1
|
Cruz Garcia Villa P, Izunza Laisequilla A, Puga Ortega E, Alaez Verson C. Prevalence of DNA-Repair Gene mutations in Mexican men with prostate cancer. Actas Urol Esp 2024; 48:588-595. [PMID: 38735436 DOI: 10.1016/j.acuroe.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/07/2024] [Indexed: 05/14/2024]
Abstract
INTRODUCTION AND OBJECTIVE Mexico reported 26,742 new cases of prostate cancer in 2020. Different risk factors have been identified in the pathogenesis of prostate cancer. Among them, genetic factors and alterations or mutations in specific genes have been described in different ethnic groups worldwide. The aim of our study is to report the prevalence of germline DNA-repair gene mutations in Mexican patients with prostate cancer. MATERIAL AND METHOD We performed germline genetic testing in 50 patients with localized prostate cancer and 50 patients with metastatic prostate cancer. Demographic, clinical, and histopathological data were collected. RESULTS Thirty-seven germline mutations were identified in 32 patients. The most commonly affected genes were ATM in 6%, followed by FANCA (5%), and ATR (4%). BRCA2 mutations were identified in 3%. The frequency of mutations was higher in the metastatic group. DISCUSSION AND CONCLUSION The results of our study show different mutations from those reported in different populations or regions. The use of PARP inhibitors is indicated in patients with germline mutations, specifically BRCA2, showing improvement in overall survival and progression free survival. To our knowledge, this is the first study reporting the prevalence of mutations in DNA-repair genes in Mexican patients with prostate cancer.
Collapse
Affiliation(s)
- P Cruz Garcia Villa
- Servicio de Urología, Hospital Regional Lic. Adolfo López Mateos, ISSSTE, Ciudad de México, Mexico.
| | - A Izunza Laisequilla
- Servicio de Urología, Hospital Regional Lic. Adolfo López Mateos, ISSSTE, Ciudad de México, Mexico
| | - E Puga Ortega
- Servicio de Urología, Hospital Regional Lic. Adolfo López Mateos, ISSSTE, Ciudad de México, Mexico
| | - C Alaez Verson
- Departamento de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica, Ciudad de México, Mexico
| |
Collapse
|
2
|
Foley GR, Marthick JR, Lucas SE, Raspin K, Banks A, Stanford JL, Ostrander EA, FitzGerald LM, Dickinson JL. Germline Sequencing of DNA Damage Repair Genes in Two Hereditary Prostate Cancer Cohorts Reveals New Disease Risk-Associated Gene Variants. Cancers (Basel) 2024; 16:2482. [PMID: 39001544 PMCID: PMC11240467 DOI: 10.3390/cancers16132482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Rare, inherited variants in DNA damage repair (DDR) genes have a recognised role in prostate cancer (PrCa) susceptibility. In addition, these genes are therapeutically targetable. While rare variants are informing clinical management in other common cancers, defining the rare disease-associated variants in PrCa has been challenging. Here, whole-genome and -exome sequencing data from two independent, high-risk Australian and North American familial PrCa datasets were interrogated for novel DDR risk variants. Rare DDR gene variants (predicted to be damaging and present in two or more family members) were identified and subsequently genotyped in 1963 individuals (700 familial and 459 sporadic PrCa cases, 482 unaffected relatives, and 322 screened controls), and association analyses accounting for relatedness (MQLS) undertaken. In the combined datasets, rare ERCC3 (rs145201970, p = 2.57 × 10-4) and BRIP1 (rs4988345, p = 0.025) variants were significantly associated with PrCa risk. A PARP2 (rs200603922, p = 0.028) variant in the Australian dataset and a MUTYH (rs36053993, p = 0.031) variant in the North American dataset were also associated with risk. Evaluation of clinicopathological characteristics provided no evidence for a younger age or higher-grade disease at diagnosis in variant carriers, which should be taken into consideration when determining genetic screening eligibility criteria for targeted, gene-based treatments in the future. This study adds valuable knowledge to our understanding of PrCa-associated DDR genes, which will underpin effective clinical screening and treatment strategies.
Collapse
Affiliation(s)
- Georgea R Foley
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - James R Marthick
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Sionne E Lucas
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Kelsie Raspin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Annette Banks
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Janet L Stanford
- Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., M4-B874, Seattle, WA 98109, USA
| | - Elaine A Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Liesel M FitzGerald
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Joanne L Dickinson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| |
Collapse
|
3
|
Piombino C, Pipitone S, Tonni E, Mastrodomenico L, Oltrecolli M, Tchawa C, Matranga R, Roccabruna S, D’Agostino E, Pirola M, Bacchelli F, Baldessari C, Baschieri MC, Dominici M, Sabbatini R, Vitale MG. Homologous Recombination Repair Deficiency in Metastatic Prostate Cancer: New Therapeutic Opportunities. Int J Mol Sci 2024; 25:4624. [PMID: 38731844 PMCID: PMC11083429 DOI: 10.3390/ijms25094624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/14/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
More than 20% of metastatic prostate cancer carries genomic defects involving DNA damage repair pathways, mainly in homologous recombination repair-related genes. The recent approval of olaparib has paved the way to precision medicine for the treatment of metastatic prostate cancer with PARP inhibitors in this subset of patients, especially in the case of BRCA1 or BRCA2 pathogenic/likely pathogenic variants. In face of this new therapeutic opportunity, many issues remain unsolved. This narrative review aims to describe the relationship between homologous recombination repair deficiency and prostate cancer, the techniques used to determine homologous recombination repair status in prostate cancer, the crosstalk between homologous recombination repair and the androgen receptor pathway, the current evidence on PARP inhibitors activity in metastatic prostate cancer also in homologous recombination repair-proficient tumors, as well as emerging mechanisms of resistance to PARP inhibitors. The possibility of combination therapies including a PARP inhibitor is an attractive option, and more robust data are awaited from ongoing phase II and phase III trials outlined in this manuscript.
Collapse
Affiliation(s)
- Claudia Piombino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Stefania Pipitone
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Elena Tonni
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Luciana Mastrodomenico
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Marco Oltrecolli
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Cyrielle Tchawa
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Rossana Matranga
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Sara Roccabruna
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Elisa D’Agostino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Marta Pirola
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Francesca Bacchelli
- Clinical Trials Office, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Cinzia Baldessari
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Maria Cristina Baschieri
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Massimo Dominici
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Roberto Sabbatini
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Maria Giuseppa Vitale
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| |
Collapse
|
4
|
Kalampokis N, Zabaftis C, Spinos T, Karavitakis M, Leotsakos I, Katafigiotis I, van der Poel H, Grivas N, Mitropoulos D. Review on the Role of BRCA Mutations in Genomic Screening and Risk Stratification of Prostate Cancer. Curr Oncol 2024; 31:1162-1169. [PMID: 38534919 PMCID: PMC10969585 DOI: 10.3390/curroncol31030086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: Somatic and germline alterations can be commonly found in prostate cancer (PCa) patients. The aim of our present study was to perform a comprehensive review of the current literature in order to examine the impact of BRCA mutations in the context of PCa as well as their significance as genetic biomarkers. (2) Methods: A narrative review of all the available literature was performed. Only "landmark" publications were included. (3) Results: Overall, the number of PCa patients who harbor a BRCA2 mutation range between 1.2% and 3.2%. However, BRCA2 and BRCA1 mutations are responsible for most cases of hereditary PCa, increasing the risk by 3-8.6 times and up to 4 times, respectively. These mutations are correlated with aggressive disease and poor prognosis. Gene testing should be offered to patients with metastatic PCa, those with 2-3 first-degree relatives with PCa, or those aged < 55 and with one close relative with breast (age ≤ 50 years) or invasive ovarian cancer. (4) Conclusions: The individualized assessment of BRCA mutations is an important tool for the risk stratification of PCa patients. It is also a population screening tool which can guide our risk assessment strategies and achieve better results for our patients and their families.
Collapse
Affiliation(s)
- Nikolaos Kalampokis
- Department of Urology, G. Hatzikosta General Hospital, 45001 Ioannina, Greece;
| | - Christos Zabaftis
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
| | - Theodoros Spinos
- Department of Urology, University of Patras Hospital, 26504 Patras, Greece;
| | - Markos Karavitakis
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
| | - Ioannis Leotsakos
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
| | - Ioannis Katafigiotis
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
| | - Henk van der Poel
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands;
| | - Nikolaos Grivas
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands;
| | - Dionysios Mitropoulos
- Department of Urology, Medical School, National & Kapodistrian University of Athens, 14122 Athens, Greece;
| |
Collapse
|
5
|
O'Malley DE, Raspin K, Melton PE, Burdon KP, Dickinson JL, FitzGerald LM. Acquired copy number variation in prostate tumours: a review of common somatic copy number alterations, how they are formed and their clinical utility. Br J Cancer 2024; 130:347-357. [PMID: 37945750 PMCID: PMC10844642 DOI: 10.1038/s41416-023-02485-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Prostate cancer is one of the most commonly diagnosed cancers in men and unfortunately, disease will progress in up to a third of patients despite primary treatment. Currently, there is a significant lack of prognostic tests that accurately predict disease course; however, the acquisition of somatic chromosomal variation in the form of DNA copy number variants may help understand disease progression. Notably, studies have found that a higher burden of somatic copy number alterations (SCNA) correlates with more aggressive disease, recurrence after surgery and metastasis. Here we will review the literature surrounding SCNA formation, including the roles of key tumour suppressors and oncogenes (PTEN, BRCA2, NKX3.1, ERG and AR), and their potential to inform diagnostic and prognostic clinical testing to improve predictive value. Ultimately, SCNAs, or inherited germline alterations that predispose to SCNAs, could have significant clinical utility in diagnostic and prognostic tests, in addition to guiding therapeutic selection.
Collapse
Affiliation(s)
- Dannielle E O'Malley
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia
| | - Kelsie Raspin
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia
| | - Phillip E Melton
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia
- School of Population and Global Health, The University of Western Australia, Crawley, WA, Australia
| | - Kathryn P Burdon
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia
| | - Joanne L Dickinson
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia
| | - Liesel M FitzGerald
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia.
| |
Collapse
|
6
|
Incorvaia L, Perez A, Marchetti C, Brando C, Gristina V, Cancelliere D, Pivetti A, Contino S, Di Giovanni E, Barraco N, Bono M, Giurintano A, Bazan Russo TD, Gottardo A, Cutaia S, Pedone E, Peri M, Corsini LR, Fanale D, Galvano A, Scambia G, Badalamenti G, Russo A, Bazan V. Theranostic biomarkers and PARP-inhibitors effectiveness in patients with non-BRCA associated homologous recombination deficient tumors: Still looking through a dirty glass window? Cancer Treat Rev 2023; 121:102650. [PMID: 37939446 DOI: 10.1016/j.ctrv.2023.102650] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/16/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023]
Abstract
Breast cancer susceptibility gene 1 (BRCA1) and breast cancer susceptibility gene 2 (BRCA2) deleterious variants were the first and, still today, the main biomarkers of poly(ADP)ribose polymerase (PARP)-inhibitors (PARPis) benefit. The recent, increased, numbers of individuals referred for counseling and multigene panel testing, and the remarkable expansion of approved PARPis, not restricted to BRCA1/BRCA2-Pathogenic Variants (PVs), produced a strong clinical need for non-BRCA biomarkers. Significant limitations of the current testing and assays exist. The different approaches that identify the causes of Homologous Recombination Deficiency (HRD), such as the germline and somatic Homologous Recombination Repair (HRR) gene PVs, the testing showing its consequences, such as the genomic scars, or the novel functional assays such as the RAD51 foci testing, are not interchangeable, and should not be considered as substitutes for each other in clinical practice for guiding use of PARPi in non-BRCA, HRD-associated tumors. Today, the deeper knowledge on the significant relationship among all proteins involved in the HRR, not limited to BRCA, expands the possibility of a successful non-BRCA, HRD-PARPi synthetic lethality and, at the same time, reinforces the need for enhanced definition of HRD biomarkers predicting the magnitude of PARPi benefit.
Collapse
Affiliation(s)
- Lorena Incorvaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Alessandro Perez
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Claudia Marchetti
- Department of Woman's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Chiara Brando
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Valerio Gristina
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Daniela Cancelliere
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Alessia Pivetti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Silvia Contino
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Emilia Di Giovanni
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Nadia Barraco
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Marco Bono
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Ambra Giurintano
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Andrea Gottardo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Sofia Cutaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Erika Pedone
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Marta Peri
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Lidia Rita Corsini
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Daniele Fanale
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Antonio Galvano
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Giovanni Scambia
- Department of Woman's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Giuseppe Badalamenti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy.
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
7
|
Rendon RA, Selvarajah S, Wyatt AW, Kolinsky M, Schrader KA, Fleshner NE, Kinnaird A, Merrimen J, Niazi T, Saad F, Shayegan B, Wood L, Chi KN. 2023 Canadian Urological Association guideline: Genetic testing in prostate cancer. Can Urol Assoc J 2023; 17:314-325. [PMID: 37851913 PMCID: PMC10581723 DOI: 10.5489/cuaj.8588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Affiliation(s)
| | - Shamini Selvarajah
- Department of Clinical Laboratory Genetics, UHN Laboratory Medicine Program, University of Toronto, Toronto, ON, Canada
| | - Alexander W. Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Michael Kolinsky
- Division of Medical Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | | | - Neil E. Fleshner
- Division of Urology, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Adam Kinnaird
- Divison of Urology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | | | - Tamim Niazi
- Division of Radiation Oncology, Department of Oncology, McGill University, Montreal, QC, Canada
| | - Fred Saad
- Division of Urology, Department of Surgery, Université de Montréal, Montreal, QC, Canada
| | - Bobby Shayegan
- Division of Urology, Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Lori Wood
- Division of Medical Oncology, Queen Elizabeth II Health Sciences Centre, Halifax, NS, Canada
| | | |
Collapse
|
8
|
Shore N, Gazi M, Pieczonka C, Heron S, Modh R, Cahn D, Belkoff LH, Berger A, Mazzarella B, Veys J, Idom C, Morris D, Jayram G, Engelman A, Bukkapatnam R, Dato P, Bevan-Thomas R, Cornell R, Wise DR, Hardwick MK, Hernandez RD, Rojahn S, Layman P, Hatchell KE, Heald B, Nussbaum RL, Nielsen SM, Esplin ED. Efficacy of National Comprehensive Cancer Network Guidelines in Identifying Pathogenic Germline Variants Among Unselected Patients with Prostate Cancer: The PROCLAIM Trial. Eur Urol Oncol 2023; 6:477-483. [PMID: 37574391 DOI: 10.1016/j.euo.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/07/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND Prostate cancer (PCa) patients with pathogenic/likely pathogenic germline variants (PGVs) in cancer predisposition genes may be eligible for U.S. Food and Drug Administration-approved targeted therapies, clinical trials, or enhanced screening. Studies suggest that eligible patients are missing genetics-informed care due to restrictive testing criteria. OBJECTIVE To establish the prevalence of actionable PGVs among prospectively accrued, unselected PCa patients, stratified by their guideline eligibility. DESIGN, SETTING, AND PARTICIPANTS Consecutive, unselected PCa patients were enrolled at 15 sites in the USA from October 2019 to August 2021, and had multigene cancer panel testing. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Correlates between the prevalence of PGVs and clinician-reported demographic and clinical characteristics were examined. RESULTS AND LIMITATIONS Among 958 patients (median [quartiles] age at diagnosis 65 [60, 71] yr), 627 (65%) had low- or intermediate-risk disease (grade group 1, 2, or 3). A total of 77 PGVs in 17 genes were identified in 74 patients (7.7%, 95% confidence interval [CI] 6.2-9.6%). No significant difference was found in the prevalence of PGVs among patients who met the 2019 National Comprehensive Cancer Network Prostate criteria (8.8%, 43/486, 95% CI 6.6-12%) versus those who did not (6.6%, 31/472, 95% CI 4.6-9.2%; odds ratio 1.38, 95% CI 0.85-2.23), indicating that these criteria would miss 42% of patients (31/74, 95% CI 31-53%) with PGVs. The criteria were less effective at predicting PGVs in patients from under-represented populations. Most PGVs (81%, 60/74) were potentially clinically actionable. Limitations include the inability to stratify analyses based on individual ethnicity due to low numbers of non-White patients with PGVs. CONCLUSIONS Our results indicate that almost half of PCa patients with PGVs are missed by current testing guidelines. Comprehensive germline genetic testing should be offered to all patients with PCa. PATIENT SUMMARY One in 13 patients with prostate cancer carries an inherited variant that may be actionable for the patient's current care or prevention of future cancer, and could benefit from expanded testing criteria.
Collapse
Affiliation(s)
- Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA.
| | - Mukaram Gazi
- University Urology Associates of New Jersey, Hamilton, NJ, USA
| | | | - Sean Heron
- Advanced Urology Institute, St. Petersburg, FL, USA
| | - Rishi Modh
- Advanced Urology Institute, St. Petersburg, FL, USA
| | | | | | - Aaron Berger
- Associated Urological Specialists, Chicago Ridge, IL, USA
| | | | | | | | | | | | | | | | - Paul Dato
- Genesis Healthcare Partners, San Diego, CA, USA
| | | | | | - David R Wise
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | | | - Ryan D Hernandez
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | | | - Robert L Nussbaum
- Invitae Corporation, San Francisco, CA, USA; Volunteer Faculty, University of California San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
9
|
Sciarra A, Santarelli V, Santodirocco L, Frisenda M, Salciccia S, Casale P, Forte F, Mariotti G, Moriconi M, Cattarino S, Sciarra B, Bevilacqua G, Gentilucci A. Is It Time to Anticipate the Use of PARP Inhibition in Prostate Cancer Patients? Curr Oncol 2023; 30:8054-8067. [PMID: 37754499 PMCID: PMC10528501 DOI: 10.3390/curroncol30090584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/28/2023] Open
Abstract
The increasing diffusion of genetic analysis regarding the pathogenetic variants (PVs) of genes involved in DNA Damage Repair (DDR) mechanisms and the development of Poly ADP ribose polymerase (PARP) inhibitors (PARPis) led to the first valid precision medicine option tailored toward metastatic prostate cancer (mPC). The concept of anticipation in the systemic treatment of mPC was initially adopted for androgen receptor signaling inhibitors (ARSIs) to describe the expansion of their indications, from the setting of the late-stage second-line treatment of metastatic castration-resistant prostate cancer (mCRPC) to first-line therapy in selected cases. There is already mounting evidence in favor of the anticipation of PARPis in the first line of mCRPC therapy, and further evidence in favor of mHSPC is emerging. Many studies have demonstrated the synergism between ARSIs and PARP inhibitors. Recent discoveries regarding the crosstalk between the androgen receptor (AR) and DNA repair mechanisms are disconnecting the use of PARPis from genetic analysis. The new message emerging is that the combination of PARPis with ARSIs may work independently of DDR mutational status. As a matter of fact, most of the recent trials analyzing the combination of PARPis with abiraterone or enzalutamide as a first-line therapy enrolled mCRPC patients irrespective of their mutational status. The PROPEL trial concluded that the advantage of the combination was independent of PV status, despite a higher advantage being reported in the BRCA1/2 mutated subgroup. The MAGNITUDE trial, however, showed a significant advantage only in the DDR mutated subgroup, and the DDR non-mutated cohort was closed for further enrollment. The combination of PARPis with ARSIs represents a significant strategy with a view to the anticipation and intensification of care in mPC. However, it should not nullify the advantages of precision medicine linked to the genetic analysis of DDR genes.
Collapse
Affiliation(s)
- Alessandro Sciarra
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Valerio Santarelli
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Lorenzo Santodirocco
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Marco Frisenda
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Stefano Salciccia
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Paolo Casale
- Urologic Division, Humanitas Hospital, Rozzano, 00100 Milan, Italy;
| | - Flavio Forte
- Urologic Division, Figliesancamillo Hospital, 00198 Rome, Italy;
| | - Gianna Mariotti
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Martina Moriconi
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Susanna Cattarino
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Beatrice Sciarra
- Department of Chemistry, University Sapienza, 00166 Rome, Italy;
| | - Giulio Bevilacqua
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Alessandro Gentilucci
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| |
Collapse
|
10
|
Rebhan K, Stelzer PD, Pradere B, Rajwa P, Kramer G, Hofmann B, Resch I, Yurdakul O, Laccone FA, Bujalkova MG, Smogavec M, Tan YY, Ristl R, Shariat SF, Egger G, Hassler MR. Performance of clinical risk scores and prediction models to identify pathogenic germline variants in patients with advanced prostate cancer. World J Urol 2023; 41:2091-2097. [PMID: 37528288 PMCID: PMC10415416 DOI: 10.1007/s00345-023-04535-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/11/2023] [Indexed: 08/03/2023] Open
Abstract
PURPOSE Determining the frequency and distribution of pathogenic germline variants (PGVs) in Austrian prostate cancer (PCa) patients and to assess the accuracy of different clinical risk scores to correctly predict PGVs. METHODS This cross-sectional study included 313 men with advanced PCa. A comprehensive personal and family history was obtained based on predefined questionnaires. Germline DNA sequencing was performed between 2019 and 2021 irrespective of family history, metastatic or castration status or age at diagnosis. Clinical risk scores for hereditary cancer syndromes were evaluated and a PCa-specific score was developed to assess the presence of PGVs. RESULTS PGV presence was associated with metastasis (p = 0.047) and castration resistance (p = 0.011), but not with personal cancer history or with relatives with any type of cancer. Clinical risk scores (Manchester score, PREMM5 score, Amsterdam II criteria or Johns Hopkins criteria) showed low sensitivities (3.3-20%) for assessing the probability of PGV presence. A score specifically designed for PCa patients stratifying patients into low- or high-risk regarding PGV probability, correctly classified all PGV carriers as high-risk, whereas a third of PCa patients without PGVs was classified as low risk of the presence of PGVs. CONCLUSION Application of common clinical risk scores based on family history are not suitable to identify PCa patients with high PGV probabilities. A PCa-specific score stratified PCa patients into low- or high-risk of PGV presence with sufficient accuracy, and germline DNA sequencing may be omitted in patients with a low score. Further studies are needed to evaluate the score.
Collapse
Affiliation(s)
- Katharina Rebhan
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Departments of Urology and Pediatric Urology, Klinik Ottakring, Vienna, Austria
| | - Philipp D Stelzer
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Benjamin Pradere
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Department of Urology, La Croix Du Sud Hospital, Quint Fonsegrives, France
| | - Pawel Rajwa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Gero Kramer
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Bernd Hofmann
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Irene Resch
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Ozan Yurdakul
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Franco A Laccone
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | | | - Mateja Smogavec
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Yen Y Tan
- Department of Obstetrics, Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Robin Ristl
- Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Shahrokh F Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Departments of Urology, Weill Cornell Medical College, New York, NY, USA
- Department of Urology, University of Texas Southwestern, Dallas, TX, USA
- Department of Urology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan
- Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Melanie R Hassler
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
11
|
Aguiar JA, Li EV, Siddiqui MR, Soliman MA, Kumar SKSR, Schaeffer EM, Keeter MK, Brown CH, Szymaniak BM, Ross AE. Utilization of genetic testing in men with advanced prostate cancer. Prostate 2023; 83:516-523. [PMID: 36591888 DOI: 10.1002/pros.24480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/10/2022] [Accepted: 12/19/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND Genetic evaluation of men with advanced prostate cancer is recognized as imperative both to guide treatment decisions and to trigger cascade genetic testing of family members. Here we investigate utilization patterns of genetic testing among a contemporary cohort of men with advanced prostate cancer at our institution. METHODS We queried the Northwestern Electronic Data Warehouse from January 2021 to present for all men diagnosed with National Comprehensive Cancer Network high-risk/very high-risk, regional, or metastatic prostate cancer. Patients were excluded from analyses if treated at an outside institution and/or presented for a second opinion evaluation. Statistics were performed using t-test, Chi-squared test, and univariable and multivariable logistic regression with significance defined as p < 0.05. RESULTS Atotal of 320 men (52.5%) had local/regional disease and 290 (47.5%) had metastatic disease, 53 (18.3%) of whom had castrate resistant prostate cancer. Rates of germline genetic testing rate were low in patients with localized disease (9.4%) and metastatic disease (34.1%). Only 19 (35.8%) men diagnosed with metastatic castrate resistant prostate cancer underwent germline genetic evaluation. Germline testing was most frequently discussed or ordered by medical oncologists (52%) followed by urologists (20%). Men who underwent germline testing were younger (p < 0.001), more likely to have Medicaid or private insurance (p = 0.002), and more likely to have metastatic disease (p < 0.001). There were no statistically significant differences in baseline PSA, ethnicity, race, or castration sensitivity status. Age (odds ratio [OR]: 0.94, 95% confidence interval [CI]: 0.91-0.97, p < 0.001) and metastatic disease (OR: 5.71, 95% CI: 3.63-9.22, p < 0.001) were significant independent predictors of genetic testing on multivariable logistic regression. CONCLUSIONS Here we report that utilization of genetic testing is associated with metastatic disease and inversely associated with age. Overall, utilization rates of genetic testing remain low in all patient groups, including in the metastatic castrate resistant setting, where genetic testing can identify patients with homologous recombination repair deficiency who may benefit from use of targeted therapeutics such as PARP inhibitors. Genetic testing in men with aggressive prostate cancer is critical and barriers to routine implementation of testing require further study to develop strategies to improve utilization rates.
Collapse
Affiliation(s)
- Jonathan A Aguiar
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Eric V Li
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mohammad R Siddiqui
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Moataz A Soliman
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Sai K S R Kumar
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Edward M Schaeffer
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mary Kate Keeter
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - C Hendricks Brown
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Brittany M Szymaniak
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ashley E Ross
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
12
|
Maslov DV, Sember Q, Cham J, Bhangoo M. A review of treatments targeting DNA-repair gene defects in metastatic castration resistant prostate cancer. Front Oncol 2023; 13:1150777. [PMID: 36998466 PMCID: PMC10046303 DOI: 10.3389/fonc.2023.1150777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
Prostate cancer is the most common cancer in men. About 6% of those diagnosed will develop metastatic disease. Unfortunately, metastatic prostate cancer is fatal. Prostate cancer can be castration sensitive or castration resistant. Many treatments have been shown to improve progression free survival and overall survival in metastatic castration resistant prostate cancer (mCRPC). In recent years, studies have been exploring targeting mutations in the DNA Damage Repair (DDR) response that may amplify oncogenes. In this paper, we aim to discuss DDR, new approved targeted therapies, and the most recent clinical trials in the setting of metastatic CRPC.
Collapse
Affiliation(s)
- Diana V. Maslov
- Department of Hematology/Oncology, Scripps Health System, San Diego, CA, United States
| | - Quinne Sember
- Department of Hematology/Oncology, Scripps Health System, San Diego, CA, United States
| | - Jason Cham
- Scripps Clinic/Green Hospital, Department of Internal Medicine, San Diego, CA, United States
| | - Munveer Bhangoo
- Department of Hematology/Oncology, Scripps Health System, San Diego, CA, United States
| |
Collapse
|
13
|
Uemura H, Oya M, Kamoto T, Sugimoto M, Shinozaki K, Morita K, Koto R, Takahashi M, Nii M, Shin E, Nonomura N. The prevalence of gene mutations in homologous recombination repair pathways in Japanese patients with metastatic castration-resistant prostate cancer in real-world clinical practice: The multi-institutional observational ZENSHIN study. Cancer Med 2023; 12:5265-5274. [PMID: 36358026 PMCID: PMC10028105 DOI: 10.1002/cam4.5333] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/07/2022] [Accepted: 09/25/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Metastatic castration-resistant prostate cancer (mCRPC) is a genetically heterogeneous disease with a poor prognosis. The prevalence of mutations in homologous recombination repair (HRR) pathway genes, including BRCA1/2, as well as treatment patterns and clinical outcomes, are not well characterized among Japanese men with mCRPC. METHODS This multicenter, noninterventional cohort study enrolled Japanese men with mCRPC from 24 institutions between 2014 and 2018. Mutations in the 15 HRR-related genes were assessed using archival primary or metastatic tumor samples. Patterns of sequential therapies for mCRPC were investigated. Patients were followed up for survival evaluation including prostate-specific antigen progression-free survival (PSA-PFS) and overall survival (OS). RESULTS Of the 143 patients analyzed, HRR-related mutations were detected in 51 patients (35.7%). The most frequently mutated genes were CDK12 (N = 19, 13.3%), followed by BRCA2 (N = 18, 12.6%), ATM (N = 8, 5.6%), and CHEK2 (N = 3, 2.1%). The most common type of first-line therapy for mCRPC was next-generation hormonal agents (NHA, 44.4%), followed by first-generation antiandrogens (FGA, 30.3%), and taxanes (22.5%). Commonly prescribed first-/second-line sequential regimens included FGA/NHA (17.6%), NHA/NHA (15.5%), and NHA/taxanes (14.1%). The median PSA-PFS and OS for the entire cohort were 5.6 and 26.1 months, respectively. Patients carrying BRCA1/2 mutations had numerically shorter PSA-PFS (median 3.3 vs. 5.9 months) and OS (median 20.7 vs. 27.3 months) than those without mutations. CONCLUSIONS In conclusion, approximately one-third of Japanese patients with mCRPC carried mutations in HRR-related genes in this study. The real-world outcomes of mCRPC are poor with conventional therapy, warranting an expansion of treatment options based on genetic abnormalities of the disease.
Collapse
Affiliation(s)
- Hiroji Uemura
- Department of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama City, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Toshiyuki Kamoto
- Department of Urology, Faculty of Medicine, Miyazaki University, Miyazaki, Japan
| | - Mikio Sugimoto
- Department of Urology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | | | | | | | | | | | | | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
14
|
Harper JB, Greenberg SE, Hunt TC, Cooney KA, O’Neil BB. Initial outcomes and insights from a novel high-risk prostate cancer screening clinic. Prostate 2023; 83:151-157. [PMID: 36207779 PMCID: PMC9772159 DOI: 10.1002/pros.24447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/21/2022] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Guidelines for germline testing in patients with prostate cancer (PCa) are identifying family members who require additional surveillance given pathogenic variants (PVs) that confer increased PCa risk. We established an interdisciplinary clinic for cancer surveillance in high-risk individuals aimed to implement screening recommendations. This study aimed to characterize the clinical features of this cohort. PATIENTS AND METHODS The Prostate Cancer Risk Clinic (PCRC) was established for unaffected individuals with germline PVs or a strong PCa family history. PCa screening, urine labs, and questionnaires were included in the visit. Individuals with BRCA1/2 PVs underwent clinical breast exam as well. Data from the initial visit were abstracted from the medical record and questionnaires for analysis. RESULTS Thirty-five individuals with increased PCa risk were followed by the PCRC with a median age of 47 years of age. Twenty individuals (57%) had a family history of PCa, and 34 (97%) had a germline PV associated with an increased risk for developing PCa. Four individuals underwent biopsy due to care in the PCRC, with one PCa identified in an individual with TP53 PV. Median patient response scores indicated mild symptoms of an enlarged prostate (AUASS), normal erectile function (SHIM), and relatively low anxiety about developing PCa (MAX-PC). However, there were notable "outlier" scores on each questionnaire. CONCLUSIONS Individuals with prostates and BRCA1/2 PVs, among other germline PVs, can benefit from a comprehensive interdisciplinary approach to high-risk management. PCa was identified in an individual with a non-BRCA PV, emphasizing the importance and need for high-risk screening guidelines across all genes with increased risk for PCa. "Outlier" patient response scores demonstrate that some participants experienced worse symptoms or anxiety than was indicated by median scores alone.
Collapse
Affiliation(s)
- Jonathan B. Harper
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Samantha E. Greenberg
- Genetic Counseling Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Corresponding author: Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, UT 84112, USA, Tel. +1-801-213-5774; Fax: +1-801-585-5763, (S.E. Greenberg)
| | - Trevor C. Hunt
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- University of Rochester Medical Center, Department of Urology, Rochester, NY, USA
| | - Kathleen A. Cooney
- Department of Medicine, Duke University School of Medicine, and the Duke Cancer Institute, Durham, NC, USA
| | - Brock B. O’Neil
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
15
|
Eickelschulte S, Riediger AL, Angeles AK, Janke F, Duensing S, Sültmann H, Görtz M. Biomarkers for the Detection and Risk Stratification of Aggressive Prostate Cancer. Cancers (Basel) 2022; 14:cancers14246094. [PMID: 36551580 PMCID: PMC9777028 DOI: 10.3390/cancers14246094] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Current strategies for the clinical management of prostate cancer are inadequate for a precise risk stratification between indolent and aggressive tumors. Recently developed tissue-based molecular biomarkers have refined the risk assessment of the disease. The characterization of tissue biopsy components and subsequent identification of relevant tissue-based molecular alterations have the potential to improve the clinical decision making and patient outcomes. However, tissue biopsies are invasive and spatially restricted due to tumor heterogeneity. Therefore, there is an urgent need for complementary diagnostic and prognostic options. Liquid biopsy approaches are minimally invasive with potential utility for the early detection, risk stratification, and monitoring of tumors. In this review, we focus on tissue and liquid biopsy biomarkers for early diagnosis and risk stratification of prostate cancer, including modifications on the genomic, epigenomic, transcriptomic, and proteomic levels. High-risk molecular alterations combined with orthogonal clinical parameters can improve the identification of aggressive tumors and increase patient survival.
Collapse
Affiliation(s)
- Samaneh Eickelschulte
- Junior Clinical Cooperation Unit, Multiparametric Methods for Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Anja Lisa Riediger
- Junior Clinical Cooperation Unit, Multiparametric Methods for Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Arlou Kristina Angeles
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Florian Janke
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Stefan Duensing
- Molecular Urooncology, Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Holger Sültmann
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Magdalena Görtz
- Junior Clinical Cooperation Unit, Multiparametric Methods for Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-6221-42-2603
| |
Collapse
|
16
|
Bernstein-Molho R, Friedman E, Evron E. Controversies and Open Questions in Management of Cancer-Free Carriers of Germline Pathogenic Variants in BRCA1/BRCA2. Cancers (Basel) 2022; 14:cancers14194592. [PMID: 36230512 PMCID: PMC9559251 DOI: 10.3390/cancers14194592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/06/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
Females harboring germline BRCA1/BRCA2 (BRCA) P/LPV are offered a tight surveillance scheme from the age of 25−30 years, aimed at early detection of specific cancer types, in addition to risk-reducing strategies. Multiple national and international surveillance guidelines have been published and updated over the last two decades from geographically diverse countries. We searched for guidelines published between 1 January 2015 and 1 May 2022. Differences between guidelines on issues such as primary prevention, mammography screening in young (<30 years) carriers, MRI screening in carriers above age 65 years, breast imaging (if any) after risk-reducing bilateral mastectomy, during pregnancy, and breastfeeding, and hormone-replacement therapy, are just a few notable examples. Beyond formal guidelines, BRCA carriers’ concerns also focus on the timing of risk-reducing surgeries, fertility preservation, management of menopausal symptoms in cancer survivors, and pancreatic cancer surveillance, issues that, for some, there are no data to support evidence-based recommendations. This review discusses these unsettled issues, emphasizing the importance of future studies to enable global guideline harmonization for optimal surveillance strategies. Moreover, it raises the unmet need for personalized risk stratification and surveillance in BRCA P/LPV carriers.
Collapse
Affiliation(s)
- Rinat Bernstein-Molho
- The Oncogenetics Unit, Chaim Sheba Medical Center, Tel-Hashomer, The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 5265601, Israel
| | - Eitan Friedman
- Assuta Medical Center, Tel-Aviv, Israel, The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 8436322, Israel
| | - Ella Evron
- Oncology, Kaplan Medical Institute, Rehovot, Hadassah Medical School, The Hebrew University, Jerusalem 9190501, Israel
- Correspondence: or ; Tel.: +972-502-056-171
| |
Collapse
|
17
|
Finch A, Clark R, Vesprini D, Lorentz J, Kim RH, Thain E, Fleshner N, Akbari MR, Cybulski C, Narod SA. An appraisal of genetic testing for prostate cancer susceptibility. NPJ Precis Oncol 2022; 6:43. [PMID: 35732815 PMCID: PMC9217944 DOI: 10.1038/s41698-022-00282-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 05/13/2022] [Indexed: 11/09/2022] Open
Abstract
Most criteria for genetic testing for prostate cancer susceptibility require a prior diagnosis of prostate cancer, in particular cases with metastatic disease are selected. Advances in the field are expected to improve outcomes through tailored treatments for men with advanced prostate cancer with germline pathogenic variants, although these are not currently offered in the curative setting. A better understanding of the value of genetic testing for prostate cancer susceptibility in screening, for early detection and prevention is necessary. We review and summarize the literature describing germline pathogenic variants in genes associated with increased prostate cancer risk and aggressivity. Important questions include: what is our ability to screen for and prevent prostate cancer in a man with a germline pathogenic variant and how does knowledge of a germline pathogenic variant influence treatment of men with nonmetastatic disease, with hormone-resistant disease and with metastatic disease? The frequency of germline pathogenic variants in prostate cancer is well described, according to personal and family history of cancer and by stage and grade of disease. The role of these genes in aggressive prostate cancer is also discussed. It is timely to consider whether or not genetic testing should be offered to all men with prostate cancer. The goals of testing are to facilitate screening for early cancers in unaffected high-risk men and to prevent advanced disease in men with cancer.
Collapse
Affiliation(s)
- Amy Finch
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
| | - Roderick Clark
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
- Division of Urology, University of Toronto, Ontario, Canada
| | - Danny Vesprini
- Department of Radiation Oncology, Sunnybrook Health Sciences Center, University of Toronto, Ontario, Canada
| | - Justin Lorentz
- Department of Radiation Oncology, Sunnybrook Health Sciences Center, University of Toronto, Ontario, Canada
| | - Raymond H Kim
- Familial Cancer Clinic, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Emily Thain
- Familial Cancer Clinic, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Neil Fleshner
- Division of Urology, Departments of Surgery and Surgical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Mohammad R Akbari
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Ontario, Canada
| | - Cezary Cybulski
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Steven A Narod
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada.
- Dalla Lana School of Public Health, University of Toronto, Ontario, Canada.
| |
Collapse
|
18
|
Kensler KH, Baichoo S, Pathania S, Rebbeck TR. The tumor mutational landscape of BRCA2-deficient primary and metastatic prostate cancer. NPJ Precis Oncol 2022; 6:39. [PMID: 35715489 PMCID: PMC9205939 DOI: 10.1038/s41698-022-00284-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 05/17/2022] [Indexed: 02/08/2023] Open
Abstract
Carriers of germline BRCA2 pathogenic sequence variants have elevated aggressive prostate cancer risk and are candidates for precision oncology treatments. We examined whether BRCA2-deficient (BRCA2d) prostate tumors have distinct genomic alterations compared with BRCA2-intact (BRCA2i) tumors. Among 2536 primary and 899 metastatic prostate tumors from the ICGC, GENIE, and TCGA databases, we identified 138 primary and 85 metastatic BRCA2d tumors. Total tumor mutation burden (TMB) was higher among primary BRCA2d tumors, although pathogenic TMB did not differ by tumor BRCA2 status. Pathogenic and total single nucleotide variant (SNV) frequencies at KMT2D were higher in BRCA2d primary tumors, as was the total SNV frequency at KMT2D in BRCA2d metastatic tumors. Homozygous deletions at NEK3, RB1, and APC were enriched in BRCA2d primary tumors, and RB1 deletions in metastatic BRCA2d tumors as well. TMPRSS2-ETV1 fusions were more common in BRCA2d tumors. These results identify somatic alterations that hallmark etiological and prognostic differences between BRCA2d and BRCA2i prostate tumors.
Collapse
Affiliation(s)
- Kevin H. Kensler
- grid.5386.8000000041936877XDepartment of Population Health Sciences, Weill Cornell Medicine, New York, NY USA
| | - Shakuntala Baichoo
- grid.45199.300000 0001 2288 9451Department of Digital Technologies, FoICDT, University of Mauritius, Réduit, Mauritius
| | - Shailja Pathania
- grid.266684.80000 0001 2184 9220Center for Personalized Cancer Therapy, University of Massachusetts, Boston, MA USA ,grid.266684.80000 0001 2184 9220Department of Biology, University of Massachusetts, Boston, MA USA
| | - Timothy R. Rebbeck
- grid.65499.370000 0001 2106 9910Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA USA
| |
Collapse
|
19
|
Papachristodoulou A, Abate-Shen C. Precision intervention for prostate cancer: Re-evaluating who is at risk. Cancer Lett 2022; 538:215709. [DOI: 10.1016/j.canlet.2022.215709] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/30/2022] [Accepted: 04/25/2022] [Indexed: 02/08/2023]
|
20
|
Hinata N, Fujisawa M. Racial Differences in Prostate Cancer Characteristics and Cancer-Specific Mortality: An Overview. World J Mens Health 2022; 40:217-227. [PMID: 35021294 PMCID: PMC8987139 DOI: 10.5534/wjmh.210070] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/27/2021] [Accepted: 08/04/2021] [Indexed: 11/26/2022] Open
Abstract
Racial differences of prostate cancer incidence and mortality among Asian, Black, and Caucasian men have been known, however, comprehensive update of this topic is not yet reported. In the present review, an overview of the racial differences in prostate cancer characteristics and cancer-specific mortality is collected and reviewed. Regarding racial differences of incidence and mortality, surprising differences in the incidence of prostate cancer are seen among different populations around the world, with some countries having rates that are 60 to 100 times higher than others. African-American men have a higher incidence of prostate cancer, higher prostate cancer mortality, and are diagnosed with prostate cancer at a younger age than Caucasian American men. Furthermore, race is gaining attention as an important factor to consider for planning active surveillance for localized prostate cancer, especially among African-Americans. In addition, the causes of these differences are being elucidated by genomic profiling. Determinants of racial disparities are multifactorial, including socioeconomic and biologic factors. Although race-specific differences in prostate cancer survival estimates appear to be narrowing over time, there is an ongoing need to continue to understand and mitigate racial factors associated with disparities in health care outcomes.
Collapse
Affiliation(s)
- Nobuyuki Hinata
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Masato Fujisawa
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
21
|
Omura M, Kosaka T, Aimono E, Nakamura K, Hongo H, Mikami S, Nishihara H, Oya M. First successful case of platinum-based chemotherapy for neuroendocrine prostate cancer with BRCA2 and PTEN alterations. IJU Case Rep 2022; 5:41-44. [PMID: 35005470 PMCID: PMC8720712 DOI: 10.1002/iju5.12383] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/14/2021] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION Deoxyribonucleic acid repair gene mutations are now being studied in a variety of solid tumors, with the hope of predicting prognosis, pathogenesis, and treatment outcomes. CASE PRESENTATION We report the case of a Japanese patient with advanced castration-resistant prostate cancer who exhibited a prominent response to platinum therapy and had coexisting BRCA2 and PTEN mutations according to retrospective multigene panel analysis. CONCLUSION Through a review of clinical outcomes and genetic/pathologic profiling, the presented case provides insights into future management strategies based on the tumor genetic status.
Collapse
Affiliation(s)
- Minami Omura
- Department of UrologyKeio University School of MedicineTokyoJapan
| | - Takeo Kosaka
- Department of UrologyKeio University School of MedicineTokyoJapan
| | - Eriko Aimono
- Genomics UnitKeio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Kohei Nakamura
- Genomics UnitKeio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Hiroshi Hongo
- Department of UrologyKeio University School of MedicineTokyoJapan
| | - Shuji Mikami
- Division of Diagnostic PathologyKeio University HospitalTokyoJapan
| | - Hiroshi Nishihara
- Genomics UnitKeio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Mototsugu Oya
- Department of UrologyKeio University School of MedicineTokyoJapan
| |
Collapse
|
22
|
Salmi F, Maachi F, Tazzite A, Aboutaib R, Fekkak J, Azeddoug H, Jouhadi H. Next-generation sequencing of BRCA1 and BRCA2 genes in Moroccan prostate cancer patients with positive family history. PLoS One 2021; 16:e0254101. [PMID: 34242281 PMCID: PMC8270444 DOI: 10.1371/journal.pone.0254101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 06/18/2021] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer is the most common male cancer in Morocco. Although sporadic forms account for a large proportion of patients, familial forms of prostate cancer are observed in 20% of cases and about 5% are due to hereditary transmission. Indeed, germline mutations in BRCA1/2 genes have been associated with prostate cancer risk. However, the spectrum of these mutations was not investigated in Moroccan Prostate cancer patients. Thereby, the aim of this study was to characterize and to estimate the prevalence of germline BRCA1/2 mutations and large rearrangements in Moroccan patients with familial prostate cancer. The entire coding regions and intron/exon boundaries of BRCA1 and BRCA2 genes have been analyzed by next generation sequencing (NGS) in a total of 30 familial prostate cancer patients. Three pathogenic mutations were detected in four unrelated patients (13.3%). One BRCA1 mutation (c.1953_1956delGAAA) and two BRCA2 mutations (c.7234_7235insG and BRCA2ΔE12). In addition, sixty-three distinct polymorphisms and unclassified variants have been found. Early identification of germline BRCA1/2 mutations may be relevant for the management of Moroccan prostate cancer patients.
Collapse
Affiliation(s)
- Fatiha Salmi
- Laboratory of Genetics and Molecular Pathology, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca, Morocco
| | - Fatima Maachi
- Helicobacter Pylori and Gastric Pathologies Laboratory, Pasteur Institute of Morocco, Casablanca, Morocco
| | - Amal Tazzite
- Laboratory of Genetics and Molecular Pathology, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca, Morocco
| | - Rachid Aboutaib
- Department of Urology, Ibn Rochd University Hospital Center, Casablanca, Morocco
| | - Jamal Fekkak
- Molecular Biology Department, Anoual Laboratory, Casablanca, Morocco
| | - Houssine Azeddoug
- Faculty of Sciences-Biochemistry and Molecular Biology Laboratory, University Hassan II Casablanca, Casablanca, Morocco
| | - Hassan Jouhadi
- Laboratory of Genetics and Molecular Pathology, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca, Morocco
- Mohammed VI Center for Cancer Treatment, Ibn Rochd University Hospital Center, Casablanca, Morocco
| |
Collapse
|
23
|
Abstract
Prostate cancer represents a significant health care burden in the United States due to its incidence, treatment-related morbidity, and cancer-specific mortality. The burden begins with prostate-specific antigen screening, which has been subject to controversy due to concerns of overdiagnosis and overtreatment. Advancements in molecular oncology have provided evidence for the inherited predisposition to prostate cancer, which could improve individualized, risk-adapted approaches to screening and mitigate the harms of routine screening. This review presents the current evidence for the genetic basis of prostate cancer and novel genetically informed, risk-adapted screening strategies for prostate cancer.
Collapse
|
24
|
Doan DK, Schmidt KT, Chau CH, Figg WD. Germline Genetics of Prostate Cancer: Prevalence of Risk Variants and Clinical Implications for Disease Management. Cancers (Basel) 2021; 13:cancers13092154. [PMID: 33947030 PMCID: PMC8124444 DOI: 10.3390/cancers13092154] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/30/2022] Open
Abstract
Prostate cancer has entered into the era of precision medicine with the recent approvals of targeted therapeutics (olaparib and rucaparib). The presence of germline mutations has important hereditary cancer implications for patients with prostate cancer, and germline testing is increasingly important in cancer screening, risk assessment, and the overall treatment and management of the disease. In this review, we discuss germline variants associated with inherited predisposition, prostate cancer risk and outcomes. We review recommendations for germline testing, available testing platforms, genetic counseling as well as discuss the therapeutic implications of germline variants relevant to prostate cancer treatments. Understanding the role of germline (heritable) mutations that affect prostate cancer biology and risk as well as the subsequent effect of these alterations on potential therapies is critical as the treatment paradigm shifts towards precision medicine. Furthermore, enhancing patient education tactics and healthcare system infrastructure is essential for the utilization of relevant predictive biomarkers and the improvement of clinical outcomes of patients with prostate cancer or at high risk of developing the disease.
Collapse
Affiliation(s)
| | - Keith T. Schmidt
- Clinical Pharmacology Program, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA;
| | - Cindy H. Chau
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA;
| | - William D. Figg
- Clinical Pharmacology Program, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA;
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA;
- Correspondence: ; Tel.: +1-240-760-6179; Fax: +1-240-858-3020
| |
Collapse
|
25
|
Vietri MT, D’Elia G, Caliendo G, Resse M, Casamassimi A, Passariello L, Albanese L, Cioffi M, Molinari AM. Hereditary Prostate Cancer: Genes Related, Target Therapy and Prevention. Int J Mol Sci 2021; 22:ijms22073753. [PMID: 33916521 PMCID: PMC8038462 DOI: 10.3390/ijms22073753] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/27/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) is globally the second most diagnosed cancer type and the most common cause of cancer-related deaths in men. Family history of PCa, hereditary breast and ovarian cancer (HBOC) and Lynch syndromes (LS), are among the most important risk factors compared to age, race, ethnicity and environmental factors for PCa development. Hereditary prostate cancer (HPCa) has the highest heritability of any major cancer in men. The proportion of PCa attributable to hereditary factors has been estimated in the range of 5–15%. To date, the genes more consistently associated to HPCa susceptibility include mismatch repair (MMR) genes (MLH1, MSH2, MSH6, and PMS2) and homologous recombination genes (BRCA1/2, ATM, PALB2, CHEK2). Additional genes are also recommended to be integrated into specific research, including HOXB13, BRP1 and NSB1. Importantly, BRCA1/BRCA2 and ATM mutated patients potentially benefit from Poly (ADP-ribose) polymerase PARP inhibitors, through a mechanism of synthetic lethality, causing selective tumor cell cytotoxicity in cell lines. Moreover, the detection of germline alterations in MMR genes has therapeutic implications, as it may help to predict immunotherapy benefits. Here, we discuss the current knowledge of the genetic basis for inherited predisposition to PCa, the potential target therapy, and the role of active surveillance as a management strategy for patients with low-risk PCa. Finally, the current PCa guideline recommendations are reviewed.
Collapse
Affiliation(s)
- Maria Teresa Vietri
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy; (A.C.); (A.M.M.)
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
- Correspondence: ; Tel.: +39-081-566-7639; Fax: +39-081-450-169
| | - Giovanna D’Elia
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| | - Gemma Caliendo
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| | - Marianna Resse
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| | - Amelia Casamassimi
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy; (A.C.); (A.M.M.)
| | - Luana Passariello
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| | - Luisa Albanese
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| | - Michele Cioffi
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| | - Anna Maria Molinari
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy; (A.C.); (A.M.M.)
- U.O.C. Clinical and Molecular Pathology, A.O.U. University of Campania “Luigi Vanvitelli”, 80138 Naple, Italy; (G.D.); (G.C.); (M.R.); (L.P.); (L.A.); (M.C.)
| |
Collapse
|
26
|
Greenberg SE, Hunt TC, Ambrose JP, Lowrance WT, Dechet CB, O'Neil BB, Tward JD. Clinical Germline Testing Results of Men With Prostate Cancer: Patient-Level Factors and Implications of NCCN Guideline Expansion. JCO Precis Oncol 2021; 5:PO.20.00432. [PMID: 34250421 PMCID: PMC8232879 DOI: 10.1200/po.20.00432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/17/2020] [Accepted: 02/02/2021] [Indexed: 12/24/2022] Open
Abstract
Germline likely pathogenic or pathogenic variants (PVs) have been identified in up to 17% of men with prostate cancer (PC) and may drive disease severity or be targetable by novel therapies. National Comprehensive Cancer Network (NCCN) guidelines encouraging germline testing in metastatic PC were recently expanded to include all men with high-risk, very high-risk, or regional PC. Our aim was to assess the impact of expanded NCCN guidelines on the detection rate of germline PVs and to determine patient-level factors associated with a PV germline testing result. PATIENTS AND METHODS Men with PC underwent multigene germline genetic testing for PVs from June 2016 to December 2018, and trends were compared. The association of patient-level factors with a PV germline testing result, where ≥ 1 PV was identified, was assessed using analysis of variance and univariate logistic regression. Sensitivity analyses were limited to clinically actionable variants and those associated with disease severity or progression (BRCA1/2 and ATM). RESULTS Of 408 men undergoing germline testing, 42 (10.3%) men had PVs and 366 (89.7%) men did not have PVs identified. The proportion of men identified with a germline PV remained stable following testing criteria expansion (9.4% v 10.6%, P = .73). No patient-level factors were significantly associated with increased odds of a PV germline testing result, including age at diagnosis, race, pretreatment prostate-specific antigen, Gleason grade group, NCCN risk group, and family history of cancer (breast and/or ovarian, prostate, or any cancer). CONCLUSION This study demonstrated a stable PV detection rate in men with PC using expanded criteria aligned to the updated NCCN testing guidelines. However, we did not find strong evidence to suggest that patient-level factors are associated with PV germline testing results. These findings support the recent expansion of NCCN germline testing guidelines in PC.
Collapse
Affiliation(s)
- Samantha E. Greenberg
- Genetic Counseling Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Trevor C. Hunt
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Jacob P. Ambrose
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - William T. Lowrance
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Christopher B. Dechet
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Brock B. O'Neil
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Jonathan D. Tward
- Department of Radiation Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| |
Collapse
|
27
|
Lewis DD, Cropp CD. The Impact of African Ancestry on Prostate Cancer Disparities in the Era of Precision Medicine. Genes (Basel) 2020; 11:E1471. [PMID: 33302594 PMCID: PMC7762993 DOI: 10.3390/genes11121471] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer disproportionately affects men of African ancestry at nearly twice the rate of men of European ancestry despite the advancement of treatment strategies and prevention. In this review, we discuss the underlying causes of these disparities including genetics, environmental/behavioral, and social determinants of health while highlighting the implications and challenges that contribute to the stark underrepresentation of men of African ancestry in clinical trials and genetic research studies. Reducing prostate cancer disparities through the development of personalized medicine approaches based on genetics will require a holistic understanding of the complex interplay of non-genetic factors that disproportionately exacerbate the observed disparity between men of African and European ancestries.
Collapse
Affiliation(s)
- Deyana D. Lewis
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, Baltimore, MD 21224, USA
| | - Cheryl D. Cropp
- Department of Pharmaceutical, Social and Administrative Sciences, Samford University McWhorter School of Pharmacy, Birmingham, AL 35229, USA;
| |
Collapse
|
28
|
Compérat E, Wasinger G, Oszwald A, Kain R, Cancel-Tassin G, Cussenot O. The Genetic Complexity of Prostate Cancer. Genes (Basel) 2020; 11:E1396. [PMID: 33255593 PMCID: PMC7760266 DOI: 10.3390/genes11121396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 01/09/2023] Open
Abstract
Prostate cancer (PCa) is a major concern in public health, with many genetically distinct subsets. Genomic alterations in PCa are extraordinarily complex, and both germline and somatic mutations are of great importance in the development of this tumor. The aim of this review is to provide an overview of genetic changes that can occur in the development of PCa and their role in potential therapeutic approaches. Various pathways and mechanisms proposed to play major roles in PCa are described in detail to provide an overview of current knowledge.
Collapse
Affiliation(s)
- Eva Compérat
- CeRePP/GRC5 Predictive Onco-Urology, Sorbonne University, 75020 Paris, France; (G.C.-T.); (O.C.)
- Department of Pathology, Hôpital Tenon, Sorbonne University, 75020 Paris, France
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (G.W.); (A.O.); (R.K.)
| | - Gabriel Wasinger
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (G.W.); (A.O.); (R.K.)
| | - André Oszwald
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (G.W.); (A.O.); (R.K.)
| | - Renate Kain
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (G.W.); (A.O.); (R.K.)
| | - Geraldine Cancel-Tassin
- CeRePP/GRC5 Predictive Onco-Urology, Sorbonne University, 75020 Paris, France; (G.C.-T.); (O.C.)
| | - Olivier Cussenot
- CeRePP/GRC5 Predictive Onco-Urology, Sorbonne University, 75020 Paris, France; (G.C.-T.); (O.C.)
- Department of Urology, Hôpital Tenon, Sorbonne University, 75020 Paris, France
| |
Collapse
|
29
|
Tang T, Wang LA, Wang P, Tong D, Liu G, Zhang J, Dai N, Zhang Y, Yuan G, Geary K, Zhang D, Liu Q, Jiang J. Case Report: Co-Existence of BRCA2 and PALB2 Germline Mutations in Familial Prostate Cancer With Solitary Lung Metastasis. Front Oncol 2020; 10:564694. [PMID: 33194641 PMCID: PMC7649358 DOI: 10.3389/fonc.2020.564694] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
Background Mutation-caused loss-of-function of factors involved in DNA damage response (DDR) is responsible for the development and progression of ~20% of prostate cancer (PCa). Some mutations can be used in cancer risk assessment and informed treatment decisions. Methods Target capture-based deep sequencing of 11 genes was conducted with total DNA purified from the proband’s peripheral blood. Sanger sequencing was conducted to screen potential germline mutations in the proband’s family members. Targeted sequencing of a panel of 1,021 genes was done with DNA purified from the tumor tissue. Results Two previously unreported germline mutations in the DDR pathway, BRCA2 (c.8474_8487delCATACCCTATACAG, p.A2825Vfs*15) and PALB2 (c.472delC, p.Q158Rfs*19) were identified in a patient with metastatic PCa. A specific therapeutic regimen including androgen deprivation therapy, locally radical radiotherapy, and systemic platinum chemotherapy worked well against his cancer. In addition, the metastatic ovarian cancer in the proband’s half-sister harboring the same BRCA2 germline mutation also responded well to platinum chemotherapy. Conclusions The newly identified germline mutations in DDR plays important role in PCa development. Since specific regimen worked well against this cancer, screening of DDR mutation could provide better management for patients with these mutation-mediated PCa.
Collapse
Affiliation(s)
- Tang Tang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Lin-Ang Wang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Peng Wang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Dali Tong
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Gaolei Liu
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jun Zhang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Nan Dai
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Yao Zhang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Gang Yuan
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Kyla Geary
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| | - Dianzheng Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| | - Qiuli Liu
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jun Jiang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
30
|
Nientiedt C, Duensing A, Zschäbitz S, Jäger D, Hohenfellner M, Stenzinger A, Duensing S. PARP inhibition in prostate cancer. Genes Chromosomes Cancer 2020; 60:344-351. [PMID: 33084183 DOI: 10.1002/gcc.22903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 12/11/2022] Open
Abstract
Defects in DNA damage repair genes are more common in prostate cancer than previously thought. These alterations provide an opportunity for precision oncology approaches and a number of studies have now shown that PARP inhibitors can have significant antitumor activity in men with DNA damage repair-deficient metastatic castration-resistant prostate cancer. This review summarizes the key clinical trials related to the use of PARP inhibitors in prostate cancer. Besides clinical outcomes, toxicity, and PARP inhibitor resistance, the role of different DNA repair genes in the response to PARP inhibition will be discussed.
Collapse
Affiliation(s)
- Cathleen Nientiedt
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Anette Duensing
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Precision Oncology of Urological Malignancies, Department of Urology, University Hospital Heidelberg, Heidelberg, Germany.,Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Stefanie Zschäbitz
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Markus Hohenfellner
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | | | - Stefan Duensing
- Department of Urology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Molecular Urooncology, Department of Urology, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
31
|
Sun P, Li Y, Chao X, Li J, Luo R, Li M, He J. Clinical characteristics and prognostic implications of BRCA-associated tumors in males: a pan-tumor survey. BMC Cancer 2020; 20:994. [PMID: 33054725 PMCID: PMC7556962 DOI: 10.1186/s12885-020-07481-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The BRCA mutation (BRCAm) in males has been reported to confer a higher risk for the development of various tumors. However, little is known about its clinicopathologic features and prognostic implications. DESIGN We conducted a retrospective pan-tumor survey on 346 cases of BRCA-associated tumors in males. Comparative analyses were conducted among male and female patients with BRCAm (n = 349), as well as in male patients without BRCAm (n = 4577). RESULTS Similar incidences of BRCAm (6.0 vs. 6.6%) and age at diagnosis of tumor (median, 65 vs. 60 years) were observed in male and female patients. Carcinomas of the lung, bladder, stomach, and cutaneous melanoma were the frequent tumors demonstrating BRCAm in males, of which the majority were stage II or III diseases with a higher frequency of BRCA2 mutations. Compared to that in the non-BRCAm group, cutaneous melanoma (16.3 vs. 5.0%), lung cancer (19.4 vs. 11.8%), bladder cancer (15.6 vs. 5.6%), and stomach cancer (11.9 vs. 5.5%) accounted for a higher proportion in the BRCAm group. Advanced disease and more mutation counts (median, 322 vs. 63 mutations) were also found in the BRCAm group. A total of 127 BRCA1 and 311 BRCA2 mutations were identified, of which 21.8 and 28.6% were deleterious, respectively. Frequent deleterious variants were identified in carcinomas of the breast (100.0%), colorectum (62.2%), prostate (43.3%), and stomach (42.9%). BRCA1 fusions with NF1, FAM134C, BECN1, or LSM12 and recurrent BRCA2 mutations at P606L/S, E832K/G, and T3033Lfs*29 were detected. Frameshift mutations in BRCA2 at N1784 (N1784Kfs*3, N1784Tfs*3) were frequently observed in both male and female patients. Compared with those in females, BRCA mutations in males were associated with decreased overall survival (OS) and progression-free survival (PFS). Male patients with deleterious BRCAm displayed increased OS compared with non-BRCAm carriers. The subgroup analysis demonstrated that BRCAm was associated with increased OS in gastric and bladder cancers, decreased PFS in prostate, esophageal, and head and neck cancers, and decreased OS in glioma/glioblastoma in males. CONCLUSION These findings provide an overview of the distinct characteristics and clinical outcomes of male patients with BRCA-associated tumors, suggesting the importance of further genetic BRCA testing in males.
Collapse
Affiliation(s)
- Peng Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China, 510060. .,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China, 510060. .,Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, 510080, Guangzhou, P. R. China.
| | - Yue Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China, 510060.,Department of Molecular Diagnosis, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China, 510060
| | - Xue Chao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China, 510060.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China, 510060
| | - Jibin Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China, 510060.,Department of Clinical Research, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China, 510060
| | - Rongzhen Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China, 510060.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China, 510060
| | - Mei Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China, 510060.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China, 510060
| | - Jiehua He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P. R. China, 510060. .,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China, 510060. .,Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, 510080, Guangzhou, P. R. China.
| |
Collapse
|
32
|
Mohler JL, Antonarakis ES, Armstrong AJ, D'Amico AV, Davis BJ, Dorff T, Eastham JA, Enke CA, Farrington TA, Higano CS, Horwitz EM, Hurwitz M, Ippolito JE, Kane CJ, Kuettel MR, Lang JM, McKenney J, Netto G, Penson DF, Plimack ER, Pow-Sang JM, Pugh TJ, Richey S, Roach M, Rosenfeld S, Schaeffer E, Shabsigh A, Small EJ, Spratt DE, Srinivas S, Tward J, Shead DA, Freedman-Cass DA. Prostate Cancer, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2020; 17:479-505. [PMID: 31085757 DOI: 10.6004/jnccn.2019.0023] [Citation(s) in RCA: 882] [Impact Index Per Article: 220.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The NCCN Guidelines for Prostate Cancer include recommendations regarding diagnosis, risk stratification and workup, treatment options for localized disease, and management of recurrent and advanced disease for clinicians who treat patients with prostate cancer. The portions of the guidelines included herein focus on the roles of germline and somatic genetic testing, risk stratification with nomograms and tumor multigene molecular testing, androgen deprivation therapy, secondary hormonal therapy, chemotherapy, and immunotherapy in patients with prostate cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Joseph E Ippolito
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | - Jesse McKenney
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | - George Netto
- University of Alabama at Birmingham Comprehensive Cancer Center
| | | | | | | | | | - Sylvia Richey
- St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | - Mack Roach
- UCSF Helen Diller Family Comprehensive Cancer Center
| | | | - Edward Schaeffer
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | - Ahmad Shabsigh
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Eric J Small
- UCSF Helen Diller Family Comprehensive Cancer Center
| | | | | | - Jonathan Tward
- Huntsman Cancer Institute at the University of Utah; and
| | | | | |
Collapse
|
33
|
Stellato M, Guadalupi V, Sepe P, Mennitto A, Claps M, Zattarin E, Verzoni E, Valdagni R, De Braud FG, Santini D, Tonini G, Procopio G. The emerging role of PARP inhibitors in prostate cancer. Expert Rev Anticancer Ther 2020; 20:715-726. [PMID: 32758032 DOI: 10.1080/14737140.2020.1797497] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION In prostate cancer , there has recently been an emerging interest in mutations in genes belonging to the homologous recombination repair (HRR) pathway and in the inhibition of poly (ADP-ribose) polymerase (PARP) proteins. AREAS COVERED Mutations in the HRR genes, including BRCA1, BRCA2, and Ataxia-Telangiesctasia mutated (ATM), have been reported in prostate cancer, with different incidence in the localized and advanced settings. The PARP enzyme complex is involved in repair of DNA damage and its inhibition causes the accumulation of DNA mutations in HRR deficient cells. Several PARP inhibitors (PARPi) are under development, such as olaparib, talazoparib, niraparib, rucaparib, and veliparib. In metastatic castration resistant prostate cancer (mCRPC), olaparib has been the most studied and its clinical efficacy has been validated in a phase III clinical trial. Rucaparib and niraparib have also shown promising results in the preliminary analyzes of two phase II trials, while talazoparib is currently under development. EXPERT OPINION PARPi have become part of the treatment of mCRPC. Early results of combination therapy with PARPi and new hormonal therapy are promising and are supported by a strong biological rationale. Current results need to be validated in randomized phase III-controlled trials in order to translate the use of PARPi into real world practice.
Collapse
Affiliation(s)
- Marco Stellato
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori , Milan, Italy
| | - Valentina Guadalupi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori , Milan, Italy
| | - Pierangela Sepe
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori , Milan, Italy
| | - Alessia Mennitto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori , Milan, Italy
| | - Mélanie Claps
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori , Milan, Italy
| | - Emma Zattarin
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori , Milan, Italy
| | - Elena Verzoni
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori , Milan, Italy
| | - Riccardo Valdagni
- Department of Oncology and Hemato-Oncology, University of Milan , Milan, Italy.,Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale Dei Tumori , Milano, Italy.,Radiation Oncology 1, Fondazione IRCCS Istituto Nazionale Dei Tumori , Milano, Italy
| | - Filippo Gm De Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Dei Tumori , Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan , Milan, Italy
| | - Daniele Santini
- Department of Medical Oncology, Campus Bio-Medico University of Rome , Rome, Italy
| | - Giuseppe Tonini
- Department of Medical Oncology, Campus Bio-Medico University of Rome , Rome, Italy
| | - Giuseppe Procopio
- Department of Medical Oncology, Campus Bio-Medico University of Rome , Rome, Italy
| |
Collapse
|
34
|
Findakly D, Wang J. Intricate Interplay of Entwined Metabolic and Inflammatory Life-threatening Processes in Tumor Lysis Syndrome Complicating Prostate Cancer: A Systematic Review with a Single Institution Experience. Cureus 2020; 12:e7395. [PMID: 32226700 PMCID: PMC7096067 DOI: 10.7759/cureus.7395] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tumor lysis syndrome (TLS) occurs in rapidly proliferating tumor cells, either spontaneously or after cytotoxic therapy. It has been well-documented in hematological diseases but is extremely rare in solid neoplasms, particularly in prostate cancer (PRCA). In the presence of risk factors, it can cause metabolic disturbances and be potentially fatal. We searched PubMed, Medline, ScienceDirect, and Scopus for "tumor lysis syndrome" and "prostate cancer" and conducted a systematic review with a pooled analysis for the published literature and cases from our institution. Twenty-two TLS cases were identified (18 published in the literature and four cases from our institution). The patients' median age was 68 years (range 16-82), and most cases were prostate adenocarcinoma. The median prostate-specific antigen (PSA) was 374 (range 66.7-10,867). Ten cases (45.5%) had spontaneous TLS (STLS) while 12 cases (54.5%) were treatment-related (TTLS). All patients had elevated lactate dehydrogenase (LDH) with other biochemical variables, and all underwent aggressive supportive therapy. Eleven patients underwent hemodialysis, 12 patients received rasburicase, while three patients received allopurinol. The mortality rate was 75% among 12 cases of TTLS, and it was 30% of the 10 cases with STLS. Among patients with PRCA, both TTLS and STLS linked to very high mortality. Early identification of TLS would substantially attain improved survival outcomes. Hence, physicians should consider TLS as a differential diagnosis when evaluating AKI and electrolyte abnormalities, particularly in patients with metastatic PRCA and high disease burden, even before the initiation of cytotoxic therapy.
Collapse
Affiliation(s)
- Dawood Findakly
- Internal Medicine, Creighton University Arizona Health Education Alliance / Valleywise Health Medical Center (formerly MIHS), Phoenix, USA
| | - Jue Wang
- Genitourinary Oncology, Creighton University School of Medicine / University of Arizona Cancer Center at Dignity Health St. Joseph's, Phoenix, USA
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW To provide the reader an understanding of the importance and limitations of prostate cancer (PCa) screening, the heritable component of PCa and the role that germline genetic markers can play in risk-adapted screening and treatment. RECENT FINDINGS Despite strong science supporting the association of germline genetic change with PCa risk and outcome, there has been a reluctance to pursue practical application of these technologies. Recent findings suggest that actionable information may now be garnered from this form of testing, which can help men at risk for and with PCa. SUMMARY This is an exciting time whereby germline genetic markers can help overcome some of the shortcomings of current PCa screening and treatment paradigms. Understanding their benefit and limitations while keeping the patient's best interest in mind will be the key for the responsible application of these exciting technologies.
Collapse
|
36
|
The Genomic and Molecular Pathology of Prostate Cancer: Clinical Implications for Diagnosis, Prognosis, and Therapy. Adv Anat Pathol 2020; 27:11-19. [PMID: 31503032 DOI: 10.1097/pap.0000000000000245] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Prostate cancer (PCa) is the most common noncutaneous malignancy affecting American men and the second most common cause of cancer death. The traditional risk classification schemes for PCa are limited due to the vast clinical and molecular heterogeneity of the disease. Fortunately, recent advancements in sequencing technologies have provided us with valuable insight into the genomics of PCa. To date, a wide array of recurrent genomic alterations in PCa have been identified. Incorporating these distinct molecular subtypes of PCa into prediction models provides opportunities for improved risk stratification and ultimately better patient outcomes. In this review, we summarize the key molecular subtypes of PCa and focus on those genomic alterations that have clinical implications for diagnosis, prognosis, and therapeutic response.
Collapse
|
37
|
Page EC, Bancroft EK, Brook MN, Assel M, Hassan Al Battat M, Thomas S, Taylor N, Chamberlain A, Pope J, Raghallaigh HN, Evans DG, Rothwell J, Maehle L, Grindedal EM, James P, Mascarenhas L, McKinley J, Side L, Thomas T, van Asperen C, Vasen H, Kiemeney LA, Ringelberg J, Jensen TD, Osther PJS, Helfand BT, Genova E, Oldenburg RA, Cybulski C, Wokolorczyk D, Ong KR, Huber C, Lam J, Taylor L, Salinas M, Feliubadaló L, Oosterwijk JC, van Zelst-Stams W, Cook J, Rosario DJ, Domchek S, Powers J, Buys S, O'Toole K, Ausems MGEM, Schmutzler RK, Rhiem K, Izatt L, Tripathi V, Teixeira MR, Cardoso M, Foulkes WD, Aprikian A, van Randeraad H, Davidson R, Longmuir M, Ruijs MWG, Helderman van den Enden ATJM, Adank M, Williams R, Andrews L, Murphy DG, Halliday D, Walker L, Liljegren A, Carlsson S, Azzabi A, Jobson I, Morton C, Shackleton K, Snape K, Hanson H, Harris M, Tischkowitz M, Taylor A, Kirk J, Susman R, Chen-Shtoyerman R, Spigelman A, Pachter N, Ahmed M, Ramon Y Cajal T, Zgajnar J, Brewer C, Gadea N, Brady AF, van Os T, Gallagher D, Johannsson O, Donaldson A, Barwell J, Nicolai N, Friedman E, Obeid E, Greenhalgh L, Murthy V, Copakova L, Saya S, McGrath J, Cooke P, Rønlund K, Richardson K, Henderson A, Teo SH, Arun B, Kast K, Dias A, Aaronson NK, Ardern-Jones A, Bangma CH, Castro E, Dearnaley D, Eccles DM, Tricker K, Eyfjord J, Falconer A, Foster C, Gronberg H, Hamdy FC, Stefansdottir V, Khoo V, Lindeman GJ, Lubinski J, Axcrona K, Mikropoulos C, Mitra A, Moynihan C, Rennert G, Suri M, Wilson P, Dudderidge T, Offman J, Kote-Jarai Z, Vickers A, Lilja H, Eeles RA. Interim Results from the IMPACT Study: Evidence for Prostate-specific Antigen Screening in BRCA2 Mutation Carriers. Eur Urol 2019; 76:831-842. [PMID: 31537406 PMCID: PMC6880781 DOI: 10.1016/j.eururo.2019.08.019] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/12/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND Mutations in BRCA2 cause a higher risk of early-onset aggressive prostate cancer (PrCa). The IMPACT study is evaluating targeted PrCa screening using prostate-specific-antigen (PSA) in men with germline BRCA1/2 mutations. OBJECTIVE To report the utility of PSA screening, PrCa incidence, positive predictive value of PSA, biopsy, and tumour characteristics after 3 yr of screening, by BRCA status. DESIGN, SETTING, AND PARTICIPANTS Men aged 40-69 yr with a germline pathogenic BRCA1/2 mutation and male controls testing negative for a familial BRCA1/2 mutation were recruited. Participants underwent PSA screening for 3 yr, and if PSA > 3.0 ng/ml, men were offered prostate biopsy. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS PSA levels, PrCa incidence, and tumour characteristics were evaluated. Statistical analyses included Poisson regression offset by person-year follow-up, chi-square tests for proportion t tests for means, and Kruskal-Wallis for medians. RESULTS AND LIMITATIONS A total of 3027 patients (2932 unique individuals) were recruited (919 BRCA1 carriers, 709 BRCA1 noncarriers, 902 BRCA2 carriers, and 497 BRCA2 noncarriers). After 3 yr of screening, 527 men had PSA > 3.0 ng/ml, 357 biopsies were performed, and 112 PrCa cases were diagnosed (31 BRCA1 carriers, 19 BRCA1 noncarriers, 47 BRCA2 carriers, and 15 BRCA2 noncarriers). Higher compliance with biopsy was observed in BRCA2 carriers compared with noncarriers (73% vs 60%). Cancer incidence rate per 1000 person years was higher in BRCA2 carriers than in noncarriers (19.4 vs 12.0; p = 0.03); BRCA2 carriers were diagnosed at a younger age (61 vs 64 yr; p = 0.04) and were more likely to have clinically significant disease than BRCA2 noncarriers (77% vs 40%; p = 0.01). No differences in age or tumour characteristics were detected between BRCA1 carriers and BRCA1 noncarriers. The 4 kallikrein marker model discriminated better (area under the curve [AUC] = 0.73) for clinically significant cancer at biopsy than PSA alone (AUC = 0.65). CONCLUSIONS After 3 yr of screening, compared with noncarriers, BRCA2 mutation carriers were associated with a higher incidence of PrCa, younger age of diagnosis, and clinically significant tumours. Therefore, systematic PSA screening is indicated for men with a BRCA2 mutation. Further follow-up is required to assess the role of screening in BRCA1 mutation carriers. PATIENT SUMMARY We demonstrate that after 3 yr of prostate-specific antigen (PSA) testing, we detect more serious prostate cancers in men with BRCA2 mutations than in those without these mutations. We recommend that male BRCA2 carriers are offered systematic PSA screening.
Collapse
Affiliation(s)
| | - Elizabeth K Bancroft
- Oncogenetics Team, Institute of Cancer Research, London, UK; Cancer Genetics Unit and Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Mark N Brook
- Oncogenetics Team, Institute of Cancer Research, London, UK
| | - Melissa Assel
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY, USA
| | | | - Sarah Thomas
- Cancer Genetics Unit and Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Natalie Taylor
- Cancer Genetics Unit and Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK
| | | | - Jennifer Pope
- Oncogenetics Team, Institute of Cancer Research, London, UK
| | | | - D Gareth Evans
- Genetic Medicine, Manchester Academic Health Sciences Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Jeanette Rothwell
- Genetic Medicine, Manchester Academic Health Sciences Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Lovise Maehle
- Department of medical genetics, Oslo University Hospital, 0424 Oslo, Norway
| | | | - Paul James
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC, Australia; Genetic Medicine, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Lyon Mascarenhas
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Joanne McKinley
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Lucy Side
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | - Tessy Thomas
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | | | - Hans Vasen
- The Foundation for the Detection of Hereditary Cancer, Leiden, The Netherlands
| | | | - Janneke Ringelberg
- The Foundation for the Detection of Hereditary Cancer, Leiden, The Netherlands
| | | | | | - Brian T Helfand
- John and Carol Walter Center for Urological Health, Division of Urology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Elena Genova
- John and Carol Walter Center for Urological Health, Division of Urology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Rogier A Oldenburg
- Department of clinical genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Cezary Cybulski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Dominika Wokolorczyk
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Kai-Ren Ong
- Clinical Genetics Unit, Birmingham Women's Hospital, Birmingham, UK
| | - Camilla Huber
- Clinical Genetics Unit, Birmingham Women's Hospital, Birmingham, UK
| | - Jimmy Lam
- Department of Urology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Louise Taylor
- Department of Urology, Repatriation General Hospital, Daw Park, SA, Australia
| | - Monica Salinas
- Hereditary Cancer Program, ICO-IDIBELL (Bellvitge Biomedical Research Institute, Catalan Institute of Oncology), CIBERONC, Barcelona, Spain
| | - Lidia Feliubadaló
- Hereditary Cancer Program, ICO-IDIBELL (Bellvitge Biomedical Research Institute, Catalan Institute of Oncology), CIBERONC, Barcelona, Spain
| | - Jan C Oosterwijk
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Jackie Cook
- Sheffield Clinical Genetics Service, Sheffield Children's Hospital, Sheffield, UK
| | | | - Susan Domchek
- Basser Research Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacquelyn Powers
- Basser Research Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Saundra Buys
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Karen O'Toole
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Margreet G E M Ausems
- Division of Laboratories, Pharmacy and Biomedical Genetics, Department of Genetics, University Medical Centre, Utrecht, The Netherlands
| | - Rita K Schmutzler
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Kerstin Rhiem
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Louise Izatt
- Clinical Genetics Service, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Vishakha Tripathi
- Clinical Genetics Service, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Manuel R Teixeira
- Genetics Department and Research Center, Portuguese Oncology Institute (IPO Porto), Porto, Portugal; Biomedical Sciences Institute (ICBAS), Porto University, Porto, Portugal
| | - Marta Cardoso
- Genetics Department and Research Center, Portuguese Oncology Institute (IPO Porto), Porto, Portugal
| | - William D Foulkes
- Cancer Research Program, Research Institute of McGill University Health Centre, Montreal, Quebec, Canada; Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Armen Aprikian
- Cancer Research Program, Research Institute of McGill University Health Centre, Montreal, Quebec, Canada
| | | | - Rosemarie Davidson
- West of Scotland Genetic Service, Queen Elizabeth University Hospital, Glasgow, UK
| | - Mark Longmuir
- West of Scotland Genetic Service, Queen Elizabeth University Hospital, Glasgow, UK
| | | | | | - Muriel Adank
- VU University Medical Center, Amsterdam, The Netherlands
| | - Rachel Williams
- Hereditary Cancer Centre, Prince of Wales Hospital, Randwick, NSW, Australia; Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Lesley Andrews
- Hereditary Cancer Centre, Prince of Wales Hospital, Randwick, NSW, Australia; Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Declan G Murphy
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC, Australia; Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Dorothy Halliday
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Lisa Walker
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Annelie Liljegren
- Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Stefan Carlsson
- Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Ashraf Azzabi
- Northern Genetics Service, Newcastle upon Tyne Hospitals, UK
| | - Irene Jobson
- Northern Genetics Service, Newcastle upon Tyne Hospitals, UK
| | - Catherine Morton
- Familial Cancer Centre, The Royal Melbourne Hospital, Grattan St, Parkville, VIC, Australia
| | - Kylie Shackleton
- Familial Cancer Centre, The Royal Melbourne Hospital, Grattan St, Parkville, VIC, Australia
| | | | | | - Marion Harris
- Familial Cancer Centre, Monash Health, Clayton, VIC, Australia
| | - Marc Tischkowitz
- East Anglian Medical Genetics Service, Cambridge University Hospitals NHS Trust, Cambridge, UK; Academic Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Level 6 Addenbrooke's Treatment Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Amy Taylor
- East Anglian Medical Genetics Service, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Judy Kirk
- Familial Cancer Service, Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, Sydney, NSW, Australia; Sydney Medical School, University of Sydney, Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, Sydney, NSW, Australia
| | - Rachel Susman
- Genetic Health Queensland, Royal Brisbane & Women's Hospital, Herston, QLD, Australia
| | | | - Allan Spigelman
- Hunter Family Cancer Service, Waratah, NSW, Australia; University of New South Wales, St Vincent's Clinical School, NSW, Australia; Cancer Genetics Clinic, The Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, NSW, Australia
| | - Nicholas Pachter
- Genetic Services of WA, King Edward Memorial Hospital, Subiaco, WA, Australia; Department of Paediatrics, University of Western Australia, Perth, WA, Australia
| | - Munaza Ahmed
- NE Thames Regional Genetics Service, Institute of Child Health, London, UK
| | | | | | - Carole Brewer
- Peninsular Genetics, Derriford Hospital, Plymouth, UK; Royal Devon and Exeter Hospital, Exeter, UK
| | - Neus Gadea
- Hospital Vall d'Hebron, Barcelona, Spain
| | - Angela F Brady
- North West Thames Regional Genetics Service, Kennedy-Galton Centre, London North West University Healthcare NHS Trust, Harrow, UK
| | - Theo van Os
- Academic Medical Center, Amsterdam, The Netherlands
| | | | - Oskar Johannsson
- Landspitali-the National University Hospital of Iceland, Reykjavik, Iceland
| | | | - Julian Barwell
- University of Leicester, Leicester, UK; University Hospitals Leicester, Leicester, UK
| | | | | | - Elias Obeid
- Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Lynn Greenhalgh
- Clinical Genetics Service, Liverpool Women's Hospital, Liverpool, UK
| | | | | | - Sibel Saya
- Oncogenetics Team, Institute of Cancer Research, London, UK
| | - John McGrath
- Royal Devon and Exeter Hospital, Exeter, UK; University of Exeter Medical School, St Luke's Campus, Exeter, UK
| | | | - Karina Rønlund
- Department of Clinical Genetics, Vejle Hospital, Vejle, Denmark
| | - Kate Richardson
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Alex Henderson
- Northern Genetics Service, Newcastle upon Tyne Hospitals, UK; West Cumberland Infirmary, Whitehaven, UK
| | - Soo H Teo
- Cancer Research Initiatives Foundation, Subang Jaya Medical Centre, Selangor, Darul Ehsan, Malaysia
| | - Banu Arun
- The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Karin Kast
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Alexander Dias
- Oncogenetics Team, Institute of Cancer Research, London, UK; Instituto Nacional de Cancer Jose de Alencar Gomes da Silva (INCA), Rio de Janeiro, Brazil
| | | | - Audrey Ardern-Jones
- Cancer Genetics Unit and Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Chris H Bangma
- Department of urology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Elena Castro
- Spanish National Cancer Research Center, Madrid, Spain
| | - David Dearnaley
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, UK
| | - Diana M Eccles
- The University of Southampton Medical School, Southampton, UK; Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | - Karen Tricker
- Genetic Medicine, Manchester Academic Health Sciences Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Jorunn Eyfjord
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | - Freddie C Hamdy
- Churchill Hospital, Headington, Oxford, UK; Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | | | - Vincent Khoo
- Cancer Genetics Unit and Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK; St George's Hospital, Tooting, London, UK; Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, UK; Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Geoffrey J Lindeman
- Familial Cancer Centre, The Royal Melbourne Hospital, Grattan St, Parkville, VIC, Australia; Department of Medicine, The University of Melbourne, Parkville, VIC, Australia; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Jan Lubinski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Karol Axcrona
- Department of Urology, Akershus University Hospital, Lørenskog, Norway
| | | | - Anita Mitra
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Clare Moynihan
- Oncogenetics Team, Institute of Cancer Research, London, UK
| | - Gadi Rennert
- CHS National Cancer Control Center, Carmel Medical Center, Haifa, Israel
| | | | | | | | - Judith Offman
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Cancer Centre, Guy's Hospital, London, UK
| | | | - Andrew Vickers
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, NY, USA
| | - Hans Lilja
- Department of Translational Medicine, Lund University, Malmö, Sweden; Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Departments of Laboratory Medicine, Surgery, and Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Rosalind A Eeles
- Oncogenetics Team, Institute of Cancer Research, London, UK; Cancer Genetics Unit and Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK.
| |
Collapse
|
38
|
Giri VN, Hegarty SE, Hyatt C, O'Leary E, Garcia J, Knudsen KE, Kelly WK, Gomella LG. Germline genetic testing for inherited prostate cancer in practice: Implications for genetic testing, precision therapy, and cascade testing. Prostate 2019; 79:333-339. [PMID: 30450585 DOI: 10.1002/pros.23739] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 10/10/2018] [Indexed: 11/05/2022]
Abstract
BACKGROUND Genetic testing capability and guidelines are rapidly expanding to assess inherited prostate cancer (PCA). Clinical genetic data from multigene testing can provide insights into the germline pathogenic variant (PV) spectrum and correlates in men with PCA unselected for metastatic disease to optimize identification of men for genetic evaluation and management. METHODS A retrospective cross-sectional analysis was conducted of de-identified clinical genetic testing data from a large commercial genetic testing laboratory in the US. ICD-10 claims codes were used to identify men with PCA, along with family history data. Gleason score was abstracted from test request forms. Overall PV rate among men with PCA was estimated, along with PVs in DNA repair genes. Family history and Gleason score association to germline DNA repair PVs was assessed using Fisher's exact test with correction for false-discovery. RESULTS As of August 2017, genetic results were available on 1328 men with PCA. Overall PV rate was 15.6%, with 10.9% of PV in DNA repair genes. PVs were most commonly identified in BRCA2 (4.5%), CHEK2 (2.2%), ATM (1.8%), and BRCA1 (1.1%). Breast cancer family history was significantly associated with germline DNA repair PVs (OR 1.89, [95%CI 1.33, 2.68], P = 0.003). Among men with Gleason score>= 6 (n = 706), Gleason> = 8 was significantly associated with DNA repair PVs (OR 1.85 [95%CI 1.22, 2.80], P = 0.004). CONCLUSIONS A substantial proportion of men with PCA unselected for metastatic disease carry germline DNA repair PVs. Breast cancer family history and high Gleason score are important predictors to identify men with PCA who may carry germline DNA repair PVs. Our findings support current NCCN guidelines and have implications for genetic assessment, therapeutic management, and cascade testing for men with PCA and their families.
Collapse
Affiliation(s)
- Veda N Giri
- Cancer Risk Assessment and Clinical Cancer Genetics, Division of Population Science, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sarah E Hegarty
- Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Colette Hyatt
- Cancer Risk Assessment and Clinical Cancer Genetics, Division of Population Science, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | | | - Karen E Knudsen
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - William K Kelly
- Division of Solid Tumor Oncology, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Leonard G Gomella
- Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
39
|
Current progress and questions in germline genetics of prostate cancer. Asian J Urol 2018; 6:3-9. [PMID: 30775244 PMCID: PMC6363602 DOI: 10.1016/j.ajur.2018.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/07/2018] [Indexed: 12/31/2022] Open
Abstract
Dramatic progress has been made in the area of germline genetics of prostate cancer (PCa) in the past decade. Both common and rare genetic variants with effects on risk ranging from barely detectable to outright practice-changing have been identified. For men with high risk PCa, the application of genetic testing for inherited pathogenic mutations is becoming standard of care. A major question exists about which additional populations of men to test, as men at all risk levels can potentially benefit by knowing their unique genetic profile of germline susceptibility variants. This article will provide a brief overview of some current issues in understanding inherited susceptibility for PCa.
Collapse
|
40
|
Dias A, Kote-Jarai Z, Mikropoulos C, Eeles R. Prostate Cancer Germline Variations and Implications for Screening and Treatment. Cold Spring Harb Perspect Med 2018; 8:a030379. [PMID: 29101112 PMCID: PMC6120689 DOI: 10.1101/cshperspect.a030379] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PCa) is a highly heritable disease, and rapid evolution of sequencing technologies has enabled marked progression of our understanding of its genetic inheritance. A complex polygenic model that involves common low-penetrance susceptibility alleles causing individually small but cumulatively significant risk and rarer genetic variants causing greater risk represent the current most accepted model. Through genome-wide association studies, more than 100 single-nucleotide polymorphisms (SNPs) associated with PCa risk have been identified. Consistent reports have identified germline mutations in the genes BRCA1, BRCA2, MMR, HOXB13, CHEK2, and NBS1 as conferring moderate risks, with some leading to a more aggressive disease behavior. Considering this knowledge, several research strategies have been developed to determine whether targeted prostate screening using genetic information can overcome the limitations of population-based prostate-specific antigen (PSA) screening. Germline DNA-repair mutations are more frequent in men with metastatic disease than previously thought, and these patients have a more favorable response to therapy with poly(adenosine diphosphate [ADP]-ribose) polymerase (PARP) inhibitors. Genomic information is a practical tool that has the potential to enable the concept of precision medicine to become a reality in all steps of PCa patient care.
Collapse
Affiliation(s)
- Alexander Dias
- The Institute of Cancer Research, Sutton, Surrey SM2 5NG, United Kingdom
- The Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Academic Urology Unit and The Oncogenetics Team, London SW3 6JJ, United Kingdom
| | - Zsofia Kote-Jarai
- The Institute of Cancer Research, Sutton, Surrey SM2 5NG, United Kingdom
| | | | - Ros Eeles
- The Institute of Cancer Research, Sutton, Surrey SM2 5NG, United Kingdom
- The Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Academic Urology Unit and The Oncogenetics Team, London SW3 6JJ, United Kingdom
| |
Collapse
|
41
|
Frank S, Nelson P, Vasioukhin V. Recent advances in prostate cancer research: large-scale genomic analyses reveal novel driver mutations and DNA repair defects. F1000Res 2018; 7. [PMID: 30135717 PMCID: PMC6073096 DOI: 10.12688/f1000research.14499.1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/24/2018] [Indexed: 12/13/2022] Open
Abstract
Prostate cancer (PCa) is a disease of mutated and misregulated genes. However, primary prostate tumors have relatively few mutations, and only three genes (
ERG,
PTEN, and
SPOP) are recurrently mutated in more than 10% of primary tumors. On the other hand, metastatic castration-resistant tumors have more mutations, but, with the exception of the androgen receptor gene (
AR), no single gene is altered in more than half of tumors. Structural genomic rearrangements are common, including
ERG fusions, copy gains involving the
MYC locus, and copy losses containing
PTEN. Overall, instead of being associated with a single dominant driver event, prostate tumors display various combinations of modifications in oncogenes and tumor suppressors. This review takes a broad look at the recent advances in PCa research, including understanding the genetic alterations that drive the disease and how specific mutations can sensitize tumors to potential therapies. We begin with an overview of the genomic landscape of primary and metastatic PCa, enabled by recent large-scale sequencing efforts. Advances in three-dimensional cell culture techniques and mouse models for PCa are also discussed, and particular emphasis is placed on the benefits of patient-derived xenograft models. We also review research into understanding how ETS fusions (in particular,
TMPRSS2-ERG) and
SPOP mutations contribute to tumor initiation. Next, we examine the recent findings on the prevalence of germline DNA repair mutations in about 12% of patients with metastatic disease and their potential benefit from the use of poly(ADP-ribose) polymerase (PARP) inhibitors and immune modulation. Lastly, we discuss the recent increased prevalence of AR-negative tumors (neuroendocrine and double-negative) and the current state of immunotherapy in PCa. AR remains the primary clinical target for PCa therapies; however, it does not act alone, and better understanding of supporting mutations may help guide the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Sander Frank
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Peter Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Departments of Medicine and Urology, University of Washington, Seattle, WA 98195, USA.,Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Valeri Vasioukhin
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Department of Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
42
|
Cheng H, Powers J, Schaffer K, Sartor O. Practical Methods for Integrating Genetic Testing Into Clinical Practice for Advanced Prostate Cancer. Am Soc Clin Oncol Educ Book 2018; 38:372-381. [PMID: 30231311 DOI: 10.1200/edbk_205441] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Recent advances clearly demonstrate the potential clinical relevance of germline genetic testing and somatic genomic profiling in identifying possible therapeutic and/or clinical trial options, particularly in advanced prostate cancer. In addition, if a germline genetic mutation/pathogenic variant is identified, there may be important family implications and possible life-saving changes to healthcare management. However, there is substantial debate and uncertainty about how best to offer genetic testing services, which tests to use, which patients to test, what sequence of testing, what timing, by whom, and with what kind of follow-up. To help address this new area of potential benefit and confusion, we provide a practical overview of recent advances, discuss options and considerations for both germline and somatic testing, and offer practical advice on what providers should understand before referring and/or ordering testing, key discussion points for patients and families, and available genetics resources.
Collapse
Affiliation(s)
- Heather Cheng
- From the University of Washington, Seattle, WA; University of Pennsylvania, Philadelphia, PA; University of Rochester, Rochester, NY; Tulane Cancer Center, New Orleans, LA
| | - Jacquelyn Powers
- From the University of Washington, Seattle, WA; University of Pennsylvania, Philadelphia, PA; University of Rochester, Rochester, NY; Tulane Cancer Center, New Orleans, LA
| | - Kerry Schaffer
- From the University of Washington, Seattle, WA; University of Pennsylvania, Philadelphia, PA; University of Rochester, Rochester, NY; Tulane Cancer Center, New Orleans, LA
| | - Oliver Sartor
- From the University of Washington, Seattle, WA; University of Pennsylvania, Philadelphia, PA; University of Rochester, Rochester, NY; Tulane Cancer Center, New Orleans, LA
| |
Collapse
|
43
|
Liu Q, Tong D, Liu G, Yi Y, Xu J, Yang X, Wang L, Zhang J, Ye J, Zhang Y, Yuan G, Wang P, Chen R, Guan Y, Yi X, Zhang D, Jiang J. A novel BRCA2 mutation in prostate cancer sensitive to combined radiotherapy and androgen deprivation therapy. Cancer Biol Ther 2018; 19:669-675. [PMID: 29580149 PMCID: PMC6067857 DOI: 10.1080/15384047.2018.1451278] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Genetic factors contribute to more than 40% of prostate cancer risk, and mutations in BRCA1 and BRCA2 are well-established risk factors. By using target capture-based deep sequencing to identify potential pathogenic germline mutations, followed by Sanger sequencing to determine the loci of the mutations, we identified a novel pathogenic BRCA2 mutation caused by a cytosine-to-guanine base substitution at position 4211, resulting in protein truncation (p.Ser1404Ter), which was confirmed by immunohistochemistry. Analysis of peripheral blood also identified benign polymorphisms in BRCA2 (c.7397T>C, p.Val2466Ala) and SRD5A2 (c.87G>C, p.Lys29Asn). Analysis of tumor tissues revealed seven somatic mutations in prostate tumor tissue and nine somatic mutations in esophageal squamous carcinoma tissue (single nucleotide polymorphisms, insertions, and deletions). Five-year follow-up results indicate that ADT combined with radiotherapy successfully treated the prostate cancer. To our knowledge, we are the first to report the germline BRCA2 mutation c.4211C>G (p.Ser1404Ter) in prostate cancer. Combined ADT and radiotherapy may be effective in treating other patients with prostate cancer caused by this or similar mutations.
Collapse
Affiliation(s)
- Qiuli Liu
- a Department of Urology , Daping Hospital/Institute of Surgery Research, Third Military Medical University , Chongqing , China
| | - Dali Tong
- a Department of Urology , Daping Hospital/Institute of Surgery Research, Third Military Medical University , Chongqing , China
| | - Gaolei Liu
- a Department of Urology , Daping Hospital/Institute of Surgery Research, Third Military Medical University , Chongqing , China
| | - Yuting Yi
- b Department of Medical Center , Geneplus-Beijing Institute , Beijing , PR China
| | - Jing Xu
- a Department of Urology , Daping Hospital/Institute of Surgery Research, Third Military Medical University , Chongqing , China
| | - Xingxia Yang
- a Department of Urology , Daping Hospital/Institute of Surgery Research, Third Military Medical University , Chongqing , China
| | - Linang Wang
- a Department of Urology , Daping Hospital/Institute of Surgery Research, Third Military Medical University , Chongqing , China
| | - Jun Zhang
- a Department of Urology , Daping Hospital/Institute of Surgery Research, Third Military Medical University , Chongqing , China
| | - Jin Ye
- a Department of Urology , Daping Hospital/Institute of Surgery Research, Third Military Medical University , Chongqing , China
| | - Yao Zhang
- a Department of Urology , Daping Hospital/Institute of Surgery Research, Third Military Medical University , Chongqing , China
| | - Gang Yuan
- a Department of Urology , Daping Hospital/Institute of Surgery Research, Third Military Medical University , Chongqing , China
| | - Peng Wang
- a Department of Urology , Daping Hospital/Institute of Surgery Research, Third Military Medical University , Chongqing , China
| | - Rongrong Chen
- b Department of Medical Center , Geneplus-Beijing Institute , Beijing , PR China
| | - Yanfang Guan
- b Department of Medical Center , Geneplus-Beijing Institute , Beijing , PR China
| | - Xin Yi
- b Department of Medical Center , Geneplus-Beijing Institute , Beijing , PR China
| | - Dianzheng Zhang
- c Department of Bio-Medical Sciences , Philadelphia College of Osteopathic Medicine , 4170 City Ave, Philadelphia , PA
| | - Jun Jiang
- a Department of Urology , Daping Hospital/Institute of Surgery Research, Third Military Medical University , Chongqing , China
| |
Collapse
|
44
|
Cui M, Gao XS, Gu X, Guo W, Li X, Ma M, Qin S, Qi X, Xie M, Peng C, Bai Y. BRCA2 mutations should be screened early and routinely as markers of poor prognosis: evidence from 8,988 patients with prostate cancer. Oncotarget 2018; 8:40222-40232. [PMID: 28410213 PMCID: PMC5522317 DOI: 10.18632/oncotarget.16712] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/21/2017] [Indexed: 12/26/2022] Open
Abstract
The aim of this study was to focus on clinicopathological characteristics and prognosis in men with prostate cancer (PCa) harboring a breast cancer 2 (BRCA2) gene mutation and to offer convincing evidence to consider BRCA2 mutation as a marker of poor prognosis in the molecular classification of PCa. We searched relevant articles from PubMed, Embase, Web of Science, and the Cochrane Library databases to evaluate the differences in the overall survival (OS) and cancer-specific survival (CSS) between BRCA2 mutation carriers and non-carriers in patients with PCa. We included 525 BRCA2 mutation-carriers and 8,463 non-carriers in total from 10 studies in our meta-analysis. The results showed that carrying a BRCA2 mutation was correlated with a reduced CSS and OS when compared with that of non-carriers, with pooled Hazard Ratios (HRs) of 2.53 (95% confidence interval (CI): 2.10–3.06, P < 0.001) and 2.21 (95% CI: 1.64–2.99, P < 0.001), respectively. The results also demonstrated that BRCA2 mutation-carriers harbored a higher Gleason Score (GS) (> 7), TNM stage (> T3, N1, M1), and risk level than non-carriers. Our meta-analysis showed that a BRCA2 mutation predicted poor survival outcomes in patients with prostate cancer, especially in those undergoing treatments with radiotherapy. Therefore, the use of BRCA2 mutation as a clinical prognostic factor could help stratify the high-risk patients and provide clinical strategies for more effective targeted treatments for patients with prostate cancer.
Collapse
Affiliation(s)
- Ming Cui
- Department of Radiation Oncology, Peking University First Hospital, Peking University, Beijing, China
| | - Xian-Shu Gao
- Department of Radiation Oncology, Peking University First Hospital, Peking University, Beijing, China
| | - Xiaobin Gu
- Department of Radiation Oncology, Peking University First Hospital, Peking University, Beijing, China
| | - Wei Guo
- Graduate School of Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Xiaoying Li
- Department of Radiation Oncology, Peking University First Hospital, Peking University, Beijing, China
| | - Mingwei Ma
- Department of Radiation Oncology, Peking University First Hospital, Peking University, Beijing, China
| | - Shangbin Qin
- Department of Radiation Oncology, Peking University First Hospital, Peking University, Beijing, China
| | - Xin Qi
- Department of Radiation Oncology, Peking University First Hospital, Peking University, Beijing, China
| | - Mu Xie
- Department of Radiation Oncology, Peking University First Hospital, Peking University, Beijing, China
| | - Chuan Peng
- Department of Radiation Oncology, Peking University First Hospital, Peking University, Beijing, China
| | - Yun Bai
- Department of Radiation Oncology, Peking University First Hospital, Peking University, Beijing, China
| |
Collapse
|
45
|
Ibrahim M, Yadav S, Ogunleye F, Zakalik D. Male BRCA mutation carriers: clinical characteristics and cancer spectrum. BMC Cancer 2018; 18:179. [PMID: 29433453 PMCID: PMC5809938 DOI: 10.1186/s12885-018-4098-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 02/05/2018] [Indexed: 01/07/2023] Open
Abstract
Background Mutations in BRCA1 and BRCA2 (BRCA1/2) genes are associated with an increased risk of breast and ovarian cancers in women. The cancer characteristics of men with BRCA1/2 mutations are less well studied. This study describes the unique cancer characteristics of male BRCA1/2 mutation carriers at our institution. Methods We performed a retrospective chart review on male patients who were seen between January 2004 and December 2014 and tested positive for a BRCA1/2 mutation. We evaluated clinical characteristics, pathology findings, treatment selection and survival. Results A total of 102 male patients were identified who tested positive for a BRCA1/2 deleterious mutation. Of these 102 patients, 33 (32%) had a diagnosis of cancer. Of these 33 patients with cancer, the majority (20 patients) were found to carry a BRCA2 mutation. Median age of cancer diagnosis was 65 years (Range: 35-75 years). Of the 33 patients diagnosed with cancer, 8 had two or more cancers, including 1 patient who had 4 cancers. Prostate cancer was the most commonly diagnosed cancer, seen in 13 patients, 11 of whom were BRCA2 positive. These cancers tended to have higher Gleason scores and elevated PSA levels. The majority of these prostate cancer patients were alive and disease free at a median follow-up of 7.4 years. Male breast cancer was the second most common cancer seen in 9 patients, all of whom were BRCA2 positive. The majority of these cancers were high grade, hormone receptor positive and associated with lymph node metastases. There were no breast cancer related deaths. Other cancers included bladder cancer, pancreatic cancer, melanoma and other skin cancers. Conclusions This study describes the cancer characteristics and outcomes of male BRCA1/2 mutation carriers. A third of male BRCA1/2 mutation carriers had a diagnosis of cancer. A significant number of patients (mostly BRCA2 mutation positive) developed multiple cancers, which may have important implications for cancer screening and prevention. Despite having high grade histology and advanced stage at diagnosis, male BRCA1/2 mutation carriers with breast and prostate cancer demonstrated a favorable 5-year survival.
Collapse
Affiliation(s)
- Mohammed Ibrahim
- Department of Hematology/Oncology, Beaumont Health, 3577 W 13 Mile Rd., Ste. 202a, Royal Oak, MI, 48073, USA. .,Oakland University William Beaumont School of Medicine, 2200 N Squirrel Rd, Rochester, MI, 48309, USA.
| | - Siddhartha Yadav
- Hematology-Oncology Fellowship Program, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Foluso Ogunleye
- Department of Hematology/Oncology, Beaumont Health, 3577 W 13 Mile Rd., Ste. 202a, Royal Oak, MI, 48073, USA.,Oakland University William Beaumont School of Medicine, 2200 N Squirrel Rd, Rochester, MI, 48309, USA
| | - Dana Zakalik
- Oakland University William Beaumont School of Medicine, 2200 N Squirrel Rd, Rochester, MI, 48309, USA.,Nancy and James Grosfeld Cancer Genetics Center, Beaumont Health, 3577 W 13 Mile Rd., Ste. 140, Royal Oak, MI, 48073, USA
| |
Collapse
|
46
|
Mikropoulos C, Selkirk CGH, Saya S, Bancroft E, Vertosick E, Dadaev T, Brendler C, Page E, Dias A, Evans DG, Rothwell J, Maehle L, Axcrona K, Richardson K, Eccles D, Jensen T, Osther PJ, van Asperen CJ, Vasen H, Kiemeney LA, Ringelberg J, Cybulski C, Wokolorczyk D, Hart R, Glover W, Lam J, Taylor L, Salinas M, Feliubadaló L, Oldenburg R, Cremers R, Verhaegh G, van Zelst-Stams WA, Oosterwijk JC, Cook J, Rosario DJ, Buys SS, Conner T, Domchek S, Powers J, Ausems MGEM, Teixeira MR, Maia S, Izatt L, Schmutzler R, Rhiem K, Foulkes WD, Boshari T, Davidson R, Ruijs M, Helderman-van den Enden ATJM, Andrews L, Walker L, Snape K, Henderson A, Jobson I, Lindeman GJ, Liljegren A, Harris M, Adank MA, Kirk J, Taylor A, Susman R, Chen-Shtoyerman R, Pachter N, Spigelman A, Side L, Zgajnar J, Mora J, Brewer C, Gadea N, Brady AF, Gallagher D, van Os T, Donaldson A, Stefansdottir V, Barwell J, James PA, Murphy D, Friedman E, Nicolai N, Greenhalgh L, Obeid E, Murthy V, Copakova L, McGrath J, Teo SH, Strom S, Kast K, Leongamornlert DA, Chamberlain A, Pope J, Newlin AC, Aaronson N, Ardern-Jones A, Bangma C, Castro E, Dearnaley D, Eyfjord J, Falconer A, Foster CS, Gronberg H, Hamdy FC, Johannsson O, Khoo V, Lubinski J, Grindedal EM, McKinley J, Shackleton K, Mitra AV, Moynihan C, Rennert G, Suri M, Tricker K, Moss S, Kote-Jarai Z, Vickers A, Lilja H, Helfand BT, Eeles RA. Prostate-specific antigen velocity in a prospective prostate cancer screening study of men with genetic predisposition. Br J Cancer 2018; 118:266-276. [PMID: 29301143 PMCID: PMC5785754 DOI: 10.1038/bjc.2017.429] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Prostate-specific antigen (PSA) and PSA-velocity (PSAV) have been used to identify men at risk of prostate cancer (PrCa). The IMPACT study is evaluating PSA screening in men with a known genetic predisposition to PrCa due to BRCA1/2 mutations. This analysis evaluates the utility of PSA and PSAV for identifying PrCa and high-grade disease in this cohort. METHODS PSAV was calculated using logistic regression to determine if PSA or PSAV predicted the result of prostate biopsy (PB) in men with elevated PSA values. Cox regression was used to determine whether PSA or PSAV predicted PSA elevation in men with low PSAs. Interaction terms were included in the models to determine whether BRCA status influenced the predictiveness of PSA or PSAV. RESULTS 1634 participants had ⩾3 PSA readings of whom 174 underwent PB and 45 PrCas diagnosed. In men with PSA >3.0 ng ml-l, PSAV was not significantly associated with presence of cancer or high-grade disease. PSAV did not add to PSA for predicting time to an elevated PSA. When comparing BRCA1/2 carriers to non-carriers, we found a significant interaction between BRCA status and last PSA before biopsy (P=0.031) and BRCA2 status and PSAV (P=0.024). However, PSAV was not predictive of biopsy outcome in BRCA2 carriers. CONCLUSIONS PSA is more strongly predictive of PrCa in BRCA carriers than non-carriers. We did not find evidence that PSAV aids decision-making for BRCA carriers over absolute PSA value alone.
Collapse
Affiliation(s)
| | - Christina G Hutten Selkirk
- The John and Carol Walter Center for Urological Health, Department of Surgery, North Shore University Health System, Evanston, IL 60201, USA
- Center for Medical Genetics, Department of Medicine, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Sibel Saya
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Elizabeth Bancroft
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham Rd, London SW3 6JJ, UK
| | - Emily Vertosick
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tokhir Dadaev
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Charles Brendler
- The John and Carol Walter Center for Urological Health, Department of Surgery, North Shore University Health System, Evanston, IL 60201, USA
| | - Elizabeth Page
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Alexander Dias
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham Rd, London SW3 6JJ, UK
| | - D Gareth Evans
- Genomic Medicine, Manchester Academic Health Sciences Centre, Division of Evolution and Genomic Sciences, University of Manchester, Central Manchester University Hospitals NHS Foundation Trust, Manchester M13 9WL, UK
| | - Jeanette Rothwell
- Genomic Medicine, Manchester Academic Health Sciences Centre, Division of Evolution and Genomic Sciences, University of Manchester, Central Manchester University Hospitals NHS Foundation Trust, Manchester M13 9WL, UK
| | - Lovise Maehle
- Department of Medical Genetics, Oslo University Hospital, Oslo 0372, Norway
| | - Karol Axcrona
- Akershus University Hospital, Lørenskog 1478, Norway
| | - Kate Richardson
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, East Melbourne, VIC 3000, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC 3010, Australia
| | - Diana Eccles
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton SO16 5YA, UK
- Cancer Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Thomas Jensen
- Department of Clinical Genetics, Vejle Hospital, Vejle 7100, Denmark
| | - Palle J Osther
- Department of Clinical Genetics, Vejle Hospital, Vejle 7100, Denmark
| | - Christi J van Asperen
- Leiden University Medical Center, Department of Clinical Genetics, Leiden, ZA 2333, The Netherlands
| | - Hans Vasen
- Netherlands Foundation for the Detection of Hereditary Tumors, Leiden, ZA 2333, The Netherlands
| | | | - Janneke Ringelberg
- Netherlands Foundation for the Detection of Hereditary Tumors, Leiden, ZA 2333, The Netherlands
| | - Cezary Cybulski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin 70-204, Poland
| | - Dominika Wokolorczyk
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin 70-204, Poland
| | - Rachel Hart
- Clinical Genetics Unit, Birmingham Women's Hospital, Birmingham B15 2TG, UK
| | - Wayne Glover
- Clinical Genetics Unit, Birmingham Women's Hospital, Birmingham B15 2TG, UK
| | - Jimmy Lam
- Department of Urology, Repatriation General Hospital, Daw Park, SA 5041, Australia
| | - Louise Taylor
- Department of Urology, Repatriation General Hospital, Daw Park, SA 5041, Australia
| | - Monica Salinas
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO-IDIBELL, CIBERONC), L’Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Lidia Feliubadaló
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO-IDIBELL, CIBERONC), L’Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Rogier Oldenburg
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam 3015 CE, The Netherlands
| | - Ruben Cremers
- Radboud University Medical Center, Nijmegen, GA 6525, The Netherlands
| | - Gerald Verhaegh
- Radboud University Medical Center, Nijmegen, GA 6525, The Netherlands
| | - Wendy A van Zelst-Stams
- Netherlands Foundation for the Detection of Hereditary Tumors, Leiden, ZA 2333, The Netherlands
| | - Jan C Oosterwijk
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
| | - Jackie Cook
- Sheffield Clinical Genetics Service, Sheffield Children's Hospital, Sheffield S10 2TH, UK
| | | | - Saundra S Buys
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT 84103, USA
| | - Tom Conner
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT 84103, USA
| | - Susan Domchek
- Basser Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jacquelyn Powers
- Basser Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Margreet GEM Ausems
- Department of Genetics, University Medical Centre Utrecht, Utrecht, CX, The Netherlands
| | - Manuel R Teixeira
- Genetics Department and Research Center, Portuguese Oncology Institute, Porto 4200-072, Portugal
- Biomedical Sciences Institute (ICBAS), Porto University, Porto 4200-072, Portugal
| | - Sofia Maia
- Genetics Department and Research Center, Portuguese Oncology Institute, Porto 4200-072, Portugal
| | - Louise Izatt
- South East Thames Genetics Service, Guy’s Hospital, London SE1 9RT, UK
| | - Rita Schmutzler
- Center of Familial Breast and Ovarian Cancer, University Hospital of Cologne, Cologne 50937, Germany
| | - Kerstin Rhiem
- Center of Familial Breast and Ovarian Cancer, University Hospital of Cologne, Cologne 50937, Germany
| | - William D Foulkes
- McGill Program in Cancer Genetics, Departments of Oncology and Human Genetics, McGill University, Montreal, QC H3A 0G4, Canada
| | - Talia Boshari
- McGill Program in Cancer Genetics, Departments of Oncology and Human Genetics, McGill University, Montreal, QC H3A 0G4, Canada
| | - Rosemarie Davidson
- Duncan Guthrie Institute of Medical Genetics, Yorkhill NHS Trust, Glasgow G38SJ, UK
| | - Marielle Ruijs
- The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | | | - Lesley Andrews
- Hereditary Cancer Clinic, Prince of Wales Hospital, Randwick, NSW 2031, Australia
| | - Lisa Walker
- Churchill Hospital, Headington, Oxford OX3 7LE, UK
| | - Katie Snape
- St George’s Hospital, Tooting, London SW17 0QT, UK
| | - Alex Henderson
- Northern Genetics Service, Newcastle upon Tyne Hospitals, Newcastle NE1 3BZ, UK
| | - Irene Jobson
- Northern Genetics Service, Newcastle upon Tyne Hospitals, Newcastle NE1 3BZ, UK
| | - Geoffrey J Lindeman
- Parkville Familial Cancer Centre, The Royal Melbourne Hospital, Grattan St, Parkville, VIC 3050, Australia
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3050, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC 3050, Australia
| | - Annelie Liljegren
- Karolinska University Hospital and Karolinska Institutet, Solna 171 77, Sweden
| | - Marion Harris
- Familial Cancer Centre, Monash Health, Clayton, VIC 3168, Australia
| | - Muriel A Adank
- VU University Medical Center, Amsterdam 1081 HV, The Netherlands
| | - Judy Kirk
- Familial Cancer Service, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, Sydney, NSW 2155, Australia
| | - Amy Taylor
- Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Rachel Susman
- Genetic Health Queensland, Royal Brisbane & Women's Hospital, Herston, QLD 4029, Australia
| | | | - Nicholas Pachter
- Genetic Services of WA, King Edward Memorial Hospital, Subiaco, WA 6008, Australia
- School of Medicine and Pharmacology, University of Western Australia, Perth, WA 6009, Australia
| | - Allan Spigelman
- Hunter Family Cancer Service, Waratah, NSW 2298, Australia
- University of New South Wales, St Vincent’s Clinical School, NSW 2052, Australia
- The Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, NSW 2010, Australia
| | - Lucy Side
- NE Thames Regional Genetics Service, Great Ormond St Hospital & UCL Institute of Women’s Health, London WC1N 3JH, UK
| | | | | | - Carole Brewer
- Peninsular Genetics, Derriford Hospital, Plymouth PL6 8DH, UK
- Royal Devon and Exeter Hospital, Exeter EX2 5DW, UK
| | - Neus Gadea
- High Risk and Cancer Prevention Clinic, Vall d'Hebron University Hospital, Barcelona 08035, Spain
| | - Angela F Brady
- North West Thames Regional Genetics Service, London North West Healthcare NHS Trust, London HA1 3UJ, UK
| | | | - Theo van Os
- Academic Medical Center, Amsterdam 1105 AZ, The Netherlands
| | | | | | - Julian Barwell
- University of Leicester, Leicester LE1 7RH, UK
- University Hospitals Leicester, Leicester LE1 5WW, UK
| | - Paul A James
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, East Melbourne, VIC 3000, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC 3010, Australia
- Genetic Medicine, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia
| | - Declan Murphy
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC 3010, Australia
| | - Eitan Friedman
- Chaim Sheba Medical Center, Tel-Hashomer 52621, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | | | - Lynn Greenhalgh
- Cheshire and Mersey Clinical Genetics Service, Liverpool Women’s Hospital, Liverpool L8 7SS, UK
| | - Elias Obeid
- Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Vedang Murthy
- Tata Memorial Centre, Mumbai, Maharashtra 400012, India
| | - Lucia Copakova
- National Cancer Institute, Bratislava 83310, Slovak Republic
| | - John McGrath
- Royal Devon and Exeter Hospital, Exeter EX2 5DW, UK
| | - Soo-Hwang Teo
- Cancer Research Initiatives Foundation, Subang Jaya Medical Centre, Subang Jaya, Selangor 47500, Darul Ehsan, Malaysia
| | - Sara Strom
- The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Karin Kast
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01069, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden 01307, Germany
- German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | | | - Anthony Chamberlain
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Jenny Pope
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Anna C Newlin
- Center for Medical Genetics, Department of Medicine, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Neil Aaronson
- The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | | | - Chris Bangma
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam 3015 CE, The Netherlands
| | - Elena Castro
- Prostate Cancer Unit, Spanish National Cancer Research Centre, Madrid 28029, Spain
| | - David Dearnaley
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham Rd, London SW3 6JJ, UK
| | - Jorunn Eyfjord
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik 101, Iceland
| | - Alison Falconer
- Imperial College Healthcare NHS Trust, London, London W2 1NY, UK
| | | | | | - Freddie C Hamdy
- Churchill Hospital, Headington, Oxford OX3 7LE, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX1 2JD, UK
| | - Oskar Johannsson
- Landspitali—the National University Hospital of Iceland, Reykjavik 101, Iceland
| | - Vincent Khoo
- Royal Marsden NHS Foundation Trust, Fulham Rd, London SW3 6JJ, UK
| | - Jan Lubinski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin 70-204, Poland
| | | | - Joanne McKinley
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, East Melbourne, VIC 3000, Australia
| | - Kylie Shackleton
- Parkville Familial Cancer Centre, The Royal Melbourne Hospital, Grattan St, Parkville, VIC 3050, Australia
| | - Anita V Mitra
- University College London Hospitals NHS Foundation Trust, London NW1 2BU, UK
| | - Clare Moynihan
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Gad Rennert
- CHS National Cancer Control Center, Carmel Medical Center, Haifa 3436212, Israel
| | - Mohnish Suri
- Nottingham City Hospital, Nottingham NG5 1PB, UK
| | - Karen Tricker
- Genomic Medicine, Manchester Academic Health Sciences Centre, Division of Evolution and Genomic Sciences, University of Manchester, Central Manchester University Hospitals NHS Foundation Trust, Manchester M13 9WL, UK
| | - The IMPACT study collaborators91
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- The John and Carol Walter Center for Urological Health, Department of Surgery, North Shore University Health System, Evanston, IL 60201, USA
- Center for Medical Genetics, Department of Medicine, NorthShore University HealthSystem, Evanston, IL 60201, USA
- Royal Marsden NHS Foundation Trust, Fulham Rd, London SW3 6JJ, UK
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Genomic Medicine, Manchester Academic Health Sciences Centre, Division of Evolution and Genomic Sciences, University of Manchester, Central Manchester University Hospitals NHS Foundation Trust, Manchester M13 9WL, UK
- Department of Medical Genetics, Oslo University Hospital, Oslo 0372, Norway
- Akershus University Hospital, Lørenskog 1478, Norway
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, East Melbourne, VIC 3000, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC 3010, Australia
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton SO16 5YA, UK
- Cancer Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
- Department of Clinical Genetics, Vejle Hospital, Vejle 7100, Denmark
- Leiden University Medical Center, Department of Clinical Genetics, Leiden, ZA 2333, The Netherlands
- Netherlands Foundation for the Detection of Hereditary Tumors, Leiden, ZA 2333, The Netherlands
- Radboud University Medical Center, Nijmegen, GA 6525, The Netherlands
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin 70-204, Poland
- Clinical Genetics Unit, Birmingham Women's Hospital, Birmingham B15 2TG, UK
- Department of Urology, Repatriation General Hospital, Daw Park, SA 5041, Australia
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO-IDIBELL, CIBERONC), L’Hospitalet de Llobregat, Barcelona 08908, Spain
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam 3015 CE, The Netherlands
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen 9713 GZ, The Netherlands
- Sheffield Clinical Genetics Service, Sheffield Children's Hospital, Sheffield S10 2TH, UK
- Royal Hallamshire Hospital, Sheffield S10 2JF, UK
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT 84103, USA
- Basser Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, University Medical Centre Utrecht, Utrecht, CX, The Netherlands
- Genetics Department and Research Center, Portuguese Oncology Institute, Porto 4200-072, Portugal
- Biomedical Sciences Institute (ICBAS), Porto University, Porto 4200-072, Portugal
- South East Thames Genetics Service, Guy’s Hospital, London SE1 9RT, UK
- Center of Familial Breast and Ovarian Cancer, University Hospital of Cologne, Cologne 50937, Germany
- McGill Program in Cancer Genetics, Departments of Oncology and Human Genetics, McGill University, Montreal, QC H3A 0G4, Canada
- Duncan Guthrie Institute of Medical Genetics, Yorkhill NHS Trust, Glasgow G38SJ, UK
- The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, HX 6229, The Netherlands
- Hereditary Cancer Clinic, Prince of Wales Hospital, Randwick, NSW 2031, Australia
- Churchill Hospital, Headington, Oxford OX3 7LE, UK
- St George’s Hospital, Tooting, London SW17 0QT, UK
- Northern Genetics Service, Newcastle upon Tyne Hospitals, Newcastle NE1 3BZ, UK
- Parkville Familial Cancer Centre, The Royal Melbourne Hospital, Grattan St, Parkville, VIC 3050, Australia
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3050, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC 3050, Australia
- Karolinska University Hospital and Karolinska Institutet, Solna 171 77, Sweden
- Familial Cancer Centre, Monash Health, Clayton, VIC 3168, Australia
- VU University Medical Center, Amsterdam 1081 HV, The Netherlands
- Familial Cancer Service, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, Sydney, NSW 2155, Australia
- Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
- Genetic Health Queensland, Royal Brisbane & Women's Hospital, Herston, QLD 4029, Australia
- The Genetic Institute, Kaplan Medical Center, Rehovot 76100, Israel
- Genetic Services of WA, King Edward Memorial Hospital, Subiaco, WA 6008, Australia
- School of Medicine and Pharmacology, University of Western Australia, Perth, WA 6009, Australia
- Hunter Family Cancer Service, Waratah, NSW 2298, Australia
- University of New South Wales, St Vincent’s Clinical School, NSW 2052, Australia
- The Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, NSW 2010, Australia
- NE Thames Regional Genetics Service, Great Ormond St Hospital & UCL Institute of Women’s Health, London WC1N 3JH, UK
- Institute of Oncology, Ljubljana 1000, Slovenia
- Hospital de Sant Pau, Barcelona 08041, Spain
- Peninsular Genetics, Derriford Hospital, Plymouth PL6 8DH, UK
- Royal Devon and Exeter Hospital, Exeter EX2 5DW, UK
- High Risk and Cancer Prevention Clinic, Vall d'Hebron University Hospital, Barcelona 08035, Spain
- North West Thames Regional Genetics Service, London North West Healthcare NHS Trust, London HA1 3UJ, UK
- St James’ Hospital, Dublin 8, Ireland
- Academic Medical Center, Amsterdam 1105 AZ, The Netherlands
- St Michael’s Hospital, Bristol BS2 8EG, UK
- Landspitali—the National University Hospital of Iceland, Reykjavik 101, Iceland
- University of Leicester, Leicester LE1 7RH, UK
- University Hospitals Leicester, Leicester LE1 5WW, UK
- Genetic Medicine, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia
- Chaim Sheba Medical Center, Tel-Hashomer 52621, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
- Istituto Nazionale dei Tumori, Milano 20133, Italy
- Cheshire and Mersey Clinical Genetics Service, Liverpool Women’s Hospital, Liverpool L8 7SS, UK
- Fox Chase Cancer Center, Philadelphia, PA 19111, USA
- Tata Memorial Centre, Mumbai, Maharashtra 400012, India
- National Cancer Institute, Bratislava 83310, Slovak Republic
- Cancer Research Initiatives Foundation, Subang Jaya Medical Centre, Subang Jaya, Selangor 47500, Darul Ehsan, Malaysia
- The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01069, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden 01307, Germany
- German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- Prostate Cancer Unit, Spanish National Cancer Research Centre, Madrid 28029, Spain
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik 101, Iceland
- Imperial College Healthcare NHS Trust, London, London W2 1NY, UK
- HCA Healthcare Laboratories, London WC1E 6JA, UK
- University Hospital, Umea 907 37, Sweden
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX1 2JD, UK
- University College London Hospitals NHS Foundation Trust, London NW1 2BU, UK
- CHS National Cancer Control Center, Carmel Medical Center, Haifa 3436212, Israel
- Nottingham City Hospital, Nottingham NG5 1PB, UK
- The IMPACT Study Collaborators List see Appendix 1
- Centre for Cancer Prevention, Queen Mary University of London, London EC1M 6BQ
- Departments of Laboratory Medicine, Surgery, and Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Translational Medicine, Lund University, Malmö 205 02, Sweden
| | - Sue Moss
- Centre for Cancer Prevention, Queen Mary University of London, London EC1M 6BQ
| | - Zsofia Kote-Jarai
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
| | - Andrew Vickers
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hans Lilja
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX1 2JD, UK
- Departments of Laboratory Medicine, Surgery, and Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Brian T Helfand
- The John and Carol Walter Center for Urological Health, Department of Surgery, North Shore University Health System, Evanston, IL 60201, USA
| | - Rosalind A Eeles
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey SM2 5NG, UK
- Royal Marsden NHS Foundation Trust, Fulham Rd, London SW3 6JJ, UK
| |
Collapse
|
47
|
Is there a difference in testosterone levels and its regulators in men carrying BRCA mutations? Oncotarget 2017; 8:103843-103850. [PMID: 29262604 PMCID: PMC5732770 DOI: 10.18632/oncotarget.21802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 09/16/2017] [Indexed: 11/25/2022] Open
Abstract
Background Male BRCA mutation carriers are at risk for an early onset aggressive prostate cancer. No data exist on the association of testosterone levels among these patients. We aimed to analyze testosterone and associated hormonal levels among male BRCA carriers and non-carriers. Patients and methods Overall 87 male carriers and 43 non-carriers aged 40-70 were prospectively enrolled. Clinical data were collected and all patients were tested for total testosterone (TT), prostate specific antigen (PSA), follicle stimulating hormone (FSH), luteinizing hormone (LH), free androgen index (FAI), sex hormone binding globulin (SHBG) and prolactin. Multivariate linear regression analysis was performed to predict TT levels. Results The median age, mean BMI, comorbidities, PSA, FSH, LH and SHBG levels in both groups were similar. However, mean TT and FAI were higher in the carriers (16.7 nmol/l vs 13.5 nmol/l, p=0.03 and 39.5 vs 34.8, p=0.05, respectively), while prolactin was significantly lower. Multivariate analysis demonstrated that while BMI was inversely correlated to TT levels in both groups, LH was a predictor only in non-carriers. Conclusions Carriers have higher TT and FAI levels and lower prolactin levels; but LH does not predict their TT levels. Further research in a larger cohort of BRCA carriers with and without prostate cancer should be performed.
Collapse
|
48
|
Abstract
Mutations in BRCA1 or BRCA2 define a subset of prostate cancer patients. Herein, we address the question whether BRCA1/2 mutations have a predictive impact on chemotherapy with docetaxel, a widely used drug in patients with metastatic castration resistant prostate cancer (mCRPC). Fifty-three men treated with docetaxel for mCRPC were tested for somatic BRCA1/2 mutations of the primary tumor. In a subgroup of patients, BRCA1/2 protein expression was tested as a potential surrogate marker for BRCA1/2 inactivation. Eight of 53 patients (15.1%) harbored a deleterious BRCA2 mutation. No BRCA1 mutation was found. Patients with a BRCA2 mutation showed a response rate of 25% to docetaxel in comparison to 71.1% in men with wildtype BRCA2 (p = 0.019). While the time to develop castration resistance was similar in both subgroups, the overall survival was significantly shorter in patients harboring a BRCA2 mutation. No correlation between the BRCA1/2 protein expression and the response to docetaxel was found. While the presence of a BRCA2 mutation does not preclude a response to docetaxel, there is overall a significant correlation between BRCA2 inactivation and a poor response rate. Our results suggest that a close oncological monitoring of patients with BRCA2 mutations for taxane resistance is warranted.
Collapse
|
49
|
Goldberg H, Grievink LS, Mano R, Ber Y, Ozalbo R, Tuval S, Baniel J, Margel D. Lower Urinary Tract Symptoms and Benign Prostate Hyperplasia Features Among Male BRCA Mutation Carriers. Urology 2017; 108:71-75. [PMID: 28577930 DOI: 10.1016/j.urology.2017.05.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/08/2017] [Accepted: 05/10/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To analyze lower urinary tract symptoms and benign prostate hyperplasia features among male BRCA1 and 2 carriers and an age-matched control group. METHODS Male BRCA carriers and noncarriers aged 40-70 years were enrolled in our cross-sectional study. Relevant clinical data were collected, and patients filled the International Prostate Symptom Score. Patients also underwent prostate-specific antigen (PSA) blood testing, digital rectal examination, uroflowmetry, and post-void residual ultrasound examination. As part of their routine follow-up, BRCA carriers underwent prostate magnetic resonance imaging. RESULTS Overall, 87 carriers and 30 noncarriers were enrolled. The median age, mean body mass index, and comorbidities in both groups were similar. Maximal flow (QMAX) was higher in the noncarrier group (16.9 mL/s vs 12 mL/s, P = .01). Mean prostate volume among all BRCA carriers was 38.8 cc (19.7), but BRCA1 patients had larger glands with higher mean PSA and PSA density than BRCA2 patients (41.8 cc vs 33.1 cc, P = .047, 1.84 ng/mL vs 1.07 ng/mL, P = .006, and .044 vs .032, P = .042, respectively). Multivariate analysis demonstrated age being the sole significant predictor of PSA density in BRCA2 patients. CONCLUSION Male carrying BRCA mutations have significantly lower QMAX than healthy men. BRCA1 patients have on average larger prostate glands and higher PSA than BRCA2 patients. Further research is required to decipher the association of lower urinary tract symptoms or benign prostate hyperplasia with BRCA carriers.
Collapse
Affiliation(s)
- Hanan Goldberg
- Institute of Urology, Rabin Medical Center, Petah Tikva; and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Liat Shavit Grievink
- Institute of Urology, Rabin Medical Center, Petah Tikva; and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Roy Mano
- Institute of Urology, Rabin Medical Center, Petah Tikva; and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yaara Ber
- Institute of Urology, Rabin Medical Center, Petah Tikva; and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rachely Ozalbo
- Institute of Urology, Rabin Medical Center, Petah Tikva; and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sivan Tuval
- Institute of Urology, Rabin Medical Center, Petah Tikva; and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jack Baniel
- Institute of Urology, Rabin Medical Center, Petah Tikva; and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - David Margel
- Institute of Urology, Rabin Medical Center, Petah Tikva; and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
50
|
Are the Pathological Characteristics of Prostate Cancer More Aggressive or More Indolent Depending upon the Patient Age? BIOMED RESEARCH INTERNATIONAL 2017; 2017:1438027. [PMID: 28265568 PMCID: PMC5318620 DOI: 10.1155/2017/1438027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 01/07/2017] [Accepted: 01/12/2017] [Indexed: 12/16/2022]
Abstract
Purpose. To identify pathological characteristics of prostate cancer according to patient age at diagnosis. Methods. A retrospective review of 2,929 men diagnosed with prostate cancer was performed. Pathological characteristics were compared across age groups: ≤55, 56–75, and >75 years. Results. The study cohort included 133 patients (4.5%), 2,033 patients (69.5%), and 763 patients (26.0%) in the three age groups, respectively. The median pathological Gleason sums in the three age groups were 8, 7, and 8, respectively. The Gleason sum, primary Gleason score, and second primary Gleason score were significantly different among the three age groups (Z = 12.975, p = 0.002; Z = 9.264, p = 0.010; Z = 6.692, p = 0.035, resp.). The percentages of Gleason pattern 5 tumors for the three age groups were 44.4%, 32.3%, and 36.8%, respectively; they were significantly different (χ2 = 11.641, p = 0.003). The percentages of tumors with Gleason score grade groups 3–5 for the three age groups were 66.9%, 60.5%, and 66.3%, respectively; they were significantly different (χ2 = 9.401, p = 0.009). Conclusions. The present study indicated that men aged ≤55 years or >75 years show higher levels of clinically significant prostate cancer compared to patients between the ages of 55 and 75 years. Younger and more elderly male patients are more likely to have a more aggressive disease.
Collapse
|