1
|
Walbaum B, García-Fructuoso I, Martínez-Sáez O, Schettini F, Sánchez C, Acevedo F, Chic N, Muñoz-Carrillo J, Adamo B, Muñoz M, Partridge AH, Bellet M, Brasó-Maristany F, Prat A, Vidal M. Hormone receptor-positive early breast cancer in young women: A comprehensive review. Cancer Treat Rev 2024; 129:102804. [PMID: 39084152 DOI: 10.1016/j.ctrv.2024.102804] [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: 05/28/2024] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 08/02/2024]
Abstract
The incidence of breast cancer in ≤ 40 yr-old women (YWBC) has been steadily increasing in recent decades. Although this group of patients represents less than 10 % of all newly diagnosed BC cases it encompasses a significant burden of disease. Usually underrepresented in clinical trials, YWBCs are also characterized by late diagnoses and poorly differentiated, aggressive-subtype disease, partly explaining its poor prognosis along with a high recurrence risk, and high mortality rates. On the other hand, YWBC treatment poses unique challenges such as preservation of fertility, and long-term toxicity and adverse events. Herein, we summarize the current evidence in hormone receptor-positive YWBC including specific risk factors, clinicopathologic and genomic features, and available evidence on response to chemotherapy and endocrine therapy. Overall, we advocate for a more comprehensive multidisciplinary healthcare model to improve the outcomes and the quality of life of this subset of younger patients.
Collapse
Affiliation(s)
- Benjamin Walbaum
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute, Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Department of Medical Oncology, Pontificia Universidad Catolica de Chile, Santiago, Chile; Faculty of Medicine, Universidad Catolica de Chile, Santiago, Chile
| | - Isabel García-Fructuoso
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute, Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Olga Martínez-Sáez
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute, Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Francesco Schettini
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute, Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - César Sánchez
- Department of Medical Oncology, Pontificia Universidad Catolica de Chile, Santiago, Chile; Faculty of Medicine, Universidad Catolica de Chile, Santiago, Chile
| | - Francisco Acevedo
- Department of Medical Oncology, Pontificia Universidad Catolica de Chile, Santiago, Chile; Faculty of Medicine, Universidad Catolica de Chile, Santiago, Chile
| | - Nuria Chic
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute, Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain
| | | | - Barbara Adamo
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute, Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Harvard Medical School, Boston, MA, USA
| | - Montserrat Muñoz
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute, Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Harvard Medical School, Boston, MA, USA
| | - Ann H Partridge
- 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
| | - Meritxell Bellet
- Oncology Department, Breast Cancer Unit, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute, Barcelona, Spain; Institute of Cancer and Blood Disorders, Hospital Clinic of Barcelona, Barcelona, Spain; Reveal Genomics, Barcelona, Spain
| | - Aleix Prat
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute, Barcelona, Spain; Institute of Cancer and Blood Disorders, Hospital Clinic of Barcelona, Barcelona, Spain; Reveal Genomics, Barcelona, Spain; Institute of Oncology (IOB)-Hospital Quironsalud, Barcelona, Spain; Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Maria Vidal
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute, Barcelona, Spain; Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain; Institute of Oncology (IOB)-Hospital Quironsalud, Barcelona, Spain; Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
2
|
Mokhber A, Hall E, Uzelac A, Salmena L, Cheung A, Lubinski J, Narod SA, Kotsopoulos J. Delineating the relationship between circulating osteoprotegerin and bone health in women with a pathogenic variant in BRCA1: A cross-sectional analysis. Bone Rep 2024; 22:101802. [PMID: 39297019 PMCID: PMC11408939 DOI: 10.1016/j.bonr.2024.101802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 09/21/2024] Open
Abstract
Purpose Osteoprotegerin (OPG) plays an important role in the inhibition of osteoclast formation and bone resorption. Studies have reported lower OPG levels among women with a pathogenic variant (mutation) in the BRCA1 gene, and thus, may be at greater risk for skeletal bone loss. Thus, we investigated the association between circulating OPG and two validated markers of bone health: 1) bone fracture risk score (FRAX) and 2) bone mineral density (BMD), among BRCA mutation carriers. Methods Women with a blood sample and clinical data were included in this analysis. An enzyme-linked immunosorbent assay (ELISA) was used to quantify serum OPG (pg/mL) and the 10-year risk of major osteoporotic fracture (FRAXmajor) and hip fracture (FRAXhip) (%) was estimated using a web-based algorithm. For a subset of women, lumbar spine BMD was previously assessed by dual x-ray absorptiometry (DXA)(T-score). A Mann-Whitney U test was used to evaluate the association between OPG and FRAX score, while linear regression was used to assess the association of OPG and BMD. Results Among 701 women with a BRCA1 mutation, there was a significant (and unexpected) positive association between OPG levels and FRAX score (FRAXmajor: 2.12 (low OPG) vs. 2.53 (high OPG) P < 0.0001; FRAXhip: 0.27 (low OPG) vs. 0.44 (high OPG) P < 0.0001). In a subset with BMD measurement (n = 50), low serum OPG was associated with a significantly lower BMD T-score (-1.069 vs. -0.318; P = 0.04). Conclusion Our findings suggest that women with inherently lower OPG may be at risk of lower BMD, the gold standard marker of bone disease. Due to the young age of our cohort, on-going studies are warranted to re-evaluate the association between OPG and FRAX in BRCA mutation carriers.
Collapse
Affiliation(s)
- Aghaghia Mokhber
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
- Queen's School of Medicine, Queen's University, Kingston, ON, Canada
| | - Elizabeth Hall
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Aleksandra Uzelac
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Leonardo Salmena
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Angela Cheung
- Osteoporosis Program, University Health Network, Centre of Excellence in Skeletal Health Assessment, University of Toronto, Toronto, ON, Canada
| | - Jan Lubinski
- Pomeranian Medical Institution, Szczecin, Poland
| | - Steven A Narod
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Joanne Kotsopoulos
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Huang S, Mei Z, Wan A, Zhao M, Qi X. Application and prospect of organoid technology in breast cancer. Front Immunol 2024; 15:1413858. [PMID: 39253075 PMCID: PMC11381393 DOI: 10.3389/fimmu.2024.1413858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
Breast cancer is the most common malignant tumor in women. Due to the high heterogeneity of breast cancer cells, traditional in vitro research models still have major limitations. Therefore, it is urgent to establish an experimental model that can accurately simulate the characteristics of human breast cancer. Breast cancer organoid technology emerged as the times required, that is, to construct tissue analogs with organ characteristics by using a patient's tumor tissue through 3D culture in vitro. Since the breast cancer organoid can fully preserve the histology and genetic characteristics of the original tumor, it provides a reliable model for preclinical drug screening, establishment of breast cancer organoid biobanks, research into the mechanisms of tumor development, and determination of cancer targets. It has promoted personalized treatment for clinical breast cancer patients. This article mainly focuses on recent research progress and applications of organoid technology in breast cancer, discussing the current limitations and prospects of breast cancer organoid technology.
Collapse
Affiliation(s)
- Shanlin Huang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| | - Zifan Mei
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| | - Andi Wan
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| | - Min Zhao
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis and Treatment of Breast cancer, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
4
|
Wunderle M, Heindl F, Behrens AS, Häberle L, Hack CC, Heusinger K, Huebner H, Gass P, Ruebner M, Schulz-Wendtland R, Erber R, Hartmann A, Beckmann MW, Dougall WC, Press MF, Fasching PA, Emons J. Correlation of RANK and RANKL with mammographic density in primary breast cancer patients. Arch Gynecol Obstet 2024; 310:1223-1233. [PMID: 38836929 PMCID: PMC11258178 DOI: 10.1007/s00404-024-07495-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/24/2024] [Indexed: 06/06/2024]
Abstract
PURPOSE The receptor activator of nuclear factor kappa B (RANK) and its ligand (RANKL) have been shown to promote proliferation of the breast and breast carcinogenesis. The objective of this analysis was to investigate whether tumor-specific RANK and RANKL expression in patients with primary breast cancer is associated with high percentage mammographic density (PMD), which is a known breast cancer risk factor. METHODS Immunohistochemical staining of RANK and RANKL was performed in tissue microarrays (TMAs) from primary breast cancer samples of the Bavarian Breast Cancer Cases and Controls (BBCC) study. For RANK and RANKL expression, histochemical scores (H scores) with a cut-off value of > 0 vs 0 were established. PMD was measured in the contralateral, non-diseased breast. Linear regression models with PMD as outcome were calculated using common predictors of PMD (age at breast cancer diagnosis, body mass index (BMI) and parity) and RANK and RANKL H scores. Additionally, Spearman rank correlations (ρ) between PMD and RANK and RANKL H score were performed. RESULTS In the final cohort of 412 patients, breast cancer-specific RANK and RANKL expression was not associated with PMD (P = 0.68). There was no correlation between PMD and RANK H score (Spearman's ρ = 0.01, P = 0.87) or RANKL H score (Spearman's ρ = 0.04, P = 0.41). RANK expression was highest in triple-negative tumors, followed by HER2-positive, luminal B-like and luminal A-like tumors, while no subtype-specific expression of RANKL was found. CONCLUSION Results do not provide evidence for an association of RANK and RANKL expression in primary breast cancer with PMD.
Collapse
Affiliation(s)
- Marius Wunderle
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Felix Heindl
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Annika S Behrens
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Lothar Häberle
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Biostatistics Unit, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Carolin C Hack
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Katharina Heusinger
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Hanna Huebner
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Paul Gass
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Matthias Ruebner
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Rüdiger Schulz-Wendtland
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Institute of Diagnostic Radiology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Ramona Erber
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Institute of Pathology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Arndt Hartmann
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Institute of Pathology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - William C Dougall
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4702, Australia
- Hematology and Oncology Research, Amgen, Inc, Seattle, WA, 98119, USA
| | - Michael F Press
- Department of Pathology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Julius Emons
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| |
Collapse
|
5
|
Winship AL, Alesi LR, Stringer JM, Cao Y, Lewis YM, Tu L, Swindells EOK, Giridharan S, Cai X, Griffiths MJ, Zerafa N, Gilham L, Hickey M, Hutt KJ. Conditional loss of Brca1 in oocytes causes reduced litter size, ovarian reserve depletion and impaired oocyte in vitro maturation with advanced reproductive age in mice. EBioMedicine 2024; 106:105262. [PMID: 39084071 PMCID: PMC11342213 DOI: 10.1016/j.ebiom.2024.105262] [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: 07/02/2023] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND An estimated 1 in 350 women carry germline BRCA1/2 mutations, which confer an increased risk of developing breast and ovarian cancer, and may also contribute to subfertility. All mature, sex steroid-producing ovarian follicles are drawn from the pool of non-renewable primordial follicles, termed the 'ovarian reserve'. The clinical implications of early ovarian reserve exhaustion extend beyond infertility, to include the long-term adverse health consequences of loss of endocrine function and premature menopause. We aimed to determine whether conditional loss of Brca1 in oocytes impacts ovarian follicle numbers, oocyte quality and fertility in mice with advancing maternal age. We also aimed to determine the utility of AMH as a marker of ovarian function, by assessing circulating AMH levels in mice and women with BRCA1/2 mutations, and correlating this with ovarian follicle counts. METHODS In this study, we addressed a longstanding question in the field regarding the functional consequences of BRCA1 inactivation in oocytes. To recapitulate loss of BRCA1 protein function in oocytes, we generated mice with conditional gene deletion of Brca1 in oocytes using Gdf9-Cre recombinase (WT: Brca1fl/flGdf9+/+; cKO: Brca1fl/flGdf9cre/+). FINDINGS While the length of the fertile lifespan was not altered between groups after a comprehensive breeding trial, conditional loss of Brca1 in oocytes led to reduced litter size in female mice. Brca1 cKO animals had a reduced ovarian reserve and oocyte maturation was impaired with advanced maternal age at postnatal day (PN)300, compared to WT animals. Serum anti-Müllerian hormone (AMH) concentrations (the gold-standard indirect marker of the ovarian reserve used in clinical practice) were not predictive of reduced primordial follicle number in Brca1 cKO mice versus WT. Furthermore, we found no correlation between follicle number or density and serum AMH concentrations in matched samples from a small cohort of premenopausal women with BRCA1/2 mutations. INTERPRETATION Together, our data demonstrate that BRCA1 is a key regulator of oocyte number and quality in females and suggest that caution should be used in relying on AMH as a reliable marker of the ovarian reserve in this context. FUNDING This work was made possible through Victorian State Government Operational Infrastructure Support and Australian Government NHMRC IRIISS. This work was supported by funding from the Australian Research Council (ALW - DE21010037 and KJH - FT190100265), as well as the National Breast Cancer Foundation (IIRS-22-092) awarded to ALW and KJH. LRA, YML, LT, EOKS and MG were supported by Australian Government Research Training Program Scholarships. LRA, YML and LT were also supported by a Monash Graduate Excellence Scholarship. YC, SG and XC were supported by Monash Biomedicine Discovery Institute PhD Scholarships. LRA was also supported by a Monash University ECPF24-6809920940 Fellowship. JMS was supported by NHMRC funding (2011299). MH was supported by an NHMRC Investigator Grant (1193838).
Collapse
Affiliation(s)
- Amy L Winship
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia.
| | - Lauren R Alesi
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Jessica M Stringer
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Yujie Cao
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Yasmin M Lewis
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Lisa Tu
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Elyse O K Swindells
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Saranya Giridharan
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Xuebi Cai
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Meaghan J Griffiths
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia; University of Edinburgh, MRC Centre for Reproductive Health, Queens Medical Research Institute, Edinburgh, UK
| | - Nadeen Zerafa
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia
| | - Leslie Gilham
- Breast Cancer Network Australia and Breast Cancer Trials Australia, Camberwell, VIC, Australia
| | - Martha Hickey
- Gynaecology Research Centre, Royal Women's Hospital, Parkville, VIC, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia
| | - Karla J Hutt
- Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
6
|
Bowen CM, Demarest K, Vilar E, Shah PD. Novel Cancer Prevention Strategies in Individuals With Hereditary Cancer Syndromes: Focus on BRCA1, BRCA2, and Lynch Syndrome. Am Soc Clin Oncol Educ Book 2024; 44:e433576. [PMID: 38913968 DOI: 10.1200/edbk_433576] [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: 06/26/2024]
Abstract
Germline pathogenic variants (PVs) in the BRCA1 and BRCA2 genes confer elevated risks of breast, ovarian, and other cancers. Lynch syndrome (LS) is associated with increased risks of multiple cancer types including colorectal and uterine cancers. Current cancer risk mitigation strategies have focused on pharmacologic risk reduction, enhanced surveillance, and preventive surgeries. While these approaches can be effective, they stand to be improved on because of either limited efficacy or undesirable impact on quality of life. The current review summarizes ongoing investigational efforts in cancer risk prevention strategies for patients with germline PVs in BRCA1, BRCA2, or LS-associated genes. These efforts span radiation, surgery, and pharmacology including vaccine strategies. Understanding the molecular events involved in the premalignant to malignant transformation in high-risk individuals may ultimately contribute significantly to novel prevention strategies.
Collapse
Affiliation(s)
- Charles M Bowen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Payal D Shah
- Perelman Center for Advanced Medicine, Abramson Cancer Center, Philadelphia, PA
| |
Collapse
|
7
|
Akdag G, Dogan A, Yildirim S, Kinikoglu O, Mokresh ME, Alomari O, Turkoglu E, Isik D, Sürmeli H, Basoglu T, Sever ON, Odabas H, Yildirim ME, Turan N. Exploring the Clinical Impact of RANK Pathway Inhibition in Advanced Breast Cancer: Insights From a Retrospective Study on CDK4/6 Inhibitors and Antiresorptive Therapy. Cureus 2024; 16:e63362. [PMID: 39070363 PMCID: PMC11283752 DOI: 10.7759/cureus.63362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Breast cancer (BC) remains a significant health concern, particularly in advanced stages where the prognosis is poor. The combination of endocrine therapy (ET) with cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) has improved outcomes for advanced BC (aBC) patients. However, resistance to CDK4/6i remains a challenge, with no validated biomarkers to predict response. The receptor activator of the nuclear factor-kB (RANK) pathway has emerged as a key player in aBC, particularly in luminal BC. RANK overexpression has been associated with aggressive phenotypes and resistance to therapy. In view of these findings, we proceeded to investigate the potential involvement of the RANK pathway in luminal BC resistance to CDK4/6i. The objective was to evaluate the effectiveness of denosumab in increasing overall survival (OS) and progression-free survival (PFS). METHODS In this retrospective analysis, 158 BC patients with bone metastases were included. Patients with human epidermal growth factor receptor-2 (HER2)-negative and hormone receptor-positive BC who received palbociclib or ribociclib in addition to antiresorptive medication were included. Patients received either denosumab or zoledronic acid (ZA) therapy. The primary endpoint was OS, with PFS as a secondary endpoint. RESULTS Although the PFS and OS of denosumab were better than ZA in this study, it did not show a significant difference between the two drugs. Meanwhile, mOS was not achievable in patients in the denosumab group, while it was 34.1 months in patients in the ZA group. The hazard ratio (HR) showed a significant improvement for the denosumab group in patients under 60 of age (HR: 0.33, p<0.01), patients with a score of 1 HER2 overexpression (HR: 0.09, p=0.01), and patients with resistant endocrine (HR: 0.42, p=0.02) compared to ZA. CONCLUSION This study highlights the potential clinical relevance of the RANK pathway in BC treatment, and our findings suggest that denosumab may offer significant benefits in terms of PFS and OS for certain subgroups, particularly those with HER2 scores of 1, patients under 60, and those with endocrine-resistant BC. In conclusion, considering that RANK pathway status may be a predictive biomarker for CDK4/6i treatment and may cause treatment resistance, our results demonstrate the clinical relevance of the combination of CDK4/6i + ET with RANKL inhibition.
Collapse
Affiliation(s)
- Goncagul Akdag
- Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Istanbul, TUR
| | - Akif Dogan
- Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Istanbul, TUR
| | - Sedat Yildirim
- Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Istanbul, TUR
| | - Oguzcan Kinikoglu
- Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Istanbul, TUR
| | - Muhammed Edib Mokresh
- Medical School, Hamidiye International School of Medicine, University of Health Sciences, Istanbul, TUR
| | - Omar Alomari
- Medical School, Hamidiye International School of Medicine, University of Health Sciences, Istanbul, TUR
| | - Ezgi Turkoglu
- Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Istanbul, TUR
| | - Deniz Isik
- Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Istanbul, TUR
| | - Heves Sürmeli
- Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Istanbul, TUR
| | - Tugba Basoglu
- Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Istanbul, TUR
| | - Ozlem N Sever
- Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Istanbul, TUR
| | - Hatice Odabas
- Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Istanbul, TUR
| | - Mahmut E Yildirim
- Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Istanbul, TUR
| | - Nedim Turan
- Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Istanbul, TUR
| |
Collapse
|
8
|
Coles CE, Earl H, Anderson BO, Barrios CH, Bienz M, Bliss JM, Cameron DA, Cardoso F, Cui W, Francis PA, Jagsi R, Knaul FM, McIntosh SA, Phillips KA, Radbruch L, Thompson MK, André F, Abraham JE, Bhattacharya IS, Franzoi MA, Drewett L, Fulton A, Kazmi F, Inbah Rajah D, Mutebi M, Ng D, Ng S, Olopade OI, Rosa WE, Rubasingham J, Spence D, Stobart H, Vargas Enciso V, Vaz-Luis I, Villarreal-Garza C. The Lancet Breast Cancer Commission. Lancet 2024; 403:1895-1950. [PMID: 38636533 DOI: 10.1016/s0140-6736(24)00747-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 12/18/2023] [Accepted: 04/09/2024] [Indexed: 04/20/2024]
Affiliation(s)
| | - Helena Earl
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Benjamin O Anderson
- Global Breast Cancer Initiative, World Health Organisation and Departments of Surgery and Global Health Medicine, University of Washington, Seattle, WA, USA
| | - Carlos H Barrios
- Oncology Research Center, Hospital São Lucas, Porto Alegre, Brazil
| | - Maya Bienz
- Mount Vernon Cancer Centre, East and North Hertfordshire NHS Trust, London, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - David A Cameron
- Institute of Genetics and Cancer and Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center/Champalimaud Foundation, Lisbon, Portugal
| | - Wanda Cui
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Prudence A Francis
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Reshma Jagsi
- Emory University School of Medicine, Atlanta, GA, USA
| | - Felicia Marie Knaul
- Institute for Advanced Study of the Americas, University of Miami, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA; Tómatelo a Pecho, Mexico City, Mexico
| | - Stuart A McIntosh
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Kelly-Anne Phillips
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Lukas Radbruch
- Department of Palliative Medicine, University Hospital Bonn, Bonn, Germany
| | | | | | - Jean E Abraham
- Department of Oncology, University of Cambridge, Cambridge, UK
| | | | | | - Lynsey Drewett
- Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | | | - Farasat Kazmi
- Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
| | | | | | - Dianna Ng
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Szeyi Ng
- The Institute of Cancer Research, London, UK
| | | | - William E Rosa
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | | | | | | | | | - Cynthia Villarreal-Garza
- Breast Cancer Center, Hospital Zambrano Hellion TecSalud, Tecnologico de Monterrey, Monterrey, Mexico
| |
Collapse
|
9
|
Domchek SM, Vonderheide RH. Advancing Cancer Interception. Cancer Discov 2024; 14:600-604. [PMID: 38571414 DOI: 10.1158/2159-8290.cd-24-0015] [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: 04/05/2024]
Abstract
Rapid advances in technology and therapeutics, along with better methods to discern who is at risk for cancer by genetic testing and other means, has enabled the development of cancer interception. Targeted therapies and "immuno-interception" may eliminate premalignant lesions and require clinical trial and treatment paradigms altogether distinct from current approaches.
Collapse
Affiliation(s)
- Susan M Domchek
- Basser Center for BRCA1/2, University of Pennsylvania, Philadelphia, Pen-nsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H Vonderheide
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Park SS, Zaman T, Kim SJ, Brooks JD, Wong AKO, Lubiński J, Narod SA, Salmena L, Kotsopoulos J. Correlates of Circulating Osteoprotegerin in Women with a Pathogenic or Likely Pathogenic Variant in the BRCA1 Gene. Cancer Epidemiol Biomarkers Prev 2024; 33:298-305. [PMID: 38015775 DOI: 10.1158/1055-9965.epi-23-0577] [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: 05/20/2023] [Revised: 07/28/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Lower levels of osteoprotegerin (OPG), the decoy receptor for receptor activator of NFκB (RANK)-ligand, have been reported among women with a BRCA1 mutation, suggesting OPG may be marker of cancer risk. Whether various reproductive, hormonal, or lifestyle factors impact OPG levels in these women is unknown. METHODS BRCA1 mutation carriers enrolled in a longitudinal study, no history of cancer, and a serum sample for OPG quantification, were included. Exposure information was collected through self-reported questionnaire at study enrollment and every 2 years thereafter. Serum OPG levels (pg/mL) were measured using an ELISA, and generalized linear models were used to assess the associations between reproductive, hormonal, and lifestyle exposures at the time of blood collection with serum OPG. Adjusted means were estimated using the fully adjusted model. RESULTS A total of 701 women with a median age at blood collection of 39.0 years (18.0-82.0) were included. Older age (Spearman r = 0.24; P < 0.001) and current versus never smoking (98.82 vs. 86.24 pg/mL; Pcat < 0.001) were associated with significantly higher OPG, whereas ever versus never coffee consumption was associated with significantly lower OPG (85.92 vs. 94.05 pg/mL; Pcat = 0.03). There were no other significant associations for other exposures (P ≥ 0.06). The evaluated factors accounted for 7.5% of the variability in OPG. CONCLUSIONS OPG is minimally influenced by hormonal and lifestyle factors among BRCA1 mutation carriers. IMPACT These findings suggest that circulating OPG levels are not impacted by non-genetic factors in high-risk women.
Collapse
Affiliation(s)
- Sarah Sohyun Park
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, Canada
| | - Tasnim Zaman
- Women's College Research Institute, Women's College Hospital, Toronto, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Shana J Kim
- Women's College Research Institute, Women's College Hospital, Toronto, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Jennifer D Brooks
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Andy Kin On Wong
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Joint Department of Medical Imaging, University Health Network, Toronto, Canada
- Osteoporosis Program, Schroeder Arthritis Institute, University Health Network, Toronto, Canada
| | - Jan Lubiński
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
- Read-Gene S.A., Grzepnica, Poland
| | - Steven A Narod
- Women's College Research Institute, Women's College Hospital, Toronto, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Leonardo Salmena
- Women's College Research Institute, Women's College Hospital, Toronto, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Joanne Kotsopoulos
- Women's College Research Institute, Women's College Hospital, Toronto, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| |
Collapse
|
11
|
Zaluzec EK, Sempere LF. Systemic and Local Strategies for Primary Prevention of Breast Cancer. Cancers (Basel) 2024; 16:248. [PMID: 38254741 PMCID: PMC10814018 DOI: 10.3390/cancers16020248] [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: 10/23/2023] [Revised: 12/29/2023] [Accepted: 12/31/2023] [Indexed: 01/24/2024] Open
Abstract
One in eight women will develop breast cancer in the US. For women with moderate (15-20%) to average (12.5%) risk of breast cancer, there are few options available for risk reduction. For high-risk (>20%) women, such as BRCA mutation carriers, primary prevention strategies are limited to evidence-based surgical removal of breasts and/or ovaries and anti-estrogen treatment. Despite their effectiveness in risk reduction, not many high-risk individuals opt for surgical or hormonal interventions due to severe side effects and potentially life-changing outcomes as key deterrents. Thus, better communication about the benefits of existing strategies and the development of new strategies with minimal side effects are needed to offer women adequate risk-reducing interventions. We extensively review and discuss innovative investigational strategies for primary prevention. Most of these investigational strategies are at the pre-clinical stage, but some are already being evaluated in clinical trials and others are expected to lead to first-in-human clinical trials within 5 years. Likely, these strategies would be initially tested in high-risk individuals but may be applicable to lower-risk women, if shown to decrease risk at a similar rate to existing strategies, but with minimal side effects.
Collapse
Affiliation(s)
- Erin K. Zaluzec
- Precision Health Program, Michigan State University, East Lansing, MI 48824, USA;
- Department of Pharmacology & Toxicology, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Lorenzo F. Sempere
- Precision Health Program, Michigan State University, East Lansing, MI 48824, USA;
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
12
|
Northey JJ, Hayward MK, Yui Y, Stashko C, Kai F, Mouw JK, Thakar D, Lakins JN, Ironside AJ, Samson S, Mukhtar RA, Hwang ES, Weaver VM. Mechanosensitive hormone signaling promotes mammary progenitor expansion and breast cancer risk. Cell Stem Cell 2024; 31:106-126.e13. [PMID: 38181747 PMCID: PMC11050720 DOI: 10.1016/j.stem.2023.12.002] [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: 06/01/2023] [Revised: 09/19/2023] [Accepted: 12/06/2023] [Indexed: 01/07/2024]
Abstract
Tissue stem-progenitor cell frequency has been implicated in tumor risk and progression, but tissue-specific factors linking these associations remain ill-defined. We observed that stiff breast tissue from women with high mammographic density, who exhibit increased lifetime risk for breast cancer, associates with abundant stem-progenitor epithelial cells. Using genetically engineered mouse models of elevated integrin mechanosignaling and collagen density, syngeneic manipulations, and spheroid models, we determined that a stiff matrix and high mechanosignaling increase mammary epithelial stem-progenitor cell frequency and enhance tumor initiation in vivo. Augmented tissue mechanics expand stemness by potentiating extracellular signal-related kinase (ERK) activity to foster progesterone receptor-dependent RANK signaling. Consistently, we detected elevated phosphorylated ERK and progesterone receptors and increased levels of RANK signaling in stiff breast tissue from women with high mammographic density. The findings link fibrosis and mechanosignaling to stem-progenitor cell frequency and breast cancer risk and causally implicate epidermal growth factor receptor-ERK-dependent hormone signaling in this phenotype.
Collapse
Affiliation(s)
- Jason J Northey
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mary-Kate Hayward
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yoshihiro Yui
- Research Institute, Nozaki Tokushukai Hospital, Tanigawa 2-10-50, Daito, Osaka 574-0074, Japan
| | - Connor Stashko
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - FuiBoon Kai
- Department of Physiology & Pharmacology, University of Calgary, Calgary, AB T2N1N4, Canada; Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, AB T2N1N4, Canada
| | - Janna K Mouw
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dhruv Thakar
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jonathon N Lakins
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alastair J Ironside
- Department of Pathology, Western General Hospital, NHS Lothian, Edinburgh EH42XU, UK
| | - Susan Samson
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Rita A Mukhtar
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Valerie M Weaver
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
13
|
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
|
14
|
Behrens A, Wurmthaler L, Heindl F, Gass P, Häberle L, Volz B, Hack CC, Emons J, Erber R, Hartmann A, Beckmann MW, Ruebner M, Dougall WC, Press MF, Fasching PA, Huebner H. RANK and RANKL Expression in Tumors of Patients with Early Breast Cancer. Geburtshilfe Frauenheilkd 2024; 84:77-85. [PMID: 38178900 PMCID: PMC10764119 DOI: 10.1055/a-2192-2998] [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: 08/25/2023] [Accepted: 10/15/2023] [Indexed: 01/06/2024] Open
Abstract
Introduction The receptor activator of nuclear factor-κB (RANK) pathway was associated with the pathogenesis of breast cancer. Several studies attempted to link the RANK/RANKL pathway to prognosis; however, with inconsistent outcomes. We aimed to further contribute to the knowledge about RANK/RANKL as prognostic factors in breast cancer. Within this study, protein expression of RANK and its ligand, RANKL, in the tumor tissue was analyzed in association with disease-free survival (DFS) and overall survival (OS) in a study cohort of patients with early breast cancer. Patients and Methods 607 samples of female primary and early breast cancer patients from the Bavarian Breast Cancer Cases and Controls Study were analyzed to correlate the RANK and RANKL expression with DFS and OS. Therefore, expression was quantified using immunohistochemical staining of a tissue microarray. H-scores were determined with the cut-off value of 8.5 for RANK and 0 for RANKL expression, respectively. Results RANK and RANKL immunohistochemistry were assessed by H-score. Both biomarkers did not correlate (ρ = -0.04). According to molecular subtypes, triple-negative tumors and HER2-positive tumors showed a higher number of RANK-positive tumors (H-score ≥ 8.5), however, no subtype-specific expression of RANKL could be detected. Higher RANKL expression tended to correlate with a better prognosis. However, RANK and RANKL expression could not be identified as statistically significant prognostic factors within the study cohort. Conclusions Tumor-specific RANK and RANKL expressions are not applicable as prognostic factors for DFS and OS, but might be associated with subtype-specific breast cancer progression.
Collapse
Affiliation(s)
- Annika Behrens
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Lena Wurmthaler
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Felix Heindl
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Paul Gass
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Lothar Häberle
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
- Biostatistics Unit, Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Bernhard Volz
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Ansbach University of Applied Sciences, Ansbach, Germany
| | - Carolin C. Hack
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Julius Emons
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Ramona Erber
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Arndt Hartmann
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Matthias W. Beckmann
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Matthias Ruebner
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - William C. Dougall
- Hematology and Oncology Research, Amgen, Inc., Seattle, WA, USA
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Michael F. Press
- Department of Pathology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Peter A. Fasching
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| | - Hanna Huebner
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Center for Cancer Research (BZKF), Erlangen, Germany
| |
Collapse
|
15
|
Xu J, Liu S. Cell origin of BRCA2-mutant breast cancer. Nat Cell Biol 2024; 26:43-44. [PMID: 38216736 DOI: 10.1038/s41556-023-01322-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2024]
Affiliation(s)
- Jiahui Xu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Cancer Institutes, State Key Laboratory of Genetic Engineering, Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Cancer Institutes, State Key Laboratory of Genetic Engineering, Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- The Shanghai Key Laboratory of Medical Epigenetics, Shanghai, China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China.
| |
Collapse
|
16
|
Narod SA. Choices for cancer prevention for women with a BRCA1 mutation? a personal view. Hered Cancer Clin Pract 2023; 21:26. [PMID: 38031144 PMCID: PMC10685461 DOI: 10.1186/s13053-023-00271-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023] Open
Abstract
With widespread testing for susceptibility genes, increasing numbers of women are being identified to carry a mutation in one of many genes which renders them susceptible to cancer. The first gene to be identified (in 1994) was BRCA1 which increases a woman's risk for breast cancer (70%) and ovarian cancer (40%). The prevalence of BRCA1 gene mutations has been studied widely and in many countries, mostly in women affected with cancer. In many settings testing is offered routinely to women with serous ovarian cancer or early-onset or triple-negative breast cancer. It is preferable to identify a mutation in a healthy women prior to the diagnosis of cancer. The basic strategies for prevention include surgical prevention, chemoprevention and screening (early detection). Much progress has been made in the past two decades evaluating the benefits of these three approaches. In this commentary I provide my personal views regarding these various interventions in the context of counselling a newly diagnosed health woman with a BRCA1 mutation.
Collapse
Affiliation(s)
- Steven A Narod
- Women's College Research Institute, University of Toronto, 790 Bay Street, Toronto, ON, Canada.
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
17
|
Zeng J, Singh S, Zhou X, Jiang Y, Casarez E, Atkins KA, Janes KA, Zong H. A genetic mosaic mouse model illuminates the pre-malignant progression of basal-like breast cancer. Dis Model Mech 2023; 16:dmm050219. [PMID: 37815460 PMCID: PMC10668031 DOI: 10.1242/dmm.050219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/11/2023] [Indexed: 10/11/2023] Open
Abstract
Basal-like breast cancer (BLBC) is highly aggressive, and often characterized by BRCA1 and p53 deficiency. Although conventional mouse models enabled the investigation of BLBC at malignant stages, its initiation and pre-malignant progression remain understudied. Here, we leveraged a mouse genetic system known as mosaic analysis with double markers (MADM) to study BLBC initiation by generating rare GFP+Brca1, p53-deficient mammary cells alongside RFP+ wild-type sibling cells. After confirming the close resemblance of mammary tumors arising in this model to human BLBC at both transcriptomic and genomic levels, we focused our studies on the pre-malignant progression of BLBC. Initiated GFP+ mutant cells showed a stepwise pre-malignant progression trajectory from focal expansion to hyper-alveolarization and then to micro-invasion. Furthermore, despite morphological similarities to alveoli, hyper-alveolarized structures actually originate from ductal cells based on twin-spot analysis of GFP-RFP sibling cells. Finally, luminal-to-basal transition occurred exclusively in cells that have progressed to micro-invasive lesions. Our MADM model provides excellent spatiotemporal resolution to illuminate the pre-malignant progression of BLBC, and should enable future studies on early detection and prevention for this cancer.
Collapse
Affiliation(s)
- Jianhao Zeng
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Shambhavi Singh
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Xian Zhou
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Ying Jiang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Eli Casarez
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Kristen A. Atkins
- Department of Pathology, University of Virginia Health System, Charlottesville, VA 22908, USA
- University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Kevin A. Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Hui Zong
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
- University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA 22908, USA
| |
Collapse
|
18
|
Ben-Chetrit N, Niu X, Sotelo J, Swett AD, Rajasekhar VK, Jiao MS, Stewart CM, Bhardwaj P, Kottapalli S, Ganesan S, Loyher PL, Potenski C, Hannuna A, Brown KA, Iyengar NM, Giri DD, Lowe SW, Healey JH, Geissmann F, Sagi I, Joyce JA, Landau DA. Breast Cancer Macrophage Heterogeneity and Self-renewal are Determined by Spatial Localization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563749. [PMID: 37961223 PMCID: PMC10634790 DOI: 10.1101/2023.10.24.563749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Tumor-infiltrating macrophages support critical steps in tumor progression, and their accumulation in the tumor microenvironment (TME) is associated with adverse outcomes and therapeutic resistance across human cancers. In the TME, macrophages adopt diverse phenotypic alterations, giving rise to heterogeneous immune activation states and induction of cell cycle. While the transcriptional profiles of these activation states are well-annotated across human cancers, the underlying signals that regulate macrophage heterogeneity and accumulation remain incompletely understood. Here, we leveraged a novel ex vivo organotypic TME (oTME) model of breast cancer, in vivo murine models, and human samples to map the determinants of functional heterogeneity of TME macrophages. We identified a subset of F4/80highSca-1+ self-renewing macrophages maintained by type-I interferon (IFN) signaling and requiring physical contact with cancer-associated fibroblasts. We discovered that the contact-dependent self-renewal of TME macrophages is mediated via Notch4, and its inhibition abrogated tumor growth of breast and ovarian carcinomas in vivo, as well as lung dissemination in a PDX model of triple-negative breast cancer (TNBC). Through spatial multi-omic profiling of protein markers and transcriptomes, we found that the localization of macrophages further dictates functionally distinct but reversible phenotypes, regardless of their ontogeny. Whereas immune-stimulatory macrophages (CD11C+CD86+) populated the tumor epithelial nests, the stroma-associated macrophages (SAMs) were proliferative, immunosuppressive (Sca-1+CD206+PD-L1+), resistant to CSF-1R depletion, and associated with worse patient outcomes. Notably, following cessation of CSF-1R depletion, macrophages rebounded primarily to the SAM phenotype, which was associated with accelerated growth of mammary tumors. Our work reveals the spatial determinants of macrophage heterogeneity in breast cancer and highlights the disruption of macrophage self-renewal as a potential new therapeutic strategy.
Collapse
Affiliation(s)
- Nir Ben-Chetrit
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- These authors contributed equally
| | - Xiang Niu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- These authors contributed equally
- Present address: Genentech, Inc., South San Francisco, CA, USA
| | - Jesus Sotelo
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Ariel D. Swett
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Vinagolu K. Rajasekhar
- Orthopedic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria S. Jiao
- Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Caitlin M. Stewart
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Priya Bhardwaj
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Sanjay Kottapalli
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Saravanan Ganesan
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Pierre-Louis Loyher
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Catherine Potenski
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Assaf Hannuna
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Neil M. Iyengar
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dilip D. Giri
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Scott W. Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - John H. Healey
- Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Frederic Geissmann
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Johanna A. Joyce
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Switzerland
| | - Dan A. Landau
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| |
Collapse
|
19
|
Khan SA. Breast Cancer Risk Reduction: Current Status and Emerging Trends to Increase Efficacy and Reduce Toxicity of Preventive Medication. Surg Oncol Clin N Am 2023; 32:631-646. [PMID: 37714633 DOI: 10.1016/j.soc.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
The primary prevention of breast cancer is a worthwhile goal for which the efficacy of antiestrogens is well established. However, implementation has been problematic related to low prioritization by providers and the reluctance of high-risk women to experience medication side effects. Emerging solutions include improved risk estimation through the use of polygenic risk scores and the application of radiomics to screening mammograms; and optimization of medication dose to limit toxicity. The identification of agents to prevent estrogen receptor negative or HER2-positive tumors is being pursued, but personalization of medical risk reduction requires the prediction of tumor subtypes.
Collapse
Affiliation(s)
- Seema Ahsan Khan
- Department of Surgery, Feinberg School of Medicine of Northwestern University, 303 East Superior Street, Chicago, IL 60614, USA.
| |
Collapse
|
20
|
Najafabadi MG, Gray GK, Kong LR, Gupta K, Perera D, Naylor H, Brugge JS, Venkitaraman AR, Shehata M. A transcriptional response to replication stress selectively expands a subset of Brca2-mutant mammary epithelial cells. Nat Commun 2023; 14:5206. [PMID: 37626143 PMCID: PMC10457340 DOI: 10.1038/s41467-023-40956-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Germline BRCA2 mutation carriers frequently develop luminal-like breast cancers, but it remains unclear how BRCA2 mutations affect mammary epithelial subpopulations. Here, we report that monoallelic Brca2mut/WT mammary organoids subjected to replication stress activate a transcriptional response that selectively expands Brca2mut/WT luminal cells lacking hormone receptor expression (HR-). While CyTOF analyses reveal comparable epithelial compositions among wildtype and Brca2mut/WT mammary glands, Brca2mut/WT HR- luminal cells exhibit greater organoid formation and preferentially survive and expand under replication stress. ScRNA-seq analysis corroborates the expansion of HR- luminal cells which express elevated transcript levels of Tetraspanin-8 (Tspan8) and Thrsp, plus pathways implicated in replication stress survival including Type I interferon responses. Notably, CRISPR/Cas9-mediated deletion of Tspan8 or Thrsp prevents Brca2mut/WT HR- luminal cell expansion. Our findings indicate that Brca2mut/WT cells activate a transcriptional response after replication stress that preferentially favours outgrowth of HR- luminal cells through the expression of interferon-responsive and mammary alveolar genes.
Collapse
Affiliation(s)
| | - G Kenneth Gray
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA, USA
| | - Li Ren Kong
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
- The Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Pharmacology, NUS School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research, National University of Singapore, Singapore, Singapore
| | - Komal Gupta
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
- The Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research, National University of Singapore, Singapore, Singapore
| | - David Perera
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
| | - Huw Naylor
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA, USA
| | - Ashok R Venkitaraman
- MRC Cancer Unit, University of Cambridge, Cambridge, UK.
- The Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- Institute of Molecular & Cellular Biology Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.
| | - Mona Shehata
- Department of Oncology, University of Cambridge, Cambridge, UK.
- MRC Cancer Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
21
|
Gomes I, Gallego-Paez LM, Jiménez M, Santamaria PG, Mansinho A, Sousa R, Abreu C, Suárez EG, Costa L, Casimiro S. Co-targeting RANK pathway treats and prevents acquired resistance to CDK4/6 inhibitors in luminal breast cancer. Cell Rep Med 2023; 4:101120. [PMID: 37451269 PMCID: PMC10439176 DOI: 10.1016/j.xcrm.2023.101120] [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: 11/22/2022] [Revised: 05/11/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023]
Abstract
The combination of endocrine therapy (ET) and cyclin-dependent kinase 4/6 (CDK4/6) inhibitors (CDK4/6i) was a hallmark in metastatic luminal breast cancer (BC). However, intrinsic and acquired resistance affects long-term efficacy. Here, we study the role of the receptor activator of nuclear factor-κB (RANK) pathway in CDK4/6i resistance. We find that RANK overexpression in luminal BC is associated with intrinsic resistance to CDK4/6i, both in vitro and in mouse xenografts, and decreased proliferation rate and chronic interferon (IFN) γ response are highlighted as resistance drivers. Gene expression data from the NeoPalAna CDK4/6i clinical trial, and studies with palbociclib-resistant cell lines, show that RANK is upregulated after treatment with CDK4/6i, supporting a role in acquired resistance. Our study shows that RANK ligand (RANKL) inhibitors can restore sensitivity to CDK4/6i and prevent acquired resistance. On the basis of these findings, we conclude that pharmacological inhibition of the RANK pathway through RANKL blocking could represent an add-on to ET + CDK4/6i, warranting further clinical studies.
Collapse
Affiliation(s)
- Inês Gomes
- Luis Costa Laboratory, Institute of Molecular Medicine (iMM), Lisbon Medical School, 1649-028 Lisbon, Portugal
| | - Lina M Gallego-Paez
- Luis Costa Laboratory, Institute of Molecular Medicine (iMM), Lisbon Medical School, 1649-028 Lisbon, Portugal
| | - Maria Jiménez
- Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | | | - André Mansinho
- Oncology Division, Hospital de Santa Maria-CHULN, 1649-028 Lisbon, Portugal
| | - Rita Sousa
- Oncology Division, Hospital de Santa Maria-CHULN, 1649-028 Lisbon, Portugal
| | - Catarina Abreu
- Oncology Division, Hospital de Santa Maria-CHULN, 1649-028 Lisbon, Portugal
| | - Eva González Suárez
- Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain
| | - Luis Costa
- Luis Costa Laboratory, Institute of Molecular Medicine (iMM), Lisbon Medical School, 1649-028 Lisbon, Portugal; Oncology Division, Hospital de Santa Maria-CHULN, 1649-028 Lisbon, Portugal.
| | - Sandra Casimiro
- Luis Costa Laboratory, Institute of Molecular Medicine (iMM), Lisbon Medical School, 1649-028 Lisbon, Portugal.
| |
Collapse
|
22
|
Kolokotroni A, Gkikopoulou E, Rinotas V, Ntari L, Zareifi D, Rouchota M, Sarpaki S, Lymperopoulos I, Alexopoulos LG, Loudos G, Denis MC, Karagianni N, Douni E. Α Humanized RANKL Transgenic Mouse Model of Progestin-Induced Mammary Carcinogenesis for Evaluation of Novel Therapeutics. Cancers (Basel) 2023; 15:4006. [PMID: 37568820 PMCID: PMC10417415 DOI: 10.3390/cancers15154006] [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: 07/16/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Receptor activator of nuclear factor-κB ligand (RANKL) is critically involved in mammary gland pathophysiology, while its pharmaceutical inhibition is being currently investigated in breast cancer. Herein, we investigated whether the overexpression of human RANKL in transgenic mice affects hormone-induced mammary carcinogenesis, and evaluated the efficacy of anti-RANKL treatments, such as OPG-Fc targeting both human and mouse RANKL or Denosumab against human RANKL. We established novel MPA/DMBA-driven mammary carcinogenesis models in TgRANKL mice that express both human and mouse RANKL, as well as in humanized humTgRANKL mice expressing only human RANKL, and compared them to MPA/DMBA-treated wild-type (WT) mice. Our results show that TgRANKL and WT mice have similar levels of susceptibility to mammary carcinogenesis, while OPG-Fc treatment restored mammary ductal density, and prevented ductal branching and the formation of neoplastic foci in both genotypes. humTgRANKL mice also developed MPA/DMBA-induced tumors with similar incidence and burden to those of WT and TgRANKL mice. The prophylactic treatment of humTgRANKL mice with Denosumab significantly prevented the rate of appearance of mammary tumors from 86.7% to 15.4% and the early stages of carcinogenesis, whereas therapeutic treatment did not lead to any significant attenuation of tumor incidence or tumor burden compared to control mice, suggesting the importance of RANKL primarily in the initial stages of tumorigenesis. Overall, we provide unique genetic tools for investigating the involvement of RANKL in breast carcinogenesis, and allow the preclinical evaluation of novel therapeutics that target hormone-related breast cancers.
Collapse
Affiliation(s)
- Anthi Kolokotroni
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Fleming 34, 16672 Vari, Greece
| | - Evi Gkikopoulou
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Fleming 34, 16672 Vari, Greece
| | - Vagelis Rinotas
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Fleming 34, 16672 Vari, Greece
| | - Lydia Ntari
- Biomedcode Hellas SA, Fleming 34, 16672 Vari, Greece (M.C.D.)
| | - Danae Zareifi
- Department of Mechanical Engineering, National Technical University of Athens, 10682 Athens, Greece
| | - Maritina Rouchota
- BIOEMTECH, Lefkippos Attica Technology Park, NCSR “Demokritos”, Ag. Paraskevi, 15343 Athens, Greece (G.L.)
| | - Sophia Sarpaki
- BIOEMTECH, Lefkippos Attica Technology Park, NCSR “Demokritos”, Ag. Paraskevi, 15343 Athens, Greece (G.L.)
| | | | - Leonidas G. Alexopoulos
- Department of Mechanical Engineering, National Technical University of Athens, 10682 Athens, Greece
| | - George Loudos
- BIOEMTECH, Lefkippos Attica Technology Park, NCSR “Demokritos”, Ag. Paraskevi, 15343 Athens, Greece (G.L.)
| | - Maria C. Denis
- Biomedcode Hellas SA, Fleming 34, 16672 Vari, Greece (M.C.D.)
| | - Niki Karagianni
- Biomedcode Hellas SA, Fleming 34, 16672 Vari, Greece (M.C.D.)
| | - Eleni Douni
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Fleming 34, 16672 Vari, Greece
| |
Collapse
|
23
|
Moyer CL, Brown PH. Targeting nuclear hormone receptors for the prevention of breast cancer. Front Med (Lausanne) 2023; 10:1200947. [PMID: 37583424 PMCID: PMC10424511 DOI: 10.3389/fmed.2023.1200947] [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: 04/05/2023] [Accepted: 06/30/2023] [Indexed: 08/17/2023] Open
Abstract
Advancements in research have led to the steady decline of breast cancer mortality over the past thirty years. However, breast cancer incidence has continued to rise, resulting in an undue burden on healthcare costs and highlighting a great need for more effective breast cancer prevention strategies, including targeted chemo preventative agents. Efforts to understand the etiology of breast cancer have uncovered important roles for nuclear receptors in the development and progression of breast cancer. Targeted therapies to inhibit estrogen receptor (ER) and progesterone receptor (PR) signaling (selective ER modulators, aromatase inhibitors and selective PR modulators) have shown great promise for the treatment and prevention of hormone receptor (HR)-positive breast cancer. However, these drugs do not prevent HR-negative disease. Therefore, recent efforts have focused on novel targeted therapies with the potential to prevent both HR-positive and HR-negative breast cancer. Among these include drugs that target other nuclear receptors, such as retinoic acid receptor (RAR), retinoid X receptor (RXR) and vitamin D receptor (VDR). In this review we provide an overview of recent preclinical and clinical trials targeting members of the nuclear receptor superfamily for the prevention of breast cancer.
Collapse
Affiliation(s)
- Cassandra L. Moyer
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Powel H. Brown
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
24
|
Kotsopoulos J, Gronwald J, Huzarski T, Aeilts A, Randall Armel S, Karlan B, Singer CF, Eisen A, Tung N, Olopade O, Bordeleau L, Eng C, Foulkes WD, Neuhausen SL, Cullinane CA, Pal T, Fruscio R, Lubinski J, Metcalfe K, Sun P, Narod SA. Tamoxifen and the risk of breast cancer in women with a BRCA1 or BRCA2 mutation. Breast Cancer Res Treat 2023:10.1007/s10549-023-06991-3. [PMID: 37432545 DOI: 10.1007/s10549-023-06991-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/24/2023] [Indexed: 07/12/2023]
Abstract
PURPOSE Chemoprevention with a selective estrogen receptor modulator (tamoxifen or raloxifene) is a non-surgical option offered to high-risk women to reduce the risk of breast cancer. The evidence for tamoxifen benefit is based on trials conducted among predominantly postmenopausal women from the general population and on studies of contralateral breast cancer in women with a pathogenic variant (mutation hereafter) in BRCA1 or BRCA2. Tamoxifen has not been assessed as a primary prevention agent in women with an inherited BRCA mutation. METHODS We conducted a prospective analysis of tamoxifen chemoprevention and the risk of breast cancer in women with a BRCA1 or BRCA2 mutation. Data on tamoxifen (and raloxifene) use was collected by questionnaire and updated biennially. Information on incident cancers was collected by self-report and was confirmed by medical record review. In a matched analysis, we estimated the hazard ratio (HR) and 95% confidence intervals (CI) for developing a first primary breast cancer associated with tamoxifen or raloxifene use, using Cox proportional hazards analysis. RESULTS There were 4578 unaffected women in the cohort, of whom 137 reported tamoxifen use (3%), 83 reported raloxifene use (2%) and 12 used both drugs (0.3%). Women who used tamoxifen or raloxifene were matched 1:3 with women who used neither drug on year of birth, country of residence, year of study entry and gene (BRCA1 or BRCA2). We generated 202 matched pairs. After a mean follow-up of 6.8 years, there were 22 incident breast cancers diagnosed among tamoxifen/raloxifene users (10.9% of users) and 71 cases diagnosed among non-users (14.3% of non-users; HR = 0.64; 95% CI 0.40-1.03; P = 0.07). CONCLUSION Chemoprevention may be an effective risk-reduction option for BRCA mutation carriers, but further studies with longer follow-up are necessary.
Collapse
Affiliation(s)
- Joanne Kotsopoulos
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Jacek Gronwald
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Tomasz Huzarski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Amber Aeilts
- Division of Human Genetics, The Ohio State University Medical Center, Comprehensive Cancer Center, Columbus, OH, USA
| | - Susan Randall Armel
- Bhalwani Familial Cancer Clinic, Princess Margaret Cancer Centre, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Beth Karlan
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Christian F Singer
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Andrea Eisen
- Toronto-Sunnybrook Regional Cancer Center, Toronto, ON, Canada
| | - Nadine Tung
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Olufunmilayo Olopade
- Department of Medicine and Human Genetics, University of Chicago, Chicago, IL, USA
| | - Louise Bordeleau
- Department of Oncology, Juravinski Cancer Centre and McMaster University, Hamilton, ON, Canada
| | - Charis Eng
- Genomic Medicine Institute and Center for Personalized Genetic Healthcare, Cleveland Clinic, Cleveland, USA
| | - William D Foulkes
- Department of Oncology, McGill Program in Cancer Genetics, McGill University, Montreal, QC, Canada
| | - Susan L Neuhausen
- Division of Biomarkers of Early Detection and Prevention, City of Hope, Duarte, USA
| | - Carey A Cullinane
- Todd Cancer Institute, Long Beach Memorial Hospital, Long Beach, CA, USA
| | - Tuya Pal
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert Fruscio
- Clinic of Obstetrics and Gynecology, Department of Medicine and Surgery, University of Milan Bicocca, Monza, Italy
| | - Jan Lubinski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Kelly Metcalfe
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
- Bloomberg School of Nursing, University of Toronto, Toronto, ON, Canada
| | - Ping Sun
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
| | - Steven A Narod
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada.
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
- Women's College Research Institute, Women's College Hospital, 76 Grenville St., 6Th Floor, Toronto, ON, M5S 1B2, Canada.
| |
Collapse
|
25
|
Kast K, John EM, Hopper JL, Andrieu N, Noguès C, Mouret-Fourme E, Lasset C, Fricker JP, Berthet P, Mari V, Salle L, Schmidt MK, Ausems MGEM, Garcia EBG, van de Beek I, Wevers MR, Evans DG, Tischkowitz M, Lalloo F, Cook J, Izatt L, Tripathi V, Snape K, Musgrave H, Sharif S, Murray J, Colonna SV, Andrulis IL, Daly MB, Southey MC, de la Hoya M, Osorio A, Foretova L, Berkova D, Gerdes AM, Olah E, Jakubowska A, Singer CF, Tan Y, Augustinsson A, Rantala J, Simard J, Schmutzler RK, Milne RL, Phillips KA, Terry MB, Goldgar D, van Leeuwen FE, Mooij TM, Antoniou AC, Easton DF, Rookus MA, Engel C. Associations of height, body mass index, and weight gain with breast cancer risk in carriers of a pathogenic variant in BRCA1 or BRCA2: the BRCA1 and BRCA2 Cohort Consortium. Breast Cancer Res 2023; 25:72. [PMID: 37340476 PMCID: PMC10280955 DOI: 10.1186/s13058-023-01673-w] [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: 12/16/2022] [Accepted: 06/10/2023] [Indexed: 06/22/2023] Open
Abstract
INTRODUCTION Height, body mass index (BMI), and weight gain are associated with breast cancer risk in the general population. It is unclear whether these associations also exist for carriers of pathogenic variants in the BRCA1 or BRCA2 genes. PATIENTS AND METHODS An international pooled cohort of 8091 BRCA1/2 variant carriers was used for retrospective and prospective analyses separately for premenopausal and postmenopausal women. Cox regression was used to estimate breast cancer risk associations with height, BMI, and weight change. RESULTS In the retrospective analysis, taller height was associated with risk of premenopausal breast cancer for BRCA2 variant carriers (HR 1.20 per 10 cm increase, 95% CI 1.04-1.38). Higher young-adult BMI was associated with lower premenopausal breast cancer risk for both BRCA1 (HR 0.75 per 5 kg/m2, 95% CI 0.66-0.84) and BRCA2 (HR 0.76, 95% CI 0.65-0.89) variant carriers in the retrospective analysis, with consistent, though not statistically significant, findings from the prospective analysis. In the prospective analysis, higher BMI and adult weight gain were associated with higher postmenopausal breast cancer risk for BRCA1 carriers (HR 1.20 per 5 kg/m2, 95% CI 1.02-1.42; and HR 1.10 per 5 kg weight gain, 95% CI 1.01-1.19, respectively). CONCLUSION Anthropometric measures are associated with breast cancer risk for BRCA1 and BRCA2 variant carriers, with relative risk estimates that are generally consistent with those for women from the general population.
Collapse
Affiliation(s)
- Karin Kast
- Center for Hereditary Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), Medical Faculty, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Esther M John
- Department of Epidemiology & Population Health and of Medicine (Oncology), Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Nadine Andrieu
- INSERM U900, Paris, France
- Institut Curie, Paris, France
- Mines Paris Tech, Fontainebleau, France
- PSL Research University, Paris, France
| | - Catherine Noguès
- Aix Marseille Université, INSERM, IRD, SESSTIM, Marseille, France
- Département d'Anticipation et de Suivi Des Cancers, Oncogénétique Clinique, Institut Paoli-Calmettes, Marseille, France
| | | | | | | | | | | | - Lucie Salle
- Oncogénétique Poitou-Charentes, Niort, France
| | - Marjanka K Schmidt
- Division of Molecular Pathology, Netherlands Cancer Institute, Antoni Van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Margreet G E M Ausems
- Department of Genetics, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Irma van de Beek
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marijke R Wevers
- Department of Clinical Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - D Gareth Evans
- The Prevent Breast Cancer Research Unit, The Nightingale Centre, Manchester University NHS Foundation Trust, Manchester, UK
- Genomic Medicine, Division of Evolution and Genomic Sciences, The University of Manchester, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Manchester Breast Centre, Oglesby Cancer Research Centre, The Christie, University of Manchester, Manchester, UK
| | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Fiona Lalloo
- Clinical Genetics Service, Manchester Centre for Genomic Medicine, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Jackie Cook
- Sheffield Clinical Genetics Service, Sheffield Children's Hospital, Sheffield, UK
| | - Louise Izatt
- Department of Clinical Genetics, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Vishakha Tripathi
- Clinical Genetics Service, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Katie Snape
- Department of Clinical Genetics, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Hannah Musgrave
- Yorkshire Regional Genetics Service, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Saba Sharif
- West Midlands Regional Genetics Service, Birmingham Women's Hospital Healthcare NHS Trust, Edgbaston, Birmingham, UK
| | - Jennie Murray
- Yorkshire Regional Genetics Service, Leeds Teaching Hospitals NHS Trust, Leeds, UK
- West Midlands Regional Genetics Service, Birmingham Women's Hospital Healthcare NHS Trust, Edgbaston, Birmingham, UK
- South East of Scotland Regional Genetics Service, Western General Hospital, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Sarah V Colonna
- Department of Medicine and Huntsman Cancer Institute, University of Utah Health, Salt Lake City, UT, USA
| | - Irene L Andrulis
- Fred A. Litwin Center for Cancer Genetics, Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Mary B Daly
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Melissa C Southey
- Precision Medicine, School of Clinical Sciences at, Monash Health Monash University, Clayton, VIC, Australia
- Department of Clinical Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - Miguel de la Hoya
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), Madrid, Spain
| | - Ana Osorio
- Familial Cancer Clinical Unit, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO) and Spanish Network On Rare Diseases (CIBERER), Madrid, Spain
| | - Lenka Foretova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Dita Berkova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Anne-Marie Gerdes
- Department of Clinical Genetics, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Edith Olah
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary
| | - Anna Jakubowska
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
- Independent Laboratory of Molecular Biology and Genetic Diagnostics, Pomeranian Medical University, Szczecin, Poland
| | - Christian F Singer
- Department of OB/GYN and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Yen Tan
- Department of OB/GYN and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Annelie Augustinsson
- Department of Oncology, Clinical Sciences in Lund, Lund University Hospital, Lund, Sweden
| | | | - Jacques Simard
- Genomics Center, Centre Hospitalier Universitaire de Québec-Université Laval Research Center, Quebec City, QC, Canada
| | - Rita K Schmutzler
- Center for Hereditary Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), Medical Faculty, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Roger L Milne
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Kelly-Anne Phillips
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Victoria, Australia
| | - Mary Beth Terry
- Department of Epidemiology, Mailman School of Public Health and the Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - David Goldgar
- Department of Dermatology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Flora E van Leeuwen
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Thea M Mooij
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Antonis C Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Matti A Rookus
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Christoph Engel
- Institute for Medical Informatics, Statistics and Epidemiology, Leipzig University, Leipzig, Germany
| |
Collapse
|
26
|
Onji M, Penninger JM. RANKL and RANK in Cancer Therapy. Physiology (Bethesda) 2023; 38:0. [PMID: 36473204 DOI: 10.1152/physiol.00020.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Receptor activator of nuclear factor-κB (RANK) and its ligand (RANKL) are key regulators of mammalian physiology such as bone metabolism, immune tolerance and antitumor immunity, and mammary gland biology. Here, we explore the multiple functions of RANKL/RANK in physiology and pathophysiology and discuss underlying principles and strategies to modulate the RANKL/RANK pathway as a therapeutic target in immune-mediated cancer treatment.
Collapse
Affiliation(s)
- Masahiro Onji
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, VBC-Vienna BioCenter, Vienna, Austria
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, VBC-Vienna BioCenter, Vienna, Austria.,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
27
|
Zeng J, Singh S, Jiang Y, Casarez E, Atkins KA, Janes KA, Zong H. A genetic mosaic mouse model illuminates the pre-malignant progression of basal-like breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.25.538333. [PMID: 37163037 PMCID: PMC10168298 DOI: 10.1101/2023.04.25.538333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Basal-like breast cancer is an aggressive breast cancer subtype, often characterized by a deficiency in BRCA1 function and concomitant loss of p53 . While conventional mouse models enable the investigation of its malignant stages, one that reveals its initiation and pre-malignant progression is lacking. Here, we leveraged a mouse genetic system known as M osaic A nalysis with D ouble M arkers (MADM) to generate rare GFP-labeled Brca1 , p53 -deficient cells alongside RFP+ wildtype sibling cells in the mammary gland. The mosaicism resembles the sporadic initiation of human cancer and enables spatially resolved analysis of mutant cells in comparison to paired wildtype sibling cells. Mammary tumors arising in the model show transcriptomic and genomic characteristics similar to human basal-like breast cancer. Analysis of GFP+ mutant cells at interval time points before malignancy revealed a stepwise progression of lesions from focal expansion to hyper-alveolarization and then to micro-invasion. These stereotyped morphologies indicate the pre-malignant stage irrespective of the time point at which it is observed. Paired analysis of GFP-RFP siblings during focal expansion suggested that hyper-alveolarized structures originate from ductal rather than alveolar cells, despite their morphological similarities to alveoli. Evidence for luminal-to-basal transition at the pre-malignant stages was restricted to cells that had escaped hyper-alveoli and progressed to micro-invasive lesions. Our MADM-based mouse model presents a useful tool for studying the pre-malignancy of basal-like breast cancer. Summary statement A mouse model recapitulates the process of human basal-like breast tumorigenesis initiated from sporadic Brca1, p53 -deficient cells, empowering spatially-resolved analysis of mutant cells during pre-malignant progression.
Collapse
|
28
|
Ciscar M, Trinidad EM, Perez‐Chacon G, Alsaleem M, Jimenez M, Jimenez‐Santos MJ, Perez‐Montoyo H, Sanz‐Moreno A, Vethencourt A, Toss M, Petit A, Soler‐Monso MT, Lopez V, Gomez‐Miragaya J, Gomez‐Aleza C, Dobrolecki LE, Lewis MT, Bruna A, Mouron S, Quintela‐Fandino M, Al‐Shahrour F, Martinez‐Aranda A, Sierra A, Green AR, Rakha E, Gonzalez‐Suarez E. RANK is a poor prognosis marker and a therapeutic target in ER-negative postmenopausal breast cancer. EMBO Mol Med 2023; 15:e16715. [PMID: 36880458 PMCID: PMC10086586 DOI: 10.15252/emmm.202216715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/24/2023] [Accepted: 02/08/2023] [Indexed: 03/08/2023] Open
Abstract
Despite strong preclinical data, the therapeutic benefit of the RANKL inhibitor, denosumab, in breast cancer patients, beyond the bone, is unclear. Aiming to select patients who may benefit from denosumab, we hereby analyzed RANK and RANKL protein expression in more than 2,000 breast tumors (777 estrogen receptor-negative, ER- ) from four independent cohorts. RANK protein expression was more frequent in ER- tumors, where it associated with poor outcome and poor response to chemotherapy. In ER- breast cancer patient-derived orthoxenografts (PDXs), RANKL inhibition reduced tumor cell proliferation and stemness, regulated tumor immunity and metabolism, and improved response to chemotherapy. Intriguingly, tumor RANK protein expression associated with poor prognosis in postmenopausal breast cancer patients, activation of NFKB signaling, and modulation of immune and metabolic pathways, suggesting that RANK signaling increases after menopause. Our results demonstrate that RANK protein expression is an independent biomarker of poor prognosis in postmenopausal and ER- breast cancer patients and support the therapeutic benefit of RANK pathway inhibitors, such as denosumab, in breast cancer patients with RANK+ ER- tumors after menopause.
Collapse
Affiliation(s)
- Marina Ciscar
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO)MadridSpain
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| | - Eva M Trinidad
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| | - Gema Perez‐Chacon
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO)MadridSpain
| | - Mansour Alsaleem
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of MedicineUniversity of Nottingham Biodiscovery Institute, University ParkNottinghamUK
- Present address:
Department of Applied Medical Science, Applied CollegeQassim UniversityUnayzahSaudi Arabia
| | - Maria Jimenez
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO)MadridSpain
| | - Maria J Jimenez‐Santos
- Bioinformatics Unit, Structural Biology, Spanish National Cancer Research Centre (CNIO)MadridSpain
| | | | - Adrian Sanz‐Moreno
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| | - Andrea Vethencourt
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
- Medical Oncology, Breast Unit, Catalan Institute of Oncology (ICO)University Hospital of BellvitgeBarcelonaSpain
| | - Michael Toss
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of MedicineUniversity of Nottingham Biodiscovery Institute, University ParkNottinghamUK
| | - Anna Petit
- Pathology DepartmentUniversity Hospital of Bellvitge, IDIBELLBarcelonaSpain
| | | | - Victor Lopez
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO)MadridSpain
| | | | - Clara Gomez‐Aleza
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| | - Lacey E Dobrolecki
- Molecular and Cellular Biology and RadiologyThe Lester and Sue Smith Breast Center, Baylor College of MedicineHoustonTexasUSA
| | - Michael T Lewis
- Molecular and Cellular Biology and RadiologyThe Lester and Sue Smith Breast Center, Baylor College of MedicineHoustonTexasUSA
| | - Alejandra Bruna
- Cancer Research UK Cambridge CentreCambridgeUK
- Present address:
Molecular Pathology DivisionCentre for Paediatric Oncology Experimental MedicineCentre for Cancer EvolutionThe Institute of Cancer ResearchLondonUK
| | - Silvana Mouron
- Breast Cancer Clinical Research Unit, Clinical Research ProgramSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Miguel Quintela‐Fandino
- Breast Cancer Clinical Research Unit, Clinical Research ProgramSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Fatima Al‐Shahrour
- Bioinformatics Unit, Structural Biology, Spanish National Cancer Research Centre (CNIO)MadridSpain
| | - Antonio Martinez‐Aranda
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
- Medical Oncology, Breast Unit, Catalan Institute of Oncology (ICO)University Hospital of BellvitgeBarcelonaSpain
| | - Angels Sierra
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
- Present address:
Laboratory of Experimental Oncological Neurosurgery, Neurosurgery ServiceHospital Clinic de Barcelona‐FCRBBarcelonaSpain
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of MedicineUniversity of Nottingham Biodiscovery Institute, University ParkNottinghamUK
| | - Emad Rakha
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of MedicineUniversity of Nottingham Biodiscovery Institute, University ParkNottinghamUK
| | - Eva Gonzalez‐Suarez
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO)MadridSpain
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| |
Collapse
|
29
|
Lambouras M, Roelofs C, Pereira M, Gruber E, Vieusseux JL, Lanteri P, Johnstone CN, Muntz F, O’Toole S, Ooms LM, Mitchell CA, Anderson RL, Britt KL. Functional and Phenotypic Characterisations of Common Syngeneic Tumour Cell Lines as Estrogen Receptor-Positive Breast Cancer Models. Int J Mol Sci 2023; 24:ijms24065666. [PMID: 36982737 PMCID: PMC10053941 DOI: 10.3390/ijms24065666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Estrogen receptor-positive breast cancers (ER+ BCas) are the most common form of BCa and are increasing in incidence, largely due to changes in reproductive practices in recent decades. Tamoxifen is prescribed as a component of standard-of-care endocrine therapy for the treatment and prevention of ER+ BCa. However, it is poorly tolerated, leading to low uptake of the drug in the preventative setting. Alternative therapies and preventatives for ER+ BCa are needed but development is hampered due to a paucity of syngeneic ER+ preclinical mouse models that allow pre-clinical experimentation in immunocompetent mice. Two ER-positive models, J110 and SSM3, have been reported in addition to other tumour models occasionally shown to express ER (for example 4T1.2, 67NR, EO771, D2.0R and D2A1). Here, we have assessed ER expression and protein levels in seven mouse mammary tumour cell lines and their corresponding tumours, in addition to their cellular composition, tamoxifen sensitivity and molecular phenotype. By immunohistochemical assessment, SSM3 and, to a lesser extent, 67NR cells are ER+. Using flow cytometry and transcript expression we show that SSM3 cells are luminal in nature, whilst D2.0R and J110 cells are stromal/basal. The remainder are also stromal/basal in nature; displaying a stromal or basal Epcam/CD49f FACS phenotype and stromal and basal gene expression signatures are overrepresented in their transcript profile. Consistent with a luminal identity for SSM3 cells, they also show sensitivity to tamoxifen in vitro and in vivo. In conclusion, the data indicate that the SSM3 syngeneic cell line is the only definitively ER+ mouse mammary tumour cell line widely available for pre-clinical research.
Collapse
Affiliation(s)
- Maria Lambouras
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Charlotte Roelofs
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
| | - Melrine Pereira
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Emily Gruber
- The Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Jessica L. Vieusseux
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Patrick Lanteri
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Cameron N. Johnstone
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3084, Australia
| | - Fenella Muntz
- The Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Sandra O’Toole
- Sydney Medical School, University of Sydney, Camperdown, NSW 2050, Australia
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- Australian Clinical Labs, Sydney, NSW 2153, Australia
| | - Lisa M. Ooms
- Cancer Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Christina A. Mitchell
- Cancer Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Robin L. Anderson
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
- The Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3084, Australia
| | - Kara L. Britt
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia
- Correspondence: ; Tel.: +61-38599-7110
| |
Collapse
|
30
|
Ishikawa T. Differences between zoledronic acid and denosumab for breast cancer treatment. J Bone Miner Metab 2023; 41:301-306. [PMID: 36879056 DOI: 10.1007/s00774-023-01408-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 03/08/2023]
Abstract
Zoledronic acid and denosumab are bone-modifying agents that are clinically important in multiple aspects of bone management for breast cancer patients. These aspects include the prevention of osteoporosis induced by cancer-treatment bone loss, treatment and prevention of bone metastasis, and improvement of survival directly or indirectly by maintaining bone health. Interestingly, zoledronic acid and denosumab have different anticancer activities, and they may be associated with the improvement of survival of breast cancer patients under different mechanisms. Zoledronic acid is the most potent bisphosphonate. It provides significant benefits for improving breast cancer mortality in patients with suppressed estrogen level such as in postmenopausal or ovarian suppression condition. Although denosumab's anticancer activity has not been clearly proven compared with zoledronic acid's anticancer activity, denosumab is promising in preventing BRCA1 mutant breast cancer because RANKL is a targetable pathway in BRCA1-associated tumorigenesis. Further studies and more effective clinical use of these agents are anticipated to contribute to the improvement of the clinical outcome of breast cancer patients.
Collapse
Affiliation(s)
- Takashi Ishikawa
- Department of Breast Oncology and Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan.
| |
Collapse
|
31
|
Wu B, Qi L, Chiang HC, Pan H, Zhang X, Greenbaum A, Stark E, Wang LJ, Chen Y, Haddad BR, Clagett D, Isaacs C, Elledge R, Horvath A, Hu Y, Li R. BRCA1 deficiency in mature CD8 + T lymphocytes impairs antitumor immunity. J Immunother Cancer 2023; 11:jitc-2022-005852. [PMID: 36731891 PMCID: PMC9896206 DOI: 10.1136/jitc-2022-005852] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2023] [Indexed: 02/04/2023] Open
Abstract
Women with BRCA1 germline mutations have approximately an 80% lifetime chance of developing breast cancer. While the tumor suppressor function of BRCA1 in breast epithelium has been studied extensively, it is not clear whether BRCA1 deficiency in non-breast somatic cells also contribute to tumorigenesis. Here, we report that mouse Brca1 knockout (KO) in mature T lymphocytes compromises host antitumor immune response to transplanted syngeneic mouse mammary tumors. T cell adoptive transfer further corroborates CD8+ T cell-intrinsic impact of Brca1 KO on antitumor adaptive immunity. T cell-specific Brca1 KO mice exhibit fewer total CD8+, more exhausted, reduced cytotoxic, and reduced memory tumor-infiltrating T cell populations. Consistent with the preclinical data, cancer-free BRCA1 mutation-carrying women display lower abundance of circulating CD8+ lymphocytes than the age-matched control group. Thus, our findings support the notion that BRCA1 deficiency in adaptive immunity could contribute to BRCA1-related tumorigenesis. We also suggest that prophylactic boosting of adaptive immunity may reduce cancer incidence among at-risk women.
Collapse
Affiliation(s)
- Bogang Wu
- Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Leilei Qi
- Department of Anatomy & Cell Biology, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Huai-Chin Chiang
- Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Haihui Pan
- Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Xiaowen Zhang
- Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Alexandra Greenbaum
- Ruth Paul Cancer Genetics and Prevention Program, Medical Faculty Associates, The George Washington University, Washington, District of Columbia, USA
| | - Elizabeth Stark
- Ruth Paul Cancer Genetics and Prevention Program, Medical Faculty Associates, The George Washington University, Washington, District of Columbia, USA
| | - Li-Ju Wang
- Department of Biostatistics, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Yidong Chen
- Department of Biostatistics, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Bassem R Haddad
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA
| | - Dionyssia Clagett
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA
| | - Claudine Isaacs
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA
| | | | - Anelia Horvath
- Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Yanfen Hu
- Department of Anatomy & Cell Biology, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Rong Li
- Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| |
Collapse
|
32
|
Takeuchi Y, Gotoh N. Inflammatory cytokine-enriched microenvironment plays key roles in the development of breast cancers. Cancer Sci 2023; 114:1792-1799. [PMID: 36704829 PMCID: PMC10154879 DOI: 10.1111/cas.15734] [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: 12/05/2022] [Revised: 12/29/2022] [Accepted: 01/20/2023] [Indexed: 01/28/2023] Open
Abstract
As the incidence of breast cancer continues to increase, it is critical to develop prevention strategies for this disease. Inflammation underlies the onset of the disease, and NF-κB is a master transcription factor for inflammation. Nuclear factor-κB (NF-κB) is activated in a variety of cell types, including normal epithelial cells, cancer cells, cancer-associated fibroblasts (CAFs), and immune cells. Ductal carcinoma in situ (DCIS) is the earliest stage of breast cancer, and not all DCIS lesions develop into invasive breast cancers (IBC). Currently, most patients with DCIS undergo surgery with postoperative therapy, although there is a risk of overtreatment. In BRCA mutants, receptor activator of NF-κB (RANK)-positive progenitors serve as the cell of origin, and treatment using the RANK monoclonal antibody reduces the risk of IBC. There is still an unmet need to diagnose malignant DCIS, which has the potential to progress to IBC, and to establish appropriate prevention strategies. We recently demonstrated novel molecular mechanisms for NF-κB activation in premalignant mammary tissues, which include DCIS, and the resultant cytokine-enriched microenvironment is essential for breast cancer development. On the early endosomes in a few epithelial cells, the adaptor protein FRS2β, forming a complex with ErbB2, carries the IκB kinase (IKK) complex and leads to the activation of NF-κB, thereby inducing a variety of cytokines. Therefore, the FRS2β-NFκB axis in the inflammatory premalignant environment could be targetable to prevent IBC. Further analysis of the molecular mechanisms of inflammation in the premalignant microenvironment is necessary to prevent the risk of IBC.
Collapse
Affiliation(s)
- Yasuto Takeuchi
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan.,Institute for Frontier Science Initiative, Kanazawa University, Kanazawa City, Japan
| | - Noriko Gotoh
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan.,Institute for Frontier Science Initiative, Kanazawa University, Kanazawa City, Japan
| |
Collapse
|
33
|
Ogony J, Hoskin TL, Stallings-Mann M, Winham S, Brahmbhatt R, Arshad MA, Kannan N, Peña A, Allers T, Brown A, Sherman ME, Visscher DW, Knutson KL, Radisky DC, Degnim AC. Immune cells are increased in normal breast tissues of BRCA1/2 mutation carriers. Breast Cancer Res Treat 2023; 197:277-285. [PMID: 36380012 PMCID: PMC10168666 DOI: 10.1007/s10549-022-06786-y] [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: 06/15/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022]
Abstract
PURPOSE Breast cancer risk is elevated in pathogenic germline BRCA 1/2 mutation carriers due to compromised DNA quality control. We hypothesized that if immunosurveillance promotes tumor suppression, then normal/benign breast lobules from BRCA carriers may demonstrate higher immune cell densities. METHODS We assessed immune cell composition in normal/benign breast lobules from age-matched women with progressively increased breast cancer risk, including (1) low risk: 19 women who donated normal breast tissue to the Komen Tissue Bank (KTB) at Indiana University Simon Cancer Center, (2) intermediate risk: 15 women with biopsy-identified benign breast disease (BBD), and (3) high risk: 19 prophylactic mastectomies from women with germline mutations in BRCA1/2 genes. We performed immunohistochemical stains and analysis to quantitate immune cell densities from digital images in up to 10 representative lobules per sample. Median cell counts per mm2 were compared between groups using Wilcoxon rank-sum tests. RESULTS Normal/benign breast lobules from BRCA carriers had significantly higher densities of immune cells/mm2 compared to KTB normal donors (all p < 0.001): CD8 + 354.4 vs 150.9; CD4 + 116.3 vs 17.7; CD68 + 237.5 vs 57.8; and CD11c + (3.5% vs 0.4% pixels positive). BBD tissues differed from BRCA carriers only in CD8 + cells but had higher densities of CD4 + , CD11c + , and CD68 + immune cells compared to KTB donors. CONCLUSIONS These preliminary analyses show that normal/benign breast lobules of BRCA mutation carriers contain increased immune cells compared with normal donor breast tissues, and BBD tissues appear overall more similar to BRCA carriers.
Collapse
Affiliation(s)
- Joshua Ogony
- Quantitative Health Sciences, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Tanya L Hoskin
- Quantitative Health Sciences, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Melody Stallings-Mann
- Department of Cancer Biology, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Stacey Winham
- Quantitative Health Sciences, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Rushin Brahmbhatt
- Department of Surgery, Mayo Clinic College of Medicine, 200 First St SW, Rochester, MN, 55905, USA
| | - Muhammad Asad Arshad
- Department of Surgery, Mayo Clinic College of Medicine, 200 First St SW, Rochester, MN, 55905, USA
| | - Nagarajan Kannan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Alvaro Peña
- Department of Surgery, Mayo Clinic College of Medicine, 200 First St SW, Rochester, MN, 55905, USA
| | - Teresa Allers
- Department of Surgery, Mayo Clinic College of Medicine, 200 First St SW, Rochester, MN, 55905, USA
| | - Alyssa Brown
- Mayo Graduate School, Mayo Clinic, Rochester, MN, USA
| | - Mark E Sherman
- Quantitative Health Sciences, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Daniel W Visscher
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Amy C Degnim
- Department of Surgery, Mayo Clinic College of Medicine, 200 First St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
34
|
Zhang Y, Liang J, Liu P, Wang Q, Liu L, Zhao H. The RANK/RANKL/OPG system and tumor bone metastasis: Potential mechanisms and therapeutic strategies. Front Endocrinol (Lausanne) 2022; 13:1063815. [PMID: 36589815 PMCID: PMC9800780 DOI: 10.3389/fendo.2022.1063815] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
With the markedly increased diagnosis and incidence of cancer in the population, tumor bone metastasis has become a frequent event in tumor patients. Healthy bone integrity is maintained by a delicate balance between bone formation and bone resorption. Unfortunately, many tumors, such as prostate and breast, often metastasize to the bone, and the alterations to the bone homeostasis can particularly favor tumor homing and consequent osteolytic or osteoblastic lesions. Receptor activator of NF-κB ligand (RANKL), its receptor RANK, and osteoprotegerin (OPG) are involved in the regulation of the activation, differentiation, and survival of osteoclasts, which play critical roles in bone metastasis formation. High rates of osteoclastic bone resorption significantly increase fracture risk, cause severe bone pain, and contribute to homing tumor cells in bone and bone marrow. Consequently, suppression of the RANK/RANKL/OPG system and osteoclastic activity can not only ameliorate bone resorption but may also prevent tumor bone metastases. This review summarizes the important role of the RANK/RANKL/OPG system and osteoclasts in bone homeostasis and its effect on tumor bone metastasis and discusses therapeutic strategies based on RANKL inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | - Hongmou Zhao
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
35
|
Trivedi MS, Arber N, Friedman E, Garber JE, Holcomb K, Horowitz NS, Wright JD, Lee JJ, Vornik LA, Abutaseh S, Castile T, Sauter ER, Dimond E, Heckman-Stoddard BM, House M, Samimi G, Brown PH, Crew KD. Lessons from the Failure to Complete a Trial of Denosumab in Women With a Pathogenic BRCA1/2 Variant Scheduling Risk-Reducing Salpingo-Oophorectomy. Cancer Prev Res (Phila) 2022; 15:721-726. [PMID: 36001346 PMCID: PMC10441620 DOI: 10.1158/1940-6207.capr-22-0051] [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: 02/18/2022] [Revised: 05/19/2022] [Accepted: 07/26/2022] [Indexed: 01/31/2023]
Abstract
Female carriers of pathogenic/likely pathogenic (P/LP) BRCA1/2 variants are at increased risk of developing breast and ovarian cancer. Currently, the only effective strategy for ovarian cancer risk reduction is risk-reducing bilateral salpingo-oophorectomy (RR-BSO), which carries adverse effects related to early menopause. There is ongoing investigation of inhibition of the RANK ligand (RANKL) with denosumab as a means of chemoprevention for breast cancer in carriers of BRCA1 P/LP variants. Through the NCI Division of Cancer Prevention (DCP) Early Phase Clinical Trials Prevention Consortia, a presurgical pilot study of denosumab was developed in premenopausal carriers of P/LP BRCA1/2 variants scheduled for RR-BSO with the goal of collecting valuable data on the biologic effects of denosumab on gynecologic tissue. The study was terminated early due to the inability to accrue participants. Challenges which impacted the conduct of this study included a study design with highly selective eligibility criteria and requirements and the COVID-19 pandemic. It is critical to reflect on these issues to enhance the successful completion of future prevention studies in individuals with hereditary cancer syndromes.
Collapse
Affiliation(s)
| | - Nadir Arber
- Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Eitan Friedman
- Chaim Sheba Medical Center, Tel-Aviv University Medical School, Tel Aviv, Israel
| | | | | | | | - Jason D. Wright
- Columbia University Irving Medical Center, New York, New York
| | - J. Jack Lee
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lana A. Vornik
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Saba Abutaseh
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tawana Castile
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Edward R. Sauter
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Eileen Dimond
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | | | - Margaret House
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Goli Samimi
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Powel H. Brown
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
36
|
Saunders CM. Breast surgery: a narrative review. Med J Aust 2022; 217:262-267. [PMID: 35988063 PMCID: PMC9541238 DOI: 10.5694/mja2.51678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022]
Abstract
Breast cancer is the commonest human cancer globally and one in seven Australian women will develop it in their lifetime. Surgery is the mainstay of management both for women who are at high risk of breast cancer and for those who have been diagnosed. Increased understanding of how to predict who is most at risk of breast cancer is leading to the possibility of risk‐based screening, allowing better and more targeted early detection for women at high risk, and contrast imaging techniques are proving more accurate in diagnosing and staging cancer. The evolution of surgical practice includes the widespread use of oncoplastic surgery, allowing better cosmetic and oncological outcomes; reconstructive surgical advances, using free flap techniques; and sequencing of systemic and local therapies to better tailor treatments to the patient’s cancer and improve outcomes. Recognition of side effects of breast cancer treatment have led to improvement in the management of conditions such as chronic pain and lymphoedema, as well as addressing the psychosocial, body image and sexual complications caused by the cancer and its treatment.
Collapse
|
37
|
Ductal keratin 15 + luminal progenitors in normal breast exhibit a basal-like breast cancer transcriptomic signature. NPJ Breast Cancer 2022; 8:81. [PMID: 35821504 PMCID: PMC9276673 DOI: 10.1038/s41523-022-00444-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/10/2022] [Indexed: 11/09/2022] Open
Abstract
Normal breast luminal epithelial progenitors have been implicated as cell of origin in basal-like breast cancer, but their anatomical localization remains understudied. Here, we combine collection under the microscope of organoids from reduction mammoplasties and single-cell mRNA sequencing (scRNA-seq) of FACS-sorted luminal epithelial cells with multicolor imaging to profile ducts and terminal duct lobular units (TDLUs) and compare them with breast cancer subtypes. Unsupervised clustering reveals eleven distinct clusters and a differentiation trajectory starting with keratin 15+ (K15+) progenitors enriched in ducts. Spatial mapping of luminal progenitors is confirmed at the protein level by staining with critical duct markers. Comparison of the gene expression profiles of normal luminal cells with those of breast cancer subtypes suggests a strong correlation between normal breast ductal progenitors and basal-like breast cancer. We propose that K15+ basal-like breast cancers originate in ductal progenitors, which emphasizes the importance of not only lineages but also cellular position within the ductal-lobular tree.
Collapse
|
38
|
Functions of Breast Cancer Predisposition Genes: Implications for Clinical Management. Int J Mol Sci 2022; 23:ijms23137481. [PMID: 35806485 PMCID: PMC9267387 DOI: 10.3390/ijms23137481] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
Approximately 5–10% of all breast cancer (BC) cases are caused by germline pathogenic variants (GPVs) in various cancer predisposition genes (CPGs). The most common contributors to hereditary BC are BRCA1 and BRCA2, which are associated with hereditary breast and ovarian cancer (HBOC). ATM, BARD1, CHEK2, PALB2, RAD51C, and RAD51D have also been recognized as CPGs with a high to moderate risk of BC. Primary and secondary cancer prevention strategies have been established for HBOC patients; however, optimal preventive strategies for most hereditary BCs have not yet been established. Most BC-associated CPGs participate in DNA damage repair pathways and cell cycle checkpoint mechanisms, and function jointly in such cascades; therefore, a fundamental understanding of the disease drivers in such cascades can facilitate the accurate estimation of the genetic risk of developing BC and the selection of appropriate preventive and therapeutic strategies to manage hereditary BCs. Herein, we review the functions of key BC-associated CPGs and strategies for the clinical management in individuals harboring the GPVs of such genes.
Collapse
|
39
|
Edaily S, Abdel-Razeq H. Management Strategies of Breast Cancer Patients with BRCA1 and BRCA2 Pathogenic Germline Variants. Onco Targets Ther 2022; 15:815-826. [PMID: 35923470 PMCID: PMC9343017 DOI: 10.2147/ott.s369844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/13/2022] [Indexed: 11/23/2022] Open
Abstract
Most of breast cancer cases are sporadic; however, 15–20% are associated with family history, and some are inherited. Among those, deleterious mutations in BRCA1 and BRCA2 tumor suppressor genes are the most commonly encountered pathogenic germline variants (PGVs). Given the availability and affordability of multi-gene panel sequencing technologies, testing for PGVs is commonly practiced. With our enhanced understanding of cancer genetics and specific molecular alterations, the better acceptance of risk-directed screening and prevention, and the recent introduction of novel targeted therapies, management of BRCA-positive breast cancers is taking a new direction, focusing more on risk-reducing interventions, including mastectomy and salpingo-oophorectomy, and incorporating special treatment regimens, including platinum-based chemotherapy, and the recently-introduced PARP (poly (ADP)-ribose polymerase) inhibitors. Given the recent advances in reproductive technology and molecular medicine, younger women with PGVs may have the option of embryo selection through preimplantation genetic testing and diagnosis, thus preventing the potential transmission of the implicated genes to the next generations. In this review, we cover the clinical implications of identifying a pathogenic germline mutation in BRCA1 and BRCA2 genes in breast cancer patients, and their relatives, across the continuum of care – from cancer prevention and early detection, through active treatment and up to survivorship issues.
Collapse
Affiliation(s)
- Sarah Edaily
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Hikmat Abdel-Razeq
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
- Correspondence: Hikmat Abdel-Razeq, Department of Internal Medicine, King Hussein Cancer Center, Queen Rania Al Abdullah Street, P.O. Box: 1269, Amman, 11941, Jordan, Tel +962-6 5300460, Ext: 1000, Email
| |
Collapse
|
40
|
Bartlett TE, Evans I, Jones A, Barrett JE, Haran S, Reisel D, Papaikonomou K, Jones L, Herzog C, Pashayan N, Simões BM, Clarke RB, Evans DG, Ghezelayagh TS, Ponandai-Srinivasan S, Boggavarapu NR, Lalitkumar PG, Howell SJ, Risques RA, Rådestad AF, Dubeau L, Gemzell-Danielsson K, Widschwendter M. Antiprogestins reduce epigenetic field cancerization in breast tissue of young healthy women. Genome Med 2022; 14:64. [PMID: 35701800 PMCID: PMC9199133 DOI: 10.1186/s13073-022-01063-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/17/2022] [Indexed: 02/08/2023] Open
Abstract
Background Breast cancer is a leading cause of death in premenopausal women. Progesterone drives expansion of luminal progenitor cells, leading to the development of poor-prognostic breast cancers. However, it is not known if antagonising progesterone can prevent breast cancers in humans. We suggest that targeting progesterone signalling could be a means of reducing features which are known to promote breast cancer formation.
Methods In healthy premenopausal women with and without a BRCA mutation we studied (i) estrogen and progesterone levels in saliva over an entire menstrual cycle (n = 20); (ii) cancer-free normal breast-tissue from a control population who had no family or personal history of breast cancer and equivalently from BRCA1/2 mutation carriers (n = 28); triple negative breast cancer (TNBC) biopsies and healthy breast tissue taken from sites surrounding the TNBC in the same individuals (n = 14); and biopsies of ER+ve/PR+ve stage T1–T2 cancers and healthy breast tissue taken from sites surrounding the cancer in the same individuals (n = 31); and (iii) DNA methylation and DNA mutations in normal breast tissue (before and after treatment) from clinical trials that assessed the potential preventative effects of vitamins and antiprogestins (mifepristone and ulipristal acetate; n = 44).
Results Daily levels of progesterone were higher throughout the menstrual cycle of BRCA1/2 mutation carriers, raising the prospect of targeting progesterone signalling as a means of cancer risk reduction in this population. Furthermore, breast field cancerization DNA methylation signatures reflective of (i) the mitotic age of normal breast epithelium and (ii) the proportion of luminal progenitor cells were increased in breast cancers, indicating that luminal progenitor cells with elevated replicative age are more prone to malignant transformation. The progesterone receptor antagonist mifepristone reduced both the mitotic age and the proportion of luminal progenitor cells in normal breast tissue of all control women and in 64% of BRCA1/2 mutation carriers. These findings were validated by an alternate progesterone receptor antagonist, ulipristal acetate, which yielded similar results. Importantly, mifepristone reduced both the TP53 mutation frequency as well as the number of TP53 mutations in mitotic-age-responders. Conclusions These data support the potential usage of antiprogestins for primary prevention of poor-prognostic breast cancers. Trial registration Clinical trial 1 Mifepristone treatment prior to insertion of a levonorgestrel releasing intrauterine system for improved bleeding control – a randomized controlled trial, clinicaltrialsregister.eu, 2009-009014-40; registered on 20 July 2009. Clinical trial 2 The effect of a progesterone receptor modulator on breast tissue in women with BRCA1 and 2 mutations, clinicaltrials.gov, NCT01898312; registered on 07 May 2013. Clinical trial 3 A pilot prevention study of the effects of the anti- progestin Ulipristal Acetate (UA) on surrogate markers of breast cancer risk, clinicaltrialsregister.eu, 2015-001587-19; registered on 15 July 2015. Supplementary Information The online version contains supplementary material available at 10.1186/s13073-022-01063-5.
Collapse
Affiliation(s)
- Thomas E Bartlett
- Department of Statistical Science, University College London, London, WC1E 7HB, UK
| | - Iona Evans
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London, WC1E 6AU, UK
| | - Allison Jones
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London, WC1E 6AU, UK
| | - James E Barrett
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London, WC1E 6AU, UK.,European Translational Oncology Prevention and Screening (EUTOPS) Institute, Universität Innsbruck, 6060, Hall in Tirol, Austria.,Research Institute for Biomedical Aging Research, Universität Innsbruck, 6020, Innsbruck, Austria
| | - Shaun Haran
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London, WC1E 6AU, UK
| | - Daniel Reisel
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London, WC1E 6AU, UK
| | - Kiriaki Papaikonomou
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Louise Jones
- Centre for Tumour Biology Department, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Chiara Herzog
- European Translational Oncology Prevention and Screening (EUTOPS) Institute, Universität Innsbruck, 6060, Hall in Tirol, Austria.,Research Institute for Biomedical Aging Research, Universität Innsbruck, 6020, Innsbruck, Austria
| | - Nora Pashayan
- Department of Applied Health Research, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK
| | - Bruno M Simões
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK, England
| | - Robert B Clarke
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK, England
| | - D Gareth Evans
- University of Manchester, St. Mary's Hospital, and University Hospital of South Manchester, Manchester, UK
| | - Talayeh S Ghezelayagh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.,Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, 98195, USA
| | - Sakthivignesh Ponandai-Srinivasan
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Nageswara R Boggavarapu
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Parameswaran G Lalitkumar
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Sacha J Howell
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK, England.,Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Rosa Ana Risques
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Angelique Flöter Rådestad
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Louis Dubeau
- Department of Pathology, Keck School of Medicine, USC/Norris Comprehensive Cancer Centre, University of Southern California, Los Angeles, USA
| | - Kristina Gemzell-Danielsson
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Martin Widschwendter
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London, WC1E 6AU, UK. .,European Translational Oncology Prevention and Screening (EUTOPS) Institute, Universität Innsbruck, 6060, Hall in Tirol, Austria. .,Research Institute for Biomedical Aging Research, Universität Innsbruck, 6020, Innsbruck, Austria. .,Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
41
|
Xia YY, Kotsopoulos J. Beyond the pill: contraception and the prevention of hereditary ovarian cancer. Hered Cancer Clin Pract 2022; 20:21. [PMID: 35668475 PMCID: PMC9169328 DOI: 10.1186/s13053-022-00227-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 05/05/2022] [Indexed: 12/04/2022] Open
Abstract
BRCA1 and BRCA2 mutation carriers face an elevated lifetime risk of developing ovarian cancer. Oral contraceptives have been shown to significantly decrease the risk of ovarian cancer by approximately 50% in this high-risk population. Changes in contraceptive formulations and patterns of use over time have introduced lower hormonal dosages, different steroid types and non-oral routes of administration. Specifically, there has been a considerable shift in patterns of contraceptive use and the increase in the uptake of non-oral, long-acting, reversible contraception (e.g., intrauterine devices, implants, injections) has corresponded to a decline in oral contraceptive pill use. Whether or not these other methods confer a protective effect against ovarian cancer in the general population is not clear. To our knowledge, there have been no such studies conducted among BRCA mutation carriers. Furthermore, the impact of these changes on the risk of developing ovarian cancer is not known. In this article, we will review the existing epidemiologic evidence regarding the role of contraceptives and the risk of ovarian cancer with a focus on women with a BRCA1 or BRCA2 mutation. We will discuss recent findings and gaps in the knowledge while extrapolating from studies conducted among women from the noncarrier population.
Collapse
Affiliation(s)
- Yue Yin Xia
- Women's College Research Institute, Women's College Hospital, 76 Grenville Street, 6th Floor, Toronto, ON, M5S 1B2, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Joanne Kotsopoulos
- Women's College Research Institute, Women's College Hospital, 76 Grenville Street, 6th Floor, Toronto, ON, M5S 1B2, Canada.
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
42
|
Lammert J, Basrai M, Struck J, Hartmann O, Engel C, Bischoff SC, Berling-Ernst A, Halle M, Kiechle M, Grill S. Associations of Plasma Bioactive Adrenomedullin Levels with Cardiovascular Risk Factors in BRCA1/2 Mutation Carriers. Geburtshilfe Frauenheilkd 2022; 82:601-609. [PMID: 35903716 PMCID: PMC9315398 DOI: 10.1055/a-1811-2164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/28/2022] [Indexed: 11/18/2022] Open
Abstract
Background
Cardiovascular disease (CVD) is an important cause of morbidity and mortality in breast cancer survivors. Effective screening modalities to identify CVD risk are lacking
in this population. Adrenomedullin (ADM) has been suggested as a biomarker for subclinical cardiac dysfunction in the general population. Levels of ADM have been proven to be responsive to
lifestyle changes that lead to improved cardiovascular health. As
BRCA1/2
mutation carriers are deemed to be at an increased risk for CVD, the aim of this study was to examine plasma
ADM levels in a cohort of
BRCA
mutation carriers and to assess their association with cardiovascular risk factors.
Methods
Plasma ADM concentrations were measured in 292 female
BRCA1/2
mutation carriers with and without a history of breast cancer. Subjects were classified into high versus
low ADM levels based on the median ADM level in the entire cohort (13.8 pg/mL). Logistic regression models were used to estimate the odds ratios (OR) of having elevated ADM levels by several
cardiovascular risk factors.
Results
Of all women (median age: 43 years), 57.5% had a previous diagnosis of breast cancer. The median time between diagnosis and study entry was three years (range: 0 – 32 years).
Women presenting with metabolic syndrome had 22-fold increased odds of having elevated ADM levels (p < 0.001). Elevated ADM levels were associated with lower cardiorespiratory fitness
(OR = 0.88, p < 0.001) and several parameters of obesity (p < 0.001). ADM levels were higher in women who have ever smoked (OR = 1.72, p = 0.02). ADM levels were not associated with a
previous diagnosis of breast cancer (p = 0.28).
Conclusions
This is the first study in
BRCA
mutation carriers that has linked circulating ADM levels to traditional cardiovascular risk factors. The long-term clinical
implications of these findings are yet to be determined.
Collapse
Affiliation(s)
- Jacqueline Lammert
- Department of Gynecology and Center for Hereditary Breast and Ovarian Cancer, University Hospital rechts der Isar, Technical University of Munich (TUM), Munich,
Germany
| | - Maryam Basrai
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | | | | | - Christoph Engel
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Stephan C. Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Anika Berling-Ernst
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Martin Halle
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Marion Kiechle
- Department of Gynecology and Center for Hereditary Breast and Ovarian Cancer, University Hospital rechts der Isar, Technical University of Munich (TUM), Munich,
Germany
| | - Sabine Grill
- Department of Gynecology and Center for Hereditary Breast and Ovarian Cancer, University Hospital rechts der Isar, Technical University of Munich (TUM), Munich,
Germany
| |
Collapse
|
43
|
Gray GK, Li CMC, Rosenbluth JM, Selfors LM, Girnius N, Lin JR, Schackmann RCJ, Goh WL, Moore K, Shapiro HK, Mei S, D'Andrea K, Nathanson KL, Sorger PK, Santagata S, Regev A, Garber JE, Dillon DA, Brugge JS. A human breast atlas integrating single-cell proteomics and transcriptomics. Dev Cell 2022; 57:1400-1420.e7. [PMID: 35617956 DOI: 10.1016/j.devcel.2022.05.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/23/2022] [Accepted: 05/02/2022] [Indexed: 12/12/2022]
Abstract
The breast is a dynamic organ whose response to physiological and pathophysiological conditions alters its disease susceptibility, yet the specific effects of these clinical variables on cell state remain poorly annotated. We present a unified, high-resolution breast atlas by integrating single-cell RNA-seq, mass cytometry, and cyclic immunofluorescence, encompassing a myriad of states. We define cell subtypes within the alveolar, hormone-sensing, and basal epithelial lineages, delineating associations of several subtypes with cancer risk factors, including age, parity, and BRCA2 germline mutation. Of particular interest is a subset of alveolar cells termed basal-luminal (BL) cells, which exhibit poor transcriptional lineage fidelity, accumulate with age, and carry a gene signature associated with basal-like breast cancer. We further utilize a medium-depletion approach to identify molecular factors regulating cell-subtype proportion in organoids. Together, these data are a rich resource to elucidate diverse mammary cell states.
Collapse
Affiliation(s)
- G Kenneth Gray
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Carman Man-Chung Li
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Jennifer M Rosenbluth
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute (DFCI), Boston, MA 02115, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura M Selfors
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Nomeda Girnius
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA; The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA
| | - Jia-Ren Lin
- The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA
| | - Ron C J Schackmann
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Walter L Goh
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Kaitlin Moore
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Hana K Shapiro
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA
| | - Shaolin Mei
- The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA
| | - Kurt D'Andrea
- Department of Medicine, Division of Translation Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katherine L Nathanson
- Department of Medicine, Division of Translation Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter K Sorger
- The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA
| | - Sandro Santagata
- The Laboratory of Systems Pharmacology (LSP), HMS, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital (BWH), Boston, MA 02115, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute (DFCI), Boston, MA 02115, USA
| | - Deborah A Dillon
- Department of Pathology, Brigham and Women's Hospital (BWH), Boston, MA 02115, USA
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School (HMS), Boston, MA 02115, USA.
| |
Collapse
|
44
|
Park SS, Uzelac A, Kotsopoulos J. Delineating the role of osteoprotegerin as a marker of breast cancer risk among women with a BRCA1 mutation. Hered Cancer Clin Pract 2022; 20:14. [PMID: 35418083 PMCID: PMC9008947 DOI: 10.1186/s13053-022-00223-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/30/2022] [Indexed: 11/26/2022] Open
Abstract
Women with a pathogenic germline mutation in the BRCA1 gene face a very high lifetime risk of developing breast cancer, estimated at 72% by age 80. Prophylactic bilateral mastectomy is the only effective way to lower their risk; however, most women with a mutation opt for intensive screening with annual MRI and mammography. Given that the BRCA1 gene was identified over 20 years ago, there is a need to identify a novel non-surgical approach to hereditary breast cancer prevention. Here, we provide a review of the emerging preclinical and epidemiologic evidence implicating the dysregulation of progesterone-mediated receptor activator of nuclear factor κB (RANK) signaling in the pathogenesis of BRCA1-associated breast cancer. Experimental studies have demonstrated that RANK inhibition suppresses Brca1-mammary tumorigenesis, suggesting a potential target for prevention. Data from studies conducted among women with a BRCA1 mutation further support this pathway in BRCA1-associated breast cancer development. Progesterone-containing (but not estrogen-alone) hormone replacement therapy is associated with an increased risk of breast cancer in women with a BRCA1 mutation. Furthermore, BRCA1 mutation carriers have significantly lower levels of circulating osteoprotegerin (OPG), the decoy receptor for RANK-ligand (RANKL) and thus endogenous inhibitor of RANK signaling. OPG levels may be associated with the risk of disease, suggesting a role of this protein as a potential biomarker of breast cancer risk. This may improve upon current risk prediction models, stratifying women at the highest risk of developing the disease, and further identify those who may be targets for anti-RANKL chemoprevention. Collectively, the evidence supports therapeutic inhibition of the RANK pathway for the primary prevention of BRCA1-associated breast cancer, which may generate unique prevention strategies (without prophylactic surgery) and enhance quality of life.
Collapse
Affiliation(s)
- Sarah Sohyun Park
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
| | - Aleksandra Uzelac
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Joanne Kotsopoulos
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada.
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
45
|
Kim S, Hwang S. G-Quadruplex Matters in Tissue-Specific Tumorigenesis by BRCA1 Deficiency. Genes (Basel) 2022; 13:genes13030391. [PMID: 35327946 PMCID: PMC8948836 DOI: 10.3390/genes13030391] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/14/2022] Open
Abstract
How and why distinct genetic alterations, such as BRCA1 mutation, promote tumorigenesis in certain tissues, but not others, remain an important issue in cancer research. The underlying mechanisms may reveal tissue-specific therapeutic vulnerabilities. Although the roles of BRCA1, such as DNA damage repair and stalled fork stabilization, obviously contribute to tumor suppression, these ubiquitously important functions cannot explain tissue-specific tumorigenesis by BRCA1 mutations. Recent advances in our understanding of the cancer genome and fundamental cellular processes on DNA, such as transcription and DNA replication, have provided new insights regarding BRCA1-associated tumorigenesis, suggesting that G-quadruplex (G4) plays a critical role. In this review, we summarize the importance of G4 structures in mutagenesis of the cancer genome and cell type-specific gene regulation, and discuss a recently revealed molecular mechanism of G4/base excision repair (BER)-mediated transcriptional activation. The latter adequately explains the correlation between the accumulation of unresolved transcriptional regulatory G4s and multi-level genomic alterations observed in BRCA1-associated tumors. In summary, tissue-specific tumorigenesis by BRCA1 deficiency can be explained by cell type-specific levels of transcriptional regulatory G4s and the role of BRCA1 in resolving it. This mechanism would provide an integrated understanding of the initiation and development of BRCA1-associated tumors.
Collapse
Affiliation(s)
- Sanghyun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Sungnam 13488, Korea;
| | - Sohyun Hwang
- Department of Biomedical Science, College of Life Science, CHA University, Sungnam 13488, Korea;
- Department of Pathology, CHA Bundang Medical Center, CHA University School of Medicine, Sungnam 13496, Korea
- Correspondence:
| |
Collapse
|
46
|
Xia YY, Gronwald J, Karlan B, Lubinski J, McCuaig JM, Brooks J, Moller P, Eisen A, Sun S, Senter L, Bordeleau L, Neuhausen SL, Singer CF, Tung N, Foulkes WD, Sun P, Narod SA, Kotsopoulos J. Contraceptive use and the risk of ovarian cancer among women with a BRCA1 or BRCA2 mutation. Gynecol Oncol 2022; 164:514-521. [DOI: 10.1016/j.ygyno.2022.01.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 01/05/2023]
|
47
|
Yue Z, Niu X, Yuan Z, Qin Q, Jiang W, He L, Gao J, Ding Y, Liu Y, Xu Z, Li Z, Yang Z, Li R, Xue X, Gao Y, Yue F, Zhang XHF, Hu G, Wang Y, Li Y, Chen G, Siwko S, Gartland A, Wang N, Xiao J, Liu M, Luo J. RSPO2/RANKL-LGR4 signaling regulates osteoclastic pre-metastatic niche formation and bone metastasis. J Clin Invest 2021; 132:144579. [PMID: 34847079 PMCID: PMC8759794 DOI: 10.1172/jci144579] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
Therapeutics targeting osteoclasts are commonly used treatments for bone metastasis; however, whether and how osteoclasts regulate pre-metastatic niche and bone tropism is largely unknown. In this study, we report that osteoclast precursors (OPs) can function as a pre-metastatic niche component that facilitates breast cancer (BCa) bone metastasis at early stages. At the molecular level, unbiased GPCR ligand/agonist screening in BCa cells suggested that R-spondin 2 (RSPO2) and RANKL, through interacting with their receptor LGR4, promoted osteoclastic pre-metastatic niche formation and enhanced BCa bone metastasis. This was achieved by RSPO2/RANKL-LGR4 signal modulating WNT inhibitor DKK1 through Gαq and β-catenin signaling. DKK1 directly facilitated OP recruitment through suppressing its receptor low-density lipoprotein-related receptors 5 (LRP5) but not LRP6, upregulating Rnasek expression via inhibiting canonical WNT signaling. In clinical samples, RSPO2, LGR4 and DKK1 expression showed positive correlation with BCa bone metastasis. Furthermore, soluble LGR4 extracellular domain (ECD) protein, acting as a decoy receptor for RSPO2 and RANKL, significantly alleviated bone metastasis and osteolytic lesions in mouse bone metastasis model. These findings provide unique insights into the functional role of OPs as key components of pre-metastatic niche for BCa bone metastasis, indicate RSPO2/RANKL-LGR4 signaling as a promising target for inhibiting BCa bone metastasis.
Collapse
Affiliation(s)
- Zhiying Yue
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xin Niu
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Zengjin Yuan
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Qin Qin
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Wenhao Jiang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, China
| | - Liang He
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, China
| | - Jingduo Gao
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yi Ding
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yanxi Liu
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Ziwei Xu
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhenxi Li
- Department of Orthopedic Oncology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhengfeng Yang
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Rong Li
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xiwen Xue
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yankun Gao
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Fei Yue
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, United States of America
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, United States of America
| | - Guohong Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai, China
| | - Yi Wang
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Yi Li
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, United States of America
| | - Geng Chen
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Stefan Siwko
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, United States of America
| | - Alison Gartland
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, United Kingdom
| | - Ning Wang
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, United Kingdom
| | - Jianru Xiao
- Department of Orthopaedic Oncology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Mingyao Liu
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jian Luo
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
48
|
Ebrahimi N, Nasr Esfahani A, Samizade S, Mansouri A, Ghanaatian M, Adelian S, Shadman Manesh V, Hamblin MR. The potential application of organoids in breast cancer research and treatment. Hum Genet 2021; 141:193-208. [PMID: 34713317 DOI: 10.1007/s00439-021-02390-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 10/16/2021] [Indexed: 12/23/2022]
Abstract
Tumor heterogeneity is a major challenge for breast cancer researchers who have struggled to find effective treatments despite recent advances in oncology. Although the use of 2D cell culture methods in breast cancer research has been effective, it cannot model the heterogeneity of breast cancer as found within the body. The development of 3D culture of tumor cells and breast cancer organoids has provided a new approach in breast cancer research, allowing the identification of biomarkers, study of the interaction of tumor cells with the microenvironment, and for drug screening and discovery. In addition, the possibility of gene editing in organoids, especially using the CRISPR/Cas9 system, is convenient, and has allowed a more detailed study of tumor behavior in models closer to the physiological condition. The present review covers the application of organoids in breast cancer research. The recent use of gene-editing systems to provide insights into therapeutic approaches for breast cancer, is highlighted. The study of organoids and the possibility of gene manipulation may be a step towards the personalized treatment of breast cancer, which has so far remained unattainable due to the high heterogeneity of breast cancer.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Division of Genetics, Department of Cell, Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Islamic Republic of Iran
| | - Alireza Nasr Esfahani
- Department of Cellular and Molecular Biology, School of Biological Sciences, Islamic Azad University of Falavarjan, Falavarjan, Iran
| | - Setare Samizade
- Department of Cellular and Molecular Biology, School of Biological Sciences, Islamic Azad University of Falavarjan, Falavarjan, Iran
| | - Atena Mansouri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Masoud Ghanaatian
- Department of Microbiology, Islamic Azad University of Jahrom, Jahrom, Fars, Iran
| | - Samaneh Adelian
- Department of Genetics, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Vida Shadman Manesh
- Medical Engineering Tissue Engineering, Department of Medical Technologies, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Michael R Hamblin
- Faculty of Health Science, Laser Research Centre, University of Johannesburg, Doornfontein, Johannesburg, 2028, South Africa.
| |
Collapse
|
49
|
Clinicopathological features and BRCA1 and BRCA2 mutation status in a prospective cohort of young women with breast cancer. Br J Cancer 2021; 126:302-309. [PMID: 34703009 DOI: 10.1038/s41416-021-01597-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 10/02/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Breast cancer in young women is more likely to have higher risk features and be associated with germline BRCA1/BRCA2 mutations. We present the clinicopathologic features of breast cancers in a prospective cohort of young women, and associations between surrogate molecular subtype and BRCA1/BRCA2 mutation status. METHODS Histopathological features, biomarker status, tumour stage and BRCA status were collected. Invasive tumours were categorised as luminal A-like (ER + and/or PR + , HER2-, grade 1/2), luminal B-like (ER + and/or PR + , HER2 + , or ER + and/or PR + , HER2-, and grade 3), HER2-enriched (ER/PR-, HER2 + ) or triple-negative. RESULTS In all, 57.3% (654/1143) of invasive tumours were high grade. In total, 32.9% were luminal A-like, 42.4% luminal B-like, 8.3% HER2-enriched, and 16.4% triple-negative. Among different age groups, there were no differences in molecular phenotype, stage, grade or histopathology. 11% (131) of tumours were from BRCA mutation carriers; 64.1% BRCA1 (63.1% triple-negative), and 35.9% BRCA2 (55.3% luminal B-like). DISCUSSION The opportunity to provide comparisons across young age groups, BRCA mutation status, surrogate molecular phenotype, and the identification of more aggressive hormone receptor-positive phenotypes in this population provides direction for future work to further understand and improve disparate outcomes for young women with luminal B-like cancers, particularly BRCA2-associated cancers, with potential implications for tailored prevention and treatment.
Collapse
|
50
|
Kim H, Moon WK. Histological Findings of Mammary Gland Development and Risk of Breast Cancer in BRCA1 Mutant Mouse Models. J Breast Cancer 2021; 24:455-462. [PMID: 34652081 PMCID: PMC8561134 DOI: 10.4048/jbc.2021.24.e44] [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: 01/09/2021] [Revised: 05/30/2021] [Accepted: 09/27/2021] [Indexed: 11/30/2022] Open
Abstract
Purpose The breast cancer susceptibility gene, BRCA1, is involved in normal development and carcinogenesis of mammary glands. Here, we aimed to evaluate the relationship between histological findings of mammary gland development and breast cancer risk in BRCA1 mutant mice. Methods Five BRCA1 mutant mice and five non-mutant FVB/NJ mice were used for each group of 1-month-old (pubertal), 3-month-old (fertile), and 8-month-old (menopausal) mice. In another experiment, 15 BRCA1 mutant mice were followed up to 8 months after birth and classified into tumor-bearing (11 mice) and tumor-free (4 mice) groups. Excised mammary gland tissues were stained with Carmine Alum, and the number of terminal end buds (or alveolar buds), branching density, and duct elongation were measured using image analysis programs. Differences between the two groups were assessed using paired t-test. Results One-month-old BRCA1 mutant mice showed a higher number of terminal end buds (23.8 ± 1.0 vs. 15.6 ± 0.8, p = 0.0002), branching density (11.7 ± 0.4 vs. 9.6 ± 0.5%, p = 0.0082), and duct elongation (9.7 ± 0.7 vs. 7.3 ± 0.4 mm, p = 0.0186) than controls. However, there was no difference between the 3- and 8-month-old groups. In BRCA1 mutant mice, the tumor-bearing group showed a significantly higher number of alveolar buds (142.7 ± 5.5 vs. 105.5 ± 5.4, p = 0.0008) and branching density (30.0 ± 1.0 vs. 24.1 ± 1.1%, p = 0.008) than the tumor-free group; however, duct elongation was not different (23.9 ± 0.6 vs. 23.6 ± 0.6 mm, p = 0.8099) between the groups. Conclusion BRCA1 mutant mice exhibited early pubertal mammary gland development and delayed age-related mammary gland involution was associated with breast cancer. Our results may have clinical implications for predicting breast cancer risk and developing prevention strategies for BRCA1 mutation carriers.
Collapse
Affiliation(s)
- Hyelim Kim
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Woo Kyung Moon
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|