Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMBINATION THERAPY CONSISTING OF ADT AND AN ANDROGEN RECEPTOR VACCINE
Document Type and Number:
WIPO Patent Application WO/2017/214562
Kind Code:
A1
Abstract:
It is disclosed herein methods of treating prostate cancer comprising administering to the subject the combination of androgen deprivation therapy (ADT) and a vaccine directed against the androgen receptor or a fragment of the androgen receptor. Also disclosed are methods of increasing the efficacy of androgen deprivation therapy in a subject with prostate cancer comprising administering to the subject an effective amount of a vaccine against the androgen receptor or fragments thereof wherein the method inhibits, delays or reduces the growth of the prostate cancer and/or the development of castration-resistant prostate cancer.

Inventors:
MCNEEL DOUGLAS (US)
OLSON BRIAN (US)
Application Number:
PCT/US2017/036843
Publication Date:
December 14, 2017
Filing Date:
June 09, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
WISCONSIN ALUMNI RES FOUND (US)
International Classes:
A61K39/00; A61K38/08; A61K38/09
Foreign References:
US20080063654A12008-03-13
US7910565B22011-03-22
US8962590B22015-02-24
Other References:
ARLEN PHILIP M ET AL: "Antiandrogen, vaccine and combination therapy in patients with nonmetastatic hormone refractory prostate cancer", JOURNAL OF UROLOGY, vol. 174, no. 2, August 2005 (2005-08-01), pages 539 - 546, XP027855927, ISSN: 0022-5347
TSE BRIAN WAN-CHI ET AL: "From Bench to Bedside: Immunotherapy for Prostate Cancer", BIOMED RESEARCH INTERNATIONAL, 2014, XP002773771, ISSN: 2314-6133
JEMAL A. ET AL.: "Cancer Statistics", CA CANCER J CLIN, vol. 55, 2005, pages 10 - 30
CA CANCER J CLIN, vol. 53, 2003, pages 5 - 26
OEFELEIN ET AL., J UROL, vol. 158, 1997, pages 1460 - 1465
"Synthetic Vaccines", vol. 1, 1987, CRC PRESS, INC., pages: 83 - 92
REKOSKE, B.T.; H.A. SMITH; B.M. OLSON; B.B. MARICQUE; D.G. MCNEEL.: "PD-1 or PD-L1 Blockade Restores Antitumor Efficacy Following SSX2 Epitope-Modified DNA Vaccine Immunization.", CANCER IMMUNOL RES., vol. 3, 2015, pages 946 - 55
OLSON ET AL., CANCER IMMUNOL, IMMUNOTHER., vol. 33, 2011, pages 639 - 647
SMITH ET AL., CANC. RES., vol. 71, 2011, pages 6785 - 6795
REKOSKE, B.T.; H.A. SMITH; B.M. OLSON; B.B. MARICQUE; D.G. MCNEEL.: "PD-1 or PD-L1 Blockade Restores Antitumor Efficacy Following SSX2 Epitope-Modified DNA Vaccine Immunization", CANCER IMMUNOL RES., vol. 3, 2015, pages 946 - 55
REKOSKE, B.T.; H.A. SMITH; B.M. OLSON; B.B. MARICQUE; D.G. MCNEEL: "PD-1 or PD-L1 Blockade Restores Antitumor Efficacy Following SSX2 Epitope-Modified DNA Vaccine Immunization", CANCER IMMUNOL RES., vol. 3, 2015, pages 946 - 55
COLLURU VT; JOHNSON LE; OLSON BM; MCNEEL DG.: "Preclinical and clinical development of DNA vaccines for prostate cancer", UROL ONCOL., vol. 34, 2016, pages 193 - 204, XP029469394, DOI: doi:10.1016/j.urolonc.2013.09.014
HODGE JW; ARDIANI A; FARSACI B; KWILAS AR; GAMEIRO SR: "The tipping point for combination therapy: cancer vaccines with radiation, chemotherapy, or targeted small molecule inhibitors", SEMIN ONCOL., vol. 39, 2012, pages 323 - 39
ARDIANI A; GAMEIRO SR; KWILAS AR; DONAHUE RN; HODGE JW.: "Androgen deprivation therapy sensitizes prostate cancer cells to T-cell killing through androgen receptor dependent modulation of the apoptotic pathway", ONCOTARGET, vol. 5, 2014, pages 9335 - 48
MERCADER M; BODNER BK; MOSER MT; KWON PS; PARK ES; MANECKE RG ET AL.: "T cell infiltration of the prostate induced by androgen withdrawal in patients with prostate cancer", PROC NATL ACAD SCI USA., vol. 98, 2001, pages 14565 - 70
MORSE MD; MCNEEL DG: "Prostate Cancer Patients Treated with Androgen Deprivation Therapy Develop Persistent Changes in Adaptive Immune Responses", HUMAN IMMUNOLOGY, vol. 71, 2010, pages 496 - 504, XP027006100
MORSE MD; MCNEEL DG: "T cells localized to the androgen-deprived prostate are TH1 and TH17 biased", PROSTATE, vol. 72, 2012, pages 1239 - 47
RODEN AC; MOSER MT; TRI SD; MERCADER M; KUNTZ SM; DONG H ET AL.: "Augmentation of T cell levels and responses induced by androgen deprivation", J IMMUNOL., vol. 173, 2004, pages 6098 - 108
MERCADER M; SENGUPTA S; BODNER BK; MANECKE RG; COSAR EF; MOSER MT ET AL.: "Early effects of pharmacological androgen deprivation in human prostate cancer", BJU INT., vol. 99, 2007, pages 60 - 7
GANNON PO; POISSON AO; DELVOYE N; LAPOINTE R; MES-MASSON AM; SAAD F: "Characterization of the intra-prostatic immune cell infiltration in androgen-deprived prostate cancer patients", JOURNAL OF IMMUNOLOGICAL METHODS, vol. 348, 2009, pages 9 - 17, XP026470608, DOI: doi:10.1016/j.jim.2009.06.004
SHEN YC; KOCHEL C; FRANCICA B; ALME A; NIRSCHL C; NIRSCHL T ET AL.: "Combining androgen deprivation with immune checkpoint blockade delays the development of castration resistance in a murine model of prostate cancer", AACR 106TH ANNUAL MEETING, 2015
AKINS EJ; MOORE ML; TANG S; WILLINGHAM MC; TOOZE JA; DUBEY P: "In situ vaccination combined with androgen ablation and regulatory T-cell depletion reduces castration-resistant tumor burden in prostate-specific pten knockout mice", CANCER RES., vol. 70, 2010, pages 3473 - 82
DRAKE CG; DOODY AD; MIHALYO MA; HUANG CT; KELLEHER E; RAVI S ET AL.: "Androgen ablation mitigates tolerance to a prostate/prostate cancer-restricted antigen", CANCER CELL, vol. 7, 2005, pages 239 - 49
KOH YT; GRAY A; HIGGINS SA; HUBBY B; KAST WM: "Androgen ablation augments prostate cancer vaccine immunogenicity only when applied after immunization", PROSTATE, 2009
ARDIANI A; FARSACI B; ROGERS CJ; PROTTER A; GUO Z; KING TH ET AL.: "Combination therapy with a second-generation androgen receptor antagonist and a metastasis vaccine improves survival in a spontaneous prostate cancer model", CLIN CANCER RES., vol. 19, 2013, pages 6205 - 18, XP002748899, DOI: doi:10.1158/1078-0432.CCR-13-1026
MCNEEL DG; SMITH HA; EICKHOFF JC; LANG JM; STAAB MJ; WILDING G ET AL.: "Phase I trial of tremelimumab in combination with short-term androgen deprivation in patients with PSA-recurrent prostate cancer", CANCER IMMUNOL IMMUNOTHER, vol. 61, 2012, pages 1137 - 47, XP035074383, DOI: doi:10.1007/s00262-011-1193-1
SMALL EJ; LANCE RS; GARDNER TA; KARSH LI; FONG L; MCCOY C ET AL.: "A Randomized Phase II Trial of Sipuleucel-T with Concurrent versus Sequential Abiraterone Acetate plus Prednisone in Metastatic Castration-Resistant Prostate Cancer", CLIN CANCER RES., vol. 21, 2015, pages 3862 - 9
ANTONARAKIS ES; KIBEL AS; YU EY; KARSH LI; ELFIKY A; SHORE ND ET AL.: "Sequencing of Sipuleucel-T and Androgen Deprivation Therapy in Men with Hormone-Sensitive Biochemically-Recurrent Prostate Cancer: A Phase II Randomized Trial", CLIN CANCER RES., 2016
MADAN RA; GULLEY JL; SCHLOM J; STEINBERG SM; LIEWEHR DJ; DAHUT WL ET AL.: "Analysis of overall survival in patients with nonmetastatic castration-resistant prostate cancer treated with vaccine, nilutamide, and combination therapy", CLIN CANCER RES., vol. 14, 2008, pages 4526 - 31, XP055022508, DOI: doi:10.1158/1078-0432.CCR-07-5048
OLSON BM; MCNEEL DG: "CD8+ T cells specific for the androgen receptor are common in patients with prostate cancer and are able to lyse prostate tumor cells", CANCER IMMUNOL IMMUNOTHER., vol. 60, 2011, pages 781 - 92, XP019906893, DOI: doi:10.1007/s00262-011-0987-5
OLSON BM; JOHNSON LE; MCNEEL DG: "The androgen receptor: a biologically relevant vaccine target for the treatment of prostate cancer", CANCER IMMUNOL IMMUNOTHER., vol. 62, 2013, pages 585 - 96
GREGORY CW; HAMIL KG; KIM D; HALL SH; PRETLOW TG; MOHLER JL ET AL.: "Androgen receptor expression in androgen-independent prostate cancer is associated with increased expression of androgen-regulated genes", CANCER RES., vol. 58, 1998, pages 5718 - 24, XP002147396
ELLIS L; LEHET K; RAMAKRISHNAN S; ADELAIYE R; PILI R: "Development of a castrate resistant transplant tumor model of prostate cancer", PROSTATE, vol. 72, 2012, pages 587 - 91
WANG S; GAO J; LEI Q; ROZENGURT N; PRITCHARD C; JIAO J ET AL.: "Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer", CANCER CELL, vol. 4, 2003, pages 209 - 21, XP008118654, DOI: doi:10.1016/S1535-6108(03)00215-0
HORNBERG E; YLITALO EB; CRNALIC S; ANTTI H; STATTIN P; WIDMARK A ET AL.: "Expression of androgen receptor splice variants in prostate cancer bone metastases is associated with castration-resistance and short survival.", PLOS ONE., vol. 6, 2011, pages e19059, XP055205987, DOI: doi:10.1371/journal.pone.0019059
WATSON PA; CHEN YF; BALBAS MD; WONGVIPAT J; SOCCI ND; VIALE A ET AL.: "Constitutively active androgen receptor splice variants expressed in castration-resistant prostate cancer require full-length androgen receptor", PROC NATL ACAD SCI USA., vol. 107, 2010, pages 16759 - 65, XP055266802, DOI: doi:10.1073/pnas.1012443107
SMITH HA; CRONK RJ; LANG JM; MCNEEL DG: "Expression and Immunotherapeutic Targeting of the SSX Family of Cancer-Testis Antigens in Prostate Cancer", CANCER RES., vol. 71, 2011, pages 6785 - 95
FUEGER BJ; CZERNIN J; HILDEBRANDT I; TRAN C; HALPERN BS; STOUT D ET AL.: "Impact of animal handling on the results of 18F-FDG PET studies in mice", J NUCL MED., vol. 47, 2006, pages 999 - 1006
DISSELHORST JA; BROM M; LAVERMAN P; SLUMP CH; BOERMAN OC; OYEN WJ ET AL.: "Image-quality assessment for several positron emitters using the NEMA NU 4-2008 standards in the Siemens Inveon small-animal PET scanner", J NUCL MED., vol. 51, 2010, pages 610 - 7
VALKENBURG KC; WILLIAMS BO: "Mouse models of prostate cancer", PROSTATE CANCER., 2011, pages 895238
WEICHERT JP; CLARK PA; KANDELA IK; VACCARO AM; CLARKE W; LONGINO MA ET AL.: "Alkylphosphocholine analogs for broad-spectrum cancer imaging and therapy", SCIENCE TRANSLATIONAL MEDICINE, vol. 6, 2014, pages 240ra75
GRAFF JN; ALUMKAL JJ; DRAKE CG; THOMAS GV; REDMOND WL; FARHAD M ET AL.: "Early evidence of anti-PD-1 activity in enzalutamide-resistant prostate cancer", ONCOTARGET, 2016
PU Y; XU M; LIANG Y; YANG K; GUO Y; YANG X ET AL.: "Androgen receptor antagonists compromise T cell response against prostate cancer leading to early tumor relapse", SCIENCE TRANSLATIONAL MEDICINE, vol. 8, 2016, pages 333ra47
KISSICK HT; SANDA MG; DUNN LK; PELLEGRINI KL; ON ST; NOEL JK ET AL.: "Androgens alter T-cell immunity by inhibiting T-helper 1 differentiation", PROC NATL ACAD SCI U S A., vol. 111, 2014, pages 9887 - 92
GARNETT CT; PALENA C; CHAKRABORTY M; TSANG KY; SCHLOM J; HODGE JW: "Sublethal irradiation of human tumor cells modulates phenotype resulting in enhanced killing by cytotoxic T lymphocytes", CANCER RES., vol. 64, 2004, pages 7985 - 94
GAMEIRO SR; JAMMEH ML; WATTENBERG MM; TSANG KY; FERRONE S; HODGE JW: "Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing", ONCOTARGET, vol. 5, 2014, pages 403 - 16
CHAKRABORTY M; ABRAMS SI; CAMPHAUSEN K; LIU K; SCOTT T; COLEMAN CN ET AL.: "Irradiation of tumor cells up-regulates Fas and enhances CTL lytic activity and CTL adoptive immunotherapy", J IMMUNOL., vol. 170, 2003, pages 6338 - 47, XP055009178, DOI: doi:10.4049/jimmunol.170.12.6338
CHAKRABORTY M; ABRAMS SI; COLEMAN CN; CAMPHAUSEN K; SCHLOM J; HODGE JW: "External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing", CANCER RES., vol. 64, 2004, pages 4328 - 37
HARRIS TJ; HIPKISS EL; BORZILLARY S; WADA S; GROSSO JF; YEN HR ET AL.: "Radiotherapy augments the immune response to prostate cancer in a time-dependent manner", PROSTATE, vol. 68, 2008, pages 1319 - 29
DEMARIA S; KAWASHIMA N; YANG AM; DEVITT ML; BABB JS; ALLISON JP ET AL.: "Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer", CLIN CANCER RES., vol. 11, 2005, pages 728 - 34
GOLDEN EB; CHHABRA A; CHACHOUA A; ADAMS S; DONACH M; FENTON-KERIMIAN M ET AL.: "Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial", THE LANCET ONCOLOGY, vol. 16, 2015, pages 795 - 803
SLOVIN SF; HIGANO CS; HAMID O; TEJWANI S; HARZSTARK A; ALUMKAL JJ ET AL.: "Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: results from an open-label, multicenter phase I/II study", ANN ONCOL., vol. 24, 2013, pages 1813 - 21, XP002768390
SPRATT DE; EVANS MJ; DAVIS BJ; DORAN MG; LEE MX; SHAH N ET AL.: "Androgen Receptor Upregulation Mediates Radioresistance after Ionizing Radiation", CANCER RES., vol. 75, 2015, pages 4688 - 96
DRAKE CG; SHARMA P; GERRITSEN W: "Metastatic castration-resistant prostate cancer: new therapies, novel combination strategies and implications for immunotherapy", ONCOGENE, vol. 33, 2014, pages 5053 - 64, XP055314313, DOI: doi:10.1038/onc.2013.497
GAN L; CHEN S; WANG Y; WATAHIKI A; BOHRER L; SUN Z ET AL.: "Inhibition of the androgen receptor as a novel mechanism of taxol chemotherapy in prostate cancer", CANCER RES., vol. 69, 2009, pages 8386 - 94, XP009175194, DOI: doi:10.1158/0008-5472.CAN-09-1504
ZHU ML; HORBINSKI CM; GARZOTTO M; QIAN DZ; BEER TM; KYPRIANOU N: "Tubulin-targeting chemotherapy impairs androgen receptor activity in prostate cancer", CANCER RES., vol. 70, 2010, pages 7992 - 8002
SWEENEY CJ; CHEN YH; CARDUCCI M; LIU G; JARRARD DF; EISENBERGER M ET AL.: "Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 373, 2015, pages 737 - 46
ROWE SP; MACURA KJ; MENA E; BLACKFORD AL; NADAL R; ANTONARAKIS ES ET AL.: "PSMA-Based [(18)F]DCFPyL PET/CT Is Superior to Conventional Imaging for Lesion Detection in Patients with Metastatic Prostate Cancer", MOLECULAR IMAGING AND BIOLOGY : MIB : THE OFFICIAL PUBLICATION OF THE ACADEMY OF MOLECULAR IMAGING, vol. 18, 2016, pages 411 - 9, XP035936491, DOI: doi:10.1007/s11307-016-0957-6
BEATTIE BJ; SMITH-JONES PM; JHANWAR YS; SCHODER H; SCHMIDTLEIN CR; MORRIS MJ ET AL.: "Pharmacokinetic assessment of the uptake of 16beta-18F-fluoro-5alpha-dihydrotestosterone (FDHT) in prostate tumors as measured by PET", J NUCL MED., vol. 51, 2010, pages 183 - 92
ANTONARAKIS ES; LU C; WANG H; LUBER B; NAKAZAWA M; ROESER JC ET AL.: "AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer", N ENGL J MED., vol. 371, 2014, pages 1028 - 38, XP055203484, DOI: doi:10.1056/NEJMoa1315815
YU Z; CHEN S; SOWALSKY AG; VOZNESENSKY OS; MOSTAGHEL EA; NELSON PS ET AL.: "Rapid induction of androgen receptor splice variants by androgen deprivation in prostate cancer", CLIN CANCER RES., vol. 20, 2014, pages 1590 - 600
Attorney, Agent or Firm:
BERSON, Bennett, J. (US)
Download PDF:
Claims:
CLAIMS

We Claim:

1. A method of eliciting an anti-tumor response in a subject having prostate cancer comprising:

a. administering to the subject androgen deprivation therapy (ADT); and

b. administering to the subject a recombinant DNA vaccine comprising a polynucleotide operably linked to a transcriptional regulatory element wherein the polynucleotide encodes a androgen receptor or a fragment of the androgen receptor,

wherein the recombinant DNA vaccine is administered in an amount effective to elicit an increased anti-tumor response to the prostate cancer, wherein the combination treatment of ADT and the recombinant DNA vaccine inhibits, delays or reduces the growth of the prostate cancer.

2. The method of claim 1, wherein the DNA vaccine comprises the polynucleotide that encodes a fragment of the androgen receptor ligand binding fragment.

3. The method of claim 1, wherein the DNA vaccine is administered every two weeks to every three months.

4. The method of any one of the preceding claims, wherein the DNA vaccine is administered for at least 6 weeks.

5. The method of any one of the preceding claims, wherein the DNA vaccine is administered at for at least 10 weeks.

6 . The method of any one of claim 3-5, wherein the DNA vaccine is administered biweekly for about 6 to about 10 weeks and subsequently administered quarterly for at least a year.

7. The method of claim 6, wherein the DNA vaccine is administered for at least 18 months.

8. The method of any one of the preceding claims, wherein the vaccine is administered in a dosage of about 10 meg to 1 mg.

9. The method of any one of the preceding claims, wherein the ADT and recombinant vaccine are administered co-currently.

10. The method of any one of claims 1-8, wherein the ADT is administered before administering the recombinant vaccine.

11. The method of any one of claims 1-8, wherein the ADT is administered after administering at least one dosage of the recombinant vaccine.

12. The method any one of the preceding claims, wherein the androgen deprivation therapy comprises administering an effective amount of at least one androgen receptor-pathway targeting drug.

13. The method of claim 12, wherein the at least one androgen receptor-pathway targeting drug is selected from the group consisting of LHRH (or GnRH) analogues, LHRH (or GnRH) antagonists, AR antagonists, androgen synthesis inhibitors, an AR degrader and combinations thereof.

14. The method of any one of the preceding claims, wherein the DNA vaccine is coadministered with an adjuvant.

15. The method of claim 14, wherein the adjuvant is GM-CSF.

16. The method of any one of the preceding claims, wherein the DNA vaccine is administered intradermally, intramuscularly, subcutaneously, intravenously or intra-arterially, with or without electroporation.

17. The method of any one of the preceding claims, wherein the DNA vaccine comprises pTVG-ARLBD.

18. The method of any one of the preceding claims, wherein the DNA vaccine comprises a polynucleotide selected from the group consisting of (i) a mammalian androgen receptor, (ii) a fragment of the androgen receptor that comprises a ligand-binding domain, (iii) a fragment of the ligand-binding domain defined by SEQ ID NO: 9, (iv) a fragment of the ligand-binding domain defined by SEQ ID NO: 10, (v) a fragment of the ligand-binding domain defined by SEQ ID NO: 11, and (vi) a fragment of the ligand-binding domain defined by SEQ ID NO: 12, whereby the DNA vaccine elicits an immune response in the subject against the androgen receptor.

19. The method of any of the preceding claims, wherein the method further comprises administering to the subject an effective amount of a PD-pathway inhibitor.

20. A method of increasing the efficacy of androgen deprivation therapy in a subject with prostate cancer comprising administering to the subject an effective amount of a recombinant DNA vaccine comprising a polynucleotide operably linked to a transcriptional regulatory element wherein the polynucleotide encodes a androgen receptor or a fragment of the androgen receptor, wherein the method inhibits, delays or reduces the growth of the prostate cancer.

21. The method of claim 20, wherein the DNA vaccine comprises pTVG-ARLBD.

22. The method of claim 21, wherein the DNA vaccine comprises a polynucleotide selected from the group consisting of (i) a mammalian androgen receptor, (ii) a fragment of the androgen receptor that comprises a ligand-binding domain, (iii) a fragment of the ligand-binding domain defined by SEQ ID NO: 9, (iv) a fragment of the ligand-binding domain defined by SEQ ID NO: 10, (v) a fragment of the ligand-binding domain defined by SEQ ID NO: 11, and (vi) a fragment of the ligand-binding domain defined by SEQ ID NO: 12, whereby the DNA vaccine elicits an immune reaction in the subject against the androgen receptor.

23. The method of any one of claims 20-22, wherein the method further comprises administering to the subject an effective amount of a PD-pathway inhibitor.

24. The method of claim 19 or 23, wherein the PD-pathway inhibitor is an anti-PD-1 blocking antibody or an anti-PD-Ll antibody.

25. The method of any one of the preceding claims, wherein the prostate cancer is recurrent and/or metastatic prostate cancer.

26. The method of any one of the preceding claims, wherein the prostate cancer is castration resistant prostate cancer (mCRPC).

27. The method of any one of claims 1-26, wherein the prostate cancer is newly diagnosed prostate cancer.

28. A kit for treating prostate cancer comprising androgen deprivation therapy and a recombinant DNA vaccine that elicits an anti-androgen receptor immune response.

29. The kit of claim 27, wherein the recombinant DNA vaccine comprising a polynucleotide operably linked to a transcriptional regulatory element wherein the polynucleotide encodes a androgen receptor or a fragment of the androgen receptor.

30. The kit of claim 27 or 28, wherein the androgen receptor therapy consists of one or more drugs that target the AR pathway by interfering with AR expression or signaling.

31. The kit of any one of claims 28-30, wherein the recombinant DNA vaccine is pTVG- ARLBD.

32. The kit of any one of claims 28-31, wherein the kit further comprises a PD-1 pathway inhibitor.

Description:
COMBINATION THERAPY CONSISTING OF ADT AND AN ANDROGEN RECEPTOR VACCINE

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This Application claims priority to U.S. Provisional Application No. 62/347,646 filed on June 9, 2016, the contents of which is incorporated by reference in its entirety.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT

[0002] This invention was made with government support under CA142608 awarded by the National Institutes of Health. The government has certain rights in the invention.

BACKGROUND OF THE INVENTION

[0003] Prostate cancer is a significant health risk for men over the age of 50, with about 200,000 newly diagnosed cases each year in the United States (Jemal A. et al., Cancer Statistics, 2005 (2005) CA Cancer J Clin, 55: 10-30). It is the most common tumor diagnosed among men and the second leading cause of male cancer-related death in the United States (Jemal et al., Cancer Statistics, 2003 (2003) CA Cancer J Clin, 53 :5-26). Despite advances in screening and early detection, approximately 30% of patients undergoing definitive prostatectomy or ablative radiation therapy will have recurrent disease at 10 years (Oefelein et al., 1997, J Urol, 158: 1460-1465).

[0004] The androgen receptor (AR) is a steroid hormone receptor that plays a crucial role in the development of normal prostate tissue, as well as in the progression of prostate cancer. Patients with metastatic disease are initially treated with androgen deprivation therapy, and androgen deprivation typically is continued indefinitely once a patient has metastatic prostate cancer. Given its use in this context for over 60 years, androgen deprivation (AD) represents one of the first truly "targeted" therapies for a solid tumor, and there are few examples in the current armamentarium of novel cancer-targeting agents with as high a response rate. However, despite the initial response to this treatment in over 80% of patients, castration resistance usually emerges, with a median time of 2- 3 years. Other groups have identified that amplification of the AR is a common, and perhaps the most common, mechanism of resistance to androgen deprivation therapy, with AR-activating mutations, overexpression and/or gene amplification occurring in over 50% of patients with castration-resistant disease. These findings underscore the importance of the AR to prostate cancer, and suggest that AR antigen-loss (a major means of resistance to immunotherapy) is less likely in human prostate cancer. Recent findings have demonstrated that AR-mediated signaling remains active in the majority of castrate-resistant tumors, and hence the preferred nomenclature is now "castrate resistant" rather than "androgen independent" as was formerly used. Because one of the central means of resistance to androgen deprivation is increased AR expression, in some cases through gene amplification, the pharmacological targeting of the AR can paradoxically cause the AR to remain a target in patients with advanced, castrate-resistant disease. Metastatic prostate cancer that is castration-resistant (mCRPC) is the lethal form of this disease. With a median life expectancy of less than 3 years for patients with mCRPC, treatments that can delay the establishment of castration resistance, or treat this stage of disease more effectively, are urgently needed.

[0005] DNA vaccines have recently been added to the arsenal of treatments against prostate cancer. Relative to other vaccine approaches, DNA vaccines are advantageous in being relatively easy and inexpensive to manufacture, and are "off-the-shelf rather than individualized. Animal studies have demonstrated that DNA vaccines lead to antigen presentation through naturally processed MHC class I and II epitopes. Several DNA vaccines are being explored by academic and industry groups as novel treatments for different cancer types, and early stage clinical trials have shown DNA vaccines can augment immune responses and show evidence of clinical responses. Our laboratory has focused recent efforts on the ligand-binding domain of the androgen receptor (AR LBD) as a biologically relevant target protein, critical for the development and progression of prostate cancer. Our laboratory has demonstrated that many patients with prostate cancer have existing humoral and cellular immune responses specific for the AR LBD, and that cytolytic CD8+ T cells specific for the AR LBD can lyse human prostate cancer cells in an MHC class I-restricted fashion. We further demonstrated that a DNA vaccine encoding the AR LBD can elicit epitope-specific cytolytic CD8+ T cells in HLA-A2 transgenic mice, and used these mice as a tumor model system to assess DNA vaccines targeting AR LBD and other antigens. Immunizing tumor-bearing mice with AR LBD DNA vaccine elicited anti-tumor responses and significantly prolonged overall survival of mice.

[0006] There is a need for new and more effective treatment for prostate cancer, especially in treatment or prevention of castrate-resistant disease. BRIEF SUMMARY

[0007] This disclosure is based on the surprising findings that the addition to androgen deprivation therapy of a vaccine directed toward the androgen receptor represses prostate tumor growth and delay onset or progression of metastatic disease.

[0008] Accordingly, in a first aspect, the disclosure encompasses a method for eliciting an antitumor response in a subject having prostate cancer comprising: a) administering to the subject androgen deprivation therapy (ADT or androgen suppression therapy); and b) administering to the subject a vaccine directed toward the androgen receptor, wherein the vaccine are administered in an amount effective to elicit an increased anti-tumor response to the prostate cancer. This results in an inhibition, delay or reduction in growth of prostate cancer or metastatic disease. The Examples demonstrate a significant delay in tumor growth when the vaccine was combined with standard ADT therapy. In one embodiment, the vaccine is a DNA vaccine comprising a polynucleotide that encodes for an androgen receptor or a fragment of the androgen receptor. In another embodiment, the vaccine is a polypeptide vaccine comprising the androgen receptor or fragments thereof.

[0009] Accordingly, in a second aspect, the disclosure encompasses a method for eliciting an antitumor response in a subject having prostate cancer comprising: a) administering to the subject ADT; and b) administering to the subject a recombinant DNA vaccine comprising a polynucleotide operably linked to a transcriptional regulatory element wherein the polynucleotide encodes a androgen receptor or a fragment of the androgen receptor, wherein the ADT and the recombinant DNA vaccine are administered in an amount effective to elicit an increased anti-tumor response to the prostate cancer, and wherein the combination delays, reduced or inhibits prostate cancer cell growth or metastasis.

[0010] In a third aspect, the disclosure encompasses a method of increasing the efficacy of androgen deprivation therapy in a subject with prostate cancer comprising administering to the subject an effective amount of a recombinant DNA vaccine comprising a polynucleotide operably linked to a transcriptional regulatory element wherein the polynucleotide encodes an androgen receptor or a fragment of the androgen receptor, wherein the method inhibits, delays or reduces the growth of the prostate cancer.

[0011] In a fourth aspect, the disclosure encompasses a method of increasing the efficacy of ADT and/or augmenting or increasing the anti-tumor response of ADT treatment by administering an effective amount of a recombinant DNA vaccine and a PD-1 pathway inhibitor in an effective amount to increase the anti-tumor efficacy of ADT and/or increase or augment the anti-tumor response to ADT treatment. This triple combination therapy results in a significant delay in prostate tumor growth and metastasis.

[0012] In a fifth aspect, the disclosure encompasses a kit for treating prostate cancer comprising androgen deprivation therapy and a vaccine that elicits an anti-androgen receptor immune response.

[0013] In yet another aspect, the disclosure encompasses a kit for treating prostate cancer comprising androgen deprivation therapy, a vaccine that elicits an anti-androgen receptor immune response and a PD-1 pathway inhibitor.

BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS

[0014] Figure 1 shows that short- or long-term androgen withdrawal increases AR protein expression in 22Rvl prostate cancer cells.

[0015] Figure 2 shows that long-term androgen deprivation increases full-length AR expression, and short-term androgen deprivation induces a transient increase in AR-V7 expression. Top Panels: relative expression (normalized to β-actin control). Bottom panels: fold induction of expression over long-term FCS-cultured 22rvl cells. * indicates p < 0.05 by Student's t-test.

[0016] Figure 3A-D shows that HLA-A2-expressing, long-term androgen-deprived 22Rvl cells have increased androgen receptor expression. 22Rvl/FCS (fetal calf serum-containing medium, containing androgen) and 22Rvl/CSS (charcoal stripped serum-containing medium, androgen depleted) cell lines (or non-HLA-A2-transfected 22 Rvl controls) were evaluated for AR protein expression by ELISA (Figure 3A), RNA expression by qRT-PCR Figure 3B), HLA-A2 expression Figure 3C), and PD-L1 expression (Figure 3D) by flow cytometry (blue: 22 Rvl/FCS; red: 22Rvl/CSS; black: wild-type 22Rvl; grey: IgG-stained 22Rvl). * indicates p < 0.05 by Student's t-test.

[0017] Figure 4A-B depict intracellular cytokine staining of splenocytes cultured with 22Rvl/FCS (4 A) or 22Rvl/CSS (blue) or 22Rvl/FCS (red) cells (mean fluorescent intensity quantified in inset - * indicates p < 0.05 by Student's t-test.).

[0018] Figure 4C depicts CD69 expression of splenocytes cultured with 22Rvl/CSS (blue) or 22Rvl/FCS (red) cells (mean fluorescent intensity quantified in inset- * indicates p, 0.05 by Student's t -test). [0019] Figure 4D depicts cytotoxicity of splenocytes cultured with 22Rvl/CSS (blue) or 22Rvl/FCS (red) cells.

[0020] Figure 5A shows CD69 expression of splenocytes cultured with 22Rvl/CSS (blue) or 22Rvl/FCS (red) cells (quantified in adjacent bar graph - * indicates p < 0.05 by Student's t-test).

[0021] Figure 5B shows intracellular cytokine staining (IFNy by T Fa) of T-cells cultured with 22Rvl/CSS (right panel) or 22Rvl/FCS (left panel) cells.

[0022] Figure 5C shows frequency of CD8+ T cells that express zero (blue), one (green), two (yellow), three (orange), or four (red) Thl related molecules (ΠΤΝΓγ, TNFa, IL-2, and/or granzyme B).

[0023] Figure 5D shows granzyme B expression by CD8+ T cells following culturing with 22Rvl/FCS (red) or 22Rvl/CSS (blue) cells (quantified in adjacent bar graph - * indicates p < 0.05 by Student's t-test). Figure 5E shows frequency of CD8+ T cells expressing the surface degranulation marker CD107a. Figure 5F shows cytotoxicity of T-cells cultured with 22Rvl/CSS (blue) or 22Rvl/FCS (red) cells.

[0024] Figure 6 A depicts a schematic of the timing of a clinical trial.

[0025] Figure 6B depicts the proposed dosage regimen of the vaccine in a clinical trial.

[0026] Figure 7A and 7B depict androgen-deprived 22Rvl human prostate cancer cells and castrate- resistant MycCaP mouse prostate cancer cells have increased AR expression after ADT treatment.

[0027] Figure 7C depicts increased AR expression in tumors in the MycCaP prostate cancer model in vivo after chemical castration using the GnRH antagonist degarelix.

[0028] Figure 7D depicts T-cell activation by CD69 expression (left panel), IFNy and TNFa cytokine expression (center panel), and cytotoxicity (right panel) that results from AR-specific CD8+ T-cell activation. In all panels, * indicates p<0.05 by Student's t-test.

[0029] Figure 8A depicts splenocytes collected and analyzed for immune responses against MycCaP/CR tumor cells by intracellular cytokine staining (left panel) and AR peptide-stimulated splenocytes from pTVG-AR-immunized mice were measured for ability to lyse MycCap/AS vs. My cCap/CR tumor cells (right panel).

[0030] Figure 8B depicts mice followed for tumor volume. In all panels, * indicates p<0.05 by Student's t-test. [0031] Figure 8C depicts tumor volume post-challenge of control (sham), degarelix + pTVG4 and degarelix + pTVG-AR.

[0032] Figure 9A shows PBMC from patients previously immunized with a PAP -targeting vaccine were cultured in vitro for 72 hours with PAP in the presence of a PD-1 -blocking antibody (or IgG control), and measured for IFNy (left panel) or granzyme B (right panel) secretion by ELISA.

[0033] Figure 9B shows PBMC obtained from patients after immunization were injected into the footpads of NOD/SCID mice with PAP protein and PD-1 blocking antibody (or IgG control), and 24 hours later, footpad swelling was measured.

[0034] Figure 9C shows that PD-L1 expression was measured on circulating tumor cells from patients with persistent PAP-specific Thl-biased immune responses (R) vs non-responders (NR) following immunization with a DNA vaccine targeting PAP. The ratio PD-L1 MFI on post- treatment samples compared to pre-treatment samples is shown. In all panels, * indicates p<0.05 by Student's t-test.

[0035] Figure 10A shows that CD8+ T cells from MycCaP tumor-bearing animals treated with ADT and immunized with pTVG-AR have elevated PD-1 expression.

[0036] Figure 10B shows that recurrent tumors had elevated PD-L1 expression.

[0037] Figure IOC depicts delay in tumor growth in mice with AR-targeted immunization combined along with a PD-1 blocking antibody compared to immunization with pTVG-AR alone.

[0038] Figure 10D shows that combining AD with AR-directed immunization and PD-1 blockade further delayed tumor growth.

[0039] Figure 11A shows prostate cell lines (immortalized human epithelial lines: RWPE-1 and PrEC-E6; androgen-independent prostate cancer cell lines: DU-145 and PC-3; and androgen- dependent prostate cancer cell lines: LNCaP and 22Rvl) cultured in either androgen-replete (FCS) or androgen-deprived (CSS) medium for one to seven days (ld-7d) or for greater than six months (long- term: LT)analyzed for androgen receptor protein expression by quantitative ELISA {panel A).

[0040] Figure 11B shows prostate cell lines cultured long-term FCS (light grey) or CSS (dark grey). Cultured cell lines were analyzed for AR expression by intracellular staining using antibodies specific for the ligand-binding domain (top panels) or amino-terminal domain (lower panels).

[0041] Figure 11C shows quantified amplitude of AR expression in cultured prostate cell lines. [0042] Figure 1 ID shows frequency of AR+ cells in cultured prostate cell lines.

[0043] Figure HE shows RNA quantified from 22RV1/CSS cells cultured for different periods of time analyzed for the presence of full-length (dark grey) or AR-V7 (light grey) AR transcripts. In all panels, * indicates p<0.05 by student's t-test and data is representative of at least two independent experiments.

[0044] Figure 1 IF shows phenotypic validation of HLA-A2-transfected 22Rvl/FCS and 22Rvl/CSS cells. 22Rvl cells cultured for greater than six months in androgen replete (22Rvl/FCS) or androgen deprived (22Rvl/CSS) medium were transfected with a lentiviral vector encoding ULA- A2. HLA-A2-expressing cells were then sorted by fluorescence-activated cell sorting, and expanded lines were evaluated for AR protein by quantitative ELISA. In all panels, * indicates p<0.05 by Student's t-test.

[0045] Figure 11 G shows expanded lines of Figure 1 IF evaluated for AR protein by qRT-PCR.

[0046] Figure 11H shows cells of Figure 11G analyzed for the expression of HLA-A2.

[0047] Figure 111 shows cells of Figure 11G analyzed for the checkpoint ligand PD-L1 by flow cytometry.

[0048] Figure 12A shows AR-specific T cells have increased recognition and lysis of androgen- deprived tumor cells. AR805-specific human T-cell cultures incubated with HLA-A2-expressing

22Rvl/FCS or 22Rvl/CSS cells were measured for surface expression of CD69.

[0049] Figure 12B show AR805-specific human T-cell cultures incubated with HLA-A2-expressing

22Rvl/FCS or 22Rvl/CSS cells and measured for intracellular cytokine expression of IFNy and/or

TNFa.

[0050] Figure 12C demonstrates polyfunctional cytokine expression quantified in AR805-specific human T -cell cultures incubated with HLA-A2-expressiong 22Rvi/FCS or 22Rvl/CSS cells.

[0051] Figure 12D shows cytolytic and degranulation activity of AR-specific T cells measured by intracellular granzyme B expression.

[0052] Figure 12E shows cytolytic and degranulation activity of AR-specific T cells measured by surface CD 107a expression.

[0053] Figure 12F shows tumor cell cytotoxicity of AR-specific T cells. [0054] Figure 12G demonstrates AR-specific T cells obtained following peptide-immunization have increased recognition and lysis of androgen-deprived prostate tumor cells. Splenocytes from AR811 peptide-immunized HLA-A2 transgenic (HHDII-DR1) mice were co-cultured with HLA-A2- expressing 22Rvl/FCS or 22Rvl/CSS cells, and measured for intracellular cytokine expression of IFNg and TNFa.

[0055] Figure 12H demonstrates AR-specific T-cells of Figure 12G express CD69 on their surface.

[0056] Figure 121 demonstrates AR- specific T cells of Figure 12G are cytotoxic.

[0057] Figure 13 A demonstrates that androgen deprivation increases AR expression in Myc-CaP tumor cells in vitro and in vivo. Androgen-sensitive (Myc-CaP/AS) and castrate-resistant (Myc-

CaP/CR) cells were analyzed for AR protein expression by quantitative ELISA.

[0058] Figure 13B shows androgen-sensitive (Myc-CaP/AS) and castrate-resistant (Myc-CaP/CR) cells analyzed for AR protein expression by intracellular staining (quantified for amplitude and frequency of expression in side panels).

[0059] Figure 13C shows RNA samples from Myc-CaP/AS and Myc-CaP/CR cells analyzed by quantitative PCR for expression of full-length or splice variants mAR V2 or mAR V4.

[0060] Figure 13D shows Myc-CaP/AS tumor-bearing FVB mice with palpable tumors were treated with degarelix (n=4) or sham-treatment (n=3) and followed for tumor growth. Results are representative of two independent studies.

[0061] Figure 13E shows recurrent tumors collected and analyzed for AR expression by intracellular staining using an antibody directed against the ligand-binding domain (amplitude and frequency quantified in side panels). In all panels, * indicates p<0.05 by student's t-test.

[0062] Figure 14A shows androgen deprivation combined with immunization using pTVG-AR delayed the recurrence of castrate-resistant Myc-CaP tumors. Myc-CaP/AS tumor-bearing mice with palpable tumors were given a sham-treatment (n=3) or degarelix along with biweekly immunization with pTVG-AR (n=5) or empty vector (n=5) and followed for tumor growth (tumor volumes). Results are representative of three independent studies.

[0063] Figure 14B shows the Kaplan Meier curve for Figure 14A. Results are representative of three independent studies. [0064] Figure 14C shows splenocytes from androgen-deprived animals immunized with pTVG4 or pTVG-AR cultured with Myc-CaP/AS or Myc-CaP/CR cells and assessed for CD4+ and CD8+ T- cell intracellular cytokine expression (with polyfunctional expression quantified in side pie charts).

[0065] Figure 14D shows splenocytes from androgen-deprived animals immunized with pTVG4 or pTVG-AR cultured with Myc-CaP/AS or Myc-CaP/CR cells and assessed for cytotoxicity.

[0066] Figure 14E demonstrates immunization with pTVG-AR delays tumor growth in the presence or absence of ADT, and results in increased tumor-infiltrating T cells. Myc-CaP/AS tumor-bearing mice were given degarelix or sham treatments, then immunized with either pTVG-AR or pTVG4 control, and assayed for tumor growth (tumor volumes).

[0067] Figure 14F shows Kaplan Meier plots for mice of Figure 14E.

[0068] Figure 14G shows analysis of tumor collected from Figure 14E for the frequency of infiltrating T cells by IHC. * indicates p<0.05 by Mann-Whitney U test.

[0069] Figure 15A shows androgen deprivation increases AR expression in PTEN-deficient tumors, and immunization with pTVG-AR delays the development of castrate-resistant prostate tumors in Pten-/- mice. PTEN-CaP8 tumor cells cultured in androgen-replete (FCS) or androgen-deprived (CSS) medium were analyzed for AR expression by intracellular staining (quantified for amplitude and expression in side panels). * indicates p<0.05 by student's t-test.

[0070] Figure 15B shows the results of PTEN-CaP8 tumor cells cultured in androgen-replete (FCS) or androgen-deprived (CSS) medium analyzed for AR expression by quantitative ELISA. * indicates p<0.05 by student's t-test.

[0071] Figure 15C shows the results of twenty -week old PbCre + PTEN^ mice given a sham treatment (n=9), or degarelix along with biweekly immunization with pTVG4 (n=13) or pTVG-AR (n=13), for five months. One week prior to initiation and completion of treatment, animals were administered 124 I-CLR1404 and PET/CT scanned 96hr post intravenous injection (PET/CT images pre- and post-treatment). Signal greater than sixty percent of the max PET signal was used to calculate the mean and max percent injected dose (%ID/gtissue) for tumor, and was normalized to background muscle uptake.

[0072] Figure 15D shows the pre-treatment mean 124 I-CLR1404 uptake for randomization of treatment groups. * indicates p<0.05 by Mann-Whitney U test. [0073] Figure 15E shows the pre-treatment maximum 124 I-CLR1404 uptake for randomization of treatment groups. * indicates p<0.05 by Mann-Whitney U test.

[0074] Figure 15F shows changes in %ID/gmean pre- to post-treatment calculated (mean values shown by solid horizontal bars). * indicates p<0.05 by Mann-Whitney U test.

[0075] Figure 15G shows %ID/gmean pre- to post-treatment calculated (mean values shown by solid horizontal bars). * indicates p<0.05 by Mann-Whitney U test.

[0076] Figure 15H shows genitourinary complex masses collected during necropsy and analyzed. * indicates p<0.05 by Mann-Whitney U test.

[0077] Figure 16 is a schematic of a treatment using an anti-androgen therapy leuprolide in combination with a DNA vaccine and an anti-PDl therapy.

DETAILED DESCRIPTION

[0078] This disclosure provides compositions and methods related to combination therapies for treating prostate cancer. The specific combination of androgen therapy and vaccine against the androgen receptor (for example a DNA vaccine) unexpectedly and synergistically improves the efficacy ADT for treatment of prostate cancer, including metastatic or castrate-resistant disease. The combination therapy results in a significant decrease in tumor growth as compared to ADT alone.

[0079] In one embodiment, the method for eliciting an anti-tumor response in a subject having prostate cancer comprising: administering to the subject ADT and administering to the subject a vaccine directed toward the androgen receptor, wherein the vaccine is administered in an amount effective to elicit an increased anti-tumor response to the prostate cancer compared to ADT treatment alone. This increased anti-tumor response to prostate cancer results in the reduction, inhibition or delay of prostate cancer cell growth and/or metastasis, prolonging subject survival. In one embodiment, the anti-tumor response of the combination treatment results in a significant delay in tumor cell growth as compared to ADT treatment alone. In one embodiment, the vaccine is a DNA vaccine comprising a polynucleotide that encodes for an androgen receptor or a fragment of the androgen receptor. In another embodiment, the vaccine is a polypeptide vaccine comprising the androgen receptor or fragment thereof.

[0080] In one embodiment, the disclosure provides a method of eliciting an anti-tumor response in a subject having prostate cancer comprising: a. administering to the subject androgen deprivation therapy (ADT); and b. administering to the subject a recombinant DNA vaccine comprising a polynucleotide operably linked to a transcriptional regulatory element wherein the polynucleotide encodes a androgen receptor or a fragment of the androgen receptor, wherein the ADT and the recombinant DNA vaccine are administered in an amount effective to elicit an anti-tumor response to the prostate cancer.

[0081] Anti-tumor response in a subject includes the reducing, repressing, delaying or preventing tumor growth, reduction of tumor volume, and/or preventing, repressing, delaying or reducing metastasis of the tumor. Anti-tumor response includes the reduction of the number of tumor cells within the subject. In some embodiments, anti -tumor response includes an immune response to tumor cells expressing the androgen receptor, for example, a cytotoxic immune reaction against cells expressing androgen receptor. For example, an anti-tumor response may include lysis of tumor cells by AR-specific CD8+ T cells. Preferably, cellular immune reactions against androgen receptor are induced, with or without humoral immune reactions.

[0082] Androgen deprivation therapies (ADT) are therapies that reduce the levels of androgen hormones, or interfere with androgen receptor function/signaling, for example by use of androgen receptor-pathway targeting (e.g. antiandrogens) or chemical castration. Androgen deprivation therapy includes administering an effective amount of at least one androgen receptor pathway- targeting drug. Suitable drugs are known to one skilled in the art and include, but are not limited to, LHRH (or GnRH) analogues (agonists), LHRH (or GnRH) antagonists, AR antagonists, androgen synthesis inhibitors, other AR degrading or blocking agents, and combinations thereof. Suitable ADT include treatment with one or more of the following drugs:

[0083] (a) AR antagonists, including, but not limited to, flutamide, nilutamide, bicalutamide, enzalutamide, apalutamide, cyproterone acetate, megestrol acetate, chlormadinone acetate, spironolactone, canrenone, drospirenone, topilutamide (fluridil), cimetidine;

[0084] (b) androgen synthesis inhibitors, including, but not limited to, (i) 5a-reductase inhibitors, which include the non-limiting examples of finasteride, dutasteride, alfatradiol, and saw palmetto extract, (ii) CYP17A1 (17a-hydroxylase, 17,20-lyase) inhibitors, which include the non-limiting examples cyproterone acetate, spironolactone, danazol, gestrinone, ketoconazole, abiraterone acetate;

(iii) 3 β-Hydroxy steroid dehydrogenase inhibitors, which include the non-limiting examples danazol, gestrinone, abiraterone acetate; (iv) 17P-Hydroxy steroid dehydrogenase inhibitors, which include the non-limiting examples danazol, simvastatin; (v) CYPl lAl (cholesterol side-chain cleavage enzyme) inhibitors, which include the non-limiting examples aminoglutethimide, danazol; and (vi) HMG-CoA reductase inhibitors, which include the non-limiting example statins (e.g., atorvastatin, simvastatin);

[0085] (c) antigonadotropins including (i) progestogens, such as the non-limiting examples including progesterone, cyproterone acetate, medroxyprogesterone acetate, megestrol acetate, chlormadinone acetate, spironolactone, drospirenone; (ii) estrogens including the non-limiting examples of estradiol, ethinyl estradiol, diethylstilbestrol, conjugated equine estrogens; (iii) GnRH analogues, for example, GnRH agonists, including non-limiting examples buserelin, deslorelin, gonadorelin, goserelin, histrelin, leuprorelin, nafarelin, triptorelin; GnRH antagonists, including non- limiting examples abarelix, cetrorelix, degarelix, ganirelix; and combinations thereof. For example, suitable ADT treatment include, but are not limited to, AR antagonists bicalutamide (Casodex, AstraZeneca®), apalutamide (ARN-509, Janssen), or enzalutamide (Xtandi, Astellas ®), GnRH antanogist degarelix (Firmagon, Ferring Pharmaceuticals ®), AR degrading agents such as galeterone (Tokai) and the like. In some embodiments, one or more suitable ADT drugs can be used, such as LHRH agonists or antagonists in combination with AR antagonists or degrading agents.

[0086] In some embodiments, androgen deprivation therapy (ADT) results in overexpression of the androgen receptor (AR) in the majority of prostate cancer patients' tumors. While this overexpression can promote the development of tumor escape variants, as the present disclosure discusses, it also causes prostate tumor cells to be more susceptible to lysis by AR-specific CD8+ T cells. The Examples demonstrate in vitro that increased AR expression in prostate tumor cells can occur following standard androgen deprivation, or following treatment with commercially available AR antagonists bicalutamide (Casodex, AstraZeneca ®) or enzalutamide (Xtandi, Astellas ®). Additionally, the Examples demonstrate in vivo that treatment with degarelix (Firmagon, Ferring Pharmaceuticals ®), a GnRH antanogist used clinically for androgen deprivation therapy, results in increased AR expression in the MycCaP prostate tumor model. Significantly, while treatment with degarelix alone resulted in a delay in tumor growth, combining this treatment with a vaccine targeting the androgen receptor (pTVG-AR) resulted in a significant delay in tumor growth compared with degarelix treatment alone. Additionally, in animals that develop recurrent disease following combination degarelix and pTVG-AR treatment, tumors have a significant decrease in AR expression, suggesting this may be a biomarker of treatment failure. [0087] Patients undergoing androgen deprivation using a variety of AR-targeting pharmaceutical agents may be immunized with DNA vaccine against the androgen receptor, for example, pTVG-AR, improving the response to these standard therapies. Improved response includes the inhibition or reduction in tumor cell growth or metastasis and/or delay in tumor cell growth or metastasis.

[0088] The ADT may be delivered by any suitable dosages and schedule known by one skilled in the art. For example, a non-limiting example includes an LHRH agonist alone or in combination with an antiandrogen (e.g., bicalutamide or enzalutamide). Another non-limiting example is the combination of LHRH agonist and abiraterone and apalutamide.

[0089] Non-limiting examples of suitable dosages for LHRH agonists include, for example, Leuprolide 20-25 mg (e.g. 22.5 mg) IM every three months; and/or Goserelin LHRH agonists are typically about 9-12 mg (e.g. 10.8 mg) sc every three months. Non-limiting examples of dosages for LHRH antagonist include Degarelix 240 mg sc once as first dose, and the 80 mg sc every 28 days subsequently; Abiraterone: 1000 mg by mouth daily; Apalutamide: 240 mg by mouth daily; Bicalutamide: 50 mg by mouth daily; and/or Enzalutamide: 160 mg by mouth daily .

[0090] In some embodiments, it is contemplated that the dosage or regimen of ADT which is known and understood in the art could possibly be varied from conventional parameters (e.g. reduced amount or frequency of dosage) when combined with the vaccine as described in the present invention.

[0091] The vaccine used in the methods of this disclosure may be a recombinant DNA vaccine that encodes the androgen receptor or fragments thereof or a peptide vaccine comprising a polypeptide androgen receptor or fragments thereof. The recombinant DNA vaccine used in the methods of this disclosure may comprise a polynucleotide that encodes a mammalian androgen receptor, the ligand binding domain of the androgen receptor, or a fragment of the androgen receptor. Suitable recombinant DNA vaccine for use is disclosed in U.S. Patents 7,910,565 and 8,962,590, entitled

"Prostate cancer vaccine," which are incorporated by reference in their entireties. In some embodiments, the recombinant DNA vaccine comprises a polynucleotide that encodes a mammalian androgen receptor, a fragment of the mammalian androgen receptor that comprises the ligand- binding domain, or certain fragments of the ligand-binding domain. The plasmid DNA vaccines, when directly introduced into subjects such as humans in vivo, induce the expression of encoded polypeptides within the subject, and cause the subjects' immune system to become reactive against the polypeptides. The vaccines may be any polynucleotides that are capable of eliciting immune responses to an encoded polypeptide.

[0092] In some embodiments, the DNA vaccine comprises pTVG-AR (pTVG-AR or pTVG- ARLBD are the same vector and used herein interchangeably). pTVG-AR is a vector comprising the coding sequence for the ligand-binding domain of the human androgen receptor gene inserted into the pTVG4 vector to create the immunization vector pTVG-AR, as disclosed in U.S. Patent 7,910,565, incorporated by reference in its entirety.

[0093] The vaccine can be administered into a subject to elicit an immune response against androgen receptor in the subject. An "effective amount" or an "immunologically effective amount" means that the administration of that amount to a subject, either in a single dose or as part of a series, is effective for inducing an immune reaction against the androgen receptor (and, therefore, against cells expressing the androgen receptor). Further, an "effective amount" as contemplated in the present invention is an amount of vaccine that augments or increases the anti-tumor efficacy of ADT, resulting a delay or inhibitor of prostate tumor growth and metastasis.

[0094] Androgen receptor genes are known and have been cloned from many species. For example, the human, mouse, rat, dog, chimpanzee, macaque, and lemur androgen receptor mRNA that correspond to cDNA along with amino acid sequences can be found at GenBank Accession Nos. M_000044 (cDNA-SEQ ID NO: l and amino acid sequence-SEQ ID NO:2), M_013476 (cDNA-

SEQ ID NO:3 and amino acid sequence-SEQ ID NO:4), M_012502 (cDNA-SEQ ID NO:5 and amino acid sequence-SEQ ID NO:6), M_001003053, M_001009012, U94179, and U94178, respectively. Androgen receptor genes from other species are also known. These species include but are not limited to Sus scrofa, Astatotilapia burtoni, Gallus gallus, Kryptolebias marmoratus,

Alligator mississippiensis, Leucoraja erinacea, Haplochromis burtoni, Pimephales promelas,

Dicentrarchus labrax, Gambusia affinis, Micropogonias undulates, Oryzias latipes, Acanthopagrus schlegelii, Rana catesbeiana, Crocuta crocuta, Eulemur fulvus collaris, and Anguilla japonica (see

GenBank Accession Nos. NM_214314 (or AF161717), AY082342, NM_001040090, DQ339105,

AB186356, DQ382340, AF121257, AY727529, AY647256, AB099303, AY701761, AB076399,

AY219702, AY324231, AY128705, U94178, and AB023960, respectively). The ligand-binding domains of androgen receptors are well known in the art. For the purpose of the present invention, the ligand-binding domain of the human androgen receptor refers to a polypeptide that starts at any amino acid from amino acid positions 651 to 681 and ends at any amino acid from amino acid positions 900 to 920. For example, human androgen receptor or a fragment of the human androgen receptor that comprises amino acids 681-900 as well as DNA vaccines containing a polynucleotide encoding the above are suitable vaccines. As will be readily recognized by one of ordinary skill in the art, any DNA sequence that encodes a ligand-binding domain or a larger fragment of an androgen receptor including the full-length receptor from one of the above species as well as other animals is suitable for the present invention.

[0095] Pharmaceutically acceptable carriers are well known to those of ordinary skill in the art (Arnon, R. (Ed.) Synthetic Vaccines 1:83-92, CRC Press, Inc., Boca Raton, Fla., 1987). They include liquid media suitable for use as vehicles to introduce the peptide or polynucleotide into a patient but should not in themselves induce the production of antibodies harmful to the individual receiving the composition. An example of such liquid media is saline solution.

[0096] Moreover, the vaccine may also contain an adjuvant for stimulating the immune response and thereby enhancing the effect of the vaccine. Suitable adjuvants are known in the art and include, but are not limited to, GM-CSF, Montanide, or saponin-derivative adjuvants.

[0097] According to another embodiment, the DNA vaccine comprises a polynucleotide operatively linked to a transcriptional regulatory element (e.g., a promoter such as a heterologous promoter) wherein the polynucleotide encodes a member selected from (i) a mammalian androgen receptor (e.g., a human androgen receptor), (ii) a fragment of the androgen receptor that comprises the ligand- binding domain, (iii) a fragment of the ligand-binding domain defined by SEQ ID NO: 9 (LLLFSIIPV, amino acids 811-819 of SEQ ID NO:2); (iv) a fragment of the ligand-binding domain defined by SEQ ID NO: 10 (RMLYFAPDLV, amino acids 761-770 of SEQ ID NO:2), (v) a fragment of the ligand-binding domain defined by SEQ ID NO: 11 (FLCMKALLL, amino acids 805-813 of SEQ ID NO:2), and (vi) a fragment of the ligand-binding domain defined by SEQ ID NO: 12 (QLTKLLDSV, amino acids 859-867 of SEQ ID NO:2), wherein administration of said vaccine to a subject induces a cytotoxic immune reaction against cells expressing androgen receptor.

[0098] The DNA vaccine may comprise the polynucleotide directly linked to a transcriptional regulatory element that promotes the expression of the protein (e.g., androgen receptor or fragment thereof) within cells of the subject. Suitable transcriptional regulatory element (e.g., a promoter such as a heterologous promoter) are known in the art and include, but are not limited to, CMV promoter, Rous sarcoma virus (RSV) promoter, the simian virus 40 (SV40) promoter, the human elongation factor- la (EF-la) promoter, and the human ubiquitin C (UbC) promoter, among others.

[0099] The vaccine is suitably administered by intradermal, intramuscular, subcutaneous, or intravascular (including intravenous and intraarterial) administration to a mammal such as a human. In another aspect, the DNA vaccine is suitable for administration by muscle or skin electroporation to increase uptake of the DNA at the site of immunization.

[00100] The vaccines can be used in a prime-boost strategy in connection with the ADT therapy to induce robust and long-lasting immune response to androgen receptor. Priming and boosting vaccination protocols based on repeated injections of the same antigenic construct are well known and result in strong CTL responses. In general, the first dose may not produce protective immunity, but only "primes" the immune system. A protective immune response develops after the second or third dose.

[00101] The vaccine described herein may be provided in an effective amount to augment or increase the efficacy of ADT treatment, which can be seen by a delay, reduction or inhibition of prostate tumor cell growth or metastasis.

[00102] In one embodiment, the vaccines may be used in a conventional prime-boost strategy, in which the same antigen is administered to the animal in multiple doses. In a preferred embodiment, the DNA or peptide vaccine is used in one or more inoculations. These boosts are performed according to conventional techniques, and can be further optimized empirically in terms of schedule of administration, route of administration, choice of adjuvant, dose, and potential sequence when administered with another vaccine, therapy or homologous vaccine.

[00103] In one embodiment, the vaccine is administered every two weeks to every three months. In some embodiments, the vaccine is administered for at least 6 weeks, alternatively for at least 10 weeks, alternatively for at least 15 weeks, alternatively for at least 20 weeks, alternatively for at least

25 weeks, alternatively for at least 30 weeks, alternatively for at least 35 weeks, alternatively for at least 40 weeks, alternatively for at least 45 weeks, alternatively at least 48 weeks, alternatively for at least 50 weeks, alternatively for at least a year, alternatively for at least 18 months, alternatively for at least 20 months and can include any time in between (for example, 16 weeks 17 weeks, 18 weeks,

19 weeks, 24 weeks, etc). In some embodiments, the vaccine is administered biweekly for about 6 to about 14 weeks and subsequently administered quarterly for at least a year. In some embodiments, the vaccine is administered biweekly for about 6 to about 14 weeks and subsequently administered quarterly (i.e. every three months) for at least 18 months.

[00104] Suitable dosages of the DNA vaccine are known in the art, and include, but are not limited to, about 10 meg to about 1 mg of DNA per dosage.

[00105] In some embodiments, the ADT and recombinant vaccine are administered concurrently. In other embodiments, the subject is treated with ADT and subsequently treated with the recombinant vaccine. The time period between the ADT and recombinant vaccine administration may be a short duration (e.g., hours or days) or may be of a longer duration (e.g. weeks or months). In some embodiments, the term concurrently means that the two components are administered in close timing to each other (e.g. within hours or on the same day), but may be administered by different routes of administration (e.g. ADT orally and vaccine by injection). In some embodiments, the administration are separate, e.g. separated by hours or days in between the vaccine and the ADT. In some embodiments, the vaccine and the ADT are administered over the same time period but using different regimens that require administration on different days. Suitable regimens are discussed more herein, for example, a regimen where the vaccine is administered for a period of time before beginning dose regimen for ADT.

[00106] In some embodiments, the recombinant DNA vaccine is administered prior to androgen deprivation therapy. In some embodiments, the DNA vaccine is administered every other week for

2-24 weeks before start of administration of ADT, and in some embodiments, the DNA vaccine administration is continued every 2-16 weeks during ADT therapy. Not to be bound by any theory, but the administration of the DNA vaccine prior to administration of androgen deprivation therapy may lead to preferred immune and anti-tumor responses as it has been shown that giving ADT prior to immunization may directly interfere with the priming of T cell responses. One skilled in the art will be able to determine a preferred regimen of ADT and vaccine administration.

[00107] In some embodiments, the subject is a mammal, preferably a human.

[00108] In some embodiments, the methods of the disclosure further comprise administering to the subject an effective amount of a checkpoint pathway inhibitor in addition to the vaccine against androgen receptor to augment ADT treatment. In one example, the checkpoint pathway inhibitor is a

PD-pathway inhibitor. Suitable PD-pathway inhibitors are known in the art. In some embodiments, the PD-pathway inhibitor is an anti-PD-1 blocking antibody or an anti-PD-Ll antibody. [00109] Using different tumor antigen systems we have found that DNA vaccination can elicit PD- Ll expression in tumors as a result of tumor-specific T cells elicited that secrete IFNy. Specifically, tumors expressing a model antigen had an increase in PD-L1 expression following immunization with a DNA vaccine encoding that antigen (Rekoske, B.T., H.A. Smith, B.M. Olson, B.B. Maricque, and D.G. McNeel. (2015). "PD-1 or PD-L1 Blockade Restores Antitumor Efficacy Following SSX2 Epitope-Modified DNA Vaccine Immunization." Cancer Immunol Res. 3 :946-55). If the immunization was modified to elicit CD8+ T cells with higher PD-1 expression, this resulted in an inferior anti-tumor response.

[00110] The Examples below demonstrate that targeting the PD-1/PD-L1 pathway in combination with an AR-targeting vaccine may reduce or prevent tumor-mediated immune suppression. In MycCaP tumor-bearing animals treated with AD and immunized with pTVG-AR, CD8+ T cells were found to have elevated PD-1 expression (Figure 10A). Additionally, some recurrent tumors had elevated PD-L1 expression (Figure 10B). When AR-targeted immunization was combined along with a PD-1 blocking antibody, this treatment significantly delayed tumor growth compared to immunization with pTVG-AR alone (Figure IOC). Furthermore, combining ADT with AR-directed immunization and PD-1 blockade further delayed tumor growth (Figure 10D). Thus, the combination of vaccination and ADT with anti-PD-1 or anti-PD-Ll antibody treatment results in a greater anti -tumor response and may lead to the eradication of tumors. The addition of anti-PD-1 or anti-PD-Ll antibody treatment augments or increases the immune response to androgen receptor. For example, one non-limiting example of a method of treatment includes combination ADT comprising LURH, abiraterone, apalutamide or a combination thereof with the AR vaccine and anti- PD-1 antibody.

[00111] Many different possible scenarios are envisioned for the methods of the present invention including treatment regimens using the double combination (DNA vaccine and ADT) and triple combination therapy (DNA Vaccine, PD-1 pathway inhibitor and ADT) contemplated in this invention. In these scenarios the double and triple combinations do not have to be co-administered and may just be administered over the same period of time in suitable dosage regimens. In other embodiments, the start of each therapy is staggered to provide the most efficacious treatment regimen (e.g. administration of at least 2 or more vaccine doses before the start of ADT therapy). In some embodiments, the method of treating includes administering the triple combination for a period of time followed by administration of the double combination for a second period of time. In other embodiments, the method of treating includes a period of time administering the double combination followed by a period of time administering the triple combination therapy. In other embodiments, the method of treating includes a period of time administering the double or triple combination which includes within that time a period in which one or more of the treatments is not administered while the other therapies are maintained. For example, in the DNA vaccine and PD-1 pathway inhibitor may be administered weekly or biweekly for 2-12 weeks before ADT is administered, whereby DNA vaccine, PD-1 pathway inhibitor and ADT are all administered for at least 12-48 additional weeks, after which time ADT therapy may be stopped for a period of time during with booster administration of the vaccine may continue with or without the PD-1 pathway inhibitor. In some instances, ADT therapy may be re-initiated weeks to months later. Other suitable combinations of the therapies, treatment times and dosing regimens are contemplated to be determined by one skilled in the art.

[00112] In some embodiments, the double combination therapy of DNA vaccine and ADT are contemplated. In some embodiments, the DNA vaccine is administered at least once (i.e. from 1- 12 times) weekly or bi-weekly before the beginning of ADT. Starting the vaccine before ADT treatment may have some advantages in priming the immune system to be activated against cells expressing androgen receptor, which we have found is overexpressed in tumor cells after ADT treatment. This allows for a more robust and increased immune response to tumor cells resulting in a delay or decreased tumor growth. In some embodiments, ADT therapy is started before the administration of the DNA vaccine, e.g. for at least one month or more before DNA vaccine is administered.

[00113] In some embodiments, the ADT administration can also be intermittent. During intermittent ADT administration, the PSA number in a subject may be monitored to determine if ADT therapy should be stopped and/or started again. For example, ADT is stopped once the PSA number is lowered to a suitable level and stabilized; and ADT is restarted when the PSA number increases again (sometimes months, maybe years later). Further, the use of the DNA vaccine may include dosages where the vaccine is given periodically (e.g. every 2 week to every 3 months) over the first year to elicit an anti-tumor response and then administered occasionally (e.g. 3 months or more) as maintenance boosters to maintain an anti-tumor immune response in combination with continuous or intermittent ADT administration.

[00114] In some embodiments, the DNA vaccine, PD-1 pathway inhibitor, and ADT are each administered separately and each are administered over different overlapping time periods. In some instances, all three are administered over the same time period. In some embodiments, all three treatments are administered over the same time period constantly but at different timing intervals. In some embodiments, all three treatments are not constantly administered (e.g. there is period of time in which one or more of the treatments are not administered). In some embodiments, each of the treatments is provided in different dosages that are spaced out at different times after the start of treatment. For example, the DNA vaccine may be administered before the start of treatment with the PD-1 pathway inhibitor and before the start of ADT treatment. In some examples, the DNA vaccine may be administered once every 1-12 weeks, for at least 6 weeks or more, for example once every week or once every other week for 1-24 weeks followed by once every 3-8 weeks for at least an additional 24 weeks or more. In another example, the DNA vaccine and PD-1 pathway inhibitor may be administered on the same schedule of administration before the beginning of ADT treatment (e.g. every 2 weeks for 6-36 week, followed by every 4-6 weeks for at least an additional 6-36 weeks, followed by booster administration every 12-24 weeks for at least an additional year). Other suitable combinations of dosing schedules is contemplated.

[00115] In some embodiments, the length of each treatment (DNA vaccine, PD-1 pathway inhibitor and ADT) and period over which treatment is provided for at least a portion of time. In some embodiments, one or more of the treatments are administered over the same time period. For example, the DNA vaccine, PD-1 pathway inhibitor may be administered at different dosages and at different times over the course of months to years, while the ADT can be administered by known protocols over some or all of the same time period of months or years.

[00116] In some embodiments, the DNA vaccine and the PD-1 pathway inhibitor are administered in multiple dosages prior to the start of the ADT and continue during ADT administration. In some embodiments, the combination of treatment is administered as follows:

vaccine and PD-1 pathway inhibitor administered every 2-4 weeks (e.g. 2 weeks) for at least 8 to 16 weeks, followed by administration of the vaccine and PD-1 pathway inhibitor every 4 weeks for at least 8 to 16 additional weeks, followed by administration of the vaccine and PD-1 pathway inhibitor every 12 weeks (or alternatively every 3 months) for at least an additional 24 weeks; and ADT is administered beginning between week 10 and week 14 after start of the vaccine and PD-1 pathway inhibitor initial administration, and is administered every 12 weeks for at least 4 additional treatment times (i.e. for at least 48 weeks).

[00117] In a preferred embodiment, the vaccine is a DNA vaccine against an androgen receptor and the PD-1 pathway inhibitor is an anti-PD-1 antibody, and the ADT is leuprolide depot 22.5 mg intramuscular administration or goserelin 10.8 mg subcutaneous administration. A suitable dosage regimen is found in FIG. 16, wherein the DNA vaccine and PD-1 pathway inhibitor is administered every 2 weeks for the first 12 weeks, followed by administration every 4 weeks for an additional 12 weeks, and subsequently followed by administration every 12 weeks for at least an additional 24-48 weeks. In this dosage regimen, the ADT is administered at 12 weeks, 24 weeks, 36 weeks and 48 weeks after the initial vaccine/PD-1 inhibitor treatment. This regimen can be extended to at least a year or more in order to treat the tumor.

[00118] In some embodiments, the disclosure provides a method of increasing the efficacy of androgen deprivation therapy in a subject with prostate cancer comprising administering to the subject an effective amount of a recombinant DNA vaccine comprising a polynucleotide operably linked to a transcriptional regulatory element wherein the polynucleotide encodes an androgen receptor or a fragment of the androgen receptor, wherein the method inhibits, delays or reduces the growth of the prostate cancer. In some embodiments, the method further comprises administering to the subject an effective amount of a PD-pathway inhibitor.

[00119] The subject may have previously been diagnosed as having prostate cancer. In some embodiments, the prostate cancer may be in any stage, for example, early stage prostate cancer or newly diagnosed prostate cancer. In some embodiments, the prostate cancer may be metastatic prostate cancer. In another embodiment, the prostate cancer is castration resistant prostate cancer (mCRPC).

[00120] Some embodiments provide a kit for treating prostate cancer. The kit comprises androgen deprivation therapy and a vaccine (for example a DNA vaccine) that elicits an anti -androgen receptor immune response. A set of instructions on the dosages and regiments for administering the ADT and recombinant DNA vaccine may also be provided. In some embodiments, the androgen receptor therapy consists of one or more drugs that target the AR pathway by interfering with AR expression or signaling. Suitable vaccines and drugs are discussed above.

[00121] In another embodiment the kit comprises androgen deprivation therapy, a vaccine (for example a DNA vaccine) that elicits an anti-androgen receptor immune response and a PD-1 pathway inhibitor (such as a PD-1 antibody). A set of instructions on the dosages and regimen for each treatment may be provided.

[00122] The invention will be more fully understood upon consideration of the following non- limiting examples. Each publication, patent, and patent publication cited in this disclosure is incorporated in reference herein in its entirety.

Example 1

Androgen Deprivation Increases Androgen Receptor (AR) Expression and Enhances Tumor Cell Susceptibility to AR-specific T-Cell Responses

[00123] This Example demonstrates that Androgen deprivation results in increased full length AR expression in tumor cells (22Rvl cells), whether deprivation was for a short or extended period of time. Co-culture of AR peptide-specific CD8+ T cells with HLA-A2-expressing tumor cells resulted in increased T-cell activation, cytokine expression, and cytotoxicity assays.

Materials and Methods

Cell Culture

[00124] 22Rvl, LNCaP, PC3, and DU145 cells were obtained from ATCC, and their identity and lack of mycoplasma contamination was confirmed by DDC Medical. Cells were cultured in RPMI- 1640 medium with 200U/mL penicillin/streptomycin, ImM sodium pyruvate, and O. lmM β- mercaptoethanol. This base medium was supplemented with either 10% complete FCS (RPMI/FCS), or 10%) charcoal-stripped serum (RPMI/CSS) to generate androgen-deprived culture medium. Charcoal-stripped serum was generated by incubating dextran-coated charcoal with heat-inactivated FCS and incubating overnight at 4°C, followed by centrifugation and sterile filtration, followed by analysis for testosterone by Testosterone AccuBind ELISA (Monobind).

Androgen receptor enzyme-linked immunosorbent assay (ELISA)

[00125] Cultured prostate cancer cells were collected, cell lysates prepared, and analyzed for protein expression using the PathScan androgen receptor (AR) ELISA per manufacturer's instructions (Cell Signaling Technology). Briefly, microwell strips (pre-coated with anti-AR antibody) were coated with 2mg/mL protein lysates in triplicate, and incubated overnight at 4°C. The following day, AR was detected using a detection antibody followed by HRP -linked secondary antibody and TMB substrate development. A standard curve using purified AR LBD protein (Invitrogen) was generated, and used to obtain relative AR concentration per mg cell lysate.

Androgen receptor quantitative real-time PCR

[00126] Cultured prostate cancer cells were collected, RNA was prepared using Qiagen RNeasy RNA purification system, common concentrations of RNA was used to synthesize cDNA using iScript cDNA synthesis kid (BioRad) and used as a template for qPCR reactions using SsoFast qPCR supermix (BioRad). Reactions were run a Bio-Rad MyiQ thermocycler, using an annealing temperature of 60 °C and 40 cycles. Primer sets:

[00127] AR-FL_Fwd (ACATCAAGGAACTCGATCGTATCATTGC) (SEQ ID NO:7);

[00128] AR-FL_Rev (TTGGGCACTTGCACAGAGAT) (SEQ ID NO: 8);

[00129] AR-V7_Fwd (CCATCTTGTCGTCTTCGGAAATGTTATGAAGC) (SEQ ID NO: 13);

[00130] AR-V7_Rev (TTTGAATGAGGCAAGTCAGCCTTTCT) (SEQ ID NO: 14);

[00131] β-actin Fwd (TCATGAAGTGTGACGTTGACATCCGT) (SEQ ID NO: 15); [00132] β-actin Rev (CTTAGAAGCATTTGCGGTGCACGATG) (SEQ ID NO: 16).

[00133] Results were analyzed by the 2 "Δα method relative to β-actin as a control gene, and fold induction over FCS-treated cells was calculated using the 2 "ΔΔα method, as published [6].

Generation and validation of HLA-A2-expressing 22Ryl cells

[00134] 22Rvl cells cultured greater than six months in RPMI/FCS or RPMI/CSS were diluted into 96-well flat bottom plates at a concentration of 50 cells/well, and transfected with a lentivirus encoding the human HLA-A2 complex. Cells were expanded, stained with HLA-A2-FITC (Biolegend), and sorted for HLA-A2+ events (FACSAria Cell Sorter, BD Biosciences). HLA-A2+ 22Rvl/FCS and 22Rvl/CSS cells were expanded, and AR protein and mRNA expression was validated as above, and HLA-A2 and PD-L1 expression was evaluated by flow cytometry.

Mouse Immunology Assays

[00135] For mouse immunology studies, HHDII-DR1 heterozygous mice were immunized subcutaneously on the right hind flank with lOC^g of the AR811 peptide given with 200μ1 Complete Freund's Adjuvant (Sigma), as published (Olson et al., Cancer Immunol, Immunother. (2011), 33 : 639-647). Seven days later, splenocytes were collected, restimulated for six days with AR811 peptide, and used for intracellular cytokine staining assays and cytotoxicity assays. For intracellular cytokine staining, 200,000 splenocytes were stimulated for 18 hours with media alone, 2000 22Rvl/FCS cells, 2000 22Rvl/CSS cells, or a PMA/Ionomycin positive control. Cells were treated with monensin (GolgiStop, 2μΜ, BD Biosciences) for four hours at 37°C/5% CO2. Cells were then stained with fluorescently-labeled CD3, CD4, CD8, and CD69 antibodies, and after fixation and permeabilization, intracellular staining was conducted using fluorescently-labeled antibodies for IFNy and TNFa (BD Biosciences), or the corresponding isotype controls. Cells were subsequently analyzed using an LSR II flow cytometer (BD Biosciences), and events were analyzed by gating CD3+CD8+ splenocytes and analyzing this population for expression of IFNy and/or TNFa, as well as surface CD69 expression. Cytotoxicity assays were performed as has been previously described (Smith et al., Cane. Res. (2011), 71 : 6785-6795). Briefly, restimulated splenocytes were cultured with 22Rvl/FCS or 22Rvl/CSS target cell lines for 4-6 hours, after which LDH release was calculated using a variation of the Cytotox 96 Assay kid (Promega). The optical density (OD) signal contributed by the media alone was subtracted from all values. All sample conditions were evaluated in triplicate, with the standard error shown. Human Immunology Assays

[00136] For human immunology studies, human T-cell cultures were generated as has been previously described (Olson et al., Cancer Immunol, Immunother. (2011), 33 : 639-647). Briefly, PBMC samples from HLA-A2+ prostate cancer patients were cultured with irradiated peptide-pulsed antigen-presenting cells (either autologous DCs, PBMC, or lymphoblastoid B-cell lines). After 24 hours, cells were treated with lOU/mL IL-2, and restimulated weekly with irradiated peptide-pulsed APCs, and after 2-8 in vitro stimulations, cultures were tested for cytotolytic activity using cytotoxicity assays. AR805 peptide-specific T cells were subsequently used for intracellular cytokine staining assays and cytotoxicity assays. For intracellular cytokine staining, assays were conducted as above, but using fluorescently-labeled antibodies for intracellular IFNy, TNFa, IL-2, and granzyme B (GrB), or the corresponding isotype controls. Cells were subsequently analyzed using an LSR II flow cytometer, and events were analyzed by gating CD3+CD8+T-cells and analyzing this population for expression of IFNy, TNFa, IL-2, and/or GrB, as well as surface CD69 and CD 107a expression. Cytotoxicity assays were performed as above and has been previously described (Smith et al., Cane. Res. (2011), 71 : 6785-6795).

[00137] Results

[00138] Androgen deprivation increases AR protein expression in some prostate cancer cell lines in vitro. DU145, PC3, LNCaP, and 22Rvl cells were cultured under androgen-replete (FCS; media supplemented with complete FCS) or androgen-deprived (CSS; charcoal-stripped serum) conditions for 1, 3, 5, or 7 days or for at least three months (LT; long-term). Protein lysates were collected and analyzed for AR protein expression by ELISA as shown in Figure 1 A-D. * indicates p < 0.05 by Student's t-test.

[00139] Androgen deprivation induces a transient increase in AR-V7 mRNA expression and sustained overexpression of full-length AR mRNA. 22Rvl cells were cultured under androgen- replete or androgen-deprived conditions for 1, 3, 5, or 7 days or for at least three months. RNA was isolated and used to synthesize cDNA, and cDNA was used as the template for qRT-PCR reactions for either full-length AR (Figure 2A and 2C, left panels) or AR-V7 (Figure 2B and 2D, right panels) expression. Top Panels: relative expression (normalized to β-actin control). Bottom panels: fold induction of expression over long-term FCS-cultured 22rvl cells. * indicates p < 0.05 by Student's t- test.

[00140] ARLBD peptide-specific T-cells have increased levels of T-cell activation, Thl polyfunctional cytokine expression, and cytotoxicity against androgen-deprived prostate cancer cells than cells cultured in androgen-replete conditions. 22Rvl cells were cultured greater than six months in androgen-replete (FCS) or androgen-deprived (CSS) conditions, transfected with a lentivirus encoding HLA-A2, and sorted for HLA-A2-expressing cells by flow cytometry. Subsequent 22Rvl/FCS and 22Rvl/CSS cell lines (or non-HLA-A2-transfected 22 Rvl controls) were evaluated for AR protein expression by ELISA (Figure 3A), RNA expression by qRT-PCR (Figure 3B), HLA-A2 expression (Figure 3C), and PD-L1 expression (Figure 3D) by flow cytometry (blue: 22 Rvl/FCS; red: 22Rvl/CSS; black: wild-type 22Rvl; grey: IgG-stained 22Rvl). * indicates p < 0.05 by Student's t-test.

[00141] AR811 peptide-immunized mice have increased cytokine expression, T-cell activation, and cytotoxicity when exposed to androgen-deprived 22Rvl cells. Splenocytes from AR811 peptide-immunized HHDII-DRl heterozygous mice were evaluated for immune responses against

HLA-A2-expressing 22Rvl/FCS or 22Rvl/CSS cells. Figure 4A-B shows intracellular cytokine staining of splenocytes cultured with 22Rvl/FCS (Figure 4A) or 22Rvl/CSS (Figure 4B). Figure 4C depicts CD69 expression of splenocytes cultured with 22Rvl/CSS (blue) or 22Rvl/FCS (red) cells

(mean fluorescent intensity quantified in inset - * indicates p < 0.05 by Student's t-test.). Figure 4D shows cytotoxicity of splenocytes cultured with 22Rvl/CSS (blue) or 22Rvl/FCS (red) cells.

[00142] Human AR805 peptide-specific T-cells are shown to have increased levels of T-cell activation, Thl polyfunctional cytokine expression, and cytotoxicity when exposed to androgen-deprived 22Rvl prostate cancer cells. AR805 peptide-specific T-cells (previously cultured from the peripheral blood of HLA-A2+ prostate cancer patients (Smith et al., Cane. Res.

(2011), 71 : 6785-6795) were evaluated for immune responses against HLA-A2-expressing

22Rvl/FCS or 22Rvl/CSS cells. Figure 5A shows CD69 expression of splenocytes cultured with

22Rvl/CSS (blue) or 22Rvl/FCS (red) cells (quantified in adjacent bar graph - * indicates p < 0.05 by Student's t-test). Figure 5B shows intracellular cytokine staining (IFNy by T Fa) of T-cells cultured with 22Rvl/CSS (right panel) or 22Rvl/FCS (left panel) cells. Figure 5C shows frequency of CD8+ T cells that were found to express zero (blue), one (green), two (yellow), three (orange), or four (red) Thl related molecules (IFNy, T Fa, IL-2, and/or granzyme B). Figure 5D shows granzyme B expression by CD8+ T cells following culturing with 22Rvl/FCS (red) or 22Rvl/CSS (blue) cells (quantified in adjacent bar graph - * indicates p < 0.05 by Student's t-test). Figure 5E shows frequency of CD8+ T cells expressing the surface degranulation marker CD 107a. Figure 5F shows cytotoxicity of T-cells cultured with 22Rvl/CSS (blue) or 22Rvl/FCS (red) cells.

[00143] Thus, androgen deprivation increases androgen receptor expression and causes prostate tumor cells to have increased susceptibility to AR-specific T-cells.

Example 2

Phase I Clinical Trial evaluating a DNA vaccine encoding the AR-LBD in combination with androgen deprivation therapy

[00144] Figure 6 A depicts a clinical trial. Men with metastatic prostate cancer who have recently (within 1-6 months) initiated androgen deprivation therapy (ADT) are enrolled in a clinical trial evaluating the safety and immunogenicity of a DNA vaccine encoding the AR LBD (pTVG-AR). This trial seeks to capitalize on targeting one of the most common mechanisms of resistance to ADT (overexpression of the AR) by combining ADT with pTVG-AR, ideally resulting in delayed time to progression {dashed blue line) to castrate-resistant disease (CRPC). Figure 6B depicts the different ARMS of the study. Patients receive either six biweekly immunizations followed by quarterly boosters, or two biweekly immunizations every ten weeks, either alone or in combination with GM- CSF. Immunizations are continued 18 months or to disease progression. Primary endpoints are safety and ARLBD-specific immunity. Secondary objectives of this trial include evaluating which schedule of immunization is best able to elicit long-lived ARLBD-specific T-cell responses, the effect of GM-CSF in generating immune responses, and to determine the median time to PSA progression and 18-month PSA progression-free survival.

Example 3

[00145] AR overexpression occurs in human and mouse prostate tissue following AD therapies and enhances their recognition by AR-specific T-cells.

[00146] Androgen-deprived 22Rvl human prostate cancer cells and castrate-resistant MycCaP mouse prostate cancer cells have increased AR expression (Figure 7A and 7B) after ADT treatment. We also showed this occurs in the MycCaP prostate cancer model in vivo, as chemical castration using the GnRH antagonist degarelix resulted in increased AR expression in tumors (Figure 7C). This increased AR expression also caused these tumor cells to be better recognized by AR-specific CD8+ T-cells, as T-cells have higher levels of activation, cytokine expression, and cytotoxicity when cultured with androgen-deprived tumor cells (Figure 7D).

[00147] Briefly, Human 22Rvl prostate cancer cells cultured in androgen replete (FCS) or - deprived (CSS) conditions (Figure 7A), or mouse MycCaP cells serially passaged in untreated (MycCaP/AS) or castrate (MycCaP/CR) mice (Figure 7B) were collected and analyzed for AR expression by qPCR (left panels), ELISA (center panels), and intracellular staining (ICS, right panels). Figure 7C, FVB mice were challenged with MycCaP/AS tumor cells, and given degarelix or a sham treatment. At the time of outgrowth, tumors were collected and analyzed for AR expression by intracellular flow cytometry (example histogram with samples stained with IgG or AR- intracellular antibodies; quantified in right panels). Figure 7D, AR-specific CD8+ T cells were cultured with 22Rvl/FCS or 22Rvl/CSS cells, and evaluated for T-cell activation by CD69 expression (left panel), ΠΤΝΓγ and TNFa cytokine expression (center panel), and cytotoxicity (right panel). In all panels, * indicates p<0.05 by Student's t-test.

Example 4

[00148] AD with AR-directed immunization increases anti-tumor immune responses and delays tumor recurrence

[00149] This Experiment demonstrates that the combination of AD with AR-directed immunization increases anti-tumor immune response. MycCaP tumor-bearing FVB mice treated with degarelix

(ADT treatment) followed by immunization at weekly intervals with the pTVG-AR DNA vaccine had enhanced immune responses against androgen-deprived tumor cells and a delay in prostate cancer regrowth compared to controls (Figure 8). Briefly, male FVB mice (n=8) were implanted with MycCaP tumors, treated with degarelix (or sham), and immunized weekly with pTVG-AR or control pTVG4. Figure 8A, splenocytes were collected and analyzed for immune responses against

MycCaP/CR tumor cells by intracellular cytokine staining (left panel) and AR peptide-stimulated splenocytes from pTVG-AR-immunized mice were measured for ability to lyse MycCap/AS vs.

MycCap/CR tumor cells (right panel). Figure 8B depicts mice followed for tumor volume. In all panels, * indicates p<0.05 by Student's t-test. Figure 8C depicts tumor volume post-challenge of control (sham), degarelix + pTVG4 and degarelix + pTVG-AR.

Example 5

[00150] Increased androgen receptor expression in prostate cancer cells following androgen deprivation increases recognition by androgen receptor-specific T cells

[00151] This example again demonstrates that androgen deprivation increases AR expression in human and murine prostate tumor cells in vivo and in vitro that persisted over time. Increased AR expression was associated with increased recognition and cytolytic activity by AR-specific T cells. Further, ADT combined with vaccination, using a DNA vaccine encoding the ligand-binding domain of the AR, led to improved anti-tumor responses as measured by tumor volumes and delays in the emergence of castrate-resistant prostate tumors in two murine prostate cancer models (Myc-CaP and prostate-specific PTEN-deficient mice). This data supports the benefits of combining ADT with AR- directed immunotherapy over ADT combined with other immunotherapeutic approaches by specifically targeting a major mechanism of resistance, overexpression of AR.

[00152] MATERIALS AND METHODS OF EXAMPLE 5

Mice and cell lines

[00153] Human prostate cancer cells were obtained from ATCC, and cultured in RPMI-1640 medium with 200U/mL penicillin/streptomycin, ImM sodium pyruvate, and O. lmM β- mercaptoethanol. Cell identity and mycoplasma testing was confirmed by DDC Medical (Fairfield, OH). Myc-CaP/AS or Myc-CaP/CR cells (androgen-sensitive and castrate-resistant variants of the Myc-CaP parental line originally generated by Charles Sawyers) and culture conditions have been previously described (22). Both human and mouse cell lines were maintained in either 10% complete fetal calf serum (FCS) or charcoal-stripped serum (CSS) for androgen-replete or androgen- deprived conditions.

[00154] Tumor studies using Myc-CaP tumor cells were conducted in wild-type male FVB mice

(Jackson Laboratory, Bar Harbor, ME). PTEN knock-out mice were generated by crossing Pten floxed (loxp/loxp) animals with Probasin-Cre (PB-Cre4+) as has been described (23). Mice were screened by PCR for the floxed or wild-type PTEN alleles (forward primer: CAA GCA CTC TGC GAA CTG AG; reverse primer: AAG TTT TTG AAG GCA AGA TGC) and PB-Cre transgene (forward primer: CTG AAG AAT GGG ACA GGC ATT G; reverse primer: CAT CAC TCG TTG CAT CGA CC). Mice were maintained under aseptic conditions and all experiments were conducted under an IACUC-approved protocol.

Tumor studies

[00155] FVB mice were inoculated subcutaneously with 106 Myc-CaP/AS tumor cells, and followed daily for the presence of palpable tumors. Once tumors were palpable, mice were treated subcutaneously with either degarelix (25mg/kg) or a vehicle sham treatment every four weeks. For immunization studies, degarelix-treated animals were randomized to weekly immunization with lOC^g pTVG4 or pTVG-AR beginning one day after receiving degarelix. Tumor growth was measured at least three times weekly, and tumor volumes calculated as we've published (19). At the time of euthanasia, tumors and spleens were collected. For studies using PTEN-deficient mice, animals began receiving degarelix (25mg/kg) at 20 weeks (+/- two weeks) of age, followed by biweekly immunization with 100μg pTVG4 or pTVG-AR beginning one day after ADT. Animals were treated until 40 weeks of age (+/- two weeks) before tissue collection.

Androgen receptor enzyme-linked immunosorbent assay (ELISA)

[00156] Cultured prostate cancer cells were collected, cell lysates prepared, and analyzed for protein expression using the PathScan androgen receptor ELISA per manufacturer's instructions (Cell Signaling Technology, Danvers, MA). Briefly, microwell strips (pre-coated with anti-AR antibody) were coated with 2mg/mL protein lysates in triplicate, and incubated overnight. AR was detected using a detection antibody followed by HRP-linked secondary antibody and TMB substrate development. A standard curve using purified AR LBD protein (Invitrogen, Carlsbad, CA) was generated, and used to determine relative AR concentration per mg cell lysate.

Flow cytometry

[00157] For androgen receptor intracellular staining, cells were stained with a Live/Dead GhostDye 780 Live/Dead Stain (Tonbo Biosciences, San Diego, CA) and CD45 (clone 30-F11, Tonbo Biosciences) for dissociated tumor samples, and intracellularly stained with antibodies directed against the androgen receptor ligand-binding domain (clone EP670Y, Abeam, Cambridge, United Kingdom) and amino terminal domain (clone D6F11, Cell Signaling Technologies), or isotype controls. For HLA-A2 and PD-L1 expression, cells were stained with HLA-ABC (clone W6/32, eBioscience, San Diego, CA) and PD-L1 (clone MIH-5, eBioscience) antibodies.

Androgen receptor quantitative real-time PCR

[00158] Prostate tumor cells (cell lines or dissociated tumors) were collected, RNA was prepared (RNeasy RNA purification system; Qiagen, Hilden, Germany), used to synthesize cDNA (i Script cDNA synthesis kit; BioRad, Hercules, CA), and used as a template for qPCR reactions using SsoFast qPCR supermix (BioRad). Reactions were performed using a Bio-Rad MyiQ thermocycler, using an annealing temperature of 60°C and 40 cycles. Primer sets:

• full-length human androgen receptor

o forward: ACATCAAGGAACTCGATCGTATCATTGC, SEQ ID N07;

o reverse: TTGGGCACTTGCACAGAGAT, SEQ ID NO:8,

• AR-V7

o forward: CCATCTTGTCGTCTTCGGAAATGTTATGAAGC, SEQ ID NO: 13; o reverse: TTTGAATGAGGCAAGTCAGCCTTTCT, SEQ ID NO: 14,

• full length mouse AR

o forward: GGACCATGTTTTACCCATCG, SEQ ID NO: 17;

o reverse: ATCTGGTCATCCACATGCAA, SEQ ID NO: 18,

• mouse AR-V2

o forward: GGACCATGTTTTACCCATCG, SEQ ID NO: 17;

o reverse: TTGTTGTGGCAGCAGAGTTC, SEQ ID NO: 19,

• mouse AR-V4

o forward: GGACCATGTTTTACCCATCG, SEQ ID NO: 17;

o reverse: AAGTGGGGAACCACAGCAT, SEQ ID NO:20, and

• β-actin

o forward: TCATGAAGTGTGACGTTGACATCCGT, SEQ ID NO: 15;

o reverse: CTTAGAAGCATTTGCGGTGCACGATG, SEQ ID NO: 16)(24-26). Results were analyzed by the 2-ACt method relative to β-actin as a control gene, as published (26). Immunology Assays

[00159] To study immune responses, human T-cell lines or splenocytes were collected as previously described (20), and used for intracellular cytokine staining assays and cytotoxicity assays. For intracellular cytokine staining, cells were stimulated for 18 hours with media alone, an ARLBD peptide pool (a pool of 15-mer peptides, overlapping by 11 residues, and covering the entire sequence of the AR LBD; LifeTein, Somerset, NJ), tumor cells, or a PMA/Ionomycin positive control. Cells were stained using a fixable live/dead marker (Tonbo Bioscience) and extracellular and intracellular antibodies. Human antibodies: CD3 (clone UCHT1, BD Biosciences), CD4 (clone RPA-T4, BD Biosciences), CD8 (clone RPA-T8, eBioscience), CD69 (clone FN50, BD Biosciences), CD107a (clone H4-A3, BD Biosciences), IL2 (clone MQ1-17H12, eBioscience), IFNy (clone 4S.B3, BioLegend, San Diego, CA), TNFa (clone MAbl l, BD Biosciences), GrB (clone GB11, BD Biosciences). Mouse antibodies: CD3 (clone 17A2, BD Biosciences), CD4 (clone GK1.5, BD Biosciences), CD 8 (clone 53-6.7, BD Biosciences), CD45 (clone 30-F11, BD Biosciences), CD69 (clone H1.2F3, eBioscience), IFNy(clone XMG1.2, BD Biosciences), TNFa (clone MP6-XT22, BD Biosciences). Cells were subsequently analyzed using an LSR II or Fortessa flow cytometer (BD Biosciences), and events were analyzed by gating CD3+CD4+ or CD3+CD8+ cells and analyzing this population for expression of CD69, CD107a, IFNy, TNFa, IL2, and/or GrB. Cytotoxicity assays were performed as has been previously described (20). Briefly, splenocytes were restimulated for five days with an ARLBD peptide pool, and were cultured with tumor cell lines, after which LDH release was calculated using the Cytotox 96 Assay kit (Promega, Madison, WI), as previously published (19).

Immunohistochemistry

[00160] Paraffin-embedded MycCaP tumors were stained for CD3 expression by immunohistochemistry as has been described (20). Sections were stained with primary antibodies (CD3 : clone SP7, Abeam), developed using the LSAB+ System-HRP (Agilent Technologies, Santa Clara, CA) and Metal Enhanced DAB Substrate Kit DAB metal concentration (Thermo Fisher Scientific, Waltham, MA), imaged using an Olympus BX51 fluorescent microscope (Olympus, Lombard, IL) in combination with SPOT RT analysis software (SPOT Imaging Solutions, Sterling Heights, MI), and quantified by the frequency of CD3+ cells per lOx field, counting at least five fields per tumor section per animal by a blinded investigator.

Positron Emission Tomography/Computed Tomography Imaging

[00161] All mice were intravenously administered between 5-8 MBq of 124I-CLR1404 and then micro positron emission tomography/computed tomography (PET/CT) scanned 96hrs post-injection. During scanning, mice were anesthetized with 2% isoflurane inhalation gas mixed with lL/min of pure oxygen (27). Mice were scanned with the Siemens Inveon Hybrid microPET/CT (Siemens Medical Solutions, Knoxville, TN) in the prone position. Forty-million counts per mouse were collected for the PET scan to obtain adequate signal-to-noise. PET data were histogrammed into one static frame and subsequently reconstructed using ordered- sub set expectation maximization (OSEM) of three dimensions followed by the maximum a posteriori algorithm, and CT attenuation and scatter correction were applied based on the NEMA NU 4 image-quality parameters (28).

[00162] All PET and CT images were co-registered. Image data were analyzed using the General Analysis tools provided by Siemens Inveon Research Workplace (Siemens Medical Solutions). Data were identically window/leveled and scaled according to each animal's decay corrected injection activity. Based on the PET and CT images, a reference volume of interest (VOI) was drawn around each tumor and a separate background tissue VOI was drawn on muscle and liver. VOI thresholding within the reference tumor VOI was adjusted to include all signal greater than sixty percent of the maximum signal. Data were reported as percent injected dose normalized by the mass of the tissue VOI (%ID/g tissue), with the assumption that all tissue density is akin to water (lg/mL). Data were then averaged within pre- and post-treatment groups and normalized to background tissue values.

[00163] RESULTS

[00164] Androgen deprivation increases androgen receptor expression and enhances AR- specific T cell responses to androgen-deprived prostate tumor cell lines

[00165] In this Example, a panel of six prostate cell lines (two immortalized prostate epithelial lines, two androgen-independent prostate cancer lines, and two androgen-dependent prostate cancer lines) were cultured for short (one to seven days) or extended periods (greater than six months) in androgen-deprived medium and analyzed for AR expression. Androgen deprivation was found to result in an increase in AR protein expression in androgen-dependent prostate tumor cells by quantitative ELISA (Fig. 11 A), as well as by intracellular staining using antibodies directed against both the ligand-binding domain as well as the amino-terminal domain (Fig. 1 IB, with the amplitude and frequency of AR expression quantified in Fig. 11C and 11D, respectively). Analysis of 22Rvl cells (which are known to express AR-V7, an LBD-loss splice variant) showed that androgen deprivation led to a steadily increasing expression in full-length AR as well as a transient increase in

AR-V7 (Fig. HE), with no detectable expression of AR-Vl, AR567es, or other splice variants). However, AR-V7 expression was at significantly lower levels compared to full-length AR transcripts.

[00166] To determine whether this increase in AR expression following androgen deprivation resulted in enhanced AR-specific T-cell effector function against these tumor cells, 22Rvl cells were first transfected to express HLA-A2 as a model MHC molecule, and one for which AR-restricted epitopes have been previously identified (19). After generating this cell line, increased AR protein and RNA expression following androgen-deprivation observed in the parental cell lines was confirmed in these HLA-A2-expressing lines (Fig. 11F-G). These 22Rvl/FCS and 22Rvl/CSS cells were then incubated with T cell lines specific for the HLA-A2-restricted AR805 epitope. T cells cultured with the 22Rvl/CSS cell line were shown to have higher levels of T-cell activation (as measured by CD69 expression - Fig. 12 A), as well as increased expression of Thl cytokines (Fig. 12B), including CD8+ T cells with polyfunctional cytokine expression (Fig. 12C), compared to T cells that had been stimulated with 22Rvl cells cultured under androgen-replete conditions. Co- culture with 22Rvl/CSS cells also resulted in higher expression of granzyme B (Fig. 12D), the degranulation marker CD 107a (Fig. 12E), as well as increased cytotoxicity (Fig. 12F) compared to co-culture with 22Rvl/FCS cells. Similar studies, using splenocytes from HLA-A2 transgenic mice that were directly immunized with another HLA-A2 restricted epitope, AR811, replicated these results in terms of increased cytokine expression, T-cell activation, and cytotoxicity when cultured with androgen-deprived HLA-A2-expressing 22Rvl cells (Fig. 12G). These differences in T-cell recognition and cytotoxicity were likely not due to altered MHC class I nor PD-L1 expression, as 22Rvl/CSS and 22Rvl/FCS cells expressed identical levels of both HLA-A2 and PD-L1 (FIG. 11H- I).

[00167] Androgen deprivation increases AR expression in Myc-CaP tumor cells in vitro and in vivo

[00168] We have previously used the TRAMP mouse model to study the impact of vaccines targeting AR on tumor development and progression (20). However, it has previously been reported that androgen deprivation of TRAMP mice, and many other murine prostate tumor models, results in

AR loss and the development of neuroendocrine tumors (29). Consequently, we sought to evaluate other models more representative of human prostate cancer that continue to express AR following androgen deprivation. One such model is the Myc-CaP cell line, which mimics the human disease in that it maintains AR expression following castration (22). To confirm this, androgen-sensitive Myc- CaP cells (Myc-CaP/AS, generated from untreated FVB mice), and castration-resistant Myc-CaP cells (Myc-CaP/CR, generated from serial passaging of the Myc-CaP/AS cell line through castrated mice) were studied. Similar to what was observed in the human prostate cancer cell lines, the Myc- CaP/CR cell line was found to have increased full-length AR expression by both quantitative ELISA (Fig. 13 A) and intracellular staining compared to the Myc-CaP/AS cell line (Fig. 13B). While analysis of RNA transcripts showed an increase in the murine AR splice variants mAR-V2 and mAR-V4, these splice variants were similarly several fold lower in expression than the full-length AR (Fig. 13C). To study the expression of AR in vivo, FVB mice were inoculated with Myc- CaP/AS cells, and then given either a sham treatment or castration by administration of a GnRH antagonist (degarelix). Animals were followed for tumor growth (Fig. 13D), and recurrent tumors were collected and CD45- cells were analyzed for AR expression by intracellular staining. Tumors that recurred following androgen deprivation were found to have increased AR expression, both in terms of frequency of CD45- cells with detectable expression of the AR, as well as the amplitude of AR expression within these cells (Fig. 13E).

[00169] Immunization with pTVG-AR delays the growth of castration-resistant prostate tumors following androgen deprivation

[00170] This Example also demonstrates that androgen deprivation in combination with AR- targeted vaccination delays the outgrowth of castration-resistant tumors by specifically targeting cells overexpressing AR. Mice were implanted with Myc-CaP/AS tumors, and mice with established tumors were given either a sham treatment or degarelix. Mice treated with degarelix were then randomized to immunization with either a DNA vaccine encoding the AR LBD (pTVG-AR), or an empty vector control (pTVG4). The combination treatment with degarelix and pTVG-AR was found to delay tumor growth compared to treatment with degarelix and control vaccine (Fig. 14A-B). Additionally, when animals were evaluated for evidence of immune responses against the Myc- CaP/AS or Myc-CaP/CR cell lines, animals immunized with pTVG-AR were found to have increased immune responses against the castration-resistant cell line, both in terms of cytokine expression (Fig. 14C) as well as cytotoxicity (Fig. 14D). In parallel studies, we found that immunization of Myc-CaP/AS-bearing mice with pTVG-AR resulted in an increased frequency of tumor-infiltrating CD3+ T cells, and this was further increased when vaccination was combined with degarelix treatment (Fig. 14E).

[00171] Androgen deprivation increases AR expression in PTEN-deficient tumors, and immunization with pTVG-AR, combined with ADT, decreased growth of castration-resistant tumors

[00172] As an additional, relevant model of human prostate cancer, this Example utilized the PbCre PTENfl/fl mice, in which prostate-specific expression of the Cre recombinase drives deletion of the PTEN tumor suppression and the formation of autochthonous prostate tumors. The PTEN-CaP8 cell line (derived from one of these autochthonous tumors) was similarly cultured in androgen-replete or androgen-deprived medium. As shown in Fig. 15A-B, androgen-deprivation resulted in a significant increase in AR protein expression, similar to the human prostate cancer cell lines and Myc-CaP cell lines above. Twenty-week old PbCre+PTENfl/fl mice were then given either a sham treatment, or degarelix in combination with pTVG-AR vaccine or vector control. To non-invasively monitor tumor growth, as well as to randomize animals prior to treatment, we utilized microPET/CT imaging, employing the novel radiotracer 124I-CLR1404, which is a radioiodinated alkylphosphocholine (APC) analog that has shown selective tumor uptake in >95% of malignant models to date (30). Animals were intravenously administered 124I-CLR1404 and subsequently PET/CT scanned within one week prior to initiation and completion of therapy (Figure 15C), and imaging results were analyzed for mean and maximum tumor uptake. Analysis of tumors pre-treatment showed no difference between mean and maximum tumor uptake (Figure 15D-E). While some animals with large tumors died prior to the last imaging session, and hence not all animals underwent post- treatment imaging, notwithstanding, androgen deprivation was shown to result in decreased 1241- CLR1404 mean and max tumor uptake, as shown in Figure 15F and Figure 15G, respectively. No significant difference in %ID/gmean or %ID/gmax was detected post-treatment between animals receiving ADT and control vaccine versus animals receiving ADT and AR-targeted vaccine. However, as measured during necropsy, animals treated with degarelix and pTVG-AR had significantly smaller tumor volumes, as determined by genitourinary complex weight, compared to animals receiving degarelix and pTVG4 (Figure 15H).

[00173] DISCUSSION [00174] This Example demonstrates that androgen deprivation results in increased full-length AR expression in vitro and in vivo that persists over time, and that this increased AR expression is associated with these cells being better targets for AR-specific T cells. Furthermore, a DNA vaccine encoding the AR LBD enhanced immune responses that preferentially recognized and lysed castrate- resistant prostate cancer cells, and delayed the recurrence of castrate-resistant disease when combined with ADT. A vaccine targeting the AR may be preferred over other antigen-specific vaccines when specifically combined with ADT by targeting a major mechanism of resistance that drives castrate-resistant tumor growth.

[00175] In summary, this Example shows that increased AR expression in prostate cancer cells following ADT results in enhanced recognition and lysis by AR-specific T cells. The combination of ADT and AR-specific immunization in vivo enhanced anti-tumor T cell immunity, as well as delayed the recurrence of castrate-resistant tumors. These studies provide a rationale for combining ADT with AR-targeted immunization, an approach that is being evaluated in a Phase I clinical trial (NCT02411786).

Example 6

[00176] Immunization elicits PD-L1 expression in tumors and PD-1 PD-L1 blockage can increase the anti-tumor efficacy of DNA vaccination.

[00177] Using different tumor antigen systems we have found that DNA vaccination can elicit PD-

Ll expression in tumors as a result of tumor-specific T cells elicited that secrete IFNy. Specifically, we have reported that tumors expressing a model antigen had an increase in PD-L1 expression following immunization with a DNA vaccine encoding that antigen (Rekoske, B.T., H.A. Smith,

B.M. Olson, B.B. Maricque, and D.G. McNeel. (2015). "PD-1 or PD-L1 Blockade Restores

Antitumor Efficacy Following SSX2 Epitope-Modified DNA Vaccine Immunization." Cancer

Immunol Res. 3 :946-55). If the immunization was modified to elicit CD8+ T cells with higher PD-1 expression, this resulted in an inferior anti -tumor response. Combining vaccination with anti-PD-1 or anti-PD-Ll antibody treatment resulted in a greater anti -tumor response and eradication of tumors in some animals (Rekoske, B.T., H.A. Smith, B.M. Olson, B.B. Maricque, and D.G. McNeel. (2015).

"PD-1 or PD-L1 Blockade Restores Antitumor Efficacy Following SSX2 Epitope-Modified DNA

Vaccine Immunization." Cancer Immunol Res. 3 :946-55). We have recently identified that this also occurs following human immunization using cryopreserved blood samples collected from patients with advanced prostate cancer treated with a DNA vaccine encoding prostatic acid phosphatase (PAP).

[00178] Using in vitro and trans vivo methods, we found that immune responses to PAP were detected and/or augmented when combined with PD-1 blockade (Figure 9A, B). Moreover, we detected increased expression of PD-L1 on circulating tumor cells following DNA vaccination, and we found that higher expression correlated with the development of persistent antigen-specific IFNy- secreting T cell immune responses (Figure 9C). We observed similar results in blood samples from patients treated with sipuleucel-T, an FDA-approved vaccine for prostate cancer which targets the same PAP antigen (data not shown). Together, these data provide evidence to support combining anti -tumor vaccines with a PD-1 pathway inhibitor. Briefly, Figure 9 A shows PBMC from patients previously immunized with a PAP -targeting vaccine were cultured in vitro for 72 hours with PAP in the presence of a PD-1 -blocking antibody (or IgG control), and measured for IFNy (left panel) or granzyme B (right panel) secretion by ELISA. Figure 9B shows PBMC obtained from patients after immunization were injected into the footpads of NOD/SCID mice with PAP protein and PD-1 blocking antibody (or IgG control), and 24 hours later, footpad swelling was measured. Figure 9C,

PD-L1 expression was measured on circulating tumor cells from patients with persistent PAP- specific Thl-biased immune responses (R) vs. non-responders (NR) following immunization with a

DNA vaccine targeting PAP. The ratio of PD-L1 MFI on post-treatment samples compared to pre- treatment samples is shown. In all panels, * indicates p<0.05 by Student's t-test.

[00179] More recent preliminary data also suggest that targeting the PD-1/PD-L1 pathway in combination with an AR-targeting vaccine is a rationale combination to circumvent this means of tumor-mediated immune suppression. In MycCaP tumor-bearing animals treated with AD and immunized with pTVG-AR (as in Figure 8), CD8+ T cells were found to have elevated PD-1 expression (Figure 10A). Additionally, it was observed in other models following the generation of antigen-specific immune responses following immunization, some recurrent tumors had elevated PD-

Ll expression (Figure 10B). When AR-targeted immunization was combined along with a PD-1 blocking antibody, this treatment significantly delayed tumor growth compared to immunization with pTVG-AR alone (Figure IOC). Furthermore, combining ADT with AR-directed immunization and

PD-1 blockade further delayed tumor growth (Figure 10D). Together, these finding suggest that PD-

1 pathway inhibitors would be an effective means to target resistance to combined ADT and AR- directed immunization and could prevent (or significantly delay) the formation of the lethal, castrate- resistant form of prostate cancer.

[00180] Briefly, FVB mice were implanted subcutaneously with MycCaP tumor cells, treated the following day with degarelix, and the following day were immunized with pTVG4 (vector control) or pTVG-AR. At the time of tumor outgrowth, animals were analyzed for PD-1 expression on splenic CD8+ T-cells (Figure 10A) and PD-L1 expression on CD45- tumor cells (Figure 10B). For Figure IOC, FVB mice (n=5) were implanted with MycCaP tumors, and the following day immunized with pTVG-AR (repeated weekly), without castration, and each day following vaccination were treated with a PD-1 -blocking antibody or control, and followed for tumor growth. For Figure 10D, MycCaP tumor-bearing FVB mice were treated with degarelix, pTVG-AR, and anti- PD-1 (n=5) or IgG control (n=9), and followed for tumor growth. In all panels, * indicates p<0.05 by Student's t-test.

Example 7

Clinical Trial Design using AR-targeted vaccination in combination with androgen deprivation and T-cell checkpoint blockade

[00181] An open-label, randomized pilot clinical trial is to be performed, with a maximum of 50 patients with newly diagnosed prostate cancer. The patients are randomly assigned to one of three treatment arms below.

[00182] Study objectives: The primary clinical objectives of the trial is safety and pathological complete response rate per study arm. Primary objectives include evaluating the safety of combination androgen deprivation, alone or in combination pTVG-AR DNA vaccine with or without pembrolizumab, in patients with newly diagnosed prostate cancer; and to determine the pathological complete response rate in patients with prostate cancer treated with combination androgen deprivation (LHRH agonist, abiraterone acetate, and apalutamide) or with pTVG-AR, with or without pembrolizumab, prior to radical prostatectomy.

[00183] Secondary objectives include determining one year PSA progression-free survival rates; determining whether treatment with pTVG-AR, with or without pembrolizumab, elicits persistent systemic AR-specific Thl-biased T-cell responses, and determining whether treatment with pTVG-

AR, with or without pembrolizumab, elicits increased prostate tissue-infiltrating CD8+ T cells. [00184] Subject population: Eligible subjects are patients with newly diagnosed prostate cancer who are planning to undergo radical prostatectomy as extirpative treatment. Subjects need not be HLA-A2 positive, however serological typing is performed to identify patients for epitope-specific T-cell analyses. In previous trials at our institution, we have found ~ 50% of patients were HLA-A2- expressing, consequently we anticipate -50% of patients are available for these analysis.

[00185] Trial design: This will be a randomized, open-label, multi-institution pilot trial designed to evaluate the immunological and clinical effect of a DNA vaccine encoding AR with rhGM-CSF adjuvant given with or without pembrolizumab. Study arms will be defined as follows:

Arm 1 : Leuprolide depot (or equivalent) 22.5 mg intramuscular injection day 1, day 85

Abiraterone acetate 1000 mg p.o. daily, beginning day 1 until day prior to surgery

Prednisone 5 mg p.o. daily, beginning day 1 until 1 week after surgery, then taper

Apalutamide 240 mg p.o. daily, beginning day 1 until day prior to surgery

Arm 2: Leuprolide depot (or equivalent) 22.5 mg intramuscular injection day 1, day 85

Abiraterone acetate 1000 mg p.o. daily, beginning day 1 until day prior to surgery

Prednisone 5 mg p.o. daily, beginning day 1 until 1 week after surgery, then taper

Apalutamide 240 mg p.o. daily, beginning day 1 until day prior to surgery

pTVG-AR (100 μg) with rhGM-CSF (208 μg) administered intradermally (i.d.) biweekly 6 times, beginning day 1

Arm 3 : Leuprolide depot (or equivalent) 22.5 mg intramuscular injection day 1, day 85

Abiraterone acetate 1000 mg p.o. daily, beginning day 1 until day prior to surgery

Prednisone 5 mg p.o. daily, beginning day 1 until 1 week after surgery, then taper

Apalutamide 240 mg p.o. daily, beginning day 1 until day prior to surgery

pTVG-AR (100 μg) with rhGM-CSF (208 μg) administered intradermally (i.d.) every 3 weeks 8 times, beginning day 1

Pembrolizumab 2 mg/kg, administered intravenously over 30 minutes, every 3 weeks 8 times, beginning day 1, each dose following pTVG-AR vaccination [00186] A total of 50 eligible patients (10 in Arm 1, 20 in Arm 2 and 20 in Arm 3) will be randomized. All subjects are followed for adverse events; if adverse events attributed to study treatment exceed the tolerability limit (> 33% grade > 2 toxicity, or > 10% grade > 3 toxicity), further accrual would halt.

[00187] Measurement of Effect: Patients eligible for this trial will not have metastatic disease at the time of enrollment.

Pathological evaluation: Prostate tissues obtained by biopsy pre-treatment and at the time of prostatectomy, will be reviewed and graded by a single pathologist (Dr. Jiaoti Huang, MD PhD or designee) as per standard clinical pathology review. The absence of identifiable prostate cancer at the time of prostatectomy will be used to define a pathological complete response.

[00188] Serum PSA evaluation: Serum PSA is expected to be undetectable following prostatectomy in the absence of residual/recurrent disease. Hence PSA progression will be defined as a detectable PSA (above the clinical lab's lower limit of detection) at any point after 3 months after the date of prostatectomy, and confirmed by a second reading at least 2 weeks later.

[00189] Safety: All subjects are observed at every visit during the period of treatment for symptoms assessment. Laboratory analyses is performed at regular intervals for evidence of adverse events. These clinical laboratory studies include complete blood counts, creatinine, liver function tests, PSA, serum aldolase (for muscle-related toxicity assessment), and anti-nuclear antibodies.

Adverse events are graded by the current version (4.0) of the NCI Common Terminology Criteria.

The number and severity of toxicity incidents are analyzed descriptively in tabular format.

[00190] Immunological Monitoring: Blood will be collected by either peripheral blood draw (up to

210 mL) or leukapheresis (50-100 mL) pre-immunization, after 3 months of treatment, at the time of prostatectomy, and at 3 months, 6 months, and 12 months after prostatectomy, for immunological monitoring. From the heparinized blood, peripheral blood mononuclear cells (PBMC) will be prepared by density centrifugation over Ficoll-Paque using standard techniques. PBMC will be used directly for analysis, and residual material cryopreserved in liquid nitrogen using 90% autologous serum collected at the time of blood draw, or 90% fetal calf serum, and 10% DMSO. Sera will be prepared from the red-top tubes and stored in aliquots at -80°C for antibody analyses. IFNy and granzyme B ELISPOT analysis, and ELISA tests for antigen-specific antibodies, will be the primary methods of analysis. The primary antigens tested will be AR (experimental), PSA (negative control), and tetanus toxoid (positive control). The primary immune analysis will be conducted at the 6- month post-surgery time point, and compared with the pre-treatment time point, and for patients to be evaluable for immune response (primary endpoint), blood (PBMC and serum) from this time point must be available for analysis. However, immune monitoring will be conducted at the other time points indicated in secondary analyses to evaluate kinetic measures of immunity, and evaluate whether durable immune responses of particular phenotypes are elicited and/or maintained. Assays may be conducted at the time of sample collection (fresh) and/or batched and performed at one time from multiple cryopreserved samples collected at different time points. Other methods of effector and regulatory T-cell response to AR and other human tissue antigens may be used.

[00191] Quantitative assessment of AR-specific CD8+ T-cell effector immunity AR-specific IFNy- and granzyme B-secreting T-cell precursor frequency quantification by ELISPOT: ELISPOT will be used as the preferred methodology, as it permits analysis of low-frequency events (LOD ~ 1 : 100,000 cells) and also permits simultaneous analysis of cryopreserved batched specimens [22]. ΠΤΝΓγ and granzyme B will be preferred analytes evaluated, as these are specifically associated with inflammatory/tissue-destructive (Thl-type, cytolytic) immune responses. Specifically, cryopreserved PBMC from subjects at the various time points will be thawed, rested, and then transferred to 96-well nitrocellulose microtiter (ELISPOT) plates previously coated with monoclonal capture antibodies specific for IFNy or granzyme B. 10 5 cells per well will be cultured in the presence of media (RPMI 1640 supplemented with L-glutamine, penicillin/streptomycin, β- mercaptoethanol and 10% human AB serum) only (no antigen), 2 μg/ml AR protein, 2 μg/ml PSA protein (negative control), 2 μg/ml of peptide libraries specific for AR or control, 250 ng/ml tetanus toxoid, or 2.5 μg/ml PHA (positive mitogenic control) for 24-48 hours. Plates will then be washed with PBS containing 0.05% Tween-20 and incubated for 2.5 hours at room temperature with 50 μΐ/well PBS containing 5 μg/ml biotinylated detection antibodies for either IFNy or granzyme B. After incubation, wells will be washed with PBS, and further incubated with 100 μΐ/well streptavi din-labeled alkaline phosphatase (BioRad, Hercules, CA) and then developed with 100 μΐ/well BCIP/NBT colorimetric substrate (BioRad). The colorimetric reaction will be stopped by rinsing the plates under cool tap water, and wells will be allowed to dry completely before spots are enumerated with an ELISPOT automatic plate reader. [00192] REPORTING AND RESPONSE DEFINITION: Results will be presented as previously reported as the mean (+/- standard deviation) number of spot-forming-units (sfu) per 10 6 cells (frequency), calculated by subtracting the mean number of spots obtained from the no antigen control wells from the mean number obtained in the experimental wells, normalized to 10 6 starting PBMC, from 8-well replicate assays [23]. Comparison of experimental wells with control, no antigen, wells will be performed using a two-sample t-test, with p < 0.05 (two-sided) defined as a significant antigen-specific T-cell response. A significant antigen-specific response resulting from immunization will then be defined as a AR-specific response detectable at the 6-month post-surgery time point (or other post-treatment time point evaluated) that is significantly higher than to media only (as above), at least 3 -fold higher than the mean baseline value, and with a frequency > 10 per 10 6 PBMC.

[00193] Assessment of antigen-specific antibody immunity: Enzyme-linked immunosorbent assay (ELISA) for the detection of antibodies responses to AR: The presence of a coexisting humoral immune response to AR (or other antigens) will be evaluated by ELISA using an indirect method similar to that described previously [61]. Specifically, Immulon-4 ELISA plates (Dynex Technologies Inc.) will be coated with 2 μg/ml purified AR LBD protein (Research Diagnostics, Inc., or other antigens or commercial sources) in 0.1 M NaHC03/Na2C03 buffer (pH 9.6) overnight at 4°C. After blocking with PBS/1% BSA for 1 hour at room temperature, wells will be washed with PBS + 0.05%) Tween-20 (PBS-Tween) and then incubated for 1 hour with human sera diluted 1 :25, 1 :50, 1 : 100 and 1 :200. After washing, plates will then be sequentially incubated with a peroxidase- conjugated anti-human IgG detection antibody (Amersham), followed by peroxidase enzyme TMB substrate (Kierkegaard and Perry Laboratories). The color reaction will be stopped with IN H2SO4 and the optical density measured at 450 nm. Antibody titers for AR-specific IgG antibodies will be determined as previously described [61].

[00194] REPORTING AND RESPONSE DEFINITION: These are not strictly quantitative assays.

IgG response will be reported graphically demonstrating sera dilution curves, and by titer - defined as the highest sera dilution at which IgG responses are detectable above the mean + 3 standard deviations of the negative control. A positive IgG response resulting from immunization will be defined as an antigen-specific (anti-AR) IgG titer at least 4-fold higher than the baseline titer detectable at the 6-month post-treatment time point (or other post-treatment time point evaluated).

[00195] Histopathology Evaluation: Tissue biopsies obtained pre-treatment and at the time of prostatectomy will be available from all subjects. The purpose of these studies is first to determine whether treatment with androgen deprivation alone (ARM 1) results in an increase in CD8+ T cells, and whether this is further increased by the use of an AR-targeting vaccine (ARM 2) and further yet with pembrolizumab (ARM 3). This will be determined by standard immunohistochemistry, and by quantitative flow cytometry (when feasible with fresh tissue). As an exploratory method, the frequency of CD8+ T cells, relative to other populations, will also be determined by mRNA analysis of frozen or paraffin-embedded tissue samples. As a further exploratory method, the frequency of specific CD8+ T cell populations will be determined by TCR sequencing using frozen tissue samples (Adaptive Biotechnologies, Seattle, WA).

[00196] Secondary goals of the histopathology evaluation will be to determine whether immunization with pTVG-AR affects PD-L1 expression in the tumor (likely by eliciting tumor antigen-specific T cells secreting IFNy), and whether treatment increases expression of other T-cell regulatory ligands on T cells (PD-1, CTLA-4, TEVI-3, BTLA, VISTA, LAG-3) or tumors (e.g. HVEM, phosphatidyl serine, PD-L2). Consequently, biopsy specimens obtained pre-treatment and after 12 weeks will be stained with antibodies specific for CD3, CD4, CD8, FoxP3, PD-1, CTLA-4, ΤΓΜ3, BTLA, VISTA, LAG-3, PD-L1, PD-L2, phosphatidyl serine, HVEM and potentially other markers. Staining and quantification will be reviewed by a pathologist blinded to the treatment groups to determine CD8+ T cells per field, CD4+FoxP3+ (Treg):CD8+ T cell ratio, PD-L1 expression, and whether these or the expression of CD8+ T cells expressing one regulatory receptors (or tumor cells expressing one or more regulatory ligands) change from pre-treatment to the prostatectomy time point.

[00197] At the University of Wisconsin, samples will be transported to the UWCCC TRIP (Translational Research Initiatives in Pathology) lab for formalin fixation, paraffin embedding, sectioning, H&E staining, and ultimately for IHC analysis as described above.

Example 8 [00198] Similar to Example 7, a suitable dosage regimen for the combination treatment using vaccine, ADT and a PD-1 pathway blockage is diagramed in FIG. 16. Suitable testing parameters are shown under the timeline and will be performed as discussed in Example 7.

[00199] Each publication, patent, and patent publication cited in this disclosure is incorporated in reference herein in its entirety. The present invention is not intended to be limited to the foregoing examples, but encompasses all such modifications and variations as come within the scope of the appended claims.

[00200] REFERENCES FROM EXAMPLE 5, each is incorporated by reference in its entirety

1. Colluru VT, Johnson LE, Olson BM, McNeel DG. Preclinical and clinical development of DNA vaccines for prostate cancer. Urol Oncol. 2016;34: 193-204.

2. Hodge JW, Ardiani A, Farsaci B, Kwilas AR, Gameiro SR. The tipping point for combination therapy: cancer vaccines with radiation, chemotherapy, or targeted small molecule inhibitors. Semin Oncol. 2012;39:323-39.

3. Ardiani A, Gameiro SR, Kwilas AR, Donahue RN, Hodge JW. Androgen deprivation therapy sensitizes prostate cancer cells to T-cell killing through androgen receptor dependent modulation of the apoptotic pathway. Oncotarget. 2014;5:9335-48.

4. Mercader M, Bodner BK, Moser MT, Kwon PS, Park ES, Manecke RG, et al. T cell infiltration of the prostate induced by androgen withdrawal in patients with prostate cancer. Proc Natl Acad Sci U S A. 2001;98: 14565-70.

5. Morse MD, McNeel DG. Prostate Cancer Patients Treated with Androgen Deprivation Therapy Develop Persistent Changes in Adaptive Immune Responses. Human immunology. 2010;71 :496-504.

6. Morse MD, McNeel DG. T cells localized to the androgen-deprived prostate are TH1 and TH17 biased. Prostate. 2012;72: 1239-47.

7. Roden AC, Moser MT, Tri SD, Mercader M, Kuntz SM, Dong H, et al. Augmentation of T cell levels and responses induced by androgen deprivation. J Immunol. 2004; 173 :6098-108.

8. Mercader M, Sengupta S, Bodner BK, Manecke RG, Cosar EF, Moser MT, et al. Early effects of pharmacological androgen deprivation in human prostate cancer. BJU Int. 2007;99:60-7. 9. Gannon PO, Poisson AO, Delvoye N, Lapointe R, Mes-Masson AM, Saad F. Characterization of the intra-prostatic immune cell infiltration in androgen-deprived prostate cancer patients. Journal of immunological methods. 2009;348:9-17.

10. Shen YC, Kochel C, Francica B, Alme A, Nirschl C, Nirschl T, et al. Combining androgen deprivation with immune checkpoint blockade delays the development of castration resistance in a murine model of prostate cancer. AACR 106th Annual Meeting 2015; 2015; Philadelphia, PA.

11. Akins EJ, Moore ML, Tang S, Willingham MC, Tooze JA, Dubey P. In situ vaccination combined with androgen ablation and regulatory T-cell depletion reduces castration-resistant tumor burden in prostate-specific pten knockout mice. Cancer Res. 2010;70:3473-82.

12. Drake CG, Doody AD, Mihalyo MA, Huang CT, Kelleher E, Ravi S, et al. Androgen ablation mitigates tolerance to a prostate/prostate cancer-restricted antigen. Cancer cell. 2005;7:239-49.

13. Koh YT, Gray A, Higgins SA, Hubby B, Kast WM. Androgen ablation augments prostate cancer vaccine immunogenicity only when applied after immunization. Prostate. 2009.

14. Ardiani A, Farsaci B, Rogers CJ, Protter A, Guo Z, King TH, et al. Combination therapy with a second-generation androgen receptor antagonist and a metastasis vaccine improves survival in a spontaneous prostate cancer model. Clin Cancer Res. 2013; 19:6205-18.

15. McNeel DG, Smith HA, Eickhoff JC, Lang JM, Staab MJ, Wilding G, et al. Phase I trial of tremelimumab in combination with short-term androgen deprivation in patients with PSA-recurrent prostate cancer. Cancer Immunol Immunother. 2012;61 : 1137-47.

16. Small EJ, Lance RS, Gardner TA, Karsh LI, Fong L, McCoy C, et al. A Randomized Phase II Trial of Sipuleucel-T with Concurrent versus Sequential Abiraterone Acetate plus Prednisone in Metastatic Castration-Resistant Prostate Cancer. Clin Cancer Res. 2015;21 :3862-9.

17. Antonarakis ES, Kibel AS, Yu EY, Karsh LI, Elfiky A, Shore D, et al. Sequencing of Sipuleucel-T and Androgen Deprivation Therapy in Men with Hormone-Sensitive Biochemically- Recurrent Prostate Cancer: A Phase II Randomized Trial. Clin Cancer Res. 2016.

18. Madan RA, Gulley JL, Schlom J, Steinberg SM, Liewehr DJ, Dahut WL, et al. Analysis of overall survival in patients with nonmetastatic castration-resistant prostate cancer treated with vaccine, nilutamide, and combination therapy. Clin Cancer Res. 2008; 14:4526-31. 19. Olson BM, McNeel DG. CD8+ T cells specific for the androgen receptor are common in patients with prostate cancer and are able to lyse prostate tumor cells. Cancer Immunol Immunother. 2011;60:781-92.

20. Olson BM, Johnson LE, McNeel DG. The androgen receptor: a biologically relevant vaccine target for the treatment of prostate cancer. Cancer Immunol Immunother. 2013;62:585-96.

21. Gregory CW, Hamil KG, Kim D, Hall SH, Pretlow TG, Mohler JL, et al. Androgen receptor expression in androgen-independent prostate cancer is associated with increased expression of androgen-regulated genes. Cancer Res. 1998;58:5718-24.

22. Ellis L, Lehet K, Ramakrishnan S, Adelaiye R, Pili R. Development of a castrate resistant transplant tumor model of prostate cancer. Prostate. 2012;72:587-91.

23. Wang S, Gao J, Lei Q, Rozengurt N, Pritchard C, Jiao J, et al. Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer. Cancer cell. 2003;4:209- 21.

24. Hornberg E, Ylitalo EB, Crnalic S, Antti H, Stattin P, Widmark A, et al. Expression of androgen receptor splice variants in prostate cancer bone metastases is associated with castration- resistance and short survival. PloS one. 201 l;6:el9059.

25. Watson PA, Chen YF, Balbas MD, Wongvipat J, Socci ND, Viale A, et al. Constitutively active androgen receptor splice variants expressed in castration-resistant prostate cancer require full- length androgen receptor. Proc Natl Acad Sci U S A. 2010; 107: 16759-65.

26. Smith HA, Cronk RJ, Lang JM, McNeel DG. Expression and Immunotherapeutic Targeting of the SSX Family of Cancer-Testis Antigens in Prostate Cancer. Cancer Res. 2011;71 :6785-95.

27. Fueger BJ, Czernin J, Hildebrandt I, Tran C, Halpern BS, Stout D, et al. Impact of animal handling on the results of 18F-FDG PET studies in mice. J Nucl Med. 2006;47:999-1006.

28. Disselhorst JA, Brom M, Laverman P, Slump CH, Boerman OC, Oyen WJ, et al. Image- quality assessment for several positron emitters using the NEMA NU 4-2008 standards in the Siemens Inveon small-animal PET scanner. J Nucl Med. 2010;51 :610-7.

29. Valkenburg KC, Williams BO. Mouse models of prostate cancer. Prostate cancer. 2011;2011 :895238. 30. Weichert JP, Clark PA, Kandela IK, Vaccaro AM, Clarke W, Longino MA, et al. Alkylphosphocholine analogs for broad-spectrum cancer imaging and therapy. Science translational medicine. 2014;6:240ra75.

31. Graff JN, Alumkal JJ, Drake CG, Thomas GV, Redmond WL, Farhad M, et al. Early evidence of anti-PD-1 activity in enzalutami de-re si stant prostate cancer. Oncotarget. 2016.

32. Pu Y, Xu M, Liang Y, Yang K, Guo Y, Yang X, et al. Androgen receptor antagonists compromise T cell response against prostate cancer leading to early tumor relapse. Science translational medicine. 2016;8:333ra47.

33. Kissick HT, Sanda MG, Dunn LK, Pellegrini KL, On ST, Noel JK, et al. Androgens alter T- cell immunity by inhibiting T-helper 1 differentiation. Proc Natl Acad Sci U S A. 2014; 111 :9887-92.

34. Garnett CT, Palena C, Chakraborty M, Tsang KY, Schlom J, Hodge JW. Sublethal irradiation of human tumor cells modulates phenotype resulting in enhanced killing by cytotoxic T lymphocytes. Cancer Res. 2004;64:7985-94.

35. Gameiro SR, Jammeh ML, Wattenberg MM, Tsang KY, Ferrone S, Hodge JW. Radiation- induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget. 2014;5:403-16.

36. Chakraborty M, Abrams SI, Camphausen K, Liu K, Scott T, Coleman CN, et al. Irradiation of tumor cells up-regulates Fas and enhances CTL lytic activity and CTL adoptive immunotherapy. J Immunol. 2003; 170:6338-47.

37. Chakraborty M, Abrams SI, Coleman CN, Camphausen K, Schlom J, Hodge JW. External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine- mediated T-cell killing. Cancer Res. 2004;64:4328-37.

38. Harris TJ, Hipkiss EL, Borzillary S, Wada S, Grosso JF, Yen HR, et al. Radiotherapy augments the immune response to prostate cancer in a time-dependent manner. Prostate. 2008;68: 1319-29.

39. Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JP, et al. Immune- mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res. 2005; 11 :728-34.

40. Golden EB, Chhabra A, Chachoua A, Adams S, Donach M, Fenton-Kerimian M, et al. Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial. The lancet oncology. 2015; 16:795-803.

41. Slovin SF, Higano CS, Hamid O, Tejwani S, Harzstark A, Alumkal JJ, et al. Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: results from an open-label, multicenter phase I/II study. Ann Oncol. 2013;24: 1813-21.

42. Spratt DE, Evans MJ, Davis BJ, Doran MG, Lee MX, Shah N, et al. Androgen Receptor Upregulation Mediates Radioresi stance after Ionizing Radiation. Cancer Res. 2015;75:4688-96.

43. Drake CG, Sharma P, Gerritsen W. Metastatic castration-resistant prostate cancer: new therapies, novel combination strategies and implications for immunotherapy. Oncogene. 2014;33 :5053-64.

44. Gan L, Chen S, Wang Y, Watahiki A, Bohrer L, Sun Z, et al. Inhibition of the androgen receptor as a novel mechanism of taxol chemotherapy in prostate cancer. Cancer Res. 2009;69:8386- 94.

45. Zhu ML, Horbinski CM, Garzotto M, Qian DZ, Beer TM, Kyprianou N. Tubulin-targeting chemotherapy impairs androgen receptor activity in prostate cancer. Cancer Res. 2010;70:7992- 8002.

46. Sweeney CJ, Chen YH, Carducci M, Liu G, Jarrard DF, Eisenberger M, et al. Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer. The New England journal of medicine. 2015;373 :737-46.

47. Rowe SP, Macura KJ, Mena E, Blackford AL, Nadal R, Antonarakis ES, et al. PSMA-Based [(18)F]DCFPyL PET/CT Is Superior to Conventional Imaging for Lesion Detection in Patients with Metastatic Prostate Cancer. Molecular imaging and biology : MIB : the official publication of the Academy of Molecular Imaging. 2016; 18:411-9.

48. Beattie BJ, Smith-Jones PM, Jhanwar YS, Schoder H, Schmidtlein CR, Morris MJ, et al. Pharmacokinetic assessment of the uptake of 16beta-18F-fluoro-5alpha-dihydrotestosterone (FDHT) in prostate tumors as measured by PET. J Nucl Med. 2010;51 : 183-92.

49. Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, Roeser JC, et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med. 2014;371 : 1028-38. 50. Yu Z, Chen S, Sowalsky AG, Voznesensky OS, Mostaghel EA, Nelson PS, et al. Rapid induction of androgen receptor splice variants by androgen deprivation in prostate cancer. Clin Cancer Res. 2014;20: 1590-600.

[00201] This specification includes the sequence listing that is concurrently filed in computer readable form. This sequence listing is incorporated by reference herein.