101
|
Biller LH, Creedon SA, Klehm M, Yurgelun MB. Lynch Syndrome-Associated Cancers Beyond Colorectal Cancer. Gastrointest Endosc Clin N Am 2022; 32:75-93. [PMID: 34798988 DOI: 10.1016/j.giec.2021.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Lynch syndrome (LS) is a common form of inherited cancer susceptibility, which predisposes to colorectal cancer (CRC) along with a wide array of other extracolonic malignancies, including other gastrointestinal cancers, cancers of the gynecologic and genitourinary tracts, and other organ sites. Recent data have provided novel insights into patient-specific factors that can help clinicians understand an individual LS carrier's risk of extracolonic cancers, including sex, specific LS gene, age, family history of cancer, and other factors. This summary seeks to provide an update on extracolonic cancer risks in LS and provide recommendations for surveillance and risk reduction.
Collapse
Affiliation(s)
- Leah H Biller
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA; Brigham & Women's Hospital, Boston, MA 02215, USA
| | - Siobhan A Creedon
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Margaret Klehm
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Matthew B Yurgelun
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA; Brigham & Women's Hospital, Boston, MA 02215, USA.
| |
Collapse
|
102
|
Mollica V, Marchetti A, Rosellini M, Nuvola G, Rizzo A, Santoni M, Cimadamore A, Montironi R, Massari F. An Insight on Novel Molecular Pathways in Metastatic Prostate Cancer: A Focus on DDR, MSI and AKT. Int J Mol Sci 2021; 22:ijms222413519. [PMID: 34948314 PMCID: PMC8708596 DOI: 10.3390/ijms222413519] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/07/2021] [Accepted: 12/15/2021] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer is still one of the main causes of cancer-related death in the male population, regardless of the advancements in the treatment scenario. The genetic knowledge on prostate cancer is widely increasing, allowing researchers to identify novel promising molecular targets and treatment approaches. Genomic profiling has evidenced that DNA damage repair genes’ alterations are quite frequent in metastatic, castration resistant prostate cancer and specific therapies can interfere with this pathway, showing promising activity in this setting. Microsatellite instability is gaining attention as it seems to represent a predictive factor of the response to immunotherapy. Furthermore, the PTEN-PI3K-AKT pathway is another possible treatment target being investigated. In this review, we explore the current knowledge on these frequent genomic alterations of metastatic prostate cancer, their possible therapeutic repercussions and the promising future treatments under evaluation.
Collapse
Affiliation(s)
- Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni-15, 40138 Bologna, Italy; (V.M.); (A.M.); (M.R.); (G.N.); (A.R.); (F.M.)
| | - Andrea Marchetti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni-15, 40138 Bologna, Italy; (V.M.); (A.M.); (M.R.); (G.N.); (A.R.); (F.M.)
| | - Matteo Rosellini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni-15, 40138 Bologna, Italy; (V.M.); (A.M.); (M.R.); (G.N.); (A.R.); (F.M.)
| | - Giacomo Nuvola
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni-15, 40138 Bologna, Italy; (V.M.); (A.M.); (M.R.); (G.N.); (A.R.); (F.M.)
| | - Alessandro Rizzo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni-15, 40138 Bologna, Italy; (V.M.); (A.M.); (M.R.); (G.N.); (A.R.); (F.M.)
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
| | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy
- Correspondence:
| | - Rodolfo Montironi
- Molecular Medicine and Cell Therapy Foundation, Department of Clinical and Molecular Sciences, Polytechnic University of the Marche Region, 60100 Ancona, Italy;
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni-15, 40138 Bologna, Italy; (V.M.); (A.M.); (M.R.); (G.N.); (A.R.); (F.M.)
| |
Collapse
|
103
|
Clark R, Kenk M, McAlpine K, Thain E, Farncombe KM, Pritchard CC, Nussbaum R, Wyatt AW, de Bono J, Vesprini D, Bombard Y, Lorentz J, Narod S, Kim R, Fleshner N. The evolving role of germline genetic testing and management in prostate cancer: Report from the Princess Margaret Cancer Centre international retreat. Can Urol Assoc J 2021; 15:E623-E629. [PMID: 34171218 PMCID: PMC8631832 DOI: 10.5489/cuaj.7383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Prostate cancer is a significant cause of cancer mortality. It has been well-established that certain germline pathogenic variants confer both an increased risk of being diagnosed with prostate cancer and dying of prostate cancer.1 There are exciting developments in both the availability of genetic testing and opportunities for improved treatment of patients.On August 19, 2020, the Princess Margaret Cancer Centre in Toronto, Ontario, hosted a virtual retreat, bringing together international experts in urology, medical oncology, radiation oncology, medical genetics, and translational research, as well as a patient representative. We are pleased to provide this manuscript as a review of those proceedings for Canadian clinicians.We highlighted several needs for future research and policy action based on this meeting:Increased access to funding for germline testing for the common genetic disorders associated with increased risk of prostate cancer.More research into identifying genetic factors influencing risk stratification, treatment response, and outcomes of prostate cancer within Canadian populations at higher genetic risk for prostate cancer.Added awareness about genetic risk factors among the Canadian public.Development of patient-specific and reported outcomes research in tailored care for patients at increased genetic risk of prostate cancer.Creation of multidisciplinary clinics that specialize in tailored care for patients at increased genetic risk of prostate cancer.
Collapse
Affiliation(s)
- Roderick Clark
- Division of Urology, University Health Network, Toronto, ON, Canada
| | - Miran Kenk
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Kristen McAlpine
- Division of Urology, University Health Network, Toronto, ON, Canada
| | - Emily Thain
- Familial Cancer Clinic, University Health Network, Toronto, ON, Canada
| | - Kirsten M. Farncombe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Colin C. Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | | | - Alexander W. Wyatt
- Department of Urological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Johann de Bono
- Institute of Cancer Research, Royal Marsden Hospital, London, United Kingdom
| | - Danny Vesprini
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Yvonne Bombard
- Li Ka Shing Knowledge Institute, St. Michaels Hospital, Toronto, ON, Canada
| | - Justin Lorentz
- Genetics and High Risk Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Steven Narod
- Familial Breast Cancer Research Unit, Women’s College Research Institute, Toronto, ON, Canada
| | - Raymond Kim
- Department of Medical Oncology, University Health Network, Toronto, ON, Canada
| | - Neil Fleshner
- Division of Urology, University Health Network, Toronto, ON, Canada
| |
Collapse
|
104
|
Gandaglia G, Leni R, Bray F, Fleshner N, Freedland SJ, Kibel A, Stattin P, Van Poppel H, La Vecchia C. Epidemiology and Prevention of Prostate Cancer. Eur Urol Oncol 2021; 4:877-892. [PMID: 34716119 DOI: 10.1016/j.euo.2021.09.006] [Citation(s) in RCA: 277] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/10/2021] [Accepted: 09/28/2021] [Indexed: 01/04/2023]
Abstract
CONTEXT Worldwide, prostate cancer (PCa) represents the second most common solid tumor in men. OBJECTIVE To assess the geographical distribution of PCa, epidemiological differences, and the most relevant risk factors for the disease. EVIDENCE ACQUISITION Estimated incidence, mortality, and prevalence of PCa for the year 2020 in 185 countries were derived from the IARC GLOBOCAN database. A review of English-language articles published between 2010 and 2020 was conducted using MEDLINE, EMBASE, and Scopus to identify risk factors for PCa. EVIDENCE SYNTHESIS In the year 2020, there were over 1414000 estimated new cases of PCa worldwide, with an age-standardized rate (ASR) incidence of 31 per 100000 (lifetime cumulative risk: 3.9%). Northern Europe has the highest all-age incidence ASR (83), while the lowest ASR was in South-Central Asia (6.3). In the year 2020, there were over 375000 estimated deaths worldwide, and the overall mortality ASR was 7.7 per 100000, with the highest ASR in the Caribbean (28) and the lowest in South-Central Asia (3.1). Family history, hereditary syndromes, and race are the strongest risk factors for PCa. Metabolic syndrome was associated with the risk of developing PCa, high-grade disease, and adverse pathology. Diabetes and exposure to ultraviolet rays were found to be inversely associated to PCa incidence. Cigarette smoking and obesity may increase PCa-specific mortality, while regular physical activity may reduce disease progression. Although 5-alpha reductase inhibitors are known to be associated with a reduced incidence of PCa, available studies failed to show an effect on overall mortality. CONCLUSIONS Family history, race, and hereditary syndromes are well-established risk factors for PCa. Modifiable risk factors may impact the risk of developing PCa and that of dying from the disease, but little evidence exist for any clear indication for prevention other than early diagnosis to reduce PCa mortality. PATIENT SUMMARY Prostate cancer (PCa) rates vary profoundly worldwide, with incidence and mortality rates being highest in Northern Europe and Caribbean, respectively. South-Central Asia has the lowest epidemiological burden. Family history, race, and hereditary syndromes are well-established risk factors for PCa. Modifiable risk factors may impact the risk of developing PCa and that of dying from the disease itself, but little evidence exist for any clear indication for prevention other than early diagnosis to reduce PCa mortality.
Collapse
Affiliation(s)
- Giorgio Gandaglia
- Unit of Urology/Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| | - Riccardo Leni
- Unit of Urology/Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Freddie Bray
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Neil Fleshner
- Division or Urology, University of Toronto, Toronto, Ontario, Canada
| | - Stephen J Freedland
- Division of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Section of Urology, Durham VA Medical Center, Durham, NC, USA
| | - Adam Kibel
- Division of Urological Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pär Stattin
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Hendrick Van Poppel
- Department of Urology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
105
|
Sokolova AO, Marshall CH, Lozano R, Gulati R, Ledet EM, De Sarkar N, Grivas P, Higano CS, Montgomery B, Nelson PS, Olmos D, Sokolov V, Schweizer MT, Yezefski TA, Yu EY, Paller CJ, Sartor O, Castro E, Antonarakis ES, Cheng HH. Efficacy of systemic therapies in men with metastatic castration resistant prostate cancer harboring germline ATM versus BRCA2 mutations. Prostate 2021; 81:1382-1389. [PMID: 34516663 PMCID: PMC8563438 DOI: 10.1002/pros.24236] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/30/2021] [Indexed: 11/11/2022]
Abstract
BACKGROUND Among men with metastatic prostate cancer, about 10% have germline alterations in DNA damage response genes. Most studies have examined BRCA2 alone or an aggregate of BRCA1/2 and ATM. Emerging data suggest that ATM mutations may have distinct biology and warrant individual evaluation. The objective of this study is to determine whether response to prostate cancer systemic therapies differs between men with germline mutations in ATM (gATM) and BRCA2 (gBRCA2). METHODS This is an international multicenter retrospective matched cohort study of men with prostate cancer harboring gATM or gBRCA2. PSA50 response (≥50% decline in prostate-specific antigen) was compared using Fisher's exact test. RESULTS AND LIMITATIONS The study included 45 gATM and 45 gBRCA2 patients, matched on stage and year of germline testing. Patients with gATM and gBRCA2 had similar age, Gleason grade, and PSA at diagnosis. We did not observe differences in PSA50 responses to abiraterone, enzalutamide, or docetaxel in metastatic castration resistant prostate cancer between the two groups; however, 0/7 with gATM and 12/14 with gBRCA2 achieved PSA50 response to PARPi (p < .001). Median (95% confidence interval) overall survival from diagnosis to death was 10.9 years (9.5-not reached) versus 9.9 years (7.1-not reached, p = .07) for the gATM and gBRCA2 cohorts, respectively. Limitations include the retrospective design and lack of mutation zygosity data. CONCLUSIONS Conventional therapies can be effective in gATM carriers and should be considered before PARPi, which shows limited efficacy in this group. Men with gATM mutations warrant prioritization for novel treatment strategies.
Collapse
Affiliation(s)
| | - Catherine H. Marshall
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rebeca Lozano
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Genitourinary Cancer Traslational Research Group, Instituto de Investigación Biomédica de Málaga, Malaga, Spain
| | - Roman Gulati
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | - Petros Grivas
- University of Washington, Department of Medicine, Division of Medical Oncology, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Celestia S. Higano
- University of Washington, Department of Medicine, Division of Medical Oncology, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Bruce Montgomery
- University of Washington, Department of Medicine, Division of Medical Oncology, Seattle, WA, USA
| | - Peter S. Nelson
- University of Washington, Department of Medicine, Division of Medical Oncology, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - David Olmos
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Genitourinary Cancer Traslational Research Group, Instituto de Investigación Biomédica de Málaga, Malaga, Spain
| | | | - Michael T. Schweizer
- University of Washington, Department of Medicine, Division of Medical Oncology, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Todd A. Yezefski
- University of Washington, Department of Medicine, Division of Medical Oncology, Seattle, WA, USA
| | - Evan Y. Yu
- University of Washington, Department of Medicine, Division of Medical Oncology, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Channing J. Paller
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Oliver Sartor
- Tulane University School of Medicine, New Orleans, LA, USA
| | - Elena Castro
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Genitourinary Cancer Traslational Research Group, Instituto de Investigación Biomédica de Málaga, Malaga, Spain
- Hospital Universitario Virgen de la Victoria y Regional de Málaga, Spain
| | - Emmanuel S. Antonarakis
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Heather H. Cheng
- University of Washington, Department of Medicine, Division of Medical Oncology, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
106
|
Benafif S, Ni Raghallaigh H, McHugh J, Eeles R. Genetics of prostate cancer and its utility in treatment and screening. ADVANCES IN GENETICS 2021; 108:147-199. [PMID: 34844712 DOI: 10.1016/bs.adgen.2021.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Prostate cancer heritability is attributed to a combination of rare, moderate to highly penetrant genetic variants as well as commonly occurring variants conferring modest risks [single nucleotide polymorphisms (SNPs)]. Some of the former type of variants (e.g., BRCA2 mutations) predispose particularly to aggressive prostate cancer and confer poorer prognoses compared to men who do not carry mutations. Molecularly targeted treatments such as PARP inhibitors have improved outcomes in men carrying somatic and/or germline DNA repair gene mutations. Ongoing clinical trials are exploring other molecular targeted approaches based on prostate cancer somatic alterations. Genome wide association studies have identified >250 loci that associate with prostate cancer risk. Multi-ancestry analyses have identified shared as well as population specific risk SNPs. Prostate cancer risk SNPs can be used to estimate a polygenic risk score (PRS) to determine an individual's genetic risk of prostate cancer. The odds ratio of prostate cancer development in men whose PRS lies in the top 1% of the risk profile ranges from 9 to 11. Ongoing studies are investigating the utility of a prostate cancer PRS to target population screening to those at highest risk. With the advent of personalized medicine and development of DNA sequencing technologies, access to clinical genetic testing is increasing, and oncology guidelines from bodies such as NCCN and ESMO have been updated to provide criteria for germline testing of "at risk" healthy men as well as those with prostate cancer. Both germline and somatic prostate cancer research have significantly evolved in the past decade and will lead to further development of precision medicine approaches to prostate cancer treatment as well as potentially developing precision population screening models.
Collapse
Affiliation(s)
- S Benafif
- The Institute of Cancer Research, London, United Kingdom.
| | | | - J McHugh
- The Institute of Cancer Research, London, United Kingdom
| | - R Eeles
- The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
107
|
Giri VN, Walker A, Gross L, Trabulsi EJ, Lallas CD, Kelly WK, Gomella LG, Fischer C, Loeb S. Helix: A Digital Tool to Address Provider Needs for Prostate Cancer Genetic Testing in Clinical Practice. Clin Genitourin Cancer 2021; 20:e104-e113. [PMID: 35012874 DOI: 10.1016/j.clgc.2021.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/21/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Prostate cancer (PCA) germline testing (GT) is now standard-of-care for men with advanced PCA. Thousands of men may consider GT due to clinical and family history (FH) features. Identifying and consenting men for GT can be complex. Here we identified barriers and facilitators of GT across a spectrum of providers which informed the development of Helix - an educational and clinical/FH collection tool to facilitate GT in practice. MATERIALS AND METHODS A 12-question survey assessing knowledge of genetics PCA risk and FH was administered December 2017 to March 2018 in the Philadelphia area and at the Mid-Atlantic AUA meeting (March 2018). Responses were analyzed using descriptive statistics. Semi-structured interviews were conducted with medical oncologists, radiation oncologists, and urologists across practice settings from March-October 2020 as part of a larger study based on the Tailored Implementation in Chronic Diseases framework. Helix was then developed followed by user testing. RESULTS Fifty-six providers (50% urologists) responded to the survey. Multiple FH and genetic knowledge gaps were identified: only 66% collected maternal FH and 43% correctly identified BRCA2 and association to aggressive PCA. Genetic counseling gaps included low rates of discussing genetic discrimination laws (45%). Provider interviews (n = 14) identified barriers to FH intake including access to details and time needed. In user testing (n = 10), providers found Helix helpful for FH collection. All providers found Helix easy to use, suggesting expanded clinical use. CONCLUSION Helix addressed multiple GT knowledge and practice gaps across a spectrum of providers. This tool will become publicly available soon to facilitate PCA GT in clinical practice.
Collapse
Affiliation(s)
- Veda N Giri
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA; Cancer Risk Assessment and Clinical Cancer Genetics Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA; Department of Urology, Thomas Jefferson University, Philadelphia, PA.
| | | | - Laura Gross
- Cancer Risk Assessment and Clinical Cancer Genetics Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | | - Costas D Lallas
- Department of Urology, Thomas Jefferson University, Philadelphia, PA
| | - William K Kelly
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Leonard G Gomella
- Department of Urology, Thomas Jefferson University, Philadelphia, PA
| | - Corey Fischer
- Jefferson Digital Innovation and Consumer Experience, Thomas Jefferson University, Philadelphia, PA
| | - Stacy Loeb
- NYU-Langone Health, New York, NY; Manhattan Veterans Affairs Hospital, New York, NY
| |
Collapse
|
108
|
Advances in urologic oncology "OncoUrology Forum Special Edition": The best of 2020. Actas Urol Esp 2021; 46:214-222. [PMID: 34844900 DOI: 10.1016/j.acuroe.2021.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/05/2021] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To provide latest findings of Urologic Oncology on prostate, kidney, and bladder cancer, and analyze its impact on clinical practice as well as future schemes in the medium- and long-term. METHODS This document reviews the abstracts on Uro-Oncology presented at the 2020 Congresses (EUA, AUA, ASCO, ESMO and ASTRO), the publications with the highest impact and especially the new lines of development and progress in Uro-Oncology evaluated by the OncoForum committee. RESULTS The use of prostate-specific membrane antigen (PSMA) radioligands in the diagnosis of prostate cancer may have great potential and utility in the coming years due to their improved sensitivity and specificity. The genetic characterization of the tumor is important at both, germline and somatic levels, due to the significant role of BRCA2 mutations regarding risk. The cohort multiple randomised controlled trial is the most suitable study design at the genitourinary cancer level. The application of big data will lead to process improvements, savings in healthcare costs, and an empowerment of real-life studies through ease of data comparison, management, and storage. CONCLUSIONS The use of new diagnostic techniques with PSMA ligands will provide a more comprehensive diagnostic modality, increase the number of studies about tumor genetic profiling, and enhance their quality. The practical application of artificial intelligence will improve the treatment genitourinary cancer.
Collapse
|
109
|
Shah S, Rachmat R, Enyioma S, Ghose A, Revythis A, Boussios S. BRCA Mutations in Prostate Cancer: Assessment, Implications and Treatment Considerations. Int J Mol Sci 2021; 22:12628. [PMID: 34884434 PMCID: PMC8657599 DOI: 10.3390/ijms222312628] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/20/2021] [Accepted: 11/21/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer ranks fifth in cancer-related mortality in men worldwide. DNA damage is implicated in cancer and DNA damage response (DDR) pathways are in place against this to maintain genomic stability. Impaired DDR pathways play a role in prostate carcinogenesis and germline or somatic mutations in DDR genes have been found in both primary and metastatic prostate cancer. Among these, BRCA mutations have been found to be especially clinically relevant with a role for germline or somatic testing. Prostate cancer with DDR defects may be sensitive to poly(ADP-ribose) polymerase (PARP) inhibitors which target proteins in a process called PARylation. Initially they were used to target BRCA-mutated tumor cells in a process of synthetic lethality. However, recent studies have found potential for PARP inhibitors in a variety of other genetic settings. In this review, we explore the mechanisms of DNA repair, potential for genomic analysis of prostate cancer and therapeutics of PARP inhibitors along with their safety profile.
Collapse
Affiliation(s)
- Sidrah Shah
- Department of Palliative Care, Guy’s and St Thomas’ Hospital, Great Maze Pond, London SE1 9RT, UK;
| | - Rachelle Rachmat
- Department of Radiology, Guy’s and St Thomas’ Hospital, Great Maze Pond, London SE1 9RT, UK;
| | - Synthia Enyioma
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK; (S.E.); (A.R.)
| | - Aruni Ghose
- Department of Medical Oncology, Barts Cancer Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, W Smithfield, London EC1A 7BE, UK;
- Faculty of Life Sciences & Medicine, King’s College London, London WC2R 2LS, UK
| | - Antonios Revythis
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK; (S.E.); (A.R.)
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK; (S.E.); (A.R.)
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 9RT, UK
- AELIA Organization, 9th Km Thessaloniki-Thermi, 57001 Thessaloniki, Greece
| |
Collapse
|
110
|
Bancroft EK, Page EC, Brook MN, Thomas S, Taylor N, Pope J, McHugh J, Jones AB, Karlsson Q, Merson S, Ong KR, Hoffman J, Huber C, Maehle L, Grindedal EM, Stormorken A, Evans DG, Rothwell J, Lalloo F, Brady AF, Bartlett M, Snape K, Hanson H, James P, McKinley J, Mascarenhas L, Syngal S, Ukaegbu C, Side L, Thomas T, Barwell J, Teixeira MR, Izatt L, Suri M, Macrae FA, Poplawski N, Chen-Shtoyerman R, Ahmed M, Musgrave H, Nicolai N, Greenhalgh L, Brewer C, Pachter N, Spigelman AD, Azzabi A, Helfand BT, Halliday D, Buys S, Ramon Y Cajal T, Donaldson A, Cooney KA, Harris M, McGrath J, Davidson R, Taylor A, Cooke P, Myhill K, Hogben M, Aaronson NK, Ardern-Jones A, Bangma CH, Castro E, Dearnaley D, Dias A, Dudderidge T, Eccles DM, Green K, Eyfjord J, Falconer A, Foster CS, Gronberg H, Hamdy FC, Johannsson O, Khoo V, Lilja H, Lindeman GJ, Lubinski J, Axcrona K, Mikropoulos C, Mitra AV, Moynihan C, Ni Raghallaigh H, Rennert G, Collier R, Offman J, Kote-Jarai Z, Eeles RA. A prospective prostate cancer screening programme for men with pathogenic variants in mismatch repair genes (IMPACT): initial results from an international prospective study. Lancet Oncol 2021; 22:1618-1631. [PMID: 34678156 PMCID: PMC8576477 DOI: 10.1016/s1470-2045(21)00522-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/19/2021] [Accepted: 08/27/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Lynch syndrome is a rare familial cancer syndrome caused by pathogenic variants in the mismatch repair genes MLH1, MSH2, MSH6, or PMS2, that cause predisposition to various cancers, predominantly colorectal and endometrial cancer. Data are emerging that pathogenic variants in mismatch repair genes increase the risk of early-onset aggressive prostate cancer. The IMPACT study is prospectively assessing prostate-specific antigen (PSA) screening in men with germline mismatch repair pathogenic variants. Here, we report the usefulness of PSA screening, prostate cancer incidence, and tumour characteristics after the first screening round in men with and without these germline pathogenic variants. METHODS The IMPACT study is an international, prospective study. Men aged 40-69 years without a previous prostate cancer diagnosis and with a known germline pathogenic variant in the MLH1, MSH2, or MSH6 gene, and age-matched male controls who tested negative for a familial pathogenic variant in these genes were recruited from 34 genetic and urology clinics in eight countries, and underwent a baseline PSA screening. Men who had a PSA level higher than 3·0 ng/mL were offered a transrectal, ultrasound-guided, prostate biopsy and a histopathological analysis was done. All participants are undergoing a minimum of 5 years' annual screening. The primary endpoint was to determine the incidence, stage, and pathology of screening-detected prostate cancer in carriers of pathogenic variants compared with non-carrier controls. We used Fisher's exact test to compare the number of cases, cancer incidence, and positive predictive values of the PSA cutoff and biopsy between carriers and non-carriers and the differences between disease types (ie, cancer vs no cancer, clinically significant cancer vs no cancer). We assessed screening outcomes and tumour characteristics by pathogenic variant status. Here we present results from the first round of PSA screening in the IMPACT study. This study is registered with ClinicalTrials.gov, NCT00261456, and is now closed to accrual. FINDINGS Between Sept 28, 2012, and March 1, 2020, 828 men were recruited (644 carriers of mismatch repair pathogenic variants [204 carriers of MLH1, 305 carriers of MSH2, and 135 carriers of MSH6] and 184 non-carrier controls [65 non-carriers of MLH1, 76 non-carriers of MSH2, and 43 non-carriers of MSH6]), and in order to boost the sample size for the non-carrier control groups, we randomly selected 134 non-carriers from the BRCA1 and BRCA2 cohort of the IMPACT study, who were included in all three non-carrier cohorts. Men were predominantly of European ancestry (899 [93%] of 953 with available data), with a mean age of 52·8 years (SD 8·3). Within the first screening round, 56 (6%) men had a PSA concentration of more than 3·0 ng/mL and 35 (4%) biopsies were done. The overall incidence of prostate cancer was 1·9% (18 of 962; 95% CI 1·1-2·9). The incidence among MSH2 carriers was 4·3% (13 of 305; 95% CI 2·3-7·2), MSH2 non-carrier controls was 0·5% (one of 210; 0·0-2·6), MSH6 carriers was 3·0% (four of 135; 0·8-7·4), and none were detected among the MLH1 carriers, MLH1 non-carrier controls, and MSH6 non-carrier controls. Prostate cancer incidence, using a PSA threshold of higher than 3·0 ng/mL, was higher in MSH2 carriers than in MSH2 non-carrier controls (4·3% vs 0·5%; p=0·011) and MSH6 carriers than MSH6 non-carrier controls (3·0% vs 0%; p=0·034). The overall positive predictive value of biopsy using a PSA threshold of 3·0 ng/mL was 51·4% (95% CI 34·0-68·6), and the overall positive predictive value of a PSA threshold of 3·0 ng/mL was 32·1% (20·3-46·0). INTERPRETATION After the first screening round, carriers of MSH2 and MSH6 pathogenic variants had a higher incidence of prostate cancer compared with age-matched non-carrier controls. These findings support the use of targeted PSA screening in these men to identify those with clinically significant prostate cancer. Further annual screening rounds will need to confirm these findings. FUNDING Cancer Research UK, The Ronald and Rita McAulay Foundation, the National Institute for Health Research support to Biomedical Research Centres (The Institute of Cancer Research and Royal Marsden NHS Foundation Trust; Oxford; Manchester and the Cambridge Clinical Research Centre), Mr and Mrs Jack Baker, the Cancer Council of Tasmania, Cancer Australia, Prostate Cancer Foundation of Australia, Cancer Council of Victoria, Cancer Council of South Australia, the Victorian Cancer Agency, Cancer Australia, Prostate Cancer Foundation of Australia, Asociación Española Contra el Cáncer (AECC), the Instituto de Salud Carlos III, Fondo Europeo de Desarrollo Regional (FEDER), the Institut Català de la Salut, Autonomous Government of Catalonia, Fundação para a Ciência e a Tecnologia, National Institutes of Health National Cancer Institute, Swedish Cancer Society, General Hospital in Malmö Foundation for Combating Cancer.
Collapse
Affiliation(s)
- Elizabeth K Bancroft
- Oncogenetics Team, Institute of Cancer Research, London, UK; Cancer Genetics Unit & Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK
| | | | - Mark N Brook
- Oncogenetics Team, Institute of Cancer Research, London, UK
| | - Sarah Thomas
- Cancer Genetics Unit & Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Natalie Taylor
- Cancer Genetics Unit & Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Jennifer Pope
- Oncogenetics Team, Institute of Cancer Research, London, UK
| | - Jana McHugh
- Oncogenetics Team, Institute of Cancer Research, London, UK
| | | | | | - Susan Merson
- Oncogenetics Team, Institute of Cancer Research, London, UK
| | - Kai Ren Ong
- Clinical Genetics Unit, Birmingham Women's Hospital, Birmingham, UK
| | - Jonathan Hoffman
- Clinical Genetics Unit, Birmingham Women's Hospital, Birmingham, UK
| | - Camilla Huber
- Clinical Genetics Unit, Birmingham Women's Hospital, Birmingham, UK
| | - Lovise Maehle
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | | | - Astrid Stormorken
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - D Gareth Evans
- Genomic Medicine, Division of Evolution and Genomic Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jeanette Rothwell
- Genomic Medicine, Division of Evolution and Genomic Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Fiona Lalloo
- Genomic Medicine, Division of Evolution and Genomic Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Angela F Brady
- North West Thames Regional Genetics Service, London North West University Healthcare NHS Trust, Harrow, UK
| | - Marion Bartlett
- North West Thames Regional Genetics Service, London North West University Healthcare NHS Trust, Harrow, UK
| | | | | | - Paul James
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia; Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Joanne McKinley
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Lyon Mascarenhas
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Sapna Syngal
- Division of Population Sciences, Dana Farber Cancer Institute, Boston, MA, USA; Brigham and Women's Hospital, Boston, MA, USA
| | - Chinedu Ukaegbu
- Division of Population Sciences, Dana Farber Cancer Institute, Boston, MA, USA
| | - Lucy Side
- University Hospital Southampton, Southampton, UK; Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | - Tessy Thomas
- University Hospital Southampton, Southampton, UK; Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | - Julian Barwell
- Department of Genetics, University of Leicester, Leicester, UK; University Hospitals Leicester, Leicester, UK
| | - Manuel R Teixeira
- Genetics Department and Research Center, Portuguese Oncology Institute (IPO Porto), Porto, Portugal; Biomedical Sciences Institute (ICBAS), Porto University, Porto, Portugal
| | - Louise Izatt
- Clinical Genetics Service, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Mohnish Suri
- Clinical Genetics Service, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Finlay A Macrae
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia; Parkville Familial Cancer Centre, The Royal Melbourne Hospital, Parkville, VIC, Australia; Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Nicola Poplawski
- Adult Genetics Unit, Royal Adelaide Hospital, Adelaide, SA, Australia; Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Rakefet Chen-Shtoyerman
- The Genetic Institute, Kaplan Medical Center, Rehovot, Israel; Biology Department, Ariel University, Ariel, Israel
| | - Munaza Ahmed
- North East Thames Regional Genetics Service, Institute of Child Health, London, UK
| | - Hannah Musgrave
- Yorkshire Regional Genetics Service, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Nicola Nicolai
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Lynn Greenhalgh
- Clinical Genetics Service, Liverpool Women's Hospital, Liverpool, UK
| | - Carole Brewer
- Peninsular Genetics, Derriford Hospital, Plymouth, UK; Royal Devon and Exeter Hospital, Exeter, UK
| | - Nicholas Pachter
- Genetic Services of Western Australia, King Edward Memorial Hospital, Subiaco, WA, Australia; Department of Paediatrics, University of Western Australia, Perth, WA, Australia
| | - Allan D 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
| | - Ashraf Azzabi
- Northern Genetics Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Brian T Helfand
- John and Carol Walter Center for Urological Health, Division of Urology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Dorothy Halliday
- Oxford Centre for Genomic Medicine, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Saundra Buys
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | | | - Kathleen A Cooney
- Duke Cancer Institute and Duke University School of Medicine, Durham, NC, USA
| | - Marion Harris
- Monash Health, Clayton, VIC, Australia; Monash University, Clayton, VIC, Australia
| | - John McGrath
- Royal Devon and Exeter Hospital, Exeter, UK; University of Exeter Medical School, St Luke's Campus, Exeter, UK
| | - Rosemarie Davidson
- West of Scotland Genetic Service, Queen Elizabeth University Hospital, Glasgow, UK
| | - Amy Taylor
- East Anglian Medical Genetics Service, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | | | - Kathryn Myhill
- Cancer Genetics Unit & Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Matthew Hogben
- Cancer Genetics Unit & Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Neil K Aaronson
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Audrey Ardern-Jones
- Cancer Genetics Unit & Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Chris H Bangma
- Department of Urology, Erasmus Cancer Institute, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Elena Castro
- Spanish National Cancer Research Center, Madrid, Spain
| | - David Dearnaley
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Alexander Dias
- Instituto Nacional de Cancer Jose de Alencar Gomes da Silva INCA, Rio de Janeiro, Brazil
| | | | - Diana M Eccles
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK; Faculty of Medicine, University of Southampton, Southampton, UK
| | - Kate Green
- Genomic Medicine, Division of Evolution and Genomic Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester University 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
| | - Oskar Johannsson
- Landspitali - the National University Hospital of Iceland, Reykjavik, Iceland
| | - Vincent Khoo
- Cancer Genetics Unit & Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK; St George's Hospital, Tooting, London, UK; Department of Medicine, The University of Melbourne, Parkville, VIC, Australia; Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Hans Lilja
- Department of Translational Medicine, Lund University, Malmö, Sweden; Department of Laboratory Medicine, Department of Surgery, and Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Geoffrey J Lindeman
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia; Parkville Familial Cancer Centre, The Royal Melbourne Hospital, 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 V Mitra
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Clare Moynihan
- Oncogenetics Team, Institute of Cancer Research, London, UK
| | | | - Gad Rennert
- CHS National Cancer Control Center, Carmel Medical Center, Haifa, Israel
| | - Rebecca Collier
- Clinical Genetics Service, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Judith Offman
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, Guy's Cancer Centre, Guy's Hospital, London, UK
| | | | - Rosalind A Eeles
- Oncogenetics Team, Institute of Cancer Research, London, UK; Cancer Genetics Unit & Academic Urology Unit, Royal Marsden NHS Foundation Trust, London, UK.
| |
Collapse
|
111
|
Gandaglia G, Briganti A, Montorsi F. Reimagining prostate cancer screening: the IMPACT of germline mutations. Lancet Oncol 2021; 22:1491-1492. [PMID: 34678157 DOI: 10.1016/s1470-2045(21)00571-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 01/17/2023]
Affiliation(s)
- Giorgio Gandaglia
- Unit of Urology and Division of Oncology, IRCCS Ospedale San Raffaele, Via Olgettina 58,20132, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Alberto Briganti
- Unit of Urology and Division of Oncology, IRCCS Ospedale San Raffaele, Via Olgettina 58,20132, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Montorsi
- Unit of Urology and Division of Oncology, IRCCS Ospedale San Raffaele, Via Olgettina 58,20132, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
112
|
Horton R, Pharoah P, Hayward J, Lucassen A. Care of men with cancer-predisposing BRCA variants. BMJ 2021; 375:n2376. [PMID: 34649841 PMCID: PMC7612259 DOI: 10.1136/bmj.n2376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Rachel Horton
- Clinical Ethics and Law, Faculty of Medicine, University of Southampton, Southampton, UK
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | - Paul Pharoah
- Department of Public Health and Primary Care, Department of Oncology, Cambridge Cancer Centre, University of Cambridge, UK
| | - Judith Hayward
- Yorkshire Regional Genetics Service, Leeds, UK
- Shipley Medical Practice, Affinity Care, Shipley, UK
| | - Anneke Lucassen
- Clinical Ethics and Law, Faculty of Medicine, University of Southampton, Southampton, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
113
|
Bancroft EK, Raghallaigh HN, Page EC, Eeles RA. Updates in Prostate Cancer Research and Screening in Men at Genetically Higher Risk. CURRENT GENETIC MEDICINE REPORTS 2021; 9:47-58. [PMID: 34790437 PMCID: PMC8585808 DOI: 10.1007/s40142-021-00202-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Prostate cancer (PrCa) is the most common cancer in men in the western world and is a major source of morbidity and mortality. Currently, general population PrCa screening is not recommended due to the limitations of the prostate-specific antigen (PSA) test. As such, there is increasing interest in identifying and screening higher-risk groups. The only established risk factors for PrCa are age, ethnicity, and having a family history of PrCa. A significant proportion of PrCa cases are caused by genetic factors. RECENT FINDINGS Several rare germline variants have been identified that moderately increase risk of PrCa, and targeting screening to these men is proving useful at detecting clinically significant disease. The use of a "polygenic risk score" (PRS) that can calculate a man's personalized risk based on a number of lower-risk, but common genetic variants is the subject of ongoing research. Research efforts are currently focusing on the utility of screening in specific at-risk populations based on ethnicity, such as men of Black Afro-Caribbean descent. Whilst most screening studies have focused on use of PSA testing, the incorporation of additional molecular and genomic biomarkers alongside increasingly sophisticated imaging modalities is being designed to further refine and individualise both the screening and diagnostic pathway. Approximately 10% of men with advanced PrCa have a germline genetic predisposition leading to the opportunity for novel, targeted precision treatments. SUMMARY The mainstreaming of genomics into the PrCa screening, diagnostic and treatment pathway will soon become standard practice and this review summarises current knowledge on genetic predisposition to PrCa and screening studies that are using genomics within their algorithms to target screening to higher-risk groups of men. Finally, we evaluate the importance of germline genetics beyond screening and diagnostics, and its role in the identification of lethal PrCa and in the selection of targeted treatments for advanced disease.
Collapse
Affiliation(s)
- Elizabeth K. Bancroft
- Urology Genetics, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, UK
- Oncogenetics Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Holly Ni Raghallaigh
- Urology Genetics, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, UK
- Oncogenetics Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Elizabeth C. Page
- Urology Genetics, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, UK
- Oncogenetics Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Rosalind A. Eeles
- Urology Genetics, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, UK
- Oncogenetics Team, The Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| |
Collapse
|
114
|
Mata LA, Retamero JA, Gupta RT, García Figueras R, Luna A. Artificial Intelligence-assisted Prostate Cancer Diagnosis: Radiologic-Pathologic Correlation. Radiographics 2021; 41:1676-1697. [PMID: 34597215 DOI: 10.1148/rg.2021210020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The classic prostate cancer (PCa) diagnostic pathway that is based on prostate-specific antigen (PSA) levels and the findings of digital rectal examination followed by systematic biopsy has shown multiple limitations. The use of multiparametric MRI (mpMRI) is now widely accepted in men with clinical suspicion for PCa. In addition, clinical information, PSA density, risk calculators, and genomic and other "omics" biomarkers are being used to improve risk stratification. On the basis of mpMRI and MRI-targeted biopsies (MRI-TBx), new diagnostic pathways have been established, aiming to improve the limitations of the classic diagnostic approach. However, these pathways still show limitations associated with mpMRI and MRI-TBx. Definitive PCa diagnosis is made on the basis of histopathologic Gleason grading, which has demonstrated an excellent correlation with clinical outcomes. However, Gleason grading is done subjectively by pathologists and involves poor reproducibility, and PCa may have a heterogeneous distribution of histologic patterns. Thus, important discrepancies persist between biopsy tumor grading and final whole-organ pathologic assessment after radical prostatectomy. PCa offers a unique opportunity to establish a real radiologic-pathologic correlation, as whole-mount radical prostatectomy specimens permit a complete spatial relationship with mpMRI. Artificial intelligence is increasingly being applied to radiologic and pathologic images to improve clinical accuracy and efficiency in PCa diagnosis. This review delineates current PCa diagnostic pathways, with a focus on the role of mpMRI, MRI-TBx, and pathologic analysis. An overview of the expected improvements in PCa diagnosis derived from the use of artificial intelligence, integrated radiologic-pathologic systems, and decision support tools for multidisciplinary teams is provided. An invited commentary by Purysko is available online. Online supplemental material is available for this article. ©RSNA, 2021.
Collapse
Affiliation(s)
- Lidia Alcalá Mata
- From the Department of Radiology, Clínica Las Nieves, HT Médica, Calle Carmelo Torres Núm 2, 23007 Jaén, Spain (L.A.M., A.L.); Paige.AI, New York, NY (J.A.R.); Department of Radiology, Duke University Medical Center, Durham, NC (R.T.G.); and Department of Radiology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain (R.G.F.)
| | - Juan Antonio Retamero
- From the Department of Radiology, Clínica Las Nieves, HT Médica, Calle Carmelo Torres Núm 2, 23007 Jaén, Spain (L.A.M., A.L.); Paige.AI, New York, NY (J.A.R.); Department of Radiology, Duke University Medical Center, Durham, NC (R.T.G.); and Department of Radiology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain (R.G.F.)
| | - Rajan T Gupta
- From the Department of Radiology, Clínica Las Nieves, HT Médica, Calle Carmelo Torres Núm 2, 23007 Jaén, Spain (L.A.M., A.L.); Paige.AI, New York, NY (J.A.R.); Department of Radiology, Duke University Medical Center, Durham, NC (R.T.G.); and Department of Radiology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain (R.G.F.)
| | - Roberto García Figueras
- From the Department of Radiology, Clínica Las Nieves, HT Médica, Calle Carmelo Torres Núm 2, 23007 Jaén, Spain (L.A.M., A.L.); Paige.AI, New York, NY (J.A.R.); Department of Radiology, Duke University Medical Center, Durham, NC (R.T.G.); and Department of Radiology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain (R.G.F.)
| | - Antonio Luna
- From the Department of Radiology, Clínica Las Nieves, HT Médica, Calle Carmelo Torres Núm 2, 23007 Jaén, Spain (L.A.M., A.L.); Paige.AI, New York, NY (J.A.R.); Department of Radiology, Duke University Medical Center, Durham, NC (R.T.G.); and Department of Radiology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain (R.G.F.)
| |
Collapse
|
115
|
Avances en Uro-Oncología «OncoUrology Forum Special Edition»: lo mejor del 2020. Actas Urol Esp 2021. [DOI: 10.1016/j.acuro.2021.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
116
|
Reichl F, Muhr D, Rebhan K, Kramer G, Shariat SF, Singer CF, Tan YY. Cancer Spectrum, Family History of Cancer and Overall Survival in Men with Germline BRCA1 or BRCA2 Mutations. J Pers Med 2021; 11:jpm11090917. [PMID: 34575694 PMCID: PMC8466243 DOI: 10.3390/jpm11090917] [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: 08/06/2021] [Revised: 09/06/2021] [Accepted: 09/12/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND: Men with germline BRCA1/2 mutations are not well studied compared to their female counterparts. This study evaluates the cancer characteristics, family history of cancer, and outcomes of male BRCA1/2 mutation carriers. METHODS: All men with germline BRCA1/2 mutations who attended genetic assessment between October 1995 and October 2019 at the Medical University of Vienna were identified. Clinicohistopathological features, family history of cancer, and outcomes were assessed by mutation status. RESULTS: Of the 323 men included, 45 (13.9%) had a primary cancer diagnosis, many of whom were BRCA2 carriers (75.5%). Breast cancer (BC) was the most common cancer (57.8%) followed by prostate cancer (15.6%). Invasive ductal carcinoma and hormone receptor positive tumors were the most common. Among 26 BC-affected patients, 42% did not have any relatives with cancer. Parent of origin was only known in half of the 26 men, with 42% of them inherited through the maternal lineage versus 8% through the paternal. BRCA2 carriers and those with a family history of BC had worse overall survival (20 y vs. 23 y BRCA1 carriers; P = 0.007; 19 y vs. 21 y for those without family history of BC; P = 0.036). CONCLUSION: Male BRCA2 carriers were most likely to develop cancer and had worse prognosis. In our dataset, BC was the most common cancer, likely due to referral bias. Not all mutation carriers present with BC or have a family history of cancer to warrant genetic testing.
Collapse
Affiliation(s)
- Florian Reichl
- Department of Obstetrics, Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (F.R.); (D.M.); (C.F.S.)
| | - Daniela Muhr
- Department of Obstetrics, Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (F.R.); (D.M.); (C.F.S.)
| | - Katharina Rebhan
- Department of Urology, Medical University of Vienna, Waeringer Guertel 18-20, 1090 Vienna, Austria; (K.R.); (G.K.); (S.F.S.)
| | - Gero Kramer
- Department of Urology, Medical University of Vienna, Waeringer Guertel 18-20, 1090 Vienna, Austria; (K.R.); (G.K.); (S.F.S.)
| | - Shahrokh F. Shariat
- Department of Urology, Medical University of Vienna, Waeringer Guertel 18-20, 1090 Vienna, Austria; (K.R.); (G.K.); (S.F.S.)
- Institute for Urology and Reproductive Health, Sechenov University, 119991 Moscow, Russia
- Department of Urology, Weill Cornell Medical College, New York, NY 10065, USA
- Department of Urology, University of Texas Southwestern, Dallas, TX 75390, USA
- Department of Urology, Second Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Christian F. Singer
- Department of Obstetrics, Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (F.R.); (D.M.); (C.F.S.)
| | - Yen Y. Tan
- Department of Obstetrics, Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (F.R.); (D.M.); (C.F.S.)
- Correspondence:
| |
Collapse
|
117
|
Okoye JO. Testing for BRCA1/2 and ataxiatelangiectasia mutated in men with high prostate indices: An approach to reducing prostate cancer mortality in Asia and Africa. Asian J Urol 2021; 8:335-336. [PMID: 34401341 PMCID: PMC8356039 DOI: 10.1016/j.ajur.2020.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/28/2020] [Accepted: 06/05/2020] [Indexed: 11/25/2022] Open
Affiliation(s)
- Jude Ogechukwu Okoye
- Histopathology Unit, Department of Medical Laboratory Science, Faculty of Health Sciences and Technology, College of Medicine, Nnamdi Azikiwe University, Nnewi Campus, Anambra State, Nigeria
| |
Collapse
|
118
|
PALB2 mutations and prostate cancer risk and survival. Br J Cancer 2021; 125:569-575. [PMID: 34006922 PMCID: PMC8368211 DOI: 10.1038/s41416-021-01410-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 04/06/2021] [Accepted: 04/15/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND The objective of this study was to establish the contribution of PALB2 mutations to prostate cancer risk and to estimate survival among PALB2 carriers. METHODS We genotyped 5472 unselected men with prostate cancer and 8016 controls for two Polish founder variants of PALB2 (c.509_510delGA and c.172_175delTTGT). In patients with prostate cancer, the survival of carriers of a PALB2 mutation was compared to that of non-carriers. RESULTS A PALB2 mutation was found in 0.29% of cases and 0.21% of controls (odds ratio (OR) = 1.38; 95% confidence interval (CI) 0.70-2.73; p = 0.45). PALB2 mutation carriers were more commonly diagnosed with aggressive cancers of high (8-10) Gleason score than non-carriers (64.3 vs 18.1%, p < 0.0001). The OR for high-grade prostate cancer was 8.05 (95% CI 3.57-18.15, p < 0.0001). After a median follow-up of 102 months, the age-adjusted hazard ratio for all-cause mortality associated with a PALB2 mutation was 2.52 (95% CI 1.40-4.54; p = 0.0023). The actuarial 5-year survival was 42% for PALB2 carriers and was 72% for non-carriers (p = 0.006). CONCLUSION In Poland, PALB2 mutations predispose to an aggressive and lethal form of prostate cancer.
Collapse
|
119
|
Van Poppel H, Hogenhout R, Albers P, van den Bergh RCN, Barentsz JO, Roobol MJ. A European Model for an Organised Risk-stratified Early Detection Programme for Prostate Cancer. Eur Urol Oncol 2021; 4:731-739. [PMID: 34364829 DOI: 10.1016/j.euo.2021.06.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/10/2021] [Accepted: 06/23/2021] [Indexed: 11/16/2022]
Abstract
CONTEXT Overdiagnosis as the argument to stop prostate cancer (PCa) screening is less valid since the introduction of new technologies such as risk calculators (RCs) and magnetic resonance imaging (MRI). These new technologies result in fewer unnecessary biopsy procedures and fewer cases of both overdiagnosis and underdetection. Therefore, we can now adequately respond to the growing and urgent need for a structured risk assessment to detect PCa early. OBJECTIVE To provide expert discussion on the existing evidence for a previously published risk-stratified strategy regarding an organised population-based early detection programme for PCa. EVIDENCE ACQUISITION The proposed algorithm for early detection of PCa emerged from expert consensus by the authors based on available evidence derived from a nonsystematic review of the current literature using Medline/PubMed, Cochrane Library database, ClinicalTrials.gov, ISRCTN Registry, and the European Association of Urology guidelines on PCa. EVIDENCE SYNTHESIS Although not confirmed by the highest level of evidence, current literature and guidelines point towards an algorithm for early detection of PCa that starts with risk-based prostate-specific antigen (PSA) testing, followed by multivariable risk stratification with RCs. All men who are classified to be at intermediate and high risk are then offered prostate MRI. The combined data from RCs and MRI results can be used to select men for prostate biopsy. Low-risk men return to a risk-based safety net that includes individualised PSA-interval tests and, if necessary, repeated MRI. Depending on local availability, the use of the different risk stratification tools may be adapted. CONCLUSIONS We present a risk-stratified algorithm for an organised population-based early detection programme for clinically significant PCa. Although the proposed strategy has not yet been analysed prospectively, it exploits and may even improve the most important available benefits of "PSA-only" screening studies, while at the same time reduces unnecessary biopsies and overdiagnosis by using new risk stratification tools. PATIENT SUMMARY This paper presents a personalised strategy that enables selective early detection of prostate cancer by combining prostate-specific antigen (interval) testing' prediction models (risk calculators), and magnetic resonance imaging scans. This will likely lead to reduced prostate cancer-related morbidity and mortality, while reducing the need for prostate biopsy and limiting overdiagnosis.
Collapse
Affiliation(s)
- Hendrik Van Poppel
- Department of Development and Regeneration, University Hospital KU Leuven, Leuven, Belgium.
| | - Renée Hogenhout
- Department of Urology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Peter Albers
- Department of Urology, Heinrich-Heine University Medical Faculty, Düsseldorf, Germany; Division of Personalized Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Jelle O Barentsz
- Department of Medical Imaging, Radboudumc, Nijmegen, The Netherlands
| | - Monique J Roobol
- Department of Urology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
120
|
Sessine MS, Das S, Park B, Salami SS, Kaffenberger SD, Kasputis A, Solorzano M, Luke M, Vince RA, Kaye DR, Borza T, Stoffel EM, Cobain E, Merajver SD, Jacobs MF, Milliron KJ, Caba L, van Neste L, Mondul AM, Morgan TM. Initial Findings from a High Genetic Risk Prostate Cancer Clinic. Urology 2021; 156:96-103. [PMID: 34280438 DOI: 10.1016/j.urology.2021.05.078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To improve prostate cancer screening for high-risk men, we developed an early detection clinic for patients at high genetic risk of developing prostate cancer. Despite the rapidly growing understanding of germline variants in driving aggressive prostate cancer and the increased availability of genetic testing, there is little evidence surrounding how best to screen these men. METHODS We are reporting on the first 45 patients enrolled, men between the ages of 35-75, primarily with known pathogenic germline variants in prostate cancer susceptibility genes. Screening consists of an intake lifestyle survey, PSA, DRE, and SelectMDx urine assay. A biopsy was recommended for any of the following indications: 1) abnormal DRE, 2) PSA above threshold, or 3) SelectMDx above threshold. The primary outcomes were number needed to screen, and number needed to biopsy to diagnose a patient with prostate cancer. RESULTS Patients enrolled in the clinic included those with BRCA1 (n=7), BRCA2 (n=16), Lynch Syndrome (n=6), and CHEK2 (n = 4) known pathogenic germline variants. The median age and PSA were 58 (range 35-71) and 1.4 ng/ml (range 0.1-11.4 ng/ml), respectively. 12 patients underwent a prostate needle biopsy and there were 4positive biopsies for prostate cancer. CONCLUSION These early data support the feasibility of opening a dedicated clinic for men at high genetic risk of prostate cancer. This early report on the initial enrollment of our long-term study will help optimize early detection protocols and provide evidence for personalized prostate cancer screening in men with key pathogenic germline variants.
Collapse
Affiliation(s)
| | - Sanjay Das
- Department of Urology, Michigan Medicine
| | - Bumsoo Park
- Department of Urology, Michigan Medicine; Department of Family Medicine, Michigan Medicine
| | | | | | | | | | | | | | | | - Tudor Borza
- Department of Urology, University of Wisconsin School of Medicine and Public Health; Division of Urology, William S Middleton Memorial Veterans Hospital
| | | | - Erin Cobain
- Medical Genetics, Rogel Cancer Center, Michigan Medicine
| | | | | | | | | | | | | | | |
Collapse
|
121
|
Prostate Cancer Biomarkers: From diagnosis to prognosis and precision-guided therapeutics. Pharmacol Ther 2021; 228:107932. [PMID: 34174272 DOI: 10.1016/j.pharmthera.2021.107932] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/23/2022]
Abstract
Prostate cancer (PCa) is one of the most commonly diagnosed malignancies and among the leading causes of cancer-related death worldwide. It is a highly heterogeneous disease, ranging from remarkably slow progression or inertia to highly aggressive and fatal disease. As therapeutic decision-making, clinical trial design and outcome highly depend on the appropriate stratification of patients to risk groups, it is imperative to differentiate between benign versus more aggressive states. The incorporation of clinically valuable prognostic and predictive biomarkers is also potentially amenable in this process, in the timely prevention of metastatic disease and in the decision for therapy selection. This review summarizes the progress that has so far been made in the identification of the genomic events that can be used for the classification, prediction and prognostication of PCa, and as major targets for clinical intervention. We include an extensive list of emerging biomarkers for which there is enough preclinical evidence to suggest that they may constitute crucial targets for achieving significant advances in the management of the disease. Finally, we highlight the main challenges that are associated with the identification of clinically significant PCa biomarkers and recommend possible ways to overcome such limitations.
Collapse
|
122
|
Abstract
Prostate cancer (PCa) is one of the most common cancers in developed countries. The results of large trials indicate that the proportion of PCa attributable to hereditary factors is as high as 15%, highlighting the importance of genetic testing. Despite improved understanding of the prevalence of pathogenic variants among men with PCa, it remains unclear which men will most benefit from genetic testing. In this review, we summarize recent evidence on genetic testing in primary PCa and its impact on routine clinical practice. We outline current guideline recommendations on genetic testing, most importantly, for mutations in BRCA1/2, MMR, CHEK2, PALB2, and HOXB13 genes, as well as various single nucleotide polymorphisms associated with an increased risk of developing PCa. The implementation of genetic testing in clinical practice, especially in young patients with aggressive tumors or those with positive family history, represents a new challenge for the coming years and will identify men with pathogenic variants who may benefit from early screening/intervention and specific therapeutic options.
Collapse
|
123
|
Mark JR, McDougall C, Giri VN. Genetic Testing Guidelines and Education of Health Care Providers Involved in Prostate Cancer Care. Urol Clin North Am 2021; 48:311-322. [PMID: 34210487 DOI: 10.1016/j.ucl.2021.03.003] [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] [Indexed: 01/18/2023]
Abstract
Germline testing for prostate cancer (PCA) is revolutionizing PCA care. Two PARP inhibitors are FDA approved for men with metastatic, castration-resistant disease after progression on first-line therapies. In the screening setting, genetic test results may inform initiation and screening strategies. For men with early-stage disease, literature is emerging on the possible role of germline testing in active surveillance discussions. As such, urologists and oncologists must gain working knowledge of the principles and practice of germline testing and hereditary cancer implications for responsible implementation. Here the authors outline key learning areas and practice strategies for responsible dissemination of PCA germline testing.
Collapse
Affiliation(s)
- James Ryan Mark
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Carey McDougall
- Cancer Risk Assessment and Clinical Cancer Genetics, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA; Cancer Risk Assessment and Clinical Cancer Genetics, Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Veda N Giri
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA; Cancer Risk Assessment and Clinical Cancer Genetics, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA; Cancer Risk Assessment and Clinical Cancer Genetics, Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
124
|
Abstract
Recent studies show that the prevalence of germline pathogenic and likely pathogenic variants (also known as mutations) in DNA repair genes in metastatic prostate cancer is higher than previously recognized and higher than in unaffected men. Specific gene dysfunction is important in prostate cancer initiation and/or evolution to metastases. This article reviews key literature on individual genes, recognizing BRCA2 as the gene most commonly altered in the metastatic setting. This article discusses the importance of representative and diverse inclusion, and efforts to advance management for at-risk carrier populations to maximize clinical benefit.
Collapse
Affiliation(s)
- Alexandra O Sokolova
- Department of Medicine (Div. Oncology), University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; VA Puget Sound Health Care System, Seattle, WA, USA
| | | | - Heather H Cheng
- Department of Medicine (Div. Oncology), University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| |
Collapse
|
125
|
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
|
126
|
The role of ALDH2 in tumorigenesis and tumor progression: Targeting ALDH2 as a potential cancer treatment. Acta Pharm Sin B 2021; 11:1400-1411. [PMID: 34221859 PMCID: PMC8245805 DOI: 10.1016/j.apsb.2021.02.008] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
A major mitochondrial enzyme for protecting cells from acetaldehyde toxicity is aldehyde dehydrogenase 2 (ALDH2). The correlation between ALDH2 dysfunction and tumorigenesis/growth/metastasis has been widely reported. Either low or high ALDH2 expression contributes to tumor progression and varies among different tumor types. Furthermore, the ALDH2∗2 polymorphism (rs671) is the most common single nucleotide polymorphism (SNP) in Asia. Epidemiological studies associate ALDH2∗2 with tumorigenesis and progression. This study summarizes the essential functions and potential ALDH2 mechanisms in the occurrence, progression, and treatment of tumors in various types of cancer. Our study indicates that ALDH2 is a potential therapeutic target for cancer therapy.
Collapse
Key Words
- 4-HNE, 4-hydroxy-2-nonenal
- ALD, alcoholic liver disease
- ALDH2
- ALDH2, aldehyde dehydrogenase 2
- AMPK, AMP-activated protein kinase
- Acetaldehyde
- BCa, bladder cancer
- COUP-TF, chicken ovalbumin upstream promoter-transcription factor
- CRC, colorectal cancer
- CSCs, cancer stem cells
- Cancer
- Cancer therapy
- DFS, disease-free survival
- EC, esophageal cancer
- FA, Fanconi anemia
- FANCD2, Fanconi anemia protein
- GCA, gastric cancer
- HCC, hepatocellular carcinoma
- HDACs, histone deacetylases
- HNC, head and neck cancer
- HNF-4, hepatocyte nuclear factor 4
- HR, homologous recombination
- LCSCs, liver cancer stem cells
- MDA, malondialdehyde
- MDR, multi-drug resistance
- MN, micronuclei
- Metastasis
- NAD, nicotinamide adenine dinucleotide
- NCEs, normochromic erythrocytes
- NER, nucleotide excision repair pathway
- NF-κB, nuclear factor-κB
- NHEJ, non-homologous end-joining
- NRF2, nuclear factor erythroid 2 (NF-E2)-related factor 2
- NRRE, nuclear receptor response element
- NSCLC, non-small-cell lung
- NeG, 1,N2-etheno-dGuo
- OPC, oropharyngeal cancer
- OS, overall survival
- OvCa, ovarian cancer
- PBMC, peripheral blood mononuclear cell
- PC, pancreatic cancer
- PdG, N2-propano-2′-deoxyguanosine
- Polymorphism
- Progression
- REV1, Y-family DNA polymerase
- SCC, squamous cell carcinoma
- TGF-β, transforming growth factor β
- Tumorigenesis
- VHL, von Hippel-Lindau
- ccRCC, clear-cell renal cell carcinomas
- εPKC, epsilon protein kinase C
Collapse
|
127
|
King B, McHugh J, Snape K. A Case-Based Clinical Approach to the Investigation, Management and Screening of Families with BRCA2 Related Prostate Cancer. APPLICATION OF CLINICAL GENETICS 2021; 14:255-266. [PMID: 34295175 PMCID: PMC8290889 DOI: 10.2147/tacg.s261737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/02/2021] [Indexed: 12/02/2022]
Abstract
BRCA2 is the most commonly implicated DNA damage repair gene associated with inherited prostate cancer. BRCA2 deficient prostate cancer typically presents at a younger age, is more poorly differentiated, and is associated with worse survival outcomes than non-BRCA2 associated prostate cancer. Despite these unfavourable prognostic implications, poly-ADP ribose polymerase inhibitors and platinum-based chemotherapy have been identified as potent targeted therapeutic agents towards BRCA1/2 deficient cancer cells. This review article explores the literature surrounding BRCA2-related prostate cancer through a familial clinical scenario. The investigation, diagnosis and management of BRCA2 deficient prostate cancer will be explored, alongside the implications of the identification of a germline pathogenic BRCA2 variant within a family, cascade screening and prostate cancer surveillance in unaffected male BRCA2 carriers. A greater understanding of the molecular pathogenesis of DNA damage repair gene deficient prostate cancer, coupled with new treatment paradigms and widened access to both somatic and germline genetic analysis for prostate cancer patients and their families will hopefully enable the robust implementation of high quality evidence-based clinical pathways for both the management and identification of BRCA2 deficient prostate cancer and improved screening, early detection and prevention strategies for individuals at increased genetic risk of prostate cancer.
Collapse
Affiliation(s)
- Bradley King
- Institute of Medical and Biomedical Education, St. George's, University of London, London, UK
| | - Jana McHugh
- Department of Oncogenomics, Institute of Cancer Research, London, UK
| | - Katie Snape
- Department of Clinical Genetics, St George's University Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
128
|
Leon P, Cancel-Tassin G, Bourdon V, Buecher B, Oudard S, Brureau L, Jouffe L, Blanchet P, Stoppa-Lyonnet D, Coulet F, Sobol H, Cussenot O. Bayesian predictive model to assess BRCA2 mutational status according to clinical history: Early onset, metastatic phenotype or family history of breast/ovary cancer. Prostate 2021; 81:318-325. [PMID: 33599307 DOI: 10.1002/pros.24109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/15/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Mutations of the BRCA2 gene are the most frequent alterations found in germline DNA from men with prostate cancer (PrCa), but clinical parameters that could better orientate for BRCA2 mutation screening need to be established. METHODS Germline DNA from 325 PrCa patients (median age at diagnosis: 57 years old) was screened for BRCA2 mutation. The mutation frequency was compared between three subgroups: patients with an age at diagnosis at 55 years old and under (Group I); a personal or family history of breast, uterine or ovarian cancer (Group II); or a metastatic disease (Group III). Frequency of BRCA2 mutations was established for each combination of phenotypes, and compared between patients meeting or not the criteria for each subgroup using Fisher's exact test. Mutual information, direct effect, elasticity and contribution to the mutational status of each phenotype, taking into account overlap between subgroups, were also estimated using Bayesian algorithms. RESULTS The proportion of BRCA2 mutation was 5.9% in Group I, 10.9% in Group II and 6.9% in Group III. The frequency of BRCA2 mutation was significantly higher among patients of Group II (p = .006), and reached 15.6% among patients of this group who presented a metastatic disease. Mutual information, direct effect, elasticity and contribution to the mutational status were the highest for phenotype II. Fifteen (71.4%) of the 21 BRCA2 mutation carriers had an aggressive form of the disease. Four (19%) of them died from PrCa after a median follow-up duration of 64.5 months. CONCLUSIONS Our results showed that a higher frequency of BRCA2 mutation carriers is observed, not only among PrCa patients with young onset or a metastatic disease, but also with a personal or a familial history of breast cancer.
Collapse
Affiliation(s)
- Priscilla Leon
- Department of Urology, Clinique Pasteur, Royan, France
- GRC n°5 Predictive Onco-Urology, Tenon Hospital, AP-HP, Sorbonne University, Paris, France
| | - Geraldine Cancel-Tassin
- GRC n°5 Predictive Onco-Urology, Tenon Hospital, AP-HP, Sorbonne University, Paris, France
- CeRePP, Paris, France
| | - Violaine Bourdon
- Department of Prevention and Screening Genetic Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Bruno Buecher
- Department of Genetics, Institut Curie, Paris, France
| | - Stephane Oudard
- Department of Oncology Unit, Georges Pompidou European Hospital, APHP, Paris, France
| | - Laurent Brureau
- Department of Urology, Pointe-à-Pitre/Abymes University Hospital, Pointe a Pitre, Guadeloupe
- UMR_S 1085, EHESP, Research Institute in Health, Environment and Work (IRSET), Inserm, Pointe-à-Pitre, Guadeloupe
| | | | - Pascal Blanchet
- Department of Urology, Pointe-à-Pitre/Abymes University Hospital, Pointe a Pitre, Guadeloupe
- UMR_S 1085, EHESP, Research Institute in Health, Environment and Work (IRSET), Inserm, Pointe-à-Pitre, Guadeloupe
| | | | - Florence Coulet
- Department of Genetics, Oncogenetics Consulting, Oncogenetics Functional Unit, Groupe Hospitalier Pitié-Salpêtrière APHP, Paris, France
| | - Hagay Sobol
- Department of Prevention and Screening Genetic Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Olivier Cussenot
- GRC n°5 Predictive Onco-Urology, Tenon Hospital, AP-HP, Sorbonne University, Paris, France
- CeRePP, Paris, France
| |
Collapse
|
129
|
Psutka SP, Singer EA, Gore J. A 25-year perspective on advances in the study of the epidemiology, disparities, and outcomes of urologic cancers. Urol Oncol 2021; 39:595-601. [PMID: 33934967 DOI: 10.1016/j.urolonc.2021.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 11/28/2022]
Abstract
In this narrative review, key developments in epidemiologic and clinical outcomes-based research from eminent historical data sources over the past quarter century are summarized. We then describe the rise of secondary and administrative datasets (AD), summarizing the predominant types of available secondary datasets for contemporary research and describe the benefits and inherent limitations in working with secondary data. We review the methodological advances that permit researchers to capitalize on the full capability of secondary data while also addressing the limitations inherent in utilizing these data for the purposes of epidemiologic and outcomes research. Finally, we present candidate strategies to perpetuate this momentum towards optimizing the development of clinical research infrastructure that harnesses the full potential of the ADs to further clinical and epidemiological research, advancing data analysis, and address the many unanswered questions that remain.
Collapse
Affiliation(s)
- Sarah P Psutka
- Department of Urology, University of Washington, Seattle, WA.
| | - Eric A Singer
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - John Gore
- Department of Urology, University of Washington, Seattle, WA
| |
Collapse
|
130
|
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: 85] [Impact Index Per Article: 21.3] [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
|
131
|
Woodward ER, van Veen EM, Evans DG. From BRCA1 to Polygenic Risk Scores: Mutation-Associated Risks in Breast Cancer-Related Genes. Breast Care (Basel) 2021; 16:202-213. [PMID: 34248461 DOI: 10.1159/000515319] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Background There has been huge progress over the last 30 years in identifying the familial component of breast cancer. Summary Currently around 20% is explained by the high-risk genes BRCA1 and BRCA2, a further 2% by other high-penetrance genes, and around 5% by the moderate risk genes ATM and CHEK2. In contrast, the more than 300 low-penetrance single-nucleotide polymorphisms (SNP) now account for around 28% and they are predicted to account for most of the remaining 45% yet to be found. Even for high-risk genes which confer a 40-90% risk of breast cancer, these SNP can substantially affect the level of breast cancer risk. Indeed, the strength of family history and hormonal and reproductive factors is very important in assessing risk even for a BRCA carrier. The risks of contralateral breast cancer are also affected by SNP as well as by the presence of high or moderate risk genes. Genetic testing using gene panels is now commonplace. Key-Messages There is a need for a more parsimonious approach to panels only testing those genes with a definite 2-fold increased risk and only testing those genes with challenging management implications, such as CDH1 and TP53, when there is strong clinical indication to do so. Testing of SNP alongside genes is likely to provide a more accurate risk assessment.
Collapse
Affiliation(s)
- Emma R Woodward
- Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom.,Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Elke M van Veen
- Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom.,Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - D Gareth Evans
- Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom.,Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom.,PREVENT Breast Cancer Prevention Centre, Nightingale Centre, Manchester Universities Foundation Trust, Wythenshawe Hospital, Manchester, United Kingdom.,Manchester Breast Centre, Manchester Cancer Research Centre, The Christie, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
132
|
Teyssonneau D, Margot H, Cabart M, Anonnay M, Sargos P, Vuong NS, Soubeyran I, Sevenet N, Roubaud G. Prostate cancer and PARP inhibitors: progress and challenges. J Hematol Oncol 2021; 14:51. [PMID: 33781305 PMCID: PMC8008655 DOI: 10.1186/s13045-021-01061-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/10/2021] [Indexed: 12/22/2022] Open
Abstract
Despite survival improvements achieved over the last two decades, prostate cancer remains lethal at the metastatic castration-resistant stage (mCRPC) and new therapeutic approaches are needed. Germinal and/or somatic alterations of DNA-damage response pathway genes are found in a substantial number of patients with advanced prostate cancers, mainly of poor prognosis. Such alterations induce a dependency for single strand break reparation through the poly(adenosine diphosphate-ribose) polymerase (PARP) system, providing the rationale to develop PARP inhibitors. In solid tumors, the first demonstration of an improvement in overall survival was provided by olaparib in patients with mCRPC harboring homologous recombination repair deficiencies. Although this represents a major milestone, a number of issues relating to PARP inhibitors remain. This timely review synthesizes and discusses the rationale and development of PARP inhibitors, biomarker-based approaches associated and the future challenges related to their prescription as well as patient pathways.
Collapse
Affiliation(s)
- Diego Teyssonneau
- Department of Medical Oncology, Institut Bergonie, Bordeaux, France.
| | - Henri Margot
- Department of Genetic, Institut Bergonie, Bordeaux, France
| | - Mathilde Cabart
- Department of Medical Oncology, Institut Bergonie, Bordeaux, France
| | - Mylène Anonnay
- Department of Medical Oncology, Institut Bergonie, Bordeaux, France
| | - Paul Sargos
- Department of Radiotherapy, Institut Bergonie, Bordeaux, France
| | - Nam-Son Vuong
- Department of Urology, Clinique Saint-Augustin, Bordeaux, France
| | | | | | - Guilhem Roubaud
- Department of Medical Oncology, Institut Bergonie, Bordeaux, France
| |
Collapse
|
133
|
Cortesi L, Domati F, Guida A, Marchi I, Toss A, Barbieri E, Marcheselli L, Venturelli M, Piana S, Cirilli C, Federico M. BRCA mutation rate and characteristics of prostate tumor in breast and ovarian cancer families: analysis of 6,591 Italian pedigrees. Cancer Biol Med 2021; 18:j.issn.2095-3941.2020.0481. [PMID: 33710808 PMCID: PMC8185862 DOI: 10.20892/j.issn.2095-3941.2020.0481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE As prostate cancer (PrC) shows a BRCA mutation rate as high as 30%, it becomes crucial to find the optimal selection criteria for genetic testing. The primary objective of this study was to evaluate the BRCA mutation rate in families with PrC associated with breast and/or ovarian cancers; secondary aims were to compare the characteristics of families and BRCA-related PrC outcome among BRCA1 and BRCA2 carriers. METHODS Following the Modena criteria for the BRCA test, we evaluated the mutation rate in families with breast and/or ovarian cancer with a Gleason score ≥7 PrCs, by testing breast or ovarian cases and inferring the mutation in the prostate cases. The characteristics of families and BRCA-related PrC outcomes were measured using the chi-square (χ2) test and Kaplan-Meier methods, respectively. RESULTS Among 6,591 families, 580 (8.8%) with a Gleason score ≥ 7 PrCs were identified, of which 332 (57.2%) met the Modena selection criteria for BRCA testing. Overall, 215 breast or ovarian cancer probands (64.8%) were tested, of which 41 resulted positive for BRCA and one for CHEK2 genes (19.5%). No statistically significant differences were found in BRCA-related PrC prognosis and in the characteristics of families among BRCA1, BRCA2 and non-tested patients. Ten of 23 (44%) mutations in the BRCA2 gene fell in the prostate cancer cluster region (PCCR) at the 3´ terminal of the 7914 codon. CONCLUSIONS It appears the Modena criteria are very useful for BRCA testing selection in families with breast and/or ovarian cancer and PrC. A trend toward a worse prognosis has been found in BRCA2 carriers.
Collapse
Affiliation(s)
- Laura Cortesi
- Department of Oncology and Hematology, University Hospital of Modena, Modena 41124, Italy
| | - Federica Domati
- Department of Medical and Surgical Sciences for Children & Adults, Division of Medical Oncology, University Hospital of Modena, Modena 41124, Italy
| | - Annalisa Guida
- Department of Medical and Surgical Sciences for Children & Adults, Division of Medical Oncology, University Hospital of Modena, Modena 41124, Italy
| | - Isabella Marchi
- Department of Oncology and Hematology, University Hospital of Modena, Modena 41124, Italy
| | - Angela Toss
- Department of Medical and Surgical Sciences for Children & Adults, Division of Medical Oncology, University Hospital of Modena, Modena 41124, Italy
| | - Elena Barbieri
- Department of Oncology and Hematology, University Hospital of Modena, Modena 41124, Italy
| | - Luigi Marcheselli
- Department of Medical and Surgical Sciences for Children & Adults, Division of Medical Oncology, University Hospital of Modena, Modena 41124, Italy
| | - Marta Venturelli
- Department of Medical and Surgical Sciences for Children & Adults, Division of Medical Oncology, University Hospital of Modena, Modena 41124, Italy
| | - Simonetta Piana
- Pathology Unit, Azienda USL Reggio Emilia, IRCCS, Reggio Emilia 42123, Italy
| | - Claudia Cirilli
- Modena Cancer Registry, Public Health Department, AUSL Modena 41126, Italy
| | - Massimo Federico
- Department of Medical and Surgical Sciences for Children & Adults, Division of Medical Oncology, University Hospital of Modena, Modena 41124, Italy
| |
Collapse
|
134
|
Patuleia SIS, Hagenaars SC, Moelans CB, Ausems MGEM, van Gils CH, Tollenaar RAEM, van Diest PJ, Mesker WE, van der Wall E. Lessons Learned from Setting Up a Prospective, Longitudinal, Multicenter Study with Women at High Risk for Breast Cancer. Cancer Epidemiol Biomarkers Prev 2021; 30:441-449. [PMID: 33082203 DOI: 10.1158/1055-9965.epi-20-0770] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/30/2020] [Accepted: 10/09/2020] [Indexed: 11/16/2022] Open
Abstract
Women identified with an increased risk of breast cancer due to mutations in cancer susceptibility genes or a familial history of breast cancer undergo tailored screening with the goal of detecting tumors earlier, when potential curative interventions are still possible. Ideally, screening would identify signs of carcinogenesis even before a tumor is detectable by imaging. This could be achieved by timely signaling of altered biomarker levels for precancerous processes in liquid biopsies. Currently, the Nipple Aspirate Fluid (NAF) and the Trial Early Serum Test BREAST cancer (TESTBREAST), both ongoing, prospective, multicenter studies, are investigating biomarkers in liquid biopsies to improve breast cancer screening in high-risk women. The NAF study focuses on changes over time in miRNA expression levels both in blood and NAF samples, whereas the TESTBREAST study analyzes changes in protein levels in blood samples at sequential interval timepoints. These within-subject changes are studied in relation to later occurrence of breast cancer using a nested case-control design. These longitudinal studies face their own challenges in execution, such as hindrances in logistics and in sample processing that were difficult to anticipate. This article offers insight into those challenges and concurrently aims to provide useful strategies for the set-up of similar studies.See related commentary by Sauter, p. 429.
Collapse
Affiliation(s)
- Susana I S Patuleia
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sophie C Hagenaars
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Cathy B Moelans
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Margreet G E M Ausems
- Department of Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Carla H van Gils
- Department of Epidemiology of the Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Rob A E M Tollenaar
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Wilma E Mesker
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Elsken van der Wall
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
135
|
Ridgway AJ, Aning JJ. Role of primary care in the management of prostate cancer. ACTA ACUST UNITED AC 2021. [DOI: 10.1002/psb.1892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Alexander J Ridgway
- Alexander J Ridgway is a Core Trainee in Urology at Bristol Urological Institute, Southmead Hospital, Bristol
| | - Jonathan J Aning
- Jonathan J Aning is a Consultant Urological Surgeon at Bristol Urological Institute and Honorary Senior Lecturer at the University of Bristol
| |
Collapse
|
136
|
Hemal S, DeWitt-Foy M, Klein EA. Management of a Prostate Cancer Patient With Inherited Germline BRCA1 and BRCA2 Mutations: A Case Report. Urology 2021; 153:129-131. [PMID: 33556450 DOI: 10.1016/j.urology.2020.11.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 11/15/2022]
Abstract
The Breast Cancer Gene (BRCA) confers an 8.6-fold higher risk of developing prostate cancer in men ≤ 65 years of age and portends a worse prognosis as compared to noncarriers even in patients with low volume, localized disease. The BRCA2 gene, in particular, imparts a more biologically aggressive form of prostate cancer and a higher prostate cancer specific mortality. From a treatment standpoint, this translates to worse overall clinical outcomes for such patients. The most appropriate screening and management strategy for germline BRCA mutation carriers with prostate cancer is not known. Herein, we present an incidentally discovered prostate cancer in a 61-year-old BRCA1 and BRCA2 germline mutation carrier who was screened and managed using an individualized treatment approach.
Collapse
Affiliation(s)
- Sij Hemal
- Department of Urology, Glickman Urologic and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH.
| | - Molly DeWitt-Foy
- Department of Urology, Glickman Urologic and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Eric A Klein
- Department of Urology, Glickman Urologic and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH
| |
Collapse
|
137
|
Rebello RJ, Oing C, Knudsen KE, Loeb S, Johnson DC, Reiter RE, Gillessen S, Van der Kwast T, Bristow RG. Prostate cancer. Nat Rev Dis Primers 2021. [PMID: 33542230 DOI: 10.1038/s41572-020-0024.3-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2023]
Abstract
Prostate cancer is a complex disease that affects millions of men globally, predominantly in high human development index regions. Patients with localized disease at a low to intermediate risk of recurrence generally have a favourable outcome of 99% overall survival for 10 years if the disease is detected and treated at an early stage. Key genetic alterations include fusions of TMPRSS2 with ETS family genes, amplification of the MYC oncogene, deletion and/or mutation of PTEN and TP53 and, in advanced disease, amplification and/or mutation of the androgen receptor (AR). Prostate cancer is usually diagnosed by prostate biopsy prompted by a blood test to measure prostate-specific antigen levels and/or digital rectal examination. Treatment for localized disease includes active surveillance, radical prostatectomy or ablative radiotherapy as curative approaches. Men whose disease relapses after prostatectomy are treated with salvage radiotherapy and/or androgen deprivation therapy (ADT) for local relapse, or with ADT combined with chemotherapy or novel androgen signalling-targeted agents for systemic relapse. Advanced prostate cancer often progresses despite androgen ablation and is then considered castration-resistant and incurable. Current treatment options include AR-targeted agents, chemotherapy, radionuclides and the poly(ADP-ribose) inhibitor olaparib. Current research aims to improve prostate cancer detection, management and outcomes, including understanding the fundamental biology at all stages of the disease.
Collapse
Affiliation(s)
- Richard J Rebello
- Cancer Research UK Manchester Institute, University of Manchester, Manchester Cancer Research Centre, Manchester, UK
| | - Christoph Oing
- Cancer Research UK Manchester Institute, University of Manchester, Manchester Cancer Research Centre, Manchester, UK
- Department of Oncology, Haematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Centre Eppendorf, Hamburg, Germany
| | - Karen E Knudsen
- Sidney Kimmel Cancer Center at Jefferson Health and Thomas Jefferson University, Philadelphia, PA, USA
| | - Stacy Loeb
- Department of Urology and Population Health, New York University and Manhattan Veterans Affairs, Manhattan, NY, USA
| | - David C Johnson
- Department of Urology, University of North Carolina, Chapel Hill, NC, USA
| | - Robert E Reiter
- Department of Urology, Jonssen Comprehensive Cancer Center UCLA, Los Angeles, CA, USA
| | | | - Theodorus Van der Kwast
- Laboratory Medicine Program, Princess Margaret Cancer Center, University Health Network, Toronto, Canada
| | - Robert G Bristow
- Cancer Research UK Manchester Institute, University of Manchester, Manchester Cancer Research Centre, Manchester, UK.
| |
Collapse
|
138
|
Abstract
Prostate cancer is a complex disease that affects millions of men globally, predominantly in high human development index regions. Patients with localized disease at a low to intermediate risk of recurrence generally have a favourable outcome of 99% overall survival for 10 years if the disease is detected and treated at an early stage. Key genetic alterations include fusions of TMPRSS2 with ETS family genes, amplification of the MYC oncogene, deletion and/or mutation of PTEN and TP53 and, in advanced disease, amplification and/or mutation of the androgen receptor (AR). Prostate cancer is usually diagnosed by prostate biopsy prompted by a blood test to measure prostate-specific antigen levels and/or digital rectal examination. Treatment for localized disease includes active surveillance, radical prostatectomy or ablative radiotherapy as curative approaches. Men whose disease relapses after prostatectomy are treated with salvage radiotherapy and/or androgen deprivation therapy (ADT) for local relapse, or with ADT combined with chemotherapy or novel androgen signalling-targeted agents for systemic relapse. Advanced prostate cancer often progresses despite androgen ablation and is then considered castration-resistant and incurable. Current treatment options include AR-targeted agents, chemotherapy, radionuclides and the poly(ADP-ribose) inhibitor olaparib. Current research aims to improve prostate cancer detection, management and outcomes, including understanding the fundamental biology at all stages of the disease.
Collapse
|
139
|
de Polo A, Labbé DP. Diet-Dependent Metabolic Regulation of DNA Double-Strand Break Repair in Cancer: More Choices on the Menu. Cancer Prev Res (Phila) 2021; 14:403-414. [PMID: 33509805 DOI: 10.1158/1940-6207.capr-20-0470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/27/2020] [Accepted: 01/21/2021] [Indexed: 11/16/2022]
Abstract
Despite several epidemiologic and preclinical studies supporting the role of diet in cancer progression, the complexity of the diet-cancer link makes it challenging to deconvolute the underlying mechanisms, which remain scantly elucidated. This review focuses on genomic instability as one of the cancer hallmarks affected by diet-dependent metabolic alterations. We discuss how altered dietary intake of metabolites of the one-carbon metabolism, including methionine, folate, and vitamins B and C, can impact the methylation processes and thereby tumorigenesis. We present the concept that the protumorigenic effect of certain diets, such as the Western diet, is in part due to a diet-induced erosion of the DNA repair capacity caused by altered epigenetic and epitranscriptomic landscapes, while the protective effect of other dietary patterns, such as the Mediterranean diet, can be partly explained by their ability to sustain a proficient DNA repair. In particular, considering that diet-dependent alterations of the one-carbon metabolism can impact the rate of methylation processes, changes in dietary patterns can affect the activity of writers and erasers of histone and RNA methyl marks and consequently impair their role in ensuring a proficient DNA damage repair.
Collapse
Affiliation(s)
- Anna de Polo
- Division of Urology, Department of Surgery, McGill University and Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - David P Labbé
- Division of Urology, Department of Surgery, McGill University and Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| |
Collapse
|
140
|
Borque-Fernando A, Espílez R, Miramar D, Corbatón D, Rodríguez A, Castro E, Mateo J, Rello L, Méndez A, Gil Sanz MJ. Genetic counseling in prostate cancer: How to implement it in daily clinical practice? Actas Urol Esp 2021; 45:8-20. [PMID: 33059945 DOI: 10.1016/j.acuro.2020.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 08/22/2020] [Indexed: 11/16/2022]
Abstract
Prostate cancer plays an undeniably prominent role in public health in our days and health systems. Its epidemiological impact is quantitatively very close to that of other tumors such as colon cancer and breast cancer, in which genetic counseling is part of their routine clinical practice, both in the initial evaluation and in the selection of therapeutic strategies. Hereditary cancer syndromes, breast/ovarian and Lynch syndrome are part of genetic counseling in these tumors. Currently, we also know that they can be associated to prostate cancer. The time has come to implement genetic counseling in prostate cancer from the earliest stages of its approach, from initial suspicion to the most advanced tumors. We present an updated review carried out by our interdisciplinary working group on scientific literature, clinical practice guidelines and consensus documents, aimed at the creation and drafting of a'Protocol for genetic counseling in prostate cancer' for the study of germline, with easy application in different healthcare settings. This protocol is currently being implemented in our routine practice and provides answers to 3 specific questions: Who should receive genetic counseling for prostate cancer? Which gene panel should be analyzed? How should counseling be done according to the results obtained? Other aspects about who should perform genetic counseling, ethical considerations and regulations are also collected.
Collapse
Affiliation(s)
- A Borque-Fernando
- Servicio de Urología, Hospital Universitario Miguel Servet, IIS-Aragón, Zaragoza, España.
| | - R Espílez
- Servicio de Urología, Hospital Universitario Miguel Servet, IIS-Aragón, Zaragoza, España
| | - D Miramar
- Servicio de Bioquímica, Unidad de Genética, Hospital Universitario Miguel Servet, Zaragoza, España
| | - D Corbatón
- Servicio de Urología, Hospital Universitario Miguel Servet, IIS-Aragón, Zaragoza, España
| | - A Rodríguez
- Servicio de Bioquímica, Unidad de Genética, Hospital Universitario Miguel Servet, Zaragoza, España
| | - E Castro
- Departamento de Oncología Médica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga, Málaga, España
| | - J Mateo
- Instituto de Oncología Vall d'Hebron y Hospital Universitario Vall d'Hebron, Barcelona, España
| | - L Rello
- Servicio de Bioquímica, Unidad de Genética, Hospital Universitario Miguel Servet, Zaragoza, España
| | - A Méndez
- Servicio de Oncología Radioterápica, Hospital Universitario Miguel Servet, Zaragoza, España
| | - M J Gil Sanz
- Servicio de Urología, Hospital Universitario Miguel Servet, IIS-Aragón, Zaragoza, España
| |
Collapse
|
141
|
Sigorski D, Iżycka-Świeszewska E, Bodnar L. Poly(ADP-Ribose) Polymerase Inhibitors in Prostate Cancer: Molecular Mechanisms, and Preclinical and Clinical Data. Target Oncol 2020; 15:709-722. [PMID: 33044685 PMCID: PMC7701127 DOI: 10.1007/s11523-020-00756-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genomic instability is one of the hallmarks of cancer. The incidence of genetic alterations in homologous recombination repair genes increases during cancer progression, and 20% of prostate cancers (PCas) have defects in DNA repair genes. Several somatic and germline gene alterations drive prostate cancer tumorigenesis, and the most important of these are BRCA2, BRCA1, ATM and CHEK2. There is a group of BRCAness tumours that share phenotypic and genotypic properties with classical BRCA-mutated tumours. Poly(ADP-ribose) polymerase inhibitors (PARPis) show synthetic lethality in cancer cells with impaired homologous recombination genes, and patients with these alterations are candidates for PARPi therapy. Androgen deprivation therapy is the mainstay of PCa therapy. PARPis decrease androgen signalling by interaction with molecular mechanisms of the androgen nuclear complex. The PROFOUND phase III trial, comparing olaparib with enzalutamide/abiraterone therapy, revealed increased radiological progression-free survival (rPFS) and overall survival (OS) among patients with metastatic castration-resistant prostate cancer (mCRPC) with BRCA1, BRCA2 or ATM mutations. The clinical efficacy of PARPis has been confirmed in ovarian, breast, pancreatic and recently also in a subset of PCa. There is growing evidence that molecular tumour boards are the future of the oncological therapeutic approach in prostate cancer. In this review, we summarise the data concerning the molecular mechanisms and preclinical and clinical data of PARPis in PCa.
Collapse
Affiliation(s)
- Dawid Sigorski
- Department of Oncology, Collegium Medicum, University of Warmia and Mazury, Al. Wojska Polskiego 37, 10-228, Olsztyn, Poland.
- Clinical Department of Oncology and Immuno-Oncology, Warmian-Masurian Cancer Center of The Ministry of The Interior and Administration's Hospital, Olsztyn, Poland.
| | - Ewa Iżycka-Świeszewska
- Department of Pathology and Neuropathology, Medical University of Gdańsk, Gdańsk, Poland
| | - Lubomir Bodnar
- Department of Oncology, Collegium Medicum, University of Warmia and Mazury, Al. Wojska Polskiego 37, 10-228, Olsztyn, Poland
- Clinical Department of Oncology and Immuno-Oncology, Warmian-Masurian Cancer Center of The Ministry of The Interior and Administration's Hospital, Olsztyn, Poland
| |
Collapse
|
142
|
Enikeev D, Morozov A, Taratkin M, Barret E, Kozlov V, Singla N, Rivas JG, Podoinitsin A, Margulis V, Glybochko P. Active Surveillance for Intermediate-Risk Prostate Cancer: Systematic Review and Meta-analysis of Current Protocols and Outcomes. Clin Genitourin Cancer 2020; 18:e739-e753. [PMID: 32768356 DOI: 10.1016/j.clgc.2020.05.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/06/2020] [Accepted: 05/12/2020] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Current guidelines allow active surveillance for intermediate-risk prostate cancer patients but do not provide comprehensive recommendations for selection. We performed a systematic review and meta-analysis of outcomes for active surveillance in intermediate- and low-risk groups. METHODS We performed a systematic literature search of intermediate-risk localized prostate cancer patients undergoing active surveillance using 3 literature search engines (Medline, Web of Science, and Scopus) over the past 10 years. The primary outcome was the percentage of patients who remain under surveillance. Secondary outcomes included cancer-specific survival, overall survival, and metastasis-free survival. For articles including both low- and intermediate-risk patients undergoing active surveillance, comparisons between the two groups were made. RESULTS The proportion of patients who remained on active surveillance was comparable between the low- and intermediate-risk groups after 10 and 15 years' follow-up (odds ratio [OR], 0.97; 95% confidence interval [CI], 0.83-1.14; and OR, 0.86; 95% CI, 0.65-1.13). Cancer-specific survival was worse in the intermediate-risk group after 10 years (OR, 0.47; 95% CI, 0.31-0.69) and 15 years (OR, 0.34; 95% CI, 0.2-0.58). The overall survival rate showed no statistical difference at 5 years' follow-up (OR, 0.84; 95% CI, 0.45-1.57) but was worse in the intermediate-risk group after 10 years (OR, 0.43; 95% CI, 0.35-0.53). Metastases-free survival did not significantly differ after 5 years (OR, 0.55; 95% CI, 0.2-1.53) and was worse in the intermediate-risk group after 10 years (OR, 0.46; 95% CI, 0.28-0.77). CONCLUSION Active surveillance could be offered to patients with intermediate-risk prostate cancer. However, they should be informed of the need for regular monitoring and the possibility of discontinuation as a result of a higher rate of progression. Available data indicate that 5-year survival rates between intermediate- and low-risk patients do not differ; 10-year survival rates are worse. To assess the long-term effectiveness and safety of active surveillance, it is necessary to develop unified algorithms for patient selection and management, and to prospectively conduct studies with long-term surveillance.
Collapse
Affiliation(s)
- Dmitry Enikeev
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia.
| | - Andrey Morozov
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Mark Taratkin
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Eric Barret
- Department of Urology, Institut Mutualiste Montsouris, Paris, France
| | - Vasiliy Kozlov
- Department of Public Health and Healthcare, Sechenov University, Moscow, Russia
| | - Nirmish Singla
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Juan Gomez Rivas
- Department of Urology, La Paz University Hospital, Madrid, Spain
| | - Alexey Podoinitsin
- Moscow Regional Research and Clinical Institute MONIKI n.a. M.F. Vladimirskiy, Moscow, Russia
| | - Vitaly Margulis
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Petr Glybochko
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| |
Collapse
|
143
|
Mottet N, van den Bergh RCN, Briers E, Van den Broeck T, Cumberbatch MG, De Santis M, Fanti S, Fossati N, Gandaglia G, Gillessen S, Grivas N, Grummet J, Henry AM, van der Kwast TH, Lam TB, Lardas M, Liew M, Mason MD, Moris L, Oprea-Lager DE, van der Poel HG, Rouvière O, Schoots IG, Tilki D, Wiegel T, Willemse PPM, Cornford P. EAU-EANM-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer-2020 Update. Part 1: Screening, Diagnosis, and Local Treatment with Curative Intent. Eur Urol 2020; 79:243-262. [PMID: 33172724 DOI: 10.1016/j.eururo.2020.09.042] [Citation(s) in RCA: 1785] [Impact Index Per Article: 357.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To present a summary of the 2020 version of the European Association of Urology (EAU)-European Association of Nuclear Medicine (EANM)-European Society for Radiotherapy and Oncology (ESTRO)-European Society of Urogenital Radiology (ESUR)-International Society of Geriatric Oncology (SIOG) guidelines on screening, diagnosis, and local treatment of clinically localised prostate cancer (PCa). EVIDENCE ACQUISITION The panel performed a literature review of new data, covering the time frame between 2016 and 2020. The guidelines were updated and a strength rating for each recommendation was added based on a systematic review of the evidence. EVIDENCE SYNTHESIS A risk-adapted strategy for identifying men who may develop PCa is advised, generally commencing at 50 yr of age and based on individualised life expectancy. Risk-adapted screening should be offered to men at increased risk from the age of 45 yr and to breast cancer susceptibility gene (BRCA) mutation carriers, who have been confirmed to be at risk of early and aggressive disease (mainly BRAC2), from around 40 yr of age. The use of multiparametric magnetic resonance imaging in order to avoid unnecessary biopsies is recommended. When a biopsy is performed, a combination of targeted and systematic biopsies must be offered. There is currently no place for the routine use of tissue-based biomarkers. Whilst prostate-specific membrane antigen positron emission tomography computed tomography is the most sensitive staging procedure, the lack of outcome benefit remains a major limitation. Active surveillance (AS) should always be discussed with low-risk patients, as well as with selected intermediate-risk patients with favourable International Society of Urological Pathology (ISUP) 2 lesions. Local therapies are addressed, as well as the AS journey and the management of persistent prostate-specific antigen after surgery. A strong recommendation to consider moderate hypofractionation in intermediate-risk patients is provided. Patients with cN1 PCa should be offered a local treatment combined with long-term hormonal treatment. CONCLUSIONS The evidence in the field of diagnosis, staging, and treatment of localised PCa is evolving rapidly. The 2020 EAU-EANM-ESTRO-ESUR-SIOG guidelines on PCa summarise the most recent findings and advice for their use in clinical practice. These PCa guidelines reflect the multidisciplinary nature of PCa management. PATIENT SUMMARY Updated prostate cancer guidelines are presented, addressing screening, diagnosis, and local treatment with curative intent. These guidelines rely on the available scientific evidence, and new insights will need to be considered and included on a regular basis. In some cases, the supporting evidence for new treatment options is not yet strong enough to provide a recommendation, which is why continuous updating is important. Patients must be fully informed of all relevant options and, together with their treating physicians, decide on the most optimal management for them.
Collapse
Affiliation(s)
- Nicolas Mottet
- Department of Urology, University Hospital, St. Etienne, France.
| | | | | | | | | | - Maria De Santis
- Department of Urology, Charité Universitätsmedizin, Berlin, Germany; Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Stefano Fanti
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Italy
| | - Nicola Fossati
- Unit of Urology/Division of Oncology, URI, IRCCS Ospedale San Raffaele, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Giorgio Gandaglia
- Unit of Urology/Division of Oncology, URI, IRCCS Ospedale San Raffaele, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland; Università della Svizzera Italiana, Lugano, Switzerland; Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Nikos Grivas
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jeremy Grummet
- Department of Surgery, Central Clinical School, Monash University, Caulfield North, Victoria, Australia
| | - Ann M Henry
- Leeds Cancer Centre, St. James's University Hospital and University of Leeds, Leeds, UK
| | | | - Thomas B Lam
- Academic Urology Unit, University of Aberdeen, Aberdeen, UK; Department of Urology, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Michael Lardas
- Department of Urology, Metropolitan General Hospital, Athens, Greece
| | - Matthew Liew
- Department of Urology, Wrightington, Wigan and Leigh NHS Foundation Trust, Wigan, UK
| | - Malcolm D Mason
- Division of Cancer and Genetics, School of Medicine Cardiff University, Velindre Cancer Centre, Cardiff, UK
| | - Lisa Moris
- Department of Urology, University Hospitals Leuven, Leuven, Belgium; Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - Daniela E Oprea-Lager
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU Medical Center, Amsterdam, The Netherlands
| | - Henk G van der Poel
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Olivier Rouvière
- Hospices Civils de Lyon, Department of Urinary and Vascular Imaging, Hôpital Edouard Herriot, Lyon, France; Faculté de Médecine Lyon Est, Université de Lyon, Université Lyon 1, Lyon, France
| | - Ivo G Schoots
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Wiegel
- Department of Radiation Oncology, University Hospital Ulm, Ulm, Germany
| | - Peter-Paul M Willemse
- Department of Urology, Cancer Center University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
144
|
Avanesyan AA, Sokolenko AP, Ivantsov AO, Kleshchev MA, Maydin MA, Bizin IV, Raskin GA, Shelekhova KV, Gorodnova TV, Bessonov AA, Anisimova EI, Volynshchikova OA, Romanko AA, Ni VI, Broyde RV, Tkachenko OB, Whitehead AJ, Scherbakov AM, Imyanitov EN. Gastric Cancer in BRCA1 Germline Mutation Carriers: Results of Endoscopic Screening and Molecular Analysis of Tumor Tissues. Pathobiology 2020; 87:367-374. [PMID: 33161400 DOI: 10.1159/000511323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION There is some evidence suggesting a link between BRCA1/2 germline mutations and increased risk of gastric cancer. METHODS Endoscopic screening for stomach malignancies was performed in 120 BRCA1 mutation carriers in order to evaluate the probability of detecting the tumor disease. RESULTS No instances of gastric cancer were revealed at the first visit. The analysis of atrophic changes performed by OLGA (Operative Link for Gastritis Assessment) criteria revealed that OLGA stages I-IV alterations were observed in 26 of 41 (63%) subjects aged >50 years as compared to 29 of 79 (37%) in younger subjects (p = 0.007, χ2 test). One BRCA1 mutation carrier developed gastric cancer 4 years after the first visit for endoscopic examination. We performed next-generation sequencing analysis for this tumor and additional 4 archival gastric cancers obtained from BRCA1/2 mutation carriers. Somatic loss of the remaining BRCA1/2 allele was observed in 3 out of 5 tumors analyzed; all of these carcinomas, but none of the malignancies with the retained BRCA1/2 copy, showed chromosomal instability. CONCLUSION Taken together, these data justify further studies on the relationships between the BRCA1/2 and gastric cancer.
Collapse
Affiliation(s)
- Albina A Avanesyan
- Department of Endoscopy, City Cancer Center, Saint Petersburg, Russian Federation.,Department of Endoscopy, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation
| | - Anna P Sokolenko
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation, .,Department of Medical Genetics, St. Petersburg Pediatric Medical University, Saint Petersburg, Russian Federation,
| | - Alexandr O Ivantsov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation
| | - Maxim A Kleshchev
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation
| | - Mikhail A Maydin
- Department of Carcinogenesis and Oncogerontology, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation
| | - Ilya V Bizin
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation
| | - Grigory A Raskin
- A.M. Granov Center for Radiology and Surgical Technologies, Saint Petersburg, Russian Federation
| | - Ksenia V Shelekhova
- Department of Pathology, City Cancer Center, Saint Petersburg, Russian Federation
| | - Tatiana V Gorodnova
- Department of Oncogynecology, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation
| | - Alexandr A Bessonov
- Department of Mammology, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation
| | - Elena I Anisimova
- Leningrad Regional Oncology Hospital, Saint Petersburg, Russian Federation
| | - Olga A Volynshchikova
- Department of Clinical Management and Control, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation
| | - Alexandr A Romanko
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation
| | - Valeria I Ni
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation
| | - Robert V Broyde
- Department of Endoscopy, City Cancer Center, Saint Petersburg, Russian Federation
| | - Oleg B Tkachenko
- Department of Endoscopy, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation
| | - Aldon J Whitehead
- Medicine Internal Medicine Residency Program, The University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Alexandr M Scherbakov
- Department of Endoscopy, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation
| | - Evgeny N Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, Saint Petersburg, Russian Federation.,Department of Medical Genetics, St. Petersburg Pediatric Medical University, Saint Petersburg, Russian Federation.,Department of Oncology, I.I. Mechnikov North-Western Medical University, Saint Petersburg, Russian Federation
| |
Collapse
|
145
|
Recommandations françaises du Comité de cancérologie de l’AFU – actualisation 2020–2022 : cancer de la prostate. Prog Urol 2020; 30:S136-S251. [DOI: 10.1016/s1166-7087(20)30752-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
146
|
Chung JS, Morgan TM, Hong SK. Clinical implications of genomic evaluations for prostate cancer risk stratification, screening, and treatment: a narrative review. Prostate Int 2020; 8:99-106. [PMID: 33102389 PMCID: PMC7557186 DOI: 10.1016/j.prnil.2020.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 09/06/2020] [Indexed: 02/08/2023] Open
Abstract
New classification systems based on molecular features have been introduced to improve precision medicine for prostate cancer (PCa). This review covers the increasing risk of PCa and the differences in response to targeted therapy that are related to specific gene variations. We believe that genomic evaluations will be useful for guiding PCa risk stratification, screening, and treatment. We searched the PubMed and MEDLINE databases for articles related to genomic testing for PCa that were published in 2020 or earlier. There is increasing evidence that germline mutations in DNA repair genes, such as BRCA1/2 or ATM, are closely related to the development and aggressiveness of PCa. Targeted prostate-specific antigen screening based on the presence of germline alterations in DNA repair genes is recommend to achieve an early diagnosis of PCa. In cases of localized PCa, even if it has a favorable risk classification, patients under active surveillance with these gene alterations are likely to develop aggressive PCa. Thus, active treatment may be preferable to active surveillance for these patients. In cases of metastatic castration–resistant PCa, BRCA1/2 and DNA mismatch repair genes may be useful biomarkers for predicting the response to androgen receptor–targeting agents, poly (ADP-ribose) polymerase inhibitors, platinum chemotherapy, prostate-specific membrane antigen–targeted therapy, immunotherapy, and radium-223. Genomic evaluations may allow for risk stratification of patients with PCa based on their molecular features, which may help guide precision medicine for treating PCa.
Collapse
Affiliation(s)
- Jae-Seung Chung
- Department of Urology, Inje University Haeundae Paik Hospital, Busan, Korea
| | - Todd M Morgan
- Department of Urology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI, USA
| | - Sung Kyu Hong
- Department of Urology, Seoul National University College of Medicine, Seoul, Korea.,Department of Urology, Seoul National University Bundang Hospital, Seongnam-si, Korea
| |
Collapse
|
147
|
Segal N, Ber Y, Benjaminov O, Tamir S, Yakimov M, Kedar I, Rosenbaum E, Sela S, Ozalvo R, Shavit-Grievink L, Keder D, Baniel J, Margel D. Imaging-based prostate cancer screening among BRCA mutation carriers-results from the first round of screening. Ann Oncol 2020; 31:1545-1552. [PMID: 32958357 DOI: 10.1016/j.annonc.2020.06.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Male-carriers of BRCA1/2 gene mutations have an increased risk of prostate cancer (PCa) with a more aggressive phenotype. Current screening-guidelines suggest the use of prostate-specific antigen (PSA) only among BRCA2 carriers. Female carriers have extensive guidelines that include imaging. Our objective was to test the prevalence of PCa among BRCA carriers and examine screening strategies, using PSA and multiparametric magnetic resonance imaging (mpMRI). PATIENTS AND METHODS We recruited men aged 40-70 years with BRCA1/2 germline mutations and no prior history of prostate biopsy. All men underwent an initial round of screening which included PSA, and prostate mpMRI. PSA was considered elevated using an age-stratified threshold of ≥1 ng/ml for 40-50 years of age, ≥2 ng/ml for 50-60 years of age, and 2.5 ng/ml for 60-70 years of age. Men with elevated PSA and/or suspicious lesion on mpMRI were offered a prostate biopsy. PSA levels, MRI findings, PCa incidence, and tumor characteristics were evaluated. Decision curve analysis was used to compare screening strategies. RESULTS We recruited 188 men (108 BRCA1, 80 BRCA2), mean age 54 years (9.8). One hundred and ten (57%) had either elevated age-stratified PSA (75; 40%), a suspicious MRI lesion (67; 36%), or both (32; 17%). Of these, 92 (85%) agreed to perform a prostate biopsy. Sixteen (8.5%) were diagnosed with PCa; 44% of the tumors were classified as intermediate- or high-risk disease. mpMRI-based screening missed only one of the cancers (6%), while age-stratified PSA would have missed five (31%). Decision curve analysis showed that mpMRI screening, regardless of PSA, had the highest net benefit for PCa diagnosis, especially among men younger than 55 years of age. We found no difference in the risk of PCa between BRCA1 and BRCA2 (8.3% versus 8.7%, P = 0.91). Ninety percent had a Jewish founder mutation, thus the results cannot be generalized to all ethnic groups. CONCLUSIONS PCa is prevalent among BRCA carriers. Age may affect screening strategy for PCa in this population. Young carriers could benefit from initial MRI screening. BRCA carriers aged older than 55 years should use PSA and be referred to mpMRI if elevated. TRIAL REGISTRATION ClinicalTrial.gov ID: NCT02053805.
Collapse
Affiliation(s)
- N Segal
- Division of Urology, Rabin Medical Center, Petah-Tikva, Israel
| | - Y Ber
- Division of Urology, Rabin Medical Center, Petah-Tikva, Israel
| | - O Benjaminov
- Division of Imaging, Shaare Zedek Medical Center, Jerusalem, Israel
| | - S Tamir
- Department of Imaging, Rabin Medical Center, Petah-Tikva, Israel
| | - M Yakimov
- Department of Pathology, Rabin Medical Center, Petah-Tikva, Israel
| | - I Kedar
- The Raphael Recanati Genetic Institute, Rabin Medical Center, Petah-Tikva, Israel
| | - E Rosenbaum
- Davidoff Cancer Centre, Rabin Medical Center, Petach Tikva, Israel
| | - S Sela
- Division of Urology, Rabin Medical Center, Petah-Tikva, Israel
| | - R Ozalvo
- Division of Urology, Rabin Medical Center, Petah-Tikva, Israel
| | | | - D Keder
- Division of Urology, Rabin Medical Center, Petah-Tikva, Israel
| | - J Baniel
- Division of Urology, Rabin Medical Center, Petah-Tikva, Israel; Department of Surgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - D Margel
- Division of Urology, Rabin Medical Center, Petah-Tikva, Israel; Department of Surgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
148
|
Wokołorczyk D, Kluźniak W, Huzarski T, Gronwald J, Szymiczek A, Rusak B, Stempa K, Gliniewicz K, Kashyap A, Morawska S, Dębniak T, Jakubowska A, Szwiec M, Domagała P, Lubiński J, Narod SA, Akbari MR, Cybulski C. Mutations in ATM, NBN and BRCA2 predispose to aggressive prostate cancer in Poland. Int J Cancer 2020; 147:2793-2800. [PMID: 32875559 DOI: 10.1002/ijc.33272] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 01/04/2023]
Abstract
In designing national strategies for genetic testing, it is important to define the full spectrum of pathogenic mutations in prostate cancer (PCa) susceptibility genes. To investigate the frequency of mutations in PCa susceptibility genes in Polish familial PCa cases and to estimate gene-related PCa risks and probability of aggressive disease, we analyzed the coding regions of 14 genes by exome sequencing in 390 men with familial prostate cancer and 308 cancer-free controls. We compared the mutation frequencies between PCa cases and controls. We also compared clinical characteristics of prostate cancers between mutation carriers and noncarriers. Of the 390 PCa cases, 76 men (19.5%) carried a mutation in BRCA1, BRCA2, NBN, ATM, CHEK2, HOXB13, MSH2 or MSH6 genes. No mutations were found in BRIP1, PTEN, TP53, MLH1, PMS2 and SPOP. Significant associations with familial PCa risk were observed for CHEK2, NBN, ATM, and HOXB13. High-grade (Gleason 8-10) tumors were seen in 56% of BRCA2, NBN or ATM carriers, compared to 21% of patients who tested negative for mutations in these genes (OR = 4.7, 95% CI 2.0-10.7, P = .0003). In summary, approximately 20% of familial prostate cancer cases in Poland can be attributed to mutations in eight susceptibility genes. Carriers of mutations in BRCA2, NBN and ATM develop aggressive disease and may benefit from intensified screening and/or chemotherapy.
Collapse
Affiliation(s)
- Dominika Wokołorczyk
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Wojciech Kluźniak
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Tomasz Huzarski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland.,Department of Clinical Genetics and Pathology, University of Zielona Góra, Poland
| | - Jacek Gronwald
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Agata Szymiczek
- Women's College Research Institute, Women's College Hospital, University of Toronto, Toronto, Canada
| | - Bogna Rusak
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Klaudia Stempa
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Katarzyna Gliniewicz
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Aniruddh Kashyap
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Sylwia Morawska
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Tadeusz Dębniak
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Anna Jakubowska
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland.,Independent Laboratory of Molecular Biology and Genetic Diagnostics, Pomeranian Medical University, Szczecin, Poland
| | - Marek Szwiec
- Clinics of Oncology, University Hospital in Zielona Góra, Zielona Góra, Poland
| | - Paweł Domagała
- Department of Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Jan Lubiński
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Steven A Narod
- Women's College Research Institute, Women's College Hospital, University of Toronto, Toronto, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Mohammad R Akbari
- Women's College Research Institute, Women's College Hospital, University of Toronto, Toronto, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Cezary Cybulski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | | |
Collapse
|
149
|
Huynh-Le MP, Fan CC, Karunamuni R, Walsh EI, Turner EL, Lane JA, Martin RM, Neal DE, Donovan JL, Hamdy FC, Parsons JK, Eeles RA, Easton DF, Kote-Jarai ZS, Al Olama AA, Garcia SB, Muir K, Gronberg H, Wiklund F, Aly M, Schleutker J, Sipeky C, Tammela TLJ, Nordestgaard BG, Key TJ, Travis RC, Pharoah PDP, Pashayan N, Khaw KT, Thibodeau SN, McDonnell SK, Schaid DJ, Maier C, Vogel W, Luedeke M, Herkommer K, Kibel AS, Cybulski C, Wokolorczyk D, Kluzniak W, Cannon-Albright LA, Brenner H, Schöttker B, Holleczek B, Park JY, Sellers TA, Lin HY, Slavov CK, Kaneva RP, Mitev VI, Batra J, Clements JA, Spurdle AB, Australian Prostate Cancer BioResource (APCB), Teixeira MR, Paulo P, Maia S, Pandha H, Michael A, Mills IG, Andreassen OA, Dale AM, Seibert TM, PRACTICAL Consortium. A Genetic Risk Score to Personalize Prostate Cancer Screening, Applied to Population Data. Cancer Epidemiol Biomarkers Prev 2020; 29:1731-1738. [PMID: 32581112 PMCID: PMC7483627 DOI: 10.1158/1055-9965.epi-19-1527] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/25/2020] [Accepted: 06/15/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND A polygenic hazard score (PHS), the weighted sum of 54 SNP genotypes, was previously validated for association with clinically significant prostate cancer and for improved prostate cancer screening accuracy. Here, we assess the potential impact of PHS-informed screening. METHODS United Kingdom population incidence data (Cancer Research United Kingdom) and data from the Cluster Randomized Trial of PSA Testing for Prostate Cancer were combined to estimate age-specific clinically significant prostate cancer incidence (Gleason score ≥7, stage T3-T4, PSA ≥10, or nodal/distant metastases). Using HRs estimated from the ProtecT prostate cancer trial, age-specific incidence rates were calculated for various PHS risk percentiles. Risk-equivalent age, when someone with a given PHS percentile has prostate cancer risk equivalent to an average 50-year-old man (50-year-standard risk), was derived from PHS and incidence data. Positive predictive value (PPV) of PSA testing for clinically significant prostate cancer was calculated using PHS-adjusted age groups. RESULTS The expected age at diagnosis of clinically significant prostate cancer differs by 19 years between the 1st and 99th PHS percentiles: men with PHS in the 1st and 99th percentiles reach the 50-year-standard risk level at ages 60 and 41, respectively. PPV of PSA was higher for men with higher PHS-adjusted age. CONCLUSIONS PHS provides individualized estimates of risk-equivalent age for clinically significant prostate cancer. Screening initiation could be adjusted by a man's PHS. IMPACT Personalized genetic risk assessments could inform prostate cancer screening decisions.
Collapse
Affiliation(s)
- Minh-Phuong Huynh-Le
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, USA
| | - Chun Chieh Fan
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, USA
| | - Roshan Karunamuni
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, USA
| | - Eleanor I. Walsh
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, UK
| | - Emma L. Turner
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, UK
| | - J. Athene Lane
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Richard M. Martin
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- National Institute for Health Research (NIHR) Bristol Biomedical Research Centre, University Hospitals Bristol NHS Foundation Trust and the University of Bristol, Bristol, UK
| | - David E. Neal
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- Department of Oncology, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Cambridge UK
| | - Jenny L. Donovan
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Freddie C. Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- Faculty of Medical Science, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - J. Kellogg Parsons
- Department of Urology, University of California, San Diego, La Jolla, CA, USA
| | - Rosalind A. Eeles
- The Institute of Cancer Research, London, UK
- Royal Marsden NHS Foundation Trust, London, UK
| | - Douglas F. Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | | | - Ali Amin Al Olama
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
- Department of Clinical Neurosciences, Stroke Research Group, University of Cambridge, Cambridge, UK
| | - Sara Benlloch Garcia
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Kenneth Muir
- Division of Population Health, Health Services Research and Primary Care, University of Manchester, Oxford Road, Manchester, UK
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Henrik Gronberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Fredrik Wiklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Markus Aly
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Department of Urology, Karolinska University Hospital, Stockholm, Sweden
| | - Johanna Schleutker
- Institute of Biomedicine, University of Turku, Turku Finland
- Department of Medical Genetics, Genomics, Laboratory Division, Turku University Hospital, Turku, Finland
| | - Csilla Sipeky
- Institute of Biomedicine, University of Turku, Turku Finland
| | - Teuvo LJ Tammela
- Faculty of Medicine and Health Technology, Prostate Cancer Research Center, FI-33014 Tampere University, Finland
- Department of Urology, University of Tampere, Finland
| | - Børge G. Nordestgaard
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Copenhagen, Denmark
| | | | | | - Paul D. P. Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Strangeways Laboratory, Cambridge, UK
| | - Nora Pashayan
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Strangeways Laboratory, Cambridge, UK
- University College London, Department of Applied Health Research, London, UK
| | - Kay-Tee Khaw
- Clinical Gerontology Unit, University of Cambridge, Cambridge, UK
| | - Stephen N. Thibodeau
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Shannon K. McDonnell
- Division of Biomedical Statistics & Informatics, Mayo Clinic, Rochester, MN, USA
| | - Daniel J. Schaid
- Division of Biomedical Statistics & Informatics, Mayo Clinic, Rochester, MN, USA
| | | | - Walther Vogel
- Institute for Human Genetics, University Hospital Ulm, Ulm, Germany
| | | | - Kathleen Herkommer
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Urology, Munich, Germany
| | - Adam S. Kibel
- Division of Urologic Surgery, Brigham and Womens Hospital, Boston, MA, USA
| | - Cezary Cybulski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Dominika Wokolorczyk
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Wojciech Kluzniak
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Lisa A. Cannon-Albright
- Division of Genetic Epidemiology, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Bernd Holleczek
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Saarland Cancer Registry, D-66119 Saarbrücken, Germany
| | - Jong Y. Park
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Thomas A. Sellers
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Hui-Yi Lin
- School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Chavdar Kroumov Slavov
- Department of Urology and Alexandrovska University Hospital, Medical University of Sofia, Sofia, Bulgaria
| | - Radka P. Kaneva
- Molecular Medicine Center, Department of Medical Chemistry and Biochemistry, Medical University of Sofia, Sofia, Bulgaria
| | - Vanio I. Mitev
- Molecular Medicine Center, Department of Medical Chemistry and Biochemistry, Medical University of Sofia, Sofia, Bulgaria
| | - Jyotsna Batra
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre-Qld, Translational Research Institute, Brisbane, Queensland, Australia
| | - Judith A. Clements
- Australian Prostate Cancer Research Centre-Qld, Translational Research Institute, Brisbane, Queensland, Australia
- Translational Research Institute, Brisbane, Queensland, Australia
| | - Amanda B. Spurdle
- Molecular Cancer Epidemiology Laboratory, QIMR Berghofer Institute of Medical Research, Brisbane, Australia
| | | | - Manuel R. Teixeira
- Department of Genetics, Portuguese Oncology Institute, Porto, Portugal
- Biomedical Sciences Institute (ICBAS), University of Porto, Porto, Portugal
| | - Paula Paulo
- Department of Genetics, Portuguese Oncology Institute, Porto, Portugal
- Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Sofia Maia
- Department of Genetics, Portuguese Oncology Institute, Porto, Portugal
- Cancer Genetics Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | | | | | - Ian G. Mills
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Ole A. Andreassen
- NORMENT, KG Jebsen Centre, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Anders M. Dale
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California San Diego, La Jolla, CA, USA
| | - Tyler M. Seibert
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | | |
Collapse
|
150
|
López-Campos F, Linares-Espinós E, Maldonado Pijoan X, Sancho Pardo G, Morgan TM, Martínez-Ballesteros C, Martínez-Salamanca J, Couñago F. Genetic testing for the clinician in prostate cancer. Expert Rev Mol Diagn 2020; 20:933-946. [PMID: 32885704 DOI: 10.1080/14737159.2020.1816170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 08/24/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Prostate cancer (PCa) is one of the most common cancers worldwide and a leading cause of cancer-related mortality. Although the diagnosis and treatment of prostate cancer has improved substantially in recent years, new molecular biomarkers are needed to further prolong survival and improve the quality of life in these patients. AREAS COVERED This review analyzes the current evidence for prognostic and predictive molecular biomarkers that can be applied across different clinical scenarios, ranging from localized disease to metastatic castration-resistant PCa, with a particular emphasis on the biomarkers likely to become available in routine clinical practice in the near future. EXPERT OPINION There is a growing need for molecular testing to identify the most indolent types of prostate cancer to help optimize treatment strategies and spare treatment in these patients when possible. Current trends in the treatment of prostate cancer underscore the unmet clinical need for biomarkers to improve decision-making in a challenging clinical setting.
Collapse
Affiliation(s)
| | - Estefanía Linares-Espinós
- Urology Department, Hospital Universitario La Paz , Madrid, Spain
- Urology Department, Lyx Institute of Urology , Madrid, Spain
- Urology Department, Francisco De Vitoria University , Madrid, Spain
| | | | - Gemma Sancho Pardo
- Radiation Oncology Department, Hospital De La Santa Creu I Sant Pau , Barcelona, Spain
| | - Todd Mathew Morgan
- Urology Department. Michigan Center for Translational Pathology. Comprehensive Cancer Center, Cancer Center Floor B1 Reception C , Ann Arbor, MI, USA
| | - Claudio Martínez-Ballesteros
- Urology Department, Lyx Institute of Urology , Madrid, Spain
- Urology Department, Hospital Universitario Puerta De Hierro Majadahonda , Majadahonda, Spain
| | - Juan Martínez-Salamanca
- Urology Department, Lyx Institute of Urology , Madrid, Spain
- Urology Department, Francisco De Vitoria University , Madrid, Spain
- Urology Department, Hospital Universitario Puerta De Hierro Majadahonda , Majadahonda, Spain
| | - Felipe Couñago
- Radiation Oncology Department, Hospital Universitario Quirón Salud Madrid , Madrid, Spain
- Hospital de La Luz. Madrid
- Universidad Europea de Madrid
| |
Collapse
|