A phase II single-arm study of pembrolizumab with enzalutamide in men with metastatic castration-resistant prostate cancer progressing on enzalutamide alone

Julie N Graff, Tomasz M Beer, Joshi J Alumkal, Rachel E Slottke, William L Redmond, George V Thomas, Reid F Thompson, Mary A Wood, Yoshinobu Koguchi, Yiyi Chen, Emile Latour, Raymond C Bergan, Charles G Drake, Amy E Moran, Julie N Graff, Tomasz M Beer, Joshi J Alumkal, Rachel E Slottke, William L Redmond, George V Thomas, Reid F Thompson, Mary A Wood, Yoshinobu Koguchi, Yiyi Chen, Emile Latour, Raymond C Bergan, Charles G Drake, Amy E Moran

Abstract

Background: Checkpoint inhibitors can induce profound anticancer responses, but programmed cell death protein-1 (PD-1) inhibition monotherapy has shown minimal activity in prostate cancer. A published report showed that men with prostate cancer who were resistant to the second-generation androgen receptor inhibitor enzalutamide had increased programmed death-ligand 1 (PD-L1) expression on circulating antigen-presenting cells. We hypothesized that the addition of PD-1 inhibition in these patients could induce a meaningful cancer response.

Methods: We evaluated enzalutamide plus the PD-1 inhibitor pembrolizumab in a single-arm phase II study of 28 men with metastatic castration-resistant prostate cancer (mprogressing on enzalutamide alone. Pembrolizumab 200 mg intravenous was given every 3 weeks for four doses with enzalutamide. The primary endpoint was prostate-specific antigen (PSA) decline of ≥50%. Secondary endpoints were objective response, PSA progression-free survival (PFS), time to subsequent treatment, and time to death. Baseline tumor biopsies were obtained when feasible, and samples were sequenced and evaluated for the expression of PD-L1, microsatellite instability (MSI), mutational and neoepitope burdens.

Results: Five (18%) of 28 patients had a PSA decline of ≥50%. Three (25%) of 12 patients with measurable disease at baseline achieved an objective response. Of the five responders, two continue with PSA and radiographic response after 39.3 and 37.8 months. For the entire cohort, median follow-up was 37 months, and median PSA PFS time was 3.8 months (95% CI: 2.8 to 9.9 months). Time to subsequent treatment was 7.21 months (95% CI: 5.1 to 11.1 months). Median overall survival for all patients was 21.9 months (95% CI: 14.7 to 28 .4 months), versus 41.7 months (95% CI: 22.16 to not reached (NR)) in the responders. Of the three responders with baseline biopsies, one had MSI high disease with mutations consistent with DNA-repair defects. None had detectable PD-L1 expression.

Conclusions: Pembrolizumab has activity in mCRPC when added to enzalutamide. Responses were deep and durable and did not require tumor PD-L1 expression or DNA-repair defects.

Trial registration number: clinicaltrials.gov (NCT02312557).

Keywords: biomarkers; combination; drug therapy; immunotherapy; lymphocytes; prostatic neoplasms; tumor; tumor-infiltrating.

Conflict of interest statement

Competing interests: JG has received research funding from Astellas/Medivation, Merck, Sanofi, Janssen Biotech, and travel support from Sanofi. CD has received research funding from Bristol Myers Squibb (BMS). He has received consulting fees from BMS, Merck, AstraZeneca (AZ) and Medimmune. He has patents licensed to AZ, BMS and Medimmune. WLR has received research grants, consulting fees, and/or royalties from Bristol-Myers Squibb, Merck, Galectin Therapeutics, and Nektar Therapeutics. TMB has research funding from Astellas/Medivation, Alliance Foundation Trials, Boehringer Ingelheim, Corcept Therapeutics, Endocyte Inc, Janssen Research & Development, OncoGenex, Sotio, and Theraclone Sciences/OncoResponse. TMB receives consulting fees from AbbVie, AstraZeneca, Astellas Pharma, Bayer, Boehringer Ingelheim, Clovis Oncology, GlaxoSmithKline, Janssen Biotech, Janssen Japan, Merck, and Pfizer and has stock ownership in Salarius Pharmaceuticals, and Arvinas Inc. JJA, GVT, and RS have no conflicts. RCB is a co-owner of Third Coast Therapeutics Inc, which has an option to license patents of which he is an owner. AEM has a sponsored research agreement with AstraZeneca and has received reagents from Genetech. JJA has received consulting fees from Merck.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Radiographic and PSA changes. (A) Radiographs of the responders who had measurable disease. (B) Spider plot for changes in radiographic response for participants with measurable disease. (C) Changes in PSA for all participants. PSA, prostate-specific antigen; RECIST PD, RECIST progressive disease; RECIST SD, RECIST stable disease.
Figure 2
Figure 2
Distribution of mutational burden in responder’s versus non-responders. Boxplot showing the distributions of mutational burden (left) and neoepitope count (right) for responders versus non-responders. The center line in each box represents the median, with bottom and top boundaries of the box representing the first and third quartiles, respectively. Lines extending down and up from the box represent the minimum and maximum values, respectively. Non-responders are shown in light blue, and responders are shown in dark blue. There was no statistically significant difference between responders and non-responders in terms of mutational burden (median 6.5 vs 4.9 variants per megabase pair covered, p=0.42) or predicted neoepitope burden (679 vs 405.5 neoepitopes, p=0.42).
Figure 3
Figure 3
Pembrolizumab plus enzalutamide treatment induced systemic immune responses. (A) Serum CXCL10 levels pretreatment (C1D1; Cycle 1 Day 1) and post-treatment (C2D1, Cycle 2 Day1; C3D1, Cycle 3 Day 1) are shown (n=22). (B) The percent expression of CD38, HLA-DR, or Ki-67 in EM CD4 or CD8 T cells is shown (n=18). EM, effector memory; NS, not significant.
Figure 4
Figure 4
The functional status of EM CD8 T cells as a potential immune correlate with clinical response and outcome. (A) The percent expression of GzmB or perforin in EM CD8 T cells is compared between non-responders (NR; n=14) and responders (R; n=4). (B) Kaplan-Meier curves demonstrating that patients with higher median percentage of perforin, but not GzmB, EM CD8 T cells at baseline had prolonged PFS. (C) The frequency of MDSCs at baseline did not correlate with clinical response or outcome. EM, effector memory; GzmB, granzyme B; MDSCs, myeloid-derived suppressor cells; PFS, progression-free survival.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin 2018;68:7–30. 10.3322/caac.21442
    1. Beer TM, Armstrong AJ, Rathkopf DE, et al. . Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med 2014;371:424–33. 10.1056/NEJMoa1405095
    1. de Bono JS, Logothetis CJ, Molina A, et al. . Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med 2011;364:1995–2005. 10.1056/NEJMoa1014618
    1. Ryan CJ, Smith MR, de Bono JS, et al. . Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med 2013;368:138–48. 10.1056/NEJMoa1209096
    1. Scher HI, Fizazi K, Saad F, et al. . Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med 2012;367:1187–97. 10.1056/NEJMoa1207506
    1. Tannock IF, de Wit R, Berry WR, et al. . Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med 2004;351:1502–12. 10.1056/NEJMoa040720
    1. de Bono JS, Oudard S, Ozguroglu M, et al. . Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet 2010;376:1147–54. 10.1016/S0140-6736(10)61389-X
    1. Kantoff PW, Higano CS, Shore ND, et al. . Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411–22. 10.1056/NEJMoa1001294
    1. Parker C, Nilsson S, Heinrich D, et al. . Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med 2013;369:213–23. 10.1056/NEJMoa1213755
    1. Beer TM, Kwon ED, Drake CG, et al. . Randomized, double-blind, phase III trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic Chemotherapy-Naive castration-resistant prostate cancer. J Clin Oncol 2017;35:40–7. 10.1200/JCO.2016.69.1584
    1. Kwon ED, Drake CG, Scher HI, et al. . Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol 2014;15:700–12. 10.1016/S1470-2045(14)70189-5
    1. Topalian SL, Hodi FS, Brahmer JR, et al. . Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366:2443–54. 10.1056/NEJMoa1200690
    1. Hansen AR, Massard C, Ott PA, et al. . Pembrolizumab for advanced prostate adenocarcinoma: findings of the KEYNOTE-028 study. Ann Oncol 2018;29:1807–13. 10.1093/annonc/mdy232
    1. De Bono JS, Goh JCH, Ojamaa K, et al. . KEYNOTE-199: pembrolizumab (pembro) for docetaxel-refractory metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol 2018;36:5007 10.1200/JCO.2018.36.15_suppl.5007
    1. Karzai F, VanderWeele D, Madan RA, et al. . Activity of durvalumab plus olaparib in metastatic castration-resistant prostate cancer in men with and without DNA damage repair mutations. J Immunother Cancer 2018;6:141. 10.1186/s40425-018-0463-2
    1. Bishop JL, Sio A, Angeles A, et al. . PD-L1 is highly expressed in enzalutamide resistant prostate cancer. Oncotarget 2015;6:234–42. 10.18632/oncotarget.2703
    1. Martin AM, Nirschl TR, Nirschl CJ, et al. . Paucity of PD-L1 expression in prostate cancer: innate and adaptive immune resistance. Prostate Cancer Prostatic Dis 2015;18:325–32. 10.1038/pcan.2015.39
    1. Haffner MC, Guner G, Taheri D, et al. . Comprehensive evaluation of programmed Death-Ligand 1 expression in primary and metastatic prostate cancer. Am J Pathol 2018;188:1478–85. 10.1016/j.ajpath.2018.02.014
    1. Graff JN, Drake CG, Beer TM. Complete biochemical (prostate-specific antigen) response to sipuleucel-T with enzalutamide in castration-resistant prostate cancer: a case report with implications for future research. Urology 2013;81:381–3. 10.1016/j.urology.2012.10.044
    1. U.S. Food & Drug Administration FDA grants accelerated approval to pembrolizumab for first tissue/site agnostic indication, 2017. Available:
    1. Knight Diagnostic Laboratory’s (KDL) GeneTrailsTM solid tumor panel.
    1. Personal Genome Diagnostics.
    1. Salipante SJ, Scroggins SM, Hampel HL, et al. . Microsatellite instability detection by next generation sequencing. Clin Chem 2014;60:1192–9. 10.1373/clinchem.2014.223677
    1. Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 2010;26:841–2. 10.1093/bioinformatics/btq033
    1. Szolek A, Schubert B, Mohr C, et al. . OptiType: precision HLA typing from next-generation sequencing data. Bioinformatics 2014;30:3310–6. 10.1093/bioinformatics/btu548
    1. BMH EK, Saksena G, Covington KR, et al. . Automating somatic mutation calling for ten thousand tumor Exomes.
    1. McKenna A, Hanna M, Banks E, et al. . The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 2010;20:1297–303. 10.1101/gr.107524.110
    1. Edge P, Bafna V, Bansal V. HapCUT2: robust and accurate haplotype assembly for diverse sequencing technologies. Genome Res 2017;27:801–12. 10.1101/gr.213462.116
    1. Wood MA, Nguyen A, Struck AJ, et al. . neoepiscope improves neoepitope prediction with multivariant phasing. Bioinformatics 2020;36:713–20. 10.1093/bioinformatics/btz653
    1. Sun J-M, Zhou W, Choi Y-L, et al. . Prognostic significance of PD-L1 in patients with non-small cell lung cancer: a large cohort study of surgically resected cases. J Thorac Oncol 2016;11:1003–11. 10.1016/j.jtho.2016.04.007
    1. Koguchi Y, Gonzalez IL, Meeuwsen TL, et al. . A semi-automated approach to preparing antibody cocktails for immunophenotypic analysis of human peripheral blood. J Vis Exp 2016;108:e53485. 10.3791/53485
    1. Franciszkiewicz K, Boissonnas A, Boutet M, et al. . Role of chemokines and chemokine receptors in shaping the effector phase of the antitumor immune response. Cancer Res 2012;72:6325–32. 10.1158/0008-5472.CAN-12-2027
    1. Pritchard CC, Morrissey C, Kumar A, et al. . Complex MSH2 and MSH6 mutations in hypermutated microsatellite unstable advanced prostate cancer. Nat Commun 2014;5:4988. 10.1038/ncomms5988
    1. Hause RJ, Pritchard CC, Shendure J, et al. . Classification and characterization of microsatellite instability across 18 cancer types. Nat Med 2016;22:1342–50. 10.1038/nm.4191
    1. Wu Y-M, Cieślik M, Lonigro RJ, et al. . Inactivation of CDK12 delineates a distinct immunogenic class of advanced prostate cancer. Cell 2018;173:e14:1770–82. 10.1016/j.cell.2018.04.034
    1. Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther 2015;14:847–56. 10.1158/1535-7163.MCT-14-0983
    1. Boudadi K, Suzman DL, Anagnostou V, et al. . Ipilimumab plus nivolumab and DNA-repair defects in AR-V7-expressing metastatic prostate cancer. Oncotarget 2018;9:28561–71. 10.18632/oncotarget.25564
    1. Sharma P, Pachynski RK, Narayan V, et al. . Initial results from a phase II study of nivolumab (NIVO) plus ipilimumab (IPI) for the treatment of metastatic castration-resistant prostate cancer (mCRPC; CheckMate 650). J Clin Oncol 2019;37:142 10.1200/JCO.2019.37.7_suppl.142
    1. Pichler R, Horninger W, Heidegger I. ASCO 2018: highlights of urothelial cancer and prostate cancer. Memo 2018;11:284–90. 10.1007/s12254-018-0422-0
    1. Karzai F, Madan R, Owens H, et al. . Combination of PDL-1 and PARP inhibition in an unselected population with metastatic castrate-resistant prostate cancer (mCRPC). ASCO Annual Meeting; June 2–6, 2017.

Source: PubMed

3
Tilaa