Skip to main content
Erschienen in: memo - Magazine of European Medical Oncology 1/2024

Open Access 03.11.2023 | short review

Genetic testing and management of prostate cancer patients with pathogenic germline variants

verfasst von: Katharina Reiter, Melanie R. Hassler

Erschienen in: memo - Magazine of European Medical Oncology | Ausgabe 1/2024

Summary

Prostate cancer (PCa) is an androgen-receptor signaling-dependent disease with a subset of patients harboring pathogenic germline variants (PGVs) in genes essential for DNA repair. In the last decade, several guidelines and recommendations have been developed to define which PCa patients should receive genetic testing to identify individuals at higher risk due to inherited alterations and to facilitate personalized treatment strategies. Notably, the presence of specific germline alterations in carriers undergoing PCa screening has implications for screening strategies, and PGV carriers with advanced disease are eligible to receive targeted therapies such as poly-ADP-ribose polymerase inhibitors (PARPi) or immune checkpoint inhibitors (CKI) depending on the alterations encountered. Although less information is available on carriers with localized disease, several trials are addressing this specific patient population and will help to collect data and improve clinical management of PCa patients with PGVs.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Take-home message
  • Approximately 5% of localized and up to 12% of metastatic prostate cancer (PCa) patients carry pathogenic germline variants, with BRCA2 being the most prevalent alteration.
  • Different guidelines recommend germline sequencing in patients with metastatic disease, a family history of PCa or specific high-risk features in order to optimize treatment, assess personal cancer risk and prognosis and further guide family counselling for cancer predisposition syndromes.
  • In localized disease, underlying pathogenic germline variants should trigger shared decision-making when deciding on active surveillance and (intensified) curative treatment options.
  • In metastatic PCa patients with BRCA1/2 or ATM alterations, targeted treatment with poly-ADP-ribose polymerase inhibitors (PARP) inhibitors offers promising options. Additionally, a subset of metastatic PCa patients with mismatch repair (MMR) alterations could benefit from immune checkpoint inhibition.

Germline alterations in PCa

Prostate cancer (PCa) is the second most common cancer in men and significantly impacts global health as a leading cause of cancer-related deaths [1]. Recent studies have shown that a subset of PCa patients harbor pathogenic germline variants (PGVs) affecting DNA damage repair mechanisms, which play a distinct role in disease development and differentiation [2]. Detection and understanding the implications of these PGVs is crucial for improved clinical management with potentially beneficial outcomes for these patients.
In PCa, approximately 4–5% of patients with localized PCa and 12% of patients with metastasized PCa carry a PGV in genes essential for homologous DNA repair (DDR) or mismatch repair (MMR) [24]. Genes involved in DDR comprise BRCA1, BRCA2, ATM, CHEK2, and others. Among these, BRCA2 is the most frequently found PGV, with a prevalence of 44% among all PCa mutation carriers [2, 4]. BRCA1/2 germline mutations are causing hereditary breast and ovarian cancer (HBOC) syndrome, which increases the lifetime risk of developing breast and ovarian cancer to 40–70% [5]. For male BRCA2 carriers, the risk of developing PCa is approximately 15–30% compared to 10–15% in the general population, whereas the risk with a BRCA1 or other PGV may only be slightly elevated [6, 7].
Genes involved in MMR comprise MSH2, MSH6, MLH1 and PMS2 or EPCAM—an epigenetic silencer of MSH2. These genes predispose affected individuals to a high lifetime risk of developing colon, endometrial, and other cancers (Lynch syndrome [LS]) [8, 9]. Although only a subset of PCa patients harbor germline alterations in these genes (3–4% of all PGV carriers), their presence has a significant impact on the molecular alterations present in PCa tumors, as deficiencies in the DNA mismatch repair system can lead to a phenomenon called microsatellite instability (MSI) [2, 8]. Tumors with high MSI are characterized by high mutation rates and may have increased responses to immune checkpoint inhibitor (CKI) therapy, which otherwise has limited effects in unselected PCa patients [10, 11].

Indications for genetic testing

Currently, there are several guidelines regarding which PCa patients should receive genetic testing for hereditary cancer syndromes (Table 1). The testing criteria encompass personal cancer history, cancer features and pathology, family history, and precision therapy indications [1215]. All guidelines recommend germline testing of men with metastatic PCa, as the prevalence of PGVs in this population is over 10% [2]. In addition, guidelines state that a family history of PCa or the presence of two or more cancers within the HBOC or LS spectrum in relatives on the same family side should trigger genetic testing. Furthermore, some guidelines state that patients with high-risk (T3a or ISUP 4 or PSA > 20 ng/ml) or very-high risk (T3b-T4 or primary Gleason pattern 5 or > 4 cores with ISUP 4 or 5) localized PCa, presence of intraductal, ductal or cribriform histology, an Ashkenazi Jewish ancestry, or a diagnosis of PCa < 60 years can be offered genetic testing [12, 15, 16]. There is no consensus set of genes that must be included in PCa germline testing assays, but in general, multiple genes that may be relevant to optimize targeted therapy approaches or account for the patient’s cancer and family history are analyzed. Typical panels always include BRCA1/2 but vary in coverage regarding other homologous DNA repair or DNA mismatch genes, which should be considered when ordering these tests [12, 13, 15]. It is important to note that patients undergoing germline testing need pretest and posttest genetic counseling on potential test results and their impact on further management, and conducting genetic tests and counseling is often regulated by national laws [17].
Table 1
Indications for genetic testing in prostate cancer (PCa). Summary of current guidelines and consensus statements
Philadelphia prostate cancer consensus conference 2019 [15]
Recommended:
Metastatic PCa
Men with first-degree relatives or two or more male relatives with either:
PCa diagnosis at age < 60 years
PCa-related death
Metastatic PCa
Consider:
Advanced stage of disease (≥ T3a)
Intraductal/ductal pathology
PCa with Gleason pattern ≥ 8
≥ 2 cancers in HBOC or Lynch syndrome in any relatives within one family trait
Ashkenazi Jewish ancestry
NCCN prostate cancer guidelines [12]
Recommended:
Metastatic PCa
Regional, node-positive PCa
High-risk or very high-risk localized PCa
Personal history of breast cancer
≥ 1 first-, second-, or third-degree relative with breast, endometrial or colorectal at age ≤ 50 years or male breast, ovarian, exocrine pancreatic cancer or advanced PCa at any age
≥ 1 first-degree relative with PCA diagnosis at age ≤ 60 years
≥ 2 first-, second- or third-degree relatives with breast or PCa at any age
≥ 3 first- or second-degree relatives with Lynch syndrome-related cancer
Known FH or familial PGVs
Ashkenazi Jewish ancestry
Consider:
Intermediate-risk PCa with intraductal/cribriform pathology
Personal history of exocrine pancreatic, colorectal, gastric, melanoma, upper tract urothelial, glioblastoma, biliary tract or small intestinal cancer
EAU guidelines on prostate cancer [14]
Weak strength rating:
Metastatic PCa
High-risk PCa with FH of PCa at age < 60
Any PCa with multiple family members < 60 years or FH with PCa-related death
FH of high-risk PGVs
FH of multiple cancers within one family trait
AUA guidelines on localized and advanced prostate cancer [16]
Recommended:
Metastatic PCa
Consider:
Adverse tumor characteristics
Strong personal history of associated cancers
Strong FH of PCa
Strong FH of associated cancers
Known PGVs
Ashkenazi Jewish ancestry
AUA American Urological Association, EAU European Association of Urology, FH family history, HBOC hereditary breast and ovarian cancer, NCCN National Comprehensive Cancer Network®, PGV pathogenic variant

Clinical management of PCa patients with germline mutations

PCa screening

PCa screening for the early detection of PCa in the population of PGV carriers is an intense subject of debate due to potentially increased rates of unnecessary biopsies and their association with overdiagnosis and overtreatment [18]. Several prospective studies have been evaluating PCa screening among PGV carriers [6, 1921]. The IMPACT trial is evaluating targeted PCa screening in men with and without germline BRCA1/2 and MMR alterations [19]. In the study, men aged 40–69 underwent prostate-specific antigen (PSA) screening for 3 years, and if their PSA was higher than 3.0 ng/ml, they were offered prostate biopsy. The study reported interim results in 2019, and after 3 years of screening, BRCA2 mutation carriers had a higher incidence of PCa, were younger at diagnosis, and had more clinically significant tumors [19]. Based on this, PSA screening for men with BRCA2 mutations should start at the age of 40 or 10 years before the youngest relative was diagnosed with PCa [12, 13]. For BRCA1 carriers, no significant differences were detected compared to BRCA1 noncarriers. However, guidelines recommend that men with BRCA1 alterations should consider a screening approach similar to BRCA2 carriers [12, 13]. For carriers of MSH2 and MSH6 PGVs, a higher incidence of PCa compared with age-matched noncarriers has been reported after the first PSA screening round, indicating that PSA screening in MSH2/MSH6 carriers should start at a similar age as for BRCA2 carriers [20].
In contrast to the IMPACT study, the current practice uses imaging-based PCa examinations before definitive biopsy. This approach is tested for BRCA1/2 carriers using a combination of multiparametric magnetic resonance imaging (mpMRI)-based and PSA-based screening [21]. So far, interim results have shown that initial mpMRI-based screening may be beneficial, especially for BRCA carriers younger than 55 years, compared to PSA screening alone [21]. No data have demonstrated a benefit from prophylactic prostatectomy in PGV carriers, but clinical trials are in discussion [22]. Thus, for optimized screening approaches in this population, new biomarkers and risk calculators are urgently needed to prevent unnecessary biopsies and improve the detection of clinically significant PCa.

Active surveillance

Active surveillance (AS) intends to spare PCa patients from aggressive interventions such as surgery or radiation therapy and is indicated when the disease is low-risk after biopsy and clinical staging. However, patients with PGVs and low-risk PCa might carry a higher risk of upstaging than noncarrier patients. Indeed, a study evaluating BRCA1/2 and ATM germline mutation carriers with PCa under AS reported higher tumor grade reclassification rates than in noncarriers (hazard ratio [HR] 2.74, 95% confidence interval [CI] 1.26–5.96; p = 0.01) [23]. However, reclassification rates are similar to rates described in other AS cohorts [24]. Another study in 15 patients with low-risk PCa and DDR mutations under AS for a median of 28 months reported a PCa reclassification rate of 20%, similar to that in the general AS population [25]. Based on this limited evidence, AS is, in principle, feasible among PGV carriers and could reduce overtreatment, but given the significant reclassification rates, shared decision-making with the option of local treatment for patients with BRCA2 mutations is recommended [18].

Localized prostate cancer

After local therapy (radical prostatectomy or external-beam radiotherapy), BRCA1/2 PGV carriers have been reported to have a higher risk of metastasis and mortality with a cancer-specific survival of 61% versus 85% in noncarriers (p < 0.001) at 10 years [26]. The difference in this retrospective cohort was especially noted for patients after radiation therapy, as surgery reduced PCa-specific mortality by 48%. Patients in the radiotherapy cohort had more aggressive disease than those in the surgery cohort, which limits the direct comparison of the two groups. Among the 2019 patients enrolled in PCa screening trials, the rates of high-risk PCa, lymph node involvement (15% vs. 5%), and metastasis (18% vs. 9%) were higher in BRCA1/2 carriers than in noncarriers [4]. In addition, lower cancer-specific survival was observed in carriers than in noncarriers after standard-of-care therapy (8.6 years versus 15.7 years), and median overall survival was also decreased in carriers compared to noncarriers (8.1 years vs. 12.9 years). In a study on neoadjuvant androgen deprivation therapy followed by radical prostatectomy, a similar pathological response and no difference in the 3‑year biochemical recurrence-free survival rate was reported between DDR germline mutation carriers and noncarriers [27]. Of note, BRCA2 carriers represented only one-third of the DDR cohort. Whether a specific therapeutic approach, therapy intensification, or adjuvant therapy offers therapeutic advantages for high-risk localized PCa patients with PGVs is currently unclear, but ongoing clinical trials are addressing this pending question (NCT03432897, NCT04030559, NCT04037254).

Metastatic prostate cancer

For patients with metastatic castration-resistant (mCRPC) disease and BRCA1/2, other DDR alterations or PGVs in DNA mismatch genes, targeted therapies are available in the context of precision medicine approaches.
Several trials have shown a benefit in survival when treating DDR carriers with PARP inhibitors (PARPi), either as monotherapy or in combination with androgen receptor inhibitors (ARIs) [2832]. The PARPi olaparib significantly prolonged survival compared to ARIs in mCRPC patients with somatic BRCA1/2 or ATM mutations after previous treatment with a different ARI [28]. Another PARPi, rucaparib, has been tested as monotherapy in a phase III study in patients with somatic BRCA1/2 or ATM mutations evaluating rucaparib or physician’s choice of abiraterone, enzalutamide or docetaxel after progression on ARIs. Recent results show that rucaparib significantly prolongs progression-free survival (PFS) compared to ARIs or docetaxel in this setting [30]. The PROpel trial reported that concurrent olaparib plus abiraterone versus abiraterone alone in first-line mCRPC treatment leads to improved PFS regardless of mutation status. However, subgroup analysis showed that the effect was more pronounced in patients with somatic DDR mutations [29]. Improvement of PFS was also observed in mCRPC patients receiving talazoparib plus enzalutamide versus enzalutamide in patients with and without alterations, with a noteworthy benefit especially among patients with somatic BRCA2 alterations [31]. In contrast to these unselected populations, a trial comparing niraparib and abiraterone versus abiraterone alone in first-line mCRPC observed a significant improvement in PFS solely in patients with somatic DDR alterations [32]. These trials demonstrate a significant benefit from PARPi therapy in mCRPC patients with PGVs or somatic alterations in BRCA1/2. Several other trials with PARPi in earlier-stage PCa are ongoing, i.e., the NADIR trial in high-risk localized PCa, the AMPLITUDE trial in metastatic castration-sensitive PCa, and a trial evaluating olaparib monotherapy in biochemical recurrence after radical prostatectomy in patients with and without BRCA1/2 alterations, which may lead to further indications for PARPi in PCa [3335].
A subset of mCRPC patients with MMR alterations may benefit from CKI therapies. In a basket trial by Le et al., CKI therapy with pembrolizumab was tested in MMR-deficient cancers across 12 advanced cancer types, including PCa [36]. In this heavily pretreated and heterogenous cohort, CKI treatment showed an overall response rate of 53% and a complete response rate of 21%, which led to US Food and Drug Administration (FDA) approval for pembrolizumab for all solid tumors with MMR deficiency or MSI high without satisfactory alternative treatment options [37]. A case series of PCa patients with tumor and germline sequencing has shown that approximately 3% of PCa patients have MMR deficient tumors and that 25% carry a germline mutation in MMR genes. The retrospective study further showed that although these alterations were found in a small subset of PCa patients, approximately 45% of PCa MSI-high tumors clinically benefited from pembrolizumab treatment [38].

Conclusion

Management of prostate cancer (PCa) patients with germline mutations is essential in PCa screening, active surveillance, and localized or metastatic PCa treatment (Table 2). Given the recent recommendations for genetic testing and subsequent identification of mutation carriers, data on the best management of PCa patients with pathogenic germline variants (PGVs) at different stages during disease development is still scarce. In particular, experience how to manage patients with less well-studied but still rather frequently encountered alterations such as CHEK2 or ATM is limited, and further studies are needed to offer optimal clinical management for these patients.
Table 2
Selected studies with impact on clinical management of prostate cancer patients with germline mutations
Study
Patients included
Genes tested
Type of testing
Study design
Impact on clinical management
Screening
IMPACT [19, 20]
3027
BRCA1/2 + additional 3 MMR genes
Germline
Prospective
PSA screening is indicated for BRCA2, MSH2 and MSH6 PGV carriers
Active surveillance
Carter et al. [23]
1211
BRCA 1/2, ATM + additional 51 DNA repair genes (n = 54)
Germline
Prospective
PGVs in BRCA1/2 and ATM are associated with aggressive PCa
Localized prostate cancer
Castro et al. [26]
1302
BRCA1/2 (n = 2)
Germline
Retrospective
Outcomes for conventional treatment of localized PCa was worse in BRCA 1/2 carriers compared to noncarriers
Metastatic castration-resistant prostate cancer
De Bono et al. PROfound: olaparib [28]
4425
BRCA 1/2, ATM + additional 12 other genes
Somatic
Randomized phase III
PFS was superior in patients with HRR alterations treated with olaparib compared to enzalutamide or abiraterone
Fizazi et al. TRITON3: rucaparib [30]
4855
BRCA 1/2, ATM
Germline + somatic
Randomized phase III
PFS was longer in PCa patients with underlying BRCA alterations treated with rucaparib compared to control medication
Clarke et al. PROPel: olaparib + abiraterone [29]
796
BRCA 1/2, ATM + additional 11 genes
Somatic
Randomized phase III
Combination of abiraterone and olaparib prolonged PFS irrespective of HRR alteration status
Chi et al. MAGNITUDE: niraparib + abiraterone [32]
742
BRCA 1/2, ATM + additional 6 genes
Somatic
Randomized phase III
Niraparib + abiraterone prolonged PFS in patients with underlying HRR alterations
Agarwal et al. TALAPRO-2: talazoparib + enzalutamide (ongoing) [31]
805
BRCA 1/2, ATM + additional 9 genes
Somatic
Randomized phase III
PFS improved in patients treated with talazoparib + enzalutamide irrespective of HRR gene alterations
Le et al. [36]
86
MLH1, MSH2, MSH6, PMS2
Germline + somatic
Nonrandomized phase II
MMR-deficient cancers responded to PD‑1 blockage irrespective of tumor type
HRR homologous recombination repair, MMR mismatch repair, PCa prostate cancer, PFS progression-free survival

Conflict of interest

K. Reiter and M.R. Hassler declare that they have no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Abo für kostenpflichtige Inhalte

Literatur
1.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.CrossRefPubMed Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.CrossRefPubMed
2.
Zurück zum Zitat Pritchard CC, Mateo J, Walsh MF, De Sarkar N, Abida W, Beltran H, et al. Inherited DNA-repair gene mutations in men with metastatic prostate cancer. N Engl J Med. 2016;375(5):443–53.CrossRefPubMedPubMedCentral Pritchard CC, Mateo J, Walsh MF, De Sarkar N, Abida W, Beltran H, et al. Inherited DNA-repair gene mutations in men with metastatic prostate cancer. N Engl J Med. 2016;375(5):443–53.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Oh M, Alkhushaym N, Fallatah S, Althagafi A, Aljadeed R, Alsowaida Y, et al. The association of BRCA1 and BRCA2 mutations with prostate cancer risk, frequency, and mortality: A meta-analysis. Prostate. 2019;79(8):880–95.CrossRefPubMed Oh M, Alkhushaym N, Fallatah S, Althagafi A, Aljadeed R, Alsowaida Y, et al. The association of BRCA1 and BRCA2 mutations with prostate cancer risk, frequency, and mortality: A meta-analysis. Prostate. 2019;79(8):880–95.CrossRefPubMed
4.
Zurück zum Zitat Castro E, Goh C, Olmos D, Saunders E, Leongamornlert D, Tymrakiewicz M, et al. Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer. J Clin Oncol. 2013;31(14):1748–57.CrossRefPubMedPubMedCentral Castro E, Goh C, Olmos D, Saunders E, Leongamornlert D, Tymrakiewicz M, et al. Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer. J Clin Oncol. 2013;31(14):1748–57.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Antoniou A, Pharoah PD, Narod S, Risch HA, Eyfjord JE, Hopper JL, et al. Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case Series unselected for family history: a combined analysis of 22 studies. Am J Hum Genet. 2003;72(5):1117–30.CrossRefPubMedPubMedCentral Antoniou A, Pharoah PD, Narod S, Risch HA, Eyfjord JE, Hopper JL, et al. Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case Series unselected for family history: a combined analysis of 22 studies. Am J Hum Genet. 2003;72(5):1117–30.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Nyberg T, Frost D, Barrowdale D, Evans DG, Bancroft E, Adlard J, et al. Prostate Cancer Risks for Male BRCA1 and BRCA2 Mutation Carriers: A Prospective Cohort Study. Eur Urol. 2020;77(1):24–35.CrossRefPubMedPubMedCentral Nyberg T, Frost D, Barrowdale D, Evans DG, Bancroft E, Adlard J, et al. Prostate Cancer Risks for Male BRCA1 and BRCA2 Mutation Carriers: A Prospective Cohort Study. Eur Urol. 2020;77(1):24–35.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Nyberg T, Tischkowitz M, Antoniou AC. BRCA1 and BRCA2 pathogenic variants and prostate cancer risk: systematic review and meta-analysis. Br J Cancer. 2022;126(7):1067–81.CrossRefPubMed Nyberg T, Tischkowitz M, Antoniou AC. BRCA1 and BRCA2 pathogenic variants and prostate cancer risk: systematic review and meta-analysis. Br J Cancer. 2022;126(7):1067–81.CrossRefPubMed
8.
Zurück zum Zitat Hampel H, Frankel WL, Martin E, Arnold M, Khanduja K, Kuebler P, et al. Screening for the Lynch syndrome (hereditary nonpolyposis colorectal cancer). N Engl J Med. 2005;352(18):1851–60.CrossRefPubMed Hampel H, Frankel WL, Martin E, Arnold M, Khanduja K, Kuebler P, et al. Screening for the Lynch syndrome (hereditary nonpolyposis colorectal cancer). N Engl J Med. 2005;352(18):1851–60.CrossRefPubMed
9.
Zurück zum Zitat Hampel H, Frankel W, Panescu J, Lockman J, Sotamaa K, Fix D, et al. Screening for Lynch syndrome (hereditary nonpolyposis colorectal cancer) among endometrial cancer patients. Cancer Res. 2006;66(15):7810–7.CrossRefPubMed Hampel H, Frankel W, Panescu J, Lockman J, Sotamaa K, Fix D, et al. Screening for Lynch syndrome (hereditary nonpolyposis colorectal cancer) among endometrial cancer patients. Cancer Res. 2006;66(15):7810–7.CrossRefPubMed
10.
Zurück zum Zitat Antonarakis ES, Piulats JM, Gross-Goupil M, Goh J, Ojamaa K, Hoimes CJ, et al. Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: multicohort, open-label phase II KEYNOTE-199 study. J Clin Oncol. 2020;38(5):395–405.CrossRefPubMed Antonarakis ES, Piulats JM, Gross-Goupil M, Goh J, Ojamaa K, Hoimes CJ, et al. Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: multicohort, open-label phase II KEYNOTE-199 study. J Clin Oncol. 2020;38(5):395–405.CrossRefPubMed
11.
Zurück zum Zitat Petrelli F, Ghidini M, Ghidini A, Tomasello G. Outcomes Following Immune Checkpoint Inhibitor Treatment of Patients With Microsatellite Instability-High Cancers: A Systematic Review and Meta-analysis. JAMA Oncol. 2020;6(7:1068–71.CrossRef Petrelli F, Ghidini M, Ghidini A, Tomasello G. Outcomes Following Immune Checkpoint Inhibitor Treatment of Patients With Microsatellite Instability-High Cancers: A Systematic Review and Meta-analysis. JAMA Oncol. 2020;6(7:1068–71.CrossRef
12.
Zurück zum Zitat NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines ®) Prostate Cancer (version 1.2023). NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines ®) Prostate Cancer (version 1.2023).
13.
Zurück zum Zitat NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines ®) Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic (version 1.2023). NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines ®) Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic (version 1.2023).
14.
Zurück zum Zitat Mottet N, van den Bergh RCN, Briers E, Van den Broeck T, Cumberbatch MG, De Santis M, et al. EAU-EANM-ESTRO-ESUR-SIOG guidelines on prostate cancer – 2020 update. Part 1: screening, diagnosis, and local treatment with curative intent. Eur Urol. 2021;79(2):243–62.CrossRefPubMed Mottet N, van den Bergh RCN, Briers E, Van den Broeck T, Cumberbatch MG, De Santis M, et al. EAU-EANM-ESTRO-ESUR-SIOG guidelines on prostate cancer – 2020 update. Part 1: screening, diagnosis, and local treatment with curative intent. Eur Urol. 2021;79(2):243–62.CrossRefPubMed
15.
Zurück zum Zitat Giri VN, Knudsen KE, Kelly WK, Cheng HH, Cooney KA, Cookson MS, et al. Implementation of Germline Testing for Prostate Cancer: Philadelphia Prostate Cancer Consensus Conference 2019. J Clin Oncol. 2020;38(24):2798–811.CrossRefPubMedPubMedCentral Giri VN, Knudsen KE, Kelly WK, Cheng HH, Cooney KA, Cookson MS, et al. Implementation of Germline Testing for Prostate Cancer: Philadelphia Prostate Cancer Consensus Conference 2019. J Clin Oncol. 2020;38(24):2798–811.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Eastham JA, Auffenberg GB, Barocas DA, Chou R, Crispino T, Davis JW, et al. Clinically Localized Prostate Cancer: AUA/ASTRO Guideline, Part I: Introduction, Risk Assessment, Staging, and Risk-Based Management. J Urol. 2022;208(1):10–8.CrossRefPubMed Eastham JA, Auffenberg GB, Barocas DA, Chou R, Crispino T, Davis JW, et al. Clinically Localized Prostate Cancer: AUA/ASTRO Guideline, Part I: Introduction, Risk Assessment, Staging, and Risk-Based Management. J Urol. 2022;208(1):10–8.CrossRefPubMed
17.
Zurück zum Zitat Abacan M, Alsubaie L, Barlow-Stewart K, Caanen B, Cordier C, Courtney E, et al. The global state of the genetic counseling profession. Eur J Hum Genet. 2019;27(2):183–97.CrossRefPubMed Abacan M, Alsubaie L, Barlow-Stewart K, Caanen B, Cordier C, Courtney E, et al. The global state of the genetic counseling profession. Eur J Hum Genet. 2019;27(2):183–97.CrossRefPubMed
18.
Zurück zum Zitat Rajwa P, Quhal F, Pradere B, Gandaglia G, Ploussard G, Leapman MS, et al. Prostate cancer risk, screening and management in patients with germline BRCA1/2 mutations. Nat Rev Urol. 2023;20(4):205–16.CrossRefPubMed Rajwa P, Quhal F, Pradere B, Gandaglia G, Ploussard G, Leapman MS, et al. Prostate cancer risk, screening and management in patients with germline BRCA1/2 mutations. Nat Rev Urol. 2023;20(4):205–16.CrossRefPubMed
19.
Zurück zum Zitat Page EC, Bancroft EK, Brook MN, Assel M, Al Battat HM, Thomas S, et al. Interim Results from the IMPACT Study: Evidence for Prostate-specific Antigen Screening in BRCA2 Mutation Carriers. Eur Urol. 2019;76(6):831–42.CrossRefPubMedPubMedCentral Page EC, Bancroft EK, Brook MN, Assel M, Al Battat HM, Thomas S, et al. Interim Results from the IMPACT Study: Evidence for Prostate-specific Antigen Screening in BRCA2 Mutation Carriers. Eur Urol. 2019;76(6):831–42.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Bancroft EK, Page EC, Brook MN, Thomas S, Taylor N, Pope J, et al. A prospective prostate cancer screening programme for men with pathogenic variants in mismatch repair genes (IMPACT): initial results from an international prospective study. Lancet Oncol. 2021;22(11):1618–31.CrossRefPubMedPubMedCentral Bancroft EK, Page EC, Brook MN, Thomas S, Taylor N, Pope J, et al. A prospective prostate cancer screening programme for men with pathogenic variants in mismatch repair genes (IMPACT): initial results from an international prospective study. Lancet Oncol. 2021;22(11):1618–31.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Segal N, Ber Y, Benjaminov O, Tamir S, Yakimov M, Kedar I, et al. Imaging-based prostate cancer screening among BRCA mutation carriers-results from the first round of screening. Ann Oncol. 2020;31(11):1545–52.CrossRefPubMed Segal N, Ber Y, Benjaminov O, Tamir S, Yakimov M, Kedar I, et al. Imaging-based prostate cancer screening among BRCA mutation carriers-results from the first round of screening. Ann Oncol. 2020;31(11):1545–52.CrossRefPubMed
22.
Zurück zum Zitat Clark R, McAlpine K, Fleshner N. A Clinical Trial of Prophylactic Prostatectomy for BRCA2 Mutation Carriers. Is Now the Time? Eur Urol Focus. 2021;7(3):506–7.CrossRefPubMed Clark R, McAlpine K, Fleshner N. A Clinical Trial of Prophylactic Prostatectomy for BRCA2 Mutation Carriers. Is Now the Time? Eur Urol Focus. 2021;7(3):506–7.CrossRefPubMed
23.
Zurück zum Zitat Carter HB, Helfand B, Mamawala M, Wu Y, Landis P, Yu H, et al. Germline Mutations in ATM and BRCA1/2 Are Associated with Grade Reclassification in Men on Active Surveillance for Prostate Cancer. Eur Urol. 2019;75(5):743–9.CrossRefPubMed Carter HB, Helfand B, Mamawala M, Wu Y, Landis P, Yu H, et al. Germline Mutations in ATM and BRCA1/2 Are Associated with Grade Reclassification in Men on Active Surveillance for Prostate Cancer. Eur Urol. 2019;75(5):743–9.CrossRefPubMed
24.
Zurück zum Zitat Rajwa P, Pradere B, Quhal F, Mori K, Laukhtina E, Huebner NA, et al. Reliability of Serial Prostate Magnetic Resonance Imaging to Detect Prostate Cancer Progression During Active Surveillance: A Systematic Review and Meta-analysis. Eur Urol. 2021;80(5):549–63.CrossRefPubMed Rajwa P, Pradere B, Quhal F, Mori K, Laukhtina E, Huebner NA, et al. Reliability of Serial Prostate Magnetic Resonance Imaging to Detect Prostate Cancer Progression During Active Surveillance: A Systematic Review and Meta-analysis. Eur Urol. 2021;80(5):549–63.CrossRefPubMed
25.
Zurück zum Zitat Halstuch D, Ber Y, Kedar D, Golan S, Baniel J, Margel D. Short-Term Outcomes of Active Surveillance for Low Risk Prostate Cancer among Men with Germline DNA Repair Gene Mutations. J Urol. 2020;204(4):707–13.CrossRefPubMed Halstuch D, Ber Y, Kedar D, Golan S, Baniel J, Margel D. Short-Term Outcomes of Active Surveillance for Low Risk Prostate Cancer among Men with Germline DNA Repair Gene Mutations. J Urol. 2020;204(4):707–13.CrossRefPubMed
26.
Zurück zum Zitat Castro E, Goh C, Leongamornlert D, Saunders E, Tymrakiewicz M, Dadaev T, et al. Effect of BRCA Mutations on Metastatic Relapse and Cause-specific Survival After Radical Treatment for Localised Prostate Cancer. Eur Urol. 2015;68(2):186–93.CrossRefPubMed Castro E, Goh C, Leongamornlert D, Saunders E, Tymrakiewicz M, Dadaev T, et al. Effect of BRCA Mutations on Metastatic Relapse and Cause-specific Survival After Radical Treatment for Localised Prostate Cancer. Eur Urol. 2015;68(2):186–93.CrossRefPubMed
27.
Zurück zum Zitat Berchuck JE, Zhang Z, Silver R, Kwak L, Xie W, Lee GM, et al. Impact of Pathogenic Germline DNA Damage Repair alterations on Response to Intense Neoadjuvant Androgen Deprivation Therapy in High-risk Localized Prostate Cancer. Eur Urol. 2021;80(3):295–303.CrossRefPubMed Berchuck JE, Zhang Z, Silver R, Kwak L, Xie W, Lee GM, et al. Impact of Pathogenic Germline DNA Damage Repair alterations on Response to Intense Neoadjuvant Androgen Deprivation Therapy in High-risk Localized Prostate Cancer. Eur Urol. 2021;80(3):295–303.CrossRefPubMed
28.
Zurück zum Zitat de Bono J, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med. 2020;382(22):2091–102.CrossRefPubMed de Bono J, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med. 2020;382(22):2091–102.CrossRefPubMed
29.
Zurück zum Zitat Clarke N, Wiechno P, Alekseev B, Sala N, Jones R, Kocak I, et al. Olaparib combined with abiraterone in patients with metastatic castration-resistant prostate cancer: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 2018;19(7):975–86.CrossRefPubMed Clarke N, Wiechno P, Alekseev B, Sala N, Jones R, Kocak I, et al. Olaparib combined with abiraterone in patients with metastatic castration-resistant prostate cancer: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 2018;19(7):975–86.CrossRefPubMed
30.
Zurück zum Zitat Fizazi K, Piulats JM, Reaume MN, Ostler P, McDermott R, Gingerich JR, et al. Rucaparib or physician’s choice in metastatic prostate cancer. N Engl J Med. 2023;388(8):719–32.CrossRefPubMedPubMedCentral Fizazi K, Piulats JM, Reaume MN, Ostler P, McDermott R, Gingerich JR, et al. Rucaparib or physician’s choice in metastatic prostate cancer. N Engl J Med. 2023;388(8):719–32.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Chi KN, Sandhu S, Smith MR, Attard G, Saad M, Olmos D, et al. Niraparib plus abiraterone acetate with prednisone in patients with metastatic castration-resistant prostate cancer and homologous recombination repair gene alterations: second interim analysis of the randomized phase III MAGNITUDE trial. Ann Oncol. 2023; https://doi.org/10.1016/j.annonc.2023.06.009.CrossRefPubMed Chi KN, Sandhu S, Smith MR, Attard G, Saad M, Olmos D, et al. Niraparib plus abiraterone acetate with prednisone in patients with metastatic castration-resistant prostate cancer and homologous recombination repair gene alterations: second interim analysis of the randomized phase III MAGNITUDE trial. Ann Oncol. 2023; https://​doi.​org/​10.​1016/​j.​annonc.​2023.​06.​009.CrossRefPubMed
33.
Zurück zum Zitat Zumsteg Z, Karrison T, Michaelson MD, Tran P, Kudchadker R, Feng F. 689TiP NRG Oncology’s GU007 (NADIR): A randomized phase II trial of niraparib with standard combination androgen deprivation therapy (ADT) and radiotherapy (RT) in high-risk prostate cancer (PC) (with initial phase I). Annals of Oncology, Volume 31, Supplement 4, S546, September 2020. Zumsteg Z, Karrison T, Michaelson MD, Tran P, Kudchadker R, Feng F. 689TiP NRG Oncology’s GU007 (NADIR): A randomized phase II trial of niraparib with standard combination androgen deprivation therapy (ADT) and radiotherapy (RT) in high-risk prostate cancer (PC) (with initial phase I). Annals of Oncology, Volume 31, Supplement 4, S546, September 2020.
34.
Zurück zum Zitat Rathkopf DE, Chi KN, Olmos D, Cheng HH, Agarwal N, Graff JN, et al. AMPLITUDE: A study of niraparib in combination with abiraterone acetate plus prednisone (AAP) versus AAP for the treatment of patients with deleterious germline or somatic homologous recombination repair (HRR) gene-altered metastatic castration-sensitive prostate cancer (mCSPC). Journal of Clinical Oncology 39, no. 6_suppl. Rathkopf DE, Chi KN, Olmos D, Cheng HH, Agarwal N, Graff JN, et al. AMPLITUDE: A study of niraparib in combination with abiraterone acetate plus prednisone (AAP) versus AAP for the treatment of patients with deleterious germline or somatic homologous recombination repair (HRR) gene-altered metastatic castration-sensitive prostate cancer (mCSPC). Journal of Clinical Oncology 39, no. 6_suppl.
35.
Zurück zum Zitat Antonarakis ES, Wang H, Teply BA, Kelly WK, Willms J, Sullivan R, et al. Interim results from a phase 2 study of olaparib (without ADT) in men with biochemically-recurrent prostate cancer after prostatectomy, with integrated biomarker analysis.: Journal of Clinical Oncology 37, no. 15_suppl (May 20, 2019) 5045–5045. Antonarakis ES, Wang H, Teply BA, Kelly WK, Willms J, Sullivan R, et al. Interim results from a phase 2 study of olaparib (without ADT) in men with biochemically-recurrent prostate cancer after prostatectomy, with integrated biomarker analysis.: Journal of Clinical Oncology 37, no. 15_suppl (May 20, 2019) 5045–5045.
36.
Zurück zum Zitat Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD‑1 blockade. Science. 2017;357(6349):409–13.ADSCrossRefPubMedPubMedCentral Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD‑1 blockade. Science. 2017;357(6349):409–13.ADSCrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Marcus L, Lemery SJ, Keegan P, Approval Summary PRFDA. Pembrolizumab for the Treatment of Microsatellite Instability-High Solid Tumors. Clin. Cancer Res. 2019;25(13):3753–8. Marcus L, Lemery SJ, Keegan P, Approval Summary PRFDA. Pembrolizumab for the Treatment of Microsatellite Instability-High Solid Tumors. Clin. Cancer Res. 2019;25(13):3753–8.
38.
Zurück zum Zitat Abida W, Cheng ML, Armenia J, Middha S, Autio KA, Vargas HA, et al. Analysis of the prevalence of microsatellite instability in prostate cancer and response to immune checkpoint blockade. JAMA Oncol. 2019;5(4):471–8.CrossRefPubMed Abida W, Cheng ML, Armenia J, Middha S, Autio KA, Vargas HA, et al. Analysis of the prevalence of microsatellite instability in prostate cancer and response to immune checkpoint blockade. JAMA Oncol. 2019;5(4):471–8.CrossRefPubMed
Metadaten
Titel
Genetic testing and management of prostate cancer patients with pathogenic germline variants
verfasst von
Katharina Reiter
Melanie R. Hassler
Publikationsdatum
03.11.2023
Verlag
Springer Vienna
Erschienen in
memo - Magazine of European Medical Oncology / Ausgabe 1/2024
Print ISSN: 1865-5041
Elektronische ISSN: 1865-5076
DOI
https://doi.org/10.1007/s12254-023-00921-1

Weitere Artikel der Ausgabe 1/2024

memo - Magazine of European Medical Oncology 1/2024 Zur Ausgabe