Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMBINATION THERAPY FOR TREATMENT OF CANCER
Document Type and Number:
WIPO Patent Application WO/2021/026349
Kind Code:
A1
Abstract:
Pharmaceutical compositions comprising: one or more bromodomain and extra terminal domain (BET) proteolysis targeting chimera (PROTAC) (BET-PROTAC) therapeutic agents or one or more cyclin-dependent kinase 9 (CDK9) PROTAC (CDK9-PROTAC) therapeutic agents; and one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors; and methods of treating cancer in a human patient by administering such pharmaceutical compositions are described herein.

Inventors:
DUNCAN JAMES S (US)
Application Number:
PCT/US2020/045197
Publication Date:
February 11, 2021
Filing Date:
August 06, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
INSTITUTE FOR CANCER RES D/B/A THE RES INSTITUTE OF FOX CHASE CANCER CENTER (US)
International Classes:
A61P35/00; C07D401/14; C07D417/04
Domestic Patent References:
WO2019079701A12019-04-25
WO2019075114A12019-04-18
WO2017212329A12017-12-14
Foreign References:
US20180228907A12018-08-16
US20190175612A12019-06-13
US20180161298A12018-06-14
Other References:
PEI ET AL.: "Small molecule PROTACs: an emerging technology for targeted therapy in drug discovery", RSC ADVANCES, vol. 9, no. 30, 30 May 2019 (2019-05-30), pages 16967 - 16976, XP055790796
OLSON ET AL.: "Pharmacological perturbation of CDK9 using selective CDK9 inhibition or degradation", NATURE CHEMICAL BIOLOGY, vol. 14, no. 2, 1 February 2018 (2018-02-01), pages 163 - 170, XP055790797
ROBB ET AL.: "Chemically induced degradation of CDK9 by a proteolysis targeting chimera (PROTAC", CHEMICAL COMMUNICATIONS ( CAMBRIDGE, ENGLAND, vol. 53, no. 54, 4 July 2017 (2017-07-04), pages 7577 - 7580, XP055618975, DOI: 10.1039/C7CC03879H
See also references of EP 4010081A4
Attorney, Agent or Firm:
LEGAARD, Paul K. (US)
Download PDF:
Claims:
What Is Claimed Is:

1. A pharmaceutical composition comprising: one or more bromodomain and extra terminal domain (BET) proteolysis targeting chimera (PROTAC) (BET-PROTAC) therapeutic agents or one or more cyclin-dependent kinase 9 (CDK9) PROTAC (CDK9-PROTAC) therapeutic agents, or a combination thereof; and one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof.

2. The pharmaceutical composition according to claim 1, comprising one or more BET- PROTAC therapeutic agents.

3. The pharmaceutical composition according to claim 2, wherein the BET-PROTAC therapeutic agent is BRD2, BRD3, or BRD4 PROTAC, or any combination thereof.

4. The pharmaceutical composition according to claim 2, wherein the BET-PROTAC therapeutic agent is a VHL ligand-based BET-PROTAC.

5. The pharmaceutical composition according to claim 4, wherein the BET-PROTAC therapeutic agent is MZ1, ARV-771, ATI, MZP-61, MZP-51, or MZP-55, or any combination thereof.

6. The pharmaceutical composition according to claim 5, wherein the BET-PROTAC therapeutic agent is MZ1.

7. The pharmaceutical composition according to claim 2, wherein the BET-PROTAC therapeutic agent is a CRBN ligand-based BET-PROTAC.

8. The pharmaceutical composition according to claim 7, wherein the BET-PROTAC therapeutic agent is ARV-825, dBET6, dBET57, dBET23, ZXH 3-26, BETd246, BETd260, QCA570, or ARCC-29, or any combination thereof.

9. The pharmaceutical composition according to claim 8, wherein the BET-PROTAC therapeutic agent is ARV-825.

10. The pharmaceutical composition according to claim 2, wherein the BET-PROTAC therapeutic agent is a phthalimide-based BET-PROTAC.

11. The pharmaceutical composition according to claim 10, wherein the phthalimide-based BET-PROTAC therapeutic agent is dBETl.

12. The pharmaceutical composition according to claim 2, wherein the BET-PROTAC therapeutic agent is a Nutlin-based BET-PROTAC.

13. The pharmaceutical composition according to claim 12, wherein the Nutlin-based BET- PROTAC therapeutic agent is A1874. 14. The pharmaceutical composition according to claim 2, wherein the BET-PROTAC therapeutic agent is CFT-2718.

15. The pharmaceutical composition according to any onje of claims 1 to 14, comprising one or more CDK9-PROTAC therapeutic agents.

16. The pharmaceutical composition according to claim 15, wherein the CDK9-PROTAC therapeutic agent comprises a CDK9 inhibitor.

17. The pharmaceutical composition according to claim 16, wherein the CDK9 inhibitor is NVP-2.

18. The pharmaceutical composition according to claim 15, wherein the CDK9-PROTAC therapeutic agent is Thai SNS 032.

19. The pharmaceutical composition according to claim 15, wherein the CDK9-PROTAC therapeutic agent is CDK9 Degrader- 1.

20. The pharmaceutical composition according to any one of claims 1 to 19, comprising one or more kinase inhibitors.

21. The pharmaceutical composition according to claim 20, wherein the kinase inhibitor is an RTK signaling pathway inhibitor.

22. The pharmaceutical composition according to claim 21, wherein the RTK signaling pathway inhibitor is an INSR/IGF1R inhibitor, an EGFR/ERBB/HER2/HER3/HER4 inhibitor, an RTK inhibitor, an MERTK inhibitor, an MST1R inhibitor, or an FGFR1 inhibitor, or any combination thereof.

23. The pharmaceutical composition according to claim 22, wherein the INSR/IGF1R inhibitor is GSK1904529 A, the EGFR/ERBB/HER2/HER3/HER4 inhibitor is lapatinib, the RTK inhibitor is imatinib, the MERTK inhibitor is MRX-2843, the MST1R inhibitor is LY2801653 dihydrochloride, and the FGFR1 inhibitor is PD173074.

24. The pharmaceutical composition according to claim 20, wherein the kinase inhibitor is an MTOR signaling pathway inhibitor.

25. The pharmaceutical composition according to claim 24, wherein the MTOR signaling pathway inhibitor is an mTORCl inhibitor, a PI3K inhibitor, a P70S6K inhibitor, a PI4K2A inhibitor, or a CDK9 inhibitor, or any combination thereof.

26. The pharmaceutical composition according to claim 25, wherein the mTORCl inhibitor is RAD001 or Torin-1, the PI3K inhibitor is GDC-0941, the P70S6K inhibitor is LY2584702, the PI4K2A inhibitor is PI-273, and the CDK9 inhibitor is NVP-2.

27. The pharmaceutical composition according to claim 20, wherein the kinase inhibitor is a CDK7 inhibitor, CK2, AKT1/2, AAK1, FAK1, MEK, and RAF inhibitor. 28. The pharmaceutical composition according to claim 24, wherein the MTOR inhibitor is Dactolisib (BEZ235 or NVP-BEZ235), Rapamycin (Sirolimus), Everolimus (RAD001), AZD8055, Temsirolimus (CCI-779 or NSC 683864), PI-103, KU-0063794, Torkinib (PP242), Tacrolimus (FK506), Ridaforolimus (Deforolimus or MK-8669), Sapanisertib (INK 128, MLN0128, or TAK-228), Voxtalisib (SAR245409 orXL765) Analogue, Torin 1, Omipalisib (GSK2126458 or GSK458), OSI-027, PF-04691502, Apitolisib (GDC-0980 or RG7422),

GSK1059615, WYE-354, Gedatolisib (PF-05212384 or PKI-587), Torin 2, WYE-125132 (WYE-132), Vistusertib (AZD2014), BGT226 (NVP-BGT226), Palomid 529 (P529), PP121, WYE-687, WAY-600, ETP-46464, GDC-0349, XL388, Zotarolimus (ABT-578), LY3023414, CC-115, MHY1485, CZ415, GDC-0084, Voxtalisib (XL765 or SAR245409), 3BDO,

Bimirabsib (PQR309), CC-223, or SF2523.

29. The pharmaceutical composition according to any one of claims 1 to 19, comprising one or more KRAS inhibitors.

30. The pharmaceutical composition according to claim 29, wherein the KRAS inhibitor is ARS-1620 or MRTX849.

31. The pharmaceutical composition according to any one of claims 1 to 19, comprising one or more autophagy inhibitors.

32. The pharmaceutical composition according to claim 31, wherein the autophagy inhibitor is SAR405, Autophinib, PIK-III, LYN-1604, SBI-0206965, MRT68921 HC1, ULK-101, or hydroxychloroquine.

33. The pharmaceutical composition according to claim 1, comprising one or more BET- PROTAC therapeutic agents and one or more MTOR inhibitors or one or more KRAS inhibitors.

34. The pharmaceutical composition according to claim 1, comprising one or more CDK9- PROTAC therapeutic agents and one or more MTOR inhibitors.

35. The pharmaceutical composition according to claim 1, comprising MZ1 and one or more of GSK1904529A, lapatinib, and RAD001.

36. A method for augmenting the therapeutic effect in a human cancer patient undergoing treatment with a BET-PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent comprising administering one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof, to the patient.

37. The method according to claim 36, wherein the BET-PROTAC therapeutic agent the human cancer patient is undergoing treatment with is a BRD2, BRD3, or BRD4 PROTAC, or any combination thereof. 38. The method according to claim 36, wherein the BET-PROTAC therapeutic agent the human cancer patient is undergoing treatment with is a VHL ligand-based BET-PROTAC.

39. The method according to claim 38, wherein the VHL ligand-based BET-PROTAC is MZ1, ARV-771, ATI, MZP-61, MZP-51, or MZP-55, or any combination thereof.

40. The method according to claim 39, wherein the VHL ligand-based BET-PROTAC is MZ1.

41. The method according to claim 36, wherein the BET-PROTAC therapeutic agent the human cancer patient is undergoing treatment with is a CRBN ligand-based BET-PROTAC.

42. The method according to claim 41, wherein the CRBN ligand-based BET-PROTAC is ARV-825, dBET6, dBET57, dBET23, ZXH 3-26, BETd246, BETd260, QCA570, or ARCC-29, or any combination thereof.

43. The method according to claim 42, wherein the CRBN ligand-based BET-PROTAC is ARV-825.

44. The method according to claim 36, wherein the BET-PROTAC therapeutic agent the human cancer patient is undergoing treatment with is a phthalimide-based BET-PROTAC.

45. The method according to claim 44, wherein the phthalimide-based BET-PROTAC therapeutic agent is dBETl.

46. The method according to claim 36, wherein the BET-PROTAC therapeutic agent the human cancer patient is undergoing treatment with is a Nutlin-based BET-PROTAC.

47. The method according to claim 46, wherein the Nutlin-based BET-PROTAC therapeutic agent is A1874.

48. The method according to claim 36, wherein the BET-PROTAC therapeutic agent is CFT-2718.

49. The method according to claim 36, wherein the CDK9-PROTAC therapeutic agent the human cancer patient is undergoing treatment with comprises a CDK9 inhibitor.

50. The method according to claim 49, wherein the CDK9 inhibitor is NVP-2.

51. The method according to claim 36, wherein the CDK9-PROTAC therapeutic agent the human cancer patient is undergoing treatment with is Thai SNS 032.

52. The method according to claim 36, wherein the CDK9-PROTAC therapeutic agent the human cancer patient is undergoing treatment with is PROTAC CDK9 Degrader- 1.

53. The method according to any one of claims 36 to 52, wherein the human cancer patient is administered one or more kinase inhibitors.

54. The method according to claim 53, wherein the kinase inhibitor is an RTK signaling pathway inhibitor. 55. The method according to claim 54, wherein the RTK signaling pathway inhibitor is an INSR/IGF1R inhibitor, an EGFR/ERBB/HER2/HER3/HER4 inhibitor, an RTK inhibitor, an MERTK inhibitor, an MST1R inhibitor, or an FGFR1 inhibitor, or any combination thereof.

56. The method according to claim 55, wherein the INSR/IGF1R inhibitor is GSK1904529A, the EGFR/ERBB/HER2/HER3/HER4 inhibitor is lapatinib, the RTK inhibitor is imatinib, the MERTK inhibitor is MRX-2843, the MST1R inhibitor is LY2801653 dihydrochloride, and the FGFR1 inhibitor is PD173074.

57. The method according to claim 53, wherein the kinase inhibitor is an MTOR signaling pathway inhibitor.

58. The method according to claim 57, wherein the MTOR signaling pathway inhibitor is an mTORCl inhibitor, a PI3K inhibitor, a P70S6K inhibitor, a PI4K2A inhibitor, or a CDK9 inhibitor, or any combination thereof.

59. The method according to claim 58, wherein the mTORCl inhibitor is RAD001 or Torin-1, the PI3K inhibitor is GDC-0941, the P70S6K inhibitor is LY2584702, the PI4K2A inhibitor is PI-273, and the CDK9 inhibitor is NVP-2.

60. The method according to claim 57, wherein the MTOR inhibitor is Dactolisib (BEZ235 or NVP-BEZ235), Rapamycin (Sirolimus), Everolimus (RAD001), AZD8055, Temsirolimus (CCI-779 or NSC 683864), PI-103, KU-0063794, Torkinib (PP242), Tacrolimus (FK506), Ridaforolimus (Deforolimus or MK-8669), Sapanisertib (INK 128, MLN0128, or TAK-228), Voxtalisib (SAR245409 or XL765) Analogue, Torin 1, Omipalisib (GSK2126458 or GSK458), OSI-027, PF-04691502, Apitolisib (GDC-0980 or RG7422), GSK1059615, WYE-354, Gedatolisib (PF-05212384 or PKI-587), Torin 2, WYE-125132 (WYE-132), Vistusertib (AZD2014), BGT226 (NVP-BGT226), Palomid 529 (P529), PP121, WYE-687, WAY-600,

ETP -46464, GDC-0349, XL388, Zotarolimus (ABT-578), LY3023414, CC-115, MHY1485, CZ415, GDC-0084, Voxtalisib (XL765 or SAR245409), 3BDO, Bimiralisib (PQR309), CC-223, or SF2523.

61. The method according to any one of claims 36 to 52, wherein the human cancer patient is administered one or more KRAS inhibitors.

62. The method according to claim 61, wherein the KRAS inhibitor is ARS-1620 or MRTX849.

63. The method according to any one of claims 36 to 52, wherein the human cancer patient is administered one or more autophagy inhibitors. 64. The method according to claim 63, wherein the autophagy inhibitor is SAR405, Autophinib, PIK-III, LYN-1604, SBI-0206965, MRT68921 HC1, ULK-101, or hydroxychloroquine.

65. The method according to any one of claims 29 to 51, wherein the amount of the BET- PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human cancer patient receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is reduced compared to the amount of the BET- PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human cancer patient in the absence of receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors.

66. The method according to any one of claims 36 to 65, wherein the cancer is acute myeloid leukemia, acute monocytic leukemia, prostatic adenocarcinoma, ovarian carcinoma, or epithelial ovarian cancer.

67. The method according to claim 66, wherein the epithelial ovarian cancer is High-Grade Serous Ovarian Carcinoma (HGSOC).

68. The method according to any one of claims 36 to 65, wherein the cancer is a KRAS cancer, PIK3CA cancer, or PTEN cancer.

69. A method of treating cancer in a human patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a pharmaceutical composition comprising: one or more BET-PROTAC therapeutic agents or one or more CDK9-PROTAC therapeutic agents, or a combination thereof; and one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof.

70. The method according to claim 69, wherein the human patient is administered one or more BET-PROTAC therapeutic agents.

71. The method according to claim 70, wherein the BET-PROTAC therapeutic agent is BRD2, BRD3, or BRD4 PROTAC, or any combination thereof.

72. The method according to claim 70, wherein the BET-PROTAC therapeutic agent is a VHL ligand-based BET-PROTAC.

73. The method according to claim 72, wherein the BET-PROTAC therapeutic agent is MZ1, ARV-771, ATI, MZP-61, MZP-51, or MZP-55, or any combination thereof.

74. The method according to claim 73, wherein the BET-PROTAC therapeutic agent is

MZ1. 75. The method according to claim 70, wherein the BET-PROTAC therapeutic agent is a CRBN ligand-based BET-PROTAC.

76. The method according to claim 75, wherein the BET-PROTAC therapeutic agent is ARV-825, dBET6, dBET57, dBET23, ZXH 3-26, BETd246, BETd260, QCA570, or ARCC-29, or any combination thereof.

77. The method according to claim 76, wherein the BET-PROTAC therapeutic agent is ARV-825.

78. The method according to claim 70, wherein the BET-PROTAC therapeutic agent is a phthalimide-based BET-PROTAC.

79. The method according to claim 78, wherein the phthalimide-based BET-PROTAC therapeutic agent is dBETl.

80. The method according to claim 70, wherein the BET-PROTAC therapeutic agent is a Nutlin-based BET-PROTAC.

81. The method according to claim 80, wherein the Nutlin-based BET-PROTAC therapeutic agent is A1874.

82. The method according to claim 70, wherein the BET-PROTAC therapeutic agent is CFT-2718.

83. The method according to claim 69, wherein the human patient is administered one or more CDK9-PROTAC therapeutic agents.

84. The method according to claim 83, wherein the CDK9-PROTAC therapeutic agent is a CDK9 inhibitor.

85. The method according to claim 84, wherein the CDK9 inhibitor is NVP-2.

86. The method according to claim 83, wherein the CDK9-PROTAC therapeutic agent is Thai SNS 032.

87. The method according to claim 83, wherein the CDK9-PROTAC therapeutic agent is PROTAC CDK9 Degrader- 1.

88. The method according to any one of claims 69 to 87, wherein the human patient is administered one or more kinase inhibitors.

89. The method according to claim 80, wherein the kinase inhibitor is an RTK signaling pathway inhibitor.

90. The method according to claim 89, wherein the RTK signaling pathway inhibitor is an INSR/IGF1R inhibitor, an EGFR/ERBB/HER2/HER3/HER4 RBB inhibitor, an RTK inhibitor, an MERTK inhibitor, an MST1R inhibitor, or an FGFR1 inhibitor, or any combination thereof. 91. The method according to claim 90, wherein the INSR/IGF1R inhibitor is GSK1904529A, the EGFR/ERBB/HER2/HER3/HER4 inhibitor is lapatinib, the RTK inhibitor is imatinib, the MERTK inhibitor is MRX-2843, the MST1R inhibitor is LY2801653 dihydrochloride, and the FGFR1 inhibitor is PD173074.

92. The method according to claim 88, wherein the kinase inhibitor is an MTOR signaling pathway inhibitor.

93. The method according to claim 92, wherein the MTOR signaling pathway inhibitor is an mTORCl inhibitor, a PI3K inhibitor, a P70S6K inhibitor, a PI4K2A inhibitor, or a CDK9 inhibitor, or any combination thereof.

94. The method according to claim 93, wherein the mTORCl inhibitor is RAD001 or Torin-1, the PI3K inhibitor is GDC-0941, the P70S6K inhibitor is LY2584702, the PI4K2A inhibitor is PI-273, and the CDK9 inhibitor is NVP-2.

95. The method according to claim 92, wherein the mTOR inhibitor is Dactolisib (BEZ235 or NVP-BEZ235), Rapamycin (Sirolimus), Everolimus (RAD001), AZD8055, Temsirolimus (CCI-779 or NSC 683864), PI-103, KU-0063794, Torkinib (PP242), Tacrolimus (FK506), Ridaforolimus (Deforolimus or MK-8669), Sapanisertib (INK 128, MLN0128, or TAK-228), Voxtalisib (SAR245409 or XL765) Analogue, Torin 1, Omipalisib (GSK2126458 or GSK458), OSI-027, PF-04691502, Apitolisib (GDC-0980 or RG7422), GSK1059615, WYE-354, Gedatolisib (PF-05212384 or PKI-587), Torin 2, WYE-125132 (WYE-132), Vistusertib (AZD2014), BGT226 (NVP-BGT226), Palomid 529 (P529), PP121, WYE-687, WAY-600,

ETP -46464, GDC-0349, XL388, Zotarolimus (ABT-578), LY3023414, CC-115, MHY1485, CZ415, GDC-0084, Voxtalisib (XL765 or SAR245409), 3BDO, Bimiralisib (PQR309), CC-223, or SF2523.

96. The method according to any one of claims 69 to 87, wherein the human patient is administered one or more KRAS inhibitors.

97. The method according to claim 96, wherein the KRAS inhibitor is ARS-1620 or MRTX849.

98. The method according to any one of claims 69 to 87, wherein the human patient is administered one or more autophagy inhibitors.

99. The method according to claim 98, wherein the autophagy inhibitor is SAR405, Autophinib, PIK-III, LYN-1604, SBI-0206965, MRT68921 HC1, ULK-101, or hydroxychloroquine. 100. The method according to claim 69, wherein the human patient is administered one or more BET-PROTAC therapeutic agents and one or more MTOR inhibitors or one or more KRAS inhibitors.

101. The method according to claim 69, wherein the human patient is administered one or more CDK9-PROTAC therapeutic agents and one or more mTOR inhibitors.

102. The method according to claim 69, wherein the human patient is administered MZ1 and one or more of GSK1904529A, lapatinib, and RAD001.

103. The method according to any one of claims 69 to 102, wherein the amount of the BET- PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human patient receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is reduced compared to the amount of the BET-PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human patient in the absence of receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors. 104. The method according to any one of claims 69 to 103, wherein the cancer is acute myeloid leukemia, acute monocytic leukemia, prostatic adenocarcinoma, ovarian carcinoma, or epithelial ovarian cancer.

105. The method according to claim 104, wherein the epithelial ovarian cancer is High- Grade Serous Ovarian Carcinoma (HGSOC). 106. The method according to any one of claims 69 to 103, wherein the cancer is a KRAS cancer, PIK3CA cancer, or PTEN cancer.

Description:
Combination Therapy for Treatment of Cancer

Reference To Government Grants

This invention was made with government support under Grant Nos. CA211670 and CA006927 awarded by the National Instututes of Health. The government has certain rights in the invention.

Field

The present disclosure is directed to pharmaceutical compositions comprising: one or more bromodomain and extra terminal domain (BET) proteolysis targeting chimera (PROTAC) (BET-PROTAC) therapeutic agents or one or more cyclin-dependent kinase 9 (CDK9) PROTAC (CDK9-PROTAC) therapeutic agents; and one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors; and methods of treating cancer in a human patient by administering such pharmaceutical compositions.

Background

Ovarian cancer represents the fifth-leading cause of death in women in the United States. High-Grade Serous Ovarian Carcinoma (HGSOC) is the most common and deadly subtype of epithelial ovarian cancer but currently has limited effective molecular targeted therapies. Recently, PROTAC (Proteolysis-Targeting Chimeras) degraders have emerged as promising anti-cancer drugs through utilization of the cancer cells own protein destruction machinery to selectively degrade essential tumor drivers. A number of BET protein degraders (BBD) have been identified including MZ1, which uses JQ1 as the bromodomain targeting molecule and recruits E3 ligases using a VHL-binding moiety, and ARV825 with OTX015 as the warhead and Cereblon (CRBN)-binding moiety for E3 ligase recruitment. Targeted degradation of BET proteins has been shown to cause significant tumor growth inhibition or regression in mouse models of prostate cancer, demonstrating the in vivo therapeutic potential of BBD. Notably, recent studies have suggested BET protein degradation differs from BET protein inhibition, where BBD treatment distinctly blocks transcriptional elongation in cancer cells providing superior anti-proliferative and apoptosis inducing effects in cancer cells. Furthermore, BBD have been reported to have greater efficacy than traditional BBI in numerous cancer models, suggesting BET degradation may represent the more favorable method for targeting BET proteins in cancer. However, chronic exposure to BBD therapies triggers kinome reprogramming in cancer cells, which thus acquire resistance to BBD treatment thereby limiting the effectiveness of this therapy. Accordingly, there is a need for therapeutic approaches that overcome resistance to BBD treatment to maintain its efficiency.

Summary

The present disclosure provides pharmaceutical compositions comprising: one or more bromodomain and extra terminal domain (BET) proteolysis targeting chimera (PROTAC) (BET- PROTAC) therapeutic agents or one or more cyclin-dependent kinase 9 (CDK9) PROTAC (CDK9-PROTAC) therapeutic agents, or a combination thereof; and one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof.

The present disclosure also provides methods for augmenting the therapeutic effect in a human cancer patient undergoing treatment with a BET-PROTAC therapeutic agent or CDK9- PROTAC therapeutic agent comprising administering one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof, to the patient.

The present disclosure also provides methods of treating cancer in a human patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a pharmaceutical composition comprising: one or more BET-PROTAC therapeutic agents or one or more CDK9-PROTAC therapeutic agents, or a combination thereof; and one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof.

Brief Description Of The Drawings

Figure 1A shows BBI, JQ1 or BBD, MZ1, which uses JQ1 as bromodomain targeting molecule, were used for proteogenomics studies.

Figure IB shows that knockdown of BRD4 induced apoptosis in OVCAR8 cells confirming BRD4-dependency; OVCAR8 cells were transfected with siRNAs targeting BRD2, BRD3, BRD4 or control siRNA and cultured for 120 hours. Apoptosis was measured by assessing cleaved PARP by Western blot.

Figure 1C shows that treatment of OVCAR8 cells with JQ1 or MZ1 but not cis-MZl blocked cell viability; cells were treated with increasing doses of drug for 5 days and viability was assessed by CellTiter-Glo assays; Cis-MZl was used as a negative control which cannot recruit VHL. Figure ID shows that BBD-treatment reduces BET proteins inducing apoptosis to a greater extent than BBI-treatment; OVCAR8 cells were treated with increasing doses of JQ1 and BET protein levels was determined by Western blot; apoptosis was measured by assessing cleaved PARP levels by blot.

Figure IE shows that BBD-treatment reduces BET proteins inducing apoptosis to a greater extent than BBI-treatment; OVCAR8 cells were treated with increasing doses of MZ1 and BET protein levels was determined by Western blot; apoptosis was measured by assessing cleaved PARP levels by blot.

Figure IF shows that Short-term BBD-treatment induces differential proteome response than BBI-treatment; hierarchical clustering of protein log2 LFQ z-scores of OVCAR8 cells treated with DMSO, BBIs (JQ1 or OTX015) or BBDs (MZ1 or ARV825) for 48 hours; heat map depicts log2 LFQ z-scores for 4604 proteins measured by single-run proteome analysis.

Figure 1G shows that BBD and BBI-induced proteome responses exhibit poor correlation; Pearson correlations of single-run proteome profiles of OVCAR8 cells treated with BBIs (JQ1 or OTX015) or BBDs (MZ1 or ARV825) for 48 hours; correlation plots of SILAC- determined log2-fold changes in protein levels as a ratio of (drug/control) were generated using Perseus Software.

Figure 1H shows Pearson correlation analysis of log2-fold changes in RNA and those proteins exhibiting statistically significant changes following BBI therapies was determined using Perseus Software; BBD-induced proteome and RNA response exhibits poor correlation in contrast to BBI-therapy; OVCAR8 cells were treated with 500 nM JQ1 or MZ1 for 48 hours and global changes in protein or RNA levels was determined by single-run proteome and RNA-seq analysis.

Figure II shows Pearson correlation analysis of log2-fold changes in RNA and those proteins exhibiting statistically significant changes following BBD therapies was determined using Perseus Software.

Figure 1J shows that BBD-therapy uniquely represses translation markers, translational elongation and MYC protein levels; characterization of BBD or BBI therapies on translation, BET proteins, BRD4-targets and MYC signaling were determined by Western blot; OVCAR8 cells were treated with 500 nM MZ1, JQ1, cis-MZl or DMSO for 48 hours and characterization of BBD or BBI therapies on translation, BET proteins, BRD4-targets and MYC signaling determined by Western blot. Figure IK shows that distinct remodeling of the kinome to 48 hours MZ1 or JQ1 (500 nM) treatment in OVCAR8 cells; unsupervised hierarchical clustering of SILAC-determined log2-fold changes in MIB binding of 237 kinases as a ratio of drug/DMSO.

Figure 1L shows Pearson correlations plots of kinome response signatures to MZ1 vs JQ1 or ARV825 vs OTX015; heat map and correlation plots were generated in Perseus Software.

Figure 1M shows the kinases distinctly induced or repressed by BBD therapy; MIB-MS kinome profile of OVCAR8 cells following a 48 hour treatment with BBDs (MZ1, ARV825) or BBIs (JQ1, OTX015); volcano plot depicts SILACdetermined log2-fold changes in MIB binding as a ratio of BBDs/BBIs; statistical changes in SILAC determined MIB-binding were determined by paired T-Test (P < 0.05) using Perseus Software.

Figure IN shows that BBD-treatment blocks kinome remodeling observed with BBI- therapies; OVCAR8 cells were treated with 500 nM of JQ1 or MZ1 for 48 hours and kinase protein levels determined by Western blot.

Figure 2A shows that OVCAR8 cells acquire resistance to MZ1 or ARV825; parental and BBD-resistant cells were treated with escalating doses of MZ1 for 5 days and cell viability assessed by CellTiter-Glo; BBD-R treated cell viabilities normalized to DMSO treated BBD-R cells.

Figure 2B shows that OVCAR8 cells acquire resistance to MZ1 or ARV825; parental and BBD-resistant cells were treated with escalating doses of ARV825 for 5 days and cell viability assessed by CellTiter-Glo; BBD-R treated cell viabilities normalized to DMSO treated BBD-R cells.

Figure 2C shows that escalating doses of BBDs fails to promote degradation of BET proteins in BBD-R cells; OVCAR8 parental cells were treated with escalating doses of MZ1 for 24 hours and BET proteins and BRD4-target FOSL1 were determined by Western blot.

Figure 2D shows that escalating doses of BBDs fails to promote degradation of BET proteins in BBD-R cells; OVCAR8 parental cells were treated with escalating doses of ARV825 for 24 hours and BET proteins and BRD4-target FOSL1 were determined by Western blot.

Figure 2E shows that BET proteins remained reduced in cells chronically exposed to BBD-therapies; BET protein levels were determined by blot in parental cells, parental cells treated with MZ1 (100 nM), MZ1-R or in MZ1-R cells where MZ1 was removed from media for 48 hours.

Figure 2F shows BET protein levels were determined by blot in parental cells, parental cells treated with ARV825 (100 nM), ARV-R or in ARV-R cells where MZ1 was removed from media for 48 hours. Figure 2G shows that Cells resistant to BBI or BBDs are dependent on BRD4 for growth; OVCAR8 parental, JQ1-R, MZ1-R or ARV-R cells were transfected with siRNAs targeting individual BRD2, BRD3 or BRIM, combinations of BRDs or control siRNA, cultured for 120 hours and viability was assessed by CellTiter-Glo.

Figure 2H shows that cells resistant to MZ1 require BRD4 for MYC signaling and cell survival; OVCAR8 MZ1-R cells were transfected with siRNAs targeting individual BRD2, BRIM, BRIM, BRD2/3/4 or control siRNA, cultured for 120 hours and apoptosis and BRD4- targets were assessed by Western blot.

Figure 21 and shows that cells resistant to MZ1 are cross-resistant to VHL-based BBDs but sensitive to CRBN-based BBDs; OVCAR8 parental or MZ1-R cells were treated with increasing doses of VHL-based BBD ARV-771 for 5 days and cell viability was assessed by CellTiter-Glo.

Figure 21 shows that cells resistant to MZ1 are cross-resistant to VHL-based BBDs but sensitive to CRBN-based BBDs; OVCAR8 parental or MZ1-R cells were treated with increasing doses of CRBN-based BBD, ARV825 for 5 days and cell viability was assessed by CellTiter- Glo.

Figure 2K shows that treatment of MZ1-R cells with CRBN-based BBD ARV825 promotes degradation of BET proteins, reduces BRD4-target FOSL1 and induces apoptosis; MZ1-R cells were treated with escalating doses of ARV825 and protein levels assessed by Western blot.

Figure 2L shows that that cells resistant to Cells resistant to ARV825 show cross resistance to CRBN and VHL-based BBDs; ARV-R cells were treated with escalating doses of VHL-based BBD ARV-771 for 5 days and cell viability was assessed by CellTiter-Glo.

Figure 2M shows that that cells resistant to Cells resistant to ARV825 show cross resistance to CRBN and VHL-based BBDs; ARV-R cells were treated with escalating doses of CRBN-based BBD, ARV825 for 5 days and cell viability was assessed by CellTiter-Glo.

Figure 2N shows that treatment of ARV-R cells with VHL or CRBN-based BBDs fails to induce apoptosis observed with ARV825-treatment of parental cells as determined measuring cleaved PARP via Western blot.

Figure 20 shows that BBD-R cells exhibit sensitivity towards BBI therapies; OVCAR8 MZ1-R cells were treated with escalating doses of IQ1 for 5 days and cell viability was assessed by CellTiter-Glo. Figure 2P shows that BBD-R cells exhibit sensitivity towards BBI therapies; OVCAR8 ARV-R cells were treated with escalating doses of JQ1 for 5 days and cell viability was assessed by CellTiter-Glo.

Figure 2Q shows that BBI-R cells exhibit sensitivity to BBD-treatment; OVCAR8 JQ1- R cells were treated with escalating doses of BBIs, JQ1, OTX015, AZD5153, INC54 or MZ1 for 5 days and cell viability was assessed by CellTiter-Glo.

Figure 2R shows OVCAR8 JQ1-R cells were treated with increasing doses of MZ1 for 48 hours and apoptosis determined by Western blot through monitoring of cleaved PARP levels.

Figure 3 A shows principal component analysis of MIB-MS kinome profiles of OVCAR8 parental, JQ1-R, MZ1-R or ARV-R cells; kinase log2 SILAC ratios were determined by comparing ratio of ratios sample/s-SILAC reference.

Figure 3B shows Pearson correlation analysis of MIB-determined kinome profiles of MZ1-R relative to JQ1-R were determined using Perseus Software.

Figure 3C shows kinome remodeling to chronic MZ1 treatment; Volcano plot depicts SILAC-determined log2-fold changes in MIB-binding as a ratio of MZl-R/Parental; statistical changes in MIB-binding were determined by paired T-Test (P < 0.05) using Perseus Software.

Figure 3D shows remodeling of the kinome similar following continuous exposure to VHL or CRBN-based BBDs; scatterplot depicts SILAC-determined log2-fold changes in MIB- binding as a ratio of MZl-R/Parental or ARV-R/Parental; statistical changes in MIB-binding were determined by paired T-Test (P < 0.05) and Pearson correlation of MZ1-R and Pearson correlation of MZ1-R and ARV-R kinome responses determined using using Perseus Software.

Figure 3E shows volcano plot depicting kinases predicted to be activated or inhibited in response to chronic MZ1 treatment using Kinase Substrate Enrichment Analysis (KSEA); phosphoproteomics datasets were queried using PhosphoSitePlus at p value cutoff (0.05) and NetworkKIN score cutoff of 2; statistical differences in log2 ratios of phosphosites comparing MZl-R/s-SILAC relative to parental/s-SILAC were determined by paired T-Test, Benjamini- Hochberg adjusted p values at FDR of <0.05.

Figure 3F shows upregulation of INSR and ERBB3 phosphorylation in MZl-resistant cells; changes in RTK tyrosine phosphorylation comparing MZ1-R and Parental cells was assessed by RTK array.

Figure 3G shows increased INSR and downstream mTOR signaling in MZ1-R cells; parental and MZ1-R OVCAR8 cells protein levels were determined by Western blot.

Figure 3H shows candidate therapeutic kinase targets in BBD-R cells; kinome activation signature of OVCAR8 BBD-R cells was determined by MIB-MS and KSEA. Figure 4A shows that Chronic MZ1 -treatment promotes increased sensitivity towards inhibitors targeting INSR, ErbB and MTOR pathways; parental, JQ1-R or MZ1-R cells were treated with escalating doses of inhibitors for 5 days and cell viability was assessed by CellTiter- Glo.

Figure 4B shows that MZ1-R cells are more sensitive to INSR, ErbB or MTORC1 inhibition than JQ1-R or parental cells; parental, JQ1-R or MZ1-R OVCAR8 cells were treated with escalating doses of GSK1904529A for 5 days and cell viability was assessed by CellTiter- Glo; MZ1-R or JQ1-R inhibitor treated cell viabilities normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully.

Figure 4C shows parental, JQ1-R or MZ1-R OVCAR8 cells treated with escalating doses of lapatinib for 5 days and cell viability was assessed by CellTiter-Glo; MZ1-R or JQ1-R inhibitor treated cell viabilities normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully.

Figure 4D shows parental, JQ1-R or MZ1-R OVCAR8 cells treated with escalating doses of or RAD001 for 5 days and cell viability was assessed by CellTiter-Glo; MZ1-R or JQ1- R inhibitor treated cell viabilities normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully.

Figure 4E shows parental, JQ1-R or MZ1-R OVCAR8 cells treated with escalating doses of or MK2206 for 5 days and cell viability was assessed by CellTiter-Glo; MZ1-R or JQ1- R inhibitor treated cell viabilities normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully.

Figure 4F shows parental, JQ1-R or MZ1-R OVCAR8 cells treated with escalating doses of or NVP-2 for 5 days and cell viability was assessed by CellTiter-Glo; MZ1-R or JQ1-R inhibitor treated cell viabilities normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully.

Figure 4G shows acquired dependency on INSR, ErbB, MTOR and AKT signaling in MZ1-R cells; parental OVCAR8 cells or MZ1-R OVCAR8 cells were transfected with siRNAs targeting EGFR, ERBB2, INSR, IGF1R, MTOR or AKT1/2 and cultured for 72 hours; MZ1-R knockdown cells were normalized to MZ1-R cells transfected with non-targeting siRNA.

Figure 4H shows that GSK529A-treatment blocks INSR and downstream MTOR,

BRAF and cell cycle signaling; MIB-MS kinome profiles of OVCAR8 MZ1-R cells treated with GSK529A (1 mM) or DMSO for 48 hours; heat map depicts kinase log2 LFQ z-scores; statistical differences in kinase log2 LFQ z-scores comparing GSK529A-treated to control treated cells was determined by paired T-Test Benjamini-Hochberg adjusted p values at FDR of <0.05 using Perseus Software. Figure 41 shows treatment of MZ1-R cells with GSK529A blocks INSR and downstream MTOR, AKT and MAPK signaling; OVCAR8 MZ1-R cells were treated with escalating doses of GSK529A or DMSO for 48 hours and the activity of kinases was assessed by Western blot using phospho-antibodies.

Figure 5A shows that blockade of INSR signaling reduces BRD2 and BRD4, as well as BRD4-target MYC protein levels; OVCAR8 MZ1-R cells were treated with escalating doses of GSK529A or DMSO for 48 hours and protein levels were determined by Western blot.

Figure 5B shows depletion of BRD2 and BRD4 protein levels by GSK529A distinct to MZ1-R cells; parental, MZ1-R or JQ1-R cells were treated with escalating doses of GSK529A or DMSO and BET protein levels were determined by Western blot.

Figure 5C shows that inhibition of INSR, ErbB or MTOR commonly reduce BRD2 and BRD4 proteins; MZ1-R OVCAR8 cells were treated with 1 mM of kinase inhibitors or DMSO for 4 hours and BET protein levels were determined by western blot.

Figure 5D shows ARV-R OVCAR8 cells treated with 1 pM of kinase inhibitors or DMSO for 4 hours and BET protein levels were determined by Western blot.

Figure 5E shows that inhibition of MTORC1 promotes BET protein loss selectively in MZ1-R cells; OVCAR8 MZ1-R cells were treated with escalating doses of RAD001 or DMSO for 48 hours and protein levels were determined by Western blot.

Figure 5F shows that inhibition of MTOR does not influence BRD4 RNA levels; OVCAR8 MZ1-R or parental cells were treated with DMSO or (1 pM) of GSK529A, lapatinib or RAD001 for 48 hours and BRD4 RNA levels were determined by qRT-PCR.

Figure 5G shows that inhibition of proteasome rescues GSK529A or RAD001 -mediated reduction of BET proteins in MZ1-R cells; OVCAR8 MZ1-R cells were treated with GSK529A (2 pM), bortezominb (10 nM) or the combination for 4 or 24 hours and BET protein levels were determined by Western blot.

Figure 5H shows OVCAR8 MZ1-R cells treated with RAD001 (2 pM), bortezominb (10 nM) or the combination for 4 or 48 hours and BET protein levels were determined by Western blot.

Figure 6A shows that KURAMOCHI cells acquire resistance to MZ1; parental and MZ1 -resistant cells were treated with escalating doses of MZ1 for 5 days and cell viability was assessed by CellTiter-Glo; MZ1-R treated cell viabilities normalized to DMSO treated MZ1-R cells.

Figure 6B shows that COV362 cells acquire resistance to MZ1; parental and MZ1- resistant cells were treated with escalating doses of MZ1 for 5 days and cell viability was assessed by CellTiter-Glo; MZ1-R treated cell viabilities normalized to DMSO treated MZ1-R cells.

Figure 6C shows that OVCAR5 EOC cells acquire resistance to MZ1; parental and MZ1 -resistant cells were treated with escalating doses of MZ1 for 5 days and cell viability was assessed by CellTiter-Glo; MZ1-R treated cell viabilities normalized to DMSO treated MZ1-R cells.

Figure 6D shows that escalating doses of MZ1 fails to promote degradation of BRD4 in MZ1-R cells; KURAMOCHI parental or MZ1-R cells were treated with escalating doses of MZ1 for 24 hours and BET proteins levels were determined by Western blot.

Figure 6E shows COV362 parental or MZ1-R cells treated with escalating doses of MZ1 for 24 hours and BET proteins levels were determined by Western blot.

Figure 6F shows OVCAR5 parental or MZ1-R cells treated with escalating doses of MZ1 for 24 hours and BET proteins levels were determined by Western blot.

Figure 6G shows that MZ1-R cells exhibit increased sensitivity towards INSR, ErbB or MTORC1 inhibition; KURAMOCHI parental or MZ1-R cells were treated with escalating doses of GSK1904529A, lapatinib or RAD001 for 5 days and cell viability was assessed by CellTiter- Glo; MZ1-R or treated cell viabilities normalized to DMSO treated MZ1-R cells, respectfully.

Figure 6H shows COV362 parental or MZ1-R cells treated with escalating doses of GSK1904529A, lapatinib or RAD001 for 5 days and cell viability was assessed by CellTiter- Glo; MZ1-R or treated cell viabilities normalized to DMSO treated MZ1-R cells, respectfully.

Figure 61 shows OVCAR5 parental or MZ1-R cells treated with escalating doses of GSK1904529A, lapatinib or RAD001 for 5 days and cell viability was assessed by CellTiter- Glo; MZ1-R or treated cell viabilities normalized to DMSO treated MZ1-R cells, respectfully.

Figure 6J shows that inhibition of INSR, ErbB, or MTORC1 promotes BET protein loss in MZ1-R cells; KURAMOCHI MZ1-R cells were treated with escalating doses of GSK529A, lapatinib, RAD001 or DMSO for 48 hours and protein levels were determined by Western blot.

Figure 6K shows COV362 MZ1-R cells treated with escalating doses of GSK529A, lapatinib, RAD001 or DMSO for 48 hours and protein levels were determined by Western blot.

Figure 6L shows OVCAR5 MZ1-R cells treated with escalating doses of GSK529A, lapatinib, RAD001 or DMSO for 48 hours and protein levels were determined by Western blot.

Figure 7A shows that combination therapies involving GSK529A, lapatinib or RAD001 and MZ1 but not JQ1 improve growth repression of OVCAR8 cells; OVCAR8 cells were treated with 100 nM JQ1, MZ1 or cis-MZl, escalating doses of GSK529A, lapatinib or RAD001, or the combination of JQ1, MZ1 or cis-MZl and kinase inhibitors for 5 days and cell viability was assessed by CellTiter-Glo.

Figure 7B shows that CRBN-based BBDs synergize with GSK529A, lapatinib or RAD001; OVCAR8 cells were treated with 100 nM dBET6, escalating doses of GSK529A, lapatinib or RAD001, or the combination of dBET6 and kinase inhibitors for 5 days and cell viability was assessed by CellTiter-Glo.

Figure 7C shows that treatment of cells with GSK529A, lapatinib or RAD001 and MZ1 but not JQ1 increased apoptosis and reduced BET proteins to a greater extent than MZ1- treatment alone; OVCAR8 cells were treated with DMSO, 100 nM MZ1, (1 mM) GSK529A, RAD001, Lapatinib or the combination of MZ1 or JQ1 and kinase inhibitors; BET protein levels and the apoptotic marker cleaved PARP were determined by Western blot.

Figure 7D shows OVCAR8 cells were treated with DMSO, 100 nM JQ1, (1 pM) GSK529A, RAD001, Lapatinib or the combination of MZ1 or JQ1 and kinase inhibitors; BET protein levels and the apoptotic marker cleaved PARP were determined by Western blot.

Figure 7E shows that co-treating cells with BBDs and GSK529A or RAD001 enhances PROTAC degradation of BRD4, and increases apoptosis; OVCAR8 cells were treated with DMSO, 50 nM MZ1, (1 pM) GSK529A, or MZ1 and escalating doses of GSK529A or RAD001 and BRD4 levels and the apoptotic marker cleaved PARP were determined by Western blot.

Figure 7F shows OVCAR8 cells treated with DMSO, 50 nM MZ1, (1 pM) RAD001, or MZ1 and escalating doses of GSK529A or RAD001 and BRD4 levels and the apoptotic marker cleaved PARP were determined by Western blot.

Figure 7G shows selective BRD4 degradation but not BET protein inhibition sensitizes OC cells to RAD001, GSK529A or Lapatinib; OC cell lines were treated with 0.1 pM MZ1, 0.1 pM JQ1, escalating doses of GSK529A, Lapatinib, RAD001 or the combination of 0.1 pM MZ1 or JQ1 and escalating doses of GSK529A, Lapatinib or RAD001 for 5 days and cell viability determined by CellTiter-Glo.

Figure 7H shows that KURAMOCHI cells exhibit pronounced growth inhibition in response to dBET6 in combination with GSK529A; KURAMOCHI cells were treated with 100 nM GSK529A together with dBET6 or JQ1 for 5 days and cell viability was assessed by CellTiter-Glo.

Figure 71 shows KURAMOCHI cells treated with 100 nM RAD001 together with dBET6 or JQ1 for 5 days and cell viability was assessed by CellTiter-Glo.

Figure 7J shows a model of kinome adaptive bypass remodeling underlying protection of BRD4 from PROTAC-mediated degradation in OC cells and promoting resistance to BBDs. Figure 8 A shows BBD ARV825 with OTX015 as the warhead and a Cerebl on-binding moiety for E3 ligase recruitment.

Figure 8B shows that BBD-treatment reduces BET proteins inducing apoptosis to a greater extent than BBI-treatment; OVCAR8 cells were treated with increasing doses of ARV- 825 and BET protein levels were determined by Western blot; apoptosis was measured by assessing cleaved PARP levels by blot.

Figure 8C shows a more pronounced cleavage of PARP at lower doses with BBD- treatment single-run proteome analysis.

Figure 8D shows that BRD2, BRD3 and BRD4 proteins were reduced by BBD but not BBI-treatment, and induced protein levels of HEXIM1 was observed only with BBI-treatment.

Figure 8E shows reactome pathways upregulated by 48 hours of BBD threapy.

Figure 8F shows reactome pathways upregulated by 48 hours of BBI threapy.

Figure 8G shows analysis of global expression changes in response to MZ1 or JQ1 by RNA-seq.

Figure 8H shows analysis of global expression changes in response to MZ1 or JQ1 by RNA-seq.

Figure 81 shows proteins significantly repressed by 48 hours of BBD-treatment.

Figure 8J shows BET inhibitor MIB-signature (P<0.05).

Figure 8K shows PROTAC MIB-signature.

Figure 8L shows kinases exhibitng increased MIB-binding in response to BBI treatment.

Figure 8M shows common MIB-MS & RNA-seq response signatures for MZ1.

Figure 8N shows common MIB-MS & RNA-seq response signatures for JQ1.

Figure 80 shows RTK reprogramming induced by JQ1 -treatment are blocked by using BBDs; KURAMOCHI cells were treated with DMSO, JQ1 (500 nM) or MZ1 (500 nM) for 48 hours and protein levels determined by Western blot.

Figure 8P shows RTK reprogramming induced by JQ1 -treatment are blocked by using BBDs; COV362 cells were treated with DMSO, JQ1 (500 nM) or MZ1 (500 nM) for 48 hours and protein levels determined by Western blot.

Figure 9A shows that cells chronically exposed to JQ1 (JQ1-R) were unaffected by increasing doses of JQ1.

Figure 9B shows that JQl-treatment failed to reduce BRD4 target FOSL1.

Figure 9C shows that some degree of PROTAC degradation occurs in ARV-R cells.

Figure 9D shows that some degree of PROTAC degradation occurs in MZ1-R cells. Figure 9E shows RNA changes in BBD-R cells relative to parental cells.

Figure 9F shows protein levels changes in BBD-R cells relative to parental cells.

Figure 9G shows that BRD2 may function in contributing to MZ1 resistance through promoting MAPK signaling.

Figure 9H shows knockdown of BRD2 reduces RAF-MEK-ERK signaling in MZ1-R cells, as shown by Western blot; OVCAR8 MZ1-R cells were transfected with siRNAs targeting BRD2 or control siRNA, cultured for 120 hours, and kinase activities assessed by Western blot using phospho-antibodies.

Figure 91 shows MZ1-R cells display sensitivity towards CRBN-based BBD, dBET6; OVCAR8 parental or MZ1-R cells were treated with increasing doses of dBET6 for 5 days and cell viability assessed by CellTiter-Glo.

Figure 9J shows BET proteins reduced by CRBN-based dBET6 but not VHL-based, ARV-771; OVCAR8 parental cells were treated with 100 nM MZ1 and compared to MZ1-R cells treated with 100 or 500 nM of MZ1, ARV-771 or dBET6 and BET protein levels determined by Western blot.

Figure 9K shows ARV-R cells are cross-resistant to MZ1 -treatment; OVCAR8 parental ARV-R cells were treated with increasing doses of MZ1 for 5 days and cell viability assessed by CellTiter-Glo.

Figure 9L shows JQ1-R cells exhibit sensitivity to BBD-treatment; PEOl JQ1-R cells were treated with escalating doses of BBIs, JQ1, OTX015, AZD5153, or ARV825 for 5 days and cell viability assessed by CellTiter-Glo.

Figure 10A shows ARV-R I Parental MIB-signature.

Figure 10B shows JQ1-R I Parental MIB-signature.

Figure IOC shows ARV-R and MZ1-R MIB-MS kinome profiles cluster together and are distinct from JQ1-R or parental cells as shown by PC A and hierarchical clustering.

Figure 10D shows KEGG pathway analysis of kinases exhibiting induced MIB-binding in JQ1-R cells relative to parental cells.

Figure 10E shows common MIB-signature MZ1-R and JQ1-R.

Figure 10F shows KEGG pathway analysis of kinases exhibiting induced MIB-binding in MZ1-R cells relative to parental cells.

Figure 10G shows KEGG pathway analysis of proteins reduced in MZ1-R cells relative to parental cells (P < 0.05).

Figure 11A shows parental, JQ1-R or MZ1-R OVCAR8 cells were treated with escalating doses of LP-935509 for 5 days and cell viability was assessed by CellTiter-Glo; MZ1- R or JQ1-R inhibitor treated cell viabilities were normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully.

Figure 11B shows parental, JQ1-R or MZ1-R OVCAR8 cells treated with escalating doses of PND 11 for 5 days and cell viability was assessed by CellTiter-Glo; MZ1-R or JQ1-R inhibitor treated cell viabilities were normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully.

Figure 11C shows parental, JQ1-R or MZ1-R OVCAR8 cells treated with escalating doses of or Trametinib for 5 days and cell viability was assessed by CellTiter-Glo; MZ1-R or JQ1-R inhibitor treated cell viabilities were normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully.

Figure 11D shows parental, JQ1-R or MZ1-R OVCAR8 cells treated with escalating doses of or Y3009120 for 5 days and cell viability was assessed by CellTiter-Glo; MZ1-R or JQ1-R inhibitor treated cell viabilities were normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully.

Figure 11E shows parental, JQ1-R or MZ1-R OVCAR8 cells treated with escalating doses of or GDC0941 for 5 days and cell viability was assessed by CellTiter-Glo; MZ1-R or JQ1-R inhibitor treated cell viabilities were normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully.

Figure 1 IF shows parental, JQ1-R or MZ1-R OVCAR8 cells treated with escalating doses of or Cx-4945 for 5 days and cell viability was assessed by CellTiter-Glo; MZ1-R or JQ1- R inhibitor treated cell viabilities were normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully.

Figure 11G shows parental, ARV -R cells treated with escalating doses of GSK1904529A for 5 days and cell viability was assessed by CellTiter-Glo; ARV-R inhibitor treated cell viabilities were normalized to DMSO treated ARV-R cells, respectfully.

Figure 11H shows parental, ARV-R cells treated with escalating doses of lapatinib for 5 days and cell viability was assessed by CellTiter-Glo; ARV-R inhibitor treated cell viabilities were normalized to DMSO treated ARV-R cells, respectfully.

Figure 111 shows parental, ARV-R cells treated with escalating doses of or RAD001 for 5 days and cell viability was assessed by CellTiter-Glo; ARV-R inhibitor treated cell viabilities were normalized to DMSO treated ARV-R cells, respectfully.

Figure 11 J shows parental, ARV-R cells treated with escalating doses of MK2206 for 5 days and cell viability was assessed by CellTiter-Glo; ARV-R inhibitor treated cell viabilities were normalized to DMSO treated ARV-R cells, respectfully. Figure 11K shows parental, ARV-R cells treated with escalating doses of or LP-935509 for 5 days and cell viability was assessed by CellTiter-Glo; ARV-R inhibitor treated cell viabilities were normalized to DMSO treated ARV-R cells, respectfully.

Figure 11L ARV-R cells are more sensitive to MTORC1 inhibition than parental cells; parental or MZ1-R OVCAR8 cells were treated with escalating doses of GSK1904529A for 5 days and cell viability assessed by CellTiter-Glo; ARV-R inhibitor treated cell viabilities normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully.

Figure 12A shows proteome response to GSK529A in MZ1-R cells.

Figure 12B shows MZ1-R OVCAR8 cells with escalating does of Ck-4945 or DMSO for 4 hours and BET protein levels were determined by Western blot.

Figure 12C shows ARV-R OVCAR8 cells with 1 mM of kinase inhibitors or DMSO for 4 hours and BET protein levels were determined by Western blot.

Figure 12D shows MZ1-R OVCAR8 cells with 1 pM of kinase inhibitors or DMSO for 4 hours and BET protein levels were determined by Western blot.

Figure 12E shows JQ1-R OVCAR8 cells with 1 pM of kinase inhibitors or DMSO for 4 hours and BET protein levels were determined by Western blot.

Figure 13 A shows that knockdown of BRD4 and to a lesser extent BRD2 inhibited cell viability of KURAMOCHI MZ1-R cells.

Figure 13B shows that knockdown of BRD4 and to a lesser extent BRD2 inhibited cell viability of COV362 MZ1-R cells.

Figure 13C shows that knockdown of BRD4 and to a lesser extent BRD2 inhibited cell viability of OVCAR5 MZ1-R cells.

Figure 13D shows growth curves afret treatment of OVCAR5 JQ1-R cells with GSK529A.

Figure 13E shows growth curves afret treatment of OVCAR5 JQ1-R cells with lapatinib.

Figure 13F shows growth curves afret treatment of OVCAR5 JQ1-R cells with RADOOl.

Figure 14A shows combined treatment of OVCAR8 cells with JQ1 or MZ1 and Vx-90.

Figure 14B shows combined treatment of OVCAR8 cells with JQ1 or MZ1 and GDC0941.

Figure 14C shows combined treatment of OVCAR8 cells with JQ1 or MZ1 and trametinib.

Figure 14D shows combined treatment of OVCAR8 cells with JQ1 or MZ1 and THZ-1. Figure 14E shows combined treatment of OVCAR8 cells with JQ1 or MZ1 and NVP-2.

Figure 14F shows combined treatment of OVCAR8 cells with JQ1 or MZ1 and CX-

4945.

Figure 14G shows combined treatment of OVCAR8 cells with JQ1 or MZ1 and RADOOl.

Figure 14H shows combined treatment of OVCAR8 cells with JQ1 or MZ1 and GSK529A.

Figure 141 shows BDT4 protein levels in OVCAR8 cells following combined treatment of with JQ1 and GSK529A.

Figure 14J shows treatment of RAS-altered EOC cell lines OVCAR5, JHOS-2, A1847, COV362 and CAOV3, or PIK3CA mutant SKOV3 cells or KURMAOCHI with JQ1 or MZ1 and GSK1904529 A.

Figure 14K shows treatment of RAS-altered EOC cell lines OVCAR5, JHOS-2, A1847, COV362 and CAOV3, or PIK3CA mutant SKOV3 cells or KURAMOCHI with JQ1 or MZ1 and Lapatinib.

Figure 14L shows treatment of RAS-altered EOC cell lines OVCAR5, JHOS-2, A1847, COV362 and CAOV3, or PIK3CA mutant SKOV3 cells or KURMAOCHI with JQ1 or MZ1 and RADOOL

Figure 14M shows OVSAHO have the highest protein levels of BRD4 and they exhibit exhibit sensitivity towards the MZ1 and MTOR combination.

Figure 14N shows treatment of OVSAHO cells with MZ1, GSK529A, lapatinib, or RADOOl alone, or a combination of MZ1 with each of GSK529A, lapatinib, or RADOOl.

Figure 15A shows treatment of OVCAR8 cells with CDK9-degrader Thai SNS 032 reduced CDK9 protein levels as determined by Western blot.

Figure 15B shows chronic exposure to Thai SNS 032 failed to degrade CDK9 that is overcome by GSK529A, lapatinib or RADOOl treatment, where protein levels were assessed by Western blot.

Figure 15C shows Thal-R cells were more sensitive to INSR inhibition than parental cells, where cell viability assessed by CellTiter-Glo.

Figure 15D shows Thal-R cells were more sensitive to ErbB inhibition than parental cells, where cell viability assessed by CellTiter-Glo.

Figure 15E shows Thal-R cells were more sensitive to MTORC1 inhibition than parental cells, where cell viability assessed by CellTiter-Glo. Figure 15F shows therapies involving GSK529A alone and the combination of GSK529A and Thai SNS 032, which improved growth repression of OVCAR8 cells, where cell viability was assessed by CellTiter-Glo.

Figure 15G shows therapies involving RAD001 alone and the combination of RAD001 and Thai SNS 032, which improved growth repression of OVCAR8 cells, where cell viability was assessed by CellTiter-Glo.

Figure 16 shows MTORC1 inhibition enhances PROTAC-meditated degradation of BRD4, CDK9 or FAK1 proteins in cancer cells: treated with DMSO, RAD001, increasing doses of dBET6 or RAD001 + increasing doses of dBET6 and BRD4 protein assessed by WB (Panel A); treated with DMSO, RAD001, increasing doses of Thai SNS 032 or RAD001 + increasing doses of Thai SNS 032 and CDK9 protein assessed by WB (Panel B); and treated with DMSO, RAD001, increasing doses of FAK-Protac or RAD001 + increasing doses of FAK-Protac and CDK9 protein assessed by WB (Panel C).

Figure 17 shows the blockade of MTORC1 signaling sensitizes cancer cells resistant to BET-PROTACs by: a dose response matrix of RAD001 or dBET6 in A1847 cells (Panel A); immunoblot analysis of BRD4 protein levels (Panel B); drug synergy analysis of RAD001 and dBET6 in DLD-1 cells (Panel C); and spheroid assays (Panel D).

Figure 18 shows activation of K-ras signaling promotes resistance to BET-PROTACs that can be reversed by MTORC1 inhibitors; expression of K-ras mutants promote resistance to MZ1 therapy (Panel A); immunoblot depicts activation of translation by forced expression of K- ras G12D mutants in SW48 cells (Panel B); immunoblot analysis of BRD4 protein levels (Panel C); dose synergy analysis of RAD001 or MZ1 in K-ras G12D mutant SW48 cells (Panels D and E); immunoblot for BRD4 protein levels in response to combination (Panel F); dose synergy analysis of RAD001 or MZ1 in K-ras wild-type SW48 cells (Panels G and H); and immunoblot for BRD4 protein levels in response to combination (Panel I).

Description Of Embodiments

Various terms relating to aspects of the present disclosure are used throughout the specification and claims. Such terms are to be given their ordinary meaning in the art, unless otherwise indicated. Other specifically defined terms are to be construed in a manner consistent with the definitions provided herein.

Unless otherwise expressly stated, it is in no way intended that any method or aspect set forth herein be construed as requiring that its steps be performed in a specific order.

Accordingly, where a method claim does not specifically state in the claims or descriptions that the steps are to be limited to a specific order, it is in no way intended that an order be inferred, in any respect. This holds for any possible non-expressed basis for interpretation, including matters of logic with respect to arrangement of steps or operational flow, plain meaning derived from grammatical organization or punctuation, or the number or type of aspects described in the specification.

As used herein, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise.

As used herein, the term “about” means that the recited numerical value is approximate and small variations would not significantly affect the practice of the disclosed embodiments. Where a numerical value is used, unless indicated otherwise by the context, the term “about” means the numerical value can vary by ±10% and remain within the scope of the disclosed embodiments.

As used herein, the term “comprising” may be replaced with “consisting” or “consisting essentially of’ in particular embodiments as desired.

In the claims, as well as in the specification above, all transitional phrases such as “comprising,” “including,” “carrying,” “having,” “containing,” “involving,” “holding,” “composed of,” and the like are to be understood to be open-ended, i.e., to mean including but not limited to.

It should also be understood that, in certain methods described herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited unless the context indicates otherwise.

As used herein, the terms “subject” and “patient” are used interchangeably. A subject may include any animal, including mammals. Mammals include, but are not limited to, farm animals (such as, for example, horse, cow, pig), companion animals (such as, for example, dog, cat), laboratory animals (such as, for example, mouse, rat, rabbits), and non-human primates. In some embodiments, the subject is a human.

The terms “cancer” and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth/proliferation. A “tumor” comprises one or more cancerous cells. Examples of cancer are provided elsewhere herein.

As used herein in the specification and in the claims, the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from anyone or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements. This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified. Thus, as a nonlimiting example, “at least one of A and B” (or, equivalently, “at least one of A or B,” or, equivalently “at least one of A and/or B”) can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.

As used herein, unless defined otherwise in a claim, the term “optionally” means that the subsequently described event(s) may or may not occur, and includes both event(s) that occur and event(s) that do not occur.

As used herein, unless defined otherwise, the phrase “optionally substituted”, “substituted” or variations thereof denote an optional substitution, including multiple degrees of substitution, with one or more substituent group, for example, one, two or three. The phrase should not be interpreted as duplicative of the substitutions herein described and depicted.

It should be appreciated that particular features of the disclosure, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment. Conversely, various features of the disclosure which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable subcombination.

It should be understood that stereoisomers (including diastereomers and enantiomers) of the compounds described herein, as well as mixtures thereof, are within the scope of the present disclosure. By way of non-limiting example, the mixture may be a racemate or the mixture may comprise unequal proportions of one particular stereoisomer over the other. Additionally, the compounds can be provided as a substantially pure stereoisomers. Diastereomers include, for example, cis-trans isomers, E-Z isomers, conformers, and rotamers. Methods of preparation of stereoisomers are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated herein. Cis and trans geometric isomers of the compounds are also included herein and can be isolated as a mixture of isomers or as separated isomeric forms. Where a compound capable of stereoisomerism or geometric isomerism is designated in its structure or name without reference to specific R/S or cis/trans configurations, it is intended that all such isomers are contemplated.

Appropriate compounds described herein may also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Examples of prototropic tautomers include, but are not limited to, ketone-enol pairs, amide-imidic acid pairs, lactam-lactim pairs, amide-imidic acid pairs, enamine-imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system including, but not limited to, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 1,2, 4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.

The compounds described herein also include hydrates and solvates, as well as anhydrous and non-solvated forms.

The compounds described herein also include derivatives referred to as prodrugs, which can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds. Examples of prodrugs include compounds as described herein that contain one or more molecular moieties appended to a hydroxyl, amino, sulfhydryl, or carboxyl group of the compound, and that when administered to a patient, cleaves in vivo to form the free hydroxyl, amino, sulfhydryl, or carboxyl group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups in the compounds described herein. Preparation and use of prodrugs is discussed in T. Higuchi et al, “Pro-drugs as Novel Delivery Systems,” Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.

The present disclosure is based on a surprising and unexpected discovery that resistance of cancer to bromodomain and extra terminal domain (BET) proteolysis targeting chimera (PROTAC) (BET-PROTAC) therapeutic agents or one or more cyclin-dependent kinase 9 (CDK9) PROTAC (CDK9-PROTAC) therapeutic agents can be overcome by inhibiting one or more cell signaling pathways including RTK signaling pathway, mTOR signaling pathway, CDK7 signaling pathway, KRAS signaling pathway or an autophagy signaling pathway. The term “PROTAC” refers to proteolysis-targeting chimera molecules having generally three components, an E3 ubiquitin ligase binding group (E3LB), a linker L, and a protein binding group (PB).

The term “linker”, “linker unit”, or “link” as used herein means a chemical moiety comprising a chain of atoms that covalently attaches a PROTAC moiety to an antibody, or a component of a PROTAC to another component of the PROTAC. In some embodiments, a linker is a divalent radical.

The term “ubiquitin ligase” refers to a family of proteins that facilitate the transfer of ubiquitin to a specific substrate protein, targeting the substrate protein for degradation. For example, cereblon is an E3 ubiquitin ligase protein that alone or in combination with an E2 ubiquitin-conjugating enzyme causes the attachment of ubiquitin to a lysine on a target protein, and subsequently targets the specific protein substrates for degradation by the proteasome. Thus, E3 ubiquitin ligase alone or in complex with an E2 ubiquitin conjugating enzyme is responsible for the transfer of ubiquitin to targeted proteins. In general, the ubiquitin ligase is involved in polyubiquitination such that a second ubiquitin is attached to the first; a third is attached to the second, and so forth. Polyubiquitination marks proteins for degradation by the proteasome. However, there are some ubiquitination events that are limited to mono-ubiquitination, in which only a single ubiquitin is added by the ubiquitin ligase to a substrate molecule. Mono- ubiquitinated proteins are not targeted to the proteasome for degradation, but may instead be altered in their cellular location or function, for example, via binding other proteins that have domains capable of binding ubiquitin. Further complicating matters, different lysines on ubiquitin can be targeted by an E3 to make chains. The most common lysine is Lys48 on the ubiquitin chain. This is the lysine used to make polyubiquitin, which is recognized by the proteasome.

MTORC1 inhibition may represent a general therapeutic strategy to improve PROTAC- induced degradation of protein targets and improve efficacy of PROTACs by minimizing concentrations of PROTACs required for reduction of protein targets in cells. Without being limited to any particular theory, it is believed that chronic exposure to BET-PROTAC (which targets BRD4 for degradation) triggers reprogramming of cancer cell kinome via CDK9- dependent upregulation of BRD4 transcription and activation of INSR, ErbB and mTORCl- signaling, thereby protecting BRD4 from PROT AC-mediated degradation. This effect is believed to underlie BET-PROTAC therapy resistance in cancer cells. It is believed that inhibiting these signaling pathways may suppress this resistance mechanism thereby restoring cancer cell sensitivity to BRD4 for degradation. Thus, cancer cells intrinsically resistant to BET- PROTACs can be made sensitive by blocking mTORCl signaling, supporting combination therapies involving MTORCl and BET protein degraders for the treatment of these types of cancers.

Accordingly, the present disclosure provides pharmaceutical compositions comprising: one or more bromodomain and extra terminal domain (BET) proteolysis targeting chimera (PROTAC) (BET-PROTAC) therapeutic agents or one or more cyclin-dependent kinase 9 (CDK9) PROTAC (CDK9-PROTAC) therapeutic agents, or a combination thereof; and one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof.

Proteolysis targeting chimeras are described, for example, in U. S. Patent Application Publication No. US 2019/0175612, U.S. Pat. No. 7,208,157, and PCT Publications No. WO2013/106643, WO2013/106646, and WO2015/. PROTACs have a general structure E3LB- L-PB; wherein, E3LB is an E3 ligase binding group covalently bound to L; L is a linker covalently bound to E3LB and PB; PB is a protein binding group covalently bound to L.

E3 ubiquitin ligases (of which over 600 are known in humans) confer substrate specificity for ubiquitination. There are known ligands which bind to these ligases. As described herein, an E3 ubiquitin ligase binding group is a peptide or small molecule that can bind an E3 ubiquitin ligase.

Specific E3 ubiquitin ligases include: von Hippel-Lindau (VHL); cereblon, XIAP, E3A; MDM2; Anaphase-promoting complex (APC); UBR5 (EDD1); SOCS/BC- box/eloBC/CUL5/RING; LNXp80; CBX4; CBLL1; HACE1; HECTD1; HECTD2; HECTD3; HECW1; HECW2; HERC1; HERC2; HERC3; HERC4; HUWE1; ITCH; NEDD4; NEDD4L; PPIL2; PRPF19; PIAS1; PIAS2; PIAS3; PIAS4; RANBP2; RNF4; RBX1; SMURF1; SMURF2; STUB1; TOPOR5; TRIP12; UBE3A; UBE3B; UBE3C; UBE4A; UBE4B; UBOX5; UBR5; WWP1; WWP2; Parkin; A20/TNFAIP3; AMFR/gp78; ARA54; beta-TrCPl/BTRC; BRCA1; CBL; CHIP/STUB 1; E6; E6AP/UBE3A; F-box protein 15/FBX015; FBXW7/Cdc4; GRAII/RNF128; HOIP/RNF31; cIAP-l/HIAP-2; cIAP-2/HIAP-l; cIAP (pan); ITCH/AIP4; KAP1; MARCH8; Mind Bomb 1/MIBl; Mind Bomb 2/MIB2; MuRFl/TRIM63; NDFIP1; NEDD4; NleL; Parkin; RNF2; RNF4; RNF8; RNF168; RNF43; SART1; Skp2; SMURF2; TRAF-1; TRAF-2; TRAF-3; TRAF-4; TRAF-5; TRAF-6; TRIM5; TRIM21; TRIM32; UBR5; and ZNRF3.

In some embodiments, E3 ubiquitin ligase is von Hippel-Lindau (VHL) tumor suppressor, the substrate recognition subunit of the E3 ligase complex VCB, which also consists of elongins B and C, Cul2 and Rbxl. The primary substrate of VHL is Hypoxia Inducible Factor 1a (HIF-Ia), a transcription factor that upregulates genes such as the pro-angiogenic growth factor VEGF and the red blood cell inducing cytokine erythropoietin in response to low oxygen levels. Compounds that bind VHL may be hydroxyproline compounds such as those disclosed in WO2013/106643, and other compounds described in US2016/0045607, WO2014187777, US20140356322, and U.S. Pat. No. 9,249,.

In some embodiments, E3 ubiquitin ligase is X-linked inhibitor of apoptosis (XIAP). XIAP is a protein that stops apoptotic cell death. Deregulation of XIAP has been associated with cancer, neurodegenerative disorders and autoimmunity. In the development of lung cancer, the overexpression of XIAP inhibits caspases. In developing prostate cancer, XIAP is one of four IAPs overexpressed in the prostatic epithelium. Mutations in the XIAP gene can result in a severe and rare type of inflammatory bowel disease. Defects in the XIAP gene can also result in an extremely rare condition called X-linked lymphoproliferative disease. Degradation of XIAP can enhance apoptosis by preventing XIAP from binding to caspases. This allows normal caspase activity to proceed.

Examples of small molecular binding compounds for XIAP include compounds disclosed in U.S. Pat. No. 9,096,544; WO 2015187998; WO 2015071393; U.S. Pat. Nos. 9,278,978; 9,249,151; US 20160024055; US 20150307499; US 20140135270; US 20150284427; US 20150259359; US 20150266879; US 20150246882; US 20150252072; US 20150225449; U.S. Pat. No. 8,883,771 , J. Med. Chem, 2015, 58(16) 6574-6588 and Small- molecule Pan-IAP Antagonists: A Patent Review (2010) Expert Opin Ther Pat; 20: 251-67 (Flygare & Fairbrother).

In some embodiments, E3 ubiquitin ligase is MDM2. Examples of small molecular binding compounds for MDM2 include the “nutlin” compounds, e.g., nutlin 3a and nutlin 3.

Thalidomide, lenalidomide, pomalidomide and analogs thereof are known to bind to cereblon. The crystal structure of cereblon (CRBN) with thalidomide and derivative compounds are described in US2015/0374678. Other small molecule compounds that bind to cereblon are also known, e.g., the compounds disclosed as an in US2016/0058872 and US2015/0291562. Further, phthalimide conjugation with binders, such as antagonists, of BET bromodomains can provide PROTACs with highly-selective cereblon-dependent BET protein degradation. Winter et al, Science, Jun. 19, 2015, 1376. Such PROTACs can be conjugated to an antibody as described herein to form a PAC. Additional E3 ligase binding groups are described, for example, in the U. S. Patent Application Publication No. 2019/0175612.

The PB component is a group which binds to a target protein intended to be degraded. The term “protein” includes oligopeptides and polypeptide sequences of sufficient length that they can bind to a PB group. Any protein in a eukaryotic system or a microbial system, including a virus, bacteria or fungus, as otherwise described herein, are targets for ubiquitination mediated by the compounds described herein.

PB groups include, for example, any moiety which binds to a protein specifically (binds to a target protein) and includes the following non-limiting examples of small molecule target protein moieties: Hsp90 inhibitors, kinase inhibitors (such as CDK9 inhibitors), MDM2 inhibitors, compounds targeting Human BET Bromodomain-containing proteins, HD AC inhibitors, human lysine methyltransferase inhibitors, angiogenesis inhibitors, immunosuppressive compounds, and compounds targeting the aryl hydrocarbon receptor (AHR), among numerous others.

In some embodiments, PB group specifically binds one or more Bromodomain and Extra-Terminal motif (BET) proteins. BET is a subfamily of proteins responsible for recognition acetylated lysine residues, such as those on the N-terminal tails of histones, and include BRD2, BRD3, BRD4 and BRDT (see WO 2011/143669). In some embodiments, the BET-specific PB group comprises an anti-BET antibody. In some embodiments the BET-specific PB group comprises a BET inhibitor that is specific for one or more BET proteins. Numerous BET inhibitors are known in the art and are described, for example in Doroshow et al, Annals of Oncology, 2017, 28, 1776-1787; Perez-Salviaa and Esteller Epigenetics, 2017, 12, 323-339; Klein, RMD Open, 2018, 4:e000744; Ocana et al, Oncotarget, 2017, 8, 71285-71291; Hogg et al, Blood, 2017, 130, 2537; U.S. Patent Application Publications 2018/0117030, 2016/0095867, 2018/0117165, PCT Publications WO 2011/054843, WO 2009/084693, 20180117165, and JP2008-156311.

In some embodiments, the BET inhibitors include, without limitation, olinone, JQ1, iBET, RVX-208, PF-1, ABBV-075, BAY1238097, BI 894999, BMS-986158, CPI-0610, FT- 1101, GS-5829, GSK525762/I-BET762, GSK2820151/I-BET151, INCB054329, OTX015/MK- 8628, PLX51107, R06870810/TEN-010, ZEN003694, CPI203, PFI-1, MS436, RVX2135, BAY-299, 1-BET762, RVX297, SF1126, INCB054329, INCB057643, R06870810, LY294002, AZD5153, MT-1, MS645 and RG6146.

In some embodiments PB group specifically binds Cyclin-Dependent Kinase 9 (CDK9) Protein. In some embodiments, the CDK9-specific PB group comprises an anti-CDK9 antibody. In some embodiments the CDK9-specific PB group comprises a CDK9 inhibitor. Numerous BET inhibitors are described, for example in Krystof, Medicinal Research Reviews, 2009, DOI 10.1002/med.24172; U.S. Patent Application Publications 2017/0304315, 2017/0173021, 2015/0329537, 2014/0287454, and PCT Publcations WO 2009/047359, WO 2010/003133, WO 2008/79933 and WO 2011/012661.

In some embodiments, the CDK9 inhibitors include, without limitation, NVP-2, LDC000067, SNS-032 (BMS-387032), AT7519, P-276-00, AZD5438, PHA-767491, PHA- 793887, PHA-848125, BAY 1143572, BAY 1112054, Cdk9 inhibitor II (CAS 140651-18-9 from Calbiochem), DRB, AZD-5438, SNS-032, dinaciclib, LY2857785, flavopiridol, purvalanol B, CDKI-71, CDKI-73, CAN508, FIT-039, CYC065, Ro-3306, 3,4-dimethyl-5-[2-(4-piperazin-l- yl-phenylamino)-pyrimidin-4-yl]-3H-thiazol-2-one, wogonin, apigenin, chrysin, luteolin, 4- methyl-5-[2-(3-nitroanilino)pyrimidin-4-yl]-l,3-thiazol-2-am ine, 1073485-20-7P and Cpd Bl.

The E3LB and PB groups of PROTACs as described herein can be connected with linker (L). In certain embodiments, the linker group L is a group comprising one or more covalently connected structural units of A (e.g., -Ai . . . Aq-), wherein Ai is a group coupled to at least one of a E3LB, a PB, or a combination thereof. In certain embodiments, Ai links a E3LB, a PB, or a combination thereof directly to another E3LB, PB, or combination thereof. In other embodiments, Ai links a EL3B, a PB, or a combination thereof indirectly to another E3LB, PB, or combination thereof through A q .

In certain embodiments, q is an integer greater than or equal to 0. In certain embodiments, q is an integer greater than or equal to 1.

In some embodiments, e.g., where q is greater than 2, A q is a group which is connected to an E3LB moiety, and Ai and A q are connected via structural units of A (number of such structural units of A: q-2). In some embodiments, e.g., where q is 2, A q is a group which is connected to Ai and to an E3LB moiety. In some embodiments, e.g., where q is 1, the structure of the linker group L-Ai-, and Ai is a group which is connected to an E3LB moiety and a PB moiety.

In additional embodiments, q is an integer from 1 to 100, 1 to 90, 1 to 80, 1 to 70, 1 to 60, 1 to 50, 1 to 40, 1 to 30, 1 to 20, or 1 to 10.

In some embodiments, the linker group is an optionally substituted (poly)ethyleneglycol having from 1 to about 100 ethylene glycol units, from 1 to about 50 ethylene glycol units, from 1 to about 25 ethylene glycol units, from 1 to about 10 ethylene glycol units, from 1 to about 8 ethylene glycol units, and from 1 to about 6 ethylene glycol units, from 2 to 4 ethylene glycol units, or optionally substituted alkyl groups interdispersed with optionally substituted, O, N, S, P or Si atoms. In some embodiments, the linker is substituted with an aryl, phenyl, benzyl, alkyl, alkylene, or heterocycle group. In some embodiments, the linker may be asymmetric or symmetrical. In any of the embodiments of the compounds described herein, the linker group may be any suitable moiety as described herein. In some embodiments, the linker is a substituted or unsubstituted polyethylene glycol group ranging in size from about 1 to about 12 ethylene glycol units, from 1 to about 10 ethylene glycol units, from 2 to about 6 ethylene glycol units, from 2 to about 5 ethylene glycol units, or from 2 to 4 ethylene glycol units.

Although the E3LB group and PB group may be covalently linked to the linker group through any group which is appropriate and stable to the chemistry of the linker. The linker can be independently covalently bonded to the E3LB group and the PB group preferably through an amide, ester, thioester, keto group, carbamate (urethane), carbon or ether, each of which groups may be inserted anywhere on the E3LB group and PB group to provide maximum binding of the E3LB group on the ubiquitin ligase and the PB group on the target protein to be degraded. In some embodiments where the PB group is an E3LB group, the target protein for degradation may be the ubiquitin ligase itself. In some embodiments, the linker may be linked to an optionally substituted alkyl, alkylene, alkene or alkyne group, an aryl group or a heterocyclic group on the E3LB and/or PB groups. It is noted that an E3LB group or a PB group may need to be derivatized to make a chemical functional group that is reactive with a chemical functional group on the linker. Alternately, the linker may need to be derivatized to include a chemical functional group that can react with a functional group found on E3LB and/or PB.

Although the E3LB group and PB group may be covalently linked to the linker group through any group which is appropriate and stable to the chemistry of the linker, in preferred aspects, the linker is independently covalently bonded to the E3LB group and the PB group through an amide, ester, thioester, keto group, carbamate (urethane) or ether, each of which groups may be inserted anywhere on the E3LB group and PB group to allow binding of the E3LB group to the ubiquitin ligase and the PB group to the target protein to be degraded. In other words, as shown herein, the linker can be designed and connected to E3LB and PB to minimize, eliminate, or neutralize any impact its presence might have on the binding of E3LB and PB to their respective binding partners. In certain aspects, the targeted protein for degradation may be a ubiquitin ligase.

Additional linkers L are disclosed in US Application Publication Nos. 2019/0175612, 2016/0058872; 2016/0045607; 2014/0356322; and 2015/0291562, and W02014/063061.

Multiple BET-specific POTACS assembled from, inter alia, the components described above are described in, for example in Sun et al, Leukemia, 2018, 32, 343-352; Zhang et al, Mol. Cancer Ther., 2019, 18, 1302-1311; Yang et al, Drug Discovery Today: Technologies, 2019, 31, 43-51; Raina et al, PNAS, 2016, 113, 7124-7129; and Zou et al, Cell Biochem Fund, 2019, 37, 21-30; An and Fu, EBioMedicine, 2018, 36, 553-562.

In some embodiments, the BET -PROTACS include, without limitation, MZ1, ARV- 771, ATI, MZP-61, MZP-51, MZP-55, ARV825, dBET6, dBET57, dBET23, ZXH 3-26, BETd246, BETd260, QCA570, or ARCC-29, A1874, and CFT-2718.

Multiple CDK9-specific POTACS assembled, inter alia, from the components described above are described in, for example in Olson et al, Nat. Chem. Biol., 2018, 14, 163-170 and Robb et al, Chem. Commun. (Camb), 2017, 53, 7577-7580.

In some embodiments, the CDK9-PROTACS include, without limitation, THAL SNS 032, PROTAC3, and CDK9 Degrader- 1.

In some embodiments, the compositions comprise a single PROTAC therapeutic agent. In some embodiments, the compositions comprise multiple PROTAC agents comprising any combination of the PROTAC agents described herein.

In some embodiments, the compositions further comprise one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors. In some embodiments the kinase pathway inhibitors inhibit kinase signaling pathways including, without limitation, a receptor tyrosine kinase (RTK) pathway inhibitor, a mammalian target of rapamycin (MTOR) pathway inhibitor, or a CDK7 pathway inhibitor.

RTK family of kinases are described, for example, by Bertrand et al, The International Journal of Developmental Biology, 61(10-11-12), 697-722 and Regad, Cancers 2015, 7, 1758- 1784. In addition to RTK, the family includes, inter alia, Insulin Receptor (INSR/IGF1R), Epidermal Growth Factor Receptor family (EGFR/ERBB/HER2/ HER3/HER4), Proto- Oncogene Tyrosine-Protein Kinase (MERTK), Macrophage-Stimulating Protein Receptor 1 (MST1R) and Fibroblast Growth Factor Receptor 1 (FGFR1), any of which can be targeted for inhibition according to the methods of the present disclosure. In some embodiments, the RTK pathway inhibitor targets RTK directly. RTK inhibitors include, without limitation, GTP 14564,

R 1530, imatinib, Sorafenib, Pazopanib, Cabozantinib, Sunitinib, Crizitonib, Regorafenib, and Dovitinib. In some embodiments, the RTK pathway inhibitor is an EGFR inhibitor. EGFR inhibitors include, without limitation, Trastuzumab, Panitumumab, Cetuximab, Afatinib, Sapitinib, Neratinib, Theliatinib, Avitinib, Canertinib, AG-490, CP-724714, Dacomitinib, WZ4002, CUDC-101, AG-1478, PD153035, Pelitinib, AC480, AEE788, OSI-420, WZ3146, ARRY-380, AST-1306, Rociletinib, Genistein, Varlitinib, Icotinib, TAK-285, WHI-P154, Daphnetin, PD168393, Tyrphostin 9, CNX-2006, AG-18, AZ5104, Osimertinib, CL-387785, Olmutinib, (-)-Epigallocatechin Gallate(EGCG), AZD3759, Poziotinib, Naquotinib, Chrysophanol, Nazartinib, Norcantharidin, Lifirafenib, Lidocaine hydrochloride, Butein,

EAI045, NSC228155, Erlotinib, Gefitinib, and Lapatinib. In some embodiments, the RTK pathway inhibitor is an MERTK inhibitor. MERTK inhibitors include, without limitation, UNC1062, MRX-2843, RXDX-106, UNC2541, and unc2025. In some embodiments, the RTK pathway inhibitor is an MST1R inhibitor. MST1R inhibitors include, without limitation, LY2801653 dihydrochloride, BMS777607, and PHA 665752. In some embodiments, the RTK pathway inhibitor is an FGFR1 inhibitor. FGFR1 inhibitors include, without limitation, Ponatinib, BGJ398, Nintedanib, PD173074, Dovitinib, Alofanib, Gambogenic Acid, Derazantinib, Nintedanib, AZD4547, Danusertib, Brivanib, Dovitinib Dilactic acid, Dovitinib Lactate, MK- 2461, SSR128129E, LY2874455, H3B-6527, Erdafitinib, NSC12, S49076, BLU-554, PRN1371, PD-166866, PD-166866, FIIN-2, CH5183284, BLU9931, and SUN11602. mTOR pathway inhibitors are described, for example, by Harter et al., PLoS ONE,

2015, 10, e0127123. In addition to MTOR, the signaling pathway includes, inter alia, Phosphoinositide 3-Kinase (PI3K), p70S6 kinase (P70S6K), Phosphatidylinositol 4-Kinase Type 2 Alpha (PI4K2A), and CDK9, any of which can be targeted for inhibition according to the methods of the present disclosure. In some embodiments, the mTOR inhibitor targets the mTOR kinase directly. mTOR inhibitors include, without limitation, Dactolisib (BEZ235 or NVP- BEZ235), Rapamycin (Sirolimus), Everolimus (RAD001), AZD8055, Temsirolimus (CCI-779 or NSC 683864), PI-103, KU-0063794, Torkinib (PP242), Tacrolimus (FK506), Ridaforolimus (Deforolimus or MK-8669), Sapanisertib (INK 128, MLN0128, or TAK-228), Voxtalisib (SAR245409 or XL765) Analogue, Torin 1, Omipalisib (GSK2126458 or GSK458), OSI-027, PF-04691502, Apitolisib (GDC-0980 or RG7422), GSK1059615, WYE-354, Gedatolisib (PF- 05212384 or PKI-587), Torin 2, WYE-125132 (WYE-132), Vistusertib (AZD2014), BGT226 (NVP-BGT226), Palomid 529 (P529), PP121, WYE-687, WAY-600, ETP-46464, GDC-0349, XL388, Zotarolimus (ABT-578), LY3023414, CC-115, MHY1485, CZ415, GDC-0084, Voxtalisib (XL765 or SAR245409), 3BDO, Bimiralisib (PQR309), CC-223, and SF2523. In some embodiments, the mTOR pathway inhibitor is an PI3K inhibitor. PI3K inhibitors include, without limitation, compound 7n, Dactolisib, Pictilisib (GDC-0941), LY294002, Buparlisib, IC- 87114, Wortmannin, XL147 analogue, ZSTK474, Apitolisib, AS-605240, 3-Methyladenine, PIK-90, PF-04691502, AZD6482, Apitolisib, GSK1059615, Duvelisib, Gedatolisib, TG100-115, AS-252424, BGT226, CUDC-907, AS-604850, GSK2636771, Copanlisib, CH5132799, CAY10505, PIK-293, PKI-402, TGI 00713, VS-5584, Taselisib, CZC24832, SF2523, AZD8835, AMG319, Seletalisib, TGR-1202, Pilaralisib, Bimiralisib, IPI-549, GDC-0084, Voxtalisib, HS- 173, PF-4989216, Tenalisib, 740 Y-P, Leniolisib, GNE-317, LY3023414, GSK2292767, AZD8186, 2-D08, Nemiralisib, PI-103, Vistusertib, BGT226, and CC-223. In some embodiments, the mTOR pathway inhibitor is an P70S6K inhibitor. P70S6K inhibitors include, without limitation, BI-D1870, AT7867, PF-4708671, AD80, AT13148, LY2584702, and LY2584702 Tosylate. In some embodiments, the mTOR pathway inhibitor is an PI4K2A inhibitor. PI4K2A inhibitors include, without limitation, PIK-93, PI-273, NA04, NB04, NE02, NC02, NC03, NB02, NC04, ND02, NE03, NF03, NF04, NG02, NG03, NH02, NC02-567, and NC02-770 (see Sengupta et al, 2019 The Journal of Lipid Research, 60(3):683-693). In some embodiments, the mTOR pathway inhibitor is an CDK9 inhibitor. CDK9 inhibitors include, without limitation, NVP-2, LDC000067, SNS-032 (BMS-387032), AT7519, P-276-00, AZD5438, PHA-767491, PHA-793887, PHA-848125, BAY 1143572, BAY 1112054, Cdk9 inhibitor II (CAS 140651-18-9 from Calbiochem), DRB, AZD-5438, SNS-032, dinaciclib, LY2857785, flavopiridol, purvalanol B, CDKI-71, CDKI-73, CAN508, FIT-039, CYC065, Ro- 3306, 3,4-dimethyl-5-[2-(4-piperazin-l-yl-phenylamino)-pyrimidin-4 -yl]-3H-thiazol-2-one, wogonin, apigenin, chrysin, luteolin, 4-methyl-5-[2-(3-nitroanilino)pyrimidin-4-yl]-l,3-thiazol- 2-amine, 1073485-20-7P and Cpd Bl.

The combination therapies described herein are considered to be global to improve the efficacy of PROTACs independent of the therapeutic target. Thus, MTOR inhibitors can be combined with any and all PROTACs, not just BET protein or CDK9 PROTACs.

In some embodiments, the kinase inhibitor is a CDK7 inhibitor. CDK7 inhibitors include, without limitation, THZ1, AT7519, LDC4297, LY2857785, BS-181 HC, SNS-032, R547, Flavopiridol, AT7519, PHA-793887, and Flavopiridol-HCl.

In some embodiments, the kinase inhibitor is a Casein kinase 2 (CK2) inhibitor. CK2 inhibitors include, without limitation, Silmitasertib, GSK269962, and TBB.

In some embodiments, the kinase inhibitor is a RAC-alpha serine/threonine-protein kinase 1 and 2 (AKT1/2) inhibitor. AKT1/2 inhibitors include, without limitation, Akti-1/2, MK-2206, Perifosine, GSK690693, Ipatasertib, AZD5363, AT7867, Triciribine, CCT128930, A- 674563, Miltefosine, TIC10, SC66, Afuresertib, AT13148, Uprosertib, , SC79, and ARQ 092.

In some embodiments, the kinase inhibitor is an Adaptor-associated kinase 1 (AAK1) inhibitor. AAK1 inhibitors include, without limitation, LP-935509, LP-922761, BMT-090605, BMT-124110, LP-927443, and BMS-901715 (see, Kostich et al., J. Pharmacol. Exp. Ther., 2016, 358, 371-386).

In some embodiments, the kinase inhibitor is a focal adhesion kinase (FAK1) inhibitor. FAK1 inhibitors include, without limitation, PF-00562271, PF-573228, TAE226, PF-03814735, Defactinib, GSK2256098, PF-431396, Y15, and PND-1186. In some embodiments, the kinase inhibitor is a mitogen-activated protein kinase kinase (MEK) inhibitor. MEK inhibitors include, without limitation, Binimetinib, Selumetinib, PD0325901, Trametinib, U0126-EtOH, PD184352, PD98059, Pimasertib, TAK-733, AZD8330, PD318088, SL327, Refametinib, GDC-0623, Cobimetinib, and BI-847325.

In some embodiments, the kinase inhibitor is a Rapidly Accelerated Fibrosarcoma (RAF) kinase inhibitor. RAF kinase inhibitors include, without limitation, Sorafenib, Vemurafenib, Regorafenib, Sorafenib Tosylate, Dabrafenib, AZ304, Belvarafenib, ERK-IN-1, PLX8394, Doramapimod, PLX-4720, LY3009120, RAF265, GW 5074, Dabrafenib Mesylate, LXH254, Agerafenib, GDC-0879, AZ 628, Ro 5126766, TAK-632, Regorafenib Hydrochloride, PLX7904, CCT196969, HG6-64-1, TAK-580, ZM 336372, AD80, SB-590885, Regorafenib monohydrate, Lifirafenib, L-779450, P-0850, B-Raf inhibitor 1, Belvarafenib, B-RafIN 1, LUT014, BI-882370, Agerafenib hydrochloride, and B-Raf inhibitor 1 dihydrochloride.

In some embodiments, the compositions further comprise a KRAS inhibitor. KRAS inhibitors include, without limitation, K-Ras(G12C) inhibitors 1-12, Olaparib, AMG-510, Deltarasin, 6H05, ARS-1620, KRpep-2d, ARS-853, Lonafamib, MRTX-1257, PHT-7.3, ARS- 1323 and MRTX849.

In some embodiments, the compositions further comprise an autophagy inhibitor. Autophagy inhibitors include, without limitation, Nimodipine, Lucanthone, Liensinine, Autophinib, DC661, EAD1, Spautin-1, ROC-325, PIK-III, PHY34, MHY1485, Hydroxychloroquine Sulfate, CA-5f, Bafilomycin Al, Daurisoline, 3BDO, SAR405,

Elaiophylin, Autogramin-2, Lys05, DC661, IITZ-01, SBI-0206965, AS1842856, Chloroquine, 3- Methyladenine, ULK-101, J22352, LYN-1604, MRT68921 HC1, and hydroxychloroquine.

In some embodiments, the compositions comprise one or more BET-PROTAC therapeutic agents and one or more MTOR signaling pathway inhibitors. In some embodiments, the compositions comprise MZ1 and one or more MTOR signaling pathway inhibitors. In some embodiments, the compositions comprise MZ1 and one or more of RAD001, Torin-1, GDC- 0941, LY2584702, PI-273, orNVP-2. In some embodiments, the compositions comprise MZ1 and GDC-0941. In some embodiments, the compositions comprise MZ1 and lapatinib. In some embodiments, the compositions comprise MZ1 and RAD001. In some embodiments, the compositions comprise MZ1 and LY2584702. In some embodiments, the compositions comprise MZ1 and PI-273. In some embodiments, the compositions comprise MZ1 and NVP-2. In some embodiments, the compositions comprise ARV825 and one or more MTOR signaling pathway inhibitors. In some embodiments, the compositions comprise ARV825 and one or more of RAD001, Torin-1, GDC-0941, LY2584702, PI-273, orNVP-2. In some embodiments, the compositions comprise ARV825 and GDC-0941. In some embodiments, the compositions comprise ARV825 and lapatinib. In some embodiments, the compositions comprise ARV825 and RAD001. In some embodiments, the compositions comprise ARV825 and LY2584702. In some embodiments, the compositions comprise ARV825 and PI-273. In some embodiments, the compositions comprise ARV825 and NVP-2. In some embodiments, the compositions comprise dBETl and one or more MTOR signaling pathway inhibitors. In some embodiments, the compositions comprise dBETl and one or more of RAD001, Torin-1, GDC-0941, LY2584702, PI-273, or NVP-2. In some embodiments, the compositions comprise dBETl and GDC-0941.

In some embodiments, the compositions comprise dBETl and lapatinib. In some embodiments, the compositions comprise dBETl and RADOOl. In some embodiments, the compositions comprise dBETl and LY2584702. In some embodiments, the compositions comprise dBETl and PI-273. In some embodiments, the compositions comprise dBETl and NVP-2. In some embodiments, the compositions comprise A1874 and one or more MTOR signaling pathway inhibitors. In some embodiments, the compositions comprise A1874 and one or more of RADOOl, Torin-1, GDC-0941, LY2584702, PI-273, or NVP-2. In some embodiments, the compositions comprise A1874 and GDC-0941. In some embodiments, the compositions comprise A1874 and lapatinib. In some embodiments, the compositions comprise A1874 and RADOOl. In some embodiments, the compositions comprise A1874 and LY2584702. In some embodiments, the compositions comprise A1874 and PI-273. In some embodiments, the compositions comprise A1874 and NVP-2. In some embodiments, the compositions comprise CFT-2718 and one or more MTOR signaling pathway inhibitors. In some embodiments, the compositions comprise CFT-2718 and one or more of RADOOl, Torin-1, GDC-0941, LY2584702, PI-273, or NVP-2. In some embodiments, the compositions comprise CFT-2718 and GDC-0941. In some embodiments, the compositions comprise CFT-2718 and lapatinib. In some embodiments, the compositions comprise CFT-2718 and RADOOl. In some embodiments, the compositions comprise CFT-2718 and LY2584702. In some embodiments, the compositions comprise CFT-2718 and PI-273. In some embodiments, the compositions comprise CFT-2718 and NVP-2.

In some embodiments, the compositions comprise one or more BET-PROTAC therapeutic agents and one or more RTK signaling pathway inhibitors. In some embodiments, the compositions comprise MZ1 and one or more RTK signaling pathway inhibitors. In some embodiments, the compositions comprise MZ1 and one or more of GSK1904529A, lapatinib, imatinib, MRX-2843, LY2801653 dihydrochloride and PD173074. In some embodiments, the compositions comprise MZ1 and GSK1904529A. In some embodiments, the compositions comprise MZ1 and lapatinib. In some embodiments, the compositions comprise MZ1 and imatinib. In some embodiments, the compositions comprise MZ1 and MRX-2843. In some embodiments, the compositions comprise MZ1 and LY2801653 dihydrochloride. In some embodiments, the compositions comprise MZ1 and PD173074. In some embodiments, the compositions comprise ARV825 and one or more RTK signaling pathway inhibitors. In some embodiments, the compositions comprise ARV825 and one or more of GSK1904529A, lapatinib, imatinib, MRX-2843, LY2801653 dihydrochloride and PD173074. In some embodiments, the compositions comprise ARV825 and GSK1904529A. In some embodiments, the compositions comprise ARV825 and lapatinib. In some embodiments, the compositions comprise ARV825 and imatinib. In some embodiments, the compositions comprise ARV825 and MRX-2843. In some embodiments, the compositions comprise ARV825 and LY2801653 dihydrochloride. In some embodiments, the compositions comprise ARV825 and PD173074. In some embodiments, the compositions comprise dBETl and one or more RTK signaling pathway inhibitors. In some embodiments, the compositions comprise dBETl and one or more of GSK1904529A, lapatinib, imatinib, MRX-2843, LY2801653 dihydrochloride and PD173074. In some embodiments, the compositions comprise dBETl and GSK1904529A. In some embodiments, the compositions comprise dBETl and lapatinib. In some embodiments, the compositions comprise dBETl and imatinib. In some embodiments, the compositions comprise dBETl and MRX-2843. In some embodiments, the compositions comprise dBETl and LY2801653 dihydrochloride. In some embodiments, the compositions comprise dBETl and PD173074. In some embodiments, the compositions comprise A1874 and one or more RTK signaling pathway inhibitors. In some embodiments, the compositions comprise A1874 and one or more of GSK1904529A, lapatinib, imatinib, MRX-2843, LY2801653 dihydrochloride and PD173074. In some embodiments, the compositions comprise A1874 and GSK1904529A. In some embodiments, the compositions comprise A1874 and lapatinib. In some embodiments, the compositions comprise A1874 and imatinib. In some embodiments, the compositions comprise A1874 and MRX-2843. In some embodiments, the compositions comprise A1874 and LY2801653 dihydrochloride. In some embodiments, the compositions comprise A1874 and PD173074. In some embodiments, the compositions comprise CFT-2718 and one or more RTK signaling pathway inhibitors. In some embodiments, the compositions comprise CFT-2718 and one or more of GSK1904529A, lapatinib, imatinib, MRX-2843, LY2801653 dihydrochloride and PD173074. In some embodiments, the compositions comprise CFT-2718 and GSK1904529A. In some embodiments, the compositions comprise CFT-2718 and lapatinib. In some embodiments, the compositions comprise CFT-2718 and imatinib. In some embodiments, the compositions comprise CFT-2718 and MRX-2843. In some embodiments, the compositions comprise CFT-2718 and LY2801653 dihydrochloride. In some embodiments, the compositions comprise CFT-2718 and PD 173074.

In some embodiments, the compositions comprise one or more BET-PROTAC therapeutic agents and one or more KRAS inhibitors. In some embodiments, the compositions comprise MZ1 and one or more KRAS inhibitors. In some embodiments, the compositions comprise MZ1 and one or more of ARS-1620 and MRTX849. In some embodiments, the compositions comprise MZ1 and ARS-1620. In some embodiments, the compositions comprise MZ1 and MRTX849. In some embodiments, the compositions comprise ARV825 and one or more of ARS-1620 and MRTX849. In some embodiments, the compositions comprise ARV825 and ARS-1620. In some embodiments, the compositions comprise ARV825 and MRTX849. In some embodiments, the compositions comprise dBETl and one or more KRAS inhibitors. In some embodiments, the compositions comprise dBETl and one or more of ARS-1620 and MRTX849. In some embodiments, the compositions comprise dBETl and ARS-1620. In some embodiments, the compositions comprise dBETl and MRTX849. In some embodiments, the compositions comprise A1874 and one or more KRAS inhibitors. In some embodiments, the compositions comprise A1874 and one or more of ARS-1620 and MRTX849. In some embodiments, the compositions comprise A1874 and ARS-1620. In some embodiments, the compositions comprise A1874 and MRTX849. In some embodiments, the compositions comprise CFT-2718 and one or more KRAS inhibitors. In some embodiments, the compositions comprise CFT-2718 and one or more of ARS-1620 and MRTX849. In some embodiments, the compositions comprise CFT-2718 and ARS-1620. In some embodiments, the compositions comprise CFT-2718 and MRTX849.

In some embodiments, the compositions comprise one or more BET-PROTAC therapeutic agents and one or more autophagy inhibitors. In some embodiments, the compositions comprise MZ1 and one or more autophagy inhibitors. In some embodiments, the compositions comprise MZ1 and one or more of SAR405, Autophinib, PIK-III, LYN-1604, SBI- 0206965, MRT68921 HC1, ULK-101, and hydroxychloroquine. In some embodiments, the compositions comprise MZ1 and SAR405. In some embodiments, the compositions comprise MZ1 and Autophinib. In some embodiments, the compositions comprise MZ1 and PIK-III. In some embodiments, the compositions comprise MZ1 and LYN-1604. In some embodiments, the compositions comprise MZ1 and SBI-0206965. In some embodiments, the compositions comprise MZ1 and MRT68921 HC1. In some embodiments, the compositions comprise MZ1 and ULK-101. In some embodiments, the compositions comprise MZ1 and hydroxychloroquine. In some embodiments, the compositions comprise ARV825 and one or more autophagy inhibitors. In some embodiments, the compositions comprise ARV825 and one or more of SAR405, Autophinib, PIK-III, LYN-1604, SBI-0206965, MRT68921 HC1, ULK-101, and hydroxychloroquine. In some embodiments, the compositions comprise ARV825 and SAR405. In some embodiments, the compositions comprise ARV825 and Autophinib. In some embodiments, the compositions comprise ARV825 and PIK-III. In some embodiments, the compositions comprise ARV825 and LYN-1604. In some embodiments, the compositions comprise ARV825 and SBI-0206965. In some embodiments, the compositions comprise ARV825 and MRT68921 HC1. In some embodiments, the compositions comprise ARV825 and ULK-101. In some embodiments, the compositions comprise ARV825 and hydroxychloroquine. In some embodiments, the compositions comprise dBETl and one or more autophagy inhibitors. In some embodiments, the compositions comprise dBETl and one or more of SAR405, Autophinib, PIK-III, LYN-1604, SBI-0206965, MRT68921 HC1, ULK-101, and hydroxychloroquine. In some embodiments, the compositions comprise dBETl and SAR405.

In some embodiments, the compositions comprise dBETl and Autophinib. In some embodiments, the compositions comprise dBETl and PIK-III. In some embodiments, the compositions comprise dBETl and LYN-1604. In some embodiments, the compositions comprise dBETl and SBI-0206965. In some embodiments, the compositions comprise dBETl and MRT68921 HC1. In some embodiments, the compositions comprise dBET and ULK-101. In some embodiments, the compositions comprise dBET and hydroxychloroquine. In some embodiments, the compositions comprise A1874 and one or more autophagy inhibitors. In some embodiments, the compositions comprise A1874 and one or more of SAR405, Autophinib, PIK- III, LYN-1604, SBI-0206965, MRT68921 HC1, ULK-101, and hydroxychloroquine. In some embodiments, the compositions comprise A1874 and SAR405. In some embodiments, the compositions comprise A1874 and Autophinib. In some embodiments, the compositions comprise A1874 and PIK-III. In some embodiments, the compositions comprise A1874 and LYN-1604. In some embodiments, the compositions comprise A1874 and SBI-0206965. In some embodiments, the compositions comprise A1874 and MRT68921 HC1. In some embodiments, the compositions comprise A1874 and ULK-101. In some embodiments, the compositions comprise A1874 and hydroxychloroquine. In some embodiments, the compositions comprise CFT-2718 and one or more autophagy inhibitors. In some embodiments, the compositions comprise CFT-2718 and one or more of SAR405, Autophinib, PIK-III, LYN-1604, SBI- 0206965, MRT68921 HC1, ULK-101, and hydroxychloroquine. In some embodiments, the compositions comprise CFT-2718 and SAR405. In some embodiments, the compositions comprise CFT-2718 and Autophinib. In some embodiments, the compositions comprise CFT- 2718 and PIK-III. In some embodiments, the compositions comprise CFT-2718 and LYN-1604. In some embodiments, the compositions comprise CFT-2718 and SBI-0206965. In some embodiments, the compositions comprise CFT-2718 and MRT68921 HC1. In some embodiments, the compositions comprise CFT-2718 and ULK-101. In some embodiments, the compositions comprise CFT-2718 and hydroxychloroquine.

In some embodiments, the compositions comprise one or more CDK9-PROTAC therapeutic agents and one or more MTOR signaling pathway inhibitors. In some embodiments, the compositions comprise THAL SNS 032 and one or more MTOR signaling pathway inhibitors. In some embodiments, the compositions comprise THAL SNS 032 and one or more of RAD001, Torin-1, GDC-0941, LY2584702, PI-273, or NVP-2. In some embodiments, the compositions comprise THAL SNS 032 and GDC-0941. In some embodiments, the compositions comprise THAL SNS 032 and lapatinib. In some embodiments, the compositions comprise THAL SNS 032 and RAD001. In some embodiments, the compositions comprise THAL SNS 032 and LY2584702. In some embodiments, the compositions comprise THAL SNS 032 and PI-273. In some embodiments, the compositions comprise THAL SNS 032 and NVP-2. In some embodiments, the compositions comprise NVP-2 and one or more MTOR signaling pathway inhibitors. In some embodiments, the compositions comprise NVP-2 and one or more of RAD001, Torin-1, GDC-0941, LY2584702, or PI-273. In some embodiments, the compositions comprise NVP-2 and GDC-0941. In some embodiments, the compositions comprise NVP-2 and lapatinib. In some embodiments, the compositions comprise NVP-2 and RAD001. In some embodiments, the compositions comprise NVP-2 and LY2584702. In some embodiments, the compositions comprise NVP-2 and PI-273. In some embodiments, the compositions comprise CDK9 Degrader- 1 and one or more MTOR signaling pathway inhibitors. In some embodiments, the compositions comprise CDK9 Degrader- 1 and one or more of RAD001, Torin-1, GDC-0941, LY2584702, PI-273, or NVP-2. In some embodiments, the compositions comprise CDK9 Degrader- 1 and GDC-0941. In some embodiments, the compositions comprise CDK9 Degrader- 1 and lapatinib. In some embodiments, the compositions comprise CDK9 Degrader- 1 and RAD001. In some embodiments, the compositions comprise CDK9 Degrader-1 and LY2584702. In some embodiments, the compositions comprise CDK9 Degrader-1 and PI-273. In some embodiments, the compositions comprise CDK9 Degrader- 1 and NVP-2.

In some embodiments, the compositions comprise one or more CDK9-PROTAC therapeutic agents and one or more RTK signaling pathway inhibitors. In some embodiments, the compositions comprise THAL SNS 032 and one or more RTK signaling pathway inhibitors. In some embodiments, the compositions comprise THAL SNS 032 and one or more of GSK1904529A, lapatinib, imatinib, MRX-2843, LY2801653 dihydrochloride and PD173074. In some embodiments, the compositions comprise THAL SNS 032 and GSK1904529A. In some embodiments, the compositions comprise THAL SNS 032 and lapatinib. In some embodiments, the compositions comprise THAL SNS 032 and imatinib. In some embodiments, the compositions comprise THAL SNS 032 and MRX-2843. In some embodiments, the compositions comprise THAL SNS 032 and LY2801653 dihydrochloride. In some embodiments, the compositions comprise THAL SNS 032 and PD173074. In some embodiments, the compositions comprise NVP-2 and one or more RTK signaling pathway inhibitors. In some embodiments, the compositions comprise NVP-2 and one or more of GSK1904529A, lapatinib, imatinib, MRX-2843, LY2801653 dihydrochloride and PD173074. In some embodiments, the compositions comprise NVP-2 and GSK1904529 A. In some embodiments, the compositions comprise NVP-2 and lapatinib. In some embodiments, the compositions comprise NVP-2 and imatinib. In some embodiments, the compositions comprise NVP-2 and MRX-2843. In some embodiments, the compositions comprise NVP-2 and LY2801653 dihydrochloride. In some embodiments, the compositions comprise NVP-2 and PD173074. In some embodiments, the compositions comprise CDK9 Degrader-1 and one or more RTK signaling pathway inhibitors. In some embodiments, the compositions comprise CDK9 Degrader- 1 and one or more of GSK1904529 A, lapatinib, imatinib, MRX-2843, LY2801653 dihydrochloride and PD173074. In some embodiments, the compositions comprise CDK9 Degrader- 1 and GSK1904529A. In some embodiments, the compositions comprise CDK9 Degrader- 1 and lapatinib. In some embodiments, the compositions comprise CDK9 Degrader- 1 and imatinib. In some embodiments, the compositions comprise CDK9 Degrader- 1 and MRX-2843. In some embodiments, the compositions comprise CDK9 Degrader-1 and LY2801653 dihydrochloride. In some embodiments, the compositions comprise CDK9 Degrader-1 and PD173074.

In some embodiments, the compositions comprise one or more CDK9-PROTAC therapeutic agents and one or more KRAS inhibitors. In some embodiments, the compositions comprise THAL SNS 032 and one or more KRAS inhibitors. In some embodiments, the compositions comprise THAL SNS 032 and one or more of ARS-1620 and MRTX849. In some embodiments, the compositions comprise THAL SNS 032 and ARS-1620. In some embodiments, the compositions comprise THAL SNS 032 and MRTX849. In some embodiments, the compositions comprise NVP-2 and one or more of ARS-1620 and MRTX849. In some embodiments, the compositions comprise NVP-2 and ARS-1620. In some embodiments, the compositions comprise NVP-2 and MRTX849. In some embodiments, the compositions comprise CDK9 Degrader- 1 and one or more KRAS inhibitors. In some embodiments, the compositions comprise CDK9 Degrader- 1 and one or more of ARS-1620 and MRTX849. In some embodiments, the compositions comprise CDK9 Degrader-1 and ARS- 1620. In some embodiments, the compositions comprise CDK9 Degrader- 1 and MRTX849.

In some embodiments, the compositions comprise one or more CDK9-PROTAC therapeutic agents and one or more autophagy inhibitors. In some embodiments, the compositions comprise THAL SNS 032 and one or more autophagy inhibitors. In some embodiments, the compositions comprise THAL SNS 032 and one or more of SAR405, Autophinib, PIK-III, LYN-1604, SBI-0206965, MRT68921 HC1, ULK-101, and hydroxychloroquine. In some embodiments, the compositions comprise THAL SNS 032 and SAR405. In some embodiments, the compositions comprise THAL SNS 032 and Autophinib.

In some embodiments, the compositions comprise THAL SNS 032 and PIK-III. In some embodiments, the compositions comprise THAL SNS 032 and LYN-1604. In some embodiments, the compositions comprise THAL SNS 032 and SBI-0206965. In some embodiments, the compositions comprise THAL SNS 032 and MRT68921 HC1. In some embodiments, the compositions comprise THAL SNS 032 and ULK-101. In some embodiments, the compositions comprise THAL SNS 032 and hydroxychloroquine. In some embodiments, the compositions comprise NVP-2 and one or more autophagy inhibitors. In some embodiments, the compositions comprise NVP-2 and one or more of SAR405, Autophinib, PIK-III, LYN-1604, SBI-0206965, MRT68921 HC1, ULK-101, and hydroxychloroquine. In some embodiments, the compositions comprise NVP-2 and SAR405. In some embodiments, the compositions comprise NVP-2 and Autophinib. In some embodiments, the compositions comprise NVP-2 and PIK-III. In some embodiments, the compositions comprise NVP-2 and LYN-1604. In some embodiments, the compositions comprise NVP-2 and SBI-0206965. In some embodiments, the compositions comprise NVP-2 and MRT68921 HC1. In some embodiments, the compositions comprise NVP-2 and ULK-101. In some embodiments, the compositions comprise NVP-2 and hydroxychloroquine. In some embodiments, the compositions comprise CDK9 Degrader- 1 and one or more autophagy inhibitors. In some embodiments, the compositions comprise CDK9 Degrader-1 and one or more of SAR405, Autophinib, PIK-III, LYN-1604, SBI-0206965, MRT68921 HC1, ULK-101, and hydroxychloroquine. In some embodiments, the compositions comprise CDK9 Degrader-1 and SAR405. In some embodiments, the compositions comprise CDK9 Degrader- 1 and Autophinib. In some embodiments, the compositions comprise CDK9 Degrader-1 and PIK-III. In some embodiments, the compositions comprise CDK9 Degrader- 1 and LYN-1604. In some embodiments, the compositions comprise CDK9 Degrader-1 and SBI- 0206965. In some embodiments, the compositions comprise CDK9 Degrader-1 and MRT68921 HC1. In some embodiments, the compositions comprise CDK9 Degrader-1 and ULK-101. In some embodiments, the compositions comprise CDK9 Degrader-1 and hydroxychloroquine.

The present disclosure further provides methods for augmenting the therapeutic effect in a human cancer patient undergoing treatment with a BET-PROTAC therapeutic agent or CDK9- PROTAC therapeutic agent comprising administering one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof, to the patient.

As used herein, the term “augment” or “augmenting” as used herein, for therapeutic purposes, generally refers to an improvement in the pharmacodynamic effect (referred to as the efficacy) of a therapeutic agent. Thus, the term “augment” refers to the ability of the kinase inhibitors, KRAS inhibitors, or autophagy inhibitors to raise the efficacy of a PROTACs leading to the killing of a greater number of cancer cells over the same unit of time (e.g., 24, 48, or 72 hour period) when the kinase inhibitors, KRAS inhibitors, or autophagy inhibitors are administered prior to, along with, or after the PROTACs as compared to the PROTACs alone.

The present disclosure also provides methods of treating cancer in a human patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a pharmaceutical composition comprising one or more BET-PROTAC therapeutic agents or one or more CDK9-PROTAC therapeutic agents, or a combination thereof and one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof.

The present disclosure also provides methods of overcoming resistance to BET- PROTAC therapeutic agents or CDK9-PROTAC therapeutic in a human patient, the method comprising administering to the patient a therapeutically effective amount of a pharmaceutical composition comprising one or more BET-PROTAC therapeutic agents or one or more CDK9- PROTAC therapeutic agents, or a combination thereof and one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof.

In some embodiments, the BET-PROTAC or CDK9-PROTAC is administered prior to the administration of the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors or after administration of the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors. In some embodiments, the BET- PROTAC or CDK9-PROTAC is administered prior to the administration of the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors. In some embodiments, the BET-PROTAC or CDK9-PROTAC is administered after administration of the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors. In some embodiments, the BET-PROTAC or CDK9-PROTAC is administered concurrently with administration of the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof

As used herein, “treatment” (and grammatical variations thereof such as “treat” or “treating”) refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. In some embodiments, compositions described herein are used to delay development of a disease or to slow the progression of a disease. In some embodiments, compositions described herein are used to increase survival of a patient having a disease. In some embodiments, compositions described herein are used to increase or extend survival of a patient having a disease.

As used herein, the term “survival” refers to the patient remaining alive, and includes disease free survival (DFS), progression free survival (PFS) and overall survival (OS). Survival can be estimated by the Kaplan-Meier method, and any differences in survival are computed using the stratified log-rank test.

As used herein, the term “progression-Free Survival” (PFS) is the time from the first day of treatment to documented disease progression (including isolated CNS progression) or death from any cause on study, whichever occurs first.

As used herein, the term “disease free survival (DFS)” refers to the patient remaining alive, without return of the cancer, for a defined period of time such as about 1 year, about 2 years, about 3 years, about 4 years, about 5 years, about 10 years, etc., from initiation of treatment or from initial diagnosis. In one aspect of the subject matter described herein, DFS is analyzed according to the intent-to-treat principle, i.e., patients are evaluated on the basis of their assigned therapy. The events used in the analysis of DFS can include local, regional and distant recurrence of cancer, occurrence of secondary cancer, and death from any cause in patients without a prior event (e.g, breast cancer recurrence or second primary cancer). As used herein, the term “overall survival” refers to the patient remaining alive for a defined period of time, such as about 1 year, about 2 years, about 3 years, about 4 years, about 5 years, about 10 years, etc., from initiation of treatment or from initial diagnosis.

By “extending survival” is meant increasing DFS and/or OS in a treated patient relative to an untreated patient, or relative to a control treatment protocol. Survival is monitored for at least about six months, or at least about 1 year, or at least about 2 years, or at least about 3 years, or at least about 4 years, or at least about 5 years, or at least about 10 years, etc., following the initiation of treatment or following the initial diagnosis.

A drug that is administered “concurrently” with one or more other drugs is administered during the same treatment cycle, on the same day of treatment as the one or more other drugs, and, optionally, at the same time as the one or more other drugs. For instance, for cancer therapies given every 3 weeks, the concurrently administered drugs are each administered on day-1 of a 3-week cycle.

The term “effective” is used to describe an amount of a compound, composition or component which, when used within the context of its intended use, effects an intended result. The term effective subsumes all other effective amount or effective concentration terms, which are otherwise described or used in the present application.

An “effective amount” of an agent, e.g., a pharmaceutical formulation, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result. For example, an effective amount of the drug for treating cancer may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer. To the extent the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic. The effective amount may extend progression free survival (e.g. as measured by Response Evaluation Criteria for Solid Tumors, RECIST, or CA-125 changes), result in an objective response (including a partial response, PR, or complete response, CR), increase overall survival time, and/or improve one or more symptoms of cancer (e.g. as assessed by FOSI).

As used herein, the term “therapeutically effective amount” means any amount which, as compared to a corresponding subject who has not received such amount, results in treatment of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder. The term also includes within its scope amounts effective to enhance normal physiological function. For use in therapy, therapeutically effective amounts of a PAC, as well as salts thereof, may be administered as the raw chemical. Additionally, the active ingredient may be presented as a pharmaceutical composition.

The present disclosure also provides methods for killing or inhibiting growth of a cancer cell comprising comprising contacting the cancer cell with any of the combinations of compounds, or compositions comprising the same as described herein. In some embodiments, one or more compounds may be combined in the same composition for any of the methods disclosed herein.

Thus, the compounds and compositions can be used as anti-cancer and anti-tumor agents, e.g., the compounds can kill or inhibit the growth of cancer cells. The compounds and compositions can also be used in methods of reducing cancer in an animal, or in methods of treating or preventing the spread or metastasis of cancer in an animal, or in methods of treating an animal afflicted with cancer. The compounds and compositions can also be used in methods of killing or inhibiting the growth of a cancer cell, or in methods of inhibiting tumor growth.

Cancers that are treatable are broadly divided into the categories of carcinoma, lymphoma and sarcoma. Examples of carcinomas include, but are not limited to: adenocarcinoma, acinic cell adenocarcinoma, adrenal cortical carcinomas, alveoli cell carcinoma, anaplastic carcinoma, basaloid carcinoma, basal cell carcinoma, bronchiolar carcinoma, bronchogenic carcinoma, renaladinol carcinoma, embryonal carcinoma, anometroid carcinoma, fibrolamolar liver cell carcinoma, follicular carcinomas, giant cell carcinomas, hepatocellular carcinoma, intraepidermal carcinoma, intraepithelial carcinoma, leptomanigio carcinoma, medullary carcinoma, melanotic carcinoma, menigual carcinoma, mesometonephric carcinoma, oat cell carcinoma, squamal cell carcinoma, sweat gland carcinoma, transitional cell carcinoma, and tubular cell carcinoma. Sarcomas include, but are not limited to: amelioblastic sarcoma, angiolithic sarcoma, botryoid sarcoma, endometrial stroma sarcoma, ewing sarcoma, fascicular sarcoma, giant cell sarcoma, granulositic sarcoma, immunoblastic sarcoma, juxaccordial osteogenic sarcoma, coppices sarcoma, leukocytic sarcoma (leukemia), lymphatic sarcoma (lympho sarcoma), medullary sarcoma, myeloid sarcoma (granulocitic sarcoma), austiogenci sarcoma, periosteal sarcoma, reticulum cell sarcoma (histiocytic lymphoma), round cell sarcoma, spindle cell sarcoma, synovial sarcoma, and telangiectatic audiogenic sarcoma. Lymphomas include, but are not limited to: Hodgkin’s disease and lymphocytic lymphomas, such as Burkitt’s lymphoma, NPDL, NML, NH and diffuse lymphomas.

Examples of cancers which may be treated by the compositions include, but are not limited to acute myeloid leukemia, acute monocytic leukemia, prostatic adenocarcinoma, ovarian carcinoma, or epithelial ovarian cancer, such as High-Grade Serous Ovarian Carcinoma (HGSOC). In some embodiments, the cancers which may be treated by the compositions include KRAS cancers (including, but not limited to non-small cell lung cancer (NSCLC), colorectal cancer, and pancreatic cancer), PIK3CA cancers (including, but not limited to breast cancer, colon cancer, endometrial cancer, glioblastoma multiformes, epidermal nevi, seborrheic keratoses (SK), ovarian cancer, gastric cancer, squamous cell carcinoma, thyroid cancer, oral squamous cell carcinoma, nasopharyngeal carcinoma, cervical cancer, papillary mucinous carcinoma of the pancreas, squamous cell carcinoma of the esophagus, adenocarcinomas of the esophagus, gallbladder carcinoma, cholangiocarcinoma, invasive pituitary tumors, penile tumors, bladder cancer, and diffuse large B cell lymphomas) or PTEN cancers (including, but not limited to glioblastoma, endometrial cancer, breast cancer, Endometrial Cancer, and prostate cancer). Thus, cancer cells harboring K-Ras mutations or other mutations (such as Kras-amplification, PIK3CA-mutations, PTEN-loss, EGFR-mutants, HER2 overexpression, and AKT amplification) that strongly activate mTORCl signaling can promote intrinsic resistance to BBDs representing a patient population that may benefit from combined mTORCl and PROTAC -treatment.

The compounds and compositions can be used in methods of killing or inhibiting the growth of cancer cells, either in vivo or in vitro, or inhibiting the growth of a cancerous tumor.

In some embodiments, the compounds and compositions are used in conjunction with other therapies, such as standard immunotherapy, neoadjuvant therapy, radiotherapy, tumor surgery, and conventional chemotherapy directed against solid tumors and for the control of establishment of metastases. Additionally, the compounds and compositions can be administered after surgery where solid tumors have been removed as a prophylaxis against metastasis. Cytotoxic or chemotherapeutic agents include, but are ot limited to, aziridine thiotepa, alkyl sulfonate, nitrosoureas, platinum complexes, NO classic alkylators, folate analogs, purine analogs, adenosine analogs, pyrimidine analogs, substituted urea, antitumor antibiotics, microtubulle agents, and asprignase.

In some embodiments, the subject is also administered radiation therapy, immunotherapy, and/or neoadjuvant therapy. In some embodiments, the subject is also administered radiation therapy. In some embodiments, the subject is also administered immunotherapy. In some embodiments, the subject is also administered neoadjuvant therapy.

The present disclosure provides pharmaceutical formulations comprising one or more bromodomain and extra terminal domain (BET) proteolysis targeting chimera (PROTAC) (BET- PROTAC) therapeutic agents or one or more cyclin-dependent kinase 9 (CDK9) PROTAC (CDK9-PROTAC) therapeutic agents, or a combination thereof; one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof and a pharmaceutically acceptable carrier or excipient.

In some embodiments, the ratio of the BET-PROTAC or CDK9-PROTAC to the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is from about 0.01:1 to about 100:1 (w/w), from about 0.1:1 to about 10:1 (w/w), or from about 1:1 to about 5:1 (w/w). In some embodiments, the ratio of the BET-PROTAC or CDK9-PROTAC to the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is from about 0.01:1 to about 100:1 (w/w). In some embodiments, the ratio of the BET-PROTAC or CDK9-PROTAC to the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is from about 0.1:1 to about 10:1 (w/w). In some embodiments, the ratio of the BET-PROTAC or CDK9-PROTAC to the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is from about 1 : 1 to about 5:1 (w/w).

In some embodiments, the BET-PROTAC or CDK9-PROTAC is present in an amount from about 1 mg to about 100 mg, from about 5 mg to about 75 mg, from about 10 mg to about 60 mg, or from about 12.5 mg to about 50 mg, and the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is present in an amount from about 1 mg to about 500 mg, from about 50 mg to about 400 mg, from about 75 mg to about 300 mg, or from about 100 mg to about 200 mg. In some embodiments, the BET-PROTAC or CDK9- PROTAC is present in an amount from about 1 mg to about 100 mg. In some embodiments, the BET-PROTAC or CDK9-PROTAC is present in an amount from about 5 mg to about 75 mg. In some embodiments, the BET-PROTAC or CDK9-PROTAC is present in an amount from about 10 mg to about 60 mg. In some embodiments, the BET-PROTAC or CDK9-PROTAC is present in an amount from about 12.5 mg to about 50 mg. In some embodiments, the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is present in an amount from about 1 mg to about 500 mg. In some embodiments, the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is present in an amount from about 50 mg to about 400 mg. In some embodiments, the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is present in an amount from about 75 mg to about 300 mg. In some embodiments, the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is present in an amount from about 100 mg to about 200 mg.

In some embodiments, the amount of the BET-PROTAC therapeutic agent or CDK9- PROTAC therapeutic agent administered to the human patient receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is reduced compared to the amount of the BET-PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human patient in the absence of receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors. In some embodiments, the amount of the BET-PROTAC therapeutic agent or CDK9- PROTAC therapeutic agent administered to the human patient receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is reduced by 10%. In some embodiments, the amount of the BET-PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human patient receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is reduced by 15%. In some embodiments, the amount of the BET-PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human patient receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is reduced by 20%. In some embodiments, the amount of the BET-PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human patient receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is reduced by 25%. In some embodiments, the amount of the BET-PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human patient receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is reduced by 30%. In some embodiments, the amount of the BET-PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human patient receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is reduced by 35%. In some embodiments, the amount of the BET- PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human patient receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is reduced by 40%. In some embodiments, the amount of the BET-PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human patient receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is reduced by 45%. In some embodiments, the amount of the BET-PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human patient receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors is reduced by 50% compared to the amount of the BET-PROTAC therapeutic agent or CDK9-PROTAC therapeutic agent administered to the human patient in the absence of receiving administration of one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors.

The term “pharmaceutical formulation” refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.

A “pharmaceutically acceptable excipient” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject. A pharmaceutically acceptable excipient includes, but is not limited to, a buffer, carrier, stabilizer, or preservative.

The phrase “pharmaceutically acceptable salt,” as used herein, refers to pharmaceutically acceptable organic or inorganic salts of a molecule. Exemplary salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, and pamoate (i.e., l,l'-methylene-bis-(2-hydroxy-3- naphthoate)) salts. A pharmaceutically acceptable salt may involve the inclusion of another molecule such as an acetate ion, a succinate ion or other counterion. The counterion may be any organic or inorganic moiety that stabilizes the charge on the parent compound. Furthermore, a pharmaceutically acceptable salt may have more than one charged atom in its structure. Instances where multiple charged atoms are part of the pharmaceutically acceptable salt can have multiple counter ions. Hence, a pharmaceutically acceptable salt can have one or more charged atoms and/or one or more counterion.

Other salts, which are not pharmaceutically acceptable, may be useful in the preparation of compounds of described herein and these should be considered to form a further aspect of the subject matter. These salts, such as oxalic or trifluoroacetate, while not in themselves pharmaceutically acceptable, may be useful in the preparation of salts useful as intermediates in obtaining the compounds described herein and their pharmaceutically acceptable salts.

In some embodiments, the BET-PROTAC or CDK9-PROTAC and the one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors are co administered to the subject together in a single pharmaceutical composition. In some embodiments, the single pharmaceutical composition is an oral dosage form, an intravenous dosage form, a topical dosage form, an intraperitoneal dosage form, or an intrathecal dosage form. In some embodiments, the single pharmaceutical composition is an oral dosage form or an intravenous dosage form. In some embodiments, the single pharmaceutical composition is an oral dosage form. In some embodiments, the single pharmaceutical composition is an intravenous dosage form. In some embodiments, the oral dosage form is a pill, tablet, capsule, gel-cap, or liquid. In some embodiments, the oral dosage form is a pill. In some embodiments, the oral dosage form is a tablet. In some embodiments, the oral dosage form is a capsule. In some embodiments, the oral dosage form is a gel-cap. In some embodiments, the oral dosage form is a liquid.

The terms “co-administration” and “co-administering” or “combination therapy” refer to both concurrent administration (administration of two or more therapeutic agents at the same time) and time varied administration (administration of one or more therapeutic agents at a time different from that of the administration of an additional therapeutic agent or agents), as long as the therapeutic agents are present in the patient to some extent, preferably at effective amounts, at the same time. In certain preferred aspects, one or more of the present compounds described herein, are coadministered in combination with at least one additional bioactive agent, especially including an anticancer agent. In particularly preferred aspects, the co-administration of compounds results in synergistic activity and/or therapy, including anticancer activity.

In some embodiments, the pharmaceutical composition is an oral dosage form, an intravenous dosage form, a topical dosage form, an intraperitoneal dosage form, or an intrathecal dosage form. In some embodiments, the pharmaceutical composition is an oral dosage form or an intravenous dosage form. In some embodiments, the pharmaceutical composition is an oral dosage form.

In some embodiments, the oral dosage form is a pill, tablet, capsule, cachet, gel-cap, pellet, powder, granule, or liquid. In some embodiments, the oral dosage form is a pill, tablet, capsule, gel-cap, or liquid. In some embodiments, the oral dosage form is a pill. In some embodiments, the oral dosage form is a tablet. In some embodiments, the oral dosage form is a capsule. In some embodiments, the oral dosage form is a gel-cap. In some embodiments, the oral dosage form is a liquid.

In some embodiments, the oral dosage form is protected from light and present within a blister pack, bottle, or intravenous bag. In some embodiments, the oral dosage form is present within a blister pack, bottle, or intravenous bag. In some embodiments, the oral dosage form is present within a blister pack. In some embodiments, the oral dosage form is present within a bottle. In some embodiments, the oral dosage form is present within an intravenous bag.

The compounds and compositions described herein can be administered by any route of administration including, but not limited to, oral, intravenous, topical, intraperitoneal, and intrathecal. In some embodiments, the administration is oral, intravenous, intraperitoneal, or intrathecal. In some embodiments, the administration is oral, intravenous, or intraperitoneal. In some embodiments, the administration is oral or intravenous. In some embodiments, the administration is oral or topical. In some embodiments, the administration is oral or intraperitoneal. In some embodiments, the administration is oral or intrathecal. The route of administration can depend on the particular disease, disorder, or condition being treated and can be selected or adjusted by the clinician according to methods known to the clinician to obtain desired clinical responses. Methods for administration are known in the art and one skilled in the art can refer to various pharmacologic references for guidance (see, for example, Modem Pharmaceutics, Banker & Rhodes, Marcel Dekker, Inc. (1979); and Goodman & Gilman’s The Pharmaceutical Basis of Therapeutics, 6th Edition, MacMillan Publishing Co., New York (1980)).

In some embodiments, it may be desirable to administer one or more compounds, or a pharmaceutically acceptable salt thereof, or compisition(s) comprising the same to a particular area in need of treatment. This may be achieved, for example, by local infusion (for example, during surgery), topical application (for example, with a wound dressing after surgery), or by injection (for example, by depot injection). Formulations for injection can be presented in unit dosage form, such as in ampoules or in multi-dose containers, with an added preservative.

The compounds and compositions described herein can be formulated for parenteral administration by injection, such as by bolus injection or continuous infusion. The compounds and compositions can be administered by continuous infusion subcutaneously over a period of about 15 minutes to about 24 hours. The compositions can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents. In some embodiments, the injectable is in the form of short-acting, depot, or implant and pellet forms injected subcutaneously or intramuscularly. In some embodiments, the parenteral dosage form is the form of a solution, suspension, emulsion, or dry powder.

For oral administration, the compounds and compositions described herein can be formulated by combining the compounds with pharmaceutically acceptable carriers. Such carriers enable the compounds to be formulated as tablets, pills, dragees, capsules, emulsions, liquids, gels, syrups, caches, pellets, powders, granules, slurries, lozenges, aqueous or oily suspensions, and the like, for oral ingestion by a subject to be treated. Pharmaceutical preparations for oral use can be obtained by, for example, adding a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include, but are not limited to, fillers such as sugars, including, but not limited to, lactose, sucrose, mannitol, and sorbitol; cellulose preparations including, but not limited to, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and polyvinylpyrrolidone (PVP). If desired, disintegrating agents can be added, including, but not limited to, the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.

Orally administered compounds and compositions can contain one or more optional agents, for example, sweetening agents such as fructose, aspartame or saccharin; flavoring agents such as peppermint, oil of wintergreen, or cherry; coloring agents; and preserving agents, to provide a pharmaceutically palatable preparation. Moreover, when in tablet or pill form, the compositions may be coated to delay disintegration and absorption in the gastrointestinal tract thereby providing a sustained action over an extended period of time. Selectively permeable membranes surrounding an osmotically active driving compound are also suitable for orally administered compounds. Oral compositions can include standard vehicles such as, for example, mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Such vehicles are suitably of pharmaceutical grade.

Dragee cores can be provided with suitable coatings. For this purpose, concentrated sugar solutions can be used, which can optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments can be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.

Pharmaceutical preparations which can be used orally include, but are not limited to, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers can be added.

In transdermal administration, the compounds and compositions can be applied to a plaster, or can be applied by transdermal, therapeutic systems that are consequently supplied to the organism. In some embodiments, the compounds and compositions are present in creams, solutions, powders, fluid emulsions, fluid suspensions, semi-solids, ointments, pastes, gels, jellies, and foams, or in patches containing any of the same.

The compounds and compositions described herein can also be formulated as a depot preparation. Such long acting formulations can be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Depot injections can be administered at about 1 to about 6 months or longer intervals. Thus, for example, the compounds and compositions 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.

In some embodiments, the compounds and compositions can be delivered in a controlled release system. In some embodiments, a pump may be used (see Langer, supra;

Sefton, CRC Crit. Ref. Biomed. Eng., 1987, 14, 201; Buchwald et al, Surgery, 1980, 88, 507 Saudek et al, N. Engl. J. Med., 1989, 321, 574). In some embodiments, polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974); Controlled Drug Bioavailabibty, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger et al, J. Macromol. Sci. Rev. Macromol. Chern, 1983, 23, 61; see, also Levy et al, Science, 1985, 228, 190; During et al, Ann. Neurol., 1989, 25, 351; Howard et al., J. Neurosurg., 1989, 71, 105). In some embodiments, a controlled-release system can be placed in proximity of the target of the compounds described herein, such as the liver, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)). Other controlled-release systems discussed in the review by Langer, Science, 1990, 249, 1527-1533) may be used.

The compounds and compositions described herein can be contained in formulations with pharmaceutically acceptable diluents, fillers, disintegrants, binders, lubricants, surfactants, hydrophobic vehicles, water soluble vehicles, emulsifiers, buffers, humectants, moisturizers, solubilizers, preservatives and the like. The pharmaceutical compositions can also comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include, but are not limited to, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols. In some embodiments, the compounds described herein can be used with agents including, but not limited to, topical analgesics (e.g., lidocaine), barrier devices (e.g., GelClair), or rinses (e.g., Caphosol). Pharmaceutical carriers can be liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil, and the like. The pharmaceutical carriers can also be saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea, and the like. In addition, auxiliary, stabilizing, thickening, lubricating and coloring agents can be used.

In some embodiments, the compounds and compositions described herein can be delivered in a vesicle, in particular a liposome (see, Langer, Science, 1990, 249, 1527-1533; Treat et al, in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.).

The compositions described herein can be administered either alone (as a single composition comprising the compounds described herein) or in combination (concurrently or serially) with other pharmaceutical agents. For example, the compounds and compositions can be administered in combination with anti-cancer or anti-neoplastic agents (for example, methotrexate, taxol, mercaptopurine, thioguanine, hydroxyurea, cytarabine, cyclophosphamide, ifosfamide, nitrosoureas, cisplatin, carboplatin, mitomycin, dacarbazine, procarbizine, etoposides, campathecins, bleomycin, doxorubicin, idarubicin, daunorubicin, dactinomycin, plicamycin, mitoxantrone, asparaginase, vinblastine, vincristine, vinorelbine, paclitaxel, and docetaxel) or therapies (for example, surgery or radiotherapy).

The amount of any particular compound to be administered may be that amount which is therapeutically effective. The dosage to be administered may depend on the characteristics of the subject being treated, e.g., the particular animal treated, age, weight, health, types of concurrent treatment, if any, and frequency of treatments, and on the nature and extent of the disease, condition, or disorder, and can be easily determined by one skilled in the art (e.g., by the clinician). The selection of the specific dose regimen can be selected or adjusted or titrated by the clinician according to methods known to the clinician to obtain the desired clinical response. In addition, in vitro or in vivo assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the compositions may also depend on the route of administration, and should be decided according to the judgment of the practitioner and each patient’s circumstances.

Suitable compositions include, but are not limited to, oral non-absorbed compositions. Suitable compositions also include, but are not limited to saline, water, cyclodextrin solutions, and buffered solutions of pH 3-9.

The compounds and compositions described herein can be formulated with numerous excipients including, but not limited to, purified water, propylene glycol, PEG 400, glycerin, DMA, ethanol, benzyl alcohol, citric acid/sodium citrate (pH3), citric acid/sodium citrate (pH5), tris(hydroxymethyl)amino methane HC1 (pH7.0), 0.9% saline, and 1.2% saline, and any combination thereof. In some embodiments, excipient is chosen from propylene glycol, purified water, and glycerin.

In some embodiments, the excipient is a multi-component system chosen from 20% w/v propylene glycol in saline, 30% w/v propylene glycol in saline, 40% w/v propylene glycol in saline, 50% w/v propylene glycol in saline, 15% w/v propylene glycol in purified water, 30% w/v propylene glycol in purified water, 50% w/v propylene glycol in purified water, 30% w/v propylene glycol and 5 w/v ethanol in purified water, 15% w/v glycerin in purified water, 30% w/v glycerin in purified water, 50% w/v glycerin in purified water, 20% w/v Kleptose in purified water, 40% w/v Kleptose in purified water, and 25% w/v Captisol in purified water. In some embodiments, the excipient is chosen from 50% w/v propylene glycol in purified water, 15% w/v glycerin in purified water, 20% w/v Kleptose in purified water, 40% w/v Kleptose in purified water, and 25% w/v Captisol in purified water. In some embodiments, the excipient is chosen from 20% w/v Kleptose in purified water, 20% w/v propylene glycol in purified water, and 15% w/v glycerin in purified water.

In some embodiments, the compounds and compositions described herein can be lyophilized to a solid and reconstituted with, for example, water prior to use.

When administered to a human, the compounds and compositions can be sterile. Water is a suitable carrier when the compound and composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical carriers also include excipients such as starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The present compositions, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.

The compositions described herein can take the form of a solution, suspension, emulsion, tablet, pill, pellet, capsule, capsule containing a liquid, powder, sustained-release formulation, aerosol, spray, or any other form suitable for use. Examples of suitable pharmaceutical carriers are described in Remington’s Pharmaceutical Sciences, A.R. Gennaro (Editor) Mack Publishing Co.

In some embodiments, the compounds and compositions are formulated in accordance with routine procedures as pharmaceutical compositions adapted for administration to humans. Typically, compounds are solutions in sterile isotonic aqueous buffer. Where necessary, the compositions can also include a solubilizing agent. Compositions for intravenous administration may optionally include a local anesthetic such as lidocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the compound or composition is to be administered by infusion, it can be dispensed, for example, with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the compound or composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.

The pharmaceutical compositions can be in unit dosage form. In such form, the composition can be divided into unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of the preparations, for example, packeted tablets, capsules, and powders in vials or ampules. The unit dosage form can also be a capsule, cachet, or tablet itself, or it can be the appropriate number of any of these packaged forms.

In some embodiments, a composition is in the form of a liquid wherein the active agents are present in solution, in suspension, as an emulsion, or as a solution/suspension. In some embodiments, the liquid composition is in the form of a gel. In other embodiments, the liquid composition is aqueous. In other embodiments, the composition is in the form of an ointment.

In some embodiments, the composition is an in situ gellable aqueous solution, suspension or solution/suspension, comprising about from 0.2% to about 3% or from about 0.5% to about 1% by weight of a gelling polysaccharide, chosen from gellan gum, alginate gum and chitosan, and about 1% to about 50% of a water-soluble film-forming polymer, preferably selected from alkylcelluloses (e.g., methylcellulose, ethylcellulose), hydroxyalkylcelluloses (e.g., hydroxy ethylcellulose, hydroxypropyl methylcellulose), hyaluronic acid and salts thereof, chondroitin sulfate and salts thereof, polymers of acrylamide, acrylic acid and poly cyanoacrylates, polymers of methyl methacrylate and

2-hydroxyethyl methacrylate, poly dextrose, cyclodextrins, polydextrin, maltodextrin, dextran, poly dextrose, gelatin, collagen, natural gums (e.g., xanthan, locust bean, acacia, tragacanth and carrageenan gums and agar), polygalacturonic acid derivatives (e.g., pectin), polyvinyl alcohol, polyvinylpyrrolidone and polyethylene glycol. The composition can optionally contain a gel- promoting counterion such as calcium in latent form, for example encapsulated in gelatin.

In some embodiments, the composition is an in situ gellable aqueous solution, suspension or solution/suspension comprising about 0.1% to about 5% of a carrageenan gum, e.g., a carrageenan gum having no more than 2 sulfate groups per repeating disaccharide unit, such as e.g., kappa-carrageenan, having 18-25% ester sulfate by weight, iota-carrageenan, having 25-34% ester sulfate by weight, and mixtures thereof.

Optionally one or more stabilizers can be included in the compositions to enhance chemical stability where required. Suitable stabilizers include, but are not limited to, chelating agents or complexing agents, such as, for example, the calcium complexing agent ethylene diamine tetraacetic acid (EDTA). For example, an appropriate amount of EDTA or a salt thereof, e.g., the disodium salt, can be included in the composition to complex excess calcium ions and prevent gel formation during storage. EDTA or a salt thereof can suitably be included in an amount of about 0.01% to about 0.5%. In those embodiments containing a preservative other than EDTA, the EDTA or a salt thereof, more particularly disodium EDTA, can be present in an amount of about 0.025% to about 0.1% by weight.

The present disclosure also provides combinations of a BET-PROTAC or a CDK9- PROTAC, or a pharmaceutically acceptable salt thereof, and one or more kinase inhibitors, one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof, or a pharmaceutically acceptable salt thereof, for use in the manufacture of a medicament for treating cancer. Any of the combinations described herein can be used in the manufacture of a medicament for treating any of the cancers described herein.

The present disclosure also provides uses of a pharmaceutical composition comprising a BET-PROTAC or a CDK9-PROTAC, or a pharmaceutically acceptable salt thereof, and one or more KRAS inhibitors, or one or more autophagy inhibitors, or any combination thereof, or a pharmaceutically acceptable salt thereof, for treating cancer. Any of the combinations described herein can be used for treating any of the cancers described herein.

In order that the subject matter disclosed herein may be more efficiently understood, examples are provided below. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting the claimed subject matter in any manner. Throughout these examples, molecular cloning reactions, and other standard recombinant DNA techniques, were carried out according to methods described in Maniatis et al, Molecular Cloning - A Laboratory Manual, 2nd ed., Cold Spring Harbor Press (1989), using commercially available reagents, except where otherwise noted. Examples

Example 1: BET Degrader Therapies Uniquely Block Translation Signaling in OC Cells Promoting Kinome Destabilization and Apoptosis

To functionally explore the distinct methods of targeting BET proteins in ovarian cancer, a detailed proteomics characterization of OC cells treated with BBDs (MZ1 or ARV825) or BBIs (JQ1, or OTX015) was carried out. The BBDs used in the study were, MZ1, which uses JQ1 as the bromodomain targeting molecule and recruits E3 ligases using a VHL-binding moiety, and ARV825 with OTX015 as the warhead and a Cereblon-binding moiety for E3 ligase recruitment (Figures 1A and 8A). The established epithelial ovarian cancer cell line OVCAR8 was selected for the studies as it has previously been shown to require BRD4 for tumor formation. Knockdown of BRD4 induced apoptosis in OVCAR8 cells confirming BRD4- dependency (Figure IB).

Dose escalation of JQ1 or MZ1 but not cis-MZl, a negative control unable to recruit VHL, in OVCAR8 cells inhibited cell viability similarly, with GI50 values 133 nM (JQ1) and 142 nM (MZ1) (Figure 1C). BBDs MZ1 or ARV825 degraded BET proteins in a concentration- dependent manner in OVCAR8 cells (Figures ID, IE and 8B). Treatment with BBIs or BBDs induced apoptosis in OVCAR8 cells, however, a more pronounced cleavage of PARP at lower doses was observed with BBD-treatment (Figures ID, IE and 8C), demonstrating BET protein degradation was superior at inducing apoptosis in cancer cells than BET bromodomain inhibition.

Global proteomic characterization of BBD or BBI treated OVCAR8 cells using single run proteome analysis demonstrated MZ1 or ARV825 promoted distinct changes in global protein levels compared to BBI treatment (Figures IF and 1G). As expected, BRD2, BRD3 and BRD4 proteins were reduced by BBD but not BBI-treatment, and induced protein levels of HEXIM1 was observed only with BBI-treatment, consistent with previous reports (Figure 8D). Short-term exposure to BBI or BBD therapies commonly induced proteins involved in metabolism signaling, including citric acid (TCA) cycle and pyruvate metabolism (Figures 8E and 8F).

Notably, MZ1 or ARV825 -treatment uniquely downregulated proteins involved in ribosome and translation signaling, in addition to those regulating transcription and cell cycle signaling observed with BBI-treatment. Enrichment maps of pathways repressed by short-term BBD generated from single-run proteome profiles of OVCAR8 cells treated with BBDs (MZ1 or ARV825) for 48 hours were prepared (data not shown). Statistical differences in protein log2 LFQ z-scores comparing BBDs or BBIs to control treated cells was determined by paired T-Test Benjamini-Hochberg adjusted p values at FDR of <0.05 using Perseus Software. Proteins significantly repressed by BBD or BBI therapies were then analyzed by gProfiler to determine pathway enrichments (BH P<05). Enrichment maps were generated in Cytoscape using the Enrichment Map app v2.2.1. Enrichment maps of pathways repressed by short-term BBI generated from single-run proteome profiles of OVCAR8 cells treated with BBIs (JQ1 or OTX015) for 48 hours wer also prepred (data not shown). Statistical differences in protein log2 LFQ z-scores comparing BBDs or BBIs to control treated cells was determined.

Analysis of global expression changes in response to MZ1 or JQ1 by RNA-seq showed the treatments differentially altered the transcriptome landscape (Figures 8G and 8H). Comparison of global protein and RNA changes following BBD or BBI-treatment, revealed many of the proteins altered by BBI-treatment were commonly induced or repressed at the RNA level (r = 0.565) (Figure 1H). In contrast, BBD-induced changes in RNA and protein levels were less correlated (r = 0.330), with many genes downregulated at the protein level but not RNA, suggesting a significant component of the molecular response to BET protein degradation occurred post-transcriptionally (Figures II and 81).

Western blot analysis of JQ1 or MZ1 treated OVCAR8 cells confirmed the repression of translational signaling by BBD therapies. MZ1 -treatment distinctly reduced phosphorylation of FOX01/3 (T24/32), P70S6 Kinase (T389), pEIF4E (S209) and r4EBR1 (T37/46) relative to cis-MZl or JQ1 treatment (Figure 1J). Previous studies have shown BRD4 degradation blocks RNA pol II (S2) phosphorylation inhibiting transcriptional elongation. Consistent with these findings, BBD-treatment of OVCAR8 cells reduced RNA pol II (S2) phosphorylation that was not observed with JQ1. Moreover, in contrast to JQ1 therapy, MZ1 -treatment reduced MYC protein levels, demonstrating BRD4 degradation but not inhibition can block MYC signaling in OVCAR8 cells.

Kinome profiling of BBD or BBI treated OC cells using MIB-MS showed dynamic reprogramming of kinase signaling was similar within BBDs (ARV825 and MZ1) or BBIs (JQ1 and OTX015), but less similar amongst BBD and BBI (Figures IK andlL). Consistent with BBD-mediated translational repression, the majority of kinases induced by BBI treatment were unaltered or inhibited by BBD treatment, many encompassing the published JQ1 -kinome response signature (Figure 8J). MZl-treatment uniquely increased MIB-binding of INSR, CDK7, EPHA4 and EPHA2, as well as established BRD4 interacting proteins CSNK2A1, CSNK2A2, and CDK9 (Figure 1M). Overall, BBD-treatment significantly reduced kinase MIB-binding relative to BBI therapy, including the inhibition of established OC driver kinases EGFR, FGFR1, FGFR3, MET, BRAF, PTK2, PAK4, CHEK1 and RPS6KB1. MZ1 -induced changes in kinase RNA and MIB-binding exhibited poor correlation (r =0.394) compared to JQ1 -treatment (r = 0.595) illustrating the profound impact of BET protein degradation on kinase protein stability in OVCAR8 cells (Figures 8K and 8L). Western blot analysis of JQ1 or MZ1 treated OVCAR8 cells confirmed the distinct consequence of BET degradation, including the MZ1 -specific reduction of RTKs, EGFR, ERBB2, ERBB3, FGFR1, and IGF1R (Figure IN). Similarly, the majority of kinases induced by BBI treatment in additional OC cell lines KURAMOCHI or COV362 were either unaltered or reduced by BBD therapy, demonstrating BBD-treatment uniquely blocks compensatory kinome reprogramming observed with BBI-treatment (Figures 8M and 8N). Ovarian cancer cell lines KURAMOCHI and COV362 were treated with BET PROTAC MZ1 for 48 hours, which causes downregulation of RTKs protein levels such as FGFR1, FGFR2, FGFR3, IGF1R and ERBB4 that was not observed with BET inhibitor JQ1 (Figures 80 and 8P).

Together, proteogenomic characterization of BET protein degradation or inhibition in BRD4-driven OVCAR8 cells revealed BBD therapies uniquely inhibited translation, blocked kinome reprogramming observed with BBI therapies, as well as reduced MYC protein levels leading to enhanced apoptosis of OVCAR8 cells.

Example 2: Chronic Exposure to BBDs Fails to Maintain Sufficient Degradation of BET Proteins in OC Cells

To explore acquired resistance mechanisms to BBD or BBI therapies, resistant OVCAR8 cells through chronic exposure to increasing doses of JQ1, MZ1 or ARV825 were generated. The chronically exposed OVCAR8 cells (MZ1-R) or (ARV-R) were more resistant to BBD than treatment naive (i.e., parental cells), where they showed a rightward shift in MZ1 or ARV825 dose-response cell viability curves (Figures 2A and 2B). In contrast to parental OVCAR8 cells, treatment of MZ1-R or ARV-R cells with increasing doses of MZ1 or ARV825, respectfully, was insufficient to degrade BRD2, BRD3 or BRD4, as well as did not reduce BRD4-target FOSL1 protein levels to extent of parent cells (Figures 2C and 2D). Notably, ARV- R cells showed minimal levels of BRD3 following chronic exposure (Figure 2D). Cells chronically exposed to JQ1 (JQ1-R) were unaffected by increasing doses of JQ1, and JQ1- treatment failed to reduce BRD4 target FOSL1 (Figures 9A and 9B).

Characterization of BBD-R cells revealed BRD4 protein levels remained reduced in MZ1-R cells and BRD2, BRD3 and BRD4 reduced in ARV-R cells relative to parental cells (Figures 2E and 2F). Removal of BBDs from MZ1-R or ARV-R cells restored BET protein levels to those observed in parental cells and knockdown of CRBN (ARV-R) or VHL (MZ1-R) restored BET protein levels in BBD-R cells, demonstrating some degree of PROTAC degradation was still occurring in BBD-R cells (Figures 2E, 2F, 9C, and 9D). Genetic deletion of CRBN or CUL2, which is ubiquitin ligase recruited by VHL-based BBDs, was recently reported as resistance mechanisms to BBDs. Here, no significant reductions in RNA levels of CRBN were observed in ARV-R cells, though a slight reduction in CRBN protein relative to parental cells was detected by western blot (Figues 9E and 9F). CUL2 protein levels were unchanged in MZ1-R cells, while VHL RNA and protein levels showed a minor reduction relative to parental cells. No change in the CRBN-recruiting ubiquitin ligase CUL4 protein levels were detected in ARV-R cells (Figues 9E and 9F).

An increase in BRD2, BRD3 and BRD4 RNA levels was observed in MZ1-R and ARV- R cells relative to parental cells, while JQ1-R cells showed elevated BRD2 and BRD3 RNA levels, demonstrating chronic BBD treatment uniquely induced transcription of BRD4 (Figure 9F). Importantly, MZ1-R and ARV-R cells retained reliance on residual BRD4 protein and acquired dependency on BRD2, where knockdown of BRD2, BRD4 or both significantly reduced cell viability in BBD-R cells (Figure 2G). Moreover, depletion of BRD2 and/or BRD4 induced apoptosis and blocked BRD4-regulated MYC and FOSL1 signaling in MZ1-R cells (Figure 2H). Knockdown of BRD2 reduced RAF, MEK and ERK activity in MZ1-R cells, suggesting BRD2 may have unique function contributing to MZ1 resistance through promoting MAPK signaling (Figure 9G). Consistent with previous studies, knockdown of BRD4 in JQ1-R cells and to a lesser extent BRD2 also inhibited cell growth, demonstrating acquired resistance to either BBD or BBIs required BRD4 protein (Figure 2G).

Further analysis of BBD-R cells revealed MZ1-R cells were cross-resistant to ARV- 771, another VHL-based BET-PROTAC that utilized OTX015 as a warhead (Figure 21). Of particular interest, treatment of MZ1-R cells with CRBN-based PROTACs ARV825 (OTX015- based) or dBET6 (JQ1 -based) reduced BET proteins, inhibited cell viability and promoted apoptosis, demonstrating the CRBN-based degradation machinery remained functional and could be exploited to degrade BRD4 overcoming MZ1 resistance (Figures 2J, 2K, 9H, and 91). Similarly, ARV-R cells also showed cross resistance to other CRBN-based BBDs, dBET6, however, in contrast to MZ1-R cells, treatment of ARV-R cells with VHL-based MZ1 or ARV- 771 failed to reduce BET proteins, block growth or induce apoptosis, demonstrating ARV-R cells were resistant to both VHL and CRBN-based BET PROTACs (Figures 2L, 2M, 2N, and 9J).

MZ1-R or ARV-R cells were shown to be sensitive to BET inhibition by JQ1, where increasing concentrations of JQ1 reduced cell viability to a greater extent than parental cells, suggesting the BET bromodomain function of BRD2 and BRD4 was essential for cell growth and survival of BBD-R cells (Figures 20, 2P, and 9J). Resistance to BBI has been shown to occur though BET bromodomain independent functions such as interactions with MED1 rendering BBI-resistant cells sensitive to BRD4 knockdown. Treatment of JQ1- resistant OC cells with MZ1 or ARV825 degraded BET proteins, inhibited cell viability, reduced MYC/FOSL1 protein levels and induced apoptosis; demonstrating BBDs can overcome acquired resistance to BBIs (Figures 2Q, 2R, and 9K). PEOl ovarian cancer cells that have acquired resistance to BET inhibitor JQ1 can be killed by treatment with BET PROTAC MZ1 but not other BET inhibitors such as JQ1, AZD5153 or OTX015 (Figure 9L).

Example 3: Chronic BBD Therapy Remodels Kinome in OC Cells Upregulating INSR and mTOR/AKT Signaling

To explore the mechanistic basis for acquired resistance to BBDs, kinome, phosphoproteome and global proteome profiling of BBD and BBI-resistant OVCAR8 cells were carried out. PC A and Pearson correlation analysis revealed MZ1-R and ARV-R kinome signatures were overall distinct from JQ1-R or parental OVCAR8 cells (Figures 3 A and 3B). Volcano plots depict kinases exhibiting statistically significant changes in MIB-binding following chronic exposure to MZ1, ARV825 or JQ1 relative to parental OVCAR8 cells (Figures 3C, 10A, and 10B). Notably, MZ1-R and ARV-R kinome signatures were highly similar (r = 0.680), with the majority of kinases commonly induced or repressed, demonstrating resistance to CRBN or VHL-based BET PROTACs elicited similar kinome adaptations (Figues 3D and IOC).

Chronic exposure to JQ1 increased MIB-binding of kinases associated with IGF1R, PI3K, mTOR, MAPK, and ErbB signaling (Figure 10D). Similarly, increased MIB-binding of MTOR, RPS6KB1, BRAF, and MAPK1 were detected in BBD-R cells relative to parental cells, demonstrating mTOR and MAPK signaling were commonly activated by chronic BBI or BBD treatment (Figure 10E). KEGG and STRING pathway analysis of kinases commonly upregulated in MZ1-R and ARV-R cells revealed a network enriched in mTOR signaling, insulin signaling, regulation of actin cytoskeleton, PI3K-AKT signaling, and FoxO signaling, as well as LKB1/AMPK, NF-KB, environmental stress, hippo signaling, golgi trafficking, endocytosis and autophagy signaling (data not shown). Consistent MIB-MS profiles, kinase substrate enrichment analysis (KSEA) of alterations in global phosphorylation following chronic exposure to MZ1, predicted activation of kinases involved in PI3K/AKT signaling (AKT2/3), AMPK signaling (PRKAA1/PRKAA2), hippo signaling (STK3/MST4), cell cycle signaling (PIM1, PLK2 and NEK1), as well as several kinases involved in environmental stress (MAP2K6, MAP2K3, MAPK8, MAPK14 and MAPK10) (Figure 3E). Similarly, single-run proteome profiling of MZ1-R or parental cells showed chronic MZ1 exposure induced proteins involved in translation, mitochondrial translation, metabolism of proteins, membrane trafficking, vesicle-mediated transport, clathrin-mediated endocytosis, RHO GTPase signaling, cellular response to stress, infection signaling, and trans-golgi network vesicle budding. Proteins reduced by chronic MZ1- treatment were enriched in metabolism of RNA, RNA transport, RNA splicing, ribosome, and proteasome signaling (Figure 10G).

Receptor tyrosine kinase (RTK) array profiling and Western blot analysis confirmed activation of INSR in MZ1-R cells relative to parental cells (Figures 3F and 3G). An increase in EGFR, ERBB3 and AXL tyrosine phosphorylation was also observed by RTK array analysis in MZ1-R cells relative to parental cells (Figure 3H). Elevated phosphorylation of MTOR (S2448) and translation components p70S6K (T359), pEIF4E (S209) and r4EBR1 (T37/46), as well AKT-substrate FOX01 (T24/32) were confirmed by western blot, consistent with predicted activation of mTOR/PI3K and translation signaling by proteomics analysis (Figure 3G). Additionally, increased protein levels of MAP3K3 and phosphorylated c-Jun (S73), as well as PRKAA1 (AMPKa) were detected in MZ1-R cells, signifying increased cellular stress response and AMPK signaling. Phosphorylated RNA pol II at (S2), MYC and FOSL1, which were rapidly lost by short-term MZ1 -treatment, were detected by blot at similar levels in MZ1-R and parental cells, consistent with the return of BRD4 function in MZ1-R cells.

Collectively, proteomic analysis revealed chronic exposure to BBDs remodeled the kinome of OVCAR8 cells distinctly from BBI therapies. BBD-resistant cells activated a network of kinases involved in insulin, PI3K/mTOR, translation, MAPK signaling, environmental stress, endocytosis, autophagy and AMPK signaling, representing potential therapeutic avenues to block or overcome BBD-resistance in OC cells (Figure 3H).

Example 4: Chronic Exposure to BBDs Promotes Enhanced Sensitivity Towards INSR, ErbB and MTOR Inhibitors

To define growth and survival functions of kinases remodeled by chronic BBD treatment, an inhibitor screen in MZ1-R, JQ1-R or parental OVCAR8 cells was carried out (Figure 4A). The screen revealed MZ1-R cells were more sensitive to inhibitors targeting INSR/IGF1R (GSK1904529A), ErbB family (lapatinib), AKT (MK-2206), mTORCl (RAD001), AAK1 (LP-935509) and FAK1 (PND-1186) relative to JQ1-R cells or parental cells, consistent with predicted activation/upregulation of these kinases by MIB-MS and KSEA profiling (Figures 4B, 4C, 4D, 4E, 11A, and 1 IB). Inhibition of MEK (trametinib) or RAF (LY3009120) had the most profound impact on JQ1-R cell viability, while both MZ1-R and JQ1-R cells were more sensitive to PI3K inhibitor (GDC-0941), CDK9 inhibitor (NVP-2), or CK2 inhibitor (Cx-4945) than parental cells (Figures 4F, 11C, 11D, 11E, and 1 IF).

Additionally, MZ1-R and JQ1-R cells were shown to be more sensitive to PARP inhibitor (olaparib), as well as the autophagy inhibitor (hydroxychloroquine). These findings show BRD4 functions as a positive regulator of homologous recombination (HR), as well as negative regulator of autophagy in cancer cells (Figure 4A).

Knockdown of INSR EGFR, ERBB2, MTOR or AKT1/2 reduced cell viability in MZ1- R cells to a greater extent than parental cells, suggesting BBD-R cells acquired greater dependency on these kinases for cell growth and survival in the presence of continued BBD exposure (Figure 4G). Treatment of ARV-R cells with GSK1904529 A, lapatinib, RAD001, MK2206 and LP-935509 also reduced cell viability more than in parental cells, demonstrating VHL or CRBN-based BBD-resistant cells acquire common kinase dependencies (Figures 1 IF,

11G, and 11H). Moreover, treatment of ARV-R or MZ1-R cells with GSK1904529A (GSK529A) inhibited colony formation to a greater extent than parental cells (Figure 1 II). MZ1- R cells were more sensitive to AKT inhibition than JQ1-R or parental cells. Parental, JQ1-R or MZ1-R OVCAR8 cells were treated with escalating doses of MK2206 for 5 days and cell viability assessed by CellTiter-Glo. MZ1-R or JQ1-R inhibitor treated cell viabilities normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully (Figure 11J). MZ1-R cells were more sensitive to AAK1 inhibition than JQ1-R or parental cells. Parental, JQ1-R or MZ1-R OVCAR8 cells were treated with escalating doses of LP-935509 for 5 days and cell viability assessed by CellTiter-Glo. MZ1-R or JQ1-R inhibitor treated cell viabilities normalized to DMSO treated JQ1-R or MZ1-R cells, respectfully (Figure 11K). GSK529A-treatment blocked colony formation of MZ1-R and ARV-R cells to a greater extent than parental cells. Long-term 14-day colony formation assay of OVCAR8 parental, MZ1-R or ARV-R were treated with escalating doeses of GSK529A or DMSO. Colony formation was assessed by crystal violet staining (Figure 11L).

MIB-MS analysis of GSK529A-treated MZ1-R cells showed reduced MIB-binding of drug targets INSR and IGF1R and established downstream INSR signaling kinases MTOR, RPS6KB1, BRAF, MAP2K1/2 and MAPK3 (ERK1) (Figure 4H). Reduced MIB-binding of kinases involved in cell cycle (WEE1, CHEK1, CDK2, CDK6 and PLK1), as well as kinases required for translation (EIF2AK1, EIF2AK2, and EIF2AK4) were also observed following GSK529A treatment. Inhibition of INSR/IGF1R, MTOR, P70S6K, BRAF and ERK activity in MZ1-R cells was confirmed by phospho-blot (Figure 41). Single-run proteome analysis of GSK529A treated MZ1-R cells showed inhibition of INSR signaling repressed mRNA processing, RNA splicing, translation initiation and mitosis signaling, consistent with inhibition of mTOR signaling and cycle arrest. Enrichment maps of pathways repressed by INSR inhibition generated from MIB-MS profiles of OVCAR8 cells treated with GSK529A (1 mM) treatment for 48 hours. Statistical differences in protein log2 LFQ z-scores comparing GSK529A to control treated MZ1-R cells was determined by paired T-Test Benjamini- Hochberg adjusted p values at FDR of <0.05 using Perseus Software. Proteins significantly repressed by GSK529A therapies were then analyzed by gProfiler to determine pathway enrichments (BH P<05). Enrichment maps were generated in Cytoscape using the Enrichment Map app v2.2.1. (data not shown).

Example 5: Blockade of MTOR Signaling Destabilizes BET Proteins Overcoming Resistance to BET Degraders

Proteomics profiling and Western blot analysis showed BRD2 and BRD4 protein levels were significantly reduced by GSK529A treatment in MZ1-R cells, suggesting inhibition of INSR and downstream mTOR signaling may impact BRD2 and BRD4 protein stability in BBD- R cells (Figures 5 A and 12A). Moreover, a dose dependent reduction in MYC protein levels and an induction of apoptosis was also observed in response to GSK529A-treatment that coincided with BET protein loss. Treatment of parental cells or JQ1-R cells with GSK529A did not reduce BET protein levels, suggesting the response uniquely occurred in BBD-R cells (Figure 5B).

Additionally, treatment of MZ1-R or ARV-R cells with lapatinib (ErbB), RAD001 (mTORCl), or Torin-1 (mTORCl/2) but not CDK9, CDK7, FGFR CK2 or AKT inhibitors, reduced BRD2 and BRD4 levels in MZ1-R and ARV-R cells (Figures 5C, 5D, 12B, 12C, and 12D). Notably, GSK529A, lapatinib, RAD001 and Torin-1 treatment reduced MTOR-mediated phosphorylation of P70S6K (T359), suggesting these inhibitors commonly blocked MTOR signaling in BBD-R cells. Treatment of MZ1-R but not parental or JQ1-R cells with RAD001 reduced BRD4 protein levels, signifying the MTOR-dependent protection of BRD2/4 was distinct to cells chronically exposed to BBDs (Figures 5E and 12E). BRD2, BRD3 or BRD4 RNA levels were unaffected by GSK529A, Lapatinib or RAD001 treatment in MZ1-R cells, demonstrating the reduced levels of BRD2 and BRD4 were not due to blockade of transcription (Figure 5F). The reduction in BET proteins mediated by 4 or 48 hours GSK529A or RAD001 treatment was rescued by proteasome inhibitor treatment, signifying blockade of MTOR activity impacts BRD4 post transcriptionally (Figures 5G and 5H). Furthermore, inhibition of phosphorylated RNA pol II (S2) was observed in MZ1-R cells following a 48 hour treatment with RAD001 that could be reversed by bortezomib treatment, consistent with MTOR signaling regulating BRD4 protein function in MZ1-R cells.

Example 6: MTOR-driven Stabilization of BRD4 A Global Mechanism of Resistance to BBD Therapies in OC Cells

To further define the role of MTOR signaling in resistance to BBD therapies, additional BBD-R OC cell lines were characterized. The chronically exposed MZ1-R cells (KURAMOCHI, COV362 and OVCAR5) were more resistant to BBD than treatment naive (i.e., parental cells), where they showed a rightward shift in MZ1 dose-response cell viability curves and treatment with MZ1 failed to mediated degradation of BET proteins (Figures 6A, 6B, 6C, 6D, 6E, and 6F). Consistent with OVCAR8 MZ1-R cells, knockdown of BRD4 and to a lesser extent BRD2 inhibited cell viability of OVCAR5, COV362 or KURAMOCHI MZ1-R cells, confirming the dependency of MZ1-R cells for residual BRD4 protein levels (Figures 13A, 13B, and 13C). Treatment of OVCAR5, COV362 and KURAMOCHI MZ1-R cells with GSK529A, lapatinib or RAD001 significantly blocked cell growth to a greater extent than parental cells (Figures 6G,

6H, and 61). Treatment of OVCAR5 JQ1-R cells with GSK529A, lapatinib or RADOOl showed minimal growth inhibition, further demonstrating exquisite sensitivity to INSR/IGF1R, EGFR and mTORCl inhibition was uniquely acquired by chronic BBD treatment but not BBI therapies (Figures 13D, 13E, and 13F). Lastly, GSK529A, lapatinib or RADOOl treatment in COV362, OVCAR5 and KURAMOCHI MZ1-R cells reduced BRD2 and BRD4 proteins, suggesting the INSR, ErbB and mTORCl -mediated stabilization of BET proteins in response to chronic exposure to BBD was a common mechanism of resistance to BBD therapies amongst OC cell lines (Figures 6J, 6K, and 6L).

Example 7: INSR, ErbB and/or mTORCl Inhibition Improves PROT AC-mediated Degradation of BRD4 Providing Superior Growth Repression and Apoptosis of OC Cells

Combined treatment of OVCAR8 cells with either JQlor MZ1 and KIs targeting ATR, PI3K, MEK, CK2, CDK7, or CDK9 enhanced growth repression, demonstrating BBDs similarly synergizes with KIs previously shown to improve JQ1 efficacy in OC cells (Figures 14A, 14B, 14C, 14D, 14E, and 14F). Co-treatment of OVCAR8 cells with MZ1 or GSK529A, lapatinib or RADOOl uniquely enhanced growth inhibition and increased apoptosis relative to single agent therapies, whereas combined treatment with JQ1 or cis-MZl showed no synergy (Figure 7A). Moreover, increasing JQ1 concentrations to 500 nM did not improve synergy with GSK529A, lapatinib or RADOOl, demonstrating the combination therapies were only active using BBDs (Figures 14G and 14H). The drug synergy was also observed when combining CRBN-based BBD, dBET6, demonstrating either VHL or CRBN-based BBD therapies can be used for the kinase inhibitor combination therapies (Figure 7B).

Increased apoptosis and a complete reduction in BET proteins was observed in OVCAR8 cells when MZ1 was combined with GSK529A, RAD001, or lapatinib, suggesting blockade of INSR, ErbB and/or MTOR enhanced MZ1 -mediated degradation of BET proteins improving OC cell killing (Figures 7C and 7D). The benefit of co- treating cells with MZ1 and GSK529A, RAD001, or lapatinib was further illustrated, where dose escalation of RAD001 or GSK529A promoted enhanced degradation of BRD4, as well as increased levels of cleaved PARP than single agent therapies (Figures 7E and 7F). These findings suggest MTOR signaling may be promoting BRD4 stability following short-term BBD-therapy, similar to that observed when OC cells were chronically exposed to BBDs. Contrary, co-treatment of OVCAR8 cells with JQ1 and RAD001 or GSK529A had no impact on BRD4 protein levels (Figure 141).

Combination therapies involving GSK529A, lapatinib or RAD001 and MZ1 but not JQ1 significantly enhanced growth repression across a panel EOC cell lines harboring genomic alterations frequently observed in OC patient tumors (Figures 7G, 14J, 14K, and 14L). Cancer cell lines harboring MYC amplifications have been shown to exhibit enhanced sensitivity towards BBI therapies. Here, MYC-amplified HGSOC cell lines KURMAOCHI, OVCAR4, and COV362 showed pronounced synergy to combinations of BBDs and GSK529A, lapatinib, or RAD001 (Figures 7G and 7H). Moreover, BRD4 amplification and/or overexpression has been associated with poor prognosis and survival in HGSOC. Importantly, doses as low as 10 nM of MZ1 were sufficient to induce sensitivity towards GSK529A, lapatinib or RAD001 in BRD4 overexpressing HGSOC cell line OVSAHO (Figures 14M and 14N). Alterations in PI3K and RAS signaling including activating KRAS or PIK3CA mutations have been shown to promote intrinsic resistance to BBI therapies. Notably, treatment of RAS-altered EOC cell lines OVCAR5, JHOS-2, A1847, COV362 and CAOV3, or PIK3CA mutant SKOV3 cells with the BBD/kinase combination therapies significantly enhanced growth inhibition compared to single agents, without the requirement of MEK or PI3K inhibitors (Figures 7G, 14J, 14K, and 14L). KURAMOCHI cells were sensitive to the combination of dBET6 and MTOR inhibitor RAD001, while single agents were less effective. KURAMOCHI cells were treated with 100 nM dBET6, 100 nM JQ1 or the combination of dBET6 and RAD001 or JQ1 and RAD001 for 5 days and cell viability assessed by CellTitr Glo (Figure 71). The proposed model of the protection of BRD4 from PROTAC-degradation by MTOR signaling in ovarian cancer cells is shown in Figure 7J. PROTAC-mediated degradation of BRD4 induces RTK signaling resulting in increased MTOR signaling which prevents the degradation of BRD4 in cells resistant to BET PROTACs.

Overall, combining BBDs and kinase inhibitors GSK529A, lapatinib or RAD001 was superior at blocking growth of EOC cells than BBI-KI combinations or single agent therapies, representing promising BBD-selective combination therapies for the treatment of ovarian cancer. In contrast to BBI therapy, BET degradation uniquely blocked translation signaling, preventing kinome reprogramming observed with BBI-treatments, leading to superior blockade of MYC signaling and induction of apoptosis. OC cells overcame chronic BBD therapy via CDK9- dependent upregulation of BRD4 transcription and activation of INSR, ErbB and mTORCl- signaling, which protected BRD4 from PROTAC -mediated degradation. Inhibition of INSR, ErbB or mTORCl in BBD-R cells resulted in proteasome-dependent degradation of BRD4 and subsequent apoptosis, while inhibition of CDK9 blocked BRD4 transcription in BBD-R cells overcoming BBD-resistance. Moreover, combination therapies involving BBDs and INSR, ErbB, or mTORCl inhibitors improved PROTAC -mediated degradation of BRD4 in parental OC cells enhancing BBD-induced apoptosis, representing a promising therapy for the treatment of OC.

Example 8: CDK9 PROTAC Resistance is Overcome by Co-administration of Kinase Inhibitors

Chronic exposure of OVCAR8 cells to CDK9 PROTAC (Thai SNS 032) sensitized cells to RAD001 or GSK1904529A. Treatment of OVCAR8 Thai SNS 032 resistant cells with RAD001 or GSK529A resulted in downregulation of CDK9, demonstrating blockade of MTOR signaling re-sensistizes OVCAR8 cells to PROTAC-degradation. Moreover, these studies show both BET PROTACs and CDK9 PROTACs elicit a similar dependency on MTOR signaling to promote drug resistance. Co-treatment of parental OVCAR8 cells with Thai SNS 032 and RAD001 or GSK1904529A reduced cell viability to a greater extent than single agents alone, signifying blockade of MTOR signaling can improve the efficacy of CDK9-PROTAC degradation. In particular, OVCAR8 cells were treated with CDK9-degrader Thai SNS 032, resulting in the reduction of CDK9 protein levels. OVCAR8 cells were treated with escalating doses of Thai SNS 032 and CDK9 protein levels were determined by Western blot (Figure 15A). Chronic exposure to Thai SNS 032 failed to degrade CDK9 that is overcome by GSK529A, lapatinib or RAD001 treatment. OVCAR8 Thal-R cells were treated with Thai (500 nM) or (2 mM) GSK529A, lapatinib, or RAD001 for 4 hours and protein levels were assessed by Western blot. (Figure 15B). Thal-R cells were more sensitive to INSR, ErbB or MTORCl inhibition than parental cells. Parental or Thal-R OVCAR8 cells were treated with escalating doses of GSK1904529A (Figure 15C), lapatinib (Figure 15D), or RAD001 (Figure 15E) for 5 days and cell viability was assessed by CellTiter-Glo. Thal-R inhibitor treated cell viabilities were normalized to DMSO treated Thal-R cells. Combination therapies involving GSK529A or RAD001 and Thai SNS 032 improved growth repression of OVCAR8 cells. OVCAR8 cells were treated with 100 nM Thai SNS 032, escalating doses of GSK529A or RAD001, or the combination of Thai SNS 032 and kinase inhibitors, for 5 days and cell viability was assessed by CellTiter-Glo (Figures 15F and 15G).

Example 9: MTORC1 Inhibition Enhances PROTAC-Mediated Degradation of Protein Targets

MTORC1 inhibition enhanced PROTAC-meditated degradation of BRD4, CDK9 or FAK1 proteins in cancer cells. Briefly, A1847 cells were treated with DMSO, 2 mM RAD001, increasing doses of dBET6 or 2 pM RAD001 + increasing doses of dBET6 for 4 hours and BRD4 protein was assessed by Western blot (Figure 16, Panel A). A1847 cells were treated with DMSO, 2 pM RAD001, increasing doses of Thai SNS 032 or 2 pM RAD001 + increasing doses of Thai SNS 032 for 4 hours and CDK9 protein was assessed by Western blot (Figure 16, Panel B). A1847 cells were treated with DMSO, 2 pM RAD001, increasing doses of FAK-Protac or 2 pM RAD001 + increasing doses of FAK-Protac for 4 hours and CDK9 protein was assessed by Western blot (Figure 16, Panel C).

Immunoblot analysis showed inhibition of mTORCl signaling by RAD001 improved MZ1 or dBET6-mediated degradation of BRD4 lowering the concentration of BBD required to achieve maximal protein degradation (Figure lg, Panel A). Notably, a 100-fold reduction in concentration required to degrade BRD4 was observed when dBET6 was combined with RAD001, while potent enhancement was observed with MZ1 -treatment. Importantly, the improved degradation at lower concentration of BBDs was also observed in additional cancer cell lines MOLT4, SUM159 and HCT116. Furthermore, blockade of mTORCl also enhanced degradation of CDK9 by Thai SNS 032, where RAD001 co-treatment significantly reduced the concentration of Thai SNS 032 required to achieve maximal degradation of CDK9 in A1847 cells (Figure 16, Panel B). Notably, RAD001 -treatment improved degradation efficiency of other PROTACs targeting FAK1 (FAK-PROTAC), suggesting blockade of mTORCl may enhance degradation of PROTACs targets, reducing the concentration of degraders required to deplete proteins (Figure 16, Panel C). Thus, MTORCl inhibition may represent a general therapeutic strategy to improve PROTAC -induced degradation of protein targets and improve efficacy of PROTACs by minimizing concentrations of PROTACs required for reduce protein targets in cells.

Example 10: MTORC1 Inhibition Facilitates Degradation of Protein Targets in Cancer Cells Intrinsically Resistant to PROTAC Therapy

Blockade of MTORC1 signaling sensitizes cancer cells resistant to BET-PROTACs. Briefly, a Cell-Titer Glo assay was performed for cell viability of A1847 cell lines treated with increasing concentrations of RAD001, dBET6 or the combination of RAD001 and dBET6 and cultured for 120 hours. Data were analyzed as % cell viability of DMSO control, presented as means of 3 independent assays. A dose response matrix of RAD001 or dBET6 in A1847 cells is shown in Figure 17, Panel A. DLD-1 cells were treated with DMSO, 2 mM RAD001, increasing doses of dBET6 or 2 pM RAD001 + increasing doses of dBET6 for 4 hours and BRD4 protein assessed by Western blot. An immunoblot analysis of BRD4 protein levels is shown in Figure 17, Panel B. A drug synergy analysis of RAD001 and dBET6 in DLD-1 cells (N=3) is shown in Figure 17, Panel C. Spheroid assays were performed wherein DLD-1 cells were treated with DMSO, 2 pM RAD001, 1 nM dBET6 or 2 pM RAD001 + 1 nM dBET6 and spheroid formation was assessed (see, Fure 17, Panel D).

Several cancer cell lines have been reported to be intrinsically resistant to BBDs, including the colorectal cancer cell line DLD-1. Here, DLD-1 cells were shown to be resistant to dBET6 (Figure 17, anel A) with GI50’s > 1 pM requiring high concentrations of dBET6 to reduce BET proteins and inhibit protein synthesis (Figure 17, Panel B). DLD-1 cells harbor activating mutations in the K-Ras and PIK3CA genes, both known to strongly potentiate mTORCl signaling. Notably, combined treatment of RAD001 and dBET6 enhanced the degradation of BRD4 in DLD-1 cells, markedly reducing BRD4 levels, compared to treatment with single agent dBET6 (Figure 17, Panel B). Strong drug synergy was observed in RADOOl and dBET6 treated cells with Bliss synergy score of 24.17 (Figure 17, Panel C) and the combination blocked spheroid growth in 3D assays while single agents were ineffective, demonstrating RADOOl -treatment can sensitize resistant cells to BBDs (Figure 17, Panel D). Thus, cancer cells intrinsically resistant to BET protein PROTACs can be made sensitive by blocking mTORCl signaling, supporting combination therapies involving MTORCl and BET protein degraders for the treatment of these types of cancers. Example 11: Activation of K-ras Signaling Promotes Resistance to BET-PROTACs that can be Reversed by MTORC1 Inhibitors

K-ras mutations promote resistance to BET protein degraders that can be overcome by MTORC1 inhibitors. Briefly, isogenic SW48 colorectal cancer cells expressing wild-type or mutant (G12D) K-ras were treated with increasing doses of MZ1 and cell viability was assessed. Expression of K-ras mutants promote resistance to MZ1 therapy (Figure 18, Panel A). Immunoblot analysis depicts activation of translation by forced expression of K-ras G12D mutants in SW48 cells (Figure 18, Panel B). SW48 cells expressing wild-type or K-ras G12D mutants were treated with DMSO, 2 mM RAD001, increasing doses of MZ1 or 2 mM RAD001 + increasing doses of MZ1 for 4 hours and BRD4 protein assessed by Western blot. Immunoblot analysis of BRD4 protein levels are shown in Figure 18, Panel C. A Cell-Titer Glo assay was performed for cell viability of SW48 (KRAS G12D) cell lines treated with increasing concentrations of RAD001, MZ1 or the combination of RAD001 and MZ1 and cultured for 120 hours. Data were analyzed as % cell viability of DMSO control, presented as means of 3 independent assays. A dose synergy analysis of RAD001 or MZ1 in K-ras G12D mutant SW48 cells is shown in Figure 18, Panels D and E. Immunoblot analysis for BRD4 protein levels in response to combination is shown in Figure 18, Panel F. A Cell-Titer Glo assay was performed for cell viability of SW48 (KRAS WT) cell lines treated with increasing concentrations of RAD001, MZ1 or the combination of RAD001 and MZ1 and cultured for 120 hours. Data were analyzed as % cell viability of DMSO control, presented as means of 3 independent assays. A dose synergy analysis of RADOOl or MZ1 in K-ras wild-type SW48 cells is shown in Figure 18, Panels G and H. An immunoblot analysis for BRD4 protein levels in response to combination is shown in Figure 18, Panel I.

To explore the impact of K-ras mutations on BBD responses, isogenic colorectal cancer cell lines harboring a knockin G12D K-Ras mutation or wild-type K-Ras were treated with MZ1 (Figure 18, Panel A). Cells harboring K-Ras mutations displayed elevated translation signaling (Figure 18, Panel B), and were less sensitive to MZ1 than parental cells exhibiting a 10-fold shift in GI50 from 0.63 to 63 nM. Moreover, knock-in of the K-Ras mutation impaired the degradation of BRD4 by MZ1 relative to parental cells, requiring higher concentrations of MZ1 to achieve complete degradation of BRD4 (Figure 18, Panel C). Inhibition of mTORCl signaling with RADOOl improved MZl-mediated degradation of BRD4 in K-Ras expressing cells lowering the concentration of MZ1 required to achieve maximal degradation. Combined treatment of K-Ras isogeneic cells with RADOOl and MZ1 was highly synergistic at inhibiting cell viability with a Bliss synergy score of 21.041, as well as improved degradation of BRD4 relative to single agents (Figure 18, Panels D, E, and F). In contrast, the combination therapy displayed less synergy (bliss synergy score of 7.59) in K-Ras WT cells, as single agent MZ1 inhibited viability > 50% at low concentrations (Figure 18, Panels G, H, and I). To summarize, these experiments indicate that cancer cells harboring K-Ras mutations or other mutations that strongly activate mTORCl signaling can promote intrinsic resistance to BBDs representing a patient population that may benefit from combined mTORCl and PROT AC -treatment.

Various modifications of the described subject matter, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference (including, but not limited to, journal articles, U.S. and non-U.S. patents, patent application publications, international patent application publications, gene bank accession numbers, and the like) cited in the present application is incorporated herein by reference in its entirety.