51
|
Narod SA. Choices for cancer prevention for women with a BRCA1 mutation? a personal view. Hered Cancer Clin Pract 2023; 21:26. [PMID: 38031144 PMCID: PMC10685461 DOI: 10.1186/s13053-023-00271-3] [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: 10/30/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023] Open
Abstract
With widespread testing for susceptibility genes, increasing numbers of women are being identified to carry a mutation in one of many genes which renders them susceptible to cancer. The first gene to be identified (in 1994) was BRCA1 which increases a woman's risk for breast cancer (70%) and ovarian cancer (40%). The prevalence of BRCA1 gene mutations has been studied widely and in many countries, mostly in women affected with cancer. In many settings testing is offered routinely to women with serous ovarian cancer or early-onset or triple-negative breast cancer. It is preferable to identify a mutation in a healthy women prior to the diagnosis of cancer. The basic strategies for prevention include surgical prevention, chemoprevention and screening (early detection). Much progress has been made in the past two decades evaluating the benefits of these three approaches. In this commentary I provide my personal views regarding these various interventions in the context of counselling a newly diagnosed health woman with a BRCA1 mutation.
Collapse
Affiliation(s)
- Steven A Narod
- Women's College Research Institute, University of Toronto, 790 Bay Street, Toronto, ON, Canada.
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
52
|
Filser M, Schwartz M, Merchadou K, Hamza A, Villy MC, Decees A, Frouin E, Girard E, Caputo SM, Renault V, Becette V, Golmard L, Servant N, Stoppa-Lyonnet D, Delattre O, Colas C, Masliah-Planchon J. Adaptive nanopore sequencing to determine pathogenicity of BRCA1 exonic duplication. J Med Genet 2023; 60:1206-1209. [PMID: 37263769 PMCID: PMC10715497 DOI: 10.1136/jmg-2023-109155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/30/2023] [Indexed: 06/03/2023]
Abstract
BRCA1 and BRCA2 are tumour suppressor genes that have been characterised as predisposition genes for the development of hereditary breast and ovarian cancers among other malignancies. The molecular diagnosis of this predisposition syndrome is based on the detection of inactivating variants of any type in those genes. But in the case of structural variants, functional consequences can be difficult to assess using standard molecular methods, as the precise resolution of their sequence is often impossible with short-read next generation sequencing techniques. It has been recently demonstrated that Oxford Nanopore long-read sequencing technology can accurately and rapidly provide genetic diagnoses of Mendelian diseases, including those linked to pathogenic structural variants. Here, we report the accurate resolution of a germline duplication event of exons 18-20 of BRCA1 using Nanopore sequencing with adaptive sampling target enrichment. This allowed us to classify this variant as pathogenic within a short timeframe of 10 days. This study provides a proof-of-concept that nanopore adaptive sampling is a highly efficient technique for the investigation of structural variants of tumour suppressor genes in a clinical context.
Collapse
Affiliation(s)
- Mathilde Filser
- Genetics Department, Institut Curie, Paris, France
- PSL Research University, Paris, France
| | - Mathias Schwartz
- Genetics Department, Institut Curie, Paris, France
- PSL Research University, Paris, France
| | - Kevin Merchadou
- PSL Research University, Paris, France
- Clinical Bioinformatics Unit, Institut Curie, Paris, France
| | - Abderaouf Hamza
- Genetics Department, Institut Curie, Paris, France
- PSL Research University, Paris, France
| | - Marie-Charlotte Villy
- Oncogenetic Clinic Unit, Institut Curie, Paris, France
- SIREDO Oncology Centre, Institut Curie, Paris, France
| | - Antoine Decees
- Genetics Department, Institut Curie, Paris, France
- PSL Research University, Paris, France
| | - Eléonore Frouin
- PSL Research University, Paris, France
- Clinical Bioinformatics Unit, Institut Curie, Paris, France
| | - Elodie Girard
- PSL Research University, Paris, France
- INSERM U900, Institut Curie, Paris, France
| | - Sandrine M Caputo
- Genetics Department, Institut Curie, Paris, France
- PSL Research University, Paris, France
| | - Victor Renault
- PSL Research University, Paris, France
- Clinical Bioinformatics Unit, Institut Curie, Paris, France
| | - Véronique Becette
- PSL Research University, Paris, France
- Anatomo- and Cyto-pathology, Institut Curie, Saint-Cloud, France
| | - Lisa Golmard
- Genetics Department, Institut Curie, Paris, France
- PSL Research University, Paris, France
| | - Nicolas Servant
- PSL Research University, Paris, France
- INSERM U900, Institut Curie, Paris, France
| | - Dominique Stoppa-Lyonnet
- Genetics Department, Institut Curie, Paris, France
- SIREDO Oncology Centre, Institut Curie, Paris, France
| | - Olivier Delattre
- Genetics Department, Institut Curie, Paris, France
- Inserm U830, PSL University, Research Center, Institut Curie, Paris, France
| | - Chrystelle Colas
- PSL Research University, Paris, France
- Oncogenetic Clinic Unit, Institut Curie, Paris, France
| | | |
Collapse
|
53
|
Cao S, Liu M, Guo Z, Li Y. Long noncoding RNA KCNMB2-AS1 acts as an oncogene in ovarian cancer. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1844-1846. [PMID: 37710992 PMCID: PMC10679871 DOI: 10.3724/abbs.2023228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/15/2023] [Indexed: 09/16/2023] Open
Affiliation(s)
- Siyu Cao
- Department of Gynecologic OncologyFudan University Shanghai Cancer CenterShanghai200032China
| | - Min Liu
- Lab for Noncoding RNA and CancerSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Ziyi Guo
- Lab for Noncoding RNA and CancerSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Yanli Li
- Lab for Noncoding RNA and CancerSchool of Life SciencesShanghai UniversityShanghai200444China
| |
Collapse
|
54
|
Muhammad N, Azeem A, Arif S, Naeemi H, Masood I, Hassan U, Ijaz B, Hanif F, Syed AA, Yusuf MA, Rashid MU. Prevalence of BRCA1 and BRCA2 germline variants in an unselected pancreatic cancer patient cohort in Pakistan. Hered Cancer Clin Pract 2023; 21:22. [PMID: 37951914 PMCID: PMC10640758 DOI: 10.1186/s13053-023-00269-x] [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: 05/18/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND BRCA1 and BRCA2 (BRCA1/2) are the most frequently investigated genes among Caucasian pancreatic cancer patients, whereas limited reports are available among Asians. We aimed to investigate the prevalence of BRCA1/2 germline variants in Pakistani pancreatic cancer patients. METHODS One hundred and fifty unselected and prospectively enrolled pancreatic cancer patients were comprehensively screened for BRCA1/2 germline variants using denaturing high-performance liquid chromatography and high-resolution melting analyses, followed by DNA sequencing of the variant fragments. The novel variants were analyzed for their pathogenic effect using in-silico tools. Potentially functional variants were further screened in 200 cancer-free controls. RESULTS Protein truncating variant was detected in BRCA2 only, with a prevalence of 0.7% (1/150). A frameshift BRCA2 variant (p.Asp946Ilefs*14) was identified in a 71-year-old male patient of Pathan ethnicity, with a family history of abdominal cancer. Additionally, we found a novel variant in BRCA2 (p.Glu2650Gln), two previously reported variants in BRCA1 (p.Thr293Ser) and BRCA2 (p.Ile2296Leu) and a recurrent nonsense variant in BRCA2 (p.Lys3326Ter). These variants were classified as variants of uncertain significance (VUS). It is noteworthy that none of these VUS carriers had a family history of pancreatic or other cancers. CONCLUSIONS In this first study, BRCA1/2 pathogenic variant is identified with a low frequency in pancreatic cancer patients from Pakistan. Comprehensive multigene panel testing is recommended in the Pakistani pancreatic cancer patients to enhance genetic understanding in this population.
Collapse
Affiliation(s)
- Noor Muhammad
- Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC), Lahore, Pakistan
- Laboratory of Applied and Functional Genomics, National Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Ayesha Azeem
- Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC), Lahore, Pakistan
| | - Shumaila Arif
- Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC), Lahore, Pakistan
| | - Humaira Naeemi
- Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC), Lahore, Pakistan
| | - Iqra Masood
- Clinical Research Office, SKMCH&RC, Lahore, Pakistan
| | - Usman Hassan
- Department of Pathology, SKMCH&RC, Lahore, Pakistan
| | - Bushra Ijaz
- Laboratory of Applied and Functional Genomics, National Center of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Faisal Hanif
- Department of Surgical Oncology, SKMCH&RC, Lahore, Pakistan
- Centre for Liver and Biliary Sciences, Bahria International Hospital, Lahore, Pakistan
| | - Aamir Ali Syed
- Department of Surgical Oncology, SKMCH&RC, Lahore, Pakistan
| | | | - Muhammad Usman Rashid
- Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC), Lahore, Pakistan.
| |
Collapse
|
55
|
Nebgen DR, Domchek SM, Kotsopoulos J, de Hullu JA, Crosbie EJ, Paramanandam VS, Brood-van Zanten MMA, Norquist BM, Guise T, Rozenberg S, Kurian AW, Pederson HJ, Yuksel N, Michaelson-Cohen R, Bober SL, da Silva Filho AL, Johansen N, Guidozzi F, Evans DG, Menon U, Kingsberg SA, Powell CB, Grandi G, Marchetti C, Jacobson M, Brennan DJ, Hickey M. Care after premenopausal risk-reducing salpingo-oophorectomy in high-risk women: Scoping review and international consensus recommendations. BJOG 2023; 130:1437-1450. [PMID: 37132126 DOI: 10.1111/1471-0528.17511] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 05/04/2023]
Abstract
Women at high inherited risk of ovarian cancer are offered risk-reducing salpingo-oophorectomy (RRSO) from age 35 to 45 years. Although potentially life-saving, RRSO may induce symptoms that negatively affect quality of life and impair long-term health. Clinical care following RRSO is often suboptimal. This scoping review describes how RRSO affects short- and long-term health and provides evidence-based international consensus recommendations for care from preoperative counselling to long-term disease prevention. This includes the efficacy and safety of hormonal and non-hormonal treatments for vasomotor symptoms, sleep disturbance and sexual dysfunction and effective approaches to prevent bone and cardiovascular disease.
Collapse
Affiliation(s)
- Denise R Nebgen
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Susan M Domchek
- Basser Center for BRCA, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joanne Kotsopoulos
- Women's College Research Institute, Women's College Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Joanne A de Hullu
- Department of Obstetrics and Gynaecology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Emma J Crosbie
- Division of Cancer Sciences, University of Manchester, St Mary's Hospital, Manchester, UK
| | - Vincent Singh Paramanandam
- Department of Obstetrics and Gynaecology, The Royal Women's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - Monique M A Brood-van Zanten
- Department of Gynecology, The Netherlands Cancer Institute, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Barbara M Norquist
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - Theresa Guise
- Department of Endocrine Neoplasia and Hormone Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Serge Rozenberg
- Department of Obstetrics and Gynaecology, Universite Libre de Bruxelles, Brussels, Belgium
| | - Allison W Kurian
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Holly J Pederson
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio, USA
| | - Nese Yuksel
- Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Rachel Michaelson-Cohen
- Department of Gynaecology and Medical Genetics Institute, Hebrew University Faculty of Medicine, Shaare Zedek Medical Centre, Jerusalem, Israel
| | - Sharon L Bober
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Nora Johansen
- Department of Gynaecology and Obstetrics, Sørlandet Hospital HF Arendal, Arendal, Norway
| | - F Guidozzi
- Deparment of Obstetrics and Gynaecology, University of Witwatersrand, Johanesburg, South Africa
| | - D Gareth Evans
- University of Manchester, Prevent Breast Cancer Centre, Manchester, UK
| | - Usha Menon
- MRC Clinical Trials Unit, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Sheryl A Kingsberg
- University Hospitals Cleveland Medical Center, Case Western University School of Medicine, Cleveland, Ohio, USA
| | - C Bethan Powell
- Kaiser Permanente Northern California, Hereditary Cancer Program, San Francisco, California, USA
| | - Giovanni Grandi
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Modena, Italy
| | - Claudia Marchetti
- Department of Women's and Children's Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS-Catholic University Sacred Heart, Rome, Italy
| | - Michelle Jacobson
- Women's College Hospital and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Donal J Brennan
- UCD Gynaecological Oncology Group, UCD School of Medicine, Mater University Hospital, Dublin, Ireland
| | - Martha Hickey
- Department of Obstetrics and Gynaecology, Research Precinct, Level 7, The Royal Women's Hospital, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
56
|
Ghouil R, Miron S, Sato K, Ristic D, van Rossum-Fikkert SE, Legrand P, Ouldali M, Winter JM, Ropars V, David G, Arteni AA, Wyman C, Knipscheer P, Kanaar R, Zelensky AN, Zinn-Justin S. BRCA2-HSF2BP oligomeric ring disassembly by BRME1 promotes homologous recombination. SCIENCE ADVANCES 2023; 9:eadi7352. [PMID: 37889963 PMCID: PMC10610910 DOI: 10.1126/sciadv.adi7352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023]
Abstract
In meiotic homologous recombination (HR), BRCA2 facilitates loading of the recombinases RAD51 and DMC1 at the sites of double-strand breaks (DSBs). The HSF2BP-BRME1 complex interacts with BRCA2. Its absence causes a severe reduction in recombinase loading at meiotic DSB. We previously showed that, in somatic cancer cells ectopically producing HSF2BP, DNA damage can trigger HSF2BP-dependent degradation of BRCA2, which prevents HR. Here, we report that, upon binding to BRCA2, HSF2BP forms octameric rings that are able to interlock into a large ring-shaped 24-mer. Addition of BRME1 leads to dissociation of both of these ring structures and cancels the disruptive effect of HSF2BP on cancer cell resistance to DNA damage. It also prevents BRCA2 degradation during interstrand DNA crosslink repair in Xenopus egg extracts. We propose that, during meiosis, the control of HSF2BPBRCA2 oligomerization by BRME1 ensures timely assembly of the ring complex that concentrates BRCA2 and controls its turnover, thus promoting HR.
Collapse
Affiliation(s)
- Rania Ghouil
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Simona Miron
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Koichi Sato
- Oncode Institute, Hubrecht Institute–KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Dejan Ristic
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA, Rotterdam, Netherlands
| | - Sari E. van Rossum-Fikkert
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA, Rotterdam, Netherlands
| | - Pierre Legrand
- Synchrotron SOLEIL, HelioBio group, L’Orme des Merisiers, Gif sur-Yvette, France
| | - Malika Ouldali
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | | | - Virginie Ropars
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Gabriel David
- Synchrotron SOLEIL, HelioBio group, L’Orme des Merisiers, Gif sur-Yvette, France
| | - Ana-Andreea Arteni
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Claire Wyman
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA, Rotterdam, Netherlands
- Department of Radiation Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA, Rotterdam, Netherlands
| | - Puck Knipscheer
- Oncode Institute, Hubrecht Institute–KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Roland Kanaar
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA, Rotterdam, Netherlands
| | - Alex N. Zelensky
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3000 CA, Rotterdam, Netherlands
| | - Sophie Zinn-Justin
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| |
Collapse
|
57
|
Li J, Zheng H, Hou J, Chen J, Zhang F, Yang X, Jin F, Xi Y. X-linked RBBP7 mutation causes maturation arrest and testicular tumors. J Clin Invest 2023; 133:e171541. [PMID: 37843278 PMCID: PMC10575721 DOI: 10.1172/jci171541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/24/2023] [Indexed: 10/17/2023] Open
Abstract
Maturation arrest (MA) is a subtype of non-obstructive azoospermia, and male infertility is a known risk factor for testicular tumors. However, the genetic basis for many affected individuals remains unknown. Here, we identified a deleterious hemizygous variant of X-linked retinoblastoma-binding protein 7 (RBBP7) as a potential key cause of MA, which was also found to be associated with the development of Leydig cell tumors. This mutation resulted in premature protein translation termination, affecting the sixth WD40 domain of the RBBP7 and the interaction of the mutated RBBP7 with histone H4. Decreased BRCA1 and increased γH2AX were observed in the proband. In mouse spermatogonial and pachytene spermatocyte-derived cells, deprivation of rbbp7 led to cell cycle arrest and apoptosis. In Drosophila, knockdown of RBBP7/Caf1-55 in germ cells resulted in complete absence of germ cells and reduced testis size, whereas knockdown of RBBP7/Caf1-55 in cyst cells resulted in hyperproliferative testicular cells. Interestingly, male infertility caused by Caf1-55 deficiency was rescued by ectopic expression of wild-type human RBBP7 but not mutant variants, suggesting the importance of RBBP7 in spermatogenesis. Our study provides insights into the mechanisms underlying the co-occurrence of MA and testicular tumors and may pave the way for innovative genetic diagnostics of these 2 diseases.
Collapse
Affiliation(s)
- Jingping Li
- Department of Reproductive Endocrinology and
| | - Huimei Zheng
- Division of Human Reproduction and Developmental Genetics, Reproductive Medicine Center, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaru Hou
- Division of Human Reproduction and Developmental Genetics, Reproductive Medicine Center, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianhua Chen
- Department of Pathology, Reproductive Medicine Center, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Xiaohang Yang
- Division of Human Reproduction and Developmental Genetics, Reproductive Medicine Center, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, China
| | - Fan Jin
- Department of Reproductive Endocrinology and
| | - Yongmei Xi
- Division of Human Reproduction and Developmental Genetics, Reproductive Medicine Center, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
58
|
Arnon J, Tabi M, Rottenberg Y, Zick A, Blumenfeld P, Hamburger T, Pikarsky E, Avraham E, Levine L, Popovtzer A, Yablonski-Peretz T, Kadouri L, Nechushtan H. Clinical Characteristics, Response to Platinum-Based Chemotherapy and Poly (Adenosine Phosphate-Ribose) Polymerase Inhibitors in Advanced Lung Cancer Patients Harboring BRCA Mutations. Cancers (Basel) 2023; 15:4733. [PMID: 37835426 PMCID: PMC10571558 DOI: 10.3390/cancers15194733] [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: 08/23/2023] [Revised: 09/16/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
The oncogenic role and clinical relevance of BRCA mutations in NSCLC remain unclear. We aim to evaluate the characteristics and clinical outcomes of patients with NSCLC harboring BRCA mutations treated at Hadassah Medical Center (HMC). We retrospectively assessed all patients with advanced NSCLC who underwent next-generation sequencing (NGS) and were found to have pathogenic somatic BRCA mutations (p-BRCA). We compared clinical outcomes in NSCLC patients with wild-type BRCA (wt-BRCA) matched by age, stage, gender, smoking, PDL-1 and driver mutations. Between 2015 and 2022, we evaluated 598 patients with advanced NSCLC using NGS and found 26 patients with p-BRCA, of whom 17 (65.4%) were carriers of germline BRCA variants and represented 1% of all BRCA carriers HMC. The median age of diagnosis was 67 years old (40-78), 13 patients (50%) had a history of smoking and 9 patients (34.6%) had additional driver mutations (EGFR, ALK, BRAF, MET or ERBB2). Objective response rate and median progression-free survival (PFS) for first-line platinum-based chemotherapy in the p-BRCA group compared to wt-BRCA controls were 72.2% and 16 months (CI 95%, 5-22), compared to 47.4% and 7 months (CI 95%, 5-9), respectively, and HR for PFS was 0.41 (CI 95%, 0.17-0.97). Six patients in the p-BRCA group were treated with advanced-line poly (adenosine-phosphate-ribose) polymerase inhibitors (PARPi), with a durable response observed in four patients (66%). In this cohort, patients with NSCLC harboring p-BRCA exhibit high-sensitivity PARPi and a prolonged response to platinum, suggesting some oncogenic role for BRCA mutations in NSCLC. The results support further prospective trials of the treatment of NSCLC harboring p-BRCA with PARPi.
Collapse
Affiliation(s)
- Johnathan Arnon
- Sharett Institute of Oncology, Hadassah Medical Center, Jerusalem 91120, Israel (L.K.); (H.N.)
- Factuality of Medicine, Hebrew University of Jerusalem, Jerusalem 12272, Israel
| | - Michael Tabi
- Factuality of Medicine, Hebrew University of Jerusalem, Jerusalem 12272, Israel
| | - Yakir Rottenberg
- Sharett Institute of Oncology, Hadassah Medical Center, Jerusalem 91120, Israel (L.K.); (H.N.)
- Factuality of Medicine, Hebrew University of Jerusalem, Jerusalem 12272, Israel
| | - Aviad Zick
- Sharett Institute of Oncology, Hadassah Medical Center, Jerusalem 91120, Israel (L.K.); (H.N.)
- Factuality of Medicine, Hebrew University of Jerusalem, Jerusalem 12272, Israel
| | - Philip Blumenfeld
- Sharett Institute of Oncology, Hadassah Medical Center, Jerusalem 91120, Israel (L.K.); (H.N.)
- Factuality of Medicine, Hebrew University of Jerusalem, Jerusalem 12272, Israel
| | - Tamar Hamburger
- Sharett Institute of Oncology, Hadassah Medical Center, Jerusalem 91120, Israel (L.K.); (H.N.)
- Factuality of Medicine, Hebrew University of Jerusalem, Jerusalem 12272, Israel
| | - Eli Pikarsky
- Factuality of Medicine, Hebrew University of Jerusalem, Jerusalem 12272, Israel
- Department of Pathology, Hadassah Medical Center, Jerusalem 91120, Israel
| | - Eti Avraham
- Department of Pathology, Hadassah Medical Center, Jerusalem 91120, Israel
| | - Leeby Levine
- Stern College for Women, Yeshiva University, New York, NY 10033, USA
| | - Aron Popovtzer
- Sharett Institute of Oncology, Hadassah Medical Center, Jerusalem 91120, Israel (L.K.); (H.N.)
- Factuality of Medicine, Hebrew University of Jerusalem, Jerusalem 12272, Israel
| | - Tamar Yablonski-Peretz
- Sharett Institute of Oncology, Hadassah Medical Center, Jerusalem 91120, Israel (L.K.); (H.N.)
- Factuality of Medicine, Hebrew University of Jerusalem, Jerusalem 12272, Israel
| | - Luna Kadouri
- Sharett Institute of Oncology, Hadassah Medical Center, Jerusalem 91120, Israel (L.K.); (H.N.)
- Factuality of Medicine, Hebrew University of Jerusalem, Jerusalem 12272, Israel
| | - Hovav Nechushtan
- Sharett Institute of Oncology, Hadassah Medical Center, Jerusalem 91120, Israel (L.K.); (H.N.)
- Factuality of Medicine, Hebrew University of Jerusalem, Jerusalem 12272, Israel
| |
Collapse
|
59
|
Wang QL, Zhang Y, Zeng E, Grassmann F, He W, Czene K. Risk of estrogen receptor-specific breast cancer by family history of estrogen receptor subtypes and other cancers. J Natl Cancer Inst 2023; 115:1020-1028. [PMID: 37243749 PMCID: PMC10483332 DOI: 10.1093/jnci/djad104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 05/02/2023] [Accepted: 05/25/2023] [Indexed: 05/29/2023] Open
Abstract
BACKGROUND The extent to which the risk of estrogen receptor (ER)-specific breast cancer is associated with ER status of breast cancer and other cancers among first-degree relatives is unclear. METHODS This population-based cohort included 464 707 cancer-free women in Stockholm, Sweden, during 1978-2019. For ER-negative and ER-positive breast cancers, we estimated hazard ratios (HRs) associated with ER status of female first-degree relatives with breast cancer and of other cancers in all first-degree relatives. Associations between ER-negative and ER-positive status by family cancer history were estimated using logistic regression in a case-only design. RESULTS Women with familial ER-positive breast cancer had 1.87 times (95% confidence interval [CI] = 1.77 to 1.97) higher risk of ER-positive subtype, whereas the corresponding hazard ratio for ER-negative was 2.54 (95% CI = 2.08 to 3.10) when having familial ER-negative breast cancer. The risk increased with an increasing number of female first-degree relatives having concordant subtypes and younger age at diagnosis (Ptrend <.001 for both). Nonbreast cancers among first-degree relatives were associated with both ER-positive (HR = 1.14, 95% CI = 1.10 to 1.17) and ER-negative (HR = 1.08, 95% CI = 1.01 to 1.16) breast cancers. Compared with women with ER-positive breast cancer, women with ER-negative breast cancer were more likely to have family history of liver (odds ratio [OR] = 1.33, 95% CI = 1.05 to 1.67), ovary (OR = 1.28, 95% CI = 1.01 to 1.61), and testicle cancer (OR = 1.79, 95% CI = 1.01 to 3.16) but less likely to have family history of endometrial cancer (OR = 0.77, 95% CI = 0.60 to 1.00) and leukemia (OR = 0.72, 95% CI = 0.56 to 0.91). CONCLUSIONS Risk of ER-specific breast cancer differs according to ER status of female first-degree relatives with breast cancer and some other cancers of first-degree relatives. This family history information should be considered in the individual risk prediction for ER subtypes.
Collapse
Affiliation(s)
- Qiao-Li Wang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Yuqi Zhang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Erwei Zeng
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Felix Grassmann
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Wei He
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Chronic Disease Research Institute, The Children’s Hospital, and National Clinical Research Center for Child Health, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Nutrition and Food Hygiene, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
60
|
Jia KY, Menes TS, Bernstein-Molho R, Nissan A, Zippel D. Characterization of patients with a diagnosis of breast cancer and melanoma: genetic susceptibility or increased surveillance? Eur J Cancer Prev 2023; 32:418-422. [PMID: 36912151 DOI: 10.1097/cej.0000000000000792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
BACKGROUND Breast cancer diagnosis had been linked to an increased risk of melanoma in several reports. The aim of the current study was to assess the role of genetics, increased surveillance, and radiation treatment in patients with a dual diagnosis of breast cancer and melanoma (DBM). MATERIALS AND METHODS All patients treated at Sheba Medical Center between 2007 and 2021 with DBM were included in the cohort. Data on family history, genetic tests, characteristics, and treatment of both cancers were collected. The proportion of patients with a pathogenic variant (PV) in BRCA1 and BRCA2 genes was compared to a control group of patients with breast cancer. The proportion of patients presenting with in-situ disease was compared to the national registry data. RESULTS The cohort included 222 DBM patients of whom 114 had documentation of genetic testing. Twenty patients tested positive for PVs of which 13 (11%) were in BRCA genes. This was comparable to the proportion in patients with a diagnosis of breast cancer (736; 19%). The proportion of melanoma diagnosed at stage 0 was comparable to the national proportion ( N = 40; 30% vs. 28%, respectively). In comparison to the national registry, a larger proportion of breast cancers were ductal carcinoma in situ or lobular carcinoma in situ [10% in the registry vs. 19% (22) in the cohort; P < 0.003]. CONCLUSIONS In patients with DBM we did not find an increased proportion of PVs in BRCA genes. Our findings suggest that the increased standardized incidence ratio of the dual diagnosis may be partially explained by increased surveillance and detection of earlier-stage cancers.
Collapse
Affiliation(s)
- Karen Y Jia
- Department of Surgery, Maimonides Medical Center, Brooklyn, New York, USA
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv
| | - Tehillah S Menes
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv
- Department of General and Oncological Surgery-Surgery C, Chaim Sheba Medical Center, Tel Hashomer
| | - Rinat Bernstein-Molho
- Susanne Levy Gertner Oncogenetics Unit, The Danek Gertner Institute of Human Genetics, Chaim, Israel
| | - Aviram Nissan
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv
- Department of General and Oncological Surgery-Surgery C, Chaim Sheba Medical Center, Tel Hashomer
| | - Dov Zippel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv
- Department of General and Oncological Surgery-Surgery C, Chaim Sheba Medical Center, Tel Hashomer
| |
Collapse
|
61
|
Saeidi H, Bakrin IH, Raju CS, Ismail P, Saraf M, Khairul-Asri MG. Genetic aberrations of homologous recombination repair pathways in prostate cancer: The prognostic and therapeutic implications. Adv Med Sci 2023; 68:359-365. [PMID: 37757663 DOI: 10.1016/j.advms.2023.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023]
Abstract
Prostate cancer (PC) is the second most common cancer in men worldwide. Homologous recombination repair (HRR) gene defects have been identified in a significant proportion of metastatic castration-resistant PC (mCRPC) and are associated with an increased risk of PC and more aggressive PC. Importantly, it has been well-documented that poly ADP-ribose polymerase (PARP) inhibition in cells with HR deficiency (HRD) can cause cell death. This has been exploited for the targeted treatment of PC patients with HRD by PARP inhibitors. Moreover, it has been shown that platinum-based chemotherapy is more effective in mCRPC patients with HRR gene alterations. This review highlights the prognosis and therapeutic implications of HRR gene alterations in PC.
Collapse
Affiliation(s)
- Hamidreza Saeidi
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, University of Putra Malaysia, Serdang, Malaysia.
| | - Ikmal Hisyam Bakrin
- Department of Pathology, Faculty of Medicine and Health Sciences, University of Putra Malaysia, Serdang, Malaysia
| | - Chandramathi Samudi Raju
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Patimah Ismail
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, University of Putra Malaysia, Serdang, Malaysia
| | - Mohsen Saraf
- Department of Biomedical Engineering, Faculty of Engineering, University of Isfahan, Isfahan, Iran.
| | - Mohd Ghani Khairul-Asri
- Department of Urology, Faculty of Medicine and Health Sciences, University of Putra Malaysia, Selangor, Malaysia
| |
Collapse
|
62
|
Parry PI, Lefringhausen A, Turni C, Neil CJ, Cosford R, Hudson NJ, Gillespie J. 'Spikeopathy': COVID-19 Spike Protein Is Pathogenic, from Both Virus and Vaccine mRNA. Biomedicines 2023; 11:2287. [PMID: 37626783 PMCID: PMC10452662 DOI: 10.3390/biomedicines11082287] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
The COVID-19 pandemic caused much illness, many deaths, and profound disruption to society. The production of 'safe and effective' vaccines was a key public health target. Sadly, unprecedented high rates of adverse events have overshadowed the benefits. This two-part narrative review presents evidence for the widespread harms of novel product COVID-19 mRNA and adenovectorDNA vaccines and is novel in attempting to provide a thorough overview of harms arising from the new technology in vaccines that relied on human cells producing a foreign antigen that has evidence of pathogenicity. This first paper explores peer-reviewed data counter to the 'safe and effective' narrative attached to these new technologies. Spike protein pathogenicity, termed 'spikeopathy', whether from the SARS-CoV-2 virus or produced by vaccine gene codes, akin to a 'synthetic virus', is increasingly understood in terms of molecular biology and pathophysiology. Pharmacokinetic transfection through body tissues distant from the injection site by lipid-nanoparticles or viral-vector carriers means that 'spikeopathy' can affect many organs. The inflammatory properties of the nanoparticles used to ferry mRNA; N1-methylpseudouridine employed to prolong synthetic mRNA function; the widespread biodistribution of the mRNA and DNA codes and translated spike proteins, and autoimmunity via human production of foreign proteins, contribute to harmful effects. This paper reviews autoimmune, cardiovascular, neurological, potential oncological effects, and autopsy evidence for spikeopathy. With many gene-based therapeutic technologies planned, a re-evaluation is necessary and timely.
Collapse
Affiliation(s)
- Peter I. Parry
- Children’s Health Research Clinical Unit, Faculty of Medicine, The University of Queensland, South Brisbane, QLD 4101, Australia
- Department of Psychiatry, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Astrid Lefringhausen
- Children’s Health Defence (Australia Chapter), Huskisson, NSW 2540, Australia; (A.L.); (R.C.); (J.G.)
| | - Conny Turni
- Microbiology Research, QAAFI (Queensland Alliance for Agriculture and Food Innovation), The University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Christopher J. Neil
- Department of Medicine, University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Robyn Cosford
- Children’s Health Defence (Australia Chapter), Huskisson, NSW 2540, Australia; (A.L.); (R.C.); (J.G.)
| | - Nicholas J. Hudson
- School of Agriculture and Food Science, The University of Queensland, Brisbane, QLD 4072, Australia;
| | - Julian Gillespie
- Children’s Health Defence (Australia Chapter), Huskisson, NSW 2540, Australia; (A.L.); (R.C.); (J.G.)
| |
Collapse
|
63
|
Zuhair Kassem T, Wunderle M, Kuhlmann L, Ruebner M, Huebner H, Hoyer J, Reis A, Fasching PA, Beckmann MW, Hack CC, Fietkau R, Distel L. Ex Vivo Chromosomal Radiosensitivity Testing in Patients with Pathological Germline Variants in Breast Cancer High-Susceptibility Genes BReast CAncer 1 and BReast CAncer 2. Curr Issues Mol Biol 2023; 45:6618-6633. [PMID: 37623237 PMCID: PMC10453196 DOI: 10.3390/cimb45080418] [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: 07/15/2023] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Individual radiosensitivity is an important factor in the occurrence of undesirable consequences of radiotherapy. The potential for increased radiosensitivity has been linked to highly penetrant heterozygous mutations in DNA repair genes such as BRCA1 and BRCA2. By studying the chromosomal radiosensitivity of BRCA1/2 mutation carriers compared to the general population, we study whether increased chromosomal radiation sensitivity is observed in patients with BRCA1/2 variants. METHODS Three-color-fluorescence in situ hybridization was performed on ex vivo-irradiated peripheral blood lymphocytes from 64 female patients with a heterozygous germline BRCA1 or BRCA2 mutation. Aberrations in chromosomes #1, #2 and #4 were analyzed. Mean breaks per metaphase (B/M) served as the parameter for chromosomal radiosensitivity. The results were compared with chromosomal radiosensitivity in a cohort of generally healthy individuals and patients with rectal cancer or breast cancer. RESULTS Patients with BRCA1/2 mutations (n = 64; B/M 0.47) overall showed a significantly higher chromosomal radiosensitivity than general healthy individuals (n = 211; B/M 0.41) and patients with rectal cancer (n = 379; B/M 0.44) and breast cancer (n = 147; B/M 0.45) without proven germline mutations. Chromosomal radiosensitivity varied depending on the locus of the BRCA1/2 mutation. CONCLUSIONS BRCA1/2 mutations result in slightly increased chromosomal sensitivity to radiation. A few individual patients have a marked increase in radiation sensitivity. Therefore, these patients are at a higher risk for adverse therapeutic consequences.
Collapse
Affiliation(s)
- Tara Zuhair Kassem
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 27, D-91054 Erlangen, Germany; (T.Z.K.); (L.K.); (R.F.)
| | - Marius Wunderle
- Department of Gynecology and Obstetrics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 27, D-91054 Erlangen, Germany; (M.W.); (M.R.); (H.H.); (P.A.F.); (M.W.B.); (C.C.H.)
| | - Lukas Kuhlmann
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 27, D-91054 Erlangen, Germany; (T.Z.K.); (L.K.); (R.F.)
| | - Matthias Ruebner
- Department of Gynecology and Obstetrics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 27, D-91054 Erlangen, Germany; (M.W.); (M.R.); (H.H.); (P.A.F.); (M.W.B.); (C.C.H.)
| | - Hanna Huebner
- Department of Gynecology and Obstetrics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 27, D-91054 Erlangen, Germany; (M.W.); (M.R.); (H.H.); (P.A.F.); (M.W.B.); (C.C.H.)
| | - Juliane Hoyer
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 10, D-91054 Erlangen, Germany; (J.H.); (A.R.)
| | - André Reis
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 10, D-91054 Erlangen, Germany; (J.H.); (A.R.)
- Centre for Rare Diseases Erlangen (ZSEER), Universitätsklinikum Erlangen, D-91054 Erlangen, Germany
| | - Peter A. Fasching
- Department of Gynecology and Obstetrics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 27, D-91054 Erlangen, Germany; (M.W.); (M.R.); (H.H.); (P.A.F.); (M.W.B.); (C.C.H.)
| | - Matthias W. Beckmann
- Department of Gynecology and Obstetrics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 27, D-91054 Erlangen, Germany; (M.W.); (M.R.); (H.H.); (P.A.F.); (M.W.B.); (C.C.H.)
| | - Carolin C. Hack
- Department of Gynecology and Obstetrics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 27, D-91054 Erlangen, Germany; (M.W.); (M.R.); (H.H.); (P.A.F.); (M.W.B.); (C.C.H.)
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 27, D-91054 Erlangen, Germany; (T.Z.K.); (L.K.); (R.F.)
| | - Luitpold Distel
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsstraße 27, D-91054 Erlangen, Germany; (T.Z.K.); (L.K.); (R.F.)
| |
Collapse
|
64
|
Xiong Y, Rao Y, Hu J, Luo Z, Chen C. Nanoparticle-Based Photothermal Therapy for Breast Cancer Noninvasive Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2305140. [PMID: 37561994 DOI: 10.1002/adma.202305140] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/29/2023] [Indexed: 08/12/2023]
Abstract
Rapid advancements in materials science and nanotechnology, intertwined with oncology, have positioned photothermal therapy (PTT) as a promising noninvasive treatment strategy for cancer. The breast's superficial anatomical location and aesthetic significance render breast cancer a particularly pertinent candidate for the clinical application of PTT following melanoma. This review comprehensively explores the research conducted on the various types of nanoparticles employed in PTT for breast cancer and elaborates on their specific roles and mechanisms of action. The integration of PTT with existing clinical therapies for breast cancer is scrutinized, underscoring its potential for synergistic outcomes. Additionally, the mechanisms underlying PTT and consequential modifications to the tumor microenvironment after treatment are elaborated from a medical perspective. Future research directions are suggested, with an emphasis on the development of integrative platforms that combine multiple therapeutic approaches and the optimization of nanoparticle synthesis for enhanced treatment efficacy. The goal is to push the boundaries of PTT toward a comprehensive, clinically applicable treatment for breast cancer.
Collapse
Affiliation(s)
- Yao Xiong
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, P. R. China
| | - Yan Rao
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University School of Medicine, Wuhan, Hubei, 430000, P. R. China
| | - Jiawei Hu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, P. R. China
| | - Zixuan Luo
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, P. R. China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No 238 Jiefang Road, Wuchang District, Wuhan, Hubei, 430060, P. R. China
| |
Collapse
|
65
|
Wineland D, Le AN, Hausler R, Kelly G, Barrett E, Desai H, Wubbenhorst B, Pluta J, Bastian P, Symecko H, D'Andrea K, Doucette A, Gabriel P, Reiss KA, Nayak A, Feldman M, Domchek SM, Nathanson KL, Maxwell KN. Biallelic BRCA Loss and Homologous Recombination Deficiency in Nonbreast/Ovarian Tumors in Germline BRCA1/2 Carriers. JCO Precis Oncol 2023; 7:e2300036. [PMID: 37535879 PMCID: PMC10581613 DOI: 10.1200/po.23.00036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/02/2023] [Accepted: 06/02/2023] [Indexed: 08/05/2023] Open
Abstract
PURPOSE Breast and ovarian tumors in germline BRCA1/2 carriers undergo allele-specific loss of heterozygosity, resulting in homologous recombination deficiency (HRD) and sensitivity to poly-ADP-ribose polymerase (PARP) inhibitors. This study investigated whether biallelic loss and HRD also occur in primary nonbreast/ovarian tumors that arise in germline BRCA1/2 carriers. METHODS A clinically ascertained cohort of BRCA1/2 carriers with a primary nonbreast/ovarian cancer was identified, including canonical (prostate and pancreatic cancers) and noncanonical (all other) tumor types. Whole-exome sequencing or clinical sequencing results (n = 45) were analyzed. A pan-cancer analysis of nonbreast/ovarian primary tumors from germline BRCA1/2 carriers from The Cancer Genome Atlas (TCGA, n = 73) was used as a validation cohort. RESULTS Ages of nonbreast/ovarian cancer diagnosis in germline BRCA1/2 carriers were similar to controls for the majority of cancer types. Nine of 45 (20%) primary nonbreast/ovarian tumors from germline BRCA1/2 carriers had biallelic loss of BRCA1/2 in the clinical cohort, and 23 of 73 (32%) in the TCGA cohort. In the combined cohort, 35% and 27% of primary canonical and noncanonical BRCA tumor types, respectively, had biallelic loss. High HRD scores (HRDex > 42) were detected in 81% of tumors with biallelic BRCA loss compared with 22% (P < .001) of tumors without biallelic BRCA loss. No differences in genomic profile, including mutational signatures, mutation spectrum, tumor mutational burden, or microsatellite instability, were found in primary nonbreast/ovarian tumors with or without biallelic BRCA1/2 loss. CONCLUSION A proportion of noncanonical primary tumors have biallelic loss and evidence of HRD. Our data suggest that assessment of biallelic loss and HRD could supplement identification of germline BRCA1/2 mutations in selection of patients for platinum or PARP inhibitor therapy.
Collapse
Affiliation(s)
- Dylane Wineland
- Arcadia University and Chester County Hospital, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Anh N. Le
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ryan Hausler
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Gregory Kelly
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Emanuel Barrett
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Heena Desai
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Bradley Wubbenhorst
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - John Pluta
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Paul Bastian
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Heather Symecko
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kurt D'Andrea
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Abigail Doucette
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Peter Gabriel
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kim A. Reiss
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Anupma Nayak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michael Feldman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Susan M. Domchek
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Katherine L. Nathanson
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kara N. Maxwell
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
66
|
Dholakia G, Meade J. A Pathogenic Germline BRCA1 Variant in a Patient With Cellular Congenital Mesoblastic Nephroma: A Case Report. Cureus 2023; 15:e43857. [PMID: 37736432 PMCID: PMC10510949 DOI: 10.7759/cureus.43857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2023] [Indexed: 09/23/2023] Open
Abstract
Benign cystic tumors of the kidney are well-described in infants and young children. Here we report an infant diagnosed with a cellular congenital mesoblastic nephroma (CMN) with a germline pathogenic variant in BRCA1. This finding is novel because BRCA1 is an adult-onset cancer predisposition gene causing breast, ovarian, pancreatic, and prostate cancers. However, increasing studies are indicating the presence of germline BRCA1 in both malignant and benign childhood cancers.
Collapse
Affiliation(s)
| | - Julia Meade
- Pediatrics, University of Pittsburgh Medical Center, Pittsburgh, USA
| |
Collapse
|
67
|
Ueki A, Yoshida R, Kosaka T, Matsubayashi H. Clinical risk management of breast, ovarian, pancreatic, and prostatic cancers for BRCA1/2 variant carriers in Japan. J Hum Genet 2023; 68:517-526. [PMID: 37088789 DOI: 10.1038/s10038-023-01153-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/21/2023] [Accepted: 04/13/2023] [Indexed: 04/25/2023]
Abstract
Opportunities for genetic counseling and germline BRCA1/2 (BRCA) testing are increasing in Japan owing to cancer genomic profiling testing and companion diagnostics being covered by national health insurance for patients with BRCA-related cancers. These tests are useful not only to judge whether platinum agents and PARP inhibitors are indicated but also to reveal an autosomal-dominant inherited cancer syndrome: hereditary breast and ovarian cancer. In individuals with germline BRCA variants, risk of cancers of the breast, ovary, pancreas, and prostate is significantly increased at various ages of onset, but the stomach, uterus, biliary tract, and skin might also be at risk. For women with pathogenic BRCA variants, breast awareness and image analyses should be initiated in their 20s, and risk-reducing procedures such as mastectomy are recommended starting in their 30s, with salpingo-oophorectomy in their late 30s. For male BRCA pathogenic variant carriers, prostatic surveillance should be applied using serum prostate-specific antigen starting in their 40s. For both sexes, image examinations ideally using endoscopic ultrasound and magnetic resonance cholangiopancreatography and blood testing should begin in their 50s for pancreatic surveillance. Homologous recombination pathway-associated genes are also causative candidates. Variant pathogenicity needs to be evaluated every 6-12 months when results are uncertain for clinical significance. Genetic counseling needs to be offered to the blood relatives of the pathogenic variant carriers with suitable timing. We review the recommended cross-organ BRCA risk management in Japan.
Collapse
Affiliation(s)
- Arisa Ueki
- Department of Clinical Genetics, The Cancer Institute Hospital of JFCR, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Reiko Yoshida
- Institute for Clinical Genetics and Genomics, Showa University, 1-5-8 Hatanodai Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroyuki Matsubayashi
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan.
| |
Collapse
|
68
|
Sia TY, Gordhandas SB, Birsoy O, Kemel Y, Maio A, Salo-Mullen E, Sheehan M, Hensley ML, Rubinstein M, Makker V, Grisham RN, O'Cearbhaill RE, Roche KL, Mueller JJ, Leitao MM, Sonoda Y, Chi DS, Abu-Rustum NR, Berger MF, Ellenson LH, Latham A, Stadler Z, Offit K, Aghajanian C, Weigelt B, Mandelker D, Liu YL. Germline drivers of gynecologic carcinosarcomas. Gynecol Oncol 2023; 174:34-41. [PMID: 37149903 PMCID: PMC10330315 DOI: 10.1016/j.ygyno.2023.04.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/09/2023]
Abstract
OBJECTIVES To describe the prevalence of germline pathogenic variants (gPVs) in endometrial and ovarian carcinosarcomas and determine if gPVs are drivers of carcinosarcoma. METHODS Patients with endometrial or ovarian carcinosarcomas who underwent clinical tumor-normal sequencing from 1/1/2015 to 6/1/2021 and consented to germline assessment of ≥76 cancer predisposition genes were included. In patients with gPVs, biallelic inactivation was identified through analysis of loss of heterozygosity and somatic pathogenic alterations. RESULTS Of 216 patients identified, 167 (77%) were diagnosed with endometrial carcinosarcoma and 49 (23%) with ovarian carcinosarcoma. Overall, 33 gPVs were observed in 29 patients (13%); 20 gPVs (61%) had biallelic loss in tumors. The rate of high-penetrance gPVs overall was 7% (16 of 216); 88% of high-penetrance gPVs had biallelic loss. In the endometrial carcinosarcoma cohort, 22 gPVs were found in 19 (11%) of 167 patients; 12 gPVs (55%) had biallelic loss in tumors, including 8 (89%) of 9 in high-penetrance gPVs. Among the ovarian carcinosarcoma cohort, 11 gPVs were found in 10 (20%) of 49 patients; 8 gPVs (73%) had biallelic loss in tumors, and all evaluable high-penetrance gPVs (n = 6) had biallelic loss. All gPVs in homologous recombination (BRCA1, BRCA2, RAD51C) and Lynch syndrome (MSH2, MSH6) genes had biallelic loss in tumors (n = 15). CONCLUSIONS gPVs in genes affecting homologous recombination- or Lynch-associated mismatch repair exhibited biallelic inactivation within tumors, suggesting likely drivers of gynecologic carcinosarcoma. Our data support germline testing for patients with gynecologic carcinosarcomas, given implications for treatment and risk-reduction in patients and at-risk family members.
Collapse
Affiliation(s)
- Tiffany Y Sia
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sushmita B Gordhandas
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ozge Birsoy
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yelena Kemel
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anna Maio
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Erin Salo-Mullen
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Margaret Sheehan
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martee L Hensley
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Maria Rubinstein
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Vicky Makker
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Rachel N Grisham
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Roisin E O'Cearbhaill
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Kara Long Roche
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Jennifer J Mueller
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Mario M Leitao
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Yukio Sonoda
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Dennis S Chi
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Nadeem R Abu-Rustum
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Michael F Berger
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lora H Ellenson
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alicia Latham
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Zsofia Stadler
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Kenneth Offit
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Carol Aghajanian
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Diana Mandelker
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ying L Liu
- Clinical Genetics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
69
|
Bucalo A, Conti G, Valentini V, Capalbo C, Bruselles A, Tartaglia M, Bonanni B, Calistri D, Coppa A, Cortesi L, Giannini G, Gismondi V, Manoukian S, Manzella L, Montagna M, Peterlongo P, Radice P, Russo A, Tibiletti MG, Turchetti D, Viel A, Zanna I, Palli D, Silvestri V, Ottini L. Male breast cancer risk associated with pathogenic variants in genes other than BRCA1/2: an Italian case-control study. Eur J Cancer 2023; 188:183-191. [PMID: 37262986 DOI: 10.1016/j.ejca.2023.04.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND Germline pathogenic variants (PVs) in BRCA1/2 genes are associated with breast cancer (BC) risk in both women and men. Multigene panel testing is being increasingly used for BC risk assessment, allowing the identification of PVs in genes other than BRCA1/2. While data on actionable PVs in other cancer susceptibility genes are now available in female BC, reliable data are still lacking in male BC (MBC). This study aimed to provide the patterns, prevalence and risk estimates associated with PVs in non-BRCA1/2 genes for MBC in order to improve BC prevention for male patients. METHODS We performed a large case-control study in the Italian population, including 767 BRCA1/2-negative MBCs and 1349 male controls, all screened using a custom 50 cancer gene panel. RESULTS PVs in genes other than BRCA1/2 were significantly more frequent in MBCs compared with controls (4.8% vs 1.8%, respectively) and associated with a threefold increased MBC risk (OR: 3.48, 95% CI: 1.88-6.44; p < 0.0001). PV carriers were more likely to have personal (p = 0.03) and family (p = 0.02) history of cancers, not limited to BC. PALB2 PVs were associated with a sevenfold increased MBC risk (OR: 7.28, 95% CI: 1.17-45.52; p = 0.034), and ATM PVs with a fivefold increased MBC risk (OR: 4.79, 95% CI: 1.12-20.56; p = 0.035). CONCLUSIONS This study highlights the role of PALB2 and ATM PVs in MBC susceptibility and provides risk estimates at population level. These data may help in the implementation of multigene panel testing in MBC patients and inform gender-specific BC risk management and decision making for patients and their families.
Collapse
Affiliation(s)
- Agostino Bucalo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Giulia Conti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Virginia Valentini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Carlo Capalbo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics Research Unit, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Daniele Calistri
- Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori"-IRST IRCCS, Meldola, Italy
| | - Anna Coppa
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Laura Cortesi
- Department of Oncology and Haematology, University of Modena and Reggio Emilia, Modena, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Viviana Gismondi
- Hereditary Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Siranoush Manoukian
- Unità di Genetica Medica, Dipartimento di Oncologia Medica ed Ematologia, Fondazione IRCCS Istituto Nazionale dei Tumori (INT), Milan, Italy
| | - Livia Manzella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Marco Montagna
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Paolo Peterlongo
- Genome Diagnostics Program, IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Paolo Radice
- Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Research, Fondazione IRCCS Istituto Nazionale Dei Tumori (INT), Milan, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical and Oncological Sciences, University of Palermo, Palermo, Italy
| | - Maria Grazia Tibiletti
- Dipartimento di Patologia, ASST Settelaghi and Centro di Ricerca per lo studio dei tumori eredo-familiari, Università dell'Insubria, Varese, Italy
| | - Daniela Turchetti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Alessandra Viel
- Unità di Oncogenetica e Oncogenomica Funzionale, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Ines Zanna
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Domenico Palli
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | | | - Laura Ottini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
70
|
Li Z, Jiao X, Robertson AG, Di Sante G, Ashton AW, DiRocco A, Wang M, Zhao J, Addya S, Wang C, McCue PA, South AP, Cordon-Cardo C, Liu R, Patel K, Hamid R, Parmar J, DuHadaway JB, Jones SJM, Casimiro MC, Schultz N, Kossenkov A, Phoon LY, Chen H, Lan L, Sun Y, Iczkowski KA, Rui H, Pestell RG. The DACH1 gene is frequently deleted in prostate cancer, restrains prostatic intraepithelial neoplasia, decreases DNA damage repair, and predicts therapy responses. Oncogene 2023; 42:1857-1873. [PMID: 37095257 PMCID: PMC10238272 DOI: 10.1038/s41388-023-02668-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 04/26/2023]
Abstract
Prostate cancer (PCa), the second leading cause of death in American men, includes distinct genetic subtypes with distinct therapeutic vulnerabilities. The DACH1 gene encodes a winged helix/Forkhead DNA-binding protein that competes for binding to FOXM1 sites. Herein, DACH1 gene deletion within the 13q21.31-q21.33 region occurs in up to 18% of human PCa and was associated with increased AR activity and poor prognosis. In prostate OncoMice, prostate-specific deletion of the Dach1 gene enhanced prostatic intraepithelial neoplasia (PIN), and was associated with increased TGFβ activity and DNA damage. Reduced Dach1 increased DNA damage in response to genotoxic stresses. DACH1 was recruited to sites of DNA damage, augmenting recruitment of Ku70/Ku80. Reduced Dach1 expression was associated with increased homology directed repair and resistance to PARP inhibitors and TGFβ kinase inhibitors. Reduced Dach1 expression may define a subclass of PCa that warrants specific therapies.
Collapse
Affiliation(s)
- Zhiping Li
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - A Gordon Robertson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, VSZ 4S6, Canada
- Dxige Research, Courtenay, BC, V9N 1C2, Canada
| | - Gabriele Di Sante
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Anthony W Ashton
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
- Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Wynnewood, PA, 19096, USA
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, 2006, Australia
| | - Agnese DiRocco
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Min Wang
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Jun Zhao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Sankar Addya
- Department of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - Chenguang Wang
- Department of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - Peter A McCue
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - Andrew P South
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Mt. Sinai, Hospital, 1468 Madison Ave., Floor 15, New York, NY, 10029, USA
| | - Runzhi Liu
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Kishan Patel
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Rasha Hamid
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Jorim Parmar
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - James B DuHadaway
- Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Wynnewood, PA, 19096, USA
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, VSZ 4S6, Canada
| | - Mathew C Casimiro
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
- Abraham Baldwin Agricultural College, Department of Science and Mathematics, Box 15, 2802 Moore Highway, Tifton, GA, 31794, USA
| | - Nikolaus Schultz
- Human Oncology and Pathogenesis Program, Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew Kossenkov
- Center for Systems and Computational Biology, The Wistar Institute, 3601 Spruce St., Philadelphia, PA, 19104, USA
| | - Lai Yee Phoon
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Hao Chen
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Li Lan
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Yunguang Sun
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Richard G Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA.
- The Wistar Cancer Center, Philadelphia, PA, 19104, USA.
| |
Collapse
|
71
|
Calvello M, Marabelli M, Gandini S, Marino E, Bernard L, Dal Molin M, Di Cola G, Zanzottera C, Corso G, Fazio N, Gervaso L, Fumagalli Romario U, Barberis M, Guerrieri-Gonzaga A, Bertario L, Serrano D, Bonanni B. Hereditary Gastric Cancer: Single-Gene or Multigene Panel Testing? A Mono-Institutional Experience. Genes (Basel) 2023; 14:genes14051077. [PMID: 37239438 DOI: 10.3390/genes14051077] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Gastric cancer (GC) has long been a 'Cinderella' among hereditary cancers. Until recently, single-gene testing (SGT) was the only approach to identify high-risk individuals. With the spread of multigene panel testing (MGPT), a debate arose on the involvement of other genes, particularly those pertaining to homologous recombination (HR) repair. We report our mono-institutional experience in genetic counseling and SGT for 54 GC patients, with the detection of nine pathogenic variants (PVs) (9/54:16.7%). Seven out of fifty (14%) patients who underwent SGT for unknown mutations were carriers of a PV in CDH1 (n = 3), BRCA2 (n = 2), BRCA1 (n = 1), and MSH2 (n = 1), while one patient (2%) carried two variants of unknown significance (VUSs). CDH1 and MSH2 emerged as genes involved in early-onset diffuse and later-onset intestinal GCs, respectively. We additionally conducted MGPT on 37 patients, identifying five PVs (13.5%), including three (3/5:60%) in an HR gene (BRCA2, ATM, RAD51D) and at least one VUS in 13 patients (35.1%). Comparing PV carriers and non-carriers, we observed a statistically significant difference in PVs between patients with and without family history of GC (p-value: 0.045) or Lynch-related tumors (p-value: 0.036). Genetic counseling remains central to GC risk assessment. MGPT appeared advantageous in patients with unspecific phenotypes, although it led to challenging results.
Collapse
Affiliation(s)
- Mariarosaria Calvello
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Monica Marabelli
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Sara Gandini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Elena Marino
- Clinic Unit of Oncogenomics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Loris Bernard
- Clinic Unit of Oncogenomics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Matteo Dal Molin
- Clinic Unit of Oncogenomics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giulia Di Cola
- Clinic Unit of Oncogenomics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Cristina Zanzottera
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giovanni Corso
- Division of Breast Surgery, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
- European Cancer Prevention Organization (ECP), 20122 Milan, Italy
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Lorenzo Gervaso
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Molecular Medicine Program, University of Pavia, 27100 Pavia, Italy
| | | | - Massimo Barberis
- Clinic Unit of Oncogenomics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Aliana Guerrieri-Gonzaga
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Lucio Bertario
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Davide Serrano
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| |
Collapse
|
72
|
Harrold E, Latham A, Pemmaraju N, Lieu CH. Early-Onset GI Cancers: Rising Trends, Genetic Risks, Novel Strategies, and Special Considerations. Am Soc Clin Oncol Educ Book 2023; 43:e398068. [PMID: 37235819 DOI: 10.1200/edbk_398068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Cancers in young adults (commonly described as early-onset [EO] cancer) represent a group of malignancies that have unique and challenging biology and genetic, treatment, social, and psychological implications. Even more concerning is a rising trend of EO cancers in multiple tumor types. Research and investigation in EO cancers will help elucidate mechanisms of carcinogenesis, differences in biology and response to treatment, and the need for multidisciplinary care to ensure comprehensive treatment and support for young patients.
Collapse
Affiliation(s)
- Emily Harrold
- Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Alicia Latham
- Memorial Sloan-Kettering Cancer Center, New York, NY
| | | | | |
Collapse
|
73
|
Zampiga V, Cangini I, Bandini E, Azzali I, Ravegnani M, Ravaioli A, Mancini S, Tebaldi M, Tedaldi G, Pirini F, Veneroni L, Frassineti GL, Falcini F, Danesi R, Calistri D, Arcangeli V. Prevalence of a BRCA2 Pathogenic Variant in Hereditary-Breast-and-Ovarian-Cancer-Syndrome Families with Increased Risk of Pancreatic Cancer in a Restricted Italian Area. Cancers (Basel) 2023; 15:cancers15072132. [PMID: 37046793 PMCID: PMC10093547 DOI: 10.3390/cancers15072132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/31/2023] [Accepted: 04/01/2023] [Indexed: 04/07/2023] Open
Abstract
PVs and LPVs in BRCA1/2 genes are correlated to a high risk of developing breast cancer and/or ovarian cancer (Hereditary Breast and Ovarian Cancer syndrome, HBOC); additionally, in recent years, an increasing number of BRCA 1/2 variants have been identified and associated with pancreatic cancer. Epidemiologic studies have highlighted that inherited factors are involved in 10% to 20% of PCs, mainly through deleterious variants of BRCA2. The frequency of BRCA1/2 germline alterations fluctuates quite a lot among different ethnic groups, and the estimated rate of PVs/LPVs variants in Italian HBOC families is not very accurate, according to different reports. The aim of our study is to describe the prevalence of a BRCA2 PV observed in a selected cohort of HBOC patients and their relatives, whose common origin is the eastern coast of Emilia Romagna, a region of Italy. This study provides insight into the frequency of the variant detected in this area and provides evidence of an increased risk of pancreatic and breast cancer, useful for genetic counseling and surveillance programs.
Collapse
Affiliation(s)
- Valentina Zampiga
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Ilaria Cangini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Erika Bandini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Irene Azzali
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Mila Ravegnani
- Romagna Cancer Registry, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Alessandra Ravaioli
- Romagna Cancer Registry, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Silvia Mancini
- Romagna Cancer Registry, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Michela Tebaldi
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Gianluca Tedaldi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Francesca Pirini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Luigi Veneroni
- Surgical Department, Infermi Hospital, 47923 Rimini, Italy
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Fabio Falcini
- Romagna Cancer Registry, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Rita Danesi
- Romagna Cancer Registry, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Daniele Calistri
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Valentina Arcangeli
- Romagna Cancer Registry, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| |
Collapse
|
74
|
Usui Y, Taniyama Y, Endo M, Koyanagi YN, Kasugai Y, Oze I, Ito H, Imoto I, Tanaka T, Tajika M, Niwa Y, Iwasaki Y, Aoi T, Hakozaki N, Takata S, Suzuki K, Terao C, Hatakeyama M, Hirata M, Sugano K, Yoshida T, Kamatani Y, Nakagawa H, Matsuda K, Murakami Y, Spurdle AB, Matsuo K, Momozawa Y. Helicobacter pylori, Homologous-Recombination Genes, and Gastric Cancer. N Engl J Med 2023; 388:1181-1190. [PMID: 36988593 DOI: 10.1056/nejmoa2211807] [Citation(s) in RCA: 75] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
BACKGROUND Helicobacter pylori infection is a well-known risk factor for gastric cancer. However, the contribution of germline pathogenic variants in cancer-predisposing genes and their effect, when combined with H. pylori infection, on the risk of gastric cancer has not been widely evaluated. METHODS We evaluated the association between germline pathogenic variants in 27 cancer-predisposing genes and the risk of gastric cancer in a sample of 10,426 patients with gastric cancer and 38,153 controls from BioBank Japan. We also assessed the combined effect of pathogenic variants and H. pylori infection status on the risk of gastric cancer and calculated the cumulative risk in 1433 patients with gastric cancer and 5997 controls from the Hospital-based Epidemiologic Research Program at Aichi Cancer Center (HERPACC). RESULTS Germline pathogenic variants in nine genes (APC, ATM, BRCA1, BRCA2, CDH1, MLH1, MSH2, MSH6, and PALB2) were associated with the risk of gastric cancer. We found an interaction between H. pylori infection and pathogenic variants in homologous-recombination genes with respect to the risk of gastric cancer in the sample from HERPACC (relative excess risk due to the interaction, 16.01; 95% confidence interval [CI], 2.22 to 29.81; P = 0.02). At 85 years of age, persons with H. pylori infection and a pathogenic variant had a higher cumulative risk of gastric cancer than noncarriers infected with H. pylori (45.5% [95% CI, 20.7 to 62.6] vs. 14.4% [95% CI, 12.2 to 16.6]). CONCLUSIONS H. pylori infection modified the risk of gastric cancer associated with germline pathogenic variants in homologous-recombination genes. (Funded by the Japan Agency for Medical Research and Development and others.).
Collapse
Affiliation(s)
- Yoshiaki Usui
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yukari Taniyama
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Mikiko Endo
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yuriko N Koyanagi
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yumiko Kasugai
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Isao Oze
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Hidemi Ito
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Issei Imoto
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Tsutomu Tanaka
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Masahiro Tajika
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yasumasa Niwa
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yusuke Iwasaki
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Tomomi Aoi
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Nozomi Hakozaki
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Sadaaki Takata
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Kunihiko Suzuki
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Chikashi Terao
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Masanori Hatakeyama
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Makoto Hirata
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Kokichi Sugano
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Teruhiko Yoshida
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yoichiro Kamatani
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Hidewaki Nakagawa
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Koichi Matsuda
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yoshinori Murakami
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Amanda B Spurdle
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Keitaro Matsuo
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| | - Yukihide Momozawa
- From the Laboratories for Genotyping Development (Y.U., M.E., Y.I., T.A., N.H., S.T., K. Suzuki, Y. Momozawa), Statistical and Translational Genetics (C.T.), and Cancer Genomics (H.N.), RIKEN Center for Integrative Medical Sciences, Yokohama, the Divisions of Cancer Information and Control (Y.U., Y.T., Y.N.K., H.I.) and Cancer Epidemiology and Prevention (Y. Kasugai, I.O., K. Matsuo), Department of Preventive Medicine, Aichi Cancer Center, the Divisions of Cancer Epidemiology (Y. Kasugai, K. Matsuo) and Descriptive Cancer Epidemiology (H.I.), Nagoya University Graduate School of Medicine, Aichi Cancer Center Research Institute (I.I.), and the Department of Endoscopy (T.T., M.T.), Aichi Cancer Center Hospital (Y.N.), Nagoya, the Department of Hematology, Oncology, and Respiratory Medicine, Okayama University Medical School, Okayama (Y.U.), the Laboratory of Microbial Carcinogenesis, Institute of Microbial Chemistry, Microbial Chemistry Research Foundation (M. Hatakeyama), the Department of Genetic Medicine and Services, National Cancer Center Hospital (M. Hirata, K. Sugano, T.Y.), the Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science (M. Hirata, Y. Murakami), and the Laboratories of Complex Trait Genomics (Y. Kamatani) and Clinical Genome Sequencing (K. Matsuda), Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, and the Department of Genetic Medicine, Kyoundo Hospital, Sasaki Foundation (K. Sugano), Tokyo, and the Research Center of Infection-Associated Cancer, Institute for Genetic Medicine, Hokkaido University, Sapporo (M. Hatakeyama) - all in Japan; and the Population Health Program, QIMR (Queensland Institute of Medical Research) Berghofer Medical Research Institute, Brisbane, Australia (A.B.S.)
| |
Collapse
|
75
|
Liu J, Yao L, Sun J, Hu L, Chen J, Zhang J, Xu Y, Xie Y. Cancer risk in relatives of BRCA1/2 pathogenic variant carriers in a large series of unselected patients with breast cancer. Cancer Biol Med 2023; 20:j.issn.2095-3941.2022.0593. [PMID: 36861435 PMCID: PMC9978892 DOI: 10.20892/j.issn.2095-3941.2022.0593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
OBJECTIVE The spectrum and risk of cancer in relatives of BRCA1/2 pathogenic variant carriers in the Chinese population have not been established. METHODS A family history of cancer in 9903 unselected breast cancer patients was retrospectively analyzed. BRCA1/2 status was determined for all patients and relative risks (RRs) were calculated to evaluate cancer risk in relatives of the patients. RESULTS The incidences of breast cancer in female relatives of BRCA1 carriers, BRCA2 carriers, and non-carriers were 33.0%, 32.2%, and 7.7%, respectively. The corresponding incidences of ovarian cancer were 11.5%, 2.4%, and 0.5%, respectively. The incidences of pancreatic cancer in male relatives of BRCA1 carriers, BRCA2 carriers, and non-carriers were 1.4%, 2.7%, and 0.6%, respectively. The corresponding incidences of prostate cancer were 1.0%, 2.1%, and 0.4%, respectively. The risks of breast and ovarian cancers in female relatives of BRCA1 and BRCA2 carriers were significantly higher than female relatives of non-carriers (BRCA1: RR = 4.29, P < 0.001 and RR = 21.95, P < 0.001; BRCA2: RR = 4.19, P < 0.001 and RR = 4.65, P < 0.001, respectively). Additionally, higher risks of pancreatic and prostate cancers were noted in male relatives of BRCA2 carriers than non-carriers (RR = 4.34, P = 0.001 and RR = 4.86, P = 0.001, respectively). CONCLUSIONS Female relatives of BRCA1 and BRCA2 carriers are at increased risk for breast and ovarian cancers, and male relatives of BRCA2 carriers are at increased risk for pancreatic and prostate cancers.
Collapse
Affiliation(s)
- Jiaming Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Familial & Hereditary Cancer Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lu Yao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Familial & Hereditary Cancer Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jie Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Familial & Hereditary Cancer Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Li Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Familial & Hereditary Cancer Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jiuan Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Familial & Hereditary Cancer Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Juan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Familial & Hereditary Cancer Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ye Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Familial & Hereditary Cancer Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yuntao Xie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Familial & Hereditary Cancer Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
- Correspondence to: Yuntao Xie, E-mail:
| |
Collapse
|
76
|
Imyanitov EN, Kuligina ES, Sokolenko AP, Suspitsin EN, Yanus GA, Iyevleva AG, Ivantsov AO, Aleksakhina SN. Hereditary cancer syndromes. World J Clin Oncol 2023; 14:40-68. [PMID: 36908677 PMCID: PMC9993141 DOI: 10.5306/wjco.v14.i2.40] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/09/2022] [Accepted: 02/14/2023] [Indexed: 02/21/2023] Open
Abstract
Hereditary cancer syndromes (HCSs) are arguably the most frequent category of Mendelian genetic diseases, as at least 2% of presumably healthy subjects carry highly-penetrant tumor-predisposing pathogenic variants (PVs). Hereditary breast-ovarian cancer and Lynch syndrome make the highest contribution to cancer morbidity; in addition, there are several dozen less frequent types of familial tumors. The development of the majority albeit not all hereditary malignancies involves two-hit mechanism, i.e. the somatic inactivation of the remaining copy of the affected gene. Earlier studies on cancer families suggested nearly fatal penetrance for the majority of HCS genes; however, population-based investigations and especially large-scale next-generation sequencing data sets demonstrate that the presence of some highly-penetrant PVs is often compatible with healthy status. Hereditary cancer research initially focused mainly on cancer detection and prevention. Recent studies identified multiple HCS-specific drug vulnerabilities, which translated into the development of highly efficient therapeutic options.
Collapse
Affiliation(s)
- Evgeny N Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Ekaterina S Kuligina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Anna P Sokolenko
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Evgeny N Suspitsin
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Grigoriy A Yanus
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Aglaya G Iyevleva
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Alexandr O Ivantsov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| | - Svetlana N Aleksakhina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg 197758, Russia
- Department of Clinical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg 194100, Russia
| |
Collapse
|
77
|
Paiella S, Azzolina D, Gregori D, Malleo G, Golan T, Simeone DM, Davis MB, Vacca PG, Crovetto A, Bassi C, Salvia R, Biankin AV, Casolino R. A systematic review and meta-analysis of germline BRCA mutations in pancreatic cancer patients identifies global and racial disparities in access to genetic testing. ESMO Open 2023; 8:100881. [PMID: 36822114 PMCID: PMC10163165 DOI: 10.1016/j.esmoop.2023.100881] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Germline BRCA1 and BRCA2 mutations (gBRCAm) can inform pancreatic cancer (PC) risk and treatment but most of the available information is derived from white patients. The ethnic and geographic variability of gBRCAm prevalence and of germline BRCA (gBRCA) testing uptake in PC globally is largely unknown. MATERIALS AND METHODS We carried out a systematic review and prevalence meta-analysis of gBRCA testing and gBRCAm prevalence in PC patients stratified by ethnicity. The main outcome was the distribution of gBRCA testing uptake across diverse populations worldwide. Secondary outcomes included: geographic distribution of gBRCA testing uptake, temporal analysis of gBRCA testing uptake in ethnic groups, and pooled proportion of gBRCAm stratified by ethnicity. The study is listed under PROSPERO registration number #CRD42022311769. RESULTS A total of 51 studies with 16 621 patients were included. Twelve of the studies (23.5%) enrolled white patients only, 10 Asians only (19.6%), and 29 (56.9%) included mixed populations. The pooled prevalence of white, Asian, African American, and Hispanic patients tested per study was 88.7%, 34.8%, 3.6%, and 5.2%, respectively. The majority of included studies were from high-income countries (HICs) (64; 91.2%). Temporal analysis showed a significant increase only in white and Asians patients tested from 2000 to present (P < 0.001). The pooled prevalence of gBRCAm was: 3.3% in white, 1.7% in Asian, and negligible (<0.3%) in African American and Hispanic patients. CONCLUSIONS Data on gBRCA testing and gBRCAm in PC derive mostly from white patients and from HICs. This limits the interpretation of gBRCAm for treating PC across diverse populations and implies substantial global and racial disparities in access to BRCA testing in PC.
Collapse
Affiliation(s)
- S Paiella
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona. https://twitter.com/Totuccio83
| | - D Azzolina
- Department of Environmental and Preventive Science, University of Ferrara, Ferrara
| | - D Gregori
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, Padova, Italy. https://twitter.com/gregoriDario
| | - G Malleo
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona. https://twitter.com/gimalleo
| | - T Golan
- Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - D M Simeone
- Department of Surgery, New York University, New York; Perlmutter Cancer Center, New York University, New York. https://twitter.com/MadameSurgeon
| | - M B Davis
- Department of Surgery and Surgical Oncology, Weill Cornell University, New York; Englander Institute of Precision Medicine, Weill Cornell University, New York, USA. https://twitter.com/MeliD32
| | - P G Vacca
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona. https://twitter.com/pvhdfm
| | - A Crovetto
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona. https://twitter.com/crovetto_a
| | - C Bassi
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona
| | - R Salvia
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona. https://twitter.com/SalviaRobi
| | - A V Biankin
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow; West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK; Faculty of Medicine, South Western Sydney Clinical School, University of NSW, Liverpool, Australia.
| | - R Casolino
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow.
| |
Collapse
|
78
|
Stoffel EM, Brand RE, Goggins M. Pancreatic Cancer: Changing Epidemiology and New Approaches to Risk Assessment, Early Detection, and Prevention. Gastroenterology 2023; 164:752-765. [PMID: 36804602 DOI: 10.1053/j.gastro.2023.02.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/23/2023]
Abstract
Pancreatic cancer usually results in poor survival with limited options for treatment, as most affected individuals present with advanced disease. Early detection of preinvasive pancreatic neoplasia and identifying molecular therapeutic targets provide opportunities for extending survival. Although screening for pancreatic cancer is currently not recommended for the general population, emerging evidence indicates that pancreatic surveillance can improve outcomes for individuals in certain high-risk groups. Changes in the epidemiology of pancreatic cancer, experience from pancreatic surveillance, and discovery of novel biomarkers provide a roadmap for new strategies for pancreatic cancer risk assessment, early detection, and prevention.
Collapse
Affiliation(s)
- Elena M Stoffel
- Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan.
| | - Randall E Brand
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Michael Goggins
- Departments of Medicine and Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
79
|
Allen I, Hassan H, Sofianopoulou E, Eccles D, Turnbull C, Tischkowitz M, Pharoah P, Antoniou AC. Risks of second non-breast primaries following breast cancer in women: a systematic review and meta-analysis. Breast Cancer Res 2023; 25:18. [PMID: 36765408 PMCID: PMC9912682 DOI: 10.1186/s13058-023-01610-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/25/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Second primary cancer incidence is rising among breast cancer survivors. We examined the risks of non-breast second primaries, in combination and at specific cancer sites, through a systematic review and meta-analysis. METHODS We conducted a systematic search of PubMed, Embase, and Web of Science, seeking studies published by March 2022. We included studies that reported standardized incidence ratios (SIRs), with associated standard errors, assessing the combined risk of second non-breast primaries following breast cancer. We performed meta-analyses of combined second primary risks, stratifying by age, follow-up duration, and geographic region. We also assessed second primary risks at several specific sites, stratifying by age. The inverse variance method with DerSimonian-Laird estimators was used in all meta-analyses, assuming a random-effects model. Associated biases and study quality were evaluated using the Newcastle-Ottawa scale. RESULTS One prospective and twenty-seven retrospective cohort studies were identified. SIRs for second non-breast primaries combined ranged from 0.84 to 1.84. The summary SIR estimate was 1.24 (95% CI 1.14-1.36, I2: 99%). This varied by age: the estimate was 1.59 (95% CI 1.36-1.85) when breast cancer was diagnosed before age 50, which was significantly higher than in women first diagnosed at 50 or over (SIR: 1.13, 95% CI 1.01-1.36, p for difference: < 0.001). SPC risks were also significantly higher when based on Asian, rather than European, registries (Asia-SIR: 1.47, 95% CI 1.29-1.67. Europe-SIR: 1.16, 95% CI 1.04-1.28). There were significantly increased risks of second thyroid (SIR: 1.89, 95% CI 1.49-2.38), corpus uteri (SIR: 1.84, 95% CI 1.53-2.23), ovary (SIR: 1.53, 95% CI 1.35-1.73), kidney (SIR: 1.43, 95% CI 1.17-1.73), oesophagus (SIR: 1.39, 95% CI 1.26-1.55), skin (melanoma) (SIR: 1.34, 95% CI 1.18-1.52), blood (leukaemia) (SIR: 1.30, 95% CI 1.17-1.45), lung (SIR: 1.25, 95% CI 1.03-1.51), stomach (SIR: 1.23, 95% CI 1.12-1.36) and bladder (SIR: 1.15, 95% CI 1.05-1.26) primaries. CONCLUSIONS Breast cancer survivors are at significantly increased risk of second primaries at many sites. Risks are higher for those diagnosed with breast cancer before age 50 and in Asian breast cancer survivors compared to European breast cancer survivors. This study is limited by a lack of data on potentially confounding variables. The conclusions may inform clinical management decisions following breast cancer, although specific clinical recommendations lie outside the scope of this review.
Collapse
Affiliation(s)
- Isaac Allen
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, CB1 8RN, UK.
| | - Hend Hassan
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Eleni Sofianopoulou
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Diana Eccles
- Department of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Clare Turnbull
- Translational Genetics Team, Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Marc Tischkowitz
- Department of Medical Genetics, Cambridge Biomedical Research Centre, National Institute for Health Research, University of Cambridge, Cambridge, UK
| | - Paul Pharoah
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Antonis C Antoniou
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, CB1 8RN, UK
| |
Collapse
|
80
|
Li Z, Jiao X, Robertson AG, Sante GD, Ashton AW, DiRocco A, Wang M, Zhao J, Addya S, Wang C, McCue PA, South AP, Cordon-Cardo C, Liu R, Patel K, Hamid R, Parmar J, DuHadaway JB, Jones SJ, Casimiro MC, Schultz N, Kossenkov A, Phoon LY, Chen H, Lan L, Sun Y, Iczkowski KA, Rui H, Pestell RG. The DACH1 gene is frequently deleted in prostate cancer, restrains prostatic intraepithelial neoplasia, decreases DNA damage repair, and predicts therapy responses. RESEARCH SQUARE 2023:rs.3.rs-2423179. [PMID: 36712010 PMCID: PMC9882663 DOI: 10.21203/rs.3.rs-2423179/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Prostate cancer (PCa), the second leading cause of death in American men, includes distinct genetic subtypes with distinct therapeutic vulnerabilities. The DACH1 gene encodes a winged helix/Forkhead DNA-binding protein that competes for binding to FOXM1 sites. Herein, DACH1 gene deletion within the 13q21.31-q21.33 region occurs in up to 18% of human PCa and was associated with increased AR activity and poor prognosis. In prostate OncoMice, prostate-specific deletion of the Dach1 gene enhanced prostatic intraepithelial neoplasia (PIN), and was associated with increased TGFb activity and DNA damage. Reduced Dach1 increased DNA damage in response to genotoxic stresses. DACH1 was recruited to sites of DNA damage, augmenting recruitment of Ku70/Ku80. Reduced Dach1 expression was associated with increased homology directed repair and resistance to PARP inhibitors and TGFb kinase inhibitors. Reduced Dach1 expression may define a subclass of PCa that warrants specific therapies.
Collapse
Affiliation(s)
- Zhiping Li
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - A. Gordon Robertson
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC VSZ 4S6, Canada
| | - Gabriele Di Sante
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Anthony W. Ashton
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia; Sydney Medical School Northern, University of Sydney, NSW, 2006, Australia
- Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Wynnewood, PA 19096
| | - Agnese DiRocco
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Min Wang
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Jun Zhao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Sankar Addya
- Department of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10 Street, Philadelphia, PA 19107
| | - Chenguang Wang
- Department of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10 Street, Philadelphia, PA 19107
| | - Peter A. McCue
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10 Street, Philadelphia, PA 19107
| | - Andrew P. South
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10 Street, Philadelphia, PA 19107
| | - Carlos Cordon-Cardo
- Department of Pathology, Mt. Sinai, Hospital, 1468 Madison Ave., Floor 15, New York, NY, 10029
| | - Runzhi Liu
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Kishan Patel
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Rasha Hamid
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - Jorim Parmar
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
| | - James B. DuHadaway
- Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Wynnewood, PA 19096
| | - Steven J. Jones
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC VSZ 4S6, Canada
| | - Mathew C. Casimiro
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
- Abraham Baldwin Agricultural College, Department of Science and Mathematics, Box 15, 2802 Moore Highway, Tifton, GA, 31794
| | - Nikolaus Schultz
- Human Oncology and Pathogenesis Program, Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrew Kossenkov
- Center for Systems and Computational Biology, The Wistar Institute, 3601 Spruce St., Philadelphia, PA 19104, USA
| | - Lai Yee Phoon
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA, and Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Hao Chen
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA, and Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Li Lan
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA, and Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Yunguang Sun
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Richard G. Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902 Pennsylvania
- The Wistar Cancer Center, Philadelphia, PA 19107
| |
Collapse
|
81
|
Prostate cancer risk, screening and management in patients with germline BRCA1/2 mutations. Nat Rev Urol 2023; 20:205-216. [PMID: 36600087 DOI: 10.1038/s41585-022-00680-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2022] [Indexed: 01/05/2023]
Abstract
Mutations in the BRCA1 and BRCA2 tumour suppressor genes are associated with prostate cancer risk; however, optimal screening protocols for individuals with these mutations have been a subject of debate. Several prospective studies of prostate cancer incidence and screening among BRCA1/2 mutation carriers have indicated at least a twofold to fourfold increase in prostate cancer risk among carriers of BRCA2 mutations compared with the general population. Moreover, BRCA2 mutations are associated with more aggressive, high-grade disease characteristics at diagnosis, more aggressive clinical behaviour and greater prostate cancer-specific mortality. The risk for BRCA1 mutations seems to be attenuated compared with BRCA2. Prostate-specific antigen (PSA) measurement or prostate magnetic resonance imaging (MRI) alone is an imperfect indicator of clinically significant prostate cancer; therefore, BRCA1/2 mutation carriers might benefit from refined risk stratification strategies. However, the long-term impact of prostate cancer screening is unknown, and the optimal management of BRCA1/2 carriers with prostate cancer has not been defined. Whether timely localized therapy can improve overall survival in the screened population is uncertain. Long-term results of prospective studies are awaited to confirm the optimal screening strategies and benefits of prostate cancer screening among BRCA1/2 mutation carriers, and whether these approaches ultimately have a positive impact on survival and quality of life in these patients.
Collapse
|
82
|
Mazer BL, Lee JW, Roberts NJ, Chu LC, Lennon AM, Klein AP, Eshleman JR, Fishman EK, Canto MI, Goggins MG, Hruban RH. Screening for pancreatic cancer has the potential to save lives, but is it practical? Expert Rev Gastroenterol Hepatol 2023; 17:555-574. [PMID: 37212770 PMCID: PMC10424088 DOI: 10.1080/17474124.2023.2217354] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/21/2023] [Accepted: 05/19/2023] [Indexed: 05/23/2023]
Abstract
INTRODUCTION Most patients with pancreatic cancer present with advanced stage, incurable disease. However, patients with high-grade precancerous lesions and many patients with low-stage disease can be cured with surgery, suggesting that early detection has the potential to improve survival. While serum CA19.9 has been a long-standing biomarker used for pancreatic cancer disease monitoring, its low sensitivity and poor specificity have driven investigators to hunt for better diagnostic markers. AREAS COVERED This review will cover recent advances in genetics, proteomics, imaging, and artificial intelligence, which offer opportunities for the early detection of curable pancreatic neoplasms. EXPERT OPINION From exosomes, to circulating tumor DNA, to subtle changes on imaging, we know much more now about the biology and clinical manifestations of early pancreatic neoplasia than we did just five years ago. The overriding challenge, however, remains the development of a practical approach to screen for a relatively rare, but deadly, disease that is often treated with complex surgery. It is our hope that future advances will bring us closer to an effective and financially sound approach for the early detection of pancreatic cancer and its precursors.
Collapse
Affiliation(s)
- Benjamin L. Mazer
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jae W. Lee
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nicholas J. Roberts
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Linda C. Chu
- Department of Radiology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anne Marie Lennon
- Department of Medicine, Division of Gastroenterology and Hepatology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alison P. Klein
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James R. Eshleman
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elliot K. Fishman
- Department of Radiology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marcia Irene Canto
- Department of Medicine, Division of Gastroenterology and Hepatology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael G. Goggins
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ralph H. Hruban
- The Sol Goldman Pancreatic Cancer Research Center, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
83
|
Li C, Zhang L, Zhuo Z, Su F, Li H, Xu S, Liu Y, Zhang Z, Xie Y, Yu X, Bian L, Xiao F. Artificial intelligence-based recognition for variant pathogenicity of BRCA1 using AlphaFold2-predicted structures. Theranostics 2023; 13:391-402. [PMID: 36593954 PMCID: PMC9800725 DOI: 10.7150/thno.79362] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022] Open
Abstract
With the surge of the high-throughput sequencing technologies, many genetic variants have been identified in the past decade. The vast majority of these variants are defined as variants of uncertain significance (VUS), as their significance to the function or health of an organism is not known. It is urgently needed to develop intelligent models for the clinical interpretation of VUS. State-of-the-art artificial intelligence (AI)-based variant effect predictors only learn features from primary amino acid sequences, leaving out information about the most important three-dimensional structure that is more related to its function. Methods: We proposed a deep convolutional neural network model named variant effect recognition network for BRCA1 (vERnet-B) to recognize the clinical pathogenicity of missense single-nucleotide variants in the BRCT domain of BRCA1. vERnet-B learned features associated with the pathogenicity from the tertiary protein structures of variants predicted by AlphaFold2. Results: After performing a series of validation and analyses on vERnet-B, we discovered that it exhibited significant advances over previous works. Recognizing the phenotypic consequences of VUS is one of the most daunting challenges in genetic informatics; however, we achieved 85% accuracy in recognizing disease BRCA1 variants with an ideal balance of false-positive and true-positive detection rates. vERnet-B correctly recognized the pathogenicity of variant A1708E, which was poorly predicted by AlphaFold2 as previously described. The vERnet-B web server is freely available from URL: http://ai-lab.bjrz.org.cn/vERnet. Conclusions: We applied protein tertiary structures to successfully recognize the pathogenic missense SNVs, which were difficult to be addressed by classical approaches based on sequences. Our work demonstrated that AlphaFold2-predicted structures were expected to be used for rich feature learning and revealed unique insights into the clinical interpretation of VUS in disease-related genes, using vERnet-B as a discovery tool.
Collapse
Affiliation(s)
- Chang Li
- Peking University Fifth School of Clinical Medicine, Beijing, China.,Clinical Biobank, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.,The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Lili Zhang
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.,The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhongling Zhuo
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China.,The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Su
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Hexin Li
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Siyuan Xu
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Ye Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Zaifeng Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yibo Xie
- Information Center, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xue Yu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.,✉ Corresponding authors: Prof. Xue Yu, Department of Cardiology, Beijing Hospital, Beijing, China, ; Prof. Liheng Bian, Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China, ; Prof. Fei Xiao, Peking University Fifth School of Clinical Medicine, Beijing, China, Phone: (86)10-58115083,
| | - Liheng Bian
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China.,✉ Corresponding authors: Prof. Xue Yu, Department of Cardiology, Beijing Hospital, Beijing, China, ; Prof. Liheng Bian, Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China, ; Prof. Fei Xiao, Peking University Fifth School of Clinical Medicine, Beijing, China, Phone: (86)10-58115083,
| | - Fei Xiao
- Peking University Fifth School of Clinical Medicine, Beijing, China.,Clinical Biobank, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.,The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.,✉ Corresponding authors: Prof. Xue Yu, Department of Cardiology, Beijing Hospital, Beijing, China, ; Prof. Liheng Bian, Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China, ; Prof. Fei Xiao, Peking University Fifth School of Clinical Medicine, Beijing, China, Phone: (86)10-58115083,
| |
Collapse
|
84
|
Truong H, Breen K, Nandakumar S, Sjoberg DD, Kemel Y, Mehta N, Lenis AT, Reisz PA, Carruthers J, Benfante N, Joseph V, Khurram A, Gopalan A, Fine SW, Reuter VE, Vickers AJ, Birsoy O, Liu Y, Walsh M, Latham A, Mandelker D, Stadler ZK, Pietzak E, Ehdaie B, Touijer KA, Laudone VP, Slovin SF, Autio KA, Danila DC, Rathkopf DE, Eastham JA, Chen Y, Morris MJ, Offit K, Solit DB, Scher HI, Abida W, Robson ME, Carlo MI. Gene-based Confirmatory Germline Testing Following Tumor-only Sequencing of Prostate Cancer. Eur Urol 2023; 83:29-38. [PMID: 36115772 PMCID: PMC10208030 DOI: 10.1016/j.eururo.2022.08.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Tumor-only genomic profiling is an important tool in therapeutic management of men with prostate cancer. Since clinically actionable germline variants may be reflected in tumor profiling, it is critical to identify which variants have a higher risk of being germline in origin to better counsel patients and prioritize genetic testing. OBJECTIVE To determine when variants found on tumor-only sequencing of prostate cancers should prompt confirmatory germline testing. DESIGN, SETTING, AND PARTICIPANTS Men with prostate cancer who underwent both tumor and germline sequencing at Memorial Sloan Kettering Cancer Center from January 1, 2015 to January 31, 2020 were evaluated. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Tumor and germline profiles were analyzed for pathogenic and likely pathogenic ("pathogenic") variants in 60 moderate- or high-penetrance genes associated with cancer predisposition. The germline probability (germline/germline + somatic) of a variant was calculated for each gene. Clinical and pathologic factors were analyzed as potential modifiers of germline probability. RESULTS AND LIMITATIONS Of the 1883 patients identified, 1084 (58%) had a somatic or germline pathogenic variant in one of 60 cancer susceptibility genes, and of them, 240 (22%) had at least one germline variant. Overall, the most frequent variants were in TP53, PTEN, APC, BRCA2, RB1, ATM, and CHEK2. Variants in TP53, PTEN, or RB1 were identified in 746 (40%) patients and were exclusively somatic. Variants with the highest germline probabilities were in PALB2 (69%), MITF (62%), HOXB13 (60%), CHEK2 (55%), BRCA1 (55%), and BRCA2 (47%), and the overall germline probability of a variant in any DNA damage repair gene was 40%. Limitations were that most of the men included in the cohort had metastatic disease, and different thresholds for pathogenicity exist for somatic and germline variants. CONCLUSIONS Of patients with pathogenic variants found on prostate tumor sequencing, 22% had clinically actionable germline variants, for which the germline probabilities varied widely by gene. Our results provide an evidenced-based clinical framework to prioritize referral to genetic counseling following tumor-only sequencing. PATIENT SUMMARY Patients with advanced prostate cancer are recommended to have germline genetic testing. Genetic sequencing of a patient's prostate tumor may also identify certain gene variants that are inherited. We found that patients who had variants in certain genes, such as ones that function in DNA damage repair, identified in their prostate tumor sequencing, had a high risk for having an inherited cancer syndrome.
Collapse
Affiliation(s)
- Hong Truong
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kelsey Breen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Subhiksha Nandakumar
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel D Sjoberg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yelena Kemel
- Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nikita Mehta
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew T Lenis
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Peter A Reisz
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jessica Carruthers
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicole Benfante
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vijai Joseph
- Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aliya Khurram
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anuradha Gopalan
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samson W Fine
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Victor E Reuter
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew J Vickers
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ozge Birsoy
- Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ying Liu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Walsh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alicia Latham
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Diana Mandelker
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eugene Pietzak
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Behfar Ehdaie
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karim A Touijer
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vincent P Laudone
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Susan F Slovin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karen A Autio
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel C Danila
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dana E Rathkopf
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James A Eastham
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu Chen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael J Morris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth Offit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B Solit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Howard I Scher
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wassim Abida
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark E Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria I Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Niehaus Center for Inherited Cancer Genomics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
85
|
Gastric Cancer Risk and Pathogenesis in BRCA1 and BRCA2 Carriers. Cancers (Basel) 2022; 14:cancers14235953. [PMID: 36497436 PMCID: PMC9736932 DOI: 10.3390/cancers14235953] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022] Open
Abstract
Carriers of a pathogenic germline variant (PV) in BRCA1 or BRCA2 are at increased risk for a number of malignancies, including breast, ovarian, pancreatic, and prostate cancer. In this review, we discuss emerging evidence that BRCA2 PV carriers, and likely also BRCA1 PV carriers, are also at increased risk for gastric cancer (GC), highlighting that GC may be part of the BRCA1/2 cancer risk spectrum. While the pathogenesis of GC among BRCA1/2 PV carriers remains unclear, increasing evidence reveals that GCs are often enriched with mutations in homologous recombination-associated genes such as BRCA1/2, and that GC prognosis and response to certain therapies can depend on BRCA1/2 expression. Given the strength of data published to date, a risk management strategy for GC among BRCA1/2 PV carriers is needed, and herein we also propose a potential strategy for GC risk management in this population. Moving forward, further study is clearly warranted to define the mechanistic relationship between BRCA1/2 PVs and development of GC as well as to determine how GC risk management should be factored into the clinical care of BRCA1/2 carriers.
Collapse
|
86
|
Kratz CP, Smirnov D, Autry R, Jäger N, Waszak SM, Großhennig A, Berutti R, Wendorff M, Hainaut P, Pfister SM, Prokisch H, Ripperger T, Malkin D. Heterozygous BRCA1 and BRCA2 and Mismatch Repair Gene Pathogenic Variants in Children and Adolescents With Cancer. J Natl Cancer Inst 2022; 114:1523-1532. [PMID: 35980168 DOI: 10.1093/jnci/djac151] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/21/2022] [Accepted: 07/20/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Genetic predisposition is has been identified as a cause of cancer, yet little is known about the role of adult cancer predisposition syndromes in childhood cancer. We examined the extent to which heterozygous pathogenic germline variants in BRCA1, BRCA2, PALB2, ATM, CHEK2, MSH2, MSH6, MLH1, and PMS2 contribute to cancer risk in children and adolescents. METHODS We conducted a meta-analysis of 11 studies that incorporated comprehensive germline testing for children and adolescents with cancer. ClinVar pathogenic or likely pathogenic variants (PVs) in genes of interest were compared with 2 control groups. Results were validated in a cohort of mainly European patients and controls. We employed the Proxy External Controls Association Test to account for different pipelines. RESULTS Among 3975 children and adolescents with cancer, statistically significant associations with cancer risk were observed for PVs in BRCA1 and 2 (26 PVs vs 63 PVs among 27 501 controls, odds ratio = 2.78, 95% confidence interval = 1.69 to 4.45; P < .001) and mismatch repair genes (19 PVs vs 14 PVs among 27 501 controls, odds ratio = 7.33, 95% confidence interval = 3.64 to 14.82; P <.001). Associations were seen in brain and other solid tumors but not in hematologic neoplasms. We confirmed similar findings in 1664 pediatric cancer patients primarily of European descent. CONCLUSION These data suggest that heterozygous PVs in BRCA1 and 2 and mismatch repair genes contribute with reduced penetrance to cancer risk in children and adolescents. No changes to predictive genetic testing and surveillance recommendations are required.
Collapse
Affiliation(s)
- Christian P Kratz
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Dmitrii Smirnov
- Institute of Human Genetics, School of Medicine, Technische Universität München, München, Germany.,Institute of Neurogenomics, Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Robert Autry
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.,Division of Paediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Natalie Jäger
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.,Division of Paediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Sebastian M Waszak
- Centre for Molecular Medicine Norway (NCMM), Nordic European Molecular Biology Laboratory (EMBL) Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway.,Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Anika Großhennig
- Institute of Biostatistics, Hannover Medical School, Hannover, Germany
| | - Riccardo Berutti
- Institute of Human Genetics, School of Medicine, Technische Universität München, München, Germany.,Institute of Neurogenomics, Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Mareike Wendorff
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany
| | - Pierre Hainaut
- Univ. Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, F38000, Grenoble, France
| | - Stefan M Pfister
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.,Division of Paediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Holger Prokisch
- Institute of Human Genetics, School of Medicine, Technische Universität München, München, Germany.,Institute of Neurogenomics, Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Tim Ripperger
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - David Malkin
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.,Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Paediatrics, University of Toronto, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
87
|
Lei H, Zhang M, Zhang L, Hemminki K, Wang XJ, Chen T. Overview on population screening for carriers with germline BRCA mutation in China. Front Oncol 2022; 12:1002360. [PMID: 36439508 PMCID: PMC9682265 DOI: 10.3389/fonc.2022.1002360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/24/2022] [Indexed: 12/01/2023] Open
Abstract
Carriers with BRCA1/2 germline pathogenic variants are associated with a high risk of breast and ovarian cancers (also pancreatic and prostate cancers). While the spectrum on germline BRCA mutations among the Chinese population shows ethnic specificity, the identification of carriers with germline BRCA mutation before cancer onset is the most effective approach to protect them. This review focused on the current status of BRCA1/2 screening, the surveillance and prevention measures, and discussed the issues and potential impact of BRCA1/2 population screening in China. We conducted literature research on databases PubMed and Google Scholar, as well as Chinese databases CNKI and Wangfang Med Online database (up to 31 March 2022). Latest publications on germline BRCA1/2 prevalence, spectrum, genetic screening as well as carrier counseling, surveillance and prevention were captured where available. While overall 15,256 records were retrieved, 72 publications using germline BRCA1/2 testing were finally retained for further analyses. Germline BRCA1/2 mutations are common in Chinese patients with hereditary breast, ovarian, prostate and pancreatic cancers. Within previous studies, a unique BRCA mutation spectrum in China was revealed. Next-generation sequencing panel was considered as the most common method for BRCA1/2 screening. Regular surveillance and preventive surgeries were tailored to carriers with mutated-BRCA1/2. We recommend that all Chinese diagnosed with breast, ovarian, pancreatic or prostate cancers and also healthy family members, shall undergo BRCA1/2 gene test to provide risk assessment. Subsequently, timely preventive measures for mutation carriers are recommended after authentic genetic counseling.
Collapse
Affiliation(s)
- Huijun Lei
- Department of Cancer Prevention/Zhejiang Cancer Institute, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Min Zhang
- School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Luyao Zhang
- Department of Cancer Epidemiology and Prevention, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Kari Hemminki
- Biomedical Center, Faculty of Medicine, Charles University in Pilsen, Pilsen, Czechia
- Division of Cancer Epidemiology, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld, Heidelberg, Germany
| | - Xiao-jia Wang
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Tianhui Chen
- Department of Cancer Prevention/Zhejiang Cancer Institute, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo, China
| |
Collapse
|
88
|
Familial history and prevalence of BRCA1, BRCA2 and TP53 pathogenic variants in HBOC Brazilian patients from a public healthcare service. Sci Rep 2022; 12:18629. [PMID: 36329109 PMCID: PMC9633799 DOI: 10.1038/s41598-022-23012-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
Several studies have demonstrated the cost-effectiveness of genetic testing for surveillance and treatment of carriers of germline pathogenic variants associated with hereditary breast/ovarian cancer syndrome (HBOC). In Brazil, seventy percent of the population is assisted by the public Unified Health System (SUS), where genetic testing is still unavailable. And few studies were performed regarding the prevalence of HBOC pathogenic variants in this context. Here, we estimated the prevalence of germline pathogenic variants in BRCA1, BRCA2 and TP53 genes in Brazilian patients suspected of HBOC and referred to public healthcare service. Predictive power of risk prediction models for detecting mutation carriers was also evaluated. We found that 41 out of 257 tested patients (15.9%) were carriers of pathogenic variants in the analyzed genes. Most frequent pathogenic variant was the founder Brazilian mutation TP53 c.1010G > A (p.Arg337His), adding to the accumulated evidence that supports inclusion of TP53 in routine testing of Brazilian HBOC patients. Surprisingly, BRCA1 c.5266dupC (p.Gln1756fs), a frequently reported pathogenic variant in Brazilian HBOC patients, was not observed. Regarding the use of predictive models, we found that familial history of cancer might be used to improve selection or prioritization of patients for genetic testing, especially in a context of limited resources.
Collapse
|
89
|
Mangone L, Marinelli F, Bisceglia I, Braghiroli MB, Damato A, Pinto C. Five-year relative survival by stage of breast and colon cancers in northern Italy. Front Oncol 2022; 12:982461. [PMID: 36387150 PMCID: PMC9659859 DOI: 10.3389/fonc.2022.982461] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/13/2022] [Indexed: 11/30/2022] Open
Abstract
The aim of this study is to present the 5-year relative survival by stage of breast and colorectal cancer patients in a northern Italian province. For the period 2013-2015, cases were selected from the Reggio Emilia Cancer Registry. Breast cancer patients were divided into 3 age groups: <45, 45-74 (the target screening population) and 74+. Colorectal cancers patients were classified into <50, 50-69 (the target screening population), and over 69 years. Carcinomas in situ and unknown stage were both excluded from the survival analyses. The five-year relative survival was estimated using the Pohar Perme method. During the period examined, 1,450 breast cancers and 992 colorectal cancer cases were registered. Analyzing in detail the patients with breast cancer for the entire 2013-2015 period, we noted that 50.4% were in stage I, 33.6% in stage II, 10.8% in stage III and 3.8% in stage IV. The stage was unknown in only 1.3% of patients (19 cases). The stage data of patients with colorectal cancer showed 24.5% were in stage I, 26.1% in stage II, 23.4% in stage III, and 24.6% in stage IV, and 1.4% unknown. Breast cancer 5-year survival was 100%, 89.7%, 71.4%, and 29.1% for stages I, II, III and IV, respectively and for colon cancer 96.7%, 83.4%, 70.8% and 16.2%, respectively.The presence of cancer screening, associated with effective treatments, account for the high survival rate of early-stage breast and colon cancers.
Collapse
Affiliation(s)
- Lucia Mangone
- Epidemiology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Via Amendola 2, Reggio Emilia, Italy
- *Correspondence: Lucia Mangone,
| | - Francesco Marinelli
- Epidemiology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Via Amendola 2, Reggio Emilia, Italy
| | - Isabella Bisceglia
- Epidemiology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Via Amendola 2, Reggio Emilia, Italy
| | - Maria Barbara Braghiroli
- Epidemiology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Via Amendola 2, Reggio Emilia, Italy
| | - Angela Damato
- Medical Oncology Unit, Comprehensive Cancer Center, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, Reggio Emilia, Italy
- Department of Biotechnologies, University of Siena, Siena, Italy
| | - Carmine Pinto
- Medical Oncology Unit, Comprehensive Cancer Center, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, Reggio Emilia, Italy
| |
Collapse
|
90
|
Long JM, Ebrahimzadeh J, Stanich PP, Katona BW. Endoscopic Surveillance in Patients with the Highest Risk of Gastric Cancer: Challenges and Solutions. Cancer Manag Res 2022; 14:2953-2969. [PMID: 36238953 PMCID: PMC9553156 DOI: 10.2147/cmar.s277898] [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: 08/06/2022] [Accepted: 09/22/2022] [Indexed: 11/23/2022] Open
Abstract
Gastric cancer is one of the most significant causes of cancer-related morbidity and mortality worldwide. Recognized modifiable risk factors include Helicobacter pylori infection, geographic location, select dietary factors, tobacco use and alcohol consumption. In addition, multiple hereditary cancer predisposition syndromes are associated with significantly elevated gastric cancer risk. Endoscopic surveillance in hereditary gastric cancer predisposition syndromes has the potential to identify gastric cancer at earlier and more treatable stages, as well as to prevent development of gastric cancer through identification of precancerous lesions. However, much uncertainty remains regarding use of endoscopic surveillance in hereditary gastric cancer predisposition syndromes, including whether or not it should be routinely performed, the surveillance interval and age of initiation, cost-effectiveness, and whether surveillance ultimately improves survival from gastric cancer for these high-risk individuals. In this review, we outline the hereditary gastric cancer predisposition syndromes associated with the highest gastric cancer risks. Additionally, we cover current evidence and guidelines addressing hereditary gastric cancer risk and surveillance in these syndromes, along with current challenges and limitations that emphasize a need for continued research in this field.
Collapse
Affiliation(s)
- Jessica M Long
- Division of Hematology and Oncology, Penn Medicine, Philadelphia, PA, USA
| | | | - Peter P Stanich
- Division of Gastroenterology, Hepatology & Nutrition, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
| | - Bryson W Katona
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Correspondence: Bryson W Katona, Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, 751 South Pavilion, Philadelphia, PA, 19104, USA, Tel +1-215-349-8222, Fax +1-215-349-5915, Email
| |
Collapse
|
91
|
Sunami T, Yamada A, Kondo T, Kanai M, Nagai K, Uchida Y, Yokode M, Matsumori T, Uza N, Murakami H, Yamada T, Muto M. Exceptional Response of Pancreatic Acinar Cell Carcinoma and Bile Duct Cancer to Platinum-Based Chemotherapy in a Family With a Germline BRCA2 Variant. Pancreas 2022; 51:1258-1262. [PMID: 37078954 DOI: 10.1097/mpa.0000000000002150] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
ABSTRACT Pancreatic cancer and its rare subtype, acinar cell carcinoma (PACC), frequently harbor germline and/or somatic variants in homologous recombinant genes, including BRCA2. Individuals possessing germline pathogenic BRCA2 variants are known to have a higher risk of developing various cancers, including breast, ovarian, pancreatic, and bile duct cancers (BDCs). It has been reported that tumors positive for BRCA1/2 variants are sensitive to platinum-based agents. Thus, BRCA1/2 germline testing and comprehensive genomic profiling are recommended to identify genetic susceptibility and to indicate optimal targeted therapy. Here, we report familial occurrence of PACC and BDC associated with BRCA2; both tumors responded exceptionally well to platinum-based chemotherapy. A 37-year-old man was diagnosed with unresectable PACC with a germline BRCA2 variant. He was treated with oxaliplatin-containing chemotherapy and conversion surgery, and remains alive without tumor recurrence after more than 36 months. His father also possessed the identical germline BRCA2 variant and was diagnosed with extrahepatic BDC with lymph node metastases. The tumors showed marked shrinkage upon treatment with cisplatin-containing chemotherapy. Our cases underscore the importance of comprehensive genomic profiling and genetic testing for BRCA2 to ensure optimal therapeutic options for PACC as well as to identify high-risk individuals with various cancers in the family.
Collapse
Affiliation(s)
| | | | | | - Masashi Kanai
- Department of Clinical Oncology, Kyoto University Hospital
| | - Kazuyuki Nagai
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery
| | - Yoichiro Uchida
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery
| | - Masataka Yokode
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University
| | - Tomoaki Matsumori
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University
| | - Hiromi Murakami
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
| | - Takahiro Yamada
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
| | | |
Collapse
|
92
|
Bernstein-Molho R, Friedman E, Evron E. Controversies and Open Questions in Management of Cancer-Free Carriers of Germline Pathogenic Variants in BRCA1/BRCA2. Cancers (Basel) 2022; 14:cancers14194592. [PMID: 36230512 PMCID: PMC9559251 DOI: 10.3390/cancers14194592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/06/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
Females harboring germline BRCA1/BRCA2 (BRCA) P/LPV are offered a tight surveillance scheme from the age of 25−30 years, aimed at early detection of specific cancer types, in addition to risk-reducing strategies. Multiple national and international surveillance guidelines have been published and updated over the last two decades from geographically diverse countries. We searched for guidelines published between 1 January 2015 and 1 May 2022. Differences between guidelines on issues such as primary prevention, mammography screening in young (<30 years) carriers, MRI screening in carriers above age 65 years, breast imaging (if any) after risk-reducing bilateral mastectomy, during pregnancy, and breastfeeding, and hormone-replacement therapy, are just a few notable examples. Beyond formal guidelines, BRCA carriers’ concerns also focus on the timing of risk-reducing surgeries, fertility preservation, management of menopausal symptoms in cancer survivors, and pancreatic cancer surveillance, issues that, for some, there are no data to support evidence-based recommendations. This review discusses these unsettled issues, emphasizing the importance of future studies to enable global guideline harmonization for optimal surveillance strategies. Moreover, it raises the unmet need for personalized risk stratification and surveillance in BRCA P/LPV carriers.
Collapse
Affiliation(s)
- Rinat Bernstein-Molho
- The Oncogenetics Unit, Chaim Sheba Medical Center, Tel-Hashomer, The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 5265601, Israel
| | - Eitan Friedman
- Assuta Medical Center, Tel-Aviv, Israel, The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 8436322, Israel
| | - Ella Evron
- Oncology, Kaplan Medical Institute, Rehovot, Hadassah Medical School, The Hebrew University, Jerusalem 9190501, Israel
- Correspondence: or ; Tel.: +972-502-056-171
| |
Collapse
|
93
|
Characterization of lung cancers in patients with BRCA germline variants: A multicenter series. Lung Cancer 2022; 173:67-70. [PMID: 36156322 DOI: 10.1016/j.lungcan.2022.09.002] [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: 03/03/2022] [Revised: 07/24/2022] [Accepted: 09/02/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION BRCA1 and BRCA2 (BReast CAncer susceptibility genes) are two tumor-suppressor genes associated with the hereditary breast and ovarian cancer susceptibility syndrome. Recent studies also suggest an increased lung adenocarcinoma risk in carriers. METHODS We conducted a multi-center retrospective study in 18 different French pulmonology and/or oncology departments on medico-administrative and clinical data prospectively collected in the Clinical Data Warehouse (CDW) of Greater Paris University Hospitals (Assistance Publique-Hôpitaux de Paris, AP-HP). Clinical characteristics and outcomes of patients with LC and a previously known BRCA1/2gl variant were retrospectively evaluated. RESULTS 17 patients with LC and known BRCA1/2gl variant were included. Patients were most women, former smokers with localized disease and BRCA2 variants. All LC were adenocarcinoma. For patients with medical history of cancer, median time from the first cancer in the BRCA spectrum and the LC occurrence was 20 years. Median disease-free survival (DFS) and overall survival (OS) in localized tumor (Stage I and II) was not reached and 78.6 months, respectively. In advanced cancer (Stade III and IV) median progression free survival was 9.7 months and median OS was 17.8 months. Univariate OS and DFS/PFS analyses by BRCA status did not find significant differences. CONCLUSION Results seem to show particular LC features in carriers of BRCA2 variants: adenocarcinoma subtype, woman, former or non-smoker.
Collapse
|
94
|
Saba H, Goggins M. Familial Pancreatic Cancer. Gastroenterol Clin North Am 2022; 51:561-575. [PMID: 36153110 PMCID: PMC11095833 DOI: 10.1016/j.gtc.2022.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Individuals at increased risk of developing pancreatic cancer, including those with a significant family history of the disease and those with pancreatic cancer susceptibility gene variants, can benefit from pancreas surveillance. Most pancreatic cancers diagnosed during surveillance are early-stage and such patients can achieve long-term survival. Determining who should undergo pancreas surveillance is still a work-in-progress, but the main tools clinicians use to estimate an individual's risk of pancreatic cancer are patient's age, the extent of their family history of pancreatic cancer, and whether or not they have a pancreatic cancer susceptibility gene mutation.
Collapse
Affiliation(s)
- Helena Saba
- Departments of Pathology, Johns Hopkins Medical Institutions, CRB2 351, 1550 Orleans Street, Baltimore, MD 21231, USA
| | - Michael Goggins
- Departments of Pathology, Johns Hopkins Medical Institutions, CRB2 351, 1550 Orleans Street, Baltimore, MD 21231, USA; Departments of Medicine, Johns Hopkins Medical Institutions, CRB2 351, 1550 Orleans Street, Baltimore, MD 21231, USA; Departments of Oncology, Johns Hopkins Medical Institutions, CRB2 351, 1550 Orleans Street, Baltimore, MD 21231, USA; Bloomberg School of Public Health, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, CRB2 351, 1550 Orleans Street, Baltimore, MD 21231, USA.
| |
Collapse
|
95
|
Wang L, Domchek SM, Kochman ML, Katona BW. Reaching beyond family history as inclusion criteria for pancreatic cancer surveillance in high-risk populations. Genes Cancer 2022; 13:49-51. [PMID: 36051752 PMCID: PMC9423661 DOI: 10.18632/genesandcancer.223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Indexed: 11/25/2022] Open
Affiliation(s)
| | | | | | - Bryson W. Katona
- Correspondence to:Bryson W. Katona, Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA email:
| |
Collapse
|
96
|
Li S, Silvestri V, Rebbeck TR, Neuhausen SL, Hopper JL, Nielsen HR, Ottini L, Antoniou AC. Reply to V. Fallet et al. J Clin Oncol 2022; 40:2509-2510. [PMID: 35549306 DOI: 10.1200/jco.22.00782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 11/20/2022] Open
Affiliation(s)
- Shuai Li
- Shuai Li, MD, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom, Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia; Valentina Silvestri, PhD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; Timothy R. Rebbeck, PhD, Harvard T.H. Chan School of Public Health, Boston, MA, Dana-Farber Cancer Institute, Boston, MA; Susan L. Neuhausen, PhD, Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA; John L. Hopper, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Henriette Roed Nielsen, PhD, Department of Clinical Genetics, Odense University Hospital, Odence, Denmark; Laura Ottini, MD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; and Antonis C. Antoniou, PhD, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Valentina Silvestri
- Shuai Li, MD, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom, Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia; Valentina Silvestri, PhD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; Timothy R. Rebbeck, PhD, Harvard T.H. Chan School of Public Health, Boston, MA, Dana-Farber Cancer Institute, Boston, MA; Susan L. Neuhausen, PhD, Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA; John L. Hopper, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Henriette Roed Nielsen, PhD, Department of Clinical Genetics, Odense University Hospital, Odence, Denmark; Laura Ottini, MD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; and Antonis C. Antoniou, PhD, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Timothy R Rebbeck
- Shuai Li, MD, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom, Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia; Valentina Silvestri, PhD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; Timothy R. Rebbeck, PhD, Harvard T.H. Chan School of Public Health, Boston, MA, Dana-Farber Cancer Institute, Boston, MA; Susan L. Neuhausen, PhD, Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA; John L. Hopper, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Henriette Roed Nielsen, PhD, Department of Clinical Genetics, Odense University Hospital, Odence, Denmark; Laura Ottini, MD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; and Antonis C. Antoniou, PhD, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Susan L Neuhausen
- Shuai Li, MD, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom, Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia; Valentina Silvestri, PhD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; Timothy R. Rebbeck, PhD, Harvard T.H. Chan School of Public Health, Boston, MA, Dana-Farber Cancer Institute, Boston, MA; Susan L. Neuhausen, PhD, Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA; John L. Hopper, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Henriette Roed Nielsen, PhD, Department of Clinical Genetics, Odense University Hospital, Odence, Denmark; Laura Ottini, MD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; and Antonis C. Antoniou, PhD, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - John L Hopper
- Shuai Li, MD, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom, Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia; Valentina Silvestri, PhD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; Timothy R. Rebbeck, PhD, Harvard T.H. Chan School of Public Health, Boston, MA, Dana-Farber Cancer Institute, Boston, MA; Susan L. Neuhausen, PhD, Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA; John L. Hopper, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Henriette Roed Nielsen, PhD, Department of Clinical Genetics, Odense University Hospital, Odence, Denmark; Laura Ottini, MD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; and Antonis C. Antoniou, PhD, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Henriette Roed Nielsen
- Shuai Li, MD, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom, Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia; Valentina Silvestri, PhD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; Timothy R. Rebbeck, PhD, Harvard T.H. Chan School of Public Health, Boston, MA, Dana-Farber Cancer Institute, Boston, MA; Susan L. Neuhausen, PhD, Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA; John L. Hopper, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Henriette Roed Nielsen, PhD, Department of Clinical Genetics, Odense University Hospital, Odence, Denmark; Laura Ottini, MD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; and Antonis C. Antoniou, PhD, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Laura Ottini
- Shuai Li, MD, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom, Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia; Valentina Silvestri, PhD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; Timothy R. Rebbeck, PhD, Harvard T.H. Chan School of Public Health, Boston, MA, Dana-Farber Cancer Institute, Boston, MA; Susan L. Neuhausen, PhD, Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA; John L. Hopper, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Henriette Roed Nielsen, PhD, Department of Clinical Genetics, Odense University Hospital, Odence, Denmark; Laura Ottini, MD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; and Antonis C. Antoniou, PhD, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Antonis C Antoniou
- Shuai Li, MD, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom, Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia; Valentina Silvestri, PhD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; Timothy R. Rebbeck, PhD, Harvard T.H. Chan School of Public Health, Boston, MA, Dana-Farber Cancer Institute, Boston, MA; Susan L. Neuhausen, PhD, Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA; John L. Hopper, PhD, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Henriette Roed Nielsen, PhD, Department of Clinical Genetics, Odense University Hospital, Odence, Denmark; Laura Ottini, MD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy; and Antonis C. Antoniou, PhD, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
97
|
Jürgens H, Roht L, Leitsalu L, Nõukas M, Palover M, Nikopensius T, Reigo A, Kals M, Kallak K, Kütner R, Budrikas K, Kuusk S, Valvere V, Laidre P, Toome K, Rekker K, Tooming M, Ülle Murumets, Kahre T, Kruuv-Käo K, Õunap K, Padrik P, Metspalu A, Esko T, Fischer K, Tõnisson N. Precise, Genotype-First Breast Cancer Prevention: Experience With Transferring Monogenic Findings From a Population Biobank to the Clinical Setting. Front Genet 2022; 13:881100. [PMID: 35938029 PMCID: PMC9355130 DOI: 10.3389/fgene.2022.881100] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Although hereditary breast cancer screening and management are well accepted and established in clinical settings, these efforts result in the detection of only a fraction of genetic predisposition at the population level. Here, we describe our experience from a national pilot study (2018-2021) in which 180 female participants of Estonian biobank (of >150,000 participants in total) were re-contacted to discuss personalized clinical prevention measures based on their genetic predisposition defined by 11 breast cancer-related genes. Our results show that genetic risk variants are relatively common in the average-risk Estonian population. Seventy-five percent of breast cancer cases in at-risk subjects occurred before the age of 50 years. Only one-third of subjects would have been eligible for clinical screening according to the current criteria. The participants perceived the receipt of genetic risk information as valuable. Fluent cooperation of project teams supported by state-of-art data management, quality control, and secure transfer can enable the integration of research results to everyday medical practice in a highly efficient, timely, and well-accepted manner. The positive experience in this genotype-first breast cancer study confirms the value of using existing basic genomic data from population biobanks for precise prevention.
Collapse
Affiliation(s)
- Hannes Jürgens
- Tartu University Hospital, Clinic of Hematology and Oncology, Tartu, Estonia
- University of Tartu, Clinic of Hematology and Oncology, Tartu, Estonia
| | - Laura Roht
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - Liis Leitsalu
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Margit Nõukas
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Marili Palover
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tiit Nikopensius
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Anu Reigo
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mart Kals
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Kersti Kallak
- Tartu University Hospital, Clinic of Hematology and Oncology, Tartu, Estonia
- University of Tartu, Clinic of Hematology and Oncology, Tartu, Estonia
| | - Riina Kütner
- North-Estonian Medical Center, Oncology and Haematology Clinic, Tallinn, Estonia
| | - Kai Budrikas
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Saskia Kuusk
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Vahur Valvere
- North-Estonian Medical Center, Oncology and Haematology Clinic, Tallinn, Estonia
| | - Piret Laidre
- Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - Kadri Toome
- Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - Kadri Rekker
- Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - Mikk Tooming
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - Ülle Murumets
- Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - Tiina Kahre
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - Krista Kruuv-Käo
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
- Tartu University Hospital, Tartu, Estonia
| | - Katrin Õunap
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - Peeter Padrik
- Tartu University Hospital, Clinic of Hematology and Oncology, Tartu, Estonia
- University of Tartu, Clinic of Hematology and Oncology, Tartu, Estonia
- Antegenes, Tartu, Estonia
| | - Andres Metspalu
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tõnu Esko
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Krista Fischer
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Neeme Tõnisson
- Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
- Estonian Biobank, Institute of Genomics, University of Tartu, Tartu, Estonia
| |
Collapse
|
98
|
Yurgelun MB. Building on More Than 20 Years of Progress in Pancreatic Cancer Surveillance for High-Risk Individuals. J Clin Oncol 2022; 40:3230-3234. [PMID: 35862875 DOI: 10.1200/jco.22.01287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Matthew B Yurgelun
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| |
Collapse
|
99
|
Rolfes M, Borde J, Möllenhoff K, Kayali M, Ernst C, Gehrig A, Sutter C, Ramser J, Niederacher D, Horváth J, Arnold N, Meindl A, Auber B, Rump A, Wang-Gohrke S, Ritter J, Hentschel J, Thiele H, Altmüller J, Nürnberg P, Rhiem K, Engel C, Wappenschmidt B, Schmutzler RK, Hahnen E, Hauke J. Prevalence of Cancer Predisposition Germline Variants in Male Breast Cancer Patients: Results of the German Consortium for Hereditary Breast and Ovarian Cancer. Cancers (Basel) 2022; 14:3292. [PMID: 35805063 PMCID: PMC9265404 DOI: 10.3390/cancers14133292] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023] Open
Abstract
Male breast cancer (mBC) is associated with a high prevalence of pathogenic variants (PVs) in the BRCA2 gene; however, data regarding other BC predisposition genes are limited. In this retrospective multicenter study, we investigated the prevalence of PVs in BRCA1/2 and 23 non-BRCA1/2 genes using a sample of 614 patients with mBC, recruited through the centers of the German Consortium for Hereditary Breast and Ovarian Cancer. A high proportion of patients with mBC carried PVs in BRCA2 (23.0%, 142/614) and BRCA1 (4.6%, 28/614). The prevalence of BRCA1/2 PVs was 11.0% in patients with mBC without a family history of breast and/or ovarian cancer. Patients with BRCA1/2 PVs did not show an earlier disease onset than those without. The predominant clinical presentation of tumor phenotypes was estrogen receptor (ER)-positive, progesterone receptor (PR)-positive, and HER2-negative (77.7%); further, 10.2% of the tumors were triple-positive, and 1.2% were triple-negative. No association was found between ER/PR/HER2 status and BRCA1/2 PV occurrence. Comparing the prevalence of protein-truncating variants (PTVs) between patients with mBC and control data (ExAC, n = 27,173) revealed significant associations of PTVs in both BRCA1 and BRCA2 with mBC (BRCA1: OR = 17.04, 95% CI = 10.54−26.82, p < 10−5; BRCA2: OR = 77.71, 95% CI = 58.71−102.33, p < 10−5). A case-control investigation of 23 non-BRCA1/2 genes in 340 BRCA1/2-negative patients and ExAC controls revealed significant associations of PTVs in CHEK2, PALB2, and ATM with mBC (CHEK2: OR = 3.78, 95% CI = 1.59−7.71, p = 0.002; PALB2: OR = 14.77, 95% CI = 5.02−36.02, p < 10−5; ATM: OR = 3.36, 95% CI = 0.89−8.96, p = 0.04). Overall, our findings support the benefit of multi-gene panel testing in patients with mBC irrespective of their family history, age at disease onset, and tumor phenotype.
Collapse
Affiliation(s)
- Muriel Rolfes
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany; (M.R.); (J.B.); (M.K.); (C.E.); (K.R.); (B.W.); (R.K.S.); (J.H.)
| | - Julika Borde
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany; (M.R.); (J.B.); (M.K.); (C.E.); (K.R.); (B.W.); (R.K.S.); (J.H.)
| | - Kathrin Möllenhoff
- Mathematisches Institut, Heinrich-Heine-Universität Duesseldorf, 40225 Duesseldorf, Germany;
| | - Mohamad Kayali
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany; (M.R.); (J.B.); (M.K.); (C.E.); (K.R.); (B.W.); (R.K.S.); (J.H.)
| | - Corinna Ernst
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany; (M.R.); (J.B.); (M.K.); (C.E.); (K.R.); (B.W.); (R.K.S.); (J.H.)
| | - Andrea Gehrig
- Institute of Human Genetics, University Wuerzburg, 97074 Wuerzburg, Germany;
| | - Christian Sutter
- Institute of Human Genetics, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Juliane Ramser
- Department of Gynecology and Obstetrics, Technical University Munich, 80333 Munich, Germany;
| | - Dieter Niederacher
- Department of Gynecology and Obstetrics, University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Judit Horváth
- Institute for Human Genetics, University Hospital Muenster, 48149 Muenster, Germany;
| | - Norbert Arnold
- Institute of Clinical Molecular Biology, Department of Gynecology and Obstetrics, University Hospital of Schleswig-Holstein, Campus Kiel, Christian-Albrechts University Kiel, 24105 Kiel, Germany;
| | - Alfons Meindl
- Department of Gynecology and Obstetrics, LMU Munich, University Hospital Munich, 80337 Munich, Germany;
| | - Bernd Auber
- Department of Human Genetics, Hannover Medical School, 30645 Hannover, Germany;
| | - Andreas Rump
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, TU Dresden, 01062 Dresden, Germany;
| | - Shan Wang-Gohrke
- Department of Gynecology and Obstetrics, University of Ulm, 89075 Ulm, Germany;
| | - Julia Ritter
- Institute of Medical and Human Genetics, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany;
| | - Julia Hentschel
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, 04103 Leipzig, Germany;
| | - Holger Thiele
- Cologne Center for Genomics (CCG) and Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; (H.T.); (J.A.); (P.N.)
| | - Janine Altmüller
- Cologne Center for Genomics (CCG) and Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; (H.T.); (J.A.); (P.N.)
- Core Facility Genomics, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics (CCG) and Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; (H.T.); (J.A.); (P.N.)
| | - Kerstin Rhiem
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany; (M.R.); (J.B.); (M.K.); (C.E.); (K.R.); (B.W.); (R.K.S.); (J.H.)
| | - Christoph Engel
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, 04107 Leipzig, Germany;
| | - Barbara Wappenschmidt
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany; (M.R.); (J.B.); (M.K.); (C.E.); (K.R.); (B.W.); (R.K.S.); (J.H.)
| | - Rita K. Schmutzler
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany; (M.R.); (J.B.); (M.K.); (C.E.); (K.R.); (B.W.); (R.K.S.); (J.H.)
| | - Eric Hahnen
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany; (M.R.); (J.B.); (M.K.); (C.E.); (K.R.); (B.W.); (R.K.S.); (J.H.)
| | - Jan Hauke
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany; (M.R.); (J.B.); (M.K.); (C.E.); (K.R.); (B.W.); (R.K.S.); (J.H.)
| |
Collapse
|
100
|
A Genome-First Approach to Estimate Prevalence of Germline Pathogenic Variants and Risk of Pancreatic Cancer in Select Cancer Susceptibility Genes. Cancers (Basel) 2022; 14:cancers14133257. [PMID: 35805029 PMCID: PMC9265005 DOI: 10.3390/cancers14133257] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 12/20/2022] Open
Abstract
Patients with germline pathogenic variants (GPV) in cancer predisposition genes are at increased risk of pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer. The genes most frequently found to harbor GPV in unselected PDAC cases are ATM, BRCA1, BRCA2, CDKN2A, CHEK2, and PALB2. However, GPV prevalence and gene-specific associations have not been extensively studied in the general population. To further explore these associations, we analyzed genomic and phenotypic data obtained from the UK Biobank (UKB) and Geisinger MyCode Community Health Initiative (GHS) cohorts comprising 200,600 and 175,449 participants, respectively. We estimated the frequency and calculated relative risks (RRs) of heterozygotes in both cohorts and a subset of individuals with PDAC. The combined frequency of heterozygous carriers of GPV in the general population ranged from 1.22% for CHEK2 to 0.05% for CDKN2A. The frequency of GPV in PDAC cases varied from 2.38% (ATM) to 0.19% (BRCA1 and CDKN2A). The RRs of PDAC were elevated for all genes except for BRCA1 and varied widely by gene from high (ATM) to low (CHEK2, BRCA2). This work expands our understanding of the frequencies of GPV heterozygous carriers and associations between PDAC and GPV in several important PDAC susceptibility genes.
Collapse
|