Ipilimumab plus nivolumab and DNA-repair defects in AR-V7-expressing metastatic prostate cancer

Karim Boudadi, Daniel L Suzman, Valsamo Anagnostou, Wei Fu, Brandon Luber, Hao Wang, Noushin Niknafs, James R White, John L Silberstein, Rana Sullivan, Donna Dowling, Rana Harb, Thomas R Nirschl, Brendan A Veeneman, Scott A Tomlins, Yipeng Wang, Adam Jendrisak, Ryon P Graf, Ryan Dittamore, Michael A Carducci, Mario A Eisenberger, Michael C Haffner, Alan K Meeker, James R Eshleman, Jun Luo, Victor E Velculescu, Charles G Drake, Emmanuel S Antonarakis, Karim Boudadi, Daniel L Suzman, Valsamo Anagnostou, Wei Fu, Brandon Luber, Hao Wang, Noushin Niknafs, James R White, John L Silberstein, Rana Sullivan, Donna Dowling, Rana Harb, Thomas R Nirschl, Brendan A Veeneman, Scott A Tomlins, Yipeng Wang, Adam Jendrisak, Ryon P Graf, Ryan Dittamore, Michael A Carducci, Mario A Eisenberger, Michael C Haffner, Alan K Meeker, James R Eshleman, Jun Luo, Victor E Velculescu, Charles G Drake, Emmanuel S Antonarakis

Abstract

AR-V7-expressing metastatic prostate cancer is an aggressive phenotype with poor progression-free survival (PFS) and overall survival (OS). Preliminary evidence suggests that AR-V7-positive tumors may be enriched for DNA-repair defects, perhaps rendering them more sensitive to immune-checkpoint blockade. We enrolled 15 metastatic prostate cancer patients with AR-V7-expressing circulating tumor cells into a prospective phase-2 trial. Patients received nivolumab 3 mg/kg plus ipilimumab 1 mg/kg every 3 weeks for four doses, then maintenance nivolumab 3 mg/kg every 2 weeks. Targeted next-generation sequencing was performed to determine DNA-repair deficiency (DRD) status. Outcomes included PSA response rates, objective response rates (ORR), PSA progression-free survival (PSA-PFS), clinical/radiographic PFS and OS. Median age of participants was 65, median PSA was 115 ng/mL, 67% had visceral metastases, and 60% had ≥4 prior systemic therapies. Six of 15 men (40%) had DRD mutations (three in BRCA2, two in ATM, one in ERCC4; none had microsatellite instability). Overall, the PSA response rate was 2/15 (13%), ORR was 2/8 (25%) in those with measurable disease, median PSA-PFS was 3.0 (95%CI 2.1-NR) months, PFS was 3.7 (95%CI 2.8-7.5) months, and OS was 8.2 (95%CI 5.5-10.4) months. Outcomes appeared generally better in DRD+ vs. DRD- tumors with respect to PSA responses (33% vs. 0%; P=0.14, nonsignificant), ORR (40% vs. 0%; P=0.46, nonsignificant), PSA-PFS (HR 0.19; P<0.01, significant), PFS (HR 0.31; P=0.01, significant), and OS (HR 0.41; P=0.11, nonsignificant). There were no new safety concerns. Ipilimumab plus nivolumab demonstrated encouraging efficacy in AR-V7-positive prostate cancers with DRD mutations, but not in the overall study population.

Keywords: AR-V7; DNA repair; ipilimumab; nivolumab; prostate cancer.

Conflict of interest statement

CONFLICTS OF INTEREST E.S.A. is a paid consultant/advisor to Janssen, Astellas, Sanofi, Dendreon, Medivation, ESSA, AstraZeneca, Clovis and Merck; has received research funding to his institution from Janssen, Johnson & Johnson, Sanofi, Dendreon, Genentech, Novartis, Tokai, Bristol Myers-Squibb, AstraZeneca, Clovis and Merck; and is the co-inventor of a biomarker technology that has been licensed to Qiagen. S.A.T. has served as a consultant and received honoraria from Roche/ Ventana Medical Systems, Almac Diagnostics, Janssen, AbbVie and Astellas/Medivation; he is also a co-founder of, consultant for and Laboratory Director of Strata Oncology. Y.W., A.J., R.P.G. and R.D. are employees of Epic Sciences. V.E.V. is a founder of Personal Genome Diagnostics (PGDx), is a member of its Scientific Advisory Board and Board of Directors, and owns PGDx stock, which is subject to certain restrictions under university policy; he is also on the Scientific Advisory Board for Ignyta; the terms of these arrangements are managed by the Johns Hopkins University in accordance with its conflict of interest policies. The remaining authors disclose no relevant conflicts of interest.

Figures

Figure 1. PSA responses and radiographic responses…
Figure 1. PSA responses and radiographic responses according to DRD status
(A) Waterfall plot showing PSA responses according to DRD status. The two patients with PSA50 responses (#4 and #8) both had biallelic BRCA2 gene mutations. Patient #4 had a mixed soft-tissue response (some measurable lesions decreased while others increased) and achieved a durable PFS. Patient #8 did not have any measurable disease, but also achieved a durable PFS, and experienced complete resolution of malignant bone pain (pain score 7/10 decreased to 0/10 after 12 weeks of therapy); he is still alive after 17.5+ months of follow-up. (B) Waterfall plot showing objective RECIST responses according to DRD status. The two patients with soft-tissue responses (#6 and #14) had mutations in ATM and ERCC4, respectively. Patient #6 achieved a durable PFS, and is still alive after 17.9+ months of follow-up. (C) CT scan of radiographic response for patient #6 (with somatic ATM mutation) at baseline and after 24 weeks of treatment. The sum diameter of his target lesions decreased by 52% at the time of his best response. (D). CT scan of radiographic response for patient #14 (with somatic ERCC4 mutation) at baseline and after 9 weeks of treatment. The sum diameter of his target lesions decreased by 75% at the time of his best response.
Figure 2. Time-to-event outcomes, according to DRD…
Figure 2. Time-to-event outcomes, according to DRD status
(A) PSA-PFS, according to DRD status [HR 0.19, 95%CI 0.06–0.62, P=0.0003]. (B) PFS, according to DRD status [HR 0.31, 95%CI 0.10–0.92, P=0.014]. (C) OS, according to DRD status [HR 0.41, 95%CI 0.14–1.21, P=0.11].
Figure 3. Clinical outcomes, according to Shannon…
Figure 3. Clinical outcomes, according to Shannon index (low vs. high)
(A) PSA responses, according to Shannon Index. (B) RECIST responses, according to Shannon index. (C) PSA-PFS, according to Shannon index [HR 0.67, 95%CI 0.23–1.99, P=0.44]. (D) PFS, according to Shannon index [HR 0.43, 95%CI 0.15–1.22, P=0.11]. (E) OS, according to Shannon index [HR 0.34, 95%CI 0.11–0.99, P=0.07].

References

    1. Antonarakis ES, Armstrong AJ, Dehm SM, Luo J. Androgen receptor variant-driven prostate cancer: clinical implications and therapeutic targeting. Prostate Cancer Prostatic Dis. 2016;19:231–41. doi: 10.1038/pcan.2016.17.
    1. Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, Roeser JC, Chen Y, Mohammad TA, Chen Y, Fedor HL, Lotan TL, Zheng Q, De Marzo AM, et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med. 2014;371:1028–38. doi: 10.1056/NEJMoa1315815.
    1. Antonarakis ES, Lu C, Luber B, Wang H, Chen Y, Zhu Y, Silberstein JL, Taylor MN, Maughan BL, Denmeade SR, Pienta KJ, Paller CJ, Carducci MA, et al. Clinical significance of androgen receptor splice variant-7 mRNA detection in circulating tumor cells of men with metastatic castration-resistant prostate cancer treated with first- and second-line abiraterone and enzalutamide. J Clin Oncol. 2017;35:2149–56. doi: 10.1200/JCO.2016.70.1961.
    1. Antonarakis ES, Lu C, Luber B, Wang H, Chen Y, Nakazawa M, Nadal R, Paller CJ, Denmeade SR, Carducci MA, Eisenberger MA, Luo J. Androgen receptor splice variant 7 and efficacy of taxane chemotherapy in patients with metastatic castration-resistant prostate cancer. JAMA Oncol. 2015;1:582–91. doi: 10.1001/jamaoncol.2015.1341.
    1. Scher HI, Lu D, Schreiber NA, Louw J, Graf RP, Vargas HA, Johnson A, Jendrisak A, Bambury R, Danila D, McLaughlin B, Wahl J, Greene SB, et al. Association of AR-V7 on circulating tumor cells as a treatment-specific biomarker with outcomes and survival in castration-resistant prostate cancer. JAMA Oncol. 2016;2:1441–49. doi: 10.1001/jamaoncol.2016.1828.
    1. Topalian SL. Targeting immune checkpoints in cancer therapy. JAMA. 2017;318:1647–48. doi: 10.1001/jama.2017.14155.
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, Ferrucci PF, Hill A, Wagstaff J, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373:23–34. doi: 10.1056/NEJMoa1504030.
    1. Hammers HJ, Plimack ER, Infante JR, Rini BI, McDermott DF, Lewis LD, Voss MH, Sharma P, Pal SK, Razak AR, Kollmannsberger C, Heng DY, Spratlin J, et al. Safety and efficacy of nivolumab in combination with ipilimumab in metastatic renal cell carcinoma: the CheckMate 016 study. J Clin Oncol. 2017;35:3851–58. doi: 10.1200/JCO.2016.72.1985.
    1. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS, Wong F, Azad NS, Rucki AA, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–13. doi: 10.1126/science.aan6733.
    1. McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK, Jamal-Hanjani M, Wilson GA, Birkbak NJ, Hiley CT, Watkins TB, Shafi S, Murugaesu N, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351:1463–69. doi: 10.1126/science.aaf1490.
    1. Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, Mosquera JM, Montgomery B, Taplin ME, Pritchard CC, Attard G, Beltran H, Abida W, Bradley RK, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;161:1215–28. . Erratum in: Cell. 2015; 162:454.
    1. Beer TM, Kwon ED, Drake CG, Fizazi K, Logothetis C, Gravis G, Ganju V, Polikoff J, Saad F, Humanski P, Piulats JM, Gonzalez Mella P, Ng SS, 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;35:40–47. doi: 10.1200/JCO.2016.69.1584.
    1. Hansen A, Massard C, Ott PA, Haas N, Lopez J, Ejadi S, Wallmark J, Keam B, Delord JP, Aggarwal R, Gould M, Qiu P, Saraf S, et al. Pembrolizumab for patients with advanced prostate adenocarcinoma: preliminary results from the KEYNOTE-028 study. Ann Oncol. 2016;27(suppl_6):vi243–65. doi: 10.1093/annonc/mdw372.09.
    1. Joshi H, Pinski JK. Association of AR-V7 expression with molecular and clinical characteristics in prostate cancer. J Clin Oncol. 2016;34:109.
    1. Lokhandwala PM, Riel SL, Haley L, Lu C, Chen Y, Silberstein J, Zhu Y, Zheng G, Lin MT, Gocke CD, Partin AW, Antonarakis ES, Luo J, Eshleman JR. Analytical validation of androgen receptor splice variant 7 detection in a Clinical Laboratory Improvement Amendments (CLIA) laboratory setting. J Mol Diagn. 2017;19:115–25. doi: 10.1016/j.jmoldx.2016.08.003.
    1. Markowski MC, Silberstein JL, Eshleman JR, Eisenberger MA, Luo J, Antonarakis ES. Clinical utility of CLIA-grade AR-V7 testing in patients with metastatic castration-resistant prostate cancer. JCO Precis Oncol. 2017:1–9. doi: 10.1200/PO.17.00127.
    1. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, Verweij J, Van Glabbeke M, van Oosterom AT, Christian MC, Gwyther SG. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000;92:205–16. doi: 10.1093/jnci/92.3.205.
    1. Scher HI, Halabi S, Tannock I, Morris M, Sternberg CN, Carducci MA, Eisenberger MA, Higano C, Bubley GJ, Dreicer R, Petrylak D, Kantoff P, Basch E, et al. Prostate Cancer Clinical Trials Working Group Design and end points of clinical trials for patients with progressive prostate cancer and castrate levels of testosterone: recommendations of the Prostate Cancer Clinical Trials Working Group. J Clin Oncol. 2008;26:1148–59. doi: 10.1200/JCO.2007.12.4487.
    1. Werner SL, Graf RP, Landers M, Valenta DT, Schroeder M, Greene SB, Bales N, Dittamore R, Marrinucci D. Analytical validation and capabilities of the Epic CTC platform: enrichment-free circulating tumour cell detection and characterization. J Circ Biomark. 2015;4:3. doi: 10.5772/60725.
    1. Scher HI, Graf RP, Schreiber NA, McLaughlin B, Jendrisak A, Wang Y, Lee J, Greene S, Krupa R, Lu D, Bamford P, Louw JE, Dugan L, et al. Phenotypic heterogeneity of circulating tumor cells informs clinical decisions between AR signaling inhibitors and taxanes in metastatic prostate cancer. Cancer Res. 2017;77:5687–98. doi: 10.1158/0008-5472.CAN-17-1353.
    1. Abida W, Armenia J, Gopalan A, Brennan R, Walsh M, Barron D, Danila D, Rathkopf D, Morris M, Slovin S, McLaughlin B, Curtis K, Hyman DM, et al. Prospective genomic profiling of prostate cancer across disease states reveals germline and somatic alterations that may affect clinical decision making. JCO Precis Oncol. 2017:1–16. doi: 10.1200/PO.17.00029.
    1. Mateo J, Carreira S, Sandhu S, Miranda S, Mossop H, Perez-Lopez R, Nava Rodrigues D, Robinson D, Omlin A, Tunariu N, Boysen G, Porta N, Flohr P, et al. DNA-repair defects and olaparib in metastatic prostate cancer. N Engl J Med. 2015;373:1697–708. doi: 10.1056/NEJMoa1506859.
    1. Teply BA, Antonarakis ES. Treatment strategies for DNA repair-deficient prostate cancer. Expert Rev Clin Pharmacol. 2017;10:889–98. doi: 10.1080/17512433.2017.1338138.
    1. Schweizer MT, Antonarakis ES. Prognostic and therapeutic implications of DNA repair gene mutations in advanced prostate cancer. Clin Adv Hematol Oncol. 2017;15:785–95.
    1. Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, Berent-Maoz B, Pang J, Chmielowski B, Cherry G, Seja E, Lomeli S, Kong X, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell. 2016;165:35–44. . Erratum in: Cell. 2017; 168:542.
    1. Taylor RA, Fraser M, Livingstone J, Espiritu SM, Thorne H, Huang V, Lo W, Shiah YJ, Yamaguchi TN, Sliwinski A, Horsburgh S, Meng A, Heisler LE, et al. Germline BRCA2 mutations drive prostate cancers with distinct evolutionary trajectories. Nat Commun. 2017;8:13671. doi: 10.1038/ncomms13671.
    1. Nolan E, Savas P, Policheni AN, Darcy PK, Vaillant F, Mintoff CP, Dushyanthen S, Mansour M, Pang JB, Fox SB, Perou CM, Visvader JE, Gray DHD, Kathleen Cuningham Foundation Consortium for Research into Familial Breast Cancer (kConFab) Combined im mune checkpoint blockade as a therapeutic strategy for BRCA1-mutated breast cancer. Sci Transl Med. 2017:9. doi: 10.1126/scitranslmed.aal4922.
    1. Strickland KC, Howitt BE, Shukla SA, Rodig S, Ritterhouse LL, Liu JF, Garber JE, Chowdhury D, Wu CJ, D’Andrea AD, Matulonis UA, Konstantinopoulos PA. Association and prognostic significance of BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer. Oncotarget. 2016;7:13587–98. doi: 10.18632/oncotarget.7277.
    1. Karzai F, Madan RA, Owens H, Couvillon A, Hankin A, Williams M, Bilusic M, Cordes LM, Trepel JB, Killian K, Meltzer PS, Gulley JL, Lee JM, Dahut WL. A phase 2 study of olaparib and durvalumab in metastatic castrate-resistant prostate cancer (mCRPC) in an unselected population. J Clin Oncol. 2018;36:163. doi: 10.1200/JCO.2018.36.6_suppl.163.
    1. Teo MY, Seier K, Ostrovnaya I, Regazzi AM, Kania BE, Moran MM, Cipolla CK, Bluth MJ, Chaim J, Al-Ahmadie H, Snyder A, Carlo MI, Solit DB, et al. Alterations in DNA damage response and repair genes as potential marker of clinical benefit from PD-1/PD-L1 blockade in advanced urothelial cancers. J Clin Oncol. 2018;36:1685–1694. doi: 10.1200/JCO.2017.75.7740.

Source: PubMed

3
購読する