1
|
Abubakar M, Duggan MA, Fan S, Pfeiffer RM, Lawrence S, Mutreja K, Klein A, Koka H, Ahearn TU, Henry JE, Sprague BL, Vacek PM, Weaver DL, Richert-Boe K, Kimes TM, Titiloye N, Edusei L, Figueroa JD, Yang XR, Garcia-Closas M, Rohan TE, Gierach GL. Unraveling the role of stromal disruption in aggressive breast cancer etiology and outcomes. J Natl Cancer Inst 2025:djaf070. [PMID: 40366376 DOI: 10.1093/jnci/djaf070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/01/2024] [Accepted: 03/05/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Aggressive (typically high-grade) breast cancers (BCs) remain major contributors to BC-related mortality globally. The tissue changes underpinning their etiology and outcomes, however, remain poorly characterized. METHODS Spatially resolved machine-learning algorithms were used to characterize "stromal disruption" as a morphological metric of reduced/altered extracellular matrix and increased immune, inflammatory, and/or wound response-related processes in normal, benign breast disease (BBD), and invasive hematoxylin and eosin (H&E)-stained breast tissues. Associations of stromal disruption with BC etiologic factors were assessed among 4023 healthy breast tissue donors, its impact on BC incidence was assessed among 974 BBD patients in a nested case-control study, while its prognostic associations were assessed in 4 BC patient cohorts (n = 4223). RESULTS Epidemiologic risk factors for aggressive BC, including younger age, multiparity, Black race, obesity, and family history, demonstrated strong associations with increasing stromal disruption in H&E sections prior to tumor development. Substantial stromal disruption in BBD H&E was associated with ∼4-fold increased risk of aggressive (high-grade) BC and ∼3 years shorter latency from BBD to BC diagnosis, independently of BBD histology. Across BC cohorts, stromal disruption in H&E was associated with aggressive (mostly high-grade) tumor phenotypes and with markedly poor prognosis among ER-positive patients, irrespective of histology. The immunobiology of stromal disruption reflected heightened innate (CD68+), adaptive (CD3+CD4+, CD3+CD8+), immunoregulatory (CD3+CD4+FOXP3+), immune escape (PD1+PDL1+), endothelial (CD31+), and myofibroblast (α-SMA+) marker expression. CONCLUSION Our findings highlight the active stromal role in aggressive BC etiology and outcomes, opening possibilities for readily identifying high-risk women across the BC continuum that may benefit from stroma-centric preventative or therapeutic strategies.
Collapse
Affiliation(s)
- Mustapha Abubakar
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Bethesda, MD, United States
| | - Máire A Duggan
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Shaoqi Fan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Bethesda, MD, United States
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Bethesda, MD, United States
| | - Scott Lawrence
- Molecular and Digital Pathology Laboratory, Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Karun Mutreja
- Molecular and Digital Pathology Laboratory, Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Alyssa Klein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Bethesda, MD, United States
| | - Hela Koka
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Bethesda, MD, United States
| | - Thomas U Ahearn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Bethesda, MD, United States
| | - Jill E Henry
- Biospecimen Collection and Banking Core, Susan G. Komen Tissue Bank at the IU Simon Comprehensive Cancer Center, Indianapolis, IN, United States
| | - Brian L Sprague
- University of Vermont College of Medicine and Vermont Cancer Center, Burlington, VT, United States
| | - Pamela M Vacek
- University of Vermont College of Medicine and Vermont Cancer Center, Burlington, VT, United States
| | - Donald L Weaver
- University of Vermont College of Medicine and Vermont Cancer Center, Burlington, VT, United States
| | | | - Teresa M Kimes
- Kaiser Permanente Center for Health Research, Portland, OR, United States
| | | | | | - Jonine D Figueroa
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Bethesda, MD, United States
| | - Xiaohong R Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Bethesda, MD, United States
| | - Montserrat Garcia-Closas
- Division of Cancer Genetics and Epidemiology, Institute of Cancer Research London, Sutton, United Kingdom
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Gretchen L Gierach
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
2
|
Spijkervet JRJAH, Lanjouw L, Berger LPV, Dorrius MD, van der Vegt B, de Bock GH. Clinicopathological Characteristics of Ovarian and Breast Cancer in PALB2, RAD51C, and RAD51D Germline Pathogenic Variant Carriers. Genes (Basel) 2025; 16:556. [PMID: 40428378 PMCID: PMC12111020 DOI: 10.3390/genes16050556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Germline pathogenic variants (GPVs) in PALB2, RAD51C, and RAD51D increase breast cancer (BC) and ovarian cancer (OC) risk. Limited data on clinicopathological characteristics of BC and OC in women with these GPVs hamper guideline development. Therefore, this study aims to describe these characteristics in a consecutive series of female PALB2, RAD51C, and RAD51D GPV carriers. Methods: Women with a PALB2, RAD51C, or RAD51D GPV determined before July 2023 at the University Medical Center Groningen were included. Cancer diagnoses were obtained through linkage with the Dutch Nationwide Pathology Databank (Palga). Median onset age and histopathological subtypes were compared to the data of The Netherlands Cancer Registry (NCR). Results: Among 164 GPV carriers (125 PALB2, 30 RAD51C, and 9 RAD51D), 54 BC and 6 OC cases were identified. The median BC onset age was 52 (n = 50), 71 (n = 3), and 43 years (n = 1) for PALB2, RAD51C, and RAD51D, respectively, compared with 62 years in the NCR. No BC histological subtype differences were observed in PALB2 carriers. The populations of RAD51C and RAD51D carriers were too small to compare to NCR data. No OC cases occurred in PALB2 carriers. The median OC onset age was 66 (n = 4) and 56 years (n = 2) for RAD51C and RAD51D carriers, respectively, versus 67 years in the NCR. All RAD51D carriers had high-grade serous carcinoma, compared to 51.5% in the NCR. Conclusions: Differences in onset age and histological subtypes were observed between GPV carriers and national data. Further research on cancer characteristics is needed to optimize counseling and cancer prevention in these women.
Collapse
Affiliation(s)
- Jella-Rike J. A. H. Spijkervet
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - L. Lanjouw
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - L. P. V. Berger
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - M. D. Dorrius
- Department of Radiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - B. van der Vegt
- Department of Pathology & Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - G. H. de Bock
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
3
|
Arafa AT, Yadav S, Marshall CH, Mauer E, Huang M, Yilma B, van der Pol Y, Fragkogianni S, Teslow EA, Kellen S, Boytim E, Luo C, Ludwig M, Zhang W, Jayaraj A, Armstrong DK, Isaacs WB, Drake JM, Nguyen HD, Huang RS, Chao CY, Lou E, Dehm SM, Couch FJ, Hwang JH, Antonarakis ES. Germline-Somatic Interactions in BRCA-Associated Cancers: Unique Molecular Profiles and Clinical Outcomes Linking ATM to TP53 Synthetic Essentiality. Clin Cancer Res 2025; 31:1730-1745. [PMID: 40019487 PMCID: PMC12045718 DOI: 10.1158/1078-0432.ccr-24-2058] [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: 06/29/2024] [Revised: 09/27/2024] [Accepted: 02/27/2025] [Indexed: 03/01/2025]
Abstract
PURPOSE Germline alterations in homologous recombination repair (gHRR) genes affect the pathogenesis, treatment options, and survival of patients with cancer. However, distinct gHRR gene alterations may differentially affect treatment response and oncogenic signaling. In this study, we interrogated genomic and transcriptomic data and assessed clinical outcomes of patients with gHRR mutations across four BRCA-associated cancers (breast, ovarian, pancreatic, and prostate cancers) to identify therapeutic vulnerabilities. EXPERIMENTAL DESIGN We assessed 24,309 patients undergoing matched tumor/normal next-generation DNA and RNA sequencing. Annotated gHRR gene variants [germline BRCA1, germline BRCA2, germline PALB2, germline ATM (gATM), and germline CHEK2] were analyzed. HRs were used to assess survival outcomes comparing germline versus sporadic groups. Somatic alterations and their frequencies were compared across gHRR-altered groups. Differential gene expression and gene set enrichment analysis were used to compare transcriptomic profiles. RESULTS Somatic TP53 mutations were depleted in gATM carriers (P < 0.05) across all four BRCA-associated cancers by up to 2.5-fold. Tumors with germline BRCA1/2 mutations were associated with improved survival in patients with ovarian cancer and had consistent enrichment of TP53 mutations in all four cancers. gATM mutations displayed elevated p53 transcriptional activity in all four cancers, with significance reached in breast and prostate cancers (P < 0.01). In breast, ovarian, and prostate cancers, gATM tumors demonstrated significantly increased inflammatory pathways (P < 0.001). Finally, using gene dependency data, we found that cell lines that were highly dependent on ATM were co-dependent on canonical p53 function. CONCLUSIONS gATM-associated cancers seem to require intact p53 activity and this synthetic essentiality may be used to guide targeted therapies that perturb canonical TP53 function.
Collapse
Affiliation(s)
- Ali T. Arafa
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Catherine H. Marshall
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | - Samuel Kellen
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ella Boytim
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Christine Luo
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Megan Ludwig
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Weijie Zhang
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Deborah K. Armstrong
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William B. Isaacs
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Justin M. Drake
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Hai Dang Nguyen
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - R. Stephanie Huang
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Emil Lou
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Scott M. Dehm
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Fergus J. Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Justin H. Hwang
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | | |
Collapse
|
4
|
Johansen Taber K, Hughes E, Gutin A, DeHart WB, Becker L, Jasper J, Ratzel S, Miller DC, Chawla D, Morin P, Certa J, Kurian AW. Association of Polygenic-Based Breast Cancer Risk Prediction With Patient Management. JCO Precis Oncol 2025; 9:e2400716. [PMID: 40334154 PMCID: PMC12068550 DOI: 10.1200/po-24-00716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/07/2025] [Accepted: 03/21/2025] [Indexed: 05/09/2025] Open
Abstract
PURPOSE Breast cancer (BC) risk prediction is more accurate when clinical and polygenic factors are combined (combined risk score [CRS]), but little is known about how CRS results affect real-world patient management. METHODS Deidentified medical and pharmacy claims data were linked with Tyrer-Cuzick (TC) and CRS results and evaluated for BC risk management. Patients were divided into subcohorts on the basis of lifetime risk predicted by CRS and by TC ("+": ≥20% risk, "-": <20%): CRS+ TC+, CRS+ TC-, CRS- TC+, and CRS- TC-. Claims data related to screening mammography (SM) in patients younger than 40 years, breast magnetic resonance imaging (MRI), and genetic counseling (GC) were compared 360 days before and after CRS testing. Differences in pre- and post-CRS management were evaluated using McNemar tests, and post-CRS management of subcohorts was compared using multivariable logistic regression. RESULTS After CRS testing, the CRS+ TC+, CRS+ TC-, and CRS- TC+ subcohorts had 1.6-2.2-fold increases in SM in patients younger than 40 years (all P < .02) and 4.7-5.6-fold increases in breast MRI (all P < .001). The CRS+ TC+ and CRS+ TC- subcohorts had 1.9-2.3-fold increases in GC (both P < .001). SM in those younger than 40 years, breast MRI, and GC did not increase in the CRS- TC- subcohort. After CRS testing, compared with the CRS- TC- subcohort, the CRS+ TC+, CRS+ TC-, and CRS- TC+ subcohorts had significantly higher odds of receiving SM before age 40 years (odds ratio [OR], 3.80-5.19), breast MRI (OR, 11.55-23.09), and GC (OR, 2.03-2.91; all P < .001). CONCLUSION Patients with ≥20% lifetime risk predicted by either CRS or TC were more likely to receive enhanced management compared with those who had <20% lifetime risk, suggesting that clinicians considered the CRS in BC risk management.
Collapse
|
5
|
Wong SM, Apostolova C, Ferroum A, Alhassan B, Prakash I, Basik M, Martel K, Meterissian S, Fleiszer D, Wong N, Sadinsky MB, Malagon T, Boileau JF, Foulkes WD. Chemotherapy receipt in affected BRCA1/2 and PALB2 carriers with operable breast cancer: the impact of early detection and pre-diagnostic awareness on clinical outcomes and treatment. Hered Cancer Clin Pract 2025; 23:14. [PMID: 40275390 PMCID: PMC12020017 DOI: 10.1186/s13053-025-00314-x] [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: 09/30/2024] [Accepted: 04/18/2025] [Indexed: 04/26/2025] Open
Abstract
PURPOSE While enhanced breast screening of germline pathogenic variant (GPV) carriers results in earlier stage at diagnosis, the impact of tumour biology and GPV on chemotherapy receipt in early-stage disease remains understudied. METHODS We retrospectively reviewed treatment administered following a first diagnosis of BRCA1/2- and PALB2-associated breast cancer between 2002 and 2022. Chemotherapy receipt was compared according to tumor size, biologic subtype, and GPV. Subgroup analyses were performed in women with T1N0 disease and in those with pre-diagnostic awareness of their GPV. RESULTS Overall, 309 affected BRCA1/2 and PALB2 carriers with a median age of 43 years at breast cancer diagnosis (range, 19-80 years) were included; 160 (51.8%) BRCA1, 130 (42.1%) BRCA2, and 19 (6.1%) PALB2 carriers. Chemotherapy was administered in 70.9% of index breast cancer cases and was significantly associated with younger age, tumor size, histologic grade, nodal status, and biologic subtype (all p < 0.05). Chemotherapy receipt was 80.6% in BRCA1-associated breast cancers compared to 56.9% in BRCA2 and 84.2% in PALB2 associated breast cancers (p < 0.001). In subgroup analysis of early stage, T1N0 disease, chemotherapy was administered in 78.9% BRCA1 and 59.5% BRCA2/PALB2 patients (p = 0.04). Pre-diagnostic awareness of a GPV in BRCA1/2 or PALB2 was associated with smaller invasive tumors (%T1, 50% vs. 32.9%; p = 0.002) and node-negative invasive disease (87.1% vs. 72.2%), as well as a reduced likelihood of chemotherapy (59.7% vs. 74.3%, p = 0.02). CONCLUSION Chemotherapy receipt is high in BRCA1/2 and PALB2-associated breast cancers including in early stage, node-negative disease. Pre-diagnostic awareness is associated with a lower likelihood of requiring chemotherapy for a breast cancer diagnosis.
Collapse
Affiliation(s)
- Stephanie M Wong
- Department of Surgery, McGill University, Montreal, QC, Canada.
- Stroll Cancer Prevention Centre, Jewish General Hospital, Montreal, QC, Canada.
- Department of Oncology, McGill University, Montreal, QC, Canada.
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.
- Segal Cancer Centre, Jewish General Hospital, 3755 Cote Ste Catherine, E713, Montreal, QC, H3T1E2, Canada.
| | - Carla Apostolova
- Department of Surgery, McGill University, Montreal, QC, Canada
- Stroll Cancer Prevention Centre, Jewish General Hospital, Montreal, QC, Canada
| | - Amina Ferroum
- Department of Surgery, McGill University, Montreal, QC, Canada
- Stroll Cancer Prevention Centre, Jewish General Hospital, Montreal, QC, Canada
| | - Basmah Alhassan
- Department of Surgery, McGill University, Montreal, QC, Canada
- Department of Oncology, McGill University, Montreal, QC, Canada
| | - Ipshita Prakash
- Department of Surgery, McGill University, Montreal, QC, Canada
- Department of Oncology, McGill University, Montreal, QC, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Mark Basik
- Department of Surgery, McGill University, Montreal, QC, Canada
- Department of Oncology, McGill University, Montreal, QC, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Karyne Martel
- Department of Surgery, McGill University, Montreal, QC, Canada
| | - Sarkis Meterissian
- Department of Surgery, McGill University, Montreal, QC, Canada
- Department of Oncology, McGill University, Montreal, QC, Canada
| | - David Fleiszer
- Department of Surgery, McGill University, Montreal, QC, Canada
- Department of Oncology, McGill University, Montreal, QC, Canada
| | - Nora Wong
- Stroll Cancer Prevention Centre, Jewish General Hospital, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Michaela Bercovitch Sadinsky
- Stroll Cancer Prevention Centre, Jewish General Hospital, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Talia Malagon
- Department of Oncology, McGill University, Montreal, QC, Canada
- St Mary's Research Centre, Montreal West Island Integrated University Health and Social Services Centre, Montreal, QC, Canada
| | | | - William D Foulkes
- Stroll Cancer Prevention Centre, Jewish General Hospital, Montreal, QC, Canada
- Department of Oncology, McGill University, Montreal, QC, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| |
Collapse
|
6
|
Abubakar M, Ahearn TU, Duggan MA, Lawrence S, Adjei EK, Clegg-Lamptey JN, Yarney J, Wiafe-Addai B, Awuah B, Wiafe S, Nyarko K, Aitpillah FS, Ansong D, Hewitt SM, Brinton LA, Figueroa JD, Garcia-Closas M, Edusei L, Titiloye N, for the Ghana Breast Health Study Team. Contribution of Prediagnostic Host Factors to Shaping the Stromal Microenvironment of Breast Cancer among Sub-Saharan African Women. Cancer Epidemiol Biomarkers Prev 2025; 34:462-473. [PMID: 38958945 PMCID: PMC11966112 DOI: 10.1158/1055-9965.epi-24-0390] [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: 03/11/2024] [Revised: 05/31/2024] [Accepted: 07/01/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND The stromal microenvironment (SME) is integral to breast cancer biology, impacting metastatic proclivity and treatment response. Emerging data indicate that host factors may impact the SME, but the relationship between prediagnostic host factors and SME phenotype remains poorly characterized, particularly among women of African ancestry. METHODS We conducted a case-only analysis involving 792 patients with breast cancer (17-84 years) from the Ghana Breast Health Study. High-accuracy machine-learning algorithms were applied to standard H&E-stained images to characterize SME phenotypes [including percent tumor-associated connective tissue stroma, Ta-CTS (%); tumor-associated stromal cellular density, Ta-SCD (%)]. Associations between prediagnostic host factors and SME phenotypes were assessed in multivariable linear regression models. RESULTS Decreasing Ta-CTS and increasing Ta-SCD were associated with aggressive, mostly high-grade tumors (P-value < 0.001). Several prediagnostic host factors were associated with Ta-SCD independently of tumor characteristics. Compared with nulliparous women, parous women had higher levels of Ta-SCD [mean (standard deviation, SD) = 31.3% (7.6%) vs. 28.9% (7.1%); P-value = 0.01]. Similarly, women with a positive family history of breast cancer had higher levels of Ta-SCD than those without family history [mean (SD) = 33.0% (7.5%)] vs. 30.9% (7.6%); P-value = 0.03]. Conversely, increasing body size was associated with decreasing Ta-SCD [mean (SD) = 31.6% (7.4%), 31.4% (7.3%), and 30.1% (8.0%) for slight, average, and large body sizes, respectively; P-value = 0.005]. CONCLUSIONS Epidemiological risk factors were associated with varying degrees of stromal cellularity in tumors, independently of clinicopathological characteristics. IMPACT The findings raise the possibility that epidemiological risk factors may partly influence tumor biology via the stromal microenvironment. See related In the Spotlight, p. 459.
Collapse
Affiliation(s)
- Mustapha Abubakar
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Thomas U. Ahearn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Maire A. Duggan
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Canada
| | - Scott Lawrence
- Molecular and Digital Pathology Laboratory, Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick, Maryland
| | | | | | | | | | | | - Seth Wiafe
- Loma Linda University, School of Public Health, Loma Linda, California
| | | | | | - Daniel Ansong
- Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Stephen M. Hewitt
- Center for cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Louise A. Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jonine D. Figueroa
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Usher Institute and Cancer Research UK Edinburgh Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Montserrat Garcia-Closas
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | | |
Collapse
|
7
|
Hahnen E, Hauke J, Gelmon K, Marmé F, Ernst C, Martin M, Untch M, Bonnefoi H, Knudsen E, Im SA, DeMichele A, Van't Veer L, Kim SB, Bear H, McCarthy N, Rhiem K, Turner N, Witkiewicz A, Rojo F, Filipits M, Martin LA, Fasching PA, Schem C, Becker K, García-Sáenz JA, Kelly CM, Reimer T, Toi M, Rugo HS, Denkert C, Gnant M, Makris A, Liu Y, Valota O, Felder B, Weber K, Nekljudova V, Loibl S. BRCA1/2 and Other Predisposition Genes in High-Risk Hormone Receptor+/Human Epidermal Growth Factor Receptor 2- Breast Cancer Treated With Endocrine Therapy With or Without Palbociclib: A Secondary PENELOPE-B Study Analysis. JCO Precis Oncol 2025; 9:e2400742. [PMID: 40209141 PMCID: PMC12005863 DOI: 10.1200/po-24-00742] [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: 10/14/2024] [Revised: 01/17/2025] [Accepted: 03/03/2025] [Indexed: 04/12/2025] Open
Abstract
PURPOSE The PENELOPE-B trial (ClinicalTrials.gov identifier: NCT01864746) recruited patients with hormone receptor+/human epidermal growth factor receptor 2- early breast cancer without a pathological complete response after taxane-containing neoadjuvant chemotherapy and at a high risk of relapse. Patients were randomly assigned (1:1) to receive 13 cycles of palbociclib once daily or placebo on days 1-21 in a 28-day cycle in addition to endocrine therapy (ET). PENELOPE-B did not show improved invasive disease-free survival (iDFS) after adding palbociclib to ET. This retrospective analysis investigated the impact of germline pathogenic variant (PV) status of BRCA1/2 and non-BRCA1/2 cancer predisposition genes on the outcomes of PENELOPE-B trial patients. METHODS In total, 445 patients were sampled following a case-cohort design and 442 were analyzed for germline PVs. Statistical analyses were performed for time-to-event end points (iDFS, distant disease-free survival [DDFS], and overall survival [OS]). RESULTS Of the 442 patients, 42 carried PVs in any cancer predisposition gene; 15 carried BRCA1/2 PVs. Irrespective of the treatment arms, PV status was not a prognostic factor. Regarding the treatment arms in BRCA1/2 PV carriers, numerically better 3-year outcomes were observed in the palbociclib arm (iDFS, 95%; DDFS, 95%; OS, 100%) than in the placebo arm (iDFS, 72.8%; DDFS, 72.8%; OS, 87.5%; hazard ratios palbociclib v placebo 0.349 [iDFS] and 0.562 [DDFS], not calculated for OS, too few events). In patients without BRCA1/2 PVs, the differences in 3-year outcomes were negligible. PVs in non-BRCA1/2 cancer predisposition genes did not influence the efficacy of palbociclib, although gene-specific effects could not be excluded. CONCLUSION Patients with BRCA1/2 PVs had numerically better outcomes after palbociclib. However, the number of BRCA1/2 carriers was small. Larger randomized clinical trials should consider the PV status to further evaluate whether BRCA1/2 PV carriers benefit from cyclin-dependent kinase 4 and 6 inhibitor treatment.
Collapse
Affiliation(s)
- Eric Hahnen
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Jan Hauke
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | | | | | - Corinna Ernst
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Miguel Martin
- Instituto de Investigacion Sanitaria Gregorio Marañon, CIBERONC, Universidad Complutense, Madrid, Spain
| | | | - Hervé Bonnefoi
- Institut Bergonié and Université de Bordeaux INSERM U916, Bordeaux, France
| | - Erik Knudsen
- Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Seock-Ah Im
- Seoul National University Hospital, Seoul, Korea
| | | | | | - Sung-Bae Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Harry Bear
- Division of Surgical Oncology, Massey Comprehensive Cancer Center, Virginia Commonwealth University, VCU Health, Richmond, VA
| | - Nicole McCarthy
- Breast Cancer Trials Australia and New Zealand and University of Queensland, Brisbane, Australia
| | - Kerstin Rhiem
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Nicholas Turner
- The Institute of Cancer Research: Royal Cancer Hospital, London, Great Britain
| | | | - Federico Rojo
- Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Martin Filipits
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Lesley-Ann Martin
- Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, United Kingdom
| | | | | | - Kerstin Becker
- Cologne Center for Genomics, Medical Faculty, University of Cologne, Cologne, Germany
| | | | - Catherine M. Kelly
- Trinity St James's Cancer Centre at St James's Hospital Dublin, Dublin, Ireland
| | - Toralf Reimer
- Department of Obstetrics and Gynecology, University of Rostock, Rostock, Germany
| | - Masakazu Toi
- Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hope S. Rugo
- University of California San Francisco Comprehensive Cancer Center, San Francisco, CA
| | - Carsten Denkert
- Institut für Pathologie, Philipps-Universität Marburg, Marburg, Germany
| | - Michael Gnant
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | - Yuan Liu
- Pfizer, Pfizer Italia s.r.l., Milan, Italy
| | | | | | | | | | | |
Collapse
|
8
|
Rodriguez-Hernandez A, Martínez-Sáez O, Brasó-Maristany F, Conte B, Gómez R, García-Fructuoso I, Fratini B, Segui E, Potrony M, Sanfeliu E, Cobo S, Galvan P, Moreno L, Grau E, Aceituno MR, Puig-Butille JA, Oriola J, Goberna G, Blasco P, Castillo O, Sirenko V, Aguirre A, Vidal M, Muñoz M, Ramon Y Cajal T, Balaguer F, Prat A, Adamo B. Prevalence and clinical impact of germline pathogenic variants in breast cancer: a descriptive large single-center study. ESMO Open 2025; 10:104543. [PMID: 40209283 PMCID: PMC12008705 DOI: 10.1016/j.esmoop.2025.104543] [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: 12/06/2024] [Revised: 02/14/2025] [Accepted: 03/08/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Germline (likely) pathogenic variants (PVs) are identified in 5%-10% of patients with breast cancer (BC) and play a critical role in guiding clinical management, including the use of targeted therapies such as poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi). High-risk genes such as BRCA1, BRCA2, and PALB2, and moderate-risk genes such as CHEK2 and ATM, influence BC risk and treatment decisions. This study evaluates the prevalence and clinical impact of PVs in a large consecutive cohort. MATERIALS AND METHODS A retrospective analysis was conducted on 912 individuals with BC who underwent germline testing at the Hospital Clinic of Barcelona from 2016 to 2023. Genetic testing for 14 BC and Lynch syndrome genes was carried out using the TruSight Hereditary Cancer Panel. Statistical analyses were carried out to assess associations between germline results and clinical characteristics, including eligibility for PARPi therapy. RESULTS Of the 912 individuals, 129 (14.1%) had a PV, with BRCA2 (31.8%) and BRCA1 (24%) being the most frequently altered genes. Additionally, 16.2% carried variants of uncertain significance, most commonly in ATM and BRCA2 genes. Patients with PV were younger compared with PV-negative individuals (median age: 43.5 versus 48.2 years, P = 0.006), more likely to have bilateral BC (13.3% versus 5.8%, P = 0.002), and more frequently diagnosed with triple-negative BC (TNBC; 28.7% versus 20.8%, P = 0.046). Of those with PVs, 39.1% completed a bilateral mastectomy, 36.7% had a risk-reducing salpingo-oophorectomy, and 22.7% had both surgeries. PV detection was associated with higher stages at diagnosis (stage IV: 13.0% versus 5.9%, P < 0.001). In the metastatic cohort, 12.9% received PARPi therapy, with 80.7% harboring BRCA1/2 PVs. In early BC, 13.1% met the criteria for adjuvant PARPi. CONCLUSIONS The identification of germline PVs significantly influences surgical decisions and systemic therapies. Genetic testing for patients with BC optimizes care, particularly in selecting candidates for PARPi in both early and advanced BC, improving management and prevention strategies.
Collapse
Affiliation(s)
- A Rodriguez-Hernandez
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Facultat de Medicina i Ciències de la Salud, Universitat de Barcelona (UB), Barcelona, Spain. https://twitter.com/AdelaRodrguezH1
| | - O Martínez-Sáez
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Facultat de Medicina i Ciències de la Salud, Universitat de Barcelona (UB), Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain
| | - F Brasó-Maristany
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors lab, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Cancer Institute, Hospital Clinic of Barcelona, Barcelona, Spain; Reveal Genomics, S.L Barcelona, Barcelona, Spain
| | - B Conte
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain
| | - R Gómez
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain
| | - I García-Fructuoso
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain
| | - B Fratini
- Medical Oncology Unit 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - E Segui
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Facultat de Medicina i Ciències de la Salud, Universitat de Barcelona (UB), Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain
| | - M Potrony
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - E Sanfeliu
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Department of Pathology, Biomedical Diagnostic Center, Hospital Clinic of Barcelona, Barcelona, Spain
| | - S Cobo
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - P Galvan
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - L Moreno
- Facultat de Medicina i Ciències de la Salud, Universitat de Barcelona (UB), Barcelona, Spain; Gastroenterology Department, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - E Grau
- Facultat de Medicina i Ciències de la Salud, Universitat de Barcelona (UB), Barcelona, Spain; Gastroenterology Department, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - M R Aceituno
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - J A Puig-Butille
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, Barcelona, Spain
| | - J Oriola
- Biochemistry and Molecular Genetics Department, Hospital Clínic of Barcelona, IDIBAPS, Barcelona, Spain
| | - G Goberna
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - P Blasco
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - O Castillo
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - V Sirenko
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - A Aguirre
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - M Vidal
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain
| | - M Muñoz
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain
| | - T Ramon Y Cajal
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - F Balaguer
- Facultat de Medicina i Ciències de la Salud, Universitat de Barcelona (UB), Barcelona, Spain; Gastroenterology Department, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - A Prat
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Facultat de Medicina i Ciències de la Salud, Universitat de Barcelona (UB), Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain; Reveal Genomics, S.L Barcelona, Barcelona, Spain; Institute of Oncology (IOB)-Hospital Quirónsalud, Barcelona, Spain.
| | - B Adamo
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Facultat de Medicina i Ciències de la Salud, Universitat de Barcelona (UB), Barcelona, Spain; SOLTI Cooperative Group, Barcelona, Spain. https://twitter.com/badamo20
| |
Collapse
|
9
|
Torrisi R, Gerosa R, Miggiano C, Saltalamacchia G, Benvenuti C, Santoro A. Beyond failure of endocrine-based therapies in HR+/HER2 negative advanced breast cancer: What before chemotherapy? A glimpse into the future. Crit Rev Oncol Hematol 2025; 208:104634. [PMID: 39900320 DOI: 10.1016/j.critrevonc.2025.104634] [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: 11/04/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Despite the impressive improvements achieved by endocrine therapy and CDK4/6 inhibitors (CDK4/6i) and the forthcoming availability of alternative endocrine manipulations and targeted therapies, hormone-receptor positive/HER2 negative (HR+/HER2-) advanced breast cancer (ABC) is almost inevitably destined to become endocrine- refractory. At this time chemotherapy has been recently challenged and partly replaced by new targeted options as antibody-drug conjugated (ADCs). Trastuzumab-deruxtecan has been proven meaningfully superior to chemotherapy either in 1st and later lines after progression to CDK4/6i in HER2-low ABC and results with other ADCs as Sacituzumab Govitecan and Datopotamab-deruxtecan are promising, but the definition of cross-resistance between these drugs sharing either antibody or payload is crucial before implementing them in a useful sequence. While PARP inhibitors are the standard 2nd line in patients with gBRCA mutation, it is not still known whether patients with mutations of PALB2 or of other homologous recombinant defect (HRD)-related genes will benefit of the same treatment. On the other hand, the results obtained with immune checkpoint inhibitors (ICIs) in HR+ /HER2-ABC contrarily to the early setting are disappointing up to now, but investigations of ICIs in combination with other targeted drugs which may increase immune response and the search for better markers of activity are under way. Moreover the anticipation in upfront treatment of ADCs or PARPi in patients with features of putative endocrine resistance and/or of less sensitiviy to CDK4/6i and the choice of therapy in patients recurring during or soon after adjuvant CDK4/6i and olaparib represent further challenges for the future.
Collapse
Affiliation(s)
- Rosalba Torrisi
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy.
| | - Riccardo Gerosa
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Chiara Miggiano
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Giuseppe Saltalamacchia
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy
| | - Chiara Benvenuti
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Armando Santoro
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| |
Collapse
|
10
|
Carvalho E, Canberk S, Schmitt F, Vale N. Molecular Subtypes and Mechanisms of Breast Cancer: Precision Medicine Approaches for Targeted Therapies. Cancers (Basel) 2025; 17:1102. [PMID: 40227634 PMCID: PMC11987866 DOI: 10.3390/cancers17071102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/20/2025] [Accepted: 03/23/2025] [Indexed: 04/15/2025] Open
Abstract
Breast cancer remains one of the most prevalent diseases worldwide, primarily affecting women. Its heterogeneous nature poses a significant challenge in the development of effective and targeted treatments. Molecular characterization has enabled breast cancer to be classified into four main subtypes: luminal A, luminal B, HER2-positive, and triple-negative breast cancer, based on hormone receptor expression and HER2 status. A deeper understanding of these molecular markers and their associated signaling pathways, such as MAPK and PI3K/AKT, is essential for improving prognosis and optimizing treatment strategies. Currently, several therapeutic agents are utilized in neoadjuvant and adjuvant therapies, often in combination with surgical interventions. However, emerging evidence highlights the growing challenge of drug resistance, which significantly limits the efficacy of existing treatments. Addressing this issue may require innovative approaches, including combination therapies and precision medicine strategies, tailored to the molecular profile of each patient. Therefore, a comprehensive understanding of the pathophysiologic mechanisms driving breast cancer progression and resistance is crucial for the development of advanced targeted therapies with greater precision and efficacy. This review aims to explore recent advancements in molecular research related to breast cancer subtypes and provide a critical analysis of current therapeutic approaches within the framework of precision medicine.
Collapse
Affiliation(s)
- Eduarda Carvalho
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
| | - Sule Canberk
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
- RISE-Health, Department of Pathology, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Fernando Schmitt
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
- RISE-Health, Department of Pathology, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (E.C.); (S.C.); (F.S.)
- RISE-Health, Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
11
|
Viart NM, Renault AL, Eon-Marchais S, Jiao Y, Fuhrmann L, El Houdigui SM, Le Gal D, Cavaciuti E, Dondon MG, Beauvallet J, Raynal V, Stoppa-Lyonnet D, Vincent-Salomon A, Andrieu N, Southey MC, Lesueur F. Breast tumors from ATM pathogenic variant carriers display a specific genome-wide DNA methylation profile. Breast Cancer Res 2025; 27:36. [PMID: 40069712 PMCID: PMC11899765 DOI: 10.1186/s13058-025-01988-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND The ataxia-telangiectasia mutated (ATM) kinase phosphorylates and activates several downstream targets that are essential for DNA damage repair, cell cycle inhibition and apoptosis. Germline biallelic inactivation of the ATM gene causes ataxia-telangiectasia (A-T), and heterozygous pathogenic variant (PV) carriers are at increased risk of cancer, notably breast cancer. This study aimed to investigate whether DNA methylation profiling can be useful as a biomarker to identify tumors arising in ATM PV carriers, which may help for the management and optimal tailoring of therapies of these patients. METHODS Breast tumor enriched DNA was prepared from 2 A-T patients, 27 patients carrying an ATM PV, 6 patients carrying a variant of uncertain clinical significance and 484 noncarriers enrolled in epidemiological studies conducted in France and Australia to investigate genetic and nongenetic factors involved in breast cancer susceptibility. Genome-wide DNA methylation analysis was performed using the Illumina Infinium HumanMethylation EPIC and 450K BeadChips. Correlation between promoter methylation and gene expression was assessed for 10 tumors for which transcriptomic data were available. RESULTS We found that the ATM promoter was hypermethylated in 62% of tumors of heterozygous PV carriers compared to the mean methylation level of ATM promoter in tumors of noncarriers. Gene set enrichment analyses identified 47 biological pathways enriched in hypermethylated genes involved in neoplastic, neurodegenerative and metabolic-related pathways in tumor of PV carriers. Among the 327 differentially methylated promoters, promoters of ARHGAP40, SCGB3A1 (HIN-1), and CYBRD1 (DCYTB) were hypermethylated and associated with a lower gene expression in these tumors. Moreover, using three different deep learning algorithms (logistic regression, random forest and XGBoost), we identified a set of 27 additional biomarkers predictive of ATM status, which could be used in the future to provide evidence for or against pathogenicity in ATM variant classification strategies. CONCLUSIONS We showed that breast tumors that arise in women who carry an ATM PV display a specific genome-wide DNA methylation profile. Specifically, the methylation pattern of 27 key gene promoters was predictive of ATM PV status of the women. These genes may also represent new medical prevention and therapeutic targets for these women.
Collapse
Affiliation(s)
- Nicolas M Viart
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | - Anne-Laure Renault
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
- Monash University, Clayton, VIC; University of Melbourne, Parkville, VIC, Australia
| | | | - Yue Jiao
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | | | | | - Dorothée Le Gal
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | - Eve Cavaciuti
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | | | - Juana Beauvallet
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | - Virginie Raynal
- ICGex Next-Generation Sequencing Platform, Institut Curie, PSL University, Paris, France
| | | | | | - Nadine Andrieu
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | - Melissa C Southey
- Monash University, Clayton, VIC; University of Melbourne, Parkville, VIC, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Fabienne Lesueur
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France.
| |
Collapse
|
12
|
Hemminki K, Försti A, Hemminki O, Scott RJ, Hemminki A. Age-specific familial risks in cancer as clues to germline genetic and environmental causes: focus on colorectal, endometrial, prostate, kidney, breast and lung cancers. Hered Cancer Clin Pract 2025; 23:7. [PMID: 39985094 PMCID: PMC11844152 DOI: 10.1186/s13053-024-00301-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 11/18/2024] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND The Swedish Family-Cancer Database (FCD) is the largest source of data on familial cancer in the world, including practically complete family structures and individual cancer diagnoses from the high-quality cancer registry. We present a novel application of FCD by analyzing age-specific familial risks and interpreting them through likely causes, such as germline pathogenic variants and/or environmental exposures. MAIN BODY The basic assumption for this approach is that a discrete familial clustering in a narrow age-interval is not random but may provide causal clues. For this analysis we selected reasonably common cancers to meaningfully scrutinize familial risk through adulthood in which cancers are diagnosed, that included colorectal (CRC) and endometrial cancers, prostate and kidney cancers and breast and lung cancers. The interpretation is based on the literature. The highest familial relative risks for CRC and endometrial cancers were found at ages 40-44 years, matching the peak impact of mismatch repair gene mutations. However endometrial cancer showed also a small early onset component which could not be explained. Age-related familial risks for breast, prostate and kidney cancers also matched data from large-scale sequencing; these included the early onset component in kidney cancer which was likely due to VHL mutations. Age distribution of familial lung cancer was unique in showing a wide peak extending from middle to old ages, which would be consistent with a combination of direct genetic effects and indirect influence on inheritance of smoking dependence. CONCLUSIONS The present review of age-specific familial risks and age-of-onset data from the literature may allow an interpretation that the familial and germline landscapes are reasonably harmonious for relatively early onset cancers but at higher ages no discrete peaks can be found which may implicate attenuated impact of high-risk genes and polygenic influence.
Collapse
Affiliation(s)
- Kari Hemminki
- Biomedical Center, Faculty of Medicine, Charles University, Pilsen, 30605, Czech Republic.
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg, 69120, Germany.
| | - Asta Försti
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Cancer Consortium (DKTK), Heidelberg, Germany
| | - Otto Hemminki
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Rodney J Scott
- The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
13
|
Weis LN, Bychkovsky BL, Hernandez AR, Barroso-Sousa R, Sandoval RL. CHEK2-related breast cancer: real-world challenges. Fam Cancer 2025; 24:23. [PMID: 39966186 DOI: 10.1007/s10689-025-00448-w] [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: 09/19/2024] [Accepted: 02/09/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE Management of cancer risks associated with the CHEK2 gene, a moderate penetrance breast cancer gene, is challenging in real-world practice. Family history, traditional breast cancer risk factors, and specific genetic CHEK2 variants are risk modifiers in this setting and add complexity for surveillance and risk-reduction decisions. Here, we present a case series of Brazilian CHEK2 carriers affected by breast cancer. METHODS Patients evaluated in the Oncogenetics Department of Hospital Sírio-Libanês (Brasília, Brazil) between November 2017 and September 2021, who had a personal history of breast cancer and a germline genetic test with a pathogenic or likely pathogenic CHEK2 variant, were selected for case description. Clinical pearls and knowledge gaps were highlighted for each case. RESULTS Twelve women were included in this descriptive analysis. All patients had early-stage breast cancer. Most of them were diagnosed with breast cancer prior to age 50 (9/12) and had a close relative affected by breast cancer (9/12). Seven patients harbored intronic pathogenic variants. Clinical pearls included the following: lack of risk estimates for intronic CHEK2 variants among non-European ancestry CHEK2 carriers, environmental exposures as a risk modifier, notable non-breast cancer diagnosis at young ages, incidental germline finding during tumor profiling, breast cancer diagnosis before the recommended age of breast cancer screening, family history of breast cancer as a risk modifier, and clinical outcomes after breast cancer treatment. CONCLUSIONS Improvements in cancer risk assessment and cancer prevention for CHEK2 carriers are still needed to overcome current clinical challenges on the management of these patients.
Collapse
Affiliation(s)
- Luiza N Weis
- Dasa Oncology, Hospital Brasília, Brasília, Brazil
| | - Brittany L Bychkovsky
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, SGAS 613, S/N 94, Via L2 Sul, 70.200 - Asa Sul Brasília-DF, Boston, MA, 70200-730, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Adela Rodríguez Hernandez
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, SGAS 613, S/N 94, Via L2 Sul, 70.200 - Asa Sul Brasília-DF, Boston, MA, 70200-730, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | | | - Renata L Sandoval
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, SGAS 613, S/N 94, Via L2 Sul, 70.200 - Asa Sul Brasília-DF, Boston, MA, 70200-730, USA.
- Hospital Sírio-Libanês, Oncology Center, Brasília, Brazil.
| |
Collapse
|
14
|
Togashi Y, Nagahashi M, Kashima M, Okada C, Kinjo C, Miyazaki A, Ueda M, Tsubamoto H, Sawai H, Miyoshi Y. Germline variants detected by multigene panel testing in patients with suspected hereditary breast cancer. Surg Today 2025:10.1007/s00595-025-02994-3. [PMID: 39831988 DOI: 10.1007/s00595-025-02994-3] [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: 10/04/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025]
Abstract
PURPOSE To clarify the status of multigene panel testing for suspected hereditary breast cancer in our institute, and disclose the characteristics of the variants detected. METHODS This was a retrospective study of individuals who underwent next-generation sequencing-based multigene panel testing at our institute to investigate hereditary genetic variants for suspected hereditary breast cancer. RESULTS We identified 36 women who underwent multigene panel testing: 8 (22.2%) had a pathogenic variant, with or without other variants of uncertain significance (VUSs); 15 (41.7%) had VUSs only; and 13 (36.1%) had negative genetic test results. Of the eight pathogenic variants, five were BRCA2 variants and one each were BRCA1, MLH1, and RINT1 variants. The VUSs included BRCA1 and BRCA2, as well as other breast cancer-associated genes, such as ATM, CDH1, CHEK2, and PALB2. Referring to the latest ClinVar database, one of the variants identified as a VUS at diagnosis was re-determined as likely pathogenic, and three of the variants identified as VUSs at diagnosis were re-determined as benign. CONCLUSION VUSs are frequently identified during testing and it is important to monitor these individuals because VUS evaluations can change over time.
Collapse
Affiliation(s)
- Yusa Togashi
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Masayuki Nagahashi
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan.
- Department of Clinical Genetics, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan.
| | - Mina Kashima
- Department of Clinical Genetics, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Chiho Okada
- Department of Clinical Genetics, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Chinatsu Kinjo
- Department of Clinical Genetics, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Ayako Miyazaki
- Department of Clinical Genetics, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Mako Ueda
- Department of Clinical Genetics, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Hiroshi Tsubamoto
- Department of Clinical Genetics, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
- Department of Obstetrics and Gynecology, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Hideaki Sawai
- Department of Clinical Genetics, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
- Department of Obstetrics and Gynecology, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Yasuo Miyoshi
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| |
Collapse
|
15
|
Pal T, Schon KR, Astiazaran-Symonds E, Balmaña J, Foulkes WD, James P, Klugman S, Livinski AA, Mak JS, Ngeow J, Voian N, Wick MJ, Hanson H, Stewart DR, Tischkowitz M. Management of individuals with heterozygous germline pathogenic variants in ATM: A clinical practice resource of the American College of Medical Genetics and Genomics (ACMG). Genet Med 2025; 27:101243. [PMID: 39636577 DOI: 10.1016/j.gim.2024.101243] [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: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 12/07/2024] Open
Abstract
PURPOSE ATM germline pathogenic variants (GPVs) are associated with a moderately increased risk of female breast cancer, pancreatic cancer, and prostate cancer. Resources for managing ATM heterozygotes in clinical practice are limited. METHODS An international workgroup developed a clinical practice resource to guide management of ATM heterozygotes using peer-reviewed publications and expert opinion. RESULTS Although ATM is a moderate (intermediate) penetrance gene, cancer risks may be considered as a continuous variable, influenced by family history and other modifiers. ATM GPV heterozygotes should generally be offered enhanced breast surveillance according to their personalized risk estimate and country-specific guidelines and, generally, risk-reducing mastectomy is not recommended. Prostate cancer surveillance should be considered. Pancreatic cancer surveillance should be considered based on assessment of family history, ideally as part of a clinical trial, with existence of country-specific guidelines. For ATM GPV heterozygotes who develop cancer, radiation therapy decisions should not be influenced by the genetic result. Although poly-adenosine diphosphate ribose polymerase inhibitors are licensed for use in metastatic castration-resistant prostate cancer and ATM GPVs, the evidence-base is currently weak. CONCLUSION Systematic prospective data collection is needed to establish the spectrum of ATM-associated cancer and determine the outlines of surveillance, response to cancer treatment, and survival.
Collapse
Affiliation(s)
- Tuya Pal
- Department of Medicine, Vanderbilt University Medical Center/Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Katherine R Schon
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | | | - Judith Balmaña
- Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Medical Oncology Department, Hospital Universitari Vall d'Hebron, Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - William D Foulkes
- Departments of Human Genetics, Oncology and Medicine, McGill University, Montréal, Québec, Canada
| | - Paul James
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia; Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Susan Klugman
- Division of Reproductive & Medical Genetics, Department of Obstetrics and Gynecology and Women's Health, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY
| | - Alicia A Livinski
- National Institutes of Health Library, Office of Research Services, OD, NIH, Bethesda, MD
| | - Julie S Mak
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
| | - Joanne Ngeow
- Genomic Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Nicoleta Voian
- Providence Genetic Risk Clinic, Providence Cancer Institute, Portland, OR
| | - Myra J Wick
- Departments of Obstetrics and Gynecology and Clinical Genomics, Mayo Clinic, Rochester, MN
| | - Helen Hanson
- Peninsula Clinical Genetics, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom; Faculty of Health and Life Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Douglas R Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
16
|
Shah PD. Estrogen Receptor Status of Contralateral Breast Cancers-Value Added or Pathology Without a Purpose? JAMA Netw Open 2024; 7:e2452107. [PMID: 39786409 DOI: 10.1001/jamanetworkopen.2024.52107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Affiliation(s)
- Payal D Shah
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| |
Collapse
|
17
|
Goldfeld EI, Kelly BE, Ring KL. What About the Others? Clinical Management of Gynecologic Cancer Risk in Patients With Moderate-Risk Hereditary Cancer Genes ( ATM , BRIP1 , RAD51C , RAD51D , and PALB2 ). Clin Obstet Gynecol 2024; 67:696-701. [PMID: 39324947 DOI: 10.1097/grf.0000000000000897] [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: 09/27/2024]
Abstract
Hereditary cancer syndromes associated with gynecologic malignancies account for up to 18% of all cases of ovarian, uterine, and cervical cancers, and identification of these syndromes has implications for cancer screening and risk reduction techniques in affected patients. The associated cancer risks with moderate-penetrance genes are rapidly evolving and present variable risks for the provider counseling the patient. In this review, we detail the cancer risk and management of patients with germline PV in the moderate-risk hereditary cancer genes ATM , BRIP1 , RAD51C , RAD51D , and PALB2 .
Collapse
Affiliation(s)
- Ester I Goldfeld
- Department of Obstetrics and Gynecology, University of Virginia Health System, Charlottesville, Virginia
| | | | | |
Collapse
|
18
|
Teruel I, Castellà E, Recalde S, Viñas G, Petit A, Trigueros M, Martínez-Balibrea E, Felip E, Bergamino M, Bernat-Peguera A, Cirauqui B, Quiroga V, Ferrando-Díez A, Pous A, López A, Boronat L, Soler G, Recuero J, Romeo M, Guillén P, Mesía R, Ballana E, Martínez-Cardús A, Margelí M. Assessing the Prognostic Value of Cytoplasmic and Stromal Caveolin-1 in Early Triple-Negative Breast Cancer Undergoing Neoadjuvant Chemotherapy. Int J Mol Sci 2024; 25:12241. [PMID: 39596307 PMCID: PMC11594706 DOI: 10.3390/ijms252212241] [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: 09/17/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype with limited therapeutic options, leading to higher relapse rates and mortality. Identifying prognostic biomarkers like caveolin-1 (CAV1) is crucial for personalized treatment. CAV1 influences tumor progression and chemotherapy response, particularly through its interaction with the tumor microenvironment (TME) and cancer metabolism. Understanding the prognostic value of CAV1 in different cellular compartments is essential for its clinical application in TNBC. In the methods section CAV1 gene expression in TNBC was evaluated using in silico analysis, followed by the immunohistochemical staining of tumor cytoplasm (cCAV1) and stromal cells (sCAV1) in 58 early-stage TNBC patients. Statistical analyses were performed to correlate CAV1 expression with clinicopathological features and survival. In the results section, in silico analysis revealed higher CAV1 expression in TNBC, correlating with shorter overall survival. In the patient samples, cCAV1 was observed in 10.3% of cases, and was associated with larger tumors, higher grades, and poorer prognoses. sCAV1 was detected in 42% of cases, associated with less proliferative and less aggressive tumors, but did not significantly impact prognoses. In conclusion, cCAV1 expression is a significant prognostic marker in early-stage TNBC, highlighting the importance of assessing CAV1 in different cellular compartments. Further research is needed to explore the mechanisms and clinical implications of cCAV1.
Collapse
Affiliation(s)
- Iris Teruel
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Eva Castellà
- Department of Pathology, Hospital Germans Trias i Pujol, IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain;
| | - Sabela Recalde
- Department of Medical Oncology-Breast Cancer Unit, Institut Català d’Oncologia (ICO)-H.U.Bellvitge, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, 08907 Barcelona, Spain;
| | - Gemma Viñas
- Department of Medical Oncology-Breast Cancer Unit, Institut Català d’Oncologia (ICO)-H.U.Doctor Josep Trueta, Precision Oncology Group (OncoGIR-Pro), Institut d’Investigació Biomèdica de Girona (IDIBGI), Universitat de Girona, 17007 Girona, Spain;
| | - Anna Petit
- Departament of Pathology, Hospital Universitari de Bellvitge, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet del Llobregat, 08908 Barcelona, Spain;
| | - Macedonia Trigueros
- AIDS Research Institute-IrsiCaixa, Health Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Universitat Autònoma de Barcelona, 08916 Badalona, Spain (E.B.)
| | - Eva Martínez-Balibrea
- ProCURE Program, Institut Català d’Oncologia (ICO) and CARE Program, Health Research Institute Germans Trias i Pujol (IGTP), 08916 Badalona, Spain;
| | - Eudald Felip
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Milana Bergamino
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Adrià Bernat-Peguera
- CARE Program, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), 08916 Badalona, Spain;
| | - Beatriz Cirauqui
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Vanesa Quiroga
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Angelica Ferrando-Díez
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Anna Pous
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Assumpció López
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Laia Boronat
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Gemma Soler
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Jordi Recuero
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Margarita Romeo
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Pau Guillén
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Ricard Mesía
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| | - Ester Ballana
- AIDS Research Institute-IrsiCaixa, Health Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Universitat Autònoma de Barcelona, 08916 Badalona, Spain (E.B.)
| | - Anna Martínez-Cardús
- CARE Program, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), 08916 Badalona, Spain;
| | - Mireia Margelí
- Medical Oncology Department, Catalan Institut of Oncology (ICO)-Badalona, B-ARGO (Badalona Applied Research Group in Oncology) and IGTP (Health Research Institute Germans Trias i Pujol), Universitat Autònoma de Barcelona, 08916 Badalona, Spain; (I.T.); (E.F.); (M.B.); (B.C.); (V.Q.); (A.F.-D.); (A.P.); (A.L.); (L.B.); (G.S.); (J.R.); (P.G.); (R.M.)
| |
Collapse
|
19
|
Bottosso M, Sandoval RL, Verret B, Polidorio N, Caron O, Gennari A, Bychkovsky BL, Cahill SH, Achatz MI, Guarneri V, André F, Garber JE. HER2 status and response to neoadjuvant anti-HER2 treatment among patients with breast cancer and Li-Fraumeni syndrome. Eur J Cancer 2024; 211:114307. [PMID: 39260016 DOI: 10.1016/j.ejca.2024.114307] [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: 05/22/2024] [Revised: 07/28/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Breast cancer (BC) is the most common cancer among females with Li-Fraumeni syndrome (LFS), but available data on LFS-related BC characteristics are derived from small retrospective cohorts. Prior work has demonstrated a high proportion of HER2-positive BCs, but our understanding of how HER2-positive LFS BCs respond to anti-HER2 treatments is limited. METHODS BCs diagnosed in patients with germline TP53 variants between 2002-2022 were assembled from three institutions. Hormone receptor (HR) and HER2 expression were retrieved from pathology records. Pathologic complete response (pCR) was defined as ypT0/is ypN0. RESULTS A total of 264 BCs were identified among 232 patients with LFS: 211 (79.9 %) were invasive carcinomas, of which 106 were HER2-positive. Among HER2-positive BCs, most tumors co-expressed HRs (72.6 %) and were more frequent among those diagnosed at younger age (p < 0.001). Mastectomy was the preferred surgical approach among women with nonmetastatic cancers (77.8 %) and most received anti-HER2 targeted therapy (74.7 %). Among 38 patients receiving neoadjuvant therapy with available post-treatment pathology reports, 27 (71.1 %) achieved pCR: 18/26 (69.2 %) among HR-positive and 7/10 (70.0 %) HR-negative. The rate of pCR was 84.6 % among patients treated with an anthracycline-free regimen (all received trastuzumab). Among classifiable HER2-negative BCs (n = 77), 31 (40.3 %) were HER2-low and 46 (59.7 %) HER2-zero. CONCLUSIONS Among females with LFS and BC, HER2-positive subtype was associated with younger age at diagnosis and a predominant HR-positivity. Favorable pCR rates were observed among those receiving neoadjuvant HER2-directed therapies, for both HR-positive and negative tumors. These data may inform the counseling and care of patients with LFS.
Collapse
Affiliation(s)
- Michele Bottosso
- INSERM Unit U981, Gustave Roussy Cancer Campus, Villejuif, France; Department of Surgery, Oncology and Gastroenterology, University of Padua, Italy.
| | - Renata L Sandoval
- Medical Oncology Center, Hospital Sírio-Libanês, Brasília, DF, Brazil
| | - Benjamin Verret
- INSERM Unit U981, Gustave Roussy Cancer Campus, Villejuif, France
| | - Natalia Polidorio
- Breast Surgery Department, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Olivier Caron
- Gustave Roussy, Département de médecine oncologique, F-94805 Villejuif, France
| | - Alessandra Gennari
- Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy; Medical Oncology, "Maggiore Della Carità" University Hospital, Novara, Italy
| | - Brittany L Bychkovsky
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| | - Sophie H Cahill
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maria I Achatz
- Centro de Oncologia, Hospital Sírio-Libanês, Sao Paulo, SP, Brazil
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Italy
| | - Fabrice André
- INSERM Unit U981, Gustave Roussy Cancer Campus, Villejuif, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France; PRISM, INSERM, Gustave Roussy, Villejuif, France; Paris Saclay University, Gif Sur-Yvette, France
| | - Judy E Garber
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
| |
Collapse
|
20
|
Akamandisa MP, Boddicker NJ, Yadav S, Hu C, Hart SN, Ambrosone C, Anton-Culver H, Auer PL, Bodelon C, Burnside ES, Chen F, Eliassen HA, Goldgar DE, Haiman C, Hodge JM, Huang H, John EM, Karam R, Lacey JV, Lindstroem S, Martinez E, Na J, Neuhausen SL, O'Brien KM, Olson JE, Pal T, Palmer JR, Patel AV, Pesaran T, Polley EC, Richardson ME, Ruddy K, Sandler DP, Teras LR, Trentham-Dietz A, Vachon CM, Weinberg C, Winham SJ, Yao S, Zirpoli G, Kraft P, Weitzel JN, Domchek SM, Couch FJ, Nathanson KL. Association of Gene Variant Type and Location with Breast Cancer Risk in the General Population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.11.24315237. [PMID: 39417132 PMCID: PMC11482981 DOI: 10.1101/2024.10.11.24315237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Importance Pathogenic variants (PVs) in ATM, BRCA1, BRCA2, CHEK2 , and PALB2 are associated with increased breast cancer risk. However, it is unknown whether breast cancer risk differs by PV type or location in carriers ascertained from the general population. Objective To evaluate breast cancer risks associated with PV type and location in ATM, BRCA1, BRCA2, CHEK2 , and PALB2 . Design Age adjusted case-control association analysis for all participants, subsets of PV carriers, and women with no breast cancer family history in population-based and clinical testing cohorts. Setting Twelve US population-based studies within the Cancer Risk Estimates Related to Susceptibility (CARRIERS) Consortium, and breast cancer cases from the UK-Biobank and an Ambry Genetics clinical testing cohort. Participants 32,247 women with and 32,544 age-matched women without a breast cancer diagnosis from CARRIERS; 237 and 1351 women with BRCA2 PVs and breast cancer from the UKBB and Ambry Genetics, respectively. Exposures PVs in ATM, BRCA1, BRCA2, CHEK2, and PALB2. Main Outcomes and Measures PVs were grouped by type and location within genes and assessed for risks of breast cancer (odds ratios (OR), 95% confidence intervals (CI), and p-values) using logistic regression. Mean ages at diagnosis were compared using linear regression. Results Compared to women carrying BRCA2 exon 11 protein truncating variants (PTVs) in the CARRIERS population-based study, women with BRCA2 ex13-27 PTVs (OR=2.7, 95%CI 1.1-7.9) and ex1-10 PTVs (OR=1.6, 95%CI 0.8-3.5) had higher breast cancer risks, lower rates of ER-negative breast cancer (ex13-27 OR=0.5, 95%CI 0.2-0.9; ex1-10 OR=0.5, 95%CI 0.1-1.0), and earlier age of breast cancer diagnosis (ex13-27 5.5 years, p<0.001; ex1-10 2.4 years, p=0.17). These associations with ER-negative breast cancer and age replicated in a high-risk clinical cohort and the population-based UK Biobank cohort. No differences in risk or age at diagnosis by gene region were observed for PTVs in other predisposition genes. Conclusions and Relevance Population-based and clinical high-risk cohorts establish that PTVs in exon 11 of BRCA2 are associated with reduced risk of breast cancer, later age at diagnosis, and greater risk of ER-negative disease. These differential risks may improve individualized risk prediction and clinical management for women carrying BRCA2 PTVs. Key Points Question: Does ATM , BRCA1 , BRCA2 , CHEK2 and PALB2 pathogenic variant type and location influence breast cancer risk in population-based studies? Findings: Breast cancer risk and estrogen receptor status differ based on the type and location of pathogenic variants in BRCA2 . Women carrying protein truncating variants in exon 11 have a lower breast cancer risk in the population-based cohorts, older age at diagnosis and higher rates of estrogen receptor negative breast cancer than women with exon 1-10 or exon 13-27 truncation variants in population-based and clinical testing cohorts. Meaning: Incorporating pathogenic variant type and location in cancer risk models may improve individualized risk prediction.
Collapse
|
21
|
Villacis RAR, Côrtes L, Basso TR, do Canto LM, Souza JS, Aagaard MM, da Cruz Formiga MN, Aguiar S, Achatz MI, Rogatto SR. Germline DNA Damage Repair Gene Alterations in Patients with Metachronous Breast and Colorectal Cancer. Int J Mol Sci 2024; 25:10275. [PMID: 39408606 PMCID: PMC11476855 DOI: 10.3390/ijms251910275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
A hereditary component of breast (BC) and colorectal cancer (CRC) has been described in approximately one-third of these tumor types. BC patients have an increased risk of developing CRC as a second primary tumor and vice versa. Germline genomic variants (NextSeq550, Illumina) were investigated in 24 unrelated BC and/or CRC patients and 7 relatives from 3 index patients. Fifty-six pathogenic or likely pathogenic variants were identified in 19 of 24 patients. We detected single-nucleotide variants (SNVs) in CRC predisposition genes (MLH1 and MUTYH) and other promising candidates (CDK5RAP3, MAD1L1, NOS3, and POLM). Eighteen patients presented SNVs or copy number variants (CNVs) in DNA damage repair genes. We also identified SNVs recently associated with BC or CRC predisposition (PABPC1, TYRO3, MAP3K1, SLC15A4, and LAMA1). The PABPC1c.1255C>T variant was detected in nine unrelated patients. Each patient presented at least one SNV/CNV in a candidate gene, and most had alterations in more than one gene, reinforcing a polygenic model for BC/CRC predisposition. A significant fraction of BC/CRC patients with a family history of these tumors harbored deleterious germline variants in DNA repair genes. Our findings can lead to strategies to improve the diagnosis, genetic counseling, and treatment of patients and their relatives.
Collapse
Affiliation(s)
- Rolando André Rios Villacis
- Department of Clinical Genetics, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; (R.A.R.V.); (L.C.); (T.R.B.); (L.M.d.C.); (M.M.A.)
- Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília-UnB, Brasília 70910-900, DF, Brazil
| | - Luiza Côrtes
- Department of Clinical Genetics, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; (R.A.R.V.); (L.C.); (T.R.B.); (L.M.d.C.); (M.M.A.)
- Tocogynecology Graduation Program, Medical School, São Paulo State University UNESP, Botucatu 18618-687, SP, Brazil
| | - Tatiane Ramos Basso
- Department of Clinical Genetics, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; (R.A.R.V.); (L.C.); (T.R.B.); (L.M.d.C.); (M.M.A.)
| | - Luisa Matos do Canto
- Department of Clinical Genetics, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; (R.A.R.V.); (L.C.); (T.R.B.); (L.M.d.C.); (M.M.A.)
| | | | - Mads Malik Aagaard
- Department of Clinical Genetics, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; (R.A.R.V.); (L.C.); (T.R.B.); (L.M.d.C.); (M.M.A.)
| | | | - Samuel Aguiar
- Colorectal Cancer Reference Center, A.C. Camargo Cancer Center, São Paulo 01509-010, SP, Brazil;
| | - Maria Isabel Achatz
- Cancer Genetics Unit, Oncology Branch, Hospital Sirio-Libanês, São Paulo 01308-050, SP, Brazil;
| | - Silvia Regina Rogatto
- Department of Clinical Genetics, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; (R.A.R.V.); (L.C.); (T.R.B.); (L.M.d.C.); (M.M.A.)
- Institute of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, 5000 Odense, Denmark
- Danish Colorectal Cancer Center South, 7100 Vejle, Denmark
- Botucatu Medical School Hospital, São Paulo State University UNESP, Botucatu 18618-687, SP, Brazil
| |
Collapse
|
22
|
Jinadasa M, Humphreys A, Massey EJ, Vergani P, Jacobs-Pearson M, Smith K, Vinnicombe S, Papathomas T. Triple-Negative Pleomorphic Lobular Carcinoma in a BRCA1 Mutation Carrier: A Case of Complete Pathological Response. AMERICAN JOURNAL OF CASE REPORTS 2024; 25:e943882. [PMID: 39127886 PMCID: PMC11660003 DOI: 10.12659/ajcr.943882] [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: 01/21/2024] [Revised: 07/04/2024] [Accepted: 06/11/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Hereditary breast cancer arising in BRCA1-deficient patients is commonly diagnosed as invasive carcinoma of no special type (NST) with medullary features, while invasive lobular carcinoma (ILC) appears to be significantly under-represented in BRCA1 mutation carriers. We report a case of pleomorphic ILC arising in a 28-year-old woman harboring a germline BRCA1 c.3756_3759delGTCT p.(Ser1253Argfs*10) pathogenic variant. CASE REPORT A nulliparous 28-year-old woman with a family history of BRCA1 mutation presented to the symptomatic breast clinic with a several-week history of a left 80-mm breast lump. Core biopsy established a diagnosis of a poorly differentiated triple-negative breast cancer (TNBC) of pleomorphic lobular phenotype. Her clinical diagnosis was cT3, N0, M0, cStageIIB. The MDT recommended CT staging, MRI breast imaging and neoadjuvant chemotherapy (NACT). PET CT imaging showed no evidence of distant metastatic disease. The patient had a good radiological response to NACT with a FEC-T carboplatin regimen. Post-NACT imaging showed a residual cystic mass and the patient underwent a mastectomy and sentinel lymph node biopsy with plans for a delayed latissimus dorsi reconstruction following her adjuvant radiotherapy treatment. A complete pathological response was subsequently demonstrated without any evidence of metastatic disease. CONCLUSIONS This case is the first report of pleomorphic ILC with a triple-negative receptor status and a complete pathological response in a BRCA1 mutation carrier. Our study expands the heterogeneous spectrum of TNBC and contributes to a better understanding of the molecular genetic landscape that characterizes invasive pleomorphic lobular neoplasia.
Collapse
Affiliation(s)
- Maheshika Jinadasa
- Gloucestershire Cellular Pathology Laboratory, Cheltenham General Hospital, Gloucestershire Hospitals NHS Foundation Trust, Cheltenham, United Kingdom
| | - Alex Humphreys
- Department of Surgery, Northumbria Healthcare NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Eleanore J. Massey
- Thirlestaine Breast Centre, Cheltenham General Hospital, Gloucestershire Hospitals NHS Foundation Trust, Cheltenham, United Kingdom
| | - Patricia Vergani
- Gloucestershire Cellular Pathology Laboratory, Cheltenham General Hospital, Gloucestershire Hospitals NHS Foundation Trust, Cheltenham, United Kingdom
| | - Makaela Jacobs-Pearson
- Bristol Regional Clinical Genetics Service, St Michael’s Hospital, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Katherine Smith
- Bristol Regional Clinical Genetics Service, St Michael’s Hospital, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Sarah Vinnicombe
- Department of Radiology, Cheltenham General Hospital, Gloucestershire Hospitals NHS Foundation Trust, Cheltenham, United Kingdom
| | - Thomas Papathomas
- Department of Clinical Pathology, Vestre Viken HF, Drammen, Norway
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
23
|
Yadav S, Couch FJ, Domchek SM. Germline Genetic Testing for Hereditary Breast and Ovarian Cancer: Current Concepts in Risk Evaluation. Cold Spring Harb Perspect Med 2024; 14:a041318. [PMID: 38151326 PMCID: PMC11293548 DOI: 10.1101/cshperspect.a041318] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Our understanding of hereditary breast and ovarian cancer has significantly improved over the past two decades. In addition to BRCA1/2, pathogenic variants in several other DNA-repair genes have been shown to increase the risks of breast and ovarian cancer. The magnitude of cancer risk is impacted not only by the gene involved, but also by family history of cancer, polygenic risk scores, and, in certain genes, pathogenic variant type or location. While estimates of breast and ovarian cancer risk associated with pathogenic variants are available, these are predominantly based on studies of high-risk populations with young age at diagnosis of cancer, multiple primary cancers, or family history of cancer. More recently, breast cancer risk for germline pathogenic variant carriers has been estimated from population-based studies. Here, we provide a review of the field of germline genetic testing and risk evaluation for hereditary breast and ovarian cancers in high-risk and population-based settings.
Collapse
Affiliation(s)
- Siddhartha Yadav
- Department of Oncology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55901, USA
| | - Susan M Domchek
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
24
|
Lebedeva A, Veselovsky E, Kavun A, Belova E, Grigoreva T, Orlov P, Subbotovskaya A, Shipunov M, Mashkov O, Bilalov F, Shatalov P, Kaprin A, Shegai P, Diuzhev Z, Migiaev O, Vytnova N, Mileyko V, Ivanov M. Untapped Potential of Poly(ADP-Ribose) Polymerase Inhibitors: Lessons Learned From the Real-World Clinical Homologous Recombination Repair Mutation Testing. World J Oncol 2024; 15:562-578. [PMID: 38993246 PMCID: PMC11236374 DOI: 10.14740/wjon1820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/29/2024] [Indexed: 07/13/2024] Open
Abstract
Background Testing for homologous recombination deficiency (HRD) mutations is pivotal to assess individual risk, to proact preventive measures in healthy carriers and to tailor treatments for cancer patients. Increasing prominence of poly(ADP-ribose) polymerase (PARP) inhibitors with remarkable impact on molecular-selected patient survival across diverse nosologies, ingrains testing for BRCA genes and beyond in clinical practice. Nevertheless, testing strategies remain a question of debate. While several pathogenic BRCA1/2 gene variants have been described as founder pathogenic mutations frequently found in patients from Russia, other homologous recombination repair (HRR) genes have not been sufficiently explored. In this study, we present real-world data of routine HRR gene testing in Russia. Methods We evaluated clinical and sequencing data from cancer patients who had germline/somatic next-generation sequencing (NGS) HRR gene testing in Russia (BRCA1/2/ATM/CHEK2, or 15 HRR genes). The primary objectives of this study were to evaluate the frequency of BRCA1/2 and non-BRCA gene mutations in real-world unselected patients from Russia, and to determine whether testing beyond BRCA1/2 is feasible. Results Data of 2,032 patients were collected from February 2021 to February 2023. Most had breast (n = 715, 35.2%), ovarian (n = 259, 12.7%), pancreatic (n = 85, 4.2%), or prostate cancer (n = 58, 2.9%). We observed 586 variants of uncertain significance (VUS) and 372 deleterious variants (DVs) across 487 patients, with 17.6% HRR-mutation positivity. HRR testing identified 120 (11.8%) BRCA1/2-positive, and 172 (16.9%) HRR-positive patients. With 51 DVs identified in 242 formalin-fixed paraffin-embedded (FFPE), testing for variant origin clarification was required in one case (0.4%). Most BRCA1/2 germline variants were DV (121 DVs, 26 VUS); in non-BRCA1/2 genes, VUS were ubiquitous (53 DVs, 132 VUS). In silico prediction identified additional 4.9% HRR and 1.2% BRCA1/2/ATM/CHEK2 mutation patients. Conclusions Our study represents one of the first reports about the incidence of DV and VUS in HRR genes, including genes beyond BRCA1/2, identified in cancer patients from Russia, assessed by NGS. In silico predictions of the observed HRR gene variants suggest that non-BRCA gene testing is likely to result in higher frequency of patients who are candidates for PARP inhibitor therapy. Continuing sequencing efforts should clarify interpretation of frequently observed non-BRCA VUS.
Collapse
Affiliation(s)
- Alexandra Lebedeva
- OncoAtlas LLC, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Egor Veselovsky
- OncoAtlas LLC, Moscow, Russia
- Department of Evolutionary Genetics of Development, Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | | | - Ekaterina Belova
- OncoAtlas LLC, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Tatiana Grigoreva
- OncoAtlas LLC, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Pavel Orlov
- The Federal Research Center for Fundamental and Translational Medicine (NIIECM FRC FTM), Novosibirsk, Russia
| | - Anna Subbotovskaya
- The Federal Research Center for Fundamental and Translational Medicine (NIIECM FRC FTM), Novosibirsk, Russia
| | - Maksim Shipunov
- The Federal Research Center for Fundamental and Translational Medicine (NIIECM FRC FTM), Novosibirsk, Russia
| | - Oleg Mashkov
- State Budgetary Institution of Healthcare Republican Medical Genetic Center, Ufa, Russia
| | - Fanil Bilalov
- State Budgetary Institution of Healthcare Republican Medical Genetic Center, Ufa, Russia
| | - Peter Shatalov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Andrey Kaprin
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Peter Shegai
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | | | | | | | - Vladislav Mileyko
- OncoAtlas LLC, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maxim Ivanov
- OncoAtlas LLC, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia
| |
Collapse
|
25
|
Cassoli JPB, Fernandes Í, Carvalho L, Fernandes M, Centrone AF, Taniwaki L, Lima RDC, Junior UDR, Dias IWR, Taranto P, Beal J, de Lima FT, Moura F, Cendoroglo M, Araújo SEA, Uson Junior PLS. Frequency of Deleterious Germline Variants in HER2-Low Breast Cancer Patients Using a Hereditary Multipanel Gene Testing. Curr Issues Mol Biol 2024; 46:7976-7985. [PMID: 39194688 DOI: 10.3390/cimb46080471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
HER2-Low is defined as low levels of HER2 expression, based on a score of 1+ on immunohistochemical (IHC) assay or as an IHC score of 2+ and negative results on in situ hybridization (ISH or FISH). They are a heterogeneous population of breast cancers that vary in prognosis and sensitivity to systemic treatments. The frequency and clinical characteristics of pathogenic germline variants (PGVs) in HER2-Low breast cancer (BC) patients is not defined. We analyzed results from patients with BC who underwent multi-gene panel testing (MGPT) (maximum 145 genes) between 2018-2019. We reclassified HER-2 status accordingly. Relationships between the variables of interest were assessed by adopting the proportional regression Cox models. Of a total of 167 BC patients who underwent MGPT, half were hormone-receptor-positive. The median age was 45 years. About two thirds of the patients were in the earlier stage of BC. A total of 57% of the cases were reclassified as HER-2-negative or -Low. PGVs were found in 19% of the patients overall, as follows: seven BRCA1, four BRCA2, two ATM, one ATR, two CFTR, three CHEK2, one FANCA, one MERTK, one MLH1, three MUTYH, one RAD50, three RAD51C, one RECQL4, and two TP53 mutations. In HER2-Low, 26.5% of the patients had PGVs, and in the overall cohort, this was 19.8%. In conclusion, differences in the prevalence of deleterious germline mutations in HER2-Low BC patients compared to non-HER2-Low BC patients were identified. Similar alterations in BRCA were observed in this group of patients compared to the overall cohort. Germline genetic tests should be evaluated in larger cohorts of patients with HER2-Low status to better address the findings.
Collapse
Affiliation(s)
- Janaina Pontes Batista Cassoli
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Ítalo Fernandes
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Leonardo Carvalho
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Milena Fernandes
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Ana Fernanda Centrone
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Letícia Taniwaki
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Rita de Cássia Lima
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | | | - Igor Wanderley Reis Dias
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Patrícia Taranto
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Juliana Beal
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Fernanda Teresa de Lima
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Fernando Moura
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Miguel Cendoroglo
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Sergio Eduardo Alonso Araújo
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| | - Pedro Luiz Serrano Uson Junior
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
- Department of Hematology and Oncology, Hospital Israelita Albert Einstein, São Paulo 05652000, Brazil
| |
Collapse
|
26
|
Garmendia D, Weidner A, Venton L, Pal T. Comparing Cancer Risk Management between Females with Truncating CHEK2 1100delC versus Missense CHEK2 I157T Variants. Genes (Basel) 2024; 15:881. [PMID: 39062660 PMCID: PMC11276105 DOI: 10.3390/genes15070881] [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: 05/09/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Breast cancer (BC) risks imparted by CHEK2 c.1100delC ("1100delC") germline pathogenic/likely pathogenic variant (GPV) are 20-30%, compared to CHEK2 c.470T>C ("I157T") GPV with <20%, leading to different breast screening recommendations through MRI. We compared cancer risk management (CRM) across these two GPVs. Study participants were adult females with an 1100delC or I157T GPV drawn from the Inherited Cancer Registry (ICARE) across the United States. Cancer history, clinical characteristics, and CRM were compared using chi-squared tests, t-tests, and logistic regression. Of 150 CHEK2 carriers, 40.7% had BC, with a mean age of 50. Comparing 1100delC and I157T GPVs, there were no differences in rates of (1) breast MRI among those with (65.2% versus 55.6% of 23 and 9; p = 0.612) and without (44.0% versus 44.8% of 50 and 29; p = 0.943) BC; (2) risk-reducing mastectomy among those with (50% versus 38.9% of 46 and 15; p = 0.501) and without (13.8% versus 6.5% of 58 and 31; p = 0.296) BC; and (3) risk-reducing salpingo-oophorectomy among those with (24.2% versus 22.2% of 45 and 18; p = 0.852) and without (17.5% versus 16.7% of 57 and 30; p = 0.918) BC. The results suggest over-screening with breast MRI among CHEK2 I157T GPV carriers and possible overuse of risk-reducing surgeries among CHEK2 carriers.
Collapse
Affiliation(s)
- Diego Garmendia
- Vanderbilt University Medical Center, 1500 21st Ave. So., Suite 2810, Nashville, TN 37212, USA; (D.G.); (A.W.); (L.V.)
| | - Anne Weidner
- Vanderbilt University Medical Center, 1500 21st Ave. So., Suite 2810, Nashville, TN 37212, USA; (D.G.); (A.W.); (L.V.)
| | - Lindsay Venton
- Vanderbilt University Medical Center, 1500 21st Ave. So., Suite 2810, Nashville, TN 37212, USA; (D.G.); (A.W.); (L.V.)
| | - Tuya Pal
- Vanderbilt University Medical Center, 1500 21st Ave. So., Suite 2810, Nashville, TN 37212, USA; (D.G.); (A.W.); (L.V.)
- Vanderbilt-Ingram Cancer Center, 2220 Pierce Ave, Nashville, TN 37232, USA
| |
Collapse
|
27
|
Zhang X, Chen R, Huo Z, Li W, Jiang M, Su G, Liu Y, Cai Y, Huang W, Xiong Y, Wang S. Blood-based molecular and cellular biomarkers of early response to neoadjuvant PD-1 blockade in patients with non-small cell lung cancer. Cancer Cell Int 2024; 24:225. [PMID: 38951894 PMCID: PMC11218110 DOI: 10.1186/s12935-024-03412-3] [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: 02/23/2024] [Accepted: 06/22/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Despite the improved survival observed in PD-1/PD-L1 blockade therapy, a substantial proportion of cancer patients, including those with non-small cell lung cancer (NSCLC), still lack a response. METHODS Transcriptomic profiling was conducted on a discovery cohort comprising 100 whole blood samples, as collected multiple times from 48 healthy controls (including 43 published data) and 31 NSCLC patients that under treatment with a combination of anti-PD-1 Tislelizumab and chemotherapy. Differentially expressed genes (DEGs), simulated immune cell subsets, and germline DNA mutational markers were identified from patients achieved a pathological complete response during the early treatment cycles. The predictive values of mutational markers were further validated in an independent immunotherapy cohort of 1661 subjects, and then confirmed in genetically matched lung cancer cell lines by a co-culturing model. RESULTS The gene expression of hundreds of DEGs (FDR p < 0.05, fold change < -2 or > 2) distinguished responders from healthy controls, indicating the potential to stratify patients utilizing early on-treatment features from blood. PD-1-mediated cell abundance changes in memory CD4 + and regulatory T cell subset were more significant or exclusively observed in responders. A panel of top-ranked genetic alterations showed significant associations with improved survival (p < 0.05) and heightened responsiveness to anti-PD-1 treatment in patient cohort and co-cultured cell lines. CONCLUSION This study discovered and validated peripheral blood-based biomarkers with evident predictive efficacy for early therapy response and patient stratification before treatment for neoadjuvant PD-1 blockade in NSCLC patients.
Collapse
Affiliation(s)
- Xi Zhang
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 710069, Shaanxi, Xi'an, China.
| | - Rui Chen
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Zirong Huo
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Wenqing Li
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Mengju Jiang
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Guodong Su
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Yuru Liu
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Yu Cai
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Wuhao Huang
- Department of Lung Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, 300060, China
| | - Yuyan Xiong
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 710069, Shaanxi, Xi'an, China
| | - Shengguang Wang
- Department of Lung Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, 300060, China.
| |
Collapse
|
28
|
Li J, He P, Cai Q, Chen L, Wang Y, Cai W, Qiu Y, Liu S, Guo W, Chen M, Lin Y, Wang C, Fu F. Spectrum and characteristics of germline PALB2 pathogenic variants in 1556 early-onset breast cancer patients in China. J Cancer Res Clin Oncol 2024; 150:322. [PMID: 38914840 PMCID: PMC11196361 DOI: 10.1007/s00432-024-05758-7] [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: 02/12/2024] [Accepted: 04/22/2024] [Indexed: 06/26/2024]
Abstract
PURPOSE Limited data are available regarding the partner and localizer of BRCA2 (PALB2) in Chinese patients with early breast cancer. This study aimed to assess the spectrum and characteristics of germline PALB2 pathogenic variants in this population. METHODS Peripheral blood samples were collected from 1556 patients diagnosed with BRCA1/2-negative early-onset breast cancer. All coding regions and exon‒intron boundaries of the PALB2 genes were screened through next-generation sequencing. RESULTS The prevalence of PALB2 pathogenic variants was approximately 0.77% in the cohort. Eleven PALB2 pathogenic variants were identified in twelve participants, including five frameshift mutations and six nonsense mutations. All other variants were detected once, except for PALB2 c.1056_1057del (detected twice). Two PALB2 carriers (2/12, 16.7%) have documented family history of breast cancer and/or ovarian cancer. Patients with a positive family history exhibited a threefold higher possibility of being identified as PALB2 carriers than those without a family history (2% vs. 0.69%), although the difference was not statistically significant (p = 0.178). Compared to non-carriers, PALB2 carriers has a tendency to appear in younger age (≤ 30 years) (25% vs 14.4%), human epidermal growth factor receptor-2 (HER2)-negative status (83.3% vs. 70.2%), and diagnosed with invasive micropapillary carcinoma (16.7% vs 3.1%). CONCLUSION The prevalence of the germline PALB2 pathogenic variants was approximately 0.77% in Chinese patients with BRCA1/2-negative early-onset breast cancer. Our findings is crucial for understanding population-specific genetic risks and offering insights that can enhance genetic counseling and genetic testing strategies in this population.
Collapse
Affiliation(s)
- Jing Li
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Peng He
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Qindong Cai
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Lili Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Yali Wang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Weifeng Cai
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Yibin Qiu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Shunyi Liu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Wenhui Guo
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Minyan Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Yuxiang Lin
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Chuan Wang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China.
- Breast Cancer Institute, Fujian Medical University, Fuzhou, 350001, Fujian Province, China.
| | - Fangmeng Fu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| |
Collapse
|
29
|
de Baumont AC, Cadore NA, Pedrotti LG, Curzel GD, Schuch JB, Bessel M, Bordignon C, Rosa ML, Macedo GDS, Rosa DD. Germline rare variants in HER2-positive breast cancer predisposition: a systematic review and meta-analysis. Front Oncol 2024; 14:1395970. [PMID: 38978731 PMCID: PMC11228612 DOI: 10.3389/fonc.2024.1395970] [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] [Received: 03/04/2024] [Accepted: 06/04/2024] [Indexed: 07/10/2024] Open
Abstract
Introduction Approximately 10% of breast cancer (BC) cases result from hereditary causes. Genetic testing has been widely implemented in BC care to determine hereditary cancer syndromes and personalized medicine. Thus, identification of individuals carrying germline pathogenic variants could be useful to provide appropriate prophylactic or screening measures for each BC subtype, however, there are few formal recommendations for genetic testing in this sense so far. In this study, we assessed rare germline variants in a specific group of genes in order to determine the association with human epidermal growth factor 2 enriched (HER2+) BC phenotype through a systematic review and meta-analysis comparing subtypes overexpressing HER2 with other clinically recognized subtypes of BC. This review was registered with PROSPERO (ID: CRD42023447571). Methods We conducted an online literature search in PubMed (MEDLINE), Scopus, and EMBASE databases. We included original studies that investigated germline variants in HER2+ BC patients and selected the studies that reported only rare and/or pathogenic germline variants. We assessed the risk of bias and quality of the studies using the Joanna Briggs Institute Critical Appraisal checklists and the Modified Newcastle-Ottawa Scale for Genetic Studies, respectively. Considering hormone receptor and HER2 expression status, we compared gene-based risks initially in HR-HER2-, HR+HER2-, HR+HER2+, and HR-HER2+ groups, conducting separate meta-analyses using the random effects model for each comparison, and within them for each gene. Results Of the total 36 studies describing germline variants, 11 studies provided information on the prevalence of variants in the different clinically relevant BC subtypes and allowed comparisons. Germline variants within eight genes showed significant differences when meta-analyzed between the BC groups: BRCA1, BRCA2, TP53, ATM, CHEK2, PALB2, RAD51C, and BARD1. Notably, TP53, ATM, and CHEK2 germline variants were identified as predisposing factors for HER2+ subtypes, whereas BRCA1, BRCA2, PALB2, RAD51C, and BARD1 germline variants were associated with a predisposition to low HER2 expression. Main concerns about bias and quality assessment were the lack of confounding factors control; and comparability or outcome assessment, respectively. Discussion Our findings underscore the connection between germline variants and differential expression of the HER2 protein and BC subtypes. Systematic review registration https://www.crd.york.ac.uk/PROSPERO, identifier CRD42023447571.
Collapse
Affiliation(s)
| | - Nathan Araujo Cadore
- Responsabilidade Social, Hospital Moinhos de Vento, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | | | - Marina Bessel
- Responsabilidade Social, Hospital Moinhos de Vento, Porto Alegre, RS, Brazil
| | - Cláudia Bordignon
- Responsabilidade Social, Hospital Moinhos de Vento, Porto Alegre, RS, Brazil
| | - Mahira Lopes Rosa
- Responsabilidade Social, Hospital Moinhos de Vento, Porto Alegre, RS, Brazil
| | | | - Daniela Dornelles Rosa
- Responsabilidade Social, Hospital Moinhos de Vento, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
30
|
Laws A, Leonard S, Hershey E, Stokes S, Vincuilla J, Sharma E, Milliron K, Garber JE, Merajver SD, King TA, Pilewskie ML. Upgrade Rates and Breast Cancer Development Among Germline Pathogenic Variant Carriers with High-Risk Breast Lesions. Ann Surg Oncol 2024; 31:3120-3127. [PMID: 38261128 DOI: 10.1245/s10434-024-14947-0] [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: 07/10/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024]
Abstract
BACKGROUND High-risk lesions (HRL) of the breast are risk factors for future breast cancer development and may be associated with a concurrent underlying malignancy when identified on needle biopsy; however, there are few data evaluating HRLs in carriers of germline pathogenic variants (PVs) in breast cancer predisposition genes. METHODS We identified patients from two institutions with germline PVs in high- and moderate-penetrance breast cancer predisposition genes and an HRL in an intact breast, including atypical ductal hyperplasia (ADH), flat epithelial atypia (FEA), and lobular neoplasia (LN). We calculated upgrade rates at surgical excision and used Kaplan-Meier methods to characterize 3-year breast cancer risk in patients without upgrade. RESULTS Of 117 lesions in 105 patients, 65 (55.6%) were ADH, 48 (41.0%) were LN, and 4 (3.4%) were FEA. Most PVs (83.8%) were in the BRCA1/2, CHEK2 and ATM genes. ADH and FEA were excised in most cases (87.1%), with upgrade rates of 11.8% (95% confidence interval [CI] 5.5-23.4%) and 0%, respectively. LN was selectively excised (53.8%); upgrade rate in the excision group was 4.8% (95% CI 0.8-22.7%), and with 20 months of median follow-up, no same-site cancers developed in the observation group. Among those not upgraded, the 3-year risk of breast cancer development was 13.1% (95% CI 6.3-26.3%), mostly estrogen receptor-positive (ER +) disease (89.5%). CONCLUSIONS Upgrade rates for HRLs in patients with PVs in breast cancer predisposition genes appear similar to non-carriers. HRLs may be associated with increased short-term ER+ breast cancer risk in PV carriers, warranting strong consideration of surgical or chemoprevention therapies in this population.
Collapse
Affiliation(s)
- Alison Laws
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Saskia Leonard
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Emma Hershey
- Department of Surgery, Michigan Medicine, Ann Arbor, MI, USA
| | - Samantha Stokes
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Julie Vincuilla
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Eshita Sharma
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Kara Milliron
- Breast and Ovarian Cancer Risk Evaluation Program, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI, USA
| | - Judy E Garber
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sofia D Merajver
- Breast and Ovarian Cancer Risk Evaluation Program, Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI, USA
| | - Tari A King
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
31
|
Irelli A, Patruno LV, Chiatamone Ranieri S, Di Giacomo D, Malatesta S, Alesse E, Tessitore A, Cannita K. Role of Breast Cancer Risk Estimation Models to Identify Women Eligible for Genetic Testing and Risk-Reducing Surgery. Biomedicines 2024; 12:714. [PMID: 38672070 PMCID: PMC11048717 DOI: 10.3390/biomedicines12040714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Hereditary breast and ovarian cancer (HBOC) syndrome is responsible for approximately 10% of breast cancers (BCs). The HBOC gene panel includes both high-risk genes, i.e., a four times higher risk of BC (BRCA1, BRCA2, PALB2, CDH1, PTEN, STK11 and TP53), and moderate-risk genes, i.e., a two to four times higher risk of BC (BARD1, CHEK2, RAD51C, RAD51D and ATM). Pathogenic germline variants (PGVs) in HBOC genes confer an absolute risk of BC that changes according to the gene considered. We illustrate and compare different BC risk estimation models, also describing their limitations. These models allow us to identify women eligible for genetic testing and possibly to offer surgical strategies for primary prevention, i.e., risk-reducing mastectomies and salpingo-oophorectomies.
Collapse
Affiliation(s)
- Azzurra Irelli
- Medical Oncology Unit, Department of Oncology, “Giuseppe Mazzini” Hospital, AUSL 04 Teramo, 64100 Teramo, Italy; (L.V.P.); (K.C.)
| | - Leonardo Valerio Patruno
- Medical Oncology Unit, Department of Oncology, “Giuseppe Mazzini” Hospital, AUSL 04 Teramo, 64100 Teramo, Italy; (L.V.P.); (K.C.)
| | - Sofia Chiatamone Ranieri
- Pathology Unit, Department of Services, AUSL 04 Teramo, 64100 Teramo, Italy; (S.C.R.); (D.D.G.); (S.M.)
| | - Daniela Di Giacomo
- Pathology Unit, Department of Services, AUSL 04 Teramo, 64100 Teramo, Italy; (S.C.R.); (D.D.G.); (S.M.)
| | - Sara Malatesta
- Pathology Unit, Department of Services, AUSL 04 Teramo, 64100 Teramo, Italy; (S.C.R.); (D.D.G.); (S.M.)
| | - Edoardo Alesse
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.A.); (A.T.)
| | - Alessandra Tessitore
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.A.); (A.T.)
| | - Katia Cannita
- Medical Oncology Unit, Department of Oncology, “Giuseppe Mazzini” Hospital, AUSL 04 Teramo, 64100 Teramo, Italy; (L.V.P.); (K.C.)
| |
Collapse
|
32
|
McClellan JC, Li JL, Gao G, Huo D. Expression- and splicing-based multi-tissue transcriptome-wide association studies identified multiple genes for breast cancer by estrogen-receptor status. Breast Cancer Res 2024; 26:51. [PMID: 38515142 PMCID: PMC10958972 DOI: 10.1186/s13058-024-01809-6] [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: 09/22/2023] [Accepted: 03/14/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Although several transcriptome-wide association studies (TWASs) have been performed to identify genes associated with overall breast cancer (BC) risk, only a few TWAS have explored the differences in estrogen receptor-positive (ER+) and estrogen receptor-negative (ER-) breast cancer. Additionally, these studies were based on gene expression prediction models trained primarily in breast tissue, and they did not account for alternative splicing of genes. METHODS In this study, we utilized two approaches to perform multi-tissue TWASs of breast cancer by ER subtype: (1) an expression-based TWAS that combined TWAS signals for each gene across multiple tissues and (2) a splicing-based TWAS that combined TWAS signals of all excised introns for each gene across tissues. To perform this TWAS, we utilized summary statistics for ER + BC from the Breast Cancer Association Consortium (BCAC) and for ER- BC from a meta-analysis of BCAC and the Consortium of Investigators of Modifiers of BRCA1 and BRCA2 (CIMBA). RESULTS In total, we identified 230 genes in 86 loci that were associated with ER + BC and 66 genes in 29 loci that were associated with ER- BC at a Bonferroni threshold of significance. Of these genes, 2 genes associated with ER + BC at the 1q21.1 locus were located at least 1 Mb from published GWAS hits. For several well-studied tumor suppressor genes such as TP53 and CHEK2 which have historically been thought to impact BC risk through rare, penetrant mutations, we discovered that common variants, which modulate gene expression, may additionally contribute to ER + or ER- etiology. CONCLUSIONS Our study comprehensively examined how differences in common variation contribute to molecular differences between ER + and ER- BC and introduces a novel, splicing-based framework that can be used in future TWAS studies.
Collapse
Affiliation(s)
- Julian C McClellan
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
| | - James L Li
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
| | - Guimin Gao
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA.
| | - Dezheng Huo
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA.
- Section of Hematology & Oncology, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
33
|
Rodriguez J, Grassmann F, Xiao Q, Eriksson M, Mao X, Bajalica-Lagercrantz S, Hall P, Czene K. Investigation of Genetic Alterations Associated With Interval Breast Cancer. JAMA Oncol 2024; 10:372-379. [PMID: 38270937 PMCID: PMC10811589 DOI: 10.1001/jamaoncol.2023.6287] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/16/2023] [Indexed: 01/26/2024]
Abstract
Importance Breast cancers (BCs) diagnosed between 2 screening examinations are called interval cancers (ICs), and they have worse clinicopathological characteristics and poorer prognosis than screen-detected cancers (SDCs). However, the association of rare germline genetic variants with IC have not been studied. Objective To evaluate whether rare germline deleterious protein-truncating variants (PTVs) can be applied to discriminate between IC and SDC while considering mammographic density. Design, Setting, and Participants This population-based genetic association study was based on women aged 40 to 76 years who were attending mammographic screening in Sweden. All women with a diagnosis of BC between January 2001 and January 2016 were included, together with age-matched controls. Patients with BC were followed up for survival until 2021. Statistical analysis was performed from September 2021 to December 2022. Exposure Germline PTVs in 34 BC susceptibility genes as analyzed by targeted sequencing. Main Outcomes and Measures Odds ratios (ORs) were used to compare IC with SDC using logistic regression. Hazard ratios were used to investigate BC-specific survival using Cox regression. Results All 4121 patients with BC (IC, n = 1229; SDC, n = 2892) were female, with a mean (SD) age of 55.5 (7.1) years. There were 5631 age-matched controls. The PTVs of the ATM, BRCA1, BRCA2, CHEK2, and PALB2 genes were more common in patients with IC compared with SDC (OR, 1.48; 95% CI, 1.06-2.05). This association was primarily influenced by BRCA1/2 and PALB2 variants. A family history of BC together with PTVs of any of these genes synergistically increased the probability of receiving a diagnosis of IC rather than SDC (OR, 3.95; 95% CI, 1.97-7.92). Furthermore, 10-year BC-specific survival revealed that if a patient received a diagnosis of an IC, carriers of PTVs in any of these 5 genes had significantly worse survival compared with patients not carrying any of them (hazard ratio, 2.04; 95% CI, 1.06-3.92). All of these associations were further pronounced in a subset of patients with IC who had a low mammographic density at prior screening examination. Conclusions and Relevance The results of this study may be helpful in future optimizations of screening programs that aim to lower mortality as well as the clinical treatment of patients with BC.
Collapse
Affiliation(s)
- Juan Rodriguez
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Felix Grassmann
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Health and Medical University, Potsdam, Germany
| | - Qingyang Xiao
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Eriksson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Xinhe Mao
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Södersjukhuset, Stockholm, Sweden
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Wei X, Sun L, Slade E, Fierheller CT, Oxley S, Kalra A, Sia J, Sideris M, McCluggage WG, Bromham N, Dworzynski K, Rosenthal AN, Brentnall A, Duffy S, Evans DG, Yang L, Legood R, Manchanda R. Cost-Effectiveness of Gene-Specific Prevention Strategies for Ovarian and Breast Cancer. JAMA Netw Open 2024; 7:e2355324. [PMID: 38334999 PMCID: PMC10858404 DOI: 10.1001/jamanetworkopen.2023.55324] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/16/2023] [Indexed: 02/10/2024] Open
Abstract
Importance Pathogenic variants (PVs) in BRCA1, BRCA2, PALB2, RAD51C, RAD51D, and BRIP1 cancer susceptibility genes (CSGs) confer an increased ovarian cancer (OC) risk, with BRCA1, BRCA2, PALB2, RAD51C, and RAD51D PVs also conferring an elevated breast cancer (BC) risk. Risk-reducing surgery, medical prevention, and BC surveillance offer the opportunity to prevent cancers and deaths, but their cost-effectiveness for individual CSGs remains poorly addressed. Objective To estimate the cost-effectiveness of prevention strategies for OC and BC among individuals carrying PVs in the previously listed CSGs. Design, Setting, and Participants In this economic evaluation, a decision-analytic Markov model evaluated the cost-effectiveness of risk-reducing salpingo-oophorectomy (RRSO) and, where relevant, risk-reducing mastectomy (RRM) compared with nonsurgical interventions (including BC surveillance and medical prevention for increased BC risk) from December 1, 2022, to August 31, 2023. The analysis took a UK payer perspective with a lifetime horizon. The simulated cohort consisted of women aged 30 years who carried BRCA1, BRCA2, PALB2, RAD51C, RAD51D, or BRIP1 PVs. Appropriate sensitivity and scenario analyses were performed. Exposures CSG-specific interventions, including RRSO at age 35 to 50 years with or without BC surveillance and medical prevention (ie, tamoxifen or anastrozole) from age 30 or 40 years, RRM at age 30 to 40 years, both RRSO and RRM, BC surveillance and medical prevention, or no intervention. Main Outcomes and Measures The incremental cost-effectiveness ratio (ICER) was calculated as incremental cost per quality-adjusted life-year (QALY) gained. OC and BC cases and deaths were estimated. Results In the simulated cohort of women aged 30 years with no cancer, undergoing both RRSO and RRM was most cost-effective for individuals carrying BRCA1 (RRM at age 30 years; RRSO at age 35 years), BRCA2 (RRM at age 35 years; RRSO at age 40 years), and PALB2 (RRM at age 40 years; RRSO at age 45 years) PVs. The corresponding ICERs were -£1942/QALY (-$2680/QALY), -£89/QALY (-$123/QALY), and £2381/QALY ($3286/QALY), respectively. RRSO at age 45 years was cost-effective for RAD51C, RAD51D, and BRIP1 PV carriers compared with nonsurgical strategies. The corresponding ICERs were £962/QALY ($1328/QALY), £771/QALY ($1064/QALY), and £2355/QALY ($3250/QALY), respectively. The most cost-effective preventive strategy per 1000 PV carriers could prevent 923 OC and BC cases and 302 deaths among those carrying BRCA1; 686 OC and BC cases and 170 deaths for BRCA2; 464 OC and BC cases and 130 deaths for PALB2; 102 OC cases and 64 deaths for RAD51C; 118 OC cases and 76 deaths for RAD51D; and 55 OC cases and 37 deaths for BRIP1. Probabilistic sensitivity analysis indicated both RRSO and RRM were most cost-effective in 96.5%, 89.2%, and 84.8% of simulations for BRCA1, BRCA2, and PALB2 PVs, respectively, while RRSO was cost-effective in approximately 100% of simulations for RAD51C, RAD51D, and BRIP1 PVs. Conclusions and Relevance In this cost-effectiveness study, RRSO with or without RRM at varying optimal ages was cost-effective compared with nonsurgical strategies for individuals who carried BRCA1, BRCA2, PALB2, RAD51C, RAD51D, or BRIP1 PVs. These findings support personalizing risk-reducing surgery and guideline recommendations for individual CSG-specific OC and BC risk management.
Collapse
Affiliation(s)
- Xia Wei
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - Li Sun
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - Eric Slade
- National Institute for Health and Care Excellence, London, United Kingdom
| | - Caitlin T. Fierheller
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - Samuel Oxley
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
- Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom
| | - Ashwin Kalra
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
- Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom
| | - Jacqueline Sia
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
- Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom
| | - Michail Sideris
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
- Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom
| | - W. Glenn McCluggage
- Department of Pathology, Belfast Health & Social Care Trust, Royal Victoria Hospital, Belfast, United Kingdom
| | - Nathan Bromham
- National Institute for Health and Care Excellence, London, United Kingdom
| | | | - Adam N. Rosenthal
- Department of Gynaecology, University College London Hospitals NHS Foundation trust, London, United Kingdom
- Department of Women’s Cancer, UCL EGA Institute for Women’s Health, University College London, London, United Kingdom
| | - Adam Brentnall
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - Stephen Duffy
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - D. Gareth Evans
- Manchester Centre for Genomic Medicine, Division of Evolution, Infection and Genomic Sciences, University of Manchester, MAHSC, Manchester, United Kingdom
| | - Li Yang
- School of Public Health, Peking University, Beijing, China
| | - Rosa Legood
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - Ranjit Manchanda
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
- Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials & Methodology, Faculty of Population Health Sciences, University College London, London, United Kingdom
| |
Collapse
|
35
|
Agaoglu NB, Unal B, Hayes CP, Walker M, Ng OH, Doganay L, Can ND, Rana HQ, Ghazani AA. Genomic disparity impacts variant classification of cancer susceptibility genes in Turkish breast cancer patients. Cancer Med 2024; 13:e6852. [PMID: 38308423 PMCID: PMC10905328 DOI: 10.1002/cam4.6852] [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: 05/20/2023] [Revised: 11/23/2023] [Accepted: 12/10/2023] [Indexed: 02/04/2024] Open
Abstract
OBJECTIVE Turkish genome is underrepresented in large genomic databases. This study aims to evaluate the effect of allele frequency in the Turkish population in determining the clinical utility of germline findings in breast cancer, including invasive lobular carcinoma (ILC), mixed invasive ductal and lobular carcinoma (IDC-L), and ductal carcinoma (DC). METHODS Two clinic-based cohorts from the Umraniye Research and Training Hospital (URTH) were used in this study: a cohort consisting of 132 women with breast cancer and a non-cancer cohort consisting of 492 participants. The evaluation of the germline landscape was performed by analysis of 27 cancer genes. The frequency and type of variants in the breast cancer cohort were compared to those in the non-cancer cohort to investigate the effect of population genetics. The variant allele frequencies in Turkish Variome and gnomAD were statistically evaluated. RESULTS The genetic analysis identified 121 variants in the breast cancer cohort (actionable = 32, VUS = 89) and 223 variants in the non-cancer cohort (actionable = 25, VUS = 188). The occurrence of 21 variants in both suggested a possible genetic population effect. Evaluation of allele frequency of 121 variants from the breast cancer cohort showed 22% had a significantly higher value in Turkish Variome compared to gnomAD (p < 0.0001, 95% CI) with a mean difference of 60 times (ranging from 1.37-354.4). After adjusting for variant allele frequency using the ancestry-appropriate database, 6.7% (5/75) of VUS was reclassified to likely benign. CONCLUSION To our knowledge, this is the first study of population genetic effects in breast cancer subtypes in Turkish women. Our findings underscore the need for a large genomic database representing Turkish population-specific variants. It further highlights the significance of the ancestry-appropriate population database for accurate variant assessment in clinical settings.
Collapse
Affiliation(s)
- Nihat B. Agaoglu
- Department of Medical Genetics, Division of Cancer GeneticsUmraniye Training and Research HospitalIstanbulTurkey
| | - Busra Unal
- Department of Medical Genetics, Division of Cancer GeneticsUmraniye Training and Research HospitalIstanbulTurkey
- Division of GeneticsBrigham and Women's HospitalBostonMassachusettsUSA
| | - Connor P. Hayes
- Division of GeneticsBrigham and Women's HospitalBostonMassachusettsUSA
| | - McKenzie Walker
- Division of GeneticsBrigham and Women's HospitalBostonMassachusettsUSA
| | - Ozden Hatirnaz Ng
- Department of Medical Biology, School of MedicineAcibadem UniversityIstanbulTurkey
| | - Levent Doganay
- Department of Medical Genetics, Division of Cancer GeneticsUmraniye Training and Research HospitalIstanbulTurkey
| | - Nisan D. Can
- Department of Molecular Biology Genetics and BiotechnologyIstanbul Technical UniversityIstanbulTurkey
| | - Huma Q. Rana
- Division of Cancer Genetics and PreventionDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of MedicineBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Arezou A. Ghazani
- Division of GeneticsBrigham and Women's HospitalBostonMassachusettsUSA
- Department of MedicineBrigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
36
|
Caputo A, Vipparthi K, Bazeley P, Downs-Kelly E, McIntire P, Duckworth LA, Ni Y, Hu B, Keri RA, Karaayvaz M. Spatial Transcriptomics Suggests That Alterations Occur in the Preneoplastic Breast Microenvironment of BRCA1/2 Mutation Carriers. Mol Cancer Res 2024; 22:169-180. [PMID: 37878345 PMCID: PMC10872731 DOI: 10.1158/1541-7786.mcr-23-0489] [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: 06/19/2023] [Revised: 08/28/2023] [Accepted: 10/23/2023] [Indexed: 10/26/2023]
Abstract
Breast cancer is the most common cancer in females, affecting one in every eight women and accounting for the majority of cancer-related deaths in women worldwide. Germline mutations in the BRCA1 and BRCA2 genes are significant risk factors for specific subtypes of breast cancer. BRCA1 mutations are associated with basal-like breast cancers, whereas BRCA2 mutations are associated with luminal-like disease. Defects in mammary epithelial cell differentiation have been previously recognized in germline BRCA1/2 mutation carriers even before cancer incidence. However, the underlying mechanism is largely unknown. Here, we employ spatial transcriptomics to investigate defects in mammary epithelial cell differentiation accompanied by distinct microenvironmental alterations in preneoplastic breast tissues from BRCA1/2 mutation carriers and normal breast tissues from noncarrier controls. We uncovered spatially defined receptor-ligand interactions in these tissues for the investigation of autocrine and paracrine signaling. We discovered that β1-integrin-mediated autocrine signaling in BRCA2-deficient mammary epithelial cells may differ from BRCA1-deficient mammary epithelial cells. In addition, we found that the epithelial-to-stromal paracrine signaling in the breast tissues of BRCA1/2 mutation carriers is greater than in control tissues. More integrin-ligand pairs were differentially correlated in BRCA1/2-mutant breast tissues than noncarrier breast tissues with more integrin receptor-expressing stromal cells. IMPLICATIONS These results suggest alterations in the communication between mammary epithelial cells and the microenvironment in BRCA1 and BRCA2 mutation carriers, laying the foundation for designing innovative breast cancer chemo-prevention strategies for high-risk patients.
Collapse
Affiliation(s)
- Anthony Caputo
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kavya Vipparthi
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Peter Bazeley
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Erinn Downs-Kelly
- Department of Pathology, Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Patrick McIntire
- Department of Pathology, Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lauren A. Duckworth
- Department of Pathology, Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ying Ni
- Center for Immunotherapy & Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bo Hu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ruth A. Keri
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Mihriban Karaayvaz
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
37
|
Valentini V, Bucalo A, Conti G, Celli L, Porzio V, Capalbo C, Silvestri V, Ottini L. Gender-Specific Genetic Predisposition to Breast Cancer: BRCA Genes and Beyond. Cancers (Basel) 2024; 16:579. [PMID: 38339330 PMCID: PMC10854694 DOI: 10.3390/cancers16030579] [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: 12/21/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Among neoplastic diseases, breast cancer (BC) is one of the most influenced by gender. Despite common misconceptions associating BC as a women-only disease, BC can also occur in men. Additionally, transgender individuals may also experience BC. Genetic risk factors play a relevant role in BC predisposition, with important implications in precision prevention and treatment. The genetic architecture of BC susceptibility is similar in women and men, with high-, moderate-, and low-penetrance risk variants; however, some sex-specific features have emerged. Inherited high-penetrance pathogenic variants (PVs) in BRCA1 and BRCA2 genes are the strongest BC genetic risk factor. BRCA1 and BRCA2 PVs are more commonly associated with increased risk of female and male BC, respectively. Notably, BRCA-associated BCs are characterized by sex-specific pathologic features. Recently, next-generation sequencing technologies have helped to provide more insights on the role of moderate-penetrance BC risk variants, particularly in PALB2, CHEK2, and ATM genes, while international collaborative genome-wide association studies have contributed evidence on common low-penetrance BC risk variants, on their combined effect in polygenic models, and on their role as risk modulators in BRCA1/2 PV carriers. Overall, all these studies suggested that the genetic basis of male BC, although similar, may differ from female BC. Evaluating the genetic component of male BC as a distinct entity from female BC is the first step to improve both personalized risk assessment and therapeutic choices of patients of both sexes in order to reach gender equality in BC care. In this review, we summarize the latest research in the field of BC genetic predisposition with a particular focus on similarities and differences in male and female BC, and we also discuss the implications, challenges, and open issues that surround the establishment of a gender-oriented clinical management for BC.
Collapse
Affiliation(s)
- Virginia Valentini
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.V.); (A.B.); (G.C.); (L.C.); (V.P.); (C.C.); (V.S.)
| | - Agostino Bucalo
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.V.); (A.B.); (G.C.); (L.C.); (V.P.); (C.C.); (V.S.)
| | - Giulia Conti
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.V.); (A.B.); (G.C.); (L.C.); (V.P.); (C.C.); (V.S.)
| | - Ludovica Celli
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.V.); (A.B.); (G.C.); (L.C.); (V.P.); (C.C.); (V.S.)
| | - Virginia Porzio
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.V.); (A.B.); (G.C.); (L.C.); (V.P.); (C.C.); (V.S.)
| | - Carlo Capalbo
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.V.); (A.B.); (G.C.); (L.C.); (V.P.); (C.C.); (V.S.)
- Medical Oncology Unit, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Valentina Silvestri
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.V.); (A.B.); (G.C.); (L.C.); (V.P.); (C.C.); (V.S.)
| | - Laura Ottini
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (V.V.); (A.B.); (G.C.); (L.C.); (V.P.); (C.C.); (V.S.)
| |
Collapse
|
38
|
Wang Y, Dackus GMHE, Rosenberg EH, Cornelissen S, de Boo LW, Broeks A, Brugman W, Chan TWS, van Diest PJ, Hauptmann M, Ter Hoeve ND, Isaeva OI, de Jong VMT, Jóźwiak K, Kluin RJC, Kok M, Koop E, Nederlof PM, Opdam M, Schouten PC, Siesling S, van Steenis C, Voogd AC, Vreuls W, Salgado RF, Linn SC, Schmidt MK. Long-term outcomes of young, node-negative, chemotherapy-naïve, triple-negative breast cancer patients according to BRCA1 status. BMC Med 2024; 22:9. [PMID: 38191387 PMCID: PMC10775514 DOI: 10.1186/s12916-023-03233-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 12/15/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Due to the abundant usage of chemotherapy in young triple-negative breast cancer (TNBC) patients, the unbiased prognostic value of BRCA1-related biomarkers in this population remains unclear. In addition, whether BRCA1-related biomarkers modify the well-established prognostic value of stromal tumor-infiltrating lymphocytes (sTILs) is unknown. This study aimed to compare the outcomes of young, node-negative, chemotherapy-naïve TNBC patients according to BRCA1 status, taking sTILs into account. METHODS We included 485 Dutch women diagnosed with node-negative TNBC under age 40 between 1989 and 2000. During this period, these women were considered low-risk and did not receive chemotherapy. BRCA1 status, including pathogenic germline BRCA1 mutation (gBRCA1m), somatic BRCA1 mutation (sBRCA1m), and tumor BRCA1 promoter methylation (BRCA1-PM), was assessed using DNA from formalin-fixed paraffin-embedded tissue. sTILs were assessed according to the international guideline. Patients' outcomes were compared using Cox regression and competing risk models. RESULTS Among the 399 patients with BRCA1 status, 26.3% had a gBRCA1m, 5.3% had a sBRCA1m, 36.6% had tumor BRCA1-PM, and 31.8% had BRCA1-non-altered tumors. Compared to BRCA1-non-alteration, gBRCA1m was associated with worse overall survival (OS) from the fourth year after diagnosis (adjusted HR, 2.11; 95% CI, 1.18-3.75), and this association attenuated after adjustment for second primary tumors. Every 10% sTIL increment was associated with 16% higher OS (adjusted HR, 0.84; 95% CI, 0.78-0.90) in gBRCA1m, sBRCA1m, or BRCA1-non-altered patients and 31% higher OS in tumor BRCA1-PM patients. Among the 66 patients with tumor BRCA1-PM and ≥ 50% sTILs, we observed excellent 15-year OS (97.0%; 95% CI, 92.9-100%). Conversely, among the 61 patients with gBRCA1m and < 50% sTILs, we observed poor 15-year OS (50.8%; 95% CI, 39.7-65.0%). Furthermore, gBRCA1m was associated with higher (adjusted subdistribution HR, 4.04; 95% CI, 2.29-7.13) and tumor BRCA1-PM with lower (adjusted subdistribution HR, 0.42; 95% CI, 0.19-0.95) incidence of second primary tumors, compared to BRCA1-non-alteration. CONCLUSIONS Although both gBRCA1m and tumor BRCA1-PM alter BRCA1 gene transcription, they are associated with different outcomes in young, node-negative, chemotherapy-naïve TNBC patients. By combining sTILs and BRCA1 status for risk classification, we were able to identify potential subgroups in this population to intensify and optimize adjuvant treatment.
Collapse
Affiliation(s)
- Yuwei Wang
- Division of Molecular Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Gwen M H E Dackus
- Division of Molecular Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Efraim H Rosenberg
- Division of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sten Cornelissen
- Division of Molecular Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Leonora W de Boo
- Division of Molecular Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wim Brugman
- Genomics Core Facility, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Terry W S Chan
- Division of Molecular Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michael Hauptmann
- Institute of Biostatistics and Registry Research, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Natalie D Ter Hoeve
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Olga I Isaeva
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Vincent M T de Jong
- Division of Molecular Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Katarzyna Jóźwiak
- Institute of Biostatistics and Registry Research, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Roelof J C Kluin
- Genomics Core Facility, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marleen Kok
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Esther Koop
- Department of Pathology, Gelre Ziekenhuizen, Apeldoorn, The Netherlands
| | - Petra M Nederlof
- Division of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Mark Opdam
- Division of Molecular Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Philip C Schouten
- Division of Molecular Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Sabine Siesling
- Department of Research and Development, Netherlands Comprehensive Cancer Organization, Utrecht, The Netherlands
- Department of Health Technology and Services Research, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | | | - Adri C Voogd
- Department of Epidemiology, Maastricht University, Maastricht, The Netherlands
| | - Willem Vreuls
- Department of Pathology, Canisius Wilhelmina Ziekenhuis, Nijmegen, The Netherlands
| | - Roberto F Salgado
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Center, Melbourne, Australia
| | - Sabine C Linn
- Division of Molecular Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marjanka K Schmidt
- Division of Molecular Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
39
|
Joyce R, Pascual R, Heitink L, Capaldo BD, Vaillant F, Christie M, Tsai M, Surgenor E, Anttila CJA, Rajasekhar P, Jackling FC, Trussart M, Milevskiy MJG, Song X, Li M, Teh CE, Gray DHD, Smyth GK, Chen Y, Lindeman GJ, Visvader JE. Identification of aberrant luminal progenitors and mTORC1 as a potential breast cancer prevention target in BRCA2 mutation carriers. Nat Cell Biol 2024; 26:138-152. [PMID: 38216737 DOI: 10.1038/s41556-023-01315-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 11/15/2023] [Indexed: 01/14/2024]
Abstract
Inheritance of a BRCA2 pathogenic variant conveys a substantial life-time risk of breast cancer. Identification of the cell(s)-of-origin of BRCA2-mutant breast cancer and targetable perturbations that contribute to transformation remains an unmet need for these individuals who frequently undergo prophylactic mastectomy. Using preneoplastic specimens from age-matched, premenopausal females, here we show broad dysregulation across the luminal compartment in BRCA2mut/+ tissue, including expansion of aberrant ERBB3lo luminal progenitor and mature cells, and the presence of atypical oestrogen receptor (ER)-positive lesions. Transcriptional profiling and functional assays revealed perturbed proteostasis and translation in ERBB3lo progenitors in BRCA2mut/+ breast tissue, independent of ageing. Similar molecular perturbations marked tumours bearing BRCA2-truncating mutations. ERBB3lo progenitors could generate both ER+ and ER- cells, potentially serving as cells-of-origin for ER-positive or triple-negative cancers. Short-term treatment with an mTORC1 inhibitor substantially curtailed tumorigenesis in a preclinical model of BRCA2-deficient breast cancer, thus uncovering a potential prevention strategy for BRCA2 mutation carriers.
Collapse
Affiliation(s)
- Rachel Joyce
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Rosa Pascual
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Luuk Heitink
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Bianca D Capaldo
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - François Vaillant
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Michael Christie
- Department of Anatomical Pathology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Minhsuang Tsai
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Elliot Surgenor
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Casey J A Anttila
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Pradeep Rajasekhar
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Felicity C Jackling
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Marie Trussart
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Michael J G Milevskiy
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Xiaoyu Song
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Mengbo Li
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Charis E Teh
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Daniel H D Gray
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Gordon K Smyth
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria, Australia
| | - Yunshun Chen
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Geoffrey J Lindeman
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia.
- Parkville Familial Cancer Centre and Department of Medical Oncology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, Victoria, Australia.
| | - Jane E Visvader
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
40
|
Chen C, Lin CJ, Pei YC, Ma D, Liao L, Li SY, Fan L, Di GH, Wu SY, Liu XY, Wang YJ, Hong Q, Zhang GL, Xu LL, Li BB, Huang W, Shi JX, Jiang YZ, Hu X, Shao ZM. Comprehensive genomic profiling of breast cancers characterizes germline-somatic mutation interactions mediating therapeutic vulnerabilities. Cell Discov 2023; 9:125. [PMID: 38114467 PMCID: PMC10730692 DOI: 10.1038/s41421-023-00614-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/08/2023] [Indexed: 12/21/2023] Open
Abstract
Germline-somatic mutation interactions are universal and associated with tumorigenesis, but their role in breast cancer, especially in non-Caucasians, remains poorly characterized. We performed large-scale prospective targeted sequencing of matched tumor-blood samples from 4079 Chinese females, coupled with detailed clinical annotation, to map interactions between germline and somatic alterations. We discovered 368 pathogenic germline variants and identified 5 breast cancer DNA repair-associated genes (BCDGs; BRCA1/BRCA2/CHEK2/PALB2/TP53). BCDG mutation carriers, especially those with two-hit inactivation, demonstrated younger onset, higher tumor mutation burden, and greater clinical benefits from platinum drugs, PARP inhibitors, and immune checkpoint inhibitors. Furthermore, we leveraged a multiomics cohort to reveal that clinical benefits derived from two-hit events are associated with increased genome instability and an immune-activated tumor microenvironment. We also established an ethnicity-specific tool to predict BCDG mutation and two-hit status for genetic evaluation and therapeutic decisions. Overall, this study leveraged the large sequencing cohort of Chinese breast cancers, optimizing genomics-guided selection of DNA damaging-targeted therapy and immunotherapy within a broader population.
Collapse
Affiliation(s)
- Chao Chen
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cai-Jin Lin
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Chen Pei
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ding Ma
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li Liao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Si-Yuan Li
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Fan
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Gen-Hong Di
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Song-Yang Wu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xi-Yu Liu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yun-Jin Wang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qi Hong
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
| | - Guo-Liang Zhang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lin-Lin Xu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
| | - Bei-Bei Li
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wei Huang
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jin-Xiu Shi
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Xin Hu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China.
| |
Collapse
|
41
|
Corso G, Marabelli M, Calvello M, Gandini S, Risti M, Feroce I, Mannucci S, Girardi A, De Scalzi AM, Magnoni F, Marino E, Bernard L, Veronesi P, Guerini-Rocco E, Barberis M, Guerrieri-Gonzaga A, Bonanni B. Germline pathogenic variants in metaplastic breast cancer patients and the emerging role of the BRCA1 gene. Eur J Hum Genet 2023; 31:1275-1282. [PMID: 37460658 PMCID: PMC10620155 DOI: 10.1038/s41431-023-01429-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/29/2023] [Accepted: 07/04/2023] [Indexed: 08/19/2023] Open
Abstract
Metaplastic breast cancer (MpBC) is a rare, aggressive breast cancer (BC) histotype. Scarce information is available about MpBC genetic predisposition. Previous studies, mainly consisting of case reports, retrospective reviews and others on target therapies, pointed to a possible involvement of the BRCA1 gene in increasing MpBC risk, without ever confirming it. In this study, we retrospectively reviewed all BC patients counseled at our Institute for genetic testing of at least BRCA1 or BRCA2 (BRCA) genes and we found that 23 (23/5226 = 0.4%) were affected by MpBC. About 65% (15/23) of MpBC patients harbored a germline pathogenic variant (PV): 13 in BRCA1 (86.7%), including two patients who received genetic testing for known familial PV, one in TP53 (6.7%), and one in MLH1 (6.7%). We observed a statistically different frequency of MpBC in patients who carried a PV in the BRCA genes (13/1114 = 1.2%) vs. all other BC patients (10/4112 = 0.2%) (p = 0.0002). BRCA carriers proved to have an increased risk of developing MpBC compared to all other BC patients who were tested for BRCA genes (OR = 4.47; 95% CI: 1.95-10.23). Notably, MpBCs were diagnosed in 2.1% (13/610) of BRCA1 carriers. No MpBCs were observed in BRCA2 carriers (0/498 = 0%), revealing a statistically significant difference between the prevalence of MpBCs in BRCA1 and BRCA2 carriers (p = 0.0015). Our results confirmed that BRCA1 is involved in MpBC predisposition. Further studies on unselected patients are needed to elucidate the authentic role of BRCA1 and to explore the possible implication of other genes in MpBC predisposition.
Collapse
Affiliation(s)
- Giovanni Corso
- Division of Breast Surgery, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- European Cancer Prevention Organization (ECP), Milan, Italy
| | - Monica Marabelli
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy.
| | - Mariarosaria Calvello
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Sara Gandini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Matilde Risti
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Irene Feroce
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Sara Mannucci
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Antonia Girardi
- Division of Breast Surgery, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Francesca Magnoni
- Division of Breast Surgery, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Elena Marino
- Clinic Unit of Oncogenomics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Loris Bernard
- Clinic Unit of Oncogenomics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Paolo Veronesi
- Division of Breast Surgery, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Elena Guerini-Rocco
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Massimo Barberis
- Clinic Unit of Oncogenomics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Aliana Guerrieri-Gonzaga
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
42
|
Lee EG, Lim J, Ha HI, Lim MC, Chang YJ, Won YJ, Jung SY. Characteristics of second primary breast cancer after ovarian cancer: a Korea central cancer registry retrospective study. Front Oncol 2023; 13:1208320. [PMID: 37781206 PMCID: PMC10539581 DOI: 10.3389/fonc.2023.1208320] [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: 04/18/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
Background Second primary cancer has become an important issue among cancer survivors. This study sought to determine the differences in clinicopathologic outcomes between second primary breast cancer (SPBC) after ovarian cancer and primary breast cancer (PBC) in the Republic of Korea. Methods and materials We searched the Korea Central Cancer Registry and identified 251,244 breast cancer cases that were diagnosed between 1999 and 2017. The incident rate and standardized incidence ratio (SIR) were calculated. Demographic and clinical characteristics and overall survival (OS) rates were estimated according to age, histological type, and cancer stage. Results Among the 228,329 patients included, 228,148 were patients with PBC, and 181 patients had SPBC diagnosed after ovarian cancer (OC). The mean ages at diagnosis were 56.09 ± 10.81 years for SPBC and 50.65 ± 11.40 years for PBC. Patients with SPBC were significantly less likely than patients with PBC to receive adjuvant radiotherapy (14.92% vs. 21.92%, p = 0.02) or adjuvant chemotherapy (44.75% vs. 55.69%, p < 0.01). Based on the age-standardized rate (ASR), the incidence of SPBC after OC was 293.58 per 100,000 ovarian cancer patients and the incidence of PBC was 39.13 per 100,000 women. The SIR for SPBC was 1.27 (1.09-1.46, 95% Confidence interval) in the patients overall. The 5-year OS rates were 72.88% and 89.37% for SPBC and PBC (p < 0.01). The OS rate in SPBC decreased significantly with advanced stage and older age. Conclusion The incidence of breast cancer is about 1.27 times higher in ovarian cancer patients than in healthy people. The survival outcomes were worse for SPBC than for PBC and were related to older age and advanced stage. Active screening for breast cancer is necessary in ovarian cancer patients.
Collapse
Affiliation(s)
- Eun-Gyeong Lee
- Center for Breast Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Jiwon Lim
- Division of Cancer Registration and Surveillance, National Cancer Control Institute, National Cancer Center, Goyang, Republic of Korea
| | - Hyeong In Ha
- Department of Obstetrics and Gynecology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Myong Cheol Lim
- Center for Gynecologic Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
- Department of Cancer Control and Population Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Republic of Korea
- Division of Tumor Immunology, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Yoon Jung Chang
- Department of Cancer Control and Population Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Republic of Korea
- Division of Cancer Control & Policy, National Cancer Control Institute, National Cancer Center, Goyang, Republic of Korea
| | - Young-Joo Won
- Division of Health Administration, Yonsei University, Wonju, Republic of Korea
| | - So-Youn Jung
- Center for Breast Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
- Cancer Healthcare Research Branch, Research Institute, National Cancer Center, Goyang, Republic of Korea
| |
Collapse
|
43
|
Lin PH, Tien YW, Cheng WF, Chiang YC, Wu CH, Yang K, Huang CS. Diverse genetic spectrum among patients who met the criteria of hereditary breast, ovarian and pancreatic cancer syndrome. J Gynecol Oncol 2023; 34:e66. [PMID: 37170728 PMCID: PMC10482589 DOI: 10.3802/jgo.2023.34.e66] [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: 12/31/2022] [Revised: 03/06/2023] [Accepted: 04/16/2023] [Indexed: 05/13/2023] Open
Abstract
OBJECTIVE Genetic high-risk assessment combines hereditary breast, ovarian and pancreatic cancer into one syndrome. However, there is a lack of data for comparing the germline mutational spectrum of the cancer predisposing genes between these three cancers. METHODS Patients who met the criteria of the hereditary breast, ovarian and pancreatic cancer were enrolled and received multi-gene sequencing. RESULTS We enrolled 730 probands: 418 developed breast cancer, 185 had ovarian cancer, and 145 had pancreatic cancer. Out of the 18 patients who had two types of cancer, 16 had breast and ovarian cancer and 2 had breast and pancreatic cancer. A total of 167 (22.9%) patients had 170 mutations. Mutation frequency in breast, ovarian and pancreatic cancer was 22.3%, 33.5% and 17.2%, respectively. The mutation rate was significantly higher in patients with double cancers than those with a single cancer (p<0.001). BRCA1 and BRCA2 were the most dominant genes associated with hereditary breast and ovarian cancer, whereas ATM was the most prevalent gene related to hereditary pancreatic cancer. Genes of hereditary colon cancer such as lynch syndrome were presented in a part of patients with pancreatic or ovarian cancer but seldom in those with breast cancer. Families with a history of both ovarian and breast cancer were associated with a higher mutation rate than those with other histories. CONCLUSION The mutation spectrum varies across the three cancer types and family histories. Our analysis provides guidance for physicians, counsellors, and counselees on the offer and uptake of genetic counseling.
Collapse
Affiliation(s)
- Po-Han Lin
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
- Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yun-Wen Tien
- Department of Surgery, National Taiwan University Hospital and Medical College of the National Taiwan University, Taipei, Taiwan
| | - Wen-Fang Cheng
- Department of Gynecology and Obstetrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Ying-Cheng Chiang
- Department of Gynecology and Obstetrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Chien-Huei Wu
- Department of Surgery, National Taiwan University Hospital and Medical College of the National Taiwan University, Taipei, Taiwan
| | - Karen Yang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Chiun-Sheng Huang
- Department of Surgery, National Taiwan University Hospital and Medical College of the National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
44
|
Butz H, Nagy P, Papp J, Bozsik A, Grolmusz VK, Pócza T, Oláh E, Patócs A. PALB2 Variants Extend the Mutational Profile of Hungarian Patients with Breast and Ovarian Cancer. Cancers (Basel) 2023; 15:4350. [PMID: 37686625 PMCID: PMC10487218 DOI: 10.3390/cancers15174350] [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: 07/26/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND The pathogenic/likely pathogenic (P/LP) variant detection rate and profile of PALB2, the third most important breast cancer gene, may vary between different populations. METHODS PALB2 was analyzed in peripheral blood samples of three independent cohorts: prospectively between September 2021 and March 2023 (i) in 1280 consecutive patients with breast and/or ovarian cancer (HBOC), (ii) in 568 patients with other cancers (controls), and retrospectively, (iii) in 191 young breast cancer (<33 years, yBC) patients. These data were compared with data of 134,187 non-cancer individuals retrieved from the Genome Aggregation Database. RESULTS Altogether, 235 cases (235/1280; 18.3%) carried at least one P/LP variant in one of the HBOC susceptibility genes. P/LP PALB2 variants were identified in 18 patients (1.4%; 18/1280) in the HBOC and 3 cases (1.5%; 3/191) in the yBC group. In the control group, only one patient had a disease-causing PALB2 variant (0.17%; 1/568) as a secondary finding not related to the disease, which was similar (0.15%; 205/134,187) in the non-cancer control group. The NM_024675.4:c.509_510delGA variant was the most common among our patients (33%; 6/18). We did not find a significant difference in the incidence of PALB2 disease-causing variants according to age; however, the median age of tumor onset was lower in PALB2 P/LP carriers versus wild-type patients (44 vs. 48 years). In our cohort, the odds ratio for breast cancer risk in women with PALB2 P/LP variants was between 8.1 and 9.3 compared to non-HBOC cancer patients and the non-cancer population, respectively. CONCLUSIONS PALB2 P/LP variants are not uncommon among breast and/or ovarian cancer patients. Their incidence was the same in the two breast cancer cohorts studied but may occur rarely in patients with non-breast/ovarian cancer. The c.509_510delGA variant is particularly common in the studied Hungarian patient population.
Collapse
Affiliation(s)
- Henriett Butz
- Department of Molecular Genetics, The National Tumor Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Center, 1122 Budapest, Hungary (A.B.); (V.K.G.); (T.P.); (E.O.); (A.P.)
- Department of Oncology Biobank, National Institute of Oncology, 1122 Budapest, Hungary
- Hereditary Tumours Research Group, Eötvös Loránd Research Network, 1089 Budapest, Hungary
- Department of Laboratory Medicine, Semmelweis University, 1092 Budapest, Hungary
| | - Petra Nagy
- Department of Molecular Genetics, The National Tumor Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Center, 1122 Budapest, Hungary (A.B.); (V.K.G.); (T.P.); (E.O.); (A.P.)
| | - János Papp
- Department of Molecular Genetics, The National Tumor Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Center, 1122 Budapest, Hungary (A.B.); (V.K.G.); (T.P.); (E.O.); (A.P.)
- Hereditary Tumours Research Group, Eötvös Loránd Research Network, 1089 Budapest, Hungary
| | - Anikó Bozsik
- Department of Molecular Genetics, The National Tumor Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Center, 1122 Budapest, Hungary (A.B.); (V.K.G.); (T.P.); (E.O.); (A.P.)
- Hereditary Tumours Research Group, Eötvös Loránd Research Network, 1089 Budapest, Hungary
| | - Vince Kornél Grolmusz
- Department of Molecular Genetics, The National Tumor Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Center, 1122 Budapest, Hungary (A.B.); (V.K.G.); (T.P.); (E.O.); (A.P.)
- Hereditary Tumours Research Group, Eötvös Loránd Research Network, 1089 Budapest, Hungary
| | - Tímea Pócza
- Department of Molecular Genetics, The National Tumor Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Center, 1122 Budapest, Hungary (A.B.); (V.K.G.); (T.P.); (E.O.); (A.P.)
| | - Edit Oláh
- Department of Molecular Genetics, The National Tumor Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Center, 1122 Budapest, Hungary (A.B.); (V.K.G.); (T.P.); (E.O.); (A.P.)
| | - Attila Patócs
- Department of Molecular Genetics, The National Tumor Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Center, 1122 Budapest, Hungary (A.B.); (V.K.G.); (T.P.); (E.O.); (A.P.)
- Hereditary Tumours Research Group, Eötvös Loránd Research Network, 1089 Budapest, Hungary
- Department of Laboratory Medicine, Semmelweis University, 1092 Budapest, Hungary
| |
Collapse
|
45
|
Öfverholm A, Törngren T, Rosén A, Arver B, Einbeigi Z, Haraldsson K, Ståhlbom AK, Kuchinskaya E, Lindblom A, Melin B, Paulsson-Karlsson Y, Stenmark-Askmalm M, Tham E, von Wachenfeldt A, Kvist A, Borg Å, Ehrencrona H. Extended genetic analysis and tumor characteristics in over 4600 women with suspected hereditary breast and ovarian cancer. BMC Cancer 2023; 23:738. [PMID: 37563628 PMCID: PMC10413543 DOI: 10.1186/s12885-023-11229-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND Genetic screening for pathogenic variants (PVs) in cancer predisposition genes can affect treatment strategies, risk prediction and preventive measures for patients and families. For decades, hereditary breast and ovarian cancer (HBOC) has been attributed to PVs in the genes BRCA1 and BRCA2, and more recently other rare alleles have been firmly established as associated with a high or moderate increased risk of developing breast and/or ovarian cancer. Here, we assess the genetic variation and tumor characteristics in a large cohort of women with suspected HBOC in a clinical oncogenetic setting. METHODS Women with suspected HBOC referred from all oncogenetic clinics in Sweden over a six-year inclusion period were screened for PVs in 13 clinically relevant genes. The genetic outcome was compared with tumor characteristics and other clinical data collected from national cancer registries and hospital records. RESULTS In 4622 women with breast and/or ovarian cancer the overall diagnostic yield (the proportion of women carrying at least one PV) was 16.6%. BRCA1/2 PVs were found in 8.9% of women (BRCA1 5.95% and BRCA2 2.94%) and PVs in the other breast and ovarian cancer predisposition genes in 8.2%: ATM (1.58%), BARD1 (0.45%), BRIP1 (0.43%), CDH1 (0.11%), CHEK2 (3.46%), PALB2 (0.84%), PTEN (0.02%), RAD51C (0.54%), RAD51D (0.15%), STK11 (0) and TP53 (0.56%). Thus, inclusion of the 11 genes in addition to BRCA1/2 increased diagnostic yield by 7.7%. The yield was, as expected, significantly higher in certain subgroups such as younger patients, medullary breast cancer, higher Nottingham Histologic Grade, ER-negative breast cancer, triple-negative breast cancer and high grade serous ovarian cancer. Age and tumor subtype distributions differed substantially depending on genetic finding. CONCLUSIONS This study contributes to understanding the clinical and genetic landscape of breast and ovarian cancer susceptibility. Extending clinical genetic screening from BRCA1 and BRCA2 to 13 established cancer predisposition genes almost doubles the diagnostic yield, which has implications for genetic counseling and clinical guidelines. The very low yield in the syndrome genes CDH1, PTEN and STK11 questions the usefulness of including these genes on routine gene panels.
Collapse
Affiliation(s)
- Anna Öfverholm
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Therese Törngren
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Anna Rosén
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Brita Arver
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Zakaria Einbeigi
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
- Department of Medicine and Oncology, Southern Älvsborg Hospital, Borås, Sweden
| | - Karin Haraldsson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | - Ekaterina Kuchinskaya
- Department of Clinical Pathology and Clinical Genetics, Department of Clinical Experimental Medicine, Linköping University, Linköping, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Annika Lindblom
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Beatrice Melin
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Ylva Paulsson-Karlsson
- Department of Immunology, Genetics and Pathology, Uppsala University Hospital, Uppsala, Sweden
| | - Marie Stenmark-Askmalm
- Department of Clinical Pathology and Clinical Genetics, Department of Clinical Experimental Medicine, Linköping University, Linköping, Sweden
- Department of Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden
| | - Emma Tham
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Anna von Wachenfeldt
- Department of Clinical Science and Education at Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Södersjukhuset, Stockholm, Sweden
| | - Anders Kvist
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Åke Borg
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Hans Ehrencrona
- Department of Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden.
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
46
|
Evans DG, Sithambaram S, van Veen EM, Burghel GJ, Schlecht H, Harkness EF, Byers H, Ellingford JM, Gandhi A, Howell SJ, Howell A, Forde C, Lalloo F, Newman WG, Smith MJ, Woodward ER. Differential involvement of germline pathogenic variants in breast cancer genes between DCIS and low-grade invasive cancers. J Med Genet 2023; 60:740-746. [PMID: 36442995 DOI: 10.1136/jmg-2022-108790] [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: 06/28/2022] [Accepted: 11/14/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE To investigate frequency of germline pathogenic variants (PVs) in women with ductal carcinoma in situ (DCIS) and grade 1 invasive breast cancer (G1BC). METHODS We undertook BRCA1/2 analysis in 311 women with DCIS and 392 with G1BC and extended panel testing (non-BRCA1/2) in 176/311 with DCIS and 156/392 with G1BC. We investigated PV detection by age at diagnosis, Manchester Score (MS), DCIS grade and receptor status. RESULTS 30/311 (9.6%) with DCIS and 16/392 with G1BC (4.1%) had a BRCA1/2 PV (p=0.003), and 24/176-(13.6%) and 7/156-(4.5%), respectively, a non-BRCA1/2 PV (p=0.004). Increasing MS was associated with increased likelihood of BRCA1/2 PV in both DCIS and G1BC, although the 10% threshold was not predictive for G1GB. 13/32 (40.6%) DCIS and 0/17 with G1BC <40 years had a non-BRCA1/2 PV (p<0.001). 0/16 DCIS G1 had a PV. For G2 and G3 DCIS, PV rates were 10/98 (BRCA1/2) and 9/90 (non-BRCA1/2), and 8/47 (BRCA1/2) and 8/45 (non-BRCA1/2), respectively. 6/9 BRCA1 and 3/26 BRCA2-associated DCIS were oestrogen receptor negative-(p=0.003). G1BC population testing showed no increased PV rate (OR=1.16, 95% CI 0.28 to 4.80). CONCLUSION DCIS is more likely to be associated with both BRCA1/2 and non-BRCA1/2 PVs than G1BC. Extended panel testing ought to be offered in young-onset DCIS where PV detection rates are highest.
Collapse
Affiliation(s)
- D Gareth Evans
- Division of Evolution and Genomic Science, The University of Manchester School of Health Sciences, Manchester, UK
| | - Siva Sithambaram
- Manchester Univerities Hospital NHS Foundation Trust, Manchester, UK
| | - Elke Maria van Veen
- Division of Evolution and Genomic Sciences, The University of Manchester, Manchester, UK
| | | | - Helene Schlecht
- North West Genomic Laboratory Hub, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Elaine F Harkness
- Division of Evolution and Genomic Sciences, The University of Manchester, Manchester, UK
| | - Helen Byers
- Genomic Medicine, The University of Manchester School of Health Sciences, Manchester, UK
| | - Jamie M Ellingford
- Institute of Human Development, The University of Manchester School of Health Sciences, Manchester, UK
| | - Ashu Gandhi
- Prevent Breast Cancer Centre, Wythenshawe Hospital Manchester Universities Foundation Trust, Manchester, UK
| | - Sacha J Howell
- Manchester Univerities Hospital NHS Foundation Trust, Manchester, UK
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Anthony Howell
- Manchester Foundation Trust, Prevent Breast Cancer Centre, Manchester, UK
| | - Claire Forde
- Clinical Genetics Service, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Fiona Lalloo
- Clinical Genetics Service, Manchester Centre for Genomic Medicine, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - William G Newman
- Genetics, The University of Manchester School of Health Sciences, Manchester, UK
| | - Miriam Jane Smith
- Genetic Medicine, The University of Manchester School of Health Sciences, Manchester, UK
| | | |
Collapse
|
47
|
Hu C, Nagaraj AB, Shimelis H, Montalban G, Lee KY, Huang H, Lumby CA, Na J, Susswein LR, Roberts ME, Marshall ML, Hiraki S, LaDuca H, Chao E, Yussuf A, Pesaran T, Neuhausen SL, Haiman CA, Kraft P, Lindstrom S, Palmer JR, Teras LR, Vachon CM, Yao S, Ong I, Nathanson KL, Weitzel JN, Boddicker N, Gnanaolivu R, Polley EC, Mer G, Cui G, Karam R, Richardson ME, Domchek SM, Yadav S, Hruska KS, Dolinsky J, Weroha SJ, Hart SN, Simard J, Masson JY, Pang YP, Couch FJ. Functional and Clinical Characterization of Variants of Uncertain Significance Identifies a Hotspot for Inactivating Missense Variants in RAD51C. Cancer Res 2023; 83:2557-2571. [PMID: 37253112 PMCID: PMC10390864 DOI: 10.1158/0008-5472.can-22-2319] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/07/2022] [Accepted: 05/25/2023] [Indexed: 06/01/2023]
Abstract
Pathogenic protein-truncating variants of RAD51C, which plays an integral role in promoting DNA damage repair, increase the risk of breast and ovarian cancer. A large number of RAD51C missense variants of uncertain significance (VUS) have been identified, but the effects of the majority of these variants on RAD51C function and cancer predisposition have not been established. Here, analysis of 173 missense variants by a homology-directed repair (HDR) assay in reconstituted RAD51C-/- cells identified 30 nonfunctional (deleterious) variants, including 18 in a hotspot within the ATP-binding region. The deleterious variants conferred sensitivity to cisplatin and olaparib and disrupted formation of RAD51C/XRCC3 and RAD51B/RAD51C/RAD51D/XRCC2 complexes. Computational analysis indicated the deleterious variant effects were consistent with structural effects on ATP-binding to RAD51C. A subset of the variants displayed similar effects on RAD51C activity in reconstituted human RAD51C-depleted cancer cells. Case-control association studies of deleterious variants in women with breast and ovarian cancer and noncancer controls showed associations with moderate breast cancer risk [OR, 3.92; 95% confidence interval (95% CI), 2.18-7.59] and high ovarian cancer risk (OR, 14.8; 95% CI, 7.71-30.36), similar to protein-truncating variants. This functional data supports the clinical classification of inactivating RAD51C missense variants as pathogenic or likely pathogenic, which may improve the clinical management of variant carriers. SIGNIFICANCE Functional analysis of the impact of a large number of missense variants on RAD51C function provides insight into RAD51C activity and information for classification of the cancer relevance of RAD51C variants.
Collapse
Affiliation(s)
| | | | | | - Gemma Montalban
- CHU de Quebec-Université Laval Research Center, Université Laval, Quebec City, Quebec, Canada
| | | | | | | | - Jie Na
- Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | | | | | | | | | | | | | - Peter Kraft
- T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Sara Lindstrom
- Department of Epidemiology, University of Washington, Seattle, Washington
| | - Julie R. Palmer
- Slone Epidemiology Center at Boston University, Boston, Massachusetts
| | - Lauren R. Teras
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, Georgia
| | | | - Song Yao
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Irene Ong
- University of Wisconsin-Madison, Madison, Wisconsin
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jacques Simard
- CHU de Quebec-Université Laval Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Jean Yves Masson
- CHU de Quebec-Université Laval Research Center, Université Laval, Quebec City, Quebec, Canada
| | | | | |
Collapse
|
48
|
Turchiano A, Piglionica M, Martino S, Bagnulo R, Garganese A, De Luisi A, Chirulli S, Iacoviello M, Stasi M, Tabaku O, Meneleo E, Capurso M, Crocetta S, Lattarulo S, Krylovska Y, Lastella P, Forleo C, Stella A, Bukvic N, Simone C, Resta N. Impact of High-to-Moderate Penetrance Genes on Genetic Testing: Looking over Breast Cancer. Genes (Basel) 2023; 14:1530. [PMID: 37628581 PMCID: PMC10454640 DOI: 10.3390/genes14081530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer (BC) is the most common cancer and the leading cause of cancer death in women worldwide. Since the discovery of the highly penetrant susceptibility genes BRCA1 and BRCA2, many other predisposition genes that confer a moderate risk of BC have been identified. Advances in multigene panel testing have allowed the simultaneous sequencing of BRCA1/2 with these genes in a cost-effective way. Germline DNA from 521 cases with BC fulfilling diagnostic criteria for hereditary BC were screened with multigene NGS testing. Pathogenic (PVs) and likely pathogenic (LPVs) variants in moderate penetrance genes were identified in 15 out of 521 patients (2.9%), including 2 missense, 7 non-sense, 1 indel, and 3 splice variants, as well as two different exon deletions, as follows: ATM (n = 4), CHEK2 (n = 5), PALB2 (n = 2), RAD51C (n = 1), and RAD51D (n = 3). Moreover, the segregation analysis of PVs and LPVs into first-degree relatives allowed the detection of CHEK2 variant carriers diagnosed with in situ melanoma and clear cell renal cell carcinoma (ccRCC), respectively. Extended testing beyond BRCA1/2 identified PVs and LPVs in a further 2.9% of BC patients. In conclusion, panel testing yields more accurate genetic information for appropriate counselling, risk management, and preventive options than assessing BRCA1/2 alone.
Collapse
Affiliation(s)
- Antonella Turchiano
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Marilidia Piglionica
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Stefania Martino
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Rosanna Bagnulo
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Antonella Garganese
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Annunziata De Luisi
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Stefania Chirulli
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Matteo Iacoviello
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Michele Stasi
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Ornella Tabaku
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Eleonora Meneleo
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Martina Capurso
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Silvia Crocetta
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Simone Lattarulo
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Yevheniia Krylovska
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Patrizia Lastella
- Rare Disease Center, Internal Medicine Unit “C. Frugoni”, AOU Policlinico di Bari, 70124 Bari, Italy;
| | - Cinzia Forleo
- Cardiology Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Alessandro Stella
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Nenad Bukvic
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| | - Cristiano Simone
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
- Medical Genetics, National Institute of Gastroenterology, “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Nicoletta Resta
- Medical Genetic, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.T.); (M.P.); (S.M.); (R.B.); (A.G.); (A.D.L.); (S.C.); (M.I.); (M.S.); (O.T.); (E.M.); (M.C.); (S.C.); (S.L.); (Y.K.); (A.S.); (N.B.); (C.S.)
| |
Collapse
|
49
|
Kikuchi M, Gomi N, Ueki A, Osako T, Terauchi T. Effectiveness and tasks of breast MRI surveillance for high-risk women with cancer susceptibility genes other than BRCA1/2: a single institution study. Breast Cancer 2023; 30:577-583. [PMID: 36897545 DOI: 10.1007/s12282-023-01448-3] [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: 11/24/2022] [Accepted: 03/01/2023] [Indexed: 03/11/2023]
Abstract
BACKGROUND In Japan, with the introduction of multigene panel testing, there is an urgent need to build a new medical system for hereditary breast cancer patients that covers pathogenic variants other than BRCA1/2. The aim of this study was to reveal the current status of breast MRI surveillance for high-risk breast cancer susceptibility genes other than BRCA1/2 and the characteristics of detected breast cancer. METHODS We retrospectively examined 42 breast MRI surveillance with contrast performed on patients with hereditary tumors other than BRCA1/2 pathogenic variants at our hospital from 2017 to 2021. MRI exams were evaluated independently by two radiologists. Final histopathological diagnosis for malignant lesions were obtained from surgical specimen. RESULTS A total of 16 patients included TP53, CDH1, PALB2, ATM pathogenic variants and 3 variant of unknown significance. 2 patients with TP53 pathogenic variants were detected breast cancer by annual MRI surveillance. The rate of cancer detection was 12.5% (2/16). One patient was detected synchronous bilateral breast cancer and unilateral multiple breast cancers (3 lesions in 1 patient), so there were 4 malignant lesions in total. Surgical pathology of 4 lesions were 2 ductal carcinoma in situ, 1 invasive lobular carcinoma, and 1 invasive ductal carcinoma. MRI findings of 4 malignant lesions were detected as 2 non mass enhancement, 1 focus and 1 small mass. All of 2 patients with PALB2 pathogenic variants had previously developed breast cancer. CONCLUSIONS Germline TP53 and PALB2 were strongly associated with breast cancer, suggesting that MRI surveillance is essential for breast cancer-related hereditary predisposition.
Collapse
Affiliation(s)
- Mari Kikuchi
- Department of Diagnostic Imaging, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan.
| | - Naoya Gomi
- Department of Diagnostic Imaging, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Arisa Ueki
- Department of Clinical Genetic Oncology, Cancer Institute Hospital of Japanese Foundation For Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Tomo Osako
- Division of Pathology, Cancer Institute of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Takashi Terauchi
- Department of Diagnostic Imaging, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| |
Collapse
|
50
|
Oropeza E, Seker S, Carrel S, Mazumder A, Lozano D, Jimenez A, VandenHeuvel SN, Noltensmeyer DA, Punturi NB, Lei JT, Lim B, Waltz SE, Raghavan SA, Bainbridge MN, Haricharan S. Molecular portraits of cell cycle checkpoint kinases in cancer evolution, progression, and treatment responsiveness. SCIENCE ADVANCES 2023; 9:eadf2860. [PMID: 37390209 PMCID: PMC10313178 DOI: 10.1126/sciadv.adf2860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 05/26/2023] [Indexed: 07/02/2023]
Abstract
Cell cycle dysregulation is prerequisite for cancer formation. However, it is unknown whether the mode of dysregulation affects disease characteristics. Here, we conduct comprehensive analyses of cell cycle checkpoint dysregulation using patient data and experimental investigations. We find that ATM mutation predisposes the diagnosis of primary estrogen receptor (ER)+/human epidermal growth factor (HER)2- cancer in older women. Conversely, CHK2 dysregulation induces formation of metastatic, premenopausal ER+/HER2- breast cancer (P = 0.001) that is treatment-resistant (HR = 6.15, P = 0.01). Lastly, while mutations in ATR alone are rare, ATR/TP53 co-mutation is 12-fold enriched over expected in ER+/HER2- disease (P = 0.002) and associates with metastatic progression (HR = 2.01, P = 0.006). Concordantly, ATR dysregulation induces metastatic phenotypes in TP53 mutant, not wild-type, cells. Overall, we identify mode of cell cycle dysregulation as a distinct event that determines subtype, metastatic potential, and treatment responsiveness, providing rationale for reconsidering diagnostic classification through the lens of the mode of cell cycle dysregulation..
Collapse
Affiliation(s)
- Elena Oropeza
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sinem Seker
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sabrina Carrel
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Aloran Mazumder
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Daniel Lozano
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Athena Jimenez
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | | | - Nindo B. Punturi
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jonathan T. Lei
- Lester and Sue Smith Breast Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Bora Lim
- Lester and Sue Smith Breast Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Oncology/Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Susan E. Waltz
- Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA
- Research Service, Cincinnati Veteran's Affairs Medical Center, 3200 Vine St., Cincinnati, OH, USA
| | | | | | - Svasti Haricharan
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|