof.
  • compositions of the present invention encompass compositions made by admixing one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)), one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), and a pharmaceutically acceptable carrier and/or excipient or diluent.
  • RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP
  • one or more RORy inhibitors e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small -interfering RNA)) and/or one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) are present in a composition or kit of the present invention or are administered ( e.g ., to a subject) at a concentration of about 0.1 mM, 0.2 mM, 0.3 mM, 0.4 mM, 0.5 mM, 0.6 mM, 0.7 mM, 0.8 mM
  • compositions of the present invention may be prepared by any of the methods well-known in the art of pharmacy.
  • Pharmaceutically acceptable carriers suitable for use with the present invention include any of the standard pharmaceutical carriers, buffers and excipients, including phosphate-buffered saline solution, water, and emulsions (such as an oil/water or water/oil emulsion), and various types of wetting agents and/or adjuvants. Suitable pharmaceutical carriers and their formulations are described in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, l9th ed. 1995). Preferred pharmaceutical carriers depend upon the intended mode of administration of the active agent(s).
  • compositions of the present invention can include one or more RORy inhibitors (e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) together with one or more anticancer drugs such as an anti-androgen drug (e.g, enzalutamide, abiraterone, and/or bicalutamide) and/or a chemotherapeutic agent (e.g, tamoxifen and/or a taxane such as
  • the pharmaceutical composition can include one or more RORy inhibitors and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) together with an anti-androgen drug, such as enzalutamide.
  • statins e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin
  • an anti-androgen drug such as enzalutamide.
  • a pharmaceutical composition may optionally contain other therapeutic ingredients.
  • the compounds of the present invention can be combined as the active ingredient in intimate admixture with a suitable pharmaceutical carrier and/or excipient according to conventional pharmaceutical compounding techniques. Any carrier and/or excipient suitable for the form of preparation desired for administration is contemplated for use with the compounds disclosed herein.
  • the pharmaceutical compositions comprising one or more RORy inhibitors e.g., a compound of Formula I (e.g ., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti -RORy antibody, and/or an interfering RNA (e.g., small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) and the pharmaceutical compositions comprising one or more anticancer drugs are prepared as a single medicament.
  • the pharmaceutical compositions comprising one or more RORy inhibitors and one or more statins and the pharmaceutical compositions comprising one or more anticancer drugs are prepared as separate medicaments.
  • compositions of the present invention include formulations suitable for topical, parenteral, pulmonary, nasal, rectal, or oral administration.
  • the most suitable route of administration in any given case will depend in part on the nature and severity of the cancer condition and also optionally the stage of the cancer.
  • the one or more RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small- interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) is administered in combination with an anticancer drug
  • the administration of the one or more RORy inhibitors and one or more statins and the anticancer drug may be administered using the same or a different administration route.
  • the one or more RORy inhibitors and one or more statins and the anticancer drug may be administered orally or parenterally (e.g, intravenously).
  • the one or more RORy inhibitors and one or more statins may be administered orally, while the anticancer drug may be administered parenterally (e.g, intravenously), or vice versa.
  • compositions suitable for systemic (enteral or parenteral) administration include oral, rectal, sublingual, or sublabial administration. In some embodiments, the compositions may be administered via a syringe or intravenously.
  • Compositions for pulmonary administration include, but are not limited to, dry powder compositions consisting of the powder of a compound described herein, or a salt thereof, and the powder of a suitable carrier and/or lubricant. The compositions for pulmonary administration can be inhaled from any suitable dry powder inhaler device known to a person skilled in the art.
  • compositions for systemic administration include, but are not limited to, dry powder compositions consisting of the composition as set forth herein and the powder of a suitable carrier and/or excipient.
  • the compositions for systemic administration can be represented by, but not limited to, tablets, capsules, pills, syrups, solutions, and suspensions.
  • the present invention provides compositions further including a pharmaceutical surfactant.
  • the present invention provides compositions further including a cryoprotectant.
  • the cryoprotectant is selected from the group consisting of glucose, sucrose, trehalose, lactose, sodium glutamate, PVP, HPpCD, CD, glycerol, maltose, mannitol, and saccharose.
  • the present invention provides a pharmaceutical composition including one or more RORy inhibitors (e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti- RORy antibody, and/or an interfering RNA (e.g., small-interfering RNA)), one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), and a pharmaceutically acceptable excipient.
  • RORy inhibitors e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti- RORy antibody, and/
  • the present invention provides a pharmaceutical composition including one or more RORy inhibitors, one or more statins, and one or more anticancer drugs such as an anti-androgen drug (e.g, enzalutamide, abiraterone, and/or bicalutamide) and/or a chemotherapeutic agent (e.g, tamoxifen and/or a taxane such as docetaxel), in combination with a pharmaceutically acceptable excipient.
  • the present invention provides a pharmaceutical composition including one or more RORy inhibitors, one or more statins, and an anti-androgen drug, such as enzalutamide, in combination with a pharmaceutically acceptable excipient.
  • the pharmaceutically acceptable excipient includes a salt or a diluent.
  • the present invention provides compositions including an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti-RORy antibody, and/or an interfering RNA (e.g., small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin).
  • RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti-RORy antibody, and/or an interfering RNA (e
  • the composition is formulated for oral administration or parenteral (e.g, intravenous) administration and includes one or more RORy inhibitors, one or more statins, and at least one member selected from the group consisting of an aqueous solution and a buffer solution.
  • the composition can include an effective amount of one or more RORy inhibitors, one or more statins, and one or more anticancer drugs such as an anti androgen drug (e.g, enzalutamide, abiraterone, and/or bicalutamide) and/or a chemotherapeutic agent (e.g, tamoxifen and/or a taxane such as docetaxel).
  • an anti androgen drug e.g, enzalutamide, abiraterone, and/or bicalutamide
  • a chemotherapeutic agent e.g, tamoxifen and/or a taxane such as docetaxel.
  • compositions or medicaments for use in the present invention can be formulated by standard techniques using one or more physiologically acceptable carriers or excipients. Suitable pharmaceutical carriers are described herein and in Remington: The Science and Practice of Pharmacy, 2lst Ed., University of the Sciences in Philadelphia, Lippencott Williams & Wilkins (2005).
  • a pharmaceutical composition or a medicament can take the form of, e.g, a tablet or a capsule prepared by conventional means with a pharmaceutically acceptable excipient.
  • a pharmaceutically acceptable excipient e.g., a tablet or a capsule prepared by conventional means with a pharmaceutically acceptable excipient.
  • lubricants e.g, silica, anhydrous colloidal silica, talcum, stearic acid, its magnesium or calcium salt (e.g, magnesium stearate or calcium stearate), metallic stearates, colloidal silicon dioxide, hydrogenated vegetable oil, corn starch, sodium benzoate, sodium acetate and/or polyethyleneglycol; for tablets also (c) binders, e.g, magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, polyvinylpyrrolidone and/or hydroxypropyl methylcellulose; if desired (d) disintegrants, e.g, starches
  • the tablet contains a mixture of hydroxypropyl methylcellulose, polyethyleneglycol 6000 and titatium dioxide. Tablets may be either film coated or enteric coated according to methods known in the art.
  • Liquid preparations for oral administration can take the form of, for example, solutions, syrups, or suspensions, or they can be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives, for example, suspending agents, for example, sorbitol syrup, cellulose derivatives, or hydrogenated edible fats; emulsifying agents, for example, lecithin or acacia; non-aqueous vehicles, for example, almond oil, oily esters, ethyl alcohol, or fractionated vegetable oils; and preservatives, for example, methyl or propyl-p-hydroxybenzoates or sorbic acid.
  • the preparations can also contain buffer salts, flavoring, coloring, and/or sweetening agents as appropriate. If desired, preparations for oral administration can be suitably formulated to give controlled release of the active compound.
  • Controlled release parenteral formulations of the compositions of the present invention can be made as implants, oily injections, or as particulate systems.
  • Particulate systems include microspheres, microparticles, microcapsules, nanocapsules, nanospheres, and nanoparticles.
  • Polymers can be used for ion-controlled release of compositions of the present invention.
  • Various degradable and nondegradable polymeric matrices for use in controlled drug delivery are known in the art (Langer R., Accounts Chem. Res., 26:537-542 (1993)).
  • the block copolymer, polaxamer 407 exists as a viscous yet mobile liquid at low temperatures but forms a semisolid gel at body temperature. It has shown to be an effective vehicle for formulation and sustained delivery of recombinant interleukin 2 and urease (Johnston et ak, Pharm. Res., 9:425-434 (1992); and Pec et ah, ./. Parent. Sci.
  • hydroxyapatite has been used as a microcarrier for controlled release of proteins (Ijntema et ak, Int. J Pharm., 112:215-224 (1994)).
  • liposomes are used for controlled release as well as drug targeting of the lipid-capsulated drug (Betageri et ak, LIPOSOME DRUG DELIVERY SYSTEMS, Technomic Publishing Co., Inc., Lancaster, PA (1993)).
  • Numerous additional systems for controlled delivery of therapeutic proteins are known. See, e.g., U.S. Pat. No.
  • compositions or medicaments comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g ., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti -RORy antibody, and/or an interfering RNA (e.g., small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) can be administered to a subject at a therapeutically effective dose to treat the subject’s cancer, as described herein.
  • RORy inhibitors e.g., a compound of Formula I (e.g ., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table
  • compositions or medicaments comprising one or more RORy inhibitors and one or more statins can be co- administered to a subject in combination with an effective amount of an anticancer drug at a therapeutically effective dose to treat the subject’s cancer, as described herein.
  • the pharmaceutical composition or medicament comprising one or more RORy inhibitors and one or more statins is administered to a subject in an amount sufficient to elicit an effective therapeutic response in the subject.
  • the pharmaceutical composition or medicament comprising one or more RORy inhibitors and one or more statins can be co-administered to a subject at a therapeutically effective dose in combination with an effective amount of an anticancer drug to elicit an effective therapeutic response in the subject.
  • the methods comprise first administering one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti- RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) to a patient having cancer, and then administering an anticancer drug, such as an anti-androgen drug and/or a chemotherapeutic agent, to the patient.
  • RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and
  • the methods comprise first administering an anticancer drug, such as an anti-androgen drug and/or a chemotherapeutic agent, to a patient having cancer, and then administering one or more RORy inhibitors and one or more statins to the patient.
  • the methods comprise co-administering one or more RORy inhibitors and one or more statins with an anticancer drug, such as an anti-androgen drug and/or a chemotherapeutic agent, to a patient having cancer.
  • the methods of administration comprise administering one or more RORy inhibitors (e.g., a compound of Formula I (e.g ., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g., small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), alone or in combination with enzalutamide to a patient in need thereof (e.g, a patient in need of cancer treatment).
  • RORy inhibitors e.g., a compound of Formula I (e.g ., XY018, XY063), GSK805, SR2211, and/or a
  • the methods of administration comprise administering one or more RORy inhibitors and one or more statins, alone or in combination with abiraterone to a patient in need thereof.
  • the methods comprise administering one or more RORy inhibitors and one or more statins, alone or in combination with bicalutamide to a patient in need thereof.
  • the methods comprise administering one or more RORy inhibitors and one or more statins, alone or in combination with a taxane such as docetaxel to a patient in need thereof.
  • the methods comprise administering one or more RORy inhibitors and one or more statins, alone or in combination with tamoxifen to a patient in need thereof.
  • the present invention provides a method of delivering an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) to a patient having cancer such as prostate cancer (e.g, CRPC).
  • RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43
  • the RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) described herein are useful in the manufacture of a pharmaceutical composition or a medicament.
  • a compound of Formula I e.g, XY018, XY063
  • GSK805 e.g, SR2211
  • a compound listed in Table 5 such as VTP-43742
  • an anti-RORy antibody e.g, small-interfering RNA
  • statins
  • a pharmaceutical composition or medicament can be administered to a subject in need thereof, e.g. a patient having a cancer such as breast cancer, prostate cancer (e.g ., CRPC), lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma.
  • a cancer such as breast cancer, prostate cancer (e.g ., CRPC), lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma.
  • compositions or medicaments for use in the present invention can be formulated by standard techniques using one or more physiologically acceptable carriers or excipients. Suitable pharmaceutical carriers are described herein and in "Remington's Pharmaceutical Sciences” by E.W. Martin. Compounds and agents of the present invention and their physiologically acceptable salts and solvates can be formulated for administration by any suitable route, including via inhalation, topically, nasally, orally, intravenously, parenterally, or rectally.
  • Typical formulations for topical administration include creams, ointments, sprays, lotions, and patches.
  • the pharmaceutical composition can, however, be formulated for any type of administration, e.g., intradermal, subdermal, intravenous, intramuscular, intranasal, intracerebral, intratracheal, intraarterial, intraperitoneal, intravesical, intrapleural, intracoronary or intratumoral injection, with a syringe or other devices.
  • Formulation for administration by inhalation (e.g, aerosol), or for oral or rectal administration is also contemplated.
  • Suitable formulations for transdermal application include an effective amount of one or more compounds described herein, optionally with a carrier.
  • Preferred carriers include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host.
  • transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • Matrix transdermal formulations may also be used.
  • a pharmaceutical composition or a medicament can take the form of, for example, a tablet or a capsule prepared by conventional means with a pharmaceutically acceptable excipient.
  • the present invention provides tablets and gelatin capsules comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), alone or in combination with other compounds such as anti-androgen drugs and/or docetaxel, or a dried solid powder of these drugs, together with one or more ROR
  • Tablets may be either film coated or enteric coated according to methods known in the art.
  • Liquid preparations for oral administration can take the form of, for example, solutions, syrups, or suspensions, or they can be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives, for example, suspending agents, for example, sorbitol syrup, cellulose derivatives, or hydrogenated edible fats; emulsifying agents, for example, lecithin or acacia; non-aqueous vehicles, for example, almond oil, oily esters, ethyl alcohol, or fractionated vegetable oils; and preservatives, for example, methyl or propyl-p-hydroxybenzoates or sorbic acid.
  • the preparations can also contain buffer salts, flavoring, coloring, and/or sweetening agents as appropriate. If desired, preparations for oral administration can be suitably formulated to give controlled release of the active compound(s).
  • compositions and formulations set forth herein can be formulated for parenteral administration by injection, for example by bolus injection or continuous infusion.
  • Formulations for injection can be presented in unit dosage form, for example, in ampoules or in multi-dose containers, with an added preservative.
  • Injectable compositions are preferably aqueous isotonic solutions or suspensions, and suppositories are preferably prepared from fatty emulsions or suspensions.
  • the compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers.
  • the active ingredient(s) can be in powder form for constitution with a suitable vehicle, for example, sterile pyrogen-free water, before use.
  • a suitable vehicle for example, sterile pyrogen-free water
  • they may also contain other therapeutically valuable substances.
  • the compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1 to 75%, preferably about 1 to 50%, of the active ingredient(s).
  • compositions of the present invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, for example, dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide, or other suitable gas.
  • a suitable propellant for example, dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide, or other suitable gas.
  • the dosage unit can be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, for example, gelatin for use in an inhaler or insufflator can be formulated containing a powder mix of the compound(s) and a suitable powder base, for example, lactose or starch.
  • compositions set forth herein can also be formulated in rectal compositions, for example, suppositories or retention enemas, for example, containing conventional suppository bases, for example, cocoa butter or other glycerides.
  • the active ingredient(s) can be formulated as a depot preparation.
  • Such long-acting formulations can be administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection.
  • one or more of the compounds described herein can be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • a pharmaceutical composition or medicament of the present invention can comprise (i) an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), and (ii) optionally an anticancer drug such as an anti-androgen drug (e.g, enzalutamide, abiraterone, bicalutamide), a chemotherapeutic agent such as a taxane (e
  • RORy inhibitors
  • the therapeutic agent(s) may be used individually, sequentially, or in combination with one or more other such therapeutic agents (e.g., a first therapeutic agent, a second therapeutic agent, a compound of the present invention, etc.). Administration may be by the same or different route of administration or together in the same pharmaceutical formulation.
  • compositions or medicaments can be administered to a subject at a therapeutically effective dose to prevent, treat, sensitize, or control a cancer such as prostate cancer (e.g ., CRPC), lung cancer, breast cancer (e.g, TNBC), liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma as described herein.
  • a cancer such as prostate cancer (e.g ., CRPC), lung cancer, breast cancer (e.g, TNBC), liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma as described herein.
  • the pharmaceutical composition or medicament is administered to a subject in an amount sufficient to elicit an effective therapeutic response in the subject.
  • An effective therapeutic response includes a response that at least partially arrests or slows the symptoms or complications of the cancer. An amount adequate to accomplish this is defined as a“therapeutically effective dose.”
  • a unit dosage for oral administration to a mammal of about 50 to about 70 kg may contain between about 5 and about 500 mg, about 25-200 mg, about 100 and about 1000 mg, about 200 and about 2000 mg, about 500 and about 5000 mg, or between about 1000 and about 2000 mg of the active ingredient.
  • a unit dosage for oral administration to a mammal of about 50 to about 70 kg may contain about 10 mg, 20 mg, 25 mg, 50 mg, 75 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 700 mg, 800 mg, 900 mg, 1,000 mg, 1,250 mg, 1,500 mg, 2,000 mg, 2,500 mg, 3,000 mg, or more of the active ingredient.
  • a dosage of the active compound(s) of the present invention is a dosage that is sufficient to achieve the desired effect.
  • Optimal dosing schedules can be calculated from measurements of active agent accumulation in the body of a subject. In general, dosage may be given once or more of daily, weekly, or monthly. Persons of ordinary skill in the art can easily determine optimum dosages, dosing methodologies and repetition rates.
  • Optimum dosages, toxicity, and therapeutic efficacy of the compositions of the present invention may vary depending on the relative potency of the administered composition and can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, for example, by determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio, LD50/ED50.
  • Agents that exhibit large therapeutic indices are preferred. While agents that exhibit toxic side effects can be used, care should be taken to design a delivery system that targets such agents to the site of affected tissue to minimize potential damage to normal cells and, thereby, reduce side effects.
  • Optimal dosing schedules can be calculated from measurements of active ingredient accumulation in the body of a subject. In general, dosage is from about 1 ng to about 1,000 mg per kg of body weight and may be given once or more daily, weekly, monthly, or yearly. Persons of ordinary skill in the art can easily determine optimum dosages, dosing methodologies and repetition rates.
  • RORy inhibitor e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti- RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and a statin (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) to a human being following established protocols known in the art and the disclosure herein.
  • a statin e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin
  • the data obtained from, for example, animal studies can be used to formulate a dosage range for use in humans.
  • the dosage of compounds of the present invention lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage can vary within this range depending upon the dosage form employed and the route of administration.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
  • Levels in plasma can be measured, for example, by high performance liquid chromatography (HPLC).
  • HPLC high performance liquid chromatography
  • the dose equivalent of a chimeric protein, preferably a composition is from about 1 ng/kg to about 100 mg/kg for a typical subject.
  • a typical composition of the present invention for oral or intravenous administration can be about 0.1 mg to about 10 mg of active ingredient per patient per day; about 1 mg to about 100 mg per patient per day; about 25 mg to about 200 mg per patient per day; about 50 mg to about 500 mg per patient per day; about 100 mg to about 1000 mg per patient per day; or about 1000 mg to about 2000 mg per patient per day.
  • Exemplary dosages include, but are not limited to, about 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 21 mg, 22 mg, 23 mg, 24 mg, 25 mg, 26 mg, 27 mg, 28 mg, 29 mg, 30 mg, 31 mg, 32 mg, 33 mg, 34 mg, 35 mg, 36 mg, 37 mg, 38 mg, 39 mg, 40 mg, 41 mg, 42 mg, 43 mg, 44 mg, 45 mg, 46 mg, 47 mg, 48 mg, 49 mg, 50 mg, 55 mg, 60, mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 700 mg, 800 mg, 900 mg, 1,000 mg, 1,250 mg
  • Exemplary doses of the compositions described herein include milligram or microgram amounts of the composition per kilogram of subject or sample weight (e.g. , about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 5 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram). It is furthermore understood that appropriate doses of a composition depend upon the potency of the composition with respect to the desired effect to be achieved. When one or more of these compositions is to be administered to a mammal, a physician, veterinarian, or researcher may, for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained.
  • the specific dose level for any particular mammal subject will depend upon a variety of factors including the activity of the specific composition employed, the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate of excretion, any drug combination, and the degree of expression or activity to be modulated.
  • a pharmaceutical composition or medicament of the present invention is administered, e.g. , in a daily dose in the range from about 1 mg of compound per kg of subject weight (1 mg/kg) to about lg/kg.
  • the dose is a dose in the range of about 5 mg/kg to about 500 mg/kg.
  • the dose is about 10 mg/kg to about 250 mg/kg.
  • the dose is about 25 mg/kg to about 150 mg/kg.
  • a preferred dose is about 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 18, 20, 25, 30, 40, or 50 mg/kg.
  • the daily dose can be administered once per day or divided into subdoses and administered in multiple doses, e.g., twice, three times, or four times per day.
  • compositions described herein may be administered in different amounts and at different times.
  • certain factors may influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the disease or malignant condition, previous treatments, the general health and/or age of the subject, and other diseases present.
  • treatment of a subject with a therapeutically effective amount of a composition can include a single treatment or, preferably, can include a series of treatments.
  • compositions set forth herein may be administered for at least three consecutive days, often for at least five consecutive days, more often for at least ten, and sometimes for 20, 30, 40 or more consecutive days. While consecutive daily doses are a preferred route to achieve a therapeutically effective dose, a therapeutically beneficial effect can be achieved even if the agents are not administered daily, so long as the administration is repeated frequently enough to maintain a therapeutically effective concentration of the agents in the subject. For example, one can administer the agents every other day, every third day, or, if higher dose ranges are employed and tolerated by the subject, once a week.
  • the RORy inhibitor e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small -interfering RNA)) and/or statin (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) are orally administered.
  • a compound of Formula I e.g, XY018, XY063
  • GSK805 e.g, SR2211
  • a compound listed in Table 5 such as VTP- 43742
  • an anti -RORy antibody e.g, small -interfering RNA
  • statin e.g, atorvastat
  • the RORy inhibitor and/or statin is orally administered to a subject (e.g, an adult human) at a daily dose of approximately 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 21 mg, 22 mg, 23 mg, 24 mg, 25 mg, 26 mg, 27 mg, 28 mg, 29 mg, 30 mg, 31 mg, 32 mg, 33 mg, 34 mg, 35 mg, 36 mg, 37 mg, 38 mg, 39 mg, 40 mg, 41 mg, 42 mg, 43 mg, 44 mg, 45 mg, 46 mg, 47 mg, 48 mg, 49 mg, 50 mg, 55 mg, 60, mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg,
  • the RORy inhibitor and/or statin are orally administered to a subject (e.g, an adult human) at a daily dose of between about 1,000 mg and about 2,000 mg per day. In some embodiments, the RORy inhibitor and/or statin is orally administered.
  • the RORy inhibitor and/or statin are orally administered to a subject (e.g, an adult human) at a daily dose of approximately 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 21 mg, 22 mg, 23 mg, 24 mg, 25 mg, 26 mg, 27 mg, 28 mg, 29 mg, 30 mg, 31 mg, 32 mg, 33 mg, 34 mg, 35 mg, 36 mg, 37 mg, 38 mg, 39 mg, 40 mg, 41 mg, 42 mg, 43 mg, 44 mg, 45 mg, 46 mg, 47 mg, 48 mg, 49 mg, 50 mg, 55 mg, 60, mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg,
  • RORy inhibitor and/or statin are orally administered to a subject (e.g, an adult human) at a daily dose of between 25 and 200 mg per day.
  • the RORy inhibitor and/or statin and an anti-androgen drug are orally co- administered.
  • the RORy inhibitor and/or statin can be co-administered at a daily oral dose of between about 25 mg and 1000 mg mg per day with the anti -androgen drug at a daily oral dose of between about 25 mg and 2000 mg per day.
  • the methods comprise sequentially administering one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), followed by one or more anticancer drugs such as an anti androgen drug (e.g, enzalutamide, abiraterone, bicalutamide), a chemotherapeutic agent such as a taxane (e.g, docetaxel) or tamoxifen, and combinations
  • RORy inhibitors
  • an efficacious or effective amount of an composition is determined by first administering a low dose or small amount of the composition, and then incrementally increasing the administered dose or dosages, adding a second or third medication as needed, until a desired effect of is observed in the treated subject with minimal or no toxic side effects.
  • compositions Single or multiple administrations of the compositions are administered depending on the dosage and frequency as required and tolerated by the patient.
  • the composition should provide a sufficient quantity of the compositions of this invention to effectively treat the patient.
  • the dose is sufficient to treat or ameliorate symptoms or signs of disease without producing unacceptable toxicity to the patient.
  • kits and systems can be prepared according to the present invention, depending upon the intended user of the kit and system and the particular needs of the user.
  • the present invention provides a kit that includes ( e.g ,., an effective amount of) one or more RORy inhibitors (e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin).
  • RORy inhibitors e.g., a compound of Formula I (e.g., XY018, XY063), G
  • the present invention provides a kit that includes one or more RORy inhibitors, one or more statins, and one or more anticancer drugs such as an anti-androgen drug (e.g, enzalutamide, abiraterone, and/or bicalutamide) and/or a chemotherapeutic agent (e.g, tamoxifen and/or a taxane such as docetaxel).
  • an anti-androgen drug e.g, enzalutamide, abiraterone, and/or bicalutamide
  • a chemotherapeutic agent e.g, tamoxifen and/or a taxane such as docetaxel
  • kits described herein can include a label describing a method of administering one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti- RORy antibody, and/or an interfering RNA (e.g., small-interfering RNA)), one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, and/or cerivastatin, pravastatin, pitavastatin), and/or one or more anticancer drugs (e.g, to a subject, such as a subject in need of cancer treatment).
  • RORy inhibitors e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and
  • kits described herein can include a label describing a method of treating cancer in a subject with a cancer such as breast cancer (e.g, TNBC), prostate cancer (e.g, CRPC), lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma.
  • a cancer such as breast cancer (e.g, TNBC), prostate cancer (e.g, CRPC), lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma.
  • compositions of the present invention including but not limited to, compositions comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, and/or pitavastatin), and optionally one or more anticancer drugs may, if desired, be presented in a bottle, jar, vial, ampoule, tube, or other container-closure system approved by the Food and Drug Administration (FDA) or other regulatory body, which may provide one or more dosages containing the compounds.
  • RORy inhibitors e.g., a compound of
  • the package or dispenser may also be accompanied by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, the notice indicating approval by the agency.
  • the kit may include a formulation or composition as described herein, a container closure system including the formulation or a dosage unit form including the formulation, and a notice or instructions describing a method of use as described herein.
  • the kit includes a container which is compartmentalized for holding the various elements of a formulation (e.g., the dry ingredients and the liquid ingredients) or composition, instructions for making the formulation or composition, and instructions for administering the formulation or composition for enhancing the immune response in a subject with a cancer.
  • a formulation e.g., the dry ingredients and the liquid ingredients
  • instructions for making the formulation or composition e.g., the instructions for making the formulation or composition
  • instructions for administering the formulation or composition for enhancing the immune response in a subject with a cancer e.g., the dry ingredients and the liquid ingredients
  • the kit may include the pharmaceutical preparation(s) in dehydrated or dry form, with instructions for its rehydration (or reconstitution) and administration.
  • Kits with unit doses of the compounds described herein, e.g. in oral, rectal, transdermal, or injectable doses (e.g, for intramuscular, intravenous, or subcutaneous injection), are provided.
  • an informational package insert describing the use and attendant benefits of the composition for enhancing the immune response in a subject with a cancer such as prostate cancer (e.g ., CRPC), lung cancer, breast cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma may be included in addition to the containers containing the unit doses.
  • a cancer such as prostate cancer (e.g ., CRPC), lung cancer, breast cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma
  • a cancer such as prostate cancer (e.g ., CRPC), lung cancer, breast cancer, liver cancer, ovarian
  • Some embodiments of the present invention provide packages that include one or more RORy inhibitors (e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), and optionally one or more anticancer drugs such as an anti androgen drug (e.g, enzalutamide, abiraterone, and/or bicalutamide) and/or a chemotherapeutic agent (e.g, tamoxifen and/or a taxane such
  • Metabolic reprogramming a hallmark of cancer, fuels the malignant tumor growth and survival by providing energy, reducing power and building blocks.
  • One metabolic process that is often deregulated in tumors of most cancer types is the mevalonate (MV A) pathway which produces sterols such as cholesterol, isoprenoids, and ubiquinone that are essential for tumor growth (4).
  • MV A mevalonate
  • the major regulators of the MVA pathway enzyme expression are the transcription factors sterol regulatory element-binding protein 1 and 2 (SREBP1 and -2) and LXRs (2, 5). SREBPs play a pivotal role in the maintenance of cholesterol homeostasis.
  • SREBPs In response to a low intracellular sterol level, SREBPs dissociate from the INSIGs proteins at the endoplasmic reticulum (ER) and translocate from the ER to the Golgi where they are cleaved by proteases.
  • the cleaved N-terminal portion of SREBPs translocate to the nucleus and bind to target gene promoters at SRE sites to activate the expression of MVA pathway and cholesterol synthesis enzymes such as HMGCS, HMGCR and MVK for de novo cholesterol synthesis, as well as genes involved in cholesterol homeostasis such as low density lipoprotein receptor (LDLR), which facilitates the cellular uptake of exogenous lipoprotein-derived cholesterol.
  • LDLR low density lipoprotein receptor
  • Statins are a class of cholesterol/lipid-lowering drugs for treating patients with hypercholesterolemia to reduce the associated high risk of cardiovascular disease (CVD) (1).
  • Statins are small-molecule inhibitors of 3 -hydroxy-3 -methyl glutary coenzyme A reductase (HMGCR), a rate-limiting enzyme in hepatic cholesterol biosynthesis.
  • HMGCR 3 -hydroxy-3 -methyl glutary coenzyme A reductase
  • Statin treatment strongly reduces cholesterol production initially, and, as an adaptive response, the reduced cellular cholesterol level triggers the SREBP-mediated activation of gene expression including that of LDLR in liver and other tissues, which leads to an increased uptake of LDL from the circulation, hence lowering blood cholesterol level.
  • statins are associated with a statistically significant reduction of the risk of cancer relapse and cancer-related death.
  • Such findings have prompted many clinical trials of statins as cancer therapeutics in patients with different types of cancers including lung cancer, prostate cancer and breast cancer.
  • no clinical trial has shown a clear and sustained benefit from treatment with statins in the cancer patients.
  • statins can result in inhibition of cell growth and cell death (1).
  • tumor inhibition efficacy of statins is unclear for most cancer types.
  • a few animal studies may have shown tumor inhibitory effects, the data were obtained by using very high doses of statins which are deemed unsafe and unsuitable for clinical use in cancer patients. Therefore, currently the potential value of statin use in cancer prevention and treatment is unclear.
  • statin treatment can lead to increased expression of MVA pathway genes. Due to the oncogenic regulation of SREBPs and the MVA pathway, cancer cells may have a heightened cholesterol homeostasis response mediated by SREBPs to maintain the levels of MVA pathway products following statin treatment (4). Therefore, the development of strategies to combine statins with therapeutics that block the heightened homeostasis such as inhibitors of SREBPs can be of high value in the effective treatment of many types of cancer.
  • RORy small-molecule inhibitors or genetic silencing of a member of the nuclear receptor family protein, RORy, strongly inhibit the growth of cancer cell and xenograft tumors and induce marked cell death in prostate cancer (6).
  • genetic silencing of RORy resulted in strong inhibition of growth and survival of breast cancer and lung cancer cells.
  • Targeting RORy with its inhibitors also strongly inhibited the growth and survival of several types of cancer cells. Because of the apparent function of RORy in many tumor types, we postulated that RORy might control a pathway or process common to most cancer types.
  • RORy is required for cancer cell growth and survival of different cancer types
  • the cholesterol synthesis pathway involves more than 20 different steps of enzymatic activities, with steps carried out by HMGCR and SQLE being the rate-limiting steps in the pathway.
  • HMGCR in particular, is the target of statins, a class of cholesterol/lipid-lowering drugs for treating patients with hypercholesterolemia, in order to reduce their high risk of cardiovascular diseases.
  • statins a class of cholesterol/lipid-lowering drugs for treating patients with hypercholesterolemia, in order to reduce their high risk of cardiovascular diseases.
  • FIG. 4B when examined in a TNBC cell line MDA-MB468, the mRNA expression of most of the enzyme genes in the pathway, including HMGCR and SQLE, was significantly inhibited by 2.5 mM RORy inhibitor XY018 (also known as F18) and GSK805. As shown in FIG.
  • statin treatment can induce an adaptive response in cells by SREBP-dependent activation of gene expression including that of cholesterol synthesis and LDLR.
  • SREBP-dependent activation of gene expression including that of cholesterol synthesis and LDLR.
  • FIG. 5 treatment of TNBC cells with 1.25 or 2.5 mM atorvastatin resulted in robust increases of most of the cholesterol synthesis genes (e.g., HMGCS1 HMGCR, MVD, SQLE, LSS, CYP51A1, and DHCR7).
  • treatment of the cells with the RORy inhibitors XY018 and XY063 at the same concentrations caused significant decreases in their expression.
  • the RORy inhibitors XY018 and XY063 alone did not display a strong growth inhibition, either.
  • the combination of 0.125 pM XY018 or XY063 with different concentrations of simvastatin caused marked growth inhibition that was much more than additive from either alone.
  • FIGS. 6B-6D similar synergistic effects were observed in other TNBC cells (SUM159 and MB468) and with a different statin (z.e., atorvastatin).
  • statins may have different effects on cancer cell growth and survival (1).
  • Table 3 presents the IC50 values of different statins measured by CellTiter-Glo assay, and shows than when combined with relatively low concentrations of RORy inhibitors, except that of pravastatin, the IC50 values of seven different statins (z.e., atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, and pitavastatin) are all significantly decreased (approximately 3- to lO-fold), indicating that the RORy inhibitors potently sensitize the TNBC cells to inhibition by statins.
  • statins z.e., atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, and pitavastatin
  • RORy inhibitors in combination with statins display strong synergy in the killing of cancer cells of lung cancer, gastric cancer and prostate cancer, including cancer cells that are resistant to current therapeutics
  • statins Similar to what was observed with TNBC cells, when treating lung cancer cells A549 and H358 with the different statins in combination with RORy inhibitors, the IC50 values of seven different statins (i.e., atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, and pitavastatin) are all strongly decreased (ranging from a 3- to over a 20-fold decrease), indicating that the RORy inhibitors potently sensitize lung cancer cells to inhibition by statins (Table 4).
  • statins i.e., atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, and pitavastatin
  • A549 and H358 NSCLC cells express wild type EGFR and thus are resistant to EGFR inhibitors such as gefitinib and erlotinib, which target kinase hyperactive-mutant forms of EGFR in the cancer cells.
  • EGFR inhibitors such as gefitinib and erlotinib, which target kinase hyperactive-mutant forms of EGFR in the cancer cells.
  • results shown in FIG. 9 demonstrate that, similar to the data obtained with TNBC and lung cancer cells, in models of prostate cancer (22Rvl and C4- 2B), combinations of XY018 or XY063 with different concentrations of simvastatin or atorvastatin also caused marked growth inhibition that was much more than additive from either alone. Similar results, shown in FIG. 10, were obtained with several gastric cancer cell lines (AGS, HGC-27 and MGC-803). Notably, the two prostate cancer cell lines (22Rvl and C4-2B) are castration-resistant prostate cancer (CRPC) cells, suggesting that the combination of the RORy inhibitors and statins can be effective in the treatment of CRPC.
  • CRPC castration-resistant prostate cancer
  • VTP -43742 also known as AGN 242428
  • AGN 242428 is one such compound that has been on clinical trial for its potential use in treatment of psoriasis.
  • FIG. 11 treatment of human breast cancer cells (MDA-MB468) and CRPC prostate cancer cells (C4-2B) with VTP-43742, either alone or in combination with different concentrations of statins (atorvastatin or simvastatin), resulted in strong growth inhibition.
  • statins that display strong synergism with the RORy inhibitors in killing cancer cells include all of the major statin drugs that are clinically used to treat hypercholesterolemia, including atorvastatin, simvastatin, fluvastatin, lovastatin, mevastatin, pitavastatin, and rosuvastatin.
  • HCC70, MDA-MB468, HCC1500, ZR75-1, and HCC1954 TNBC cancer cells were cultured in RPMI1640.
  • MDA-MB231, MCF-7, MDA-MB361, and BT20 cells were cultured in DMEM.
  • BT549 cells were cultured in RPMI1640 plus 1% insulin.
  • Hs578T cells were cultured in DMEM plus 1% insulin.
  • SUM149 and SUM159 were cultured in DMEM/F12 medium plus insulin and hydrocortisone. The culture media for SEIM149 and SEIM159 were supplemented with 5% FBS (Hyclone) while other media was supplemented with 10% FBS.
  • HCC70, MDA-MB468, MDA-MB231, DMEM, and BT549 were obtained from ATCC.
  • the TNBC cancer cell lines were recently authenticated by ATCC using STR profiling. Gastric cancer cells were obtained from ATCC and cultured in RPMI plus 10% FBS. Prostate cancer cell line source and culture conditions were described previously in (6). Cell lines were regularly tested for being negative for mycoplasma.
  • Sources for chemicals were as follows: GSK805 was obtained from Calbiochem or WuXi AppTec; XY018 and XY063 were obtained from WuXi AppTec; VTP-43742 was synthesized by the laboratory of one of the inventors. Statins were obtained from Selleck. Other chemicals were obtained from Sigma unless indicated otherwise. gRT-PCR and Western blottins analysis
  • siRNAs for gene knockdown were purchased from Dharmacon.
  • the siRNA target sequences for RORC were published previously (6).
  • Transfections were performed with OptiMEM (Invitrogen) and Dharmafectin #1 (Dharmacon) following the manufacturer’s instructions.
  • sgRNAs were designed using the MIT CRISPR design software (crispr.mit.edu). Oligos corresponding to the sgRNAs were synthesized and cloned into lentiCRISPR v2 vectors following lentiCRISPRv2 and lentiGuide oligo cloning protocol (Addgene, plasmid #52961). Lentiviral particles were produced in 293T cells as in our previous study (6). TNBC cancer cells were plated at 2 c 10 5 cells per well in 6-well plates. Sixteen hours later, 1 mL of virus-containing supernatant with 10 ng polybrene was added to the cells. After 4 to 6 hours, medium was changed to regular medium and cultured for another 72 hours before being harvested for cell number and protein expression analysis.
  • HCC70 cells were treated with vehicle or the antagonists XY018 or XY063 or atorvastatin, or a combination of antagonists and atorvastatin for 24 hours before RNA extraction.
  • RNA-seq libraries from 1 pg total RNA were prepared using Illumina Tru-Seq RNA Sample Prep Kit, according to the manufacturer’s instructions. Libraries were validated with an Agilent Bioanalyzer (Agilent Technologies, Palo Alto, CA). Sequencing was performed on an Illumina HiSeq 2000 sequencer at BGI Tech (Hong Kong). The FASTQ- formatted sequence data were analyzed using a standard BWA-Bowtie-Cufflinks.
  • mice having a size of six or more were estimated to have a high statistic power, based on power calculation and previous studies involving the same xenograft models.
  • the mice were randomized and then administered, via oral gavage, five times per week, 100 pL of vehicle (with a formulation of 15% Cremophor EL, Calbiochem, 82.5% PBS, and 2.5% DMSO), statin (in PBS), RORy antagonist/inhibitor (in a formulation of 15% Cremophor EL, 82.5% PBS and 2.5% DMSO), or a combination of RORy inhibitor and statin (in their respective solvents).
  • vehicle with a formulation of 15% Cremophor EL, Calbiochem, 82.5% PBS, and 2.5% DMSO
  • statin in PBS
  • RORy antagonist/inhibitor in a formulation of 15% Cremophor EL, 82.5% PBS and 2.5% DMSO
  • a combination of RORy inhibitor and statin in their respective solvents
  • Tumor volumes were monitored using calipers with volumes calculated using the equation: p/6 (length x width 2 ). Body weight during the course of the study was also monitored. At the end of the studies mice were killed and tumors were dissected and weighed. Additionally, the organs of kidney, heart, lung, liver, spleen and serum were harvested. The procedures were approved by the Institutional Animal Care and Else Committee (IACUC) of University of California, Davis.
  • IACUC Institutional Animal Care and Else Committee
  • EXAMPLE 2 Using a combination of RORy inhibitors and statins synergistically sensitizes cancer cells to statins and inhibits cancer cell growth
  • FIG. 16A shows that the combination index (Cl) for XY018 and simvastatin was 0.179, which indicates a high synergy in growth inhibition, as calculated using data shown at the bottom of FIG. 16A from single or combined simvastatin and XY018 treatment of human lung cancer cell line PC9.
  • PC9 cell viability was measured by CellTiter-Glo (Promega) after 4 days of treatment with XY018, simvastatin or their combination in the indicated concentrations (in pM).
  • FIGS. 16B and 16C human lung cancer A549 cells were treated by the indicated increasing concentrations of a statin alone (fluvastatin (FIG. 16B) or lovastatin FIG. 16C) plus vehicle control), or the different concentrations of the indicated statins in combination with RORy inhibitor/antagonist XY018 or XY063 (2.5 pM) for 4 days.
  • Cell viability was measured by CellTiter-Glo (Promega). Cell viability obtained from cells treated with vehicle only (without either the statin or the RORy inhibitor/antagonist) was set as 100. The experiments were repeated three times.
  • FIG. 17 shows that combined treatment with RORy inhibitors/antagonists and statins was synergistic in the inhibition of lung cancer tumor growth.
  • FIG. 17A Tumor volume in mean ⁇ s.e.m. is shown in FIG. 17A
  • FIG. 17B Tumor weight in mean ⁇ s.d. is shown in FIG. 17B
  • dissected tumor images, at last treatment are shown in FIG. 17C.
  • FIG. 18 shows that a combined treatment of cancer cells with a RORy inhibitor and statin synergistically inhibited oncogenic kinase signaling by receptor tyrosine kinases (RTKs) and serine/threonine kinases.
  • RTKs receptor tyrosine kinases
  • FIG. 18 A EGFR-mutant human lung cancer PC9 cells were treated for 24 hours with atorvastatin/ ATV (0, 1, 5 or 10 mM) either alone (with control vehicle), or in combination with 1.25 pM or 5 pM RORy inhibitor XY018. Cells were then harvested for preparation of cell lysates.
  • Immunoblotting analysis of the cell lysates with antibodies against specific proteins as indicated at right to each row of bands (anti-EGFR to detect total amount of EGFR, anti- p/phospho-EGFR (Tyrl068) to detect signaling-activated EGFR, anti -ART to detect total amount of ART, anti-p/phospho-ART (Ser473) to detect signaling-activated ART, anti-ERK to detect total amount of the p44/42 MAPK ERK1/2, and anti-p/phospho-ERK (Thr202/Tyr204) to detect signaling-activated ERK). Immunoblotting with anti-GAPDH protein was also performed to demonstrate loading of equal amount of cell lysates.
  • KRAS-mutant human lung cancer A549 cells were treated for 24 hours with control vehicle, XY018 (2.5 pM), XY063 (2.5 pM), atorvastatin (10 pM) or their combination in serum-free media, followed by 50 ng/mL of growth factor EGF stimulation for 15 minutes. Cells were then harvested for preparation of cell lysates and immunoblotting analysis with antibodies as above for the data shown in FIG. 18 A.
  • FIG. 19 shows biochemical analysis of mouse serum samples for the assessment of any significant alteration of key blood/serum biochemical parameters or toxicity indicators.
  • SCID mice were treated for 63 days, via oral gavage, with RORy inhibitors alone (XY018, 10 mg/kg, p.o. or XY063, 10 mg/kg, p.o) or atorvastatin (20 mg/kg, p.o) alone or RORy inhibitors combined with atorvastatin (XY018, 10 mg/kg, p.o. + atorvastatin 20 mg/kg, p.o or XY063, 10 mg/kg, p.o.
  • RORy inhibitors cause tumor regression and block metastasis.
  • mice bearing MB468-derived orthotopic xenograft tumors i.p.
  • two different doses of XY018 We found that at a relatively low dose of 2.5 mg/kg the antagonist was effective in inhibition of the tumor growth while a higher dose (5 mg/kg) displayed a complete tumor growth blockade for over 7 weeks (FIG. 20A). Similar potent tumor-inhibition activities of antagonists GSK805 and XY018 at 5 mg/kg were observed in additional TNBC HCC70 and SUM159 cell-derived xenografts (FIGS. 20B, 20C). To provide more relevant data for a clinical setting, we measured the oral dosing (p.
  • LM2 lung-metastatic model
  • NSG (JAX stock #005557) mice were purchased from the Jackson Laboratory. SCID C. B -17 mice or Balb/c nu/nu athymic mice were purchased from Envigo. Mice were housed under standard conditions with free access to food and water, under a 12 h light/l2 h dark cycle in a temperature-controlled environment. Mice were fed a standard rodent chow diet (Envigo Teklad 2918). For cell line-derived xenograft, cells (2 c 106 for each of the human cancer cells or 3 x 104 mouse 4T1) were mixed with Matrigel as 50% suspension.
  • the cells injected, in a volume of 0.1 ml, bilaterally into inguinal mammary glands of 4- 6-week-old female C.B-17-SCID mice.
  • PDX-1079 JAX ID: TM01079
  • PDX-1173 JAX ID: J000101173
  • Animal group size of six or more was estimated to have a high statistic power, based on power calculation and previous studies involving the same xenograft models.
  • the concentrations of drug and the routes of drug administration are indicated in each figure.
  • mice When the tumor volumes reached to the indicated volume, mice were randomized and then administered with 100 pl of vehicle (with a formulation of 15% Cremophor EL, Calbiochem, 82.5% PBS, and 2.5% DMSO), statins (in PBS), or RORy antagonists (in a formulation of 15% Cremophor EL, 82.5% PBS and 2.5% DMSO), or their combinations (in their respective solvent).
  • Tumor volumes were monitored using calipers with volume calculated using the equation: p/6 (length c width2). Body weight and survival during the course of the study was also monitored. At the end of the studies mice were killed and tumors were dissected and weighed. Additionally, the organs and whole blood were harvested.
  • mice For effects on lung metastasis from orthotopic 4T1 murine tumors, female Balb/c mice were inoculated with 4T1 cells (3 x 104) into the inguinal mammary gland fat pad. Once the tumors reached 250 mm3 at the primary sites they were surgically removed. Mice were then treated daily with XY018 (20 mg/kg, i.p.) or vehicle for 4 weeks. For histological analysis, mice were euthanized at the end of the treatments, and tumor nodules in the lungs were analyzed by H&E staining for morphology.
  • mice For effects on lung colonization, 2 x 105 luciferase-expressing MDA-MB231-LM2 (4175) cells were injected into each nude mouse via tail. The mice were treated daily with vehicle or 20 mg/kg XY018, i.p. for 4 weeks. Tumor growth in the lungs was monitored for bioluminescence with a Xenogen IVIS-200 system.
  • a method for preventing or treating a cancer in a subject comprising administering to the subject an effective amount of a RORy inhibitor and a statin.
  • the RORy inhibitor is a small- molecule compound, an anti -RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
  • n is an integer selected from the group consisting of 0, 1, 2, or 3;
  • Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF 3 )2, heterocyclyl, and cycloalkyl,
  • R5 is selected from the group consisting of H, and C1-C3 alkyl group
  • R 2 is selected from the group consisting of H, halogen, and alkyl
  • R 3 is selected from the group consisting of H and alkyl
  • R 4 is selected from the group consisting of C0-C4 alkylene-R 6 , C0-C4 alkylene- Rvcycloalkyl, and C 0 -C 4 alkylene-Rvheterocyclyl,
  • R 6 is selected from the group consisting of -Rx, -ORx, -CORx, -COORx, -S(0) m R 8 , cycloalkyl, and heterocyclyl, m is 0 or 2
  • R 7 is selected from the group consisting of -OR9, -C(0)R9, -NR9, -SR9, -S(0)R9, -S(0) 2 R9,
  • Rx is selected from the group consisting of H, and Ci-C 3 alkyl group, and R 9 is Ci-C 3 alkyl ene;
  • each cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, carboxy, amino, -CONFh, -COOR10, -COR10, -OR10, -NHCOR10, -NHCOOR10, and -COH(CF 3 ) 2 ,
  • each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono- , bi- or tri-cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, Ci-C 4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -CONFh, -COORio, -CORio, -ORio, -NHCORio, -NHCOORio, -COH(CF 3 )2, -C6H5R11, morpholinyl, piperidinyl, tetrahydrofuranyl, substituted pyridyl group,
  • Rio is independently selected from the group consisting of H, C 1 -C 4 alkyl, and phenyl, and
  • R 11 is independently selected from the group consisting of C 1 -C 4 alkyl, halogen, acetyl, methoxy, and ethoxy.
  • statin is selected from the group consisting of atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, pitavastatin, cerivastatin, pravastatin, and a combination thereof.
  • the cancer is resistant to an anticancer drug.
  • the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
  • the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
  • chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof.
  • cancer selected from the group consisting of breast cancer, prostate cancer, lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, and glioma.
  • TNBC triple- negative breast cancer
  • tamoxifen-resistant breast cancer tamoxifen-resistant breast cancer
  • radiation-resistant breast cancer HER2-positive breast cancer
  • ER-positive breast cancer ER-positive breast cancer
  • lung cancer is a non small-cell lung cancer (NSCLC), K-Ras mutant lung cancer, BRAF mutant lung cancer, EGFR mutant lung cancer, tyrosine kinase inhibitor-resistant lung cancer, or small cell lung cancer (SCLC).
  • NSCLC non small-cell lung cancer
  • K-Ras mutant lung cancer K-Ras mutant lung cancer
  • BRAF mutant lung cancer BRAF mutant lung cancer
  • EGFR mutant lung cancer tyrosine kinase inhibitor-resistant lung cancer
  • SCLC small cell lung cancer
  • a composition comprising a RORy inhibitor and a statin.
  • composition of embodiment 27, wherein the RORy inhibitor is a small-molecule compound, an anti-RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
  • the RORy inhibitor is selected from the group consisting of a compound according to Formula I:
  • n is an integer selected from the group consisting of 0, 1, 2, or 3;
  • Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF 3 )2, heterocyclyl, and cycloalkyl,
  • R 5 is selected from the group consisting of H, and C1-C3 alkyl group
  • R 2 is selected from the group consisting of H, halogen, and alkyl
  • R 3 is selected from the group consisting of H and alkyl
  • R 4 is selected from the group consisting of C0-C4 alkylene-R 6 , C0-C4 alkylene- Rvcycloalkyl, and C 0 -C 4 alkylene-Rvheterocyclyl,
  • R 6 is selected from the group consisting of -Rx, -ORx, -CORx, -COORx, -S(0) m R 8 , cycloalkyl, and heterocyclyl, m is 0 or 2
  • R 7 is selected from the group consisting of -OR9, -C(0)R9, -NR9, -SR9, -S(0)R9, -S(0) 2 R9,
  • Rx is selected from the group consisting of H, and Ci-C 3 alkyl group, and R 9 is Ci-C 3 alkylene;
  • each cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, carboxy, amino, -CONFh, -COOR10, -COR10, -OR10, -NHCOR10, -NHCOOR10, and -COH(CF 3 ) 2 ,
  • each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono- , bi- or tri-cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, C1-C4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -CONFh, -COOR10, -COR10, -OR10, -NHCORio, -NHCOORio, -COH(CF 3 )2, -C6H5R11, morpholinyl, piperidinyl, tetrahydrofuranyl, substituted pyridyl group,
  • Rio is independently selected from the group consisting of H, C 1 -C 4 alkyl, and phenyl, and
  • R 11 is independently selected from the group consisting of C 1 -C 4 alkyl, halogen, acetyl, methoxy, and ethoxy.
  • composition of any one of embodiments 27 to 30, wherein the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP-43742, and a combination thereof.
  • composition of embodiment 34 wherein the effective amount of the RORy inhibitor and the statin is sufficient to enhance an anticancer drug therapeutic effect and/or produce a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof.
  • composition of embodiment 36, wherein enhancing the anticancer drug therapeutic effect comprises reversing or reducing cancer cell resistance to the anticancer drug and/or sensitizing cancer cells to the anticancer drug.
  • composition of embodiment 36 or 37, wherein the enhancement of the anticancer drug therapeutic effect and/or the beneficial effect that is produced is greater when the RORy inhibitor and the statin are administered in combination compared to when the RORy inhibitor or the statin are administered alone.
  • composition of embodiment 40 wherein the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
  • the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
  • composition of embodiment 41, wherein the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
  • composition of embodiment 41, wherein the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof.
  • composition of embodiment 43 wherein the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
  • kits for preventing or treating cancer in a subject comprising a RORy inhibitor and a statin.
  • kits of embodiment 46 wherein the RORy inhibitor is a small- molecule compound, an anti- RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
  • the RORy inhibitor is a small- molecule compound, an anti- RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
  • n is an integer selected from the group consisting of 0, 1, 2, or 3;
  • Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF 3 )2, heterocyclyl, and cycloalkyl,
  • R5 is selected from the group consisting of H, and C1-C3 alkyl group
  • R 2 is selected from the group consisting of H, halogen, and alkyl
  • R 3 is selected from the group consisting of H and alkyl
  • R 4 is selected from the group consisting of C0-C4 alkylene-R 6 , C0-C4 alkylene- Rvcycloalkyl, and C 0 -C 4 alkylene-Rvheterocyclyl,
  • R 6 is selected from the group consisting of -Rx, -ORx, -CORx, -COORx, -S(0) m R 8 , cycloalkyl, and heterocyclyl, m is 0 or 2
  • R 7 is selected from the group consisting of -OR9, -C(0)R9, -NR9, -SR9, -S(0)R9, -S(0) 2 R9,
  • Rx is selected from the group consisting of H, and Ci-C 3 alkyl group, and R 9 is Ci-C 3 alkyl ene;
  • each cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, carboxy, amino, -CONFh, -COORio, -CORio, -ORio, -NHCORio, -NHCOORio, and -COH(CF 3 ) 2 ,
  • each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono- , bi- or tri-cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, Ci-C 4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -CONFh, -COORio, -CORio, -ORio, -NHCORio, -NHCOORio, -COH(CF 3 ) 2 , -C6H5R11, morpholinyl, piperidinyl, tetrahydrofuranyl, substituted pyridyl group,
  • Rio is independently selected from the group consisting of H, C 1 -C 4 alkyl, and phenyl, and
  • R 11 is independently selected from the group consisting of C 1 -C 4 alkyl, halogen, acetyl, methoxy, and ethoxy.
  • kits of any one of embodiments 46 to 48, wherein the RORy inhibitor selectively binds to RORy and inhibits RORy activity are provided.
  • kit of any one of embodiments 46 to 49, wherein the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP- 43742, and a combination thereof.
  • statin is selected from the group consisting of atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, pitavastatin, cerivastatin, pravastatin, and a combination thereof.
  • the kit comprises an effective amount of the RORy inhibitor that is sufficient to sensitize a cancer to the statin.
  • kits of embodiment 53 wherein the effective amount of the RORy inhibitor and the statin is sufficient to enhance an anticancer drug therapeutic effect and/or produce a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof.
  • kits of embodiment 55, wherein enhancing the anticancer drug therapeutic effect comprises reversing or reducing cancer cell resistance to the anticancer drug and/or sensitizing cancer cells to the anticancer drug.
  • kit of embodiment 55 or 56, wherein the enhancement of the anticancer drug therapeutic effect and/or the beneficial effect that is produced is greater when the RORy inhibitor and the statin are administered in combination compared to when the RORy inhibitor or the statin are administered alone.
  • kit of any one of embodiments 46 to 58, further comprising an anticancer drug further comprising an anticancer drug.
  • kits of embodiment 59 wherein the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
  • the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
  • kits of embodiment 60 wherein the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
  • kits of embodiment 60 wherein the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof.
  • the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
  • kit of any one of embodiments 46 to 63 further comprising a label with instructions for administering the RORy inhibitor and the statin to the subject.
  • a method for preventing or treating a cancer in a subject comprising administering to the subject an effective amount of a RORy inhibitor, wherein the RORy inhibitor is a compound listed in Table 5, a pharmaceutically acceptable salt thereof, an isomer thereof, a racemate thereof, a prodrug thereof, a co-crystalline complex thereof, a hydrate thereof, or a solvate thereof.
  • the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
  • anti -androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
  • chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof.
  • 73 The method of embodiment 72, wherein the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof. 74. The method of any one of embodiments 66 to 73, wherein the cancer is selected from the group consisting of breast cancer, prostate cancer, lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, and glioma.
  • TNBC triple- negative breast cancer
  • tamoxifen-resistant breast cancer tamoxifen-resistant breast cancer
  • radiation-resistant breast cancer HER2-positive breast cancer
  • ER-positive breast cancer ER-positive breast cancer
  • lung cancer is a non small-cell lung cancer (NSCLC), K-Ras mutant lung cancer, BRAF mutant lung cancer, EGFR mutant lung cancer, tyrosine kinase inhibitor-resistant lung cancer, or small cell lung cancer (SCLC).
  • NSCLC non small-cell lung cancer
  • K-Ras mutant lung cancer K-Ras mutant lung cancer
  • BRAF mutant lung cancer BRAF mutant lung cancer
  • EGFR mutant lung cancer tyrosine kinase inhibitor-resistant lung cancer
  • SCLC small cell lung cancer

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Emergency Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides methods for treating cancer, in which the methods comprise administering a combination of a RORγinhibitor and a statin to a subject. In some embodiments, administering the combination of the RORγinhibitor and the statin enhances the effects of an anti cancer drug such as an anti-androgen drug or a chemotherapeutic agent. In some embodiments, administering the combination of the RORγ inhibitor and the statin reverses or reduces resistance of a cancer to the anti cancer drug. In some embodiments, the cancer is sensitized to the statin.

Description

METHODS FOR TREATING CANCER WITH RORgamma
INHIBITORS AND STATINS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to U.S. Provisional Application No. 62/726,087, filed August 31, 2018, the disclosure of which is herein incorporated by reference in its entirety for all purposes.
STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT
[0002] This invention was made with government support under Grant No. R01CA206222, awarded by the National Institutes of Health. The government has certain rights in the invention.
BACKGROUND OF THE INVENTION
[0003] Cancer is a leading cause of death in the United States, and despite the development of various different treatment methods such as chemotherapy, radiation therapy, and hormone deprivation therapy, there is no 100% effective cure to these diseases. In 2012, there were approximately 14 million new cases of cancer, and approximately 8.2 deaths caused by cancer worldwide. It is expected that the number of new cases of cancer will increase from approximately 14 million in 2012 to approximately 30 million by the year 2030.
[0004] As such, there is currently a need in the art for new methods and compositions for treating cancer patients. The present invention addresses this need and provides related advantages as well.
BRIEF SUMMARY OF THE INVENTION
[0005] In a first aspect, the present invention provides a method for preventing or treating a cancer in a subject, the method comprising administering to the subject an effective amount of a RORy inhibitor and a statin. In some embodiments, the RORy inhibitor is a small- molecule compound, an anti-RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
[0006] In some embodiments, the RORy inhibitor is selected from the group consisting of a compound according to Formula I:
Figure imgf000003_0001
GSK805, SR2211, a compound listed in Table 5, pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and solvates thereof, wherein X is C(=0) or S02; n is an integer selected from the group consisting of 0, 1, 2, or 3; Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF3)2, heterocyclyl, and cycloalkyl, wherein R5 is selected from the group consisting of H, and C1-C3 alkyl group; R2 is selected from the group consisting of H, halogen, and alkyl; R3 is selected from the group consisting of H and alkyl; R4 is selected from the group consisting of C0-C4 alkylene-R6, Co- C4 alkylene-Rvcycloalkyl, and C0-C4 alkylene-Rvheterocyclyl, wherein R6 is selected from the group consisting of -Rx, -ORx, -CORx, -COORs, -S(0)mR8, cycloalkyl, and heterocyclyl, m is 0 or 2, and R7 is selected from the group consisting of -OR9, -C(0)R9, -NR9, -SR9, - S(0)R9, -S(0)2R9, wherein Rx is selected from the group consisting of H, and Ci-C3 alkyl group, and R9 is Ci-C3 alkylene; wherein each cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, carboxy, amino, -CONFh, -COOR10, - COR10, -OR10, -NHCOR10, -NHCOOR10, and -COH(CF3)2, each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono-, bi- or tri-cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, C1-C4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -CONFh, - COOR10, -COR10, -OR10, -NHCOR10, -NHCOOR10, -COH(CF3)2, -C6H5RII, morpholinyl, piperidinyl, tetrahydrofuranyl, substituted pyridyl group, wherein Rio is independently selected from the group consisting of H, C1-C4 alkyl, and phenyl, and Rn is independently selected from the group consisting of C1-C4 alkyl, halogen, acetyl, methoxy, and ethoxy. [0007] In some embodiments, the RORy inhibitor selectively binds to RORy and inhibits RORy activity. In some embodiments, the compound of Formula I is represented by a compound according to any one of Formulas Ic to Ii:
Figure imgf000004_0001
Figure imgf000004_0002
t thereof, a derivative thereof, an analog thereof, or a combination thereof.
[0008] In some embodiments, the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP-43742, and a combination thereof. In some embodiments, the statin is selected form the group consisting of atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, pitavastatin, cerivastatin, pravastatin, and a combination thereof.
[0009] In some embodiments, the cancer is resistant to an anticancer drug. In some embodiments, the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof. In some embodiments, the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof. In some embodiments, the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof. In some embodiments, the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
[0010] In some embodiments, the cancer is selected from the group consisting of breast cancer, prostate cancer, lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, and glioma. In some embodiments, the breast cancer is a triple-negative breast cancer (TNBC), tamoxifen-resistant breast cancer, radiation-resistant breast cancer, HER2-positive breast cancer, or ER-positive breast cancer.In some embodiments, the prostate cancer is a castration-resistant prostate cancer. In some embodiments, the lung cancer is a non-small-cell lung cancer (NSCLC), K-Ras mutant lung cancer, BRAF mutant lung cancer, EGFR mutant lung cancer, tyrosine kinase inhibitor- resistant lung cancer, or small cell lung cancer (SCLC).
[0011] In some embodiments, the method further comprises administering the anti-cancer drug to the subject. In some embodiments, the subject is a human in need of cancer treatment. In some embodiments, administering the RORy inhibitor and the statin enhances the therapeutic effect of the anticancer drug. In some embodiments, administering the RORy inhibitor and the statin reverses or reduces cancer cell resistance to the anti cancer drug and/or sensitizes cancer cells to the anticancer drug. In some embodiments, administering the RORy inhibitor and the statin produces a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof. In some embodiments, enhancement of the anticancer drug therapeutic effect and/or the beneficial effect that is produced are greater when the RORy inhibitor and the statin are administered in combination compared to when the RORy inhibitor or the statin are administered alone. In some embodiments, administering the RORy inhibitor and the statin in combination produces a synergistic enhancement of the anticancer drug and/or produces a synergistic beneficial effect. In some embodiments, the cancer is sensitized to the statin. [0012] In some embodiments, the RORy inhibitor and the statin are administered concomitantly. In some embodiments, the RORy inhibitor and the statin are administered sequentially.
[0013] In another aspect, the present invention provides a composition comprising a RORy inhibitor and a statin. In some embodiments, the RORy inhibitor is a small-molecule compound, an anti-RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
[0014] In some embodiments, the RORy inhibitor is selected from the group consisting of a compound according to Formula I:
Figure imgf000006_0001
GSK805, SR2211, a compound listed in Table 5, pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and solvates thereof, wherein X is C(=0) or S02; n is an integer selected from the group consisting of 0, 1, 2, or 3; Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF3)2, heterocyclyl, and cycloalkyl, wherein R5 is selected from the group consisting of H, and C1-C3 alkyl group; R2 is selected from the group consisting of H, halogen, and alkyl; R3 is selected from the group consisting of H and alkyl; R4 is selected from the group consisting of C0-C4 alkylene-R6, Co- C4 alkylene-Rvcycloalkyl, and C0-C4 alkylene-Rvheterocyclyl, wherein R6 is selected from the group consisting of -Rx, -ORx, -CORx, -COORs, -S(0)mR8, cycloalkyl, and heterocyclyl, m is 0 or 2, and R7 is selected from the group consisting of -OR9, -C(0)R9, -NR9, -SR9, - S(0)R9, -S(0)2R9, wherein Rx is selected from the group consisting of H, and Ci-C3 alkyl group, and R9 is Ci-C3 alkylene; wherein each cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, carboxy, amino, -CONFh, -COOR10, - COR10, -OR10, -NHCOR10, -NHCOOR10, and -COH(CF3)2, each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono-, bi- or tri-cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, C1-C4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -CONH2, - COOR10, -COR10, -OR10, -NHCOR10, -NHCOOR10, -COH(CF3)2, -C6H5RII, morpholinyl, piperidinyl, tetrahydrofuranyl, substituted pyridyl group, wherein Rio is independently selected from the group consisting of H, C1-C4 alkyl, and phenyl, and Rn is independently selected from the group consisting of C1-C4 alkyl, halogen, acetyl, methoxy, and ethoxy.
[0015] In some embodiments, the RORy inhibitor selectively binds to RORy and inhibits RORy activity. In some embodiments, the compound of Formula I is represented by a compound according to any one of Formulas Ic to Ii:
Figure imgf000007_0001
Figure imgf000007_0002
pharmaceutically acceptable salt thereof, a derivative thereof, an analog thereof, or a combination thereof.
[0016] In some embodiments, the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP-43742, and a combination thereof. In some embodiments, the statin is selected form the group consisting of atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, pitavastatin, cerivastatin, pravastatin, and a combination thereof.
[0017] In some embodiments, the composition comprises an effective amount of the RORy inhibitor and the statin. In some embodiments, the composition comprises an effective amount of the RORy inhibitor that is sufficient to sensitize a cancer to the statin. In some embodiments, the effective amount of the RORy inhibitor and the statin is sufficient to enhance an anticancer drug therapeutic effect and/or produce a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof. In some embodiments, enhancing the anticancer drug therapeutic effect comprises reversing or reducing cancer cell resistance to the anticancer drug and/or sensitizing cancer cells to the anticancer drug. In some embodiments, the enhancement of the anticancer drug therapeutic effect and/or the beneficial effect that is produced is greater when the RORy inhibitor and the statin are administered in combination compared to when the RORy inhibitor or the statin are administered alone. In some embodiments, the anticancer drug therapeutic effect is synergistically enhanced and/or the beneficial effect is synergistically produced when the RORy inhibitor and the statin are administered in combination.
[0018] In some embodiments, the composition further comprises an anticancer drug. In some embodiments, the anticancer drug is selected from the group consisting of an anti androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof. In some embodiments, the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof. In some embodiments, the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof. In some embodiments, the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
[0019] In some embodiments, the composition further comprises a pharmaceutically acceptable excipient or diluent. [0020] In another aspect, the present invention provides a kit comprising a RORy inhibitor and a statin. In some embodiments, the RORy inhibitor is a small-molecule compound, an anti-RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
[0021] In some embodiments, the RORy inhibitor is selected from the group consisting of a compound according to Formula I:
Figure imgf000009_0001
GSK805, SR2211, a compound listed in Table 5, pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and solvates thereof, wherein X is C(=0) or S02; n is an integer selected from the group consisting of 0, 1, 2, or 3; Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF3)2, heterocyclyl, and cycloalkyl, wherein R5 is selected from the group consisting of H, and C1-C3 alkyl group; R2 is selected from the group consisting of H, halogen, and alkyl; R3 is selected from the group consisting of H and alkyl; R4 is selected from the group consisting of C0-C4 alkylene-R6, Co- C4 alkylene-Rvcycloalkyl, and C0-C4 alkylene-Rvheterocyclyl, wherein R6 is selected from the group consisting of -Rx, -ORx, -CORx, -COORs, -S(0)mR8, cycloalkyl, and heterocyclyl, m is 0 or 2, and R7 is selected from the group consisting of -OR9, -C(0)R9, -NR9, -SR9, - S(0)R9, -S(0)2R9, wherein Rx is selected from the group consisting of H, and Ci-C3 alkyl group, and R9 is Ci-C3 alkylene; wherein each cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, carboxy, amino, -CONFh, -COOR10, - COR10, -OR10, -NHCOR10, -NHCOOR10, and -COH(CF3)2, each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono-, bi- or tri-cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, C1-C4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -CONFh, - COOR10, -COR10, -OR10, -NHCOR10, -NHCOOR10, -COH(CF3)2, -C6H5RII, morpholinyl, piperidinyl, tetrahydrofuranyl, substituted pyridyl group, wherein Rio is independently selected from the group consisting of H, C1-C4 alkyl, and phenyl, and Rn is independently selected from the group consisting of C1-C4 alkyl, halogen, acetyl, methoxy, and ethoxy.
[0022] In some embodiments, the RORy inhibitor selectively binds to RORy and inhibits RORy activity. In some embodiments, the compound of Formula I is represented by a compound according to any one of Formulas Ic to Ii:
Figure imgf000010_0001
Figure imgf000010_0002
t thereof, a derivative thereof, an analog thereof, or a combination thereof.
[0023] In some embodiments, the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP-43742, and a combination thereof. In some embodiments, the statin is selected form the group consisting of atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, pitavastatin, cerivastatin, pravastatin, and a combination thereof.
[0024] In some embodiments, the kit comprises an effective amount of the RORy inhibitor and the statin. In some embodiments, the kit comprises an effective amount of the RORy inhibitor that is sufficient to sensitize the cancer to the statin. In some embodiments, the effective amount of the RORy inhibitor and the statin is sufficient to enhance an anticancer drug therapeutic effect and/or produce a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof. In some embodiments, enhancing the anticancer drug therapeutic effect comprises reversing or reducing cancer cell resistance to the anticancer drug and/or sensitizing cancer cells to the anticancer drug. In some embodiments, the enhancement of the anticancer drug therapeutic effect and/or the beneficial effect that is produced is greater when the RORy inhibitor and the statin are administered in combination compared to when the RORy inhibitor or the statin are administered alone. In some embodiments, the anticancer drug therapeutic effect is synergistically enhanced and/or the beneficial effect is synergistically produced when the RORy inhibitor and the statin are administered in combination.
[0025] In some embodiments, the kit further comprises an anticancer drug. In some embodiments, the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof. In some embodiments, the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof. In some embodiments, the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof. In some embodiments, the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
[0026] In some embodiments, the kit further comprises a label with instructions for administering the RORy inhibitor and the statin to the subject. In some embodiments, the subject is a human in need of cancer treatment.
[0027] In another aspect the present invention provides a method for preventing or treating a cancer in a subject, the method comprising administering to the subject an effective amount of a RORy inhibitor, wherein the RORy inhibitor is a compound listed in Table 5, a pharmaceutically acceptable salt thereof, an isomer thereof, a racemate thereof, a prodrug thereof, a co-crystalline complex thereof, a hydrate thereof, or a solvate thereof. In some embodiments, the RORy inhibitor selectively binds to RORy and inhibits RORy activity. In some embodiments, the RORy inhibitor is VTP-43742.
[0028] In some embodiments, the cancer is resistant to an anticancer drug. In some embodiments, the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof. In some embodiments, the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof. In some embodiments, the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof. In some embodiments, taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
[0029] In some embodiments, the cancer is selected from the group consisting of prostate cancer, lung cancer, breast cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, and glioma. In some embodiments, the prostate cancer is a castration-resistant prostate cancer. In some embodiments, the lung cancer is a non-small-cell lung cancer (NSCLC), K-Ras mutant lung cancer, BRAF mutant lung cancer, EGFR mutant lung cancer, tyrosine kinase inhibitor-resistant lung cancer, or small cell lung cancer (SCLC). In some embodiments, the breast cancer is a triple-negative breast cancer (TNBC), tamoxifen-resistant breast cancer, radiation-resistant breast cancer, HER2-positive breast cancer, or ER-positive breast cancer.
[0030] In some embodiments, the method further comprises administering the anti-cancer drug to the subject. In some embodiments, the subject is a human in need of cancer treatment. In some embodiments, administering the RORy inhibitor enhances the therapeutic effect of the anticancer drug. In some embodiments, administering the RORy inhibitor reverses or reduces cancer cell resistance to the anticancer drug and/or sensitizes cancer cells to the anticancer drug. In some embodiments, administering the RORy inhibitor produces a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof. [0031] Other objects, features, and advantages of the present invention will be apparent to one of skill in the art from the following detailed description and figures.
BRIEF DESCRIPTION OF THE DRAWINGS
[0032] FIGS. 1A-1D show that RORy is required for cancer cell growth and survival of breast cancer. FIG. 1A: MDA-MB468 and HCC70 TNBC cells were infected with lentiviruses expressing control sgRNA against GFP or two sgRNAs against RORC, and Cas9. Three and six days later, viable cell numbers were counted. Data are shown as mean ± s. d. n = 3. FIG. 1B: The TNBC cells were infected as in FIG. 1A. Three days later, cells were harvested for Western blotting analysis of indicated proteins with specific antibodies. FIG. 1C: The TNBC cells were transfected with control siRNA (siNeal) or two siRNAs against RORC. Two and four days later, viable cell numbers were counted. Data are shown as mean ± s. d. n = 3. FIG. 1D: The HCC70 TNBC cells were transfected as in FIG. 1C. Three days later, cells were harvested for Western blotting analysis of indicated proteins with specific antibodies.
[0033] FIG. 2 shows that RORy is required for growth and survival of multiple different TNBC cell lines but not for non-malignant MCF10A cells. SUM159, MB436, Hs578T, and BT549 TNBC cells or MCF10A cells were infected with lentiviruses expressing control sgRNA against GFP or two sgRNAs against RORC , and Cas9. Three and six days later, viable cell numbers were counted. Data are shown as mean ± s. d. n = 3.
[0034] FIG. 3 shows that RORy is required for growth and survival of mutant KRAS lung cancer cells. A549 and H460 lung cancer cells that express mutant KRAS were transfected with control siRNA (siCont) or two siRNAs against RORC. Three and six days later, viable cell numbers were counted. Data are shown as mean ± s. d. n = 3.
[0035] FIGS. 4A-4C show that RORy inhibition suppresses the expression of genes controlling cholesterol synthesis. FIG. 4A: An abbreviated pathway of cholesterol synthesis showing statin and azole inhibitors and their corresponding target enzymes. FIG. 4B: Heat map display of fold changes (in log2) in gene expression of cholesterol biosynthesis pathway between cells treated by the indicated inhibitors and cells treated with vehicle control, as detected by RNA-seq analysis of HCC70 TNBC cells treated by vehicle, 2.5 mM RORy inhibitor XY018, or GSK805 for 48 hours. FIG. 4C: Western blotting analysis of proteins in cholesterol synthesis pathway with MDA-MB468 cells treated by indicated concentrations of RORy inhibitor XY018 and XY063. [0036] FIG. 5 shows that RORy inhibition abolishes statin treatment-induced up-regulation of cholesterol synthesis and homeostasis genes. Heat map display of fold changes (in log2) in gene expression of cholesterol synthesis and homeostasis pathways between cells treated by the indicated inhibitors and vehicle control, as detected by RNA-seq of HCC70 TNBC cells treated with either vehicle (DMSO), RORy inhibitor XY018 (1.25 or 2.5 mM), XY063 (1.25 or 2.5 pM), atorvastatin (1.25 or 2.5 pM) alone, or a combination of the RORy inhibitor XY018 with atorvastatin.
[0037] FIGS. 6A-6D show that RORy inhibitors in combination with statins display strong synergy in the inhibition of TNBC cancer cell growth. Breast cancer cells HCC70 (FIG. 6A), MDA-MB468 cells (FIGS. 6B and 6D), and SUM159 cells (FIG. 6C) were treated by indicated concentrations of simvastatin or atorvastatin alone, or in combination with indicated concentrations of RORy inhibitor XY018 or XY063 for 4 days. The cell viability was measured by CellTiter-Glo, with viability of cells treated with 0 pM of statin and RORy inhibitor set at 100. Data are shown as mean ± s. d. n = 3. The experiments were repeated three times. ** P < 0.01; Student’s t test.
[0038] FIGS. 7A-7D show that RORy inhibitors in combination with statins display strong synergy in the killing of TNBC cancer cells. Representative images of colony formation of TNBC HCC70 cells (FIG. 7A) and MDA-MB468 cells (FIG. 7C) treated with indicated concentrations of simvastatin (Simva) alone, or in combination with XY018 or XY063 for 14 days, after which colonies formed were enumerated. Colonies formed by HCC70 cells (FIG. 7B) and MDA-MB468 cells (FIG. 7D) after the 14 days of treatment were plotted for each of the treatments. Student’s t test, n = 3. * P < 0.001 (compared to siControl); Student’s t test.
[0039] FIGS. 8A and 8B show that RORy inhibitors in combination with statins display strong synergy in the inhibition of lung cancer cell growth. A549 lung cancer cells were treated by indicated concentrations of fluvastatin (FIG. 8A) or lovastatin (FIG. 8B), either alone or in combination with indicated concentrations of RORy inhibitor XY018 or XY063 for 4 days. The cell viability was measured by CellTiter-Glo, with viability of cells treated with 0 pM of statin and RORy inhibitor set at 100. Data are shown as mean ± s. d. n = 3. The experiments were repeated three times.
[0040] FIGS. 9A-9D show that RORy inhibitors in combination with statins display strong synergy in the inhibition of CRPC prostate cancer cell growth. CRPC prostate cancer cells 22Rvl and C4-2B cells were treated by indicated concentrations of atorvastatin (FIGS. 9B and 9D, respectively) or simvastatin (FIGS. 9 A and 9C, respectively), either alone or in combination with indicated concentrations of RORy inhibitor XY018 or XY063 for 4 days. The cell viability was measured by CellTiter-Glo, with viability of cells treated with 0 mM of statin and RORy inhibitor set at 100. Data are shown as mean ± s. d. n = 3. The experiments were repeated three times. ** P < 0.001; Student’s t test.
[0041] FIGS. 10A-10I show that RORy inhibitors in combination with statins display strong synergy in the inhibition of gastric cancer cell growth. FIGS. 10 A, 10B, and 10C show cell viability assays of AGS, HGC-27, and MGC-803 gastric cancer cells, respectively, that were treated by indicated concentrations of RORy inhibitors GSK805, XY018, or SR2211 for 4 days. The cell viability was measured by CellTiter-Glo. FIGS. 10D-10I show cell viability assays of gastric cancer cells that were treated by atorvastatin (AGS, HGC-27, and MGC-803 cells shown in FIGS. 10D, 10E, and 10F, respectively) or simvastatin (AGS, HGC-27, and MGC-803 cells shown in FIGS. 10G, 10H, and 101, respectively), either alone or in combination with indicated concentrations of RORy inhibitor XY018 for 4 days. The cell viability was measured by CellTiter-Glo, with viability of cells treated with 0 pM of statin and RORy inhibitor set at 100. Data are shown as mean ± s. d. n = 3. The experiments were repeated three times.
[0042] FIGS. 11A-11D show that RORy inhibitor VTP -43742 in combination with statins displays strong synergy in the inhibition of breast cancer and prostate cancer cell growth. FIGS. 11A and 11B: cells of the HCC70 triple-negative breast cancer (TNBC) cell model were treated by the indicated statin (simvastatin (FIG. 11 A) or atorvastatin (FIG. 11B)) at increasing concentrations (from 0.625 pM to 10 pM) either alone (plus vehicle control) or with 2.5 pM RORy inhibitor/antagonist VTP -43742. Cells were treated for 4 days. FIGS. 11C and 11D: cells of the C4-2B castration-resistant prostate cancer (CRPC) cell model were treated by the indicated statin (simvastatin (FIG. 11C) or atorvastatin (FIG. 11D)) at increasing concentrations (from 0.625 pM to 10 pM) either alone (plus vehicle control) or with 2.5 pM RORy inhibitor/antagonist VTP-43742. Cells were treated for 4 days. For data shown in all panels, cell viability was measured by CellTiter-GLO. The cell viability obtained from cells treated with vehicle only ( i.e without either the statin or the ROR-y inhibitor/antagonist) was set as 100. The experiments were repeated three times and in sexplicate. Data are shown as mean ± s.d. * P < 0.01; Student’s / test. [0043] FIGS. 12A and 12B show that inhibition of RORy with small-interfering RNA (siRNA) strongly sensitizes prostate cancer and breast cancer cells to killing by statins. Cells of the MDA-MB468 triple-negative breast cancer (TNBC) cell model or the C4-2B castration-resistant prostate cancer (CRPC) cell model were first transfected with 5 nM of control siRNA or two RORC gene siRNAs (si-RORC-l and siRORC-2) as previously reported in J. Wang et al, Nature Medicine , 22, 488-496 (2016). Cells were then seeded in 6-well plates, cultured and treated by indicated concentrations of atorvastatin for 12 days. Cells were then fixed with 10% formalin and cell colonies were stained with 0.2% crystal violet. The numbers of colonies were then counted. The experiments were performed in triplicates and repeated three times. Data shown are as mean ± s.d. * P < 0.01 (compared to siControl); Student’s / test.
[0044] FIGS. 13A-13D show that oral administration of RORy inhibitors in combination with statins suppresses breast cancer tumor growth more effectively than administration of either alone. Mammary gland-orthotopic xenograft tumors were established by implanting breast cancer MDA-MB468 cells into mammary fat pads of SCID mice. Mice were treated 5 times per week with vehicle, RORy inhibitors alone (XY018 10 mg/kg p.o. or XY063 10 mg/kg p.o.), atorvastatin alone (20 mg/kg p.o.), or a combination of RORy inhibitors with atorvastatin (XY018 10 mg/kg p.o. + Atorva. 20 mg/kg p.o. or XY063 10 mg/kg p.o. + Atorva. 20 mg/kg p.o.; n=7 mice per group) for 56 days. FIG. 13 A shows tumor volume. FIG. 13B shows tumor weight. FIG. 13C shows representative images or tumors. FIG. 13D shows mouse body weight. Significance was calculated using Student’s / test. * P < 0.001, ** P < 0.0005, *** P < 0.0001.
[0045] FIGS. 14A-14D show that RORy inhibitors in combination with statins suppress lung tumor growth more effectively than administration of either alone. A549 lung xenograft tumors were established by subcutaneous (s.c.) injection of A549 cells into NOD-SCID female mice. When tumors reached approximately 100 mm3, mice were randomized into groups for treatment by either vehicle, intraperitoneal (i.p.) injection of XY018 (5 mg/kg, five times per week), oral gavage of atorvastatin (20 mg/kg, five times per week), or a combination of XY018 and atorvastatin (five times per week). FIG. 14A shows tumor volume. FIG. 14B shows tumor weight. FIG. 14C shows representative images or tumors. FIG. 14D shows mouse body weight.
[0046] FIG. 15 shows the structures of various statins. [0047] FIGS. 16A-16C show that inhibition of RORy with antagonists XY018 and XY063 strongly sensitizes lung cancer cells to killing by different statins. FIG. 16A shows the combination index (Cl) and supporting data. FIG. 16B shows viability of A549 cells using fluvastatin and XY018 or XY063. FIG. 16C shows viability of A549 cells using lovastatin and XY018 or XY063. Data shown are mean ± s.d. ** P < 0.002; Student’s t test.
[0048] FIGS. 17A-17C show that combined treatment with RORy inhibitors/antagonists and statins is synergistic in the inhibition of lung cancer tumor growth. FIG. 17A shows tumor size data. FIG. 17B shows tumor weight data. FIG. 17C shows images of representative tumors. Significance was calculated using Student’s t test. * P < 0.05, *** P < 0 01
[0049] FIGS. 18A and 18B show that a combined treatment of cancer cells with RORy inhibitor and statin synergistically inhibits the oncogenic kinase signaling by receptor tyrosine kinase (RTK) and serine/threonine kinase. FIG. 18A shows data from PC9 cells. FIG. 18B shows data from A549 cells.
[0050] FIG. 19 shows biochemical analysis of serum samples for the assessment of significant alteration of key blood/serum biochemical parameters or toxicity indicators.
[0051] FIGS. 20A-20K shows that RORy inhibitors alone or synergize with statins strongly inhibit TNBC tumor growth in vivo. FIG. 20A: Mice bearing MDA-MB468 were treated, p.o., 5 times per week, with vehicle or indicated doses of RORy inhibitor XY018 for 52 days. Tumor volume (top), representative images (middle) and tumor weight (bottom) are shown n = 7 mice per group mpk, mg per kg. FIG. 20B: Mice bearing HCC70 were treated, i.p., 5 times per week, with vehicle or 5 mg/kg XY018 or GSK805 for 43 days. Tumor volume, representative images (left) and tumor weight (right) are shown n = 7. FIG. 20C: Mice bearing SUM159 were treated, i.p., 5 times per week, with vehicle or 5 mg/kg XY018 or GSK805 for 45 days. Tumor volume, representative images (left) and tumor weight (right) are shown n = 7. FIG. 20D: Body weight of mice bearing PDX-1079 were treated, p.o., 5 times per week, with vehicle or 50 mg/kg RORy inhibitor XY018 for 57 days n = 7. FIG. 20E: Mice bearing MCF-7 were treated, i.p., 5 times per week, with vehicle or 10 mg/kg XY018 for 42 days. Tumor volume, representative images, tumor weight and body weight are shown n = 7. FIG. 20F: Mice bearing 4T1 were treated, i.p., 5 times per week, with vehicle or 20 mg/kg XY018 for 56 days. Tumor volume and body weight are shown n = 10. FIG. 20G: Bioluminescence monitoring of MDA-MB231-4175 tumor metastasis in the lung. Nude mice were injected MDA-MB231-4175 cells carrying a luciferase gene via tail vein and treated daily with vehicle or 20 mg/kg XY018 for 4 weeks n = 6. FIGS. 20H, 201: Body weight (left) and representative tumor images (right) of mice bearing PDX-1079 or PDX- 1173 treated, 5 times per week, p.o., with vehicle, 20 mg/kg XY018 alone, 15 mg/kg ATV alone, or both XY018 + ATV for indicated days n = 7 mice per group. FIG. 20J: PDX-1079 were treated, 5 times per week, p.o., with vehicle, 20 mg/kg XY018 alone, l5mg/kg simvastatin alone (SIM) or both of XY018 + SIM for 47 days. Tumor weight is shown n = 7. FIG. 20K: Mice bearing MDA-MB468 were treated, 5 times per week, p.o., with vehicle, 20 mg/kg XY018 alone, l5mg/kg ATV alone or both XY018 + ATV for 32 days. Tumor weight is shown. n=7. Throughout, data are shown as mean ± s.e.m. Student’s t-test. * p < 0.05, ** p < 0.01, NS, not significant.
[0052] FIGS. 21A-21G show that RORy inhibitors alone or with statins cause tumor regression and block metastasis. FIG. 21A: Mice bearing PDX-1079 were treated, p.o., 5 times per week, with vehicle or 50 mg/kg XY018 for 57 days. Tumor volume and representative images are shown n = 7 mice per group. Student’s t-test. ** p < 0.0l. mpk, mg/kg. FIG. 21B: K-M survival plot of 4T1 tumor-bearing mice treated with vehicle or XY018 (20 mg/kg, i.p.) for 56 days, n = 10. FIG. 21C: Lung tumor nodules metastasized from primary sites were analyzed for 4T1 tumor-carrying mice treated with XY018 (20 mg/kg, i.p.) or vehicle daily for 4 weeks. Representative lung section images were taken with arrows indicating tumor nodules n = 6. Student’s t-test. ** p < 0.01. FIGS. 21D, 21E: PDX-1079 and PDX-1173 were treated, 5 times per week, p.o., with vehicle, 20 mg/kg XY018 alone, l5mg/kg ATV alone, or both XY018 + ATV for indicated days. Tumor volume (left) and tumor weight (right) are shown n = 7. Student’ s t-test. ** p < 0.0l. FIG. 21F: PDX-1079 were treated, 5 times per week, p.o., with vehicle, 20 mg/kg XY018 alone, l5mg/kg simvastatin alone (SIM) or both of XY018 + SIM for 47 days. Tumor volume is shown n = 7. Student’s t-test. ** p < 0.01. FIG. 21 G: Mice bearing MDA-MB468 were treated, 5 times per week, p.o., with vehicle, 20 mg/kg XY018 alone, 15 mg/kg ATV alone or both XY018 + ATV for 32 days. Tumor volume is shown n = 7. Student’s t-test. ** p < 0 01 DETAILED DESCRIPTION OF THE INVENTION
I. INTRODUCTION
[0053] Metabolic reprogramming, a hallmark of cancer, fuels malignant tumor growth and survival by providing energy, reducing power and building blocks. One metabolic process that is often deregulated in tumors of most cancer types is the mevalonate (MV A) pathway, which produces sterols such as cholesterol, isoprenoids, and ubiquinone that are essential for tumor growth (4). The major regulators of the MVA pathway enzyme expression are the transcription factors sterol regulatory element-binding protein 1 and 2 (SREBP1 and -2) and LXRs (2, 5). SREBPs play a pivotal role in maintenance of cholesterol homeostasis.
[0054] Statins are a class of cholesterol/lipid-lowering drugs used for treating patients with hypercholesterolemia to reduce the associated high risk of cardiovascular disease (CVD) (1). Statin treatment strongly reduces cholesterol production initially, and, as an adaptive response, the reduced cellular cholesterol level triggers the SREBP-mediated activation of gene expression including that of LDLR in liver and other tissues, which leads to an increased uptake of LDL from the circulation, hence lowering blood cholesterol level. Epidemiological studies comparing cancer patients receiving statins for their CVD risk to those not receiving statins suggest that the use of statins is associated with a statistically significant reduction of the risk of cancer relapse and cancer-related death.
[0055] The present invention is based, in part, on the discovery that RORy inhibitors effectively sensitize cancer cells to statin-induced cell death and growth inhibition, and that a combination treatment with RORy inhibitors and statins results in highly synergistic killing of cancer cells of multiple cancer types.
II. DEFINITIONS
[0056] It is noted here that as used in this specification and the appended claims, the singular forms“a,”“an,” and“the” include plural reference unless the context clearly dictates otherwise.
[0057] The terms“retinoic acid receptor-related orphan receptor y” and“RORy” refers to either or both isoforms encoded by the RORC (RAR-related orphan receptor C; see, e.g., NCBI Gene ID 6097 for the human gene) gene, namely RORy (also referred to as RORyl or RORC1; see, e.g, NCBI Reference Sequence NP 005051.2 for the human sequence) and RORyt (also known as RORy 2 or RORC2; see, e.g., NCBI Reference Sequence NP_001001523.1 for the human sequence).
[0058] The terms“subject”,“patient” or“individual” are used herein interchangeably to include a human or animal. For example, the animal subject may be a mammal, a primate (e.g, a monkey), a livestock animal (e.g, a horse, a cow, a sheep, a pig, or a goat), a companion animal (e.g, a dog, a cat), a laboratory test animal (e.g, a mouse, a rat, a guinea pig, a bird), an animal of veterinary significance, or an animal of economic significance.
[0059] As used herein, the term“effective amount” includes a dosage sufficient to produce a desired result with respect to the indicated disorder, condition, or mental state. The desired result may comprise a subjective or objective improvement in the recipient of the dosage. In one non-limiting example, an effective amount of a RORy inhibitor and a statin includes an amount sufficient to alleviate the signs, symptoms, or causes of a cancer, e.g., a therapeutically or prophylactically effective amount. Thus, an effective amount can be an amount that slows or reverses tumor growth (e.g, slows tumor size), increases mean time of survival, inhibits tumor progression or metastasis, or sensitizes a cancer cell to an anticancer drug to which it has become or is resistant. Also, in a second non-limiting example, an effective amount of a RORy inhibitor and a statin includes an amount sufficient to cause a substantial improvement in a subject having cancer when administered to the subject. The amount will vary with the type of cancer being treated, the stage of advancement of the cancer, the type and concentration of composition applied, and the amount of an anticancer drug (e.g, anti-androgen drug) that is also administered to the subject. In a third non-limiting example, an effective amount of a RORy inhibitor and a statin can include an amount that is effective in enhancing the therapeutic activity of anticancer drugs such as anti-androgen drugs (e.g, bicalutamide, enzalutamide, arbiraterone, etc.) and/or chemotherapeutic agents (e.g, tamoxifen and/or taxanes such as docetaxel), and/or sensitizing a cancer (e.g, a cancer cell) to an anticancer drug or other therapy. In some embodiments, an effective amount (e.g. of a RORy inhibitor) is an amount that is sufficient to sensitize a cancer to a statin.
[0060] The term“small-molecule compound” refers to a compound or drug that has a low molecular weight. The small-molecule compound may be a small organic molecule, either naturally occurring or synthetic. In some embodiments, the small-molecule compound has a molecular weight of more than about 50 Daltons and less than about 1,500 Daltons, e.g., between about 100 Daltons and about 1,000 Daltons or between about 200 Daltons and about 600 Daltons.
[0061] The term“interfering RNA” or“RNAi” or“interfering RNA sequence” as used herein includes single-stranded RNA ( e.g ., mature miRNA, ssRNAi oligonucleotides, ssDNAi oligonucleotides), double-stranded RNA (i.e., duplex RNA such as siRNA, Dicer- substrate dsRNA, shRNA, aiRNA, or pre-miRNA), a DNA-RNA hybrid (see, e.g., PCT Publication No. WO 2004/078941), or a DNA-DNA hybrid (see, e.g., PCT Publication No. WO 2004/104199) that is capable of reducing or inhibiting the expression of a target gene or sequence (e.g, by mediating the degradation or inhibiting the translation of mRNAs which are complementary to the interfering RNA sequence) when the interfering RNA is in the same cell as the target gene or sequence. Interfering RNA thus refers to the single-stranded RNA that is complementary to a target mRNA sequence or to the double-stranded RNA formed by two complementary strands or by a single, self-complementary strand. Interfering RNA may have substantial or complete identity to the target gene or sequence, or may comprise a region of mismatch (i.e., a mismatch motif). The sequence of the interfering RNA can correspond to the full-length target gene, or a subsequence thereof. In some embodiments, the interfering RNA molecules are chemically synthesized. The disclosures of each of the above patent documents are herein incorporated by reference in their entirety for all purposes.
[0062] Interfering RNA includes“small-interfering RNA” or“siRNA,” e.g., interfering RNA of about 15-60, 15-50, or 15-40 (duplex) nucleotides in length, more typically about 15- 30, 15-25, or 19-25 (duplex) nucleotides in length, and is preferably about 20-24, 21-22, or 21-23 (duplex) nucleotides in length (e.g, each complementary sequence of the double- stranded siRNA is 15-60, 15-50, 15-40, 15-30, 15-25, or 19-25 nucleotides in length, preferably about 20-24, 21-22, or 21-23 nucleotides in length, and the double-stranded siRNA is about 15-60, 15-50, 15-40, 15-30, 15-25, or 19-25 base pairs in length, preferably about 18-22, 19-20, or 19-21 base pairs in length). siRNA duplexes may comprise 3’ overhangs of about 1 to about 4 nucleotides or about 2 to about 3 nucleotides and 5’ phosphate termini. As used herein, the term“siRNA” includes RNA-RNA duplexes as well as DNA-RNA hybrids (see, e.g., PCT Publication No. WO 2004/078941).
[0063] siRNA can be chemically synthesized or can be generated by cleavage of longer dsRNA (e.g, dsRNA greater than about 25 nucleotides in length), e.g., with the E. coli RNase III or Dicer. These enzymes process the dsRNA into biologically active siRNA (see, e.g., Yang el al, Proc. Natl. Acad. Sci. USA, 99:9942-9947 (2002); Calegari el al, Proc. Natl. Acad. Sci. USA, 99: 14236 (2002); Byrom et al, Ambion TechNotes, l0(l):4-6 (2003); Kawasaki et al, Nucleic Acids Res., 31 :981-987 (2003); Knight et al, Science, 293:2269- 2271 (2001); and Robertson et al, J. Biol. Chem., 243:82 (1968)). Preferably, dsRNA are at least 50 nucleotides to about 100, 200, 300, 400, or 500 nucleotides in length. A dsRNA may be as long as 1000, 1500, 2000, 5000 nucleotides in length, or longer. The dsRNA can encode an entire gene transcript or a partial gene transcript.
[0064] As used herein, the term“mismatch motif’ or“mismatch region” refers to a portion of an interfering RNA sequence that does not have 100% complementarity to its target sequence. An interfering RNA may have at least one, two, three, four, five, six, or more mismatch regions. The mismatch regions may be contiguous or may be separated by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more nucleotides. The mismatch motifs or regions may comprise a single nucleotide or may comprise two, three, four, five, or more nucleotides.
[0065] As used herein, the term“treating” includes, but is not limited to, methods and manipulations to produce beneficial changes in a recipient's health status, e.g. , a patient’s cancer status. The changes can be either subjective or objective and can relate to features such as symptoms or signs of the cancer being treated. For example, if the patient notes decreased pain, then successful treatment of pain has occurred. For example, if a decrease in the amount of swelling has occurred, then a beneficial treatment of inflammation has occurred. Similarly, if the clinician notes objective changes, such as reducing the number of cancer cells, the growth of the cancer cells, the size of cancer tumors, or the resistance of the cancer cells to another cancer drug, then treatment of cancer has also been beneficial. Preventing the deterioration of a recipient’s status is also included by the term. Treating, as used herein, also includes administering combination of a RORy inhibitor and a statin alone with or without an anticancer drug to a subject having cancer. In certain instances, the cancer is breast cancer, prostate cancer, lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or glioblastoma multiforme.
[0066] As used herein, the term “administering” includes activities associated with providing a patient an amount of a compound described herein, e.g, a RORy inhibitor and a statin. Administering includes providing unit dosages of compositions set forth herein to a patient in need thereof. Administering includes providing effective amounts of compounds for a specified period of time, e.g, for about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or more days, or in a specified sequence, e.g. , administration of a RORy inhibitor and a statin followed by the administration of one or more anticancer drugs, or vice versa.
[0067] As used herein, the term“co-administering” includes sequential or simultaneous administration of two or more structurally different compounds. For example, two or more structurally different pharmaceutically active compounds can be co-administered by administering a pharmaceutical composition adapted for oral administration that contains two or more structurally different active pharmaceutically active compounds. As another example, two or more structurally different compounds can be co-administered by administering one compound and then administering the other compound. In some instances, the co-administered compounds are administered by the same route. In other instances, the co-administered compounds are administered via different routes. For example, one compound can be administered orally, and the other compound can be administered, e.g, sequentially or simultaneously, via intravenous or intraperitoneal injection.
[0068] As used herein, the term“cancer” refers to conditions including solid cancers, lymphomas, and leukemias. Examples of different types of cancers include, but are not limited to, prostate cancer, lung cancer (e.g, non-small cell lung cancer or NSCLC), ovarian cancer, colorectal cancer, liver cancer (i.e., hepatocarcinoma), renal cancer (i.e., renal cell carcinoma), bladder cancer, breast cancer, thyroid cancer, pleural cancer, pancreatic cancer, uterine cancer, cervical cancer, testicular cancer, anal cancer, bile duct cancer, gastrointestinal carcinoid tumors, esophageal cancer, gall bladder cancer, appendix cancer, small intestine cancer, stomach (gastric) cancer, cancer of the central nervous system, skin cancer, choriocarcinoma, head and neck cancer, blood cancer, endometrial cancer, osteogenic sarcoma, fibrosarcoma, neuroblastoma, glioma, melanoma, B-cell lymphoma, non-Hodgkin's lymphoma, Burkitfs lymphoma, Small Cell lymphoma, Large Cell lymphoma, monocytic leukemia, myelogenous leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, and multiple myeloma. In some instances, the cancer can be metastatic. In certain instances, the cancer is prostate cancer, lung cancer, breast cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma such as glioblastoma multiforme. In other instances, the cancer can be resistant to an anticancer drug, e.g, an anti-androgen-resistant cancer, a taxane-resistant cancer (e.g, docetaxel- resistant cancer), a tamoxifen-resistant cancer, a radiation-resistant cancer, or a tyrosine kinase inhibitor-resistant cancer.
[0069] As used herein, the terms“prostate cancer” or“prostate cancer cell” refer to a cancer cell or cells that reside in prostate tissue. The prostate cancer can be benign, malignant, or metastatic. The prostate cancer can be androgen-insensitive, hormone-resistant, or castrate-resistant. The prostate cancer can be“advanced stage prostate cancer” or “advanced prostate cancer.” Advanced stage prostate cancer includes a class of prostate cancers that has progressed beyond early stages of the disease. Typically, advanced stage prostate cancers are associated with a poor prognosis. Types of advanced stage prostate cancers include, but are not limited to, metastatic prostate cancer, drug-resistant prostate cancer such as anti -androgen -resistant prostate cancer ( e.g ., enzalutamide-resistant prostate cancer, abiraterone-resistant prostate cancer, bicalutamide-resistant prostate cancer, etc.), taxane-resistant prostate cancer (e.g., docetaxel-resistant prostate cancer) and the like, hormone refractory prostate cancer, castration-resistant prostate cancer (CRPC), metastatic castration-resistant prostate cancer, AR-V7-induced drug-resistant prostate cancer such as AR-V7-induced anti-androgen-resistant prostate cancer (e.g, AR-V7-induced enzalutamide- resistant prostate cancer), AKR1C3 -induced drug-resistant prostate cancer such as AKR1C3- induced anti -androgen-resistant prostate cancer (e.g, AKR1C3 -induced enzalutamide- resistant prostate cancer), and combinations thereof. In some instances, the advanced stage prostate cancers do not generally respond, or are resistant, to treatment with one or more of the following conventional prostate cancer therapies: enzalutamide, abiraterone, bicalutamide, and docetaxel. Compounds, compositions, and methods of the present invention are provided for treating prostate cancer, such as advanced stage prostate cancer, including any one or more (e.g, two, three, four, five, six, seven, eight, nine, ten, or more) of the types of advanced stage prostate cancers disclosed herein.
[0070] As used herein, the phrase“enhancing the therapeutic effects” includes any of a number of subjective or objective factors indicating a beneficial response or improvement of the condition being treated as discussed herein. For example, enhancing the therapeutic effects of an anticancer drug such as an anti -androgen drug (e.g, enzalutamide, abiraterone, or bicalutamide) or a chemotherapeutic agent such as tamoxifen or a taxane (e.g, docetaxel) includes reversing or reducing cancer cell resistance and/or sensitizing a drug-resistant cancer to the anticancer drug therapy. Also, for example, enhancing the therapeutic effects of an anticancer drug includes altering drug-resistant cancer cells so that the cells are not resistant to the anticancer drug. Also, for example, enhancing the therapeutic effects of an anticancer drug includes additively or synergistically improving or increasing the activity of the anticancer drug. In some embodiments, the enhancement includes a one-fold, two-fold, three-fold, five-fold, ten-fold, twenty-fold, fifty-fold, hundred-fold, or thousand-fold increase in the therapeutic activity of an anticancer drug used to treat cancer.
[0071] As used herein, the phrase“reversing cancer cell resistance” includes altering or modifying a cancer cell that is resistant to anticancer drug therapy so that the cell is no longer resitant to anticancer drug therapy.
[0072] As used herein, the phrase“reducing cancer cell resistance” includes increasing the therapeutic activity of an anticancer drug towards cancer cells that are, or previously were, resistant to anticancer drug therapy.
[0073] As used herein, the phrase“sensitizing cancer cell resistance” includes inducing sensitization towards anticancer drug therapy in cancer cells that are resistant to anticancer drug therapy. Sensitization as used herein includes inducing or enhancing the ability of a cancer cell to be effectively treated with an anticancer drug. Sensitization also includes reducing the dosage required to achieve a beneficial effect with an anticancer drug.
[0074] As used herein, the phrase“anti-androgen drug” includes anti-androgen compounds that alter the androgen pathway by blocking the androgen receptors, competing for binding sites on the cell’s surface, or affecting or mediating androgen production. Anti-androgen drugs are useful for treating several diseases including, but not limited to, prostate cancer. Anti-androgen drugs include, but are not limited to, enzalutamide, abiraterone, bicalutamide, flutamide, nilutamide, apalutamide, finasteride, dutasteride, alfatradiol, and combinations thereof.
[0075] As used herein, the term“androgen receptor” or“AR” includes a nuclear receptor that binds androgenic hormones, e.g., testosterone or dihydrotestosterone, in the cytoplasm and translocates to the nucleus. AR modulates, inter alia , transcription of target genes by binding to Androgen Response Elements (AREs) in the promoters of such target genes.
[0076] As used herein, the term“AR variant” includes a splice variant of full-length AR. Various AR variants are known. See , Guo el al ., Cancer Res., 69(6):2305-l3 (2009). Exemplary AR variants include, but are not limited to, variants lacking a functional ligand binding domain (LBD). An example of an AR variant that lacks an LBD is AR-V7. “AR- V7” includes androgen receptor splice variant 7, a contituitively active variant of an AR that lacks a functional ligand binding domain (LBD). See , e.g., Hu el al. , Cancer Research , 69(1): 16-22 (2009).
[0077] “Pharmaceutically acceptable” or“therapeutically acceptable” includes a substance which does not interfere with the effectiveness or the biological activity of the active ingredients and which is not toxic to the hosts in the amounts used, and which hosts may be either humans or animals to which it is to be administered.
[0078] “Alkyl” refers to a straight or branched, saturated, aliphatic radical having the number of carbon atoms indicated. Alkyl can include any number of carbons, such as Ci-2, Ci-3, Ci-4, Ci-5, Ci-6, Ci-7, Ci-8, Ci-9, Ci-io, C2-3, C2-4, C2-5, C2-6, C3-4, C3-5, C3-6, C4-5, C4-6 and C5-6. For example, Ci-6 alkyl includes, but is not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, hexyl, etc. Alkyl can also refer to alkyl groups having up to 20 carbons atoms, such as, but not limited to heptyl, octyl, nonyl, decyl, etc. Alkyl groups can be substituted or unsubstituted.
[0079] “Alkylene” refers to a straight or branched, saturated, aliphatic radical having the number of carbon atoms indicated, and linking at least two other groups, i.e., a divalent hydrocarbon radical. The two moieties linked to the alkylene can be linked to the same atom or different atoms of the alkylene group. For instance, a straight chain alkylene can be the bivalent radical of -(CH2)n- where n is 1, 2, 3, 4, 5 or 6. Representative alkylene groups include, but are not limited to, methylene, ethylene, propylene, isopropylene, butylene, isobutylene, sec-butylene, pentylene and hexylene. Alkylene groups can be substituted or unsubstituted.
[0080] “Alkenyl” refers to a straight chain or branched hydrocarbon having at least 2 carbon atoms and at least one double bond. Alkenyl can include any number of carbons, such as C2, C2-3, C2-4, C2-5, C2-6, C2-7, C2-8, C2-9, C2-10, C3, C3-4, C3-5, C3-6, C4, C4-5, C4-6, C5, C5-6, and C6. Alkenyl groups can have any suitable number of double bonds, including, but not limited to, 1, 2, 3, 4, 5 or more. Examples of alkenyl groups include, but are not limited to, vinyl (ethenyl), propenyl, isopropenyl, l-butenyl, 2-butenyl, isobutenyl, butadienyl, l-pentenyl, 2-pentenyl, isopentenyl, l,3-pentadienyl, l,4-pentadienyl, l-hexenyl, 2-hexenyl, 3-hexenyl, l,3-hexadienyl, l,4-hexadienyl, l,5-hexadienyl, 2,4-hexadienyl, or l,3,5-hexatrienyl. Alkenyl groups can be substituted or unsubstituted. [0081] “Heteroalkyl” refers to an alkyl group of any suitable length and having from 1 to 3 heteroatoms such as N, O and S. Additional heteroatoms can also be useful, including, but not limited to, B, Al, Si and P. The heteroatoms can also be oxidized, such as, but not limited to, -S(O)- and -S(0)2-. For example, heteroalkyl can include ethers, thioethers and alkyl-amines. The heteroatom portion of the heteroalkyl can replace a hydrogen of the alkyl group to form a hydroxy, thio or amino group. Alternatively, the heteroatom portion can be the connecting atom, or be inserted between two carbon atoms.
[0082] “Haloalkyl” refers to an alkyl group, where some or all of the hydrogen atoms are replaced with halogen atoms. As for alkyl group, haloalkyl groups can have any suitable number of carbon atoms, such as Ci-6. For example, haloalkyl includes trifluoromethyl, fluoromethyl, etc. In some instances, the term“perfluoro” can be used to define a compound or radical where all the hydrogens are replaced with fluorine. For example, perfluoromethyl refers to 1,1,1 -trifluoromethyl.
[0083] “Alkoxy” refers to an alkyl group having an oxygen atom that connects the alkyl group to the point of attachment: alkyl-O-. As for alkyl group, alkoxy groups can have any suitable number of carbon atoms, such as Ci-6. Alkoxy groups include, for example, methoxy, ethoxy, propoxy, iso-propoxy, butoxy, 2-butoxy, iso-butoxy, sec-butoxy, tert-butoxy, pentoxy, hexoxy, etc. The alkoxy groups can be further substituted with a variety of substituents described within. Alkoxy groups can be substituted or unsubstituted.
[0084] “Aryl” refers to an aromatic ring system having any suitable number of ring atoms and any suitable number of rings. Aryl groups can include any suitable number of ring atoms, such as, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or 16 ring atoms, as well as from 6 to 10, 6 to 12, or 6 to 14 ring members. Aryl groups can be monocyclic, fused to form bicyclic or tricyclic groups, or linked by a bond to form a biaryl group. Representative aryl groups include phenyl, naphthyl and biphenyl. Other aryl groups include benzyl, having a methylene linking group. Some aryl groups have from 6 to 12 ring members, such as phenyl, naphthyl or biphenyl. Other aryl groups have from 6 to 10 ring members, such as phenyl or naphthyl. Some other aryl groups have 6 ring members, such as phenyl. Aryl groups can be substituted or unsubstituted.
[0085] “Heteroaryl” refers to a monocyclic or fused bicyclic or tricyclic aromatic ring assembly containing 5 to 16 ring atoms, where from 1 to 5 of the ring atoms are a heteroatom such as N, O or S. Additional heteroatoms can also be useful, including, but not limited to, B, Al, Si and P. The heteroatoms can also be oxidized, such as, but not limited to, -S(O)- and -S(0)2-. Heteroaryl groups can include any number of ring atoms, such as, 3 to 6, 4 to 6, 5 to 6, 3 to 8, 4 to 8, 5 to 8, 6 to 8, 3 to 9, 3 to 10, 3 to 11, or 3 to 12 ring members. Any suitable number of heteroatoms can be included in the heteroaryl groups, such as 1, 2, 3, 4, or 5, or 1 to 2, 1 to 3, 1 to 4, 1 to 5, 2 to 3, 2 to 4, 2 to 5, 3 to 4, or 3 to 5. Heteroaryl groups can have from 5 to 8 ring members and from 1 to 4 heteroatoms, or from 5 to 8 ring members and from 1 to 3 heteroatoms, or from 5 to 6 ring members and from 1 to 4 heteroatoms, or from 5 to 6 ring members and from 1 to 3 heteroatoms. The heteroaryl group can include groups such as pyrrole, pyridine, imidazole, pyrazole, triazole, tetrazole, pyrazine, pyrimidine, pyridazine, triazine (1,2,3-, 1,2,4- and 1,3, 5-isomers), thiophene, furan, thiazole, isothiazole, oxazole, and isoxazole. The heteroaryl groups can also be fused to aromatic ring systems, such as a phenyl ring, to form members including, but not limited to, benzopyrroles such as indole and isoindole, benzopyridines such as quinoline and isoquinoline, benzopyrazine (quinoxaline), benzopyrimidine (quinazoline), benzopyridazines such as phthalazine and cinnoline, benzothiophene, and benzofuran. Other heteroaryl groups include heteroaryl rings linked by a bond, such as bipyridine. Heteroaryl groups can be substituted or unsubstituted.
[0086] “Cycloalkyl” refers to a saturated or partially unsaturated, monocyclic, fused bicyclic or bridged polycyclic ring assembly containing from 3 to 12 ring atoms, or the number of atoms indicated. Cycloalkyl can include any number of carbons, such as C3-6, C4-6, C5-6, C3-8, C4-8, C5-8, C6-8, C3-9, C3-10, C3-11, and C3-12. Saturated monocyclic cycloalkyl rings include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cyclooctyl. Saturated bicyclic and polycyclic cycloalkyl rings include, for example, norbomane, [2.2.2] bicyclooctane, decahydronaphthalene and adamantane. Cycloalkyl groups can also be partially unsaturated, having one or more double or triple bonds in the ring. Representative cycloalkyl groups that are partially unsaturated include, but are not limited to, cyclobutene, cyclopentene, cyclohexene, cyclohexadiene (1,3- and 1, 4-isomers), cycloheptene, cycloheptadiene, cyclooctene, cyclooctadiene (1,3-, 1,4- and 1, 5-isomers), norbomene, and norbomadiene. When cycloalkyl is a saturated monocyclic C3-8 cycloalkyl, exemplary groups include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. When cycloalkyl is a saturated monocyclic C3-6 cycloalkyl, exemplary groups include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Cycloalkyl groups can be substituted or unsubstituted. [0087] “Heterocycloalkyl” refers to a saturated or partially unsaturated ring system having from 3 to 13 ring members and from 1 to 4 heteroatoms of N, O and S. Heterocycloalkyl groups can include fused bi- or tri- cyclic systems, and can include one or more points of unsaturation. Additional heteroatoms can also be useful, including, but not limited to, B, Al, Si and P. The heteroatoms can also be oxidized, such as, but not limited to, -S(O)- and -S(0)2-. Heterocycloalkyl groups can include any number of ring atoms, such as, 3 to 6, 4 to 6, 5 to 6, 3 to 8, 4 to 8, 5 to 8, 6 to 8, 3 to 9, 3 to 10, 3 to 11, 3 to 12 or 3 to 13 ring members. Any suitable number of heteroatoms can be included in the heterocycloalkyl groups, such as 1, 2, 3, or 4, or 1 to 2, 1 to 3, 1 to 4, 2 to 3, 2 to 4, or 3 to 4. The heterocycloalkyl group can include groups such as aziridine, azetidine, pyrrolidine, piperidine, azepane, azocane, quinuclidine, pyrazolidine, imidazolidine, piperazine (1,2-, 1,3- and 1, 4-isomers), oxirane, oxetane, tetrahydrofuran, oxane (tetrahydropyran), oxepane, thiirane, thietane, thiolane (tetrahydrothiophene), thiane (tetrahydrothiopyran), oxazolidine, isoxazolidine, thiazolidine, isothiazolidine, dioxolane, dithiolane, or morpholine. The heterocycloalkyl groups can also be fused to aromatic or non-aromatic ring systems to form members including, but not limited to, indoline. Heterocycloalkyl groups can be unsubstituted or substituted. For example, heterocycloalkyl groups can be substituted with Ci-6 alkyl, oxo (=0), or aryl, among many others.
[0088] The heterocycloalkyl groups can be linked via any position on the ring. For example, piperidine can be 1-, 2-, 3- or 4-piperidine, pyrazolidine can be 1-, 2-, 3-, or 4- pyrazolidine, imidazolidine can be 1-, 2-, 3- or 4-imidazolidine, piperazine can be 1-, 2-, 3- or 4-piperazine, and morpholine can be 1-, 2-, 3- or 4-morpholine.
[0089] The term“synergy” or“synergistic effect” refers to an effect produced by two or more compounds ( e.g a RORy inhibitor and a statin) that is greater than the effect produced by a sum of the effects of the individual compounds (i.e., an effect that is greater than an additive effect). Several methods are available for determining whether a combination of drugs produces a synergistic effect. As a non-limiting example, the Highest Single Agent approach simply reflects that the fact that the resulting effect of a combination of drugs (EAB) is greater than the effects of the individual drugs (EA and EB). A combination index (Cl) can be calculated according to the formula:
a _ ma X(EA,Eb)
EAB [0090] As another non-limiting example, according to the Response Additivity Approach, a synergistic drug combination effect occurs when the EAB is greater than the expected additive effects of the individual drugs (EA and EB). Here, the Cl is calculated using the formula:
a _ EA +EB
[0091] As yet another non-limiting example, the Bliss Independence model is based on the principle that drug effects are the outcomes of probabilistic processes, and makes the assumption that drugs act independently such that they do not interfere with each other (i.e., different sites of action). However, the model also assumes that each drug contributes to the production of a common result. According to this method, the observed combination effect is expressed as a probability (0 < EAB £ 1) and is compared to the expected additive effect expressed as:
EA + EB (1 -EA) = EA + EB - EAEB, where 0 < EA £ 1 and 0 < EB < 1. The Cl for this method is calculated using the formula:
Cl _ EA +EB ~EAEB
[0092] Methods of identifying synergistic effects are further discussed in Foucquier J. and Guedj M. Pharmacology Research & Perspectives (2015) (3)3:e00l49, incorporated herein by reference in its entirety for all purposes.
III. DESCRIPTION OF THE EMBODIMENTS
[0093] The present invention provides compositions, methods, and kits comprising one or more RORy inhibitors (e.g., a small-molecule compound such as a compound of Formula I ( e.g. , XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statin drugs (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) that are useful for treating cancer, e.g. , prostate cancer, such as castration-resistant prostate cancer (CRPC), and numerous other types of cancer including lung cancer, breast cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, and glioma. In some embodiments, the compositions, methods and kits further comprise an anticancer drug such as an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen- specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, or a combination thereof. In some embodiments, the compositions, methods, and kits comprise an effective amount of the RORy inhibitor, statin, and/or anticancer drug. In some embodiments, RORy inhibitors used in compositions, methods, and kits of the present invention specifically bind to RORy and inhibit RORy activity.
A. Small-Molecule RORy Inhibitors
[0094] Receptor-related orphan receptor y (RORy) inhibitors include small-molecule compounds that inhibit retinoic acid receptor-related orphan receptor y (RORy) transcription, translation, stability, and/or activity. In certain embodiments, a RORy inhibitor binds to RORy and inhibits the activity of the receptor. In other embodiments, a RORy inhibitor selectively binds to RORy. In some embodiments, a RORy inhibitor selectively binds to RORy relative to RORa and/or RORp. In some embodiments, a RORy inhibitor selectively binds to RORy and inhibits RORy activity. In some embodiments, a RORy inhibitor inhibits RORy activity relative to RORa and/or RORp. In some instances, inhibition of RORy activity includes inhibition of recruitment of coactivators such as SRC-l and/or SRC-3 to an androgen receptor (AR) ROR response element (RORE). In other instances, inhibition of RORy activity includes inhibition of transcription of the AR gene and/or a variant thereof such as AR-V7. As used herein, the terms“RORy inhibitor,”“RORy antagonist,” and “RORy inverse agonist” are used interchangeably.
[0095] Non-limiting examples of small-molecule RORy inhibitors include compounds of Formula I ( e.g ., those listed in Table 2, such as XY018 and XY063), GSK805, SR2211, and those described in Pandya et al, J Med. Chem. (2018) (DOI: l0. l02l/acs.jmedchem.8b00588), e.g., those listed in Table 5, such as VTP -43742. Any combination of RORy inhibitors may be used. In some embodiments, the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP-43742, and a combination thereof.
Figure imgf000031_0001
[0096] Compounds of Formula I (e.g, XY018 or XY063) can be used in methods of the present invention as RORy inhibitors or RORy antagonists. [0097] In some embodiments, compounds of Formula I ( e.g ., XY018 or XY063) include inverse agonists that bind to RORy and decrease its activity below a constitutive (e.g., intrinsic or basal) level of activity in the absence of any ligand. In some embodiments, compounds of Formula I include pharmaceutically acceptable salts, derivatives, analogs, isomers, racemates, prodrugs, co-crystalline complexes, hydrates, and solvates thereof.
[0098] In certain embodiments, the compound of Formula I (e.g, XY018 or XY063) has a half-maximal inhibitory concentration (IC50) value of from about 100 nM to about 100 mM, e.g, from about 100 nM to about 50 pM, from about 100 nM to about 25 pM, from about 100 nM to about 10 pM, from about 500 nM to about 100 pM, from about 500 nM to about 50 pM, from about 500 nM to about 25 pM, from about 500 nM to about 10 pM, from about 1 pM to about 100 pM, from about 1 pM to about 50 pM, from about 1 pM to about 25 pM, from about 1 pM to about 10 pM, or about 100 nM, 200 nM, 300 nM, 400 nM, 500 nM, 600 nM, 700 nM, 800 nM, 900 nM, 1 pM, 2 pM, 3 pM, 4 pM, 5 pM, 6 pM, 7 pM, 8 pM, 9 pM, or 10 pM. In some instances, the IC50 value for a specific compound of Formula I is measured using an in vitro assay in cancer cells that have been incubated with the compound. The IC50 value can be determined based on the effect of the compound of Formula I in inhibiting the survival of cancer cells such as cells from a cancer cell line or primary tumor cells. In other embodiments, the compound of Formula I has an inhibitor constant (Kp that is essentially the same numerical value as the IC50 value, or is about one-half the value of the IC50 value.
[0099] In particular embodiments, the compound of Formula I has the following structure:
Figure imgf000032_0001
or a pharmaceutically acceptable salt, isomer, racemate, prodrug, co- crystalline complex, hydrate, or solvate thereof, wherein
X is C(=0) or SO¾
n is an integer selected from the group consisting of 0, 1, 2, or 3,
Ki is selected from the group consisting of FI, halo, alkyl, trifluoromethyl, cyano, -COQRs, -COR.5, -ORs, -CQH(CF3)2, heterocydyi, and cycloalkyl,
wherein R5 is selected from the group consisting of H, and Ci-C3 alkyl group; R2 is selected from the group consisting of FI, halogen, and alkyl;
R3 is selected from the group consisting of H and alkyl; R4 is selected from the group consisting of C0-C4 alkylene-R6, C0-C4 alkylene- R7-cycloa3ky3, and C0-C4 alkylene-R7-heterocyclyl,
wherein Re is selected from the group consisting of -Rg, -ORg, -CQRg, -COOR8, -S(0)mR8, cycloalkyl, and heterocyclyl, m is 0 or 2, and R? is selected from the group consisting of -OR9, -C(0)3¾, -NR9, -SR9, ~S(())R9, -S(0)2R9,
wherein Rg is selected from the group consisting of H, and C1-C3 alkyl group, and R9 is C1-C3 alkyl ene;
wherein each cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, cafboxy, amino, -CONH2, -COOR10, -CQR10, -OR10, -NHCOR10, -NHCOOR10, and -COH(CF3)2,
each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono- , bi- or tri -cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, C1-C4 alkyl, trifluoromethyl, cyano, carhoxy, nitro, amino, -CON! l·, -CQOR10, -COR10, -QR10, -NHCOR10, -NHCOOR10, -COH(CF3)2, -CeHsRn, morphoiinyl, piperidinyl, tetrahydrofuranyl, substituted pyridyl group,
wherein Rio is independently selected from the group consisting of H, C I -C I alkyl, and phenyl, and
R11 is independently selected from the group consisting of C1-C4 alkyl, halogen, acetyl, methoxy, and ethoxy.
[0100] In particular embodiments, Ra is selected from the group consisting of H, methyl, ethyl, propyl, fluoro, chloro, bromo, trifluoromethyl, cyano, -COH(CF3)2, -COOR5, -COR5, -ORs, heterocyclyl, and cycloalkyl, wherein R5 is selected from the group consisting of H, methyl, ethyl, and propyl.
[0101] In certain embodiments, R2 is selected from the group consisting of H, methyl, ethyl, propyl, fluoro, chloro, and bromo.
[0102] In some embodiments, R3 is selected from the group consisting of H, methyl, ethyl, propyl, and isopropyl.
[0103] In some embodiments, R4 is selected from the group consisting of: (1) (':.··( Ί alkylene-Re, wherein R6 is selected from the group consisting of -R8, -ORg, -CORg, -COORg, -S(0)mR8, cycloalkyl, and heterocyclyl, m is 0 or 2, and Rg is selected from the group consisting of H, methyl, ethyl, and propyl group;
(2) Co-C4 aikyiene-R7-cycioaikyi,
wherein said eycioalkyi group is selected from the group consisting of cyclobutane, cyclopentane, cyclohexane, cycloheptane, phenyl, and naphthyl, and said eycioalkyi group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, carboxy, amino, -COM ! ·· -COOR10, -COR10, -OR10, -NHCOR10, -NHCOOR10, and -COH(CF3)2, wherein Rio is selected from the group consisting of methyl, ethyl, propyl, isopropyl, and phenyl; and
(3) C0-C4 alkyiene-R7-heterocyciyl,
wlierein said heterocyclyl group is selected from the group consisting of imidazoly!, triazolyl, pyrazolyl, thienyl, oxazoly!, isoxazoiyl, pyrazinyi, pyridazinyi, pyrimidinyl, pyrrol yl, piperazinyl, tetrahydro-pyrrolyl, piped dinyl, morpholinyl, 1,3- dioxolany!, isoquinolinyl, indoiine group, 1H- indazolyl, 1H- benzo [d] imidazolyl, 1 H- indolyl, benzo [d] [1,3] dioxoie, benzo [d] thiazolyi, and a member of the moieties shown in Table 1, and said heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, C1-C4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -CONH2, -COOR10, -COR10, -OR10, -NHCOR10, -NHCOOR10, -COH(CF3)2, -CsH Ru, morpholinyl, piperidinyl, tetrahydrofuranyl, and a substituted pyridyl group;
wherein R~ is selected from the group consisting of -ORg, -C(0)R9, -NR9, -SR9, -S(0)R9, -S(0)2R9,
R9 is C1-C3 alkylene,
Rio is independently selected from the group consisting of H, methyl, ethyl, propyl, isopropyl, and phenyl,
Rn is independently selected from the group consisting of methyl, ethyl, propyl, isopropyl, fluoro, chloro, bromo, acetyl, methoxy, and ethoxy, and
R12 is selected from the group consisting of H, methyl, ethyl , propyl, and isopropyl. Table 1: Possible Heterocyclic Moieties
Figure imgf000035_0002
[0104] In particular embodiments, the RORy inhibitor compound of Formula I is represented by a compound according to Formula la:
Figure imgf000035_0001
wherein
Ri is selected from the group consisting of H, methyl, ethyl, propyl, fluoro, ch!oro, bromo, trifluoro ethyl, cyano, -COH(CF3)2, -COORs, -CORs, -OR5, heterocyclyl, and cycloalkyl, wherein R5 is selected from the group consisting of FI, and Ci-saikyl group;
R2 is selected from the group consisting of H, fluoro, chloro, bromo;
R3 is selected from the group consisting of H, methyl, ethyl, propyl, and isopropyl;
i is selected from the group consisting of
(1) C0-C4 alkylene-Rs, wherein R& is selected from the group consisting of -Rs, -ORg, -CORs, -COORg, -S(0)mRs, cycloalkyl, and heterocyclyl , m is 0 or 2, and Rg is selected from the group consisting of H, and Ci-3alkyl;
(2) C0-C4 alkylene-R--cycloalkyl, wherein R? is selected from the group consisting of -OR9, -C(0)R9, -NR9, -SR9, -S(0)Rg, -S(0)2R9, wherein
R9 is selected from the group consisting of Cl-3aikylene, heterocyclyl, and cycloalkyl,
wherein each cycloalkyi group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyi group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethy!, cyano, carboxy, amino, -CONH2, -CQORio, -CORio, -ORl0, -NHCORio, -NHCOORio, and -COI Kdrik
each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono- , bi- or tri -cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, Ci-C4 alkyl, trifluoromethyi, cyano, carboxy, nitro, amino, -COM l·, -COORio, -CORl0, -ORl0, -NHCORio, -NHCOORio, -COH(CF3)2, -CcHsRn, morpholinyl, piperidinyl, tetrahydrofuranyl , substituted pyridyl group,
wherein Rl0 is independently selected from the group consisting of C1-C4 alkyl, and phenyl, and
Rn is independently selected from the group consisting of C1-C4 alkyl, halogen, acetyl, methoxy, and ethoxy.
[0105] In particular embodiments, Ri is a cycloalkyl group having an un saturated ring structure of 6 carbon atoms (e.g, a phenyl group), wherein the cycloalkyl group is substituted with a fluoro group (e.g., at the 2' position) and a 1,1, 1,3,3, 3-hexafluoro-2-hydroxypropan-2- yl group (e.g, at the 4' position). In some embodiments, R2 is H. In other embodiments, R3 is H.
[0106] In certain embodiments, the RORy inhibitor compound of Formula I is represented by a compound according to Formula lb:
Figure imgf000036_0001
wherein
R is selected from the group consisting of H, methyl, ethyl, propyl, fluoro, chloro, bromo, trifluoromethyi, cyano, -COH(CF3)2, -COQRs, -CORs, -OR5, heterocyclyl, and cycloalkyl, wherein
R5 is selected from the group consisting of H, and Ci-3 alkyl group, and said cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms,
said cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C -C4 alkyl group. trifluoromethyl, cyano, carboxy, amino, -CONH2, -COOR10, -COR10, -ORl0, -NHCORio,
-NHCOOR10, and -COH(CF3)2,
said heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono-, bi- or tri~ cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, (), and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, C1-C4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -COM l·, -COOR10, -CORl0, -ORl0, -NHCORio, -NHCOOR10, -COH(CF3)2, -CeHjRn, morpholinyl, piperidinyl, tetrahydrofuranyl , substituted pyridyl group;
R2 is selected from the group consisting of H, fluoro, chloro, bromo;
Ri is selected from the group consisting of H, methyl, ethyl, propyl, and isopropyl;
R4 is selected from the group consisting of
(1) C0-C4 alkylene-Re, wherein R6 is selected from the group consisting of -Rg, -ORg, and cycloalkyl, and Rg is selected from the group consisting of H, and Cusalkyl;
(2) C0-C4 alkyl ene-R?- heterocyclyl, wherein R? is selected from the group consisting of -OR9, -C(0)R9, -NR·,·. -SR9, -S(0)R9, -S(0)2R9, and R9 is selected from the group consisting of Ciualkyiene, heterocyclyl, and cycloalkyl,
wherein said heterocyclyl group is selected from the group consisting of imidazolyl, triazolyl, pyrazolyl, thienyl, oxazolyl, isoxazolyl, pyrazinyl, pyridazinyl, pyrimidinyl, pyrrolyl, piperazinyl, tetrahydro-pyrroly! , piperidinyl, morpholinyl, 1,3- dioxolany!, isoquinolinyl, indoline group, 1H- indazolyl, 1H- benzo [d] imidazolyl, IH- indoly!, benzo [d] [1,3] dioxole, benzo [d] thiazo!yl, and a member of the moieties shown in Table 1, and said heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, Cj-C4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -CONH2, -COOR10, -COR10, -OR10, -NHCORio, -NHCOOR10, -COH(CF3)2, -CfHsRn, morpholinyl, piperidinyl, tetrahydrofuranyl, substituted pyridyl group;
Rl0 is independently selected from the group consisting of Ch-4 alkyl and
RJ J is independently selected from the group consisting of methyl, ethyl, propyl, isopropyl, halogen, acetyl, methoxy, and ethoxy, and
R12 is selected from the group consisting of H, methyl, ethyl , propyl, isopropyl. [0107] In particular embodiments, the compound of Formula I is selected from the group consisting of l-ethyl -N- (2- fluoro-4- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) phenyl) -2- oxo-l, 2- dihydro-benzo [cd] indole-6-sulfonamide, N- (2- fluoro-4- (1, 1, 1,3,3, 3-hexafluoro- 2-hydroxy-2-yl) phenyl) -4,4-dimethyl-2-oxo- l,2,3,4-tetrahydro-quinolin-6-sulfonamide, N- (2- fluoro-4- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) phenyl) - 2-oxo-l -propyl- 1, 2,3,4- tetrahydro-quinolin-6-sulfonamide, N- (2- fluoro-4- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) phenyl) - 2-oxo-3 -propyl -2, 3-benzo [d] oxazole -6- sulfonamide, N- (2- fluoro-4- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) phenyl) -2-oxo-indoline-5-sulfonamide, N- (2- fluoro-4- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) phenyl) -2,4-dioxo-l,3- propyl-l, 2,3,4- tetrahydro-quinazolin-6-sulfonamide, N- (2- fluoro-4- (1, 1, 1,3,3, 3-hexafluoro-2-hydroxy-2- yl) phenyl) -2-oxo-l, 2, 3, 4 - tetrahydro-quinolin-6-sulfonamide, l-ethyl -N- (2- fluoro-4- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1,1 '-biphenyl] 4-yl) -2-oxo-l, 2-dihydro-benzo [cd] indole-6-sulfonamide, N- (2- fluoro-4- (l, l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [I, G- biphenyl] -4-yl) 4,4-dimethyl-2-oxo-l,2,3,4-tetrahydro-quinolin-6-sulfonamide, N- (2- fluoro-4- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) phenyl) - [l,l'-biphenyl] -4 - yl) -2-oxo-l- propyl-l,2,3,4-tetrahydro-quinolin-6-sulfonamide, N- (2- fluoro-4- (1,1, 1,3,3, 3-hexafluoro-2- hydroxy-2-yl) - [l, l'-biphenyl] -4-yl) 2-oxo-3 -propyl-2,3 -benzo [d] oxazole -6- sulfonamide, N- (2- fluoro-4- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl] -4-yl) 2-oxo- indoline-5-sulfonamide, N- (2- fluoro-4- (l, l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [I, G- biphenyl] -4-yl) 2,4-dioxo-l,3-dipropyl-l,2,3,4-tetrahydro-quinazolin-6-sulfonamide, N- (2- fluoro-4- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl] -4-yl) 2,4-dioxo-l,2,3,4- tetrahydro-quinazolin-6-sulfonamide, N- (2- fluoro-4- (1, 1, 1,3,3, 3-hexafluoro-2-hydroxy-2- yl) - [l, l'-biphenyl] -4-yl) 2-oxo-2, 3-benzo [d] oxazole -6- sulfonamide, N- (2- fluoro-4- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l, l'-biphenyl] -4-yl) 2-oxo-l, 2,3, 4-tetrahydro- quinolin-6-sulfonamide, N- (2- fluoro-4- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [I,G- biphenyl] -4-yl) 3-methyl-2-oxo-l,2,3,4-tetrahydro-quinazolin-6-sulfonamide, l-acetyl— N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [I,G- phenyl] -4-yl) indoline-5- sulfonamide, N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l, l'-biphenyl] -4 yl) -2, 2-dimethyl-3-oxo-3, 4-dihydro -2H- benzo [b] [1,4] oxazine-6-sulfonamide, 4-acetyl— N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [I, G- phenyl] -4-yl) -2,2-dihydro- 3, 4-dihydro -2H- benzo [b] [1,4] oxazine-6-sulfonamide, N- (2'- fluoro-4 '- (1, 1,1, 3,3,3- hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl] -4 yl) -3 -oxo-3, 4-dihydro -2H- benzo [b] [1,4] oxazine-6-sulfonamide, N- (2'- fluoro-4 '- (l, l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [I, G- biphenyl] -4 yl) -2-oxo-2, 4-dihydro -1H- benzo [d] [1,3] oxazin-7-sulfonamide, N- (2'- fluoro-4 (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1, 1 '-biphenyl] -4 yl) -4-methyl-3-oxo- 3, 4-dihydro -2H- benzo [b] [1,4] oxazine-6-sulfonamide, 4-acetyl— N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [I,G- phenyl] -4-yl) -3,4-dihydro -2H- benzo [b] [1,4] oxazine-6-sulfonamide, 4-fluoro— N- (2'- fluoro-4 '- (1,1, 1,3,3, 3-hexafluoro-2-hydroxy- 2-yl) - [l, l'-biphenyl ] -4-yl) benzenesulfonamide, 2,4-Difluoro -N- (2'- fluoro-4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [I, G- biphenyl] -4-yl) benzenesulfonamide, N- (2 - fluoro-4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l, l'-biphenyl] -4 yl) thiophene-2- sulfonamide, N- (2'- fluoro-4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl] -4 yl) -2,3-dihydro-benzo [b] [1,4] dioxin-6-sulfonamide, N- (2'- fluoro-4 '- (1, 1, 1, 3,3,3- hexafluoro-2-hydroxy-2-yl) - [l, l'-biphenyl] -4 yl) -4-phenoxy-benzenesulfonamide, N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl] -4 yl) -4-phenyl- morpholin-benzenesulfonamide. N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl] -4-yl ) -2- (p-toluene-yl) acetamide, 2- (3,4-dimethoxyphenyl) -N- (2'- fluoro- 4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl ) - [l, l'-biphenyl] -4-yl) -2-acetamide, 2- (4- chlorophenyl) -N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1, G-biphenyl] - 4-yl) -2-acetamide, 2- (4-bromophenyl) -N- (2'- fluoro-4 '- (1,1, 1,3,3, 3-hexafluoro-2- hydroxy-2-yl) - [1, G-biphenyl] -4-yl) -2-acetamide, 2- (3-chlorophenyl) -N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1, G-biphenyl] -4-yl) -2-acetamide, N- (2'- fluoro- 4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl] -4-yl ) -2- (4-nitrophenyl) acetamide, N- (2'- fluoro-4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl] -4-yl ) -2- (2-nitrophenyl) acetamide, N- (2'- fluoro-4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl] -4-yl ) -2- (4- (trifluorom ethyl) phenyl) acetamide, N- (2'- fluoro-4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [I, G -biphenyl] -4-yl ) - pentanamide, N- (2'- fluoro-4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl] -4-yl ) -3,3- dimethylbutanamide, 2- (3,4-dichlorophenyl) -N- (2'- fluoro-4 '- (1,1, 1,3,3, 3-hexafluoro-2- hydroxy-2-yl) - [l,l'-biphenyl] -4-yl) acetamide, N- (2'- fluoro-4 '- (1, 1, 1,3,3, 3-hexafluoro-2- hydroxy-2-yl) - [l,l'-biphenyl] -4-yl ) benzamide, N- (2'- fluoro-4 '- (l, l, l,3,3,3-hexafluoro- 2-hydroxy-2-yl) - [l,l'-biphenyl] -4-yl ) -2- (p-tolyl) benzenesulfonamide, 4 - ((N- (2- fluoro- 4- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) phenyl) sulfamoyl) methyl) benzoic acid methyl, 4 - ((N- (2- fluoro-4- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [ 1,1' -biphenyl] -4 - yl) sulfamoyl) methyl) benzoic acid, 4 - ((N- (2- fluoro-4- (1,1, 1,3,3, 3-hexafluoro-2-hydroxy-2- yl) phenyl) sulfamoyl) methyl) benzoic acid, 4 - ((N- (2- fluoro-4- (1,1, 1,3,3, 3-hexafluoro-2- hydroxy-2-yl) - [l,l'-biphenyl] -4 - yl) -N- propyl-sulfamoyl-yl) methyl) benzoate, 4 - ((N - ([I,G- biphenyl] -4-yl) sulfamoyl) methyl) benzoate 4 - ((N- (4- acetylphenyl) sulfamoyl) methyl) benzoate, 4 - ((N- (3,4- dimethoxyphenyl) amino) methyl) benzoate, 4 - ((N- (2- oxo-l -propyl- 1,2, 3, 4-tetrahydro-quinolin-6-yl) sulfamoyl) methyl) benzoic acid, N- (2'- fluoro-4 (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1,1 '-biphenyl] -4-yl ) -1- (4- (methyl sulfonyl) phenyl) methanesulfonamide, N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1, 1 '-biphenyl] -4-yl ) -4- (trifluoromethyl) benzamide, 2 - ((N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1, l'-biphenyl] 4-yl) sulfamoyl) methyl) benzoate, N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1, G-biphenyl] -4-yl ) -1- (3-nitrophenyl) methyl sulfonamide, 3 - ((N- (2'- fluoro-4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1, G-biphenyl] 4-yl) sulfamoyl) methyl) benzoate, 1- (4-cyanophenyl) -N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1 , l'- biphenyl] -4-yl) methyl sulfonamide, N- (2'- fluoro-4 '- (1,1, 1,3,3, 3-hexafluoro-2-hydroxy-2- yl) - [1, G-biphenyl] -4-yl ) -1- (2-nitrophenyl) methyl sulfonamide, N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1, G-biphenyl] -4-yl ) -1- (4-nitrophenyl) methyl sulfonamide, 4 - ((N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1, l'-biphenyl] 4-yl) amino) methyl) benzoate, 3 - ((N- (2'- fluoro-4 '- (1,1, 1,3,3, 3-hexafluoro-2-hydroxy-2- yl) - [1, G-biphenyl] 4-yl) sulfamoyl) methyl) benzoic acid
2 - ((N- (2'- fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1, G-biphenyl] 4-yl) sulfamoyl) methyl) benzoic acid, 4 - ((N- (2'- fluoro-4 '- (1,1, 1,3,3, 3-hexafluoro-2-hydroxy-2- yl) - [1, G-biphenyl] 4-yl) sulfamoyl) methyl) -N- methylbenzamide, 4 - ((N- (2'- fluoro-4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1, G-biphenyl] 4-yl) sulfamoyl) methyl) benzamide, 4 - ((N- (3'- fluoro - [1, G-biphenyl] -4-yl) sulfamoyl) methyl) benzoate, 4 - ((N- (4'- fluoro - [1, G-biphenyl] -4-yl) sulfamoyl) methyl) benzoate, 4 - ((N- (4'- chloro-biphenyl- 4-yl) sulfamoyl) methyl) benzoate, 4 - ((N- (3'- chloro [1, G-biphenyl] -4-yl) sulfamoyl) methyl) benzoate, 4 - ((N- (3 ', 4'- difluoro [1, G-biphenyl] -4-yl) sulfamoyl) methyl) benzoate, 4 - ((N- (2 '- (trifluoromethoxy) - [1, G-biphenyl] -4-yl) sulfamoyl) methyl) benzoate, 4 - ((N- (3'- methoxy [1, G-biphenyl] -4-yl) sulfamoyl) methyl) benzoate, 4 - ((N- (4'- methoxy - [1, G-biphenyl] -4-yl) sulfamoyl) methyl) benzoate, 4 - ((N- (2'- fluoro - [1, l'- biphenyl] -4-yl) sulfamoyl) methyl) benzoate, N- (2'- fluoro-4 '- (1,1, 1,3,3, 3-hexafluoro-2- hydroxy-2-yl) - [1, G-biphenyl] -4-yl ) benzenesulfonamide, N- (2'- fluoro-4 '- (1, 1,1, 3,3,3- hexafluoro-2-hydroxy-2-yl) - [1, G-biphenyl] -4-yl ) -2,3-dihydro-benzo [b] [1,4] dioxin-6- carboxamide, N- (2'- fluoro-4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1, l'-biphenyl] -4- yl ) -4-phenoxy-benzamide, 4 - ((2'-fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1, l'-biphenyl] -4- yl) carbamoyl) benzoate, N- (2'- fluoro-4 '- (1, 1, 1,3,3, 3-hexafluoro-2- hydroxy-2-yl) - [1, G-biphenyl] -4-yl ) -3 -phenyl-propanamide, 4,4,4-trifluoro -N- (2'- fluoro- 4 (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1,1 biphenyl] -4-yl) butanamide, N- (2'- fluoro-4 (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [1,1 '-biphenyl] -4-yl ) heptanamide, 4- cyclohexyl -N- (2'- fluoro-4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl ] -4- yl) butanamide, Methyl-4 - ((2'-fluoro-4 '- (l, l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [I,G- biphenyl] 4-yl) amino) -4-oxobutanoic acid, Methyl-5 - ((2'-fluoro-4 '- (1, 1,1, 3,3,3- hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl] 4-yl) amino) -5-oxo-pentanoic acid, Methyl-6 - ((2'-fluoro-4 '- (l,l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl] 4-yl) amino) -6-oxo- hexanoate, N- (2'- fluoro-4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l,l'-biphenyl] -4-yl ) -5-oxo-hexanamide, N- (2'- fluoro-4 '- (l, l, l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [I, G- biphenyl] -4-yl ) -1- (4- (trifluorom ethyl) phenyl) methanesulfonamide, 4 - ((N- (2'- fluoro-4 '- (l, l,l,3,3,3-hexafluoro-2-hydroxy-2-yl) - [l, l'-biphenyl] 4-yl) sulfamoyl) methyl) benzoate, and a combination thereof.
[0108] In some embodiments, the RORy inhibitor compound of Formula I is a compound in Table 2.
Table 2: Exemplary Structures of RORy inhibitor compounds of Formula I
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0002
[0109] In particular embodiments, the RORy inhibitor compound of Formula I is represented by a compound according to any one of Formulas Ic to Ii:
Figure imgf000053_0001
Figure imgf000054_0001
or a pharmaceutically acceptable salt thereof, a derivative thereof, an analog thereof, or a combination thereof.
[0110] The compound of Formula Ic is also called XY018 or F18 and corresponds to Structure No. 37 in Table 2. The compound of Formula Id is also called F17 and corresponds to Structure No. 36 in Table 2. The compound of Formula Ie is also called F62 and corresponds to Structure No. 80 in Table 2. The compound of Formula If is also called XY063 or F63 and corresponds to Structure No. 81 in Table 2. The compound of Formula Ig is also called F64 and corresponds to Structure No. 82 in Table 2. The compound of Formula Ih is also called F65 and corresponds to Structure No. 83 in Table 2. The compound of Formula Ii is also called F68 and corresponds to Structure No. 86 in Table 2. [0111] In some embodiments, the RORy inhibitor compound of Formula I is a compound disclosed in Chinese Patent Application No. 201410344302.0, filed July 18, 2014, the disclosure of which is hereby incorporated by reference in its entirety for all purposes.
[0112] In some embodiments, the RORy inhibitor compound of Formula I is a compound disclosed in Chinese Patent Publication No. CN 105272904, the disclosure of which is hereby incorporated by reference in its entirety for all purposes.
2. GSK805
[0113] The term“GSK805” refers to N-[2,6-dichloro-2'-(trifluoromethoxy)[l,r-biphenyl]- 4-yl]-4-(ethylsulfonyl)-benzeneacetamide, which has the following structure:
Figure imgf000055_0001
as well as pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and/or solvates thereof.
[0114] GSK805 is an orally bioavailable retinoic acid receptor-related orphan receptor gΐ (RORyt) inverse agonist that directly and reversibly interacts with the receptor's putative ligand binding domain to block its effects on transcription without exerting significant effects on DNA binding. GSK805 inhibits the expression of IL-17 (e.g., at a concentration of 0.5 mM) in naive CD4+ T cells activated under Thl 7-cell-polarizing conditions and affects the broader RORyt-dependent gene network, inhibiting the development and pathogenic function of Thl7 cells.
3. SR2211
[0115] The term “SR2211” refers to 2-fluoro-4’-[[4-(4-pyridinylmethyl)-l- piperazinyl]methyl]-a,a-Z>A(trifluoromethyl)-[l,r-biphenyl]-4-methanol, which as the following structure:
Figure imgf000056_0001
as well as pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and/or solvates thereof.
[0116] SR2211 selectively binds RORy (Ki = 105 nM) and acts as an inverse agonist of constitutive in vitro RORy activity (ICso = 320 nM). SR2211 exerts minimal effects on the activity of RORa, LXRa, and FXR. SR 2211 significantly inhibits gene expression of IL-17 and IL-23 receptor in activated EL-4 mouse T lymphocytes, e.g., when given at a concentration of 5 mM.
4. Additional RORy inhibitors
[0117] Additional examples of suitable RORy inhibitors (i.e., that can be used in methods of the present invention, e.g., either alone or with a statin as described herein) are described in Pandya et al., J Med. Chem. (2018) (DOI: l0. l02l/acs.jmedchem.8b00588; hereby incorporated by reference in its entirety for all purposes), some of which are discussed below.
Table 5 Exemplary additional RORy inhibitors.
Figure imgf000056_0002
Figure imgf000057_0001
Figure imgf000058_0002
[0118] In some embodiments, the RORy inhibitor is a sulfonamide of a cyclic amine. In some embodiments, the sulfonamide of a cyclic amine has the following structure:
Figure imgf000058_0001
wherein R13 is H or F.
[0119] In some embodiments, the sulfonamide of a cyclic amine is N-((R)-2-fluoro-3- hydroxy-3 -methylbutyl)-2-((S)- 1 -((4-fluorophenyl)sulfonyl)-6-( 1 , 1 , 1 ,3 ,3 ,3 -hexafluoro-2- hydroxypropan-2-yl)-l,2,3,4-tetrahydroquinolin-2-yl)acetamide (chemical formula: C25H26F8N2O5S, molecular weight: 618.54), which has the following structure:
Figure imgf000059_0001
[0120] In some embodiments, the sulfonamide of a cyclic amine has the following structure:
Figure imgf000059_0002
wherein R14 is CFh or O.
[0121] In some embodiments, the sulfonamide of a cyclic amine is 2-chloro-6-fluoro-N-(2- ((3-(trifluoromethyl)phenyl)sulfonyl)-2,3,4,5-tetrahydro-lH-benzo[c]azepin-8-yl)benzamide (chemical formula: C24H19CIF4N2O3S, molecular weight: 526.93), which has the following structure:
Figure imgf000059_0003
[0122] In some embodiments, the RORy inhibitor is an aryl sulfonyl compound. Within these compounds, a sulfonyl group attached to an aryl or heteroaryl ring can participate in hydrogen bond interactions ( e.g with Arg367 and/or Leu287 of RORy) that contribute to RORy inhibitory potency. In some embodiments, the aryl sulfonyl compound is a cyclopropyl derivative, e.g., N-(3,5-dichloro-4-(l-(5-(4-fluorophenyl)-l,2,4-oxadiazol-3- yl)cyclopropyl)phenyl)-2-(4-(ethylsulfonyl)phenyl)acetamide (chemical formula: C27H22CI2FN3O4S, molecular weight: 574.45), which has the following structure:
Figure imgf000060_0001
[0123] The cyclopropyl ring (e.g, 1,1 -di substituted cyclopropyl ring) can, at least in some instances, project an attached aryl or heteroaryl group such that occupancy of the hydrophobic pocket of a RORy ligand binding domain is improved.
[0124] In some embodiments, the aryl sulfonyl compound contains a hydroxymethyl group, e.g, (S)-7-ethyl-N-((R,4Z,6E)-6-(ethylsulfonyl)-l-hydroxyocta-4,6-dien-2-yl)-6-(((lr,4S)-4- (trifluoromethyl) cyclohexyl)methyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridine-3-carboxamide (chemical formula: C28H36F3N3O4S, molecular weight: 567.67), which has the following structure:
Figure imgf000060_0002
[0125] In some embodiments, the aryl sulfonyl compound is (S)-N-((5- (ethylsulfonyl)pyridin-2-yl)methyl)-7-isopropyl-6-(((lr,4S)-4-(trifluoromethyl) cyclohexyl) methyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridine-3 -carboxamide (also known as VTP -43742) (chemical formula: C27H35F3N4O3S, molecular weight: 552.66), which has the following structure:
Figure imgf000061_0001
[0126] In some embodiments, the aryl sulfonyl compound contains a terminal CF3 group that interacts with His479 and/or Trp3 l7 of RORy, thus creating a steric clash that destabilizes helix 12 of RORy (i.e., in the agonist position). In some embodiments, the aryl sulfonyl compound inhibits RORy by trapping a water molecule, thus forming a hydrogen bond to Tyr502 of RORy. In some embodiments, connectivity of the carboxamide group to the central core of the aryl sulfonyl compound is inverted ( e.g ., (S)-7-ethyl-N-((R,4Z,6E)-6- (ethylsulfonyl)-l-hydroxyocta-4,6-dien-2-yl)-6-(((lr,4S)-4-(trifluoromethyl) cyclohexyl) methyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridine-3 -carboxamide or (S)-N-((5-
(ethylsulfonyl)pyridin-2-yl)methyl)-7-isopropyl-6-(((lr,4S)-4-(trifluoromethyl) cyclohexyl) methyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridine-3-carboxamide).
[0127] In some embodiments, the aryl sulfonyl compound is an isoindoline derivative, e.g., (R)-2-acetyl-N-(4-(l,l,l,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl)-5-(methylsulfonyl) isoindoline-l-carboxamide (chemical formula: C21H18F6N2O5S, molecular weight: 524.43), which has the following structure:
Figure imgf000061_0002
[0128] In some embodiments, the aryl sulfonyl compound contains an N-acetyl group comprising an oxygen atom that hydrogen bonds with the RORy ligand binding domain. In some embodiments, the aryl sulfonyl compound contains a hexafluoroisopropanol moiety that interacts with helices 11 and/or 12 of RORy.
[0129] In some embodiments, the RORy inhibitor is an amide/bis-amide derivative. In some embodiments, the amide/bis-amide derivative contains one or two amide groups that either directly or indirectly ( e.g water-mediated) participate in hydrogen bond interactions with Phe 377, Glu379, and/or His479 of RORy.
[0130] In some embodiments, the amide/bis-amide derivative is a piperazine derivative, e-g (S)-N-(8-((4-(cyclopentanecarbonyl)-3-methylpiperazin-l-yl)methyl)-7- methylimidazo[l,2-a]pyridin-6-yl)-2-methylpyrimidine-5-carboxamide (chemical formula: C26H33N7O2, molecular weight: 457.60), which has the following structure:
Figure imgf000062_0001
[0131] In some embodiments, the amide/bis-amide derivative contains a pyrimidine ring comprising two nitrogen atoms that form water-mediated hydrogen bonds to residues Arg367, Leu287, and/or Glu379 of RORy. In some embodiments, the amide/bis-amide derivative contains an NH group that participates in a hydrogen bond to Phe377 of RORy.
[0132] In some embodiments, the amide/bis-amide derivative is 2-((lS,3s)-3-((R)-5-((7- fluoro-l,l-dimethyl-2,3-dihydro-lH-inden-5-yl)carbamoyl)-2-methoxy-5,6,7,8-tetrahydro- l,6-naphthyridine-6-carbonyl)cyclobutyl)acetic acid (chemical formula: C28H32FN3O5, molecular weight: 509.58), which has the following structure:
Figure imgf000062_0002
[0133] In some embodiments, the amide/bis-amide derivative contains an amide group that participates in a hydrogen bond interaction with Phe377 of RORy, an amide group that participates in a hydrogen bond interaction with Glu379 of RORy, a pyridine ring comprising a nitrogen atom that participates in a hydrogen bond interaction with Gln286 of RORy, and/or a methoxy group that participates in a hydrogen bond interaction with Arg364 of RORy. [0134] In some embodiments, the amide/bis-amide derivative is 3-cyano-N-(3-(l- (cyclopentanecarbonyl)piperidin-4-yl)-l-methyl-4-(trifluoromethyl)-lH-pyrrolo[2,3-b] pyridin-5-yl)benzamide (chemical formula: C28H28F3N5O2, molecular weight: 523.56), which has the following structure:
Figure imgf000063_0001
[0135] In some embodiments, the RORy inhibitor is an indole derivative. In some embodiments, the indole derivative is (2,4-dichloro-3-((l,4-dimethyl-6-(trifluoromethyl)-lH- indol-2-yl)methyl)phenyl)(4-(oxetan-3-yl)piperazin-l-yl)methanone (chemical formula: C26H26CI2F3N3O2, molecular weight: 540.41), which has the following structure:
Figure imgf000063_0002
B. Statins
[0136] Statins are a group of compounds that are 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase inhibitors (also known as HMG-CoA inhibitors or HMGCR inhibitors) that function as lipid-lowering compounds. HMG-CoA is an enzyme {i.e., NADH-dependent (EC 1.1.1.88), NADPH-dependent (EC 1.1.1.34)) that is the rate-limiting enzyme of the mevalonate pathway, i.e., the metabolic pathway that produces cholesterol and other isoprenoids. HMG-CoA is normally suppressed by cholesterol that is derived from the internalization and degradation of low density lipoprotein (LDL) via the LDL receptor, as well as oxidized species of cholesterol. Competitive inhibition of HMG-CoA by statins initially reduces cholesterol production, and, as an adaptive response, the reduced cellular cholesterol level triggers the sterol regulatory element-binding protein (SREBP)-mediated activation of gene expression, including that of LDL receptors (LDLRs) in the liver and other tissues, which leads to an increased uptake of LDL from the circulation, hence lowering blood cholesterol levels.
[0137] Non-limiting examples of statins suitable for use in compositions, methods, and kits of the present invention include atorvastatin, cerivastatin, fluvastatin, lovastatin, mevastatin, pitavastatin, pravastatin, rosuvastatin, and simvastatin, the chemical structures of which are shown in FIG. 15. Furthermore, it is understood that combinations of statins can be used in compositions, methods, and kits of the present invention.
C. Anticancer Drugs
[0138] In certain embodiments, the RORy inhibitor (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and the statin (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) can be used in combination with an anticancer drug to reduce or reverse cancer cell resistance to the anticancer drug, e.g, by sensitizing the cancer cell to the anticancer drug.
[0139] Non-limiting examples of anticancer drugs include anti-androgen drugs, chemotherapeutic agents, radiotherapeutic agents, antigen-specific immunotherapeutic agents, endocrine therapies, tyrosine kinase inhibitors, and combinations thereof.
1. Anti-Androgen Drugs
[0140] Anti-androgen drugs are compounds that inhibit the transcription, translation, stability, and/or activity of androgen receptors (AR) or variants thereof (e.g. AR-V7). Inhibition of AR activity can include inhibition of recruitment of AR to Androgen Response Elements (AREs). In some embodiments, inhibition of AR activity can include inhibition of recruitment of AR to the PSA promoter. In some embodiments, inhibition of AR activity can include inhibition of AR-induced activation of the PSA promoter. In some embodiments, inhibition of AR activity can include inhibition of AR-induced PSA production. For example, inhibition of AR can include inhibition of production of PSA in the absence of DHT.
[0141] Anti-androgen drugs include, but are not limited to, enzalutamide, abiraterone, bicalutamide, flutamide, nilutamide, apalutamide, finasteride, dutasteride, alfatradiol, and combinations thereof. [0142] In some embodiments, the present invention provides a composition comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more anti-androgen drugs. In certain instances, the composition further comprises a pharmaceutically acceptable excipient or diluent. In other instances, the composition is formulated for oral or parenteral administration.
[0143] In other embodiments, the present invention provides a method for treating cancer in a subject comprising administering to the subject an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more anti-androgen drugs. In certain instances, the effective amount of the RORy inhibitor and the statin is an amount sufficient to sensitize an anti-androgen drug-resistant cancer such as anti -androgen drug-resistant prostate cancer (e.g, castration-resistant prostate cancer) to anti -androgen drug treatment. Combinations of RORy inhibitors and statins with anti-androgen drugs can be delivered to a subject via the same route of administration (e.g, orally or parenterally) or via different routes of administration (e.g, intravenously for RORy inhibitors and/or a statins and orally for anti -androgen drugs, or vice versa).
2. Chemotherapeutic Agents
[0144] Chemotherapeutic agents are well known in the art and include, but are not limited to, anthracenediones (anthraquinones) such as anthracy clines (e.g, daunorubicin (daunomycin; rubidomycin), doxorubicin, epirubicin, idarubicin, and valrubicin), mitoxantrone, and pixantrone; platinum-based agents (e.g, cisplatin, carboplatin, oxaliplatin, satraplatin, picoplatin, nedaplatin, triplatin, and lipoplatin); tamoxifen and metabolites thereof such as 4-hydroxytamoxifen (afimoxifene) and N-desmethyl-4-hydroxytamoxifen (endoxifen); taxanes such as paclitaxel (taxol), docetaxel, cabazitaxel, hongdoushan A, hongdoushan B, hongdoushan C, baccatin I, baccatin II, and lO-deacetylbaccatin; alkylating agents ( e.g ., nitrogen mustards such as mechlorethamine (HN2), cyclophosphamide, ifosfamide, melphalan (L-sarcolysin), and chlorambucil); ethylenimines and methylmelamines (e.g., hexamethylmelamine, thiotepa, alkyl sulphonates such as busulfan, nitrosoureas such as carmustine (BCNU), lomustine (CCNLJ), semustine (methyl-CCN-U), and streptozoein (streptozotocin), and triazenes such as decarbazine (DTIC; dimethyltriazenoimidazolecarboxamide)); antimetabolites (e.g, folic acid analogs such as methotrexate (amethopterin), pyrimidine analogs such as fluorouracil (5-fluorouracil; 5-FU), floxuridine (fluorodeoxyuridine; FUdR), and cytarabine (cytosine arabinoside), and purine analogs and related inhibitors such as mercaptopurine (6-mercaptopurine; 6-MP), thioguanine (6-thioguanine; 6-TG), and pentostatin (2'-deoxycofonnycin)); natural products (e.g, vinca alkaloids such as vinblastine (VLB) and vincristine, epipodophyllotoxins such as etoposide and teniposide, and antibiotics such as dactinomycin (actinomycin D), bleomycin, plicamycin (mithramycin), and mitomycin (mitomycin Q); enzymes such as L-asparaginase; biological response modifiers such as interferon alpha); substituted ureas such as hydroxyurea; methyl hydrazine derivatives such as procarbazine (N-methylhydrazine; MIH); adrenocortical suppressants such as mitotane (o,p'-DDD) and aminoglutethimide; analogs thereof; derivatives thereof; and combinations thereof.
[0145] In some embodiments, the present invention provides a composition comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more chemotherapeutic agents. In certain instances, the composition further comprises a pharmaceutically acceptable excipient or diluent. In other instances, the composition is formulated for oral or parenteral administration.
[0146] In other embodiments, the present invention provides a method for treating cancer in a subject comprising administering to the subject an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more chemotherapeutic agents. In certain instances, the effective amount of the one or more RORy inhibitors and one or more statins is an amount sufficient to sensitize a chemotherapy drug-resistant cancer such as a tamoxifen- resistant cancer ( e.g ., tamoxifen -resistant breast cancer) or a taxane-resistant cancer (e.g, docetaxel-resistant prostate cancer) to chemotherapy drug treatment. Combinations of RORy inhibitors, statins, and chemotherapeutic agents can be delivered to a subject via the same route of administration (e.g, orally or parenterally) or via different routes of administration (e.g, intravenously for RORy inhibitors and statins and orally for chemotherapeutic agents, or vice versa).
3. Radiotherapeutic Agents
[0147] Radiotherapeutic agents are well known in the art and can comprise external-beam radiation therapy and/or internal radiation therapy. External beam radiation therapy delivers radioactive beams of high energy X-rays and/or gamma rays to a patient’s tumor, whereas internal radiation therapy delivers radioactive atoms to a patient’s tumor. Both external beam radiation therapy and internal radiation therapy are used to suppress tumor growth or kill cancer cells by delivering a sufficient quantity of radioactivity to the target site. In some embodiments, the radiotherapeutic agent comprises a radioactive atom and is complexed with a biologic or synthetic agent to increase delivery to the target site. Such biologic or synthetic agents are known in the art. Suitable radioactive atoms for use with the RORy inhibitors (e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) include any of the radionuclides described herein, or any other isotope which emits enough energy to destroy a targeted tissue or cell. In some embodiments, radiotherapeutic agents may be coupled to targeting moieties, such as antibodies, to improve the localization of radiotherapeutic agents to cancerous cells.
[0148] The term “radionuclide” is intended to include any nuclide that exhibits radioactivity. A“nuclide” refers to a type of atom specified by its atomic number, atomic mass, and energy state, such as carbon 14 (14C). “Radioactivity” refers to the radiation, including alpha particles, beta particles, nucleons, electrons, positrons, neutrinos, and gamma rays, emitted by a radioactive substance. Examples of radionuclides suitable for use in the present invention include, but are not limited to, fluorine 18 (18F), fluorine 19 (19F), phosphorus 32 (32P), scandium 47 (47Sc), cobalt 55 (55Co), copper 60 (60Cu), copper 61 (61Cu), copper 62 (62Cu), copper 64 (64Cu), gallium 66 (66Ga), copper 67 (67Cu), gallium 67 (67Ga), gallium 68 (68Ga), rubidium 82 (82Rb), yttrium 86 (86Y), yttrium 87 (87Y), strontium 89 (89Sr), yttrium 90 (90Y), rhodium 105 (105Rh), silver 111 (luAg), indium 111 (U1ln), iodine 124 (124I), iodine 125 (125I), iodine 131 (131I), tin H7m (117mSn), technetium 99m ("mTc) promethium 149 (149Pm), samarium 153 (153Sm), holmium 166 (166HO), lutetium 177 (177LU), rhenium 186 (186Re), rhenium 188 (188Re), thallium 201 (201Tl), astatine 211 (211At), and bismuth 212 (212Bi). As used herein, the“m” in 117mSn and 99mTc stands for the meta state. Additionally, naturally-occurring radioactive elements such as uranium, radium, and thorium, which typically represent mixtures of radioisotopes, are suitable examples of radionuclides. 67Cu, 131I, 177Lu, and 186Re are beta- and gamma-emitting radionuclides. 212Bi is an alpha- and beta-emitting radionuclide. 211At is an alpha-emitting radionuclide. 32P, 47Sc, 89Sr, 90Y, 105Rh, luAg, 117mSn, 149Pm, 153Sm, 166HO, and 188Re are examples of beta- emitting radionuclides. 67Ga, U1ln, 99mTc, and 201Tl are examples of gamma-emitting radionuclides. 55Co, 60Cu, 61Cu, 62Cu, 66Ga, 68Ga, 82Rb, and 86Y are examples of positron- emitting radionuclides. 64Cu is a beta- and positron-emitting radionuclide.
[0149] In some embodiments, the present invention provides a composition comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more radiotherapeutic agents. In certain instances, the composition further comprises a pharmaceutically acceptable excipient or diluent. In other instances, the composition is formulated for oral or parenteral administration.
[0150] In other embodiments, the present invention provides a method for treating cancer in a subject comprising administering to the subject an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more radiotherapeutic agents. In certain instances, the effective amount of the one or more RORy inhibitors and one or more statins is an amount sufficient to sensitize a radiation-resistant cancer such as a radiation-resistant breast cancer to radiation treatment. One or more RORy inhibitors, one or more statins, and radiotherapeutic agents can be delivered to a subject via the same route of administration (e.g, orally or parenterally) or via different routes of administration (e.g, intravenously for RORy inhibitors and/or statins and orally for radiotherapeutic agents, or vice versa).
4. Endocrine Therapies
[0151] Endocrine therapy is the manipulation of the endocrine system through the administration of specific hormones or drugs which inhibit or decrease the production or activity of targeted hormones or alter the gene expression pattern of targeted cells. Endocrine therapy is particularly useful in certain types of cancer, including breast cancer. Any known hormone antagonist or modulator may be used in the present invention. Endocrine therapies useful in the present invention include, but are not limited to, aromatase inhibitors (e.g. letrozole), megestrol acetate, flutamide, tamoxifen, raloxifene, lasofoxifene, bazedoxifene, bazedoxifene/conjugated estrogens, and combinations thereof.
[0152] In some embodiments, the present invention provides a composition comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more endocrine therapies. In certain instances, the composition further comprises a pharmaceutically acceptable excipient or diluent. In other instances, the composition is formulated for oral or parenteral administration.
[0153] In other embodiments, the present invention provides a method for treating cancer in a subject comprising administering to the subject an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more endocrine therapies. In certain instances, the effective amount of the one or more RORy inhibitors and one or more statins is an amount sufficient to sensitize an endocrine therapy-resistant cancer such as a tamoxifen -resistant breast cancer to endocrine therapy. RORy inhibitors, statins, and endocrine therapies can be delivered to a subject via the same route of administration ( e.g ., orally or parenterally) or via different routes of administration (e.g., intravenously for RORy inhibitors and/or statins and orally for endocrine therapies, or vice versa).
5. Tyrosine Kinase Inhibitors
[0154] Tyrosine kinase inhibitors are small-molecules that inhibit tyrosine kinase proteins. Tyrosine kinases are enzymes that activate many proteins in cellular signal transduction cascades by addition of a phosphate group to the protein. High expression and aberrant activation, of tyrosine kinase proteins can cause undesirable“switching on” of cellular signaling pathways that can result in uncontrolled cellular proliferation associated with cancerous cellular phenotypes. Various forms of cancer are currently treated by inhibiting or reducing the activity of poorly regulated tyrosine kinase proteins with tyrosine kinase inhibitors. Treatment regimens with tyrosine kinase inhibitors can suppress, reduce the incidence, reduce the severity, or inhibit the progression of cancer. Examples of tyrosine kinase inhibitors include, but are not limited to, gefitinib, erlotinib, sorafenib, sunitinib, dasatinib, lapatinib, nilotinib, bortezomib, salinomycin, and combinations thereof.
[0155] In some embodiments, the present invention provides a composition comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more tyrosine kinase inhibitors. In certain instances, the composition further comprises a pharmaceutically acceptable excipient or diluent. In other instances, the composition is formulated for oral or parenteral administration.
[0156] In other embodiments, the present invention provides a method for treating cancer in a subject comprising administering to the subject an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more tyrosine kinase inhibitors. In certain instances, the effective amount of the one or more RORy inhibitors and one or more statins is an amount sufficient to sensitize a tyrosine kinase inhibitor-resistant cancer such as a tyrosine kinase inhibitor-resistant non-small-cell lung cancer (NSCLC) to tyrosine kinase inhibitor therapy. RORy inhibitors, statins, and tyrosine kinase inhibitors can be delivered to a subject via the same route of administration ( e.g ., orally or parenterally) or via different routes of administration (e.g., intravenously for RORy inhibitors and/or statins and orally for tyrosine kinase inhibitors, or vice versa).
6. Antigen-Specific or Tumor Microenvironment-Targeting
Immunotherapeutic Agents
[0157] In some embodiments, antigen-specific immunotherapeutic agents include compounds and compositions designed to stimulate the immune system to specifically recognize antigens expressed or overexpressed by cancerous cells. In other embodiments, antigen-specific immunotherapeutic agents include compounds and compositions that will specifically recognize antigens expressed or overexpressed by cancerous ceils. In some embodiments, tumor microenvironment-targeting agents are used that may include, for example, compounds, antibodies, and other compositions that specifically recognize immune- modulating cytokines and suppress their tumor-promoting activities. Non-limiting examples of antigen-specific or tumor microenvironment-targeting immunotherapeutic agents include vaccines (e.g., peptide vaccines), antibodies, cytotoxic T cell lymphocytes (CTLs), chimeric antigen receptor T cells (CAR-T cells), immune checkpoints (e.g, CTLA-4, PD-l, and PD- Ll), antibodies against immune modulating cytokines (e.g, IL-6, IL-23 and IL-17), and combinations thereof. In particular embodiments, the antigens presented by cancerous cells are highly specific to each cancer type, and the vaccines, antibodies, CTLs, and/or CAR-T cells used is dependent on the cancer type being treated.
[0158] A vaccine can stimulate the immune system to specifically recognize and attack antigens presented by cancerous cells. Vaccines can comprise one or more peptides, peptide fragments, fusion peptides, DNA, RNA, other biologic or non-biologic material, or combinations thereof.
[0159] In some embodiments, one or more peptides, peptide fragments, or fusion peptides may be used for a peptide vaccine. The peptides may be harvested from an endogenous source or chemically synthesized. The peptides chosen are specific for the type of cancer being treated. For example, when targeting cancer cells, some commonly targeted proteins include GM-CSF, IL-l3Ra2, EphA2, and Survivin; however, specific cancer types will have specifically preferred peptides used for targeting afflicted cells. In some embodiments, the one or more peptides in the peptide vaccine are free soluble peptides. In other embodiments, the one or more peptides in the peptide vaccine are tethered together using any means known in the art.
[0160] In some embodiments, vaccines include cancer vaccines such as, e.g., tecemotide (L-BLP25), oncophage, sipuleucel-T, and combinations thereof. Tecemotide (L-BLP25) is a liposomal antigen-specific cancer immunotherapy that contains 25 amino acids from the immunogenic tandem-repeat region of MUC1 (see, e.g, Mehta NR el al. , Clin. Cancer Res., 18:2861-2871 (2012)).
[0161] Antibodies can recognize antigens expressed or overexpressed by cancerous cells Antigens recognized by these antibodies can be proteins expressed, activated, or overexpressed on the cell surface or proteins secreted into the extracellular fluid. In some embodiments, antibodies can be used to target human effector ceils (e.g., macrophages) against the cancerous cells. In some embodiments, antibodies are used to inhibit the normal function of cell surface receptors. In some embodiments, antibodies bind to the ligands of eeli surface receptors to block the cellular signaling cascade. Antibodies used as antigen- specific immunotherapeutic agents can be monoclonal or polyclonal antibodies as well as chimeric, humanized, or human antibodies, and can be previously isolated from the patient or produced from another biologic source. Methods of producing antibodies are well known in the art, and may be made by any known means. For example, antibodies described herein can be produced by conventional monoclonal antibody methodology e.g, the standard somatic cell hybridization technique of Kohler and Milstein, Nature 256: 495 (1975), the contents of which are herein incorporated by reference for all purposes. In some embodiments, antibodies useful in the treatment of cancer include immune checkpoint inhibitors. In particular embodiments, antibodies useful in the treatment of cancer include, but are not limited to, alemtuzumab. bevacizumab, cetuximab, ipilimumab, nivolumab, ofatumumab, panitumumab, pembrolizumab, atezolizumab, rituximab, trastuzumab, and combinations thereof.
[0162] The use of CTLs and CAR-T cells as antigen-specific immunotherapeutic agents is a form of adoptive T cell transfer therapy. Adoptive T cell transfer therapy is a technique that can boost the natural immune system’s ability to combat cancer by enriching for and/or designing T cells that are able to effectively recognize, bind, and kill a diseased cell. CTLs can recognize and bind cancerous cells using T-cell receptors (TCR). TCRs contain a highly variable binding region that allows them to recognize a large range of antigens. TCRs bind to the major histocompatibility complex I (MHC I) of cancerous cells presenting an appropriate antigen. TCR binding is highly specific, so only a small number of CTLs will be able to recognize a particular antigen. Once an antigen is recognized by CTLs binding to the MHC I complex of the cancerous cell, they activate to induce cellular death. Activated CTLs proliferate to fight the detected cancer.
[0163] CTLs administered in this therapy may be derived from the subject or may be derived from other biological sources. Methods for producing CTLs directed to a particular antigen are well known in the art, and can be harvested from an individual possessing a CTL directed to a particular antigen or produced outside of the body (ex vivo). For example, when treating cancer, cytotoxic T cells from a subject’s tumor are isolated, the cytotoxic T cells with the greatest antitumor activity are identified, the identified cytotoxic T cells are cultured to produce large amounts of the most effective cells, and the cultured cytotoxic T cells are reintroduced into the subject to treat the cancer. CTLs can also be produced in healthy individuals using ex vivo techniques described in U.S. Patent No. 5,962,318, and U.S. Patent Application Publication No. 2009/0324539, the contents of which are herein incorporated by reference for all purposes. The ex vivo methods described herein can be useful for individuals both before cancer onset or after cancer onset.
[0164] CAR-T cells are modified T cells which have been engineered to possess a cellular specificity domain that has not been produced naturally. The natural specificity domain of T cells are T-cell receptors that recognize a particular antigen presented on MHC class I molecules. In some embodiments, CAR-T cells possess a T-cell receptor that has not been naturally produced in a subject’s body. In some embodiments, the cellular specificity domain is a monoclonal antibody that is specific for the targeted cells or tissue. CAR-T cells can be produced using any means known in the art. In some embodiments, cytotoxic T cells are harvested from a subject’s blood, the cytotoxic T cells are genetically modified by inserting a gene that encodes for a receptor that recognizes an antigen specific to the cancer affecting the subject, the CAR-T cells are cultured and can be stored for later use or reintroduced into the subject’s body to treat the cancer. For more information on the details of producing CAR-T cells, see, e.g., U.S. Patent No. 9,102,760, U.S. Patent No. 8,399,645, U.S. Patent No. 8,975,071, and U.S. Patent No. 8,916,381, the contents of which are herein incorporated by reference for all purposes.
[0165] In some embodiments, the present invention provides a composition comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more antigen-specific immunotherapeutic agents. In certain instances, the composition further comprises a pharmaceutically acceptable excipient or diluent. In some instances, the composition is formulated for oral or parenteral administration.
[0166] In other embodiments, the present invention provides a method for treating cancer in a subject comprising administering to the subject an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) in combination with one or more antigen-specific immunotherapeutic agents. In certain instances, the effective amount of the one or more RORy inhibitors and one or more statins is an amount sufficient to sensitize a cancer that is resistant to treatment with antigen- specific immunotherapeutic agents to such treatment. RORy inhibitors, statins, and antigen- specific immunotherapeutic agents can be delivered to a subject via the same route of administration (e.g, orally or parenterally) or via different routes of administration (e.g, intravenously for RORy inhibitors and/or statins and orally for antigen-specific immunotherapeutic agents, or vice versa).
D. Diseases and Conditions
[0167] In certain aspects, a cancer can be treated or prevented by administering one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin). In some embodiments, the one or more RORy inhibitors and one or more statins are administered in combination with an anticancer drug. Cancer generally includes any of various malignant neoplasms characterized by the proliferation of anaplastic cells that tend to invade surrounding tissue and metastasize to new body sites. Non-limiting examples of different types of cancer suitable for treatment using the compositions of the present invention include breast cancer, prostate cancer (including drug- resistant prostate cancers, e.g., apalutamide-resistant prostate cancer), lung cancer, ovarian cancer, bladder cancer, thyroid cancer, liver cancer, pleural cancer, pancreatic cancer, cervical cancer, testicular cancer, colon cancer, anal cancer, bile duct cancer, gastrointestinal carcinoid tumors, esophageal cancer, gall bladder cancer, rectal cancer, appendix cancer, small intestine cancer, stomach (gastric) cancer, renal cancer ( i.e ., renal cell carcinoma), cancer of the central nervous system, skin cancer, choriocarcinomas, head and neck cancers, bone cancer, osteogenic sarcomas, fibrosarcoma, neuroblastoma, glioma, endometrial cancer, melanoma, leukemia (e.g., acute lymphocytic leukemia, chronic lymphocytic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, or hairy cell leukemia), lymphoma (e.g, non-Hodgkin's lymphoma, Hodgkin's lymphoma, B-cell lymphoma, or Burkitt's lymphoma), and multiple myeloma.
[0168] In particular embodiments, the cancer is an epithelial cancer (e.g, prostate cancer, ovarian cancer, breast cancer, and the like), or a blood cancer (e.g, leukemia, lymphoma, multiple myeloma). In some embodiments, the cancer is a prostate cancer. In certain embodiments, the prostate cancer is an advanced stage prostate cancer selected from one or more of metastatic prostate cancer, drug-resistant prostate cancer (e.g, anti -androgen- resistant prostate cancer such as enzalutamide-resistant prostate cancer, abiraterone-resistant prostate cancer, bicalutamide-resistant prostate cancer, etc.; taxane-resistant prostate cancer; docetaxel-resistant prostate cancer; and the like), hormone refractory prostate cancer, castration-resistant prostate cancer (CRPC), metastatic castration-resistant prostate cancer, AR-V7-induced drug-resistant prostate cancer such as AR-V7-induced anti-androgen- resistant prostate cancer (e.g, AR-V7-induced enzalutamide-resistant prostate cancer), AKR1C3 -induced drug-resistant prostate cancer such as AKR1C3 -induced anti-androgen- resistant prostate cancer (e.g, AKR1C3 -induced enzalutamide-resistant prostate cancer), and combinations thereof. In certain embodiments, the prostate cancer is an advanced stage prostate cancer selected from drug-resistant tumors with neuroendocrine (NE) phenotypes or NEPC. [0169] In other embodiments, the cancer is a lung cancer, breast cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma. In certain instances, the lung cancer is a non-small-cell lung cancer (NSCLC), K-Ras mutant lung cancer, BRAF mutant lung cancer, tyrosine kinase inhibitor-resistant lung cancer, small cell lung cancer (SCLC), adenocarcinoma ( e.g ., adenocarcinoma in situ), squamous cell carcinoma, large cell carcinoma, bronchial carcinoid, or combinations thereof. In certain instances, the breast cancer is triple-negative breast cancer (TNBC), tamoxifen- resistant breast cancer, radiation-resistant breast cancer, ductal carcinoma in situ , invasive ductal carcinoma, HER2-positive breast cancer, ER-positive breast cancer, inflammatory breast cancer, metastatic breast cancer, medullary carcinoma, tubular carcinoma, mucinous carcinoma (colloid), or combinations thereof. In certain instances, the liver cancer is a hepatocellular carcinoma (HCC), cholangiocarcinoma (bile duct cancer), angiosarcoma, hepatoblastoma, or combinations thereof. In certain instances, the glioma is an ependymoma, astrocytoma (e.g., glioblastoma multiforme), oligodendroglioma, brainstem glioma, optic nerve glioma, or combinations thereof (e.g, mixed glioma). In certain instances, the gastric (stomach) cancer is an adenocarcinoma of the distal esophagus, gastroesophageal junction and/or stomach, a gastrointestinal carcinoid tumor, a gastrointestinal stromal tumor, an associated lymphoma, a cancer linked to infection with H. pylori , or combinations thereof.
E. Pharmaceutical Compositions
[0170] The pharmaceutical compositions of the present invention encompass compositions made by admixing one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)), one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), and a pharmaceutically acceptable carrier and/or excipient or diluent. Such compositions are suitable for pharmaceutical use in an animal or a human.
[0171] In some embodiments, one or more RORy inhibitors (e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small -interfering RNA)) and/or one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) are present in a composition or kit of the present invention or are administered ( e.g ., to a subject) at a concentration of about 0.1 mM, 0.2 mM, 0.3 mM, 0.4 mM, 0.5 mM, 0.6 mM, 0.7 mM, 0.8 mM, 0.9 mM, 1 mM, 1.5 mM, 2 mM, 2.5 mM, 3 mM, 3.5 mM, 4 mM, 4.5 mM, 5 mM, 5.5 mM, 6 mM, 6.5 mM, 7 mM, 7.5 mM, 8 mM, 8.5 mM, 9 mM, 9.5 mM, 10 mM, 11 mM, 12 mM, 13 mM, 14 mM, 15 mM, 16 mM, 17 mM, 18 mM, 19 mM, 20 mM, 25 mM, 30 mM, 35 mM, 40 mM, 45 mM, 50 mM, 55 mM, 60 mM, 65 mM, 70 mM, 75 mM, 80 mM, 85 mM, 90 mM, 95 mM, or 100 mM.
[0172] The pharmaceutical compositions of the present invention may be prepared by any of the methods well-known in the art of pharmacy. Pharmaceutically acceptable carriers suitable for use with the present invention include any of the standard pharmaceutical carriers, buffers and excipients, including phosphate-buffered saline solution, water, and emulsions (such as an oil/water or water/oil emulsion), and various types of wetting agents and/or adjuvants. Suitable pharmaceutical carriers and their formulations are described in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, l9th ed. 1995). Preferred pharmaceutical carriers depend upon the intended mode of administration of the active agent(s).
[0173] The pharmaceutical compositions of the present invention can include one or more RORy inhibitors (e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) together with one or more anticancer drugs such as an anti-androgen drug (e.g, enzalutamide, abiraterone, and/or bicalutamide) and/or a chemotherapeutic agent (e.g, tamoxifen and/or a taxane such as docetaxel and cabazitaxel), or any pharmaceutically acceptable salts thereof, as an active ingredient, and a pharmaceutically acceptable carrier and/or excipient or diluent. In particular embodiments, the pharmaceutical composition can include one or more RORy inhibitors and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) together with an anti-androgen drug, such as enzalutamide. A pharmaceutical composition may optionally contain other therapeutic ingredients.
[0174] The compounds of the present invention can be combined as the active ingredient in intimate admixture with a suitable pharmaceutical carrier and/or excipient according to conventional pharmaceutical compounding techniques. Any carrier and/or excipient suitable for the form of preparation desired for administration is contemplated for use with the compounds disclosed herein.
[0175] In some embodiments, the pharmaceutical compositions comprising one or more RORy inhibitors (e.g., a compound of Formula I ( e.g ., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti -RORy antibody, and/or an interfering RNA (e.g., small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) and the pharmaceutical compositions comprising one or more anticancer drugs are prepared as a single medicament. In other embodiments, the pharmaceutical compositions comprising one or more RORy inhibitors and one or more statins and the pharmaceutical compositions comprising one or more anticancer drugs are prepared as separate medicaments.
[0176] The pharmaceutical compositions of the present invention include formulations suitable for topical, parenteral, pulmonary, nasal, rectal, or oral administration. The most suitable route of administration in any given case will depend in part on the nature and severity of the cancer condition and also optionally the stage of the cancer.
[0177] In embodiments where the one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small- interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) is administered in combination with an anticancer drug, the administration of the one or more RORy inhibitors and one or more statins and the anticancer drug may be administered using the same or a different administration route. For example, in some embodiments, the one or more RORy inhibitors and one or more statins and the anticancer drug may be administered orally or parenterally (e.g, intravenously). For example, in other embodiments, the one or more RORy inhibitors and one or more statins may be administered orally, while the anticancer drug may be administered parenterally (e.g, intravenously), or vice versa.
[0178] Other preferred compositions include compositions suitable for systemic (enteral or parenteral) administration. Systemic administration includes oral, rectal, sublingual, or sublabial administration. In some embodiments, the compositions may be administered via a syringe or intravenously. [0179] Compositions for pulmonary administration include, but are not limited to, dry powder compositions consisting of the powder of a compound described herein, or a salt thereof, and the powder of a suitable carrier and/or lubricant. The compositions for pulmonary administration can be inhaled from any suitable dry powder inhaler device known to a person skilled in the art.
[0180] Compositions for systemic administration include, but are not limited to, dry powder compositions consisting of the composition as set forth herein and the powder of a suitable carrier and/or excipient. The compositions for systemic administration can be represented by, but not limited to, tablets, capsules, pills, syrups, solutions, and suspensions.
[0181] In some embodiments, the present invention provides compositions further including a pharmaceutical surfactant. In other embodiments, the present invention provides compositions further including a cryoprotectant. In some embodiments, the cryoprotectant is selected from the group consisting of glucose, sucrose, trehalose, lactose, sodium glutamate, PVP, HPpCD, CD, glycerol, maltose, mannitol, and saccharose.
[0182] In some embodiments, the present invention provides a pharmaceutical composition including one or more RORy inhibitors (e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti- RORy antibody, and/or an interfering RNA (e.g., small-interfering RNA)), one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), and a pharmaceutically acceptable excipient. In some embodiments, the present invention provides a pharmaceutical composition including one or more RORy inhibitors, one or more statins, and one or more anticancer drugs such as an anti-androgen drug (e.g, enzalutamide, abiraterone, and/or bicalutamide) and/or a chemotherapeutic agent (e.g, tamoxifen and/or a taxane such as docetaxel), in combination with a pharmaceutically acceptable excipient. In particular embodiments, the present invention provides a pharmaceutical composition including one or more RORy inhibitors, one or more statins, and an anti-androgen drug, such as enzalutamide, in combination with a pharmaceutically acceptable excipient. In some of these embodiments, the pharmaceutically acceptable excipient includes a salt or a diluent.
[0183] In some embodiments, the present invention provides compositions including an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti-RORy antibody, and/or an interfering RNA (e.g., small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin). In some embodiments, the composition is formulated for oral administration or parenteral (e.g, intravenous) administration and includes one or more RORy inhibitors, one or more statins, and at least one member selected from the group consisting of an aqueous solution and a buffer solution. In some embodiments, the composition can include an effective amount of one or more RORy inhibitors, one or more statins, and one or more anticancer drugs such as an anti androgen drug (e.g, enzalutamide, abiraterone, and/or bicalutamide) and/or a chemotherapeutic agent (e.g, tamoxifen and/or a taxane such as docetaxel).
[0184] Pharmaceutical compositions or medicaments for use in the present invention can be formulated by standard techniques using one or more physiologically acceptable carriers or excipients. Suitable pharmaceutical carriers are described herein and in Remington: The Science and Practice of Pharmacy, 2lst Ed., University of the Sciences in Philadelphia, Lippencott Williams & Wilkins (2005).
[0185] For oral administration, a pharmaceutical composition or a medicament can take the form of, e.g, a tablet or a capsule prepared by conventional means with a pharmaceutically acceptable excipient. Preferred are tablets and gelatin capsules comprising the active ingredient(s), together with (a) diluents or fillers, e.g. , lactose, dextrose, sucrose, mannitol, sorbitol, cellulose (e.g., ethyl cellulose, microcrystalline cellulose), glycine, pectin, polyacrylates and/or calcium hydrogen phosphate, calcium sulfate, (b) lubricants, e.g, silica, anhydrous colloidal silica, talcum, stearic acid, its magnesium or calcium salt (e.g, magnesium stearate or calcium stearate), metallic stearates, colloidal silicon dioxide, hydrogenated vegetable oil, corn starch, sodium benzoate, sodium acetate and/or polyethyleneglycol; for tablets also (c) binders, e.g, magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, polyvinylpyrrolidone and/or hydroxypropyl methylcellulose; if desired (d) disintegrants, e.g, starches (e.g, potato starch or sodium starch), glycolate, agar, alginic acid or its sodium salt, or effervescent mixtures; (e) wetting agents, e.g, sodium lauryl sulfate, and/or (f) absorbents, colorants, flavors and sweeteners. In some embodiments, the tablet contains a mixture of hydroxypropyl methylcellulose, polyethyleneglycol 6000 and titatium dioxide. Tablets may be either film coated or enteric coated according to methods known in the art. [0186] Liquid preparations for oral administration can take the form of, for example, solutions, syrups, or suspensions, or they can be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives, for example, suspending agents, for example, sorbitol syrup, cellulose derivatives, or hydrogenated edible fats; emulsifying agents, for example, lecithin or acacia; non-aqueous vehicles, for example, almond oil, oily esters, ethyl alcohol, or fractionated vegetable oils; and preservatives, for example, methyl or propyl-p-hydroxybenzoates or sorbic acid. The preparations can also contain buffer salts, flavoring, coloring, and/or sweetening agents as appropriate. If desired, preparations for oral administration can be suitably formulated to give controlled release of the active compound.
[0187] Controlled release parenteral formulations of the compositions of the present invention can be made as implants, oily injections, or as particulate systems. For a broad overview of delivery systems see, Banga, A.J., THERAPEUTIC PEPTIDES AND PROTEINS: FORMULATION, PROCESSING, AND DELIVERY SYSTEMS, Technomic Publishing Company, Inc., Lancaster, PA, (1995) incorporated herein by reference. Particulate systems include microspheres, microparticles, microcapsules, nanocapsules, nanospheres, and nanoparticles.
[0188] Polymers can be used for ion-controlled release of compositions of the present invention. Various degradable and nondegradable polymeric matrices for use in controlled drug delivery are known in the art (Langer R., Accounts Chem. Res., 26:537-542 (1993)). For example, the block copolymer, polaxamer 407 exists as a viscous yet mobile liquid at low temperatures but forms a semisolid gel at body temperature. It has shown to be an effective vehicle for formulation and sustained delivery of recombinant interleukin 2 and urease (Johnston et ak, Pharm. Res., 9:425-434 (1992); and Pec et ah, ./. Parent. Sci. Tech., 44(2):58 65 (1990)). Alternatively, hydroxyapatite has been used as a microcarrier for controlled release of proteins (Ijntema et ak, Int. J Pharm., 112:215-224 (1994)). In yet another aspect, liposomes are used for controlled release as well as drug targeting of the lipid-capsulated drug (Betageri et ak, LIPOSOME DRUG DELIVERY SYSTEMS, Technomic Publishing Co., Inc., Lancaster, PA (1993)). Numerous additional systems for controlled delivery of therapeutic proteins are known. See, e.g., U.S. Pat. No. 5,055,303, 5,188,837, 4,235,871, 4,501,728, 4,837,028 4,957,735 and 5,019,369, 5,055,303; 5,514,670; 5,413,797; 5,268,164; 5,004,697; 4,902,505; 5,506,206, 5,271,961; 5,254,342 and 5,534,496, each of which is incorporated herein by reference.
F. Methods of Administration
[0189] Pharmaceutical compositions or medicaments comprising one or more RORy inhibitors (e.g., a compound of Formula I ( e.g ., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti -RORy antibody, and/or an interfering RNA (e.g., small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) can be administered to a subject at a therapeutically effective dose to treat the subject’s cancer, as described herein. In some embodiments, pharmaceutical compositions or medicaments comprising one or more RORy inhibitors and one or more statins can be co- administered to a subject in combination with an effective amount of an anticancer drug at a therapeutically effective dose to treat the subject’s cancer, as described herein. In some embodiments, the pharmaceutical composition or medicament comprising one or more RORy inhibitors and one or more statins is administered to a subject in an amount sufficient to elicit an effective therapeutic response in the subject. In some embodiments, the pharmaceutical composition or medicament comprising one or more RORy inhibitors and one or more statins can be co-administered to a subject at a therapeutically effective dose in combination with an effective amount of an anticancer drug to elicit an effective therapeutic response in the subject.
[0190] In certain methods of treating cancer, set forth herein, the methods comprise first administering one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti- RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) to a patient having cancer, and then administering an anticancer drug, such as an anti-androgen drug and/or a chemotherapeutic agent, to the patient. In certain methods of treating cancer, set forth herein, the methods comprise first administering an anticancer drug, such as an anti-androgen drug and/or a chemotherapeutic agent, to a patient having cancer, and then administering one or more RORy inhibitors and one or more statins to the patient. In certain methods of treating cancer, set forth herein, the methods comprise co-administering one or more RORy inhibitors and one or more statins with an anticancer drug, such as an anti-androgen drug and/or a chemotherapeutic agent, to a patient having cancer.
[0191] In some embodiments, the methods of administration comprise administering one or more RORy inhibitors (e.g., a compound of Formula I ( e.g ., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g., small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), alone or in combination with enzalutamide to a patient in need thereof (e.g, a patient in need of cancer treatment). In other embodiments, the methods of administration comprise administering one or more RORy inhibitors and one or more statins, alone or in combination with abiraterone to a patient in need thereof. In yet other embodiments, the methods comprise administering one or more RORy inhibitors and one or more statins, alone or in combination with bicalutamide to a patient in need thereof. In still yet other embodiments, the methods comprise administering one or more RORy inhibitors and one or more statins, alone or in combination with a taxane such as docetaxel to a patient in need thereof. In further embodiments, the methods comprise administering one or more RORy inhibitors and one or more statins, alone or in combination with tamoxifen to a patient in need thereof.
[0192] In certain embodiments, the present invention provides a method of delivering an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) to a patient having cancer such as prostate cancer (e.g, CRPC).
[0193] The RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) described herein are useful in the manufacture of a pharmaceutical composition or a medicament. A pharmaceutical composition or medicament can be administered to a subject in need thereof, e.g. a patient having a cancer such as breast cancer, prostate cancer ( e.g ., CRPC), lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma.
[0194] Pharmaceutical compositions or medicaments for use in the present invention can be formulated by standard techniques using one or more physiologically acceptable carriers or excipients. Suitable pharmaceutical carriers are described herein and in "Remington's Pharmaceutical Sciences" by E.W. Martin. Compounds and agents of the present invention and their physiologically acceptable salts and solvates can be formulated for administration by any suitable route, including via inhalation, topically, nasally, orally, intravenously, parenterally, or rectally.
1. Routes of Administration
[0195] Typical formulations for topical administration include creams, ointments, sprays, lotions, and patches. The pharmaceutical composition can, however, be formulated for any type of administration, e.g., intradermal, subdermal, intravenous, intramuscular, intranasal, intracerebral, intratracheal, intraarterial, intraperitoneal, intravesical, intrapleural, intracoronary or intratumoral injection, with a syringe or other devices. Formulation for administration by inhalation (e.g, aerosol), or for oral or rectal administration is also contemplated.
[0196] Suitable formulations for transdermal application include an effective amount of one or more compounds described herein, optionally with a carrier. Preferred carriers include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host. For example, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin. Matrix transdermal formulations may also be used.
[0197] For oral administration, a pharmaceutical composition or a medicament can take the form of, for example, a tablet or a capsule prepared by conventional means with a pharmaceutically acceptable excipient. The present invention provides tablets and gelatin capsules comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), alone or in combination with other compounds such as anti-androgen drugs and/or docetaxel, or a dried solid powder of these drugs, together with (a) diluents or fillers, e.g ., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose (e.g., ethyl cellulose, microcrystalline cellulose), glycine, pectin, polyacrylates and/or calcium hydrogen phosphate, calcium sulfate, (b) lubricants, e.g, silica, talcum, stearic acid, magnesium or calcium salt, metallic stearates, colloidal silicon dioxide, hydrogenated vegetable oil, corn starch, sodium benzoate, sodium acetate and/or polyethyleneglycol; for tablets also (c) binders, e.g, magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, polyvinylpyrrolidone and/or hydroxypropyl methylcellulose; if desired (d) disintegrants, e.g, starches (e.g, potato starch or sodium starch), glycolate, agar, alginic acid or its sodium salt, or effervescent mixtures; (e) wetting agents, e.g, sodium lauryl sulphate, and/or (f) absorbents, colorants, flavors and sweeteners.
[0198] Tablets may be either film coated or enteric coated according to methods known in the art. Liquid preparations for oral administration can take the form of, for example, solutions, syrups, or suspensions, or they can be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives, for example, suspending agents, for example, sorbitol syrup, cellulose derivatives, or hydrogenated edible fats; emulsifying agents, for example, lecithin or acacia; non-aqueous vehicles, for example, almond oil, oily esters, ethyl alcohol, or fractionated vegetable oils; and preservatives, for example, methyl or propyl-p-hydroxybenzoates or sorbic acid. The preparations can also contain buffer salts, flavoring, coloring, and/or sweetening agents as appropriate. If desired, preparations for oral administration can be suitably formulated to give controlled release of the active compound(s).
[0199] The compositions and formulations set forth herein can be formulated for parenteral administration by injection, for example by bolus injection or continuous infusion. Formulations for injection can be presented in unit dosage form, for example, in ampoules or in multi-dose containers, with an added preservative. Injectable compositions are preferably aqueous isotonic solutions or suspensions, and suppositories are preferably prepared from fatty emulsions or suspensions. The compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. Alternatively, the active ingredient(s) can be in powder form for constitution with a suitable vehicle, for example, sterile pyrogen-free water, before use. In addition, they may also contain other therapeutically valuable substances. The compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1 to 75%, preferably about 1 to 50%, of the active ingredient(s).
[0200] For administration by inhalation, the compositions of the present invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, for example, dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide, or other suitable gas. In the case of a pressurized aerosol, the dosage unit can be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, for example, gelatin for use in an inhaler or insufflator can be formulated containing a powder mix of the compound(s) and a suitable powder base, for example, lactose or starch.
[0201] The compositions set forth herein can also be formulated in rectal compositions, for example, suppositories or retention enemas, for example, containing conventional suppository bases, for example, cocoa butter or other glycerides.
[0202] Furthermore, the active ingredient(s) can be formulated as a depot preparation. Such long-acting formulations can be administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, one or more of the compounds described herein can be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
[0203] In particular embodiments, a pharmaceutical composition or medicament of the present invention can comprise (i) an effective amount of one or more RORy inhibitors (e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), and (ii) optionally an anticancer drug such as an anti-androgen drug (e.g, enzalutamide, abiraterone, bicalutamide), a chemotherapeutic agent such as a taxane (e.g, docetaxel) or tamoxifen, and combinations thereof. The therapeutic agent(s) may be used individually, sequentially, or in combination with one or more other such therapeutic agents (e.g., a first therapeutic agent, a second therapeutic agent, a compound of the present invention, etc.). Administration may be by the same or different route of administration or together in the same pharmaceutical formulation.
2. Dosage
[0204] Pharmaceutical compositions or medicaments can be administered to a subject at a therapeutically effective dose to prevent, treat, sensitize, or control a cancer such as prostate cancer ( e.g ., CRPC), lung cancer, breast cancer (e.g, TNBC), liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma as described herein. The pharmaceutical composition or medicament is administered to a subject in an amount sufficient to elicit an effective therapeutic response in the subject. An effective therapeutic response includes a response that at least partially arrests or slows the symptoms or complications of the cancer. An amount adequate to accomplish this is defined as a“therapeutically effective dose.”
[0205] The dosage of active agents administered is dependent on the subject’s body weight, age, individual condition, surface area or volume of the area to be treated and on the form of administration. The size of the dose also will be determined by the existence, nature, and extent of any adverse effects that accompany the administration of a particular formulation in a particular subject. A unit dosage for oral administration to a mammal of about 50 to about 70 kg may contain between about 5 and about 500 mg, about 25-200 mg, about 100 and about 1000 mg, about 200 and about 2000 mg, about 500 and about 5000 mg, or between about 1000 and about 2000 mg of the active ingredient. A unit dosage for oral administration to a mammal of about 50 to about 70 kg may contain about 10 mg, 20 mg, 25 mg, 50 mg, 75 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 700 mg, 800 mg, 900 mg, 1,000 mg, 1,250 mg, 1,500 mg, 2,000 mg, 2,500 mg, 3,000 mg, or more of the active ingredient. Typically, a dosage of the active compound(s) of the present invention is a dosage that is sufficient to achieve the desired effect. Optimal dosing schedules can be calculated from measurements of active agent accumulation in the body of a subject. In general, dosage may be given once or more of daily, weekly, or monthly. Persons of ordinary skill in the art can easily determine optimum dosages, dosing methodologies and repetition rates.
[0206] Optimum dosages, toxicity, and therapeutic efficacy of the compositions of the present invention may vary depending on the relative potency of the administered composition and can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, for example, by determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio, LD50/ED50. Agents that exhibit large therapeutic indices are preferred. While agents that exhibit toxic side effects can be used, care should be taken to design a delivery system that targets such agents to the site of affected tissue to minimize potential damage to normal cells and, thereby, reduce side effects.
[0207] Optimal dosing schedules can be calculated from measurements of active ingredient accumulation in the body of a subject. In general, dosage is from about 1 ng to about 1,000 mg per kg of body weight and may be given once or more daily, weekly, monthly, or yearly. Persons of ordinary skill in the art can easily determine optimum dosages, dosing methodologies and repetition rates. One of skill in the art will be able to determine optimal dosing for administration of a RORy inhibitor (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti- RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and a statin (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) to a human being following established protocols known in the art and the disclosure herein.
[0208] The data obtained from, for example, animal studies (e.g, rodents and monkeys) can be used to formulate a dosage range for use in humans. The dosage of compounds of the present invention lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage can vary within this range depending upon the dosage form employed and the route of administration. For any composition for use in the methods of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma can be measured, for example, by high performance liquid chromatography (HPLC). In general, the dose equivalent of a chimeric protein, preferably a composition is from about 1 ng/kg to about 100 mg/kg for a typical subject. [0209] A typical composition of the present invention for oral or intravenous administration can be about 0.1 mg to about 10 mg of active ingredient per patient per day; about 1 mg to about 100 mg per patient per day; about 25 mg to about 200 mg per patient per day; about 50 mg to about 500 mg per patient per day; about 100 mg to about 1000 mg per patient per day; or about 1000 mg to about 2000 mg per patient per day. Exemplary dosages include, but are not limited to, about 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 21 mg, 22 mg, 23 mg, 24 mg, 25 mg, 26 mg, 27 mg, 28 mg, 29 mg, 30 mg, 31 mg, 32 mg, 33 mg, 34 mg, 35 mg, 36 mg, 37 mg, 38 mg, 39 mg, 40 mg, 41 mg, 42 mg, 43 mg, 44 mg, 45 mg, 46 mg, 47 mg, 48 mg, 49 mg, 50 mg, 55 mg, 60, mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 700 mg, 800 mg, 900 mg, 1,000 mg, 1,250 mg, 1,500 mg, 2,000 mg, 2,500 mg, 3,000 mg, or more of the active ingredient per patient per day. Methods for preparing administrable compositions will be known or apparent to those skilled in the art and are described in more detail in such publications as Remington: The Science and Practice of Pharmacy, 2 Ist Ed., ETniversity of the Sciences in Philadelphia, Lippencott Williams & Wilkins (2005).
[0210] Exemplary doses of the compositions described herein include milligram or microgram amounts of the composition per kilogram of subject or sample weight ( e.g. , about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 5 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram). It is furthermore understood that appropriate doses of a composition depend upon the potency of the composition with respect to the desired effect to be achieved. When one or more of these compositions is to be administered to a mammal, a physician, veterinarian, or researcher may, for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained. In addition, it is understood that the specific dose level for any particular mammal subject will depend upon a variety of factors including the activity of the specific composition employed, the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate of excretion, any drug combination, and the degree of expression or activity to be modulated.
[0211] In some embodiments, a pharmaceutical composition or medicament of the present invention is administered, e.g. , in a daily dose in the range from about 1 mg of compound per kg of subject weight (1 mg/kg) to about lg/kg. In another embodiment, the dose is a dose in the range of about 5 mg/kg to about 500 mg/kg. In yet another embodiment, the dose is about 10 mg/kg to about 250 mg/kg. In another embodiment, the dose is about 25 mg/kg to about 150 mg/kg. A preferred dose is about 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 18, 20, 25, 30, 40, or 50 mg/kg. The daily dose can be administered once per day or divided into subdoses and administered in multiple doses, e.g., twice, three times, or four times per day. However, as will be appreciated by a skilled artisan, compositions described herein may be administered in different amounts and at different times. The skilled artisan will also appreciate that certain factors may influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the disease or malignant condition, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of a composition can include a single treatment or, preferably, can include a series of treatments.
[0212] To achieve the desired therapeutic effect, compounds or agents described herein may be administered for multiple days at the therapeutically effective daily dose. Thus, therapeutically effective administration of compounds to treat prostate cancer in a subject may require periodic (e.g, daily) administration that continues for a period ranging from three days to two weeks or longer. Compositions set forth herein may be administered for at least three consecutive days, often for at least five consecutive days, more often for at least ten, and sometimes for 20, 30, 40 or more consecutive days. While consecutive daily doses are a preferred route to achieve a therapeutically effective dose, a therapeutically beneficial effect can be achieved even if the agents are not administered daily, so long as the administration is repeated frequently enough to maintain a therapeutically effective concentration of the agents in the subject. For example, one can administer the agents every other day, every third day, or, if higher dose ranges are employed and tolerated by the subject, once a week.
[0213] In some embodiments, the RORy inhibitor (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small -interfering RNA)) and/or statin (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin) are orally administered. In some embodiments, the RORy inhibitor and/or statin is orally administered to a subject (e.g, an adult human) at a daily dose of approximately 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 21 mg, 22 mg, 23 mg, 24 mg, 25 mg, 26 mg, 27 mg, 28 mg, 29 mg, 30 mg, 31 mg, 32 mg, 33 mg, 34 mg, 35 mg, 36 mg, 37 mg, 38 mg, 39 mg, 40 mg, 41 mg, 42 mg, 43 mg, 44 mg, 45 mg, 46 mg, 47 mg, 48 mg, 49 mg, 50 mg, 55 mg, 60, mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg,
200 mg, 225 mg, 250 mg, 275mg, 300, 325 mg, 350 mg, 375 mg, 400 mg, 500 mg, 600 mg,
700 mg, 800 mg, 900 mg, 1,000 mg, 1,250 mg, 1,500 mg, 1,750 mg, 2,000 mg, 2,500 mg, 3,000 mg, 3,500 mg, 4,000 mg, 4,500 mg, 5,000 mg or more per day. In some embodiments, the RORy inhibitor and/or statin are orally administered to a subject (e.g, an adult human) at a daily dose of between about 1,000 mg and about 2,000 mg per day. In some embodiments, the RORy inhibitor and/or statin is orally administered. In some embodiments, the RORy inhibitor and/or statin are orally administered to a subject (e.g, an adult human) at a daily dose of approximately 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 21 mg, 22 mg, 23 mg, 24 mg, 25 mg, 26 mg, 27 mg, 28 mg, 29 mg, 30 mg, 31 mg, 32 mg, 33 mg, 34 mg, 35 mg, 36 mg, 37 mg, 38 mg, 39 mg, 40 mg, 41 mg, 42 mg, 43 mg, 44 mg, 45 mg, 46 mg, 47 mg, 48 mg, 49 mg, 50 mg, 55 mg, 60, mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg,
200 mg, 225 mg, 250 mg, 275mg, 300, 325 mg, 350 mg, 375 mg, 400 mg, 500 mg or more per day. In some embodiments, RORy inhibitor and/or statin are orally administered to a subject (e.g, an adult human) at a daily dose of between 25 and 200 mg per day. In some embodiments, the RORy inhibitor and/or statin and an anti-androgen drug are orally co- administered. For example, the RORy inhibitor and/or statin can be co-administered at a daily oral dose of between about 25 mg and 1000 mg mg per day with the anti -androgen drug at a daily oral dose of between about 25 mg and 2000 mg per day.
[0214] In some embodiments, the methods comprise sequentially administering one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), followed by one or more anticancer drugs such as an anti androgen drug (e.g, enzalutamide, abiraterone, bicalutamide), a chemotherapeutic agent such as a taxane (e.g, docetaxel) or tamoxifen, and combinations thereof. In some embodiments, the methods comprise sequentially administering one or more anticancer drugs followed by one or more RORy inhibitors and one or more statins.
[0215] Following successful treatment, it may be desirable to have the subject undergo maintenance therapy to prevent the recurrence of the cancer.
[0216] Determination of an effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. Generally, an efficacious or effective amount of an composition is determined by first administering a low dose or small amount of the composition, and then incrementally increasing the administered dose or dosages, adding a second or third medication as needed, until a desired effect of is observed in the treated subject with minimal or no toxic side effects.
[0217] Single or multiple administrations of the compositions are administered depending on the dosage and frequency as required and tolerated by the patient. In any event, the composition should provide a sufficient quantity of the compositions of this invention to effectively treat the patient. Generally, the dose is sufficient to treat or ameliorate symptoms or signs of disease without producing unacceptable toxicity to the patient.
G. Kits, Containers, Devices, and Systems
[0218] A wide variety of kits and systems can be prepared according to the present invention, depending upon the intended user of the kit and system and the particular needs of the user. In some embodiments, the present invention provides a kit that includes ( e.g ,., an effective amount of) one or more RORy inhibitors (e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin). In other aspects, the present invention provides a kit that includes one or more RORy inhibitors, one or more statins, and one or more anticancer drugs such as an anti-androgen drug (e.g, enzalutamide, abiraterone, and/or bicalutamide) and/or a chemotherapeutic agent (e.g, tamoxifen and/or a taxane such as docetaxel).
[0219] Some of the kits described herein can include a label describing a method of administering one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti- RORy antibody, and/or an interfering RNA (e.g., small-interfering RNA)), one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, and/or cerivastatin, pravastatin, pitavastatin), and/or one or more anticancer drugs (e.g, to a subject, such as a subject in need of cancer treatment). Some of the kits described herein can include a label describing a method of treating cancer in a subject with a cancer such as breast cancer (e.g, TNBC), prostate cancer (e.g, CRPC), lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma.
[0220] The compositions of the present invention, including but not limited to, compositions comprising one or more RORy inhibitors (e.g., a compound of Formula I (e.g, XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP- 43742, an anti -RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, and/or pitavastatin), and optionally one or more anticancer drugs may, if desired, be presented in a bottle, jar, vial, ampoule, tube, or other container-closure system approved by the Food and Drug Administration (FDA) or other regulatory body, which may provide one or more dosages containing the compounds. The package or dispenser may also be accompanied by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, the notice indicating approval by the agency. In certain aspects, the kit may include a formulation or composition as described herein, a container closure system including the formulation or a dosage unit form including the formulation, and a notice or instructions describing a method of use as described herein.
[0221] In some embodiments, the kit includes a container which is compartmentalized for holding the various elements of a formulation (e.g., the dry ingredients and the liquid ingredients) or composition, instructions for making the formulation or composition, and instructions for administering the formulation or composition for enhancing the immune response in a subject with a cancer.
[0222] In certain embodiments, the kit may include the pharmaceutical preparation(s) in dehydrated or dry form, with instructions for its rehydration (or reconstitution) and administration.
[0223] Kits with unit doses of the compounds described herein, e.g. in oral, rectal, transdermal, or injectable doses (e.g, for intramuscular, intravenous, or subcutaneous injection), are provided. In such kits, an informational package insert describing the use and attendant benefits of the composition for enhancing the immune response in a subject with a cancer such as prostate cancer ( e.g ., CRPC), lung cancer, breast cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, or a glioma may be included in addition to the containers containing the unit doses.
[0224] Some embodiments of the present invention provide packages that include one or more RORy inhibitors (e.g., a compound of Formula I (e.g., XY018, XY063), GSK805, SR2211, and/or a compound listed in Table 5 such as VTP-43742, an anti-RORy antibody, and/or an interfering RNA (e.g, small-interfering RNA)) and one or more statins (e.g, atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, cerivastatin, pravastatin, and/or pitavastatin), and optionally one or more anticancer drugs such as an anti androgen drug (e.g, enzalutamide, abiraterone, and/or bicalutamide) and/or a chemotherapeutic agent (e.g, tamoxifen and/or a taxane such as docetaxel).
IV. EXAMPLES
[0225] The present invention will be described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the invention in any manner. Those of skill in the art will readily recognize a variety of noncritical parameters which can be changed or modified to yield essentially the same results.
EXAMPLE 1 Targeting tumor metabolism with a combination of RQRY inhibitors and statins as a new strategy for effective treatment of therapeutic resistant cancers
Introduction
[0226] Metabolic reprogramming, a hallmark of cancer, fuels the malignant tumor growth and survival by providing energy, reducing power and building blocks. One metabolic process that is often deregulated in tumors of most cancer types is the mevalonate (MV A) pathway which produces sterols such as cholesterol, isoprenoids, and ubiquinone that are essential for tumor growth (4). The major regulators of the MVA pathway enzyme expression are the transcription factors sterol regulatory element-binding protein 1 and 2 (SREBP1 and -2) and LXRs (2, 5). SREBPs play a pivotal role in the maintenance of cholesterol homeostasis. In response to a low intracellular sterol level, SREBPs dissociate from the INSIGs proteins at the endoplasmic reticulum (ER) and translocate from the ER to the Golgi where they are cleaved by proteases. The cleaved N-terminal portion of SREBPs translocate to the nucleus and bind to target gene promoters at SRE sites to activate the expression of MVA pathway and cholesterol synthesis enzymes such as HMGCS, HMGCR and MVK for de novo cholesterol synthesis, as well as genes involved in cholesterol homeostasis such as low density lipoprotein receptor (LDLR), which facilitates the cellular uptake of exogenous lipoprotein-derived cholesterol. Upon the increase in cellular cholesterol level, the associations of SREBPs with INSIGs at the ER are stabilized and SREBP activation of gene expression is decreased.
[0227] Statins are a class of cholesterol/lipid-lowering drugs for treating patients with hypercholesterolemia to reduce the associated high risk of cardiovascular disease (CVD) (1). Statins are small-molecule inhibitors of 3 -hydroxy-3 -methyl glutary coenzyme A reductase (HMGCR), a rate-limiting enzyme in hepatic cholesterol biosynthesis. Statin treatment strongly reduces cholesterol production initially, and, as an adaptive response, the reduced cellular cholesterol level triggers the SREBP-mediated activation of gene expression including that of LDLR in liver and other tissues, which leads to an increased uptake of LDL from the circulation, hence lowering blood cholesterol level. Interestingly, epidemiological studies comparing cancer patients receiving statins for their CVD risk to those not receiving statins suggest that the use of statins is associated with a statistically significant reduction of the risk of cancer relapse and cancer-related death. Such findings have prompted many clinical trials of statins as cancer therapeutics in patients with different types of cancers including lung cancer, prostate cancer and breast cancer. However, thus far no clinical trial has shown a clear and sustained benefit from treatment with statins in the cancer patients.
[0228] On the other hand, preclinical studies using in vitro cell culture have demonstrated that treatment of cancer cells with high concentrations of statins can result in inhibition of cell growth and cell death (1). However, in animal models, the tumor inhibition efficacy of statins, as a single agent or in combination with other drugs, is unclear for most cancer types. Although a few animal studies may have shown tumor inhibitory effects, the data were obtained by using very high doses of statins which are deemed unsafe and unsuitable for clinical use in cancer patients. Therefore, currently the potential value of statin use in cancer prevention and treatment is unclear.
[0229] Recent preclinical and clinical studies suggest that statin treatment can lead to increased expression of MVA pathway genes. Due to the oncogenic regulation of SREBPs and the MVA pathway, cancer cells may have a heightened cholesterol homeostasis response mediated by SREBPs to maintain the levels of MVA pathway products following statin treatment (4). Therefore, the development of strategies to combine statins with therapeutics that block the heightened homeostasis such as inhibitors of SREBPs can be of high value in the effective treatment of many types of cancer.
[0230] We recently found that small-molecule inhibitors or genetic silencing of a member of the nuclear receptor family protein, RORy, strongly inhibit the growth of cancer cell and xenograft tumors and induce marked cell death in prostate cancer (6). Here, we report that genetic silencing of RORy resulted in strong inhibition of growth and survival of breast cancer and lung cancer cells. Targeting RORy with its inhibitors also strongly inhibited the growth and survival of several types of cancer cells. Because of the apparent function of RORy in many tumor types, we postulated that RORy might control a pathway or process common to most cancer types. Indeed, we found that genes of the MVA-cholesterol pathway are strongly affected by the RORy inhibitors and that their inhibitory effects on cancer cell growth can be rescued by supplying cells with exogenous cholesterol. More importantly, we found that the RORy inhibitors effectively sensitize cancer cells to statin-induced cell death and growth inhibition and that a combination treatment with the RORy inhibitors and statins resulted in highly synergistic killing of cancer cells of multiple cancer types.
Results
RORy is required for cancer cell growth and survival of different cancer types
[0231] We previously found that RORy plays an important function in prostate cancer. To examine whether RORy plays a role in other cancer types, we performed genetic deletion/knockout of the RORy gene RORC using CRISPR-Cas9 or genetic silencing/knockdown of RORC expression using siRNA. As shown in FIG. 1 A, knockout of the RORy gene by two different RORC sgRNA-directed CRISPR-Cas9 constructs, but not the control sgGFP construct, markedly inhibited the growth of triple-negative breast cancer (TNBC) cells HCC70 and MDA-MB468. As shown in FIG. 1B, Western blotting analysis demonstrated that the knockout caused a significant decrease in RORy protein expression in the two cells, a decrease of anti-apoptotic protein MCL-l expression and an increase of cleaved PARP-l, a marker of apoptotic cell death. As shown in FIG. 1C, siRNA silencing of RORC expression also significantly inhibited the growth of the breast cancer cells. As shown in FIG. 1D, similar to the effects seen in RORC knockout cells, the siRNA silencing resulted in decreased expression of RORy and MCL-l and an increased level of cleaved PARP-l. The strong silencing also led to significant decreased expression of proteins that are important for cell proliferation, growth and survival, such as ANCCA/ATAD2, c-MYC, CDK4, Cyclin Dl, and BCL-2. Moreover, as shown in FIG. 2, RORC knockout caused similar growth inhibition in other TNBC cell lines including SUM159, MB436, Hs578T and BT549. Interestingly, RORC knockout did not significantly affect the growth of MCF-10A, a non-malignant human breast epithelial cell line, suggesting that RORy does not play a significant role in growth and survival of normal or non-malignant cells. To explore further the function of RORy, we performed siRNA knockdown in lung cancer cells. As shown in FIG. 3, silencing of RORC with the two siRNAs markedly inhibited the growth of A549 and H460 cells. The two lung cancer cell lines express mutant KRAS and are insensitive to EGFR inhibitor drugs.
Inhibition of RORy suppresses the expression of senes controlling cholesterol synthesis
[0232] The fact that RORy is required for the growth and survival of multiple different types of cancer cells prompted us to examine mechanisms that are common to the different cancers. Members of the nuclear receptor family such as PPARs and LXRs play important functions in the control of lipid metabolism in cancers. Studies with RORy gene knockout mice showed that RORy regulates glucose and lipid metabolism in mouse liver and other tissues (3). We thus interrogated the gene expression profiling data we recently published in our study of RORy in prostate cancer (6), for major pathways or processes altered by RORy inhibition. In addition to the androgen receptor (AR) pathway we reported (6), we identified cholesterol synthesis and homeostasis as another major pathway affected by RORy inhibition.
[0233] As shown in FIG. 4A, the cholesterol synthesis pathway involves more than 20 different steps of enzymatic activities, with steps carried out by HMGCR and SQLE being the rate-limiting steps in the pathway. HMGCR, in particular, is the target of statins, a class of cholesterol/lipid-lowering drugs for treating patients with hypercholesterolemia, in order to reduce their high risk of cardiovascular diseases. As shown in FIG. 4B, when examined in a TNBC cell line MDA-MB468, the mRNA expression of most of the enzyme genes in the pathway, including HMGCR and SQLE, was significantly inhibited by 2.5 mM RORy inhibitor XY018 (also known as F18) and GSK805. As shown in FIG. 4C, Western blotting showed that protein levels of many of the enzymes such as HMGCS1, HMGCR, SQLE, MVK, MVD, FDFT1, FDPS and EBP are all markedly decreased in cells treated by the RORy inhibitors XY018 and XY063.
Inhibition of RORy effectively abolishes statin treatment-induced up-regulation of cholesterol synthesis senes and the statin effects on cholesterol uptake and efflux senes
[0234] As mentioned above, as a part of its mechanism of action, statin treatment can induce an adaptive response in cells by SREBP-dependent activation of gene expression including that of cholesterol synthesis and LDLR. As shown in FIG. 5, treatment of TNBC cells with 1.25 or 2.5 mM atorvastatin resulted in robust increases of most of the cholesterol synthesis genes (e.g., HMGCS1 HMGCR, MVD, SQLE, LSS, CYP51A1, and DHCR7). In contrast, treatment of the cells with the RORy inhibitors XY018 and XY063 at the same concentrations caused significant decreases in their expression. Interestingly, treatment with both a RORy inhibitor and a statin resulted in inhibitory effects that were largely the same as those by RORy inhibitors alone, indicating that inhibition of RORy effectively abolishes statin treatment-induced upregulation of cholesterol synthesis. Moreover, similar effects were seen with LDLR, a cholesterol uptake protein. On the other hand, the cholesterol efflux transporter gene ABCA1 was decreased by the statin but strongly increased by the RORy inhibitors either alone or in combination with the statin. Together, these results suggest that inhibition of RORy in TNBC cells can potently inhibit the cholesterol pathways and also effectively abolish statin treatment-induced adaptive response of cholesterol homeostasis.
RORy inhibitors in combination with statins display strons synergy in the killins of TNBC cancer cells
[0235] Having demonstrated that RORy inhibition can effectively abolish a statin-induced adaptive response of cholesterol homeostasis, we next examined whether treatment of cancer cells with RORy inhibitors in combination with statins can have therapeutic effects that are advantageous over either alone. As shown in FIG. 6A, with cell viability measured using a CellTiter-Glo assay, at 1.25 pM or lower concentrations, simvastatin did not cause significant growth inhibition of HCC70 TNBC cells after 4 days of treatment. At a concentration that is less than 1/10th of their IC50, such as 0.125 pM, the RORy inhibitors XY018 and XY063 alone (i.e., when 0 pM statin was used) did not display a strong growth inhibition, either. However, the combination of 0.125 pM XY018 or XY063 with different concentrations of simvastatin caused marked growth inhibition that was much more than additive from either alone. As shown in FIGS. 6B-6D, similar synergistic effects were observed in other TNBC cells (SUM159 and MB468) and with a different statin (z.e., atorvastatin).
[0236] Different statins may have different effects on cancer cell growth and survival (1). Table 3 presents the IC50 values of different statins measured by CellTiter-Glo assay, and shows than when combined with relatively low concentrations of RORy inhibitors, except that of pravastatin, the IC50 values of seven different statins (z.e., atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, and pitavastatin) are all significantly decreased (approximately 3- to lO-fold), indicating that the RORy inhibitors potently sensitize the TNBC cells to inhibition by statins.
Table 3 RORy inhibitors in combination with different statins display strong synergy in the killing of TNBC cancer cells
Figure imgf000099_0001
[0237] We next measured the treatment effects on cell survival by a 2D colony formation assay. As shown in FIGS. 7 A and 7B, at 1 mM or lower concentrations, simvastatin did not cause a significant decrease in survival (measured by colony numbers) of HCC70 and MB468 TNBC cells. Likewise, low concentrations of the RORy inhibitors XY018 and XY063 alone also did not lead to a strong survival inhibition. However, the combination of XY018 or XY063 with different concentrations of simvastatin caused a marked synergistic inhibitory effect on the survival of TNBC cells.
RORy inhibitors in combination with statins display strong synergy in the killing of cancer cells of lung cancer, gastric cancer and prostate cancer, including cancer cells that are resistant to current therapeutics
[0238] The remarkably synergistic effects on TNBC cells elicited by the combination of RORy inhibitors and statins encouraged us to examine whether a similar synergy could be obtained in other cancer types. As shown in FIGS. 8A and 8B, at 2.5 mM or lower concentrations, fluvastatin or lovastatin did not cause significant growth inhibition of A549 lung adenocarcinoma cells after 4 days of treatment. At 2.5 mM, the RORy inhibitors XY018 and XY063 alone also did not display any significant growth inhibition. However, the combination of 2.5 mM XY018 or XY063 with different concentrations of simvastatin caused marked growth inhibition that was much more than additive from either alone.
[0239] Similar to what was observed with TNBC cells, when treating lung cancer cells A549 and H358 with the different statins in combination with RORy inhibitors, the IC50 values of seven different statins (i.e., atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, and pitavastatin) are all strongly decreased (ranging from a 3- to over a 20-fold decrease), indicating that the RORy inhibitors potently sensitize lung cancer cells to inhibition by statins (Table 4). A549 and H358 NSCLC cells express wild type EGFR and thus are resistant to EGFR inhibitors such as gefitinib and erlotinib, which target kinase hyperactive-mutant forms of EGFR in the cancer cells.
Table 4 RORy inhibitors in combination with different statins display strong synergy in killing of lung cancer cells
Figure imgf000100_0001
[0240] To further our studies, we also extended our analysis to other cancer types such as prostate cancer and gastric cancer. Results shown in FIG. 9 demonstrate that, similar to the data obtained with TNBC and lung cancer cells, in models of prostate cancer (22Rvl and C4- 2B), combinations of XY018 or XY063 with different concentrations of simvastatin or atorvastatin also caused marked growth inhibition that was much more than additive from either alone. Similar results, shown in FIG. 10, were obtained with several gastric cancer cell lines (AGS, HGC-27 and MGC-803). Notably, the two prostate cancer cell lines (22Rvl and C4-2B) are castration-resistant prostate cancer (CRPC) cells, suggesting that the combination of the RORy inhibitors and statins can be effective in the treatment of CRPC.
[0241] Many RORy small-molecule inhibitors (also known as inverse agonists) have been identified that exhibit high potency in the treatment of autoimmune diseases in in vitro and in vivo preclinical models. VTP -43742 (also known as AGN 242428) is one such compound that has been on clinical trial for its potential use in treatment of psoriasis. As shown in FIG. 11, treatment of human breast cancer cells (MDA-MB468) and CRPC prostate cancer cells (C4-2B) with VTP-43742, either alone or in combination with different concentrations of statins (atorvastatin or simvastatin), resulted in strong growth inhibition. Importantly, the growth inhibition caused by combinations of VTP-43742 with different concentrations of statins was much more than additive as compared to either alone, indicating that a combination treatment of cancer cells with VTP-43742 and a statin is synergistic in killing the cancer cells.
RORy inhibition by siRNAs in combination with statins display strong synergy in the killing of cancer cells
[0242] To examine whether RORy inhibition strategies other than the use of small- molecule inhibitors can also elicit strong synergy with statins in killing cancer cells, we treated human breast cancer cells (MDA-MB468) and CRPC prostate cancer cells (C4-2B) with control siRNA or siRNAs against the human RORC gene, either alone or in combination with different concentrations of a statin (atorvastatin). As shown in FIG. 12A, atorvastatin treatment alone at even the highest concentration of 0.5 mM resulted in a cell survival/colony number decrease from control of approximately 360 to about 300, while siRNA knockdown alone of the RORC gene resulted in a decrease in number from approximately 360 to about 330. However, a combined treatment of the cells with both the statin and the RORC siRNAs resulted in a dramatic decrease of cell survival/colony formation to approximately 130. Similar results were obtained by using the siRNAs against the RORC gene having different targeting sequences as reported previously in Wang, J et al, Nature Medicine , 22, 488-496 (2016). Moreover, a similar synergistic inhibition of cancer cell survival by a combination of siRNA against RORC and a statin was also observed in C4-2B human CRPC prostate cancer cells (FIG. 12B).
Oral administration of RORy inhibitors in combination with statins display strong synergy in inhibition of tumor growth
[0243] We next evaluated the therapeutic potential of combining RORy inhibitors with statins using several human cancer cell line-derived xenograft models. First, we used MDA- MB468 TNBC cells to establish xenograft tumors in female SCID mice. When tumors reached approximately 50 mm3 size, mice were randomized to treatment groups. RORy inhibitors XY018 and XY063 or atorvastatin, or vehicle were administered, through oral gavage (p.o.), to the mice at 10 or 20 mg/kg, five times per week. As shown in FIGS. 13A, 13B and 13C, although the RORy inhibitor or atorvastatin alone displayed significant inhibition effects on the tumor growth, the combined treatment of the RORy inhibitor with the statin display more effective inhibition of tumor growth than either alone. In fact, the combined treatment resulted in almost complete suppression of the tumor growth. Importantly, neither the single nor combined treatments significantly affected the animal body weight over the course of the treatment (FIG. 13D), suggesting that the combined treatment strategy is relatively safe.
[0244] Next, we performed similar experiments using a xenograft tumor model of lung cancer. First, we generated xenograft tumors using A549 cells injected into NOD-SCID female mice. When tumors reached approximately 100 mm3 in size, mice were randomized to groups for treatment by either vehicle, intraperitoneal (i.p.) injection of XY018 (5 mg/kg five times per week), oral gavage of atorvastatin (20 mg/kg five times per week), or a combination of XY018 and atorvastatin (five times per week). Similar to that observed with the TNBC model, tumor growth inhibition was observed when XY018 alone was used; however, more significant tumor suppression was observed when mice were co-treated with XY018 and atorvastatin (FIGS. 14A-14C). Moreover, the treatments did not significantly affect the animal body weight (FIG. 14D). Together, these results strongly suggest that a combination of small-molecule inhibitors of RORy and statins to target the aberrant mevalonate-cholesterol pathway can be a new strategy for effective treatment of a broad spectrum of solid tumors.
Discussion
[0245] We believe that there are several novel or unique aspects about the results from this study. First, currently there is no report about combining the use of statins with RORy modulators for the treatment of any disease including cancer. Our findings indicate a new way to sensitize tumors to statins and to RORy modulators. Secondly, we found that in multiple different cancer types ( e.g ., breast cancer, prostate cancer, gastric cancer, and lung cancer), combining statins with RORy inhibitors can result in strong synergistic effect of cancer cell killing. Moreover, we observed similar synergistic effects in cancer subtypes that currently do not have effective therapy, which include KRAS-mutant lung cancer, gastric cancer, and TNBC. Likewise, we demonstrated a synergistic effect of cancer cell killing by combining statins and RORy inhibitors in cancer cells or tumor models that are resistant to current therapeutics, including chemotherapy drugs and targeted therapy drugs. Therefore, a combined treatment with small-molecule inhibitors of RORy and statins can be applied to a broad spectrum of human cancers.
[0246] Our notion that a combined treatment with small-molecule inhibitors of RORy and statins can have a broad utility in the treatment of human cancers is also supported by our results obtained using statins or RORy inhibitors with different chemical structures. Namely, the RORy inhibitors that can synergize with statins in cancer cell killing are not limited to one particular chemical scaffold/structure. Thus, we found that in addition to XY018 and XY063, RORy inhibitors such as GSK805 and SR2211 developed for potential use in treating autoimmune diseases also display the synergism, although the level of synergism is lower than that of the XY018 or XY063. The statins that display strong synergism with the RORy inhibitors in killing cancer cells include all of the major statin drugs that are clinically used to treat hypercholesterolemia, including atorvastatin, simvastatin, fluvastatin, lovastatin, mevastatin, pitavastatin, and rosuvastatin.
[0247] Maintenance of cholesterol homeostasis is crucial for the growth and function of both normal and cancer cells. Therefore, one concern is that targeting the tumor aberrant mevalonate-cholesterol pathway by a combination of small-molecule inhibitors of RORy and statins could cause significant side effects on normal tissues. However, with respect to this concern, we observed that the combined treatments did not significantly affect the animal body weight over the course of the treatment. Moreover, we have performed studies that have shown that the combined treatment also did not cause any significant alteration in circulating cholesterol levels or any significant liver or kidney toxicity. Together, these data suggest that the combined treatment strategy is relatively safe.
Materials and Methods
Cell culture
[0248] HCC70, MDA-MB468, HCC1500, ZR75-1, and HCC1954 TNBC cancer cells were cultured in RPMI1640. MDA-MB231, MCF-7, MDA-MB361, and BT20 cells were cultured in DMEM. BT549 cells were cultured in RPMI1640 plus 1% insulin. Hs578T cells were cultured in DMEM plus 1% insulin. SUM149 and SUM159 were cultured in DMEM/F12 medium plus insulin and hydrocortisone. The culture media for SEIM149 and SEIM159 were supplemented with 5% FBS (Hyclone) while other media was supplemented with 10% FBS. Cells were grown at 37 °C in 5% C02 incubators. HCC70, MDA-MB468, MDA-MB231, DMEM, and BT549 were obtained from ATCC. The TNBC cancer cell lines were recently authenticated by ATCC using STR profiling. Gastric cancer cells were obtained from ATCC and cultured in RPMI plus 10% FBS. Prostate cancer cell line source and culture conditions were described previously in (6). Cell lines were regularly tested for being negative for mycoplasma.
Chemicals
[0249] Sources for chemicals were as follows: GSK805 was obtained from Calbiochem or WuXi AppTec; XY018 and XY063 were obtained from WuXi AppTec; VTP-43742 was synthesized by the laboratory of one of the inventors. Statins were obtained from Selleck. Other chemicals were obtained from Sigma unless indicated otherwise. gRT-PCR and Western blottins analysis
[0250] Total RNA was isolated from cells in 6-well or lO-cm plates, and the cDNA was prepared, amplified and measured in the presence of SYBR. Briefly, the fluorescent values were collected and a melting curve analysis was performed. Fold difference was calculated. The experiments were performed at least three times with data presented as mean values ± s.d. Cell lysates were analyzed by immunoblotting with antibodies specifically recognizing RORy and other indicated proteins.
Cell viability and colony formation
[0251] For cell viability, cells were seeded in 96-well plates at 1,500-2,500 cells per well (optimum density for growth) in a total volume of 100 pL media. Serially diluted compounds in 100 pL of media were added to the cells 12 hours later. After 4 days of incubation, Cell- Titer GLO reagents (Promega) were added and luminescence was measured using a GLOMAX microplate luminometer (Promega) according to the manufacturer’s instructions. All experimental points were set up as sextuplicate as biological replication and the entire experiments were repeated three times. The data are presented as percentage of viable cells with vehicle treated cells set as 100. The estimated in vitro IC50 values were calculated using GraphPad Prism 6 software.
[0252] For colony formation, 800 cells were seeded in a well of 6-well plates and cultured for 14 days with the medium changed every 3 days. When the cell clone grew visible, the medium was removed and the cells were fixed with 10% formalin for 10 minutes. Then the plates were washed twice with PBS and the cell colonies were stained with 0.2% crystal violet (in 10% formalin) for 15 minutes. The numbers of cell colonies were counted after being washed 5 times with PBS. The above assays were performed in triplicate and the entire experiments were repeated three times. siRNA transfection
[0253] siRNAs for gene knockdown were purchased from Dharmacon. The siRNA target sequences for RORC were published previously (6). Transfections were performed with OptiMEM (Invitrogen) and Dharmafectin #1 (Dharmacon) following the manufacturer’s instructions.
CRISP R/Cas9 sgRNA design, lentivirus production and infection
[0254] sgRNAs were designed using the MIT CRISPR design software (crispr.mit.edu). Oligos corresponding to the sgRNAs were synthesized and cloned into lentiCRISPR v2 vectors following lentiCRISPRv2 and lentiGuide oligo cloning protocol (Addgene, plasmid #52961). Lentiviral particles were produced in 293T cells as in our previous study (6). TNBC cancer cells were plated at 2 c 105 cells per well in 6-well plates. Sixteen hours later, 1 mL of virus-containing supernatant with 10 ng polybrene was added to the cells. After 4 to 6 hours, medium was changed to regular medium and cultured for another 72 hours before being harvested for cell number and protein expression analysis.
RNA-seq data analysis
[0255] HCC70 cells were treated with vehicle or the antagonists XY018 or XY063 or atorvastatin, or a combination of antagonists and atorvastatin for 24 hours before RNA extraction. RNA-seq libraries from 1 pg total RNA were prepared using Illumina Tru-Seq RNA Sample Prep Kit, according to the manufacturer’s instructions. Libraries were validated with an Agilent Bioanalyzer (Agilent Technologies, Palo Alto, CA). Sequencing was performed on an Illumina HiSeq 2000 sequencer at BGI Tech (Hong Kong). The FASTQ- formatted sequence data were analyzed using a standard BWA-Bowtie-Cufflinks.
Xenograft tumor models and chemical compound treatments
[0256] Four-week-old female SCID C.B17 mice or B ALB/c nu/nu athymic mice were purchased from Envigo. Breast cancer MDA-MB468 cells from cell culture (2 x 106 cells) were mixed with Matrigel as a 50% suspension and then injected, in a volume of 0.1 ml, bilaterally into inguinal mammary glands of 4-6-week-old C.B-17-SCID mice. A549 lung xenograft tumors were established by subcutaneous (s.c.) injection of A549 cell suspensions in Matrigel into the flanks of NOD-SCID female mice. Animal groups having a size of six or more was estimated to have a high statistic power, based on power calculation and previous studies involving the same xenograft models. When the tumor volumes reached the volumes indicated in the figures, the mice were randomized and then administered, via oral gavage, five times per week, 100 pL of vehicle (with a formulation of 15% Cremophor EL, Calbiochem, 82.5% PBS, and 2.5% DMSO), statin (in PBS), RORy antagonist/inhibitor (in a formulation of 15% Cremophor EL, 82.5% PBS and 2.5% DMSO), or a combination of RORy inhibitor and statin (in their respective solvents). Tumor volumes were monitored using calipers with volumes calculated using the equation: p/6 (length x width2). Body weight during the course of the study was also monitored. At the end of the studies mice were killed and tumors were dissected and weighed. Additionally, the organs of kidney, heart, lung, liver, spleen and serum were harvested. The procedures were approved by the Institutional Animal Care and Else Committee (IACUC) of University of California, Davis.
Statistical Analysis
[0257] Cell culture-based experiments were performed three times or more with assay points triplicated or sextuplicated, as indicated. The data are presented as mean values ± s.d.
EXAMPLE 2 Using a combination of RORy inhibitors and statins synergistically sensitizes cancer cells to statins and inhibits cancer cell growth
[0258] As shown in FIG. 16, inhibition of RORy with the antagonists XY018 and XY063 strongly sensitized lung cancer cells to killing by different statins. The top of FIG. 16A shows that the combination index (Cl) for XY018 and simvastatin was 0.179, which indicates a high synergy in growth inhibition, as calculated using data shown at the bottom of FIG. 16A from single or combined simvastatin and XY018 treatment of human lung cancer cell line PC9. For data shown at the bottom of FIG. 16A, PC9 cell viability was measured by CellTiter-Glo (Promega) after 4 days of treatment with XY018, simvastatin or their combination in the indicated concentrations (in pM).
[0259] As shown in FIGS. 16B and 16C, human lung cancer A549 cells were treated by the indicated increasing concentrations of a statin alone (fluvastatin (FIG. 16B) or lovastatin FIG. 16C) plus vehicle control), or the different concentrations of the indicated statins in combination with RORy inhibitor/antagonist XY018 or XY063 (2.5 pM) for 4 days. Cell viability was measured by CellTiter-Glo (Promega). Cell viability obtained from cells treated with vehicle only (without either the statin or the RORy inhibitor/antagonist) was set as 100. The experiments were repeated three times.
[0260] Further studying the combination treatment effects on lung cancer cells, FIG. 17 shows that combined treatment with RORy inhibitors/antagonists and statins was synergistic in the inhibition of lung cancer tumor growth. SCID female mice bearing EGFR-mutant, PC9 human lung cancer cell xenograft tumors subcutaneously were randomized for treatment, via oral gavage, with RORy inhibitors alone (XY018, 10 mg/kg, p.o.), atorvastatin (ATV, 10 mg/kg, p.o.) alone, or a combination of the RORy inhibitor and atorvastatin (XY018, 10 mg/kg, p.o. + ATV 10 mg/kg, p.o.), or vehicle (n=7 mice per group) for 27 days, 5 times per week. Tumor volume in mean ± s.e.m. is shown in FIG. 17A, tumor weight in mean ± s.d. is shown in FIG. 17B, and dissected tumor images, at last treatment, are shown in FIG. 17C.
[0261] Based on the high synergistic index (CI=0. l79) and data interpretation similar to that shown in FIG. 6, the results in FIG. 16 indicate that inhibition of RORy with small- molecule antagonists strongly sensitizes lung cancer cells to killing by different statins.
[0262] Turning to mechanism, FIG. 18 shows that a combined treatment of cancer cells with a RORy inhibitor and statin synergistically inhibited oncogenic kinase signaling by receptor tyrosine kinases (RTKs) and serine/threonine kinases. As shown in FIG. 18 A, EGFR-mutant human lung cancer PC9 cells were treated for 24 hours with atorvastatin/ ATV (0, 1, 5 or 10 mM) either alone (with control vehicle), or in combination with 1.25 pM or 5 pM RORy inhibitor XY018. Cells were then harvested for preparation of cell lysates. Immunoblotting analysis of the cell lysates with antibodies against specific proteins as indicated at right to each row of bands (anti-EGFR to detect total amount of EGFR, anti- p/phospho-EGFR (Tyrl068) to detect signaling-activated EGFR, anti -ART to detect total amount of ART, anti-p/phospho-ART (Ser473) to detect signaling-activated ART, anti-ERK to detect total amount of the p44/42 MAPK ERK1/2, and anti-p/phospho-ERK (Thr202/Tyr204) to detect signaling-activated ERK). Immunoblotting with anti-GAPDH protein was also performed to demonstrate loading of equal amount of cell lysates.
[0263] As shown in FIG. 18B, KRAS-mutant human lung cancer A549 cells were treated for 24 hours with control vehicle, XY018 (2.5 pM), XY063 (2.5 pM), atorvastatin (10 pM) or their combination in serum-free media, followed by 50 ng/mL of growth factor EGF stimulation for 15 minutes. Cells were then harvested for preparation of cell lysates and immunoblotting analysis with antibodies as above for the data shown in FIG. 18 A.
[0264] For the data shown in FIG. 18 A, based on the markedly decreased detection (as measured by intensities of the bands) of the phospho-forms of EGFR, AKT and ERK, in cells treated with both XY018 and atorvastatin if compared to cells treated with the vehicle control or either alone, the PC9 cancer cell-intrinsic phosphorylation levels of EGFR, AKT and ERK were strongly inhibited by the combined treatment of cells with the RORy inhibitor and atorvastatin, even at their low concentrations (i.e., 1.25 mM XY018 plus 1 mM atorvastatin). For the data shown in FIG. 18B, based on the markedly decreased intensities of the bands of the phospho-forms of EGFR and AKT in cells treated with both XY018 or XY063 and atorvastatin/ATV if compared to cells treated with the vehicle control or either alone, the EGF growth factor-stimulated phosphorylation levels of EGFR and AKT were strongly inhibited by the combined treatment of cells with the RORy inhibitor and atorvastatin.
[0265] FIG. 19 shows biochemical analysis of mouse serum samples for the assessment of any significant alteration of key blood/serum biochemical parameters or toxicity indicators. SCID mice were treated for 63 days, via oral gavage, with RORy inhibitors alone (XY018, 10 mg/kg, p.o. or XY063, 10 mg/kg, p.o) or atorvastatin (20 mg/kg, p.o) alone or RORy inhibitors combined with atorvastatin (XY018, 10 mg/kg, p.o. + atorvastatin 20 mg/kg, p.o or XY063, 10 mg/kg, p.o. + atorvastatin 20 mg/kg, p.o) or vehicle (n=7 mice per group) for 63 days. 5 times per week. The reference ranges were provided by Comparative Pathology Lab at ETC Davis (cpl.ucdavis.edu/).
[0266] By comparing to the reference ranges of B ALB/c mice, data in FIG. 19 show that the treatment of mice with the RORy inhibitors XY018 or XY063 alone, or in combination with a statin (atorvastatin) did not cause any significant alteration of key blood/serum biochemical parameters or any significant liver or kidney toxicity.
Example 3 Analysis of the effects of RORy inhibitors and statins on tumor growth and metastasis in vivo
RORy inhibitors cause tumor regression and block metastasis.
[0267] To examine the therapeutic value of RORy antagonists, we first treated mice bearing MB468-derived orthotopic xenograft tumors, i.p., with two different doses of XY018. We found that at a relatively low dose of 2.5 mg/kg the antagonist was effective in inhibition of the tumor growth while a higher dose (5 mg/kg) displayed a complete tumor growth blockade for over 7 weeks (FIG. 20A). Similar potent tumor-inhibition activities of antagonists GSK805 and XY018 at 5 mg/kg were observed in additional TNBC HCC70 and SUM159 cell-derived xenografts (FIGS. 20B, 20C). To provide more relevant data for a clinical setting, we measured the oral dosing (p. o.) efficacy of XY018 in a PDX model of TNBC and found that oral administration of XY018 (50 mg/kg) resulted in tumor regression (FIGS. 21A, 20D). XY018 treatment via i.p. (10 mg/kg) did not suppress the growth of MCF-7-derived orthotopic xenografts (FIG. 20E).
[0268] Next, we examined whether RORy antagonists have effects on tumor growth and metastasis in an immune-intact host environment with 4T1 syngeneic tumors. Similar to the effect in the xenograft models, the RORy antagonist strongly inhibited 4T1 tumor growth, and more importantly, dramatically extended host animal survival (FIGS. 21B, 20F). It is well known that the 4T1 primary tumor cells at mammary gland metastasize with high efficiency to the lung. To examine the potential effect of the antagonist on tumor metastasis, we surgically removed the primary tumors at mammary glands and then treated the mice with XY018 daily for 4 weeks. Histological analysis revealed that tumor nodules formed in the lung were significantly reduced (FIG. 21C). To further establish the anti -metastasis activity of targeting RORy, we used the lung-metastatic model (LM2) of MD A-MB231. After the animals received LM2 cells via tail vein, we treated them with XY018 for 4 weeks before measuring tumor cell bioluminescence and observed a strong reduction of LM2 lung colonization in the treated mice (FIG. 20G).
RORy inhibitors in combination with statins regress tumors
[0269] We next evaluated the therapeutic potential of combining RORy inhibitors with statins using multiple TNBC models. First, in two PDX models (1079 and 1173), oral administration of antagonist XY018 alone (20 mg/kg) significantly inhibited the tumor growth, whereas ATV alone (15 mg/kg, p.o.) did not display any significant effect. Remarkably, their combined treatment caused tumor regression (FIG. 21D, FIG. 21E, FIG. 20H, FIG. 201). Moreover, in PDX- 1079 bearing animals, combined treatment of the RORy antagonist with a different statin drug (SIM) elicited essentially the same strong synergy (FIG. 21F, FIG. 20J). Finally, in the MDA-MB468 cell-derived xenografts, the combined treatment also displayed a synergistic tumor growth inhibition (FIG. 21G, FIG. 20K). Importantly, neither the single nor the combined treatments affected the animal body weight over the course of the treatment, suggesting that the RORy inhibitor or the combined treatment strategy is relatively safe. Together, the results from our animal experiments strongly suggest that RORy inhibitor alone or its combination with statin can be a new strategy for effective treatment of TNBC.
Methods— Mouse models and treatments
[0270] NSG (JAX stock #005557) mice were purchased from the Jackson Laboratory. SCID C. B -17 mice or Balb/c nu/nu athymic mice were purchased from Envigo. Mice were housed under standard conditions with free access to food and water, under a 12 h light/l2 h dark cycle in a temperature-controlled environment. Mice were fed a standard rodent chow diet (Envigo Teklad 2918). For cell line-derived xenograft, cells (2 c 106 for each of the human cancer cells or 3 x 104 mouse 4T1) were mixed with Matrigel as 50% suspension. Then the cells injected, in a volume of 0.1 ml, bilaterally into inguinal mammary glands of 4- 6-week-old female C.B-17-SCID mice. For the patient-derived xenograft, PDX-1079 (JAX ID: TM01079) and PDX-1173 (JAX ID: J000101173) were purchased from the Jackson Laboratory. PDXs were propagated by inserting ~ 2mm3 into inguinal mammary glands of 4-6-week-old female C.B-17-SCID mice. Animal group size of six or more was estimated to have a high statistic power, based on power calculation and previous studies involving the same xenograft models. The concentrations of drug and the routes of drug administration are indicated in each figure. When the tumor volumes reached to the indicated volume, mice were randomized and then administered with 100 pl of vehicle (with a formulation of 15% Cremophor EL, Calbiochem, 82.5% PBS, and 2.5% DMSO), statins (in PBS), or RORy antagonists (in a formulation of 15% Cremophor EL, 82.5% PBS and 2.5% DMSO), or their combinations (in their respective solvent). Tumor volumes were monitored using calipers with volume calculated using the equation: p/6 (length c width2). Body weight and survival during the course of the study was also monitored. At the end of the studies mice were killed and tumors were dissected and weighed. Additionally, the organs and whole blood were harvested.
[0271] For effects on lung metastasis from orthotopic 4T1 murine tumors, female Balb/c mice were inoculated with 4T1 cells (3 x 104) into the inguinal mammary gland fat pad. Once the tumors reached 250 mm3 at the primary sites they were surgically removed. Mice were then treated daily with XY018 (20 mg/kg, i.p.) or vehicle for 4 weeks. For histological analysis, mice were euthanized at the end of the treatments, and tumor nodules in the lungs were analyzed by H&E staining for morphology.
[0272] For effects on lung colonization, 2 x 105 luciferase-expressing MDA-MB231-LM2 (4175) cells were injected into each nude mouse via tail. The mice were treated daily with vehicle or 20 mg/kg XY018, i.p. for 4 weeks. Tumor growth in the lungs was monitored for bioluminescence with a Xenogen IVIS-200 system.
V. REFERENCES
1. Clendening, J. W., and Penn, L. Z. (2012). Oncogene 37, 4967-4978.
2. Jeon, T. L, and Osborne, T. F. (2012). Trends in endocrinology and metabolism: TEM 23, 65-72.
3. Jetten, A. M., Kang, H. S., and Takeda, Y. (2013). Frontiers in endocrinology 4, 1.
4. Mullen, P. J., Yu, R., Longo, J., Archer, M. C., and Penn, L. Z. (2016). Nature reviews Cancer 16, 718-731.
5. Shimano, H., and Sato, R. (2017). Nat Rev Endocrinol 13, 710-730.
6. Wang, J., Zou, J. X., Xue, X., Cai, D., Zhang, Y., Duan, Z., Xiang, Q., Yang, J. C., Louie, M. C., Borowsky, A. D„ et al. (2016). Nat Med 22, 488-496.
VI. EXEMPLARY EMBODIMENTS
[0273] Exemplary embodiments provided in accordance with the presently disclosed subject matter include, but are not limited to, the claims and the following embodiments:
1. A method for preventing or treating a cancer in a subject, the method comprising administering to the subject an effective amount of a RORy inhibitor and a statin. 2. The method of embodiment 1, wherein the RORy inhibitor is a small- molecule compound, an anti -RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
3. The method of embodiment 1 or 2, wherein the RORy inhibitor is selected from the group consisting of a compound according to Formula I:
Figure imgf000112_0001
GSK805, SR2211, a compound listed in Table 5, pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and solvates thereof, wherein
X is C(=0) or S02;
n is an integer selected from the group consisting of 0, 1, 2, or 3;
Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF3)2, heterocyclyl, and cycloalkyl,
wherein R5 is selected from the group consisting of H, and C1-C3 alkyl group; R2 is selected from the group consisting of H, halogen, and alkyl;
R3 is selected from the group consisting of H and alkyl;
R4 is selected from the group consisting of C0-C4 alkylene-R6, C0-C4 alkylene- Rvcycloalkyl, and C0-C4 alkylene-Rvheterocyclyl,
wherein R6 is selected from the group consisting of -Rx, -ORx, -CORx, -COORx, -S(0)mR8, cycloalkyl, and heterocyclyl, m is 0 or 2, and R7 is selected from the group consisting of -OR9, -C(0)R9, -NR9, -SR9, -S(0)R9, -S(0)2R9,
wherein Rx is selected from the group consisting of H, and Ci-C3 alkyl group, and R9 is Ci-C3 alkyl ene;
wherein each cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, carboxy, amino, -CONFh, -COOR10, -COR10, -OR10, -NHCOR10, -NHCOOR10, and -COH(CF3)2,
each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono- , bi- or tri-cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, Ci-C4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -CONFh, -COORio, -CORio, -ORio, -NHCORio, -NHCOORio, -COH(CF3)2, -C6H5R11, morpholinyl, piperidinyl, tetrahydrofuranyl, substituted pyridyl group,
wherein Rio is independently selected from the group consisting of H, C1-C4 alkyl, and phenyl, and
R11 is independently selected from the group consisting of C1-C4 alkyl, halogen, acetyl, methoxy, and ethoxy.
4. The method of any one of embodiments 1 to 3, wherein the RORy inhibitor selectively binds to RORy and inhibits RORy activity.
5. The method of any one of embodiments 1 to 4, wherein the compound of Formula I is represented by a compound according to any one of Formulas Ic to Ii:
Figure imgf000113_0001
Figure imgf000114_0001
or a pharmaceutically acceptable salt thereof, a derivative thereof, an analog thereof, or a combination thereof.
6. The method of any one of embodiments 1 to 4, wherein the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP- 43742, and a combination thereof.
7. The method of any one of embodiments 1 to 6, wherein the statin is selected from the group consisting of atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, pitavastatin, cerivastatin, pravastatin, and a combination thereof.
8. The method of any one of embodiments 1 to 7, wherein the cancer is resistant to an anticancer drug. 9. The method of embodiment 8, wherein the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
10. The method of embodiment 9, wherein the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
11. The method of embodiment 9, wherein the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof.
12. The method of embodiment 11, wherein the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
13. The method of any one of embodiments 1 to 12, wherein the cancer is selected from the group consisting of breast cancer, prostate cancer, lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, and glioma.
14. The method of embodiment 13, wherein the breast cancer is a triple- negative breast cancer (TNBC), tamoxifen-resistant breast cancer, radiation-resistant breast cancer, HER2-positive breast cancer, or ER-positive breast cancer.
15. The method of embodiment 13, wherein the prostate cancer is a castration-resistant prostate cancer.
16. The method of embodiment 13, wherein the lung cancer is a non small-cell lung cancer (NSCLC), K-Ras mutant lung cancer, BRAF mutant lung cancer, EGFR mutant lung cancer, tyrosine kinase inhibitor-resistant lung cancer, or small cell lung cancer (SCLC).
17. The method of any one of embodiments 8 to 14, further comprising administering the anti-cancer drug to the subject.
18. The method of any one of embodiments 1 to 17, wherein the subject is a human in need of cancer treatment. 19. The method of any one of embodiments 8 to 18, wherein administering the RORy inhibitor and the statin enhances the therapeutic effect of the anticancer drug.
20. The method of embodiment 19, wherein administering the RORy inhibitor and the statin reverses or reduces cancer cell resistance to the anti cancer drug and/or sensitizes cancer cells to the anticancer drug.
21. The method of any one of embodiments 1 to 20, wherein administering the RORy inhibitor and the statin produces a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof.
22. The method of any one of embodiments 19 to 21, wherein enhancement of the anticancer drug therapeutic effect and/or the beneficial effect that is produced are greater when the RORy inhibitor and the statin are administered in combination compared to when the RORy inhibitor or the statin are administered alone.
23. The method of embodiment 22, wherein administering the RORy inhibitor and the statin in combination produces a synergistic enhancement of the anticancer drug and/or produces a synergistic beneficial effect.
24. The method of any one of embodiments 1 to 23, wherein the cancer is sensitized to the statin.
25. The method of any one of embodiments 1 to 24, wherein the RORy inhibitor and the statin are administered concomitantly.
26. The method of any one of embodiments 1 to 24, wherein the RORy inhibitor and the statin are administered sequentially.
27. A composition comprising a RORy inhibitor and a statin.
28. The composition of embodiment 27, wherein the RORy inhibitor is a small-molecule compound, an anti-RORy antibody, a small-interfering RNA (siRNA), or a combination thereof. 29. The composition of embodiment 27 or 28, wherein the RORy inhibitor is selected from the group consisting of a compound according to Formula I:
Figure imgf000117_0001
GSK805, SR2211, a compound listed in Table 5, pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and solvates thereof, wherein
X is C(=0) or S02;
n is an integer selected from the group consisting of 0, 1, 2, or 3;
Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF3)2, heterocyclyl, and cycloalkyl,
wherein R5 is selected from the group consisting of H, and C1-C3 alkyl group; R2 is selected from the group consisting of H, halogen, and alkyl;
R3 is selected from the group consisting of H and alkyl;
R4 is selected from the group consisting of C0-C4 alkylene-R6, C0-C4 alkylene- Rvcycloalkyl, and C0-C4 alkylene-Rvheterocyclyl,
wherein R6 is selected from the group consisting of -Rx, -ORx, -CORx, -COORx, -S(0)mR8, cycloalkyl, and heterocyclyl, m is 0 or 2, and R7 is selected from the group consisting of -OR9, -C(0)R9, -NR9, -SR9, -S(0)R9, -S(0)2R9,
wherein Rx is selected from the group consisting of H, and Ci-C3 alkyl group, and R9 is Ci-C3 alkylene;
wherein each cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, carboxy, amino, -CONFh, -COOR10, -COR10, -OR10, -NHCOR10, -NHCOOR10, and -COH(CF3)2,
each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono- , bi- or tri-cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, C1-C4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -CONFh, -COOR10, -COR10, -OR10, -NHCORio, -NHCOORio, -COH(CF3)2, -C6H5R11, morpholinyl, piperidinyl, tetrahydrofuranyl, substituted pyridyl group,
wherein Rio is independently selected from the group consisting of H, C1-C4 alkyl, and phenyl, and
R11 is independently selected from the group consisting of C1-C4 alkyl, halogen, acetyl, methoxy, and ethoxy.
30. The composition of any one of embodiments 27 to 29, wherein the RORy inhibitor selectively binds to RORy and inhibits RORy activity.
31. The composition of any one of embodiments 27 to 30, wherein the compound of Formula I is represented by a compound according to any one of Formulas Ic to Ii:
Figure imgf000118_0001
Figure imgf000119_0001
or a pharmaceutically acceptable salt thereof, a derivative thereof, an analog thereof, or a combination thereof.
32. The composition of any one of embodiments 27 to 30, wherein the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP-43742, and a combination thereof.
33. The composition of any one of embodiments 27 to 32, wherein the statin is selected from the group consisting of atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, pitavastatin, cerivastatin, pravastatin, and a combination thereof.
34. The composition of any one of embodiments 27 to 33, wherein the composition comprises an effective amount of the RORy inhibitor and the statin.
35. The composition of any one of embodiments 27 to 34, wherein the composition comprises an effective amount of the RORy inhibitor that is sufficient to sensitize a cancer to the statin.
36. The composition of embodiment 34, wherein the effective amount of the RORy inhibitor and the statin is sufficient to enhance an anticancer drug therapeutic effect and/or produce a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof.
37. The composition of embodiment 36, wherein enhancing the anticancer drug therapeutic effect comprises reversing or reducing cancer cell resistance to the anticancer drug and/or sensitizing cancer cells to the anticancer drug.
38. The composition of embodiment 36 or 37, wherein the enhancement of the anticancer drug therapeutic effect and/or the beneficial effect that is produced is greater when the RORy inhibitor and the statin are administered in combination compared to when the RORy inhibitor or the statin are administered alone.
39. The composition of embodiment 38, wherein the anticancer drug therapeutic effect is synergistically enhanced and/or the beneficial effect is synergistically produced when the RORy inhibitor and the statin are administered in combination.
40. The composition of any one of embodiments 27 to 39, further comprising an anticancer drug.
41. The composition of embodiment 40, wherein the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
42. The composition of embodiment 41, wherein the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
43. The composition of embodiment 41, wherein the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof.
44. The composition of embodiment 43, wherein the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof. 45. The composition of any one of embodiments 27 to 44, further comprising a pharmaceutically acceptable excipient or diluent.
46. A kit for preventing or treating cancer in a subject, the kit comprising a RORy inhibitor and a statin.
47. The kit of embodiment 46, wherein the RORy inhibitor is a small- molecule compound, an anti- RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
48. The kit of embodiment 46 or 47, wherein the RORy inhibitor is selected from the group consisting of a compound according to Formula I:
Figure imgf000121_0001
GSK805, SR2211, a compound listed in Table 5, pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and solvates thereof, wherein
X is C(=0) or S02;
n is an integer selected from the group consisting of 0, 1, 2, or 3;
Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF3)2, heterocyclyl, and cycloalkyl,
wherein R5 is selected from the group consisting of H, and C1-C3 alkyl group; R2 is selected from the group consisting of H, halogen, and alkyl;
R3 is selected from the group consisting of H and alkyl;
R4 is selected from the group consisting of C0-C4 alkylene-R6, C0-C4 alkylene- Rvcycloalkyl, and C0-C4 alkylene-Rvheterocyclyl,
wherein R6 is selected from the group consisting of -Rx, -ORx, -CORx, -COORx, -S(0)mR8, cycloalkyl, and heterocyclyl, m is 0 or 2, and R7 is selected from the group consisting of -OR9, -C(0)R9, -NR9, -SR9, -S(0)R9, -S(0)2R9,
wherein Rx is selected from the group consisting of H, and Ci-C3 alkyl group, and R9 is Ci-C3 alkyl ene;
wherein each cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, carboxy, amino, -CONFh, -COORio, -CORio, -ORio, -NHCORio, -NHCOORio, and -COH(CF3)2,
each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono- , bi- or tri-cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, Ci-C4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -CONFh, -COORio, -CORio, -ORio, -NHCORio, -NHCOORio, -COH(CF3)2, -C6H5R11, morpholinyl, piperidinyl, tetrahydrofuranyl, substituted pyridyl group,
wherein Rio is independently selected from the group consisting of H, C1-C4 alkyl, and phenyl, and
R11 is independently selected from the group consisting of C1-C4 alkyl, halogen, acetyl, methoxy, and ethoxy.
49. The kit of any one of embodiments 46 to 48, wherein the RORy inhibitor selectively binds to RORy and inhibits RORy activity.
50. The kit of any one of embodiments 46 to 49, wherein the compound of Formula I is represented by a compound according to any one of Formulas Ic to Ii:
Figure imgf000122_0001
Figure imgf000123_0001
or a pharmaceutically acceptable salt thereof, a derivative thereof, an analog thereof, or a combination thereof.
51. The kit of any one of embodiments 46 to 49, wherein the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP- 43742, and a combination thereof.
52. The kit of any one of embodiments 46 to 51, wherein the statin is selected from the group consisting of atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, pitavastatin, cerivastatin, pravastatin, and a combination thereof.
53. The kit of any one of embodiments 46 to 52, wherein the kit comprises an effective amount of the RORy inhibitor and the statin. 54. The kit of any one of embodiments 46 to 53, wherein the kit comprises an effective amount of the RORy inhibitor that is sufficient to sensitize a cancer to the statin.
55. The kit of embodiment 53, wherein the effective amount of the RORy inhibitor and the statin is sufficient to enhance an anticancer drug therapeutic effect and/or produce a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof.
56. The kit of embodiment 55, wherein enhancing the anticancer drug therapeutic effect comprises reversing or reducing cancer cell resistance to the anticancer drug and/or sensitizing cancer cells to the anticancer drug.
57. The kit of embodiment 55 or 56, wherein the enhancement of the anticancer drug therapeutic effect and/or the beneficial effect that is produced is greater when the RORy inhibitor and the statin are administered in combination compared to when the RORy inhibitor or the statin are administered alone.
58. The kit of embodiment 57, wherein the anticancer drug therapeutic effect is synergistically enhanced and/or the beneficial effect is synergistically produced when the RORy inhibitor and the statin are administered in combination.
59. The kit of any one of embodiments 46 to 58, further comprising an anticancer drug.
60. The kit of embodiment 59, wherein the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
61. The kit of embodiment 60, wherein the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
62. The kit of embodiment 60, wherein the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof. 63. The kit of embodiment 62, wherein the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
64. The kit of any one of embodiments 46 to 63, further comprising a label with instructions for administering the RORy inhibitor and the statin to the subject.
65. The kit of any one of embodiments 46 to 64, wherein the subject is a human in need of cancer treatment.
66. A method for preventing or treating a cancer in a subject, the method comprising administering to the subject an effective amount of a RORy inhibitor, wherein the RORy inhibitor is a compound listed in Table 5, a pharmaceutically acceptable salt thereof, an isomer thereof, a racemate thereof, a prodrug thereof, a co-crystalline complex thereof, a hydrate thereof, or a solvate thereof.
67. The method of embodiment 66, wherein the RORy inhibitor selectively binds to RORy and inhibits RORy activity.
68. The method of embodiment 66 or 67, wherein the RORy inhibitor is
VTP-43742.
69. The method of any one of embodiments 66 to 68, wherein the cancer is resistant to an anticancer drug.
70. The method of embodiment 69, wherein the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
71. The method of embodiment 70, wherein the anti -androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
72. The method of embodiment 70, wherein the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof.
73. The method of embodiment 72, wherein the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof. 74. The method of any one of embodiments 66 to 73, wherein the cancer is selected from the group consisting of breast cancer, prostate cancer, lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, and glioma.
75. The method of embodiment 74, wherein the breast cancer is a triple- negative breast cancer (TNBC), tamoxifen-resistant breast cancer, radiation-resistant breast cancer, HER2-positive breast cancer, or ER-positive breast cancer.
76. The method of embodiment 74, wherein the prostate cancer is a castration-resistant prostate cancer.
77. The method of embodiment 74, wherein the lung cancer is a non small-cell lung cancer (NSCLC), K-Ras mutant lung cancer, BRAF mutant lung cancer, EGFR mutant lung cancer, tyrosine kinase inhibitor-resistant lung cancer, or small cell lung cancer (SCLC).
78. The method of any one of embodiments 69 to 75, further comprising administering the anti-cancer drug to the subject.
79. The method of any one of embodiments 66 to 78, wherein the subject is a human in need of cancer treatment.
80. The method of any one of embodiments 69 to 79, wherein administering the RORy inhibitor enhances the therapeutic effect of the anticancer drug.
81. The method of embodiment 80, wherein administering the RORy inhibitor and reverses or reduces cancer cell resistance to the anticancer drug and/or sensitizes cancer cells to the anticancer drug.
[0274] Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, one of skill in the art will appreciate that certain changes and modifications may be practiced within the scope of the appended claims. In addition, each reference provided herein is incorporated by reference in its entirety to the same extent as if each reference was individually incorporated by reference.

Claims

WHAT IS CLAIMED IS:
1. A method for preventing or treating a cancer in a subject, the method comprising administering to the subject an effective amount of a RORy inhibitor and a statin.
2. The method of claim 1, wherein the RORy inhibitor is a small- molecule compound, an anti-RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
3. The method of claim 1, wherein the RORy inhibitor is selected from the group consisting of a compound according to Formula I:
Figure imgf000127_0001
GSK805, SR2211, a compound listed in Table 5, pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and solvates thereof, wherein
X is C(=0) or S02;
n is an integer selected from the group consisting of 0, 1, 2, or 3;
Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF3)2, heterocyclyl, and cycloalkyl,
wherein R5 is selected from the group consisting of H, and C1-C3 alkyl group; R2 is selected from the group consisting of H, halogen, and alkyl;
R3 is selected from the group consisting of H and alkyl;
R4 is selected from the group consisting of C0-C4 alkylene-R6, C0-C4 alkylene- Rvcycloalkyl, and C0-C4 alkylene-Rvheterocyclyl,
wherein R6 is selected from the group consisting of -Rx, -ORx, -CORx, -COORx, -S(0)mR8, cycloalkyl, and heterocyclyl, m is 0 or 2, and R7 is selected from the group consisting of -OR9, -C(0)R9, -NR9, -SR9, -S(0)R9, -S(0)2R9,
wherein Rx is selected from the group consisting of H, and Ci-C3 alkyl group, and R9 is Ci-C3 alkyl ene;
wherein each cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, carboxy, amino, -CONFh, -COORio, -CORio, -ORio, -NHCORio, -NHCOORio, and -COH(CF3)2,
each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono- , bi- or tri-cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, Ci-C4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -CONFh, -COORio, -CORio, -ORio, -NHCORio, -NHCOORio, -COH(CF3)2, -C6H5R11, morpholinyl, piperidinyl, tetrahydrofuranyl, substituted pyridyl group,
wherein Rio is independently selected from the group consisting of H, C1-C4 alkyl, and phenyl, and
R11 is independently selected from the group consisting of C1-C4 alkyl, halogen, acetyl, methoxy, and ethoxy.
4. The method of claim 1, wherein the RORy inhibitor selectively binds to RORy and inhibits RORy activity.
5. The method of claim 3, wherein the compound of Formula I is represented by a compound according to any one of Formulas Ic to Ii:
Figure imgf000128_0001
Figure imgf000129_0001
or a pharmaceutically acceptable salt thereof, a derivative thereof, an analog thereof, or a combination thereof.
6. The method of claim 1, wherein the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP -43742, and a combination thereof.
7. The method of claim 1, wherein the statin is selected from the group consisting of atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, pitavastatin, cerivastatin, pravastatin, and a combination thereof.
8. The method of claim 1, wherein the cancer is resistant to an anticancer drug.
9. The method of claim 8, wherein the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
10. The method of claim 9, wherein the anti-androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
11. The method of claim 9, wherein the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof.
12. The method of claim 11, wherein the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
13. The method of claim 1, wherein the cancer is selected from the group consisting of breast cancer, prostate cancer, lung cancer, liver cancer, ovarian cancer, endometrial cancer, bladder cancer, colon cancer, gastric cancer, lymphoma, and glioma.
14. The method of claim 13, wherein the breast cancer is a triple-negative breast cancer (TNBC), tamoxifen-resistant breast cancer, radiation-resistant breast cancer, HER2-positive breast cancer, or ER-positive breast cancer.
15. The method of claim 13, wherein the prostate cancer is a castration- resistant prostate cancer.
16. The method of claim 13, wherein the lung cancer is a non-small-cell lung cancer (NSCLC), K-Ras mutant lung cancer, BRAF mutant lung cancer, EGFR mutant lung cancer, tyrosine kinase inhibitor-resistant lung cancer, or small cell lung cancer (SCLC).
17. The method of claim 8, further comprising administering the anti cancer drug to the subject.
18. The method of claim 1, wherein the subject is a human in need of cancer treatment.
19. The method of claim 8, wherein administering the RORy inhibitor and the statin enhances the therapeutic effect of the anticancer drug.
20. The method of claim 19, wherein administering the RORy inhibitor and the statin reverses or reduces cancer cell resistance to the anticancer drug and/or sensitizes cancer cells to the anticancer drug.
21. The method of claim 1, wherein administering the RORy inhibitor and the statin produces a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof.
22. The method of claim 19, wherein enhancement of the anticancer drug therapeutic effect and/or the beneficial effect that is produced are greater when the RORy inhibitor and the statin are administered in combination compared to when the RORy inhibitor or the statin are administered alone.
23. The method of claim 22, wherein administering the RORy inhibitor and the statin in combination produces a synergistic enhancement of the anticancer drug and/or produces a synergistic beneficial effect.
24. The method of claim 1, wherein the cancer is sensitized to the statin.
25. The method of claim 1, wherein the RORy inhibitor and the statin are administered concomitantly.
26. The method of claim 1, wherein the RORy inhibitor and the statin are administered sequentially.
27. A composition comprising a RORy inhibitor and a statin.
28. The composition of claim 27, wherein the RORy inhibitor is a small- molecule compound, an anti-RORy antibody, a small-interfering RNA (siRNA), or a combination thereof.
29. The composition of claim 27, wherein the RORy inhibitor is selected from the group consisting of a compound according to Formula I:
Figure imgf000132_0001
GSK805, SR2211, a compound listed in Table 5, pharmaceutically acceptable salts thereof, isomers thereof, racemates thereof, prodrugs thereof, co-crystalline complexes thereof, hydrates thereof, and solvates thereof, wherein
X is C(=0) or S02;
n is an integer selected from the group consisting of 0, 1, 2, or 3;
Ri is selected from the group consisting of H, halo, alkyl, trifluoromethyl, cyano, -COORs, -COR5, -OR5, -COH(CF3)2, heterocyclyl, and cycloalkyl,
wherein R5 is selected from the group consisting of H, and C1-C3 alkyl group; R2 is selected from the group consisting of H, halogen, and alkyl;
R3 is selected from the group consisting of H and alkyl;
R4 is selected from the group consisting of C0-C4 alkylene-R6, C0-C4 alkylene- Rvcycloalkyl, and C0-C4 alkylene-Rvheterocyclyl,
wherein R6 is selected from the group consisting of -Rx, -ORx, -CORx, -COORx, -S(0)mR8, cycloalkyl, and heterocyclyl, m is 0 or 2, and R7 is selected from the group consisting of -OR9, -C(0)R9, -NR9, -SR9, -S(0)R9, -S(0)2R9,
wherein Rx is selected from the group consisting of H, and Ci-C3 alkyl group, and R9 is Ci-C3 alkyl ene;
wherein each cycloalkyl group is a saturated or unsaturated ring structure ranging from 3 to 10 carbon atoms, and each cycloalkyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from the group consisting of halogen, C1-C4 alkyl group, trifluoromethyl, cyano, carboxy, amino, -CONFh, -COOR10, -COR10, -OR10, -NHCOR10, -NHCOOR10, and -COH(CF3)2,
each heterocyclyl group is a 5 to 12 membered saturated or unsaturated mono- , bi- or tri-cyclic structure comprising from 1 to 3 heteroatoms independently selected from the group consisting of N, O, and S, and each heterocyclyl group is optionally substituted with 0, 1, 2 or 3 substituents independently selected from halogen, C1-C4 alkyl, trifluoromethyl, cyano, carboxy, nitro, amino, -CONFh, -COOR10, -COR10, -OR10, -NHCORio, -NHCOORio, -COH(CF3)2, -C6H5R11, morpholinyl, piperidinyl, tetrahydrofuranyl, substituted pyridyl group,
wherein Rio is independently selected from the group consisting of H, C1-C4 alkyl, and phenyl, and
R11 is independently selected from the group consisting of C1-C4 alkyl, halogen, acetyl, methoxy, and ethoxy.
30. The composition of claim 27, wherein the RORy inhibitor selectively binds to RORy and inhibits RORy activity.
31. The composition of claim 27, wherein the compound of Formula I is represented by a compound according to any one of Formulas Ic to Ii:
Figure imgf000133_0001
Figure imgf000134_0001
or a pharmaceutically acceptable salt thereof, a derivative thereof, an analog thereof, or a combination thereof.
32. The composition of claim 27, wherein the RORy inhibitor is selected from the group consisting of XY018, XY063, GSK805, SR2211, VTP-43742, and a combination thereof.
33. The composition of claim 27, wherein the statin is selected from the group consisting of atorvastatin, simvastatin, lovastatin, fluvastatin, rosuvastatin, mevastatin, pitavastatin, cerivastatin, pravastatin, and a combination thereof.
34. The composition of claim 27, wherein the composition comprises an effective amount of the RORy inhibitor and the statin.
35. The composition of claim 27, wherein the composition comprises an effective amount of the RORy inhibitor that is sufficient to sensitize a cancer to the statin.
36. The composition of claim 34, wherein the effective amount of the RORy inhibitor and the statin is sufficient to enhance an anticancer drug therapeutic effect and/or produce a beneficial effect selected from the group consisting of inhibiting cancer cell growth, inhibiting cancer cell metastasis, decreasing tumor size, increasing survival time of the subject, ameliorating one or more signs and/or symptoms of cancer, and a combination thereof.
37. The composition of claim 36, wherein enhancing the anticancer drug therapeutic effect comprises reversing or reducing cancer cell resistance to the anticancer drug and/or sensitizing cancer cells to the anticancer drug.
38. The composition of claim 36, wherein the enhancement of the anticancer drug therapeutic effect and/or the beneficial effect that is produced is greater when the RORy inhibitor and the statin are administered in combination compared to when the RORy inhibitor or the statin are administered alone.
39. The composition of claim 38, wherein the anticancer drug therapeutic effect is synergistically enhanced and/or the beneficial effect is synergistically produced when the RORy inhibitor and the statin are administered in combination.
40. The composition of claim 27, further comprising an anticancer drug.
41. The composition of claim 40, wherein the anticancer drug is selected from the group consisting of an anti-androgen drug, a chemotherapeutic agent, a radiotherapeutic agent, an antigen-specific immunotherapeutic agent, an endocrine therapy, a tyrosine kinase inhibitor, and a combination thereof.
42. The composition of claim 41, wherein the anti -androgen drug is selected from the group consisting of enzalutamide, bicalutamide, arbiraterone, nilutamide, flutamide, apalutamide, finasteride, dutasteride, alfatradiol, and a combination thereof.
43. The composition of claim 41, wherein the chemotherapeutic agent is selected from the group consisting of tamoxifen, a taxane, and a combination thereof.
44. The composition of claim 43, wherein the taxane is selected from the group consisting of paclitaxel, docetaxel, and a combination thereof.
45. The composition of claim 27, further comprising a pharmaceutically acceptable excipient or diluent.
PCT/US2019/049178 2018-08-31 2019-08-30 Methods for treating cancer with rorgamma inhibitors and statins Ceased WO2020047487A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862726087P 2018-08-31 2018-08-31
US62/726,087 2018-08-31

Publications (1)

Publication Number Publication Date
WO2020047487A1 true WO2020047487A1 (en) 2020-03-05

Family

ID=69643067

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/049178 Ceased WO2020047487A1 (en) 2018-08-31 2019-08-30 Methods for treating cancer with rorgamma inhibitors and statins

Country Status (1)

Country Link
WO (1) WO2020047487A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021228217A1 (en) * 2020-05-15 2021-11-18 上海辉启生物医药科技有限公司 ANILINE COMPOUND USED AS RORγ REGULATOR
US20210353621A1 (en) * 2020-05-12 2021-11-18 Arvinas Operations, Inc. Methods of treating prostate cancer
WO2022122740A1 (en) * 2020-12-07 2022-06-16 Diaccurate Ror-gamma inhibitors for the treatment of ror-gamma-dependent cancer
US11952347B2 (en) 2016-10-11 2024-04-09 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of androgen receptor
US12043612B2 (en) 2020-05-09 2024-07-23 Arvinas Operations, Inc. Methods of manufacturing a bifunctional compound, ultrapure forms of the bifunctional compound, and dosage forms comprising the same
WO2024255845A1 (en) * 2023-06-14 2024-12-19 中国科学院上海药物研究所 Protein degradation agent, and pharmaceutical composition and use thereof
US12496301B2 (en) 2023-12-08 2025-12-16 Arvinas Operations, Inc. Use of androgen receptor degrader for the treatment of spinal and bulbar muscular atrophy

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170159057A1 (en) * 2004-07-01 2017-06-08 Dan Littman Compositions and methods for modulation of rorgammat functions
WO2017127442A1 (en) * 2016-01-18 2017-07-27 The Regents Of The University Of California Methods for treating cancer with rorgamma inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170159057A1 (en) * 2004-07-01 2017-06-08 Dan Littman Compositions and methods for modulation of rorgammat functions
WO2017127442A1 (en) * 2016-01-18 2017-07-27 The Regents Of The University Of California Methods for treating cancer with rorgamma inhibitors

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
JETTEN ET AL.: "Retinoic acid-related orphan receptor gamma (RORgamma): Connecting sterol metabolism to regulation of the immune system and autoimmune disease", CURRENT OPINION IN TOXICOLOGY, vol. 8, 1 April 2018 (2018-04-01), pages 66 - 80, XP055697486 *
SCHWEIZER ET AL.: "Targeting intratumoral androgens: statins and beyond", MEDICAL ONCOLOGY, vol. 8, 11 May 2016 (2016-05-11), pages 388 - 395, XP055697492 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11952347B2 (en) 2016-10-11 2024-04-09 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of androgen receptor
US11964945B2 (en) 2016-10-11 2024-04-23 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of androgen receptor
US12077509B2 (en) 2016-10-11 2024-09-03 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of androgen receptor
US12043612B2 (en) 2020-05-09 2024-07-23 Arvinas Operations, Inc. Methods of manufacturing a bifunctional compound, ultrapure forms of the bifunctional compound, and dosage forms comprising the same
US20210353621A1 (en) * 2020-05-12 2021-11-18 Arvinas Operations, Inc. Methods of treating prostate cancer
WO2021228217A1 (en) * 2020-05-15 2021-11-18 上海辉启生物医药科技有限公司 ANILINE COMPOUND USED AS RORγ REGULATOR
CN113773251A (en) * 2020-05-15 2021-12-10 上海辉启生物医药科技有限公司 Anilines useful as ROR gamma modulators
CN113773251B (en) * 2020-05-15 2024-06-11 励缔(杭州)医药科技有限公司 Anilines useful as rory modulators
WO2022122740A1 (en) * 2020-12-07 2022-06-16 Diaccurate Ror-gamma inhibitors for the treatment of ror-gamma-dependent cancer
WO2024255845A1 (en) * 2023-06-14 2024-12-19 中国科学院上海药物研究所 Protein degradation agent, and pharmaceutical composition and use thereof
US12496301B2 (en) 2023-12-08 2025-12-16 Arvinas Operations, Inc. Use of androgen receptor degrader for the treatment of spinal and bulbar muscular atrophy

Similar Documents

Publication Publication Date Title
WO2020047487A1 (en) Methods for treating cancer with rorgamma inhibitors and statins
US10959984B2 (en) Methods for treating cancer with RORγ inhibitors
Garg et al. The pleiotropic role of transcription factor STAT3 in oncogenesis and its targeting through natural products for cancer prevention and therapy
US11215617B2 (en) Treatment of metastatic prostate cancer
US20210161837A1 (en) Methods for treating cancer with rorgamma inhibitors
Mimeault et al. Hypoxia‐inducing factors as master regulators of stemness properties and altered metabolism of cancer‐and metastasis‐initiating cells
ES2877629T3 (en) Administration of hypoxia-activated prodrugs and antiangiogenic agents for the treatment of cancer
US20160074390A1 (en) Human dosing of phosphatase inhibitor
AU2014344789B2 (en) Pharmaceutical combinations for the treatment of cancer
US12152036B2 (en) Substituted pyrrolo[1,2-α]quinoxalin-4(5H)-ones as CX3CR1 antagonists
RU2014150860A (en) DOSING MODE PI-3 KINASES
JP6373252B2 (en) Methods of treating cancer using aurora kinase inhibitors
WO2019113155A1 (en) Oxabicycloheptanes for treatment of secondary acute myeloid leukemia
US20180153850A1 (en) Compositions and methods for treatment of cancer
WO2014091486A1 (en) Combination therapy for cancer
JP2024023269A (en) Antitumor agents and combination drugs
JP7541512B2 (en) Prodrugs and their medical uses
JP2024538411A (en) How to protect your heart
HK1222548B (en) Pharmaceutical combinations for the treatment of cancer
HK1228747B (en) Treatment of metastatic prostate cancer
HK1228747A1 (en) Treatment of metastatic prostate cancer

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: 19855850

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19855850

Country of ref document: EP

Kind code of ref document: A1