451
|
Genetic medicine is accelerating in Japan. Breast Cancer 2022; 29:659-665. [PMID: 35191009 PMCID: PMC9225975 DOI: 10.1007/s12282-022-01342-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 02/09/2022] [Indexed: 11/24/2022]
Abstract
Background In 2018, BRACAnalysis® was covered by medical insurance in Japan as a companion diagnostic test for the poly ADP-ribose polymerase inhibitor olaparib. In April 2020, eligibility for BRCA1/2 genetic testing was expanded to the diagnosis of hereditary breast and ovarian cancer syndrome, and medical management including prophylactic surgery and surveillance were covered by public insurance for BRCA1/2 mutation carriers who developed breast or ovarian cancer. The amount of BRCA1/2 genetic testing has been increasing recently, but the number of subjects and the impact of testing for patients’ outcomes remain unclear. Patients and methods This study explored the potential number of patients who will be eligible for new insurance coverage for BRCA1/2 genetic testing. We analyzed 868 patients from 938 surgeries between January 2014 and September 2020 from our database. Results Overall, 372 patients (43%) were eligible for new insurance coverage for BRCA1/2 genetic testing. The most common category was family history of breast or ovarian cancer within third-degree relatives. We found that 202 patients (23%) had family history of breast or ovarian cancer. In addition, the progression-free survival was significantly lower in triple-negative breast cancer patients aged 60 years or younger compared with the other patients (P = 0.0005). Conclusion The genetic medicine for primary breast cancer patients with BRCA1/2 germline mutation is accelerating rapidly in Japan. Therefore, establishing a system for the genetic medicine would be urgent. Supplementary Information The online version contains supplementary material available at 10.1007/s12282-022-01342-4.
Collapse
|
452
|
Bokkers K, Vlaming M, Engelhardt EG, Zweemer RP, van Oort IM, Kiemeney LALM, Bleiker EMA, Ausems MGEM. The Feasibility of Implementing Mainstream Germline Genetic Testing in Routine Cancer Care-A Systematic Review. Cancers (Basel) 2022; 14:cancers14041059. [PMID: 35205807 PMCID: PMC8870548 DOI: 10.3390/cancers14041059] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/10/2022] [Accepted: 02/17/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Germline genetic testing for patients with cancer can have important implications for treatment, preventive options, and for family members. In a mainstream genetic testing pathway, pre-test counseling is performed by non-genetic healthcare professionals, thereby making genetic testing more accessible to all patients who might benefit from it. These mainstream genetic testing pathways are being implemented in different hospitals around the world, and for different cancer types. It is important to evaluate how a mainstream genetic testing pathway can be made sustainable and if quality of genetic care is maintained. We show in this systematic review that it is feasible to incorporate a mainstream genetic testing pathway into routine cancer care while maintaining quality of care. A training procedure for non-genetic healthcare professionals and a close collaboration between genetics and other clinical departments are highly recommended to ensure sustainability. Abstract Background: Non-genetic healthcare professionals can provide pre-test counseling and order germline genetic tests themselves, which is called mainstream genetic testing. In this systematic review, we determined whether mainstream genetic testing was feasible in daily practice while maintaining quality of genetic care. Methods: PubMed, Embase, CINAHL, and PsychINFO were searched for articles describing mainstream genetic testing initiatives in cancer care. Results: Seventeen articles, reporting on 15 studies, met the inclusion criteria. Non-genetic healthcare professionals concluded that mainstream genetic testing was possible within the timeframe of a routine consultation. In 14 studies, non-genetic healthcare professionals completed some form of training about genetics. When referral was coordinated by a genetics team, the majority of patients carrying a pathogenic variant were seen for post-test counseling by genetic healthcare professionals. The number of days between cancer diagnosis and test result disclosure was always lower in the mainstream genetic testing pathway than in the standard genetic testing pathway (e.g., pre-test counseling at genetics department). Conclusions: Mainstream genetic testing seems feasible in daily practice with no insurmountable barriers. A structured pathway with a training procedure is desirable, as well as a close collaboration between genetics and other clinical departments.
Collapse
Affiliation(s)
- Kyra Bokkers
- Division Laboratories, Pharmacy and Biomedical Genetics, Department of Genetics, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (K.B.); (M.V.)
| | - Michiel Vlaming
- Division Laboratories, Pharmacy and Biomedical Genetics, Department of Genetics, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (K.B.); (M.V.)
| | - Ellen G. Engelhardt
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (E.G.E.); (E.M.A.B.)
| | - Ronald P. Zweemer
- Department of Gynecological Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands;
| | - Inge M. van Oort
- Department of Urology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands; (I.M.v.O.); (L.A.L.M.K.)
| | - Lambertus A. L. M. Kiemeney
- Department of Urology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands; (I.M.v.O.); (L.A.L.M.K.)
- Department for Health Evidence, Radboud University Medical Center, Geert Grooteplein Zuid 21, 6525 EZ Nijmegen, The Netherlands
| | - Eveline M. A. Bleiker
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; (E.G.E.); (E.M.A.B.)
- Department of Clinical Genetics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Family Cancer Clinic, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Margreet G. E. M. Ausems
- Division Laboratories, Pharmacy and Biomedical Genetics, Department of Genetics, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (K.B.); (M.V.)
- Correspondence: ; Tel.: +31-88-75-538-00
| |
Collapse
|
453
|
Sandru F, Petca RC, Carsote M, Petca A, Dumitrascu M, Ghemigian A. Adrenocortical carcinoma: Pediatric aspects (Review). Exp Ther Med 2022; 23:287. [DOI: 10.3892/etm.2022.11216] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/24/2021] [Indexed: 11/05/2022] Open
Affiliation(s)
- Florica Sandru
- Department of Dermatology, ‘Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Răzvan-Cosmin Petca
- Department of Urology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Mara Carsote
- Department of Endocrinology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Aida Petca
- Department of Obstetrics and Gynecology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Mihai Dumitrascu
- Department of Obstetrics and Gynecology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Adina Ghemigian
- Department of Endocrinology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
454
|
Fanale D, Pivetti A, Cancelliere D, Spera A, Bono M, Fiorino A, Pedone E, Barraco N, Brando C, Perez A, Guarneri MF, Russo TDB, Vieni S, Guarneri G, Russo A, Bazan V. BRCA1/2 variants of unknown significance in hereditary breast and ovarian cancer (HBOC) syndrome: looking for the hidden meaning. Crit Rev Oncol Hematol 2022; 172:103626. [PMID: 35150867 DOI: 10.1016/j.critrevonc.2022.103626] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/28/2022] [Accepted: 02/07/2022] [Indexed: 01/04/2023] Open
Abstract
Hereditary breast and ovarian cancer syndrome is caused by germline mutations in BRCA1/2 genes. These genes are very large and their mutations are heterogeneous and scattered throughout the coding sequence. In addition to the above-mentioned mutations, variants of uncertain/unknown significance (VUSs) have been identified in BRCA genes, which make more difficult the clinical management of the patient and risk assessment. In the last decades, several laboratories have developed different databases that contain more than 2000 variants for the two genes and integrated strategies which include multifactorial prediction models based on direct and indirect genetic evidence, to classify the VUS and attribute them a clinical significance associated with a deleterious, high-low or neutral risk. This review provides a comprehensive overview of literature studies concerning the VUSs, in order to assess their impact on the population and provide new insight for the appropriate patient management in clinical practice.
Collapse
Affiliation(s)
- Daniele Fanale
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Alessia Pivetti
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Daniela Cancelliere
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Antonio Spera
- Department of Radiotherapy, San Giovanni di Dio Hospital, ASP of Agrigento, Agrigento, Italy
| | - Marco Bono
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Alessia Fiorino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Erika Pedone
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Nadia Barraco
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Chiara Brando
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Alessandro Perez
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | | | - Tancredi Didier Bazan Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Salvatore Vieni
- Division of General and Oncological Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Italy
| | - Girolamo Guarneri
- Gynecology Section, Mother - Child Department, University of Palermo, 90127 Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy.
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
455
|
Layman RM, Lin H, Gutierrez Barrera AM, Karuturi MS, Yam C, Arun BK. Clinical outcomes and Oncotype DX Breast Recurrence Score® in early-stage BRCA-associated hormone receptor-positive breast cancer. Cancer Med 2022; 11:1474-1483. [PMID: 35128817 PMCID: PMC8921901 DOI: 10.1002/cam4.4566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/20/2021] [Accepted: 12/07/2021] [Indexed: 12/20/2022] Open
Abstract
Background BRCA‐associated breast cancers tend to have distinctive features compared to sporadic breast cancers; further characterization can aid in optimizing treatment. Methods The study evaluated a patient cohort with early‐stage estrogen receptor positive, HER2 negative invasive breast cancer who had Oncotype DX Breast Recurrence Score® analysis and genetic testing for hereditary breast and ovarian cancer syndrome. Data on patients and their breast cancers with outcomes were collected and analyzed. Results 745 patients were included, of whom 33 had pathogenic BRCA mutations (8 BRCA1, 25 BRCA2). Patients with BRCA mutations were younger and received more adjuvant chemotherapy, but less endocrine therapy and radiation therapy. BRCA‐associated breast cancers had less progesterone receptor expression, higher nuclear grade, and higher Oncotype DX Breast Recurrence Scores® with median Recurrence Score® 29, compared to 16 in cancers without mutations (p < 0.0001). Breast cancer recurrence developed in 18% of patients with BRCA mutations and 9% of patient without mutations, although multivariate analysis of relapse‐free survival was not significant, HR 1.519 (95% confidence interval [CI] 0.64–3.58; p = 0.3401). After adjusting for Recurrence Score®, overall survival by BRCA status was improved HR 0.448 (95% CI 0.06–3.34; p = 0.4333). Conclusions BRCA‐associated early‐stage hormone receptor‐positive breast cancers have higher Oncotype DX Breast Recurrence Score® compared to those without mutations. BRCA status did not significantly impact relapse‐free survival and overall survival. Larger clinical trials are needed to further assess the findings, and if confirmed, could impact clinical management of BRCA‐associated breast cancers.
Collapse
Affiliation(s)
- Rachel M Layman
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Heather Lin
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Meghan S Karuturi
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Clinton Yam
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Banu K Arun
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
456
|
Vaughan H, Rugo HS, Haemel A. Risk-Based Screening for Cancer in Patients With Dermatomyositis: Toward a More Individualized Approach. JAMA Dermatol 2022; 158:244-247. [PMID: 35107570 DOI: 10.1001/jamadermatol.2021.5841] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Heidi Vaughan
- School of Medicine, University of California, San Francisco
| | - Hope S Rugo
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco Comprehensive Cancer Center, San Francisco
| | - Anna Haemel
- Department of Dermatology, University of California, San Francisco
| |
Collapse
|
457
|
Gynecological Management of the Breast Cancer Survivor. Best Pract Res Clin Obstet Gynaecol 2022; 82:69-80. [DOI: 10.1016/j.bpobgyn.2022.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/31/2022] [Accepted: 01/31/2022] [Indexed: 12/30/2022]
|
458
|
“It was a no-brainer”: A Qualitative Study of Factors Driving Previvors’ Decision-Making when Considering Risk-Reducing Salpingectomy with Delayed Oophorectomy. Gynecol Oncol Rep 2022; 40:100948. [PMID: 35265744 PMCID: PMC8898920 DOI: 10.1016/j.gore.2022.100948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/30/2022] Open
Abstract
Self-advocacy and building rapport with providers facilitated decision-making for RRS-DO-focused previvors. Avoiding menopause through RRS was a key motivator for RRS-DO-focused previvors. RRS-DO-focused previvors viewed RRS as a stepwise approach to risk reduction. Understanding previvor priorities and experiences can help optimize shared decision-making.
Objective Previvors are becoming more aware of the option of risk-reducing salpingectomy with delayed oophorectomy (RRS-DO) to mitigate their risk of ovarian cancer. In this qualitative study, we explored the clinical and non-clinical factors that impacted previvors’ decision-making to pursue RRS-DO as a risk reduction strategy. Methods Semi-structured telephone interviews were conducted with previvors and transcribed verbatim. Using ATLAS.ti® software, two primary investigators interpreted data through thematic analysis. After coding four interviews, the investigators discussed discrepancies between codes with a moderator and resolved and refined code. The investigators applied the universal codebook to all interviews and revised the codebook using an iterative approach. Examining codes within and across interviews allowed for major themes and patterns to emerge. Results Interviews were conducted with seventeen previvors (ages 31–46). 6 (25%) previvors had a BRCA1 mutation, 7 (41%), a BRCA2 mutation, 3 (13%), a Lynch-related mutation, and 1 (6%), other (MUTYH mutation). At the time of interview, 12 previvors (71%) were planning (6) or had undergone (6) RRS-DO, 4 (23%) were planning (1) or had undergone (3) risk reducing salpingo-oophorectomy (RRSO), and 1 (6%) was undecided. Three major themes emerged: motivating factors for selecting surgical risk reduction option, barriers complicating surgical decision-making, and facilitating factors for surgical decision-making. RRS-DO-focused previvors prioritized avoiding menopause, and they also emphasized that self-advocacy and building rapport with providers facilitated their decision-making. Conclusion By understanding previvors’ priorities and experiences, physicians can better partner with previvors as they navigate their ovarian cancer risk reduction journey. This will ultimately optimize shared decision-making.
Collapse
|
459
|
Petrova D, Cruz M, Sánchez MJ. BRCA1/2 testing for genetic susceptibility to cancer after 25 years: A scoping review and a primer on ethical implications. Breast 2022; 61:66-76. [PMID: 34920368 PMCID: PMC8686063 DOI: 10.1016/j.breast.2021.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Mutations in the genes called BRCA1 and BRCA2 are associated with significantly elevated lifetime risk of developing breast and ovarian cancer. This year marks 25 years since genetic tests for BRCA1/2 mutations became available to the public. Currently, comprehensive guidelines exist regarding BRCA1/2 testing and preventive measures in mutation carriers. As such, BRCA1/2 testing represents a precedent not only in genetic testing and management of genetic cancer risk, but also in bioethics. The goal of the current research was to offer a review and an ethical primer of the main ethical challenges related to BRCA testing. METHOD A systematic scoping review was undertaken following the PRISMA Extension for Scoping Reviews (PRISMA-ScR). Four databases were searched and 18 articles that met the inclusion criteria were synthetized narratively into a conceptual map. RESULTS Ethical discussions revolved around the BRCA1/2 gene discovery, how tests are distributed for clinical use, the choice to undergo testing, unresolved issues in receiving and disclosing test results, reproductive decision-making, and culture-specific ethics. Several unique properties of the latest developments in testing circumstances (e.g., incorporation of BRCA1/2 testing in multi-gene or whole genome sequence panels and tests sold directly to consumers) significantly raised the complexity of ethical debates. CONCLUSIONS Multidisciplinary ethical discussion is necessary to guide not only individual decision making but also societal practices and medical guidelines in light of the new technologies available and the latest results regarding psychological, social, and health outcomes in cancer previvors and survivors affected by BRCA mutations.
Collapse
Affiliation(s)
- Dafina Petrova
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Escuela Andaluza de Salud Pública, Granada, Spain; CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain.
| | - Maite Cruz
- Escuela Andaluza de Salud Pública, Granada, Spain
| | - María-José Sánchez
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Escuela Andaluza de Salud Pública, Granada, Spain; CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain; Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain
| |
Collapse
|
460
|
Watanabe T, Soeda S, Endo Y, Okabe C, Sato T, Kamo N, Ueda M, Kojima M, Furukawa S, Nishigori H, Takahashi T, Fujimori K. Rare Hereditary Gynecological Cancer Syndromes. Int J Mol Sci 2022; 23:1563. [PMID: 35163487 PMCID: PMC8835983 DOI: 10.3390/ijms23031563] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 12/04/2022] Open
Abstract
Hereditary cancer syndromes, which are characterized by onset at an early age and an increased risk of developing certain tumors, are caused by germline pathogenic variants in tumor suppressor genes and are mostly inherited in an autosomal dominant manner. Therefore, hereditary cancer syndromes have been used as powerful models to identify and characterize susceptibility genes associated with cancer. Furthermore, clarification of the association between genotypes and phenotypes in one disease has provided insights into the etiology of other seemingly different diseases. Molecular genetic discoveries from the study of hereditary cancer syndrome have not only changed the methods of diagnosis and management, but have also shed light on the molecular regulatory pathways that are important in the development and treatment of sporadic tumors. The main cancer susceptibility syndromes that involve gynecologic cancers include hereditary breast and ovarian cancer syndrome as well as Lynch syndrome. However, in addition to these two hereditary cancer syndromes, there are several other hereditary syndromes associated with gynecologic cancers. In the present review, we provide an overview of the clinical features, and discuss the molecular genetics, of four rare hereditary gynecological cancer syndromes; Cowden syndrome, Peutz-Jeghers syndrome, DICER1 syndrome and rhabdoid tumor predisposition syndrome 2.
Collapse
Affiliation(s)
- Takafumi Watanabe
- Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima 960-1295, Japan; (S.S.); (Y.E.); (C.O.); (T.S.); (N.K.); (M.U.); (M.K.); (S.F.); (K.F.)
| | - Shu Soeda
- Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima 960-1295, Japan; (S.S.); (Y.E.); (C.O.); (T.S.); (N.K.); (M.U.); (M.K.); (S.F.); (K.F.)
| | - Yuta Endo
- Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima 960-1295, Japan; (S.S.); (Y.E.); (C.O.); (T.S.); (N.K.); (M.U.); (M.K.); (S.F.); (K.F.)
| | - Chikako Okabe
- Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima 960-1295, Japan; (S.S.); (Y.E.); (C.O.); (T.S.); (N.K.); (M.U.); (M.K.); (S.F.); (K.F.)
| | - Tetsu Sato
- Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima 960-1295, Japan; (S.S.); (Y.E.); (C.O.); (T.S.); (N.K.); (M.U.); (M.K.); (S.F.); (K.F.)
| | - Norihito Kamo
- Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima 960-1295, Japan; (S.S.); (Y.E.); (C.O.); (T.S.); (N.K.); (M.U.); (M.K.); (S.F.); (K.F.)
| | - Makiko Ueda
- Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima 960-1295, Japan; (S.S.); (Y.E.); (C.O.); (T.S.); (N.K.); (M.U.); (M.K.); (S.F.); (K.F.)
| | - Manabu Kojima
- Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima 960-1295, Japan; (S.S.); (Y.E.); (C.O.); (T.S.); (N.K.); (M.U.); (M.K.); (S.F.); (K.F.)
| | - Shigenori Furukawa
- Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima 960-1295, Japan; (S.S.); (Y.E.); (C.O.); (T.S.); (N.K.); (M.U.); (M.K.); (S.F.); (K.F.)
| | - Hidekazu Nishigori
- Fukushima Medical Center for Children and Women, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan; (H.N.); (T.T.)
| | - Toshifumi Takahashi
- Fukushima Medical Center for Children and Women, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan; (H.N.); (T.T.)
| | - Keiya Fujimori
- Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima 960-1295, Japan; (S.S.); (Y.E.); (C.O.); (T.S.); (N.K.); (M.U.); (M.K.); (S.F.); (K.F.)
| |
Collapse
|
461
|
Lebedeva A, Shaykhutdinova Y, Seriak D, Ignatova E, Rozhavskaya E, Vardhan D, Manicka S, Sharova M, Grigoreva T, Baranova A, Mileyko V, Ivanov M. Incidental germline findings during molecular profiling of tumor tissues for precision oncology: molecular survey and methodological obstacles. J Transl Med 2022; 20:29. [PMID: 35033101 PMCID: PMC8760669 DOI: 10.1186/s12967-022-03230-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 01/05/2022] [Indexed: 12/13/2022] Open
Abstract
Background A fraction of patients referred for complex molecular profiling of biopsied tumors may harbor germline variants in genes associated with the development of hereditary cancer syndromes (HCS). Neither the bioinformatic analysis nor the reporting of such incidental germline findings are standardized. Methods Data from Next-Generation Sequencing (NGS) of biopsied tumor samples referred for complex molecular profiling were analyzed for germline variants in HCS-associated genes. Analysis of variant origin was performed employing bioinformatic algorithms followed by manual curation. When possible, the origin of the variant was validated by Sanger sequencing of the sample of normal tissue. The variants’ pathogenicity was assessed according to ACMG/AMP. Results Tumors were sampled from 183 patients (Males: 75 [41.0%]; Females: 108 [59.0%]; mean [SD] age, 57.7 [13.3] years) and analysed by targeted NGS. The most common tumor types were colorectal (19%), pancreatic (13%), and lung cancer (10%). A total of 56 sequence variants in genes associated with HCS were detected in 40 patients. Of them, 17 variants found in 14 patients were predicted to be of germline origin, with 6 variants interpreted as pathogenic (PV) or likely pathogenic (LPV), and 9 as variants of uncertain significance (VUS). For the 41 out of 42 (97%) missense variants in HCS-associated genes, the results of computational prediction of variant origin were concordant with that of experimental examination. We estimate that Sanger sequencing of a sample of normal tissue would be required for ~ 1–7% of the total assessed cases with PV or LPV, when necessity to follow with genetic counselling referral in ~ 2–15% of total assessed cases (PV, LPV or VUS found in HCS genes). Conclusion Incidental findings of pathogenic germline variants are common in data from cancer patients referred for complex molecular profiling. We propose an algorithm for the management of patients with newly detected variants in genes associated with HCS.
Collapse
Affiliation(s)
- Alexandra Lebedeva
- Atlas Oncodiagnostics, LLC, Moscow, Russia. .,Sechenov University, Moscow, Russia.
| | | | | | - Ekaterina Ignatova
- Atlas Oncodiagnostics, LLC, Moscow, Russia.,Department of chemotherapy №2, Federal State Budgetary Institution «N.N. Blokhin National Medical Research Center of Oncology» of the Ministry of Health of the Russian Federation, Moscow, Russia.,Department of Oncogenetics, Institute of Higher and Additional Professional Education, Research Centre for Medical Genetics, Moscow, Russia
| | | | | | - Sofia Manicka
- School of Systems Biology, George Mason University, Mannas, VA, USA
| | | | | | - Ancha Baranova
- School of Systems Biology, George Mason University, Mannas, VA, USA
| | | | - Maxim Ivanov
- Atlas Oncodiagnostics, LLC, Moscow, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia
| |
Collapse
|
462
|
Beyond BRCA1/2: Homologous Recombination Repair Genetic Profile in a Large Cohort of Apulian Ovarian Cancers. Cancers (Basel) 2022; 14:cancers14020365. [PMID: 35053526 PMCID: PMC8773795 DOI: 10.3390/cancers14020365] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/29/2021] [Accepted: 01/07/2022] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Ovarian cancer (OC) is the second most common gynecologic malignancy and the most common cause of death among women with gynecologic cancer. Despite significant improvements having been made over the past decades, OC remains one of the most challenging malignancies to treat. Targeted therapies, such as PARPi, have emerged as one of the most interesting treatments for OC, particularly in women with BRCA1 or BRCA2 mutations. or those with a dysfunctional homologous recombination repair pathway. The purpose of our study is to address the role of NGS-targeted resequencing in the clinical routine of OC, focusing not only on BRCA1/2 but also on the homologous recombination repair genetic profile. Abstract Background: Pathogenic variants in homologous recombination repair (HRR) genes other than BRCA1/2 have been associated with a high risk of ovarian cancer (OC). In current clinical practice, genetic testing is generally limited to BRCA1/2. Herein, we investigated the mutational status of both BRCA1/2 and 5 HRR genes in 69 unselected OC, evaluating the advantage of multigene panel testing in everyday clinical practice. Methods: We analyzed 69 epithelial OC samples using an NGS custom multigene panel of the 5 HRR pathways genes, beyond the genetic screening routine of BRCA1/2 testing. Results: Overall, 19 pathogenic variants (27.5%) were detected. The majority (21.7%) of patients displayed a deleterious mutation in BRCA1/2, whereas 5.8% harbored a pathogenic variant in one of the HRR genes. Additionally, there were 14 (20.3%) uncertain significant variants (VUS). The assessment of germline mutational status showed that a small number of variants (five) were not detected in the corresponding blood sample. Notably, we detected one BRIP1 and four BRCA1/2 deleterious variants in the low-grade serous and endometrioid histology OC, respectively. Conclusion: We demonstrate that using a multigene panel beyond BRCA1/2 improves the diagnostic yield in OC testing, and it could produce clinically relevant results.
Collapse
|
463
|
Papageorgiou GI, Fergadis E, Skouteris N, Christakos E, Tsakatikas SA, Lianos E, Kosmas C. Case Report: Combination of Olaparib With Chemotherapy in a Patient With ATM-Deficient Colorectal Cancer. Front Oncol 2022; 11:788809. [PMID: 35004311 PMCID: PMC8728007 DOI: 10.3389/fonc.2021.788809] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/01/2021] [Indexed: 12/18/2022] Open
Abstract
Poly-ADP ribose polymerase (PARP) inhibitors are constantly increasing in their indications for use as anti-cancer treatment in various neoplasms, the majority of which are linked with BRCA deficiency. Preclinical data support the investigation of PARP inhibitors in other neoplasms exhibiting “BRCAness” or homologous recombination deficiency (HRD) as monotherapy as well as in combination with chemotherapy. With the current report we present the case of a heavily pretreated 55-year-old male patient diagnosed with stage IV ATM-deficient CRC, who was effectively treated with an off-label olaparib-irinotecan combination after exhaustion of all available treatment choices; furthermore, we discuss the existing data providing evidence for the use of PARP inhibitors in ATM-deficient CRC and encourage the implementation of next-generation sequencing (NGS) in patients with no other available treatment options.
Collapse
Affiliation(s)
- Georgios I Papageorgiou
- Division of Medical Oncology & Hematopoietic Cell Transplant Unit, Department of Medicine, ''Metaxa'' Cancer Hospital, Piraeus, Greece
| | - Evangelos Fergadis
- Division of Medical Oncology & Hematopoietic Cell Transplant Unit, Department of Medicine, ''Metaxa'' Cancer Hospital, Piraeus, Greece
| | - Nikos Skouteris
- Division of Medical Oncology & Hematopoietic Cell Transplant Unit, Department of Medicine, ''Metaxa'' Cancer Hospital, Piraeus, Greece
| | - Evridiki Christakos
- Division of Medical Oncology & Hematopoietic Cell Transplant Unit, Department of Medicine, ''Metaxa'' Cancer Hospital, Piraeus, Greece
| | - Sergios A Tsakatikas
- Division of Medical Oncology & Hematopoietic Cell Transplant Unit, Department of Medicine, ''Metaxa'' Cancer Hospital, Piraeus, Greece
| | - Evangelos Lianos
- Division of Medical Oncology & Hematopoietic Cell Transplant Unit, Department of Medicine, ''Metaxa'' Cancer Hospital, Piraeus, Greece
| | - Christos Kosmas
- Division of Medical Oncology & Hematopoietic Cell Transplant Unit, Department of Medicine, ''Metaxa'' Cancer Hospital, Piraeus, Greece
| |
Collapse
|
464
|
Elkefi S, Choudhury A, Strachna O, Asan O. Impact of Health Perception and Knowledge on Genetic Testing Decisions Using the Health Belief Model. JCO Clin Cancer Inform 2022; 6:e2100117. [PMID: 34990211 PMCID: PMC9848547 DOI: 10.1200/cci.21.00117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
PURPOSE Early detection of cancer risk is essential as it is associated with a higher chance of survival, more successful treatment, and improved quality of life. Genetic testing helps at-risk patients estimate the likelihood of developing cancer in a lifetime. This study aims to indentify the factors (perceived susceptibility, severity, benefits, and self-efficacy) that impact one's decision to take the genetic test. METHODS We examined the impacts of different factors of the health belief model on the engagement of patients in genetic testing using data from the National Cancer Institute's 2020 cross-sectional nationally representative data published in 2021. Complete surveys were answered by 3,865 participants (weighted population size = 253,815,197). All estimates were weighted to be nationally representative of the US population using the jackknife weighting method for parameter estimation. We used multivariable logistic regression to test our hypotheses for patients who have taken the genetic test for cancer risk detection. We adjusted the multivariate model for age, education, income, race, sex, cancer history, familial cancer history, and education. RESULTS We tested five hypotheses using the health belief model. Respondents who had genetic testing were more likely to rely on their health care providers and genetic counselors to make their decisions. Respondents who had genetic tests also reported less reliability on other sources than doctors: for the internet and social media (odds ratio = 0.33; P < .001) and for journals and magazines (odds ratio = 0.48; P = .007). CONCLUSION The findings show that patients generally rely on suggestions from their health care providers and counselors in genetic testing decisions. These findings also indicate that health care providers play a critical role in helping patients decide whether to use genetic testing to detect cancer risk in the early stages.
Collapse
Affiliation(s)
- Safa Elkefi
- Stevens Institute of Technology, School of Systems and Enterprises, Hoboken, NJ
| | - Avishek Choudhury
- Stevens Institute of Technology, School of Systems and Enterprises, Hoboken, NJ
| | - Olga Strachna
- Stevens Institute of Technology, School of Systems and Enterprises, Hoboken, NJ
| | - Onur Asan
- Stevens Institute of Technology, School of Systems and Enterprises, Hoboken, NJ,Onur Asan, PhD, School of Systems and Enterprises, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ 07030; e-mail:
| |
Collapse
|
465
|
Henderson V, Strayhorn SM, Bergeron NQ, Strahan DC, Ganschow PS, Khanna AS, Watson K, Hoskins K, Molina Y. Healthcare Predictors of Information Dissemination About Genetic Risks. Cancer Control 2022; 29:10732748221104666. [PMID: 35658635 PMCID: PMC9174561 DOI: 10.1177/10732748221104666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objectives Despite the benefits of genetic counseling and testing (GCT), utilization is
particularly low among African American (AA) women who exhibit breast cancer
features that are common in BRCA-associated cancer. Underutilization is
especially problematic for AA women who are more likely to die from breast
cancer than women from any other race or ethnicity. Due to medical mistrust,
fear, and stigma that can be associated with genetic services among
racial/ethnic minorities, reliance on trusted social networks may be an
impactful strategy to increase dissemination of knowledge about hereditary
cancer risk. Informed by the social cognitive theory, the purpose of this
study is to determine: 1) which AA patients diagnosed with breast cancer and
with identified hereditary risk are sharing information about hereditary
risk with their networks; 2) the nature of the information dissemination;
and 3) if personal GCT experiences is associated with dissemination of
information about hereditary risk. Methods Among consented participants (n = 100) that completed an interview
administered using a 202-item questionnaire consisting of open- and
closed-ended questions, 62 patients were identified to be at higher risk for
breast cancer. Descriptive statistics, bivariable chi-square, Pearson’s
exact tests, and regression analyses were conducted to examine differences
in characteristics between high-risk participants who disseminated
hereditary risk information and participants who did not. Results Among high-risk participants, 25 (40%) indicated they had disseminated
information about hereditary risk to at least one member in their
family/friend network and 37 (60%) had not. Receipt of both provider
recommendations and receipt of GCT services was associated with greater odds
of disseminating information about hereditary risk with networks, OR = 4.53,
95%CI [1.33, 15.50], p = .02. Conclusion Interventions that increase self-efficacy gained through additional
personalized knowledge and experience gained through provider
recommendations and by undergoing GCT may facilitate information
dissemination among social/familial networks.
Collapse
Affiliation(s)
- Vida Henderson
- Fred Hutchinson Cancer Research Center, Public Health Sciences Division, Seattle, WA, USA.,14681Community Health Sciences Division, University of Illinois Chicago, Chicago, IL, USA
| | - Shaila M Strayhorn
- 14621School of Health and Applied Human Sciences, University of North Carolina Wilmington, Wilmington, NC, USA
| | - Nyahne Q Bergeron
- 14681Community Health Sciences Division, University of Illinois Chicago, Chicago, IL, USA
| | - Desmona C Strahan
- 14681Community Health Sciences Division, University of Illinois Chicago, Chicago, IL, USA
| | - Pamela S Ganschow
- College of Medicine, University of Illinois Chicago, Chicago, IL, USA.,Cancer Center, University of Illinois Chicago, Chicago, IL, USA
| | - Aditya S Khanna
- Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
| | - Karriem Watson
- All of Us Program, National Institutes of Health, Bethesda, MD, USA
| | - Kent Hoskins
- College of Medicine, University of Illinois Chicago, Chicago, IL, USA.,Cancer Center, University of Illinois Chicago, Chicago, IL, USA
| | - Yamile Molina
- 14681Community Health Sciences Division, University of Illinois Chicago, Chicago, IL, USA.,Cancer Center, University of Illinois Chicago, Chicago, IL, USA
| |
Collapse
|
466
|
Physicians' perceptions of the factors influencing disclosure of secondary findings in tumour genomic profiling in Japan: a qualitative study. Eur J Hum Genet 2022; 30:88-94. [PMID: 34400810 PMCID: PMC8738764 DOI: 10.1038/s41431-021-00944-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/18/2021] [Accepted: 08/05/2021] [Indexed: 01/03/2023] Open
Abstract
Tumour genomic profiling (TGP), conducted in search of therapeutics, sometimes reveals potentially pathogenic germline variants as secondary findings (SFs). Physicians involved in TGP are often specialised in oncology and not in clinical genetics. To better utilise SFs, we explored issues physicians have during disclosure and the potential for collaborations with clinical genetics professionals. Semi-structured interviews were conducted with 14 physicians who had experience in handling outpatient TGP at designated core hospitals for cancer genomic medicine in Japan. The data were analysed thematically. The difficulties physicians experienced during informed consent (IC) included educating patients about SFs, providing detailed information on SFs, and explaining the impact of SFs on patients' family members. When SFs were detected, physicians had reservations regarding the relevance of the disclosure criteria. Confirmatory germline tests were performed using peripheral blood when tumour-only tests detected suspected SFs. Some physicians had reservations about the necessity of confirmatory tests when they did not affect the patients' treatment options. To encourage patients to receive confirmatory tests, improvements are necessary in the healthcare system, such as insurance reimbursements, education for physicians so that they can provide a better explanation to their patients, and genetic literacy of physicians and patients. The physicians offered insights into the challenges they experienced related to IC, disclosure of SFs, and expectations for active collaborations with clinical genetics professionals. Wider healthcare insurance coverage and better genetic literacy of the population may lead to more patients taking confirmatory tests when SFs are suspected.
Collapse
|
467
|
Shahani SA, Marcotte EL. Landscape of germline cancer predisposition mutations testing and management in pediatrics: Implications for research and clinical care. Front Pediatr 2022; 10:1011873. [PMID: 36225340 PMCID: PMC9548803 DOI: 10.3389/fped.2022.1011873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
As germline genetic testing capacities have improved over the last two decades, increasingly more people are newly diagnosed with germline cancer susceptibility mutations. In the wake of this growth, there remain limitations in both testing strategies and translation of these results into morbidity- and mortality-reducing practices, with pediatric populations remaining especially vulnerable. To face the challenges evoked by an expanding diversity of germline cancer mutations, we can draw upon a model cancer-associated genetic condition for which we have developed a breadth of expertise in managing, Trisomy 21. We can additionally apply advances in other disciplines, such as oncofertility and pharmacogenomics, to enhance care delivery. Herein, we describe the history of germline mutation testing, epidemiology of known germline cancer mutations and their associations with childhood cancer, testing limitations, and future directions for research and clinical care.
Collapse
Affiliation(s)
- Shilpa A Shahani
- Department of Pediatrics, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Erin L Marcotte
- Division of Epidemiology and Clinical Research, Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
468
|
da Silva Fontinele DR, Assunção Ribeiro da Costa RE, da Silva Magalhães MK, Vieira SC. Genetic testing for hereditary predisposition to breast cancer in the real world: Initial experience. Breast Dis 2022; 41:249-254. [PMID: 35570472 DOI: 10.3233/bd-210082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Around 5%-10% of breast cancers are due to hereditary breast and ovarian cancer syndrome. Genetic testing is important to identify these cases, enabling the adoption of specific risk-reducing treatment strategies. OBJECTIVE To analyze the performance of genetic testing and its implications in patients with indication of genetic testing to identify hereditary predisposition to breast cancer. METHODS This is a retrospective observational cross-sectional study, including 176 patients with clinical indication of genetic testing for pathogenic variants related to breast, ovarian and pancreatic cancers (among others), managed from 1999 to 2021 in an Oncology private clinic located in the city of Teresina (PI), Brazil. RESULTS There was a predominance of female patients (98.9%) and those with a family (91.0%) and personal history (64.2%) of cancer. In the study, 102 patients (57.9%) received genetic testing. BRCA1 and BRCA2 pathogenic variants occurred in 26 cases (90%). Another three PALB2 and TP53 pathogenic variants were detected. Eleven pathogenic variant carriers (38%) underwent risk-reducing surgeries. CONCLUSIONS BRCA1/BRCA2 pathogenic variants occurred in around 25% of tested patients. Approximately 42.0% of the patients did not undergo genetic testing, despite clinical indication.
Collapse
|
469
|
Jeong J, Park B, Kim D, Kim J, Lee BY, Yoon J, Kim SW. Sex Differences in Attitudes Toward Marriage and Childbearing Based on the Assumption of Being BRCA1/2 Mutation Carriers Among Young People. J Breast Cancer 2022; 25:233-243. [PMID: 35775702 PMCID: PMC9250881 DOI: 10.4048/jbc.2022.25.e25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 04/04/2022] [Accepted: 05/03/2022] [Indexed: 12/04/2022] Open
Abstract
Purpose This study investigated changes in attitudes toward marriage and childbearing assuming a BRCA1/2 mutation carrier status among healthy, unmarried individuals in Korea. Methods A nationally representative sample of healthy, unmarried individuals aged 20–39 years was surveyed. A questionnaire on marriage and childbearing intentions was administered to the participants before and after providing them with information on BRCA1/2 mutation carriers’ breast and ovarian cancer risks and their autosomal dominant inheritance pattern. The participants were asked about their attitudes toward childbearing through preimplantation genetic diagnosis (PGD). Results Of the participants who initially wanted to marry, the assumption that they or their partners had BRCA1/2 mutation caused 25.3% to no longer want to get married and 36.2% to change their attitude from wanting to bear children to no longer wanting them. Females were more likely than males to change their attitudes toward marriage and childbearing. The participants who had negative attitudes toward genetic testing were more likely to change their attitudes regarding marriage and childbearing than those who were favorable toward both disclosure and testing. More than 50% of the participants who did not want children were willing to bear children through PGD when it was assumed that they were BRCA mutation carriers. Conclusion On the assumption of being carriers, general, young, and healthy females were more likely than males to negatively change their attitudes toward marriage and childbearing. Public education on the implications of living with mutation carriers and reproductive options may be required.
Collapse
Affiliation(s)
- Jiwon Jeong
- Department of Internal Medicine, Daerim St. Mary’s Hospital, Seoul, Korea
| | - Boyoung Park
- Department of Preventive Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Dongwon Kim
- Department of Preventive Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Jiyoung Kim
- Department of Surgery, Daerim St. Mary’s Hospital, Seoul, Korea
| | - Bom-Yi Lee
- Department of Surgery, Daerim St. Mary’s Hospital, Seoul, Korea
| | - Junghyun Yoon
- Department of Health Sciences, Hanyang University College of Medicine, Seoul, Korea
| | - Sung-Won Kim
- Department of Surgery, Daerim St. Mary’s Hospital, Seoul, Korea
| |
Collapse
|
470
|
Rustgi SD, Hilfrank KJ, Kastrinos F. Familial Predisposition and Genetic Risk Factors Associated with Pancreatic Cancer. Gastrointest Endosc Clin N Am 2022; 32:1-12. [PMID: 34798979 DOI: 10.1016/j.giec.2021.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer (PC) is a highly lethal cancer and projected to be the second leading cause of cancer death by 2030. Multigene panel testing has facilitated the identification of germline variants associated with an increased risk of PC. Precision treatment has led to improved outcomes for patients with these findings. Because of these improved outcomes as well as the implications for at-risk family members who may benefit from additional cancer screening, the NCCN recommends universal genetic testing for newly diagnosed PC patients. This review describes the most common heritable conditions associated with PC and those who may benefit from screening.
Collapse
Affiliation(s)
- Sheila D Rustgi
- Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, 630 West 168th Street, Box 83, Room P&S 3-401, New York, NY 10032, USA
| | - Kimberly J Hilfrank
- Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, 630 West 168th Street, Box 83, Room P&S 3-401, New York, NY 10032, USA
| | - Fay Kastrinos
- Division of Digestive & Liver Diseases, Columbia University Irving Medical Center, 630 West 168th Street, Box 83, Room P&S 3-401, New York, NY 10032, USA.
| |
Collapse
|
471
|
Carleton N, Nasrazadani A, Gade K, Beriwal S, Barry PN, Brufsky AM, Bhargava R, Berg WA, Zuley ML, van Londen GJ, Marroquin OC, Thull DL, Mai PL, Diego EJ, Lotze MT, Oesterreich S, McAuliffe PF, Lee AV. Personalising therapy for early-stage oestrogen receptor-positive breast cancer in older women. THE LANCET. HEALTHY LONGEVITY 2022; 3:e54-e66. [PMID: 35047868 PMCID: PMC8765742 DOI: 10.1016/s2666-7568(21)00280-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Age is one of the most important risk factors for the development of breast cancer. Nearly a third of all breast cancer cases occur in older women (aged ≥70 years), with most cases being oestrogen receptor-positive (ER+). Such tumours are often indolent and unlikely to be the ultimate cause of death for older women, particularly when considering other comorbidities. This Review focuses on unique clinical considerations for screening, detection, and treatment regimens for older women who develop ER+ breast cancers-specifically, we focus on recent trends for de-implementation of screening, staging, surgery, and adjuvant therapies along the continuum of care. Additionally, we also review emerging basic and translational research that will further uncover the unique underlying biology of these tumours, which develop in the context of systemic age-related inflammation and changing hormone profiles. With prevailing trends of clinical de-implementation, new insights into mechanistic biology might provide an opportunity for precision medicine approaches to treat patients with well tolerated, low-toxicity agents to extend patients' lives with a higher quality of life, prevent tumour recurrences, and reduce cancer-related burdens.
Collapse
Affiliation(s)
- Neil Carleton
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Azadeh Nasrazadani
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Kristine Gade
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Sushil Beriwal
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Parul N Barry
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Adam M Brufsky
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Rohit Bhargava
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Wendie A Berg
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Margarita L Zuley
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - G J van Londen
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Oscar C Marroquin
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Darcy L Thull
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Phuong L Mai
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Emilia J Diego
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Michael T Lotze
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Steffi Oesterreich
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Priscilla F McAuliffe
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| | - Adrian V Lee
- (N Carleton BS, Prof S Oesterreich PhD, P F McAuliffe MD, Prof A V Lee PhD) (S Beriwal MD, P N Barry MD), (N Carleton, Prof S Oesterreich, P F McAuliffe, Prof A V Lee); (A Nasrazadani MD, K Gade MD, Prof A M Brufksy MD, G J van Londen MD), (Prof R Bhargava MD), (D L Thull MS, P L Mai MD), (E J Diego MD, Prof M T Lotze MD, P F McAuliffe), (Prof M T Lotze), (Prof M T Lotze), (Prof S Oesterreich, Prof A V Lee), (Prof W A Berg MD, Prof M L Zuley MD); (O C Marroquin MD)
| |
Collapse
|
472
|
Liu X, Yue J, Pervaiz R, Zhang H, Wang L. Association between fertility treatments and breast cancer risk in women with a family history or BRCA mutations: a systematic review and meta-analysis. Front Endocrinol (Lausanne) 2022; 13:986477. [PMID: 36176466 PMCID: PMC9513064 DOI: 10.3389/fendo.2022.986477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
UNLABELLED Women with hereditary breast cancer factors are more likely to be infertile and tend to receive fertility treatments. The safety of fertility treatments that contain hormone-related medications for ovarian stimulation has gained wide attention; however, evidence of the safety of fertility treatments is limited. This study aims to assess the association between fertility treatments and the incidence rate of breast cancer in women with a family history of breast cancer or BRCA mutations. A literature search was conducted in PubMed, Cochrane Library, and Embase. Studies concerning the effect of fertility treatments on breast cancer risk in genetically susceptible women were included. The fixed and random effects models were used to estimate the summary effects. Risk Of Bias In Non-randomized Studies - of Interventions instrument was used to assess the risk of bias in the included studies. A total of 5,282 studies were screened. Five cohort studies and three case-control studies were included. Breast cancer risk was not significantly increased by fertility treatments in general genetically susceptible women [pooled odds ratio (OR) 1.18, 95% confidence interval (CI) 0.96-1.45], women with a family history of breast cancer (pooled OR 1.35, 95% CI 0.97-1.89), or women with BRCA mutations (pooled OR 1.02, 95% CI 0.74-1.4). In subgroup analyses, there was no significant increase in breast cancer risk whether in BRCA1 mutation carriers (pooled OR 1.18, 95% CI 0.81-1.72), BRCA2 mutation carriers (pooled OR 0.54, 95% CI 0.09-3.34), or in the women treated with in vitro fertilization (pooled OR 0.75, 95% CI 0.51-1.1), clomiphene citrate (pooled OR 1.07, 95% CI 0.78-1.45) or gonadotropins (pooled OR 1.32, 95% CI 0.8-2.18). This is the first meta-analysis concerning the impact of fertility treatments on breast cancer risk in genetically susceptible women. Despite the finding that fertility treatment did not significantly increase breast cancer risk in genetically susceptible women, large prospective cohorts with more detailed information are required. Further investigations are needed to explore subtypes of breast cancer, genetic background of hormone-related breast cancer, and the association between BRCA mutations and the incidence of hormone receptor-positive breast cancer. REGISTRATION NUMBER PROSPERO(CRD42021281336).
Collapse
Affiliation(s)
- Xiaojing Liu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Jing Yue
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Ruqiya Pervaiz
- Faculty of Chemical and Life Science, Department of Zoology, Abdul Wali Khan University, Mardan, Pakistan
| | - Hanwang Zhang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Lan Wang, ; Hanwang Zhang,
| | - Lan Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Lan Wang, ; Hanwang Zhang,
| |
Collapse
|
473
|
Truong H, Carlo MI. Hereditary Cancer and Genetics in Renal Cell Carcinoma. Urol Oncol 2022. [DOI: 10.1007/978-3-030-89891-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
474
|
Sekine M, Enomoto T, Arai M, Den H, Nomura H, Ikeuchi T, Nakamura S. Differences in age at diagnosis of ovarian cancer for each BRCA mutation type in Japan: optimal timing to carry out risk-reducing salpingo-oophorectomy. J Gynecol Oncol 2022; 33:e46. [PMID: 35557031 PMCID: PMC9250856 DOI: 10.3802/jgo.2022.33.e46] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 12/10/2021] [Accepted: 03/05/2022] [Indexed: 12/04/2022] Open
Abstract
Objective BRCA1 and BRCA2 mutation carriers are recommended to undergo risk-reducing salpingo-oophorectomy (RRSO) by age 40 and 45, respectively. However, the carriers have a different way of thinking about their life plan. We aimed to investigate the distribution of age at diagnosis of ovarian cancer (OC) patients to examine the optimal timing of RRSO in the carriers. Methods We examined a correlation between age at diagnosis of OC and common mutation types in 3,517 probands that received BRCA genetic testing. Among them, germline BRCA1 mutation (gBRCA1m), germline BRCA2 mutation (gBRCA2m) and germline BRCA wild-type (gBRCAwt) were found in 185, 42 and 241 OC patients, respectively. Results The average age at diagnosis of OC in gBRCA1m and gBRCA2m was 51.3 and 58.3 years, respectively, and the difference from gBRCAwt (53.8 years) was significant. The gBRCA2m carriers did not develop OC under the age of 40. The average age was 50.1 years for L63X and 52.8 years for Q934X in BRCA1, and 55.1 years for R2318X and 61.1 years for STOP1861 in BRCA2. The age at diagnosis in L63X or R2318X carriers was relatively younger than other BRCA1 or BRCA2 carriers, however their differences were not significant. With L63X and R2318X carriers, 89.4% (42/47) and 100% (7/7) of women were able to prevent the development of OC, respectively, when RRSO was performed at age 40. Conclusion There appears to be no difference in the age at diagnosis of OC depending on the type of BRCA common mutation. Further analysis would be needed. This is the first report to present the optimal timing of risk-reducing salpingo-oophorectomy for each BRCA mutation type in Japan. The average age at diagnosis of ovarian cancer (OC) in germline BRCA2 mutation (gBRCA2m) is higher than that in germline BRCA wild-type. The gBRCA2m carriers did not develop OC under the age of 40. The average age at diagnosis in L63X or R2318X carriers was relatively younger than other BRCA mutation carriers, though the differences were not significant.
Collapse
Affiliation(s)
- Masayuki Sekine
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Masami Arai
- Clinical Genetics, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Hiroki Den
- Department of Hygiene, Public Health, and Preventative Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hiroyuki Nomura
- Department of Obstetrics and Gynecology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Seigo Nakamura
- Division of Breast Surgical Oncology, Department of Surgery, Showa University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
475
|
Mampel A, Sottile ML, Denita-Juárez SP, Vargas AL, Vargas-Roig LM. Double heterozygous pathogenic variants in the BRCA1 and BRCA2 genes in a patient with bilateral metachronous breast cancer. Cancer Genet 2022; 260-261:14-17. [PMID: 34801929 DOI: 10.1016/j.cancergen.2021.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/24/2021] [Accepted: 11/08/2021] [Indexed: 12/20/2022]
Abstract
Double heterozygosity pathogenic variants in BRCA1 and BRCA2 genes are a very rare finding, particularly in non-Ashkenazi individuals. We described the first case of double heterozygosity variants in a non-Ashkenazi Argentinean woman with metachronous bilateral breast cancer. The proband is a 65-year-old female diagnosed with invasive ductal carcinoma in the left breast at 45 years old and invasive carcinoma in the right breast at 65 years old. She underwent a multi-gene panel testing indicating the presence of two concurrent heterozygous germline deleterious variants NM_007300.4(BRCA1):c.4201C>T (p.Gln1401Ter), and NM_000059.3(BRCA2):c.5146_5149del (p.Tyr1716fs). . The patient's son (40 years-old) was found to have the inherited pathogenic variant in BRCA2 gene. There are few reports of double heterozygosity variants in BRCA1 and BRCA2 genes in Latin America. The two pathogenic variants identified in our patient have not been described together so far.
Collapse
Affiliation(s)
- Alejandra Mampel
- University Hospital, Mendoza, Argentina; Medical School, National University of Cuyo, Mendoza, Argentina.
| | - Mayra L Sottile
- Tumor Biology Laboratory, Institute of Medicine and Experimental Biology of Cuyo, National Research Council of Argentine-National University of Cuyo, Mendoza, Argentina; Medical School, University of Mendoza, Mendoza, Argentina
| | | | - Ana L Vargas
- University Hospital, Mendoza, Argentina; Medical School, National University of Cuyo, Mendoza, Argentina
| | - Laura M Vargas-Roig
- Medical School, National University of Cuyo, Mendoza, Argentina; Tumor Biology Laboratory, Institute of Medicine and Experimental Biology of Cuyo, National Research Council of Argentine-National University of Cuyo, Mendoza, Argentina
| |
Collapse
|
476
|
Kurian AW, Abrahamse P, Ward KC, Hamilton AS, Deapen D, Berek JS, Hoang L, Yussuf A, Dolinsky J, Brown K, Slavin T, Hofer TP, Katz SJ. Association of Family Cancer History With Pathogenic Variants in Specific Breast Cancer Susceptibility Genes. JCO Precis Oncol 2021; 5:PO.21.00261. [PMID: 34977446 PMCID: PMC8710333 DOI: 10.1200/po.21.00261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/12/2021] [Accepted: 11/22/2021] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Family cancer history is an important component of genetic testing guidelines that estimate which patients with breast cancer are most likely to carry a germline pathogenic variant (PV). However, we do not know whether more extensive family history is differentially associated with PVs in specific genes. METHODS All women diagnosed with breast cancer in 2013-2017 and reported to statewide SEER registries of Georgia and California were linked to clinical genetic testing results and family history from two laboratories. Family history was defined as strong (suggestive of PVs in high-penetrance genes such as BRCA1/2 or TP53, including male breast, ovarian, pancreatic, sarcoma, or multiple female breast cancers), moderate (any other cancer history), or none. Among established breast cancer susceptibility genes (ATM, BARD1, BRCA1, BRCA2, CDH1, CHEK2, NF1, PALB2, PTEN, RAD51C, RAD51D, and TP53), we evaluated PV prevalence according to family history extent and breast cancer subtype. We used a multivariable model to test for interaction between affected gene and family history extent for ATM, BRCA1/2, CHEK2, and PALB2. RESULTS A total of 34,865 women linked to genetic results. Higher PV prevalence with increasing family history extent (P < .001) was observed only with BRCA1 (3.04% with none, 3.22% with moderate, and 4.06% with strong history) and in triple-negative breast cancer with PALB2 (0.75% with none, 2.23% with moderate, and 2.63% with strong history). In a multivariable model adjusted for age and subtype, there was no interaction between family history extent and PV prevalence for any gene except PALB2 (P = .037). CONCLUSION Extent of family cancer history is not differentially associated with PVs across established breast cancer susceptibility genes and cannot be used to personalize genes selected for testing.
Collapse
Affiliation(s)
- Allison W. Kurian
- Departments of Medicine and of Epidemiology & Population Health, Stanford University, Stanford, CA,Allison W. Kurian, MD, MSc, Department of Medicine and of Epidemiology & Population Health, Stanford University School of Medicine, HRP Redwood Building, Room T254A, 150 Governor's Lane, Stanford, CA 94305; e-mail:
| | - Paul Abrahamse
- Department of Health Management and Policy, School of Public Health, University of Michigan, Ann Arbor, MI,Department of Biostatistics, University of Michigan, Ann Arbor, MI
| | - Kevin C. Ward
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Ann S. Hamilton
- Department of Preventive Medicine in the Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Dennis Deapen
- Department of Preventive Medicine in the Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Jonathan S. Berek
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, and Stanford Women's Cancer Center, Stanford University, Stanford, CA
| | | | | | | | | | | | - Timothy P. Hofer
- Department of Internal Medicine, University of Michigan and Center for Clinical Management Research, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI
| | - Steven J. Katz
- Department of Internal Medicine, University of Michigan and Center for Clinical Management Research, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI
| |
Collapse
|
477
|
Advances in the Aetiology & Endoscopic Detection and Management of Early Gastric Cancer. Cancers (Basel) 2021; 13:cancers13246242. [PMID: 34944861 PMCID: PMC8699285 DOI: 10.3390/cancers13246242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Gastric adenocarcinoma has remained a highly lethal disease. Awareness and recognition of preneoplastic conditions (including gastric atrophy and intestinal metaplasia) using high-resolution white-light endoscopy as well as chromoendoscopy is therefore essential. Helicobacter pylori, a class I carcinogen, remains the main contributor to the development of sporadic distal gastric neoplasia. Management of early gastric neoplasia with endoscopic resections should be in line with standard indications. A multidisciplinary approach to any case of an early gastric neoplasia is imperative. Hereditary forms of gastric cancer require a tailored approach and individua-lized surveillance. Abstract The mortality rates of gastric carcinoma remain high, despite the progress in research and development in disease mechanisms and treatment. Therefore, recognition of gastric precancerous lesions and early neoplasia is crucial. Two subtypes of sporadic gastric cancer have been recognized: cardia subtype and non-cardia (distal) subtype, the latter being more frequent and largely associated with infection of Helicobacter pylori, a class I carcinogen. Helicobacter pylori initiates the widely accepted Correa cascade, describing a stepwise progression through precursor lesions from chronic inflammation to gastric atrophy, gastric intestinal metaplasia and neoplasia. Our knowledge on He-licobacter pylori is still limited, and multiple questions in the context of its contribution to the pathogenesis of gastric neoplasia are yet to be answered. Awareness and recognition of gastric atrophy and intestinal metaplasia on high-definition white-light endoscopy, image-enhanced endoscopy and magnification endoscopy, in combination with histology from the biopsies taken accurately according to the protocol, are crucial to guiding the management. Standard indications for endoscopic resections (endoscopic mucosal resection and endoscopic submucosal dissection) of gastric dysplasia and intestinal type of gastric carcinoma have been recommended by multiple societies. Endoscopic evaluation and surveillance should be offered to individuals with an inherited predisposition to gastric carcinoma.
Collapse
|
478
|
Su R, Liu Y, Wu X, Xiang J, Xi X. Dynamically Accumulating Homologous Recombination Deficiency Score Served as an Important Prognosis Factor in High-Grade Serous Ovarian Cancer. Front Mol Biosci 2021; 8:762741. [PMID: 34869593 PMCID: PMC8640082 DOI: 10.3389/fmolb.2021.762741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/25/2021] [Indexed: 11/30/2022] Open
Abstract
Background: The homologous recombination (HR) pathway defects in cancers induced abrogation of cell cycle checkpoints, resulting in the accumulation of DNA damage, mitotic catastrophe, and cell death. Cancers with BRCA1/2 loss and other accumulation of similar genomic scars resulting in HRD displayed increased sensitivity to chemotherapy. Our study aimed to explore HRD score genetic mechanisms and subsequent clinical outcomes in human cancers, especially ovarian cancer. Methods: We analyzed TCGA data of HRD score in 33 cancer types and evaluated HRD score distribution and difference among tumor stages and between primary and recurrent tumor tissues. A weighted gene co-expression network analysis (WGCNA) was performed to identify highly correlated genes representing essential modules contributing to the HRD score and distinguish the hub genes and significant pathways. We verified HRD status predicting roles in patients’ overall survival (OS) with univariate and multivariate Cox regression analyses and built the predicting model for patient survival. Results: We found that the HRD score increased with the rise in tumor stage, except for stage IV. The HRD score tended to grow up higher in recurrent tumor tissue than in their primary counterparts (p = 0.083). We constructed 15 co-expression modules with WGCNA, identified co-expressed genes and pathways impacting the HRD score, and concluded that the HRD score was tightly associated with tumor cells replication and proliferation. A combined HRD score ≥42 was associated with shorter OS in 33 cancer types (HR = 1.010, 95% CI: 1.008–1.011, p < 0.001). However, in ovarian cancer, which ranked the highest HRD score among other cancers, HRD ≥42 cohort was significantly associated with longer OS (HR = 0.99, 95% CI: 0.98–0.99, p < 0.0001). We also built a predicting model for 3 and 5 years survival in HGSC patients. Conclusion: A quantitative HRD score representing the accumulated genomic scars was dynamically increasing in proliferating tumor cells since the HRD score was tightly correlated to tumor cell division and replication. We highlighted HRD score biomarker role in prognosis prediction of ovarian cancer.
Collapse
Affiliation(s)
- Rongjia Su
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuan Liu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaomei Wu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiangdong Xiang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaowei Xi
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
479
|
Garcia-Pelaez J, Barbosa-Matos R, São José C, Sousa S, Gullo I, Hoogerbrugge N, Carneiro F, Oliveira C. Gastric cancer genetic predisposition and clinical presentations: Established heritable causes and potential candidate genes. Eur J Med Genet 2021; 65:104401. [PMID: 34871783 DOI: 10.1016/j.ejmg.2021.104401] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 11/10/2021] [Accepted: 11/28/2021] [Indexed: 12/12/2022]
Abstract
Tumour risk syndromes (TRS) are characterized by an increased risk of early-onset cancers in a familial context. High cancer risk is mostly driven by loss-of-function variants in a single cancer-associated gene. Presently, predisposition to diffuse gastric cancer (DGC) is explained by CDH1 and CTNNA1 pathogenic and likely pathogenic variants (P/LP), causing Hereditary Diffuse Gastric Cancer (HDGC); while APC promoter 1B single nucleotide variants predispose to Gastric Adenocarcinoma and Proximal Polyposis of the Stomach (GAPPS). Familial Intestinal Gastric Cancer (FIGC), recognized as a GC-predisposing disease, remains understudied and genetically unsolved. GC can also occur in the spectrum of other TRS. Identification of heritable causes allows defining diagnostic testing criteria, helps to clinically classify GC families into the appropriate TRS, and allows performing pre-symptomatic testing identifying at-risk individuals for downstream surveillance, risk reduction and/or treatment. However, most of HDGC, some GAPPS, and most FIGC patients/families remain unsolved, expecting a heritable factor to be discovered. The missing heritability in GC-associated tumour risk syndromes (GC-TRS) is likely explained not by a single major gene, but by a diversity of genes, some, predisposing to other TRS. This would gain support if GC-enriched small families or apparently isolated early-onset GC cases were hiding a family history compatible with another TRS. Herein, we revisited current knowledge on GC-TRS, and searched in the literature for individuals/families bearing P/LP variants predisposing for other TRS, but whose probands display a clinical presentation and/or family history also fitting GC-TRS criteria. We found 27 families with family history compatible with HDGC or FIGC, harbouring 28 P/LP variants in 16 TRS-associated genes, mainly associated with DNA repair. PALB2 or BRCA2 were the most frequently mutated candidate genes in individuals with family history compatible with HDGC and FIGC, respectively. Consolidation of PALB2 and BRCA2 as HDGC- or FIGC-associated genes, respectively, holds promise and worth additional research. This analysis further highlighted the influence, that proband's choice and small or unreported family history have, for a correct TRS diagnosis, genetic screening, and disease management. In this review, we provide a rational for identification of particularly relevant candidate genes in GC-TRS.
Collapse
Affiliation(s)
- José Garcia-Pelaez
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; Doctoral Programme in Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rita Barbosa-Matos
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; International Doctoral Programme in Molecular and Cellular Biotechnology Applied to Health Sciences from Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Celina São José
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; Doctoral Programme in Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Sónia Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Irene Gullo
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; FMUP - Faculty of Medicine of the University of Porto, Porto, Portugal; Centro Hospitalar e Universitário S. João, Porto, Portugal
| | - Nicoline Hoogerbrugge
- Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Fátima Carneiro
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; FMUP - Faculty of Medicine of the University of Porto, Porto, Portugal; Centro Hospitalar e Universitário S. João, Porto, Portugal
| | - Carla Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal; FMUP - Faculty of Medicine of the University of Porto, Porto, Portugal.
| |
Collapse
|
480
|
Martinez-Cannon BA, Barragan-Carrillo R, Villarreal-Garza C. Young Women with Breast Cancer in Resource-Limited Settings: What We Know and What We Need to Do Better. BREAST CANCER (DOVE MEDICAL PRESS) 2021; 13:641-650. [PMID: 34880675 PMCID: PMC8648095 DOI: 10.2147/bctt.s303047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/23/2021] [Indexed: 06/13/2023]
Abstract
Young women with breast cancer (YWBC) account for a variable proportion of patients diagnosed with breast cancer around the globe, with a higher prevalence in resource-limited settings than in high-income countries. This group represents a unique population that warrants special attention due to specific biological considerations and age-specific supportive care issues. This review aims to explore existing knowledge regarding YWBC's needs, particularly in resource-restricted settings. To date, scarce information regarding the care of YWBC in resource-constrained countries is available, with most reports describing suboptimal care in terms of survivorship needs. Health care providers should implement actions to improve endocrine treatment adherence, referrals for fertility counseling and preservation, contraceptive use compliance, timely body image and sexual function interventions, comprehensive genetic risk assessments, and early quality of life and psychosocial health interventions. While high costs act as a barrier for optimal care in resource-limited settings, improving patient education represents a promising and cost-effective solution to improve patient care. Future research on developing tailored educational resources for YWBC in resource-limited settings should be considered a priority.
Collapse
Affiliation(s)
- Bertha Alejandra Martinez-Cannon
- Hematology-Oncology Department, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
- Joven & Fuerte: Programa para la Atencion e Investigacion de Mujeres Jovenes con Cancer de Mama en Mexico, Mexico City, Mexico
| | - Regina Barragan-Carrillo
- Hematology-Oncology Department, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
- Joven & Fuerte: Programa para la Atencion e Investigacion de Mujeres Jovenes con Cancer de Mama en Mexico, Mexico City, Mexico
| | - Cynthia Villarreal-Garza
- Joven & Fuerte: Programa para la Atencion e Investigacion de Mujeres Jovenes con Cancer de Mama en Mexico, Mexico City, Mexico
- Breast Cancer Center, Hospital Zambrano Hellion TecSalud, Tecnologico de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| |
Collapse
|
481
|
Karia PS, Joshu CE, Visvanathan K. Uptake and Predictors of Opportunistic Salpingectomy for Ovarian Cancer Risk Reduction in the United States. Cancer Prev Res (Phila) 2021; 14:1101-1110. [PMID: 34413116 PMCID: PMC8756422 DOI: 10.1158/1940-6207.capr-21-0121] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/30/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022]
Abstract
Evidence suggesting that high-grade serous ovarian cancers originate in the fallopian tubes has led to the emergence of opportunistic salpingectomy (OS) as an approach to reduce ovarian-cancer risk. In the U.S., some national societies now recommend OS in place of tubal ligation for sterilization or during a benign hysterectomy in average-risk women. However, limited data exist on the dissemination of OS in clinical practice. We examined the uptake and predictors of OS in a nationwide sample of inpatient and outpatient claims (N = 48,231,235) from 2010 to 2017. Incidence rates of OS were calculated, and an interrupted time-series analysis was used to quantify changes in rates before (2010-2013) and after (2015-2017) national guideline release. Predictors of OS use were examined using Poisson regression. From 2010 to 2017, the age-adjusted incidence rate of OS for sterilization and OS during hysterectomy increased 17.8-fold [95% confidence interval (CI), 16.2-19.5] and 7.6-fold (95% CI, 5.5-10.4), respectively. The rapid increase (age-adjusted increase in quarterly rates of between 109% and 250%) coincided with the time of national guideline release. In multivariable-adjusted analyses, OS use was more common in young women and varied significantly by geographic region, rurality, family history/genetic susceptibility, surgical indication, inpatient/outpatient setting, and underlying comorbidities. Similar differences in OS uptake were noted in analyses limited to women with a family history/genetic susceptibility to breast/ovarian cancer. Our results highlight significant differences in OS uptake in both high- and average-risk women. Defining subsets of women who would benefit most from OS and identifying barriers to equitable OS uptake is needed. PREVENTION RELEVANCE: Opportunistic salpingectomy for ovarian-cancer risk reduction has been rapidly adopted in the U.S., with significant variation in uptake by demographic and clinical factors. Studies examining barriers to opportunistic salpingectomy access and the long-term effectiveness and potential adverse effects of opportunistic salpingectomy are needed.
Collapse
Affiliation(s)
- Pritesh S Karia
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York
| | - Corinne E Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, Maryland
| | - Kala Visvanathan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, Maryland
| |
Collapse
|
482
|
Reisel D, Baran C, Manchanda R. Preventive population genomics: The model of BRCA related cancers. ADVANCES IN GENETICS 2021; 108:1-33. [PMID: 34844711 DOI: 10.1016/bs.adgen.2021.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Preventive population genomics offers the prospect of population stratification for targeting screening and prevention and tailoring care to those at greatest risk. Within cancer, this approach is now within reach, given our expanding knowledge of its heritable components, improved ability to predict risk, and increasing availability of effective preventive strategies. Advances in technology and bioinformatics has made population-testing technically feasible. The BRCA model provides 30 years of insight and experience of how to conceive of and construct care and serves as an initial model for preventive population genomics. Population-based BRCA-testing in the Jewish population is feasible, acceptable, reduces anxiety, does not detrimentally affect psychological well-being or quality of life, is cost-effective and is now beginning to be implemented. Population-based BRCA-testing and multigene panel testing in the wider general population is cost-effective for numerous health systems and can save thousands more lives than the current clinical strategy. There is huge potential for using both genetic and non-genetic information in complex risk prediction algorithms to stratify populations for risk adapted screening and prevention. While numerous strides have been made in the last decade several issues need resolving for population genomics to fulfil its promise and potential for maximizing precision prevention. Healthcare systems need to overcome significant challenges associated with developing delivery pathways, infrastructure expansion including laboratory services, clinical workforce training, scaling of management pathways for screening and prevention. Large-scale real world population studies are needed to evaluate context specific population-testing implementation models for cancer risk prediction, screening and prevention.
Collapse
Affiliation(s)
- Dan Reisel
- EGA Institute for Women's Health, University College London, London, United Kingdom
| | - Chawan Baran
- Wolfson Institute of Preventive Medicine, CRUK Barts Centre, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Ranjit Manchanda
- Wolfson Institute of Preventive Medicine, CRUK Barts Centre, Queen Mary University of London, Charterhouse Square, London, United Kingdom; Department of Gynaecological Oncology, St Bartholomew's Hospital, London, United Kingdom; Department of Health Services Research, London School of Hygiene & Tropical Medicine, London, United Kingdom.
| |
Collapse
|
483
|
Creating Breast and Gynecologic Cancer Guidelines for Transgender Patients With BRCA Mutations. Obstet Gynecol 2021; 138:911-917. [PMID: 34735408 DOI: 10.1097/aog.0000000000004597] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
More than 1.5 million individuals in the United States identify as transgender. Transgender individuals have lower rates of health care utilization and higher rates of health care discrimination than cisgender patients. With a growing interest in providing comprehensive and compassionate care to the transgender community, there has been a concurrent increase in research on transgender health. However, lack of long-term data limits understanding the effects of hormone therapy on cancer risk factors in this population. This is particularly relevant for patients with hormonally mediated cancers and those at elevated risk from hereditary breast and ovarian cancer syndromes. Few cancer-screening and management guidelines currently exist for this population. Specific practices guided by the nuances of gender identity and gender-affirming care are essential to improve clinical management and to avoid further alienating a population that is already marginalized from the health care system. This commentary summarizes screening, management, and surveillance strategies devised for cisgender patients to offer corresponding recommendations tailored for transgender BRCA mutation carriers. In doing so, it highlights critical unanswered questions pertaining to the care of these patients. To address these questions, we must prioritize this population and adopt more inclusive frameworks in medicine and research.
Collapse
|
484
|
Advances in urologic oncology "OncoUrology Forum Special Edition": The best of 2020. Actas Urol Esp 2021; 46:214-222. [PMID: 34844900 DOI: 10.1016/j.acuroe.2021.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/05/2021] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To provide latest findings of Urologic Oncology on prostate, kidney, and bladder cancer, and analyze its impact on clinical practice as well as future schemes in the medium- and long-term. METHODS This document reviews the abstracts on Uro-Oncology presented at the 2020 Congresses (EUA, AUA, ASCO, ESMO and ASTRO), the publications with the highest impact and especially the new lines of development and progress in Uro-Oncology evaluated by the OncoForum committee. RESULTS The use of prostate-specific membrane antigen (PSMA) radioligands in the diagnosis of prostate cancer may have great potential and utility in the coming years due to their improved sensitivity and specificity. The genetic characterization of the tumor is important at both, germline and somatic levels, due to the significant role of BRCA2 mutations regarding risk. The cohort multiple randomised controlled trial is the most suitable study design at the genitourinary cancer level. The application of big data will lead to process improvements, savings in healthcare costs, and an empowerment of real-life studies through ease of data comparison, management, and storage. CONCLUSIONS The use of new diagnostic techniques with PSMA ligands will provide a more comprehensive diagnostic modality, increase the number of studies about tumor genetic profiling, and enhance their quality. The practical application of artificial intelligence will improve the treatment genitourinary cancer.
Collapse
|
485
|
Rahman WT, Helvie MA. Breast cancer screening in average and high-risk women. Best Pract Res Clin Obstet Gynaecol 2021; 83:3-14. [PMID: 34903436 DOI: 10.1016/j.bpobgyn.2021.11.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/28/2022]
Abstract
Breast cancer is the most common cancer among females worldwide with rising incidence. In the United States, screening mammography and advances in therapy have lowered mortality by 41% since 1990. Screening mammography is supported by randomized control trials (RCT), observational studies, and computer model data. Digital breast tomosynthesis is a new technology that addresses limitations in mammography resulting from overlapping breast tissue, improving its sensitivity and specificity. Patients at high risk for breast cancer include those with a ≥20% lifetime risk, high-risk germline mutation, or history of thoracic radiation treatment between 10-30 years of age. Such patients are recommended to undergo annual screening mammography and adjunctive annual screening breast MRI. Patients unable to undergo MRI may undergo whole breast ultrasound or contrast-enhanced mammography. Pregnant and lactating patients at average risk for breast cancer are recommended to undergo age-appropriate screening mammography.
Collapse
Affiliation(s)
- W Tania Rahman
- Department of Radiology, Division of Breast Imaging, 1500 E. Medical Center Dr., Ann Arbor, MI, 48109, USA.
| | - Mark A Helvie
- Department of Radiology, Division of Breast Imaging, 1500 E. Medical Center Dr., Ann Arbor, MI, 48109, USA
| |
Collapse
|
486
|
Dillon J, Ademuyiwa FO, Barrett M, Moss HA, Wignall E, Menendez C, Hughes KS, Plichta JK. Disparities in Genetic Testing for Heritable Solid-Tumor Malignancies. Surg Oncol Clin N Am 2021; 31:109-126. [PMID: 34776060 DOI: 10.1016/j.soc.2021.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Genetic testing offers providers a potentially life saving tool for identifying and intervening in high-risk individuals. However, disparities in receipt of genetic testing have been consistently demonstrated and undoubtedly have significant implications for the populations not receiving the standard of care. If correctly used, there is the potential for genetic testing to play a role in decreasing health disparities among individuals of different races and ethnicities. However, if genetic testing continues to revolutionize cancer care while being disproportionately distributed, it also has the potential to widen the existing mortality gap between various racial and ethnic populations.
Collapse
Affiliation(s)
- Jacquelyn Dillon
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Foluso O Ademuyiwa
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Megan Barrett
- Department of Obstetrics & Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Haley A Moss
- Department of Obstetrics & Gynecology, Duke University Medical Center, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA. https://twitter.com/haleyarden1
| | | | - Carolyn Menendez
- Department of Surgery, Duke University Medical Center, Durham, NC, USA; Clinical Cancer Genetics, Duke Cancer Institute, Durham, NC, USA. https://twitter.com/@CSMenendez
| | - Kevin S Hughes
- Surgical Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Jennifer K Plichta
- Department of Surgery, Duke University Medical Center, Durham, NC, USA; Department of Population Health Sciences, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
487
|
A Review of Breast Cancer Risk Factors in Adolescents and Young Adults. Cancers (Basel) 2021; 13:cancers13215552. [PMID: 34771713 PMCID: PMC8583289 DOI: 10.3390/cancers13215552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Cancer diagnosed in patients between the ages of 15 and 39 deserves special consideration. Diagnoses within this cohort of adolescents and young adults include childhood cancers which present at an older age than expected, or an early presentation of cancers that are typically observed in older adults, such as breast cancer. Cancers within this age group are associated with worse disease-free and overall survival rates, and the incidence of these cases are rising. Knowing an individual’s susceptibility to disease can change their clinical management and allow for the risk-testing of relatives. This review discusses the risk factors that contribute to breast cancer in this unique cohort of patients, including inherited genetic risk factors, as well as environmental and lifestyle factors. We also describe risk models that allow clinicians to quantify a patient’s lifetime risk of developing disease. Abstract Cancer in adolescents and young adults (AYAs) deserves special consideration for several reasons. AYA cancers encompass paediatric malignancies that present at an older age than expected, or early-onset of cancers that are typically observed in adults. However, disease diagnosed in the AYA population is distinct to those same cancers which are diagnosed in a paediatric or older adult setting. Worse disease-free and overall survival outcomes are observed in the AYA setting, and the incidence of AYA cancers is increasing. Knowledge of an individual’s underlying cancer predisposition can influence their clinical care and may facilitate early tumour surveillance strategies and cascade testing of at-risk relatives. This information can further influence reproductive decision making. In this review we discuss the risk factors contributing to AYA breast cancer, such as heritable predisposition, environmental, and lifestyle factors. We also describe a number of risk models which incorporate genetic factors that aid clinicians in quantifying an individual’s lifetime risk of disease.
Collapse
|
488
|
Uptake and timing of risk-reducing salpingo-oophorectomy among patients with BRCA1 and BRCA2 mutations. Am J Obstet Gynecol 2021; 225:508.e1-508.e10. [PMID: 34171390 DOI: 10.1016/j.ajog.2021.06.070] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND In women with BRCA mutations, risk-reducing bilateral salpingo-oophorectomy has been shown to decrease gynecologic cancer-specific and overall mortality. The National Comprehensive Cancer Network recommends that patients with BRCA mutations undergo risk-reducing bilateral salpingo-oophorectomy between the ages of 35 and 40 years for BRCA1 mutation carriers and between the ages of 40 and 45 years for BRCA2 mutation carriers or after childbearing is complete. Currently, uptake and timing of risk-reducing bilateral salpingo-oophorectomy and reasons for delays in risk-reducing bilateral salpingo-oophorectomy are not well understood. OBJECTIVE We sought to evaluate uptake and timing of risk-reducing bilateral salpingo-oophorectomy among women with BRCA1 and BRCA2 mutations concerning the National Comprehensive Cancer Network guidelines and reasons for delays in risk-reducing bilateral salpingo-oophorectomy. STUDY DESIGN In this retrospective chart review, we identified women with BRCA1 and BRCA2 mutations who discussed risk-reducing bilateral salpingo-oophorectomy with a provider between 2012 and 2021. Uptake of risk-reducing bilateral salpingo-oophorectomy was documented, and patients were classified as having timely or delay in risk-reducing bilateral salpingo-oophorectomy based on the National Comprehensive Cancer Network guidelines. For those with delay in risk-reducing bilateral salpingo-oophorectomy, reasons cited for delay were collected. Comparative statistical analyses were performed to evaluate characteristics of those with timely vs delayed risk-reducing bilateral salpingo-oophorectomy. A multivariable logistic regression model was used to evaluate the associations among factors related to timing of risk-reducing bilateral salpingo-oophorectomy. RESULTS We identified 638 BRCA1 and BRCA2 mutation carriers seen between 2012 and 2021. Of these patients, 306 (48.0%) had undergone risk-reducing bilateral salpingo-oophorectomy and 332 (52.0%) had not. When evaluating the timing of risk-reducing bilateral salpingo-oophorectomy, 136 (21.3%) underwent timely risk-reducing bilateral salpingo-oophorectomy, 239 (37.5%) had delays in risk-reducing bilateral salpingo-oophorectomy, and 263 (41.2%) had not undergone risk-reducing bilateral salpingo-oophorectomy but were younger than the National Comprehensive Cancer Network age guidelines; therefore, they were neither timely nor delayed. Patients with delay in risk-reducing bilateral salpingo-oophorectomy were significantly older at the time of genetic testing than those with timely risk-reducing bilateral salpingo-oophorectomy (mean, 49.8 vs 36.3 years; P<.001). Of the 306 patients who underwent risk-reducing bilateral salpingo-oophorectomy, those with delayed risk-reducing bilateral salpingo-oophorectomy had a significantly shorter interval between BRCA identification and risk-reducing bilateral salpingo-oophorectomy than those with timely risk-reducing bilateral salpingo-oophorectomy (median, 8.7 vs 17.6 months; P<.001). Patients with delay in risk-reducing bilateral salpingo-oophorectomy were more likely to have a personal history of cancer than those with timely risk-reducing bilateral salpingo-oophorectomy (49.8% vs 37.5%; P=.028). Of the 239 women with delay in risk-reducing bilateral salpingo-oophorectomy, 188 (78.7%) had delayed BRCA mutation identification, 29 (12.1%) had menopausal concerns, 17 (7.1%) had ongoing cancer treatment, 12 (5.0%) had coordination with breast surgery, 20 (8.4%) had miscellaneous reasons, and 19 (7.9%) had no reason documented. In the multivariate model, older age at BRCA diagnosis (odds ratio, 0.73; 95% confidence interval, 0.68-0.78; P<.001) was significantly associated with delayed risk-reducing bilateral salpingo-oophorectomy timing; those with BRCA2 mutation type were 7.54 times as likely to have timely risk-reducing bilateral salpingo-oophorectomy than BRCA1 mutation carriers (odds ratio, 7.54; 95% confidence, 3.70-16.42; P<.001). CONCLUSION Nearly 38% of BRCA1 and BRCA2 mutation carriers undergo or have yet to undergo risk-reducing bilateral salpingo-oophorectomy over the recommended National Comprehensive Cancer Network age. The most common reason for the delay in risk-reducing bilateral salpingo-oophorectomy was delayed identification of BRCA mutation, noted in 79% of patients with delayed risk-reducing bilateral salpingo-oophorectomy. Timely genetic testing for eligible patients can increase appropriately timed risk-reducing bilateral salpingo-oophorectomy for the prevention of ovarian cancer and reduction of mortality in BRCA mutation carriers.
Collapse
|
489
|
Schwartz AN, Hyman SR, Stokes SM, Castillo D, Tung NM, Weitzel JN, Rana HQ, Garber JE. Evaluation of TP53 Variants Detected on Peripheral Blood or Saliva Testing: Discerning Germline From Somatic TP53 Variants. JCO Precis Oncol 2021; 5:1677-1686. [PMID: 34994652 DOI: 10.1200/po.21.00278] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/15/2021] [Accepted: 09/24/2021] [Indexed: 01/04/2023] Open
Abstract
PURPOSE Multigene panel testing (MGPT) identifies TP53 pathogenic or likely pathogenic (P/LP) variants in patients with diverse phenotypes, of which only one is classic Li-Fraumeni syndrome. Low variant allelic fraction (VAF) in TP53 found on germline testing may suggest aberrant clonal expansion or constitutional mosaicism. We evaluated TP53-positive probands seen in a cancer genetics program to determine germline versus somatic status. METHODS We reviewed TP53-positive probands from 2012 to 2019 identified by MGPT on blood or saliva (N = 84). Available VAFs were collected. Probands with a familial variant, who met Li-Fraumeni syndrome testing criteria or who carried a founder variant, were considered germline. For those with uncertain germline status, TP53 variants were further examined using ancillary data of family members and somatic tissue. RESULTS Of the 84 probands, 54.7% had germline variants with 33.3% meeting criteria for germline status and 21.4% confirmed through ancillary testing. Aberrant clonal expansion comprised 13.1% with clonal hematopoiesis of indeterminate potential and 2.4% with a hematologic malignancy. Constitutional mosaicism was confirmed in 8.3% probands. Definitive status could not be determined in 3.6% despite ancillary assessment, and 17.9% did not have ancillary testing. CONCLUSION A TP53 P/LP variant found on peripheral blood or saliva MGPT does not always originate in the germline. In a clinical cancer genetics cohort, approximately half of the patients had TP53 P/LP germline variants; these patients plus those with constitutional mosaicism require intensified surveillance. A framework of multiple strategies enables discernment of germline from constitutional mosaic and acquired variants, which is essential for appropriate management.
Collapse
Affiliation(s)
- Alison N Schwartz
- Division of Cancer Genetics and Prevention, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Sophie R Hyman
- Division of Cancer Genetics and Prevention, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Samantha M Stokes
- Division of Cancer Genetics and Prevention, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Danielle Castillo
- Division of Clinical Cancer Genomics, Beckman Research Institute, City of Hope, Duarte, CA
| | - Nadine M Tung
- Harvard Medical School, Boston, MA
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Boston, MA
| | | | - Huma Q Rana
- Division of Cancer Genetics and Prevention, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Judy E Garber
- Division of Cancer Genetics and Prevention, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
490
|
Lai E, Ziranu P, Spanu D, Dubois M, Pretta A, Tolu S, Camera S, Liscia N, Mariani S, Persano M, Migliari M, Donisi C, Demurtas L, Pusceddu V, Puzzoni M, Scartozzi M. BRCA-mutant pancreatic ductal adenocarcinoma. Br J Cancer 2021; 125:1321-1332. [PMID: 34262146 PMCID: PMC8575931 DOI: 10.1038/s41416-021-01469-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 05/28/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
Despite continued research, pancreatic ductal adenocarcinoma (PDAC) remains one of the main causes of cancer death. Interest is growing in the role of the tumour suppressors breast cancer 1 (BRCA1) and BRCA2-typically associated with breast and ovarian cancer-in the pathogenesis of PDAC. Indeed, both germline and sporadic mutations in BRCA1/2 have been found to play a role in the development of PDAC. However, data regarding BRCA1/2-mutant PDAC are lacking. In this review, we aim to outline the specific landscape of BRCA-mutant PDAC, focusing on heritability, clinical features, differences between BRCA1 and 2 mutations and between germline and sporadic alterations, as well as established therapeutic strategies and those that are still under evaluation.
Collapse
Affiliation(s)
- Eleonora Lai
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | - Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | - Dario Spanu
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | - Marco Dubois
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
- Medical Oncology Unit, Sapienza University of Rome, Rome, Italy
- Department of Medical Oncology, Institut Jules Bordet-Université Libre de Bruxelles (ULB), Brussells, Belgium
| | - Simona Tolu
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
- Medical Oncology Unit, Sapienza University of Rome, Rome, Italy
| | - Silvia Camera
- Department of Medical Oncology, San Raffaele Scientific Institute, Milan, Italy
| | - Nicole Liscia
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
- Medical Oncology Unit, Sapienza University of Rome, Rome, Italy
| | - Stefano Mariani
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | - Mara Persano
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | - Marco Migliari
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | - Clelia Donisi
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | - Laura Demurtas
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | - Valeria Pusceddu
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | - Marco Puzzoni
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy.
| |
Collapse
|
491
|
Furniss CS, Yurgelun MB, Ukaegbu C, Constantinou PE, Lafferty CC, Talcove-Berko ER, Schwartz AN, Stopfer JE, Underhill-Blazey M, Kenner B, Nelson SH, Okumura S, Law S, Zhou AY, Coffin TB, Rodriguez NJ, Uno H, Ocean AJ, McAllister F, Lowy AM, Lippman SM, Klein AP, Madlensky L, Petersen GM, Garber JE, Goggins MG, Maitra A, Syngal S. Novel Models of Genetic Education and Testing for Pancreatic Cancer Interception: Preliminary Results from the GENERATE Study. Cancer Prev Res (Phila) 2021; 14:1021-1032. [PMID: 34625409 PMCID: PMC8563400 DOI: 10.1158/1940-6207.capr-20-0642] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/20/2021] [Accepted: 07/09/2021] [Indexed: 12/13/2022]
Abstract
Up to 10% of patients with pancreatic ductal adenocarcinoma (PDAC) carry underlying germline pathogenic variants in cancer susceptibility genes. The GENetic Education Risk Assessment and TEsting (GENERATE) study aimed to evaluate novel methods of genetic education and testing in relatives of patients with PDAC. Eligible individuals had a family history of PDAC and a relative with a germline pathogenic variant in APC, ATM, BRCA1, BRCA2, CDKN2A, EPCAM, MLH1, MSH2, MSH6, PALB2, PMS2, STK11, or TP53 genes. Participants were recruited at six academic cancer centers and through social media campaigns and patient advocacy efforts. Enrollment occurred via the study website (https://GENERATEstudy.org) and all participation, including collecting a saliva sample for genetic testing, could be done from home. Participants were randomized to one of two remote methods that delivered genetic education about the risks of inherited PDAC and strategies for surveillance. The primary outcome of the study was uptake of genetic testing. From 5/8/2019 to 5/6/2020, 49 participants were randomized to each of the intervention arms. Overall, 90 of 98 (92%) of randomized participants completed genetic testing. The most frequently detected pathogenic variants included those in BRCA2 (N = 15, 17%), ATM (N = 11, 12%), and CDKN2A (N = 4, 4%). Participation in the study remained steady throughout the onset of the Coronavirus disease (COVID-19) pandemic. Preliminary data from the GENERATE study indicate success of remote alternatives to traditional cascade testing, with genetic testing rates over 90% and a high rate of identification of germline pathogenic variant carriers who would be ideal candidates for PDAC interception approaches. PREVENTION RELEVANCE: Preliminary data from the GENERATE study indicate success of remote alternatives for pancreatic cancer genetic testing and education, with genetic testing uptake rates over 90% and a high rate of identification of germline pathogenic variant carriers who would be ideal candidates for pancreatic cancer interception.
Collapse
Affiliation(s)
- C Sloane Furniss
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Matthew B Yurgelun
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Pamela E Constantinou
- Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | | | | | - Scott H Nelson
- Pancreatic Cancer Action Network Volunteer, Patient Advocate, and Pancreatic Cancer Survivor, St. Anthony, Minnesota
| | | | | | | | | | - Nicolette J Rodriguez
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Brigham and Women's Hospital, Boston, Massachusetts
| | - Hajime Uno
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | | | - Florencia McAllister
- Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Andrew M Lowy
- Moores Cancer Center, UC San Diego, San Diego, California
| | | | - Alison P Klein
- Johns Hopkins University, Sol Goldman Pancreatic Cancer Research Center, Baltimore, Maryland
| | - Lisa Madlensky
- Moores Cancer Center, UC San Diego, San Diego, California
| | | | - Judy E Garber
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Brigham and Women's Hospital, Boston, Massachusetts
| | - Michael G Goggins
- Johns Hopkins University, Sol Goldman Pancreatic Cancer Research Center, Baltimore, Maryland
| | - Anirban Maitra
- Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Sapna Syngal
- Dana-Farber Cancer Institute, Boston, Massachusetts.
- Harvard Medical School, Boston, Massachusetts
- Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
492
|
Marques CAV, Figueiredo END, Gutiérrez MGRD. Breast cancer screening program for risk groups: facts and perspectives. Rev Bras Enferm 2021; 75:e20210050. [PMID: 34669830 DOI: 10.1590/0034-7167-2021-0050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/22/2021] [Indexed: 12/09/2022] Open
Abstract
OBJECTIVES to measure the frequency and compliance of breast cancer screening, according to the risk for this disease. METHODS a cross-sectional study with 950 female users of 38 public Primary Health Care services in São Paulo, between October and December 2013. According to UHS criteria, participants were grouped into high risk and standard risk, and frequency, association (p≤0.05), and screening compliance were measured. RESULTS 6.7% had high risk and 93.3% standard risk, respectively; in these groups, the frequency and compliance of clinical breast examination were 40.3% and 37.1%, and 43.5% and 43.0% (frequency p=0.631, compliance p=0.290). Mammograms were 67.7% and 35.5% for participants at high risk, and 57.4% and 25.4% for those at standard risk (frequency p=0.090, compliance p=0.000). CONCLUSIONS in the groups, attendance and conformity of the clinical breast exam were similar; for mammography, it was higher in those at high risk, with assertiveness lower than the 70% set in UHS.
Collapse
|
493
|
Agaoglu NB, Doganay L. Concurrent pathogenic variations in patients with hereditary cancer syndromes. Eur J Med Genet 2021; 64:104366. [PMID: 34637943 DOI: 10.1016/j.ejmg.2021.104366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/26/2021] [Accepted: 10/08/2021] [Indexed: 01/13/2023]
Abstract
Cancer is a multifactorial disorder; however, 5-10% of all cancers show hereditary background. In recent years many targeted next generation sequencing panels comprising cancer predisposition genes have been developed and used for diagnostic purposes in patients with increased cancer risk. Screening multiple genes at a time allows multiple variants in different genes to be detected as well. This study aims to determine the cases with concurrent mutations in different hereditary cancer predisposition genes and how they are clinically affected. Here, we screened 1090 index cases by next generation sequencing based hereditary cancer panels and evaluated the reflection of multiple variations on the phenotype. We detected 11 (1%) cases with pathogenic variants in more than one gene. These concurrent variations occurred mostly in BRCA1/2 (7/11) accompanied with MUTYH, ATM, CHECK2, NBN, and RAD50. In addition, MUTYH&ATM, NBN&MSH6, MUTYH&CHEK2 double heterozygous cases were detected. Moreover, we identified a case with three heterozygous variations in CDH1, MUTYH, and CHEK2. These patients presented malignancies that were mostly related to pathogenic variations they carried. Although they are rare, defining double heterozygous cases is important for managing appropriate therapy and accurate genetic consulting for the patients and family members.
Collapse
Affiliation(s)
- Nihat Bugra Agaoglu
- Genomic Laboratory (GLAB), Umraniye Training and Research Hospital, University of Health Sciences, Istanbul, Turkey; Department of Medical Genetics, Umraniye Training and Research Hospital, University of Health Sciences, Istanbul, Turkey.
| | - Levent Doganay
- Genomic Laboratory (GLAB), Umraniye Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
494
|
Uson PLS, Samadder NJ, Riegert-Johnson D, Boardman L, Borad MJ, Ahn D, Sonbol MB, Faigel DO, Fukami N, Pannala R, Kunze K, Golafshar M, Klint M, Esplin ED, Nussbaum RL, Stewart AK, Bekaii-Saab T. Clinical Impact of Pathogenic Germline Variants in Pancreatic Cancer: Results From a Multicenter, Prospective, Universal Genetic Testing Study. Clin Transl Gastroenterol 2021; 12:e00414. [PMID: 34620795 PMCID: PMC8500569 DOI: 10.14309/ctg.0000000000000414] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION To report the prevalence and outcomes of unselected pancreatic cancer (PC) patients with pathogenic/likely pathogenic germline variants (PGVs) detected using a universal testing approach. METHODS We undertook a prospective, multisite study of germline sequencing using a >80 gene next-generation sequencing platform among 250 patients with PC (not selected for age or family history of cancer) between April 1, 2018, and March 31, 2020. Demographic, tumor characteristics, and clinical outcomes were compared between PGV carriers and noncarriers. RESULTS Of 250 patients, the mean age was 65 years (SD 8.7), 56% was male, 83.6% was White, and 65.6% had advanced disease (stages III and IV). PGVs were found in 15.2% (N = 38) of patients, and 2 patients had more than 1 PGV. Variants of uncertain significance were found in 44.4% (N = 111). Family history of cancer (odds ratio: 2.36, 95% confidence interval: 1.14-5.19, P = 0.025) was associated with a higher risk of PGV. In a median follow-up of 16.5 months, the median overall survival was 16.8 months in PGV carriers compared with 16.5 months in noncarriers (hazard ratio: 0.51, 95% confidence interval: 0.25-1.01, P = 0.05). Higher levels of carbohydrate antigen 19-9 and advanced disease stages (III and IV) were associated with worse outcomes in both groups. Overall, 68% of PGV carriers had mutations in homologous recombination repair genes, including BRCA1, BRCA2, PALB2, ATM, CHEK2, NBN, and RAD51C. DISCUSSION Universal multigene panel testing in PC reveals that 1 in 6 patients are carriers of PGV. Multigene germline testing should be used to aid in treatment selection, prognostication, and familial cancer counseling.
Collapse
Affiliation(s)
- Pedro L. S. Uson
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - N. Jewel Samadder
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Douglas Riegert-Johnson
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Lisa Boardman
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Mitesh J. Borad
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Daniel Ahn
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Mohamad B. Sonbol
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Douglas O. Faigel
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Norio Fukami
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Rahul Pannala
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Katie Kunze
- Department of Biostatistics, Mayo Clinic, Scottsdale, Arizona, USA
| | | | - Margaret Klint
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - A. Keith Stewart
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Tanios Bekaii-Saab
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| |
Collapse
|
495
|
Hur YM, Mun J, Kim MK, Lee M, Kim YH, Kim SC. Disparities between Uptake of Germline BRCA1/ 2 Gene Tests and Implementation of Post-test Management Strategies in Epithelial Ovarian, Fallopian Tube, or Primary Peritoneal Cancer Patients. J Korean Med Sci 2021; 36:e241. [PMID: 34609091 PMCID: PMC8490789 DOI: 10.3346/jkms.2021.36.e241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/08/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND To assess the rate of germline BRCA gene tests in epithelial ovarian cancer (EOC) patients and uptake of post-test risk management strategies in BRCA1/2-mutated patients. METHODS Institutional databases were searched to identify patients who were diagnosed with epithelial ovarian, fallopian tube, or primary peritoneal cancer (EOC) between 2009 and 2019 in two academic hospitals. Retrospective review on medical records was performed to collect clinico-pathologic variables, including performance of germline BRCA gene test and its results, as well as conduct of breast cancer screening tests and cascade testing. If annual mammography +/- breast ultrasonography was performed, it was considered that regular breast cancer surveillance was done. RESULTS A total of 840 women with EOC were identified during the study period. Of these, 454 patients (54.0%) received BRCA gene testing and 106 patients (106/454, 23.3%) were positive for BRCA1/2 mutations. The rate of BRCA tests has markedly increased from 25.8% in 2009-2012 to 62.7% in 2017-2019. Among the 93 patients with BRCA1/2 mutation without previous personal breast cancer history, 20 patients (21.5%) received annual mammography with or without breast ultrasonography for regular surveillance. Among the 106 BRCA1/2-mutated EOC patients, cascade testing on family members was performed only in 13 patients (12.3%). CONCLUSION Although BRCA1/2 gene tests have been substantially expanded, the uptake of post-test risk management strategies, including breast cancer screening for BRCA1/2-mutated patients and cascade testing for family members, has remained low. Strategies to increase its uptake and education about the importance of post-test risk managements are needed.
Collapse
Affiliation(s)
- Young Min Hur
- Department of Obstetrics and Gynecology, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul, Korea
| | - Jaehee Mun
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Mi-Kyung Kim
- Department of Obstetrics and Gynecology, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul, Korea.
| | - Maria Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Yun Hwan Kim
- Department of Obstetrics and Gynecology, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul, Korea
| | - Seung-Cheol Kim
- Department of Obstetrics and Gynecology, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul, Korea
| |
Collapse
|
496
|
Chiang YC, Lin PH, Lu TP, Kuo KT, Tai YJ, Hsu HC, Wu CY, Lee CY, Shen H, Chen CA, Cheng WF. A DNA Damage Response Gene Panel for Different Histologic Types of Epithelial Ovarian Carcinomas and Their Outcomes. Biomedicines 2021; 9:biomedicines9101384. [PMID: 34680501 PMCID: PMC8533221 DOI: 10.3390/biomedicines9101384] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/20/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022] Open
Abstract
DNA damage response (DDR) is important for maintaining genomic integrity of the cell. Aberrant DDR pathways lead to accumulation of DNA damage, genomic instability and malignant transformations. Gene mutations have been proven to be associated with epithelial ovarian cancer, and the majority of the literature has focused on BRCA. In this study, we investigated the somatic mutation of DNA damage response genes in epithelial ovarian cancer patients using a multiple-gene panel with next-generation sequencing. In all, 69 serous, 39 endometrioid and 64 clear cell carcinoma patients were enrolled. Serous carcinoma patients (69.6%) had higher percentages of DDR gene mutations compared with patients with endometrioid (33.3%) and clear cell carcinoma (26.6%) (p < 0.001, chi-squared test). The percentages of DDR gene mutations in patients with recurrence (53.9 vs. 32.9% p = 0.006, chi-squared test) or cancer-related death (59.2 vs. 34.4% p = 0.001, chi-squared test) were higher than those without recurrence or living patients. In endometrioid carcinoma, patients with ≥2 DDR gene mutations had shorter PFS (p = 0.0035, log-rank test) and OS (p = 0.015, log-rank test) than those with one mutation or none. In clear cell carcinoma, patients with ≥2 DDR gene mutations had significantly shorter PFS (p = 0.0056, log-rank test) and OS (p = 0.0046, log-rank test) than those with 1 DDR mutation or none. In the EOC patients, somatic DDR gene mutations were associated with advanced-stage tumor recurrence and tumor-related death. Type I EOC patients with DDR mutations had an unfavorable prognosis, especially for clear cell carcinoma.
Collapse
Affiliation(s)
- Ying-Cheng Chiang
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei 100226, Taiwan;
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 100226, Taiwan; (Y.-J.T.); (C.-Y.W.)
| | - Po-Han Lin
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 100226, Taiwan;
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100025, Taiwan
| | - Tzu-Pin Lu
- Institute of Epidemiology and Preventive Medicine, Department of Public Health, National Taiwan University, Taipei 100025, Taiwan;
| | - Kuan-Ting Kuo
- Department of Pathology, College of Medicine, National Taiwan University, Taipei 100225, Taiwan;
| | - Yi-Jou Tai
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 100226, Taiwan; (Y.-J.T.); (C.-Y.W.)
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100225, Taiwan; (H.-C.H.); (C.-Y.L.); (H.S.)
| | - Heng-Cheng Hsu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100225, Taiwan; (H.-C.H.); (C.-Y.L.); (H.S.)
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu 30059, Taiwan
| | - Chia-Ying Wu
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 100226, Taiwan; (Y.-J.T.); (C.-Y.W.)
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100225, Taiwan; (H.-C.H.); (C.-Y.L.); (H.S.)
| | - Chia-Yi Lee
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100225, Taiwan; (H.-C.H.); (C.-Y.L.); (H.S.)
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu 30059, Taiwan
| | - Hung Shen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100225, Taiwan; (H.-C.H.); (C.-Y.L.); (H.S.)
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu 30059, Taiwan
| | - Chi-An Chen
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei 100226, Taiwan;
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 100226, Taiwan; (Y.-J.T.); (C.-Y.W.)
- Correspondence: (C.-A.C.); (W.-F.C.); Tel.: +886-2-2312-3456 (ext. 71964) (W.-F.C.)
| | - Wen-Fang Cheng
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei 100226, Taiwan;
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 100226, Taiwan; (Y.-J.T.); (C.-Y.W.)
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100225, Taiwan; (H.-C.H.); (C.-Y.L.); (H.S.)
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei 100025, Taiwan
- Correspondence: (C.-A.C.); (W.-F.C.); Tel.: +886-2-2312-3456 (ext. 71964) (W.-F.C.)
| |
Collapse
|
497
|
Avances en Uro-Oncología «OncoUrology Forum Special Edition»: lo mejor del 2020. Actas Urol Esp 2021. [DOI: 10.1016/j.acuro.2021.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
498
|
Fasaye GA, Liu Y, Calzone K. Nurse practitioners have a vital role in achieving health equity in clinical cancer genetics. J Am Assoc Nurse Pract 2021; 33:763-765. [PMID: 35280937 PMCID: PMC8916687 DOI: 10.1097/jxx.0000000000000648] [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: 11/27/2022]
Abstract
Long-standing and persistent racial inequities exist in cancer prevention, diagnosis, treatment, and outcomes. Genetic medicine has the promise to significantly advance the identification of at-risk individuals and facilitate prevention, early detection, and treatment of cancer. Genetic testing is increasingly becoming incorporated into the screening-to-treatment continuum of care for cancer. Although genetic technologies are relatively new to the cancer care landscape, racial inequities already exist in awareness, access, referral, and uptake. Nurses play a vital role in achieving health equity, but success requires that nurses understand, recognize and take action to overcome the factors that have fostered health inequities.
Collapse
Affiliation(s)
- Grace-Ann Fasaye
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Genetics Branch
| | - Yi Liu
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Genetics Branch
| | - Kathleen Calzone
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Genetics Branch
| |
Collapse
|
499
|
Briceño-Morales X, Briceño-Morales C, Guerrero-Macías SI, Pedroza-Durán AM, Súarez-Rodríguez RA. Revisiting the indication for prophylactic contralateral mastectomy in patients with Li-Fraumeni syndrome and breast cancer. Case report. REVISTA COLOMBIANA DE OBSTETRICIA Y GINECOLOGIA 2021; 72:307-318. [PMID: 34851573 PMCID: PMC8603825 DOI: 10.18597/rcog.3690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 08/09/2021] [Indexed: 11/04/2022]
Abstract
Objective To describe the case of a patient with Li-Fraumeni syndrome (LFS) and breast cancer in whom the benefit of contralateral prophylactic mastectomy (CPM) was challenged; and to offer a critical discussion regarding the evidence supporting this procedure in this patient population. Case presentation A 37-year-old woman with breast cancer and a family history of multiple early onset cancer of the LFS spectrum in whom a pathogenic variant of the TP53 gene was confirmed during adjuvant hormonal therapy. The case was presented during the multidisciplinary meeting of the Breast Service of a referral oncology center in Colombia, in order to discuss the benefit of CPM. The decision of the board meeting was not to perform CPM. After 30 months of follow-up, the patient is disease-free. Conclusion There is no evidence on the impact of CPM on survival of patients with LFS and breast cancer in particular. However, in light of the current knowledge, it is not possible to generalize the approach of withholding this prophylactic surgery. It is important to report those cases in which the decision is made to either perform or omit this procedure in order to increase the body of evidence, considering the limitations that make it difficult to build large cohorts or conduct trials exclusively for this genetic disorder.
Collapse
|
500
|
Trend and survival benefit of contralateral prophylactic mastectomy among men with stage I-III unilateral breast cancer in the USA, 1998-2016. Breast Cancer Res Treat 2021; 190:503-515. [PMID: 34554371 DOI: 10.1007/s10549-021-06397-z] [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: 06/30/2021] [Accepted: 09/11/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Our study aimed to explore temporal trends and survival benefit of contralateral prophylactic mastectomy (CPM) in male breast cancer (MBC). METHODS Men with stage I-III unilateral breast cancer between 1998 and 2016 were identified from the surveillance, epidemiology, and end results (SEER). We compared CPM rate over the study period using the Cochrane-Armitage test for trend. Logistic regression model was used to test for factors predicting CPM. Survival analysis was conducted in patients who underwent CPM or unilateral mastectomy (UM) with a first diagnosis of unilateral breast cancer. Kaplan-Meier curve and univariate and multivariable Cox proportional hazards regression analyses were performed to compare overall survival (OS) and breast cancer-specific survival (BCSS) between CPM and UM groups. Propensity score matching was adopted to balance baseline characteristics. RESULTS 5118 MBC cases were included in the present study, with 4.1% (n = 209) patients underwent CPM. The proportion of men undergoing CPM increased from 1.7 in 1998 to 6.3% in 2016 (P < 0.0001). Young age, recent years of diagnosis, higher tumor grade and lower T stage were significantly associated with CPM. A cohort of 3566 patients were enrolled in survival analysis with a median follow-up of 65 months. CPM was associated with better OS (HR 0.58, 95% CI 0.37-0.89, P = 0.022) rather than BCSS (HR 0.57, 95% CI 0.29-1.11, P = 0.153) compared with UM. In propensity score-matched model, CPM was not an independent prognostic factor for OS (HR 0.83, 95% CI 0.46-1.52, P = 0.553) and BCSS (HR 0.98, 95% CI 0.39-2.47, P = 0.970). CONCLUSION Our study revealed a dramatic increase in CPM utilization among MBC, especially in young patients. However, CPM provides no survival benefit for MBC compared with UM, indicating the decision of CPM should be fully discussed.
Collapse
|