Olaparib in patients with metastatic castration-resistant prostate cancer with DNA repair gene aberrations (TOPARP-B): a multicentre, open-label, randomised, phase 2 trial

Joaquin Mateo, Nuria Porta, Diletta Bianchini, Ursula McGovern, Tony Elliott, Robert Jones, Isabel Syndikus, Christy Ralph, Suneil Jain, Mohini Varughese, Omi Parikh, Simon Crabb, Angus Robinson, Duncan McLaren, Alison Birtle, Jacob Tanguay, Susana Miranda, Ines Figueiredo, George Seed, Claudia Bertan, Penny Flohr, Berni Ebbs, Pasquale Rescigno, Gemma Fowler, Ana Ferreira, Ruth Riisnaes, Rita Pereira, Andra Curcean, Robert Chandler, Matthew Clarke, Bora Gurel, Mateus Crespo, Daniel Nava Rodrigues, Shahneen Sandhu, Aude Espinasse, Peter Chatfield, Nina Tunariu, Wei Yuan, Emma Hall, Suzanne Carreira, Johann S de Bono, Joaquin Mateo, Nuria Porta, Diletta Bianchini, Ursula McGovern, Tony Elliott, Robert Jones, Isabel Syndikus, Christy Ralph, Suneil Jain, Mohini Varughese, Omi Parikh, Simon Crabb, Angus Robinson, Duncan McLaren, Alison Birtle, Jacob Tanguay, Susana Miranda, Ines Figueiredo, George Seed, Claudia Bertan, Penny Flohr, Berni Ebbs, Pasquale Rescigno, Gemma Fowler, Ana Ferreira, Ruth Riisnaes, Rita Pereira, Andra Curcean, Robert Chandler, Matthew Clarke, Bora Gurel, Mateus Crespo, Daniel Nava Rodrigues, Shahneen Sandhu, Aude Espinasse, Peter Chatfield, Nina Tunariu, Wei Yuan, Emma Hall, Suzanne Carreira, Johann S de Bono

Abstract

Background: Metastatic castration-resistant prostate cancer is enriched in DNA damage response (DDR) gene aberrations. The TOPARP-B trial aims to prospectively validate the association between DDR gene aberrations and response to olaparib in metastatic castration-resistant prostate cancer.

Methods: In this open-label, investigator-initiated, randomised phase 2 trial following a selection (or pick-the-winner) design, we recruited participants from 17 UK hospitals. Men aged 18 years or older with progressing metastatic castration-resistant prostate cancer previously treated with one or two taxane chemotherapy regimens and with an Eastern Cooperative Oncology Group performance status of 2 or less had tumour biopsies tested with targeted sequencing. Patients with DDR gene aberrations were randomly assigned (1:1) by a computer-generated minimisation method, with balancing for circulating tumour cell count at screening, to receive 400 mg or 300 mg olaparib twice daily, given continuously in 4-week cycles until disease progression or unacceptable toxicity. Neither participants nor investigators were masked to dose allocation. The primary endpoint of confirmed response was defined as a composite of all patients presenting with any of the following outcomes: radiological objective response (as assessed by Response Evaluation Criteria in Solid Tumors 1.1), a decrease in prostate-specific antigen (PSA) of 50% or more (PSA50) from baseline, or conversion of circulating tumour cell count (from ≥5 cells per 7·5 mL blood at baseline to <5 cells per 7·5 mL blood). A confirmed response in a consecutive assessment after at least 4 weeks was required for each component. The primary analysis was done in the evaluable population. If at least 19 (43%) of 44 evaluable patients in a dose cohort responded, then the dose cohort would be considered successful. Safety was assessed in all patients who received at least one dose of olaparib. This trial is registered at ClinicalTrials.gov, NCT01682772. Recruitment for the trial has completed and follow-up is ongoing.

Findings: 711 patients consented for targeted screening between April 1, 2015, and Aug 30, 2018. 161 patients had DDR gene aberrations, 98 of whom were randomly assigned and treated (49 patients for each olaparib dose), with 92 evaluable for the primary endpoint (46 patients for each olaparib dose). Median follow-up was 24·8 months (IQR 16·7-35·9). Confirmed composite response was achieved in 25 (54·3%; 95% CI 39·0-69·1) of 46 evaluable patients in the 400 mg cohort, and 18 (39·1%; 25·1-54·6) of 46 evaluable patients in the 300 mg cohort. Radiological response was achieved in eight (24·2%; 11·1-42·3) of 33 evaluable patients in the 400 mg cohort and six (16·2%; 6·2-32·0) of 37 in the 300 mg cohort; PSA50 response was achieved in 17 (37·0%; 23·2-52·5) of 46 and 13 (30·2%; 17·2-46·1) of 43; and circulating tumour cell count conversion was achieved in 15 (53·6%; 33·9-72·5) of 28 and 13 (48·1%; 28·7-68·1) of 27. The most common grade 3-4 adverse event in both cohorts was anaemia (15 [31%] of 49 patients in the 300 mg cohort and 18 [37%] of 49 in the 400 mg cohort). 19 serious adverse reactions were reported in 13 patients. One death possibly related to treatment (myocardial infarction) occurred after 11 days of treatment in the 300 mg cohort.

Interpretation: Olaparib has antitumour activity against metastatic castration-resistant prostate cancer with DDR gene aberrations, supporting the implementation of genomic stratification of metastatic castration-resistant prostate cancer in clinical practice.

Funding: Cancer Research UK, AstraZeneca, Prostate Cancer UK, the Prostate Cancer Foundation, the Experimental Cancer Medicine Centres Network, and the National Institute for Health Research Biomedical Research Centres.

Copyright © 2020 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
(A) Trial profile and (B) DDR gene alterations in the intention-to-treat population (n=98) DDR=DNA damage response. CTC=circulating tumour cell. *Non-mutually exclusive subgroups: one patient had BRCA1/2, CDK12, and other mutations, and two patients had both PALB2 and other mutations (included in each subgroup). †The BRCA2 K3226* variant is supposedly non-pathogenic and was therefore not considered sufficient for patients to be considered eligible; however, one patient with a BRCA2 K3226* variant was included because of evidence of concomitant loss of the contralateral allele.
Figure 2
Figure 2
Antitumour activity by allocated dose cohort (intention-to-treat population) (A) Best percentage change from baseline in PSA during treatment. (B) Best percentage change from baseline in the sum of target lesions (Response Evaluation Criteria in Solid Tumors 1·1) during treatment. (C) Radiographic progression-free survival. (D) Swimmers plot of time on treatment for each patient, indicating periods of treatment interruptions, dose reductions, and, in the 300 mg cohort, dose escalations. Treatment periods of ≥6 months and ≥12 months are highlighted. PSA=prostate-specific antigen. PSA50=decrease in prostate-specific antigen of ≥50%.
Figure 3
Figure 3
Antitumour activity by gene aberration subgroup (intention-to-treat population, pooled 300 mg and 400 mg cohorts) (A) Maximum percentage change from baseline in PSA during treatment. (B) Maximum percentage change from baseline in the sum of target lesions (Response Evaluation Criteria in Solid Tumors 1·1) during treatment. (C) Radiographic progression-free survival. (D) Swimmers plot of time on treatment for each patient. ITT=intention-to-treat. NE=not estimable. PSA=prostate-specific antigen. PSA50=decrease in prostate-specific antigen of ≥50%. *Patients presenting with mutliple mutations are represented in a single subgroup.

References

    1. Attard G, Parker C, Eeles RA. Prostate cancer. Lancet. 2016;387:70–82.
    1. Armenia J, Wankowicz SAM, Liu D. The long tail of oncogenic drivers in prostate cancer. Nat Genet. 2018;50:645–651.
    1. Robinson D, Van Allen EM, Wu YM. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;162:454.
    1. Quigley DA, Dang HX, Zhao SG. Genomic hallmarks and structural variation in metastatic prostate cancer. Cell. 2018;174:758. 69.e9.
    1. Farmer H, McCabe N, Lord CJ. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature. 2005;434:917–921.
    1. Bryant HE, Schultz N, Thomas HD. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature. 2005;434:913–917.
    1. Kaufman B, Shapira-Frommer R, Schmutzler RK. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol. 2015;33:244–250.
    1. Pujade-Lauraine E, Ledermann JA, Selle F. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2017;18:1274–1284.
    1. Ledermann J, Harter P, Gourley C. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol. 2014;15:852–861.
    1. Murai J, Huang SY, Renaud A. Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib. Mol Cancer Ther. 2014;13:433–443.
    1. Murai J, Huang SY, Das BB. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res. 2012;72:5588–5599.
    1. Mateo J, Carreira S, Sandhu S. DNA-repair defects and olaparib in metastatic prostate cancer. N Engl J Med. 2015;373:1697–1708.
    1. Scher HI, Halabi S, Tannock I. 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;26:1148–1159.
    1. Eisenhauer EA, Therasse P, Bogaerts J. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45:228–247.
    1. Mateo J, Moreno V, Gupta A. An adaptive study to determine the optimal dose of the tablet formulation of the PARP inhibitor olaparib. Target Oncol. 2016;11:401–415.
    1. Seed G, Yuan W, Mateo J. Gene copy number estimation from targeted next-generation sequencing of prostate cancer biopsies: analytic validation and clinical qualification. Clin Cancer Res. 2017;23:6070–6077.
    1. Scher HI, Heller G, Molina A. Circulating tumor cell biomarker panel as an individual-level surrogate for survival in metastatic castration-resistant prostate cancer. J Clin Oncol. 2015;33:1348–1355.
    1. Yap C, Pettitt A, Billingham L. Screened selection design for randomised phase II oncology trials: an example in chronic lymphocytic leukaemia. BMC Med Res Methodol. 2013;13:87.
    1. Meeks HD, Song H, Michailidou K. BRCA2 polymorphic stop codon K3326X and the risk of breast, prostate, and ovarian cancers. J Natl Cancer Inst. 2016;108
    1. Goodall J, Mateo J, Yuan W. Circulating cell-free DNA to guide prostate cancer treatment with PARP inhibition. Cancer Discov. 2017;7:1006–1017.
    1. Sandhu SK, Schelman WR, Wilding G. The poly(ADP-ribose) polymerase inhibitor niraparib (MK4827) in BRCA mutation carriers and patients with sporadic cancer: a phase 1 dose-escalation trial. Lancet Oncol. 2013;14:882–892.
    1. de Bono J, Ramanathan RK, Mina L. Phase I, dose-escalation, two-part trial of the PARP inhibitor talazoparib in patients with advanced germline BRCA1/2 mutations and selected sporadic cancers. Cancer Discov. 2017;7:620–629.
    1. Fong PC, Boss DS, Yap TA. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med. 2009;361:123–134.
    1. Wu YM, Cieslik M, Lonigro RJ. Inactivation of CDK12 delineates a distinct immunogenic class of advanced prostate cancer. Cell. 2018;173:1770. 82.e14.
    1. Tutt A, Robson M, Garber JE. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial. Lancet. 2010;376:235–244.
    1. Audeh MW, Carmichael J, Penson RT. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial. Lancet. 2010;376:245–251.
    1. Pritchard CC, Mateo J, Walsh MF. Inherited DNA-repair gene mutations in men with metastatic prostate cancer. N Engl J Med. 2016;375:443–453.
    1. Alexandrov LB, Nik-Zainal S, Wedge DC. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421.
    1. Cruz C, Castroviejo-Bermejo M, Gutiérrez-Enríquez S. RAD51 foci as a functional biomarker of homologous recombination repair and PARP inhibitor resistance in germline BRCA-mutated breast cancer. Ann Oncol. 2018;29:1203–1210.
    1. Abida W, Bryce AH, Vogelzang NJ. Preliminary results from TRITON2: a phase 2 study of rucaparib in patients with metastatic castration-resistant prostate cancer (mCRPC) associated with homologous recombination repair (HRR) gene alterations. Ann Oncol. 2018;29(suppl 8):vii271–vii302.
    1. Clarke N, Wiechno P, Alekseev B. Olaparib combined with abiraterone in patients with metastatic castration-resistant prostate cancer: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 2018;19:975–986.

Source: PubMed

3
Předplatit