You are on page 1of 13

REVIEwS

Germline testing and genetic


counselling in prostate cancer
Jessica Russo1 and Veda N. Giri   1,2 ✉
Abstract | Genetic testing for prostate cancer is rapidly growing and is increasingly being driven
by precision medicine. Rates of germline pathogenic variants have been reported in up to 15% of
men with prostate cancer, particularly in metastatic disease, and results of genetic testing could
uncover options for precision therapy along with a spectrum of hereditary cancer-​predisposition
syndromes with unique clinical features that have complex management options. Thus, the
pre-​test discussion, whether delivered by genetic counsellors or by health-​care professionals in
hybrid models, involves information on hereditary cancer risk, extent of gene testing, purpose of
testing, medical history and family history, potential types of results, additional cancer risks that
might be uncovered, genetically based management and effect on families. Understanding
precision medicine, personalized cancer risk management and syndrome-​related cancer risk
management is important in order to develop collaborative strategies with genetic counselling
for optimal care of patients and their families.

Prostate cancer is a common cancer in men, and a subset men who carry mutations in DNA repair genes such as
of men can have a hereditary predisposition to develop- in BRCA1, BRCA2, ATM, or the DNA mismatch repair
ing this disease1,2. Germline testing (hereditary cancer genes1–7. Thus, pre-​test discussions with patients need
genetic testing) encompasses testing for genes linked to address the potential effects of treatment or screen-
to hereditary syndromes, such as hereditary breast and ing, hereditary cancer risk and implications for blood
ovarian cancer (HBOC) syndrome, Lynch syndrome relatives1–4,8–10.
and hereditary prostate cancer1–4. Furthermore, germline Many thousands of men could now be eligible for
testing now involves multigene testing of a host of addi- germline testing and, therefore, strategies for clinical
tional genes, such as DNA repair genes, which might also genetic evaluation need to be implemented. Genetics
confer increased risk of additional cancers and be impor- care delivery can occur through genetic counselling or
tant for therapeutic determination1–7. Germline testing by employing a hybrid model (a health-​care provider–
for inherited mutations is important to estimate cancer genetic counselling collaborative approach). Genetic
risks above the general population, with magnitude counselling is a specialized field and genetic counsel-
of risk being gene specific1,8. Population risk for devel- lors are professionals trained in the principles and prac-
oping prostate cancer is 11%, whereas men with specific tice of genetic testing, hereditary cancer assessment,
genetic mutations (pathogenic or probable pathogenic informed decision-​making for genetic testing and the
variants) can have a 2-​fold to 10-​fold increase in lifetime implications of genetic testing for patients and their
risk of developing prostate cancer, such as for mutations families10–13 (Box 1). They perform an assessment of a
1
Cancer Risk Assessment in BRCA2 or HOXB13 (ref.1). Risk-​based screening for patient’s medical history and family history, discuss can-
and Clinical Cancer Genetics, prostate cancer is evolving, with current guidelines from cer heredity and options for genetic testing, implications
Sidney Kimmel Cancer Center,
Thomas Jefferson University,
the National Comprehensive Cancer Network (NCCN) of test results, and the benefits and risks of testing so
Philadelphia, PA, USA. recommending initiation of prostate cancer screening that patients can make an informed decision10–13. Upon
2
Departments of Medical for BRCA2 carriers at the age of 40 years with consid- return of the results, genetic counsellors help patients to
Oncology, Cancer Biology, eration of the same for BRCA1 carriers9. Furthermore, understand their results and discuss recommendations
and Urology, Sidney Kimmel prostate cancer germline testing came to the forefront in based on the results and the patient’s medical history
Cancer Center, Thomas the precision medicine era1,2,4–6. Multiple targeted agents, and family history. They also coordinate cascade test-
Jefferson University,
Philadelphia, PA, USA.
such as poly(ADP-​ribose) polymerase (PARP) inhibi- ing (testing of blood relatives in families with a known
✉e-​mail: Veda.Giri@ tors and immunotherapeutic drugs, are FDA approved genetic mutation) or additional genetic testing in a
jefferson.edu or have FDA designations for use in men with metastatic family9–12.
https://doi.org/10.1038/ castration-​resistant prostate cancer (mCRPC) owing to In the current precision medicine era, in which
s41585-022-00580-7 demonstrated clinical responses, particularly among genetic mutations might guide options for targeted

Nature Reviews | Urology

0123456789();:
Reviews

Key points are tumour suppressor genes involved in DNA homol-


ogous recombination repair18–20. HBOC is inherited in
• Germline (hereditary) genetic testing is rising in importance for treatment, screening an autosomal-​dominant manner (in which a single copy
and risk assessment of prostate cancer. of a mutation on a non-​sex chromosome is sufficient
• Multiple hereditary cancer syndromes might be associated with prostate cancer, to predispose to disease)19,20 and is associated with
might confer risk of other cancerous and non-​cancerous conditions, and can have breast cancer (male and female), ovarian cancer, pan-
hereditary cancer implications for family members. The rates of these syndromes can creatic cancer, prostate cancer and melanoma9. Up to
vary based upon the attributed genetic mutations.
15% of men with metastatic prostate cancer have been
• Multiple aspects of germline testing should be discussed in the pre-​test setting reported to carry germline mutations in DNA repair
for men to make an informed decision, including the purpose of genetic testing, the
genes such as BRCA1 and BRCA2, whereas ~5–7% of
benefits and risks of testing, hereditary cancer risk, identification of additional cancer
risks, familial implications and the state of genetic discrimination protections.
men with early-​stage prostate cancer are carriers1,2,21–23.
BRCA2 mutations are more strongly associated with
• Genetic evaluation can be conducted by genetic counsellors or a hybrid model can
be employed, in which health-​care providers deliver pre-​test informed consent
prostate cancer than BRCA1 mutations, with an approxi-
for testing, order testing and then determine referral to genetic counselling for mately eightfold versus approximately threefold increase
appropriate patients. in risk, respectively1,24. Furthermore, BRCA2 mutations
• Precision medicine is increasingly driving decisions for germline testing. Poly(ADP- have been associated with aggressive prostate cancer
ribose) polymerase (PARP) inhibitors, immune checkpoint inhibitors and various other with decreased prostate cancer-​specific survival1,25–27.
agents now in clinical trials have clinical activity in patients with certain hereditary Individuals with Ashkenazi Jewish ancestry are at an
cancer gene mutations, such as in DNA repair genes. increased risk of carrying a mutation in BRCA1 or
• Patients’ experiences with germline testing can be variable; taking the patient’s BRCA2 owing to three known founder mutations in this
current experience into account, considering referral to genetic counselling when population: 185delAG (BRCA1), 5382insC (BRCA1) and
needed and offering germline testing for eligible men at repeated intervals if initially 6174delT (BRCA2)28,29. The combined population risk
declined are important. of carrying one of these variants is 1 in 40 for Ashkenazi
Jewish individuals compared with 1 in 400 for the
therapies and with the expanded indications for germline general population28,30.
testing, a need for alternative delivery of germline testing Hereditary prostate cancer, with generational and/or
initiated by health-​care providers has arisen given the young onset of disease, is another hereditary syn-
relative shortage of genetic counsellors and the need to drome associated with prostate cancer development1,31.
limit additional visits and reduce barriers13–17. Hybrid HOXB13, the first classic hereditary prostate cancer
models have emerged in practice to expand access to gene identified in 2012 is associated with an approxi-
germline testing, in which joint care strategies are imple- mately eightfold increased risk of prostate cancer and
mented between health-​care providers and genetic approximately tenfold increased risk of early-​onset
counsellors to address pre-​test consent strategies, man- disease1,24,32. HOXB13 is currently not known to confer
agement of men with pathogenic variants, management increased risk of developing other cancers. The G84E
of subsets of men with variants of uncertain significance pathogenic variant of HOXB13 has an established asso-
(VUS), management of subsets of men with negative ciation with prostate cancer; its overall frequency was
results with strong family cancer history, and cascade reported to be 1.34% among men with prostate cancer in
testing2,14. a pooled analysis across multiple studies33, whereas the
Given the growth of indications for germline test- highest carrier rate of 6.25% has been reported in men
ing in prostate cancer7,9, this Review addresses major with prostate cancer in Finland34. HOXB13 codes for a
autosomal-​dominant hereditary cancer syndromes homeobox transcription factor and is located on chromo­
linked with prostate cancer, germline testing criteria, some 17 (ref.34); this gene is part of a large group of tran-
genetic testing strategies, genetically informed prostate scription factors called the homeobox protein family,
cancer screening and precision management, delivery which has a role in prostate development35. HOXB13
of genetic counselling or alternative genetic services and mutations are inherited in an autosomal-​dominant
special considerations for this population. All of these manner1,32.
issues are now of crucial relevance for optimal cancer Prostate cancer is also within the spectrum of Lynch
risk assessment, screening and treatment of patients with syndrome cancers1,24,36. Lynch syndrome, classically
prostate cancer or at risk of developing prostate cancer known as hereditary non-​polyposis colorectal cancer
guided by genetic information. syndrome, predisposes to development of cancers of
the colon and/or rectum, pancreas, ovary, uterus, upper
Inherited cancer syndromes contributing to bowel and urinary tract, and sebaceous carcinoma36,37.
prostate cancer Lynch syndrome is associated with mutations in the fol-
Multiple hereditary cancer syndromes can be associ- lowing genes involved in DNA mismatch repair (MMR):
ated with prostate cancer, such as HBOC syndrome and MLH1 (chromosome 3), MSH2 (chromosome 2),
Lynch syndrome, which have specific clinical manifes- MSH6 (chromosome 2) and PMS2 (chromosome 7)
tations and management1,2 (Table 1). These syndromes and is inherited in an autosomal-​dominant manner36,37.
require unique attention when performing genetic coun- Certain deletions in the EPCAM gene have also been
selling and/or pre-​test informed consent and germline shown to cause Lynch syndrome by causing an MSH2
testing (Table 1). HBOC is associated with mutations in epimutation37. However, associated cancer risks vary
BRCA1 and BRCA2 (ref.9). BRCA1 and BRCA2, which based on the study and DNA MMR genes studied1,36–39.
are located on chromosomes 17 and 13, respectively, Overall, mutations in the DNA MMR genes, particularly

www.nature.com/nrurol

0123456789();:
Reviews

MSH2 and MSH6, are associated with an approximately characteristics and manifestations, and additional
threefold increased risk of developing prostate cancer1,38. cancers1,7,9,24; thus, multigene testing for prostate cancer
Other genes, such as CHEK2 or NBN, have vary­ has become a standard of genetic testing practice1,2,7.
ing degrees of association with prostate cancer, but Several of these syndromes have established guidelines
are included in prostate cancer testing panels owing to inform hereditary cancer assessment in families and
to implications for therapy1,2. Multiple other genes guide screening for at-​risk individuals7,9,39 (Table 1).
involved in DNA repair, such as ATM, PALB2, RAD51C
and RAD51D, could also be important for testing to Germline testing criteria and approach to testing
inform response to precision therapies such as PARP for prostate cancer
inhibitors1,2,4,5. Germline testing criteria for prostate cancer encom-
These data show that multiple genes linked with a passes personal cancer history, cancer features and
spectrum of hereditary cancer syndromes are associated pathology, family history and precision therapy
with varying degrees of prostate cancer risk, disease indications 7,9. The NCCN publishes guidelines for
consideration of germline testing across multiple can-
Box 1 | Basic elements of genetics care delivery and germline testing cer types, including prostate cancer7,9. Germline test-
ing criteria for prostate cancer have been provided
Aspects of germline genetic testing by multiple NCCN panels7,9, an international expert
• Testing DNA from non-​cancerous cells to detect variation associated with potential
consensus statement from the Philadelphia Prostate
risk of disease or effect on treatment.
Cancer Consensus Conference 2019 (ref.2), American
• Disease risk is predominantly determined by the effect that the variant has on the
Urological Association/American Society for Radiation
ability of the gene to translate into a functioning protein.
Oncology/Society of Urologic Oncology (AUA/ASTRO/
• The goal of germline genetic testing in oncology is to assess an individual’s cancer
SUO) 2020 guideline for advanced prostate cancer40,
predisposition and understand why a specific cancer developed based on pathogenic
and the European Association of Urology (EAU) 2020
variants in genes implicated in important cellular processes.
guideline2,7,9,41 (Box 2). Prostate cancer germline testing
• Germline testing can also be conducted in oncology to inform targeted therapy
criteria from the NCCN are uniform among guidelines
options, particularly in the metastatic setting.
regarding cancer features (stage, grade) and ancestry;
• Variants detected in germline testing can be passed down or inherited by offspring.
however, they differ by family history criteria7,9. NCCN
Germline testing differs from somatic genetic testing of a tumour assessing for somatic
variants, which are usually acquired in tumour formation. Confirmatory germline guidelines agree regarding recommending germline
testing of somatic variants might be indicated to determine hereditary nature. testing for men with any one of the following: meta-
static prostate cancer, regional or node-​positive disease,
Aspects of genetic counselling very high-​risk or high-​risk prostate cancer (defined by
• Evaluating a patient’s medical and family history to assess the likelihood of a genetic grade group, T stage, and serum PSA levels at diagnosis),
predisposition and advise appropriate genetic testing. intraductal or cribriform histology, Ashkenazi Jewish
• Providing education regarding genetic testing, inherited health risk, or effect on ancestry, or family history of a relative or relatives with a
treatment. germline mutation7,9. Eliciting family history of prostate
• Supporting patients in making genetic testing decisions. cancer (age at diagnosis, known metastatic disease, death
• Interpreting genetic testing results and helping patients to understand and adjust to from prostate cancer) is important to inform for suspi-
potential medical, familial and psychosocial implications. cion of hereditary prostate cancer and to help to inform
Clinical scope of a genetic counsellor regarding the full scope of genes to test2,7,9. Furthermore,
knowing the family history of cancers of the breast, ovary,
• A medical professional with specialized training in advanced genetics and counselling.
pancreas, colon and/or rectum, uterus, small bowel,
• Genetic counsellors often work in a clinic or hospital setting along with physicians
urinary tract and skin is important7. Clinical practice
and other health-​care providers as part of a patient’s care team.
might not be straightforward, but clinicians can refer
• They can specialize in different areas of genetics including, but not limited to,
to all guidelines to determine which family history cri­
oncological, paediatric, prenatal or preconception, cardiovascular and neurological.
teria might match their patient’s family history to deter­
Aspects of a hybrid genetics care delivery model mine eligibility for germline testing7,9. Thus, health-care
• A collaborative care model between health-​care providers and genetic counsellors. providers need to become familiar with the nuances of
• Health-​care providers deliver pre-​test informed consent. these guidelines and remain current, as the guidelines
• Health-​care providers order germline testing, particularly focusing on the effect on are regularly updated.
treatment or management. Multiple professional organizations address germline
• Upon return of the results, health-​care providers discuss implications for prostate testing for prostate cancer in various ways (Table 1; Box 2).
cancer treatment, management or screening. Appropriate patients are referred to The EAU 2020 guideline41 recommends conducting
genetic counsellors. germline testing for men with metastatic prostate can-
-- Results of pathogenic or probable pathogenic variants cer. This guideline also recommends performing genetic
-- Results of variants of uncertain significance with strong family cancer history testing for men with mCRPC for DNA repair mutations
-- Men with negative results with strong family cancer history to determine eligibility for PARP inhibitors. The AUA/
-- Complex genetic results ASTRO/SUO 2020 advanced prostate cancer guideline40
• Some patients might need to be referred to genetic counselling upfront. states that patients with metastatic prostate cancer
-- Men who prefer to see a genetic counsellor should be offered genetic counselling and genetic testing
-- Men with a high level of anxiety or psychosocial issues regardless of age and family history30. It also recommends
-- Men with known mutations in the family
offering PARP inhibitors to patients with deleterious or
-- Men with rare or complex syndromes
suspected deleterious germline or somatic homologous

Nature Reviews | Urology

0123456789();:
Reviews

Table 1 | Major hereditary cancer syndromes linked with prostate cancer


gene Syndrome Clinical manifestations Prostate cancer management considerations by selected guidelines for patients with
affecting cancer mutationsb
management
(patient and familial
implications)a
BRCA1 HBOC Female and male breast NCCN guidelines genetic/Familial High-​risk Assessment: Breast, ovarian, and
cancer Pancreatic Version 1.2022 (ref.9)
BRCA2
Ovarian cancer Prostate cancer: PSA screening starting at the age of 40 years with annual intervals
Recommend screening for BRCA2 carriers and consider screening for BRCA1 carriers
Prostate cancer
NCCN Prostate Cancer (Version 2.2021)7
Pancreatic cancer
For mCRPC, DNA genes such as BRCA1, BRCA2, ATM, etc. might inform early platinum therapy
Melanoma
or PARP inhibitor therapy
NCCN Prostate Cancer Early Detection (Version 2.2020)49
Consider shared decision-​making for men with BRCA1 or BRCA2 mutations regarding prostate
cancer screening starting at the age of 40 years and to consider annual versus biannual
screening
Philadelphia Prostate Cancer Consensus Conference 2019 (ref.2)
Recommend precision medicine clinical trials
mCRPC: BRCA2 and BRCA1 inform PARP inhibitor therapy; also inform use of platinum
chemotherapy. Consider immunotherapy for DNA MMR gene mutation carriers
Consider BRCA2 in active surveillance discussions
Prostate cancer screening at the age of 40 years or 10 years before the youngest person in
the family was diagnosed with prostate cancer and continue screening with annual intervals
(recommended for BRCA2 carriers with consideration for BRCA1 carriers)
AUA/ASTro/SUo Advanced Prostate Cancer 2020 (ref.40)
Offer PARP inhibitors to patients with deleterious or suspected deleterious germline or
somatic homologous recombination repair gene-​mutated mCRPC following previous
treatment with enzalutamide or abiraterone acetate, and/or a taxane-​based chemotherapy.
Consider platinum-​based chemotherapy as an alternative for patients who cannot use or
obtain a PARP inhibitor
EAU Prostate Cancer 2020 (ref.41)
Genetic testing for DNA repair mutations for PARP inhibitor therapy in mCRPC
MLH1 Lynch Colorectal NCCN Prostate Cancer (Version 2.2021)7
syndrome
MSH2 Endometrial For mCRPC, consider dMMR or MSI-​H status for pembrolizumab
MSH6 Ovarian Philadelphia Prostate Cancer Consensus Conference 2019 (ref.2)
PMS2 Urothelial cancer (renal Consider DNA MMR gene mutations for pembrolizumab
pelvis, ureteral cancers)
EPCAM Consider prostate cancer screening at age 40 years or 10 years before the youngest person in
Gastric and/or small the family was diagnosed with prostate cancer and continue screening with annual intervals
bowel
AUA/ASTro/SUo Advanced Prostate Cancer 2020 (ref.40)
Pancreatic
In patients with dMMR or MSI-​H mCRPC, offer pembrolizumab
Prostate
Brain
HOXB13 Hereditary Prostate cancer Philadelphia Prostate Cancer Consensus Conference 2019 (ref.2)
prostate
Consider prostate cancer screening at age 40 years or 10 years before the youngest person in
cancer
the family was diagnosed with prostate cancer and continue screening with annual intervals
ASTRO, American Society for Radiation Oncology; AUA, American Urological Association; dMMR, mismatch repair deficiency; EAU, European Association of Urology;
HBOC, hereditary breast and ovarian cancer syndrome; mCRPC, metastatic castration-​resistant prostate cancer; MMR, mismatch repair; MSI-​H, microsatellite
instability high; NCCN, National Comprehensive Cancer Network; PARP, poly(ADP-​ribose) polymerase; SUO, Society of Urologic Oncology. aClinical trials are
important for patients with mutations concerning treatment and management of prostate cancer. Many of the hereditary syndromes in this table have cancerous
and non-​cancerous disease features beyond prostate cancer that also require management. bMultiple NCCN guidelines address management of cancer risks, risk
reduction and treatment affecting men and their families.

recombination repair gene-​mutated mCRPC following Clinical germline testing has now progressed from
treatment with enzalutamide or abiraterone acetate, and/or single gene testing to multigene testing options for
a taxane-​b ased chemotherapy. It also recommends patients2,3,42. Multiple genetic testing laboratories offer
considering platinum-​based chemotherapy as an alter- germline testing options for patients with prostate can-
native for patients who cannot use or obtain a PARP cer or those concerned about risk of prostate cancer.
inhibitor and offering pembrolizumab to patients with In general, testing options include guideline-​focused
MMR-​deficient or microsatellite instability-​high mCRPC. panels, tumour-​specific panels and large comprehensive

www.nature.com/nrurol

0123456789();:
Reviews

cancer panels2,3 (Table 2). Furthermore, some genetic of test ordering for the test to be cost free to patients in
testing laboratories have the option of initially conduct- the USA.
ing testing with a small set of genes, and then reflex test- Multigene panel testing has become the standard of
ing a larger set of genes if initial testing does not reveal care for many genetic testing indications7,9; thus, dedi-
pathogenic and/or probable pathogenic variants in the cated discussion of the considerations of panel tests is
genes tested2. These reflex options typically need to be needed before a patient pursues genetic testing2,42. The
conducted within the laboratory-​specified time frame size and availability of multiple gene panels varies by
laboratory and the process of helping the patient choose
the best panel for them can be complex2,42,43. A thorough
Box 2 | Examples of germline testing criteria for prostate cancer
discussion of benefits and limitations of panel testing is
NCCN guidelines genetic/Familial High-​risk Assessment: Breast, ovarian and necessary before pursuing multigene panel testing, with
Pancreatic (Version 1.2022)9 recognition that these discussions need to be tailored
• Personal history of prostate cancer at any age with to the purpose of testing2,7,9,42,43. For example, multigene
-- Metastatic, intraductal or cribriform histology, or high-​risk or very-​high-​risk group testing in a patient with metastatic prostate cancer might
(see NCCN Prostate Cancer guideline7 criteria below) regardless of age
include large panel testing to optimize targeted therapy
• Any NCCN risk group (see NCCN Prostate Cancer guideline7 below) with the following or clinical trial options2,4; however, in early-​stage pros-
-- ≥1 close blood relative diagnosed with breast cancer at ≤50 years old, pancreatic,
tate cancer, genetic testing might be more tailored and
ovarian, or metastatic, intraductal or cribriform prostate cancer at any age
-- ≥2 close blood relatives with either breast or prostate cancer at any age
encompass genes that account for the patient’s cancer
-- Ashkenazi Jewish ancestry and family history2. Patients need to understand the
benefits and limitations of various multigene panel test-
NCCN guidelines Prostate Cancer (Version 2.2021)7
ing options2,42,43. Benefits of large-​panel testing include
Regardless of age
potentially increased chance of uncovering a mutation,
• Metastatic prostate cancer
cost efficiency, and reduced testing fatigue, which can
• Very high risk: T3b–T4 or primary Gleason pattern 5 or >4 cores with grade group 4 or 5
occur with repeat testing42,43. Limitations of large-​panel
• High risk: T3a or grade group 4 or 5 or PSA >20 ng/ml
testing include an increased rate of detecting VUS, unex-
Any other risk group with any of the following pected secondary findings, mutations in genes in which
• Ashkenazi Jewish ancestry the cancer risk is unknown or not well established, and
• Prostate cancer family history: brother, father, or multiple family members finding mutations in genes without current management
diagnosed with prostate cancer at <60 years old or died from prostate cancer guidelines2,3,7,9,42.
(grade groups 2–5)
Acceptable specimens for germline genetic testing
• ≥3 cancers on the same side of the family (especially if diagnosed at ≤50 years old): bile at most laboratories include peripheral blood, saliva,
duct, breast, colorectal, endometrial, gastric, kidney, melanoma, ovarian, pancreatic,
and cheek and/or buccal swab. For some patients, such
prostate (grade groups 2–5), small bowel, or urothelial
as those with certain haematological malignancies or a
Philadelphia Prostate Cancer Consensus Conference 2019 (ref.2) history of transplant, a skin punch biopsy sample might
Recommend to be needed to avoid testing tumour cells for genetic muta-
• Men with metastatic prostate cancer tions. Germline genetic testing through most commer-
• Men with a family history of prostate cancer: men with one first-​degree relative cial laboratories generally has a 3–4-​week turnaround
or ≥2 male relatives with one of the following: time depending on testing techniques, sample used and
-- Diagnosed with prostate cancer at the age of <60 years billing practices. Each laboratory has individual practice
-- Any of whom died from prostate cancer or had metastatic prostate cancer
that providers should become familiar with.
Consider for
Importantly, the difference between ‘medical genetic
• Men with non-​metastatic prostate cancer with one of the following
-- Diagnosed with prostate cancer at <60 years old testing’ or ‘clinical genetic testing’ and ‘recreational
-- Advanced disease (T3a or higher) genetic testing’ or ‘direct-​to-​consumer’ testing needs to
-- Intraductal or ductal pathology be distinguished44–47. Medical or clinical genetic testing
-- Grade group 4 (Gleason sum 8) or above is a comprehensive process that involves extensive inter-
• Men with Ashkenazi Jewish ancestry rogation of multiple genes or genetic regions to detect
• Men with two or more cancers within the hereditary breast and ovarian cancer or thousands of pathogenic variants in disease-​related
Lynch syndrome spectrum in any relatives on the same side of the family (especially if genes of interest. The technology can detect multiple
diagnosed at <50 years old) types of mutations and deletions and/or duplications
and/or rearrangements. Genetic evaluation is guided by
AUA/ASTro/SUo Clinically localized Prostate Cancer 2017 (ref.89)
a genetics professional or health-​care professional and
Referral to genetic counselling should be considered for patients (and their families)
with high-​risk localized prostate cancer and a strong family history of specific cancers
is tailored to the patient’s medical history, family his-
(such as breast, ovarian, pancreatic, other gastrointestinal tumours) tory and personal preferences2,7,9,42. Recreational genetic
tests or direct-​to-​consumer tests are primarily designed
AUA/ASTro/SUo Advanced Prostate Cancer 2020 (ref.40)
for population-​level testing, with some advantages and
Patients with metastatic hormone-​sensitive prostate cancer should be offered genetic
important disadvantages to consider45–47. The main
counselling and genetic testing regardless of age and family history
advantage is increased access to genetic testing with-
EAU guidelines: Prostate Cancer41 out practice barriers or delays that can be involved in
Conduct germline testing for men with metastatic prostate cancer
scheduling appointments with genetic counselling15,45,46.
ASTRO, American Society for Radiation Oncology; AUA, American Urological Association; However, multiple important considerations need to be
EAU, European Association of Urology; NCCN, National Comprehensive Cancer Network; noted: the testing might not be suited to a particular
SUO, Society of Urologic Oncology.
patient’s medical condition or address family cancer

Nature Reviews | Urology

0123456789();:
Reviews

Table 2 | Prostate cancer panels available through experienced USA-​based clinical laboratories
Testing Multigene panels and genes Considerations
laboratory
Invitae Prostate cancer panel: ATM, BRCA1, BRCA2, Ability to customize gene panelsa
CHEK2, EPCAM, HOXB13, MLH1, MSH2, MSH6,
Reflex testing is available within a specific
NBN, PMS2, TP53
time frame
Prostate cancer HRR panel: ATM, BARD1,
Offers paired RNA testingb
BRCA1, BRCA2, BRIP1, CHEK2, FANCL, PALB2,
RAD51C, RAD51D Accepts skin punch biopsy specimens for testing
Ambry Genetics ProstateNext panel: ATM, BRCA1, BRCA2, Ability to customize gene panelsa
CHEK2, EPCAM, HOXB13, MLH1, MSH2, MSH6,
Reflex testing is available within a specific
NBN, PALB2, PMS2, RAD51D, TP53
time frame
Offers paired RNA testing (RNAinsight)b
Accepts skin punch biopsy specimens for testing
GeneDx Hereditary prostate cancer panel: ATM, BRCA1, Ability to customize gene panelsa
BRCA2, BRIP1, CHEK2, EPCAM, HOXB13, MLH1,
Reflex testing is available
MSH2, MSH6, NBN, PALB2, PMS2, RAD51C,
RAD51D, TP53 Accepts skin punch biopsy specimens for testing
Myriad Genetics myRisk panel: APC, ATM, AXIN2, BARD1, HOXB13 sequencing only
BMPR1A, BRCA1, BRCA2, BRIP1, CDH1, CDK4,
Reflex testing from small panels to largest panel
CDKN2A, CHEK2, EPCAM, GREM1, HOXB13
(Myriad myRisk) is available, but might require an
GALNT12, MLH1, MSH2, MSH3, MSH6, MUTYH,
additional specimen
NBN, NTHL1, PALB2, PMS2, POLE, POLD1, PTEN,
RAD51C, RAD51D, RNF43, RPS20, SMAD4, Accepts skin punch biopsy specimens for testing
STK11, TP53
Color Hereditary cancer panel: APC, ATM, BAP1, PMS2: only covers exons 1–11. Variant
BARD1, BMPR1A, BRCA1, BRCA2, BRIP1, CDH1, interpretation outside of this area of the gene
CDK4, CDKN2A, CHEK2, EPCAM, GREM1, MITF, is difficult owing to a pseudogene PMS2CL that
MLH1, MSH2, MSH6, MUTYH, NBN, PALB2, shares a similar coding sequence
PMS2, POLE, POLD1, PTEN, RAD51C, RAD51D,
HOXB13 not included
SMAD4, STK11, TP53
As of August 2021; furthermore, multiple additional cancer panels are available at these laboratories. HRR, homologous recombination
repair. aCustomization involves being able to choose the genes to test rather than a premade gene panel. bRNA testing involves
assessing the function of DNA variants usually reported as variants of uncertain significance (VUS). These VUS are usually in genes
associated with hereditary cancer and disrupt RNA splicing88.

history; the testing might be incomplete compared with Management of prostate cancer based on
clinical genetic testing (testing only a few mutations germline mutations
versus complete sequencing and alteration assessment; Historically, germline testing for cancer predispo-
limited genes tested)44–47; if test results return with no sition syndromes was performed to inform cancer
pathogenic variants, individuals might have false reas- risk, screening and cancer risk-​reduction measures44.
surance of no hereditary cancer condition in themselves However, advances in precision medicine have heralded
or their families, with subsequent missed hereditary a new era of expanded therapeutic utility of germline
cancer management and lack of appropriate follow-​up testing, which is now highly relevant to prostate
care4,44–47; misunderstanding of various types of genetic cancer1,2,4–7,9 (Table 1). Hereditary cancer management
results (such as VUS) can occur, with propagation of for syndrome-​associated cancers including and beyond
misinformation in families, potential adverse effects prostate cancer is also crucial7,9.
on life insurance or long-​term care insurance for muta- The influence of genetic testing in prostate can-
tion carriers, and adverse psychological consequences cer screening has been of long-​standing interest, with
without preparatory pre-​test genetic counselling44–47. emerging data influencing screening approaches2,7,48.
Furthermore, patients might present to their physicians Updated results from the IMPACT prostate cancer
or genetic counsellors with results of at-​home genetic screening trial were reported in 2019 (ref.48). This study
tests, which could then require repeat or confirmatory included men aged 40–69 years, 919 of whom were
germline testing, often at a cost to the patient. BRCA1 carriers, 709 of whom were BRCA1 non-​carriers,
Overall, criteria for genetic testing for prostate can- 902 of whom were BRCA2 carriers, and of whom 497
cer are tailored to the clinical features, pathological were BRCA2 non-​c arriers, who underwent 3 years
features and family history7,9. Genetic testing options of screening48. Higher prostate cancer incidence was
have expanded and providers should have an under- observed among BRCA2 carriers than non-​carriers
standing of the benefits and limitations of genetic test (19.4 versus 12.0; P = 0.03). BRCA2 carriers were also
options2,7,9,42. Close collaboration between health-​care diagnosed with prostate cancer at a younger age (61
providers and genetic counsellors is important for opti- versus 64 years; P = 0.04) and were more likely to have
mal testing and comprehensive recommendations to clinically significant disease than BRCA2 non-​carriers
patients2,7,9,14,42. (77% versus 40%; P = 0.01)48. Cancer incidences per

www.nature.com/nrurol

0123456789();:
Reviews

1,000 person-​years were 19 in BRCA2 carriers, 12 in BRCA2 Initially, data from phase II trials led to FDA des-
non-​carriers, 14 in BRCA1 carriers and 11 in BRCA1 non- ignations for PARP inhibitors including olaparib,
carriers48. Thus, the NCCN Genetic/Familial High Risk rucaparib and niraparib for men with mCRPC with
Assessment: Breast, Ovarian, and Pancreatic (Version germline or somatic alterations in DNA repair genes
1.2022) guideline recommends that men with BRCA2 involved in homologous recombination repair (such
mutations start PSA screening at the age of 40 and that as BRCA1 and BRCA2) and DNA damage signalling
men with BRCA1 mutations consider the same9. The and checkpoint regulation (for example, ATM)53–55,61,62.
NCCN Prostate Cancer Early Detection Guideline In 2020, the FDA approved two PARP inhibitors, olap-
(Version 2.2020) recommends considering shared arib and rucaparib, for men with mCRPC with specific
decision-​making for men carrying BRCA1 or BRCA2 DNA repair defects on progression after standard lines
mutations regarding prostate cancer screening starting of therapy5,6. PARP inhibitors work by using the con-
at the age of 40 years and to consider annual versus every cept of ‘synthetic lethality’, in which the co-​ordinated
other year screening49 (Table 1). Initial results from pros- effort of repairing single-​strand DNA breaks by PARP1
tate cancer screening in men with pathogenic variants and double-​strand breaks by homologous recombina-
in DNA MMR genes were also reported in the IMPACT tion repair is compromised62. In individuals who carry
study in 2021 (ref.50). Among 962 men enrolled, includ- BRCA1 or BRCA2 mutations, homologous recombina-
ing those carrying and not carrying DNA MMR muta- tion repair is defective and, therefore, PARP inhibition
tions, prostate cancer incidence (using a PSA threshold has clinical activity62. Rucaparib was granted acceler-
of >3.0 ng/ml) was higher in MSH2 carriers than in ated approval for BRCA1-​mutated or BRCA2-​mutated
MSH2 non-​carriers (4.3% versus 0.5%; P = 0.011) and mCRPC with previous treatment with androgen
in MSH6 carriers than in MSH6 non-​carriers (3.0% receptor-​directed therapy and taxane-​based chemo-
versus 0%; P = 0.034)50. The overall positive predictive therapy based on demonstrated clinical responses in
value of biopsy using a PSA threshold of 3.0 ng/ml was the TRITON2 study6. In TRITON2, overall response
51.4% (95% CI 34.0–68.6)50. To inform further strategies rates were reported to be 43.5% in men with BRCA1
for prostate cancer screening and to encompass other or BRCA2 mutations with measurable disease (95% CI
genes now available on prostate cancer multigene pan- 31.0–56.7%) and PSA response rate was 54.8% (95% CI
els, recommendations from the Philadelphia Prostate 45.2–64.1%)6. Olaparib was FDA approved for the treat-
Cancer Consensus Conference 2019 were published ment of mCRPC in men with deleterious or suspected
for prostate cancer screening based on mutation status deleterious germline or somatic homologous recombina-
and age at diagnosis of prostate cancer in the family2 tion repair gene mutations who had progressed follow-
(Table 1). In these recommendations, agreement to start ing previous treatment with enzalutamide or abiraterone
PSA screening at the age of 40 years or 10 years before based on the PROfound study5. Men with BRCA1 or
the youngest age at prostate cancer diagnosis among BRCA2 or ATM (cohort A) or multiple other DNA repair
men carrying BRCA2 mutations, and to consider the mutations (BARD1, BRIP1, CDK12, CHEK1, CHEK2,
same in men with BRCA1, ATM, HOXB13 and DNA FANCL, PALB2, PP2R2A, RAD51B, RAD51C, RAD51D,
MMR mutations is strong2. Currently, prophylactic RAD54L) (cohort B) were randomized to receive olap-
prostatectomy is not indicated for mutation carriers. arib 300 mg twice a day or enzalutamide or abiraterone.
Genetically based management of men with Progression-​free survival was significantly longer for
early-​stage prostate cancer is also evolving regarding men in cohort A treated with olaparib than treatment
active surveillance discussions51. In one study, 6 of with abiraterone or enzalutamide (median 7.4 versus
11 men with BRCA2 mutations on active surveillance 3.6 months; P < 0.001)5. Furthermore, overall survival
had significant upgrading of biopsy samples, either was also superior for men in cohort A treated with olap-
scheduled or prompted by serum PSA levels, compared arib to those who received abiraterone or enzalutamide
with 283 of 1,200 non-​BRCA2 carriers (adjusted HR 2.74; (median 19.1 versus 14.7 months, P = 0.02)64. Multiple
95% CI 1.26–5.96, P = 0.01)51. Surveillance biopsies were PARP inhibitors are being studied for the treatment
performed at 1–2 years after prostate cancer diagnosis of prostate cancer56. These trials include PARP inhib-
based on the cohorts included in the analysis51. These itors alone or in combination with androgen receptor
early results point to the potential need to include signalling inhibitors, immune checkpoint inhibitors,
germline test results, particularly for BRCA2 muta- chemotherapy or other agents56.
tions, in active surveillance discussions; further data Immunotherapy with pembrolizumab for prostate
are needed for definitive recommendations and current cancer is another area of advance in treatment for which
practice is evolving based on expert consensus2,52. assessment of DNA MMR deficiency is performed on
Prostate cancer is an important urological malig- tumour tissue, potentially leading to suspicion of Lynch
nancy with increasing options for precision medi- syndrome7,57,58. The mechanism of action is complex;
cine for men with mCRPC2,4–7,53–64. Genetic results are agents such as pembrolizumab promote tumour cell
increasingly influencing choice of therapy, such as death by binding to T cell PD1 receptors and disrupt-
PARP inhibitors for men with mCRPC with mutations ing interaction with PDL1 molecules on tumour cells,
in DNA repair genes, particularly BRCA1 and/or BRCA2 enabling immune attack on a tumour57,63. In 2017,
(refs4–7,53–56,62). Genetic mutations that influence treat- the FDA approved pembrolizumab for the treatment
ment decisions can be from germline testing or somatic of all solid tumours, including prostate cancer, that
testing and, therefore, might be based on hereditary or exhibit MMR deficiency and/or exhibit microsatellite
acquired genetic mutations, respectively7. instability57,58. Approximately 5–10% of prostate cancers

Nature Reviews | Urology

0123456789();:
Reviews

exhibit MMR deficiency and, therefore, a subset of these testing (genes to include, panel to use, etc.) to fully assess
patients with prostate cancer might be candidates for for an associated cancer predisposition syndrome8,10–12.
pembrolizumab59. Clinical trials are ongoing and fur- Some ‘red flags’ indicating a suspected hereditary cancer
ther data are emerging regarding additional biomarkers syndrome include personal and/or family history of can-
of response to immune checkpoint inhibitors in men cers and other clinical features known to be associated
with prostate cancer58,60. Pembrolizumab eligibility is with specific genetic syndromes, cancers diagnosed at
based on MMR deficiency or microsatellite instability unusually young ages, multiple blood relatives across
status determined in tumours, but the somatic genomic generations diagnosed with similar or genetically linked
analysis results might point to the need to evaluate the cancers, a personal history of bilateral and/or multiple
germline for Lynch syndrome39. primary cancers, and an ethnic background known to
NCCN guidelines address precision management of increase the risk of hereditary cancer (such as Ashkenazi
prostate cancer and are regularly updated7. The NCCN Jewish ancestry)8–12,28,29.
Prostate Cancer (Version 2.2021) guideline states that Genetic counsellors typically have an education-​
for men with mCRPC, alterations in DNA repair genes oriented approach to the counselling session that enables
such as BRCA1, BRCA2 and ATM might indicate PARP the patient to make an informed decision while address-
inhibitor therapy7. This guideline further suggests con- ing psychosocial concerns10. The pre-​test discussion
sideration of MMR deficiency or microsatellite status for comprises multiple components, including the purpose
pembrolizumab candidacy7. of testing (precision therapy, surgical decision-​making,
Overall, genetic information has become central risk assessment, effect on screening, cascade testing),
to management approaches for prostate cancer, with a estimates of cancer risk, risks and benefits of testing,
rising role in determining which precision medicine to types of test results that could be returned (pathogenic
choose, early-​stage disease management and screening7,9. or probable pathogenic variant, VUS, negative), options
Olaparib and rucaparib are FDA approved for the treat- for multigene panel testing, cost of genetic testing, and
ment of men with mCRPC after progression on various psychological counselling and support8,10–13. Communi­
standard therapies, and have unique approvals based cation of genetic risk for at-​risk individuals also involves
on a spectrum of germline mutations5,6. Thus, genetic a discussion of statistical probabilities for mutation pre­
testing and genetic counselling need to be considered sence and implications for personal and family cancer
by urologists and oncologists for appropriate patients risk. In the advanced-​cancer setting, genetic counsellors
during the course of management or treatment. need to adapt counselling models towards how genetic
testing might inform options for targeted therapy and
Genetics care delivery guide panel testing2,4,7,14.
Traditionally, genetics care delivery has been conducted Another component of pre-​test counselling is discus-
through genetic counselling by specialist genetics coun- sion of the benefits and limitations of genetic testing2,8–13.
sellors; however, novel approaches are also being used to Benefits of genetic testing include information for
improve accessibility and speed. medical management, precision therapy options, iden-
tification of additional cancer risks to inform screening,
Genetic counselling. Many patients with prostate cancer and identification of at-​risk family members to address
or at risk of developing prostate cancer are now increas- cancer risks. Limitations of genetic testing include tech-
ingly in need of germline testing2,7,9, necessitating pre-​ nical issues (such as the inability to detect certain genetic
test informed decision-​making for germline testing that variants with standard testing technology), limited
addresses precision management and screening and knowledge of cancer risk or management for some genes,
encompasses hereditary cancer management. Genetics identification of VUS and complex genetic findings8–13.
care delivery can occur through the traditional approach The chances of having a VUS reported are increased
of referral to genetic counselling or, increasingly, by when an increased number of genes are tested 2,65.
using hybrid approaches to handle the rising volume of Furthermore, VUS rates are increased in patient popu­
patients in need of germline testing and reduce barriers lations with diverse race or ethnic backgrounds65.
to care14–17. Health-​care providers and genetic counsel- Additional considerations include genetic discrimina-
lors need to have a working knowledge of hereditary tion protections and gaps in protection under the 2008
cancer assessment, the nuances of genetic evaluation, Genetic Information Nondiscrimination Act (GINA)66.
and prostate cancer management across the stage and The GINA law provides protection related to health
risk spectrum (Fig. 1; Box 1). insurance and employment discrimination for muta-
The generally accepted model of genetic counselling tion carriers in most scenarios. The GINA law does not
can be divided into pre-​test counselling (before genetic provide protections for military health insurance and
testing) and post-​test counselling (after test results employment, life insurance, disability and long-​term
return)8,10–12 (Fig. 1). The pre-​test counselling visit entails care insurance66. Currently, this law only applies to adults
gaining knowledge of a patient’s personal medical his- who have been tested; how the law will apply to blood
tory, family history and previous genetic testing in the relatives regarding effect on insurance might evolve
family8,10–12. A three-​generation pedigree, including over time.
the patient’s medical history, cancers in male and female In a post-​test disclosure session, genetic test results
relatives, and ethnicity, is gathered at the pre-​test session. are discussed with the patient along with recommen-
This information is used to assess the patient’s risk of dations. Genetic results are reviewed to enhance under-
carry­ing a mutation and informs the strategy for germline standing of associated medical implications for patients

www.nature.com/nrurol

0123456789();:
Reviews

Pre-test counselling and pre-test informed consent Genetic testing Post-test disclosure

Genetic evaluation

Pre-test discussion elements Genetic testing considerations Post-test recommendations


• Take personal cancer history, pathology, • Focused versus cancer-specific • Review genetic test results
ancestry and syndromic features versus comprehensive cancer • Pathogenic or probably pathogenic: gene-based,
• Take family history panel versus reflex panel personal-based and family history-based
• Obtain genetic and somatic testing records • Laboratory with experienced recommendations (Table 1)
testing and variant • VUS and negative: family history-based and
• Address purpose of genetic testing
reclassification programmes personal history-based recommendations
• Estimate cancer risk
• Data privacy policy of laboratory • Discuss genetic testing in family
• Discuss benefits and limitations of testing
• If pathogenic or probably pathogenic variant:
• Discuss options for multipanel testing
cascade testing for this variant in blood relatives
• Discuss cost of genetic testing
• Provide resources or strategies for patients to
• Discuss types of genetic test results discuss genetic results with children, siblings,
• Discuss effect on family cancer risks parents and extended blood relatives
• Consider psychosocial factors • If VUS or negative: testing family members might
• Discuss GINA law be appropriate based on family history
• Assess psychological effect (anxiety, worry, guilt,
coping with cancer treatment)
• Provide results to full care team

Strategies

Approaches Sample collection Approaches


• Genetic counselling or hybrid model between • Saliva • Genetic counselling or hybrid model between
health-care provider and genetic counsellor • Blood (mobile phlebotomy) health-care provider and genetic counsellor
• Use online tools (such as My Family Health • Skin punch biopsies under • Alternative delivery: telehealth-based or
Portrait) to obtain family history telephone-based delivery
certain circumstances
• Video to deliver pre-test information • Artificial intelligence (chatbots)
• Alternative delivery: telehealth-based
(web-based live video conferencing) or
telephone-based delivery
• Artificial intelligence (chatbots)

Clinical and psychosocial concerns that might influence patient counselling experience and decision-making for prostate cancer genetic testing
• Clinical concerns: new prostate cancer • Psychosocial or quality-of-life concerns: urinary incontinence, erectile dysfunction, ADT-related hot
diagnosis, development of metastatic flushes and gynaecomastia, chemotherapy effects; difficulty coping with new cancer diagnosis or disease
disease, progression of disease, candidacy progression; guilt over possible hereditary cancer risk in family; being overwhelmed regarding multiple
for targeted therapies aspects of health-care decision-making

Fig. 1 | genetic evaluation process for patients with or at risk of developing prostate cancer. The process and
elements involved in pre-​test counselling and informed consent, genetic testing and post-​test disclosure. Special
considerations in each step in the process and unique clinical and psychosocial concerns that can influence decision-​
making are shown. ADT, androgen-​deprivation therapy; GINA, Genetic Information Nondiscrimination Act; VUS, variants
of uncertain significance.

and their family members while also addressing emo- pancreatic cancer screening (if a family history of pan-
tional and psychosocial needs8–13. Medical management creatic cancer exists) with a gastroenterologist and be
discussion is based on genetic testing results and the evaluated by a dermatologist for risk of melanoma9.
patient’s specific family history. Pathogenic or probable These visits can be coordinated by genetic counsellors,
pathogenic variants can influence medical manage- the patient’s primary-​care doctor or be the responsibility
ment for urological malignancies, cancer risk recom- of the patient. Improved support for patients who carry
mendations and cascade testing of blood relatives2,7,9. mutations to have multidisciplinary screening is needed.
Coordination of cancer screening for mutations can VUS are also reported in genetic test results; patients
be complex, as multiple cancer risks might need to be need to understand how to interpret these findings. VUS
addressed. For example, men with BRCA2 mutations do not affect management at the time of reporting2,10.
without prostate cancer are usually recommended to VUS are followed over time by genetic testing laboratories
start prostate cancer screening at the age of 40 years by for evidence in support of or against pathogenicity67,68.
their urologist or their primary-​care doctor9; they are A 2018 retrospective cohort study including over 1 mil-
also recommended to have clinical breast examinations lion genetic tests showed that 7.7% of VUS results were
by their physician starting at the age of 35 years, discuss reclassified, of which 91.2% were downgraded to benign

Nature Reviews | Urology

0123456789();:
Reviews

or probably benign68. Thus, counselling sessions need to counselling has needed to be adapted for increased access
provide this information to patients and include plans to testing, rapid return of results, and to mitigate the rela­
to recontact patients if VUS reclassification updates tive shortage of genetic counselling14–16. Adaptations to
occur, to provide revised recommendations as needed. the traditional genetic counselling model have, therefore,
For patients who receive VUS results or totally negative been emerging, in which oncologists and urologists are
genetic results, referral to genetic counselling might be becoming more involved in both pre-​test and post-​test
needed, and, if family cancer history is strong, they might contexts (Fig. 1; Box 1). In this hybrid, point-​of-​care
need to receive comprehensive family history-​based model, the physician performs the pre-​test consent and
recommendations or to determine whether additional orders genetic testing. When results are returned, they
relatives need germline testing1,2,4. review the results with the patient regarding effect on
Cascade testing in a family of a patient with a patho- treatment and then refer a subset of patients to a genetic
genic variant or probable pathogenic variant (mutation) counsellor to discuss genetic results, provide full recom-
takes co-​ordinated effort between a genetic counsellor, mendations based on personal and family history, and
the patient, and their family. Once a patient is identified discuss cascade testing or further testing in the family
as having a pathogenic variant or probable pathogenic as indicated14,17. This approach facilitates rapid return of
variant, the genetic counsellor typically needs to obtain genetic results, but some complexities in practice need
a release of information from the patient to use this to be considered and addressed proactively (Box 1). One
information to guide genetic testing of blood relatives. consideration is the time needed to perform appropriate
The patient needs to communicate information of their pre-​test informed consent for genetic testing. Multiple
genetic test results to blood relatives, who then need to elements need to be discussed and understood by
contact their local genetics services to make an appoint- patients to make an informed decision for testing2,7,9,10.
ment for genetic counselling and genetic testing. These To facilitate pre-​test information delivery, videos are
genetic counselling sessions for relatives will include being studied in which patients view a genetics educa-
the genetic results of the proband (original patient), the tion video to understand the genetic testing process and
relative’s medical history and family history, discussion considerations before pursuing genetic testing72. Patient-​
of pre-​test elements, and optimal testing approach10. The reported outcomes in a male population undergoing
relative will next undergo genetic testing and then return prostate cancer germline testing from one study (n = 127)
for disclosure of results and recommendations based on revealed that most men (71%) chose a pre-​test video
their test results10. compared with genetic counselling (29%) (P < 0.001),
Communication of genetic results and recommenda- with no negative effect on patient satisfaction, decisional
tions in families can be challenging. Providing resources conflict for genetic testing, cancer genetics knowledge or
or strategies for patients to discuss genetic results with uptake of genetic testing72.
children, siblings, parents and extended blood relatives is In a hybrid model, intake of a three-​generation ped-
an important part of genetic counselling69,70. Some con- igree to accurately determine extent of testing might be
troversies that genetic counsellors can encounter centre challenging in busy clinics. One approach to increase the
around difficult family dynamics, when family members yield of full family history intake in the pre-​test setting
do not wish to discuss their genetic results with relatives includes use of online tools (such as My Family Health
or have little contact with family members69,70. In such Portrait from the Centers for Disease Control and
cases, balancing a patient’s autonomy and confidentiality Prevention)73 into which patients can input family his-
with a duty to warn relatives of the potential genetic risk tory at home before their appointments. Furthermore,
becomes difficult69–71. The American Society of Human consideration of optimal panels for testing by ordering
Genetics permits a provider to disclose information to physicians, such as focused, guideline-​based, compre-
a third party only under specific circumstances, such hensive or reflex panels, which cover the patient’s med-
as when encouragement to disclose to family members ical history, family history and take patient preferences
has failed and a serious risk that is identifiable and can into account, becomes important2. In particular, reflex
be prevented, treated or reduced by early monitoring testing might be preferable in hybrid models to enable
exists71. flexibility for upfront ordering and expanded testing
In summary, the genetic counselling process encom- based on full family history information obtained along
passes pre-​test and post-​test sessions, focuses on edu- the care pathway2. In the USA, insurance coverage can
cating and aiding the patient in choosing appropriate also be a limiting factor as some policies require the
genetic testing, and considers psychosocial concerns that patient to undergo genetic counselling with a certified
might influence decision-​making for genetic testing8–13. genetics professional before undergoing genetic test-
Genetic test results are not always straightforward as ing in order for insurance to cover the cost of testing.
genetic testing technology is limited and guidelines can Various countries have national or private policies that
vary7,9,10,39. Results and implications for patients and also need to be followed. Urologists or oncologists seek-
family members are important to address, along with ing to employ hybrid genetics care delivery models in
sharing of results with family members to enable cascade their practices are encouraged to have increased work-
testing10. ing knowledge of the principles and practice of genetic
counselling and genetic testing2,74. Finally, proactive
Novel genetics care delivery approaches. In the era of development of genetics care delivery and referral cri-
expanded germline testing and precision medicine, the teria between health-​care providers and genetic coun-
traditional model of referral of all patients to genetic sellors is encouraged when instituting hybrid models

www.nature.com/nrurol

0123456789();:
Reviews

in practice to streamline pre-​test and post-​test care and Unique aspects of genetic counselling for men
optimize genetic testing14 (Box 1; Fig. 1). with prostate cancer
To enhance access to genetic counselling, novel With the expansion of genetic testing criteria for pros-
technology-​based approaches are in use or under devel- tate cancer7,9, improved insight is needed in to how men
opment. Telephone-​based and telehealth-​based (also process the pre-​test counselling discussion, understand
called telegenetics when in the context of providing their genetic results and handle the anxiety or guilt of
web-​based video-​conferencing genetic consultation) are having an inherited genetic mutation. Many men might
alternative ways of disseminating genetic counselling75,76. have high satisfaction with their genetic evaluation
More research is needed concerning the use of these experience, but some men may have difficulty with the
approaches in patients with prostate cancer, but pre- process. Patients might need additional resources or
vious research in women primarily engaged in genetic support to understand their genetic results. For exam-
counselling for HBOC reported that telephone-​based ple, results are emerging that report that some men who
genetic counselling was not inferior to standard genetic undergo prostate cancer genetic testing might have lim-
counselling for patient-​reported outcomes77,78. Results ited understanding of VUS82. Given that ~30% of men
of one randomized trial that included 669 women undergoing prostate cancer germline testing will have
undergoing genetic counselling for HBOC showed the VUS reported1,83, this proportion represents a substan-
non-​inferiority of telephone-​based genetic counselling tial number of men in need of reinforcement of infor­
versus in-​person genetic counselling for knowledge, mation to enhance understanding of results and to limit
satisfaction, decision conflict, distress and quality of propagation of misinformation in families82.
life77. Results of another study showed that anxiety, Furthermore, understanding quality-​of-​life issues
cancer-​specific distress, perceived personal control, and for men dealing with prostate cancer treatment across
decisional conflict for genetic testing were not inferior the stage spectrum is needed when discussing germline
at the 1-​year follow-​up point between telephone-​based testing. Men might be dealing with treatment-​related
counselling and in-​person counselling among women effects, such as urinary incontinence, erectile dysfunc-
undergoing evaluation for HBOC78. Web-​based telege- tion, fatigue, hot flushes, weight gain, gynaecomastia, or
netics has also been reported to have high satisfaction loss of libido and/or sexual dysfunction caused by treat-
rates among patients undergoing genetic counselling and ments including prostatectomy, radiotherapy, androgen
genetic testing76. A randomized trial of 130 patients (90% deprivation therapy or chemotherapy84–86. Men on active
of whom were women) undergoing cancer genetic coun- surveillance might fear disease progression and men
selling primarily for HBOC or Lynch syndrome reported with biochemical recurrence might fear disease recur-
no difference in patient satisfaction between telegenet- rence (Fig. 1). Clinical experience shows that most of the
ics and in-​person genetic counselling (P = 0.03)76. Key time these issues are not barriers for men to undergo
barriers to address include comfort with and access to germline testing, but occasionally current quality-​of-​life
technology, reimbursement, co-​ordination of sample issues might make processing genetic information diffi-
collection for genetic testing, and privacy and ethical cult or overwhelming87. Difficulty coping with progres-
issues. In the virtual era of the COVID-19 pandemic, sive metastatic disease, new development of metastatic
the importance of remote genetics care delivery, such as disease, or a new diagnosis of prostate cancer with the
telegenetic visits, and remote sample collection, such multitude of treatment or management options could
as through mobile phlebotomy, have increased 79,80. make informed decision-​making for germline test-
Finally, chatbots are also being studied in genetic edu- ing challenging for some men. If men initially decline
cation, consent and counselling delivery81. A chatbot is germline testing, repeated offering of testing to eligible
a computer programme that simulates human conver­ men would be encouraged. Dedicated patient-​reported
sations and enables communication of information by outcomes data in male populations undergoing genetic
messages or voice command81. Chatbots have been devel- testing for prostate cancer are needed. A patient-​driven
oped to aid delivery of genetic testing consent, follow-up registry (PROGRESS Registry) is currently actively
monitoring and cascade testing of family members, recruiting men in the USA who have had prostate
with patients supporting this technology from focus cancer-​related germline testing to garner patient expe-
group data81. In one study of focus group participants riences to support development of resources for men and
enrolled in a genomics research programme in which their families.
chatbots were used, participants were supported using Overall, identifying and addressing the clinical,
chatbots to consent to genomics research and to inter- genetic and psychosocial issues for men is important
act with health-​care providers for recommendations for facilitating decision-​making on genetic testing
and co-​ordination of care, as well as to share genetic and for supporting men in their cancer treatment and
information with relatives81. Further research is needed genetic evaluation experience.
among men with prostate cancer regarding utility and
satisfaction with genetics care delivery models and tools Conclusions
for male-​specific patient-​reported outcomes. Genetic evaluation for men with prostate cancer or at
Overall, with the rising volume of patients in need risk of prostate cancer development is a specialized
of genetic counselling and germline testing, current and area requiring knowledge of pre-​test genetic informa-
future models of genetic counselling need to be adapted tion, genetic testing options and approaches, complex
to provide responsible delivery of genetic counselling to hereditary cancer syndromes, and unique considerations
patients on an individualized basis. important for patients and providers to understand.

Nature Reviews | Urology

0123456789();:
Reviews

Genetic counselling, hybrid models, and technology- contributing to prostate cancer across diverse popu­
based approaches are essential to provide access to lations to enhance the genetic evaluation impact and
genetic testing for men and their families. This popu- experience for men and their families.
lation of patients deserves dedicated studies of patient-
reported outcomes and increased knowledge of genetics Published online xx xx xxxx

1. National Cancer Institute. Genetics of Prostate 18. Sztupinszki, Z. et al. Detection of molecular signatures 40. Lowrance, W et al. Advanced prostate cancer: AUA/
Cancer (PDQ®) – Health Professional Version. of homologous recombination deficiency in prostate ASTRO/SUO guideline. AUA www.auanet.org/
NIH https://www.cancer.gov/types/prostate/hp/ cancer with or without BRCA1/2 mutations. Clin. Cancer guidelines/advanced-​prostate-cancer (2020).
prostate-genetics-pdq (2022). Res. 26, 2673–2680 (2020). 41. Mottet, N et al. EAU guidelines: prostate cancer. EAU
2. Giri, V. N. et al. Implementation of germline testing 19. National Human Genome Research Institute. Talking https://uroweb.org/guideline/prostatecancer (2020).
for prostate cancer: Philadelphia Prostate Cancer glossary of genetic terms. NIH https://www.genome.gov/ 42. Domchek, S. M., Bradbury, A., Garber, J. E., Offit, K.
Consensus Conference 2019. J. Clin. Oncol. 38, genetics-​glossary (2022). & Robson, M. E. Multiplex genetic testing for cancer
2798–2811 (2020). 20. Welcsh, P. L. & King, M.-C. BRCA1 and BRCA2 and the susceptibility: out on the high wire without a net?
This paper provides a comprehensive review of genetics of breast and ovarian cancer. Hum. Mol. Genet. J. Clin. Oncol. 31, 1267–1270 (2013).
genes involved in prostate cancer risk, screening 10, 705–713 (2001). 43. Lynce, F. & Isaacs, C. How far do we go with genetic
and therapy, along with a genetic evaluation 21. Pritchard, C. C. et al. Inherited DNA-​repair gene evaluation. Am. Soc. Clin. Oncol. Educ. Book. 35,
framework for practice. mutations in men with metastatic prostate cancer. e72–e78 (2016).
3. LaDuca, H. et al. A clinical guide to hereditary cancer N. Engl. J. Med. 375, 443–453 (2016). 44. Cancer Genetics Risk Assessment and Counseling.
panel testing: evaluation of gene-​specific cancer This was a seminal paper describing rates of National Cancer Institute PDQ®. NIH https://
associations and sensitivity of genetic testing criteria germline mutations in men with mCRPC that www.cancer.gov/about-​cancer/causes-​prevention/
in a cohort of 165,000 high-​risk patients. Genet. Med. supported expansion of germline testing in men genetics/risk-​assessment-pdq (2022).
22, 407–415 (2020). with mCRPC. 45. Oh, B. Direct-​to-consumer genetic testing: advantages
4. Cheng, H. H. et al. Germline and somatic mutations in 22. Nicolosi, P. et al. Prevalence of germline variants in and pitfalls. Genomics Inform. 17, e33 (2019).
prostate cancer for the clinician. J. Natl Compr. Cancer prostate cancer and implications for current genetic 46. Horton, R. et al. Direct-​to-consumer genetic testing.
Netw. 17, 515–521 (2019). testing guidelines. JAMA Oncol. 5, 523–528 (2019). BMJ 367, l5688 (2019).
5. de Bono, J. et al. Olaparib for metastatic castration-​ 23. Giri, V. N. et al. Inherited mutations in males 47. Roberts, J. S. & Ostergren, J. Direct-​to-consumer
resistant prostate cancer. N. Engl. J. Med. 382, undergoing multigene panel testing for prostate genetic testing and personal genomics services:
2091–2102 (2020). cancer — emerging implications for personalized a review of recent empirical studies. Curr. Genet.
This paper details a phase III randomized trial of prostate cancer genetic evaluation. J. Clin. Oncol. Med. Rep. 1, 182–200 (2013).
the clinical activity of olaparib in men with mCRPC Precis. Oncol. 1, 1–17 (2017). 48. Page, E. C. et al. Interim results from the IMPACT study:
and DNA repair mutations and led to FDA approval 24. Giri, V. N. & Beebe-​Dimmer, J. L. Familial prostate evidence for prostate-​specific antigen screening in
for men with mCRPC and DNA repair mutations cancer. Semin. Oncol. 43, 560–565 (2016). BRCA2 mutation carriers. Eur. Urol. 76, 831–842
after progressing on initial lines of therapy. 25. Castro, E. et al. Germline BRCA mutations are (2019).
6. Abida W., et al. Rucaparib in men with metastatic associated with higher risk of nodal involvement, distant 49. National Comprehensive Cancer Network Clinical
castration-​resistant prostate cancer harboring a metastasis, and poor survival outcomes in prostate Guidelines in Oncology (NCCN Guidelines®): prostate
BRCA1 or BRCA2 gene alteration. J. Clin. Oncol. cancer. J. Clin. Oncol. 31, 1748–1757 (2013). cancer early detection (Version 2.2020) (NCCN, 2020).
https://doi.org/10.1200/JCO.20.01035 (2020). 26. Akbari, M. R. et al. The impact of a BRCA2 mutation 50. Bancroft, E. K. et al. A prospective prostate cancer
This paper details results of clinical activity of on mortality from screen-​detected prostate cancer. screening programme for men with pathogenic variants
rucaparib in men with mCRPC and BRCA mutations Br. J. Cancer 111, 1238–1240 (2014). in mismatch repair genes (IMPACT): initial results from
and led to FDA approval for men with mCRPC and 27. Castro, E. et al. Effect of BRCA mutations on metastatic an international prospective study. Lancet Oncol. 22,
BRCA mutations after progressing on initial lines relapse and cause-​specific survival after radical 1618–1631 (2021).
of therapy. treatment for localised prostate cancer. Eur. Urol. 68, 51. Carter, H. B. et al. Germline mutations in ATM and
7. National Comprehensive Cancer Network. Clinical 186–193 (2015). BRCA1/2 are associated with grade reclassification
Guidelines in Oncology (NCCN Guidelines®): Prostate 28. King, M. C. et al. Breast and ovarian cancer risks due in men on active surveillance for prostate cancer.
Cancer (Version 2.2021). NCCN https://www.nccn.org/ to inherited mutations in BRCA1 and BRCA2. Science Eur. Urol. 75, 743–749 (2019).
guidelines/guidelines-​detail?category=1&id=1459 02, 643–646 (2003). 52. Loeb, S. & Giri, V. N. Clinical implications of germline
(2021). 29. Kirchhoff, T. et al. BRCA mutations and risk of prostate testing in newly diagnosed prostate cancer. Eur. Urol.
8. National Cancer Institute. Cancer Genetics Overview cancer in Ashkenazi Jews. Clin. Cancer Res. 10, Oncol. 4, 1–9 (2021).
(PDQ®) — Health Professional Version. NIH https:// 2918–2921 (2004). 53. Mateo, J. et al. DNA-​repair defects and olaparib in
www.cancer.gov/about-​cancer/causes-​prevention/ 30. Abul-​Husn, N. S. et al. Exome sequencing reveals a metastatic prostate cancer. N. Engl. J. Med. 373,
genetics/overview-​pdq (2022). high prevalence of BRCA1 and BRCA2 founder variants 1697–1708 (2015).
9. National Comprehensive Cancer Network. National in a diverse population-​based biobank. Genome Med. 54. Abida, W. et al. Preliminary results from TRITON2:
Comprehensive Cancer Network Clinical Guidelines 12, 2 (2020). a phase 2 study of rucaparib in patients with mCRPC
in Oncology (NCCN Guidelines®): Genetic/Familial 31. Carter, B. S. et al. Hereditary prostate cancer: associated with homologous recombination repair
High-​Risk Assessment: Breast, Ovarian, and Pancreatic epidemiologic and clinical features. J. Urol. 150, gene alterations. Ann. Oncol. 29 (Suppl. 8),
(Version 1.2022). NCCN https://www.nccn.org/ 797–802 (1993). viii271–viii302 (2018).
guidelines/guidelines-​detail?category=2&id=1503 32. Ewing, C. M. et al. Germline mutations in HOXB13 55. Smith, M. R. et al. Pre-​specified interim analysis
(2022). and prostate-​cancer risk. N. Engl. J. Med. 366, of GALAHAD: a phase II study of niraparib in patients
10. Riley, B. D. et al. Essential elements of genetic cancer 141–149 (2012). (pts) with metastatic castration-​resistant prostate
risk assessment, counseling, and testing: updated This paper established HOXB13 as a hereditary cancer (mCRPC) and biallelic DNA-​repair gene
recommendations of the National Society of Genetic prostate cancer gene. defects (DRD). Ann. Oncol. 30 (Suppl. 5), V884–V885
Counselors. J. Genet. Couns. 21, 151–161 (2012). 33. Witte, J. S. et al. HOXB13 mutation and prostate (2019).
This paper highlights principles of genetic cancer: studies of siblings and aggressive disease. 56. Virtanen, V. et al. PARP inhibitors in prostate cancer —
counselling that are important in clinical genetic Cancer Epidemiol. Biomark. Prev. 22, 675–680 the preclinical rationale and current clinical
evaluation practice regarding genetic testing for (2013). development. Genes 10, 565 (2019).
prostate cancer. 34. Laitinen, V. H. et al. HOXB13 G84E mutation in 57. Le, D. T. et al. Mismatch repair deficiency predicts
11. Resta, R. et al. A new definition of genetic counseling: Finland: population-​based analysis of prostate, breast, response of solid tumors to PD-1 blockade. Science
national society of genetic counselors’ task force and colorectal cancer risk. Cancer Epidemiol. Biomark. 357, 409–413 (2017).
report. J. Genet. Couns. 15, 77–83 (2006). Prev. 22, 452–460 (2013). 58. Graff, J. N. et al. Early evidence of anti-​PD-1 activity in
12. Uhlmann, W. R., Schuette, J. L., & Yashar, B. A Guide to 35. Brechka, H., Bhanvadia, R. R., VanOpstall, C. enzalutamide-​resistant prostate cancer. Oncotarget 7,
Genetic Counseling 2nd edn (Wiley-​Blackwell, 2009). & Vander Griend, D. J. HOXB13 mutations and 52810–52817 (2016).
13. Schneider, K. A. Counseling about Cancer: Strategies binding partners in prostate development and cancer: 59. Cheng, H. H. The resounding effect of DNA repair
for Genetic Counseling 3rd edn (Wiley-​Blackwell, 2009). function, clinical significance, and future directions. deficiency in prostate cancer. Urol. Oncol. 36,
14. Giri, V. N., Hyatt, C. & Gomella, L. G. Germline testing Genes Dis. 4, 75–87 (2017). 385–388 (2018).
for men with PCA: navigating an expanding new world 36. Raymond, V. M. et al. Elevated risk of prostate cancer 60. Antonarakis, E. S. et al. Pembrolizumab for treatment-​
of genetic evaluation for precision therapy and precision among men with Lynch syndrome. J. Clin. Oncol. 31, refractory metastatic castration-​resistant prostate
management. J. Clin. Oncol. 37, 1455–1459 (2019). 1713–1718 (2013). cancer: multicohort, open-​label phase II KEYNOTE-199
15. Hughes, K. S. Genetic testing: what problem are 37. Lynch, H. et al. Milestones of Lynch syndrome: study. J. Clin. Oncol. 38, 395–405 (2020).
we trying to solve? J. Clin. Oncol. 35, 3789–3791 1895–2015. Nat. Rev. Cancer 15, 181–194 (2015). 61. Maréchal, A. & Zou, L. DNA damage sensing by the
(2017). 38. Dominguez-​Valentin, M. et al. Cancer risks by gene, ATM and ATR kinases. Cold Spring Harb. Perspect. Biol.
16. Abacan, M. et al. The global state of the genetic age, and gender in 6350 carriers of pathogenic 5, a012716 (2013).
counseling profession. Eur. J. Hum. Genet. 27, mismatch repair variants: findings from the Prospective 62. Mateo, J. et al. A decade of clinical development
183–197 (2019). Lynch Syndrome Database. Genet. Med. 22, 15–25 of PARP inhibitors in perspective. Ann. Oncol. 30,
17. Thompson, M. A. et al. Coordinating an oncology (2020). 437–1447 (2019).
precision medicine clinic within an integrated health 39. National Comprehensive Cancer Network. Clinical 63. Boyiadzis, M. M. et al. Significance and implications of
system: lessons learned in year one. J. Patient Cent. Guidelines in Oncology (NCCN Guidelines®): Lynch FDA approval of pembrolizumab for biomarker-​defined
Res. Rev. 6, 36–45 (2019). syndrome (Version 2.2021) (NCCN, 2021). disease. J. Immunother. Cancer 6, 35 (2018).

www.nature.com/nrurol

0123456789();:
Reviews

64. Hussain, M. et al. Survival with olaparib in metastatic counseling: keeping up in the era of precision medicine. 86. Sharpley, C. F., Birsika, V. & Denham, J. W. Factors
castration-​resistant prostate cancer. N. Engl. J. Med. Am. J. Med. Genet. 178C, 24–37 (2018). associated with feelings of loss of masculinity in men
383, 2345–2357 (2020). 76. Buchanan, A. H. et al. Randomized trial of telegenetics with prostate cancer in the RADAR trial.
65. Caswell-​Jin, J. L. et al. Racial/ethnic differences in vs. in-​person cancer genetic counseling: cost, patient Psychooncology 23, 524–530 (2014).
multiple-​gene sequencing results for hereditary cancer satisfaction and attendance. J. Genet. Couns. 24, 87. Bester, J., Cole, C. & Kodish, E. The limits of informed
risk. Genet. Med. 20, 234–239 (2018). 961–970 (2015). consent for an overwhelmed patient: clinicians’ role
66. U.S. Government Publishing Office Genetic Information 77. Schwartz, M. D. et al. Randomized noninferiority trial in protecting patients and preventing overwhelm.
Nondiscrimination Act of 2008 Public Law 110–233, of telephone versus in-​person genetic counseling for AMA J. Ethics 18, 869–886 (2016).
122 Stat. 881 (2008). hereditary breast and ovarian cancer. J. Clin. Oncol. 88. Karam, R. et al. Assessment of diagnostic outcomes
67. Richards, S. et al. Standards and guidelines for the 32, 618–626 (2014). of RNA genetic testing for hereditary cancer.
interpretation of sequence variants: a joint consensus 78. Kinney, A. Y. et al. Randomized noninferiority trial JAMA Netw. Open 2, e1913900 (2019).
recommendation of the American College of Medical of telephone delivery of BRCA1/2 genetic counseling 89. Sanda, M. G. et al. Clinically Localized Prostate
Genetics and Genomics and the Association for compared with in-​person counseling: 1-year follow-​up. Cancer: AUA/ASTRO/SUO Guideline (2017). AUA
Molecular Pathology. Genet. Med. 17, 405–424 J. Clin. Oncol. 34, 2914–2924 (2016). https://www.auanet.org/guidelines/guidelines/
(2015). 79. Gadzinski, A. J. & Ellimoottil, C. Telehealth in urology prostate-​cancer-clinically-​localized-guideline (2017).
68. Mersch, J. et al. Prevalence of variant reclassification after the COVID-19 pandemic. Nat. Rev. Urol. 17,
following hereditary cancer genetic testing. JAMA 363–364 (2020). Author contributions
320, 1266–1274 (2018). 80. Mauer, C. et al. Adapting genetic counseling operations Both authors made substantial contributions to discussion of
69. Gallo, A. M., Angst, D. B. & Knafl, K. A. Disclosure amidst the COVID-19 pandemic. J. Genet. Couns. 30, content, researched data for the article, wrote the manuscript
of genetic information within families. Am. J. Nurs. 949–955 (2021). and reviewed and edited the article before submission.
109, 65–69 (2009). 81. Schmidlen, T., Schwartz, M., DiLoreto, K., Kirchner, H. L.
70. Daly, M. B. A family-​centered model for sharing & Sturm, A. C. Patient assessment of chatbots for the Competing interests
genetic risk. J. Law Med. Ethics 43, 545–551 (2015). scalable delivery of genetic counseling. J. Genet. Couns. The authors declare no competing interests.
71. Perry, T. J. et al. The duty to warn at-​risk relatives — 28, 1166–1177 (2019).
the experience of genetic counselors and medical 82. Giri, V. N. et al. Understanding of multigene Peer review information
geneticists. Am. J. Med. Genet. 182, 314–321 test results among males undergoing germline Nature Reviews Urology thanks Olivier Cussenot, George
(2019). testing for inherited prostate cancer: implications Netto, Celestia Higano, Alexandra Sokolova, Rana McKay
72. Russo, J. et al. Pretest genetic education video vs. for genetic counseling. Prostate 78, 879–888 and the other, anonymous, reviewer(s) for their contribution
genetic counseling for men considering prostate (2018). to the peer review of this work.
cancer germline testing: a patient-​choice study to 83. Giri, V. N. et al. Germline genetic testing for inherited
address urgent practice needs. J. Clin. Oncol. Precis. prostate cancer in practice: implications for genetic Publisher’s note
Oncol. 5, PO.21.00238 (2021). testing, precision therapy, and cascade testing. Prostate Springer Nature remains neutral with regard to jurisdictional
73. Centers for Disease Control and Prevention. My Family 79, 333–339 (2019). claims in published maps and institutional affiliations.
Health Portrait: a tool from Surgeon General. CDC 84. Zeliadt, S. B. et al. Why do men choose one treatment
https://phgkb.cdc.gov/FHH/html/index.html (2022). over another?: a review of patient decision making for
74. Mark, J. R., McDougall, C. & Giri, V. N. Genetic testing localized prostate cancer. Cancer 106, 1865–1874 Related links
guidelines and education of health care providers (2006). My Family Health Portrait: https://www.cdc.gov/genomics/
involved in prostate cancer care. Urol. Clin. North. Am. 85. Taylor, L. G., Canfield, S. E. & Du, X. L. Review of major famhistory/knowing_not_enough.htm
48, 311–322 (2021). adverse effects of androgen-​deprivation therapy in PROGRess Registry: www.progressregistry.com
75. Stoll, K., Kubendran, S. & Cohen, S. A. The past, men with prostate cancer. Cancer 115, 2388–2399
present and future of service delivery in genetic (2009). © Springer Nature Limited 2022

Nature Reviews | Urology

0123456789();:

You might also like