The Case for Platinum Therapy in Prostate Cancer

Publication
Article
Oncology Live Urologists in Cancer Care®April 2018
Volume 7
Issue 2

The arsenal of therapeutic options for men with castration-resistant prostate cancer has steadily grown as ongoing research uncovers more about prostate cancer biology and resistance mechanisms.

William Oh, MD

Past Transplant Experience

As ongoing research continues to deepen our understanding of prostate cancer biology and resistance mechanisms, our arsenal of therapeutic options for men with castration-resistant prostate cancer (CRPC) has steadily grown. Four general classes of therapeutic agents have emerged so far: cytotoxic chemotherapy (docetaxel [Taxotere] and cabazitaxel [Jevtana]), androgen receptor (AR)-pathway directed therapies (abiraterone [Zytiga], enzalutamide [Xtandi], and apalutamide [Erleada]), bone-targeted therapy (radium-223 [Xofigo]), and active cellular immunotherapy (sipuleucel-T [Provenge]). Each of the drugs in these classes has demonstrated significant clinical benefits in randomized phase III clinical trials in CRPC,1-9 but the duration of response for each remains relatively shortlived and the survival benefits are moderate. There remains a great need for additional treatment options. Here we reexamine the case for platinum therapy.Platinum agents have long been studied in metastatic CRPC (mCRPC), but in patient cohorts that are largely molecularly unselected (Table). Trial experience with single-agent carboplatin,10-13 cisplatin,14,15 and oxaliplatin16 have generally yielded modest rates of prostatespecific antigen (PSA) decline, improvement in pain, and radiographic response.17 Although platinum doublet combinations with a taxane,18-20 anthracycline,21,22 etoposide,23,24 or gemcitabine25 are usually associated with slightly higher rates of PSA decline, they are also associated with increased toxicity. Triplet combination regimens, such as paclitaxel/ estramustine/carboplatin26 or docetaxel/estramustine/ carboplatin27-29 have been found to have significant clinical activity. In one study, docetaxel/estramustine/carboplatin was associated with a ≥50% decline in PSA in 95% of enrolled patients.29 However, these studies were relatively small and these regimens have not been further developed. Of the 3 platinum drugs, cisplatin and oxaliplatin use is often limited by renal impairment and neuropathic toxicity. Carboplatin is generally well tolerated, but has a heavier footprint in terms of myelosuppressive effects.

Satraplatin, a fourth-generation oral platinum analogue, was tested in the randomized phase III SPARC study. A total of 950 men with chemotherapy- refractory mCRPC were enrolled and received either satraplatin plus prednisone or placebo plus prednisone. Although satraplatin was associated with a small, but significant, benefit in progression-free survival (11.1 vs 9.7 weeks; HR, 0.67; 95% CI, 0.57-0.77; P <.001), improvement in time to pain progression (66.1 vs 22.3 weeks; HR, 0.64; 95% CI, 0.51-0.79; P <.001), and PSA response (25.4% vs 12.4%; P <.001), there was no demonstrated difference in median overall survival (OS) compared with placebo (61.3 vs 61.4 weeks; HR, 0.98; 95% CI, 0.84-1.15; P = .80).30

Although SPARC was ultimately a negative trial and further development of satraplatin stopped, the results again confirm that a subset of patients with mCRPC is responsive to platinum therapy, even as a single agent. Unfortunately, predictive markers to identify the cohort most likely to benefit from platinum therapy could not be developed from SPARC, as correlative and translational studies were not included in the study design.

Table. Platinum Agents Effective in Treating Subtypes of Prostate Cancer

Neuroendocrine Prostate Cancer

Currently, outside of small cell carcinoma of the prostate, where platinum therapy is considered standard offering based on limited study findings31-33 and an extrapolation of efficacy data in small cell lung cancer, platinum compounds have not been formally adopted into any professional practice guidelines. This is due to a lack of defined OS benefit in the general nonselected population with prostate cancer. It could be argued that adoption of platinum drugs has also been poor due to inadequate characterization of the subtype of patients with CRPC who could benefit from platinum agents. Neuroendocrine prostate cancer (NEPC), a heterogeneous histology34 that most commonly transdifferentiates from preexisting adenocarcinoma as an adaptive response to long-term exposure to androgendeprivation therapy (ADT) and AR blockade,35,36 is classically considered to be responsive to platinumbased therapies. The response rates are as high as 70% in those with poorly differentiated histology.37

DNA Damage Repair Pathway

Histologically, NEPC is archetypally characterized as the presence of small round blue neuroendocrine cells with a lack of AR expression. Phenotypically, NEPC frequently metastasizes to visceral organs, has disease burden out of proportion to serum PSA levels, and may express elevated serum neuroendocrine markers such as chromogranin A, synaptophysin, and neuron specific enolase. Clinically speaking, these findings are neither specific nor sensitive, so more definitive molecular characterization is needed to recognize this subgroup of patients who may be good candidates for platinum compounds.Genomic instability has long been known to be a hallmark of cancer, leading to genomic heterogeneity that gives variable clinical courses and response to treatment. With recent advances in genomic profiling, changes to the cancer genome—both germline and somatic&mdash;have been better characterized.

Germline mutations that disrupt DNA damage repair, especially the process of homologous recombination, such as BRCA1, BRCA2, ATM, CHEK2, PALB2, and RAD51D, have prognostic consequence. Various study results show them to be associated with increased risk of prostate cancer diagnosis at a younger age, higher histologic grade on diagnosis, more aggressive clinical course, and increased risk of cancer-specific mortality.38-41 It has also been suggested that homologous recombination is associated with relative insensitivity and worse outcomes with ADT.

The presence of germline mutations in DNA damage repair genes had previously been thought to be relatively low, with The Cancer Genome Atlas reporting a prevalence of 4.6% in men with localized prostate cancer.42 However, recent study results demonstrate a significantly higher proportion of men with mCRPC harboring these germline mutations, 11.8%, irrespective of family history of cancer.43 When considering both germline and somatic genomic alterations, the prevalence may be as high as 20% in mCRPC.44-46 Outside of specific mutations in the defined DNA damage repair pathway genes, a “BRCAness” genomic signature may exist for an even wider population of patients with mCRPC.

These genomic findings have therapeutic implications in that there has been evidence to suggest increased sensitivity to platinum therapy in ovarian47,48 and breast cancers49,50 with germline BRCA1/2 mutations. Platinum compounds exert their antineoplastic activity by forming covalent DNA adducts, causing double-stranded DNA breaks. Loss-of-function mutations or alterations to BRCA1, BRCA2, ATM, CHEK2, PALB2, and RAD51D, among other genes, all of which serve important roles in the repair process for double-stranded DNA breaks— including homologous repair, nonhomologous end joining, or altered DNA damage checkpoints&mdash;put the disease at a handicap in overcoming the effects of platinum agents.45,51

Early evidence suggests that prostate cancers that harbor deficiencies in the DNA damage repair pathway are also associated with a higher likelihood of response to platinum agents and poly (ADP-ribose) polymerase (PARP) inhibitors.52-54 In a retrospective analysis at Dana-Farber Cancer Institute of 141 men with CRPC who received at least 2 cycles of carboplatin/docetaxel chemotherapy, 75% (6/8) of men carrying deleterious BRCA2 variants experienced a >50% decline in PSA within 12 weeks of treatment initiation compared with 17% (23/133) in those who lacked the BRCA2 variant (P <.001).52 Twenty-five percent of BRCA2 carriers had a >90% PSA decline versus 4.5% of noncarriers.52 Median OS from time of platinum therapy initiation was 18.9 months for carriers versus 9.5 months for noncarriers (P = .03). Prospective studies are needed to fully realize the benefits of platinum compounds in molecularly selected patients (NCT02311764, NCT02598895).

Prostate Cancer Intensive Non-Cross Reactive Therapy for CRPC

Given the higher than expected prevalence of germline mutations and their prognostic and therapeutic implications, the question arises whether it would be appropriate to actively screen all men with mCRPC for these mutations. If so, what type of testing should be done? Would germline testing be sufficient by itself? Should somatic mutations be screened for as well? The prospect of obtaining metastatic biopsies on all patients with mCRPC, for the purpose of genomic testing, may prove to be a difficult task, especially as some lesions, such as osseous metastases, may not lend themselves to easy collection. Next-generation sequencing of circulating tumor cells or cell-free DNA analysis could play a role in identifying patients at higher likelihood of benefit with a platinum-based regimen.In the absence of having genomic data to optimally guide treatment selection, is there still a place for the routine use of platinum therapy in the standard management of mCRPC? Emerging data suggest that prolonged therapeutic exposure to potent AR-pathway— targeting agents places selective pressures on the tumor, inducing adaptive changes and leading to the emergence of “treatment-induced lineage crisis,” characterized by poorly differentiated and aggressive prostate cancers, visceral and bulky metastasis, and low PSA production.55 Lineage crisis often occurs in the form of treatment-induced neuroendocrine differentiation or epithelial-to-mesenchymal transition. There may be a role for platinum-based agents in maintaining lineage plasticity and collateral sensitivity and preventing the onset of treatment-induced lineage crisis and therapeutic resistance.

Currently enrolling at the Mount Sinai Health System is the Prostate Cancer Intensive Non—Cross Reactive Therapy (PRINT) for CRPC trial (NCT02903160), a single-arm investigator-initiated phase II study aimed at evaluating the clinical benefits of treating an intrinsically heterogeneous tumor with a rapidly cycling, non–cross reactive regimen of FDA-approved prostate cancer therapeutic agents. While optimal timing and sequence of different drugs in mCRPC remains a topic of debate, the standard approach so far has been to treat with a single agent until resistance develops, at which point the patient is transitioned to a different agent. Inherent genetic instability will lead to substantial tumor heterogeneity56 along with varied amounts of genetic dysregulation and varied patterns of antineoplastic resistance.57 This argues that a one-size-fits-all, sequential, single-agent approach will ultimately be met with limited success and subpopulations resilient to single-agent therapies will be identified, leading to a multidrug-refractory phenotype.

Our rapidly-cycling treatment regimen contains 3 consecutive treatment modules, each lasting 12 weeks:

  1. Abiraterone
  2. Cabazitaxel + carboplatin
  3. Enzalutamide + radium-223

By switching between non—cross reactive therapies, the selective pressure exerted by any single therapeutic agent’s intermittent use is limited, allowing resistance rates to stabilize or decrease in its absence. Additionally, the therapeutic agents are delivered as rationally designed combinations to achieve optimal therapeutic dosing and minimize overlapping adverse events. The rationale for inclusion of carboplatin in this regimen is 2-fold: to improve disease response and to help maintain lineage plasticity.

Future Directions

The primary endpoint of this study is time to disease progression following completion of the study regimen. If this novel treatment strategy significantly lengthens the time until disease progression, this trial would serve as proof-of-principle for future treatment regimen design in mCRPC. Additionally, as this regimen was designed to only include fully approved therapies, it would be immediately accessible to any treating oncologist.Platinum compounds remain relevant in mCRPC. Going forward, next-generation sequencing will be needed to further develop and expand on the development of biomarkers to predict for platinum responsiveness and to better identify patients appropriate for prospective trials involving platinum-based regimens. A better understanding of whether biomarkers associated with platinum sensitivity and their association with response to PARP inhibitors, and vice versa, will be important. It’s also important to discover how these 2 classes of drugs with similar mechanisms of actions, but potentially different mechanisms of resistance, should interplay.

References

  1. Tannock IF, de Wit R, Berry WR, et al; TAX 327 Investigators. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med. 2004;351(15):1502-1512. doi: 10.1056/NEJMoa040720.
  2. Kantoff PW, Higano CS, Shore ND, et al; IMPACT Study Investigators. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363(5):411-422. doi: 10.1056/NEJMoa1001294.
  3. de Bono JS, Oudard S, Ozguroglu M, et al; TROPIC Investigators. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet. 2010;376(9747):1147-1154. doi: 10.1016/S0140-6736(10)61389-X.
  4. de Bono JS, Logothetis CJ, Molina A, et al; COU-AA-301 Investigators. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med. 2011;364(21):1995-2005. doi: 10.1056/NEJMoa1014618.
  5. Scher HI, Fizazi K, Saad F, et al; AFFIRM Investigators. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 2012;367(13):1187-1197. doi: 10.1056/NEJMoa1207506.
  6. Ryan CJ, Smoth MR, de Bono JS, et al; COU-AA-302 Investigators. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med. 2013;368(2):138-148. doi: 10.1056/NEJMoa1209096.
  7. Parker C, Nilsson S, Heinrich D, et al; ALSYMPCA Investigators. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med. 2013;369(3):213-223. doi: 10.1056/NEJMoa1213755.
  8. Beer TM, Armstron AJ, Rathkopf DE, et al; PREVAIL Investigators. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. 2014;371:424-433. doi: 10.1056/NEJMoa1405095.
  9. Smith MR, Saad F, Chowdhury S, et al; SPARTAN Investigators. Apalutamide treatment and metastasis-free survival in prostate cancer [published online February 8, 2018]. N Engl J Med. doi: 10.1056/NEJMoa1715546.
  10. Trump DL, Marsh JC, Kvols LK, et al. A phase II trial of carboplatin (NSC 241240) in advanced prostate cancer, refractory to hormonal therapy. An Eastern Cooperative Oncology Group pilot study. Invest New Drugs. 1990;(8 suppl 1):S91-S94.
  11. Miglietta L, Cannobbio L, Boccardo F. Assessment of response to carboplatin in patients with hormone-refractory prostate cancer: a critical analysis of drug activity. Anticancer Res. 1995;15(6B):2825-2828.
  12. Canobbio L, Guarneri D, Miglietta L, Decensi A, Oneto F, Boccardo F. Carboplatin in advanced hormone refractory prostatic cancer patients. Eur J Cancer. 1993;29A(15):2094-2096.
  13. Jungi WF, Bernhard J, Hürny C, et al. Effect of carboplatin on response and palliation in hormone-refractory prostate cancer. Swiss Group for Clinical Cancer Research (SAKK). Support Care Cancer. 1998;6(5):462-468.
  14. Loening SA, Beckley S, Brady MF, et al. Comparison of estramustine phosphate, methotrexate and cis-platinum in patients with advanced, hormone refractory prostate cancer. J Urol. 1983;129(5):1001-1006.
  15. Buonerba C, Federico P, D'Aniello C, et al. Phase II trial of cisplatin plus prednisone in docetaxel-refractory castration-resistant prostate cancer patients. Cancer Chemother Pharmacol. 2011;67(6):1455-1461. doi: 10.1007/s00280-011-1594-z.
  16. Droz JP, Muracciole X, Mottet N, et al. Phase II study of oxaliplatin versus oxaliplatin combined with infusional 5-fluorouracil in hormone refractory metastatic prostate cancer patients. Ann Oncol. 2003;14(8):1291-1298.
  17. Oh WK, Tay MH, Huang J. Is there a role for platinum chemotherapy in the treatment of patients with hormone-refractory prostate cancer? Cancer. 2007;109(3):477-486. doi: 10.1002/cncr.22439.
  18. Culine S, El Demery M, Lamy PJ, Iborra F, Avancès C, Pinguet F. Docetaxel and cisplatin in patients with metastatic androgen independent prostate cancer and circulating neuroendocrine markers. J Urol. 2007;178(3 Pt 1):844-848; discussion 848.
  19. Cabrespine A, Guy L, Khenifar E, et al. Randomized phase II study comparing paclitaxel and carboplatin versus mitoxantrone in patients with hormone-refractory prostate cancer. Urology. 2006;67(2):354-359. goldjournal.net/article/S0090-4295(05)01205-7/fulltext. Published 2006. Accessed March 12, 2018.
  20. Kentepozidis N, Soultati A, Giassas S, et al. Paclitaxel in combination with carboplatin as salvage treatment in patients with castration-resistant prostate cancer: a Hellenic oncology research group multicenter phase II study. Cancer Chemother Pharmacol. 2012;70(1):161-168. doi: 10.1007/s00280-012-1896-9.
  21. Citrin DL, Hogan TF. A phase II evaluation of adriamycin and cis-platinum in hormone resistant prostate cancer. Cancer. 1982;50(2):201-206.
  22. Huan SD, Stewart DJ, Aitken SE, Segal R, Yau JC. Combination of epirubicin and cisplatin in hormone-refractory metastatic prostate cancer. Am J Clin Oncol. 1999;22(5):471-474.
  23. Fléchon A, Pouessel D, Ferlay C, et al. Phase II study of carboplatin and etoposide in patients with anaplastic progressive metastatic castration-resistant prostate cancer (mCRPC) with or without neuroendocrine differentiation: results of the French Genito-Urinary Tumor Group (GETUG) P01 trial. Ann Oncol. 2011;22(11):2476-2481. doi: 10.1093/annonc/mdr004.
  24. Loriot Y, Massard C, Gross-Goupil M, et al. Combining carboplatin and etoposide in docetaxel-pretreated patients with castration-resistant prostate cancer: a prospective study evaluating also neuroendocrine features. Ann Oncol. 2009;20(4):703-708. doi: 10.1093/annonc/mdn694.
  25. Lee JL, Ahn JH, Choi MK, et al. Gemcitabine-oxaliplatin plus prednisolone is active in patients with castration-resistant prostate cancer for whom docetaxel-based chemotherapy failed. Br J Cancer. 2014;110(10):2472-2478.
  26. Regan MM, O'Donnell EK, Kelly WK, et al. Efficacy of carboplatin-taxane combinations in the management of castration-resistant prostate cancer: a pooled analysis of seven prospective clinical trials. Ann Oncol. 2010;21(2):312-318. doi: 10.1093/annonc/mdp308.
  27. Oh WK, Hagmann E, Manola J, et al. A phase I study of estramustine, weekly docetaxel, and carboplatin chemotherapy in patients with hormone-refractory prostate cancer. Clin Cancer Res. 2005;11(1):284-289.
  28. Oh WK, Halabi S, Kelly WK, et al; Cancer and Leukemia Group B 99813. A phase II study of estramustine, docetaxel, and carboplatin with granulocyte-colony-stimulating factor support in patients with hormone-refractory prostate carcinoma: Cancer and Leukemia Group B 99813. Cancer. 2003;98(12):2592-2598.
  29. Kikuno N, Urakami S, Nakamura S, et al. Phase-II study of docetaxel, estramustine phosphate, and carboplatin in patients with hormone-refractory prostate cancer. Eur Urol. 2007;51(5):1252-1258. doi: 10.1016/j.eururo.2006.12.030.
  30. Sternberg CN, Petrylak DP, Sartor O, et al. Multinational, double-blind, phase III study of prednisone and either satraplatin or placebo in patients with castrate-refractory prostate cancer progressing after prior chemotherapy: the SPARC trial. J Clin Oncol. 2009;27(32):5431-5438. doi: 10.1200/JCO.2008.20.1228.
  31. Amato RJ, Logothetis CJ, Hallinan R, Ro JY, Sella A, Dexeus FH. Chemotherapy for small cell carcinoma of prostatic origin. J Urol. 1992;147(3 Pt 2):935-937.
  32. Papandreou CN, Daliani DD, Thall PF, et al. Results of a phase II study with doxorubicin, etoposide, and cisplatin in patients with fully characterized small-cell carcinoma of the prostate. J Clin Oncol. 2002;20(14):3072-3080. doi: 10.1200/JCO.2002.12.065.
  33. Moore SR, Reinberg Y, Zhang G. Small cell carcinoma of prostate: effectiveness of hormonal versus chemotherapy. Urology. 1992;39(5):411-416.
  34. Beltran H, Prandi D, Mosquera JM, et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat Med. 2016;22(3):298-305. doi: 10.1038/nm.4045.
  35. Jongsma J, Oomen MH, Noordzij MA, et al. Androgen deprivation of the PC-310 [correction of prohormone convertase-310] human prostate cancer model system induces neuroendocrine differentiation. Cancer Res. 2000;60(3):741-748.
  36. Jiborn T, Bjartell A, Abrahamsson PA. Neuroendocrine differentiation in prostatic carcinoma during hormonal treatment. Urology. 1998;51(4):585-589.
  37. Spigel DR, Hainsworth JD, Greco FA. Neuroendocrine carcinoma of unknown primary site. Semin Oncol. 2009;36(1):52-59. doi: 10.1053/j.seminoncol.2008.10.003.
  38. Kote-Jarai Z, Leongamornlert D, Saunders E, et al. BRCA2 is a moderate penetrance gene contributing to young-onset prostate cancer: implications for genetic testing in prostate cancer patients. Br J Cancer. 2011;105(8):1230-1234. doi: 10.1038/bjc.2011.383.
  39. Gallagher DJ, Gaudet MM, Pal P, et al. Germline BRCA mutations denote a clinicopathologic subset of prostate cancer. Clin Cancer Res. 2010;16(7):2115-2121. doi: 10.1158/1078-0432.CCR-09-2871.
  40. Castro E, Goh C, Olmos D, et al. Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer. J Clin Oncol. 2013;31(14):1748-1757. doi: 10.1200/JCO.2012.43.1882.
  41. Leongamornlert D, Saunders E, Dadaev T, et al. Frequent germline deleterious mutations in DNA repair genes in familial prostate cancer cases are associated with advanced disease. Br J Cancer. 2014;110(6):1663-1672. doi: 10.1038/bjc.2014.30.
  42. Cancer Genome Atlas Research Network. The molecular taxonomy of primary prostate cancer. Cell. 2015;163(4):1011-1025. doi: 10.1016/j.cell.2015.10.025.
  43. Pritchard CC, Mateo J, Walsh MF, et al. Inherited DNA-repair gene mutations in men with metastatic prostate cancer. N Engl J Med. 2016;375(5):443-453. doi: 10.1056/NEJMoa1603144.
  44. Robinson D, Van Allen EM, Wu YM, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;161(5):1215-1228. doi: 10.1016/j.cell.2015.05.001.
  45. Grasso CS, Wu YM, Robinson DR, et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature. 2012;487(7406):239-243. doi: 10.1038/nature11125.
  46. Beltran H, Yelensky R, Frampton GM, et al. Targeted next-generation sequencing of advanced prostate cancer identifies potential therapeutic targets and disease heterogeneity. Eur Urol. 2013;63(5):920-926. doi: 10.1016/j.eururo.2012.08.053.
  47. Tan DS, Rothermundt C, Thomas K, et al. "BRCAness" syndrome in ovarian cancer: a case-control study describing the clinical features and outcome of patients with epithelial ovarian cancer associated with BRCA1 and BRCA2 mutations. J Clin Oncol. 2008;26(34):5530-5536. doi: 10.1200/JCO.2008.16.1703.
  48. Sun C, Li N, Ding D, et al. The role of BRCA status on the prognosis of patients with epithelial ovarian cancer: a systematic review of the literature with a meta-analysis. PLoS One. 2014;9(5):e95285. doi: 10.1371/journal.pone.0095285.
  49. Telli ML, Jensen KC, Vinayak S, et al. Phase II study of gemcitabine, carboplatin, and iniparib as neoadjuvant therapy for triple-negative and BRCA1/2 mutation-associated breast cancer with assessment of a tumor-based measure of genomic instability: PrECOG 0105. J Clin Oncol. 2015;33(17):1895-1901. doi: 10.1200/JCO.2014.57.0085.
  50. Isakoff SJ, Mayer EL, He L, et al. TBCRC009: a multicenter phase II clinical trial of platinum monotherapy with biomarker assessment in metastatic triple-negative breast cancer. J Clin Oncol. 2015;33(17):1902-1909. doi: 10.1200/JCO.2014.57.6660.
  51. Eeles R, Goh C, Castro E, et al. The genetic epidemiology of prostate cancer and its clinical implications. Nat Rev Urol. 2014;11(1):18-31. doi: 10.1038/nrurol.2013.266.
  52. Pomerantz MM, Spisák S, Jia L, 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. doi: 10.1002/cncr.30808.
  53. Mateo J, Carreira S, Sandhu S, et al. DNA-repair defects and olaparib in metastatic prostate cancer. N Engl J Med. 2015;373(18):1697-1708. doi: 10.1056/NEJMoa1506859.
  54. Lord CJ, Ashworth A. BRCAness revisited. Nat Rev Cancer. 2016;16(2):110-120. doi: 10.1038/nrc.2015.21.
  55. Roubaud G, Liaw BC, Oh WK, Mulholland DJ. Strategies to avoid treatment-induced lineage crisis in advanced prostate cancer. Nat Rev Clin Oncol. 2017;14(5):269-283. doi: 10.1038/nrclinonc.2016.181.
  56. Cheng L, Bostwick DG, Li G, et al. Allelic imbalance in the clonal evolution of prostate carcinoma. Cancer. 1999;85(9):2017-2022.
  57. Mitsiades N, Sung CC, Schultz N, et al. Distinct patterns of dysregulated expression of enzymes involved in androgen synthesis and metabolism in metastatic prostate cancer tumors. Cancer Res. 2012;72(23):6142-6152. doi: 10.1158/0008-5472.CAN-12-1335.
  58. Aparicio AM, Harzstark AL, Corn PG, et al. Platinum-based chemotherapy for variant castrate-resistant prostate cancer. Clin Cancer Res. 2013;19(13):3621-3630. doi: 10.1158/1078-0432.CCR-12-3791.
Related Videos
Emmanuel Antonarakis, MD, associate director, Translational Research, Masonic Cancer Center, University of Minnesota, Clark Endowed Professor of Medicine, University of Minnesota Medical School
Gautam Jha, MD, medical director, M Health Fairview Masonic Cancer Clinic and the Advanced Treatment Center at the M Health Fairview Clinics and Surgery Center—Minneapolis, chair, cancer committee, M Health Fairview Ridges Hospital
Minesh Mehta, MD
Minesh Mehta, MD
Ruben Olivares, MD
Phillip J. Koo, MD
Daniel Spratt, MD
Daniel Spratt, MD
Philip J. Koo, MD
Anthony D'Amico MD, PhD