1
|
Fallone F, Rebeaud M, Bouche C, Fontaine J, Arellano C, Ducoux-Petit M, Orgerit L, Deudon R, Nicolle R, Franchet C, Estève D, Mouton-Barbosa E, Dauvillier S, Moutahir M, Burlet-Schiltz O, Bouloumié A, Vaysse C, Muller C. Lack of fibro-inflammatory response in human mammary adipose tissue in obesity. Int J Obes (Lond) 2024:10.1038/s41366-024-01705-1. [PMID: 39738492 DOI: 10.1038/s41366-024-01705-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Understanding how obesity impacts human mammary adipose tissue (MAT) biology is crucial for deciphering its role in mammary epithelium during both physiological and pathophysiological processes, including breast cancer. Hypertrophic mammary adipocytes and Crown-Like Structures are present in MAT of patients with obesity but whether these changes initiate a fibro-inflammatory response at the tissue level remains insufficiently explored. OBJECTIVE We investigated the markers of adipose tissue dysfunction (immune cell infiltration, secretion pattern and fibrosis) in tumor-free MAT of patients with obesity versus patients who are lean. METHODS Tumor-free MAT were obtained from 96 women with (n = 43) or without (n = 53) obesity who underwent mastectomy for breast cancer risk reduction or treatment. Immune and non-immune cell infiltration were determined using flow cytometry. Bulk transcriptomic was used to characterize the phenotype of CD206+ macrophages whose infiltration is increased in patients with obesity. Conditioned-medium were prepared from MAT to characterize their secretome and dose adipokines and cytokines by ELISA assay. The extra-cellular matrix (ECM) deposition was evaluated by Masson trichrome staining on cross-stained sections, 3D imaging of red picrosirius-stained tissues and measure of hydroxyproline content. RESULTS We observed an increase of CD206+/HLA-DR+ macrophages in the stromal vascular fraction of MAT from patients with obesity compared to patients who are lean. Other immune cell infiltration and endothelial or adipose progenitor cell numbers were similar between groups. Bulk transcriptomics on CD206+ macrophages revealed a significant decrease in ECM component expression and processing in obesity. In addition, no heightened secretion of pro-inflammatory cytokines, TGF-β1 or MCP-1 was observed in the samples from patients with obesity. ECM characterization revealed an absence of fibrosis, with MAT of patients with obesity showing even a slightly reduced collagen secretion and deposition compared with their lean counterparts. CONCLUSIONS Obesity is not associated with inflammation nor fibrosis in MAT, highlighting its unique behavior.
Collapse
Affiliation(s)
- Frédérique Fallone
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France.
| | - Marie Rebeaud
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
| | - Caroline Bouche
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Jessica Fontaine
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université de Toulouse, INSERM, UPS, Toulouse, France
| | - Carlo Arellano
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Manuelle Ducoux-Petit
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Infrastructure Nationale de Protéomique, ProFI, FR 2048, Toulouse, France
| | - Lucyle Orgerit
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Rémi Deudon
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Rémy Nicolle
- Université Paris Cité, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018, Paris, France
| | - Camille Franchet
- Département d'Anatomo-Pathologie, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - David Estève
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
| | - Emmanuelle Mouton-Barbosa
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Infrastructure Nationale de Protéomique, ProFI, FR 2048, Toulouse, France
| | - Stéphanie Dauvillier
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
| | - Mohamed Moutahir
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Infrastructure Nationale de Protéomique, ProFI, FR 2048, Toulouse, France
| | - Anne Bouloumié
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université de Toulouse, INSERM, UPS, Toulouse, France
| | - Charlotte Vaysse
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Catherine Muller
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France.
| |
Collapse
|
2
|
Tan L, Mei J, Tang R, Huang D, Qi K, Ossowski Z, Wang X. Can exercise as a complementary technique manage inflammatory markers in women with breast cancer who are overweight and obese? A systematic review and meta-analysis. Complement Ther Med 2024; 88:103119. [PMID: 39710346 DOI: 10.1016/j.ctim.2024.103119] [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: 09/24/2024] [Revised: 12/05/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Inflammation can result in the development of breast cancer in women with overweight and obese, and also affects the outcome and prognosis of breast cancer patients, thereby decreasing the cure and survival rates of breast cancer patients. Exercise may benefit breast cancer patients as a supplement to conventional treatments. However, research on the effects of exercise on inflammatory markers in women with breast cancer who are overweight and obese remains incomplete. OBJECTIVE A systematic review and meta-analysis were used to study the effects of exercise on inflammatory markers in women with breast cancer who are overweight and obese. METHOD Literature up to May 2024 was searched from databases such as Cochrane, Embase, Pubmed, Web of Science, and EBSCO, and English-language randomized controlled trials (RCTs) that met the inclusion criteria were screened. The screening criteria were as follows (A) written in English; (B) RCT; (C) studied in women with overweight obese and breast cancer; (D) outcome measures: inflammatory markers; (E) the duration of the exercise intervention was unlimited. RESULTS A total of 14 articles and 1064 participants were included. Exercise significantly reduced C-reactive protein (CRP) (MD: -0.52, 95 % CI: -0.94 to -0.11; p = 0.01; heterogeneity p < 0.1), interleukin-6 (IL-6) (MD: -0.87, 95 % CI: -1.62 to -0.11; p = 0.02; heterogeneity p < 0.1), and leptin (MD: -0.92, 95 % CI: -1.71 to -0.13; p = 0.02; heterogeneity p < 0.1) levels and exercise significantly increased adiponectin levels (MD: 0.89, 95 % CI: 0.03-1.75, p = 0.04; heterogeneity p < 0.1) but had no effect on tumor necrosis factor-α (TNF-α) (MD: -0.26, 95 % CI: -0.82-0.29; p = 0.35; heterogeneity p < 0.1) and IL-10 (MD: 0.14, 95 % CI: -0.17-0.45; p = 0.37; heterogeneity p = 0.45) were not significant. In addition, subgroup analyses suggest that combination training (CE) may be the most recommended type of exercise to decrease pro-inflammatory markers, and increase anti-inflammatory markers in women with overweight obesity, and have breast cancer. CONCLUSION Exercise significantly reduced CRP, IL-6, and leptin levels and overall increased adiponectin levels in women with overweight obese, and breast cancer. However, the effects on TNF-α and IL-10 levels were not significant. CE may be the most recommended type of exercise for reducing pro-inflammatory factors and increasing anti-inflammatory factors. Therefore, this study considers exercise as an effective complementary approach to managing inflammatory markers in women with breast cancer who are overweight and obese. Future researchers may consider exploring the combined effects of exercise and dietary control, weight loss, and other factors, and formulate a comprehensive treatment plan accordingly.
Collapse
Affiliation(s)
- Liang Tan
- Gdansk University of Physical Education and Sport, Gdansk 80-336, Poland.
| | - Jinyu Mei
- Department of Physical Education, Harbin Institute of Technology (Weihai), Weihai 264209, China.
| | - Ruihong Tang
- Education University of Hong Kong (EdUHK), Hongkong, 999077, China; Hunan First Normal University, Changsha 410002, China.
| | - Duo Huang
- Shangrao Normal University, Shangrao 334001, China.
| | - Kai Qi
- Gdansk University of Physical Education and Sport, Gdansk 80-336, Poland.
| | - Zbigniew Ossowski
- Gdansk University of Physical Education and Sport, Gdansk 80-336, Poland.
| | - Xiaoning Wang
- School of Physical Education, Shandong University, Jinan 250061, China.
| |
Collapse
|
3
|
Javed SR, Skolariki A, Zameer MZ, Lord SR. Implications of obesity and insulin resistance for the treatment of oestrogen receptor-positive breast cancer. Br J Cancer 2024; 131:1724-1736. [PMID: 39251829 PMCID: PMC11589622 DOI: 10.1038/s41416-024-02833-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 09/11/2024] Open
Abstract
Breast cancer is the most common cancer in women, and incidence rates are rising, it is thought in part, due to increasing levels of obesity. Endocrine therapy (ET) remains the cornerstone of systemic therapy for early and advanced oestrogen receptor-positive (ER + ) breast cancer, but despite treatment advances, it is becoming more evident that obesity and insulin resistance are associated with worse outcomes. Here, we describe the current understanding of the relationship between both obesity and diabetes and the prevalence and outcomes for ER+ breast cancer. We also discuss the mechanisms associated with resistance to ET and the relationship to treatment toxicity.
Collapse
Affiliation(s)
| | | | | | - Simon R Lord
- Department of Oncology, University of Oxford, Oxford, UK.
| |
Collapse
|
4
|
Krishnan VD, Kostev K, Kalder M. Is there an association between mastitis and breast cancer? a retrospective cohort study from Germany. Cancer Causes Control 2024; 35:1517-1523. [PMID: 39207599 PMCID: PMC11564347 DOI: 10.1007/s10552-024-01909-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE The aim of the study was to explore the association between mastitis and subsequent breast cancer. METHODS This retrospective cohort study included women aged ≥ 18 years with an initial mastitis diagnosis from 315 office-based gynecologists in Germany between January 2005 and December 2021. Women without mastitis were matched to women with mastitis using propensity score matching based on age, index year, average yearly consultation frequency during the follow-up period, and coexisting diseases such as obesity, benign mammary dysplasia, hypertrophy of the breast, unspecified lump of breast, and other disorders of the breast. The 10-year cumulative incidence of breast cancer for the mastitis-cohort and non-mastitis-cohort was studied with Kaplan-Meier curves using the log-rank test. The association between mastitis and breast cancer was studied separately for four age groups with univariable Cox regression analyses. RESULTS In the follow-up period of 7 months to 10 years after the index date, 2.9% of mastitis patients and 2.4% of matched non-mastitis patients were diagnosed with breast cancer. A Cox regression analysis revealed a significant association between mastitis and subsequent breast cancer (HR: 1.37; 95% CI: 1.11-1.70). According to the age-stratified analyses, a strong and significant association was only observed in the age group > 50 years (HR: 1.73; 95% 1.25-2.40). CONCLUSION The findings of our retrospective cohort study support an association between mastitis and subsequent breast cancer diagnoses in women aged > 50 years. The pathophysiological basis and possibility of confounders however requires further investigation.
Collapse
Affiliation(s)
- Vedanth D Krishnan
- Department of Gynecology and Obstetrics, University Hospital Marburg, Philipps-University, Marburg, Germany
| | - Karel Kostev
- Department of Gynecology and Obstetrics, University Hospital Marburg, Philipps-University, Marburg, Germany.
- IQVIA, Frankfurt, Germany.
| | - Matthias Kalder
- Department of Gynecology and Obstetrics, University Hospital Marburg, Philipps-University, Marburg, Germany
| |
Collapse
|
5
|
Kruglikov IL, Scherer PE. Is the endotoxin-complement cascade the major driver in lipedema? Trends Endocrinol Metab 2024; 35:769-780. [PMID: 38688780 PMCID: PMC11387139 DOI: 10.1016/j.tem.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024]
Abstract
Lipedema is a poorly understood disorder of adipose tissue characterized by abnormal but symmetrical deposition of subcutaneous white adipose tissue (WAT) in proximal extremities. Here, we propose that the underlying cause for lipedema could be triggered by a selective accumulation of bacterial lipopolysaccharides (LPS; also known as endotoxin) in gluteofemoral WAT. Together with a malfunctioning complement system, this induces low-grade inflammation in the depot and raises its uncontrollable expansion. Correspondingly, more attention should be paid in future research to the endotoxemia prevalent in patients with lipedema. We would like to propose that proper management of endotoxemia can reduce the progression and even improve the state of disease in patients with lipedema.
Collapse
Affiliation(s)
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA.
| |
Collapse
|
6
|
Gogg S, Nerstedt A, Smith U, Hansson E. Breast volume in non-obese females is related to breast adipose cell hypertrophy, inflammation, and COX2 expression. J Plast Surg Hand Surg 2024; 59:83-88. [PMID: 38967364 DOI: 10.2340/jphs.v59.40754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/13/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Breast hypertrophy seems to be a risk factor for breast cancer and the amount and characteristics of breast adipose tissue may play important roles. The main aim of this study was to investigate associations between breast volume in normal weight women and hypertrophic adipose tissue and inflammation. METHODS Fifteen non-obese women undergoing breast reduction surgery were examined. Breast volume was measured with plastic cups and surgery was indicated if the breast was 800 ml or larger according to Swedish guidelines. We isolated adipose cells from the breasts and ambient subcutaneous tissue to measure cell size, cell inflammation and other known markers of risk of developing breast cancer including COX2 gene activation and MAPK, a cell proliferation regulator. RESULTS Breast adipose cell size was characterized by cell hypertrophy and closely related to breast volume. The breast adipose cells were also characterized by being pro-inflammatory with increased IL-6, IL-8, IL-1β, CCL-2, TNF-a and an increased marker of cell senescence GLB1/β-galactosidase, commonly increased in hypertrophic adipose tissue. The prostaglandin synthetic marker COX2 was also increased in the hypertrophic cells and COX2 has previously been shown to be an important marker of risk of developing breast cancer. Interestingly, the phosphorylation of the proliferation marker MAPK was also increased in the hypertrophic adipose cells. CONCLUSION Taken together, these findings show that increased breast volume in non-obese women is associated with adipose cell hypertrophy and dysfunction and characterized by increased inflammation and other markers of increased risk for developing breast cancer. TRIAL REGISTRATION Projektdatabasen FoU i VGR, project number: 249191 (https://www.researchweb.org/is/vgr/project/249191).
Collapse
Affiliation(s)
- Silvia Gogg
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Annika Nerstedt
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf Smith
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Institute of Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Emma Hansson
- Department of Plastic Surgery, Institute of Clinical Sciences, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Plastic Surgery, Gothenburg, Sweden.
| |
Collapse
|
7
|
Rebeaud M, Lacombe M, Fallone F, Milhas D, Roumiguié M, Vaysse C, Attané C, Muller C. Specificities of mammary and periprostatic adipose tissues: A perspective from cancer research. ANNALES D'ENDOCRINOLOGIE 2024; 85:220-225. [PMID: 38871505 DOI: 10.1016/j.ando.2024.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
In addition to the major subcutaneous and visceral adipose tissues (AT), other adipose depots are dispersed throughout the body and are found in close interaction with proximal organs such as mammary and periprostatic AT (MAT and PPAT respectively). These ATs have an effect on proximal organ function during physiological processes and diseases such as cancer. We highlighted here some of their most distinctive features in terms of tissular organization and responses to external stimuli and discussed how obesity affects them based on our current knowledge.
Collapse
Affiliation(s)
- Marie Rebeaud
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France
| | - Mathilde Lacombe
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France
| | - Frédérique Fallone
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France
| | - Delphine Milhas
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France
| | - Mathieu Roumiguié
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France; Département d'urologie, CHU de Toulouse, 1, avenue du Professeur-Jean-Poulhès, 31400 Toulouse, France
| | - Charlotte Vaysse
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France; Département de chirurgie gynécologique-oncologique, institut universitaire du cancer de Toulouse-Oncopole, CHU de Toulouse, 1, avenue Irène-Joliot-Curie, 31059 Toulouse cedex 9, France
| | - Camille Attané
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France
| | - Catherine Muller
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France.
| |
Collapse
|
8
|
Anazco D, Acosta A, Cathcart-Rake EJ, D'Andre SD, Hurtado MD. Weight-centric prevention of cancer. OBESITY PILLARS 2024; 10:100106. [PMID: 38495815 PMCID: PMC10943063 DOI: 10.1016/j.obpill.2024.100106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
Background The link between excess adiposity and carcinogenesis has been well established for multiple malignancies, and cancer is one of the main contributors to obesity-related mortality. The potential role of different weight-loss interventions on cancer risk modification has been assessed, however, its clinical implications remain to be determined. In this clinical review, we present the data assessing the effect of weight loss interventions on cancer risk. Methods In this clinical review, we conducted a comprehensive search of relevant literature using MEDLINE, Embase, Web of Science, and Google Scholar databases for relevant studies from inception to January 20, 2024. In this clinical review, we present systematic reviews and meta-analysis, randomized clinical trials, and prospective and retrospective observational studies that address the effect of different treatment modalities for obesity in cancer risk. In addition, we incorporate the opinions from experts in the field of obesity medicine and oncology regarding the potential of weight loss as a preventative intervention for cancer. Results Intentional weight loss achieved through different modalities has been associated with a reduced cancer incidence. To date, the effect of weight loss on the postmenopausal women population has been more widely studied, with multiple reports indicating a protective effect of weight loss on hormone-dependent malignancies. The effect of bariatric interventions as a protective intervention for cancer has been studied extensively, showing a significant reduction in cancer incidence and mortality, however, data for the effect of bariatric surgery on certain specific types of cancer is conflicting or limited. Conclusion Medical nutrition therapy, exercise, antiobesity medication, and bariatric interventions, might lead to a reduction in cancer risk through weight loss-dependent and independent factors. Further evidence is needed to better determine which population might benefit the most, and the amount of weight loss required to provide a clinically significant preventative effect.
Collapse
Affiliation(s)
- Diego Anazco
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Maria D. Hurtado
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
9
|
Forma A, Grunwald A, Zembala P, Januszewski J, Brachet A, Zembala R, Świątek K, Baj J. Micronutrient Status and Breast Cancer: A Narrative Review. Int J Mol Sci 2024; 25:4968. [PMID: 38732186 PMCID: PMC11084730 DOI: 10.3390/ijms25094968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Breast cancer is one of the most common cancers worldwide, at the same time being one of the most prevalent causes of women's death. Many factors such as alcohol, weight fluctuations, or hormonal replacement therapy can potentially contribute to breast cancer development and progression. Another important factor in breast cancer onset includes micronutrient status. In this narrative review, we analyzed 23 micronutrients and their possible influence on breast cancer onset and progression. Further, the aim of this study was to investigate the impact of micronutrient status on the prevention of breast cancer and its possible influence on various therapeutic pathways. We researched meta-analyses, systemic and narrative reviews, retrospective studies, as well as original studies on human and animal models. The results of these studies indicate a possible correlation between the different levels of micronutrients and a decreased risk of breast cancer as well as a better survival rate. However, further studies are necessary to establish adequate doses of supplementation of the chosen micronutrients and the exact mechanisms of micronutrient impact on breast cancer therapy.
Collapse
Affiliation(s)
- Alicja Forma
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (A.G.); (A.B.)
| | - Arkadiusz Grunwald
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (A.G.); (A.B.)
| | - Patryk Zembala
- Faculty of Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland;
| | - Jacek Januszewski
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (J.J.); (K.Ś.); (J.B.)
| | - Adam Brachet
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (A.G.); (A.B.)
| | - Roksana Zembala
- Faculty of Medicine, Cardinal Stefan Wyszynski University in Warsaw, Wóycickiego 1/3, 01-938 Warsaw, Poland;
| | - Kamila Świątek
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (J.J.); (K.Ś.); (J.B.)
| | - Jacek Baj
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (J.J.); (K.Ś.); (J.B.)
| |
Collapse
|
10
|
Lim PW, Stucky CCH, Wasif N, Etzioni DA, Harold KL, Madura JA, Ven Fong Z. Bariatric Surgery and Longitudinal Cancer Risk: A Review. JAMA Surg 2024; 159:331-338. [PMID: 38294801 DOI: 10.1001/jamasurg.2023.5809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Importance Cancer is one of the leading causes of death in the United States, with the obesity epidemic contributing to its steady increase every year. Recent cohort studies find an association between bariatric surgery and reduced longitudinal cancer risk, but with heterogeneous findings. Observations This review summarizes how obesity leads to an increased risk of developing cancer and synthesizes current evidence behind the potential for bariatric surgery to reduce longitudinal cancer risk. Overall, bariatric surgery appears to have the strongest and most consistent association with decreased incidence of developing breast, ovarian, and endometrial cancers. The association of bariatric surgery and the development of esophageal, gastric, liver, and pancreas cancer is heterogenous with studies showing either no association or decreased longitudinal incidences. Conversely, there have been preclinical and cohort studies implying an increased risk of developing colon and rectal cancer after bariatric surgery. A review and synthesis of the existing literature reveals epidemiologic shortcomings of cohort studies that potentially explain incongruencies observed between studies. Conclusions and Relevance Studies examining the association of bariatric surgery and longitudinal cancer risk remain heterogeneous and could be explained by certain epidemiologic considerations. This review provides a framework to better define subgroups of patients at higher risk of developing cancer who would potentially benefit more from bariatric surgery, as well as subgroups where more caution should be exercised.
Collapse
Affiliation(s)
- Pei-Wen Lim
- Division of General Surgery, Department of Surgery, Mayo Clinic Arizona, Phoenix
| | - Chee-Chee H Stucky
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, Mayo Clinic Arizona, Phoenix
| | - Nabil Wasif
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, Mayo Clinic Arizona, Phoenix
| | - David A Etzioni
- Division of General Surgery, Department of Surgery, Mayo Clinic Arizona, Phoenix
| | - Kristi L Harold
- Division of General Surgery, Department of Surgery, Mayo Clinic Arizona, Phoenix
| | - James A Madura
- Division of General Surgery, Department of Surgery, Mayo Clinic Arizona, Phoenix
| | - Zhi Ven Fong
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, Mayo Clinic Arizona, Phoenix
| |
Collapse
|
11
|
Zwager MC, Holt-Kedde I, Timmer-Bosscha H, de Bock GH, Werker PMN, Schröder CP, van der Vegt B, Arjaans M. Presence of crown-like structures in breast adipose tissue; differences between healthy controls, BRCA1/2 gene mutation carriers and breast cancer patients. Breast Cancer Res Treat 2024; 204:27-37. [PMID: 38057686 DOI: 10.1007/s10549-023-07169-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023]
Abstract
PURPOSE Crown-like structures (CLS) in breast adipose tissue are associated with inflammation and a potential factor in breast cancer behaviour. Whether this effect varies between breast cancer subtypes and is influenced by BMI and BRCA mutation status is presently unknown. Therefore, we compared CLS presence between adipose tissue of healthy controls, BRCA1/2 gene mutation carriers and breast cancer patients, and assessed the relation of CLS with clinical outcome in breast cancer patients. METHODS Immunohistochemical staining for CD68 was performed on breast adipose tissue sections of 48 healthy controls, 78 BRCA1/2 gene mutation carriers and 259 breast cancer patients. CLS presence and index (CLS/cm2) were correlated with BMI, BRCA status, tumour presence, intrinsic tumour subtype and tumour characteristics. Associations with clinical outcome were assessed. RESULTS CLS were more often present in breast cancer patients compared to BRCA carriers and healthy controls. CLS presence was associated with the presence of breast cancer and high BMI. CLS were more often present in Luminal-B-like tumours compared to the other subtypes. No correlations between CLS and BRCA status or age was found. In TNBC, CLS were related to lymphovascular invasion. No association with survival was found. CONCLUSION In conclusion, CLS were more frequently present in breast adipose tissue of breast cancer patients compared to BRCA1/2 gene mutation carriers and healthy controls. Furthermore, our study provides evidence of the association between obesity and presence of CLS. The prognostic significance and impact on clinical outcome of differences in CLS numbers should be further assessed in prospective studies.
Collapse
Affiliation(s)
- Mieke C Zwager
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Iris Holt-Kedde
- Department of Plastic Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hetty Timmer-Bosscha
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Geertruida H de Bock
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Paul M N Werker
- Department of Plastic Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Carolien P Schröder
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marlous Arjaans
- Department of Plastic Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Plastic Surgery, OLVG Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Shea AA, Heffron CL, Grieco JP, Roberts PC, Schmelz EM. Obesity modulates the cellular and molecular microenvironment in the peritoneal cavity: implication for ovarian cancer risk. Front Immunol 2024; 14:1323399. [PMID: 38264656 PMCID: PMC10803595 DOI: 10.3389/fimmu.2023.1323399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/12/2023] [Indexed: 01/25/2024] Open
Abstract
Introduction Abdominal obesity increases the risk of developing ovarian cancer but the molecular mechanisms of how obesity supports ovarian cancer development remain unknown. Here we investigated the impact of obesity on the immune cell and gene expression profiles of distinct abdominal tissues, focusing on the peritoneal serous fluid (PSF) and the omental fat band (OFB) as critical determinants for the dissemination of ovarian metastases and early metastatic events within the peritoneal cavity. Methods Female C57BL/6 mice were fed a low-fat (LFD) or a high-fat diet (HFD) for 12 weeks until the body weights in the HFD group were significantly higher and the mice displayed an impaired glucose tolerance. Then the mice were injected with the murine ovarian cancer cells (MOSE-LTICv) while remaining on their diets. After 21 days, the mice were sacrificed, tumor burden was evaluated and tissues were harvested. The immune cell composition of abdominal tissues and changes in gene expression in the PSF and OFB were evaluated by flow cytometry and qPCR RT2-profiler PCR arrays and confirmed by qRT-PCR, respectively. Other peritoneal adipose tissues including parametrial and retroperitoneal white adipose tissues as well as blood were also investigated. Results While limited effects were observed in the other peritoneal adipose tissues, feeding mice the HFD led to distinct changes in the immune cell composition in the PSF and the OFB: a depletion of B cells but an increase in myeloid-derived suppressor cells (MDSC) and mono/granulocytes, generating pro-inflammatory environments with increased expression of cyto- and chemokines, and genes supporting adhesion, survival, and growth, as well as suppression of apoptosis. This was associated with a higher peritoneal tumor burden compared to mice fed a LFD. Changes in cellular and genetic profiles were often exacerbated by the HFD. There was a large overlap in genes that were modulated by both the HFD and the cancer cells, suggesting that this 'genetic fingerprint' is important for ovarian metastases to the OFB. Discussion In accordance with the 'seed and soil' theory, our studies show that obesity contributes to the generation of a pro-inflammatory peritoneal environment that supports the survival of disseminating ovarian cancer cells in the PSF and the OFB and enhances the early metastatic adhesion events in the OFB through an increase in extracellular matrix proteins and modulators such as fibronectin 1 and collagen I expression as well as in genes supporting growth and invasion such as Tenacin C. The identified genes could potentially be used as targets for prevention strategies to lower the ovarian cancer risk in women with obesity.
Collapse
Affiliation(s)
- Amanda A. Shea
- Department of Human Nutrition, Foods and Exercise, Virginia Tech, Blacksburg, VA, United States
| | - Connie Lynn Heffron
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, United States
| | - Joseph P. Grieco
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, United States
| | - Paul C. Roberts
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, United States
| | - Eva M. Schmelz
- Department of Human Nutrition, Foods and Exercise, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
13
|
Engin AB, Engin A. Next-Cell Hypothesis: Mechanism of Obesity-Associated Carcinogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:727-766. [PMID: 39287871 DOI: 10.1007/978-3-031-63657-8_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Higher body fat content is related to a higher risk of mortality, and obesity-related cancer represents approximately 40% of all cancer patients diagnosed each year. Furthermore, epigenetic mechanisms are involved in cellular metabolic memory and can determine one's predisposition to being overweight. Low-grade chronic inflammation, a well-established characteristic of obesity, is a central component of tumor development and progression. Cancer-associated adipocytes (CAA), which enhance inflammation- and metastasis-related gene sets within the cancer microenvironment, have pro-tumoral effects. Adipose tissue is a major source of the exosomal micro ribonucleic acids (miRNAs), which modulate pathways involved in the development of obesity and obesity-related comorbidities. Owing to their composition of cargo, exosomes can activate receptors at the target cell or transfer molecules to the target cells and thereby change the phenotype of these cells. Exosomes that are released into the extracellular environment are internalized with their cargo by neighboring cells. The tumor-secreted exosomes promote organ-specific metastasis of tumor cells that normally lack the capacity to metastasize to a specific organ. Therefore, the communication between neighboring cells via exosomes is defined as the "next-cell hypothesis." The reciprocal interaction between the adipocyte and tumor cell is realized through the adipocyte-derived exosomal miRNAs and tumor cell-derived oncogenic miRNAs. The cargo molecules of adipocyte-derived exosomes are important messengers for intercellular communication involved in metabolic responses and have very specific signatures that direct the metabolic activity of target cells. RNA-induced silencing regulates gene expression through various mechanisms. Destabilization of DICER enzyme, which catalyzes the conversion of primary miRNA (pri-miRNA) to precursor miRNA (pre-miRNA), is an important checkpoint in cancer development and progression. Interestingly, adipose tissue in obesity and tumors share similar pathogenic features, and the local hypoxia progress in both. While hypoxia in obesity leads to the adipocyte dysfunction and metabolic abnormalities, in obesity-related cancer cases, it is associated with worsened prognosis, increased metastatic potential, and resistance to chemotherapy. Notch-interleukin-1 (IL-1)-Leptin crosstalk outcome is referred to as "NILCO effect." In this chapter, obesity-related cancer development is discussed in the context of "next-cell hypothesis," miRNA biogenesis, and "NILCO effect."
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
14
|
Engin A. Obesity-Associated Breast Cancer: Analysis of Risk Factors and Current Clinical Evaluation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:767-819. [PMID: 39287872 DOI: 10.1007/978-3-031-63657-8_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Several studies show that a significantly stronger association is obvious between increased body mass index (BMI) and higher breast cancer incidence. Additionally, obese and postmenopausal women are at higher risk of all-cause and breast cancer-specific mortality compared with non-obese women with breast cancer. In this context, increased levels of estrogens, excessive aromatization activity of the adipose tissue, overexpression of pro-inflammatory cytokines, insulin resistance, adipocyte-derived adipokines, hypercholesterolemia, and excessive oxidative stress contribute to the development of breast cancer in obese women. Genetic evaluation is an integral part of diagnosis and treatment for patients with breast cancer. Despite trimodality therapy, the four-year cumulative incidence of regional recurrence is significantly higher. Axillary lymph nodes as well as primary lesions have diagnostic, prognostic, and therapeutic significance for the management of breast cancer. In clinical setting, because of the obese population primary lesions and enlarged lymph nodes could be less palpable, the diagnosis may be challenging due to misinterpretation of physical findings. Thereby, a nomogram has been created as the "Breast Imaging Reporting and Data System" (BI-RADS) to increase agreement and decision-making consistency between mammography and ultrasonography (USG) experts. Additionally, the "breast density classification system," "artificial intelligence risk scores," ligand-targeted receptor probes," "digital breast tomosynthesis," "diffusion-weighted imaging," "18F-fluoro-2-deoxy-D-glucose positron emission tomography," and "dynamic contrast-enhanced magnetic resonance imaging (MRI)" are important techniques for the earlier detection of breast cancers and to reduce false-positive results. A high concordance between estrogen receptor (ER) and progesterone receptor (PR) status evaluated in preoperative percutaneous core needle biopsy and surgical specimens is demonstrated. Breast cancer surgery has become increasingly conservative; however, mastectomy may be combined with any axillary procedures, such as sentinel lymph node biopsy (SLNB) and/or axillary lymph node dissection whenever is required. As a rule, SLNB-guided axillary dissection in breast cancer patients who have clinically axillary lymph node-positive to node-negative conversion following neoadjuvant chemotherapy is recommended, because lymphedema is the most debilitating complication after any axillary surgery. There is no clear consensus on the optimal treatment of occult breast cancer, which is much discussed today. Similarly, the current trend in metastatic breast cancer is that the main palliative treatment option is systemic therapy.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
15
|
Pérez-Stuardo D, Frazão M, Ibaceta V, Brianson B, Sánchez E, Rivas-Pardo JA, Vallejos-Vidal E, Reyes-López FE, Toro-Ascuy D, Vidal EA, Reyes-Cerpa S. KLF17 is an important regulatory component of the transcriptomic response of Atlantic salmon macrophages to Piscirickettsia salmonis infection. Front Immunol 2023; 14:1264599. [PMID: 38162669 PMCID: PMC10755876 DOI: 10.3389/fimmu.2023.1264599] [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: 07/21/2023] [Accepted: 11/07/2023] [Indexed: 01/03/2024] Open
Abstract
Piscirickettsia salmonis is the most important health problem facing Chilean Aquaculture. Previous reports suggest that P. salmonis can survive in salmonid macrophages by interfering with the host immune response. However, the relevant aspects of the molecular pathogenesis of P. salmonis have been poorly characterized. In this work, we evaluated the transcriptomic changes in macrophage-like cell line SHK-1 infected with P. salmonis at 24- and 48-hours post-infection (hpi) and generated network models of the macrophage response to the infection using co-expression analysis and regulatory transcription factor-target gene information. Transcriptomic analysis showed that 635 genes were differentially expressed after 24- and/or 48-hpi. The pattern of expression of these genes was analyzed by weighted co-expression network analysis (WGCNA), which classified genes into 4 modules of expression, comprising early responses to the bacterium. Induced genes included genes involved in metabolism and cell differentiation, intracellular transportation, and cytoskeleton reorganization, while repressed genes included genes involved in extracellular matrix organization and RNA metabolism. To understand how these expression changes are orchestrated and to pinpoint relevant transcription factors (TFs) controlling the response, we established a curated database of TF-target gene regulatory interactions in Salmo salar, SalSaDB. Using this resource, together with co-expression module data, we generated infection context-specific networks that were analyzed to determine highly connected TF nodes. We found that the most connected TF of the 24- and 48-hpi response networks is KLF17, an ortholog of the KLF4 TF involved in the polarization of macrophages to an M2-phenotype in mammals. Interestingly, while KLF17 is induced by P. salmonis infection, other TFs, such as NOTCH3 and NFATC1, whose orthologs in mammals are related to M1-like macrophages, are repressed. In sum, our results suggest the induction of early regulatory events associated with an M2-like phenotype of macrophages that drives effectors related to the lysosome, RNA metabolism, cytoskeleton organization, and extracellular matrix remodeling. Moreover, the M1-like response seems delayed in generating an effective response, suggesting a polarization towards M2-like macrophages that allows the survival of P. salmonis. This work also contributes to SalSaDB, a curated database of TF-target gene interactions that is freely available for the Atlantic salmon community.
Collapse
Affiliation(s)
- Diego Pérez-Stuardo
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- Programa de Doctorado en Genómica Integrativa, Vicerrectoría de Investigación, Universidad Mayor, Santiago, Chile
| | - Mateus Frazão
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| | - Valentina Ibaceta
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| | - Bernardo Brianson
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| | - Evelyn Sánchez
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- Programa de Doctorado en Genómica Integrativa, Vicerrectoría de Investigación, Universidad Mayor, Santiago, Chile
- Agencia Nacional de Investigación y Desarrollo (ANID) Millennium Science Initiative Program-Millennium Institute for Integrative Biology (iBio), Santiago, Chile
| | - J. Andrés Rivas-Pardo
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| | - Eva Vallejos-Vidal
- Núcleo de Investigaciones Aplicadas en Ciencias Veterinarias y Agronómicas, Facultad de Medicina Veterinaria y Agronomía, Universidad De Las Américas, La Florida, Santiago, Chile
- Centro de Biotecnología Acuícola, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Centro de Nanociencia y Nanotecnología (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Felipe E. Reyes-López
- Centro de Biotecnología Acuícola, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Daniela Toro-Ascuy
- Laboratorio de Virología, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Elena A. Vidal
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- Agencia Nacional de Investigación y Desarrollo (ANID) Millennium Science Initiative Program-Millennium Institute for Integrative Biology (iBio), Santiago, Chile
| | - Sebastián Reyes-Cerpa
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| |
Collapse
|
16
|
Pu Q, Gao H. The Role of the Tumor Microenvironment in Triple-Positive Breast Cancer Progression and Therapeutic Resistance. Cancers (Basel) 2023; 15:5493. [PMID: 38001753 PMCID: PMC10670777 DOI: 10.3390/cancers15225493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/26/2023] [Accepted: 11/18/2023] [Indexed: 11/26/2023] Open
Abstract
Breast cancer (BRCA) is a highly heterogeneous systemic disease. It is ranked first globally in the incidence of new cancer cases and has emerged as the primary cause of cancer-related death among females. Among the distinct subtypes of BRCA, triple-positive breast cancer (TPBC) has been associated with increased metastasis and invasiveness, exhibiting greater resistance to endocrine therapy involving trastuzumab. It is now understood that invasion, metastasis, and treatment resistance associated with BRCA progression are not exclusively due to breast tumor cells but are from the intricate interplay between BRCA and its tumor microenvironment (TME). Accordingly, understanding the pathogenesis and evolution of the TPBC microenvironment demands a comprehensive approach. Moreover, addressing BRCA treatment necessitates a holistic consideration of the TME, bearing significant implications for identifying novel targets for anticancer interventions. This review expounds on the relationship between critical cellular components and factors in the TPBC microenvironment and the inception, advancement, and therapeutic resistance of breast cancer to provide perspectives on the latest research on TPBC.
Collapse
Affiliation(s)
- Qian Pu
- Department of Breast Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China;
- Oncology Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| | - Haidong Gao
- Department of Breast Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China;
- Oncology Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| |
Collapse
|
17
|
Nahmias-Blank D, Maimon O, Meirovitz A, Sheva K, Peretz-Yablonski T, Elkin M. Excess body weight and postmenopausal breast cancer: Emerging molecular mechanisms and perspectives. Semin Cancer Biol 2023; 96:26-35. [PMID: 37739109 DOI: 10.1016/j.semcancer.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
Postmenopausal, obese women have a significantly higher risk of developing estrogen receptor-positive (ER+) breast tumors, that are resistant to therapies and are associated with higher recurrence and death rates. The global prevalence of overweight/obese women has reached alarming proportions and with postmenopausal ER+ breast carcinoma (BC) having the highest incidence among the three obesity-related cancers in females (i.e., breast, endometrial and ovarian), this is of significant concern. Elucidation of the precise molecular mechanisms underlying the pro-cancerous action of obesity in ER+BC is therefore critical for disease prevention and novel treatment initiatives. Interestingly, accumulating data has shown opposing relationships between obesity and cancer in either pre- or post-menopausal women. Excess body weight is associated with an increased risk of breast cancer in postmenopausal women and a decreased risk in pre-menopausal women. Moreover, excess adiposity during early life appears to be protective against postmenopausal breast cancer, including both ER+ and ER negative BC subtypes. Overall, estrogen-dependent mechanisms have been implicated as the main driving force in obesity-related breast tumorigenesis. In the present review we discuss the epidemiologic and mechanistic aspects of association between obesity and breast tumors after menopause, mainly in the context of hormone dependency. Molecular and cellular events underlying this association present as potential avenues for both therapeutic intervention as well as the prevention of BC-promoting processes linked to excess adiposity, which is proving to be vital in an increasingly obese global population.
Collapse
Affiliation(s)
- Daniela Nahmias-Blank
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ofra Maimon
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amichay Meirovitz
- Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Be'er Sheva 84101, Israel
| | - Kim Sheva
- Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Be'er Sheva 84101, Israel
| | - Tamar Peretz-Yablonski
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; Hebrew University Medical School, Jerusalem 91120, Israel
| | - Michael Elkin
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; Hebrew University Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
18
|
Abubakar M, Klein A, Fan S, Lawrence S, Mutreja K, Henry JE, Pfeiffer RM, Duggan MA, Gierach GL. Host, reproductive, and lifestyle factors in relation to quantitative histologic metrics of the normal breast. Breast Cancer Res 2023; 25:97. [PMID: 37582731 PMCID: PMC10426057 DOI: 10.1186/s13058-023-01692-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/29/2023] [Indexed: 08/17/2023] Open
Abstract
BACKGROUND Emerging data indicate that variations in quantitative epithelial and stromal tissue composition and their relative abundance in benign breast biopsies independently impact risk of future invasive breast cancer. To gain further insights into breast cancer etiopathogenesis, we investigated associations between epidemiological factors and quantitative tissue composition metrics of the normal breast. METHODS The study participants were 4108 healthy women ages 18-75 years who voluntarily donated breast tissue to the US-based Susan G. Komen Tissue Bank (KTB; 2008-2019). Using high-accuracy machine learning algorithms, we quantified the percentage of epithelial, stromal, adipose, and fibroglandular tissue, as well as the proportion of fibroglandular tissue that is epithelium relative to stroma (i.e., epithelium-to-stroma proportion, ESP) on digitized hematoxylin and eosin (H&E)-stained normal breast biopsy specimens. Data on epidemiological factors were obtained from participants using a detailed questionnaire administered at the time of tissue donation. Associations between epidemiological factors and square root transformed tissue metrics were investigated using multivariable linear regression models. RESULTS With increasing age, the amount of stromal, epithelial, and fibroglandular tissue declined and adipose tissue increased, while that of ESP demonstrated a bimodal pattern. Several epidemiological factors were associated with individual tissue composition metrics, impacting ESP as a result. Compared with premenopausal women, postmenopausal women had lower ESP [β (95% Confidence Interval (CI)) = -0.28 (- 0.43, - 0.13); P < 0.001] with ESP peaks at 30-40 years and 60-70 years among pre- and postmenopausal women, respectively. Pregnancy [β (95%CI) vs nulligravid = 0.19 (0.08, 0.30); P < 0.001] and increasing number of live births (P-trend < 0.001) were positively associated with ESP, while breastfeeding was inversely associated with ESP [β (95%CI) vs no breastfeeding = -0.15 (- 0.29, - 0.01); P = 0.036]. A positive family history of breast cancer (FHBC) [β (95%CI) vs no FHBC = 0.14 (0.02-0.26); P = 0.02], being overweight or obese [β (95%CI) vs normal weight = 0.18 (0.06-0.30); P = 0.004 and 0.32 (0.21-0.44); P < 0.001, respectively], and Black race [β (95%CI) vs White = 0.12 (- 0.005, 0.25); P = 0.06] were positively associated with ESP. CONCLUSION Our findings revealed that cumulative exposure to etiological factors over the lifespan impacts normal breast tissue composition metrics, individually or jointly, to alter their dynamic equilibrium, with potential implications for breast cancer susceptibility and tumor etiologic heterogeneity.
Collapse
Affiliation(s)
- Mustapha Abubakar
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Shady Grove, Bethesda, MD, 20850, USA.
| | - Alyssa Klein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Shady Grove, Bethesda, MD, 20850, USA
| | - Shaoqi Fan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Shady Grove, Bethesda, MD, 20850, USA
| | - Scott Lawrence
- Molecular and Digital Pathology Laboratory, Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Karun Mutreja
- Molecular and Digital Pathology Laboratory, Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick, MD, 21702, USA
| | - Jill E Henry
- Biospecimen Collection and Banking Core, Susan G. Komen Tissue Bank at the IU Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Shady Grove, Bethesda, MD, 20850, USA
| | - Maire A Duggan
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, T2N2Y9, Canada
| | - Gretchen L Gierach
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Shady Grove, Bethesda, MD, 20850, USA
| |
Collapse
|
19
|
Kuziel G, Moore BN, Arendt LM. Obesity and Fibrosis: Setting the Stage for Breast Cancer. Cancers (Basel) 2023; 15:cancers15112929. [PMID: 37296891 DOI: 10.3390/cancers15112929] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Obesity is a rising health concern and is linked to a worsened breast cancer prognosis. Tumor desmoplasia, which is characterized by elevated numbers of cancer-associated fibroblasts and the deposition of fibrillar collagens within the stroma, may contribute to the aggressive clinical behavior of breast cancer in obesity. A major component of the breast is adipose tissue, and fibrotic changes in adipose tissue due to obesity may contribute to breast cancer development and the biology of the resulting tumors. Adipose tissue fibrosis is a consequence of obesity that has multiple sources. Adipocytes and adipose-derived stromal cells secrete extracellular matrix composed of collagen family members and matricellular proteins that are altered by obesity. Adipose tissue also becomes a site of chronic, macrophage-driven inflammation. Macrophages exist as a diverse population within obese adipose tissue and mediate the development of fibrosis through the secretion of growth factors and matricellular proteins and interactions with other stromal cells. While weight loss is recommended to resolve obesity, the long-term effects of weight loss on adipose tissue fibrosis and inflammation within breast tissue are less clear. Increased fibrosis within breast tissue may increase the risk for tumor development as well as promote characteristics associated with tumor aggressiveness.
Collapse
Affiliation(s)
- Genevra Kuziel
- Cancer Biology Graduate Program, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Brittney N Moore
- Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706, USA
| | - Lisa M Arendt
- Cancer Biology Graduate Program, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706, USA
| |
Collapse
|
20
|
Luo J, Zhang M, Deng Y, Li H, Bu Q, Liu R, Yu J, Liu S, Zeng Z, Sun W, Gui G, Qian X, Li Y. Copper nanoparticles lead to reproductive dysfunction by affecting key enzymes of ovarian hormone synthesis and metabolism in female rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 254:114704. [PMID: 36898311 DOI: 10.1016/j.ecoenv.2023.114704] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/07/2023] [Accepted: 02/26/2023] [Indexed: 06/18/2023]
Abstract
Studies on the general toxicity of copper nanoparticles (Cu NPs) have been conducted extensively, but their effects on reproductive toxicity remain unclear. In this study, we evaluated the toxic effect of Cu NPs on pregnant rats and their litter. The comparative in vivo toxicity of Cu ions, Cu NPs, and Cu microparticles (MPs) was studied in a 17-day repeated oral-dose experiment at the doses of 60, 120, and 180 mg/kg/day in pregnant rats. The pregnancy rate, mean live litter size, and number of dams decreased when exposed to Cu NPs. Moreover, Cu NPs caused a dose-dependent increase in ovarian Cu levels. The metabolomics results showed that Cu NPs caused reproductive dysfunction by altering sex hormones. In addition, in vivo and in vitro experiments showed that the ovarian cytochrome P450 enzymes (CYP450), responsible for hormone production, were significantly upregulated, whereas the enzymes responsible for hormone metabolism were significantly inhibited, resulting in a metabolic imbalance in some ovarian hormones. Furthermore, the results revealed that the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways significantly participated in the regulation of ovarian CYP enzyme expression. Overall, the results of the in vivo and in vitro toxicity experiments with Cu ions, Cu NPs, and Cu MPs suggested that toxicity from nanoscale Cu particles poses a more serious reproductive threat than microscale Cu as Cu NPs could directly damage the ovary and affect the metabolism of ovarian hormones.
Collapse
Affiliation(s)
- Jie Luo
- National Ethnic Affairs Commission Key Open Laboratory of Traditional Chinese Veterinary Medicine, Tongren Polytechnic College, Tongren 554300, China; College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; National and Local Engineering Research Centre for Separation and Purification Ethnic Chinese Veterinary Herbs, Tongren Polytechnic College, Tongren 554300, China; College of Agriculture, Tongren Polytechnic College, Tongren 554300, China; Engineering Research Center of Safe and Efficient Application of Guizhou Province Feed Forage, Tongren 554300, China
| | - Mingzhi Zhang
- Meishan Food and Drug Inspection and Testing Center, Meishan 611330, China
| | - Yang Deng
- Chengdu Animal Genetic Resources Protection Center, Chengdu 611130, China
| | - Haohuan Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qinglong Bu
- Department of Pet Technology, Shandong Vocational Animal Science and Veterinary College, Weifang 261061, China
| | - Rui Liu
- National Ethnic Affairs Commission Key Open Laboratory of Traditional Chinese Veterinary Medicine, Tongren Polytechnic College, Tongren 554300, China; National and Local Engineering Research Centre for Separation and Purification Ethnic Chinese Veterinary Herbs, Tongren Polytechnic College, Tongren 554300, China; College of Agriculture, Tongren Polytechnic College, Tongren 554300, China; Engineering Research Center of Safe and Efficient Application of Guizhou Province Feed Forage, Tongren 554300, China
| | - Jiansheng Yu
- National Ethnic Affairs Commission Key Open Laboratory of Traditional Chinese Veterinary Medicine, Tongren Polytechnic College, Tongren 554300, China; National and Local Engineering Research Centre for Separation and Purification Ethnic Chinese Veterinary Herbs, Tongren Polytechnic College, Tongren 554300, China
| | - Shanshan Liu
- National Ethnic Affairs Commission Key Open Laboratory of Traditional Chinese Veterinary Medicine, Tongren Polytechnic College, Tongren 554300, China; National and Local Engineering Research Centre for Separation and Purification Ethnic Chinese Veterinary Herbs, Tongren Polytechnic College, Tongren 554300, China; College of Agriculture, Tongren Polytechnic College, Tongren 554300, China
| | - Ze Zeng
- National Ethnic Affairs Commission Key Open Laboratory of Traditional Chinese Veterinary Medicine, Tongren Polytechnic College, Tongren 554300, China; National and Local Engineering Research Centre for Separation and Purification Ethnic Chinese Veterinary Herbs, Tongren Polytechnic College, Tongren 554300, China; College of Agriculture, Tongren Polytechnic College, Tongren 554300, China; Engineering Research Center of the Medicinal Diet Industry, Tongren Polytechnic College, Tongren 554300, China
| | - Wei Sun
- College of Agriculture, Tongren Polytechnic College, Tongren 554300, China
| | - Ganbei Gui
- National Ethnic Affairs Commission Key Open Laboratory of Traditional Chinese Veterinary Medicine, Tongren Polytechnic College, Tongren 554300, China; National and Local Engineering Research Centre for Separation and Purification Ethnic Chinese Veterinary Herbs, Tongren Polytechnic College, Tongren 554300, China; College of Agriculture, Tongren Polytechnic College, Tongren 554300, China; Department of Pet Technology, Shandong Vocational Animal Science and Veterinary College, Weifang 261061, China
| | - Xicheng Qian
- National Ethnic Affairs Commission Key Open Laboratory of Traditional Chinese Veterinary Medicine, Tongren Polytechnic College, Tongren 554300, China; National and Local Engineering Research Centre for Separation and Purification Ethnic Chinese Veterinary Herbs, Tongren Polytechnic College, Tongren 554300, China
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
21
|
Rebeaud M, Bouche C, Dauvillier S, Attané C, Arellano C, Vaysse C, Fallone F, Muller C. A novel 3D culture model for human primary mammary adipocytes to study their metabolic crosstalk with breast cancer in lean and obese conditions. Sci Rep 2023; 13:4707. [PMID: 36949082 PMCID: PMC10033714 DOI: 10.1038/s41598-023-31673-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/15/2023] [Indexed: 03/24/2023] Open
Abstract
Obesity is a negative prognosis factor for breast cancer. Yet, the biological mechanisms underlying this effect are still largely unknown. An emerging hypothesis is that the transfer of free fatty acids (FFA) between adipocytes and tumor cells might be altered under obese conditions, contributing to tumor progression. Currently there is a paucity of models to study human mammary adipocytes (M-Ads)-cancer crosstalk. As for other types of isolated white adipocytes, herein, we showed that human M-Ads die within 2-3 days by necrosis when grown in 2D. As an alternative, M-Ads were grown in a fibrin matrix, a 3D model that preserve their distribution, integrity and metabolic function for up to 5 days at physiological glucose concentrations (5 mM). Higher glucose concentrations frequently used in in vitro models promote lipogenesis during M-Ads culture, impairing their lipolytic function. Using transwell inserts, the matrix embedded adipocytes were cocultured with breast cancer cells. FFA transfer between M-Ads and cancer cells was observed, and this event was amplified by obesity. Together these data show that our 3D model is a new tool for studying the effect of M-Ads on tumor cells and beyond with all the components of the tumor microenvironment including the immune cells.
Collapse
Affiliation(s)
- Marie Rebeaud
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
| | - Caroline Bouche
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
- Département de Chirurgie Gynécologique oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, 1 avenue Irène Joliot-Curie, 31059, Toulouse Cedex 9, France
| | - Stéphanie Dauvillier
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
| | - Camille Attané
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
| | - Carlo Arellano
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
- Département de Chirurgie Gynécologique oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, 1 avenue Irène Joliot-Curie, 31059, Toulouse Cedex 9, France
| | - Charlotte Vaysse
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
- Département de Chirurgie Gynécologique oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, 1 avenue Irène Joliot-Curie, 31059, Toulouse Cedex 9, France
| | - Frédérique Fallone
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
| | - Catherine Muller
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France.
| |
Collapse
|
22
|
Terrisse S, Zitvogel L, Kroemer G. Impact of microbiota on breast cancer hormone therapy. Cell Stress 2023; 7:12-19. [PMID: 36926118 PMCID: PMC10012050 DOI: 10.15698/cst2023.03.277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/12/2023] Open
Abstract
Recent observations indicate that the pathogenesis and prognosis of hormone-receptor breast cancer is not only dictated by the properties of the malignant cells but also by immune and microbial parameters. Thus, the immunosurveillance system retards the development of hormone-positive breast cancer and contributes to the therapeutic efficacy of estrogen receptor antagonists and aromatase inhibitors. Moreover, the anticancer immune response is profoundly modulated by the local and intestinal microbiota, which influences cancer cell-intrinsic signaling pathways, affects the composition and function of the immune infiltrate present in the tumor microenvironment and modulates the metabolism of estrogens. Indeed, specific bacteria in the gut produce enzymes that affect the enterohepatic cycle of estrogen metabolites, convert estrogens into androgens or generate estrogen-like molecules. The knowledge of these circuitries is in its infancy, calling for further in-depth analyses.
Collapse
Affiliation(s)
| | - Laurence Zitvogel
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France.,University Paris Saclay, Gif-sur-Yvette, France.,Gustave Roussy, ClinicObiome, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Institut Universitaire de France, Inserm U1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
23
|
Bhardwaj P, Iyengar NM, Zahid H, Carter KM, Byun DJ, Choi MH, Sun Q, Savenkov O, Louka C, Liu C, Piloco P, Acosta M, Bareja R, Elemento O, Foronda M, Dow LE, Oshchepkova S, Giri DD, Pollak M, Zhou XK, Hopkins BD, Laughney AM, Frey MK, Ellenson LH, Morrow M, Spector JA, Cantley LC, Brown KA. Obesity promotes breast epithelium DNA damage in women carrying a germline mutation in BRCA1 or BRCA2. Sci Transl Med 2023; 15:eade1857. [PMID: 36812344 PMCID: PMC10557057 DOI: 10.1126/scitranslmed.ade1857] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 01/23/2023] [Indexed: 02/24/2023]
Abstract
Obesity, defined as a body mass index (BMI) ≥ 30, is an established risk factor for breast cancer among women in the general population after menopause. Whether elevated BMI is a risk factor for women with a germline mutation in BRCA1 or BRCA2 is less clear because of inconsistent findings from epidemiological studies and a lack of mechanistic studies in this population. Here, we show that DNA damage in normal breast epithelia of women carrying a BRCA mutation is positively correlated with BMI and with biomarkers of metabolic dysfunction. In addition, RNA sequencing showed obesity-associated alterations to the breast adipose microenvironment of BRCA mutation carriers, including activation of estrogen biosynthesis, which affected neighboring breast epithelial cells. In breast tissue explants cultured from women carrying a BRCA mutation, we found that blockade of estrogen biosynthesis or estrogen receptor activity decreased DNA damage. Additional obesity-associated factors, including leptin and insulin, increased DNA damage in human BRCA heterozygous epithelial cells, and inhibiting the signaling of these factors with a leptin-neutralizing antibody or PI3K inhibitor, respectively, decreased DNA damage. Furthermore, we show that increased adiposity was associated with mammary gland DNA damage and increased penetrance of mammary tumors in Brca1+/- mice. Overall, our results provide mechanistic evidence in support of a link between elevated BMI and breast cancer development in BRCA mutation carriers. This suggests that maintaining a lower body weight or pharmacologically targeting estrogen or metabolic dysfunction may reduce the risk of breast cancer in this population.
Collapse
Affiliation(s)
- Priya Bhardwaj
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Neil M. Iyengar
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Heba Zahid
- Department of Medical Laboratory Technology, College of Applied Medical Science, Taibah University, Medina 42353, Saudi Arabia
| | | | - Dong Jun Byun
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Man Ho Choi
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Qi Sun
- Computational Biology Service Unit of Life Sciences Core Laboratories Center, Cornell University, Ithaca, NY 14853, USA
| | - Oleksandr Savenkov
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, USA
| | - Charalambia Louka
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Catherine Liu
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Phoebe Piloco
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Monica Acosta
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Rohan Bareja
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Miguel Foronda
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lukas E. Dow
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sofya Oshchepkova
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Dilip D. Giri
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael Pollak
- Departments of Medicine and Oncology, McGill University, Montreal, Canada
| | - Xi Kathy Zhou
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY 10065, USA
| | - Benjamin D. Hopkins
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ashley M. Laughney
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Melissa K. Frey
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lora Hedrick Ellenson
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Monica Morrow
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jason A. Spector
- Laboratory of Bioregenerative Medicine and Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lewis C. Cantley
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
24
|
Pati S, Irfan W, Jameel A, Ahmed S, Shahid RK. Obesity and Cancer: A Current Overview of Epidemiology, Pathogenesis, Outcomes, and Management. Cancers (Basel) 2023; 15:485. [PMID: 36672434 PMCID: PMC9857053 DOI: 10.3390/cancers15020485] [Citation(s) in RCA: 186] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Obesity or excess body fat is a major global health challenge that has not only been associated with diabetes mellitus and cardiovascular disease but is also a major risk factor for the development of and mortality related to a subgroup of cancer. This review focuses on epidemiology, the relationship between obesity and the risk associated with the development and recurrence of cancer and the management of obesity. METHODS A literature search using PubMed and Google Scholar was performed and the keywords 'obesity' and cancer' were used. The search was limited to research papers published in English prior to September 2022 and focused on studies that investigated epidemiology, the pathogenesis of cancer, cancer incidence and the risk of recurrence, and the management of obesity. RESULTS About 4-8% of all cancers are attributed to obesity. Obesity is a risk factor for several major cancers, including post-menopausal breast, colorectal, endometrial, kidney, esophageal, pancreatic, liver, and gallbladder cancer. Excess body fat results in an approximately 17% increased risk of cancer-specific mortality. The relationship between obesity and the risk associated with the development of cancer and its recurrence is not fully understood and involves altered fatty acid metabolism, extracellular matrix remodeling, the secretion of adipokines and anabolic and sex hormones, immune dysregulation, and chronic inflammation. Obesity may also increase treatment-related adverse effects and influence treatment decisions regarding specific types of cancer therapy. Structured exercise in combination with dietary support and behavior therapy are effective interventions. Treatment with glucagon-like peptide-1 analogues and bariatric surgery result in more rapid weight loss and can be considered in selected cancer survivors. CONCLUSIONS Obesity increases cancer risk and mortality. Weight-reducing strategies in obesity-associated cancers are important interventions as a key component of cancer care. Future studies are warranted to further elucidate the complex relationship between obesity and cancer with the identification of targets for effective interventions.
Collapse
Affiliation(s)
- Sukanya Pati
- College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | | | - Ahmad Jameel
- College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Shahid Ahmed
- College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
- Saskatoon Cancer Center, Saskatchewan Cancer Agency, Saskatoon, SK S7N 4H4, Canada
| | - Rabia K. Shahid
- College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
25
|
Krum-Hansen S, Standahl Olsen K, Anderssen E, Frantzen JO, Lund E, Paulssen RH. Associations of breast cancer related exposures and gene expression profiles in normal breast tissue-The Norwegian Women and Cancer normal breast tissue study. Cancer Rep (Hoboken) 2023; 6:e1777. [PMID: 36617746 PMCID: PMC10075301 DOI: 10.1002/cnr2.1777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 11/11/2022] [Accepted: 12/12/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Normal breast tissue is utilized in tissue-based studies of breast carcinogenesis. While gene expression in breast tumor tissue is well explored, our knowledge of transcriptomic signatures in normal breast tissue is still incomplete. The aim of this study was to investigate variability of gene expression in a large sample of normal breast tissue biopsies, according to breast cancer related exposures (obesity, smoking, alcohol, hormone therapy, and parity). METHODS We analyzed gene expression profiles from 311 normal breast tissue biopsies from cancer-free, post-menopausal women, using Illumina bead chip arrays. Principal component analysis and K-means clustering was used for initial analysis of the dataset. The association of exposures and covariates with gene expression was determined using linear models for microarrays. RESULTS Heterogeneity of the breast tissue and cell composition had the strongest influence on gene expression profiles. After adjusting for cell composition, obesity, smoking, and alcohol showed the highest numbers of associated genes and pathways, whereas hormone therapy and parity were associated with negligible gene expression differences. CONCLUSION Our results provide insight into associations between major exposures and gene expression profiles and provide an informative baseline for improved understanding of exposure-related molecular events in normal breast tissue of cancer-free, post-menopausal women.
Collapse
Affiliation(s)
- Sanda Krum-Hansen
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Karina Standahl Olsen
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Endre Anderssen
- Genomics Support Center Tromsø (GSCT), UiT The Arctic University of Norway, Tromsø, Norway
| | - Jan Ole Frantzen
- Narvik Hospital, University Hospital of North Norway, Narvik, Norway
| | - Eiliv Lund
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ruth H Paulssen
- Genomics Support Center Tromsø (GSCT), UiT The Arctic University of Norway, Tromsø, Norway.,Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
26
|
Clapp B, Portela R, Sharma I, Nakanishi H, Marrero K, Schauer P, Halfdanarson TR, Abu Dayyeh B, Kendrick M, Ghanem OM. Risk of non-hormonal cancer after bariatric surgery: meta-analysis of retrospective observational studies. Br J Surg 2022; 110:24-33. [PMID: 36259310 DOI: 10.1093/bjs/znac343] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/18/2022] [Accepted: 09/20/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Obesity is associated with an increased incidence of at least 13 types of cancer. Although bariatric surgery has been associated with a reduced risk of hormonal cancers, data for non-hormonal cancers are scarce. The aim of this study was to evaluate the effect of bariatric surgery on the incidence of non-hormonal cancers. METHODS Cochrane, Embase, PubMed, Scopus, and Web of Science were searched for articles between 1984 and 2018, following the PRISMA system. Meta-analysis was conducted using a random-effect model with subgroup analysis by procedure and cancer type. RESULTS From 2526 studies screened, 15 were included. There were a total of 18 583 477 patients, 947 787 in the bariatric group and 17 635 690 in the control group. In comparison to the non-surgical group, the bariatric group had a lower incidence of cancer (OR .65 (95 per cent c.i. 0.53 to 0.80); P < 0.002). In the subgroup analysis, Roux-en-Y gastric bypass and sleeve gastrectomy were associated with decreased risk of developing cancer, while no difference was observed with adjustable gastric banding. When evaluated by cancer type, liver (OR 0.417 (95 per cent c.i. 0.323 to 0.538)), colorectal (OR 0.64 (95 per cent c.i. 0.49 to 0.84)), kidney and urinary tract cancer (OR 0.77 (95 per cent c.i. 0.72 to 0.83)), oesophageal (OR 0.60 (95 per cent c.i. 0.43 to 0.85)), and lung cancer (OR 0.796 (95 per cent c.i. 0.45 to 0.80)) also presented a lower cancer incidence in the bariatric group. CONCLUSION Bariatric surgery is related to an almost 50 per cent reduction in the risk of non-hormonal cancers.
Collapse
Affiliation(s)
- Benjamin Clapp
- Department of Surgery, Texas Tech HSC Paul Foster School of Medicine, El Paso, Texas, USA
| | - Ray Portela
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ishna Sharma
- Department of Surgery, University of Connecticut, Farmington, Connecticut, USA
| | | | - Katie Marrero
- Department of Surgery, Carle Foundation Hospital General Surgery Residency, Champaign, Illinois, USA
| | - Philip Schauer
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | | | - Barham Abu Dayyeh
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Omar M Ghanem
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
27
|
Hassan NF, Hassan AH, El-Ansary MR. Cytokine modulation by etanercept ameliorates metabolic syndrome and its related complications induced in rats administered a high-fat high-fructose diet. Sci Rep 2022; 12:20227. [PMID: 36418417 PMCID: PMC9684438 DOI: 10.1038/s41598-022-24593-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
The aim of the present study was to investigate the effect of etanercept (ETA)-an anti-tumor necrosis factor α (TNF-α) monoclonal antibody-on metabolic disorders such as obesity, hypertension, dyslipidemia, and insulin resistance associated with the metabolic syndrome (MS). MS was induced in rats via high-fat high-fructose (HFHF) administration for 8 weeks. Rats were divided into three groups: negative control, HFHF model, and ETA-treated groups [HFHF + ETA (0.8 mg/kg/twice weekly, subcutaneously) administered in the last 4 weeks]. ETA effectively diminished the prominent features of MS via a significant reduction in the percent body weight gain along with the modulation of adipokine levels, resulting in a significant elevation of serum adiponectin consistent with TNF-α and serum leptin level normalization. Moreover, ETA enhanced dyslipidemia and the elevated blood pressure. ETA managed the prominent features of MS and its associated complications via the downregulation of the hepatic inflammatory pathway that induces nonalcoholic steatohepatitis (NASH)-from the expression of Toll-like receptor 4, nuclear factor kappa B, and TNF-α until that of transforming growth factor-in addition to significant improvements in glucose utilization, insulin sensitivity, and liver function parameter activity and histopathological examination. ETA was effective for the treatment of all prominent features of MS and its associated complications, such as type II diabetes mellitus and NASH.
Collapse
Affiliation(s)
- Noha F. Hassan
- grid.440876.90000 0004 0377 3957Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Azza H. Hassan
- grid.7776.10000 0004 0639 9286Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Mona R. El-Ansary
- grid.440876.90000 0004 0377 3957Department of Biochemistry, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| |
Collapse
|
28
|
Buono G, Arpino G, Del Mastro L, Fabi A, Generali D, Puglisi F, Zambelli A, Cinieri S, Nuzzo F, Di Lauro V, Vigneri P, Bianchini G, Montemurro F, Gennari A, De Laurentiis M. Extended adjuvant endocrine treatment for premenopausal women: A Delphi approach to guide clinical practice. Front Oncol 2022; 12:1032166. [PMID: 36387212 PMCID: PMC9645191 DOI: 10.3389/fonc.2022.1032166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/22/2022] [Indexed: 10/02/2023] Open
Abstract
The use of an aromatase inhibitor (AI) in combination with ovarian function suppression (OFS) has become the mainstay of adjuvant endocrine therapy in high-risk premenopausal patients with hormone receptor-positive breast cancer. Although five years of such therapy effectively reduces recurrence rates, a substantial risk of late recurrence remains in this setting. Multiple trials have shown that extending AI treatment beyond five years could offer further protection. However, as these studies comprised only postmenopausal patients, no direct evidence currently exists to inform about the potential benefits and/or side effects of extended AI + OFS therapies in premenopausal women. Given these grey areas, we conducted a Delphi survey to report on the opinion of experts in breast cancer treatment and summarize a consensus on the discussed topics. A total of 44 items were identified, all centred around two main themes: 1) defining reliable prognostic factors to pinpoint premenopausal patients eligible for endocrine therapy extension; 2) designing how such therapy should optimally be administered in terms of treatment combinations and duration based on patients' menopausal status. Each item was separately discussed and anonymously voted by 12 experts representing oncological institutes spread across Italy. The consensus threshold was reached in 36 out of 44 items (82%). Herein, we discuss the levels of agreement/disagreement achieved by each item in relation to the current body of literature. In the absence of randomized trials to guide the tailoring of extended AI treatment in premenopausal women, conclusions from our study provide a framework to assist routine clinical practice.
Collapse
Affiliation(s)
- Giuseppe Buono
- Department of Breast and Thoracic Oncology, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione “G. Pascale”, Naples, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples “Federico II”, Naples, Italy
| | - Lucia Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy
- Department of Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “San Martino” General Hospital, Genoa, Italy
| | - Alessandra Fabi
- Precision Medicine in Breast Cancer, Scientific Directorate, Department of Women Child and Public Health, Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Daniele Generali
- Breast Cancer Unit, Azienda Socio-Sanitaria Territoriale di Cremona, Cremona, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Fabio Puglisi
- Department of Medicine, University of Udine, Udine, Italy
- Department of Medical Oncology, Centro di Riferimento Oncologico (CRO) Aviano, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Alberto Zambelli
- Medical Oncology, “Papa Giovanni XXIII” Hospital, Bergamo, Italy
| | - Saverio Cinieri
- Medical Oncology Division and Breast Unit, “Senatore Antonio Perrino” Hospital, Brindisi, Italy
| | - Francesco Nuzzo
- Department of Breast and Thoracic Oncology, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione “G. Pascale”, Naples, Italy
| | - Vincenzo Di Lauro
- Department of Breast and Thoracic Oncology, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione “G. Pascale”, Naples, Italy
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giampaolo Bianchini
- Department of Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “San Raffaele” Hospital, Milan, Italy
| | - Filippo Montemurro
- Breast Unit, Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia - Istituto di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), Candiolo, Italy
| | - Alessandra Gennari
- Medical Oncology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Michelino De Laurentiis
- Department of Breast and Thoracic Oncology, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione “G. Pascale”, Naples, Italy
| |
Collapse
|
29
|
Buonaiuto R, Napolitano F, Parola S, De Placido P, Forestieri V, Pecoraro G, Servetto A, Formisano L, Formisano P, Giuliano M, Arpino G, De Placido S, De Angelis C. Insight on the Role of Leptin: A Bridge from Obesity to Breast Cancer. Biomolecules 2022; 12:biom12101394. [PMID: 36291602 PMCID: PMC9599120 DOI: 10.3390/biom12101394] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/21/2022] [Accepted: 09/25/2022] [Indexed: 11/26/2022] Open
Abstract
Leptin is a peptide hormone, mainly known for its role as a mediator of adipose tissue endocrine functions, such as appetite control and energy homeostasis. In addition, leptin signaling is involved in several physiological processes as modulation of innate and adaptive immune responses and regulation of sex hormone levels. When adipose tissue expands, an imbalance of adipokines secretion may occur and increasing leptin levels contribute to promoting a chronic inflammatory state, which is largely acknowledged as a hallmark of cancer. Indeed, upon binding its receptor (LEPR), leptin activates several oncogenic pathways, such as JAK/STAT, MAPK, and PI3K/AKT, and seems to affect cancer immune response by inducing a proinflammatory immune polarization and eventually enhancing T-cell exhaustion. In particular, obesity-associated hyperleptinemia has been related to breast cancer risk development, although the underlying mechanism is yet to be completely clarified and needs to be deemed in light of multiple variables, such as menopausal state and immune response. The aim of this review is to provide an overview of the potential role of leptin as a bridge between obesity and breast cancer and to establish the physio-pathological basis of the linkage between these major health concerns in order to identify appropriate and novel therapeutic strategies to adopt in daily clinical practice.
Collapse
Affiliation(s)
- Roberto Buonaiuto
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Fabiana Napolitano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Sara Parola
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Valeria Forestieri
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Giovanna Pecoraro
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Alberto Servetto
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Pietro Formisano
- Department of Translational Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
- Correspondence:
| |
Collapse
|
30
|
Wu Y, Li X, Li Q, Cheng C, Zheng L. Adipose tissue-to-breast cancer crosstalk: Comprehensive insights. Biochim Biophys Acta Rev Cancer 2022; 1877:188800. [PMID: 36103907 DOI: 10.1016/j.bbcan.2022.188800] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
The review focuses on mechanistic evidence for the link between obesity and breast cancer. According to the IARC study, there is sufficient evidence that obesity is closely related to a variety of cancers. Among them, breast cancer is particularly disturbed by adipose tissue due to the unique histological structure of the breast. The review introduces the relationship between obesity and breast cancer from two aspects, including factors that promote tumorigenesis or metastasis. We summarize alterations in adipokines and metabolic pathways that contribute to breast cancer development. Breast cancer metastasis is closely related to obesity-induced pro-inflammatory microenvironment, adipose stem cells, and miRNAs. Based on the mechanism by which obesity causes breast cancer, we list possible therapeutic directions, including reducing the risk of breast cancer and inhibiting the progression of breast cancer. We also discussed the risk of autologous breast remodeling and fat transplantation. Finally, the causes of the obesity paradox and the function of enhancing immunity are discussed. Evaluating the balance between obesity-induced inflammation and enhanced immunity warrants further study.
Collapse
Affiliation(s)
- Yuan Wu
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Xu Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Qiong Li
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Chienshan Cheng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Lan Zheng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China.
| |
Collapse
|
31
|
Hillers-Ziemer LE, Kuziel G, Williams AE, Moore BN, Arendt LM. Breast cancer microenvironment and obesity: challenges for therapy. Cancer Metastasis Rev 2022; 41:627-647. [PMID: 35435599 PMCID: PMC9470689 DOI: 10.1007/s10555-022-10031-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023]
Abstract
Women with obesity who develop breast cancer have a worsened prognosis with diminished survival rates and increased rates of metastasis. Obesity is also associated with decreased breast cancer response to endocrine and chemotherapeutic treatments. Studies utilizing multiple in vivo models of obesity as well as human breast tumors have enhanced our understanding of how obesity alters the breast tumor microenvironment. Changes in the complement and function of adipocytes, adipose-derived stromal cells, immune cells, and endothelial cells and remodeling of the extracellular matrix all contribute to the rapid growth of breast tumors in the context of obesity. Interactions of these cells enhance secretion of cytokines and adipokines as well as local levels of estrogen within the breast tumor microenvironment that promote resistance to multiple therapies. In this review, we will discuss our current understanding of the impact of obesity on the breast tumor microenvironment, how obesity-induced changes in cellular interactions promote resistance to breast cancer treatments, and areas for development of treatment interventions for breast cancer patients with obesity.
Collapse
Affiliation(s)
- Lauren E Hillers-Ziemer
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Genevra Kuziel
- Program in Cancer Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Abbey E Williams
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Brittney N Moore
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Lisa M Arendt
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Program in Cancer Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Dr. Rm 4354A, Madison, WI, 53706, USA.
| |
Collapse
|
32
|
Sherman ME, de Bel T, Heckman MG, White L, Ogony J, Stallings-Mann M, Hilton T, Degnim AC, Vierkant RA, Hoskin T, Jensen M, Pacheco-Spann L, Henry JE, Storniolo AM, Carter JM, Winham SJ, Radisky DC, van der Laak J. Serum hormone levels and normal breast histology among premenopausal women. Breast Cancer Res Treat 2022; 194:149-158. [PMID: 35503494 PMCID: PMC9869890 DOI: 10.1007/s10549-022-06600-9] [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: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 01/26/2023]
Abstract
PURPOSE Breast terminal duct lobular units (TDLUs) are the main source of breast cancer (BC) precursors. Higher serum concentrations of hormones and growth factors have been linked to increased TDLU numbers and to elevated BC risk, with variable effects by menopausal status. We assessed associations of circulating factors with breast histology among premenopausal women using artificial intelligence (AI) and preliminarily tested whether parity modifies associations. METHODS Pathology AI analysis was performed on 316 digital images of H&E-stained sections of normal breast tissues from Komen Tissue Bank donors ages ≤ 45 years to assess 11 quantitative metrics. Associations of circulating factors with AI metrics were assessed using regression analyses, with inclusion of interaction terms to assess effect modification. RESULTS Higher prolactin levels were related to larger TDLU area (p < 0.001) and increased presence of adipose tissue proximate to TDLUs (p < 0.001), with less significant positive associations for acini counts (p = 0.012), dilated acini (p = 0.043), capillary area (p = 0.014), epithelial area (p = 0.007), and mononuclear cell counts (p = 0.017). Testosterone levels were associated with increased TDLU counts (p < 0.001), irrespective of parity, but associations differed by adipose tissue content. AI data for TDLU counts generally agreed with prior visual assessments. CONCLUSION Among premenopausal women, serum hormone levels linked to BC risk were also associated with quantitative features of normal breast tissue. These relationships were suggestively modified by parity status and tissue composition. We conclude that the microanatomic features of normal breast tissue may represent a marker of BC risk.
Collapse
Affiliation(s)
- Mark E Sherman
- Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida, USA
| | - Thomas de Bel
- Department of Pathology, Radboud University Medical Center,Radboud Institute of Health Sciences, Nijmegen, The Netherlands
| | | | - Launia White
- Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida, USA
| | - Joshua Ogony
- Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Tracy Hilton
- Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida, USA
| | - Amy C. Degnim
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Tanya Hoskin
- Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida, USA
| | - Matthew Jensen
- Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Jill E. Henry
- Susan G. Komen Tissue Bank at the IU Simon Cancer Center, Indiana University School of Medicine
| | - Anna Maria Storniolo
- Susan G. Komen Tissue Bank at the IU Simon Cancer Center, Indiana University School of Medicine
| | - Jodi M. Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Stacey J. Winham
- Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida, USA
| | - Derek C. Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jeroen van der Laak
- Department of Pathology, Radboud University Medical Center,Radboud Institute of Health Sciences, Nijmegen, The Netherlands,Center for Medical Image Science and Visualization, Linköping University, Linköping, Sweden
| |
Collapse
|
33
|
Chen Y, Han X, Wang L, Wen Q, Li L, Sun L, Chen Q. Multiple roles of ghrelin in breast cancer. Int J Biol Markers 2022; 37:241-248. [PMID: 35763463 DOI: 10.1177/03936155221110247] [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: 11/17/2022]
Abstract
Breast cancer is one of the most threatening malignant tumors in women worldwide; hence, investigators are continually performing novel research in this field. However, an accurate prediction of its prognosis and postoperative recovery remains difficult. The severity of breast cancer is patient-specific and affected by several health factors; thus, unknown mechanisms may affect its progression. This article analyzes existing literature on breast cancer, ranging from the discovery of ghrelin to its present use, and aims to provide a reference for future research into breast cancer mechanisms and treatment-plan improvement. Various parts of ghrelin have been associated with breast cancer by direct or indirect evidence. The ghrelin system may encompass the direction of expanding breast cancer treatment methods and prognostic indicators. Therefore, we compiled almost all studies on the relationship between the ghrelin system and breast cancer, including unacylated ghrelin, its GHRL gene, ghrelin O-acyltransferase, the receptor growth hormone secretagogue receptor, and several splice variants of ghrelin to lay the foundation for future research.
Collapse
Affiliation(s)
- Yiding Chen
- 176759Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuke Han
- 176759Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lan Wang
- 176759Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qing Wen
- 176759Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liufu Li
- Pengshan District People's Hospital of Meishan City, Meishan, China
| | - Lisha Sun
- 176759Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiu Chen
- 176759Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
34
|
Ku HC, Cheng CF. Role of adipocyte browning in prostate and breast tumor microenvironment. Tzu Chi Med J 2022; 34:359-366. [PMID: 36578640 PMCID: PMC9791856 DOI: 10.4103/tcmj.tcmj_62_22] [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: 03/22/2022] [Revised: 04/06/2022] [Accepted: 04/15/2022] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer (PC) and breast cancer (BC) are the most common cancers in men and women, respectively, in developed countries. The increased incidence of PC and BC largely reflects an increase in the prevalence of obesity and metabolic syndrome. In pathological conditions involving the development and progression of PC and BC, adipose tissue plays an important role via paracrine and endocrine signaling. The increase in the amount of local adipose tissue, specifically periprostatic adipose tissue, may be a key contributor to the PC pathobiology. Similarly, breast adipose tissue secretion affects various aspects of BC by influencing tumor progression, angiogenesis, metastasis, and microenvironment. In this context, the role of white adipose tissue (WAT) has been extensively studied. However, the influence of browning of the WAT on the development and progression of PC and BC is unclear and has received less attention. In this review, we highlight that adipose tissue plays a vital role in the regulation of the tumor microenvironment in PC or BC and highlight the probable underlying mechanisms linking adipose tissue with PC or BC. We further discuss whether the browning of WAT could be a therapeutic strategy for the treatment of PC and BC.
Collapse
Affiliation(s)
- Hui-Chen Ku
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Ching-Feng Cheng
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan,Department of Pediatrics, School of Medicine, Tzu Chi University, Hualien, Taiwan,Address for correspondence: Dr. Ching-Feng Cheng, Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 289, Jianguo Road, Xindian District, New Taipei, Taiwan. E-mail:
| |
Collapse
|
35
|
Song H, Jeong A, Tran TXM, Lee J, Kim M, Park B. Association between Micronutrient Intake and Breast Cancer Risk According to Body Mass Index in South Korean Adult Women: A Cohort Study. Nutrients 2022; 14:nu14132644. [PMID: 35807825 PMCID: PMC9268499 DOI: 10.3390/nu14132644] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 02/07/2023] Open
Abstract
This study investigated the association between micronutrient intake and breast cancer risk in South Korean adult women. This association was stratified according to body mass index (BMI) categories. Data from the Korean Genome and Epidemiology Study (KoGES) and the Health Examinee Study were analyzed. Altogether, 63,337 individuals (aged ≥40 years) completed the baseline and first follow-up surveys; 40,432 women without a history of cancer at baseline were included in this study. The association between micronutrient intake and breast cancer was determined by estimating the hazard ratio (HR) and 95% confidence interval (CI) using the Cox proportional hazard regression model. A stratified analysis by BMI (<25 kg/m2 and ≥25 kg/m2) was performed. The an analysis of 15 micronutrients and breast cancer risk revealed that none of the micronutrients were associated with breast cancer risk after adjusting for covariates. In obese women, the risk of breast cancer was significantly reduced in the group that consumed vitamin C more than the recommended level (HR = 0.54, 95% CI: 0.31−0.93) and vitamin B6 levels above the recommended level (HR = 0.48, 95% CI: 0.25−0.89). In obese women, exceeding the recommended daily intake levels of vitamin C and vitamin B6 was associated with a lower risk of breast cancer. However, other micronutrients were not associated with breast cancer risk in these women.
Collapse
Affiliation(s)
- Huiyeon Song
- Graduate School of Public Health, Hanyang University, Seoul 04763, Korea; (H.S.); (A.J.)
| | - Ansun Jeong
- Graduate School of Public Health, Hanyang University, Seoul 04763, Korea; (H.S.); (A.J.)
| | - Thi Xuan Mai Tran
- Department of Preventive Medicine, Hanyang University College of Medicine, Seoul 04763, Korea; (T.X.M.T.); (J.L.); (M.K.)
| | - Jiseon Lee
- Department of Preventive Medicine, Hanyang University College of Medicine, Seoul 04763, Korea; (T.X.M.T.); (J.L.); (M.K.)
| | - Mikyung Kim
- Department of Preventive Medicine, Hanyang University College of Medicine, Seoul 04763, Korea; (T.X.M.T.); (J.L.); (M.K.)
| | - Boyoung Park
- Department of Preventive Medicine, Hanyang University College of Medicine, Seoul 04763, Korea; (T.X.M.T.); (J.L.); (M.K.)
- Department of Medicine, Hanyang University College of Medicine, 222 Wangsimni-ro, Seoul 04763, Korea
- Correspondence: ; Tel.: +82-2-2220-0682; Fax: +82-31-2220-0699
| |
Collapse
|
36
|
Devericks EN, Carson MS, McCullough LE, Coleman MF, Hursting SD. The obesity-breast cancer link: a multidisciplinary perspective. Cancer Metastasis Rev 2022; 41:607-625. [PMID: 35752704 PMCID: PMC9470704 DOI: 10.1007/s10555-022-10043-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/31/2022] [Indexed: 12/12/2022]
Abstract
Obesity, exceptionally prevalent in the USA, promotes the incidence and progression of numerous cancer types including breast cancer. Complex, interacting metabolic and immune dysregulation marks the development of both breast cancer and obesity. Obesity promotes chronic low-grade inflammation, particularly in white adipose tissue, which drives immune dysfunction marked by increased pro-inflammatory cytokine production, alternative macrophage activation, and reduced T cell function. Breast tissue is predominantly composed of white adipose, and developing breast cancer readily and directly interacts with cells and signals from adipose remodeled by obesity. This review discusses the biological mechanisms through which obesity promotes breast cancer, the role of obesity in breast cancer health disparities, and dietary interventions to mitigate the adverse effects of obesity on breast cancer. We detail the intersection of obesity and breast cancer, with an emphasis on the shared and unique patterns of immune dysregulation in these disease processes. We have highlighted key areas of breast cancer biology exacerbated by obesity, including incidence, progression, and therapeutic response. We posit that interception of obesity-driven breast cancer will require interventions that limit protumor signaling from obese adipose tissue and that consider genetic, structural, and social determinants of the obesity–breast cancer link. Finally, we detail the evidence for various dietary interventions to offset obesity effects in clinical and preclinical studies of breast cancer. In light of the strong associations between obesity and breast cancer and the rising rates of obesity in many parts of the world, the development of effective, safe, well-tolerated, and equitable interventions to limit the burden of obesity on breast cancer are urgently needed.
Collapse
Affiliation(s)
- Emily N Devericks
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Meredith S Carson
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lauren E McCullough
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Michael F Coleman
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephen D Hursting
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
37
|
Kang DW, Wilson RL, Gonzalo-Encabo P, Norris MK, Hans M, Tahbaz M, Dawson J, Nguyen D, Normann AJ, Yunker AG, Sami N, Uno H, Ligibel JA, Mittelman SD, Dieli-Conwright CM. Targeting Adiposity and Inflammation With Movement to Improve Prognosis in Breast Cancer Survivors (The AIM Trial): Rationale, Design, and Methods. Front Oncol 2022; 12:896995. [PMID: 35795051 PMCID: PMC9251632 DOI: 10.3389/fonc.2022.896995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/27/2022] [Indexed: 11/21/2022] Open
Abstract
Background Obesity is a significant contributor to breast cancer recurrence and mortality. A central mechanism by which obesity stimulates cancer progression is through chronic, low-grade inflammation in adipose tissue. Exercise interventions to target chronic inflammation has a potential to improve obesity- and breast cancer-related outcomes; however, no studies have investigated the roles of exercise in modulating adipose tissue inflammation in breast cancer survivors. Also, it is unclear which exercise prescription would be optimal to maximize the outcomes. Therefore, we designed a randomized controlled trial (Taking AIM at Breast Cancer: Targeting Adiposity and Inflammation with Movement to Improve Prognosis in Breast Cancer Survivors [AIM] Trial) to examine the mechanisms by which different modalities of exercise impact chronic inflammation as a biomarker of breast cancer prognosis. Methods The AIM trial is a prospective, three-armed, phase II randomized controlled trial investigating the effects of a 16-week supervised circuit aerobic and resistance exercise (CARE) program versus a traditional aerobic and resistance exercise (TARE) program and attention control (AC) on adipose tissue inflammation in breast cancer survivors. 276 patients who are diagnosed with stage 0-III breast cancer, post-treatment, sedentary, and centrally obese are randomized to one of the three groups. The CARE and TARE groups participate in thrice-weekly supervised exercise sessions for 16 weeks. The AC group are offered the CARE program after the intervention period. The primary endpoint is adipose tissue inflammation assessed by core biopsy and blood draw. The secondary and tertiary endpoints are sarcopenic obesity, physical fitness and function, and patient reported outcomes. The exploratory outcomes are long-term breast cancer outcomes. Discussion This is the first randomized controlled trial examining the effects of exercise on adipose tissue inflammation in obese, breast cancer survivors. Our findings are anticipated to contribute to a better understanding of exercise modalities and mechanisms on adipose tissue inflammation that can potentially improve breast cancer prognosis. Clinical Trial Registration https://clinicaltrials.gov/ct2/show/NCT03091842 identifier [NCT#03091842].
Collapse
Affiliation(s)
- Dong-Woo Kang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Rebekah L. Wilson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Paola Gonzalo-Encabo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Mary K. Norris
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Marybeth Hans
- Division of Breast Surgery, Brigham and Women’s Hospital, Boston, MA, United States
| | - Meghan Tahbaz
- Department of Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Jackie Dawson
- Department of Physical Therapy, California State University, Long Beach, Long Beach, CA, United States
| | - Danny Nguyen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Amber J. Normann
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Health Sciences, Boston University, Boston, MA, United States
| | - Alexandra G. Yunker
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Nathalie Sami
- Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Hajime Uno
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Jennifer A. Ligibel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Steven D. Mittelman
- Children’s Discovery and Innovations Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Christina M. Dieli-Conwright
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
38
|
Gonzalo-Encabo P, Wilson RL, Kang DW, Norris MK, Uno H, Christopher CN, Chow C, Sami N, Fox FS, Ligibel JA, Dieli-Conwright CM. Reducing Metabolic Dysregulation in Obese Latina and/or Hispanic Breast Cancer Survivors Using Physical Activity (ROSA) Trial: A Study Protocol. Front Oncol 2022; 12:864844. [PMID: 35619910 PMCID: PMC9128380 DOI: 10.3389/fonc.2022.864844] [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: 01/28/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background Latina and Hispanic breast cancer survivors (LHBCS) are at increased risk for long-term complications and poorer metabolic health, including metabolic dysregulation (MetD) before and following breast cancer diagnosis. MetD can increase risk of cancer recurrence, death, and comorbid conditions by increasing inflammation and cancer cell proliferation. While exercise improves physical fitness and metabolic outcomes in breast cancer survivors, there is a lack of studies including underrepresented and disadvantaged minority groups such as LHBCS. Methods Our 12-month randomized (exercise or attention control) controlled trial (the ROSA trial) aims to utilize a progressive combined aerobic and resistance exercise program to improve MetD, insulin resistance, and visceral adiposity among obese LHBCS. We aim to recruit 160 women with Stage I-III breast cancer who are sedentary, centrally obese, and have completed treatment (e.g., surgery, radiation, chemotherapy) prior to enrollment. Participants randomized to the exercise group receive 16-weeks of virtually supervised aerobic and resistance training, followed by 16-weeks of unsupervised home-based aerobic and resistance exercise, and 16-weeks of follow-up. The attention control group receive a 12-month home-based stretching program. Primary and secondary outcomes are measured every 4-weeks during study visits. Discussion The ROSA trial is the first exercise oncology trial targeting high-risk sedentary, obese LHBCS to improve MetD-related outcomes. Results of this trial will help illuminate how exercise impacts health-related outcomes, survivorship, and recurrence, and inform future exercise oncology guidelines to reduce health disparities among minority cancer survivors.
Collapse
Affiliation(s)
- Paola Gonzalo-Encabo
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Rebekah L Wilson
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Dong-Woo Kang
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Mary K Norris
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Hajime Uno
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Cami N Christopher
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Christina Chow
- UCSF School of Dentistry, University of California, San Francisco, San Francisco, CA, United States
| | - Nathalie Sami
- Department of Internal Medicine, Los Angeles County-University of Southern California (LAC+USC) Medical Center, Keck School of Medicine, Los Angeles, CA, United States
| | - Frank S Fox
- Gerson Lehrman Group, New York, NY, United States
| | - Jennifer A Ligibel
- Department of Medicine, Harvard Medical School, Boston, MA, United States.,Division of Breast Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Christina M Dieli-Conwright
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Department of Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
39
|
Vitamin D deficiency: a potential risk factor for cancer in obesity? Int J Obes (Lond) 2022; 46:707-717. [PMID: 35027681 DOI: 10.1038/s41366-021-01045-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/25/2021] [Accepted: 11/26/2021] [Indexed: 12/20/2022]
Abstract
Obesity is considered an abnormal or excessive accumulation of adipose tissue, due to a prolonged positive energy balance that arises when energy intake is greater than energy expenditure, leading to an increased risk for the individual health and for the development of metabolic chronic diseases including several different types of cancer. Vitamin D deficiency is a metabolic alteration, which is often associated with the obesity condition. Vitamin D is a liposoluble vitamin, which plays a pivotal role in calcium-phosphate metabolism but extraskeletal effects have also been described. Among these, it plays an important role also in adipocyte physiology and glucose metabolism, typically dysregulated in subjects affected by obesity. Moreover, it is now recognized that Vitamin D also influences the processes of cell proliferation, differentiation, adhesion potentially leading to carcinogenesis. Indeed, data indicate a potential link between vitamin D levels and cancer, and higher vitamin D concentrations have been associated with a lower risk of developing different kinds of tumors, including breast, colon, lymphoma, lung, and prostate cancers. Thus, this review will revise the literature regarding this issue investigating and highlighting the potential mechanism of action, which might lead to new therapeutical options.
Collapse
|
40
|
Xiao M, He J, Yin L, Chen X, Zu X, Shen Y. Tumor-Associated Macrophages: Critical Players in Drug Resistance of Breast Cancer. Front Immunol 2022; 12:799428. [PMID: 34992609 PMCID: PMC8724912 DOI: 10.3389/fimmu.2021.799428] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022] Open
Abstract
Drug resistance is one of the most critical challenges in breast cancer (BC) treatment. The occurrence and development of drug resistance are closely related to the tumor immune microenvironment (TIME). Tumor-associated macrophages (TAMs), the most important immune cells in TIME, are essential for drug resistance in BC treatment. In this article, we summarize the effects of TAMs on the resistance of various drugs in endocrine therapy, chemotherapy, targeted therapy, and immunotherapy, and their underlying mechanisms. Based on the current overview of the key role of TAMs in drug resistance, we discuss the potential possibility for targeting TAMs to reduce drug resistance in BC treatment, By inhibiting the recruitment of TAMs, depleting the number of TAMs, regulating the polarization of TAMs and enhancing the phagocytosis of TAMs. Evidences in our review support it is important to develop novel therapeutic strategies to target TAMs in BC to overcome the treatment of resistance.
Collapse
Affiliation(s)
- Maoyu Xiao
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jun He
- Department of Spine Surgery, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Liyang Yin
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiguan Chen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yingying Shen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
41
|
Barone I, Caruso A, Gelsomino L, Giordano C, Bonofiglio D, Catalano S, Andò S. Obesity and endocrine therapy resistance in breast cancer: Mechanistic insights and perspectives. Obes Rev 2022; 23:e13358. [PMID: 34559450 PMCID: PMC9285685 DOI: 10.1111/obr.13358] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/07/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022]
Abstract
The incidence of obesity, a recognized risk factor for various metabolic and chronic diseases, including numerous types of cancers, has risen dramatically over the recent decades worldwide. To date, convincing research in this area has painted a complex picture about the adverse impact of high body adiposity on breast cancer onset and progression. However, an emerging but overlooked issue of clinical significance is the limited efficacy of the conventional endocrine therapies with selective estrogen receptor modulators (SERMs) or degraders (SERDs) and aromatase inhibitors (AIs) in patients affected by breast cancer and obesity. The mechanisms behind the interplay between obesity and endocrine therapy resistance are likely to be multifactorial. Therefore, what have we actually learned during these years and which are the main challenges in the field? In this review, we will critically discuss the epidemiological evidence linking obesity to endocrine therapeutic responses and we will outline the molecular players involved in this harmful connection. Given the escalating global epidemic of obesity, advances in understanding this critical node will offer new precision medicine-based therapeutic interventions and more appropriate dosing schedule for treating patients affected by obesity and with breast tumors resistant to endocrine therapies.
Collapse
Affiliation(s)
- Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Amanda Caruso
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| |
Collapse
|
42
|
Cho BA, Iyengar NM, Zhou XK, Mendieta H, Winston L, Falcone DJ, Landa J, Morrow M, Dannenberg AJ. Increased trunk fat is associated with altered gene expression in breast tissue of normal weight women. NPJ Breast Cancer 2022; 8:15. [PMID: 35087024 PMCID: PMC8795267 DOI: 10.1038/s41523-021-00369-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Increased trunk fat is associated with an elevated risk of breast cancer in normal-weight postmenopausal women. The main objective of this study was to determine whether levels of trunk fat are associated with changes in breast gene expression in normal-weight women. Non-tumorous breast tissue was collected from 32 normal BMI women who underwent mastectomy for breast cancer risk reduction or treatment. Body composition was measured by dual-energy x-ray absorptiometry. High levels of trunk fat were associated with a large number of differentially expressed genes and changes in multiple pathways and processes potentially linked to breast cancer pathogenesis. High levels of trunk fat were also associated with an elevated immune score and increased levels of leptin, CCL2, VEGF-C, IL6, and aromatase. Collectively, these results help to explain why high levels of trunk fat are associated with an increased risk of breast cancer in normal BMI women.
Collapse
Affiliation(s)
- Byuri Angela Cho
- 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.
| | - Xi Kathy Zhou
- Department of Population Health Sciences, Weill Cornell Medical College, New York, NY, USA
| | - Hillary Mendieta
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lisle Winston
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Domenick J Falcone
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Jonathan Landa
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Monica Morrow
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
43
|
Cava E, Marzullo P, Farinelli D, Gennari A, Saggia C, Riso S, Prodam F. Breast Cancer Diet "BCD": A Review of Healthy Dietary Patterns to Prevent Breast Cancer Recurrence and Reduce Mortality. Nutrients 2022; 14:nu14030476. [PMID: 35276833 PMCID: PMC8839871 DOI: 10.3390/nu14030476] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/12/2022] [Accepted: 01/15/2022] [Indexed: 01/27/2023] Open
Abstract
Breast cancer (BC) represents the most common cancer in women, while overweight and obesity are the second preventable cause of cancer. Weight gain and fat accumulation are common after BC diagnosis; moreover, weight gain during the treatment decreases the survival rate and increases the risk of recurrence in breast cancer survivors (BCS). To reduce the risk of second primary cancer or BC recurrence, and all-cause mortality in BCS, multiple interventions have been investigated to obtain reduction in weight, BMI and/or waist circumference. The aim of this narrative review is to analyze evidence on BCS for their risk of recurrence or mortality related to increased weight or fat deposition, and the effects of interventions with healthy dietary patterns to achieve a proper weight and to reduce fat-related risk. The primary focus was on dietary patterns instead of single nutrients and supplements, as the purpose was to investigate on secondary prevention in women free from disease at the end of their cancer treatment. In addition, BC relation with insulin resistance, dietary carbohydrate, and glycemic index/glycemic load is discussed. In conclusion, obesity and overweight, low rates of physical activity, and hormone receptor-status are associated with poorer BC-treatment outcomes. To date, there is a lack of evidence to suggest which dietary pattern is the best approach for weight management in BCS. In the future, multimodal lifestyle interventions with dietary, physical activity and psychological support after BC diagnosis should be studied with the aim of reducing the risk of BC recurrence or mortality.
Collapse
Affiliation(s)
- Edda Cava
- Unit of Dietetic and Clinical Nutrition, University Hospital “Maggiore della Carità”, Corso Mazzini 18, 28100 Novara, Italy; (D.F.); (S.R.)
- Correspondence: ; Tel.: +39-0321-373-3275 (ext. 2108)
| | - Paolo Marzullo
- SCDU Endocrinology, Department of Translational Medicine, University of Eastern Piedmont, Via Solaroli 17, 28100 Novara, Italy; (P.M.); (F.P.)
- Laboratory of Metabolic Research, IRCCS Istituto Auxologico Italiano, 28824 Piancavallo, Italy
| | - Deborah Farinelli
- Unit of Dietetic and Clinical Nutrition, University Hospital “Maggiore della Carità”, Corso Mazzini 18, 28100 Novara, Italy; (D.F.); (S.R.)
| | - Alessandra Gennari
- Division of Oncology, University Hospital “Maggiore della Carità”, Department of Translational Medicine, University of Eastern Piedmont, Via Solaroli 17, 28100 Novara, Italy; (A.G.); (C.S.)
| | - Chiara Saggia
- Division of Oncology, University Hospital “Maggiore della Carità”, Department of Translational Medicine, University of Eastern Piedmont, Via Solaroli 17, 28100 Novara, Italy; (A.G.); (C.S.)
| | - Sergio Riso
- Unit of Dietetic and Clinical Nutrition, University Hospital “Maggiore della Carità”, Corso Mazzini 18, 28100 Novara, Italy; (D.F.); (S.R.)
| | - Flavia Prodam
- SCDU Endocrinology, Department of Translational Medicine, University of Eastern Piedmont, Via Solaroli 17, 28100 Novara, Italy; (P.M.); (F.P.)
- Department of Health Sciences, SCDU Endocrinology, Department of Translational Medicine, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| |
Collapse
|
44
|
Coradini D. De novo cholesterol biosynthesis: an additional therapeutic target for the treatment of postmenopausal breast cancer with excessive adipose tissue. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:841-852. [PMID: 36654818 PMCID: PMC9834634 DOI: 10.37349/etat.2022.00116] [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: 08/23/2022] [Accepted: 11/08/2022] [Indexed: 12/29/2022] Open
Abstract
The onset and development of breast cancer in postmenopausal women are associated with closely related individual-dependent factors, including weight gain and high levels of circulating androgens. Adipose tissue is the most peripheral site of aromatase enzyme synthesis; therefore, the excessive accumulation of visceral fat results in increased androgens aromatization and estradiol production that provides the microenvironment favorable to tumorigenesis in mammary epithelial cells expressing estrogen receptors (ERs). Moreover, to meet the increased requirement of cholesterol for cell membrane assembly and the production of steroid hormones to sustain their proliferation, ER-positive cells activate de novo cholesterol biosynthesis and subsequent steroidogenesis. Several approaches have been followed to neutralize the de novo cholesterol synthesis, including specific enzyme inhibitors, statins, and, more recently, metformin. Cumulating evidence indicated that inhibiting cholesterol biosynthesis by statins and metformin may be a promising therapeutic strategy to block breast cancer progression. Unlike antiestrogens and aromatase inhibitors (AIs) which compete for binding to ER and inhibit androgens aromatization, respectively, statins block the production of mevalonic acid by inhibiting the activity of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, and metformin hampers the activation of the sterol regulatory element-binding protein 2 (SREBP2) transcription factor, thus inhibiting the synthesis of several enzymes involved in cholesterol biosynthesis. Noteworthy, statins and metformin not only improve the prognosis of overweight patients with ER-positive cancer but also improve the prognosis of patients with triple-negative breast cancer, the aggressive tumor subtype that lacks, at present, specific therapy.
Collapse
Affiliation(s)
- Danila Coradini
- Department of Clinical Sciences and Community Health, Campus Cascina Rosa, University of Milan, 20133 Milan, Italy,Correspondence: Danila Coradini, Department of Clinical Sciences and Community Health, Campus Cascina Rosa, University of Milan, Via Vanzetti 5, 20133 Milan, Italy.
| |
Collapse
|
45
|
Holm JB, Rosendahl AH, Borgquist S. Local Biomarkers Involved in the Interplay between Obesity and Breast Cancer. Cancers (Basel) 2021; 13:cancers13246286. [PMID: 34944905 PMCID: PMC8699696 DOI: 10.3390/cancers13246286] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Breast cancer is the second most common cancer in women worldwide. The risk of developing breast cancer depends on various mechanisms, such as age, heredity, reproductive factors, physical inactivity, and obesity. Obesity increases the risk of breast cancer and worsens outcomes for breast cancer patients. The rate of obesity is increasing worldwide, stressing the need for awareness of the association between obesity and breast cancer. In this review, we outline the biomarkers—including cellular and soluble factors—in the breast, associated with obesity, that affect the risk of breast cancer and breast cancer prognosis. Through these biomarkers, we aim to better identify patients with obesity with a higher risk of breast cancer and an inferior prognosis. Abstract Obesity is associated with an increased risk of breast cancer, which is the most common cancer in women worldwide (excluding non-melanoma skin cancer). Furthermore, breast cancer patients with obesity have an impaired prognosis. Adipose tissue is abundant in the breast. Therefore, breast cancer develops in an adipose-rich environment. During obesity, changes in the local environment in the breast occur which are associated with breast cancer. A shift towards a pro-inflammatory state is seen, resulting in altered levels of cytokines and immune cells. Levels of adipokines, such as leptin, adiponectin, and resistin, are changed. Aromatase activity rises, resulting in higher levels of potent estrogen in the breast. Lastly, remodeling of the extracellular matrix takes place. In this review, we address the current knowledge on the changes in the breast adipose tissue in obesity associated with breast cancer initiation and progression. We aim to identify obesity-associated biomarkers in the breast involved in the interplay between obesity and breast cancer. Hereby, we can improve identification of women with obesity with an increased risk of breast cancer and an impaired prognosis. Studies investigating mammary adipocytes and breast adipose tissue in women with obesity versus women without obesity are, however, sparse and further research is needed.
Collapse
Affiliation(s)
- Jonas Busk Holm
- Department of Oncology, Aarhus University Hospital, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
- Correspondence: (J.B.H.); (S.B.)
| | - Ann H. Rosendahl
- Department of Clinical Sciences Lund, Oncology, Lund University, Skåne University Hospital, Barngatan 4, 221 85 Lund, Sweden;
| | - Signe Borgquist
- Department of Oncology, Aarhus University Hospital, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
- Department of Clinical Sciences Lund, Oncology, Lund University, Skåne University Hospital, Barngatan 4, 221 85 Lund, Sweden;
- Correspondence: (J.B.H.); (S.B.)
| |
Collapse
|
46
|
Tapia E, Villa-Guillen DE, Chalasani P, Centuori S, Roe DJ, Guillen-Rodriguez J, Huang C, Galons JP, Thomson CA, Altbach M, Trujillo J, Pinto L, Martinez JA, Algotar AM, Chow HHS. A randomized controlled trial of metformin in women with components of metabolic syndrome: intervention feasibility and effects on adiposity and breast density. Breast Cancer Res Treat 2021; 190:69-78. [PMID: 34383179 PMCID: PMC8560579 DOI: 10.1007/s10549-021-06355-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 08/06/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE Obesity is a known risk factor for post-menopausal breast cancer and may increase risk for triple negative breast cancer in premenopausal women. Intervention strategies are clearly needed to reduce obesity-associated breast cancer risk. METHODS We conducted a Phase II double-blind, randomized, placebo-controlled trial of metformin in overweight/obese premenopausal women with components of metabolic syndrome to assess the potential of metformin for primary breast cancer prevention. Eligible participants were randomized to receive metformin (850 mg BID, n = 76) or placebo (n = 75) for 12 months. Outcomes included breast density, assessed by fat/water MRI with change in percent breast density as the primary endpoint, anthropometric measures, and intervention feasibility. RESULTS Seventy-six percent in the metformin arm and 83% in the placebo arm (p = 0.182) completed the 12-month intervention. Adherence to study agent was high with more than 80% of participants taking ≥ 80% assigned pills. The most common adverse events reported in the metformin arm were gastrointestinal in nature and subsided over time. Compared to placebo, metformin intervention led to a significant reduction in waist circumference (p < 0.001) and waist-to-hip ratio (p = 0.019). Compared to placebo, metformin did not change percent breast density and dense breast volume but led to a numerical but not significant decrease in non-dense breast volume (p = 0.070). CONCLUSION We conclude that metformin intervention resulted in favorable changes in anthropometric measures of adiposity and a borderline decrease in non-dense breast volume in women with metabolic dysregulation. More research is needed to understand the impact of metformin on breast cancer risk reduction. TRIAL REGISTRATION ClinicalTrials.gov NCT02028221. Registered January 7, 2014, https://clinicaltrials.gov/ct2/show/NCT02028221.
Collapse
Affiliation(s)
- Edgar Tapia
- University of Arizona Cancer Center, University of Arizona, 1515 N Campbell Ave, Tucson, AZ, 85724, USA
| | | | - Pavani Chalasani
- University of Arizona Cancer Center, University of Arizona, 1515 N Campbell Ave, Tucson, AZ, 85724, USA
- Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Sara Centuori
- University of Arizona Cancer Center, University of Arizona, 1515 N Campbell Ave, Tucson, AZ, 85724, USA
- Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Denise J Roe
- University of Arizona Cancer Center, University of Arizona, 1515 N Campbell Ave, Tucson, AZ, 85724, USA
- Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ, USA
| | - Jose Guillen-Rodriguez
- University of Arizona Cancer Center, University of Arizona, 1515 N Campbell Ave, Tucson, AZ, 85724, USA
| | - Chuan Huang
- Department of Radiology, Stony Brook University, Stony Brook, NY, USA
| | - Jean-Phillippe Galons
- University of Arizona Cancer Center, University of Arizona, 1515 N Campbell Ave, Tucson, AZ, 85724, USA
- Department of Medical Imaging, University of Arizona, Tucson, AZ, USA
| | - Cynthia A Thomson
- University of Arizona Cancer Center, University of Arizona, 1515 N Campbell Ave, Tucson, AZ, 85724, USA
- Department of Health Promotion Sciences, University of Arizona, Tucson, AZ, USA
| | - Maria Altbach
- University of Arizona Cancer Center, University of Arizona, 1515 N Campbell Ave, Tucson, AZ, 85724, USA
- Department of Medical Imaging, University of Arizona, Tucson, AZ, USA
| | - Jesse Trujillo
- University of Arizona Cancer Center, University of Arizona, 1515 N Campbell Ave, Tucson, AZ, 85724, USA
| | - Liane Pinto
- University of Arizona Cancer Center, University of Arizona, 1515 N Campbell Ave, Tucson, AZ, 85724, USA
| | - Jessica A Martinez
- University of Arizona Cancer Center, University of Arizona, 1515 N Campbell Ave, Tucson, AZ, 85724, USA
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA
| | - Amit M Algotar
- University of Arizona Cancer Center, University of Arizona, 1515 N Campbell Ave, Tucson, AZ, 85724, USA
- Department of Family and Community Medicine, University of Arizona, Tucson, AZ, USA
| | - H-H Sherry Chow
- University of Arizona Cancer Center, University of Arizona, 1515 N Campbell Ave, Tucson, AZ, 85724, USA.
- Department of Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
47
|
Cho BA, Iyengar NM, Zhou XK, Morrow M, Giri DD, Verma A, Elemento O, Pollak M, Dannenberg AJ. Blood biomarkers reflect the effects of obesity and inflammation on the human breast transcriptome. Carcinogenesis 2021; 42:1281-1292. [PMID: 34314488 PMCID: PMC8546933 DOI: 10.1093/carcin/bgab066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/15/2021] [Accepted: 07/26/2021] [Indexed: 11/14/2022] Open
Abstract
Obesity is a risk factor for the development of post-menopausal breast cancer. Breast white adipose tissue (WAT) inflammation, which is commonly found in women with excess body fat, is also associated with increased breast cancer risk. Both local and systemic effects are probably important for explaining the link between excess body fat, adipose inflammation and breast cancer. The first goal of this cross-sectional study of 196 women was to carry out transcriptome profiling to define the molecular changes that occur in the breast related to excess body fat and WAT inflammation. A second objective was to determine if commonly measured blood biomarkers of risk and prognosis reflect molecular changes in the breast. Breast WAT inflammation was assessed by immunohistochemistry. Bulk RNA-sequencing was carried out to assess gene expression in non-tumorous breast. Obesity and WAT inflammation were associated with a large number of differentially expressed genes and changes in multiple pathways linked to the development and progression of breast cancer. Altered pathways included inflammatory response, complement, KRAS signaling, tumor necrosis factor α signaling via NFkB, interleukin (IL)6-JAK-STAT3 signaling, epithelial mesenchymal transition, angiogenesis, interferon γ response and transforming growth factor (TGF)-β signaling. Increased expression of several drug targets such as aromatase, TGF-β1, IDO-1 and PD-1 were observed. Levels of various blood biomarkers including high sensitivity C-reactive protein, IL6, leptin, adiponectin, triglycerides, high-density lipoprotein cholesterol and insulin were altered and correlated with molecular changes in the breast. Collectively, this study helps to explain both the link between obesity and breast cancer and the utility of blood biomarkers for determining risk and prognosis.
Collapse
Affiliation(s)
- Byuri Angela Cho
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Neil M Iyengar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xi Kathy Zhou
- Department of Population Health Sciences, Weill Cornell Medical College, New York, NY, USA
| | - Monica Morrow
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dilip D Giri
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Akanksha Verma
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Michael Pollak
- Department of Medicine and Oncology, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
48
|
Olatunde A, Nigam M, Singh RK, Panwar AS, Lasisi A, Alhumaydhi FA, Jyoti Kumar V, Mishra AP, Sharifi-Rad J. Cancer and diabetes: the interlinking metabolic pathways and repurposing actions of antidiabetic drugs. Cancer Cell Int 2021; 21:499. [PMID: 34535145 PMCID: PMC8447515 DOI: 10.1186/s12935-021-02202-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/06/2021] [Indexed: 12/18/2022] Open
Abstract
Cancers are regarded as one of the main causes of death and result in high health burden worldwide. The management of cancer include chemotherapy, surgery and radiotherapy. The chemotherapy, which involves the use of chemical agents with cytotoxic actions is utilised as a single treatment or combined treatment. However, these managements of cancer such as chemotherapy poses some setbacks such as cytotoxicity on normal cells and the problem of anticancer drug resistance. Therefore, the use of other therapeutic agents such as antidiabetic drugs is one of the alternative interventions used in addressing some of the limitations in the use of anticancer agents. Antidiabetic drugs such as sulfonylureas, biguanides and thiazolidinediones showed beneficial and repurposing actions in the management of cancer, thus, the activities of these drugs against cancer is attributed to some of the metabolic links between the two disorders and these includes hyperglycaemia, hyperinsulinemia, inflammation, and oxidative stress as well as obesity. Furthermore, some studies showed that the use of antidiabetic drugs could serve as risk factors for the development of cancerous cells particularly pancreatic cancer. However, the beneficial role of these chemical agents overweighs their detrimental actions in cancer management. Hence, the present review indicates the metabolic links between cancer and diabetes and the mechanistic actions of antidiabetic drugs in the management of cancers.
Collapse
Affiliation(s)
- Ahmed Olatunde
- Department of Biochemistry, Abubakar Tafawa Balewa University, Bauchi, 740272, Nigeria
| | - Manisha Nigam
- Department of Biochemistry, School of Life Sciences, Hemvati Nandan Bahuguna Garhwal University, Srinagar, Garhwal, Uttarakhand, 246174, India.
| | - Rahul Kunwar Singh
- Department of Microbiology, School of Life Sciences, Hemvati Nandan Bahuguna Garhwal University, Srinagar, Garhwal, Uttarakhand, 246174, India
| | - Abhaya Shikhar Panwar
- Department of Biochemistry, School of Life Sciences, Hemvati Nandan Bahuguna Garhwal University, Srinagar, Garhwal, Uttarakhand, 246174, India
| | - Abdulwahab Lasisi
- Maidstone and Tunbridge Wells NHS Trust, Hermitage Lane, Maidstone, Kent, ME169QQ, UK
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Vijay Jyoti Kumar
- Department of Pharmaceutical Sciences, Hemvati Nandan Bahuguna Garhwal University, Garhwal, Srinagar, Uttarakhand, 246174, India
| | - Abhay Prakash Mishra
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Science, University of Free State, 205, Nelson Mandela Drive, Park West, Bloemfontein, 9300, South Africa
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
49
|
Hao Y, Jiang M, Miao Y, Li X, Hou C, Zhang X, Chen H, Zhong X, Li J. Effect of long-term weight gain on the risk of breast cancer across women's whole adulthood as well as hormone-changed menopause stages: A systematic review and dose-response meta-analysis. Obes Res Clin Pract 2021; 15:439-448. [PMID: 34456166 DOI: 10.1016/j.orcp.2021.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 02/05/2023]
Abstract
Adult weight gain is a good indicator of excess body fatness for breast cancer risk. However, little is known about the effect of weight gain during other special periods in women's lifetime. A publication search in PubMed and Embase through April 2020 was conducted. A primary meta-analysis comparing the highest and lowest category and a secondary meta-analysis based on dose-response meta-analysis were performed to calculate risk estimates with 95% confidence intervals using a random-effects model. For postmenopausal breast cancer, the relative risk for highest vs. lowest category of adult weight gain and weight gain since menopause were 1.55 and 1.59 (RR = 1.55, 95% CI: 1.40, 1.71; RR = 1.59, 95% CI: 1.23, 2.05). For per 5 kg increase in adult weight gain, the summary RR of postmenopausal breast cancer was 1.08 (RR = 1.08, 95% CI: 1.07, 1.09), which is much stronger in Asian women (RR = 1.34, 95% CI: 1.22, 1.47). There was no significant finding among premenopausal women (RR = 1.00, 95% CI: 0.83, 1.21). Same as adult weight gain, weight gain since menopause might be an equivalent predictor for postmenopausal breast cancer risk. More studies are warranted to confirm the magnitude of this association further.
Collapse
Affiliation(s)
- Yu Hao
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Non-communicable Diseases Research Center, West China-PUMC C.C. Chen Institute of Health, Sichuan Univetsity, Chengdu, Sichuan, China
| | - Menglu Jiang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Non-communicable Diseases Research Center, West China-PUMC C.C. Chen Institute of Health, Sichuan Univetsity, Chengdu, Sichuan, China
| | - Yunqi Miao
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Non-communicable Diseases Research Center, West China-PUMC C.C. Chen Institute of Health, Sichuan Univetsity, Chengdu, Sichuan, China
| | - Xu Li
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Non-communicable Diseases Research Center, West China-PUMC C.C. Chen Institute of Health, Sichuan Univetsity, Chengdu, Sichuan, China
| | - Can Hou
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Non-communicable Diseases Research Center, West China-PUMC C.C. Chen Institute of Health, Sichuan Univetsity, Chengdu, Sichuan, China
| | - Xiaofan Zhang
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Non-communicable Diseases Research Center, West China-PUMC C.C. Chen Institute of Health, Sichuan Univetsity, Chengdu, Sichuan, China
| | - Hui Chen
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Non-communicable Diseases Research Center, West China-PUMC C.C. Chen Institute of Health, Sichuan Univetsity, Chengdu, Sichuan, China
| | - Xiaorong Zhong
- Department of Head, Neck and Mammary Gland Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiayuan Li
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Non-communicable Diseases Research Center, West China-PUMC C.C. Chen Institute of Health, Sichuan Univetsity, Chengdu, Sichuan, China.
| |
Collapse
|
50
|
Rubinstein MM, Brown KA, Iyengar NM. Targeting obesity-related dysfunction in hormonally driven cancers. Br J Cancer 2021; 125:495-509. [PMID: 33911195 PMCID: PMC8368182 DOI: 10.1038/s41416-021-01393-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 03/05/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity is a risk factor for at least 13 different types of cancer, many of which are hormonally driven, and is associated with increased cancer incidence and morbidity. Adult obesity rates are steadily increasing and a subsequent increase in cancer burden is anticipated. Obesity-related dysfunction can contribute to cancer pathogenesis and treatment resistance through various mechanisms, including those mediated by insulin, leptin, adipokine, and aromatase signalling pathways, particularly in women. Furthermore, adiposity-related changes can influence tumour vascularity and inflammation in the tumour microenvironment, which can support tumour development and growth. Trials investigating non-pharmacological approaches to target the mechanisms driving obesity-mediated cancer pathogenesis are emerging and are necessary to better appreciate the interplay between malignancy, adiposity, diet and exercise. Diet, exercise and bariatric surgery are potential strategies to reverse the cancer-promoting effects of obesity; trials of these interventions should be conducted in a scientifically rigorous manner with dose escalation and appropriate selection of tumour phenotypes and have cancer-related clinical and mechanistic endpoints. We are only beginning to understand the mechanisms by which obesity effects cell signalling and systemic factors that contribute to oncogenesis. As the rates of obesity and cancer increase, we must promote the development of non-pharmacological lifestyle trials for the treatment and prevention of malignancy.
Collapse
Affiliation(s)
- Maria M. Rubinstein
- grid.51462.340000 0001 2171 9952Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Kristy A. Brown
- grid.5386.8000000041936877XDepartment of Biochemistry in Medicine, Weill Cornell Medical College, New York, NY USA
| | - Neil M. Iyengar
- grid.51462.340000 0001 2171 9952Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY USA
| |
Collapse
|