The ProBio trial: molecular biomarkers for advancing personalized treatment decision in patients with metastatic castration-resistant prostate cancer

Alessio Crippa, Bram De Laere, Andrea Discacciati, Berit Larsson, Jason T Connor, Erin E Gabriel, Camilla Thellenberg, Elin Jänes, Gunilla Enblad, Anders Ullen, Marie Hjälm-Eriksson, Jan Oldenburg, Piet Ost, Johan Lindberg, Martin Eklund, Henrik Grönberg, Alessio Crippa, Bram De Laere, Andrea Discacciati, Berit Larsson, Jason T Connor, Erin E Gabriel, Camilla Thellenberg, Elin Jänes, Gunilla Enblad, Anders Ullen, Marie Hjälm-Eriksson, Jan Oldenburg, Piet Ost, Johan Lindberg, Martin Eklund, Henrik Grönberg

Abstract

Background: Multiple therapies exist for patients with metastatic castration-resistant prostate cancer (mCRPC). However, their improvement on progression-free survival (PFS) remains modest, potentially explained by tumor molecular heterogeneity. Several prognostic molecular biomarkers have been identified for mCRPC that may have predictive potential to guide treatment selection and prolong PFS. We designed a platform trial to test this hypothesis.

Methods: The Prostate-Biomarker (ProBio) study is a multi-center, outcome-adaptive, multi-arm, biomarker-driven platform trial for tailoring treatment decisions for men with mCRPC. Treatment decisions in the experimental arms are based on biomarker signatures defined as mutations in certain genes/pathways suggested in the scientific literature to be important for treatment response in mCRPC. The biomarker signatures are determined by targeted sequencing of circulating tumor and germline DNA using a panel specifically designed for mCRPC.

Discussion: Patients are stratified based on the sequencing results and randomized to either current clinical practice (control), where the treating physician decides treatment, or to molecularly driven treatment selection based on the biomarker profile. Outcome-adaptive randomization is implemented to early identify promising treatments for a biomarker signature. Biomarker signature-treatment combinations graduate from the platform when they demonstrate 85% probability of improving PFS compared to the control arm. Graduated combinations are further evaluated in a seamless confirmatory trial with fixed randomization. The platform design allows for new drugs and biomarkers to be introduced in the study.

Conclusions: The ProBio design allows promising treatment-biomarker combinations to quickly graduate from the platform and be confirmed for rapid implementation in clinical care.

Trial registration: ClinicalTrials.gov Identifier NCT03903835. Date of registration: April 4, 2019. Status: Recruiting.

Keywords: Clinical trial platform; Genetic biomarker; Precision medicine; Prostate cancer.

Conflict of interest statement

Henrik Grönberg has received honoraria for giving talks at Janssen, Bayer, and Astellas. All other authors declare no conflict of interest.

Figures

Fig. 1
Fig. 1
Genomic profiling in the ProBio platform trial. Two 10-ml tubes of blood are drawn from each study participant and plasma is enriched. Extraction is performed to obtain cell-free DNA from plasma and germline DNA from white blood cells. Targeted sequencing is applied on both cell-free and germline DNA using the ProBio-panel. The ProBio panel covers mutations in 78 genes, structural variants in 11 genes and allows for interrogation of genome-wide copy-number alterations, microsatellite instability, and hypermutation. Sequence data is processed using an in-house developed bioinformatics infrastructure (https://autoseq-docs.readthedocs.io). All variants are manually examined to remove false positive calls. This information is condensed into a report which contains the biomarker profile for subsequent randomization of the study participants. Study participants that progress are reanalyzed and re-randomized
Fig. 2
Fig. 2
Study design of the ProBio platform trial. Participants who meet the inclusion criteria and agreed to participate in the study are genotyped and their biomarker profile is derived. Based on their biomarker subgroup combination they are randomized to either the control group (standard-of-care) or one of the active arms. Patients are regularly followed through the study. Their outcome data is used to adapt the randomization probabilities, assigning more patients to more beneficial therapies within a biomarker signature. Upon the first progression in the study, patients will be re-genotyped and re-randomized to an alternative arm based on their updated biomarker profile
Fig. 3
Fig. 3
Life cycle of the ProBio platform trial. After written informed consent, the biomarker subgroup combination of the patient is determined and used for randomization to either the control group (standard of care) or one of the experimental arms. Outcome data are updated monthly throughout the trial and will be used to calculate the probabilities of superiority for the active arms over the control group for each biomarker signature of interest. Based on the selected threshold, a decision to continue enrollment or to terminate (for futility or superiority) each treatment-biomarker signature will be made. As treatment-biomarker signatures leave the platform, new treatments can possibly entry in the study. The outcome data is also used to update the randomization probabilities within the biomarker subgroup combinations. Graduating treatment-biomarker signatures will enter a confirmatory trial to validate the hypotheses generated from the platform

References

    1. Nuhn P, De Bono JS, Fizazi K, Freedland SJ, Grilli M, Kantoff PW, et al. Update on systemic prostate cancer therapies: management of metastatic castration-resistant prostate vancer in the era of precision oncology. Eur Urol. 2019;75(1):88–99.
    1. Cornford P, Bellmunt J, Bolla M, Briers E, De Santis M, Gross T, et al. EAU-ESTRO-SIOG guidelines on prostate cancer. Part II: treatment of relapsing, metastatic, and castration-resistant prostate cancer. Eur Urol. 2017;71(4):630–642.
    1. Barbieri CE, Chinnaiyan AM, Lerner SP, Swanton C, Rubin MA. The emergence of precision urologic oncology: a collaborative review on biomarker-driven therapeutics. Eur Urol. 2017;71(2):237–246.
    1. Tan DSW, Thomas GV, Garrett MD, Banerji U, de Bono JS, Kaye SB, et al. Biomarker-driven early clinical trials in oncology: a paradigm shift in drug development. Cancer J. 2009;15(5):406–420.
    1. Simon R. Biomarker based clinical trial design. Chin Clin Oncol. 2014;3(3):8.
    1. Berry SM, Connor JT, Lewis RJ. The platform trial: an efficient strategy for evaluating multiple treatments. JAMA. 2015;313(16):1619–1620.
    1. Berry DA. Adaptive clinical trials in oncology. Nat Rev Clin Oncol. 2012;9(4):199–207.
    1. Culp MB, Soerjomataram I, Efstathiou JA, Bray F, Jemal A. Recent global patterns in prostate cancer incidence and mortality rates. Eur Urol. 2020;77(1):38-52.
    1. Scher HI, Sawyers CL. Biology of progressive, castration-resistant prostate cancer: directed therapies targeting the androgen-receptor signaling axis. J Clin Oncol. 2005;23(32):8253–8261.
    1. Crawford ED, Higano CS, Shore ND, Hussain M, Petrylak DP. Treating patients with metastatic castration resistant prostate cancer: a comprehensive review of available therapies. J Urol. 2015; [cited 2019 Apr 12]; Available from: .
    1. Spritzer CE, Afonso PD, Vinson EN, Turnbull JD, Morris KK, Foye A, et al. Bone marrow biopsy: RNA isolation with expression profiling in men with metastatic castration-resistant prostate cancer--factors affecting diagnostic success. Radiology. 2013;269(3):816–823.
    1. Bova GS, Kallio HML, Annala M, Kivinummi K, Högnäs G, Häyrynen S, et al. Integrated clinical, whole-genome, and transcriptome analysis of multisampled lethal metastatic prostate cancer. Cold Spring Harb Mol Case Stud. 2016;2(3):78–86. Available from: . [cited 2019 Jul 22].
    1. Gundem G, Van Loo P, Kremeyer B, Alexandrov LB, Tubio JMC, Papaemmanuil E, et al. The evolutionary history of lethal metastatic prostate cancer. Nature. 2015;520(7547):353–357.
    1. Siravegna G, Marsoni S, Siena S, Bardelli A. Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol. 2017;14(9):531–548.
    1. Wan JCM, Massie C, Garcia-Corbacho J, Mouliere F, Brenton JD, Caldas C, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer. 2017;17(4):223–238.
    1. Wyatt AW, Annala M, Aggarwal R, Beja K, Feng F, Youngren J, et al. Concordance of circulating tumor DNA and matched metastatic tissue biopsy in prostate cancer. J Natl Cancer Inst. 2017;109(12):78–86.
    1. Romanel A, Tandefelt DG, Conteduca V, Jayaram A, Casiraghi N, Wetterskog D, et al. Plasma AR and abiraterone-resistant prostate cancer. Sci Transl Med. 2015;7(312):312re10.
    1. Goodall J, Mateo J, Yuan W, Mossop H, Porta N, Miranda S, et al. Circulating cell-free DNA to guide prostate cancer treatment with PARP inhibition. Cancer Discov. 2017;7(9):1006–1017.
    1. Frenel JS, Carreira S, Goodall J, Roda D, Perez-Lopez R, Tunariu N, et al. Serial next generation sequencing of circulating cell free DNA evaluating tumour clone response to molecularly targeted drug administration. Clin Cancer Res. 2015;21(20):4586–4596.
    1. Terada N, Akamatsu S, Kobayashi T, Inoue T, Ogawa O, Antonarakis ES. Prognostic and predictive biomarkers in prostate cancer: latest evidence and clinical implications. Ther Adv Med Oncol. 2017;9(8):565–573.
    1. Prostate Cancer Biomarker Enrichment and Treatment Selection - Full Text View - [Internet]. [cited 2019 Oct 21]. Available from: .
    1. Berry DA. The Brave New World of clinical cancer research: adaptive biomarker-driven trials integrating clinical practice with clinical research. Mol Oncol. 2015;9(5):951–959.
    1. Kaplan R, Maughan T, Crook A, Fisher D, Wilson R, Brown L, et al. Evaluating many treatments and biomarkers in oncology: a new design. J Clin Oncol. 2013;31(36):4562–4568.
    1. Saville BR, Berry SM. Efficiencies of platform clinical trials: a vision of the future. Clinical Trials. 2016;13(3):358–366.
    1. Berry DA. Bayesian clinical trials. Nat Rev Drug Discov. 2006;5(1):27–36.
    1. Mota JM, Barnett E, Nauseef JT, Nguyen B, Stopsack KH, Wibmer A, et al. Platinum-based chemotherapy in metastatic prostate cancer with DNA repair gene alterations. JCO Precision Oncol. 2020;16 [cited 2020 May 12]; Available from: .
    1. Mayrhofer M, De Laere B, Whitington T, Van Oyen P, Ghysel C, Ampe J, et al. Cell-free DNA profiling of metastatic prostate cancer reveals microsatellite instability, structural rearrangements and clonal hematopoiesis. Genome Med. 2018;21:10.
    1. De Laere B, Oeyen S, Mayrhofer M, Whitington T, van Dam P-J, Van Oyen P, et al. TP53 outperforms other androgen receptor biomarkers to predict abiraterone or enzalutamide outcome in metastatic castration-resistant prostate cancer. Clin Cancer Res. 2019;25(6):1766–1773.
    1. Robinson D, Van Allen EM, Wu Y-M, Schultz N, Lonigro RJ, Mosquera J-M, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;162(2):454.
    1. Haffner MC, Mosbruger T, Esopi DM, Fedor H, Heaphy CM, Walker DA, et al. Tracking the clonal origin of lethal prostate cancer. J Clin Invest. 2013;123(11):4918–4922.
    1. 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(11):1028–1038.
    1. Armstrong AJ, Halabi S, Luo J, Nanus DM, Giannakakou P, Szmulewitz RZ, et al. Prospective multicenter validation of androgen receptor splice variant 7 and hormone therapy resistance in high-risk castration-resistant prostate cancer: the PROPHECY study. JCO. 2019;37(13):1120–1129.
    1. Plymate SR, Sharp A, de Bono JS. Nuclear circulating tumor cell androgen receptor variant 7 in castration-resistant prostate cancer: the devil is in the detail. JAMA Oncol. 2018;4(9):1187–1188.
    1. Bernemann C, Schnoeller TJ, Luedeke M, Steinestel K, Boegemann M, Schrader AJ, et al. Expression of AR-V7 in circulating tumour cells does not preclude response to next generation androgen deprivation therapy in patients with castration resistant prostate cancer. Eur Urol. 2017;71(1):1–3.
    1. Antonarakis ES, Lu C, Luber B, Wang H, Chen Y, Zhu Y, 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(19):2149–2156.
    1. Mateo J, Carreira S, Sandhu S, Miranda S, Mossop H, Perez-Lopez R, et al. DNA-repair defects and olaparib in metastatic prostate cancer. N Engl J Med. 2015;373(18):1697–1708.
    1. Pomerantz MM, Spisák S, Jia L, Cronin AM, Csabai I, Ledet E, et al. The association between germline BRCA2 variants and sensitivity to platinum-based chemotherapy among men with metastatic prostate cancer. Cancer. 2017;123(18):3532–3539.
    1. Cheng HH, Pritchard CC, Boyd T, Nelson PS, Montgomery B. Biallelic inactivation of BRCA2 in platinum-sensitive metastatic castration-resistant prostate cancer. Eur Urol. 2016;69(6):992–995.
    1. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–2520.
    1. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357(6349):409–413.
    1. Robinson DR, Wu Y-M, Lonigro RJ, Vats P, Cobain E, Everett J, et al. Integrative clinical genomics of metastatic cancer. Nature. 2017;548(7667):297–303.
    1. Cabel L, Loir E, Gravis G, Lavaud P, Massard C, Albiges L, et al. Long-term complete remission with Ipilimumab in metastatic castrate-resistant prostate cancer: case report of two patients. J Immunother Cancer. 2017;18:5.
    1. Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med. 2017;23(6):703–713.
    1. Abida W, Cheng ML, Armenia J, Middha S, Autio KA, Vargas HA, et al. Analysis of the prevalence of microsatellite instability in prostate cancer and response to immune checkpoint blockade. JAMA Oncol. 2019;5(4):471–478.
    1. Abeshouse A, Ahn J, Akbani R, Ally A, Amin S, Andry CD, Annala M, Aprikian A, Armenia J, Arora A, Auman JT. The molecular taxonomy of primary prostate cancer. Cell. 2015;163(4):1011–25.
    1. Rajpar S, Carmel A, Merabet Z, Vielh P, Foulon S, Lesaunier F, et al. The benefit of combining docetaxel to androgen deprivation therapy in localized and metastatic castration-sensitive prostate cancer as predicted by ERG status: An analysis of two GETUG phase III trials. JCO. 2017;35(15_suppl):5012.
    1. Chung JH, Dewal N, Sokol E, Mathew P, Whitehead R, Millis SZ, et al. Prospective comprehensive genomic profiling of primary and metastatic prostate tumors. JCO Precision Oncol. 2019;10 [cited 2019 Sep 17]; Available from: .
    1. Barker AD, Sigman CC, Kelloff GJ, Hylton NM, Berry DA, Esserman LJ. I-SPY 2: an adaptive breast cancer trial design in the setting of neoadjuvant chemotherapy. Clin Pharmacol Ther. 2009;86(1):97–100.
    1. Annala M, Vandekerkhove G, Khalaf D, Taavitsainen S, Beja K, Warner EW, et al. Circulating tumor DNA genomics correlate with resistance to abiraterone and enzalutamide in prostate cancer. Cancer Discov. 2018;8(4):444–457.
    1. Scher HI, Morris MJ, Stadler WM, Higano C, Basch E, Fizazi K, et al. Trial design and objectives for castration-resistant prostate cancer: updated recommendations from the prostate cancer clinical trials working group 3. J Clin Oncol. 2016;34(12):1402–1418.
    1. Korn EL, Freidlin B. Outcome-adaptive randomization: is it useful? J Clin Oncol. 2011;29(6):771–776.
    1. Wathen JK, Thall PF. A simulation study of outcome adaptive randomization in multi-arm clinical trials. Clin Trials. 2017;14(5):432–440.
    1. Buyse M, Saad ED, Burzykowski T. Adaptive randomization of neratinib in early breast cancer. N Engl J Med. 2016;375(16):1591–1592.
    1. Viele K, Broglio K, McGlothlin A, Saville BR. Comparison of methods for control allocation in multiple arm studies using response adaptive randomization. Clin Trials. 2020;17(1):52–60.
    1. Wason JMS, Trippa L. A comparison of Bayesian adaptive randomization and multi-stage designs for multi-arm clinical trials. Stat Med. 2014;33(13):2206–2221.
    1. Trippa L, Lee EQ, Wen PY, Batchelor TT, Cloughesy T, Parmigiani G, et al. Bayesian adaptive randomized trial design for patients with recurrent glioblastoma. J Clin Oncol. 2012;30(26):3258–3263.
    1. Harrington D, Parmigiani G. Adaptive randomization of neratinib in early breast cancer. N Engl J Med. 2016;375(16):1593–1594.
    1. London AJ. Learning health systems, clinical equipoise and the ethics of response adaptive randomisation. J Med Ethics. 2018;44(6):409–415.

Source: PubMed

3
Abonner