51
|
Smith AJB, O'Brien C, Haggerty A, Ko EM, Rendle KA. "Having cancer is very expensive": A qualitative study of patients with ovarian cancer and PARP inhibitor treatment. Gynecol Oncol 2024; 186:170-175. [PMID: 38691987 DOI: 10.1016/j.ygyno.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/26/2024] [Accepted: 04/21/2024] [Indexed: 05/03/2024]
Abstract
OBJECTIVE To examine patient barriers and facilitators to PARP inhibitor (PARP-I) maintenance therapy in ovarian cancer. PARP-I improves survival in ovarian cancer, but these multi-year therapies cost around $100,000 annually and are under-prescribed. METHODS We recruited patients with ovarian cancer treated with PARP-I maintenance therapy at an academic health system for a semi-structured interview. Patient demographics, including genetics and PARP-I cost, were self-reported. We assessed patient experiences with barriers and facilitators of PARP-I usage. Two team members used a thematic approach to analyze and identify key themes. RESULTS In May 2022, we interviewed 10 patients (mean age = 65 years; 80% White; 60% with a germline genetic mutation). Patients paid on average $227.50 monthly for PARP-I, straining resources for some participants. While sampled patients were insured, all patients identified having no or inadequate insurance as a major barrier to PARP-I. At the same time, all participants prioritized clinical effectiveness over costs of care. Patients identified PARP-I delivery from specialty pharmacies, separate and different from other medications, as a potential barrier, but each had been able to navigate delivery. Patients expressed significant initial side effects of PARP-I as a potential barrier yet reported clinician communication and prompt dose reduction as facilitating continuation. CONCLUSIONS Patients identified cost, restrictive pharmacy benefits, and initial side effects as barriers to PARP-I usage. Having insurance and a supportive care team were identified as facilitators. Enhancing communication about PARP-I cost and side effects could improve patient experience and receipt of evidence-based maintenance therapy in ovarian cancer.
Collapse
Affiliation(s)
- Anna Jo Bodurtha Smith
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Pennsylvania Health Systems, Philadelphia, PA, United States of America; Department of Obstetrics and Gynecology, University of Pennsylvania Health Systems, Philadelphia, PA, United States of America; Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA, United States of America; Penn Center for Cancer Care Innovation, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States of America; Center for Health Incentives and Behavioral Economics, University of Pennsylvania, Philadelphia, PA, United States of America.
| | - Caroline O'Brien
- Department of Family Medicine and Community Health, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Ashley Haggerty
- Division of Gynecologic Oncology, Hackensack Meridian Health, Red Bank, NJ, United States of America
| | - Emily M Ko
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Pennsylvania Health Systems, Philadelphia, PA, United States of America; Department of Obstetrics and Gynecology, University of Pennsylvania Health Systems, Philadelphia, PA, United States of America; Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA, United States of America; Penn Center for Cancer Care Innovation, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Katharine A Rendle
- Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA, United States of America; Penn Center for Cancer Care Innovation, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States of America; Department of Family Medicine and Community Health, University of Pennsylvania, Philadelphia, PA, United States of America
| |
Collapse
|
52
|
Kwinten KJJ, Lemain VA, de Hullu JA, Leenders WPJ, Steenbeek MP, van Altena AM, Pijnenborg JMA. Cervicovaginal specimen biomarkers for early detection of ovarian and endometrial cancer: A review. Cancer Med 2024; 13:e70000. [PMID: 39031958 PMCID: PMC11259558 DOI: 10.1002/cam4.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/17/2024] [Accepted: 06/30/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND In the last decade, technical innovations have resulted in the development of several minimally invasive diagnostic cancer tools. Within women at high risk of developing ovarian or endometrial cancer (EC) due to hereditary cancer syndrome, there is an urgent need for minimally invasive and patient-friendly methods to detect ovarian cancer and EC at an early stage. MATERIALS AND METHODS We performed a systematic search of studies using DNA methylation or mutation analysis, microbiome, or proteomics performed on cervicovaginal specimens (smear, swab, or tampon) intended to detect ovarian and EC published until January 2024. RESULTS Included studies (n = 36) showed high heterogeneity in terms of biomarkers used and outcomes, and only a few studies reported on the detection of biomarkers in high-risk subgroups. CONCLUSION Based on the findings in this review, DNA methylation techniques seem to be the most promising for detecting ovarian and EC at early stages in the general population. Future validation of cervicovaginal DNA methylation techniques is needed to determine whether this technique might be beneficial in hereditary high-risk subgroups.
Collapse
Affiliation(s)
- Kevin J. J. Kwinten
- Department of Obstetrics and GynecologyRadboud University Medical CenterNijmegenThe Netherlands
- Department of Obstetrics and GynecologyCanisius Wilhelmina HospitalNijmegenThe Netherlands
| | - Victor A. Lemain
- Department of Obstetrics and GynecologyRadboud University Medical CenterNijmegenThe Netherlands
| | - Joanne A. de Hullu
- Department of Obstetrics and GynecologyRadboud University Medical CenterNijmegenThe Netherlands
| | | | - Miranda P. Steenbeek
- Department of Obstetrics and GynecologyRadboud University Medical CenterNijmegenThe Netherlands
- Department of Obstetrics and GynecologyCatharina Hospital EindhovenEindhovenThe Netherlands
| | - Anne M. van Altena
- Department of Obstetrics and GynecologyRadboud University Medical CenterNijmegenThe Netherlands
| | | |
Collapse
|
53
|
Patel MM, Adrada BE. Hereditary Breast Cancer: BRCA Mutations and Beyond. Radiol Clin North Am 2024; 62:627-642. [PMID: 38777539 DOI: 10.1016/j.rcl.2023.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Hereditary breast cancers are manifested by pathogenic and likely pathogenic genetic mutations. Penetrance expresses the breast cancer risk associated with these genetic mutations. Although BRCA1/2 are the most widely known genetic mutations associated with breast cancer, numerous additional genes demonstrate high and moderate penetrance for breast cancer. This review describes current genetic testing, details the specific high and moderate penetrance genes for breast cancer and reviews the current approach to screening for breast cancer in patients with these genetic mutations.
Collapse
Affiliation(s)
- Miral M Patel
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, CPB5.3208, Houston, TX 77030, USA.
| | - Beatriz Elena Adrada
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe, CPB5.3208, Houston, TX 77030, USA
| |
Collapse
|
54
|
Samadder NJ, Gay E, Lindpere V, Bublitz ML, Bandel LA, Armasu SM, Vierkant RA, Ferber MJ, Klee EW, Larson NB, Kruisselbrink TM, Egan JB, Kemppainen JL, Bidwell JS, Anderson JL, McAllister TM, Walker TS, Kunze KL, Golafshar MA, Klint MA, Presutti RJ, Bobo WV, Sekulic A, Summer-Bolster JM, Willman CL, Lazaridis KN. Exome Sequencing Identifies Carriers of the Autosomal Dominant Cancer Predisposition Disorders Beyond Current Practice Guideline Recommendations. JCO Precis Oncol 2024; 8:e2400106. [PMID: 39013133 DOI: 10.1200/po.24.00106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/28/2024] [Accepted: 05/17/2024] [Indexed: 07/18/2024] Open
Abstract
PURPOSE The autosomal dominant cancer predisposition disorders hereditary breast and ovarian cancer (HBOC) and Lynch syndrome (LS) are genetic conditions for which early identification and intervention have a positive effect on the individual and public health. The goals of this study were to determine whether germline genetic screening using exome sequencing could be used to efficiently identify carriers of HBOC and LS. METHODS Participants were recruited from three geographically and racially diverse sites in the United States (Rochester, MN; Phoenix, AZ; Jacksonville, FL). Participants underwent Exome+ sequencing (Helix Inc, San Mateo, CA) and return of results for specific genetic findings: HBOC (BRCA1 and BRCA1) and LS (MLH1, MSH2, MSH6, PMS2, and EPCAM). Chart review was performed to collect demographics and personal and family cancer history. RESULTS To date, 44,306 participants have enrolled in Tapestry. Annotation and interpretation of all variants in genes for HBOC and LS resulted in the identification of 550 carriers (prevalence, 1.24%), which included 387 with HBOC (27.2% BRCA1, 42.8% BRCA2) and 163 with LS (12.3% MSH6, 8.8% PMS2, 4.5% MLH1, 3.8% MSH2, and 0.2% EPCAM). More than half of these participants (52.1%) were newly diagnosed carriers with HBOC and LS. In all, 39.2% of HBOC/LS carriers did not satisfy National Comprehensive Cancer Network (NCCN) criteria for genetic evaluation. NCCN criteria were less commonly met in underrepresented minority populations versus self-reported White race (51.5% v 37.5%, P = .028). CONCLUSION Our results emphasize the need for wider utilization of germline genetic sequencing for enhanced screening and detection of individuals who have LS and HBOC cancer predisposition syndromes.
Collapse
Affiliation(s)
- N Jewel Samadder
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Phoenix, AZ
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Comprehensive Cancer Center, Mayo Clinic, Rochester, MN
| | - Emily Gay
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
- Genetic Counseling Program, Arizona State University, Tucson, AZ
| | - Vanda Lindpere
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ
| | | | | | | | - Robert A Vierkant
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Matthew J Ferber
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Eric W Klee
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | | | - Jan B Egan
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | - Jessa S Bidwell
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | | | - T'Nita S Walker
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Katie L Kunze
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ
| | | | | | - Richard J Presutti
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Family Medicine, Mayo Clinic, Jacksonville, FL
| | - William V Bobo
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Psychiatry & Psychology, Mayo Clinic, Jacksonville, FL
| | - Aleksander Sekulic
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Dermatology, Mayo Clinic, Scottsdale, AZ
| | | | - Cheryl L Willman
- Comprehensive Cancer Center, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Konstantinos N Lazaridis
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Comprehensive Cancer Center, Mayo Clinic, Rochester, MN
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
55
|
Vellan CJ, Islam T, De Silva S, Mohd Taib NA, Prasanna G, Jayapalan JJ. Exploring novel protein-based biomarkers for advancing breast cancer diagnosis: A review. Clin Biochem 2024; 129:110776. [PMID: 38823558 DOI: 10.1016/j.clinbiochem.2024.110776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/26/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
This review provides a contemporary examination of the evolving landscape of breast cancer (BC) diagnosis, focusing on the pivotal role of novel protein-based biomarkers. The overview begins by elucidating the multifaceted nature of BC, exploring its prevalence, subtypes, and clinical complexities. A critical emphasis is placed on the transformative impact of proteomics, dissecting the proteome to unravel the molecular intricacies of BC. Navigating through various sources of samples crucial for biomarker investigations, the review underscores the significance of robust sample processing methods and their validation in ensuring reliable outcomes. The central theme of the review revolves around the identification and evaluation of novel protein-based biomarkers. Cutting-edge discoveries are summarised, shedding light on emerging biomarkers poised for clinical application. Nevertheless, the review candidly addresses the challenges inherent in biomarker discovery, including issues of standardisation, reproducibility, and the complex heterogeneity of BC. The future direction section envisions innovative strategies and technologies to overcome existing challenges. In conclusion, the review summarises the current state of BC biomarker research, offering insights into the intricacies of proteomic investigations. As precision medicine gains momentum, the integration of novel protein-based biomarkers emerges as a promising avenue for enhancing the accuracy and efficacy of BC diagnosis. This review serves as a compass for researchers and clinicians navigating the evolving landscape of BC biomarker discovery, guiding them toward transformative advancements in diagnostic precision and personalised patient care.
Collapse
Affiliation(s)
- Christina Jane Vellan
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Tania Islam
- Department of Surgery, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Sumadee De Silva
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Sri Lanka
| | - Nur Aishah Mohd Taib
- Department of Surgery, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Galhena Prasanna
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Sri Lanka
| | - Jaime Jacqueline Jayapalan
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia; Universiti Malaya Centre for Proteomics Research (UMCPR), Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
56
|
Austin JD, James E, Perez RL, Mazza GL, Kling JM, Fraker J, Mina L, Banerjee I, Sharpe R, Patel BK. Factors influencing U.S. women's interest and preferences for breast cancer risk communication: a cross-sectional study from a large tertiary care breast imaging center. BMC Womens Health 2024; 24:359. [PMID: 38907193 PMCID: PMC11191185 DOI: 10.1186/s12905-024-03197-7] [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] [Received: 04/17/2024] [Accepted: 06/10/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Breast imaging clinics in the United States (U.S.) are increasingly implementing breast cancer risk assessment (BCRA) to align with evolving guideline recommendations but with limited uptake of risk-reduction care. Effectively communicating risk information to women is central to implementation efforts, but remains understudied in the U.S. This study aims to characterize, and identify factors associated with women's interest in and preferences for breast cancer risk communication. METHODS This is a cross-sectional survey study of U.S. women presenting for a mammogram between January and March of 2021 at a large, tertiary breast imaging clinic. Survey items assessed women's interest in knowing their risk and preferences for risk communication if considered to be at high risk in hypothetical situations. Multivariable logistic regression modeling assessed factors associated with women's interest in knowing their personal risk and preferences for details around exact risk estimates. RESULTS Among 1119 women, 72.7% were interested in knowing their breast cancer risk. If at high risk, 77% preferred to receive their exact risk estimate and preferred verbal (52.9% phone/47% in-person) vs. written (26.5% online/19.5% letter) communications. Adjusted regression analyses found that those with a primary family history of breast cancer were significantly more interested in knowing their risk (OR 1.5, 95% CI 1.0, 2.1, p = 0.04), while those categorized as "more than one race or other" were significantly less interested in knowing their risk (OR 0.4, 95% CI 0.2, 0.9, p = 0.02). Women 60 + years of age were significantly less likely to prefer exact estimates of their risk (OR 0.6, 95% CI 0.5, 0.98, p < 0.01), while women with greater than a high school education were significantly more likely to prefer exact risk estimates (OR 2.5, 95% CI 1.5, 4.2, p < 0.001). CONCLUSION U.S. women in this study expressed strong interest in knowing their risk and preferred to receive exact risk estimates verbally if found to be at high risk. Sociodemographic and family history influenced women's interest and preferences for risk communication. Breast imaging centers implementing risk assessment should consider strategies tailored to women's preferences to increase interest in risk estimates and improve risk communication.
Collapse
Affiliation(s)
- Jessica D Austin
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, 13400 E. Shea Blvd, Scottsdale, AZ, 85259, USA.
| | - Emily James
- Mayo Clinic College of Medicine of Medicine and Science, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Rachel L Perez
- Mayo Clinic College of Medicine of Medicine and Science, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Gina L Mazza
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, 13400 E. Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Juliana M Kling
- Women's Health Internal Medicine, Department of Internal Medicine, Mayo Clinic, 13400 E. Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Jessica Fraker
- Women's Health Internal Medicine, Department of Internal Medicine, Mayo Clinic, 13400 E. Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Lida Mina
- Department of Internal Medicine, Division of Medical Oncology, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Imon Banerjee
- Department of Diagnostic Radiology, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Richard Sharpe
- Department of Diagnostic Radiology, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ, 85054, USA
| | - Bhavika K Patel
- Department of Diagnostic Radiology, Mayo Clinic, 5777 E Mayo Blvd, Phoenix, AZ, 85054, USA
| |
Collapse
|
57
|
Dominguez-Ortiz J, Álvarez-Gómez RM, Montiel-Manríquez R, Cedro-Tanda A, Alcaraz N, Castro-Hernández C, Bautista-Hinojosa L, Contreras-Espinosa L, Torres-Maldonado L, Fragoso-Ontiveros V, Sánchez-Contreras Y, González-Barrios R, la Fuente-Hernández MAD, Mejía-Aguayo MDLL, Juárez-Figueroa U, Padua-Bracho A, Sosa-León R, Obregon-Serrano G, Vidal-Millán S, Núñez-Martínez PM, Pedroza-Torres A, Nicasio-Arzeta S, Rodríguez A, Luna F, Cisneros-Soberanis F, Frías S, Arriaga-Canon C, Herrera-Montalvo LA. A Molecular Characterization of the Allelic Expression of the BRCA1 Founder Δ9-12 Pathogenic Variant and Its Potential Clinical Relevance in Hereditary Cancer. Int J Mol Sci 2024; 25:6773. [PMID: 38928478 PMCID: PMC11204022 DOI: 10.3390/ijms25126773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Hereditary breast and ovarian cancer (HBOC) syndrome is a genetic condition that increases the risk of breast cancer by 80% and that of ovarian cancer by 40%. The most common pathogenic variants (PVs) causing HBOC occur in the BRCA1 gene, with more than 3850 reported mutations in the gene sequence. The prevalence of specific PVs in BRCA1 has increased across populations due to the effect of founder mutations. Therefore, when a founder mutation is identified, it becomes key to improving cancer risk characterization and effective screening protocols. The only founder mutation described in the Mexican population is the deletion of exons 9 to 12 of BRCA1 (BRCA1Δ9-12), and its description focuses on the gene sequence, but no transcription profiles have been generated for individuals who carry this gene. In this study, we describe the transcription profiles of cancer patients and healthy individuals who were heterozygous for PV BRCA1Δ9-12 by analyzing the differential expression of both alleles compared with the homozygous BRCA1 control group using RT-qPCR, and we describe the isoforms produced by the BRCA1 wild-type and BRCA1Δ9-12 alleles using nanopore long-sequencing. Using the Kruskal-Wallis test, our results showed a similar transcript expression of the wild-type allele between the healthy heterozygous group and the homozygous BRCA1 control group. An association between the recurrence and increased expression of both alleles in HBOC patients was also observed. An analysis of the sequences indicated four wild-type isoforms with diagnostic potential for discerning individuals who carry the PV BRCA1Δ9-12 and identifying which of them has developed cancer.
Collapse
Affiliation(s)
- Julieta Dominguez-Ortiz
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (J.D.-O.); (R.M.-M.); (C.C.-H.); (L.B.-H.); (L.C.-E.); (R.G.-B.); (F.L.)
- Instituto Nacional de Cancerología, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Mexico City 04510, Mexico
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Av. San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (R.M.Á.-G.); (V.F.-O.); (Y.S.-C.); (M.A.D.l.F.-H.); (M.d.l.L.M.-A.); (A.P.-B.); (R.S.-L.); (G.O.-S.); (S.V.-M.); (P.M.N.-M.); (A.P.-T.)
| | - Rosa M. Álvarez-Gómez
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Av. San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (R.M.Á.-G.); (V.F.-O.); (Y.S.-C.); (M.A.D.l.F.-H.); (M.d.l.L.M.-A.); (A.P.-B.); (R.S.-L.); (G.O.-S.); (S.V.-M.); (P.M.N.-M.); (A.P.-T.)
| | - Rogelio Montiel-Manríquez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (J.D.-O.); (R.M.-M.); (C.C.-H.); (L.B.-H.); (L.C.-E.); (R.G.-B.); (F.L.)
| | - Alberto Cedro-Tanda
- Núcleo B de Innovación en Medicina de Precisión, Instituto Nacional de Medicina Genómica, Periférico Sur 4809, Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico;
| | - Nicolás Alcaraz
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3A, 2200 Copenhagen, Denmark;
| | - Clementina Castro-Hernández
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (J.D.-O.); (R.M.-M.); (C.C.-H.); (L.B.-H.); (L.C.-E.); (R.G.-B.); (F.L.)
| | - Luis Bautista-Hinojosa
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (J.D.-O.); (R.M.-M.); (C.C.-H.); (L.B.-H.); (L.C.-E.); (R.G.-B.); (F.L.)
| | - Laura Contreras-Espinosa
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (J.D.-O.); (R.M.-M.); (C.C.-H.); (L.B.-H.); (L.C.-E.); (R.G.-B.); (F.L.)
| | - Leda Torres-Maldonado
- Instituto Nacional de Pediatría, Insurgentes Sur No. 3700-C. Coyoacán, Mexico City 04530, Mexico; (L.T.-M.); (U.J.-F.); (A.R.); (S.F.)
| | - Verónica Fragoso-Ontiveros
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Av. San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (R.M.Á.-G.); (V.F.-O.); (Y.S.-C.); (M.A.D.l.F.-H.); (M.d.l.L.M.-A.); (A.P.-B.); (R.S.-L.); (G.O.-S.); (S.V.-M.); (P.M.N.-M.); (A.P.-T.)
| | - Yuliana Sánchez-Contreras
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Av. San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (R.M.Á.-G.); (V.F.-O.); (Y.S.-C.); (M.A.D.l.F.-H.); (M.d.l.L.M.-A.); (A.P.-B.); (R.S.-L.); (G.O.-S.); (S.V.-M.); (P.M.N.-M.); (A.P.-T.)
| | - Rodrigo González-Barrios
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (J.D.-O.); (R.M.-M.); (C.C.-H.); (L.B.-H.); (L.C.-E.); (R.G.-B.); (F.L.)
| | - Marcela Angélica De la Fuente-Hernández
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Av. San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (R.M.Á.-G.); (V.F.-O.); (Y.S.-C.); (M.A.D.l.F.-H.); (M.d.l.L.M.-A.); (A.P.-B.); (R.S.-L.); (G.O.-S.); (S.V.-M.); (P.M.N.-M.); (A.P.-T.)
| | - María de la Luz Mejía-Aguayo
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Av. San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (R.M.Á.-G.); (V.F.-O.); (Y.S.-C.); (M.A.D.l.F.-H.); (M.d.l.L.M.-A.); (A.P.-B.); (R.S.-L.); (G.O.-S.); (S.V.-M.); (P.M.N.-M.); (A.P.-T.)
| | - Ulises Juárez-Figueroa
- Instituto Nacional de Pediatría, Insurgentes Sur No. 3700-C. Coyoacán, Mexico City 04530, Mexico; (L.T.-M.); (U.J.-F.); (A.R.); (S.F.)
| | - Alejandra Padua-Bracho
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Av. San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (R.M.Á.-G.); (V.F.-O.); (Y.S.-C.); (M.A.D.l.F.-H.); (M.d.l.L.M.-A.); (A.P.-B.); (R.S.-L.); (G.O.-S.); (S.V.-M.); (P.M.N.-M.); (A.P.-T.)
| | - Rodrigo Sosa-León
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Av. San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (R.M.Á.-G.); (V.F.-O.); (Y.S.-C.); (M.A.D.l.F.-H.); (M.d.l.L.M.-A.); (A.P.-B.); (R.S.-L.); (G.O.-S.); (S.V.-M.); (P.M.N.-M.); (A.P.-T.)
| | - Gabriela Obregon-Serrano
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Av. San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (R.M.Á.-G.); (V.F.-O.); (Y.S.-C.); (M.A.D.l.F.-H.); (M.d.l.L.M.-A.); (A.P.-B.); (R.S.-L.); (G.O.-S.); (S.V.-M.); (P.M.N.-M.); (A.P.-T.)
| | - Silvia Vidal-Millán
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Av. San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (R.M.Á.-G.); (V.F.-O.); (Y.S.-C.); (M.A.D.l.F.-H.); (M.d.l.L.M.-A.); (A.P.-B.); (R.S.-L.); (G.O.-S.); (S.V.-M.); (P.M.N.-M.); (A.P.-T.)
| | - Paulina María Núñez-Martínez
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Av. San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (R.M.Á.-G.); (V.F.-O.); (Y.S.-C.); (M.A.D.l.F.-H.); (M.d.l.L.M.-A.); (A.P.-B.); (R.S.-L.); (G.O.-S.); (S.V.-M.); (P.M.N.-M.); (A.P.-T.)
| | - Abraham Pedroza-Torres
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Av. San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (R.M.Á.-G.); (V.F.-O.); (Y.S.-C.); (M.A.D.l.F.-H.); (M.d.l.L.M.-A.); (A.P.-B.); (R.S.-L.); (G.O.-S.); (S.V.-M.); (P.M.N.-M.); (A.P.-T.)
| | - Sergio Nicasio-Arzeta
- Natural Resource Ecology Laboratory, Colorado State University, Fort Collins, CO 80521, USA;
| | - Alfredo Rodríguez
- Instituto Nacional de Pediatría, Insurgentes Sur No. 3700-C. Coyoacán, Mexico City 04530, Mexico; (L.T.-M.); (U.J.-F.); (A.R.); (S.F.)
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), México City 04510, Mexico
| | - Fernando Luna
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (J.D.-O.); (R.M.-M.); (C.C.-H.); (L.B.-H.); (L.C.-E.); (R.G.-B.); (F.L.)
| | - Fernanda Cisneros-Soberanis
- Wellcome Trust Centre for Cell Biology, ICB, University of Edinburgh, Michael Swann Building, King’s Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK;
| | - Sara Frías
- Instituto Nacional de Pediatría, Insurgentes Sur No. 3700-C. Coyoacán, Mexico City 04530, Mexico; (L.T.-M.); (U.J.-F.); (A.R.); (S.F.)
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), México City 04510, Mexico
| | - Cristian Arriaga-Canon
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (J.D.-O.); (R.M.-M.); (C.C.-H.); (L.B.-H.); (L.C.-E.); (R.G.-B.); (F.L.)
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64710, Mexico
| | - Luis A. Herrera-Montalvo
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico; (J.D.-O.); (R.M.-M.); (C.C.-H.); (L.B.-H.); (L.C.-E.); (R.G.-B.); (F.L.)
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64710, Mexico
| |
Collapse
|
58
|
Xu C, Li X. Trends and frontiers of maintenance therapy for ovarian cancer over the past 20 years: a bibliometric analysis. Future Oncol 2024; 20:1925-1942. [PMID: 38864301 PMCID: PMC11497917 DOI: 10.1080/14796694.2024.2357378] [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] [Received: 09/29/2022] [Accepted: 05/15/2024] [Indexed: 06/13/2024] Open
Abstract
Aim: To summarize and analyze the research trends and frontiers in maintenance therapy for ovarian cancer over the past 20 years.Methods: Relevant articles were identified in Web of Science, and analyzed using CiteSpace and Excel.Results: 1204 articles from 61 countries led by the USA and England were included. 6821 authors, 2345 institutions and 292 journals have participated in the publication of papers, but the collaboration between them was not very close. The annual publication volume has increased significantly since 2015. Drug combination therapy, genetic testing, management of adverse reaction and prognostic factors are research trends and frontiers of this field.Conclusion: This is the first bibliometric research in this field, which can provide some references for researchers.
Collapse
Affiliation(s)
- Chunju Xu
- Department of Gynecology, Qianjiang Central Hospital of Chongqing, Qianjiang District, Chongqing, China
- Department of Gynecology, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Xia Li
- Department of Gynecology, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
59
|
Abdel-Razeq H, Tamimi F, Iweir S, Sharaf B, Abdel-Razeq S, Salama O, Edaily S, Bani Hani H, Azzam K, Abaza H. Genetic counseling and genetic testing for pathogenic germline mutations among high-risk patients previously diagnosed with breast cancer: a traceback approach. Sci Rep 2024; 14:12820. [PMID: 38834641 DOI: 10.1038/s41598-024-63300-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/27/2024] [Indexed: 06/06/2024] Open
Abstract
Genetic counseling and testing are more accessible than ever due to reduced costs, expanding indications and public awareness. Nonetheless, many patients missed the opportunity of genetic counseling and testing due to barriers that existed at that time of their cancer diagnoses. Given the identified implications of pathogenic mutations on patients' treatment and familial outcomes, an opportunity exists to utilize a 'traceback' approach to retrospectively examine their genetic makeup and provide consequent insights to their disease and treatment. In this study, we identified living patients diagnosed with breast cancer (BC) between July 2007 and January 2022 who would have been eligible for testing, but not tested. Overall, 422 patients met the eligibility criteria, 282 were reached and invited to participate, and germline testing was performed for 238, accounting for 84.4% of those invited. The median age (range) was 39.5 (24-64) years at BC diagnosis and 49 (31-75) years at the date of testing. Genetic testing revealed that 25 (10.5%) patients had pathogenic/likely pathogenic (P/LP) variants; mostly in BRCA2 and BRCA1. We concluded that long overdue genetic referral through a traceback approach is feasible and effective to diagnose P/LP variants in patients with history of BC who had missed the opportunity of genetic testing, with potential clinical implications for patients and their relatives.
Collapse
Affiliation(s)
- Hikmat Abdel-Razeq
- Department of Internal Medicine, King Hussein Cancer Center, 202 Queen Rania Al Abdullah Street, P.O. Box: 1269, Amman, 11941, Jordan.
- School of Medicine, The University of Jordan, Amman, Jordan.
| | - Faris Tamimi
- Department of Internal Medicine, King Hussein Cancer Center, 202 Queen Rania Al Abdullah Street, P.O. Box: 1269, Amman, 11941, Jordan
| | - Sereen Iweir
- Department of Internal Medicine, King Hussein Cancer Center, 202 Queen Rania Al Abdullah Street, P.O. Box: 1269, Amman, 11941, Jordan
- CRDF Global, Global Health Mission Area, Amman, Jordan
| | - Baha Sharaf
- Department of Internal Medicine, King Hussein Cancer Center, 202 Queen Rania Al Abdullah Street, P.O. Box: 1269, Amman, 11941, Jordan
| | | | - Osama Salama
- Department of Internal Medicine, King Hussein Cancer Center, 202 Queen Rania Al Abdullah Street, P.O. Box: 1269, Amman, 11941, Jordan
| | - Sarah Edaily
- Department of Internal Medicine, King Hussein Cancer Center, 202 Queen Rania Al Abdullah Street, P.O. Box: 1269, Amman, 11941, Jordan
| | - Hira Bani Hani
- Department of Internal Medicine, King Hussein Cancer Center, 202 Queen Rania Al Abdullah Street, P.O. Box: 1269, Amman, 11941, Jordan
| | - Khansa Azzam
- Department of Internal Medicine, King Hussein Cancer Center, 202 Queen Rania Al Abdullah Street, P.O. Box: 1269, Amman, 11941, Jordan
| | - Haneen Abaza
- Office of Scientific Affairs and Research, King Hussein Cancer Center, Amman, Jordan
| |
Collapse
|
60
|
Oliva R, Grassi S, Marchetti C, Cazzato F, Marinelli R, Scambia G, Fagotti A. Medicolegal and insurance issues regarding BRCA1 and BRCA2 gene tests in high income countries. Int J Gynecol Cancer 2024; 34:935-941. [PMID: 38642925 DOI: 10.1136/ijgc-2023-005225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2024] Open
Abstract
Hereditary breast and ovarian cancer syndrome is an autosomal dominant cancer susceptibility syndrome mainly due to variants in BRCA1 or BRCA2 genes. Patients presenting with BRCA1 or BRCA2 gene mutations have a lifetime risk of developing breast or ovarian cancer (80% and 40%, respectively). Genetic testing to explore the predisposition to develop cancer represents a pivotal factor in such cases, and this review wants to explore the main implications in terms of medicolegal liability and insurance issues. Medicolegal issues related to these diagnostic processes include: (a) failure to recommend the test; (b) failure to properly interpret the test; (c) failure to correctly translate results into clinical practice; (d) lack of informed consent; and (e) failure to refer patients to specialized genetic counseling. Such errors may lead to compensation since the legal burden inherent in the efficacy of prophylactic interventions is a proof that requires the so-called 'preponderance of the evidence'. Concerning insurance issues, the carriers of such alleles without cancer are healthy because the genetic predisposition is not a disease per se but represents a (relevant) health risk. However, disclosure of these conditions can be impelled by insurers. It can lead to so-called 'genetic discrimination' because insurance companies might use genetic information to limit insurance options or increase their costs. Many private and public healthcare funders do not cover risk reducing surgeries, even when recommended as part of a risk reduction management plan for BRCA gene mutation carriers. Here, positions on these matters from different high income countries are discussed, stressing the importance of a common supranational or international regulatory framework to reach a trade-off between the economic interests of insurers and the rights of carriers not to disclose extremely sensitive information.
Collapse
Affiliation(s)
- Riccardo Oliva
- Department of Women's and Child Health and Public Health Sciences, Obstetrics and Gynecology, Università Cattolica del Sacro Cuore - Campus di Roma, Roma, Italy
| | - Simone Grassi
- Department of Health Sciences, Section of Forensic Medical Sciences, University of Florence, Firenze, Italy
| | - Claudia Marchetti
- Università Cattolica del Sacro Cuore - Campus di Roma, Roma, Italy
- Department of Women's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Francesca Cazzato
- Section Legal Medicine, Institute of Public Health, Università Cattolica del Sacro Cuore - Campus di Roma, Roma, Italy
| | - Roberta Marinelli
- Section Legal Medicine, Institute of Public Health, Università Cattolica del Sacro Cuore - Campus di Roma, Roma, Italy
| | - Giovanni Scambia
- Università Cattolica del Sacro Cuore - Campus di Roma, Roma, Italy
- Department of Women's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Anna Fagotti
- Università Cattolica del Sacro Cuore - Campus di Roma, Roma, Italy
- Department of Women's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| |
Collapse
|
61
|
Loizzi V, Cerbone M, Arezzo F, Silvestris E, Damiani GR, Cazzato G, Cicinelli E, Cormio G. Contraception as chemoprevention of ovarian cancer in BRCA1 and BRCA2 women. Hormones (Athens) 2024; 23:277-286. [PMID: 38112915 DOI: 10.1007/s42000-023-00519-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
Ovarian cancer is the seventh most common cancer in women in the world, with an estimated worldwide mortality of over 207'000 women every year. This cancer, due to the current lack of adequate screening techniques, is commonly diagnosed late and has a poor prognosis. The oral contraceptive pill is considered the most effective prevention strategy for ovarian cancer in the general population, being associated with a decreased incidence while also having a substantial positive impact on the mortality rate, which is reduced by up to 50%. BRCA1 and BRCA2 germline mutated women have an augmented risk of ovary and breast cancer: despite international guidelines that consider prophylactic surgery as the gold standard for ovarian cancer prevention, there are currently no effective non-invasive preventive methods. In BRCA1\2 mutated patients, clinicians should weigh the benefits of contraceptive pills against the risk of long-term thromboembolic side effects and hormonal malignancies such as breast and cervical cancer. A multidisciplinary team should counsel patients on the most appropriate risk-reduction strategy tailored to their needs and expectations, proposing the oral contraceptive pill to selected patients after balancing the risks of adverse effects and the benefits on both contraception and chemoprevention.
Collapse
Affiliation(s)
- Vera Loizzi
- S.S.D. Ginecologia Oncologica Clinicizzata, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
- Dipartimento Di Biomedicina Traslazionale E Neuroscienze (DiBraiN), University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Marco Cerbone
- Obstetrics and Gynecology Unit, University of Bari, Bari, Italy.
| | - Francesca Arezzo
- Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Erica Silvestris
- Oncology Unit IRCSS Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy
| | | | - Gerardo Cazzato
- Section of Molecular Pathology, Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Ettore Cicinelli
- Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Gennaro Cormio
- Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro", 70124, Bari, Italy
- Oncology Unit IRCSS Istituto Tumori "Giovanni Paolo II", 70124, Bari, Italy
| |
Collapse
|
62
|
Mannix J, Duke H, Almajnooni A, Ongkeko M. Imaging the Male Breast: Gynecomastia, Male Breast Cancer, and Beyond. Radiographics 2024; 44:e230181. [PMID: 38752766 DOI: 10.1148/rg.230181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2024]
Abstract
The number of men undergoing breast imaging has increased in recent years, according to some reports. Most male breast concerns are related to benign causes, most commonly gynecomastia. The range of abnormalities typically encountered in the male breast is less broad than that encountered in women, given that lobule formation rarely occurs in men. Other benign causes of male breast palpable abnormalities with characteristic imaging findings include lipomas, sebaceous or epidermal inclusion cysts, and intramammary lymph nodes. Male breast cancer (MBC) is rare, representing up to 1% of breast cancer cases, but some data indicate that its incidence is increasing. MBC demonstrates some clinical features that overlap with those of gynecomastia, including a propensity for the subareolar breast. Men with breast cancer tend to present at a later stage than do women. MBC typically has similar imaging features to those of female breast cancer, often characterized by an irregular mass that may have associated calcifications. Occasionally, however, MBC has a benign-appearing imaging phenotype, with an oval shape and circumscribed margins, and therefore most solid breast masses in men require tissue diagnosis. Histopathologic evaluation may alternatively reveal other benign breast masses found in men, including papillomas, myofibroblastomas, and hemangiomas. Radiologists must be familiar with the breadth of male breast abnormalities to meet the rising challenge of caring for these patients. ©RSNA, 2024 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Jaimee Mannix
- From the Department of Radiology, Breast Imaging Section, Mallinckrodt Institute of Radiology (J.M., H.D.) and Department of Pathology (A.A., M.O.), Washington University in St. Louis School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St. Louis, MO 63110
| | - Heather Duke
- From the Department of Radiology, Breast Imaging Section, Mallinckrodt Institute of Radiology (J.M., H.D.) and Department of Pathology (A.A., M.O.), Washington University in St. Louis School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St. Louis, MO 63110
| | - Abdullah Almajnooni
- From the Department of Radiology, Breast Imaging Section, Mallinckrodt Institute of Radiology (J.M., H.D.) and Department of Pathology (A.A., M.O.), Washington University in St. Louis School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St. Louis, MO 63110
| | - Martin Ongkeko
- From the Department of Radiology, Breast Imaging Section, Mallinckrodt Institute of Radiology (J.M., H.D.) and Department of Pathology (A.A., M.O.), Washington University in St. Louis School of Medicine, 510 S Kingshighway Blvd, Campus Box 8131, St. Louis, MO 63110
| |
Collapse
|
63
|
Summey RM, Gornstein E, Decker B, Dougherty KC, Rader JS, Hopp E. Landscape of potential germline pathogenic variants in select cancer susceptibility genes in patients with adult-type ovarian granulosa cell tumors. Cancer Med 2024; 13:e7340. [PMID: 38898688 PMCID: PMC11187164 DOI: 10.1002/cam4.7340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
OBJECTIVE The objective of this study was to assess the frequency of potential germline pathogenic variants that may contribute to risk of development of adult granulosa cell tumors (AGCT) given the paucity of germline testing guidelines for these patients. METHODS This was a retrospective cross-sectional study analyzing comprehensive genomic profiling (CGP) results of AGCT with the FOXL2 p.C134W mutation submitted to Foundation Medicine between 2012 and 2022. Cases with a potential germline pathogenic variant were identified by filtering single nucleotide variants and short indels by variant allele frequency (VAF) and presence in ClinVar for select cancer susceptibility genes. Odds ratios for AGCT risk were calculated compared to a healthy population. RESULTS Prior to analysis, 595 patients were screened and 516 with a somatic FOXL2 p.C134W mutation were included. Potential germline pathogenic variants in a DNA repair-related gene (ATM, BRCA1, BRCA2, CHEK2, PALB2, PMS2, RAD51C, or RAD51D) were found in 6.6% of FOXL2-mutated AGCT. Potential germline pathogenic CHEK2 variants were found in 3.5% (18/516) of AGCT patients, a rate that was 2.8-fold higher than Genome Aggregation Database non-cancer subjects (95% CI 1.8-4.6, p < 0.001). The founder variants p.I157T (38.9%, 7/18) and p.T367fs*15 (c.1100delC; 27.8%, 5/18) were most commonly observed. CHEK2 VAF indicated frequent loss of the wildtype copy of the gene. CONCLUSIONS These results support ongoing utilization of genomic tumor profiling and confirmatory germline testing for potential germline pathogenic variants. Further prospective investigation into the biology of germline variants in this population is warranted.
Collapse
Affiliation(s)
- Rebekah M. Summey
- Division of Gynecologic Oncology, Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | | | | | | | - Janet S. Rader
- Division of Gynecologic Oncology, Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Elizabeth Hopp
- Division of Gynecologic Oncology, Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWisconsinUSA
| |
Collapse
|
64
|
Bufman H, Faermann R, Halshtok-Neiman O, Shalmon A, Gotlieb M, Samoocha D, Yagil Y, Feldman DM, Friedman E, Sklair-Levy M. Breast cancer diagnosed after age 70 years in Israeli BRCA1/BRCA2 pathogenic sequence variant carriers: a single institution experience. Breast Cancer Res Treat 2024; 205:281-285. [PMID: 38379091 PMCID: PMC11101537 DOI: 10.1007/s10549-023-07234-1] [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] [Received: 08/09/2023] [Accepted: 12/14/2023] [Indexed: 02/22/2024]
Abstract
PURPOSE A semi-annual surveillance scheme from age 25 to 30 years is offered to BRCA1/BRCA2 pathogenic sequence variants (PSVs) carriers for early detection of breast cancer (BC). There is a paucity of data on the yield of adhering to this scheme beyond 70 years of age. METHODS Female BRCA1/BRCA2 PSV carriers followed at the Meirav high-risk clinic, Sheba Medical center, Israel were eligible. Type and frequencies if use of Imaging modalities, breast biopsies and histological outcomes for participants after age 70 years were retrieved and analyzed. RESULTS Overall, the study encompassed 88 consenting participants (46 BRCA1 carriers) mean age ± SD 73.7 ± 3.3 years (range 70-90 years), followed for an average of 3.8 years (range 1-11 years). Ten carriers (11.3%) were diagnosed with BC after age 70 years (mean age at diagnosis 72 ± 2 years) and an additional case was diagnosed with breast lymphoma. The imaging modality that has led to most diagnoses was MRI (8/11 cases). Eight of these ten cases were previously diagnosed with BC prior to age 70 and in six, BC past 70 years was in the contralateral breast. The lesions size averaged 1.29 ± 0.75 cm, with IDC and DCIS diagnosed in five cases each, and none had lymph node involvement. CONCLUSION In ~10% of BRCA1/BRCA2 PSV carriers BC is diagnosed by breast imaging after age 70 years. If these results are validated in a larger study, the guidelines for the maximum age for BC surveillance in high risk women should be revisited and set at 75 years.
Collapse
Affiliation(s)
- Hila Bufman
- The Institute of Radiology, Sheba Medical Center, 1 Emek Ha-Ella St, 5265601, Tel-Hashomer, Israel
| | - Renata Faermann
- The Institute of Radiology, Sheba Medical Center, 1 Emek Ha-Ella St, 5265601, Tel-Hashomer, Israel
- The Meirav High Risk Clinic, Sheba Medical Center, Tel-Hashomer, Israel
| | - Osnat Halshtok-Neiman
- The Institute of Radiology, Sheba Medical Center, 1 Emek Ha-Ella St, 5265601, Tel-Hashomer, Israel
- The Meirav High Risk Clinic, Sheba Medical Center, Tel-Hashomer, Israel
| | - Anat Shalmon
- The Institute of Radiology, Sheba Medical Center, 1 Emek Ha-Ella St, 5265601, Tel-Hashomer, Israel
- The Meirav High Risk Clinic, Sheba Medical Center, Tel-Hashomer, Israel
| | - Michael Gotlieb
- The Institute of Radiology, Sheba Medical Center, 1 Emek Ha-Ella St, 5265601, Tel-Hashomer, Israel
- The Meirav High Risk Clinic, Sheba Medical Center, Tel-Hashomer, Israel
| | - David Samoocha
- The Institute of Radiology, Sheba Medical Center, 1 Emek Ha-Ella St, 5265601, Tel-Hashomer, Israel
- The Meirav High Risk Clinic, Sheba Medical Center, Tel-Hashomer, Israel
| | - Yael Yagil
- The Institute of Radiology, Sheba Medical Center, 1 Emek Ha-Ella St, 5265601, Tel-Hashomer, Israel
- The Meirav High Risk Clinic, Sheba Medical Center, Tel-Hashomer, Israel
| | | | - Eitan Friedman
- The Meirav High Risk Clinic, Sheba Medical Center, Tel-Hashomer, Israel
- The School of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Center for Personalized Medicine, Assuta Medical Center, Tel Aviv, Israel
| | - Miri Sklair-Levy
- The Institute of Radiology, Sheba Medical Center, 1 Emek Ha-Ella St, 5265601, Tel-Hashomer, Israel.
- The Meirav High Risk Clinic, Sheba Medical Center, Tel-Hashomer, Israel.
- The School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
65
|
Laguna JC, Tagliamento M, Lambertini M, Hiznay J, Mezquita L. Tackling Non-Small Cell Lung Cancer in Young Adults: From Risk Factors and Genetic Susceptibility to Lung Cancer Profile and Outcomes. Am Soc Clin Oncol Educ Book 2024; 44:e432488. [PMID: 38788188 DOI: 10.1200/edbk_432488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Lung cancer has traditionally been associated with advanced age; however, its increasing incidence among young adults raises concerning questions regarding its etiology and unique considerations for this population. In contrast to the older population, the onset of lung cancer at younger age may be attributed to a complex interplay of incompletely understood individual susceptibility and prevalent environmental risk factors beyond tobacco smoke exposure, such as radon gas and air pollution, which are widespread globally. Consequently, this leads to distinct clinical and molecular profiles, requiring a tailored approach. Furthermore, a diagnosis of cancer represents a threatening event during the prime years of a young person's life, prompting concern about career development, social aspects, fertility aspirations, and physical independence. This poses significant additional challenges for health care professionals in a field that remains underexplored. This comprehensive review recognizes lung cancer in young adults as a distinct entity, exploring its clinical and molecular characteristics, diverse predisposing factors, and priorities in terms of quality of life, with the aim of providing practical support to oncologists and enhancing our understanding of this under-researched population.
Collapse
Affiliation(s)
- Juan Carlos Laguna
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
| | - Marco Tagliamento
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Laura Mezquita
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
66
|
García-Iturbide R, Velázquez JA, Lozano Guzmán I, Falcon-Molina JE, Rodríguez MA, Sánchez-Gómez A, Heras Lorenzana JR, Estrada Estrada EM. Treatment and Diagnostic Approach for Lhermitte-Duclos Disease and Suspected Cowden Syndrome. Cureus 2024; 16:e62968. [PMID: 39044874 PMCID: PMC11265621 DOI: 10.7759/cureus.62968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2024] [Indexed: 07/25/2024] Open
Abstract
Lhermitte-Duclos disease (LDD) is a rare entity, which may or may not be associated with Cowden syndrome (CS). The authors present a 26-year-old male with a history of emergency treatment due to acute obstructive hydrocephalus and apparent Chiari malformation. In posterior evaluation, mild cerebellar symptoms, mucocutaneous lesions, and a left hemispheric cerebellar lesion were evident. Initially, with the clinical evidence and the radiological study report of a cerebellar tiger-striped lesion, LDD with associated CS was suspected, and a genetic protocol was performed. The protocol included an endoscopy and thyroid ultrasound, and with symptom progression, a new neurosurgical procedure was performed. To complete the approach, we used the clinical criteria for PTEN hamartoma tumor syndrome established in 2013, and CS was diagnosed in the patient. In patients with radiological and clinical suspicion of LDD and CS, it should be mandatory to investigate the presence of other types of tumors due to their association with PTEN hamartomatous tumor syndrome, and in the absence of genetic study, the clinical criteria previously established in the literature should be sufficient to establish the diagnosis.
Collapse
Affiliation(s)
- Ricardo García-Iturbide
- Neurological Surgery, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, MEX
| | - Joel A Velázquez
- Neurological Surgery, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, MEX
| | - Isauro Lozano Guzmán
- Neurological Surgery, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, MEX
| | - Jesus E Falcon-Molina
- Neurological Surgery, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, MEX
| | - Marco A Rodríguez
- Pathology, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, MEX
| | - Adrian Sánchez-Gómez
- Neurological Surgery, Hospital Regional 1ro de Octubre, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Mexico City, MEX
| | - Jesùs R Heras Lorenzana
- Neurological Surgery, Hospital Regional de Alta Especialidad Bicentenario de la Independencia, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Tultitlán de Mariano Escobedo, MEX
| | - Eric M Estrada Estrada
- Neurological Surgery, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, MEX
| |
Collapse
|
67
|
Harrold EC, Stadler ZK. Upper Gastrointestinal Cancers and the Role of Genetic Testing. Hematol Oncol Clin North Am 2024; 38:677-691. [PMID: 38458854 DOI: 10.1016/j.hoc.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024]
Abstract
Beyond the few established hereditary cancer syndromes with an upper gastrointestinal cancer component, there is increasing recognition of the contribution of novel pathogenic germline variants (gPVs) to upper gastrointestinal carcinogenesis. The detection of gPVs has potential implications for novel treatment approaches of the index cancer patient as well as long-term implications for surveillance and risk-reducing measures for cancer survivors and far-reaching implications for the patients' family. With widespread availability of multigene panel testing, new associations may be identified with germline-somatic integration being critical to determining true causality of novel gPVs. Comprehensive cancer care should incorporate both somatic and germline testing.
Collapse
Affiliation(s)
- Emily C Harrold
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland. https://twitter.com/EmilyHarrold6
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
68
|
Mauer Hall CB, Reys BD, Gemmell AP, Campbell CL, Pirzadeh-Miller SM. Downstream Revenue Generated by Patients With Hereditary Cancer in the Multigene Panel Testing Era. JCO Oncol Pract 2024:OP2300817. [PMID: 38815190 DOI: 10.1200/op.23.00817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/16/2024] [Accepted: 04/02/2024] [Indexed: 06/01/2024] Open
Abstract
PURPOSE Patients with hereditary cancer syndromes face increased medical management recommendations to address their cancer risks. As multigene panels are the standard of testing today, more patients needing clinical intervention are being identified. This study calculates the downstream revenue (DSR) generated by patients ascertained by a genetic counselor (GC) with a hereditary cancer likely pathogenic/pathogenic variant (LPV/PV). METHODS Retrospective chart review was performed for patients seen in a high-volume cancer genetics clinic between October 1, 2009, and December 31, 2021, with LPV/PVs in hereditary cancer predisposition genes. DSR and work relative value units (wRVUs) were calculated for each patient before and after they met with a GC. Subgroup analyses calculated DSR/wRVUs from patients affected and unaffected with cancer and those whose genetic counseling visit was the first at the institution (naїve). RESULTS A total of 978 patients were available for analysis after exclusions were applied. Patients generated $73.06 million (M) in US dollars (USD) in DSR and 54,814 wRVUs after their initial genetic counseling visit. Unaffected patients (n = 370, 37.8%) generated $11.38M (USD) and 13,879 wRVUs; affected patients (n = 608, 62.2%) generated $61.68M (USD) and 40,935 wRVUs. Naïve patients (n = 367, 37.5%) generated $15.39M (USD) and 11,811 wRVUs; established patients (n = 611, 62.5%) generated $57.67M (USD) and 43,003 wRVUs. Unaffected, naïve patients (n = 204, 20.9%) generated $5.48M (USD) and 5,186 wRVUs. CONCLUSION By identifying patients with hereditary cancer, GCs can bring in substantial DSR for their institution. Naïve and unaffected patients provide the greatest GC value-add as these patients represent new business and revenue sources to the institution. As multigene panels continue to expand, the number of patients needing downstream services will increase. Recognizing patients at increased cancer risk will improve patient outcomes while simultaneously providing DSR for institutions.
Collapse
Affiliation(s)
- Caitlin B Mauer Hall
- Department of Health Care Sciences, University of Texas Southwestern Medical Center, Dallas, TX
| | - Brian D Reys
- Cancer Genetics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Amber P Gemmell
- Cancer Genetics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Connor L Campbell
- Simmons Comprehensive Cancer Center Finance, University of Texas Southwestern Medical Center, Dallas, TX
| | | |
Collapse
|
69
|
Austin S, Hanson EN, Delacroix E, Bacon E, Rice J, Gerido LH, Rizzo E, Pleasant V, Stoffel EM, Griggs JJ, Resnicow K. Impact of barriers and motivators on intention and confidence to undergo hereditary cancer genetic testing. J Genet Couns 2024. [PMID: 38803214 DOI: 10.1002/jgc4.1926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
Genetic testing for hereditary cancer syndromes can provide lifesaving information allowing for individualized cancer screening, prevention, and treatment. However, the determinants, both barriers and motivators, of genetic testing intention are not well described. A survey of barriers and motivators to genetic testing was emailed to adult patients eligible for genetic testing based on cancer diagnosis who previously have not had genetic testing (n = 201). Associations between barriers/motivators with testing intention and confidence were examined first by correlation followed by multivariable linear regression model holding constant potential covariates. Seven barrier items from two domains (logistics and genetic testing knowledge) were found to significantly negatively correlate with genetic testing intention. Unexpectedly, three barrier items had significant positive correlation with genetic testing intention; these were related to family worry (passing a condition on to future generations) and testing knowledge (needing more information on the genetic testing process and what it has to offer). Ten barrier items had significant negative correlation with confidence to get a genetic test and encompassed four domains: stigma, insurance/genetic discrimination, knowledge, and cost. All motivator items were associated with intention to get a genetic test, while none were associated with confidence. Multivariable analysis yielded six total barriers (five from the knowledge domain, one from cost domain) and two motivators (relieved to know and treatment impact) that were significantly associated with genetic testing intention or confidence when controlling for demographic characteristics. These findings indicate the need for tailored interventions to amplify motivating factors and counter-message barriers to enhance patient motivation and confidence to undergo testing.
Collapse
Affiliation(s)
- Sarah Austin
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Erika N Hanson
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Emerson Delacroix
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
- University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Elizabeth Bacon
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - John Rice
- University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | | | - Elizabeth Rizzo
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Versha Pleasant
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Elena M Stoffel
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Jennifer J Griggs
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
- University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Ken Resnicow
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| |
Collapse
|
70
|
Sanabria-Salas MC, Pedroza-Duran A, Díaz-Casas SE, Nuñez Lemus M, Grillo-Ardila CF, Briceño-Morales X, García-Mora M, Ángel-Aristizábal J, Mariño Lozano IF, Suarez Rodríguez RA, Guzmán Abisaab LH. Management and Clinical Outcomes of Breast Cancer in Women Diagnosed with Hereditary Cancer Syndromes in a Clinic-Based Sample from Colombia. Cancers (Basel) 2024; 16:2020. [PMID: 38893140 PMCID: PMC11171067 DOI: 10.3390/cancers16112020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/21/2024] Open
Abstract
This study aimed to investigate prognosis and survival differences in 82 breast cancer patients with germline pathogenic/likely pathogenic variants (PVs) treated and followed at the Breast Unit of the Instituto Nacional de Cancerología, Colombia (INC-C) between 2018 and 2021. Median age at diagnosis was 46 years, with 62.2% presenting locally advanced tumors, 47.6% histological grade 3, and 35.4% with triple-negative breast cancer (TNBC) subtype. Most carriers, 74.4% (61/82), had PVs in known breast cancer susceptibility genes (i.e., "associated gene carriers" group, considered inherited breast cancer cases): BRCA2 (30), BRCA1 (14), BARD1 (4), RAD51D (3), TP53 (2), PALB2 (2), ATM (2), CHEK2 (1), RAD51C (1), NF1 (1), and PTEN (1). BRCA1-2 represented 53.7%, and homologous recombination DNA damage repair (HR-DDR) genes associated with breast cancer risk accounted for 15.9%. Patients with PVs in non-breast-cancer risk genes were combined in a different category (21/82; 25.6%) (i.e., "non-associated gene carriers" group, considered other breast cancer cases). Median follow-up was 38.1 months, and 24% experienced recurrence, with 90% being distant. The 5-year Disease-Free Survival (DFS) for inherited breast cancer cases was 66.5%, and for other breast cancer cases it was 88.2%. In particular, for carriers of PVs in the BRCA2 gene, it was 37.6%. The 5-year Overall Survival (OS) rates ranged from 68.8% for those with PVs in BRCA2 to 100% for those with PVs in other HR-DDR genes. Further studies are crucial for understanding tumor behavior and therapy response differences among Colombian breast cancer patients with germline PVs.
Collapse
Affiliation(s)
- María Carolina Sanabria-Salas
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON M5G 2C1, Canada
| | - Ana Pedroza-Duran
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Sandra E. Díaz-Casas
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Marcela Nuñez Lemus
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Carlos F. Grillo-Ardila
- Department of Obstetrics & Gynecology, School of Medicine, Universidad Nacional de Colombia, Avenida Carrera 30 N. 45-3, Bogotá 111321, Colombia;
| | - Ximena Briceño-Morales
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Mauricio García-Mora
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Javier Ángel-Aristizábal
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Iván Fernando Mariño Lozano
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Raúl Alexis Suarez Rodríguez
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| | - Luis Hernán Guzmán Abisaab
- Instituto Nacional de Cancerología, Calle 1 N. 9-85, Bogotá 111511, Colombia; (A.P.-D.); (S.E.D.-C.); (M.N.L.); (X.B.-M.); (M.G.-M.); (J.Á.-A.); (I.F.M.L.); (R.A.S.R.); (L.H.G.A.)
| |
Collapse
|
71
|
Sierra-Díaz DC, Cabrera R, Gonzalez-Vasquez LA, Angulo-Aguado M, Llinás-Caballero K, Fonseca-Mendoza DJ, Contreras-Bravo NC, Restrepo CM, Ortega-Recalde O, Morel A. Functional Analysis of BRCA1 3'UTR Variants Predisposing to Breast Cancer. Appl Clin Genet 2024; 17:57-62. [PMID: 38803352 PMCID: PMC11129763 DOI: 10.2147/tacg.s444546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/29/2024] [Indexed: 05/29/2024] Open
Abstract
Purpose Breast Cancer (BC) is the main female cancer diagnosed worldwide, and it has been described that few genes, such as BRCA1, have a high penetrance for this type of cancer. In this manuscript, we were interested in evaluating the effect of 3'UTR variants on BRCA1 expression. Patients and Methods To accomplish this objective, Whole Exome Sequencing (WES) data of 400 patients with unselected BC was used to filter variants located in the region of interest of BRCA1 gene, finding two of them (c.*36C>G and c.*369_373del). miRGate and miRanda in silico tools were used to predict microRNA (miRNA) interaction. Results The two variants (c.*36C>G, c.*369_373del) were predicted to affect miRNA interaction. After cloning of BRCA1 3'UTR into pMIR-Report vector, the construct was transfected into two BC cell lines (MDA-MB-231 and MCF-7), and the variant c.*36C>G evidenced overexpression of reporter gene luciferase, showing that the transcript was not being degraded by the miRNA in MDA-MB-231 cells. Conclusion The variant seems to protect against Triple Negative BC probably due to the expression level of miRNA in this particular cell line (MDA-MB-231). This is consistent with the clinical history of the patients who harbor BC Hormone Receptors positive (HR+).
Collapse
Affiliation(s)
- Diana Carolina Sierra-Díaz
- Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), School of Medicine and Health Sciences, Universidad Del Rosario, Bogotá, Colombia
| | - Rodrigo Cabrera
- Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), School of Medicine and Health Sciences, Universidad Del Rosario, Bogotá, Colombia
- Laboratorio de Biología Molecular y Pruebas Diagnósticas de Alta Complejidad, Fundación Cardioinfantil-Instituto de Cardiología, Bogotá, Colombia
| | - Laura Alejandra Gonzalez-Vasquez
- Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), School of Medicine and Health Sciences, Universidad Del Rosario, Bogotá, Colombia
| | - Mariana Angulo-Aguado
- Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), School of Medicine and Health Sciences, Universidad Del Rosario, Bogotá, Colombia
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Kevin Llinás-Caballero
- Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), School of Medicine and Health Sciences, Universidad Del Rosario, Bogotá, Colombia
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| | - Dora Janeth Fonseca-Mendoza
- Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), School of Medicine and Health Sciences, Universidad Del Rosario, Bogotá, Colombia
| | - Nora Constanza Contreras-Bravo
- Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), School of Medicine and Health Sciences, Universidad Del Rosario, Bogotá, Colombia
| | - Carlos Martin Restrepo
- Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), School of Medicine and Health Sciences, Universidad Del Rosario, Bogotá, Colombia
| | - Oscar Ortega-Recalde
- Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), School of Medicine and Health Sciences, Universidad Del Rosario, Bogotá, Colombia
- Departamento de Morfología, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, D.C, Colombia
| | - Adrien Morel
- Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), School of Medicine and Health Sciences, Universidad Del Rosario, Bogotá, Colombia
| |
Collapse
|
72
|
Wang J, Fu K, Zhang M, Liang L, Ni M, Sun HX, Yin R, Tang M. Mutation characteristics of cancer susceptibility genes in Chinese ovarian cancer patients. Front Oncol 2024; 14:1395818. [PMID: 38817903 PMCID: PMC11137316 DOI: 10.3389/fonc.2024.1395818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/03/2024] [Indexed: 06/01/2024] Open
Abstract
Introduction The association between mutations in susceptibility genes and the occurrence of ovarian cancer has been extensively studied. Previous research has primarily concentrated on genes involved in the homologous recombination repair pathway, particularly BRCA1 and BRCA2. However, a wider range of genes related to the DNA damage response pathways has not been fully explored. Methods To investigate the mutation characteristics of cancer susceptibility genes in the Chinese ovarian cancer population and the associations between gene mutations and clinical data, this study initially gathered a total of 1171 Chinese ovarian cancer samples and compiled a dataset of germline mutations in 171 genes. Results In this study, it was determined that MC1R and PRKDC were high-frequency ovarian cancer susceptibility genes in the Chinese population, exhibiting notable distinctions from those in European and American populations; moreover high-frequency mutation genes, such as MC1R: c.359T>C and PRKDC: c.10681T>A, typically had high-frequency mutation sites. Furthermore, we identified c.8187G>T as a characteristic mutation of BRCA2 in the Chinese population, and the CHEK2 mutation was significantly associated with the early onset of ovarian cancer, while the CDH1 and FAM175A mutations were more prevalent in Northeast China. Additionally, Fanconi anemia pathway-related genes were significantly associated with ovarian carcinogenesis. Conclusion In summary, this research provided fundamental data support for the optimization of ovarian cancer gene screening policies and the determination of treatment, and contributed to the precise intervention and management of patients.
Collapse
Affiliation(s)
- Jie Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Genomics, Shenzhen, China
| | - Kaiyu Fu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Molecular Epidemiology of Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mengpei Zhang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Molecular Epidemiology of Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | - Meng Ni
- BGI Genomics, Shenzhen, China
| | | | - Rutie Yin
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Molecular Epidemiology of Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | | |
Collapse
|
73
|
Ando Y, Dbouk M, Yoshida T, Abou Diwan E, Saba H, Dbouk A, Yoshida K, Roberts NJ, Klein AP, Burkhart R, He J, Hruban RH, Goggins M. Germline Pathogenic Variants in Patients With Pancreatic and Periampullary Cancers. JCO Precis Oncol 2024; 8:e2400101. [PMID: 38781545 DOI: 10.1200/po.24.00101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/11/2024] [Accepted: 04/02/2024] [Indexed: 05/25/2024] Open
Abstract
PURPOSE Inherited cancer susceptibility is often not suspected in the absence of a significant cancer family history. Pathogenic germline variants in pancreatic cancer are well-studied, and routine genetic testing is recommended in the guidelines. However, data on rare periampullary cancers other than pancreatic cancer are insufficient. We compared the prevalence of germline susceptibility variants in patients with pancreatic cancer and nonpancreatic periampullary cancers. MATERIALS AND METHODS Six hundred and eight patients who had undergone pancreaticoduodenal resection at a tertiary referral hospital were studied, including 213 with pancreatic ductal adenocarcinoma, 172 with ampullary cancer, 154 with distal common bile duct cancer, and 69 with duodenal adenocarcinoma. Twenty cancer susceptibility and candidate susceptibility genes were sequenced, and variant interpretation was assessed by interrogating ClinVar and PubMed. RESULTS Pathogenic or likely pathogenic, moderate- to high-penetrant germline variants were identified in 46 patients (7.7%), including a similar percentage of patients with pancreatic (8.5%) and nonpancreatic periampullary cancer (7.1%). Low-penetrant variants were identified in an additional 11 patients (1.8%). Eighty-nine percent of the moderate- to high-penetrant variants involved the major cancer susceptibility genes BRCA2, ATM, BRCA1, CDKN2A, MSH2/MLH1, and PALB2; the remaining 11% involved other cancer susceptibility genes such as BRIP1, BAP1, and MSH6. Almost all pathogenic variant carriers had a family history of cancer. CONCLUSION Patients with pancreatic and nonpancreatic periampullary cancer have a similar prevalence of pathogenic cancer susceptibility variants. Germline susceptibility testing should be considered for patients with any periampullary cancer.
Collapse
Affiliation(s)
- Yohei Ando
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Mohamad Dbouk
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Takeichi Yoshida
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Elizabeth Abou Diwan
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Helena Saba
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Ali Dbouk
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Kanako Yoshida
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Nicholas J Roberts
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Departments of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Alison P Klein
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Departments of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- The Bloomberg School of Public Health, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Richard Burkhart
- Departments of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Jin He
- Departments of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Ralph H Hruban
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Departments of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- The Bloomberg School of Public Health, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Michael Goggins
- Departments of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Departments of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Departments of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
74
|
Laguna JC, Pastor B, Nalda I, Hijazo-Pechero S, Teixido C, Potrony M, Puig-Butillé JA, Mezquita L. Incidental pathogenic germline alterations detected through liquid biopsy in patients with solid tumors: prevalence, clinical utility and implications. Br J Cancer 2024; 130:1420-1431. [PMID: 38532104 PMCID: PMC11059286 DOI: 10.1038/s41416-024-02607-9] [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] [Received: 09/15/2023] [Revised: 01/14/2024] [Accepted: 01/25/2024] [Indexed: 03/28/2024] Open
Abstract
Liquid biopsy, a minimally invasive approach for detecting tumor biomarkers in blood, has emerged as a leading-edge technique in cancer precision medicine. New evidence has shown that liquid biopsies can incidentally detect pathogenic germline variants (PGVs) associated with cancer predisposition, including in patients with a cancer for which genetic testing is not recommended. The ability to detect these incidental PGV in cancer patients through liquid biopsy raises important questions regarding the management of this information and its clinical implications. This incidental identification of PGVs raises concerns about cancer predisposition and the potential impact on patient management, not only in terms of providing access to treatment based on the tumor molecular profiling, but also the management of revealing genetic predisposition in patients and families. Understanding how to interpret this information is essential to ensure proper decision-making and to optimize cancer treatment and prevention strategies. In this review we provide a comprehensive summary of current evidence of incidental PGVs in cancer predisposition genes identified by liquid biopsy in patients with cancer. We critically review the methodological considerations of liquid biopsy as a tool for germline diagnosis, clinical utility and potential implications for cancer prevention, treatment, and research.
Collapse
Affiliation(s)
- Juan Carlos Laguna
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
| | - Belén Pastor
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Irene Nalda
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
| | - Sara Hijazo-Pechero
- Preclinical and Experimental Research in Thoracic Tumors (PRETT), Oncobell, Bellvitge Biomedical Research Institute (IDIBELL), l'Hospitalet de Llobregat, Barcelona, Spain
| | - Cristina Teixido
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Department of Pathology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Miriam Potrony
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, Barcelona, Spain
- CIBER of Rare Diseases (CIBERER), Barcelona, Spain
| | - Joan Antón Puig-Butillé
- CIBER of Rare Diseases (CIBERER), Barcelona, Spain
- Molecular Biology CORE, Hospital Clínic of Barcelona, IDIBAPS, Barcelona, Spain
| | - Laura Mezquita
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain.
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain.
- Department of Medicine, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
75
|
Head M, Cohn B, Wernli KJ, Palazzo L, Ehrlich K, Matson A, Knerr S. Young Women's Perspectives on Being Screened for Hereditary Breast and Ovarian Cancer Risk During Routine Primary Care. Womens Health Issues 2024; 34:268-275. [PMID: 38448251 PMCID: PMC11116046 DOI: 10.1016/j.whi.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/19/2024] [Accepted: 01/29/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE The U.S. Preventive Services Task Force recommends screening women to identify individuals eligible for genetic counseling based on a priori hereditary breast and ovarian cancer syndrome (HBOC) risk (i.e., risk assessment). However, risk assessment has not been widely integrated into primary care. This qualitative study explored young women's views on implementing routine HBOC risk assessment with a focus on equity and patient-centeredness. METHODS We conducted group discussions with young women (aged 21-40 years) receiving care in an integrated health care system. Discussion groups occurred in two phases and used a modified deliberative approach that included a didactic component and prioritized developing consensus. Twenty women participated in one of three initial small group discussions (phase one). All 20 were invited to participate in a subsequent large group discussion (phase two), and 15 of them attended. FINDINGS Key themes and recommendations were as follows. Risk assessment should be accessible, contextualized, and destigmatized to encourage participation and reduce anxiety, particularly for women who do not know their family history. Providers conducting risk assessments must be equipped to address women's informational needs, relieve emotionality, and plan next steps after positive screens. Finally, to minimize differential screening uptake, health care systems must prioritize equity in program design and contribute to external educational and outreach efforts. CONCLUSION Young women see pragmatic opportunities for health systems to optimize HBOC screening implementation.
Collapse
Affiliation(s)
- Mady Head
- Genetic Counseling Graduate Program, School of Medicine, University of Washington, Seattle, Washington
| | - Betty Cohn
- Institute for Public Health Genetics, School of Public Health, University of Washington, Seattle, Washington
| | - Karen J Wernli
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | - Lorella Palazzo
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | - Kelly Ehrlich
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | - Abigail Matson
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | - Sarah Knerr
- Department of Health Systems and Population Health, School of Public Health, University of Washington, Seattle, Washington.
| |
Collapse
|
76
|
Kotsopoulos J, Narod SA. Menopausal hormone therapy for BRCA mutation carriers: A case for precision medicine. Maturitas 2024; 183:107886. [PMID: 37980268 DOI: 10.1016/j.maturitas.2023.107886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/20/2023]
Affiliation(s)
- Joanne Kotsopoulos
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Steven A Narod
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
77
|
Ceyhan-Birsoy O, Stadler ZK. Tumor-Only Sequencing: A Story Only Half Told. JCO Precis Oncol 2024; 8:e2400226. [PMID: 38810205 DOI: 10.1200/po.24.00226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 04/15/2024] [Indexed: 05/31/2024] Open
Abstract
Germline testing and tumor sequencing are often both necessary for optimal cancer treatment and management.
Collapse
Affiliation(s)
- Ozge Ceyhan-Birsoy
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
78
|
Casolino R, Beer PA, Chakravarty D, Davis MB, Malapelle U, Mazzarella L, Normanno N, Pauli C, Subbiah V, Turnbull C, Westphalen CB, Biankin AV. Interpreting and integrating genomic tests results in clinical cancer care: Overview and practical guidance. CA Cancer J Clin 2024; 74:264-285. [PMID: 38174605 DOI: 10.3322/caac.21825] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/07/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
The last decade has seen rapid progress in the use of genomic tests, including gene panels, whole-exome sequencing, and whole-genome sequencing, in research and clinical cancer care. These advances have created expansive opportunities to characterize the molecular attributes of cancer, revealing a subset of cancer-associated aberrations called driver mutations. The identification of these driver mutations can unearth vulnerabilities of cancer cells to targeted therapeutics, which has led to the development and approval of novel diagnostics and personalized interventions in various malignancies. The applications of this modern approach, often referred to as precision oncology or precision cancer medicine, are already becoming a staple in cancer care and will expand exponentially over the coming years. Although genomic tests can lead to better outcomes by informing cancer risk, prognosis, and therapeutic selection, they remain underutilized in routine cancer care. A contributing factor is a lack of understanding of their clinical utility and the difficulty of results interpretation by the broad oncology community. Practical guidelines on how to interpret and integrate genomic information in the clinical setting, addressed to clinicians without expertise in cancer genomics, are currently limited. Building upon the genomic foundations of cancer and the concept of precision oncology, the authors have developed practical guidance to aid the interpretation of genomic test results that help inform clinical decision making for patients with cancer. They also discuss the challenges that prevent the wider implementation of precision oncology.
Collapse
Affiliation(s)
- Raffaella Casolino
- Wolfson Wohl Cancer Research Center, School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Philip A Beer
- Wolfson Wohl Cancer Research Center, School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Hull York Medical School, York, UK
| | | | - Melissa B Davis
- Department of Surgery, Weill Cornell Medicine, New York City, New York, USA
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Luca Mazzarella
- Laboratory of Translational Oncology and Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori, IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Chantal Pauli
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Vivek Subbiah
- Sarah Cannon Research Institute, Nashville, Tennessee, USA
| | - Clare Turnbull
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- National Cancer Registration and Analysis Service, National Health Service (NHS) England, London, UK
- Cancer Genetics Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - C Benedikt Westphalen
- Department of Medicine III, Ludwig Maximilians University (LMU) Hospital Munich, Munich, Germany
- Comprehensive Cancer Center, LMU Hospital Munich, Munich, Germany
- German Cancer Consortium, LMU Hospital Munich, Munich, Germany
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Center, School of Cancer Sciences, University of Glasgow, Glasgow, UK
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- South Western Sydney Clinical School, Liverpool, New South Wales, Australia
| |
Collapse
|
79
|
Mars N, Kerminen S, Tamlander M, Pirinen M, Jakkula E, Aaltonen K, Meretoja T, Heinävaara S, Widén E, Ripatti S. Comprehensive Inherited Risk Estimation for Risk-Based Breast Cancer Screening in Women. J Clin Oncol 2024; 42:1477-1487. [PMID: 38422475 PMCID: PMC11095905 DOI: 10.1200/jco.23.00295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 11/24/2023] [Accepted: 12/20/2023] [Indexed: 03/02/2024] Open
Abstract
PURPOSE Family history (FH) and pathogenic variants (PVs) are used for guiding risk surveillance in selected high-risk women but little is known about their impact for breast cancer screening on population level. In addition, polygenic risk scores (PRSs) have been shown to efficiently stratify breast cancer risk through combining information about common genetic factors into one measure. METHODS In longitudinal real-life data, we evaluate PRS, FH, and PVs for stratified screening. Using FinnGen (N = 117,252), linked to the Mass Screening Registry for breast cancer (1992-2019; nationwide organized biennial screening for age 50-69 years), we assessed the screening performance of a breast cancer PRS and compared its performance with FH of breast cancer and PVs in moderate- (CHEK2)- to high-risk (PALB2) susceptibility genes. RESULTS Effect sizes for FH, PVs, and high PRS (>90th percentile) were comparable in screening-aged women, with similar implications for shifting age at screening onset. A high PRS identified women more likely to be diagnosed with breast cancer after a positive screening finding (positive predictive value [PPV], 39.5% [95% CI, 37.6 to 41.5]). Combinations of risk factors increased the PPVs up to 45% to 50%. A high PRS conferred an elevated risk of interval breast cancer (hazard ratio [HR], 2.78 [95% CI, 2.00 to 3.86] at age 50 years; HR, 2.48 [95% CI, 1.67 to 3.70] at age 60 years), and women with a low PRS (<10th percentile) had a low risk for both interval- and screen-detected breast cancers. CONCLUSION Using real-life screening data, this study demonstrates the effectiveness of a breast cancer PRS for risk stratification, alone and combined with FH and PVs. Further research is required to evaluate their impact in a prospective risk-stratified screening program, including cost-effectiveness.
Collapse
Affiliation(s)
- Nina Mars
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Sini Kerminen
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Max Tamlander
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Matti Pirinen
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Helsinki Institute for Information Technology HIIT and Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Eveliina Jakkula
- Department of Clinical Genetics, HUSLAB, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Kirsimari Aaltonen
- Department of Clinical Genetics, HUSLAB, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Tuomo Meretoja
- Breast Surgery Unit, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
- University of Helsinki, Helsinki, Finland
| | - Sirpa Heinävaara
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Finnish Cancer Registry, Cancer Society of Finland, Helsinki, Finland
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Public Health, University of Helsinki, Helsinki, Finland
| |
Collapse
|
80
|
Innella G, Ferrari S, Miccoli S, Luppi E, Fortuno C, Parsons MT, Spurdle AB, Turchetti D. Clinical implications of VUS reclassification in a single-centre series from application of ACMG/AMP classification rules specified for BRCA1/2. J Med Genet 2024; 61:483-489. [PMID: 38160042 DOI: 10.1136/jmg-2023-109694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 12/17/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND BRCA1/2 testing is crucial to guide clinical decisions in patients with hereditary breast/ovarian cancer, but detection of variants of uncertain significance (VUSs) prevents proper management of carriers. The ENIGMA (Evidence-based Network for the Interpretation of Germline Mutant Alleles) BRCA1/2 Variant Curation Expert Panel (VCEP) has recently developed BRCA1/2 variant classification guidelines consistent with ClinGen processes, specified against the ACMG/AMP (American College of Medical Genetics and Genomics/Association for Molecular-Pathology) classification framework. METHODS The ClinGen-approved BRCA1/2-specified ACMG/AMP classification guidelines were applied to BRCA1/2 VUSs identified from 2011 to 2022 in a series of patients, retrieving information from the VCEP documentation, public databases, literature and ENIGMA unpublished data. Then, we critically re-evaluated carrier families based on new results and checked consistency of updated classification with main sources for clinical interpretation of BRCA1/2 variants. RESULTS Among 166 VUSs detected in 231 index cases, 135 (81.3%) found in 197 index cases were classified by applying BRCA1/2-specified ACMG/AMP criteria: 128 (94.8%) as Benign/Likely Benign and 7 (5.2%) as Pathogenic/Likely Pathogenic. The average time from the first report as 'VUS' to classification using this approach was 49.4 months. Considering that 15 of these variants found in 64 families had already been internally reclassified prior to this work, this study provided 121 new reclassifications among the 151 (80.1%) remaining VUSs, relevant to 133/167 (79.6%) families. CONCLUSIONS These results demonstrated the effectiveness of new BRCA1/2 ACMG/AMP classification guidelines for VUS classification within a clinical cohort, and their important clinical impact. Furthermore, they suggested a cadence of no more than 3 years for regular review of VUSs, which however requires time, expertise and resources.
Collapse
Affiliation(s)
- Giovanni Innella
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Simona Ferrari
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Sara Miccoli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Elena Luppi
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Cristina Fortuno
- Population Health, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Michael T Parsons
- Population Health, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Amanda B Spurdle
- Population Health, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Daniela Turchetti
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
81
|
de Oliveira Ferreira C, Carneiro VCG, Araujo Mariz C. Germline mutations in BRCA1 and BRCA2 among Brazilian women with ovarian cancer treated in the Public Health System. BMC Cancer 2024; 24:499. [PMID: 38641594 PMCID: PMC11027424 DOI: 10.1186/s12885-024-12246-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/10/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Germline mutations in BRCA1 and BRCA2 genes are among the main causes of hereditary ovarian cancer. Identifying these mutations may reduce cancer risk, facilitate early detection, and enable personalized treatment. However, genetic testing is limited in the Brazilian Public Health System, and data regarding germline mutations in many regions are scarce. Therefore, the study aimed to investigate the prevalence of germline mutations in BRCA1 and BRCA2 in women with ovarian cancer treated in the Public Health System in Pernambuco, Brazil. METHODS A cross-sectional study was conducted in the Hereditary Cancer Program from two reference oncological centers in Pernambuco. Women (n = 45) with high-grade serous ovarian cancer underwent genetic counseling and DNA sequencing for BRCA1 and BRCA2 genes. RESULTS The prevalence of deleterious mutations in the BRCA1 and BRCA2 genes was 33%. Of the 15 germline mutations found, 13 were in BRCA1 and 2 in BRCA2; two mutations of unknown clinical significance were also found in BRCA2. Mutations c.5266dupC and c.2215 A > T were the most frequent; each was mutation observed in three patients. Additionally, the mutations c.7645dupT and c.921dupT were reported for the first time. CONCLUSION One in three women showed a pathogenic mutation, demonstrating a significant prevalence of germline mutations in this sample. Additionally, the small sample revealed an interesting number of mutations, indicating the need to explore more regions of the country.
Collapse
Affiliation(s)
| | - Vandré Cabral Gomes Carneiro
- Hospital de Câncer de Pernambuco, Recife, Pernambuco, Brazil
- Instituto de Medicina Integral Professor Fernando Figueira, Recife, Pernambuco, Brazil
| | - Carolline Araujo Mariz
- Instituto Aggeu Magalhães, FIOCRUZ, Recife, Pernambuco, Brazil.
- Faculdade de Medicina de Olinda, Olinda, Pernambuco, Brazil.
| |
Collapse
|
82
|
Li PC, Zhu YF, Pan JN, Zhu QY, Liao YY, Ding XW, Zheng LF, Cao WM. HR-positive/HER2-negative breast cancer arising in patients with or without BRCA2 mutation: different biological phenotype and similar prognosis. Ther Adv Med Oncol 2024; 16:17588359241242613. [PMID: 38606163 PMCID: PMC11008348 DOI: 10.1177/17588359241242613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/12/2024] [Indexed: 04/13/2024] Open
Abstract
Background BRCA2 plays a key role in homologous recombination. However, information regarding its mutations in Chinese patients with breast cancer remains limited. Objectives This study aimed to assess the clinicopathological characteristics of BRCA2 mutation breast cancer and explore the mutation's effect on hormone receptor (HR)-positive/human epidermal growth factor receptor 2 (HER2)-negative breast cancer survival in China. Design This hospital-based cohort study prospectively included 629 women with breast cancer diagnosed from 2008 to 2023 at Zhejiang Cancer Hospital in China. Methods We compared the clinicopathological characteristics and metastatic patterns and analysed the invasive disease-free survival (iDFS), distant relapse-free survival (DRFS) and first-line progression-free survival (PFS1) of patients with HR-positive/HER2-negative breast cancer according to BRCA2 mutations. Results Among the 629 patients, 78 had BRCA2 mutations (12.4%) and 551 did not (87.6%). The mean age at diagnosis was lower in the BRCA2 mutation breast cancer group than in the non-mutation breast cancer group (38.91 versus 41.94 years, p = 0.016). BRCA2 mutation breast cancers were more likely to be lymph node-positive than non-mutation breast cancers (73.0% versus 56.6%, p = 0.037). The pathological grade was higher in 47.1% of BRCA2 mutation breast cancers than in 29.6% of non-mutation breast cancers (p = 0.014). The proportions of patients with BRCA2 mutations who developed contralateral breast cancer (19.2% versus 8.8%, p = 0.004), breast cancer in the family (53.8% versus 38.3%, p = 0.009) and ovarian cancer in the family (7.6% versus 2.4%, p = 0.022) were higher than those of patients without the mutation. The median follow-up time was 92.78 months. Multivariate analysis showed that BRCA2 mutation was not associated with poorer iDFS [hazard ratio = 0.9, 95% confidence interval (CI) = 0.64-1.27, p = 0.56] and poorer distant relapse-free survival (DRFS) (hazard ratio = 1.09, 95% CI = 0.61-1.93, p = 0.76). There was no significant difference between the two groups with regard to metastatic patterns in the advanced disease setting. In the first-line metastatic breast cancer setting, PFS1 expression was broadly similar between the two groups irrespective of chemotherapy or endocrine therapy. Conclusion HR-positive/HER2-negative breast cancer with BRCA2 mutations differs from those without mutations in clinical behaviour and reflects more aggressive tumour behaviour. Our results indicate that BRCA2 mutations have no significant effect on the survival of Chinese women with HR-positive/HER2-negative breast cancer.
Collapse
Affiliation(s)
- Pu-Chun Li
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yi-Fan Zhu
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Jia-Ni Pan
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Qiao-Yan Zhu
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu-Yang Liao
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Xiao-Wen Ding
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Lin-Feng Zheng
- Department of Pathology, Zhejiang Cancer Hospital, 1 Banshan East Road, Hangzhou, Zhejiang 310022, China
| | - Wen-Ming Cao
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, 1 Banshan East Road, Gongsu, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
83
|
Chow RD, Parikh RB, Nathanson KL. Real-world evaluation of deep learning algorithms to classify functional pathogenic germline variants. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.05.24305402. [PMID: 38633773 PMCID: PMC11023677 DOI: 10.1101/2024.04.05.24305402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Deep learning models for variant pathogenicity prediction can recapitulate expert-curated annotations, but their performance remains unexplored on actual disease phenotypes in a real-world setting. Here, we apply three state-of-the-art pathogenicity prediction models to classify hereditary breast cancer gene variants in the UK Biobank. Predicted pathogenic variants in BRCA1, BRCA2 and PALB2, but not ATM and CHEK2, were associated with increased breast cancer risk. We explored gene-specific score thresholds for variant pathogenicity, finding that they could improve model performance. However, when specifically tasked with classifying variants of uncertain significance, the deep learning models were generally of limited clinical utility.
Collapse
Affiliation(s)
- Ryan D Chow
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Ravi B Parikh
- Division of Health Policy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Center for Cancer Care Innovation, Abramson Cancer Center, Philadelphia, PA, USA
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Katherine L Nathanson
- Basser Center for BRCA, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
84
|
Chiang J, Chua Z, Chan JY, Sule AA, Loke WH, Lum E, Ong MEH, Graves N, Ngeow J. Strategies to improve implementation of cascade testing in hereditary cancer syndromes: a systematic review. NPJ Genom Med 2024; 9:26. [PMID: 38570510 PMCID: PMC10991315 DOI: 10.1038/s41525-024-00412-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/13/2024] [Indexed: 04/05/2024] Open
Abstract
Hereditary cancer syndromes constitute approximately 10% of all cancers. Cascade testing involves testing of at-risk relatives to determine if they carry the familial pathogenic variant. Despite growing efforts targeted at improving cascade testing uptake, current literature continues to reflect poor rates of uptake, typically below 30%. This study aims to systematically review current literature on intervention strategies to improve cascade testing, assess the quality of intervention descriptions and evaluate the implementation outcomes of listed interventions. We searched major databases using keywords and subject heading of "cascade testing". Interventions proposed in each study were classified according to the Effective Practice and Organization of Care (EPOC) taxonomy. Quality of intervention description was assessed using the TIDieR checklist, and evaluation of implementation outcomes was performed using Proctor's Implementation Outcomes Framework. Improvements in rates of genetic testing uptake was seen in interventions across the different EPOC taxonomy strategies. The average TIDieR score was 7.3 out of 12. Items least reported include modifications (18.5%), plans to assess fidelity/adherence (7.4%) and actual assessment of fidelity/adherence (7.4%). An average of 2.9 out of 8 aspects of implementation outcomes were examined. The most poorly reported outcomes were cost, fidelity and sustainability, with only 3.7% of studies reporting them. Most interventions have demonstrated success in improving cascade testing uptake. Uptake of cascade testing was highest with delivery arrangement (68%). However, the quality of description of interventions and assessment of implementation outcomes are often suboptimal, hindering their replication and implementation downstream. Therefore, further adoption of standardized guidelines in reporting of interventions and formal assessment of implementation outcomes may help promote translation of these interventions into routine practice.
Collapse
Affiliation(s)
- Jianbang Chiang
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore, 169857, Singapore
| | - Ziyang Chua
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Jia Ying Chan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Ashita Ashish Sule
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Wan Hsein Loke
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Elaine Lum
- Health Services & Systems Research, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Marcus Eng Hock Ong
- Health Services & Systems Research, Duke-NUS Medical School, Singapore, 169857, Singapore
- Department of Emergency Medicine, Singapore General Hospital, Singapore, 169608, Singapore
| | - Nicholas Graves
- Health Services & Systems Research, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Joanne Ngeow
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore.
- Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore, 169857, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
| |
Collapse
|
85
|
Wu Z, Zhang Q, Jin Y, Zhang X, Chen Y, Yang C, Tang X, Jiang H, Wang X, Zhou X, Yu F, Wang B, Guan M. Population-based BRCA germline mutation screening in the Han Chinese identifies individuals at risk of BRCA mutation-related cancer: experience from a clinical diagnostic center from greater Shanghai area. BMC Cancer 2024; 24:411. [PMID: 38566028 PMCID: PMC10988807 DOI: 10.1186/s12885-024-12089-w] [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] [Received: 11/01/2023] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Deleterious BRCA1/2 (BRCA) mutation raises the risk for BRCA mutation-related malignancies, including breast, ovarian, prostate, and pancreatic cancer. Germline variation of BRCA exhibits substantial ethnical diversity. However, there is limited research on the Chinese Han population, constraining the development of strategies for BRCA mutation screening in this large ethnic group. METHODS We profile the BRCA mutational spectrum, including single nucleotide variation, insertion/deletion, and large genomic rearrangements in 2,080 apparently healthy Chinese Han individuals and 522 patients with BRCA mutation-related cancer, to determine the BRCA genetic background of the Chinese Han population, especially of the East Han. Incident cancer events were monitored in 1,005 participants from the healthy group, comprising 11 BRCA pathogenic/likely pathogenic (PLP) variant carriers and 994 PLP-free individuals, including 3 LGR carriers. RESULTS Healthy Chinese Han individuals demonstrated a distinct BRCA mutational spectrum compared to cancer patients, with a 0.53% (1 in 189) prevalence of pathogenic/likely pathogenic (PLP) variant, alongside a 3 in 2,080 occurrence of LGR. BRCA1 c. 5470_5477del demonstrated high prevalence (0.44%) in the North Han Chinese and penetrance for breast cancer. None of the 3 LGR carriers developed cancer during the follow-up. We calculated a relative risk of 135.55 (95% CI 25.07 to 732.88) for the development of BRCA mutation-related cancers in the BRCA PLP variant carriers (mean age 42.91 years, median follow-up 10 months) compared to PLP-free individuals (mean age 48.47 years, median follow-up 16 months). CONCLUSION The unique BRCA mutational profile in the Chinese Han highlights the potential for standardized population-based BRCA variant screening to enhance BRCA mutation-related cancer prevention and treatment.
Collapse
Affiliation(s)
- Zhiyuan Wu
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Qingyun Zhang
- Central Laboratory, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Yiting Jin
- Department of General Surgery, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Xinju Zhang
- Central Laboratory, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Yanli Chen
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Can Yang
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Xuemei Tang
- Central Laboratory, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Haowen Jiang
- Department of Urology, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Xiaoyi Wang
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Xinli Zhou
- Department of Oncology, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Feng Yu
- Health Management Center, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Bing Wang
- Health Management Center, Huashan Hospital, Fudan University, 200040, Shanghai, China.
| | - Ming Guan
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, 200040, Shanghai, China.
- Central Laboratory, Huashan Hospital, Fudan University, 200040, Shanghai, China.
| |
Collapse
|
86
|
Kotsopoulos J, Gronwald J, Huzarski T, Møller P, Pal T, McCuaig JM, Singer CF, Karlan BY, Aeilts A, Eng C, Eisen A, Bordeleau L, Foulkes WD, Tung N, Couch FJ, Fruscio R, Neuhausen SL, Zakalik D, Cybulski C, Metcalfe K, Olopade OI, Sun P, Lubinski J, Narod SA. Bilateral Oophorectomy and All-Cause Mortality in Women With BRCA1 and BRCA2 Sequence Variations. JAMA Oncol 2024; 10:484-492. [PMID: 38421677 PMCID: PMC10905374 DOI: 10.1001/jamaoncol.2023.6937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 08/22/2023] [Indexed: 03/02/2024]
Abstract
Importance Preventive bilateral salpingo-oophorectomy is offered to women at high risk of ovarian cancer who carry a pathogenic variant in BRCA1 or BRCA2; however, the association of oophorectomy with all-cause mortality has not been clearly defined. Objective To evaluate the association between bilateral oophorectomy and all-cause mortality among women with a BRCA1 or BRCA2 sequence variation. Design, Setting, and Participants In this international, longitudinal cohort study of women with BRCA sequence variations, information on bilateral oophorectomy was obtained via biennial questionnaire. Participants were women with a BRCA1 or BRCA2 sequence variation, no prior history of cancer, and at least 1 follow-up questionnaire completed. Women were followed up from age 35 to 75 years for incident cancers and deaths. Cox proportional hazards regression was used to estimate the hazard ratios (HRs) and 95% CIs for all-cause mortality associated with a bilateral oophorectomy (time dependent). Data analysis was performed from January 1 to June 1, 2023. Exposures Self-reported bilateral oophorectomy (with or without salpingectomy). Main Outcomes and Measures All-cause mortality, breast cancer-specific mortality, and ovarian cancer-specific mortality. Results There were 4332 women (mean age, 42.6 years) enrolled in the cohort, of whom 2932 (67.8%) chose to undergo a preventive oophorectomy at a mean (range) age of 45.4 (23.0-77.0) years. After a mean follow-up of 9.0 years, 851 women had developed cancer and 228 had died; 57 died of ovarian or fallopian tube cancer, 58 died of breast cancer, 16 died of peritoneal cancer, and 97 died of other causes. The age-adjusted HR for all-cause mortality associated with oophorectomy was 0.32 (95% CI, 0.24-0.42; P < .001). The age-adjusted HR was 0.28 (95% CI, 0.20-0.38; P < .001) and 0.43 (95% CI, 0.22-0.90; P = .03) for women with BRCA1 and BRCA2 sequence variations, respectively. For women with BRCA1 sequence variations, the estimated cumulative all-cause mortality to age 75 years for women who had an oophorectomy at age 35 years was 25%, compared to 62% for women who did not have an oophorectomy. For women with BRCA2 sequence variations, the estimated cumulative all-cause mortality to age 75 years was 14% for women who had an oophorectomy at age 35 years compared to 28% for women who did not have an oophorectomy. Conclusions and Relevance In this cohort study among women with a BRCA1 or BRCA2 sequence variation, oophorectomy was associated with a significant reduction in all-cause mortality.
Collapse
Affiliation(s)
- Joanne Kotsopoulos
- Women’s College Research Institute, University of Toronto, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Jacek Gronwald
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Tomasz Huzarski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Pål Møller
- Department of Tumour Biology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Tuya Pal
- Vanderbilt-Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jeanna M. McCuaig
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Christian F. Singer
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles
| | - Amber Aeilts
- Comprehensive Cancer Center, Division of Human Genetics, The Ohio State University Medical Center, Columbus
| | - Charis Eng
- Genomic Medicine Institute and Center for Personalized Genetic Healthcare, Cleveland Clinic, Cleveland, Ohio
| | - Andrea Eisen
- Sunnybrook Odette Cancer Center, Department of Medical Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Louise Bordeleau
- Department of Oncology, Juravinski Cancer Centre and McMaster University, Hamilton, Ontario, Canada
| | - William D. Foulkes
- McGill Program in Cancer Genetics, Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Nadine Tung
- Cancer Risk and Prevention Program, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Fergus J. Couch
- Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Robert Fruscio
- Clinic of Obstetrics and Gynecology, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Susan L. Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, California
| | - Dana Zakalik
- Grosfeld Cancer Genetics Center, Beaumont Health, Oakland University William Beaumont School of Medicine, Royal Oak, Michigan
| | - Cezary Cybulski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Kelly Metcalfe
- Women’s College Research Institute, University of Toronto, Toronto, Ontario, Canada
- Bloomberg School of Nursing, University of Toronto, Toronto, Ontario, Canada
| | | | - Ping Sun
- Women’s College Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Jan Lubinski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Steven A. Narod
- Women’s College Research Institute, University of Toronto, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
87
|
Yamada J, Fukuda K, Sugawara T, Makino K, Shimazu K, Yoshida T, Taguchi D, Shinozaki H, Terada Y, Nanjo H, Shibata H. A case of hereditary breast and ovarian cancer syndrome of initially presented as cancer of unknown primary with lymph node metastases unveiled by genetic analysis. Int Cancer Conf J 2024; 13:139-143. [PMID: 38524651 PMCID: PMC10957858 DOI: 10.1007/s13691-023-00652-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/27/2023] [Indexed: 03/26/2024] Open
Abstract
Cancer of unknown primary (CUP) is a heterogeneous disease concept involving various malignant tumors. Understanding its pathophysiology is often difficult, together with its treatment. Here, we present a case of CUP with abdominal lymph node enlargement and elevated carbohydrate antigen 125 levels. It initially resembled a favorable prognosis type similar to ovarian cancer, but metastases were observed in cervical lymph nodes, indicating a somewhat atypical CUP compared to the typical ovarian cancer-like CUP. We identified a germline Breast Cancer 1 (BRCA1) p.L63* variant through a family history inquiry and BRCA analysis, indicating hereditary breast and ovarian cancer syndrome. The patient achieved near-complete remission with platinum-based therapy followed by poly (ADP-ribose) polymerase (PARP) inhibitor. The variant has shown sensitivity in both clinical and pathogenic reports in the ClinVar database of the National Institutes of Health. No clinical studies reported on the efficacy of PARP inhibitors specific to this variant, but our case demonstrated the sensitivity of platinum-based therapy followed by PARP inhibitor. Reports of CUP in hereditary breast and ovarian cancer syndrome are very rare, with only a single report in the literature.
Collapse
Affiliation(s)
- Juri Yamada
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Hondo 1-1-1, Akita, Japan
| | - Koji Fukuda
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Tae Sugawara
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Kenichi Makino
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Kazuhiro Shimazu
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Hondo 1-1-1, Akita, Japan
| | - Taichi Yoshida
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Hondo 1-1-1, Akita, Japan
| | - Daiki Taguchi
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Hondo 1-1-1, Akita, Japan
| | - Hanae Shinozaki
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Hondo 1-1-1, Akita, Japan
| | - Yukihiro Terada
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Hiroshi Nanjo
- Department of Pathology, Akita University Hospital, Akita, Japan
| | - Hiroyuki Shibata
- Department of Clinical Oncology, Graduate School of Medicine, Akita University, Hondo 1-1-1, Akita, Japan
| |
Collapse
|
88
|
Pozzar RA, Seven M. Interventions to support decision making in people considering germline genetic testing for BRCA 1/2 pathogenic and likely pathogenic variants: A scoping review. J Genet Couns 2024; 33:392-401. [PMID: 37328917 DOI: 10.1002/jgc4.1738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/23/2023] [Accepted: 06/01/2023] [Indexed: 06/18/2023]
Abstract
Pathogenic and likely pathogenic variants in BRCA1 and BRCA2 (BRCA1/2) are medically actionable and may inform hereditary breast and ovarian cancer (HBOC) treatment and prevention. However, rates of germline genetic testing (GT) in people with and without cancer are suboptimal. Individuals' knowledge, attitudes, and beliefs may influence GT decisions. While genetic counseling (GC) provides decision support, the supply of genetic counselors is insufficient to meet demand. Accordingly, there is a need to explore the evidence on interventions that aim to support BRCA1/2 testing decisions. We conducted a scoping review of PubMed, CINAHL, Web of Science, and PsycINFO using search terms related to HBOC, GT, and decision making. First, we screened records to identify peer-reviewed reports that described interventions to support BRCA1/2 testing decisions. Next, we reviewed full-text reports and excluded studies that lacked statistical comparisons or enrolled previously tested individuals. Finally, we extracted study characteristics and findings into a table. All records and reports were reviewed independently by two authors; decisions were tracked in Rayyan, and discrepancies were resolved through discussion. Of 2116 unique citations, 25 met the eligibility criteria. Articles were published between 1997 and 2021 and described randomized trials and nonrandomized, quasi-experimental studies. Most studies tested technology-based (12/25, 48%) or written (9/25, 36%) interventions. Nearly half (12/25, 48%) of interventions were designed to complement traditional GC. Of the interventions compared to GC, 75% (6/8) increased or had a noninferior effect on knowledge, and 67% (4/6) decreased or had a noninferior effect on decisional conflict. Intervention effects on GT uptake were mixed, which may reflect evolving eligibility criteria for GT. Our findings suggest novel interventions may promote informed GT decision making, but many were developed to complement traditional GC. Trials that assess the effects of decision support interventions in diverse samples and evaluate implementation strategies for efficacious interventions are warranted.
Collapse
Affiliation(s)
- Rachel A Pozzar
- Phyllis F. Cantor Center for Research in Nursing and Patient Care Services, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Memnun Seven
- Elaine Marieb College of Nursing, University of Massachusetts, Amherst, Massachusetts, USA
| |
Collapse
|
89
|
Davey MG, Tormey S. Implications of Highly Penetrant Genetic Variants on Breast Surgery. Clin Breast Cancer 2024; 24:180-183. [PMID: 38218718 DOI: 10.1016/j.clbc.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/15/2024]
Abstract
It seems the most probable beneficiaries from the molecular era are those harboring hereditary genetic variants, which are responsible for 5% to 10% of all breast cancer diagnoses. There are several key implications of such variants on clinical practice, from expedited anticipation of primary cancer diagnoses, which can have their risk mitigated by risk reduction surgery, to pragmatism surrounding the management of male breast cancer patients. This communication discusses the implications of highly penetrant (or pathogenic) hereditary variants in contemporary breast surgery practice.
Collapse
Affiliation(s)
- Matthew G Davey
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Breast Surgery, University Hospital Limerick, Limerick, Ireland.
| | - Shona Tormey
- Department of Breast Surgery, University Hospital Limerick, Limerick, Ireland
| |
Collapse
|
90
|
Li R, Zhao X, Huang Y, Li C, Liu L, Wang M, Wang J, Song Z. The Survival Benefit of Pegylated Liposomal Doxorubicin-Based Neoadjuvant Chemotherapy in the Management of Breast Cancer. Cancer Biother Radiopharm 2024. [PMID: 38512710 DOI: 10.1089/cbr.2024.0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024] Open
Abstract
Purpose: This study aims to evaluate the short-term outcomes and prognosis and the cardiac safety of pegylated liposomal doxorubicin (PLD)-based neoadjuvant chemotherapy (NAC) compared with epirubicin-based therapy in breast cancer treatment. Methods: In total, 304 patients diagnosed with stages II and III breast cancer were enrolled that included 97 cases treated with PLD and 207 controls treated with epirubicin in NAC. The effectiveness of the antibreast cancer treatment was evaluated using overall survival (OS) and disease-free survival (DFS) metrics, whereas cardiac toxicity was measured through the left ventricular ejection fraction (LVEF) and electrocardiogram (ECG) assessments. Results: The 5-year DFS and OS rates in the PLD group were 84.5% and 88.7% (with 15 recurrences and 11 deaths), respectively, whereas in the control group, these rates were 72.9% and 79.2% (with 56 recurrences and 43 deaths). Regarding cardiac toxicity, there was no significant difference in ECG abnormalities or LVEF decline between the two groups. Conclusions: The study suggests that PLD-based NAC may provide substantial benefits in terms of DFS and OS, along with a safe cardiac toxicity profile, in patients with stage II-III breast cancer.
Collapse
Affiliation(s)
- Ruoyang Li
- Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xuewei Zhao
- Department of Medicine, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yunfei Huang
- Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chunxiao Li
- Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Liu
- Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Meiqi Wang
- Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiaxing Wang
- Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhenchuan Song
- Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
91
|
Sideris M, Menon U, Manchanda R. Screening and prevention of ovarian cancer. Med J Aust 2024; 220:264-274. [PMID: 38353066 DOI: 10.5694/mja2.52227] [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] [Received: 02/27/2023] [Accepted: 12/11/2023] [Indexed: 03/07/2024]
Abstract
Ovarian cancer remains the most lethal gynaecological malignancy with 314 000 cases and 207 000 deaths annually worldwide. Ovarian cancer cases and deaths are predicted to increase in Australia by 42% and 55% respectively by 2040. Earlier detection and significant downstaging of ovarian cancer have been demonstrated with multimodal screening in the largest randomised controlled trial of ovarian cancer screening in women at average population risk. However, none of the randomised trials have demonstrated a mortality benefit. Therefore, ovarian cancer screening is not currently recommended in women at average population risk. More frequent surveillance for ovarian cancer every three to four months in women at high risk has shown good performance characteristics and significant downstaging, but there is no available information on a survival benefit. Population testing offers an emerging novel strategy to identify women at high risk who can benefit from ovarian cancer prevention. Novel multicancer early detection biomarker, longitudinal multiple marker strategies, and new biomarkers are being investigated and evaluated for ovarian cancer screening. Risk-reducing salpingo-oophorectomy (RRSO) decreases ovarian cancer incidence and mortality and is recommended for women at over a 4-5% lifetime risk of ovarian cancer. Pre-menopausal women without contraindications to hormone replacement therapy (HRT) undergoing RRSO should be offered HRT until 51 years of age to minimise the detrimental consequences of premature menopause. Currently risk-reducing early salpingectomy and delayed oophorectomy (RRESDO) should only be offered to women at increased risk of ovarian cancer within the context of a research trial. Pre-menopausal early salpingectomy is associated with fewer menopausal symptoms and better sexual function than bilateral salpingo-oophorectomy. A Sectioning and Extensively Examining the Fimbria (SEE-FIM) protocol should be used for histopathological assessment in women at high risk of ovarian cancer who are undergoing surgical prevention. Opportunistic salpingectomy may be offered at routine gynaecological surgery to all women who have completed their family. Long term prospective opportunistic salpingectomy studies are needed to determine the effect size of ovarian cancer risk reduction and the impact on menopause.
Collapse
Affiliation(s)
- Michail Sideris
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Usha Menon
- Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Ranjit Manchanda
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- Institute of Clinical Trials and Methodology, University College London, London, UK
- Barts Health NHS Trust, London, UK
- London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
92
|
Hickey M, Basu P, Sassarini J, Stegmann ME, Weiderpass E, Nakawala Chilowa K, Yip CH, Partridge AH, Brennan DJ. Managing menopause after cancer. Lancet 2024; 403:984-996. [PMID: 38458217 DOI: 10.1016/s0140-6736(23)02802-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/18/2023] [Accepted: 12/11/2023] [Indexed: 03/10/2024]
Abstract
Globally, 9 million women are diagnosed with cancer each year. Breast cancer is the most commonly diagnosed cancer worldwide, followed by colorectal cancer in high-income countries and cervical cancer in low-income countries. Survival from cancer is improving and more women are experiencing long-term effects of cancer treatment, such as premature ovarian insufficiency or early menopause. Managing menopausal symptoms after cancer can be challenging, and more severe than at natural menopause. Menopausal symptoms can extend beyond hot flushes and night sweats (vasomotor symptoms). Treatment-induced symptoms might include sexual dysfunction and impairment of sleep, mood, and quality of life. In the long term, premature ovarian insufficiency might increase the risk of chronic conditions such as osteoporosis and cardiovascular disease. Diagnosing menopause after cancer can be challenging as menopausal symptoms can overlap with other common symptoms in patients with cancer, such as fatigue and sexual dysfunction. Menopausal hormone therapy is an effective treatment for vasomotor symptoms and seems to be safe for many patients with cancer. When hormone therapy is contraindicated or avoided, emerging evidence supports the efficacy of non-pharmacological and non-hormonal treatments, although most evidence is based on women older than 50 years with breast cancer. Vaginal oestrogen seems safe for most patients with genitourinary symptoms, but there are few non-hormonal options. Many patients have inadequate centralised care for managing menopausal symptoms after cancer treatment, and more information is needed about cost-effective and patient-focused models of care for this growing population.
Collapse
Affiliation(s)
- Martha Hickey
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne and the Royal Women's Hospital, Melbourne, VIC, Australia.
| | - Partha Basu
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, WHO, Lyon, France
| | - Jenifer Sassarini
- Department of Obstetrics and Gynaecology, School of Gynaecology, University of Glasgow, Glasgow, UK
| | - Mariken E Stegmann
- Department of Primary and Long-term Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | | | - Cheng-Har Yip
- Department of Surgery, University of Malaya, Kuala Lumpur, Malaysia
| | - Ann H Partridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Donal J Brennan
- Gynaecological Oncology Group, UCD School of Medicine, Mater Misericordiae University Hospital, Dublin, Ireland; Systems Biology Ireland, UCD School of Medicine, Dublin, Ireland
| |
Collapse
|
93
|
Snow S, Brezden-Masley C, Carter MD, Dhani N, Macaulay C, Ramjeesingh R, Raphael MJ, Slovinec D’Angelo M, Servidio-Italiano F. Barriers and Unequal Access to Timely Molecular Testing Results: Addressing the Inequities in Cancer Care Delays across Canada. Curr Oncol 2024; 31:1359-1375. [PMID: 38534936 PMCID: PMC10969404 DOI: 10.3390/curroncol31030103] [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] [Received: 01/30/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 05/26/2024] Open
Abstract
Genomic medicine is a powerful tool to improve diagnosis and outcomes for cancer patients by facilitating the delivery of the right drug at the right dose at the right time for the right patient. In 2023, a Canadian conference brought together leaders with expertise in different tumor types. The objective was to identify challenges and opportunities for change in terms of equitable and timely access to biomarker testing and reporting at the education, delivery, laboratory, patient, and health-system levels in Canada. Challenges identified included: limited patient and clinician awareness of genomic medicine options with need for formal education strategies; failure by clinicians to discuss genomic medicine with patients; delays in or no access to hereditary testing; lack of timely reporting of results; intra- and inter-provincial disparities in access; lack of funding for patients to access testing and for laboratories to provide testing; lack of standardized testing; and impact of social determinants of health. Canada must standardize its approach to biomarker testing across the country, with a view to addressing current inequities, and prioritize access to advanced molecular testing to ensure systems are in place to quickly bring innovation and evidence-based treatments to Canadian cancer patients, regardless of their place of residence or socioeconomic status.
Collapse
Affiliation(s)
- Stephanie Snow
- Division of Medical Oncology, QEII Health Sciences Centre, Halifax, NS B3H 2Y9, Canada
| | | | - Michael D. Carter
- Division of Anatomical Pathology, QEII Health Sciences Centre, Halifax, NS B3H 2Y9, Canada
| | - Neesha Dhani
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Cassandra Macaulay
- Colorectal Cancer Resource & Action Network (CCRAN), Toronto, ON M4W 3E2, Canada
| | - Ravi Ramjeesingh
- Division of Medical Oncology, QEII Health Sciences Centre, Halifax, NS B3H 2Y9, Canada
| | - Michael J. Raphael
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
| | | | | |
Collapse
|
94
|
Kamaraju S, Conroy M, Harris A, Georgen M, Min H, Powell M, Kurzrock R. Challenges to genetic testing for germline mutations associated with breast cancer among African Americans. Cancer Treat Rev 2024; 124:102695. [PMID: 38325071 DOI: 10.1016/j.ctrv.2024.102695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
Inequities in preventive cancer screening, diagnosis, treatment, and inferior cancer outcomes continue to pose challenges across the cancer continuum. While the exact reasons for these inferior outcomes are unknown, multiple barriers to various domains of social determinants of health (SDOH) play a vital role, leading to inequities in cancer care. These include barriers to transportation, housing, and food insecurities, contributing to delays in preventive screening and treatment. Furthermore, aggressive biologies also exist across various racial profiles with accompanying germline mutations. For example, African Americans (AAs) have a higher incidence of triple-negative breast cancer subtype and a high prevalence of BRCA1/2 gene mutations, increasing the risk of multiple cancers, warranting high-risk screening for these populations. Unfortunately, other barriers, such as financial insecurities, low health literacy rates, and lack of awareness, lead to delays in cancer screening and genetic testing, even with available high-risk screening and risk reduction procedures. In addition, physicians receive minimal interdisciplinary training to address genetic assessment, interpretation of the results, and almost no additional training in addressing the unique needs of racial minorities, leading to suboptimal delivery of genetic assessment provision resources among AAs. In this review, we discuss the confluence of factors and barriers limiting genetic testing among AAs and highlight the prevalence of germline mutations associated with increased risk of breast cancer among AAs, reflecting the need for multi-panel germline testing as well as education regarding hereditary cancer risks in underserved minorities.
Collapse
Affiliation(s)
- S Kamaraju
- Medical College of Wisconsin, Milwaukee, WI, USA; Department of Medicine, Division of Hematology-Oncology, Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA.
| | - M Conroy
- Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA
| | - A Harris
- Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA
| | - M Georgen
- Medical College of Wisconsin, Milwaukee, WI, USA; Department of Medicine, Division of Hematology-Oncology, Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA
| | - H Min
- Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA
| | - M Powell
- Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA
| | - R Kurzrock
- Medical College of Wisconsin, Milwaukee, WI, USA; Department of Medicine, Division of Hematology-Oncology, Medical College of Wisconsin, Milwaukee, WI, USA; Froedtert Hospital, Milwaukee, WI, USA
| |
Collapse
|
95
|
Allen CG, Donahue C, Coen E, Meeder K, Wallace K, Melvin C, Neelon B, Hughes K. Implementation Mapping for Managing Patients at High Risk for Hereditary Cancer. Am J Prev Med 2024; 66:503-515. [PMID: 37806365 PMCID: PMC10922485 DOI: 10.1016/j.amepre.2023.09.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/29/2023] [Accepted: 09/29/2023] [Indexed: 10/10/2023]
Abstract
INTRODUCTION Currently, no standard workflow exists for managing patients with pathogenic variants that put them at higher risk for hereditary cancers. Therefore, follow-up care for individuals with pathogenic variants is logistically challenging and results in poor guideline adherence. To address this challenge, authors created clinical management strategies for individuals identified at high risk for hereditary cancers. METHODS An implementation mapping approach was used to develop and evaluate the establishment of a Hereditary Cancer Clinic at the Medical University of South Carolina throughout in 2022. This approach consisted of 5 steps: conduct a needs assessment, identify objectives, select implementation strategies, produce implementation protocols, and develop an evaluation plan. The needs assessment consisted of qualitative interviews with patients (n=11), specialists (n=9), and members of the implementation team (n=4). Interviews were coded using the Consolidated Framework for Implementation Research to identify barriers and facilitators to establishment of the Hereditary Cancer Clinic. Objectives were identified, and then the team selected implementation strategies and produced implementation protocols to address concerns identified during the needs assessment. Authors conducted a second round of patient interviews to assess patient education materials. RESULTS The research team developed a long-term evaluation plan to guide future assessment of implementation, service, and clinical/patient outcomes. CONCLUSIONS This approach provides the opportunity for real-time enhancements and impact, with strategies for care specialists, patients, and implementation teams. Findings support ongoing efforts to improve patient management and outcomes while providing an opportunity for long-term evaluation of implementation strategies and guidelines for patients at high risk for hereditary cancers.
Collapse
Affiliation(s)
- Caitlin G Allen
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston, South Carolina.
| | - Colleen Donahue
- Department of Surgery, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Emma Coen
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Kiersten Meeder
- Division of Oncologic and Endocrine Surgery, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Kristin Wallace
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Cathy Melvin
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Brian Neelon
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Kevin Hughes
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
96
|
Taris N, Luporsi E, Osada M, Thiblet M, Mathelin C. [News in breast oncology genetics for female and male population]. GYNECOLOGIE, OBSTETRIQUE, FERTILITE & SENOLOGIE 2024; 52:149-157. [PMID: 38190969 DOI: 10.1016/j.gofs.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 12/30/2023] [Indexed: 01/10/2024]
Abstract
OBJECTIVES Breast oncology genetics emerged almost 30 years ago with the discovery of the BRCA1 and BRCA2 genes. The evolution of analytical practices has progressively allowed access to tests whose results now have a considerable impact on the management of both female and male breast cancers. The Sénologie commission of the Collège national des gynécologues et obstétriciens français (CNGOF) asked five specialists in breast surgery, oncology and oncological genetics to draw up a summary of the oncogenetic testing criteria used and the clinical implications for the female and male population of the test results, with or without an identified causal variant. In the case of proven genetic risk, surveillance, risk-reduction strategies, and the specificities of surgical and medical management (with PARP inhibitors in particular) were updated. METHODS This summary was based on national and international guidelines on the monitoring and therapeutic management of genetic risk, and a recent review of the literature covering the last five years. RESULTS Despite successive technical developments, the probability of identifying a causal variant in a situation suggestive of a predisposition to breast and ovarian cancer remains around 10% in France. The risk of breast cancer in women with a causal variant of the BRCA1, BRCA2, PALB2, TP53, CDH1 and PTEN genes is estimated at between 35% and 85% at age 70. The presence of a causal variant in one of these genes is the subject of different recommendations for men and women, concerning both surveillance, the age of onset and imaging modalities of which vary according to the genes involved, and risk-reduction surgery, which is possible for women as soon as their risk level exceeds 30% and remains exceptionally indicated for men. In the case of breast cancer, PARP inhibitors are a promising new class of treatment for BRCA germline mutations. CONCLUSION A discipline resolutely focused on understanding molecular mechanisms, screening and preventive medicine/surgery, oncology genetics is currently also involved in new medical/surgical approaches, the long-term benefits/risks of which will need to be monitored.
Collapse
Affiliation(s)
- Nicolas Taris
- Unité de génétique oncologique, ICANS, avenue Albert-Calmette, 67200 Strasbourg, France.
| | - Elisabeth Luporsi
- Service de génétique, hôpital Femme-Mère-Enfant, CHR de Metz-Thionville, Site de Mercy, 1, allée du Château, 57085 Metz cedex, France.
| | - Marine Osada
- Service de chirurgie, ICANS, avenue Albert-Calmette, 67200 Strasbourg, France; CHRU, avenue Molière, 67200 Strasbourg, France.
| | - Marie Thiblet
- Service de chirurgie, ICANS, avenue Albert-Calmette, 67200 Strasbourg, France; CHRU, avenue Molière, 67200 Strasbourg, France.
| | - Carole Mathelin
- Service de chirurgie, ICANS, avenue Albert-Calmette, 67200 Strasbourg, France; CHRU, avenue Molière, 67200 Strasbourg, France.
| |
Collapse
|
97
|
Deng T, Liang J, Yan C, Ni M, Xiang H, Li C, Ou J, Lin Q, Liu L, Tang G, Luo R, An X, Gao Y, Lin X. Development and validation of ultrasound-based radiomics model to predict germline BRCA mutations in patients with breast cancer. Cancer Imaging 2024; 24:31. [PMID: 38424620 PMCID: PMC10905812 DOI: 10.1186/s40644-024-00676-w] [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] [Received: 07/05/2023] [Accepted: 02/20/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Identifying breast cancer (BC) patients with germline breast cancer susceptibility gene (gBRCA) mutation is important. The current criteria for germline testing for BC remain controversial. This study aimed to develop a nomogram incorporating ultrasound radiomic features and clinicopathological factors to predict gBRCA mutations in patients with BC. MATERIALS AND METHODS In this retrospective study, 497 women with BC who underwent gBRCA genetic testing from March 2013 to May 2022 were included, including 348 for training (84 with and 264 without a gBRCA mutation) and 149 for validation(36 patients with and 113 without a gBRCA mutation). Factors associated with gBRCA mutations were identified to establish a clinicopathological model. Radiomics features were extracted from the intratumoral and peritumoral regions (3 mm and 5 mm) of each image. The least absolute shrinkage and selection operator regression algorithm was used to select the features and logistic regression analysis was used to construct three imaging models. Finally, a nomogram that combined clinicopathological and radiomics features was developed. The models were evaluated based on the area under the receiver operating characteristic curve (AUC), calibration, and clinical usefulness. RESULTS Age at diagnosis, family history of BC, personal history of other BRCA-related cancers, and human epidermal growth factor receptor 2 status were independent predictors of the clinicopathological model. The AUC of the imaging radiomics model combining intratumoral and peritumoral 3 mm areas in the validation set was 0.783 (95% confidence interval [CI]: 0.702-0.862), which showed the best performance among three imaging models. The nomogram yielded better performance than the clinicopathological model in validation sets (AUC: 0.824 [0.755-0.894] versus 0.659 [0.563-0.755], p = 0.007). CONCLUSION The nomogram based on ultrasound images and clinicopathological factors performs well in predicting gBRCA mutations in BC patients and may help to improve clinical decisions about genetic testing.
Collapse
Affiliation(s)
- Tingting Deng
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Jianwen Liang
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, China
| | - Cuiju Yan
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Mengqian Ni
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Huiling Xiang
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Chunyan Li
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Jinjing Ou
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Qingguang Lin
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Lixian Liu
- Department of Ultrasound, Guangdong Second Provincial General Hospital, Guangzhou, 510060, China
| | - Guoxue Tang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Rongzhen Luo
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xin An
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yi Gao
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518000, China.
| | - Xi Lin
- Department of Ultrasound, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
98
|
Roth S, Owczarzak J, Baker K, Davidson H, Jamal L. Experiences of hereditary cancer care among transgender and gender diverse people: "It's gender. It's cancer risk…it's everything". J Genet Couns 2024:10.1002/jgc4.1867. [PMID: 38342966 PMCID: PMC11316848 DOI: 10.1002/jgc4.1867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 12/15/2023] [Accepted: 12/27/2023] [Indexed: 02/13/2024]
Abstract
Transgender and gender diverse (TGD) individuals are a significant yet underrepresented population within genetic counseling research and broader LGBTQI+ health studies. This underrepresentation perpetuates a cycle of exclusion from the production of medical knowledge, impacting the quality and equity of care received by TGD individuals. This issue is particularly poignant in cancer genetic counseling, where TGD individuals with elevated cancer risk receive risk assessment, counseling, and referral to support based on risk figures and standards of care developed for cisgender individuals. The experiences of TGD individuals navigating inherited cancer syndromes remain largely undocumented in medical literature, posing challenges to the provision of inclusive care by genetics providers. To bridge this knowledge gap, we conducted a cross-sectional qualitative study. Nineteen semi-structured interviews were held with gender diverse adults having hereditary cancer syndromes, family histories of such syndromes, or personal histories of chest cancer. Our study employed thematic analysis using combined inductive and deductive methods to illuminate how hereditary cancer care intersects with participants' gender identities, gender expression, and gender-affirming care experiences. Participants reflected on care experiences that felt affirming or triggered gender dysphoria. Participants also discussed the interplay between risk-reducing mastectomy and top surgery, exploring co-emergent dynamics between cancer risk management and gender expression. Significantly, participants identified actionable strategies for healthcare providers to enhance support for gender diverse patients, including the mindful use of gendered language, collaborative decision-making, and conveying allyship. These findings offer valuable insights into tailoring genetic counseling to meet the unique needs of TGD individuals, advancing the path toward inclusive and appropriate care for LGBTQI+ individuals with hereditary cancer syndromes.
Collapse
Affiliation(s)
- Sarah Roth
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Center for Precision Health Research, NHGRI, NIH, Bethesda, Maryland, USA
| | - Jill Owczarzak
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kellan Baker
- Whitman-Walker Health, Washington, District of Columbia, USA
| | - Hannah Davidson
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Center for Precision Health Research, NHGRI, NIH, Bethesda, Maryland, USA
| | - Leila Jamal
- Department of Bioethics, NIH, Bethesda, Maryland, USA
- Center for Cancer Research, NCI, NIH, Bethesda, Maryland, USA
| |
Collapse
|
99
|
Bedrosian I, Somerfield MR, Achatz MI, Boughey JC, Curigliano G, Friedman S, Kohlmann WK, Kurian AW, Laronga C, Lynce F, Norquist BS, Plichta JK, Rodriguez P, Shah PD, Tischkowitz M, Wood M, Yadav S, Yao K, Robson ME. Germline Testing in Patients With Breast Cancer: ASCO-Society of Surgical Oncology Guideline. J Clin Oncol 2024; 42:584-604. [PMID: 38175972 DOI: 10.1200/jco.23.02225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 10/18/2023] [Indexed: 01/06/2024] Open
Abstract
PURPOSE To develop recommendations for germline mutation testing for patients with breast cancer. METHODS An ASCO-Society of Surgical Oncology (SSO) panel convened to develop recommendations based on a systematic review and formal consensus process. RESULTS Forty-seven articles met eligibility criteria for the germline mutation testing recommendations; 18 for the genetic counseling recommendations. RECOMMENDATIONS BRCA1/2 mutation testing should be offered to all newly diagnosed patients with breast cancer ≤65 years and select patients >65 years based on personal history, family history, ancestry, or eligibility for poly(ADP-ribose) polymerase (PARP) inhibitor therapy. All patients with recurrent breast cancer who are candidates for PARP inhibitor therapy should be offered BRCA1/2 testing, regardless of family history. BRCA1/2 testing should be offered to women who develop a second primary cancer in the ipsilateral or contralateral breast. For patients with prior history of breast cancer and without active disease, testing should be offered to patients diagnosed ≤65 years and selectively in patients diagnosed after 65 years, if it will inform personal and family risk. Testing for high-penetrance cancer susceptibility genes beyond BRCA1/2 should be offered to those with supportive family histories; testing for moderate-penetrance genes may be offered if necessary to inform personal and family cancer risk. Patients should be provided enough pretest information for informed consent; those with pathogenic variants should receive individualized post-test counseling. Variants of uncertain significance should not impact management, and patients with such variants should be followed for reclassification. Referral to providers experienced in clinical cancer genetics may help facilitate patient selection and interpretation of expanded testing, and provide counseling of individuals without pathogenic germline variants but with significant family history.Additional information is available at www.asco.org/breast-cancer-guidelines.
Collapse
Affiliation(s)
| | | | | | | | - Giuseppe Curigliano
- University of Milan, Italy
- European Institute of Oncology, IRCCS, Milano, Italy
| | - Sue Friedman
- FORCE (Facing Our Risk of Cancer Empowered), Tampa, FL
| | - Wendy K Kohlmann
- University of Utah Huntsman Cancer Institute, Salt Lake City, UT
| | | | | | | | | | | | - Patricia Rodriguez
- Hereditary Cancer Risk Assessment Program, Virginia Cancer Specialists, Arlington, VA
| | - Payal D Shah
- Basser Center for BRCA & Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | | | | | - Katherine Yao
- Division of Surgical Oncology at NorthShore University Health System, Evanston, IL
| | - Mark E Robson
- Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
100
|
Guo F, Adekanmbi V, Hsu CD, Berenson AB, Kuo YF, Shih YCT. Cost-Effectiveness of Population-Based Multigene Testing for Breast and Ovarian Cancer Prevention. JAMA Netw Open 2024; 7:e2356078. [PMID: 38353949 PMCID: PMC10867683 DOI: 10.1001/jamanetworkopen.2023.56078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 12/21/2023] [Indexed: 02/16/2024] Open
Abstract
Importance The current method of BRCA testing for breast and ovarian cancer prevention, which is based on family history, often fails to identify many carriers of pathogenic variants. Population-based genetic testing offers a transformative approach in cancer prevention by allowing for proactive identification of any high-risk individuals and enabling early interventions. Objective To assess the lifetime incremental effectiveness, costs, and cost-effectiveness of population-based multigene testing vs family history-based testing. Design, Setting, and Participants This economic evaluation used a microsimulation model to assess the cost-effectiveness of multigene testing (BRCA1, BRCA2, and PALB2) for all women aged 30 to 35 years compared with the current standard of care that is family history based. Carriers of pathogenic variants were offered interventions, such as magnetic resonance imaging with or without mammography, chemoprevention, or risk-reducing mastectomy and salpingo-oophorectomy, to reduce cancer risk. A total of 2000 simulations were run on 1 000 000 women, using a lifetime time horizon and payer perspective, and costs were adjusted to 2022 US dollars. This study was conducted from September 1, 2020, to December 15, 2023. Main Outcomes and Measures The main outcome measure was the incremental cost-effectiveness ratio (ICER), quantified as cost per quality-adjusted life-year (QALY) gained. Secondary outcomes included incremental cost, additional breast and ovarian cancer cases prevented, and excess deaths due to coronary heart disease (CHD). Results The study assessed 1 000 000 simulated women aged 30 to 35 years in the US. In the base case, population-based multigene testing was more cost-effective compared with family history-based testing, with an ICER of $55 548 per QALY (95% CI, $47 288-$65 850 per QALY). Population-based multigene testing would be able to prevent an additional 1338 cases of breast cancer and 663 cases of ovarian cancer, but it would also result in 69 cases of excess CHD and 10 excess CHD deaths per million women. The probabilistic sensitivity analyses show that the probability that population-based multigene testing is cost-effective was 100%. When the cost of the multigene test exceeded $825, population-based testing was no longer cost-effective (ICER, $100 005 per QALY; 95% CI, $87 601-$11 6323). Conclusions and Relevance In this economic analysis of population-based multigene testing, population-based testing was a more cost-effective strategy for the prevention of breast cancer and ovarian cancer when compared with the current family history-based testing strategy at the $100 000 per QALY willingness-to-pay threshold. These findings support the need for more comprehensive genetic testing strategies to identify pathogenic variant carriers and enable informed decision-making for personalized risk management.
Collapse
Affiliation(s)
- Fangjian Guo
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston
- Center for Interdisciplinary Research in Women’s Health, The University of Texas Medical Branch at Galveston, Galveston
| | - Victor Adekanmbi
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston
- Center for Interdisciplinary Research in Women’s Health, The University of Texas Medical Branch at Galveston, Galveston
| | - Christine D. Hsu
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston
- Center for Interdisciplinary Research in Women’s Health, The University of Texas Medical Branch at Galveston, Galveston
| | - Abbey B. Berenson
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston
- Center for Interdisciplinary Research in Women’s Health, The University of Texas Medical Branch at Galveston, Galveston
| | - Yong-Fang Kuo
- Center for Interdisciplinary Research in Women’s Health, The University of Texas Medical Branch at Galveston, Galveston
- Department of Biostatistics and Data Science, The University of Texas Medical Branch at Galveston, Galveston
- Office of Biostatistics, University of Texas Medical Branch at Galveston, Galveston
| | - Ya-Chen Tina Shih
- Program in Cancer Health Economics Research, Jonsson Comprehensive Cancer Center, and Department of Radiation Oncology, School of Medicine, University of California, Los Angeles
| |
Collapse
|