Germline Testing for Men With Prostate Cancer: Navigating an Expanding New World of Genetic Evaluation for Precision Therapy and Precision Management

Germline genetic testing is revolutionizing prostate cancer (PCA) care, with studies revealing inherited mutations (pathogenic variants) in a spectrum of cancer risk genes.1-5 Analyses from clinical cohorts of men with PCA have reported germline mutation rates of 15% to 17% regardless of stage,1,4,5 with rates of DNA repair mutations reported to be approximately 12% in men with metastatic PCA.2 Mutations in multiple genes including BRCA1, BRCA2, HOXB13, ATM, DNA mismatch repair genes, NBN, and CHEK2 confer variable estimates of risk for PCA predisposition, aggressive disease, and lifetime risk.1-5 The highest level of PCA risk has been reported for mutations in BRCA2, and HOXB13, with BRCA2 mutations associated with poor outcomes.1 Genetic testing for men with PCA is being driven increasingly by precision therapy and precision management considerations affecting oncology and urology.

For example, olaparib was given Breakthrough Therapy designation by the US Food and Drug Administration for BRCA1/2- or ATM-positive metastatic castration-resistant prostate cancer (mCRPC) on the basis of the TOPARP-A trial demonstrating improved responses particularly in patients with DNA repair mutations.6 Rucaparib was also granted Breakthrough Therapy designation for men with BRCA1/2-positive mCRPC after at least one androgen receptor–directed therapy and taxane-based chemotherapy on the basis of the TRITON2 study.7 Pembolizumab was granted US Food and Drug Administration accelerated approval for patients with unresectable or metastatic, microsatellite instability–high or mismatch repair deficient solid tumors that have progressed after prior treatment without other treatment alternatives.8 Men with mCRPC carrying BRCA or ATM mutations have been reported to have improved progression-free and overall survival with abiraterone and enzalutamide, with a subsequent study reporting improved cause-specific and progression-free survival among men with mCRPC and BRCA2 mutations treated with first-line abiraterone or enzalutamide over taxanes.9,10 Indeed, mutations in multiple genes (BRCA2, BRCA1, ATM, CHEK2, PALB2, RAD51D, NBN, MLH1, MSH2, PMS2, and MSH6) may provide clinical trial options for men with mCRPC, because genetically informed trials are expanding for PCA treatment.11 

Furthermore, clinical trials are emerging that are focused on the genetically informed response to treatment, such as with poly (ADP-ribose) polymerase inhibitors, in men with biochemical recurrence after prostatectomy (ClinicalTrials.gov identifier: NCT03047135),11 increasing the need to perform genetic testing sooner in the treatment algorithm. Genetic results may also be important for the management of men with early-stage PCA, with National Comprehensive Cancer Network (NCCN) guidelines stating that BRCA1/2 status should be considered in active surveillance discussions.12 Prior studies have reported the association of germline mutations in BRCA1, BRCA2, ATM, and CHEK2 (1100delC) with lethal PCA,13,14 with implications for early-stage management. BRCA1/2 and ATM mutations have also been associated with grade reclassification among men undergoing active surveillance.15 Thus, the clinical role of genetic testing in PCA treatment and management is expanding, requiring a greater percentage of men with PCA to undergo genetic evaluation. This demand for genetic assessment is posing a strain on genetic counseling services and genetics programs, raising the controversial question: Should oncologists and urologists perform genetic testing in their clinical practice? Given the lack of evidence-based data to answer this question, we herein discuss provider responsibilities of three potential models of genetic evaluation for men with PCA: (1) refer for genetic counseling, testing, and disclosure (model 1); (2) conduct pretest informed consent and post-test disclosure all within oncology or urology practices (model 2); (3) adopt a hybrid model of performing pretest informed consent and test ordering within treatment-based practices and post-test disclosure by cancer genetics professionals (model 3; Table 1). Oncologists and urologists need to be familiar with current NCCN PCA genetic testing guidelines.

genetic eval of men


Supported by expert consensus opinion,18 current testing criteria per NCCN guidelines may be summarized as follows: (1) all men with metastatic PCA regardless of family history; (2) all men with high-risk to regional disease per NCCN risk categories regardless of family history; (3) men with very low to unfavorable intermediate risk disease with brother, father, or multiple male relatives diagnosed with PCA at younger than 60 year of age, known germline DNA repair mutation in the family, or greater than one relative with family history suggestive of hereditary breast and ovarian cancer (HBOC) or Lynch syndrome; (4) men with Gleason score of 7 or greater with one or more blood relative with ovarian cancer, pancreatic cancer, metastatic PCA, or breast cancer younger than 50 years of age or two or more relatives with breast or prostate cancer (any grade) at any age, or Ashkenazi Jewish ancestry; or (5) BRCA mutations in tumor profiling.12,19 Therefore, in their practices, providers must perform intake of maternal and paternal family cancer history that covers a broad spectrum of cancers. Men meeting any of these criteria should consider genetic testing, necessitating that these men understand the implications of genetic testing for themselves and their families to make an informed decision, as advocated by multiple national organizations.12,19-24

There are many advantages to referring men with PCA to a cancer genetics counselor or provider (model 1) to discuss the complexities, uncertainties, and implications of genetic testing.20,25 Genetic counselors have graduate-level training in medical genetics and can discuss cancer inheritance, how genetic test results may inform cancer risk for patients and their families, and uncertainties regarding how genetic results may or may not provide information on the primary cancer of concern. Unanticipated cancer risks may be uncovered by test results (eg, male breast cancer, pancreatic cancer, or melanoma risks for men with BRCA2 mutations). Although genetic testing using a premade PCA panel is available through many commercial genetic testing laboratories and can ease test ordering, the patient’s personal or family history may dictate that additional genes be tested. Genetic counselors coordinate genetic testing, have insights into which laboratories have the highest-quality testing for the genes of interest, and discuss how insurance coverage may be affected by the choice of the testing laboratory. Counselors provide letters of preauthorization or medical necessity and discuss results with laboratories when there is a lack of clarity. Counselors also discuss the types of test results and their implications with patients. Pathogenic or likely pathogenic variants (clinically significant mutations) may be actionable in the management of PCA, may inform additional cancer screening recommendations, and may guide cascade testing in blood relatives. Variants of uncertain significance (VUS) are reported in at least 30% of men with PCA who have undergone genetic testing4,5 and do not affect recommendations at the time of reporting. However, a small fraction of VUS is reclassified as pathogenic or likely pathogenic over time.26 These reclassifications are reported back to ordering providers who have the responsibility to discuss the information and make appropriate recommendations with patients on an ongoing basis. VUS rates can also vary on the basis of assay and variant classification schema across laboratories. Finally, if a test result is negative, recommendations for cancer screening are based on family history; therefore, working knowledge of all NCCN guidelines is needed. In addition, counselors help coordinate cascade testing of relatives when a proband or familial mutation has been identified to inform important and potentially life-saving cancer risk, screening, and management recommendations across cancer types.

Of importance is the need to discuss the Genetic Information Nondiscrimination Act (GINA) of 2008, regarding its implications for men with PCA and their families, in the pretest counseling session.27 GINA generally provides protections for mutation carriers from health insurance discrimination and employment discrimination (with the exception of small businesses with fewer than 15 employees). Importantly, GINA does not apply to life, disability, or long-term care insurance. GINA also does not apply to Veterans’ health plans, federal employee health benefits plans, or Indian Health Service,27 although many of these have their own protections in place. Although these issues may not be imminently relevant for men with mCRPC, many men with earlier stage disease will enter survivorship and these issues will need to be discussed so that these men can understand the potential financial impact of genetic test results.20 This law becomes relevant when discussing genetic testing for unaffected men and cascade testing of unaffected family members.

The expansion of genetic testing in men with PCA has placed a strain on cancer genetics clinics. The United States has approximately 4,000 genetic counselors, insufficient to meet the growing demand for counseling.28 Many genetic programs have long wait times for appointments, with many comprehensive genetic services located in academic cancer centers, which may pose access challenges. The process of genetic referral, evaluation, results disclosure, and communication back to providers can also create delays in management decisions. Therefore, in the era of precision oncology and precision management, oncologists and urologists are increasingly considering performing pretest consent, ordering their own genetic testing, and discussing genetic test results to streamline the process (model 2; Table 1). Providers who opt to perform the entire genetic evaluation process must have a working knowledge of several topics related to cancer genetics and testing, knowledge of the current guidelines, and must consider the responsibilities of ordering, interpreting, and following up with test results as performed by genetic counselors and discussed earlier in the text (Table 1). Follow-up of variant reclassification also becomes the responsibility of oncologists and urologists in model 2. Resources for education, as well as certification programs, are available (Table 1), with PCA genetics education in need of development.

A hybrid model (model 3) that oncologists and urologists are discussing increasingly is performing pretest informed consent and ordering genetic testing in their practice, with referral to genetic counselors to discuss results and recommendations (Table 1). Here, providers will still need to address questions regarding cancer inheritance, genes to be tested, and limitations of tests, potential results, coordinate testing with genetic laboratories, and address the GINA law. In this model, the follow-up of variant reclassification also becomes the responsibility of oncologists and urologists. Although this may address the needs of rapid test ordering linked with disclosure of genetic results by a genetics professional, this approach can lead to miscommunication among patients and providers. Counselors may have chosen a different testing laboratory because of quality issues or have tested additional genes, which could create a potential added cost for patients. Patients may misunderstand the goals of testing or experience confusion over the interpretation of results because of multiple providers being involved in their care. These issues need to be vetted carefully before embarking on this model. Alternative delivery of genetic counseling, involving genetic education videos and telegenetics, is worth studying in this context in men with PCA.29

Regardless of the model, it is important for all providers to discuss men’s psychosocial and quality-of-life issues, which may affect their genetic evaluation experience. Although a substantial amount of research has been conducted regarding the psychosocial needs of women undergoing genetic evaluation,30-33 there exists a major gap in understanding the psychosocial needs of men. While discussing considerations of genetic testing, such as potential uncertainty with test results, limitations of testing, implications of testing for hereditary cancer risk for family members, and insurance discrimination, feelings of worry anxiety, guilt, fear, and future financial strain may be exacerbated or compounded by quality-of-life concerns that men may be experiencing. Men with early-stage PCA may be experiencing urinary incontinence and erectile dysfunction from radical prostatectomy, urinary or bowel toxicity and fatigue from radiation, and the fear of possible cancer progression with active surveillance.34 In the metastatic setting, a substantial proportion of men with PCA on androgen-deprivation therapy experience significant adverse effects,35 and those treated with chemotherapy may also deal with long-term toxicity. Many men deal with depression, a feeling of loss of masculinity, and a negative impact on sexual relationships.36 This area now deserves focused study regarding the impact of psychosocial needs and worries on men’s informed decision making for PCA genetic testing, understanding of results, and opportunities to intervene.

In summary, there is no final answer to the question: Should oncologists and urologists perform genetic testing in their practice? There are other models to consider beyond those proposed here, such as collaborative models, that also deserve study. Until evidence-based studies are available that focus on the various models of pretest and post-test genetic evaluation for men with PCA, oncologists, and urologists should start building collaborative relationships with cancer genetics professionals and counselors to establish a clinical flow for men with PCA who need genetic assessment. This brand new era of genetic testing for precision therapy and precision management is challenging the paradigm of genetic evaluation, which is in need of greater research. Thus, a more global dialogue across oncology, urology, genetic counseling, and population scientists is now imperative to meet the needs of men undergoing germline testing for inherited PCA.


Authors: Veda N. Giri, MD, Colette Hyatt, LCGC, MS and Leonard G. Gomella, MD, Thomas Jefferson University, Philadelphia, Pennsylvania, United States

Authors’ Disclosures Of Potential Conflicts Of Interest And Data Availability Statement Disclosures provided by the authors and data availability statement (if applicable) are available with this article at DOI https://doi.org/10.1200/ JCO.18.02181.
References: 
1. National Cancer Institute: Genetics of prostate cancer (PDQ)-Health professional version. https://www.cancer.gov/types/prostate/hp/prostate-genetics-pdq 
2. Pritchard CC, Mateo J, Walsh MF, et al: Inherited DNA-repair gene mutations in men with metastatic prostate cancer. N Engl J Med 375:443-453, 2016 
3. Schrader KA, Cheng DT, Joseph V, et al: Germline variants in targeted tumor sequencing using matched normal DNA. JAMA Oncol 2:104-111, 2016 [Erratum: JAMA Oncol 2:279, 2016] 
4. Giri VN, Hegarty SE, Hyatt C, et al: Germline genetic testing for inherited prostate cancer in practice: Implications for genetic testing, precision therapy, and cascade testing. Prostate 79:333-339, 2019 
5. Nicolosi P, Ledet E, Yang S, et al: Prevalence of germline variants in prostate cancer and implications for current genetic testing guidelines. JAMA Oncol https:// doi.org/10.1001/jamaoncol.2018.6760 [epub ahead of print on February 7, 2019] 
6. Mateo J, Carreira S, Sandhu S, et al: DNA-repair defects and olaparib in metastatic prostate cancer. N Engl J Med 373:1697-1708, 2015 
7. Clovis Oncology: Clovis Oncology receives breakthrough therapy designation for rubraca (rucaparib) for treatment of BRCA1/2-mutated metastatic castration-resistant prostate cancer (mCRPC). https://bit.ly/2O0Lqdq 
8. The ASCO Post: FDA grants accelerated approval to pembrolizumab for first tissue/site-agnostic indication. http://www.ascopost.com/News/55666? utm_source5TrendMD&utm_medium5cpc&utm_campaign5The_ASCO_Post_TrendMD_0 
9. Antonarakis ES, Lu C, Luber B, et al: Germline DNA-repair gene mutations and outcomes in men with metastatic castration-resistant prostate cancer receiving first-line abiraterone and enzalutamide. Eur Urol 74:218-225, 2018 
10. Castro E, Romero-Laorden N, Del Pozo A, et al: PROREPAIR-B: A prospective cohort study of the impact of germline DNA repair mutations on the outcomes of patients with metastatic castration-resistant prostate cancer. J Clin Oncol 37:490-503, 2019 
11. Carlo MI, Giri VN, Antonarakis ES, et al: Evolving intersection between inherited cancer genetics and therapeutic clinical trials in prostate cancer: A white paper from the Germline Genetics Working Group of the Prostate Cancer Clinical Trials Consortium . JCO Precis Oncol https://doi.org/10.1200/PO.18.00060 [epub ahead of print on August 16, 2018]
12. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: (NCCN Guidelines®): Prostate (Version 4.2018) https://www.nccn. org/professionals/physician_gls/pdf/prostate.pdf 
13. Na R, Zheng SL, Han M, et al: Germline mutations in ATM and BRCA1/2 distinguish risk for lethal and indolent prostate cancer and are associated with early age at death. Eur Urol 71:740-747, 2017 
14. Wu Y, Yu H, Zheng SL, et al: A comprehensive evaluation of CHEK2 germline mutations in men with prostate cancer. Prostate 78:607-615, 2018 
15. Carter HB, Helfand B, Mamawala M, et al: Germline mutations in ATM and BRCA1/2 are associated with grade reclassification in men on active surveillance for prostate cancer. Eur Urol S0302-2838:30684-30685, 2018 4 © 2019 by American Society of Clinical Oncology Giri, Hyatt, and Gomella Downloaded from ascopubs.org by 67.184.183.127 on April 17, 2019 from 067.184.183.127 Copyright © 2019 American Society of Clinical Oncology. All rights reserved. 
16. ASCO University: Cancer genetics program (2018 update). https://university.asco.org/cancer-genetics-program 
17. NCBI: ClinVar. https://www.ncbi.nlm.nih.gov/clinvar/ 
18. Giri VN, Knudsen KE, Kelly WK, et al: Role of genetic testing for inherited prostate cancer risk: Philadelphia Prostate Cancer Consensus Conference 2017. J Clin Oncol 36:414-424, 2018 
19. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: (NCCN Guidelines): Genetic/familial high-risk assessment: Breast and ovarian (version 2.2019). https://www.nccn.org/professionals/physician_gls/pdf/genetics_screening.pdf 
20. Riley BD, Culver JO, Skrzynia C, et al: Essential elements of genetic cancer risk assessment, counseling, and testing: Updated recommendations of the National Society of Genetic Counselors. J Genet Couns 
21:151-161, 2012 21. Hampel H, Bennett R, Buchanan A, Pearlman R, Wiesner G. A practice guideline from the American College of Medical Genetics and Genomics and the National Society of Genetic Counselors: Referral indications for cancer predisposition assessment. Genet Med 17:70-87, 2014 
22. Institute of Medicine (US) Committee on Assessing Genetic Risks: Andrews LB, Fullarton JE, Holtzman NA, et al (eds): Assessing Genetic Risks: Implications for Health and Social Policy. Washington, DC, National Academies Press, 1994 https://www.ncbi.nlm.nih.gov/books/NBK236044/ 
23. Robson ME, Bradbury AR, Arun B, et al: American Society of Clinical Oncology policy statement update: Genetic and genomic testing for cancer susceptibility. J Clin Oncol 33:3660-3667, 2015 
24. American Society of Clinical Oncology: Assessing your patient’s hereditary cancer risk. https://www.asco.org/practice-guidelines/cancer-care-initiatives/ genetics-toolkit/assessing-your-patient%E2%80%99s-hereditary 
25. Hall MJ, Forman AD, Pilarski R, et al: Gene panel testing for inherited cancer risk. J Natl Compr Canc Netw 12:1339-1346, 2014 
26. Mersch J, Brown N, Pirzadeh-Miller S, et al: Prevalence of variant reclassification following hereditary cancer genetic testing. JAMA 320:1266-1274, 2018 
27. Genetic Information Nondiscrimination Act. http://www.ginahelp.org/GINAhelp.pdf 
28. Abacan M, Alsubaie L, Barlow-Stewart K, et al: The global state of the genetic counseling profession. Eur J Hum Genet 27:183-197, 2019 
29. Buchanan AH, Rahm AK, Williams JL. Alternate service delivery models in cancer genetic counseling: A mini-review. Front Oncol 6:120, 2016 
30. Wevers MR, Hahn DE, Verhoef S, et al: Breast cancer genetic counseling after diagnosis but before treatment: A pilot study on treatment consequences and psychological impact. Patient Educ Couns 89:89-95, 2012 
31. Schlich-Bakker KJ, ten Kroode HF, Ausems MG: A literature review of the psychological impact of genetic testing on breast cancer patients. Patient Educ Couns 62:13-20, 2006 
32. Ringwald J, Wochnowski C, Bosse K, et al: Psychological distress, anxiety, and depression of cancer-affected BRCA1/2 mutation carriers: A systematic review. J Genet Couns 25:880-891, 2016 
33. NCBI: PDQ cancer information summaries: Cancer genetics risk assessment and counseling (PDQ): Health professional version. In: PDQ Cancer Information Summaries [Internet]. Bethesda, MD, National Cancer Institute, 2002 https://www.ncbi.nlm.nih.gov/books/NBK65817/ 
34. Zeliadt SB, Ramsey SD, Penson DF, et al: Why do men choose one treatment over another?: A review of patient decision making for localized prostate cancer. Cancer 106:1865-1874, 2006 
35. Taylor LG, Canfield SE, Du XL: Review of major adverse effects of androgen-deprivation therapy in men with prostate cancer. Cancer 115:2388-2399, 2009
36. Sharpley CF, Birsika V, Denham JW: Factors associated with feelings of loss of masculinity in men with prostate cancer in the RADAR trial. Psychooncology 23: 524-530, 2014


Veda N. Giri et al. Germline Testing for Men With Prostate Cancer: Navigating an Expanding New World of Genetic Evaluation for Precision Therapy and Precision Management Journal of Clinical Oncology Published online April 12, 2019. DOI: 10.1200/JCO.18.02181