Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
KINASE INHIBITORS FOR THE TREATMENT OF PROSTATE CANCER
Document Type and Number:
WIPO Patent Application WO/2023/039548
Kind Code:
A1
Abstract:
Treatment for prostate cancer, including castration-resistant prostate cancer, by utilizing a protein kinase inhibitor selected from an SC-1 compound, a PP121 compound, a PD166285 dihydrochloride (diHCI) compound, an NCGC00348110 compound, a Dasatinib compound, a PDK1/Akt/Fit DPI compound, a Staurosporine compound, and/or a Cdk 1/2 Inhibitor III compound, optionally in combination with an androgen receptor signaling inhibitor are described. Assays to predict in vivo effectiveness of potential therapeutics against prostate cancer are also provided.

Inventors:
GUJRAL TARANJIT (US)
BELLO THOMAS (US)
CHAN MARINA (US)
Application Number:
PCT/US2022/076237
Publication Date:
March 16, 2023
Filing Date:
September 09, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
FRED HUTCHINSON CANCER CENTER (US)
International Classes:
A61K31/519; C07D403/06; C07D487/04; A61P35/00
Domestic Patent References:
WO2017049166A12017-03-23
Foreign References:
US20190231778A12019-08-01
US20190047990A12019-02-14
US7642255B22010-01-05
Other References:
DATABASE PUBCHEM SUBSTANCE ANONYMOUS : "NCGC00167403-01", XP093047067, retrieved from PUBCHEM
Attorney, Agent or Firm:
WINGER, C. Rachal et al. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A method of treating prostate cancer (PCa) in a subject in need thereof comprising administering a therapeutically effective amount of SC-1 or PP121 to the subject thereby treating the PCa in the subject in need thereof.

2. A method of treating prostate cancer (PCa) in a subject in need thereof comprising administering a therapeutically effective amount of an SC-1 compound, a PP121 compound, a PD166285 dihydrochloride (diHCI) compound, an NCGC00348110 compound, a Dasatinib compound, a PDK1/Akt/Fit DPI compound, a Staurosporine compound, and/or a Cdk 1/2 Inhibitor III compound to the subject thereby treating the PCa in the subject in need thereof.

3. The method of claim 2, further comprising administering a therapeutically effective amount of an androgen receptor (AR) signaling inhibitor to the subject in need thereof.

4. The method of claim 2, wherein the PCa is castration-resistant PCa (CRPC).

5. The method of claim 2, wherein the kinase inhibitor comprises an SC-1 compound and/or a PP121 compound.

6. The method of claim 2, wherein the SC-1 compound comprises (Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4)ccc2C)C3)n(C)n1);

Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N6CCOCC6)CC5)c(C(F)(F)F)c 4)ccc2C)C3)n(C)n1 ;

C=CC(=O)N1CCCN(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4)ccc2C)C3)n1 C;

CCc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N(C)C)C5)c(C(F)(F)F)c4)ccc2 C)C3)n(C)n1 ;

Cc1cc(N(C)c2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N(C)C)C5)c(C(F)(F)F)c4)cc c2C)C3)n(C)n1 ;

Cc1ccc(NC(=O)c2cccc(C(F)(F)F)c2)cc1 Nc1cc(CN(C)CCO)nc(NCCCO)c1 ;

N-{3-[7-(3-Dimethylamino-phenylamino)-1-methyl-2-oxo-1 ,4-dihydro-2H-pyrimido[4,5- d]pyrimidin-3-yl]-4-methyl-phenyl}-3-(4-methyl-imidazol-1-yl)-5-trifluoromethyl-benzamide;

N-{3-[7-(3-Amino-phenylamino)-1-methyl-2-oxo-1 ,4-dihydro-2H-pyrimido[4,5-d]pyrimidin-3-yl]-4- methyl-phenyl}-3-trifluoromethyl-benzamide; or N-[4-Methyl-3-(1-methyl-7-methylamino-2,4-dioxo-1 ,4-dihydro-2H-pyrimido[4.5-d]pyrimidin-3- yl)-phenyl]-3-trifluoromethyl-benzamide.

7. The method of claim 2, wherein the PP121 compound comprises

(Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2C1 CCCC1 );

72 Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2CC0;

CC1 C0CC1 (C)n1 nc(-c2cnc3[nH]ccc3c2)c2c(N)ncnc21 ;

CCS(=0)c1 ccc(-n2nc(-c3cnc4[nH]ccc4c3)c3c(N)ncnc32)cc1 ;

Nc1ncnc2c1c(-c1 ccc(Oc3ccccc3) cc1)nn2C1CCNCC1;

CN(CC(=O)c1cnc2[nH]ccc2c1)C1CCCC1;

Cn 1 c(=O)c(c2cnc3[n H]ccc3c2)cn 1 CC(C) (C)O;

N-[4-[1-(1,4-dioxaspiro[4.5]dec-8-yl)-4-(8-oxa-3-azabicyclo[3.2.1]oct-3-yl)-1 H-pyrazolo[3,4- d]pyrimidin-6-yl]phenyl]-N'-methyl-(WYE-125132; methyl 4-(4-morpholino-1-(1-(pyridin-3-ylmethyl)piperidin-4-yl)-1H-pyrazolo[3,4-d]pyrimidin-6- yl)phenylcarbamate (WYE-687);

6-(1H-indol-5-yl)-4-morpholino-1-(1-(pyridin-3-ylmethyl)piperidin-4-yl)-1 H-pyrazolo[3,4- d]pyrimidine (WAY-600);

3-(2,4-bis((S)-3-methylmorpholino)pyrido[2,3-d]pyrimidin-7-yl)-N-methylbenzamide (AZD2014); or

5-(7-(methylsulfonyl)-2-morpholino-6,7-dihydro-5H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrimidin-2- amine (CH5132799).

8. The method of claim 3, wherein the AR signaling inhibitor comprises surgical removal of the testicles.

9. The method of claim 3, wherein the AR signaling inhibitor comprises abiraterone, apalutamide (ARN-509), AZD3514, chlormadinone acetate, darolutamide (ODM-201), EPI-001 , enzalutamide, finasteride, galeterone, izonsteride, ketoconazole, megestrol, steroidal anti-androgens, nadolol, N-butylbenzene-sulfonamide, RU58841 , Testolone (RAD140), triptophenolide, tulobuterol hydrochloride, and/or turosteride.

10. The method of claim 32, wherein the AR signaling inhibitor comprises abiraterone and/or enzalutamide.

11. A composition comprising a combination therapy comprising (i) an SC-1 compound, a PP121 compound, a PD166285 dihydrochloride (diHCI) compound, an NCGC00348110 compound, a Dasatinib compound, a PDK1/Akt/Fit DPI compound, a Staurosporine compound, or a Cdk 1/2 Inhibitor III compound; and (ii) an androgen receptor (AR) signaling inhibitor.

12. The composition of claim 11, wherein the composition comprises an SC-1 compound or a PP121 compound.

13. The composition of claim 12, wherein the SC-1 compound comprises (Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4)ccc2C)C3)n(C)n1);

Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N6CCOCC6)CC5)c(C(F)(F)F)c

73 4)ccc2C)C3)n(C)n1;

C=CC(=O)N1CCCN(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4)ccc2C)C3)n1

C;

CCc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N(C)C)C5)c(C(F)(F)F)c4)ccc2

C)C3)n(C)n1;

Cc1cc(N(C)c2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N(C)C)C5)c(C(F)(F)F)c4)cc c2C)C3)n(C)n1 ;

Cc1ccc(NC(=O)c2cccc(C(F)(F)F)c2)cc1Nc1cc(CN(C)CCO)nc(NCCCO)c1;

N-{3-[7-(3-Dimethylamino-phenylamino)-1-methyl-2-oxo-1,4-dihydro-2H-pyrimido[4,5- d]pyrimidin-3-yl]-4-methyl-phenyl}-3-(4-methyl-imidazol-1-yl)-5-trifluoromethyl-benzamide;

N-{3-[7-(3-Amino-phenylamino)-1-methyl-2-oxo-1 ,4-dihydro-2H-pyrimido[4,5-d]pyrimidin-3-yl]-4- methyl-phenyl}-3-trifluoromethyl-benzamide; or

N-[4-Methyl-3-(1-methyl-7-methylamino-2,4-dioxo-1 ,4-dihydro-2H-pyrimido[4.5-d]pyrimidin-3-yl)- phenyl]-3-trifluoromethyl-benzamide.

14. The composition of claim 12, wherein the PP121 compound comprises

(Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2C1 CCCC1 );

Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2CCO;

CC1 COCC1 (C)n1 nc(-c2cnc3[nH]ccc3c2)c2c(N)ncnc21 ;

CCS(=O)c1 ccc(-n2nc(-c3cnc4[nH]ccc4c3)c3c(N)ncnc32)cc1 ;

Nc1ncnc2c1c(-c1 ccc(Oc3ccccc3) cc1)nn2C1CCNCC1;

CN(CC(=O)c1cnc2[nH]ccc2c1)C1CCCC1;

Cn 1 c(=O)c(c2cnc3[n H]ccc3c2)cn 1 CC(C) (C)O;

N-[4-[1-(1,4-dioxaspiro[4.5]dec-8-yl)-4-(8-oxa-3-azabicyclo[3.2.1]oct-3-yl)-1 H-pyrazolo[3,4- d]pyrimidin-6-yl]phenyl]-N'-methyl-(WYE-125132; methyl 4-(4-morpholino-1-(1-(pyridin-3-ylmethyl)piperidin-4-yl)-1H-pyrazolo[3,4-d]pyrimidin-6- yl)phenylcarbamate (WYE-687);

6-(1H-indol-5-yl)-4-morpholino-1-(1-(pyridin-3-ylmethyl)piperidin-4-yl)-1 H-pyrazolo[3,4- d]pyrimidine (WAY-600);

3-(2,4-bis((S)-3-methylmorpholino)pyrido[2,3-d]pyrimidin-7-yl)-N-methylbenzamide (AZD2014); or

5-(7-(methylsulfonyl)-2-morpholino-6,7-dihydro-5H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrimidin-2- amine (CH5132799).

15. The composition of claim 11 , wherein the AR signaling inhibitor comprises abiraterone, apalutamide (ARN-509), AZD3514, chlormadinone acetate, darolutamide (ODM-201), EPI-001,

74 enzalutamide, finasteride, galeterone, izonsteride, ketoconazole, megestrol, steroidal antiandrogens, nadolol, N-butylbenzene-sulfonamide, RU58841, Testolone (RAD140), triptophenolide, tulobuterol hydrochloride, and/or turosteride.

16. A kit comprising a combination therapy comprising (i) an SC-1 compound, a PP121 compound, a PD166285 dihydrochloride (diHCI) compound, an NCGC00348110 compound, a Dasatinib compound, a PDK1/Akt/Fit DPI compound, a Staurosporine compound, and/or a Cdk 1/2 Inhibitor III compound; and (ii) an androgen receptor (AR) signaling inhibitor.

17. The kit of claim 16, wherein the kinase inhibitor comprises an SC-1 compound or a PP121 compound.

18. The kit of claim 17, wherein the SC-1 compound comprises (Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4)ccc2C)C3)n(C)n1);

Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N6CCOCC6)CC5)c(C(F)(F)F)c 4)ccc2C)C3)n(C)n1;

C=CC(=O)N1CCCN(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4)ccc2C)C3)n1 C;

CCc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N(C)C)C5)c(C(F)(F)F)c4)ccc2 C)C3)n(C)n1;

Cc1cc(N(C)c2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N(C)C)C5)c(C(F)(F)F)c4)cc c2C)C3)n(C)n1 ;

Cc1ccc(NC(=O)c2cccc(C(F)(F)F)c2)cc1Nc1cc(CN(C)CCO)nc(NCCCO)c1;

N-{3-[7-(3-Dimethylamino-phenylamino)-1-methyl-2-oxo-1,4-dihydro-2H-pyrimido[4,5- d]pyrimidin-3-yl]-4-methyl-phenyl}-3-(4-methyl-imidazol-1-yl)-5-trifluoromethyl-benzamide;

N-{3-[7-(3-Amino-phenylamino)-1-methyl-2-oxo-1,4-dihydro-2H-pyrimido[4,5-d]pyrimidin-3-yl]-4- methyl-phenyl}-3-trifluoromethyl-benzamide; or

N-[4-Methyl-3-(1-methyl-7-methylamino-2,4-dioxo-1 ,4-dihydro-2H-pyrimido[4.5-d]pyrimidin-3-yl)- phenyl]-3-trifluoromethyl-benzamide.

19. The kit of claim 17, wherein the PP121 compound comprises

(Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2C1 CCCC1 );

Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2CCO;

CC1 COCC1 (C)n1 nc(-c2cnc3[nH]ccc3c2)c2c(N)ncnc21 ;

CCS(=O)c1 ccc(-n2nc(-c3cnc4[nH]ccc4c3)c3c(N)ncnc32)cc1 ;

Nc1ncnc2c1c(-c1 ccc(Oc3ccccc3) cc1)nn2C1CCNCC1;

CN(CC(=O)c1cnc2[nH]ccc2c1)C1CCCC1;

Cn 1 c(=O)c(c2cnc3[n H]ccc3c2)cn 1 CC(C) (C)O;

75 N-[4-[1-(1 ,4-dioxaspiro[4.5]dec-8-yl)-4-(8-oxa-3-azabicyclo[3.2.1]oct-3-yl)-1 H-pyrazolo[3,4- d]pyrimidin-6-yl]phenyl]-N'-methyl- (WYE-125132; methyl 4-(4-morpholino-1-(1-(pyridin-3-ylmethyl)piperidin-4-yl)-1 H-pyrazolo[3,4-d]pyrimidin-6- yl)phenylcarbamate (WYE-687);

6-(1 H-indol-5-yl)-4-morpholino-1-(1-(pyridin-3-ylmethyl)piperidin-4-yl)-1 H-pyrazolo[3,4- d]pyrimidine (WAY-600);

3-(2,4-bis((S)-3-methylmorpholino)pyrido[2,3-d]pyrimidin-7-yl)-N-methylbenzamide (AZD2014); or 5-(7-(methylsulfonyl)-2-morpholino-6,7-dihydro-5H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrimidin-2- amine (CH5132799).

20. The kit of claim 16, wherein the AR signaling inhibitor comprises abiraterone, apalutamide (ARN-509), AZD3514, chlormadinone acetate, darolutamide (ODM-201), EPI-001 , enzalutamide, finasteride, galeterone, izonsteride, ketoconazole, megestrol, steroidal anti-androgens, nadolol, N-butylbenzene-sulfonamide, RU58841 , Testolone (RAD140), triptophenolide, tulobuterol hydrochloride, and/or turosteride.

21. The kit of claim 11 , wherein the AR signaling inhibitor comprises abiraterone and/or enzalutamide.

22. A method of predicting an in vivo therapeutic efficacy against prostate cancer (PCa) of a therapy comprising an SC-1 compound, a PP121 compound, a PD166285 dihydrochloride (diHCI) compound, an NCGC00348110 compound, a Dasatinib compound, a PDK1/Akt/Fit DPI compound, a Staurosporine compound, and/or a Cdk 1/2 Inhibitor III compound, the method comprising exposing cells of the PCa to the therapy; and determining expression levels of phosphoproteins selected from p-EGFRY1173, p-S6S235/236, p-FGFRY653/654, p-METY1349, p-IGF1 RY1131, p-AKTS473 and/or p-BADs112 within the cells of the PCa, wherein an observed decrease in one or more of the selected phosphoproteins in relation to a reference level predicts a positive in vivo therapeutic efficacy of the therapy against PCa.

23. The method of claim 22, wherein the kinase inhibitor comprises an SC-1 compound or a PP121 compound.

24. The method of claim 23, wherein the SC-1 compound comprises (Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4)ccc2C)C3)n(C)n1);

Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N6CCOCC6)CC5)c(C(F)(F)F)c 4)ccc2C)C3)n(C)n1 ;

C=CC(=O)N1CCCN(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4)ccc2C)C3)n1 C;

76 CCc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N(C)C)C5)c(C(F)(F)F)c4)ccc2 C)C3)n(C)n1;

Cc1cc(N(C)c2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N(C)C)C5)c(C(F)(F)F)c4)cc c2C)C3)n(C)n1 ;

Cc1ccc(NC(=O)c2cccc(C(F)(F)F)c2)cc1Nc1cc(CN(C)CCO)nc(NCCCO)c1;

N-{3-[7-(3-Dimethylamino-phenylamino)-1-methyl-2-oxo-1,4-dihydro-2H-pyrimido[4,5- d]pyrimidin-3-yl]-4-methyl-phenyl}-3-(4-methyl-imidazol-1-yl)-5-trifluoromethyl-benzamide; N-{3-[7-(3-Amino-phenylamino)-1-methyl-2-oxo-1 ,4-dihydro-2H-pyrimido[4,5-d]pyrimidin-3-yl]-4- methyl-phenyl}-3-trifluoromethyl-benzamide; or

N-[4-Methyl-3-(1-methyl-7-methylamino-2,4-dioxo-1 ,4-dihydro-2H-pyrimido[4.5-d]pyrimidin-3-yl)- phenyl]-3-trifluoromethyl-benzamide.

25. The method of claim 23, wherein the PP121 compound comprises

(Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2C1 CCCC1 );

Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2CCO;

CC1 COCC1 (C)n1 nc(-c2cnc3[nH]ccc3c2)c2c(N)ncnc21 ;

CCS(=O)c1 ccc(-n2nc(-c3cnc4[nH]ccc4c3)c3c(N)ncnc32)cc1 ;

Nc1ncnc2c1c(-c1 ccc(Oc3ccccc3) cc1)nn2C1CCNCC1;

CN(CC(=O)c1cnc2[nH]ccc2c1)C1CCCC1;

Cn 1 c(=O)c(c2cnc3[n H]ccc3c2)cn 1 CC(C) (C)O;

N-[4-[1-(1,4-dioxaspiro[4.5]dec-8-yl)-4-(8-oxa-3-azabicyclo[3.2.1]oct-3-yl)-1 H-pyrazolo[3,4- d]pyrimidin-6-yl]phenyl]-N'-methyl- (WYE-125132; methyl 4-(4-morpholino-1-(1-(pyridin-3-ylmethyl)piperidin-4-yl)-1H-pyrazolo[3,4-d]pyrimidin-6- yl)phenylcarbamate (WYE-687);

6-(1H-indol-5-yl)-4-morpholino-1-(1-(pyridin-3-ylmethyl)piperidin-4-yl)-1 H-pyrazolo[3,4- d]pyrimidine (WAY-600);

3-(2,4-bis((S)-3-methylmorpholino)pyrido[2,3-d]pyrimidin-7-yl)-N-methylbenzamide (AZD2014); or

5-(7-(methylsulfonyl)-2-morpholino-6,7-dihydro-5H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrimidin-2- amine (CH5132799).

26. The method of claim 22, wherein the therapy further comprises an androgen receptor (AR) signaling inhibitor.

27. The method of claim 22, further comprising exposing cells of a normal cell line to the therapy and determining the expression levels of phosphoproteins selected from p-EGFRY1173, p- S6S235/236, p_FGFF>Y653/654, p-M ETY1349, p-IGF1 RY1131 , p-AKTS473 and/or p-BADS112 to provide the

77 reference level.

28. The method of claim 22, wherein the cells of the PCa comprise a PCa cancer cell line.

29. The method of claim 28, wherein the PCa cancer cell line comprises a hormone naive cell line and/or a castration resistant cell line.

30. The method of claim 29, wherein hormone naive cell line comprises RWPE-2, LNCaP (e.g., LNCaP42D), LAPC-4, LAPC-9, VCaP, MDA PCa 2a/2b and/or LuCaP (e.g., LuCaP35CR).

31. The method of claim 29, wherein the castration resistant cell line comprises C4-2, C4-2B, 22Rv1 , ARCaP, PC3, and/or DU-145.

32. The method of claim 27, wherein the normal cell line is selected from pRNS-1-1 , RWPE-1 , BPH 1 , or PIN.

33. The method of claim 22, wherein the PCa is castration-resistant PCa (CRPC).

34. The method of claim 22, wherein the determining utilizes a reverse phase protein array (RPPA).

35. A method of predicting an in vivo therapeutic efficacy against prostate cancer (PCa) of a potential PCa therapeutic comprising obtaining at least two biological PCa samples for analysis wherein at least one biological PCa sample has been exposed to the potential PCa therapeutic and at least one biological PCa sample has not been exposed to the potential PCa therapeutic; and determining a protein concentration of p-EGFRY1173, p-S6S235/236, p-FGFRY653/654, p- METYi349, p-IGF1 RY1131, p-AKTS473 and/or p-BADS112 in the biological PCa samples; wherein a decrease in the concentration of p-EGFRY1173, p-S6S235/236, p-FGFRY653/654, p-METY1349, p-IGF1 RY1131, p-AKTS473 and/or p-BADS112 in the biological PCa sample exposed to the potential PCa therapeutic in comparison to the concentration of the same proteins in the biological PCa sample not exposed to the potential PCa therapeutic predicts the in vivo therapeutic efficacy of the potential therapeutic against PCa.

36. The method of claim 35, wherein the biological PCa samples are tissue samples.

37. The method of claim 36, wherein the tissue samples comprise cells, biopsy samples, laser capture micro-dissected cells, and/or biological fluids.

38. The method of claim 36, wherein the tissue samples are fresh, frozen, or fixed.

39. The method of claim 36, further comprising washing the tissue samples in phosphate buffered saline (PBS).

40. The method of claim 39, further comprising homogenizing the washed tissue samples in a Reverse Phase Protein Array (RPPA) lysis buffer to form tissue sample lysates.

41. The method of claim 40, further comprising serially diluting the tissue sample lysates and printing the serially diluted tissue sample lysates onto a solid phase.

42. The method of claim 41 , wherein the solid phase comprises a nitrocellulose-coated slide.

43. The method of claim 41 , further comprising probing the solid phase with a labeled primary antibody alone or a primary antibody followed by an appropriately labeled secondary antibody.

44. The method of claim 43, further comprising scanning the solid phase and quantifying resulting signals.

45. The method of claim 44, wherein the resulting signals are fluorescent, chemiluminescent, or colorimetric.

46. The method of claim 35, further comprising confirming the predicted in vivo efficacy of a tested therapeutic in an animal model of PCa.

47. The method of claim 35, wherein the therapeutic is a combination therapy.

48. The method of claim 47, wherein the combination therapy comprises a kinase inhibitor and an AR signaling inhibitor.

49. A compound or a salt thereof having the structure of Formula I or Formula II, Formula II wherein:

Ri is H, with the proviso that the compound is not pluripotin because R2 is not

50. The compound of claim 49 or a salt thereof, wherein the compound has the structure

51. The compound of claim 49 or a salt thereof, wherein the compound has the structure

52. A compound or a salt thereof having the structure of Formula III or Formula IV,

wherein R2 is a substituted alkane, a substituted tetrahydrofuran, or a substituted benzene.

53. The compound of claim 52, wherein R2 is

82

und of claim 52 or a salt thereof having the structure

Description:
KINASE INHIBITORS

FOR THE TREATMENT OF PROSTATE CANCER

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims priority to U.S. Provisional Patent Application No. 63/242,888 filed September 10, 2021, which is incorporated herein by reference in its entirety as if fully set forth herein.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT

[0002] This invention was made with government support under grants CA097186, CA201229, CA015704, and CA209923, awarded by the National Institutes of Health. The government has certain rights in the invention.

FIELD OF THE DISCLOSURE

[0003] The current disclosure provides treatments for prostate cancer, including castrationresistant prostate cancer. The treatment utilizes a protein kinase inhibitor selected from an SC-1 compound, a PP121 compound, a PD166285 dihydrochloride (diHCI) compound, an NCGC00348110 compound, a Dasatinib compound, a PDK1/Akt/Fit dual pathway inhibitor (DPI) compound, a Staurosporine compound, and/or a Cdk 1/2 Inhibitor III compound optionally in combination with an androgen receptor signaling inhibitor. Assays to predict in vivo effectiveness of potential therapeutics against prostate cancer are also provided.

BACKGROUND OF THE DISCLOSURE

[0004] Prostate cancer (PCa) is the second leading cause of cancer-related death in men in the United States, and its prevalence is increasing in developing countries. In 2020 there were more than 1.4 million cases of men diagnosed with PCa and 375,000 deaths caused by PCa. Risk factors associated with PCa include: age, race/ethnicity, geography, family history, gene mutations, diet, obesity, smoking, chemical exposure, inflammation of the prostate, sexually transmitted infections, and having had a vasectomy.

[0005] The most common type of PCa is typically classified as an adenocarcinoma which is a type of cancer that forms in mucus-secreting glands of organs. PCa originates in the prostate gland (a gland that produces seminal fluid) of the male reproductive system. Localized PCa can be successfully treated, with nearly 100% survival at 5 years from diagnosis. Some methods of treatment for PCa involve surgery, radiation, cryotherapy or hormone therapy.

[0006] Hormone therapy can provide an effective therapy against PCa because its growth is often driven by male sex hormones called androgens, which include testosterone. In these hormone therapies, androgen levels in the man’s body are reduced. Unfortunately, some patients fail hormone therapy or the treatment loses its efficacy over time. When hormone therapies are not effective to treat PCa, the PCa is referred to as a “castration-resistant” prostate cancer or CRPC. Unfortunately, when PCa becomes CRPC, the 5-year survival rate drops to 30%.

[0007] Protein kinases play a key role in nearly every biological process of both normal and altered cells, including cancer cells. These enzymes have been of interest as potential therapeutic targets since the first proto-oncogene, called sarcoma (Src), was identified as a protein kinase. Since then, 37 kinase inhibitors (KIs) have been approved by the U.S. Food and Drug Administration (FDA), most for oncology-related therapies.

SUMMARY OF THE DISCLOSURE

[0008] The current disclosure provides use of particular protein kinase inhibitors, optionally in combination with an androgen hormone therapy for treatment of prostate cancer (PCa), including castration-resistant prostate cancer (CRPC). In particular embodiments the protein kinase inhibitor includes an SC-1 compound, a PP121 compound, a PD166285 dihydrochloride (diHCI) compound, an NCGC00348110 compound, a Dasatinib compound, a PDK1/Akt/Fit dual pathway inhibitor (DPI) compound, a Staurosporine compound, and/or a Cdk 1/2 Inhibitor III compound. In particular embodiments, the androgen hormone therapy includes an androgen receptor signaling inhibitor.

[0009] The current disclosure also provides assays to assess the in vivo therapeutic efficacy of particular protein kinase inhibitor therapies using an assay that determines levels of phosphoproteins, such as p-EGFR Y1173 , p-S6 S235/236 , p-FGFR Y653/654 , p-MET Y1349 , p-IGF1 R Y1131 , p- AKT S473 and/or the anti-apoptotic protein, p-BAD S112 . An observed decrease in one or more of these assayed proteins correlates with the positive in vivo therapeutic efficacy of a protein kinase inhibitor therapy against PCa.

BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS

[00010] Some of the drawings submitted herein may be better understood in color. Applicant considers the color versions of the drawings as part of the original submission and reserves the right to present color images of the drawings in later proceedings.

[0011] FIGs. 1A-1 D. Constructing predictive Kinome Regularization (KiR) models from an in vitro kinase inhibitor screen. Increased concentrations of kinase inhibitors, Bosutinib and Gefitinib, showed inhibited growth, or reduced confluence, of PC3 cells. (1A) Schematic of KiR workflow. Previous biochemical profiling of kinase inhibitors resulted in a quantitative drug-target matrix. From this, an optimal set of 32 kinase inhibitors (KIs) are used for a small-scale drug screen. The results enable the generation of a regularized, cross-validated model that predicts the responses to over 400 KIs. (1 B) Representative images of PC3 cells after 52 hours of treatment with Bosutinib at 3.33pM (top) or 0.01 pM (bottom). Cell confluence mask was drawn and quantified via IncuCyte ZOOM® (Essen Instruments, Gottingen, Germany). Top image was quantified at 45.6% confluence, bottom image at 72.6% confluence. (1C) Full 72-hour growth curves of PC3 cells treated with various doses of Bosutinib. End point responses were taken when untreated control cells reached 75% confluence. Mean ± SEM, n = 3 wells/dose. (1 D) Representative doseresponse curves of PC3 treated with Bosutinib or Gefitinib. Curves are fitted with a 3-parameter logistic equation, and responses were interpolated at 500nM. Mean ± standard error of the mean (SEM), n = 3 wells/dose.

[0012] FIGs. 2A-2C. Constructing predictive KiR models from an in vitro kinase inhibitor screen. (2A) Heatmap of the residual kinase activity (as % untreated control) of the 32 inhibitors initially tested in PC3. (2B) Interpolated responses of PC3 cells to the inhibitors in (2A). (2C) Cross- validation regularization plot, showing the log of the elastic net regularization penalty (A) on the x- axis and the average of the leave-one-out cross-validation variance (LOOCV) mean-squared error on the y-axis.

[0013] FIG. 3. Properties of KiR models for castration-resistant prostate cancer (CRPC) cell lines. Cross validation is performed as LOOCV. Normalized root mean square error (RMSE)s are calculated by dividing the RMSE by the average of the training set responses, which are scaled to have a pseudo-maximum of 1 .00 for untreated samples. Alpha is user defined when generating the model.

[0014] FIGs. 4A-4E. (4A) Result of the KiR model, showing the predicted responses of PC3 cells to more than 400 kinase inhibitors. (4B-4E) Validation and testing of the KiR models. KiR model plots for four CRPC cell lines showing the response of each cell line to 427 KIs (different doses of the same compound are profiled separately and thus treated as separate compounds for this purpose). The circles (•) represent KiR model predictions, squares (■) represent experimentally measured values used to train the model, and diamonds (♦) represent experimentally measured values used for validation and not included in the final model construction. Dotted line indicates the threshold (<70% of control) for an inhibitor to be considered effective.

[0015] FIGs. 5A, 5B. Eight KIs showed effective reduction of cell confluence in all four cell lines. (5A) Euler diagram of the effective inhibitors in all four cell lines. Eight inhibitors reduced the change in confluence to <70% of untreated control in all four cell lines. (5B) Pearson correlation heatmap of the inhibition profiles of the eight effective inhibitors from panel (5A). [0016] FIGs. 6A-6D. PP121 and SC-1 effectively target CRPC cells in vitro. (6A, 6B) Increased dose of PP121 or SC-1 resulted in decreased confluence in all five cell lines. Cell line dose responses to two of the most effective inhibitors, (6A) PP121 and (6B) SC-1. Mean ± SEM, n = 3 wells/dose. (6C) Biochemically assayed inhibition profiles of PP121 and SC-1 towards receptors EGFR and FGFR, and downstream signaling molecules Src family kinases, Akt, and S6 kinase. (6D) Heatmaps of RPPA phosphorylation signals when PC3 and C4-2B cells are treated with PP121 or SC-1 in vitro. Signal is normalized to p-actin and presented relative to dimethyl sulfoxide (DMSO) control. *p<0.05, **p<0.01 , ***p<0.001 , ****p<0.0001 , two-way ANOVA with Holm-Sidak multiple comparisons test.

[0017] FIGs. 7A-7N. Reverse-phase protein array (RPPA) data from in vitro treatment with PP121 and SC-1. (7A) Illustration of the reverse-phase protein array assay. (7B) Representative RPPA slide image of C4-2B and PC3 samples treated for 1 hour with PP121 or SC-1 and probed with a phospho-Src family primary antibody. (7C) Growth signaling pathway diagram showing the relationships between the proteins whose phosphorylation status was assayed via RPPA. Interactions were based on those in KEGG® (Minoru Kanehisa, Kyoto-shi, Japan). (7D, 7E) Changes in phosphorylation status observed via RPPA when cells were treated with either PP121 (7D) or SC-1 (7E). (7F-7N) Heatmaps showing the changes in RPPA signal from various phosphorylation targets when PC3 and C4-2B cells are treated with PP121 or SC-1. Signal is normalized to p-actin and presented relative to untreated control. PP121 and SC-1 result in altered phosphorylation of key targets in the growth signaling pathway. These KIs resulted in a decrease in phosphorylation of FGFR, EGFR, Met, lnsR/IGF1 R, FAK, LYN, Src/SFK and an increase in phosphorylation of MEK1/2. PP121 also elicits a decrease in phosphorylation of Akt and SC-1 results in an increase in phosphorylation of S6.

[0018] FIGs. 8A-8F. PP121 and SC-1 significantly reduced tumor growth in vivo. (8A) Schematic representation of the experiment: mice were implanted with prostate cancer cells (PCa) subcutaneously; after 10 days, mice were treated with KI and the response was assessed longitudinally using a caliper (PC3) or monitoring the bioluminescent signal. (8B) Timeline of in vivo kinase inhibitors treatments. At day 10 after tumor implantation (5 x 10 6 PC3 cells/mouse and 6 x 10 6 C4-2B cells/mouse) mice were randomized and treated with 150 mg/kg PP121 or 80 mg/kg SC-1. (8C) Kinase inhibitor treatment response of PC3 and C4-2B subcutaneous tumors. Tumor growth is shown, mean + SD, n = 4-8 tumors per group. (8D) Pictures of representative tumors are shown. (8E) Tumor and (8F) mouse weight for both KI treatments were quantified at the endpoint. *p<0.05, **p<0.01 , ***p<0.001 , one- way ANOVA followed by Tukey’s HSD post- hoc test. [0019] FIGs. 9A-9D. Validation of PP121 and SC-1 efficacy in LNCaP42D and LuCaP35CR cell lines. (9A, 9B) Time course growth of LNCaP42D under treatment with (9A) PP121 or (9B) SC-1. Confluence was assessed as the percentage of the total image area occupied by cells. Dose response curves for (9C) LNCaP42D and (9D) LuCaP35CR under treatment with either PP121 or SC-1. Viability was determined using a luciferase-based assay.

[0020] FIGs. 10A-10C. PP121 treatment decreases PDX tumor slice viability. (10A) Scheme of the workflow of PDX tissue slice culture. Tumor cores were punched with a 6 mm biopsy punch and sliced into 250 pm sections using an automated vibratome. The sliced tissues were cultured on an insert prior to tissue viability measurement using a bioluminescence reagent and smallmolecule inhibitor treatment. The treated tissue slices were imaged using I VIS spectrum or plate reader to measure their response to treatment by acquiring the intensity of bioluminescence. (10B) Viability of LuCaP 147CR tissues treated with staurosporine (0.5 uM), or PP121 (0.5uM). Bars, mean of two independent slices; error bars, SEM. (10C) Viability of MDA-PCa-118b tissues treated with staurosporine (0.5 uM) and PP121 (0.5uM).

[0021] FIGs. 11 A, 11 B. (11 A) Plot showing relative changes in the expression of AR-targeted gene signatures in response to treatment with PP121 or SC-1 in C42B cells. Treatment with Enzalutamide was used as a positive control while DMSO was used a negative control. * denote p<0.05 and fold change >2-fold compared with DMSO control. (11 B) A heatmap showing changes in indicated phospho-proteins in drug treated PC3 tumors measured by RPPA.

DETAILED DESCRIPTION

[0022] Prostate cancer (PCa) is the second leading cause of cancer-related death in men in the United States, and its prevalence is increasing in developing countries. In 2020, there were more than 1.4 million cases of men diagnosed with PCa and 375,000 deaths caused by PCa. Risk factors associated with PCa include: age, race/ethnicity, geography, family history, gene mutations, diet, obesity, smoking, chemical exposure, inflammation of the prostate, sexually transmitted infections, and having had a vasectomy.

[0023] The most common type of PCa is typically classified as an adenocarcinoma which is a type of cancer that forms in mucus-secreting glands of organs. PCa originates in the prostate gland (a gland that produces seminal fluid) of the male reproductive system. Localized prostate cancer (PCa) can be successfully treated, with nearly 100% survival at 5 years from diagnosis. Some methods of treatment for PCa involve surgery, radiation, cryotherapy or hormone therapy. [0024] Hormone therapy can provide an effective therapy against PCa because its growth is often driven by male sex hormones called androgens, which include testosterone. In these hormone therapies, androgen levels in the man’s body are reduced. Androgen levels can be lowered by surgically removing the testicles, by administering drugs that prevent the production of androgens, and/or by blocking the ability of androgens to have an effect in the body. Unfortunately, most hormone dependent cancers become refractory to these types of treatments after one to three years and resume growth despite hormone therapy. When hormone therapies lose their efficacy to treat PCa, the PCa is referred to as a “castration-resistant” prostate cancer or CRPC. Unfortunately, when PCa becomes CRPC, the 5-year survival rate drops to 30%.

[0025] Progression towards late stage PCa may be marked by amplification of the androgen receptor (AR), expression of AR splice variants, stromal-mediated survival, and upregulation of compensatory kinase-mediated signaling pathways (Wise et al., Clin. Sci. 131 , 197-210 (2017); Nevedomskaya et al., International Journal of Molecular Sciences 19, 1359 (2018); Park et al., Biochimica et Biophysica Acta - Reviews on Cancer 1870, 198-206 (2018); Bluemn et al., Cancer Cell 32, 474-489. e6 (2017); Crumbaker et al., Cancers 9, (2017); Ciccarese et al., Cancer Treat. Rev. 43, 27-35 (2016); Chandrasekar et al., Transl. Androl. Urol. 4, 365-80 (2015); Drake et al, Proc. Natl. Acad. Sci. 110, E4762-E4769 (2013); Gao et al., Proc. Natl. Acad. Sci. U. S. A. 103, 14477-82 (2006); Lara et al., Lancet Oncol. 14, 1248-1249 (2013)).

[0026] Protein kinases play a key role in nearly every biological process of both normal and altered cells, including cancer cells. (Ardito et al., Int. J. Mol. Med. 40, 271-280 (2017); Fleuren et al., Nature Reviews Cancer 16, 83-98 (2016); Klaeger et al., Science 358, eaan4368 (2017); Wu et al., Trends Pharmacol. Sci. 36, 422-439 (2015); Knight et al., Nature Reviews Cancer 10, 130-137 (2010)). These enzymes have been of interest as potential therapeutic targets since the first proto-oncogene, SRC, was identified as a protein kinase (Fleuren et al., Nature Reviews Cancer 16, 83-98 (2016)). Since then, 37 kinase inhibitors (KIs) have been approved by the U.S. Food and Drug Administration (FDA), most for oncology-related therapies (Knapp et al., Br. J. Cancer 118, 936-937 (2018)). The ability to specifically target aberrant or misregulated cellular signaling via kinase inhibition has proven to be a very effective cancer therapy since Imatinib (Gleevec®, Novartis, Basel, Switzerland) was applied to target the mutant fusion kinase BCR- ABL in chronic myeloid leukemia (Wu et al., Trends Pharmacol. Sci. 36, 422-439 (2015)). Despite being labeled “targeted” or “selective”, multiple studies have demonstrated that most KIs, even ones used clinically, display some level of promiscuity in their targets (Klaeger et al., Science 358, eaan4368 (2017); Gao et al., Biochem. J. 451 , 313-328 (2013); Anastassiadis et al., Nat. Biotechnol. 29, 1039-1045 (2011)). While this can increase the potential for toxicity and side effects, this polypharmacology may also reduce the chance of de novo resistance by simultaneous inhibition of multiple parallel signaling pathways that could otherwise act as compensatory growth signals, as seen in the multitude of trials combining multiple KIs to target the same or parallel pathways (Knight et al., Nature Reviews Cancer 10, 130-137 (2010)). It also allows for the repurposing of already-approved molecules in different disease contexts where genetic drivers may differ, but the key signaling nodes remain the same (Knapp et al., Br. J. Cancer 118, 936-937 (2018); Rao et al., J. Biol. Chem. jbc.RA118.006805 (2019) doi:10.1074/jbc.RA118.006805; Ma et al., European Journal of Medicinal Chemistry 143, 449- 463 (2018); Zhou et al., American Journal of Clinical Dermatology 19, 181-193 (2018)). Finally, the recent approval of Midostaurin (Rydapt®, Novartis, Basel, Switzerland), a broad-acting derivative of Staurosporine, for treatment of FLT3-mutated acute myeloid leukemia demonstrates that selectivity is not a prerequisite for clinical utility (Levis et al., Blood 129, 3403-3406 (2017)). [0027] An unbiased approach, called Kinome Regularization (KiR), was previously established that exploits the polypharmacology of KIs to identify the kinases whose activity is most likely contributing to a measured phenotype (Gujral et al., Proc. Natl. Acad. Sci. 111 , 5048-5053 (2014)). This approach has been applied to study phenotypes ranging from cell migration to malarial liver stage infection (Gujral et al., Proc. Natl. Acad. Sci. 111 , 5048-5053 (2014); Arang et al., Nat. Commun. 8, 1232 (2017)). By combining biochemical profiling with a small-scale functional screen, computational models are generated that can predict the response of a system to over 400 KIs. Within the current disclosure, kinome regularization (KiR) was used to predict KIs that target prostate cancer, including CRPC.

[0028] The current disclosure particularly provides methods of treating PCa using a kinase inhibitor selected from an SC-1 compound, a PP121 compound, a PD166285 dihydrochloride (diHCI) compound, an NCGC00348110 compound, a Dasatinib compound, a PDK1/Akt/Fit dual pathway inhibitor (DPI) compound, a Staurosporine compound, and/or a Cdk 1/2 Inhibitor III compound. In particular embodiments, the kinase inhibitor can be part of a combination therapy with an androgen receptor signaling inhibitor. In particular embodiments, the selected kinase inhibitor abrogates canonical kinase-mediated growth factor signaling pathways in vitro, as well as reduces PCa spheroid growth. In particular embodiments, the selected kinase inhibitor includes SC-1 compounds and/or PP121 compounds.

[0029] In particular embodiments, an SC-1 compound includes: (Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4) ccc2C)C3)n(C)n1); Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N6CCOCC 6)CC5)c(C(F)(F)F)c 4)ccc2C)C3)n(C)n1 ;

C=CC(=O)N1CCCN(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C (F)(F)F)c4)ccc2C)C3)n1 C; CCc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N(C)C) C5)c(C(F)(F)F)c4)ccc2 C)C3)n(C)n1;

Cc1cc(N(C)c2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N (C)C)C5)c(C(F)(F)F)c4)cc c2C)C3)n(C)n1 ;

Cc1ccc(NC(=O)c2cccc(C(F)(F)F)c2)cc1Nc1cc(CN(C)CCO)nc(NCCC O)c1;

N-{3-[7-(3-Dimethylamino-phenylamino)-1-methyl-2-oxo-1,4- dihydro-2H-pyrimido[4,5- d]pyrimidin-3-yl]-4-methyl-phenyl}-3-(4-methyl-imidazol-1-yl )-5-trifluoromethyl-benzamide; N-{3-[7-(3-Amino-phenylamino)-1-methyl-2-oxo-1 ,4-dihydro-2H-pyrimido[4,5-d]pyrimidin-3-yl]-4- methyl-phenyl}-3-trifluoromethyl-benzamide; and N-[4-Methyl-3-(1-methyl-7-methylamino-2,4-dioxo-1 ,4-dihydro-2H-pyrimido[4.5-d]pyrimidin-3-yl)- phenyl]-3-trifluoromethyl-benzamide.

[0030] In particular embodiments, a PP121 compound includes:

(Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2C1 CCCC1 );

Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2CCO;

CC1 COCC1 (C)n1 nc(-c2cnc3[nH]ccc3c2)c2c(N)ncnc21 ;

CCS(=O)c1 ccc(-n2nc(-c3cnc4[nH]ccc4c3)c3c(N)ncnc32)cc1 ;

Nc1ncnc2c1c(-c1 ccc(Oc3ccccc3) cc1)nn2C1CCNCC1;

CN(CC(=O)c1cnc2[nH]ccc2c1)C1CCCC1;

Cn 1 c(=O)c(c2cnc3[n H]ccc3c2)cn 1 CC(C) (C)O;

N-[4-[1-(1,4-dioxaspiro[4.5]dec-8-yl)-4-(8-oxa-3-azabicyc lo[3.2.1]oct-3-yl)-1 H-pyrazolo[3,4- d]pyrimidin-6-yl]phenyl]-N'-methyl-(WYE-125132; methyl 4-(4-morpholino-1-(1-(pyridin-3-ylmethyl)piperidin-4-yl)-1H- pyrazolo[3,4-d]pyrimidin-6- yl)phenylcarbamate (WYE-687);

6-(1H-indol-5-yl)-4-morpholino-1-(1-(pyridin-3-ylmethyl)p iperidin-4-yl)-1 H-pyrazolo[3,4- d]pyrimidine (WAY-600);

3-(2,4-bis((S)-3-methylmorpholino)pyrido[2,3-d]pyrimidin- 7-yl)-N-methylbenzamide (AZD2014); and

5-(7-(methylsulfonyl)-2-morpholino-6,7-dihydro-5H-pyrrolo [2,3-d]pyrimidin-4-yl)pyrimidin-2- amine (CH5132799).

[0031] In particular embodiments the androgen receptor signaling inhibitor includes a dose of abiraterone, apalutamide (ARN-509), AZD3514, chlormadinone acetate, darolutamide (ODM- 201), EPI-001, enzalutamide, finasteride, galeterone, izonsteride, ketoconazole, megestrol, steroidal anti-androgens, nadolol, N-butylbenzene-sulfonamide, RU58841 , Testolone (RAD140), triptophenolide, tulobuterol hydrochloride, and/or turosteride that inhibits androgen receptor signaling.

[0032] The treated PCa can include CRPC and the treatment can delay or prevent metastasis.

[0033] The current disclosure also provides that monitoring the levels of phosphoproteins following exposure to a potential therapeutic compound can predict the in vivo efficacy of the compound against PCa. In particular embodiments, an observed decrease in one or more of p- EGFR Y1173 , P -S6 s235/236 , p-FGFR Y653/654 , p-MET Y1349 , p-IGF1 R Y1131 , p-AKT S473 and/or the anti- apoptotic protein, p-BAD S112 indicates that the compound will provide an effective anti-cancer compound in vivo.

[0034] Aspects of the current disclosure are now described with additional detail and options as follows: (i) Protein Kinase Inhibitors, (ii) Androgen Receptor Signaling Inhibitors, (iii) Compositions for Administration and Kits, (iv) Methods of Use, (v) Assays to Predict In Vivo Efficacy; (vi) Exemplary Embodiments, (vii) Experimental Examples, and (viii) Closing Paragraphs. While section headings are provided, these headings are for organizational purposes only and do not limit the scope or interpretation of the disclosure.

[0035] (i) Protein Kinase Inhibitors. Kinase inhibitors used within the current disclosure include an SC-1 compound, a PP121 compound, a PD166285 diHCI compound, an NCGC00348110 compound, a Dasatinib compound, a PDK1/Akt/Fit DPI compound, a Staurosporine compound, and/or a Cdk 1/2 Inhibitor III compound and/or functional derivates thereof.

[0036] In particular embodiments, the SC-1 compound includes SC-1. SC-1 (also known as Pluripotin) is a dual inhibitor of extracellular signal-regulated kinase 1 (ERK1 , mitogen-activated protein kinase (MAPK3)) and Ras GTPase activating protein (RasGAP). It has been found to have dissociation constant values for each of 98 nM and 212 nM, respectively. SC-1 has a molecular formula of C27H25F3N8O2, an IUPAC name of N-(3-(7-(1 ,3-dimethyl-1 H-pyrazol-5-ylamino)-1- methyl-2-oxo-1 ,2-dihydropyrimido[4,5-d]pyrimidin-3(4H)-yl)-4-methylphenyl) -3- (trifluoromethyl)benzamide, and a Simplified Molecular Input Line Entry System (SMILES) name of Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4)c cc2C)C3)n(C)n1. SC-1 has the following structure:

[0037] US7642255 discloses various derivatives of SC-1 having similar functional properties as

SC-1. Examples of such SC1-1 derivatives include: wherein: n is selected from 0, 1 , 2, 3 or 4;

Z is selected from N or CH;

R 1 is selected from hydrogen, -R 8 , -OR 8 , -S(0)o-2R 8 , -NR 7 R 8 and -NR 7 NR 7 R 8 ;

R 7 is independently selected from hydrogen and Ci-e alkyl;

R 8 is selected from hydrogen, Ci-e alkyl, C2-6 alkenyl, Ce-ioaryl-Co-4alkyl, Cs- heteroaryl-Co- 4 alkyl, C3-io cycloalkyl-Co-4 alkyl and Cs-io heterocycloalkyl-Co-4 alkyl; or

R 7 and R 8 together with the nitrogen atom to which R 7 and R 8 are attached form C3-10 heterocycloalkyl or C5-10 heteroaryl; wherein any alkyl or alkenyl of R 8 is optionally substituted by one to three radicals independently selected from halo, hydroxy, Ci-e alkyl and -NR 9 R 10 ; wherein any aryl, heteroaryl, cycloalkyl or heterocycloalkyl of R 8 , or the combination of R 7 and R 8 , is optionally substituted by one to three radicals selected from halo, hydroxy, cyano, Ci-e alkyl, Ci-e alkoxy, C2-6 alkenyl, halo-substituted-alkyl, halo-substituted-alkoxy, -XNR 9 R 10 , -XOXNR 9 R 10 , - XNR 9 S(0)O. 2 R 1 °, -XC(O)NR 9 R 10 , -XNR 9 C(O)XOR 9 , -XNR 9 C(O)NR 9 R 10 , -XNR 9 XNR 9 R 10 , - XC(O)NR 9 XNR 9 R 10 , -XNR 9 XOR 9 , -XOR 9 , -XNR 9 C(=NR 9 )NR 9 R 10 , -XS(0)o. 2 R 1 °, -XNR 9 C(O)R 9 , - XNR 9 C(O)XNR 9 R 10 , -XNR 9 C(O)R 11 , -XC(O)Rn, -XRn, -XC(O)OR and -XS(O) 0-2 NR 9 RI 0 ; wherein X is a bond or C1.4 alkylene; R 9 and R 10 are independently selected from hydrogen, Ci-e alkyl and Cs-i 2 cycloalkyl; and R 11 , is C3-10 heterocycloalkyl optionally substituted with 1 to 3 radicals selected from Ci- 6 alkyl, -XNR 9 R 10 , -XNR 9 XNR 9 R 9 , XNR 9 XOR 9 and -XOR 9 ; wherein X, R 9 and R 10 are as described above;

R 2 and R 3 are independently selected from hydrogen and Ci-e alkyl; or R 1 and R 2 together form =0 or =S;

R 4 is selected from hydrogen, hydroxy, amino, Ci-e alkyl, -XOR 9 , -XC(O)OR 9 , - XC(O)NR 9 R 10 , C3-10 cycloalkyl-Co-4 alkyl, C5-10 heteraryl-Co-4 alkyl, Ce- aryl-Co-4 alkyl and C3-10 heterocycloalkyl-Co-4 alkyl; wherein X, R 9 and R 10 are as described above;

R 5 is selected from Ci-ealkyl, C 2 .6 alkenyl, Ci-e alkoxy, halo-substituted-Ci-4 alkyl and halo- substituted-Ci-4 alkoxy;

R 6 is selected from -NRi 2 Y(O)Ris and -Y(O)NRI 2 RIS; wherein Y is selected from C, P(O), and S(O); R 12 is selected from hydrogen and Ci-e alkyl; and Ris is selected from Ce- aryl, C5-10 heteroaryl, C3-10 cycloalkyl, C3-10 heterocycloalkyl and Ci-e alkyl; wherein any aryl, heteroaryl, cycloalkyl or heterocycloalkyl of R 13 is optionally substituted by one to three radicals independently selected from halo, hydroxy, nitro, cyano, halo-substituted-Ci-6 alkyl, Ci-e alkyl, Ci-e alkoxy, halo- substituted-Ci.6 alkoxy, -XNR 9 R 9 , -XNR 9 XNR 9 R 9 , -XNR 9 C(O)R 9 , -XC(O)OR 9 , -XNR 9 S(O) 2 R 9 , - XNR 9 S(O)R 9 , -XNR 9 SR 9 and -XRI 4 ; wherein X and R 9 are as defined above and R14 is selected from C5-10 heteroaryl-Co-4 alkyl and C3-10 heterocycloalkyl-Co-4 alkyl; wherein any heteroaryl or heterocycloalkyl of Ri4 is optionally substituted with a radical selected from Ci-e alkyl, halo- substituted-Ci-e alkyl-NR 9 R 9 and C(O)OR 9 ; wherein R 9 is as described above.

[0038] In particular embodiments, the SC-1 compound includes a compound or salt thereof having the structure of Formula I or Formula II,

Formula II wherein:

Ri is H,

R2 is

[0039] In particular embodiments, the SC-1 compound includes:

Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N6CC OCC6)CC5)c(C(F)(F)F)c

4)ccc2C)C3)n(C)n1

C=CC(=O)N1CCCN(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C (F)(F)F)c4)ccc2C)C3)n1

C

Cc1cc(N(C)c2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N (C)C)C5)c(C(F)(F)F)c4)cc c2C)C3)n(C)n1

Cc1ccc(NC(=O)c2cccc(C(F)(F)F)c2)cc1Nc1cc(CN(C)CCO)nc(NCCC O)c1

[0040] In particular embodiments, the SC-1 compound includes the following compounds: N-{3-[7-(3-Dimethylamino-phenylamino)-1-methyl-2-oxo-1,4-dih ydro-2H-pyrimido[4,5- d]pyrimidin-3-yl]-4-methyl-phenyl}-3-(4-methyl-imidazol-1-yl )-5-trifluoromethyl-benzamide,

N-{3-[7-(3-Amino-phenylamino)-1-methyl-2-oxo-1 ,4-dihydro-2H-pyrimido[4,5-d]pyrimidin-

3-yl]-4-methyl-phenyl}-3-trifluoromethyl-benzamide,

; and

N-[4-Methyl-3-(1-methyl-7-methylamino-2,4-dioxo-1 ,4-dihydro-2H-pyrimido[4.5- d]pyrimidin-3-yl)-phenyl]-3-trifluoromethyl-benzamide,

[0041] In particular embodiments, the PP121 compound includes PP121. PP121 is a multitargeted inhibitor of platelet-derived growth factor receptor (PDGFR), hematopoietic cell kinase (Hck), mammalian target of rapamycin (mTOR), vascular endothelial growth factor receptor 2 (VEGFR2), sarcoma (Src), and Abl. PP121 has also been shown to inhibit DNA-PK with a half maximal inhibitory concentration value (IC50) of 60 nM (LIS20160009785; Apsel et al., (2008) Nat.Chem.Biol. 4 691 PMID: 18849971). PP121 has a molecular formula of C17H17N7, an International Union of Pure and Applied Chemistry (IUPAC) name of 1-cyclopentyl-3-(1 H- pyrrolo[2,3-b]pyridin-5-yl)-1 H-pyrazolo[3,4-d]pyrimidin-4-amine, and a Simplified Molecular Input Line Entry System (SMILES) name of Nc1 ncnc2c1c(-c1cnc3[nH]ccc3c1)nn2C1CCCC1. PP121 has the following structure:

[0042] US9604988 and US9604988 disclose various derivatives of PP121 having similar functional properties as PP121. Examples of derivatives of PP121 include: wherein:

An represents a substituted or unsubstituted arylene group;

Ar 2 represents a substituted or unsubstituted aryl or heteroaryl group; L represents an oxygen atom, a sulfur atom, -NH-, -NHCO-, or -CONH-;

Xi represents CH or a nitrogen atom;

X2 and X3 each independently represent CH or a nitrogen atom, provided that X2 and X3 are not the same;

Y represents a Ci-3-alkylene group;

W1, which is a substituent on the ring including Y and X 2 , each independently represents a Ci-6-alkyl group; m is an integer from 0 to 3; and

Z1 and Z2 each independently represent a hydrogen atom, a Ci-6-alkyl group, an amino-Ci. 6-alkyl group, a Ci-6-alkylamino-Ci-6-alkyl group, a di(Ci-6-alkyl)amino-Ci-6-alkyl group, a hydroxy- Ci-6-alkyl group, a Ci-6-alkoxy-Ci-6-alkyl group, a C2-7-aliphatic acyl group, a carboxy-Ci-6-alkyl group, a carbamoyl-Ci-6-alkyl group, a substituted or unsubstituted saturated heterocyclic group, a substituted or unsubstituted aryl-Ci-6-alkyl group, a substituted or unsubstituted heteroaryl-Ci-6- alkyl group, a substituted or unsubstituted arylcarbonyl group, or a substituted or unsubstituted heteroarylcarbonyl group, excluding the case in which both Z1 and Z2 represent hydrogen atoms; when X2 represents CH and X3 represents a nitrogen atom, Z1 and Z2 may form a substituted or unsubstituted, monocyclic or polycyclic heterocyclic group including s and containing as a ring member two or more hetero atoms selected from the group consisting of oxygen, sulfur, and nitrogen atoms, or when X2 represents a nitrogen atom and X3 represents CH, Zi and Z2 may form a substituted or unsubstituted, monocyclic or polycyclic heterocyclic group including Xs and containing a hetero atom selected from among oxygen, sulfur, and nitrogen atoms.

[0043] In particular embodiments, PP121 compounds include a compound or salt thereof having the structure of Formula III or Formula IV,

wherein R2 is a substituted alkane, a substituted tetrahydrofuran, or a substituted benzene. The term substituted, when used to describe a group is intended to describe any non-hydrogen moiety that formally replaces a hydrogen in that group. In particular embodiments, a substituted alkane includes a compound with the structure -CH2-CH2-OH. In particular embodiments, a compound

with the structure In particular embodiments, a substituted benzene includes a compound with the structure particular embodiments, Ri is

[0044] In particular embodiments, the PP121 compounds includes: Nc1ncnc2c1c(- c1cnc3[nH]ccc3c1)nn2CCO

CC1COCC1(C)n1nc(-c2cnc3[nH]ccc3c2)c2c(N)ncnc21

CCS(=O)c1ccc(-n2nc(-c3cnc4[nH]ccc4c3)c3c(N)ncnc32)cc1

Cn 1 c(=O)c(c2cnc3[n H]ccc3c2)cn 1 CC(C) (C)O

[0045] In particular embodiments, the PP121 compound includes the following compounds:

N-[4-[1-(1,4-dioxaspiro[4.5]dec-8-yl)-4-(8-oxa-3-azabicyc lo[3.2.1]oct-3-yl)-1 H- pyrazolo[3,4-d]pyrimidin-6-yl]phenyl]-N'-methyl- (WYE-125132), methyl 4-(4-morpholino-1-(1-(pyridin-3-ylmethyl)piperidin-4-yl)-1H- pyrazolo[3,4-d]pyrimidin-6- yl)phenylcarbamate (WYE-687),

6-(1H-indol-5-yl)-4-morpholino-1-(1-(pyridin-3-ylmethyl)p iperidin-4-yl)-1 H-pyrazolo[3,4- d]pyrimidine (WAY-600), 3-(2,4-bis((S)-3-methylmorpholino)pyrido[2,3-d]pyrimidin-7-y l)-N-methylbenzamide (AZD2014),

; and

5-(7-(methylsulfonyl)-2-morpholino-6,7-dihydro-5H-pyrrolo [2,3-d]pyrimidin-4-yl)pyrimidin-2- amine (CH5132799);

[0046] PD166285 diHCI inhibits tyrosine kinases c-Src, fibroblast growth factor receptor 1 (FGFR1), platelet-derived growth factor receptor (PDGFRP), and basic fibroblast growth factor (bFGF). (Gaffre et al., (2011) Toxicol Lett 138 3735 PMID: 21795279). PD166285 diHCI (also known as PD-166285 hydrate) has a molecular formula of C26H27CI2N5O2 2HCI and an IIIPAC Name of 6-(2,6-Dichlorophenyl)-2-[[4-[2-(diethylamino)ethoxy]phenyl] amino]-8-methyl- pyrido[2,3-d]pyrimidin-7(8H)-one dihydrochloride). PD166285 diHCI has the following structure:

[0047] US20190169569 discloses various derivatives of PD166285 diHCI having similar functional properties as PD166285 diHCI. Examples of derivatives include the following compounds:

N-[2-[[4-(Diethylamino)butyl]amino-6-(3,5-dimethoxyphenyl )pyrido[2,3-d]pyrimidin-7-yl]- N'-(1 ,1-dimethylethyl)urea (PD173074),

2-(2-Amino-3-methoxyphenyl)-4H-1-benzopyran-4-one (PD 98059),

1-tert-Butyl-3-[6-(2,6-dichlorophenyl)-2-[[4-(diethylamin o)butyl]amino]pyrido[2,3- d]pyrimidin-7-yl]urea (PD161570),

N-[2-Amino-6-(3,5-dimethoxyphenyl)pyrido[2,3-d]pyrimidin- 7-yl]-N'-(1 ,1 -dimethylethyl)- urea ( PD166866),

; and

MK-2206,

[0048] NCGC00348110 (also known as NCGC00348110-02; CHEMBL502156) has a molecular formula of C31H40FN7O2 and an IIIPAC name of 3-N-(2,6-Dimethylphenyl)-6-N-[3-fluoro-4-(2- pyrrolidin-1-ylethoxy)phenyl]-1-(3-methoxy-3-methylbutyl)pyr azolo[3,4-d]pyrimidine-3,6-diamine. NCGC00348110 has the following structure:

[0049] US7763624 discloses various derivatives of NCGC00348110 having similar functional properties as NCGC00348110. Examples of derivatives of NCGC00348110 include: wherein

R 1 is hydrogen or Ci-e alkyl optionally substituted with 1-3 substituents of halo, cyano, Ci- 6 alkyl, Ci-e haloalkyl or Ci-e alkoxy;

R 2 is Cs-w-cycloalkyl, phenyl, naphthyl, pyridyl, pyrimidinyl, pyridazinyl, triazinyl, piperidinyl, piperazinyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, pyrrolidinyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, quinazolinyl, isoquinazolinyl, tetrahydroquinazolinyl, tetrahydroisoquinazolinyl, phthalazinyl, morpholinyl, thiophenyl, furyl, dihydrofuryl, tetrahydrofuryl, thiazolyl, oxazolyl, isoxazolyl, isothiazolyl, indolyl, isoindolyl, indolinyl, benzofuranyl, dihydrobenzofuranyl, benzothiophenyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl or benzothiazolyl, each of which is optionally substituted independently with 1-3 substituents of R 7 , NR 8 R 9 , OR 10 , SR 11 , C(O)R 12 , COOR 13 , C(O)NR 8 R 9 , NR 14 C(O)R 15 , NR 14 C(O)NR 8 R 9 , OC(O)NR 8 R 9 , S(O) 2 R 16 , S(O) 2 NR 8 R 9 or NR 14 S(O) 2 R 16 ;

R 3 is optionally substituted alkyl, optionally substituted alkenyl or optionally substituted alkynyl, wherein the substituents are selected from R 17 , NR 8 R 9 , OR 10 , SR 11 , COOR 12 , C(O)R 13 , OC(O)R 13 , R 13 OR 10 , C(O)NR 8 R 9 , C(S)NR 8 R 9 , NR 14 C(O)R 15 , NR 14 C(S)R 15 , NR 14 C(O)NR 8 R 9 , NR 14 C(S)NR 8 R 9 , NR 14 (COOR 12 ), OC(O)NR 8 R 9 , S(O) 2 R 8 , S(O) 2 NR 8 R 9 , NR 14 S(O) 2 NR 8 R 9 , and NR 8 S(O) 2 R 9 ;

R 4 is hydrogen or Ci- 8 alkyl optionally substituted with 1-3 substituents of halo, cyano, Ci- 6 alkyl, Ci-e haloalkyl or Ci- 8 alkoxy;

R 5 is Cs-w-cycloalkyl, phenyl, naphthyl, pyridyl, pyrimidinyl, pyridazinyl, triazinyl, piperidinyl, piperazinyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, pyrrolidinyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, quinazolinyl, isoquinazolinyl, tetrahydroquinazolinyl, tetrahydroisoquinazolinyl, phthalazinyl, morpholinyl, thiophenyl, furyl, dihydrofuryl, tetrahydrofuryl, thiazolyl, oxazolyl, isoxazolyl, isothiazolyl, indolyl, isoindolyl, indolinyl, indazolyl, benzofuranyl, dihydrobenzofuranyl, benzothiophenyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl or benzothiazolyl, each of which is optionally substituted independently with 1-3 substituents of R 7 , NR 8 R 9 , OR 10 , R 10 OR 11 , SR 11 , C(O)R 12 , COOR 13 , C(O)NR 8 R 9 , NR 14 C(O)R 15 , NR 14 C(O)NR 8 R 9 , OC(O)NR 8 R 9 , S(O) 2 R 16 , S(O) 2 NR 8 R 9 and NR 17 S(O) 2 R 16 ;

R 6 is hydrogen, halogen or Ci. 8 alkyl optionally substituted with 1-3 substituents of halo, haloalkyl, CN, NO 2 , OH and NR 8 R 9 ;

R 7 is halo, haloalkyl, CN, NO 2 , Ci-10-alkyl, C 2 .io-alkenyl, C 2 .w-alkynyl, Cs-w-cycloalkyl or C4- w-cycloalkenyl, wherein each of the Ci-w-alkyl, C 2 -w-alkenyl, C 2 -w-alkynyl, Cs-w-cycloalkyl and C4- w-cycloalkenyl optionally consisting of 1-4 heteroatoms selected from N, O and S, or R 7 is a partially or fully saturated or unsaturated 5-8 membered monocyclic, 6-12 membered bicyclic, or 7-14 membered tricyclic ring system, the ring system formed of carbon atoms optionally including 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, and wherein each ring of the ring system is optionally substituted independently with one or more substituents of Ci- 8 -alkyl, C 2.8 -alkenyl, C 2.8 -alkynyl, NR 8 R 9 , OR 10 , SR 11 , C(O)R 12 , COOR 13 , C(O)NR 8 R 9 , NR 14 C(O)R 15 , NR 14 C(O)NR 8 R 9 , OC(O)NR 8 R 9 , S(O) 2 R 16 , S(O) 2 NR 8 R 9 or NR 14 S(O) 2 R 16 ; R 8 and R 9 are each independently H, Ci-s-alkyl, Cs-s-cycloalkyl, C2-8-alkenyl, C2-8-alkynyl, Ci-8-alkylamino-, Ci-8-dialkylamino-, Ci-s-alkoxyl, Ci-s-thioalkyl, Ci-8-alkoxy-Ci-8-alkyl, aryl, heteroaryl, or heterocyclyl;

R 10 , R 11 , and R 16 are each independently H, Ci-s-alkyl, C2-8-alkenyl, C2-8-alkynyl, C1.8- alkoxy-Ci-8-alkyl, Cs-s-cycloalkyl, aryl, heterocyclyl, Ci-8-alkyl-heterocyclyl or heterocyclyl-Ci-8- alkyl;

R 12 , R 13 , R 14 and R 15 are each independently H, Ci-s-alkyl, Cs-s-cycloalkyl, C2-8-alkenyl, C2- 8-alkynyl, Ci-8-alkylamino-, Ci-8-dialkylamino-, Ci-8-alkoxyl, Ci-s-thioalkyl, aryl, heteroaryl, heterocyclyl or alkyl-heterocyclyl; and

R 17 is halo, CN, NO2, or ring selected from Cs-w-cycloalkyl, phenyl, naphthyl, pyridyl, pyrimidinyl, pyridazinyl, triazinyl, piperidinyl, piperazinyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, pyrrolidinyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, quinazolinyl, isoquinazolinyl, tetrahydroquinazolinyl, tetrahydroisoquinazolinyl, phthalazinyl, morpholinyl, thiophenyl, furyl, dihydrofuryl, tetrahydrofuryl, thiazolyl, oxazolyl, isoxazolyl, isothiazolyl, indolyl, isoindolyl, indolinyl, benzofuranyl, dihydrobenzofuranyl, benzothiophenyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl and benzothiazolyl, each ring of which is optionally substituted independently with 1-3 substituents of halo, haloalkyl, CN, NO2, NH2, OH, Ci-s-alkyl, C2-8-alkenyl, C2-8-alkynyl, Ci-8-alkylamino-, Ci-8-alkoxyl or Ci-8-thioalkyl.

[0050] In particular embodiments, derivatives of NCGC00348110 include:

1-(but-3-enyl)-N 3 -(2,6-dimethylphenyl)-N 6 -phenyl-1 H-pyrazolo[3,4-d]pyrimidine-3,6-diamine; 4-(3-(2,6-dimethylphenylamino)-6-(phenylamino)-1 H-pyrazolo[3,4-d]pyrimidin-1-yl)butane- 1 ,2-diol;

1-(2-(1 ,3-dioxolan-4-yl)ethyl)-N3-(2,6-dimethylphenyl)-N6-phenyl-1 H-pyrazolo[3,4- d]pyrimidine-3,6-diamine;

3-(3-(2,6-dimethylphenylamino)-6-(phenylamino)-1 H-pyrazolo[3,4-d]pyrimidin-1-yl)propan-1- ol;

1-(3-(dimethylamino)propyl)-N3-(2,6-dimethylphenyl)-N6-ph enyl-1 H-pyrazolo[3,4- d]pyrimidine-3,6-diamine;

N 3 -(2,6-dimethylphenyl)-1-(3-(methylamino)propyl)-N 6 -phenyl-1 H-pyrazolo[3,4-d]pyrimidine-

3,6-diamine;

1-(3-(diethylamino)propyl)-N 3 -(2,6-dimethylphenyl)-N 6 -phenyl-1 H-pyrazolo[3,4-d]pyrimidine-

3,6-diamine;

N 3 -(2,6-dimethylphenyl)-N 6 -phenyl-1-(3-(pyrrolidin-1-yl)propyl)-1 H-pyrazolo[3,4-d]pyrimidine-

3,6-diamine; 1-(dimethylamino)-4-(3-(2,6-dimethylphenylamino)-6-(phenylam ino)-1 H-pyrazolo[3,4- d]pyrimidin-1-yl)butan-2-ol;

4-(3-(2,6-dimethylphenylamino)-6-(phenylamino)-1 H-pyrazolo[3,4-d]pyrimidin-1-yl)-1-

(pyrrolidin-1-yl)butan-2-ol;

4-(3-(2,6-dimethylphenylamino)-6-(phenylamino)-1 H-pyrazolo[3,4-d]pyrimidin-1-yl)butan-2- ol;

4-(3-(2,6-dimethylphenylamino)-6-(phenylamino)-1 H-pyrazolo[3,4-d]pyrimidin-1-yl)butan-2- ol;

N 3 -(2,6-dimethyl-phenyl)-N 6 -phenyl-1-(3-piperidin-1-yl-propyl)-1 H-pyrazolo[3,4-d]pyrimidine- 3,6-diamine;

N 3 -(2,6-dimethyl-phenyl)-N 6 -(4-piperazin-1-yl-phenyl)-1-(3-piperidin-1-yl-propyl) -1 H- pyrazolo[3,4-d]pyrimidine-3,6-diamine;

N 3 -(2,6-dimethyl-phenyl)-N 6 -[4-(4-methyl-piperazin-1-yl)-phenyl]-1-(3-piperidin-1 -yl-propyl)- 1 H-pyrazolo[3,4-d]pyrimidine-3,6-diamine;

N 3 -(2,6-dimethyl-phenyl)-1-(2-morpholin-4-yl-ethyl)-N 6 -phenyl-1 H-pyrazolo[3,4-d]pyrimidine- 3,6-diamine;

N 3 -(2,6-dimethyl-phenyl)-N 6 -[4-(2-methoxy-ethoxy)-phenyl]-1-(2-morpholin-4-yl-eth yl)-1 H- pyrazolo[3,4-d]pyrimidine-3,6-diamine;

N 3 -(2,6-dimethyl-phenyl)-N 6 -[4-(4-methyl-piperazin-1-yl)-phenyl]-1-(2-morpholin-4 -yl-ethyl)- 1 H-pyrazolo[3,4-d]pyrimidine-3,6-diamine;

N 3 -(2,6-dimethyl-phenyl)-N 6 -[4-(2-methoxy-ethoxy)-phenyl]-1-(3-methoxy-3-methyl-b utyl)-

1 H-pyrazolo[3,4-d]pyrimidine-3,6-diamine;

N 3 -(2,6-dimethyl-phenyl)-1-(3-methoxy-3-methyl-butyl)-N 6 -[4-(methylpiperazin-1-yl)phenyl]-

1 H-pyrazolo[3,4-d]pyrimidine-3,6-diamine;

N 3 -(2,6-dimethyl-phenyl)-1-(3-methoxy-3-methylbutyl)-N 6 -(4-piperazin-1-yl-phenyl)-1 H- pyrazolo[3,4-d]pyrimidine-3,6-diamine;

N 3 -(2,6-dimethylphenyl)-N 6 -[3-fluoro-4-(3-(piperidin-1-yl)propoxy)phenyl1-1-(3-m ethoxy-3- methylbutyl)-1 H-pyrazolo[3,4-d]pyrimidine-3,6-diamine;

N 3 -(2,6-dimethylphenyl)-1-(3-methoxy-3-methylbutyl)-1 H-indazole-3,6-diamine;

N 3 -(2,6-dimethylphenyl)-1-(3-methoxy-3-methylbutyl))-N 6 -phenyl-1 H-indazole-3,6-diamine;

N 3 -(2,6-dimethylphenyl)-1-N 6 -(4-(3,5-dimethylpiperazin-1-yl)phenyl)-1-(3-methoxy-3 - methylbutyl)-1 H-indazole-3,6-diamine;

N 3 -(2,6-dimethylphenyl)-1-N 6 -(4-piperazin-1-yl)phenyl)-1-((tetrahydro-2H-pyran-4-y l)methyl)- 1 H-indazole-3,6-diamine; N 3 -(2,6-dimethylphenyl)-1-N 6 -(3-fluoro-4-(piperidin-1-yl)propoxy)phenyl)-1-((tetra hydro-2H- pyran-4-yl)methyl)-1 H-indazole-3,6-diamine;

N 3 -(2,6-dimethylphenyl)-N 6 -phenyl-1-((tetrahydro-2H-pyran-4-yl)methyl)-1 H-indazole-3,6- diamine; ethyl-2-(3-(2,6-dimethylphenylamino)-6-(phenylamino)-1 H-indazole-1-yl)acetate;

(R) — N 3 -(2,6-dimethylphenyl)-N 6 -(4-fluorophenyl)-1-(3-(2-(methoxymethyl)pyrrolidin-1- yl)propyl)-1 H-indazole-3,6-diamine;

(R) — N 3 -(2,6-dimethylphenyl)-1-(3-(2-(methoxymethyl)pyrrolidi n-1-yl)propyl)-N 6 -phenyl-1 H- indazole-3,6-diamine;

(R) — N 3 -(2,6-dimethylphenyl)-1-(3-(2-(methoxymethyl)pyrrolidi n-1-yl)propyl)-N 6 -(4-piperazin- 1-yl)phenyl-1 H-indazole-3,6-diamine;

(R) — N 3 -(2,6-dimethylphenyl)-1-(3-(2-(methoxymethyl)pyrrolidi n-1-yl)propyl)-1 H-indazole- 3,6-diamine;

2-(3-(2,6-dimethylphenylamino)-6-(phenylamino)-1 H-indazol-1-yl)acetic acid;

2-(3-(2,6-dimethylphenylamino)-6-(phenylamino)-1 H-indazol-1-yl)-1-(4-methylpiperazin-1- yl)ethanone;

2-(3-(2,6-dimethylphenylamino)-6-(phenylamino)-1 H-indazol-1-yl)-1-(piperazin-1- yl)ethanone;

N 3 -(2,6-dimethylphenyl)-1-(2-(4-methylpiperazin-1-yl)eth yl)-N 6 -phenyl-1 H-indazole-3,6- diamine;

1-(3-methoxy-3-methyl-butyl)-N 3 , N 6 -diphenyl-1 H-pyrazolo[3,4-d]pyrimidine-3,6-diamine;

1-(3-methoxy-3-methyl-butyl)-N 6 -[4-(4-methyl-piperazin-1-yl)-phenyl-N 3 -phenyl-1 H- pyrazolo[3,4-d]pyrimidine-3,6-diamine;

1-(3-methoxy-3-methyl-butyl)-N 6 -phenyl-N 3 -(2,4,6-trimethyl-phenyl)-1 H-pyrazolo[3,4- d]pyrimidine-3,6-diamine;

N 3 -(4-fluoro-2,6-dimethyl-phenyl)-1-(3-methoxy-3-methyl- butyl)-N 6 -phenyl-1 H-pyrazolo[3,4- d]pyrimidine-3,6-diamine;

N 3 -(4-fluoro-2,6-dimethyl-phenyl)-1-(3-methoxy-3-methyl- butyl)-N 6 -[4-(4-methyl-piperazin-1- yl)-phenyl]-1 H-pyrazolo[3,4-d]pyrimidine-3,6-diamine;

N 3 -(2,6-dichlorophenyl)-1-(3-methoxy-3-methylbutyl)-N 6 -[4-(4-methyl-piperazin-1-yl)-phenyl]-

1 H-indazole-3,6-diamine;

9H-fluoren-9-yl)methyl 4-(4-(3-(2,6-dichlorophenylamino)-1-(3-methoxy-3-methyl-buty l)-1 H- indazol-6-ylamino)phenyl)piperazine-1 -carboxylate; N 3 -(2,6-dichlorophenyl)-N 6 -(3-fluoro-4-(2-(pyrrolidin-1-yl)ethoxy)phenyl)-1-(3-m ethoxy-3- methylbutyl)-1 H-pyrazolo[3,4-d]pyrimidine-3,6-diamine; and

N 3 -(2,6-dichlorophenyl)-1-(3-methoxy-3-methylbutyl)-N 6 -methyl-1H-pyrazolo[3,4- d]pyrimidine-3,6-diamine.

[0051] Dasatinib inhibits SRC-non-receptor tyrosine kinases and mainly targets kinases Bcr-Abl, SRC, Ephrins, and GFR (Talpaz et al., /V Engl J Med. 2006 Jun 15;354(24):2531-41; Das et al., J Med Chem. 2006 Nov 16;49(23):6819-32). Dasatinib has a molecular formula of C22H26CIN7O2S and an IIIPAC name of N-(2-chloro-6-methylphenyl)-2-[[6-[4-(2-hydroxyethyl)piperaz in-1-yl]-2- methylpyrimidin-4-yl]amino]-1,3-thiazole-5-carboxamide. Dasatinib has the following structure:

[0052] W02020033927 disclose various derivatives of Dasatinib having similar functional properties as Dasatinib. Examples of derivatives of Dasatinib include the following compounds: saracatinib, bosutinib,

[0053] PDK1/Akt/Flt (also known as Akt Inhibitor XXI, PDK1 Inhibitor I, or KP372-1) is a dual pathway inhibitor (DPI) that inhibits the activities of kinases PDK1 and Akt and blocks or reduces cellular phosphorylation of Akt at both Ser 473 and Thr 308 . PDK1/Akt/Flt has a molecular formula of C10H4N6O and an IIIPAC name of 6H-lndeno[1 ,2-e]tetrazolo[1 ,5-b][1 ,2,4]triazin-6-one & 10H- lndeno[2,1-e]tetrazolo[1 ,5-b][1 ,2,4]triazin-10-one. PDK1/Akt/Flt has the following structure:

[0054] US20030144294 discloses derivatives of PDK1/Akt/Flt having similar functional properties as PDK1/Akt/Flt. Examples of derivatives of PDK1/Akt/Flt include: wherein, independently, v, w, and x include C, N, O, H or, S; y and z are is N, C, or H, with the proviso that each of w, v, x, y and z is not simultaneously C; the ring formed from v, w, x, y and z may be saturated or unsaturated; and

R 1 , R 2 , R 3 and R 4 , independently, is hydrogen, alkyl, aryl, alkaryl, aralkyl, heteroalkyl, and heteroaryl; wherein any adjacent two of R 1 , R 2 , R 3 and R 4 may join together to form a 5, 6 or 7- membered carbocyclic or heterocyclic ring, with the proviso that each of R 1 , R 2 , R 3 and R 4 is not simultaneously hydrogen.

[0055] In particular embodiments, R 1 , R 2 , R 3 , and R 4 , independently, is hydrogen, Ci-Cealkyl or Ci-Ce heteroalkyl. In particular embodiments, at least one of R 1 , R 2 , R 3 and R 4 is a heteroalkyl group selected from groups of the formula R 5 -O- and R 5 -S- wherein R 5 is a C1-C15, a C1-C10, a C1- Ce hydrocarbyl, or heteroalkyl. In particular embodiments, at least one of R 1 , R 2 , R 3 and R 4 include a heteroalkyl group selected from fluorine, chlorine, bromine and iodine. In particular embodiments, at least one of R 1 , R 2 , R 3 and R 4 include an alkyl group selected from Ci-Cisalkyl, while in another aspect the alkyl group include Ci-Cealkyl.

[0056] Staurosporine is an alkaloidal, non-selective, inhibitor of protein kinase C (US5344926). Staurosporine has a molecular formula of C28H26N4O3 and an IIIPAC name of 2,3,10,11 ,12,13- hexahydro-10R-methoxy-9S-methyl-11 R-methylamino-9S,13R-epoxy-1 H,9H-diindolo[1 ,2,3- gh;3’,2’,1’-lm]pyrrolo[3,4-j][1 ,7]benzodiazonin-1-one. Staurosporine has the following structure:

[0057] EP0575955 discloses various derivatives of staurosporine having similar functional properties as staurosporine. Examples of derivatives of staurosporine include: wherein Q represents hydrogen or -X-(CH2) m -Y-(CH2)n-Z ; wherein X represents a single bond or -CO-; Y represents a single bond or -CH(OH)-; Z represents hydroxy, OCONR 1 R 2 , in which each of R 1 and R 2 independently represents hydrogen or an alkyl group having 1 to 6 carbon atoms, or R 1 and R 2 , combined together with the nitrogen atom adjacent thereto, form a heterocyclic group carboxyl, NR 3 R 4 , in which each of R 3 and R 4 independently represents hydrogen or an alkyl group having 1 to 6 carbon atoms, or R 3 and R 4 , combined together with the nitrogen atom adjacent thereto, form a heterocyclic group or, substituted or unsubstituted aryl, each of m and n independently represents an integer of from 0 to 6. [0058] In particular embodiments, derivatives of staurosporine include the following compound: wherein:

X represents a single bond or -CO-; Y represents a single bond or -CH(OH)-;

Z represents hydroxy, OCONR 1 R 2 in which each of

R 1 and R 2 independently represents hydrogen or an alkyl group having 1 to 6 carbon atoms, or R 1 and R 2 , combined together with the nitrogen atom adjacent thereto, form a heterocyclic group carboxyl, NR 3 R 4 , in which each of R 3 and R 4 independently represents hydrogen or an alkyl group having 1 to 6 carbon atoms, or R 3 and R 4 , combined together with the nitrogen atom adjacent thereto, form a heterocyclic group or, substituted or unsubstituted aryl, each of m and n independently represents an integer of from 0 to 6. [0059] Cdk1/2 inhibitor III was found to inhibit Cdk1/cyclin B, Cdk2/cyclin A, VEGF-R2, and GSK- 3p, with IC50 values measured at 600 pM, 500 pm, 32 nM, and 140 nM, respectively (Lin, R. et al., 2005. J. Med. Chem. 48, 4208). Cdk1/2 inhibitor III has a molecular formula of C15H13F2N7O2S2 and an IIIPAC name of 5-Amino-3-((4-(aminosulfonyl)phenyl)amino)-N-(2,6-difluoroph enyl)-1 H- 1 , 2, 4-triazole-1 -carbothioamide. Cdk1/2 inhibitor III has the following structure: [0060] US6924302 discloses various derivatives of Cdk1/2 inhibitor III having similar functional properties as Cdk1/2 inhibitor III. Examples of derivatives of Cdk1/2 inhibitor III include: wherein

R 4 is selected from the group consisting of: Ci-salkyl, which is optionally substituted on a terminal carbon with a substituent selected from the group consisting of — C(O)H, — C(O)(Ci. s)alkyl, — CO2H, — CO2(Ci-8)alkyl, amino, amino substituted with two substituents independently selected from the group consisting of hydrogen and Ci-salkyl, cyano, (halo)i-3, hydroxy, nitro, cycloalkyl, aryl, thienyl, imidazolinyl, and triazolyl; Ci-salkoxy or Ci-salkoxy substituted on a terminal carbon with a substituent selected from the group consisting of (halo)i-s and hydroxy; — C(O)H, — C(O)(Ci-s)alkyl, — CO2H, — CO2(Ci-s)alkyl; amino, or amino substituted with two substituents independently selected from the group consisting of Ci-salkyl and — SO2 — (Ci-s)alkyl; — C(O)amino, wherein amino is substituted with two substituents independently selected from the group consisting of hydrogen and Ci-salkyl; — SO2 — , substituted with one substituent selected from the group consisting of thienyl, imidazolinyl, triazolyl and amino, wherein amino is substituted with two substituents independently selected from the group consisting of hydrogen, Ci-salkyl , — Ci-salkylamino, wherein amino is substituted with two substituents independently selected from the group consisting of hydrogen and Ci-salkyl, thienyl, imidazolinyl, and triazolyl; cycloalkyl, aryl, thienyl, imidazolinyl, and triazolyl; wherein cycloalkyl, aryl, thienyl, imidazolinyl, and triazolyl are optionally substituted with 1 to 3 substituents independently selected from the group consisting of Ci-salkyl, wherein alkyl is optionally substituted on a terminal carbon with a substituent selected from the group consisting of amino, or amino substituted with two substituents independently selected from the group consisting of hydrogen and Ci-salkyl , cyano, (halo)i-s, hydroxy and nitro, Ci-salkoxy, amino, substituted with two substituents independently selected from the group consisting of hydrogen and Ci-salkyl), cyano, halo, hydroxy and nitro; and, wherein thienyl, imidazolinyl, or triazolyl are optionally substituted with 1 to 2 oxo substituents; X is selected from the group consisting of — C(O) — , — C(S) — and — SO2 — ; and, R 3 is selected from the group consisting of: Ci-salkyl, C^alkenyl, C2-salkynyl, wherein alkyl, alkenyl and alkynyl are optionally substituted on a terminal carbon with a substituent selected from the group consisting of — C(O)H, — C(O)(Ci-8)alkyl, — CO2H, — CC>2(Ci.8)alkyl, or amino substituted with two substituents independently selected from the group consisting of hydrogen, cyano, (halo)i. 3 , hydroxy, nitro, aryl and thienyl, imidazolinyl, or triazolyl; wherein aryl, thienyl, imidazolinyl, or triazolyl are optionally substituted with 1 to 3 substituents independently selected from the group consisting of Ci-salkyl, cyano, halo, (halo)i. 3 (Ci-8)alkyl, (halo)i. 3 (Ci.8)alkoxy, hydroxy, hydroxy(Ci.s)alkyl, hydroxy(Ci.s)alkoxy and nitro; cycloalkyl, thienyl, imidazolinyl, triazolyl, and aryl, wherein cycloalkyl, thienyl, imidazolinyl, triazolyl, and aryl are optionally substituted with 1 to 3 substituents independently selected from the group consisting of cyano, halo, hydroxy and nitro; and, wherein cycloalkyl, aryl, thienyl, imidazolinyl, or triazolyl are optionally substituted with 1 to 2 substituents independently selected from the group consisting of: Ci-salkyl, C^alkenyl, wherein alkyl and alkenyl are optionally substituted on a terminal carbon with a substituent selected from the group consisting of — C(O)H, — C(O)(Ci-s)alkyl, — CO2H, — CO2(Ci-8)alkyl, amino, amino substituted with two substituents independently selected from the group consisting of hydrogen and Ci-salkyl, cyano, (halo)i- 3 , hydroxy, nitro, cycloalkyl, thienyl, imidazolinyl, triazolyl, and aryl, — CH(OH) — Ci-s)alkyl, Ci-salkoxy, optionally substituted on a terminal carbon with a substituent selected from the group consisting of (halo)i- 3 and hydroxy; — C(O)H, — C(O)(Ci-s)alkyl, — CO2H, — CO2(Ci-s)alkyl, amino, or amino substituted with two substituents independently selected from the group consisting of hydrogen, Ci-salkyl and — C(O)(Ci-s)alkyl, — C(O)amino, wherein amino is substituted with two substituents independently selected from the group consisting of hydrogen and Ci-salkyl, — SO2 — , substituted with one substituent selected from the group consisting of thienyl, imidazolinyl, triazolyl and amino, wherein amino is substituted with two substituents independently selected from the group consisting of hydrogen, Ci-salkyl and — Ci-salkylamino, wherein amino is substituted with two substituents independently selected from the group consisting of hydrogen and Ci-salkyl; — NH — SO2 — (Ci- s)alkyl, cycloalkyl, thienyl, imidazolinyl, triazolyl, optionally substituted with 1 to 2 oxo substituents, aryl, thienyl, imidazolinyl, and triazolyl, with a proviso when R 3 is amino substituted with two substitutents, the two substitutents independently are not selected from hydrogen and aryl, and wherein aryl is phenyl.

[0061] The kinase inhibitors and functional derivatives described herein can include their pharmaceutically acceptable salts, solvates, prodrugs, tautomers, enantiomers, stereoisomers, and diastereoisomers. For example, kinase inhibitors include dimaleates, mesylates, hydrochlorides, dihydrochlorides, sodiums, succinates, malates, citrates, ditosylates, tosylates, and esylates of described kinase inhibitors and functional derivatives thereof.

[0062] (ii) Androgen Receptor Signaling Inhibitors. Androgen receptor signaling inhibitors, as the name suggests, are techniques and compounds that block or otherwise inhibit or reduce androgen receptor (AR) signaling. For example, surgical removal of the testicles provides one example of an AR signaling inhibitor because the body no longer produces androgens. AR signaling inhibitors can also, for example, block individual AR receptors, compete for binding sites of an AR receptor, alter nuclear translocation, affect DNA binding of the AR receptor, reduce expression of AR, or otherwise interrupt the androgen production or signaling pathway. For more information regarding mechanisms of AR signal inhibition, see US Patent No. 10,377,828.

[0063] In particular embodiments, an AR signaling inhibitor includes a dose of abiraterone, apalutamide (ARN-509), AZD3514, chlormadinone acetate, darolutamide (ODM-201), EPI-001 , enzalutamide, finasteride, galeterone, izonsteride, ketoconazole, megestrol, steroidal antiandrogens, nadolol, N-butylbenzene-sulfonamide, RU58841 , Testolone (RAD140), triptophenolide, tulobuterol hydrochloride, and/or turosteride that inhibits AR signaling. In particular embodiments, an AR signaling inhibitor includes abiraterone and/or enzalutamide.

[0064] Abiraterone inhibits 17a-hydroxylase CYP17, which is required for androgen biosynthesis and is expressed in testicular, adrenal, and prostatic tumor tissues. CYP17 serves as a catalyst for the conversion of pregnenolone and progesterone to their 17a-hydroxy derivatives and the formation of dehydroepiandrosterone (DHEA) and androstenedione. DHEA and androstenedione are precursors of testosterone.

[0065] Apalutamide is a nonsteroidal antiandrogen medication used in the treatment of prostate cancer.

[0066] AZD3514 is a small molecule androgen receptor inhibitor and selective androgen-receptor down-regulator (SARD). AZD3514 modulates AR signaling through two distinct mechanisms, an inhibition of ligand-driven nuclear translocation of AR and a downregulation of receptor levels, both of which were observed in vitro and in vivo.

[0067] Chlormadinone acetate is a progestin and steroid antiandrogen that inhibits the uptake of testosterone by epithelial cells and the binding of DHT to the androgen receptor within the cell.

[0068] Darolutamide (ODM-201) competitively inhibits androgen binding, AR nuclear translocation, and AR-mediated transcription.

[0069] EPI-001 is an antagonist of the androgen receptor (AR) that acts by binding covalently to the N-terminal domain (NTD) of the AR and blocking protein-protein interactions required for transcriptional activity of the AR and its splice variants. [0070] Enzalutamide is a nonsteroidal anti-androgen that is an oral inhibitor of AR signaling that blocks AR interaction, inhibits translocation of the AR to the nucleus, impairs AR binding to DNA and inhibits co-activator recruitment and receptor-mediated DNA transcription.

[0071] Finasteride, a specific and competitive inhibitor of type 2 5a-reductase enzyme. It inhibits the conversion of testosterone to dihydrotestosterone (DHT). In adults, DHT acts as primary androgen in prostate and hair follicles.

[0072] Galeterone, a steroidal anti-androgen, suppresses AR activity both by functioning as a CYP17A1 inhibitor and by binding directly to AR (AR antagonism and AR degradation).

[0073] Izonsteride (also known as LY-320,236) is a selective inhibitor of type I and type II 5a- reductase.

[0074] Ketoconazole is an antifungal agent that, in high doses, inhibits testicular and adrenal steroid synthesis. Ketoconazole is a potent inhibitor of the P450 series of enzymes that are essential for the production of androgens.

[0075] Megestrol has antiandrogenic and antiestrogenic effects. It is a progestin medication used as an appetite stimulant or treat cancer such as breast cancer or endometrial cancer.

[0076] Nadolol is a nonselective beta adrenal receptor blocker that is used to lower blood pressure by inhibiting both beta-1 and beta-2 receptor subtypes.

[0077] N-butylbenzene-sulfonamide is an antagonist of the human androgen receptor. It has been isolated from the plant Prunus Africana and has a role as a neurotoxin and a plant metabolite.

[0078] RU58841 is a non-steroidal specific androgen receptor antagonist. These compounds are similar in chemical structure to nilutamide, which is related to flutamide, bicalutamide, and enzalutamide, all of which are nonsteroidal antiandrogens similarly.

[0079] Testolone (RAD140) is a selective androgen receptor modulator (SARM).

[0080] Triptophenolide is a product derived from Tripterygium wilfordii. It is a pan-antagonist for wild-type and mutant androgen receptors. Triptophenolide exhibits its antiandrogenic activity through competitive binding with androgen in the hormone-binding pocket, decreasing the expression of androgen receptor, and reducing the nuclear translocation of androgen receptor.

[0081] Tulobuterol hydrochloride is a direct-acting sympathomimetic with selective action on p2- receptors. It has bronchodilatory, anti-inflammatory, and antiviral activities.

[0082] Turosteride is a selective inhibitor of enzyme 5a-reductase. 5a-reductase is the enzyme responsible for the conversion of testosterone (T) to 5a-dihydrotestosterone (DHT).

[0083] In particular embodiments, AR signaling inhibitors include cyproterone acetate, spironolactone, flutamide, bicalutamide, and/or nilutamide. Cyproterone acetate is a synthetic progesterone derivative with progesterone-like and anti-androgenic activity that directly inhibits the androgen receptor. Spironalactone has inhibitory actions on both the androgen receptor and 5a-reductase. Flutamide is a potent nonsteroidal androgen antagonist. No cases of hepatic impairment with flutamide doses of 125 mg/day or less have been reported. Bicalutamide (marketed as Casodex, Cosudex, Calutide, Kalumid) is an oral non-steroidal anti-androgen used in the treatment of prostate cancer and hirsutism. It is recommended 50 mg once daily in combination with a luteinizing hormone-releasing hormone analogue or surgical castration or upon progression after castration as a secondary hormonal maneuver. Nilutamide blocks the androgen receptor, preventing its interaction with testosterone. Nilutamide is marketed under the name Nilandron in the United States and under the name Anandron in Canada. It can be used in combination with a luteinizing hormone-releasing hormone analogue or surgical castration or upon progression after castration as a secondary hormonal maneuver.

[0084] (iii) Compositions for Administration and Kits. Kinase inhibitors optionally combined with compound-based AR signaling inhibitors can be formulated for administration to subjects in one or more pharmaceutically acceptable carriers. As indicated, salts and/or prodrugs and other appropriate forms of protein kinase inhibitors and AR signaling inhibitors described herein can also be used. The protein kinase inhibitors and AR signaling inhibitors described herein can be individually, collectively, or in grouped combinations referred to as "active ingredients". Each active ingredient can be formulated into its own composition for administration or active ingredients can be formulated into the same composition.

[0085] A pharmaceutically acceptable salt includes any salt that retains the activity of the active ingredients and is acceptable for pharmaceutical use. A pharmaceutically acceptable salt also refers to any salt which may form in vivo as a result of administration of an acid, another salt, or a prodrug which is converted into an acid or salt.

[0086] Suitable pharmaceutically acceptable acid addition salts can be prepared from an inorganic acid or an organic acid. Examples of such inorganic acids are hydrochloric, hydrobromic, hydroiodic, nitric, carbonic, sulfuric and phosphoric acid. Appropriate organic acids can be selected from aliphatic, cycloaliphatic, aromatic, arylaliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids.

[0087] Suitable pharmaceutically acceptable base addition salts include metallic salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made from N,N'-dibenzylethylene-diamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N- methylglucamine, lysine, arginine and procaine.

[0088] A prodrug includes an active ingredient which is converted to a therapeutically active compound after administration, such as by cleavage or by hydrolysis of a biologically labile group. [0089] The formulations disclosed herein can be formulated for administration by, for example, injection. For injection, formulations can be formulated as aqueous solutions, such as in buffers including Hanks' solution, Ringer's solution, saline, buffered saline or physiological saline, or in culture media, such as Iscove’s Modified Dulbecco’s Medium (IMDM). The aqueous solutions can include formulatory agents such as suspending, stabilizing, and/or dispersing agents.

[0090] Examples of suitable non-aqueous carriers which may be employed in the injectable formulations include polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyloleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of selected particle size in the case of dispersions, and by the use of surfactants.

[0091] Injectable formulations may also contain adjuvants such as preservatives, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like in the compositions.

[0092] Compositions can be provided in lyophilized form and/or provided in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. Lyophilized compositions can include less than 5% water content; less than 4.0% water content; or less than 3.5% water content.

[0093] In particular embodiments, the composition can be in a unit dosage form, such as in a suitable diluent in sterile, hermetically sealed ampoules or sterile syringes.

[0094] For oral administration, the compositions can be formulated as capsules, dragees, edibles, elixirs, emulsions, gels, gelcaps, granules, gums, juices, liquids, oils, pastes, pellets, pills, powders, rapidly-dissolving tablets, sachets, semi-solids, slurries, sprays, solutions, suspensions, syrups, and tablets. For oral solid formulations such as powders, capsules and tablets, suitable excipients include binders (gum tragacanth, acacia, cornstarch, gelatin), fillers such as sugars, e.g. lactose, sucrose, mannitol and sorbitol; dicalcium phosphate, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate; cellulose preparations such as maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxy-methylcellulose, and/or polyvinylpyrrolidone (PVP); granulating agents; and binding agents. If desired, disintegrating agents can be added, such as corn starch, potato starch, alginic acid, cross-linked polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. If desired, solid dosage forms can be sugar-coated or enteric-coated using standard techniques. Flavoring agents, such as peppermint, oil of Wintergreen, cherry flavoring, orange flavoring, etc. can also be used.

[0095] Additionally, compositions can be formulated as sustained-release systems utilizing semipermeable matrices of solid polymers including active ingredients as described herein. Various sustained-release materials have been established and are well known by those of ordinary skill in the art. Sustained-release systems may, depending on their chemical nature, release active ingredients following administration for a few weeks up to over 100 days. Sustained-release systems can be administered by injection; parenteral injection; instillation; or implantation into soft tissues, a body cavity, or occasionally into a blood vessel with injection through fine needles.

[0096] Sustained-release systems can include a variety of bioerodible polymers including poly(lactide), poly(glycolide), poly(caprolactone) and poly(lactide)-co(glycolide) (PLG) of desirable lactide:glycolide ratios, average molecular weights, polydispersities, and terminal group chemistries. Blending different polymer types in different ratios using various grades can result in characteristics that borrow from each of the contributing polymers.

[0097] Any composition disclosed herein can advantageously include any other pharmaceutically acceptable carriers which include those that do not produce significantly adverse, allergic, or other untoward reactions that outweigh the benefit of administration. Exemplary pharmaceutically acceptable carriers and formulations are disclosed in Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990. Moreover, formulations can be prepared to meet sterility, pyrogenicity, general safety, and purity standards as required by U.S. FDA Office of Biological Standards and/or other relevant foreign regulatory agencies.

[0098] Therapeutically effective amounts of active ingredients within a composition can include at least 0.1% w/v or w/w active ingredient; at least 1 % w/v or w/w active ingredient; at least 10% w/v or w/w active ingredient; at least 20% w/v or w/w active ingredient; at least 30% w/v or w/w active ingredient; at least 40% w/v or w/w active ingredient; at least 50% w/v or w/w active ingredient; at least 60% w/v or w/w active ingredient; at least 70% w/v or w/w active ingredient; at least 80% w/v or w/w active ingredient; at least 90% w/v or w/w active ingredient; at least 95% w/v or w/w active ingredient; or at least 99% w/v or w/w active ingredient.

[0099] In particular embodiments, compositions can include a combination of kinase inhibitors. The combination of kinase inhibitors can be combined in a w/w ratio of, for example, 1 :1 , 1 :2, 1 :3,

1 :4, 1 :5, 1 :6, 1 :7, 1 :8, 1 :9, 1 :10, 1 :11 , 1 :12, 1 :13, 1 :14, 1 :15, 1 :16, 1 :17, 1 :18, 1 :19, 1 :20, 1 :21 ,

1 :22, 1 :23, 1 :24, 1 :25, 1 :26, 1 :27, 1 :28, 1 :29, 1 :30, 1 :31 , 1 :32, 1 :33, 1 :34, 1 :35, 1 :36, 1 :37, 1 :38,

1 :39, 1 :40, 1 :41 , 1 :42, 1 :43, 1 :44, 1 :45, 1 :46, 1 :47, 1 :48, 1 :49, 1 :50, 1 :51 , 1 :52, 1 :53, 1 :54, 1 :55, 1:56, 1:57, 1:58, 1:59, 1:60, 1:61, 1:62, 1:63, 1:64, 1:65, 1:66, 1:67, 1:68, 1:69, 1:70, 1:71, 1:72, 1:73, 1:74, 1:75, 1:76, 1:77, 1:78, 1:79, 1:80, 1:81, 1:82, 1:83, 1:84, 1:85, 1:86, 1:87, 1:88, 1:89, 1:90, 1:91, 1:92, 1:93, 1:94, 1:95, 1:96, 1:97, 1:98, 1:99, or 1:100.

[0100] In particular embodiments, compositions can include a combination of kinase inhibitors. The combination of kinase inhibitors can be combined in a w/w/w ratio of, for example, 1:1:1, 1:1:2, 1:1:3, 1:1:4, 1:1:5, 1:1:6, 1:1:7, 1:1:8, 1:1:9, 1:1:10, 1:1:11, 1:1:12, 1:1:13, 1:1:14, 1:1:15, 1:1:16, 1:1:17, 1:1:18, 1:1:19, 1:1:20, 1:2:21, 1:5:22, 1:10:23, 1:2:24, 1:5:25, 1:10:26, 1:1:27, 1:2:28, 1:5:29, 1:10:30, 1:2:31, 1:1.5:32, 1:1:33, 1:1.5:34, 1:1:35, 1:1:36, 1:6:37, 1:12:38, 1:9:39, 1:4:40, 1:2:41, 1:5:42, 1:10:43, 1:12:44, 1:18:45, 1:1:46, 1:30:47, 1:1:48, 1:1:49, 1:1:50, 1:1:51, 1:1:52, 1:1:53, 1:1:54, 1:1:55, 1:1:56, 1:1:57, 1:1:58, 1:1:59, 1:6:60, 1:1:61, 1:3:62, 1:1:63, 1:1:64, 1:1:65, 1:1:66, 1:1:67, 1:1:68, 1:1:69, 1:6:70, 1:3:71, 1:2:72, 1:3:73, 1:2:74, 1:3:75, 1:4:76, 1:15:77, 1:6:78, 1:7:79, 1:8:80, 1:9:81, 1:10:82, 1:11:83, 1:12:84, 1:13:85, 1:14:86, 1:15:87, 1:16:88, 1:17:89, 1:18:90, 1:19:91, 1:20:92, 1:21:93, 1:22:94, 1:23:95, 1:24:96, 1:25:97, 1:26:98, 1:27:99, 1:28:100, 25:1:1, 25:1:25, 25:1:49, 25:1:99, 25:25:1, 28:25:25, 37:25:50, 25:25:99, 25:48:2, 25:50:27, 25:50:52, 25:50:99, 25:100:1, 28:100:25, 36:100:50, 25:100:99, 50:1:1, 50:1:25, 50:1:50, 50:1:100, 50:25:1, 2:1:1, 2:1:2, 50:25:99, 50:50:1, 49:50:25, 48:50:49, 50:49:100, 50:100:1, 37:100:25, 48:100:50, 50:99:100, 100:1:1, 100:1:25, 100:1:50, 100:1:100, 100:25:1, 100:28:25, 100:30:50, 99:25:100, 100:50:1, 100:49:25, 100:50:49, 99:50:100, 100:100:1, 100:99:25, 99:100:50, or 100:99:100.

[0101] In particular embodiments, protein kinase inhibitors can be co-formulated with an androgen receptor signaling inhibitor, for example, ataw/wratio of 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1:21, 1:22, 1:23, 1:24, 1:25, 1:26, 1:27, 1:28, 1:29, 1:30, 1:31, 1:32, 1:33, 1:34, 1:35, 1:36, 1:37, 1:38, 1:39, 1:40, 1:41,

1:42, 1:43, 1:44, 1:45, 1:46, 1:47, 1:48, 1:49, 1:50, 1:51, 1:52, 1:53, 1:54, 1:55, 1:56, 1:57, 1:58,

1:59, 1:60, 1:61, 1:62, 1:63, 1:64, 1:65, 1:66, 1:67, 1:68, 1:69, 1:70, 1:71, 1:72, 1:73, 1:74, 1:75,

1:76, 1:77, 1:78, 1:79, 1:80, 1:81, 1:82, 1:83, 1:84, 1:85, 1:86, 1:87, 1:88, 1:89, 1:90, 1:91, 1:92,

1:93, 1:94, 1:95, 1:96, 1:97, 1:98, 1:99, 1:100, 25:15, 25:30, 25:55, 25:70, 25:99, 56:15, 56:30, 56:55, 56:70, 56:100, 77:15, 77:30, 77:55, 77:70, 77:100, 100:1, 100:15, 100:30, 100:56, 100:70, or 100:99.

[0102] Compositions described herein can be packaged into kits for commercial sale.

[0103] (iv) Methods of Use. Methods disclosed herein include treating subjects (humans, veterinary animals (dogs, cats, reptiles, birds, etc.) livestock (horses, cattle, goats, pigs, chickens, etc.) and research animals (monkeys, rats, mice, fish, etc.) with compositions disclosed herein. Treating subjects includes delivering therapeutically effective amounts. Therapeutically effective amounts include those that provide effective amounts, prophylactic treatments, and/or therapeutic treatments.

[0104] An "effective amount" is the amount of a composition necessary to result in a desired physiological change in the subject. For example, an effective amount can provide an anti-cancer effect. Effective amounts are often administered for research purposes. Effective amounts disclosed herein can cause a statistically-significant effect in an animal model or in vitro assay relevant to the assessment of a cancer’s development or progression.

[0105] A "prophylactic treatment" includes a treatment administered to a subject who does not display signs or symptoms of a cancer or displays only early signs or symptoms of a cancer such that treatment is administered for the purpose of diminishing or decreasing the risk of developing the cancer further. Thus, a prophylactic treatment functions as a preventative treatment against a cancer. In particular embodiments, prophylactic treatments reduce, delay, or prevent metastasis from a primary cancer tumor site from occurring.

[0106] A "therapeutic treatment" includes a treatment administered to a subject who displays symptoms or signs of a cancer and is administered to the subject for the purpose of diminishing or eliminating those signs or symptoms of the cancer. The therapeutic treatment can reduce, control, or eliminate the presence or activity of the cancer and/or reduce control or eliminate side effects of the cancer.

[0107] Function as an effective amount, prophylactic treatment or therapeutic treatment are not mutually exclusive, and in particular embodiments, administered dosages may accomplish more than one treatment type.

[0108] In particular embodiments, therapeutically effective amounts provide anti-cancer effects. Anti-cancer effects include one or more of a decrease in the number of cancer cells, a decrease in tumor volume, inhibited tumor growth, induced chemo- or radiosensitivity in cancer cells, inhibited angiogenesis near cancer cells, inhibited cancer cell proliferation, reduced cancer- associated pain, prevented or reduced development of metastases, an increase in life expectancy, reduced relapse or re-occurrence of cancer following treatment, and/or prolonged subject life.

[0109] For administration, therapeutically effective amounts (also referred to herein as doses) can be initially estimated based on results from in vitro assays and/or animal model studies. Such information can be used to more accurately determine useful doses in subjects of interest. The actual dose amount administered to a particular subject can be determined by a physician, veterinarian, or researcher taking into account parameters such as physical, physiological and psychological factors including target, body weight, stage of PCa, type of PCa, previous or concurrent therapeutic interventions, idiopathy of the subject, and route of administration.

[0110] The compound(s) of the present invention can be administered in any form by any effective route, including, e.g., intra-arterial, oral, parenteral, enteral, intraperitoneal, topical, transdermal, nasally, aerosal, spray, inhalation, subcutaneous, intravenous, intramuscular, intrathecal, intratumoral, etc.

[0111] Exemplary doses of active ingredients can include 0.05 mg/kg to 5.0 mg/kg. For certain indications, the total daily dose can be 0.05 mg/kg to 30.0 mg/kg active ingredients administered to a subject one to three times a day, including administration of total daily doses of 0.05-3.0, 0.1- 3.0, 0.5-3.0, 1.0-3.0, 1.5-3.0, 2.0-3.0, 2.5-3.0, and 0.5-3.0 mg/kg/day of administration forms of active ingredients using 60-minute oral, intravenous or other dosing. In one particular example, doses can be administered once a day or twice a day (quaque die (QD) or bis in die (BID), respectively) to a subject with, e.g., total daily doses of 1.5 mg/kg, 3.0 mg/kg, or 4.0 mg/kg of a composition with up to 92-98% wt/v of kinase inhibitor.

[0112] Additional useful doses can often range from 0.1 to 5 pg/kg or from 0.5 to 1 pg/kg. In other examples, a dose can include 1 pg/kg, 5 pg/kg, 10 pg/kg, 20 pg/kg, 40 pg/kg, 80 pg/kg, 150 pg/kg, 350 pg/kg, 500 pg/kg, 0.1 to 5 mg/kg or from 0.5 to 1 mg/kg. In other examples, a dose can include 1 mg/kg, 5 mg/kg, 10 mg/kg, 20 mg/kg, 40 mg/kg, 80 mg/kg, 150 mg/kg, 350 mg/kg, 500 mg/kg, 550 mg/kg, 1000 mg/kg, or more.

[0113] Therapeutically effective amounts can be achieved by administering single or multiple doses during the course of a treatment regimen (e.g., hourly, every 2 hours, every 3 hours, every 4 hours, every 6 hours, every 9 hours, every 12 hours, every 18 hours, daily, every other day, every 3 days, every 4 days, every 5 days, every 6 days, weekly, every 2 weeks, every 3 weeks, or monthly).

[0114] In particular embodiments, a kinase inhibitor and, optionally, an AR signaling inhibitor is administered to prostate cancer cells. The administered active ingredients can be administered serially (within minutes, hours, or days of each other) or in parallel. As indicated previously, they also may be administered to a subject in a pre-mixed single composition.

[0115] Combination therapies disclosed herein can also be practiced with additional PCa therapy modalities. For example, combination therapies can be practiced in combination with radiation therapy. Radiation therapy can come from a machine outside the body (external-beam radiation therapy) or from radioactive material placed in the body (internal radiation therapy, more commonly called brachytherapy). Systemic radiation therapy uses a radioactive substance, given by mouth or into a vein that travels in the blood to tissues throughout the body. In particular embodiments, systemic radiation therapy, includes administration of a radioactive substance, such as radioactive iodine or a radioactive substance bound to a monoclonal antibody.

[0116] External beam radiation can include intensity-modulated radiation therapy (IMRT), image- guided radiation therapy (IGRT), stereotactic radiosurgery (SRS), stereotactic body radiation therapy (SBRT), and proton therapy. An example of internal radiation therapy is the MammoSite (Hologic, Inc., Marlborough, Mass.) system.

[0117] Other therapies that can be recommended or administered by a physician with the combination therapies described herein include: interferons, interleukins, colony-stimulating factors, monoclonal antibodies, vaccines, gene therapy, cellular immunotherapy, nonspecific immunomodulating agents, photodynamic therapy, laser therapy including laser-induced interstitial thermotherapy, and surgery or cryosurgery for the removal of tumors or abnormal tissue.

[0118] (v) Assays to Predict In Vivo Efficacy of Compounds Against PCa. The current disclosure also provides assays to assess and/or predict the therapeutic efficacy of particular protein kinase inhibitors using an assay that determines levels of phosphoproteins, such as p-EGFR Y1173 , p- S6 S 235/236, p_pQpRY653/654 ] p.|\/| ET Y1349 , p-IGF1 R Y1131 , p-AKT S473 and/or the anti-apoptotic protein, p-BAD S112 . As described herein, an observed decrease in one or more of these proteins in an assay correlates with the in vivo therapeutic efficacy of a protein kinase inhibitor against PCa.

[0119] Epidermal growth factor receptor (EGFR; also known as ERBB, ERBB1 , HER1) is a receptor tyrosine kinase belonging to the family of receptor tyrosine kinases, and activation by its ligands leads to trans-auto-phosphorylation at five tyrosine residues (Y992, Y1045, Y1068, Y1148 and Y1173) in the C-terminal domain. This autophosphorylation leads to downstream activation and signaling by several other proteins that associate with the phosphorylated tyrosines through phosphotyrosine-binding domains. EGFR activation results in major downstream signaling cascades including the RAS-RAF-mitogen activated protein kinase (MEK)-extracellular signal regulated kinase (ERK), phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT), phospholipase C (PLC) gamma-protein kinase C (PKC) and activator of transcription proteins (STATs). These downstream events modulate various cell functions, including migration, adhesion, and proliferation. EGFR is over-active and/or over-expressed in cancers of epithelial origin. Autophosphorylation of EGFR at Y1173 can be a surrogate marker for EGFR activity. Anti-p- EGFR Y1173 antibodies that bind to EGFR phosphorylated at Y1173 are commercially available, including polyclonal IgG anti-human p-EGFR Y1173 from R&D Systems (AF1095; Minneapolis, MN), a rabbit monoclonal anti-human p-EGFR Y1173 from Abeam (E124; Cambridge, UK), and a mouse monoclonal lgG2b anti-p-EGFR Y1173 clone 3E7.2 from Millipore Sigma (Cat # MABS829; Burlington, MA). [0120] Ribosomal protein S6 is a component of the 40S subunit of the ribosome and is involved in cell growth and proliferation. Growth factors and mitogens induce the activation of p70 S6 kinase, which can then phosphorylate ribosomal protein S6. Phosphorylation of ribosomal protein S6 correlates with an increase in translation of mRNA transcripts that contain an oligopyrimidine tract in their 5' untranslated regions (5'TOP). These 5'TOP mRNA transcripts encode ribosomal proteins and elongation factors necessary for translation, as well as proteins involved in cell cycle progression. S6 ribosomal protein phosphorylation sites include S235, S236, S240, and S244, located within a small, carboxy-terminal region of the S6 protein. Ribosomal protein S6 is dephosphorylated upon growth arrest. Anti-p-S6 S235/236 antibodies that bind S6 ribosomal protein phosphorylated at S235 and S236 are commercially available, including a rabbit polyclonal anti- p-S6 S235/236 from Cell Signaling Technology (#2211 ; Beverly, MA), a rabbit monoclonal anti-human p-S6 S235/236 clone SP50 from Roche (Basel, Switzerland), and a mouse monoclonal lgG1 anti-p- S6S235/236 C | one cupk43k (Thermo Fisher, Waltham, MA).

[0121] Fibroblast Growth Factor Receptors (FGFRs) belong to the fibroblast growth factor receptor family involved in mitogenesis, limb induction, and differentiation. FGFR family members include FGFR1 , FGFR2, FGFR3, and FGFR4, and differ from one another in their ligand affinities and tissue distribution. A full-length representative FGFR protein includes an extracellular region including Ig-like domains, a single hydrophobic membrane spanning region, and a cytoplasmic tyrosine kinase domain. The extracellular portion of FGFR interacts with both acidic and basic fibroblast growth factors. Following ligand binding and dimerization, the receptors are phosphorylated at specific tyrosine residues. Tyrosine residues including Y463, Y583, Y585, Y653, Y654, Y730, and Y766 in the cytoplasmic tail can be phosphorylated. Y653 and Y654 are important for catalytic activity and signaling of activated FGFR. The other phosphorylated tyrosine residues may provide docking sites for downstream signaling components. Anti-p-FGFR Y653/654 antibodies that bind FGFR phosphorylated at Y653 and Y654 are commercially available, including a mouse monoclonal lgG2b anti-human p-FGFR Y653/654 from Cell Signaling Technology (#3476; Beverly, MA), a rabbit polyclonal IgG anti-human p-FGFR Y653/654 from R&D Systems (AF3285; Minneapolis, MN), and a rabbit polyclonal IgG anti-p-FGFR Y653/654 from Thermo Fisher (#44-1140G; Waltham, MA).

[0122] Met is a high affinity tyrosine kinase receptor for hepatocyte growth factor (HGF; also known as scatter factor (SF)). Met exists as a disulfide-li nked heterodimer including a 45 kDa a subunit and a 145 kDa p subunit. The a subunit and the N-terminal region of the subunit form the extracellular domain. The remainder of the p subunit spans the plasma membrane and contains a cytoplasmic region with tyrosine kinase activity. Interaction of Met with HGF leads to autophosphorylation of Met at multiple tyrosines, which results in the recruitment of several downstream signaling components, including Gabi , c-Cbl, and PI3K. Phosphorylation of Y1234/1235 in the Met kinase domain is critical to kinase activation. The addition of a phosphate at cytoplasmic Y1003 is essential for Met protein ubiquitination and degradation. Phosphorylation at Y1349 in the Met cytoplasmic domain provides a direct binding site for substrates such as Gabi , Grb2, PI3K, and others. Altered Met levels and/or tyrosine kinase activities are found in several types of tumors, including renal, colon, and breast. Well-characterized downstream signalling pathways that are activated by c-Met include the ERK/MAPK, PI3K-Akt/PKB, Crk-Rap and Rac-Pak pathways, resulting in proliferation and increased cell survival. Anti-p-MET Y1349 antibodies that bind Met phosphorylated at Y1349 are commercially available, including rabbit polyclonal anti-human p-MET Y1349 from Rockland Immunochemicals, Inc. (#600-401-989S Gilbertsville, PA), rabbit monoclonal anti-p-MET Y1349 clone 130H2 from Cell Signaling Technology (#3133; Beverly, MA), and rabbit monoclonal anti-human p-MET Y1349 [EP2367Y] from Abeam (Cambridge, UK).

[0123] Insulin-like Growth Factor Receptors (IGFRs) are transmembrane tyrosine kinase receptors that play critical roles in development, cell growth and metabolism. Type I insulin-like growth factor receptor (IGF1 R) is a transmembrane receptor tyrosine kinase that is widely expressed in many cell lines and cell types within fetal and postnatal tissues. Receptor autophosphorylation follows binding of receptor ligands. Phosphorylation of these three tyrosine residues within the kinase domain (Y1131 , Y1135, and Y1136) is necessary for kinase activation and phosphorylation of appropriate substrates. Anti-p-IGF1 R Y1131 antibodies that bind IGF1 R phosphorylated atY1131 are commercially available, including rabbit polyclonal anti-p-IGF1 R Y1131 from Cell Signaling Technology (#3021 ; Beverly, MA), rabbit polyclonal anti-human p-IGF1 R Y1131 from Abeam (ab226915; Cambridge, UK), and rabbit polyclonal anti-p-IGF1 R Y1131 from AntibodyGenie (CABP0043; Dublin, Ireland).

[0124] AKT, also known as protein kinase B (PKB), is an ubiquitous serine/threonine kinase that plays an important role in diverse biological responses such as regulation of metabolism, cell survival, and growth by phosphorylating multiple proteins. This protein kinase is activated by insulin, PI3K, insulin growth factor 1 (IGF1) and various other growth and survival factors. Akt promotes cell survival by inhibiting apoptosis through phosphorylation and inactivation of several targets, including the proapoptotic protein Bad, forkhead transcription factors, c-raf, and caspase- 9. Three residues, T308 in the kinase domain, and S473 and Y474 in the C-terminal regulatory region, are phosphorylated when AKT is active. Anti-p-AKT S473 antibodies that bind AKT phosphorylated at S473 are commercially available, including a rabbit monoclonal IgG anti-p- AKT S473 clone 99H9L9 from Thermo Fisher (Waltham, MA), a rabbit monoclonal IgG anti-p- AKT S473 from Cell Signaling Technology (#4060; Beverly, MA), and mouse monoclonal lgG1 anti- p-AKT S473 clone #545007 from R&D Systems (Minneapolis, MN).

[0125] Bad (Bcl-2 antagonist of cell death) is a proapoptotic member of the Bcl-2 family that promotes cell death by forming heterodimers with Bcl-2 and Bcl-xL to reverse their death repressor activity. Bad displaces Bax interaction with Bcl-2 and Bcl-xL. Survival factors, such as interleukin (I L)-3, inhibit the apoptotic activity of Bad by activating intracellular signaling pathways that result in the phosphorylation of Bad at S112 and S136. Phosphorylation at these sites by kinases such as Akt, p90RSK, and mitochondria-anchored PKA can promote binding of Bad to 14-3-3 proteins to prevent an association between Bad with Bcl-2 and Bcl-xL. Phosphorylation at S155 in the BH3 domain by PKA plays a critical role in blocking the dimerization of Bad and Bcl- xL. Anti-p-BAD S112 antibodies that bind BAD phosphorylated at S112 are commercially available, including rabbit polyclonal anti-p-BAD S112 from Cell Signaling Technology (#9291 ; Beverly, MA), rabbit polyclonal anti-p-BAD S112 from Abeam (ab1435; Cambridge, UK), and rabbit polyclonal anti- p-BAD S112 from Abclonal (AP0010; Woburn, MA).

[0126] Cell lines used in PCa studies can include: normal cell lines (e.g., pRNS-1-1 , RWPE-1 , BPH 1 , PIN); hormone naive cell lines (e.g., RWPE-2, LNCaP (e.g., LNCaP42D), LAPC-4, LAPC- 9, VCaP, MDA PCa 2a/2b, LuCaP (e.g., LuCaP35CR)); and castration resistant cell lines (e.g., C4-2, C4-2B, 22Rv1 , ARCaP (aka MDA PCa 1), PC3, and DU-145). Additional cell lines are described in Cell L. BC Cancer Agency. 2014 Nov 21 ; Cell L. 2001.

[0127] Reverse Phase Protein Array (RPPA) is an antibody-based molecular profiling platform to analyze a large number of samples for quantitative assessment of key protein molecules in functional pathways. In particular embodiments, RPPA is similar to a miniaturized dot blot. In particular embodiments, RPPA determines levels of protein expression and modifications such as phosphorylation, cleavage, and fatty acid alteration. RPPA can be used to profile and validate signaling networks in human cancer cell lines and tumor tissue. In particular embodiments of RPPA, protein expression or modification can be compared between normal and diseased tissue or between samples that are untreated and treated with therapeutics. RPPA allows for high throughput, multiplexed, and very sensitive detection of proteins from extremely small numbers of input material. In particular embodiments, RPPA can detect picogram or attogram amounts of proteins.

[0128] Antibodies are key reagents for RPPA and can include antibodies that specifically recognize proteins acting on multiple signaling pathways, including: pathways including receptor tyrosine kinases; pathways including PI3K-AKT and mitogen-activated protein kinases (MAPK); cell metabolism pathways including liver kinase B1 (LKB1)-adenosine monophosphate-activated protein kinase (AMPK) and transforming growth factor beta (TGFP); DNA repair; cell cycle; immune responses to cancer; and/or apoptosis/autophagy. In particular embodiments, antibodies that bind to p-Src family 7416 , p-EGFR Y1173 , P -S6 s235/236 , p-S6 S240/244 , p-FAK Y925 , p-FGFR Y653/654 , p- MET Y1349 , p-IGF1 R Y1131 , p-AKT S473 , p-Erk1/2 T202/Y204 , p-BAD S112 , and/or p-MEK1/2 S217/221 can be used. In particular embodiments, an antibody to be used in RPPA is validated before use using a combination of immunohistochemistry and immunoblot analysis on formalin fixed and paraffin embedded tissues.

[0129] In particular embodiments, an RPPA can include the following steps. Tissue samples can be obtained from, e.g., intact cells, biopsy samples, laser capture micro-dissected cells, and/or biological fluids. In particular embodiments, samples can be fresh, frozen, or fixed. Tissue samples can be washed in PBS and homogenized in an RPPA lysis buffer to produce tissue lysates. In particular embodiments, an RPPA lysis buffer can include: 1% Triton X-100, 50 mmol/L HEPES (pH 7.4), 150 mmol/L NaCI, 1.5 mmol/L MgCI 2 , 1 mmol/L EGTA, 100 mmol/L NaF, 10 mmol/L NaPPi, 10% glycerol, 1 mmol/L Na 3 VO4, 1 mmol/L phenylmethylsulfonyl fluoride (PMSF), and 10 pg/mL aprotinin. Protein concentration of each sample can be determined by routine assays (e.g., Bradford assay) and adjusted accordingly for RPPA. The tissue lysates can be mixed with a sample buffer including sodium docecyl sulfate (SDS), glycerol, and 2- mercaptoethanol. Serially diluted tissue lysates can be printed on a solid phase such as nitrocellulose-coated slides with an arrayer such as the GeneTAC G3 arrayer (Genomic Solutions, Ann Arbor, Ml). In particular embodiments, each spot of an RPPA slide contains a whole cellular repertoire corresponding to a given cellular state that has been captured in the particular cell lysate. In particular embodiments, each spot is 250 to 350 pm wide.

[0130] Each RPPA slide can be probed with a labeled primary antibody alone, or a primary antibody followed by an appropriately labeled secondary antibody (such as a biotin-conjugated secondary antibody). The slides can be scanned and signals quantified. The signals can be generated by e.g., fluorescent, chemiluminescent, or colorimetric means. In particular embodiments, signals can be detected using a catalyzed signal amplification (CSA) system. This system takes advantage of tyramide amplification. In this system, a primary antibody is first detected with a peroxidase-conjugated secondary antibody. The bound peroxidase catalyzes oxidation of a fluorescein-conjugated phenol (fluorescyl-tyramide) which then precipitates onto the sample. The fluorescein in turn is detected by a peroxidase-conjugated anti-fluorescein. Staining is completed using diaminobenzidine/hydrogen peroxide as a chromogen/substrate. In particular embodiments, the tyramide signal amplification system can be biotin free. More than 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, or more antibodies can be used in an RPPA. An RPPA slide can include hundreds or thousands of samples.

[0131] Software such as MicroVigene (VigeneTech Inc. Carlisle, MA), Array Pro (Media Cybernetics, Rockville, MD), GenePix Pro (Molecular Devices, Sunnyvale, CA), or Mapix (Innopsys, Carbonne, France) can be used for automatic spot finding and background subtraction of signals on RPPA slides. In particular embodiments, the detection limit can be defined as two standard deviations above background. Quantification of protein levels of each sample can be done because the RPPA slides are spotted with a dilution series of a given sample. In particular embodiments, values representing the dilution series are in a linear range. Positive and negative controls can be included to compare against a measured spot signal. In particular embodiments, the controls can be in duplicates. In particular embodiments, a negative control can include a control antibody or serum that does not bind a protein of interest or a secondary antibody. In particular embodiments, spot intensity spatial variation can be corrected for each slide. In particular embodiments, a normalization step can be included to remove inter-array variability. In particular embodiments, a cell line panel can be spotted on each slide to allow calculation of spot sizes of a protein of interest normalized to the average spot intensity of the cell lines. In particular embodiments, protein signals are normalized to a housekeeping reference protein such as actin. In particular embodiments, the calculated data are normalized to correct for potential sources of variability that do not reflect biological differences in protein levels between the investigated samples, such as variations of total amount of protein extracts that were spotted. A common method to normalize RPPA data includes dye binding methods, i.e. , measurements of total protein on one slide per print run with dyes such as Fast Green FCF, Sypro Ruby, or colloidal gold. The signals obtained reflect the total amount of protein per spot immobilized on the solid phase. In particular embodiments, an antibody against single-stranded DNA (ssDNA) can be used as a suitable RPPA normalization parameter. RPPA is described in further detail in Paweletz et al. Oncogene 2001 ; 20: 1981-1989 and in Boellner and Becker. Microarrays (Basel). 2015; 4(2): 98- 114.

[0132] In particular embodiments, protein expression or modification in RPPA can be compared between different samples such as: normal vs. diseased tissue or untreated vs. treated samples. In particular embodiments, protein expression or modification in RPPA can be compared between cell lysates from mice tibia injected with PCa cells and the mice left untreated vs cell lysates from mice tibia injected with PCa cells and the mice treated with one or more protein kinase inhibitors. In particular embodiments, a statistically significant decrease in a protein level in a test state (e.g., treatment with one or more particular protein kinase inhibitors) as compared to the corresponding protein level in a reference state (e.g., untreated, treatment with a particular therapeutic) indicates that the protein is decreased in the test state. In particular embodiments, a statistically significant decrease includes a p value < 0.1 , < 0.09, < 0.08, < 0.07, < 0.06, < 0.05, < 0.04, < 0.03, < 0.02, < 0.01 , < 0.009, < 0.008, < 0.007, < 0.006, < 0.005, < 0.004, < 0.003, < 0.002, < 0.001 , or less. In particular embodiments, the Wilcoxon rank-sum test can be used to test group differences in the adjusted mean protein expression between a reference state and a test state. In particular embodiments, a protein level can be expressed as a normalized signal intensity against a housekeeping gene like actin.

[0133] Phosphoproteins disclosed herein can also be detected by: immunohistochemistry (e.g., on tissue microarrays); Western blots; far Western blots using “bait” proteins to probe for “prey” proteins of interest and then detecting the “bait” proteins; Src homology 2 (SH2) profiling using SH2 domains to probe for phosphotyrosine proteins; enzyme linked immunosorbent assays (ELISA); mass spectrometry-based technologies; kinase activity assays; intracellular flow cytometry and immunocytochemistry/immunohistochemistry; proteome profiling; and forward phase protein arrays, where two highly specific antibodies and high amounts of protein lysates are needed.

[0134] An “observed decrease” means that the measured level is lower than the measured level of a reference sample. As disclosed herein, decreases in one or more of the assayed proteins compared to a reference level predicts in vivo therapeutic efficacy. The predicted in vivo therapeutic efficacy can be confirmed, for example in animal models and clinical trials. For example, the effects of treatments on PCa in an animal model can be studied using protocols described herein and additionally, for example, as described in an in vivo xenograft system, where a human PCa cell line is transplanted into mice (Cunningham and You. J Biol Methods. 2015; 2(1): e17). Other in vivo xenograft models that can be used include: subcutaneous models where prostatic tissue is inserted into the shoulder of nude mice; orthotopic models where prostatic tissue is introduced into the mouse prostate; and subrenal capsule models that allow a high degree of vascularity.

[0135] In particular embodiments, the reference level is the level of the assayed proteins in a normal (i.e. , non-cancerous) sample. In particular embodiments, the reference level is the level of the assayed proteins in prostate cancer sample that is not exposed to the therapies disclosed herein.

[0136] (vi) Exemplary Embodiments.

1. A method of treating prostate cancer (PCa) in a subject in need thereof including administering a therapeutically effective amount of an SC-1 compound, a PP121 compound, a PD166285 dihydrochloride (diHCI) compound, an NCGC00348110 compound, a Dasatinib compound, a PDK1/Akt/Fit DPI compound, a Staurosporine compound, and/or a Cdk 1/2 Inhibitor III compound, thereby treating the PCa in the subject in need thereof (a “compound” can include “derivatives” disclosed herein).

2. The method of embodiment 1 , further including administering a therapeutically effective amount of an androgen receptor (AR) signaling inhibitor.

3. The method of embodiments 1 or 2, wherein the PCa is castration-resistant PCa (CRPC).

4. The method of any of embodiments 1-3, comprising administering an SC-1 compound or a PP121 compound, in certain examples, selected from (Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4) ccc2C)C3)n(C)n1);

Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N6CC OCC6)CC5)c(C(F)(F)F)c 4)ccc2C)C3)n(C)n1 ;

C=CC(=O)N1CCCN(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C (F)(F)F)c4)ccc2C)C3)n1 C;

CCc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N(C )C)C5)c(C(F)(F)F)c4)ccc2 C)C3)n(C)n1 ;

Cc1cc(N(C)c2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N (C)C)C5)c(C(F)(F)F)c4)cc c2C)C3)n(C)n1 ;

Cc1ccc(NC(=O)c2cccc(C(F)(F)F)c2)cc1 Nc1cc(CN(C)CCO)nc(NCCCO)c1 ;

N-{3-[7-(3-Dimethylamino-phenylamino)-1-methyl-2-oxo-1 ,4-dihydro-2H-pyrimido[4,5- d]pyrimidin-3-yl]-4-methyl-phenyl}-3-(4-methyl-imidazol-1-yl )-5-trifluoromethyl-benzamide;

N-{3-[7-(3-Amino-phenylamino)-1-methyl-2-oxo-1 ,4-dihydro-2H-pyrimido[4,5-d]pyrimidin-3-yl]-4- methyl-phenyl}-3-trifluoromethyl-benzamide;

N-[4-Methyl-3-(1-methyl-7-methylamino-2,4-dioxo-1 ,4-dihydro-2H-pyrimido[4.5-d]pyrimidin-3-yl)- phenyl]-3-trifluoromethyl-benzamide;

(Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2C1 CCCC1 );

Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2CCO;

CC1 COCC1 (C)n1 nc(-c2cnc3[nH]ccc3c2)c2c(N)ncnc21 ;

CCS(=O)c1 ccc(-n2nc(-c3cnc4[nH]ccc4c3)c3c(N)ncnc32)cc1 ;

Nc1ncnc2c1c(-c1 ccc(Oc3ccccc3) cc1)nn2C1CCNCC1 ;

CN(CC(=O)c1cnc2[nH]ccc2c1)C1CCCC1 ;

Cn 1 c(=O)c(c2cnc3[n H]ccc3c2)cn 1 CC(C) (C)O;

N-[4-[1-(1 ,4-dioxaspiro[4.5]dec-8-yl)-4-(8-oxa-3-azabicyclo[3.2.1]oct- 3-yl)-1 H-pyrazolo[3,4- d]pyrimidin-6-yl]phenyl]-N'-methyl-(WYE-125132; methyl 4-(4-morpholino-1-(1-(pyridin-3-ylmethyl)piperidin-4-yl)-1H- pyrazolo[3,4-d]pyrimidin-6- yl)phenylcarbamate (WYE-687);

6-(1H-indol-5-yl)-4-morpholino-1-(1-(pyridin-3-ylmethyl)p iperidin-4-yl)-1 H-pyrazolo[3,4- d]pyrimidine (WAY-600);

3-(2,4-bis((S)-3-methylmorpholino)pyrido[2,3-d]pyrimidin- 7-yl)-N-methylbenzamide (AZD2014); or

5-(7-(methylsulfonyl)-2-morpholino-6,7-dihydro-5H-pyrrolo [2,3-d]pyrimidin-4-yl)pyrimidin-2- amine (CH5132799)

And in certain examples, wherein the SC-1 compound is (Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4) ccc2C)C3)n(C)n1) or the PP121 compound is

(Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2C1 CCCC1 ).

5. The method of embodiment 2, wherein the AR signaling inhibitor includes surgical removal of the testicles.

6. The method of any of embodiments 2-5, wherein the AR signaling inhibitor includes abiraterone, apalutamide (ARN-509), AZD3514, chlormadinone acetate, darolutamide (ODM- 201), EPI-001, and/, finasteride, galeterone, izonsteride, ketoconazole, megestrol, steroidal antiandrogens, nadolol, N-butylbenzene-sulfonamide, RU58841, Testolone (RAD140), triptophenolide, tulobuterol hydrochloride, and/or turosteride.

7. The method of any of embodiments 2-6, wherein the AR signaling inhibitor includes abiraterone and/or enzalutamide.

8. A composition including a combination therapy including (i) an SC-1 compound, a PP121 compound, a PD166285 dihydrochloride (diHCI) compound, an NCGC00348110 compound, a Dasatinib compound, a PDK1/Akt/Fit DPI compound, a Staurosporine compound, a Cdk 1/2 Inhibitor III compound; and (ii) an androgen receptor (AR) signaling inhibitor, (a “compound” can include “derivatives” disclosed herein).

9. The composition of embodiment 8, wherein the composition includes a SC-1 compound or a PP121 compound, in certain examples selected from (Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4) ccc2C)C3)n(C)n1);

Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N6CC OCC6)CC5)c(C(F)(F)F)c 4)ccc2C)C3)n(C)n1;

C=CC(=O)N1CCCN(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C (F)(F)F)c4)ccc2C)C3)n1 C;

CCc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N(C )C)C5)c(C(F)(F)F)c4)ccc2 C)C3)n(C)n1;

Cc1cc(N(C)c2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N (C)C)C5)c(C(F)(F)F)c4)cc c2C)C3)n(C)n1 ;

Cc1ccc(NC(=O)c2cccc(C(F)(F)F)c2)cc1Nc1cc(CN(C)CCO)nc(NCCC O)c1;

N-{3-[7-(3-Dimethylamino-phenylamino)-1-methyl-2-oxo-1,4- dihydro-2H-pyrimido[4,5- d]pyrimidin-3-yl]-4-methyl-phenyl}-3-(4-methyl-imidazol-1-yl )-5-trifluoromethyl-benzamide;

N-{3-[7-(3-Amino-phenylamino)-1-methyl-2-oxo-1 ,4-dihydro-2H-pyrimido[4,5-d]pyrimidin-3-yl]-4- methyl-phenyl}-3-trifluoromethyl-benzamide;

N-[4-Methyl-3-(1-methyl-7-methylamino-2,4-dioxo-1 ,4-dihydro-2H-pyrimido[4.5-d]pyrimidin-3-yl)- phenyl]-3-trifluoromethyl-benzamide;

(Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2C1 CCCC1 );

Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3d )nn2CC0;

CC1 C0CC1 (C)n1 nc(-c2cnc3[nH]ccc3c2)c2c(N)ncnc21 ;

CCS(=0)c1 ccc(-n2nc(-c3cnc4[nH]ccc4c3)c3c(N)ncnc32)cc1 ;

N d ncnc2c1 c(-c1 ccc(Oc3ccccc3) cd)nn2C1CCNCC1;

CN(CC(=O)c1cnc2[nH]ccc2c1)C1CCCC1;

Cn 1 c(=O)c(c2cnc3[n H]ccc3c2)cn 1 CC(C) (C)O;

N-[4-[1-(1,4-dioxaspiro[4.5]dec-8-yl)-4-(8-oxa-3-azabicyc lo[3.2.1]oct-3-yl)-1 H-pyrazolo[3,4- d]pyrimidin-6-yl]phenyl]-N'-methyl-(WYE-125132; methyl 4-(4-morpholino-1-(1-(pyridin-3-ylmethyl)piperidin-4-yl)-1H- pyrazolo[3,4-d]pyrimidin-6- yl)phenylcarbamate (WYE-687);

6-(1H-indol-5-yl)-4-morpholino-1-(1-(pyridin-3-ylmethyl)p iperidin-4-yl)-1 H-pyrazolo[3,4- d]pyrimidine (WAY-600);

3-(2,4-bis((S)-3-methylmorpholino)pyrido[2,3-d]pyrimidin- 7-yl)-N-methylbenzamide (AZD2014); or

5-(7-(methylsulfonyl)-2-morpholino-6,7-dihydro-5H-pyrrolo [2,3-d]pyrimidin-4-yl)pyrimidin-2- amine (CH5132799)

And in certain examples, wherein the SC-1 compound is (Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4) ccc2C)C3)n(C)n1) or the PP121 compound is

(Nd ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2C1 CCCC1 ).

10. The composition of embodiments 8 or 9, wherein the AR signaling inhibitor includes abiraterone, apalutamide (ARN-509), AZD3514, chlormadinone acetate, darolutamide (ODM- 201), EPI-001, enzalutamide, finasteride, galeterone, izonsteride, ketoconazole, megestrol, steroidal anti-androgens, nadolol, N-butylbenzene-sulfonamide, RU58841 , Testolone (RAD140), triptophenolide, tulobuterol hydrochloride, and/or turosteride.

11. A kit including a combination therapy including (i) an SC-1 compound, a PP121 compound, a PD166285 dihydrochloride (diHCI) compound, an NCGC00348110 compound, a Dasatinib compound, a PDK1/Akt/Fit DPI compound, a Staurosporine compound, a Cdk 1/2 Inhibitor III compound; and (ii) an androgen receptor (AR) signaling inhibitor, (a “compound” can include “derivatives” disclosed herein).

12. The kit of embodiment 11 , wherein the kit includes a SC-1 compound or a PP121 compound in certain examples selected from (Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4) ccc2C)C3)n(C)n1);

Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N6CC OCC6)CC5)c(C(F)(F)F)c 4)ccc2C)C3)n(C)n1;

C=CC(=O)N1CCCN(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C (F)(F)F)c4)ccc2C)C3)n1 C;

CCc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N(C )C)C5)c(C(F)(F)F)c4)ccc2 C)C3)n(C)n1;

Cc1cc(N(C)c2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4ccc(CN5CCC(N (C)C)C5)c(C(F)(F)F)c4)cc c2C)C3)n(C)n1 ;

Cc1ccc(NC(=O)c2cccc(C(F)(F)F)c2)cc1Nc1cc(CN(C)CCO)nc(NCCC O)c1;

N-{3-[7-(3-Dimethylamino-phenylamino)-1-methyl-2-oxo-1,4- dihydro-2H-pyrimido[4,5- d]pyrimidin-3-yl]-4-methyl-phenyl}-3-(4-methyl-imidazol-1-yl )-5-trifluoromethyl-benzamide;

N-{3-[7-(3-Amino-phenylamino)-1-methyl-2-oxo-1,4-dihydro- 2H-pyrimido[4,5-d]pyrimidin-3-yl]-4- methyl-phenyl}-3-trifluoromethyl-benzamide;

N-[4-Methyl-3-(1-methyl-7-methylamino-2,4-dioxo-1 ,4-dihydro-2H-pyrimido[4.5-d]pyrimidin-3-yl)- phenyl]-3-trifluoromethyl-benzamide;

(Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2C1 CCCC1 );

Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2CCO;

CC1 COCC1 (C)n1 nc(-c2cnc3[nH]ccc3c2)c2c(N)ncnc21 ;

CCS(=O)c1 ccc(-n2nc(-c3cnc4[nH]ccc4c3)c3c(N)ncnc32)cc1 ;

Nc1ncnc2c1c(-c1 ccc(Oc3ccccc3) cc1)nn2C1CCNCC1;

CN(CC(=O)c1cnc2[nH]ccc2c1)C1CCCC1;

Cn 1 c(=O)c(c2cnc3[n H]ccc3c2)cn 1 CC(C) (C)O;

N-[4-[1-(1,4-dioxaspiro[4.5]dec-8-yl)-4-(8-oxa-3-azabicyc lo[3.2.1]oct-3-yl)-1 H-pyrazolo[3,4- d]pyrimidin-6-yl]phenyl]-N'-methyl-(WYE-125132; methyl 4-(4-morpholino-1-(1-(pyridin-3-ylmethyl)piperidin-4-yl)-1H- pyrazolo[3,4-d]pyrimidin-6- yl)phenylcarbamate (WYE-687);

6-(1H-indol-5-yl)-4-morpholino-1-(1-(pyridin-3-ylmethyl)p iperidin-4-yl)-1 H-pyrazolo[3,4- d]pyrimidine (WAY-600);

3-(2,4-bis((S)-3-methylmorpholino)pyrido[2,3-d]pyrimidin- 7-yl)-N-methylbenzamide (AZD2014); or

5-(7-(methylsulfonyl)-2-morpholino-6,7-dihydro-5H-pyrrolo [2,3-d]pyrimidin-4-yl)pyrimidin-2- amine (CH5132799)

And in certain examples, wherein the SC-1 compound is (Cc1cc(Nc2ncc3c(n2)N(C)C(=O)N(c2cc(NC(=O)c4cccc(C(F)(F)F)c4) ccc2C)C3)n(C)n1) or the PP121 compound is

(Nc1 ncnc2c1 c(-c1 cnc3[nH]ccc3c1 )nn2C1 CCCC1 ).

13. The kit of embodiments 11 or 12, wherein the AR signaling inhibitor includes abiraterone, apalutamide (ARN-509), AZD3514, chlormadinone acetate, darolutamide (ODM-201), EPI-001, enzalutamide, finasteride, galeterone, izonsteride, ketoconazole, megestrol, steroidal antiandrogens, nadolol, N-butylbenzene-sulfonamide, RU58841, Testolone (RAD140), triptophenolide, tulobuterol hydrochloride, and/or turosteride.

14. The kit of any of embodiments 11-13, wherein the AR signaling inhibitor includes abiraterone and/or enzalutamide.

15. A method of predicting an in vivo therapeutic efficacy against prostate cancer (PCa) of a therapy including a kinase inhibitor selected from SC-1 , PP121, PD166285 dihydrochloride (diHCI), NCGC00348110, Dasatinib, PDK1/Akt/Fit DPI, Staurosporine, Cdk 1/2 Inhibitor III, and/or derivatives thereof, including exposing cells of the PCa to the therapy and determining levels of phosphoproteins selected from p-EGFR Y1173 , p-S6 S235/236 , p- FGFR Y653/654 , p-MET Y1349 , p-IGF1 R Y1131 , p-AKT S473 and/or p-BAD S112 wherein an observed decrease in one or more of the assayed phosphoproteins in relation to a reference level predicts a positive in vivo therapeutic efficacy of the therapy against PCa.

16. The method of embodiment 15, wherein the therapy further includes an androgen receptor (AR) signaling inhibitor.

17. The method of embodiments 15 or 16, further including exposing cells of a normal cell line to the therapy and determining the levels of phosphoproteins selected from p-EGFRY1173, p- S6S235/236, p-FGFRY653/654, p-METY1349, p-IGF1RY1131, p-AKTS473 and/or p-BADS112 to provide the reference level. 18. The method of embodiment any of embodiments 15-17, wherein the cells of the PCa include a PCa cancer cell line.

19. The method of embodiment 18, wherein the PCa cancer cell line includes hormone naive cell lines and/or castration resistant cell lines.

20. The method of embodiment 19, wherein hormone naive cell line includes RWPE-2, LNCaP (e.g., LNCaP42D), LAPC-4, LAPC-9, VCaP, MDA PCa 2a/2b and/or LuCaP (e.g., LuCaP35CR).

21 . The method of embodiment 19 or 20, wherein the castration resistant cell line includes C4-2, C4-2B, 22Rv1 , ARCaP, PC3, and/or DU-145.

22. The method of embodiment 17, wherein the normal cell line is selected from pRNS-1-1 , RWPE-1 , BPH 1 , and/or PIN.

23. The method of any of embodiments 15-18, wherein the PCa is castration-resistant PCa (CRPC).

24. The method of any of embodiments 15-19, wherein the determining utilizes a reverse phase protein array (RPPA).

25. A method of predicting an in vivo therapeutic efficacy against prostate cancer (PCa) of a potential PCa therapeutic including obtaining biological PCa samples for analysis wherein at least one biological PCa sample has been exposed to the potential PCa therapeutic and at least one biological PCa sample has not been exposed to the potential PCa therapeutic; and determining a protein concentration of p-EGFRY1173, p-S6S235/236, p-FGFRY653/654, P-METY1349, p-IGF1 RY1131 , p-AKTS473 and/or p-BADS112 in the biological PCa samples; wherein a decrease in the concentration of p-EGFRY1173, p-S6S235/236, p-FGFRY653/654, p- METY1349, p-IGF1 RY1131 , p-AKTS473 and/or p-BADS112 in the biological PCa sample exposed to the potential PCa therapeutic in comparison to the concentration of the same proteins in the biological PCa sample not exposed to the potential PCa therapeutic predicts the in vivo therapeutic efficacy of the potential therapeutic against PCa.

26. The method of embodiment 25, wherein the biological PCa samples are tissues samples.

27. The method of embodiment 26, wherein the tissues samples include cells, biopsy samples, laser capture micro-dissected cells, and/or biological fluids.

28. The method of embodiments 26 or 27, wherein the tissues samples are fresh, frozen, or fixed.

29. The method of any of embodiments 26-28, further including washing the tissues samples in phosphate buffered saline (PBS).

30. The method of embodiment 29, further including homogenizing the washed tissue samples in a Reverse Phase Protein Array (RPPA) lysis buffer to form tissue sample lysates. 31 . The method of embodiment 30, further including serially diluting the tissue sample lysates and printing the serially diluted tissue sample lysates onto a solid phase.

32. The method of embodiment 31 , wherein the solid phase includes a nitrocellulose-coated slide.

33. The method of embodiments 31 or 32, further including probing the solid phase with a labeled primary antibody alone or a primary antibody followed by an appropriately labeled secondary antibody.

34. The method of embodiment 33, further including scanning the solid phase and quantifying resulting signals.

35. The method of embodiment 34, wherein the resulting signals are fluorescent, chemiluminescent, or colorimetric.

36. The method of any of embodiments 25-35, further including confirming the predicted in vivo efficacy of a tested therapeutic in an animal model of PCa.

37. The method of any of embodiments 25-36, wherein the therapeutic is a combination therapy.

38. The method of embodiment 37, wherein the combination therapy includes a kinase inhibitorand an AR signaling inhibitor.

[0137] (vii) Experimental Example. Results. Functional Kinase Inhibitor Screen Enables the Generation of Predictive Kinome Regularization (KiR) Models of Castration-Resistant Prostate Cancer (CRPC) Growth. To begin the investigation into kinase inhibitor (KI) efficacy in late-stage prostate cancer (PCa), a functional drug screen in four CRPC cell lines (PC3, C4-2B, DLI145, and 22Rv1) was performed (FIG. 1A). These lines have been used extensively in PCa research and account for a variety of biological phenotypes: both C4-2B and PC3 are derived from bone metastases, and C4-2B and 22Rv1 exhibit androgen receptor (AR) activity, while DLI145 and PC3 do not (Russell et al, Prostate Cancer Methods Protoc. 81 , 21-39 (2003); Cunningham et al., J. Biol. Methods 2, 17 (2015)). Each line was seeded in 96-well cell culture plates and treated with 50 kinase inhibitors at 7 doses (plus untreated control) ranging from 10 pM to 0.01 pM, in triplicate for each dose. Images of each well were taken every 2 hours using the IncuCyte ZOOM® (Gottingen, Germany) imaging system for 48-96 hours (FIG. 1 B). To account for varying growth rates between cell lines, the endpoint response was calculated as the net change in confluence (Aconfluence) between the beginning of the treatment and the time point when the control samples within the same plate reached 75% confluence, as quantified by IncuCyte ZOOM® and described previously (FIG. 1C) (Gujral et al., Proc. Natl. Acad. Sci. E3729-E3738 (2017) doi:10.1073/pnas.1703096114). Dose-response data for each cell line-inhibitor combination was generated, and the data was fitted with a three-parameter logistic equation. The resulting curve was interpolated at the doses for which the compounds were profiled (generally 500nM) (FIG. 1 D) (Anastassiadis et al., Nat. Biotechnol. 29, 1039-1045 (2011)). At these doses, 30% of molecules tested in each cell line displayed significant confluence reduction (at least 30% reduction in Aconfluence compared to control), and nearly 30% of all tested compounds displayed no significant effect. C4-2B was the most resilient line, with only 8 out of 43 initially tested compounds reducing Aconfluence by 30% or more. Four inhibitors in the initially tested sets were effective in all four cell lines: Cdk1/2 Inhibitor III, Staurosporine, PDK1/AKT/FLT Dual Pathway Inhibitor, and Dasatinib. The first three of these are very broad-acting kinase inhibitors that tend to inhibit most model systems, while Dasatinib is a SRC family kinase inhibitor that has been previously demonstrated to be effective in PCa (Nam et al., Cancer Res. 65, 9185-9189 (2005); Park et al., Cancer Res. 68, 3323-3333 (2008); Koreckij et al. , Br. J. Cancer 101 , 263-268 (2009)). Following the established pipeline, the resulting inputs to the KiR models were the inhibition profiles of the KIs (FIG. 2A) and the interpolated cell line responses (FIG. 2B). Elastic net regularization with leave-one-out cross validation (LOOCV) was performed to select the optimal regularization penalty (FIG. 2C). Multivariate linear models for each cell line were constructed, with residual kinase activities as the explanatory variables and Aconfluence as the response variable (Zou et al., J. R. Stat. Soc. Ser. B Stat. Methodol. Q7, 301-320 (2005)). The normalized root mean squared error (RMSE) between the predictions and the measured data ranged between 0.0291 for PC3 and 0.09191 for DLI145, giving confidence that the models fit the training set well even with the additional regularization and cross validation (FIG. 3). These KiR models enabled prediction of the response of each cell line to over 400 profiled KIs (FIG. 4A).

[0138] KiR Models Predict Effective Kinase Inhibitors Across CRPC Cell Lines. Having performed functional KI screens and generated KiR models for all four CRPC cell lines, the accuracy of the predictions was evaluated, and the potential effectiveness of the inhibitors were validated by testing 20 additional KIs not included in the training dataset. The measured responses to most inhibitors followed the model predictions, with validation set RMSEs ranging between 0.127 for DLI145 and 0.163 for 22Rv1 (FIGs. 3 and 4B-4E). While several cell-type specific inhibitors were identified, focus was placed on compounds that were effective in all four cell lines. An “effective” compound was defined as one that was observed or predicted (if measured was unavailable) to reduce Aconfluence by at least 30% compared to control at the interpolated dose. At this cutoff, the models displayed the highest accuracy, as determined by proportion of compounds correctly assigned to either above or below this threshold. A total of eight compounds met this criterion (FIG. 5A). Unsupervised hierarchical clustering of these compounds’ target profiles showed they generally fell into four clusters (FIG. 5B). The first cluster included three broad-spectrum inhibitors (CDK 1/2 inhibitor III, Staurosporine, and PDK/AKT/FLT dual pathway inhibitor) (Klnhibition Selectivity Scores (KISS) of -78.24, -83.51 , and -53.51 , respectively (Bello et al., iScience 8, 49- 53 (2018)). Dasatinib had a KISS of 2.89. PP121 (KISS of 24.05) seemed the most unique, and SC-1 (KISS of 37.80) showed slightly greater growth suppression than the molecule it clustered with (PD 166285 dihydrochloride, KISS of 29.02). Notably, treatment with PP121 and SC-1 potently inhibited the growth of all four CRPC cells lines tested, two additional AR-positive CRPC cells lines including LNCaP42D and LuCaP35CR (FIGs. 6A, 6B, and 9A-9D) and viability of organotypic tumor slices (Sivakumar et al., Oncoimmunology 2019. 8, e1670019) prepared from two independent patient-derived xenografts (PDX) CRPC models (FIGs. 10A-10C). Since both PP121 and SC-1 showed potent, dose-dependent growth suppression across all cell lines tested, these compounds (FIGs. 6A, 6B) were chosen for further validation.

[0139] PP121 was developed as a dual inhibitor of tyrosine kinases and phosphoinositide 3- kinases (PI3Ks) (Apsel et al. , Nature chemical biology2008. 4, 691-699), and was shown to inhibit PI3Ks, RTKs, and Src-family kinases. PP121 was reported to block the proliferation of glioblastoma, thyroid, and other tumor cells and inhibit multiple oncogenic or mutated kinases (Che et al., Tumor Biology 2014. 35, 8659-8664). SC-1 , also known as Pluripotin, inhibits Erk1 (MAPK3) and RasGAP (RASA1) and blocks differentiation pathways in embryonic stem cells, although its kinase inhibition profile demonstrates broader activity against many tyrosine kinases (Anastassiadis et al., Nature biotechnology 2011 . 29, 1039-1045; Chen et al., PNAS 2006. 103, 17266-1727). To further characterize the molecular effects of PP121 and SC-1 on prostate cancer cells in vitro, changes in phosphorylation of several signaling proteins were measured in response to low- or high-dose, short-term treatment with PP121 or SC1 using reverse-phase protein array (RPPA), a miniaturized dot blot technique that allows screening of multiple signaling proteins and pathways across many samples (Gujra et al., Oncogene 2013. 32, 3470-3476). PC3 and C4-2B cells were treated in vitro with 5 pM or 0.5 pM of PP121 or SC-1 (plus DMSO control) for 1 hour. RPPA results were quantified as the fluorescent signal intensity from each spot, normalized to the p-actin signal intensity from the same spot, and presented relative to DMSO controls. Phosphorylation changes were assayed in a subset of proteins involved in growth signaling, including EGFR, FGFR, Src family kinases, Akt, and small ribosomal protein S6. These proteins have been reported to play key roles in the growth and progression of late-stage prostate cancer (Bluemn et al., Cancer cell 2017. 32, 474-489. e476; Park et al., Cancer research 2008. 68, 3323- 3333; Drake et al., PNAS 2012. 109, 1643-1648; Goc et al., International journal of oncology 2011. 38, 267-277). The biochemical inhibition profiles of PP121 and SC-1 suggest these compounds effectively inhibit EGFR, FGFR, and Src family kinase signaling (FIG. 6C). The results showed a strong trend towards decreasing growth factor signaling in both cell lines, with PC3 tumors exhibiting more significant responses (FIG. 6D). The higher dose (5 pM) of PP121 significantly reduced EGFR, Akt, and S6 phosphorylation in both cell lines (PC3 cells, p<0.05, p<0.0001 , and p<0.0001 respectively; C4-2B cells, p<0.01 for all signals; FIG. 6D). SC-1 abrogated Src family kinase and Akt phosphorylation at both doses in PC3 cells (5 pM, p<0.0001 for both signals; 0.5 pM, p<0.01 and p<0.001 , respectively), while only significantly affecting EGFR phosphorylation in C4-2B cells (p<0.01 , both doses). In addition, whether PP121 or SC-1 could affect AR signaling was explored. Treatment of C4-2B cells with biologically active doses of either PP121 (1 pM) or SC-1 (1 pM) induced no or modest changes in the expression of AR- targeted genes (Bluemn et al., 2017, Cancer cell 32, 474-489), with few exceptions (FIG. 11 A). In comparison, treatment with enzalutamide (1 pM) significantly decreased expression of all AR- targeted genes suggesting the mechanism of action of both PP121 and SC-1 is likely complementary to AR-targeting agents, thus representing a potential alternative strategy in case of resistance emergence. Overall, inhibition of growth factor signaling in both PC3 and C4-2B upon PP121 and SC-1 treatment provides a molecular basis for how these compounds inhibit CRPC growth, consistent with previous reports of these proteins’ role in driving late-stage prostate cancer (Bluemn et al., 2017, Cancer cell 32, 474-489; Park et al., 2008, Cancer research 68, 3323-3333; Drake et al., 2012, Proceedings of the National Academy of Sciences 109, 1643- 1648; and Goc et al., 2011 , International journal of oncology 38, 267-277).

[0140] PP121 and SC-1 Abrogate Multiple Canonical Growth Factor Signaling Pathways. To further investigate the molecular effects of PP121 and SC-1 on PCa cells in vitro, reverse-phase protein array (RPPA) was performed. RPPA is a miniaturized dot blot technique that allows screening of multiple signaling proteins and pathways across many samples (Espina et al., Methods in Molecular Biology (Clifton N.J.) 441 , 113-128 (2008)). RPPA was previously applied to uncover the state of signaling pathway in tumor lysates and lysates from cell culture models (Gujral et al., Proc. Natl. Acad. Sci. E3729-E3738 (2017) doi:10.1073/pnas.1703096114; Gujral et al., Oncogene 32, 3470-3476 (2013)). Lysates from PC3 or C4-2B cells treated for 1 hour, with 5 different doses of each compound (plus untreated control), was collected and subjected to RPPA. Lysates were printed on nitrocellulose coated slides and probed with previously-validated antibodies to interrogate the signaling landscapes of the treated cells (FIGs. 7A, 7B). Consistent with their broad inhibitory profiles, both PP121 and SC-1 exhibited significant, dose-dependent effects on many proteins involved in canonical growth factor signaling. (FIGs. 7C-7E). Phosphorylation of the receptor tyrosine kinases (RTKs) EGFR Y1173 , FGFR Y653/Y654 , MET Y1349 , and IGF1 R/INSR Y1131/Y1146 decreased in a dose-dependent manner, as did p-SRC Y416 family kinases and Focal Adhesion Kinase (p-FAK/PTK2 Y925 ) (FIGs. 7D-7N). Significant changes in downstream effectors, such as p-ERK1/2 T202/Y204 or p-ABL Y245 , were not observed in the one-hour treatment. An increase in p-MEK1/2 S217/221 during treatment with either compound, and in p-S6 S235/236 with SC-1 treatment in C4-2B, were observed. A strong, dose-dependent decrease in p-AKT S473 under PP121 treatment (but not SC-1 treatment) was also observed. These results align with the biochemical profiles of these molecules and confirm that both PP121 and SC-1 inhibit multiple canonical growth factor proteins and pathways in these CRPC cell lines.

[0141] PP121 and SC-1 inhibit CRPC growth in vivo. To investigate PP121 and SC-1 efficacy in vivo, mice were implanted subcutaneously with luciferase- positive PC3 or C4-2B cells (FIG. 8A). Ten days post-implantation, mice were treated with PP121 (150 mg/kg) or SC-1 (80 mg/kg) 5 times/week, for 2 weeks, per os (taken orally), and tumor growth was monitored over time (FIG. 8B). C4-2B tumor growth was measured by macroscopic bioluminescence. Since PC3 cells grew at a fast rate, generating palpable tumors by day 10 already (leading to a saturated bioluminescent signal), tumor growth was measured with the help of a caliper. The data show that the administration of either KI as a single agent showed a 4-fold reduction in both PC3 and C4-2B tumor growth (SC-1 treated PC3 tumors, p<0.05; all other treatments, p<0.01 ; FIG. 8C). Weighing the tumor mass at the endpoint showed an average of over a 50% decrease in final PC3 tumor weight with either KI treatment (p<0.05) and an average of nearly 80% reduction in final C4-2B tumors weight (p<0.001 ; FIGs. 8D, 8E). Consistently, molecular analyses of the PP121 or SC-1 treated tumors revealed a reduction in phosphorylation of multiple growth factors signaling proteins, including phosphorylation of Src family kinases, EGFR, and FAK compared with vehicle- treated controls (FIG. 11 B). Notably, both compounds were well tolerated by mice, with no significant overall weight loss, as monitored at the endpoint (FIG. 8F). Other parameters evaluated during the experiments included hydration, breathing difficulties, aberrant behavior and movements, and abdominal cavity swelling, with no evidence of any of these complications. Therefore, both PP121 and SC-1 exhibit beneficial therapeutic activity in vivo.

[0142] Methods. Cell Culture. PC3, 22Rv1 , and DLI145 cells were purchased from the American Type Culture Collection (Manassas, VA). C4-2B cells were purchased from ViroMed Laboratories (Minnetonka, MN). PC3, C4-2B, and 22Rv1 cells were cultured in Roswell Park Memorial Institute (RPMI) media supplemented with 10% fetal bovine serum (FBS), 1% penicillin I streptomycin, and 1 % sodium pyruvate. DU 145 cells were cultured in Dulbecco’s Modified Eagle Medium (DMEM) media supplemented with 10% fetal bovine serum and 1% penicillin I streptomycin. Enzalutamide-resistant LNCaP42D cells (derived from LNCaP cells) were cultured in RPMI media supplemented with 10% fetal bovine serum, 1% penicillin I streptomycin, and 10 pM enzalutamide. LuCaP35CR cells were cultured in DMEM media supplemented with 10% fetal bovine serum and 1% penicillin I streptomycin and 1X GlutaMax (Thermo Fisher, Waltham, MA). PC3 dual-color cells expressing nuclear H2B-GFP and cytoplasmic DsRed2 were from Anticancer, Inc. (San Diego, CA); PC3 luciferase expressing cells were provided from the MD Anderson Cancer Center. Cells were cultivated in DMEM (Corning, Corning, NY) supplemented with 10% fetal bovine serum (Sigma-Aldrich, St. Louis, MO) and Penicillin-Streptomycin (100 pg/ml each, Sigma-Adrich). C4-2B cells (from the MD Anderson Cancer Center) expressing nuclear H2B-mCherry and cytoplasmic GFP-LifeAct or luciferase were cultivated in RPMI (Corning, Corning, NY) supplemented with 10% FBS (Sigma-Aldrich, St. Louis, MO), Penicillin- Streptomycin (100 pg/ml each, Sigma-Aldrich) and 1 % HEPES. The identity of cancer cell lines was verified by the “Characterized Cell Line Core Facility”, MD Anderson Cancer Center, through Short Tandem Repeat DNA profiling. Adipose tissue-derived mesenchymal stem cells (hMSCs, ASC52telo telomerase reverse transcriptase immortalized from ATCC) were cultivated in MEM 1X (Corning) supplemented with 17% FBS (Sigma-Aldrich), Penicillin-Streptomycin (100 pg/ml each, Sigma-Aldrich), vitamins (Sigma-Aldrich), non-essential amino acids (Sigma-Aldrich), and sodium pyruvate (Gibco). To induce differentiation towards the osteoblastic lineage, hMSCs were kept in osteogenic induction medium (DMEM 1X, supplemented with 10% fetal bovine serum, penicillin and streptomycin, 50 pg/mL ascorbic acid, 10 mM p-glycerophosphate, 0.1 pM dexamethasone from Sigma-Aldrich).

[0143] Drug Response and Quantification. Unless otherwise specified, all drug response data was collected by plating cells in 96-well tissue culture treated plates, allowing cells to adhere overnight, and treating with small molecules suspended in dimethyl sulfoxide (DMSO). Drug dilutions were made using three-fold serial dilutions in the appropriate media for each cell type, and each dose was administered to the cells in triplicate. Microscopy images were taken with an Incucyte® (Essen Instruments, Ann Arbor, Ml) using the 10X objective. Confluence as percentage of total area covered by cells was quantified using the Incucyte ZOOM® 2016B software.

[0144] Computational Analysis and Modeling. Unless otherwise specified, all dose-response plots, fitted curves, and interpolations were performed using Graphpad Prism version 7.03. Elastic net KiR models, predictions, and cross-validation plots were generated in R Studio version 1.0.153, running R version 3.4.1 “Single Candle”, using custom scripts that employ the “glmnet” package (Friedman et al., Journal of statistical software 2010. 33, 1). The input parameters to the cv. glmnet command were as follows: the residual kinase activities of the KIs in the training set as x-values, the Aconfluence of the corresponding KIs (as fraction of untreated control, ranging between 0 and 1) as y-values, no standardization of x-values, uniform observation weights and variable penalties, system-generated lambda sequence of 300 lambdas and a minimum ratio of 0.005, gaussian (quantitative) family response type with naive algorithm, number of cross- validation folds equal to the number of observations (the length of the response vector; LOOCV), and alpha values ranging from 0.0 to 1.0, incrementing by 0.1 (the command is run iteratively for each value of alpha). Additional calculations and analyses were performed in Microsoft Excel 365. Heatmaps were generated either in R Studio using the “pheatmap” package (Bello and Gujral, /science 2018. 8, 49-53), or in Graphpad Prism.

[0145] Reverse-Phase Protein Array. Protein microarrays were printed and processed as described previously (Phillips et al., Nat. Rev. Urol. 11 , 5-5 (2014); Espina et al., Methods in molecular biology (Clifton N.J.) 441 , 113-128 (2008)). Briefly, PC3 or C4-2B cells were plated in 6-well plates at 500,000 cells/well and allowed to adhere overnight. The following morning, the cells were treated with PP121 or SC-1 at 2 doses (5 pM, and 0.5 pM, plus DMSO control) for 1 hour. Cells were then washed twice in PBS and cell lysates were prepared in 2% sodium dodecyl sulfate (SDS) lysis buffers as described previously (Phillips et al., Nat. Rev. Urol. 11 , 5-5 (2014); Espina et al., Methods in molecular biology (Clifton N.J.) 441 , 113-128 (2008)). Whole cell lysates were printed onto 16-pad nitrocellulose coated slides (Grace Biolabs (Bend, OR), GBL505116) using the Aushon 2470 microarrayer (Aushon BioSystems, Billerca, MA). Each sample was printed in triplicate and slides were stored at - 20°C until processing. RPPA slides were washed with 1 M Tris-HCI (pH 9.0) for 2-4 days to remove SDS. Slides were then washed 2-3 times with PBS for 5 min each and blocked with Odyssey Blocking Buffer (OBB) (Licor (Lincoln, NE), 927- 40000) for one hour at room temperature. After blocking, arrays were incubated with primary antibodies in OBB at 4°C overnight. Th next day, arrays were washed thrice with PBS and incubated with IRDye labeled secondary antibodies in OBB for 1 hr at room temperature. Following incubation, slides were scanned using Licor Odyssey CLX Scanner (LiCOR). Total signal intensity from each spot was quantified using the Array-Pro analyzer software package (Media Cybernetics, Roper Technologies, Lakewood Ranch, FL). The measurement of a specific protein from an individual sample was normalized to total p-actin (Sigma-Aldrich, A1978).

[0146] In vivo samples: tumors were surgically extracted from mice and kept cryopreserved until use. Small pieces of the tumors were homogenized and lysed in 2% SDS lysis buffer using a BioGen PRG200 homogenizer (PRO scientific 01-01200, Cambridge, MA). Samples were then processed as described above.

[0147] Kinase Inhibitor Treatment In Vivo. Animal studies were approved by the controlling Institutional Animal Care and Use Committee and performed according to institutional guidelines for animal care and handling. Athymic nude and NOD-SCID mice were injected with luciferaseexpressing PC3 and C4-2B cells, respectively. Subcutaneous tumors: Mice were injected with 5 x 10 6 PC3 cells/mouse or 6 x 10 6 C4-2B subcutaneously, in 30% Matrigel in PBS (n=5-8 mice/group). After 10 days, mice were randomized and treatment started. The growth of C4-2B tumors was monitored through macroscopic bioluminescence imaging using an MS 200 imaging system (Perkin Elmer, Waltham, MA). The growth of PC3 tumors was monitored measuring tumor volume using a caliper. In vivo treatments: Mice received kinase inhibitor treatment 5 days/week for the following 10 days. Mice were administered a total daily dose of 150 mg/kg of PP121 (Tocris) in 10% DMSO (Sigma-Aldrich), 40% polyethylene glycol (PEG)-300 (Sigma-Aldrich), 5% TWEEN-80 (Sigma-Aldrich) and 45% PBS through oral gavage (100 pl volume/administration, 75 mg/kg twice a day) or 80 mg/kg of SC-1 (Selleckchem, Houston, TX) in 10% 2methyl2pirrolidinone (Sigma-Aldrich) and 90% PEG-300 (Sigma-Aldrich) through oral gavage (100 pl volume/administration, 40 mg/kg twice a day).

[0148] Statistical Analysis. Statistical analysis was performed using GraphPad Prism 7.0 (GraphPad Software, San Diego, CA). To address the difference among different groups, oneway ANOVA was performed followed by Tukey’s honestly significant difference (HSD) post-hoc test. All statistical tests were two-sided, and a P value of less than 0.05 was considered statistically significant. Statistical analysis of RPPA data was done in Graphpad Prism version 7.03 2-way ANOVA with the Holm-Sidak correction for multiple comparisons.

[0149] (viii) Closing Paragraphs. As used herein, a functional derivative of a kinase inhibitor will exhibit a substantially similar effect against PCa spheroid growth when combined with docetaxel as its reference compound when combined with docetaxel according to the experimental methods used to assess PCa spheroid growth in section (vii) of this disclosure.

[0150] Unless otherwise indicated, the practice of the present disclosure can employ conventional techniques of chemistry, organic chemistry, biochemistry, immunology, molecular biology, microbiology, and cell biology. These methods are described in, for example, Harcourt et al., Holt McDougal Modern Chemistry: Student Edition (2018); J. Karty, Organic Chemistry Principles and Mechanisms (2014); Nelson et al., Lehninger Principles of Biochemistry 5th edition (2008); Skoog et al., Fundamentals of Analytical Chemistry (8th Edition); Atkins et al., Atkins' Physical Chemistry (11th Edition); Sambrook, et al. Molecular Cloning: A Laboratory Manual, 2nd Edition (1989); F. M. Ausubel, et al. eds., Current Protocols in Molecular Biology, (1987); the series Methods IN Enzymology (Academic Press, Inc.); M. MacPherson, et al., PCR: A Practical Approach, IRL Press at Oxford University Press (1991); MacPherson et al., eds. PCR 2: Practical Approach, (1995); Harlow and Lane, eds. Antibodies, A Laboratory Manual, (1988); and R. I. Freshney, ed. Animal Cell Culture (1987).

[0151] As will be understood by one of ordinary skill in the art, each embodiment disclosed herein can comprise, consist essentially of or consist of its particular stated element, step, ingredient or component. Thus, the terms “include” or “including” should be interpreted to recite: “comprise, consist of, or consist essentially of.” The transition term “comprise” or “comprises” means has, but is not limited to, and allows for the inclusion of unspecified elements, steps, ingredients, or components, even in major amounts. The transitional phrase “consisting of” excludes any element, step, ingredient or component not specified. The transition phrase “consisting essentially of” limits the scope of the embodiment to the specified elements, steps, ingredients or components and to those that do not materially affect the embodiment. A material effect would cause a statistically significant reduction in the ability to obtain a claimed effect according to a relevant experimental method described in the current disclosure.

[0152] Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and so forth used in the specification and claims are to be understood as being modified in all instances by the term “about.” Accordingly, unless indicated to the contrary, the numerical parameters set forth in the specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained by the present invention. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. When further clarity is required, the term “about” has the meaning reasonably ascribed to it by a person skilled in the art when used in conjunction with a stated numerical value or range, i.e. denoting somewhat more or somewhat less than the stated value or range, to within a range of ±20% of the stated value; ±19% of the stated value; ±18% of the stated value; ±17% of the stated value; ±16% of the stated value; ±15% of the stated value; ±14% of the stated value; ±13% of the stated value; ±12% of the stated value; ±11% of the stated value; ±10% of the stated value; ±9% of the stated value; ±8% of the stated value; ±7% of the stated value; ±6% of the stated value; ±5% of the stated value; ±4% of the stated value; ±3% of the stated value; ±2% of the stated value; or ±1% of the stated value.

[0153] Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the invention are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical value, however, inherently contains certain errors necessarily resulting from the standard deviation found in their respective testing measurements.

[0154] The terms “a,” “an,” “the” and similar referents used in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention otherwise claimed. No language in the specification should be construed as indicating any non-claimed element essential to the practice of the invention.

[0155] Groupings of alternative elements or embodiments of the invention disclosed herein are not to be construed as limitations. Each group member may be referred to and claimed individually or in any combination with other members of the group or other elements found herein. It is anticipated that one or more members of a group may be included in, or deleted from, a group for reasons of convenience and/or patentability. When any such inclusion or deletion occurs, the specification is deemed to contain the group as modified thus fulfilling the written description of all Markush groups used in the appended claims.

[0156] Certain embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Of course, variations on these described embodiments will become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventor expects skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

[0157] Furthermore, numerous references have been made to patents, printed publications, journal articles and other written text throughout this specification (referenced materials herein). Each of the referenced materials are individually incorporated herein by reference in their entirety for their referenced teaching.

[0158] In closing, it is to be understood that the embodiments of the invention disclosed herein are illustrative of the principles of the present invention. Other modifications that may be employed are within the scope of the invention. Thus, by way of example, but not of limitation, alternative configurations of the present invention may be utilized in accordance with the teachings herein. Accordingly, the present invention is not limited to that precisely as shown and described.

[0159] The particulars shown herein are by way of example and for purposes of illustrative discussion of the preferred embodiments of the present invention only and are presented in the cause of providing what is believed to be the most useful and readily understood description of the principles and conceptual aspects of various embodiments of the invention. In this regard, no attempt is made to show structural details of the invention in more detail than is necessary for the fundamental understanding of the invention, the description taken with the drawings and/or examples making apparent to those skilled in the art how the several forms of the invention may be embodied in practice.

[0160] Definitions and explanations used in the present disclosure are meant and intended to be controlling in any future construction unless clearly and unambiguously modified in the examples or when application of the meaning renders any construction meaningless or essentially meaningless. In cases where the construction of the term would render it meaningless or essentially meaningless, the definition should be taken from Webster's Dictionary, 3rd Edition or a dictionary known to those of ordinary skill in the art, such as the Oxford Dictionary of Biochemistry and Molecular Biology (Eds. Attwood T et al., Oxford University Press, Oxford, 2006).