1
|
Pierro EW, Cottam MA, An H, Lehmann BD, Pietenpol JA, Wellen KE, Makowski L, Rathmell JC, Fingleton B, Hasty AH. Comparison of Lean, Obese, and Weight-Loss Models Reveals TREM2 Deficiency Attenuates Breast Cancer Growth Uniquely in Lean Mice and Alters Clonal T-cell Populations. Cancer Res 2025; 85:1219-1235. [PMID: 39841585 PMCID: PMC11968228 DOI: 10.1158/0008-5472.can-24-3511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/19/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
Obesity is an established risk factor for breast cancer development and poor prognosis. The adipose environment surrounding breast tumors, which is inflamed in obesity, has been implicated in tumor progression, and triggering receptor expressed on myeloid cells 2 (TREM2), a transmembrane receptor expressed on macrophages in adipose tissue and tumors, is an emerging therapeutic target for cancer. A better understanding of the mechanisms for the obesity-breast cancer association and the potential benefits of weight loss could help inform treatment strategies. In this study, we utilized lean, obese, and weight-loss mouse models to examine the impact of TREM2 deficiency on postmenopausal breast cancer depending on weight history conditions. Trem2 deficiency constrained tumor growth in lean, but not in obese or weight-loss, mice. Single-cell RNA sequencing, in conjunction with variable-diversity-joining sequencing, of tumor and tumor-adjacent mammary adipose tissue immune cells revealed differences in the immune landscapes across the different models. Tumors of lean TREM2-deficient mice exhibited a shift in clonal CD8+ T cells from an exhausted to an effector memory state, accompanied by increased clonality of CD4+ Th1 cells, that was not observed in any other diet-genotype group. Notably, identical T-cell clonotypes were identified in the tumor and tumor-adjacent mammary adipose tissue of the same mouse. Finally, anti-PD-1 therapy restricted tumor growth in lean and weight-loss, but not in obese, mice. These findings indicate that weight history could affect the efficacy of TREM2 inhibition in postmenopausal breast cancer. The reported immunologic interactions between tumors and the surrounding adipose tissue highlight significant differences under obese and weight-loss conditions. Significance: Weight history impacts the immunological landscape of postmenopausal breast cancer and the efficacy of TREM2 modulation and anti-PD-1 therapy, which has implications for personalized medicine.
Collapse
Affiliation(s)
- Elysa W. Pierro
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Matthew A. Cottam
- Department of Surgery, Division of Surgical Oncology and Endocrine Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Hanbing An
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN
| | - Brian D. Lehmann
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Jennifer A. Pietenpol
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Biochemistry, Vanderbilt University, Nashville, TN
| | - Kathryn E. Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Liza Makowski
- Department of Medicine, Division of Hematology-Oncology, University of Tennessee Health Science Center, Memphis, TN, 31863, USA
| | - Jeffrey C. Rathmell
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN
| | - Barbara Fingleton
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Pharmacology, Vanderbilt University, Nashville, TN
| | - Alyssa H. Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN
- Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern, Dallas, TX
| |
Collapse
|
2
|
Malon D, Molto C, Prasla S, Cuthbert D, Pathak N, Berner-Wygoda Y, Di Lorio M, Li M, Savill J, Mittal A, Amir E, Jhaveri K, Nadler MB. Steatotic liver disease in metastatic breast cancer treated with endocrine therapy and CDK4/6 inhibitor. Breast Cancer Res Treat 2025; 210:405-416. [PMID: 39720971 DOI: 10.1007/s10549-024-07578-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/04/2024] [Indexed: 12/26/2024]
Abstract
PURPOSE In early-stage breast cancer, steatotic liver disease (SLD) is associated with increased recurrence, cardiovascular events, and non-cancer death. Endocrine therapy (ET) increases the risk of SLD. The impact of cyclin-dependent kinases 4/6 inhibitors (CDK4/6i) on SLD and prognostic association in metastatic breast cancer is unknown. We characterized the presence of SLD, risk factors, and treatment outcomes of SLD in metastatic HR+/HER2- breast cancer receiving CDK4/6i. METHODS This single institution, retrospective, cohort study included patients with metastatic HR+/HER2- breast cancer receiving first-line ET and CDK4/6i from January 2018 to June 2022. SLD was defined as a Liver Attenuation Index (LAI) > 25 HU on contrast-enhanced CT scans and/or > 10 HU on plain CT scans. Univariable binary-logistic regression was used to assess associations with SLD. Time to treatment failure (TTF) and overall survival (OS) were analyzed using Cox proportional hazards modeling. RESULTS Among 87 patients with a median age of 58 years and 65.5% postmenopausal, 50 (57.5%) had SLD at anytime (24 at baseline, 26 acquired). SLD at baseline was statistically associated with post-menopausal status. It was quantitatively but not statistically associated with age > 65, diabetes, smoking, and HER2-low. SLD at anytime was statistically significantly associated with longer TTF (median 470 vs 830.5 days, HR = 0.38, p < 0.001). No significant differences in OS or grade 3/4 adverse events were observed between groups. CONCLUSION This study demonstrated a high prevalence of SLD in this population, with SLD presence correlated with longer TTF. SLD may be an indicator of better outcomes in metastatic HR+/HER2- breast cancer patients treated with CDK4/6i.
Collapse
Affiliation(s)
- Diego Malon
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre University Health Network, 700 University Ave, Toronto, ON, M5G 1Z5, Canada.
- University of Toronto, Toronto, ON, Canada.
| | - Consolacion Molto
- Department of Oncology, Queen's University, Kingston, ON, Canada
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, ON, Canada
- R.S. McLaughlin Durham Regional Cancer Centre, Oshawa, ON, Canada
| | - Shopnil Prasla
- Joint Department of Medical Imaging (JDMI), University Health Network, Toronto, ON, Canada
| | - Danielle Cuthbert
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre University Health Network, 700 University Ave, Toronto, ON, M5G 1Z5, Canada
- University of Toronto, Toronto, ON, Canada
| | - Neha Pathak
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre University Health Network, 700 University Ave, Toronto, ON, M5G 1Z5, Canada
- University of Toronto, Toronto, ON, Canada
| | - Yael Berner-Wygoda
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre University Health Network, 700 University Ave, Toronto, ON, M5G 1Z5, Canada
- University of Toronto, Toronto, ON, Canada
| | - Massimo Di Lorio
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre University Health Network, 700 University Ave, Toronto, ON, M5G 1Z5, Canada
- University of Toronto, Toronto, ON, Canada
| | - Meredith Li
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre University Health Network, 700 University Ave, Toronto, ON, M5G 1Z5, Canada
- University of Toronto, Toronto, ON, Canada
| | - Jacqueline Savill
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre University Health Network, 700 University Ave, Toronto, ON, M5G 1Z5, Canada
- University of Toronto, Toronto, ON, Canada
| | - Abhenil Mittal
- Department of Oncology, Health Sciences North, Sudbury, ON, Canada
- Division of Clinical Sciences, The Northern Ontario School of Medicine (NOSMU), Sudbury, Canada
| | - Eitan Amir
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre University Health Network, 700 University Ave, Toronto, ON, M5G 1Z5, Canada
- University of Toronto, Toronto, ON, Canada
| | - Kartik Jhaveri
- Joint Department of Medical Imaging (JDMI), University Health Network, Toronto, ON, Canada
| | - Michelle B Nadler
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre University Health Network, 700 University Ave, Toronto, ON, M5G 1Z5, Canada
- University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Din ZU, Cui B, Wang C, Zhang X, Mehmood A, Peng F, Liu Q. Crosstalk between lipid metabolism and EMT: emerging mechanisms and cancer therapy. Mol Cell Biochem 2025; 480:103-118. [PMID: 38622439 DOI: 10.1007/s11010-024-04995-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/19/2024] [Indexed: 04/17/2024]
Abstract
Lipids are the key component of all membranes composed of a variety of molecules that transduce intracellular signaling and provide energy to the cells in the absence of nutrients. Alteration in lipid metabolism is a major factor for cancer heterogeneity and a newly identified cancer hallmark. Reprogramming of lipid metabolism affects the diverse cancer phenotypes, especially epithelial-mesenchymal transition (EMT). EMT activation is considered to be an essential step for tumor metastasis, which exhibits a crucial role in the biological processes including development, wound healing, and stem cell maintenance, and has been widely reported to contribute pathologically to cancer progression. Altered lipid metabolism triggers EMT and activates multiple EMT-associated oncogenic pathways. Although the role of lipid metabolism-induced EMT in tumorigenesis is an attractive field of research, there are still significant gaps in understanding the underlying mechanisms and the precise contributions of this interplay. Further study is needed to clarify the specific molecular mechanisms driving the crosstalk between lipid metabolism and EMT, as well as to determine the potential therapeutic implications. The increased dependency of tumor cells on lipid metabolism represents a novel therapeutic target, and targeting altered lipid metabolism holds promise as a strategy to suppress EMT and ultimately inhibit metastasis.
Collapse
Affiliation(s)
- Zaheer Ud Din
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, Guangdong Medical University, Dongguan, China
| | - Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Cenxin Wang
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
| | - Xiaoyu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
| | - Arshad Mehmood
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Fei Peng
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China.
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China.
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
4
|
Pierro EW, Cottam MA, An H, Lehmann BD, Pietenpol JA, Wellen KE, Makowski L, Rathmell JC, Fingleton B, Hasty AH. Trem2 deficiency attenuates breast cancer tumor growth in lean, but not obese or weight loss, mice and is associated with alterations of clonal T cell populations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614811. [PMID: 39386686 PMCID: PMC11463595 DOI: 10.1101/2024.09.25.614811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Obesity is an established risk factor for breast cancer development and worsened prognosis; however, the mechanisms for this association - and the potential benefits of weight loss - have not been fully explored. The adipose environment surrounding breast tumors, which is inflamed in obesity, has been implicated in tumor progression. An emerging therapeutic target for cancer is TREM2, a transmembrane receptor of the immunoglobulin superfamily that is expressed on macrophages in adipose tissue and tumors. We utilized genetic loss of function (Trem2 +,+ and Trem2 -/-) models and dietary (lean, obese, and weight loss) intervention approaches to examine impacts on postmenopausal breast cancer. Remarkably, Trem2 deficiency ameliorated tumor growth in lean, but not obese or weight loss mice. Single-cell RNA sequencing, in conjunction with VDJ sequencing of tumor and tumor-adjacent mammary adipose tissue (mATTum-adj) immune cells, revealed that tumors of lean Trem2 -/- mice exhibited a shift in clonal CD8+ T cells from an exhausted to an effector memory state, accompanied with increased clonality of CD4+ Th1 cells, that was not observed in any other diet-genotype group. Notably, identical T cell clonotypes were identified in the tumor and mATTum-adj of the same mouse. Finally, an immune checkpoint study demonstrated that αPD-1 therapy restricted tumor growth in lean and weight loss, but not obese mice. We conclude that weight history is relevant when considering potential efficacy of TREM2 inhibition in postmenopausal breast cancer. This work reveals immunological interactions between tumors and surrounding adipose tissue, highlighting significant differences under obese and weight loss conditions.
Collapse
Affiliation(s)
- Elysa W. Pierro
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Matthew A. Cottam
- Department of Surgery, Division of Surgical Oncology and Endocrine Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Hanbing An
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN
| | - Brian D. Lehmann
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Jennifer A. Pietenpol
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Biochemistry, Vanderbilt University, Nashville, TN
| | - Kathryn E. Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Liza Makowski
- Department of Medicine, Division of Hematology-Oncology, University of Tennessee Health Science Center, Memphis, TN, 31863, USA
| | - Jeffrey C. Rathmell
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN
| | - Barbara Fingleton
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Pharmacology, Vanderbilt University, Nashville, TN
| | - Alyssa H. Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN
- Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern, Dallas, TX
| |
Collapse
|
5
|
Gonzalez-Pons R, Bernard JJ. Benzo[a]pyrene exposure prevents high fat diet-induced obesity in the 4T1 model of mammary carcinoma. Front Oncol 2024; 14:1394039. [PMID: 39301545 PMCID: PMC11410564 DOI: 10.3389/fonc.2024.1394039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024] Open
Abstract
Tumor metastasis is the main cause of death in triple-negative breast cancer (TNBC) patients. TNBC is the most aggressive subtype of breast cancer lacking the expression of estrogen, progesterone, and human epidermal growth factor 2 receptors, thereby rendering it insensitive to targeted therapies. It has been well-established that excess adiposity contributes to the progression of TNBC; however, due to the aggressive nature of this breast cancer subtype, it is imperative to determine how multiple factors can contribute to progression. Therefore, we aimed to investigate if exposure to an environmental carcinogen could impact a pre-existing obesity-promoted cancer. We utilized a spontaneous lung metastatic mouse model where 4T1 breast tumor cells are injected into the mammary gland of BALB/c mice. Feeding a high fat diet (HFD) in this model has been shown to promote tumor growth and metastasis. Herein, we tested the effects of both a HFD and benzo(a)pyrene (B[a]P) exposure. Our results indicate that diet and B[a]P had no tumor promotional interaction. However, unexpectedly, our findings reveal an inhibitory effect of B[a]P on body weight, adipose tissue deposition, and tumor volume at time of sacrifice specifically under HFD conditions.
Collapse
Affiliation(s)
- Romina Gonzalez-Pons
- Department of Pharmacology and Toxicology, Michigan State University,
East Lansing, MI, United States
| | - Jamie J. Bernard
- Department of Pharmacology and Toxicology, Michigan State University,
East Lansing, MI, United States
- Department of Medicine, Michigan State University, East
Lansing, MI, United States
| |
Collapse
|
6
|
Conner SJ, Borges HB, Guarin JR, Gerton TJ, Yui A, Salhany KJ, Mensah DN, Hamilton GA, Le GH, Lew KC, Zhang C, Oudin MJ. Obesity Induces Temporally Regulated Alterations in the Extracellular Matrix That Drive Breast Tumor Invasion and Metastasis. Cancer Res 2024; 84:2761-2775. [PMID: 38900938 PMCID: PMC11908800 DOI: 10.1158/0008-5472.can-23-2526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 04/16/2024] [Accepted: 06/14/2024] [Indexed: 06/22/2024]
Abstract
Obesity is associated with increased incidence and metastasis of triple-negative breast cancer, an aggressive breast cancer subtype. The extracellular matrix (ECM) is a major component of the tumor microenvironment that drives metastasis. To characterize the temporal effects of age and high-fat diet (HFD)-driven weight gain on the ECM, we injected allograft tumor cells at 4-week intervals into mammary fat pads of mice fed a control or HFD, assessing tumor growth and metastasis and evaluating the ECM composition of the mammary fat pads, lungs, and livers. Tumor growth was increased in obese mice after 12 weeks on HFD. Liver metastasis increased in obese mice only at 4 weeks, and elevated body weight correlated with increased metastasis to the lungs but not the liver. Whole decellularized ECM coupled with proteomics indicated that early stages of obesity were sufficient to induce changes in the ECM composition. Obesity led to an increased abundance of the proinvasive ECM proteins collagen IV and collagen VI in the mammary glands and enhanced the invasive capacity of cancer cells. Cells of stromal vascular fraction and adipose stem and progenitor cells were primarily responsible for secreting collagen IV and collagen VI, not adipocytes. Longer exposure to HFD increased the invasive potential of ECM isolated from the lungs and liver, with significant changes in ECM composition found in the liver with short-term HFD exposure. Together, these data suggest that changes in the breast, lungs, and liver ECM underlie some of the effects of obesity on triple-negative breast cancer incidence and metastasis. Significance: Organ-specific extracellular matrix changes in the primary tumor and metastatic microenvironment are mechanisms by which obesity contributes to breast cancer progression.
Collapse
Affiliation(s)
- Sydney J Conner
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Hannah B Borges
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Justinne R Guarin
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Thomas J Gerton
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Anna Yui
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Kenneth J Salhany
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Diamond N Mensah
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Grace A Hamilton
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Giang H Le
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Katherine C Lew
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Crystal Zhang
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Madeleine J Oudin
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|
7
|
Yücel KB, Aydos U, Sütcüoglu O, Kılıç ACK, Özdemir N, Özet A, Yazıcı O. Visceral obesity and sarcopenia as predictors of efficacy and hematological toxicity in patients with metastatic breast cancer treated with CDK 4/6 inhibitors. Cancer Chemother Pharmacol 2024; 93:497-507. [PMID: 38436714 DOI: 10.1007/s00280-024-04641-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024]
Abstract
PURPOSE We aimed to investigate whether visceral adipose tissue (VAT), subcutaneous adipose tissue (SAT), and skeletal muscle area (SMA) index are predictive for efficacy and hematological toxicity in ER + HER2-metastatic breast cancer (BC) patients who received CDK 4/6 inhibitors. METHODS This retrospective cohort study analyzed 52 patients who were treated with CDK 4/6 inhibitors between January 2018 and February 2021. The values of VAT, SAT, SMA indices and hematological parameters were noted before the start, at the third and sixth months of this treatment. The skeletal muscle area (SMA) and adipose tissue measurements were calculated at the level of the third lumbar vertebra. A SMA-index value of <40 cm2/m2 was accepted as the threshold value for sarcopenia. RESULTS Patients with sarcopenia had a worse progression-free survival (PFS) compared to patients without sarcopenia (19.6 vs. 9.0 months, p = 0.005). Patients with a high-VAT-index had a better PFS (20.4 vs. 9.3 months, p = 0.033). Only the baseline low-SMA- index (HR: 3.89; 95% CI: 1.35-11.25, p = 0.012) and baseline low-VAT-index (HR: 2.15; 95% CI: 1.02-4.53, p = 0.042) had significantly related to poor PFS in univariate analyses. The low-SMA-index was the only independent factor associated with poor PFS (HR: 3.99; 95% CI: 1.38-11.54, p = 0.011). No relationship was observed between body composition parameters and grade 3-4 hematological toxicity. CONCLUSION The present study supported the significance of sarcopenia and low visceral adipose tissue as potential early indicators of poor PFS in patients treated with CDK 4/6 inhibitors.
Collapse
Affiliation(s)
| | - Uguray Aydos
- Department of Nuclear Medicine, Gazi University, Ankara, Turkey
| | - Osman Sütcüoglu
- Department of Medical Oncology, Gazİ University, Ankara, Turkey
| | | | - Nuriye Özdemir
- Department of Medical Oncology, Gazİ University, Ankara, Turkey
| | - Ahmet Özet
- Department of Medical Oncology, Gazİ University, Ankara, Turkey
| | - Ozan Yazıcı
- Department of Medical Oncology, Gazİ University, Ankara, Turkey
| |
Collapse
|
8
|
Lagarde CB, Kavalakatt J, Benz MC, Hawes ML, Arbogast CA, Cullen NM, McConnell EC, Rinderle C, Hebert KL, Khosla M, Belgodere JA, Hoang VT, Collins-Burow BM, Bunnell BA, Burow ME, Alahari SK. Obesity-associated epigenetic alterations and the obesity-breast cancer axis. Oncogene 2024; 43:763-775. [PMID: 38310162 DOI: 10.1038/s41388-024-02954-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024]
Abstract
Both breast cancer and obesity can regulate epigenetic changes or be regulated by epigenetic changes. Due to the well-established link between obesity and an increased risk of developing breast cancer, understanding how obesity-mediated epigenetic changes affect breast cancer pathogenesis is critical. Researchers have described how obesity and breast cancer modulate the epigenome individually and synergistically. In this review, the epigenetic alterations that occur in obesity, including DNA methylation, histone, and chromatin modification, accelerated epigenetic age, carcinogenesis, metastasis, and tumor microenvironment modulation, are discussed. Delineating the relationship between obesity and epigenetic regulation is vital to furthering our understanding of breast cancer pathogenesis.
Collapse
Affiliation(s)
- Courtney B Lagarde
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Joachim Kavalakatt
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Megan C Benz
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Mackenzie L Hawes
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Carter A Arbogast
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Nicole M Cullen
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Emily C McConnell
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Caroline Rinderle
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Katherine L Hebert
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Maninder Khosla
- Department of Biochemistry and Molecular Biology, LSU Health Science Center School of Medicine, New Orleans, LA, 70112, USA
| | - Jorge A Belgodere
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- Department of Biological and Agricultural Engineering, Louisiana State University and Agricultural Center, Baton Rouge, LA, 70803, USA
| | - Van T Hoang
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Bridgette M Collins-Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Bruce A Bunnell
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Matthew E Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, LSU Health Science Center School of Medicine, New Orleans, LA, 70112, USA.
- Stanley S. Scott Cancer Center, LSU Health Science Center School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
9
|
Conner SJ, Guarin JR, Borges HB, Salhany KJ, Mensah DN, Hamilton GA, Le GH, Oudin MJ. Age and obesity-driven changes in the extracellular matrix of the primary tumor and metastatic site influence tumor invasion and metastatic outgrowth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554492. [PMID: 37662270 PMCID: PMC10473680 DOI: 10.1101/2023.08.24.554492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Younger age and obesity increase the incidence and metastasis of triple-negative breast cancer (TNBC), an aggressive subtype of breast cancer. The extracellular matrix (ECM) promotes tumor invasion and metastasis. We characterized the effect of age and obesity on the ECM of mammary fat pads, lungs, and liver using a diet-induced obesity (DIO) model. At 4 week intervals, we either injected the mammary fat pads with allograft tumor cells to characterize tumor growth and metastasis or isolated the mammary fat pads and livers to characterize the ECM. Age had no effect on tumor growth but increased lung and liver metastasis after 16 weeks. Obesity increased tumor growth starting at 12 weeks, increased liver metastasis only at 4 weeks, and weight gain correlated to increased lung but not liver metastasis. Utilizing whole decellularized ECM coupled with proteomics, we found that early stages of obesity were sufficient to induce changes in the ECM composition and invasive potential of mammary fat pads with increased abundance of pro-invasive ECM proteins Collagen IV and Collagen VI. We identified cells of stromal vascular fraction and adipose stem and progenitor cells as primarily responsible for secreting Collagen IV and VI, not adipocytes. We characterized the changes in ECM in the lungs and liver, and determined that older age decreases the metastatic potential of lung and liver ECM while later-stage obesity increases the metastatic potential. These data implicate ECM changes in the primary tumor and metastatic microenvironment as mechanisms by which age and obesity contribute to breast cancer progression.
Collapse
Affiliation(s)
- Sydney J. Conner
- Department of Biomedical Engineering, Tufts University, Medford, MA 02478
| | - Justinne R. Guarin
- Department of Biomedical Engineering, Tufts University, Medford, MA 02478
| | - Hannah B. Borges
- Department of Biomedical Engineering, Tufts University, Medford, MA 02478
| | - Kenneth J. Salhany
- Department of Biomedical Engineering, Tufts University, Medford, MA 02478
| | - Diamond N. Mensah
- Department of Biomedical Engineering, Tufts University, Medford, MA 02478
| | - Grace A. Hamilton
- Department of Biomedical Engineering, Tufts University, Medford, MA 02478
| | - Giang H. Le
- Department of Biomedical Engineering, Tufts University, Medford, MA 02478
| | - Madeleine J. Oudin
- Department of Biomedical Engineering, Tufts University, Medford, MA 02478
| |
Collapse
|
10
|
McDowell SA, Milette S, Doré S, Yu MW, Sorin M, Wilson L, Desharnais L, Cristea A, Varol O, Atallah A, Swaby A, Breton V, Arabzadeh A, Petrecca S, Loucif H, Bhagrath A, De Meo M, Lach KD, Issac MS, Fiset B, Rayes RF, Mandl JN, Fritz JH, Fiset PO, Holt PR, Dannenberg AJ, Spicer JD, Walsh LA, Quail DF. Obesity alters monocyte developmental trajectories to enhance metastasis. J Exp Med 2023; 220:e20220509. [PMID: 37166450 PMCID: PMC10182775 DOI: 10.1084/jem.20220509] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 02/27/2023] [Accepted: 04/19/2023] [Indexed: 05/12/2023] Open
Abstract
Obesity is characterized by chronic systemic inflammation and enhances cancer metastasis and mortality. Obesity promotes breast cancer metastasis to lung in a neutrophil-dependent manner; however, the upstream regulatory mechanisms of this process remain unknown. Here, we show that obesity-induced monocytes underlie neutrophil activation and breast cancer lung metastasis. Using mass cytometry, obesity favors the expansion of myeloid lineages while restricting lymphoid cells within the peripheral blood. RNA sequencing and flow cytometry revealed that obesity-associated monocytes resemble professional antigen-presenting cells due to a shift in their development and exhibit enhanced MHCII expression and CXCL2 production. Monocyte induction of the CXCL2-CXCR2 axis underlies neutrophil activation and release of neutrophil extracellular traps to promote metastasis, and enhancement of this signaling axis is observed in lung metastases from obese cancer patients. Our findings provide mechanistic insight into the relationship between obesity and cancer by broadening our understanding of the interactive role that myeloid cells play in this process.
Collapse
Affiliation(s)
- Sheri A.C. McDowell
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Physiology, McGill University, Montreal, Canada
| | - Simon Milette
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Samuel Doré
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
| | - Miranda W. Yu
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Physiology, McGill University, Montreal, Canada
| | - Mark Sorin
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
| | - Liam Wilson
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Physiology, McGill University, Montreal, Canada
| | - Lysanne Desharnais
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
| | - Alyssa Cristea
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Ozgun Varol
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Aline Atallah
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Anikka Swaby
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Valérie Breton
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
| | | | - Sarah Petrecca
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Hamza Loucif
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- McGill University Research Centre on Complex Traits, Montreal, Canada
| | - Aanya Bhagrath
- Department of Physiology, McGill University, Montreal, Canada
- McGill University Research Centre on Complex Traits, Montreal, Canada
| | - Meghan De Meo
- Department of Experimental Surgery, McGill University, Montreal, Canada
| | - Katherine D. Lach
- Department of Pathology, Faculty of Medicine, McGill University, Montreal, Canada
| | - Marianne S.M. Issac
- Department of Pathology, Faculty of Medicine, McGill University, Montreal, Canada
| | - Benoit Fiset
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
| | - Roni F. Rayes
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
| | - Judith N. Mandl
- Department of Physiology, McGill University, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- McGill University Research Centre on Complex Traits, Montreal, Canada
| | - Jörg H. Fritz
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- McGill University Research Centre on Complex Traits, Montreal, Canada
| | - Pierre O. Fiset
- Department of Pathology, Faculty of Medicine, McGill University, Montreal, Canada
| | - Peter R. Holt
- Laboratory of Biochemical Genetics and Metabolism, Rockefeller University, New Nork, NY, USA
| | - Andrew J. Dannenberg
- Department of Medicine (retired), Weill Cornell Medical College, New York, NY, USA
| | - Jonathan D. Spicer
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Canada
- Department of Surgery, McGill University Health Centre, Montreal, Canada
| | - Logan A. Walsh
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
| | - Daniela F. Quail
- Rosalind and Morris Goodman Cancer Institute, Montreal, Canada
- Department of Physiology, McGill University, Montreal, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Canada
| |
Collapse
|
11
|
Rachman A, Fiantoro ZH, Sutandyo N, Priantono D, Romadhon PZ, Jonlean R. Metabolic Profile and Negatively Association Between Insulin Resistance and Metastatic Incidence in Indonesian Primary Invasive Breast Cancer: A Cross-Sectional Study. Int J Gen Med 2023; 16:3257-3265. [PMID: 37546243 PMCID: PMC10404037 DOI: 10.2147/ijgm.s421558] [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: 05/25/2023] [Accepted: 07/21/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction Metastatic breast cancer was associated with high morbidity and mortality. Insulin resistance was hypothesized to be related to the incidence of advanced breast cancer. Homeostatic Model Assessment of Insulin Resistance (HOMA-IR) and Triglyceride/Glucose Index (TyG Index) are two metrics used to measure the degree of insulin resistance. This study aims to assess the relationship between the incidence of metastatic breast cancer and insulin resistance as reflected by both metrics. Material and Methods This study is a cross-sectional study involving 150 primary invasive breast cancer patients recruited from two hospitals of different sectors from August 2019 to April 2020. Patients with double cancer and autoimmune disorder were excluded from this study. Data obtained from the patients include age, body mass index (BMI), type 2 diabetes mellitus (T2DM) status and treatment, and low-density lipoprotein (LDL) cholesterol. The electronic medical records (EMR) was consulted to find histopathology examination result, cancer staging, and any missing data. The association between HOMA-IR and TyG with metastatic incidence was analyzed using either the Mann-Whitney test (for non-normally distributed data) or the independent-sample t-test (for normally distributed data). Results The mean of the TyG index is 8.60, and the median of HOMA-IR is 1.22. We found no significant correlation between both variables and the incidence of metastases. Conclusion Insulin resistance was not associated with metastatic breast cancer.
Collapse
Affiliation(s)
- Andhika Rachman
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
- Siloam MRCCC Semanggi Hospital, Jakarta, Indonesia
| | - Zaenal Hakiki Fiantoro
- Departement of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | | | - Dimas Priantono
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Pradana Zaky Romadhon
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Airlangga University, Surabaya, Indonesia
| | | |
Collapse
|
12
|
Parida S, Siddharth S, Gatla HR, Wu S, Wang G, Gabrielson K, Sears CL, Ladle BH, Sharma D. Gut colonization with an obesity-associated enteropathogenic microbe modulates the premetastatic niches to promote breast cancer lung and liver metastasis. Front Immunol 2023; 14:1194931. [PMID: 37503343 PMCID: PMC10369066 DOI: 10.3389/fimmu.2023.1194931] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/22/2023] [Indexed: 07/29/2023] Open
Abstract
Introduction Obesity, an independent risk factor for breast cancer growth and metastatic progression, is also closely intertwined with gut dysbiosis; and both obese state and dysbiosis promote each other. Enteric abundance of Bacteroides fragilis is strongly linked with obesity, and we recently discovered the presence of B. fragilis in malignant breast cancer. Given that enterotoxigenic B. fragilis or ETBF, which secretes B. fragilis toxin (BFT), has been identified as a procarcinogenic microbe in breast cancer, it is necessary to examine its impact on distant metastasis and underlying systemic and localized alterations promoting metastatic progression of breast cancer. Methods We used syngeneic mammary intraductal (MIND) model harboring gut colonization with ETBF to query distant metastasis of breast cancer cells. Alterations in the immune network and cytokines/chemokines in the tumor microenvironment and distant metastatic sites were examined using flow cytometry, immunohistochemistry, and multiplex arrays. Results ETBF infection initiates a systemic inflammation aiding in the establishment of the premetastatic niche formation in vital organs via increased proinflammatory and protumorigenic cytokines like IL17A, IL17E, IL27p28, IL17A/F, IL6, and IL10 in addition to creating a prometastatic immunosuppressive environment in the liver and lungs rich in myeloid cells, macrophages, and T regulatory cells. It induces remodeling of the tumor microenvironment via immune cell and stroma infiltration, increased vasculogenesis, and an EMT-like response, thereby encouraging early metastatic dissemination ready to colonize the conducive environment in liver and lungs of the breast tumor-bearing mice. Discussion In this study, we show that enteric ETBF infection concomitantly induces systemic inflammation, reshapes the tumor immune microenvironment, and creates conducive metastatic niches to potentiate early dissemination and seeding of metastases to liver and lung tissues in agreement with the "seed and soil hypothesis." Our results also support the ETBF-induced "parallel model" of metastasis that advocates for an early dissemination of tumor cells that form metastatic lesions independent of the primary tumor load.
Collapse
Affiliation(s)
- Sheetal Parida
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
| | - Sumit Siddharth
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
| | - Himavanth R. Gatla
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
| | - Shaoguang Wu
- Department of Oncology, Georgetown University, Baltimore, MD, United States
| | - Guannan Wang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kathleen Gabrielson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
- Johns Hopkins University School of Medicine, Molecular and Comparative Pathobiology, Baltimore, MD, United States
| | - Cynthia L. Sears
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
- Department of Oncology, Georgetown University, Baltimore, MD, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Brian H. Ladle
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Dipali Sharma
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
| |
Collapse
|
13
|
Barone I, Gelsomino L, Accattatis FM, Giordano F, Gyorffy B, Panza S, Giuliano M, Veneziani BM, Arpino G, De Angelis C, De Placido P, Bonofiglio D, Andò S, Giordano C, Catalano S. Analysis of circulating extracellular vesicle derived microRNAs in breast cancer patients with obesity: a potential role for Let-7a. J Transl Med 2023; 21:232. [PMID: 37004031 PMCID: PMC10064709 DOI: 10.1186/s12967-023-04075-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/22/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND The incidence of obesity, a known risk factor for several metabolic and chronic diseases, including numerous malignancies, has risen sharply in the world. Various clinical studies demonstrate that excessive Body Mass Index (BMI) may worsen the incidence, prognosis, and mortality rates of breast cancer. Thus, understanding the link tying up obesity and breast cancer onset and progression is critically important, as it can impact patients' survival and quality of life. Recently, circulating extracellular vesicle (EV) derived miRNAs have attracted much attention for their diagnostic, prognostic and therapeutic potential in oncology research. Although the potential role of EV-derived miRNAs in the early detection of breast cancer has been repeatedly mentioned, screening of miRNAs packaged within serum EVs has not yet been reported in patients with obesity. METHODS Circulating EVs were isolated from normal weight (NW), and overweight/obese (OW/Ob) breast cancer patients and characterized by Transmission Electron Microscopy (TEM), Nanoparticle Tracking Analysis (NTA), and protein marker expression. Evaluation of EV-associated miRNAs was conducted in a screening (RNA-seq) and a validation (qRT-PCR) cohort. Bioinformatic analysis was performed to uncover significantly enriched biological processes, molecular functions and pathways. ROC and Kaplain-Meier survival analyses were used for clinical significance. RESULTS Comparison of serum EV-derived miRNAs from NW and OW/Ob patients detected seven differentially expressed miRNAs (let-7a-5p, miR-122-5p, miR-30d-5p, miR-126-3p, miR-27b-3p, miR-4772-3p, and miR-10a-5p) in the screening cohort. GO analysis revealed the enrichment of protein phosphorylation, intracellular signal transduction, signal transduction, and vesicle-mediated transport among the top biological processes. In addition, the target genes were significantly enriched in pathways related to PI3K/Akt, growth hormones, and insulin signalings, which are all involved in obesity-related diseases and/or breast cancer progression. In the validation cohort, qRT-PCR confirmed a significant down-regulation of EV-derived let-7a in the serum of OW/Ob breast cancer patients compared to NW patients. Let-7a levels also exhibited a negative correlation with BMI values. Importantly, decreased let-7a miRNA expression was associated with higher tumor grade and poor survival in patients with breast cancer. CONCLUSION These results suggest that serum-EV derived miRNAs may reflect a differential profile in relation to a patient's BMI, which, once validated in larger cohorts of patients, could provide insights into novel specific biomarkers and innovative targets to prevent the progression of obesity-mediated breast cancer.
Collapse
Affiliation(s)
- Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy.
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy.
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Felice Maria Accattatis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Balazs Gyorffy
- Departments of Bioinformatics and Pediatrics, Semmelweis University, 1094, Budapest, Hungary
- TTK Cancer Biomarker Research Group, 1117, Budapest, Hungary
| | - Salvatore Panza
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy.
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy.
| |
Collapse
|
14
|
Chan DS, Vieira R, Abar L, Aune D, Balducci K, Cariolou M, Greenwood DC, Markozannes G, Nanu N, Becerra‐Tomás N, Giovannucci EL, Gunter MJ, Jackson AA, Kampman E, Lund V, Allen K, Brockton NT, Croker H, Katsikioti D, McGinley‐Gieser D, Mitrou P, Wiseman M, Cross AJ, Riboli E, Clinton SK, McTiernan A, Norat T, Tsilidis KK. Postdiagnosis body fatness, weight change and breast cancer prognosis: Global Cancer Update Program (CUP global) systematic literature review and meta-analysis. Int J Cancer 2023; 152:572-599. [PMID: 36279884 PMCID: PMC10092239 DOI: 10.1002/ijc.34322] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 07/29/2022] [Accepted: 09/05/2022] [Indexed: 02/01/2023]
Abstract
Previous evidence on postdiagnosis body fatness and mortality after breast cancer was graded as limited-suggestive. To evaluate the evidence on body mass index (BMI), waist circumference, waist-hip-ratio and weight change in relation to breast cancer prognosis, an updated systematic review was conducted. PubMed and Embase were searched for relevant studies published up to 31 October, 2021. Random-effects meta-analyses were conducted to estimate summary relative risks (RRs). The evidence was judged by an independent Expert Panel using pre-defined grading criteria. One randomized controlled trial and 225 observational studies were reviewed (220 publications). There was strong evidence (likelihood of causality: probable) that higher postdiagnosis BMI was associated with increased all-cause mortality (64 studies, 32 507 deaths), breast cancer-specific mortality (39 studies, 14 106 deaths) and second primary breast cancer (11 studies, 5248 events). The respective summary RRs and 95% confidence intervals per 5 kg/m2 BMI were 1.07 (1.05-1.10), 1.10 (1.06-1.14) and 1.14 (1.04-1.26), with high between-study heterogeneity (I2 = 56%, 60%, 66%), but generally consistent positive associations. Positive associations were also observed for waist circumference, waist-hip-ratio and all-cause and breast cancer-specific mortality. There was limited-suggestive evidence that postdiagnosis BMI was associated with higher risk of recurrence, nonbreast cancer deaths and cardiovascular deaths. The evidence for postdiagnosis (unexplained) weight or BMI change and all outcomes was graded as limited-no conclusion. The RCT showed potential beneficial effect of intentional weight loss on disease-free-survival, but more intervention trials and well-designed observational studies in diverse populations are needed to elucidate the impact of body composition and their changes on breast cancer outcomes.
Collapse
Affiliation(s)
- Doris S.M. Chan
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Rita Vieira
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Leila Abar
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Dagfinn Aune
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
- Department of NutritionBjørknes University CollegeOsloNorway
- Department of Endocrinology, Morbid Obesity and Preventive MedicineOslo University HospitalOsloNorway
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska InstitutetStockholmSweden
| | - Katia Balducci
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Margarita Cariolou
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Darren C. Greenwood
- Leeds Institute for Data Analytics, Faculty of Medicine and HealthUniversity of LeedsLeedsUK
| | - Georgios Markozannes
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
- Department of Hygiene and EpidemiologyUniversity of Ioannina Medical SchoolIoanninaGreece
| | - Neesha Nanu
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Nerea Becerra‐Tomás
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Edward L. Giovannucci
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Department of Nutrition, Harvard T. H. Chan School of Public HealthBostonMassachusettsUSA
| | - Marc J. Gunter
- Nutrition and Metabolism Section, International Agency for Research on CancerLyonFrance
| | - Alan A. Jackson
- Faculty of Medicine, School of Human Development and HealthUniversity of SouthamptonSouthamptonUK
- National Institute of Health Research Cancer and Nutrition CollaborationSouthamptonUK
| | - Ellen Kampman
- Division of Human Nutrition and HealthWageningen University & ResearchWageningenThe Netherlands
| | - Vivien Lund
- World Cancer Research Fund InternationalLondonUK
| | - Kate Allen
- World Cancer Research Fund InternationalLondonUK
| | | | - Helen Croker
- World Cancer Research Fund InternationalLondonUK
| | | | | | | | | | - Amanda J. Cross
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Elio Riboli
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
| | - Steven K. Clinton
- Division of Medical Oncology, The Department of Internal MedicineCollege of Medicine and Ohio State University Comprehensive Cancer Center, Ohio State UniversityColumbusOhioUSA
| | - Anne McTiernan
- Division of Public Health SciencesFred Hutchinson Cancer Research CenterSeattleWashingtonUSA
| | - Teresa Norat
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
- World Cancer Research Fund InternationalLondonUK
| | - Konstantinos K. Tsilidis
- Department of Epidemiology and BiostatisticsSchool of Public Health, Imperial College LondonLondonUK
- Department of Hygiene and EpidemiologyUniversity of Ioannina Medical SchoolIoanninaGreece
| |
Collapse
|
15
|
Liu Y, Han J, Kong T, Xiao N, Mei Q, Liu J. DriverMP enables improved identification of cancer driver genes. Gigascience 2022; 12:giad106. [PMID: 38091511 PMCID: PMC10716827 DOI: 10.1093/gigascience/giad106] [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: 07/19/2023] [Revised: 10/30/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Cancer is widely regarded as a complex disease primarily driven by genetic mutations. A critical concern and significant obstacle lies in discerning driver genes amid an extensive array of passenger genes. FINDINGS We present a new method termed DriverMP for effectively prioritizing altered genes on a cancer-type level by considering mutated gene pairs. It is designed to first apply nonsilent somatic mutation data, protein‒protein interaction network data, and differential gene expression data to prioritize mutated gene pairs, and then individual mutated genes are prioritized based on prioritized mutated gene pairs. Application of this method in 10 cancer datasets from The Cancer Genome Atlas demonstrated its great improvements over all the compared state-of-the-art methods in identifying known driver genes. Then, a comprehensive analysis demonstrated the reliability of the novel driver genes that are strongly supported by clinical experiments, disease enrichment, or biological pathway analysis. CONCLUSIONS The new method, DriverMP, which is able to identify driver genes by effectively integrating the advantages of multiple kinds of cancer data, is available at https://github.com/LiuYangyangSDU/DriverMP. In addition, we have developed a novel driver gene database for 10 cancer types and an online service that can be freely accessed without registration for users. The DriverMP method, the database of novel drivers, and the user-friendly online server are expected to contribute to new diagnostic and therapeutic opportunities for cancers.
Collapse
Affiliation(s)
- Yangyang Liu
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Jiyun Han
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Tongxin Kong
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Nannan Xiao
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Qinglin Mei
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Juntao Liu
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| |
Collapse
|
16
|
Olsson LT, Walens A, Hamilton AM, Benefield HC, Fleming JM, Carey LA, Hursting SD, Williams KP, Troester MA. Obesity and Breast Cancer Metastasis across Genomic Subtypes. Cancer Epidemiol Biomarkers Prev 2022; 31:1944-1951. [PMID: 35973227 PMCID: PMC9628732 DOI: 10.1158/1055-9965.epi-22-0013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/12/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Obese women have higher risk of aggressive breast tumors and distant metastasis. However, obesity has rarely been assessed in association with metastasis in diverse populations. METHODS In the Carolina Breast Cancer Study Phase 3 (2008-2013), waist-to-hip ratio (WHR), body mass index (BMI), and molecular subtype [PAM50 risk-of-recurrence (ROR) score] were assessed. Obesity measures were evaluated in association with metastasis within five years of diagnosis, overall and stratified by race and ROR score. Absolute risk of metastasis and risk differences between strata were calculated using the Kaplan-Meier estimator, adjusted for age, grade, stage, race, and ER status. Relative frequency of metastatic site and multiplicity were estimated in association with obesity using generalized linear models. RESULTS High-WHR was associated with higher risk of metastasis (5-year risk difference, RD, 4.3%; 95% confidence interval, 2.2-6.5). It was also associated with multiple metastases and metastases at all sites except brain. The 5-year risk of metastasis differed by race (11.2% and 6.9% in Black and non-Black, respectively) and ROR score (19.5% vs. 6.6% in high vs. low-to-intermediate ROR-PT). Non-Black women and those with low-to-intermediate ROR scores had similar risk in high- and low-WHR strata. However, among Black women and those with high ROR, risk of metastasis was elevated among high-WHR (RDBlack/non-Black = 4.6%, RDHigh/Low-Int = 3.1%). Patterns of metastasis were similar by BMI. CONCLUSIONS WHR is associated with metastatic risk, particularly among Black women and those with high-risk tumors. IMPACT Understanding how risk factors for metastasis interact may help in tailoring care plans and surveillance among patients with breast cancer.
Collapse
Affiliation(s)
- Linnea T Olsson
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Andrea Walens
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Alina M Hamilton
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Halei C Benefield
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jodie M Fleming
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC
| | - Lisa A Carey
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Stephen D Hursting
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Kevin P Williams
- Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, NC
- Co-senior authors
| | - Melissa A Troester
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Co-senior authors
| |
Collapse
|
17
|
Bohm MS, Sipe LM, Pye ME, Davis MJ, Pierre JF, Makowski L. The role of obesity and bariatric surgery-induced weight loss in breast cancer. Cancer Metastasis Rev 2022; 41:673-695. [PMID: 35870055 PMCID: PMC9470652 DOI: 10.1007/s10555-022-10050-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/06/2022] [Indexed: 02/07/2023]
Abstract
Obesity is a complex metabolic condition considered a worldwide public health crisis, and a deeper mechanistic understanding of obesity-associated diseases is urgently needed. Obesity comorbidities include many associated cancers and are estimated to account for 20% of female cancer deaths in the USA. Breast cancer, in particular, is associated with obesity and is the focus of this review. The exact causal links between obesity and breast cancer remain unclear. Still, interactions have emerged between body mass index, tumor molecular subtype, genetic background, and environmental factors that strongly suggest obesity influences the risk and progression of certain breast cancers. Supportive preclinical research uses various diet-induced obesity models to demonstrate that weight loss, via dietary interventions or changes in energy expenditure, reduces the onset or progression of breast cancers. Ongoing and future studies are now aimed at elucidating the underpinning mechanisms behind weight-loss-driven observations to improve therapy and outcomes in patients with breast cancer and reduce risk. This review aims to summarize the rapidly emerging literature on obesity and weight loss strategies with a focused discussion of bariatric surgery in both clinical and preclinical studies detailing the complex interactions between metabolism, immune response, and immunotherapy in the setting of obesity and breast cancer.
Collapse
Affiliation(s)
- Margaret S Bohm
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Laura M Sipe
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Madeline E Pye
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Matthew J Davis
- Division of Bariatric Surgery, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Joseph F Pierre
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
- Department of Nutritional Sciences, College of Agriculture and Life Science, The University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Liza Makowski
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
- College of Medicine, UTHSC Center for Cancer Research, The University of Tennessee Health Science Center, Cancer Research Building Room 322, 19 S Manassas Street, Memphis, TN, 38163, USA.
| |
Collapse
|
18
|
Sipe LM, Chaib M, Korba EB, Jo H, Lovely MC, Counts BR, Tanveer U, Holt JR, Clements JC, John NA, Daria D, Marion TN, Bohm MS, Sekhri R, Pingili AK, Teng B, Carson JA, Hayes DN, Davis MJ, Cook KL, Pierre JF, Makowski L. Response to immune checkpoint blockade improved in pre-clinical model of breast cancer after bariatric surgery. eLife 2022; 11:79143. [PMID: 35775614 PMCID: PMC9342954 DOI: 10.7554/elife.79143] [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: 03/31/2022] [Accepted: 06/26/2022] [Indexed: 11/27/2022] Open
Abstract
Bariatric surgery is a sustainable weight loss approach, including vertical sleeve gastrectomy (VSG). Obesity exacerbates tumor growth, while diet-induced weight loss impairs progression. It remains unknown how bariatric surgery-induced weight loss impacts cancer progression or alters response to therapy. Using a pre-clinical model of obesity followed by VSG or diet-induced weight loss, breast cancer progression and immune checkpoint blockade therapy were investigated. Weight loss by VSG or weight-matched dietary intervention before tumor engraftment protected against obesity-exacerbated tumor progression. However, VSG was not as effective as diet in reducing tumor burden despite achieving similar weight and adiposity loss. Leptin did not associate with changes in tumor burden; however, circulating IL-6 was elevated in VSG mice. Uniquely, VSG tumors displayed elevated inflammation and immune checkpoint ligand PD-L1+ myeloid and non-immune cells. VSG tumors also had reduced T lymphocytes and markers of cytolysis, suggesting an ineffective anti-tumor microenvironment which prompted investigation of immune checkpoint blockade. While obese mice were resistant to immune checkpoint blockade, anti-PD-L1 potently impaired tumor progression after VSG through improved anti-tumor immunity. Thus, in formerly obese mice, surgical weight loss followed by immunotherapy reduced breast cancer burden. Finally, we compared transcriptomic changes in adipose tissue after bariatric surgery from patients and mouse models. A conserved bariatric surgery-associated weight loss signature (BSAS) was identified which significantly associated with decreased tumor volume. Findings demonstrate conserved impacts of obesity and bariatric surgery-induced weight loss pathways associated with breast cancer progression. As the number of people classified as obese rises globally, so do obesity-related health risks. Studies show that people diagnosed with obesity have inflammation that contributes to tumor growth and their immune system is worse at detecting cancer cells. But weight loss is not currently used as a strategy for preventing or treating cancer. Surgical procedures for weight loss, also known as ‘bariatric surgeries’, are becoming increasingly popular. Recent studies have shown that individuals who lose weight after these treatments have a reduced risk of developing tumors. But how bariatric surgery directly impacts cancer progression has not been well studied: does it slow tumor growth or boost the anti-tumor immune response? To answer these questions, Sipe et al. compared breast tumor growth in groups of laboratory mice that were obese due to being fed a high fat diet. The first group of mice lost weight after undergoing a bariatric surgery in which part of their stomach was removed. The second lost the same amount of weight but after receiving a restricted diet, and the third underwent a fake surgery and did not lose any weight. The experiments found that surgical weight loss cuts breast cancer tumor growth in half compared with obese mice. But mice who lost the same amount of weight through dietary restrictions had even less tumor growth than surgically treated mice. The surgically treated mice who lost weight had more inflammation than mice in the two other groups, and had increased amounts of proteins and cells that block the immune response to tumors. Giving the surgically treated mice a drug that enhances the immune system’s ability to detect and destroy cancer cells reduced inflammation and helped shrink the mice’s tumors. Finally, Sipe et al. identified 54 genes which were turned on or off after bariatric surgery in both mice and humans, 11 of which were linked with tumor size. These findings provide crucial new information about how bariatric surgery can impact cancer progression. Future studies could potentially use the conserved genes identified by Sipe et al. to develop new ways to stimulate the anti-cancer benefits of weight loss without surgery.
Collapse
Affiliation(s)
- Laura M Sipe
- Department of Medicine, University of Tennessee Health Science Center, Memphis, United States
| | - Mehdi Chaib
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, United States
| | - Emily B Korba
- Department of Medicine, University of Tennessee Health Science Center, Memphis, United States
| | - Heejoon Jo
- Department of Medicine, University of Tennessee Health Science Center, Memphis, United States
| | - Mary Camille Lovely
- Department of Medicine, University of Tennessee Health Science Center, Memphis, United States
| | - Brittany R Counts
- Integrative Muscle Biology Laboratory, University of Tennessee Health Science Center, Memphis, United States
| | - Ubaid Tanveer
- Department of Medicine, University of Tennessee Health Science Center, Memphis, United States
| | - Jeremiah R Holt
- Department of Medicine, University of Tennessee Health Science Center, Memphis, United States
| | - Jared C Clements
- Department of Medicine, University of Tennessee Health Science Center, Memphis, United States
| | - Neena A John
- Department of Medicine, University of Tennessee Health Science Center, Memphis, United States
| | - Deidre Daria
- Office of Vice Chancellor for Research, University of Tennessee Health Science Center, Memphis, United States
| | - Tony N Marion
- Office of Vice Chancellor for Research, University of Tennessee Health Science Center, Memphis, United States
| | - Margaret S Bohm
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, United States
| | - Radhika Sekhri
- Department of Pathology, University of Tennessee Health Science Center, Memphis, United States
| | - Ajeeth K Pingili
- Department of Medicine, University of Tennessee Health Science Center, Memphis, United States
| | - Bin Teng
- Department of Medicine, University of Tennessee Health Science Center, Memphis, United States
| | - James A Carson
- Integrative Muscle Biology Laboratory, University of Tennessee Health Science Center, Memphis, United States
| | - D Neil Hayes
- Department of Medicine, University of Tennessee Health Science Center, Memphis, United States
| | - Matthew J Davis
- Department of Medicine, University of Tennessee Health Science Center, Memphis, United States
| | - Katherine L Cook
- Department of Surgery, Wake Forest University, Winston Salem, United States
| | - Joseph F Pierre
- Department of Microbiology, University of Tennessee Health Science Center, Memphis, United States
| | - Liza Makowski
- Department of Medicine, University of Tennessee Health Science Center, Memphis, United States
| |
Collapse
|
19
|
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: 54] [Impact Index Per Article: 18.0] [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
|
20
|
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
|
21
|
Hoy AJ, Nagarajan SR, Butler LM. Tumour fatty acid metabolism in the context of therapy resistance and obesity. Nat Rev Cancer 2021; 21:753-766. [PMID: 34417571 DOI: 10.1038/s41568-021-00388-4] [Citation(s) in RCA: 207] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/01/2021] [Indexed: 02/07/2023]
Abstract
Fatty acid metabolism is known to support tumorigenesis and disease progression as well as treatment resistance through enhanced lipid synthesis, storage and catabolism. More recently, the role of membrane fatty acid composition, for example, ratios of saturated, monounsaturated and polyunsaturated fatty acids, in promoting cell survival while limiting lipotoxicity and ferroptosis has been increasingly appreciated. Alongside these insights, it has become clear that tumour cells exhibit plasticity with respect to fatty acid metabolism, responding to extratumoural and systemic metabolic signals, such as obesity and cancer therapeutics, to promote the development of aggressive, treatment-resistant disease. Here, we describe cellular fatty acid metabolic changes that are connected to therapy resistance and contextualize obesity-associated changes in host fatty acid metabolism that likely influence the local tumour microenvironment to further modify cancer cell behaviour while simultaneously creating potential new vulnerabilities.
Collapse
Affiliation(s)
- Andrew J Hoy
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| | - Shilpa R Nagarajan
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Lisa M Butler
- Adelaide Medical School and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
22
|
Zhong X, Wang X, Sun Q. CCL2/ACKR2 interaction participate in breast cancer metastasis especially in patients with altered lipid metabolism. Med Hypotheses 2021; 158:110734. [PMID: 34861532 DOI: 10.1016/j.mehy.2021.110734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/05/2021] [Accepted: 10/23/2021] [Indexed: 11/23/2022]
Abstract
Breast cancer is the most prevalent cancer worldwide and metastasis accounts for the majority of associated deaths. Altered lipid metabolism has been proved to be involved with breast cancer, yet the underlying mechanism in lipid metabolism mediated metastasis in breast cancer remains inadequately understood. Evidence have indicated that adipocytes can produce chemokine ligand 2 (CCL2), and several studies have shown that tumor metastasis and patient survival is associated with atypical chemokine receptors/chemokine decoy receptors. We are interested in the factors that may influence cancer metastasis and patient prognosis, particularly in patients with altered lipid metabolism. In this paper, we propose a hypothesis that patients with increased expression levels of atypical chemokine receptor 2 (ACKR2) receptors will have less chance for tumor metastasis, whereas patients with decreased ACKR2 expression but high levels of chemokine receptor 2 positive (CCR2 + ) monocytes are more likely to develop metastasis and have worse outcomes. However, in patients with lower ACKR2 expression level but elevated level of CCR2 + NK cells, primary tumor can be suppressed and therefore present better outcomes.
Collapse
Affiliation(s)
- Xiaoying Zhong
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, China; Peking Union Medical College, MD Program, China
| | - Xuefei Wang
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, China
| | - Qiang Sun
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, China.
| |
Collapse
|
23
|
Yao H, He S. Multi‑faceted role of cancer‑associated adipocytes in the tumor microenvironment (Review). Mol Med Rep 2021; 24:866. [PMID: 34676881 PMCID: PMC8554381 DOI: 10.3892/mmr.2021.12506] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 09/15/2021] [Indexed: 01/08/2023] Open
Abstract
Adipocytes are a type of stromal cell found in numerous different tissues that serve an active role in the tumor microenvironment. Cancer-associated adipocytes (CAAs) display a malignant phenotype and are found at the invasive tumor front, which mediates the crosstalk network between adipocytes (the precursor cells that will become cancer-associated adipocytes in the future) and cancer cells. The present review covers the mechanisms of adipocytes in the development of cancer, including metabolic reprogramming, chemotherapy resistance and adipokine regulation. Furthermore, the potential mechanisms involved in the adipocyte-cancer cell cycle in various types of cancer, including breast, ovarian, colon and rectal cancer, are discussed. Deciphering the complex network of CAA-cancer cell crosstalk will provide insights into tumor biology and optimize therapeutic strategies.
Collapse
Affiliation(s)
- Huihui Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Songbing He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
24
|
La Camera G, Gelsomino L, Malivindi R, Barone I, Panza S, De Rose D, Giordano F, D'Esposito V, Formisano P, Bonofiglio D, Andò S, Giordano C, Catalano S. Adipocyte-derived extracellular vesicles promote breast cancer cell malignancy through HIF-1α activity. Cancer Lett 2021; 521:155-168. [PMID: 34425186 DOI: 10.1016/j.canlet.2021.08.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/28/2021] [Accepted: 08/17/2021] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EVs) are emerging key protagonists in intercellular communication between adipocytes and breast cancer (BC) cells. Here, we described a new mechanism by which EVs released by mature adipocytes promoted breast cancer cell malignancy "in vitro" and "in vivo". We found that adipocyte-derived EVs enhanced growth, motility and invasion, stem cell-like properties, as well as specific traits of epithelial-to-mesenchymal transition in both estrogen receptor positive and triple negative BC cells. Of note, adipocyte-derived EVs aid breast tumor cells in lung metastatic colonization after tail-vein injection in mice. These EV-mediated effects occur via the induction of HIF-1α activity, since they were abrogated by the use of the HIF-1α inhibitor KC7F2 or in cells silenced for HIF-1α expression. Moreover, using an "ex vivo" model of obese adipocytes we found that the depletion of EVs counteracted the ability of obese adipocytes to sustain pro-invasive phenotype in BC cells. Interestingly, EVs released by undifferentiated adipocytes failed to induce aggressiveness and HIF-1α expression. These findings shed new light on the role of adipocyte-derived EVs in breast cancer progression, suggesting the possibility to target HIF-1α activity to block the harmful adipocyte-tumor cell dialogue, especially in obese settings.
Collapse
Affiliation(s)
- Giusi La Camera
- Department of Pharmacy, Health and Nutritional Sciences, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy
| | - Rocco Malivindi
- Department of Pharmacy, Health and Nutritional Sciences, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy
| | - Salvatore Panza
- Department of Pharmacy, Health and Nutritional Sciences, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy
| | - Daniela De Rose
- Department of Pharmacy, Health and Nutritional Sciences, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy; Centro Sanitario, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy
| | - Vittoria D'Esposito
- Department of Translational Medicine, Federico II University of Naples, 80131, Naples, Italy
| | - Pietro Formisano
- Department of Translational Medicine, Federico II University of Naples, 80131, Naples, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy; Centro Sanitario, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy; Centro Sanitario, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy; Centro Sanitario, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy.
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy; Centro Sanitario, Via P. Bucci, University of Calabria, 87036, Arcavacata di Rende (CS), Italy.
| |
Collapse
|
25
|
Gaggianesi M, Di Franco S, Pantina VD, Porcelli G, D'Accardo C, Verona F, Veschi V, Colarossi L, Faldetta N, Pistone G, Bongiorno MR, Todaro M, Stassi G. Messing Up the Cancer Stem Cell Chemoresistance Mechanisms Supported by Tumor Microenvironment. Front Oncol 2021; 11:702642. [PMID: 34354950 PMCID: PMC8330815 DOI: 10.3389/fonc.2021.702642] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the recent advances in cancer patient management and in the development of targeted therapies, systemic chemotherapy is currently used as a first-line treatment for many cancer types. After an initial partial response, patients become refractory to standard therapy fostering rapid tumor progression. Compelling evidence highlights that the resistance to chemotherapeutic regimens is a peculiarity of a subpopulation of cancer cells within tumor mass, known as cancer stem cells (CSCs). This cellular compartment is endowed with tumor-initiating and metastasis formation capabilities. CSC chemoresistance is sustained by a plethora of grow factors and cytokines released by neighboring tumor microenvironment (TME), which is mainly composed by adipocytes, cancer-associated fibroblasts (CAFs), immune and endothelial cells. TME strengthens CSC refractoriness to standard and targeted therapies by enhancing survival signaling pathways, DNA repair machinery, expression of drug efflux transporters and anti-apoptotic proteins. In the last years many efforts have been made to understand CSC-TME crosstalk and develop therapeutic strategy halting this interplay. Here, we report the combinatorial approaches, which perturb the interaction network between CSCs and the different component of TME.
Collapse
Affiliation(s)
- Miriam Gaggianesi
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Simone Di Franco
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Vincenzo Davide Pantina
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | - Gaetana Porcelli
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Caterina D'Accardo
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Francesco Verona
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Veronica Veschi
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| | | | - Naida Faldetta
- Department of Surgery, Villa Sofia-Cervello Hospital, Palermo, Italy
| | - Giuseppe Pistone
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Maria Rita Bongiorno
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Matilde Todaro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Giorgio Stassi
- Department of Surgical Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, Palermo, Italy
| |
Collapse
|
26
|
Avşar Abdik E. Differentiated pre-adipocytes promote proliferation, migration and epithelial-mesenchymal transition in breast cancer cells of different p53 status. Mol Biol Rep 2021; 48:5187-5198. [PMID: 34213707 DOI: 10.1007/s11033-021-06521-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/25/2021] [Indexed: 12/11/2022]
Abstract
Breast cancer progression and metastasis are associated with stromal cells in the tumor microenvironment. Adipocytes are the most abundant cells surrounding breast stromal tissue, promote tumor progression through the induction of Epithelial-to-Mesenchymal Transition (EMT) which is negatively regulated by tumor suppressor protein p53. In this study aimed to investigate the role of p53 in the progression of breast cancer after mature adipocyte-conditioned medium (CM) application. The proliferative effect of CM obtained from differentiated pre-adipocytes were assessed by MTS assay. 20% CM increased cell proliferation in breast cancer cells, T-47D (mutant p53) and MCF-7 (wild-type p53). The migration and invasion capacity were evaluated by scratch and transwell assays, respectively. CM significantly enhanced migration and invasion capacity in T-47D compared to MCF-7. Gene and protein expressions were detected by qRT-PCR and Western Blot analysis, respectively. CM markedly increased expression levels of Cyclin D1, PI3K, MMP9, Snail and Twist in T-47D compared to MCF-7. However, CM did not change E-Cadherin level in T-47D while downregulated in MCF-7 cells. Also, the protein levels of NFκB p65, p-Akt, Snail, and Vimentin were upregulated in both cells. Overall, the findings highlight how the p53 status affects mature adipocyte-mediated proliferation, migration, and aggressive behavior of breast cancer cell lines. Targeting the tumor microenvironment may represent a promising approach for preventing breast cancer progression and metastasis.
Collapse
Affiliation(s)
- Ezgi Avşar Abdik
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, 26 Ağustos Campus, Kayisdagi, Istanbul, Turkey.
| |
Collapse
|
27
|
Abstract
AbstractThe world is in the grip of an obesity pandemic, with tripling of obesity rates since 1975; it is predicted that one-third of people on Earth will be obese by 2025. The health consequences of obesity are primarily thought to be related to cardiometabolic disorders such as diabetes and cardiovascular diseases. It is less well appreciated that obesity has been related to at least 13 different cancers and in future, (with increasing control over tobacco misuse and infections), obesity will be the main cause of cancers. While this is an area of active research, there are large gaps in the definition of what is an obesity related cancer (JRC) and more importantly, what are the underlying mechanisms. To an extent, this is due to the controversy on what constitutes “unhealthy obesity” which is further related to the causes of obesity. This narrative review examines the causes and measurement of obesity, the types of obesity-related cancers and possible mechanisms. The information has wide implications ranging from prevention, screening, prognosis and therapeutic strategies. Obesity related cancers should be an area of high-priority research. Oncologists can contribute by spreading awareness and instituting management measures for individual patients in their care.
Collapse
Affiliation(s)
- Ajit Venniyoor
- National Oncology Centre, The Royal Hospital, Muscat, Sultanate of Oman
| |
Collapse
|
28
|
McDowell SAC, Luo RBE, Arabzadeh A, Doré S, Bennett NC, Breton V, Karimi E, Rezanejad M, Yang RR, Lach KD, Issac MSM, Samborska B, Perus LJM, Moldoveanu D, Wei Y, Fiset B, Rayes RF, Watson IR, Kazak L, Guiot MC, Fiset PO, Spicer JD, Dannenberg AJ, Walsh LA, Quail DF. Neutrophil oxidative stress mediates obesity-associated vascular dysfunction and metastatic transmigration. ACTA ACUST UNITED AC 2021; 2:545-562. [DOI: 10.1038/s43018-021-00194-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/10/2021] [Indexed: 12/22/2022]
|
29
|
Bevinakoppamath S, Saleh Ahmed AM, Ramachandra SC, Vishwanath P, Prashant A. Chemopreventive and Anticancer Property of Selenoproteins in Obese Breast Cancer. Front Pharmacol 2021; 12:618172. [PMID: 33935708 PMCID: PMC8087246 DOI: 10.3389/fphar.2021.618172] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/05/2021] [Indexed: 12/24/2022] Open
Abstract
Obesity is a significant risk factor for various cancers including breast cancer resulting in an increased risk of recurrence as well as morbidity and mortality. Extensive studies on various pathways have been successful in establishing a biological relationship between obesity and breast cancer. The molecular classification of breast cancer includes five groups each having different responses to treatment. Increased levels of inflammatory cytokines seen in obese conditions drive the pro-proliferative pathways, such as the influx of macrophages, angiogenesis, and antiapoptotic pathways. Increased peripheral aromatization of androgens by aromatase increases the circulating estrogen levels which are also responsible for the association of obesity with breast cancer. Also, increased oxidative stress due to chronic low-grade inflammation in obese women plays an important role in carcinogenesis. Despite the availability of safe and effective treatment options for breast cancer, obese women are at increased risk of adverse outcomes including treatment-related toxicities. In the recent decade, selenium compounds have gained substantial interest as chemopreventive and anticancer agents. The chemical derivatives of selenium include inorganic and organic compounds that exhibit pro-oxidant properties and alter cellular redox homeostasis. They target more than one metabolic pathway by thiol modifications, induction of reactive oxygen species, and chromatin modifications to exert their chemopreventive and anticancer activities. The primary functional effectors of selenium that play a significant role in human homeostasis are selenoproteins like glutathione peroxidase, thioredoxin reductase, iodothyronine deiodinases, and selenoprotein P. Selenoproteins play a significant role in adipose tissue physiology by modulating preadipocyte proliferation and adipogenic differentiation. They correlate negatively with body mass index resulting in increased oxidative stress that may lead to carcinogenesis in obese individuals. Methylseleninic acid effectively suppresses aromatase activation thus reducing the estrogen levels and acting as a breast cancer chemopreventive agent. Adipose-derived inflammatory mediators influence the selenium metabolites and affect the proliferation and metastatic properties of cancer cells. Recently selenium nanoparticles have shown potent anticancer activity which may lead to a major breakthrough in the management of cancers caused due to multiple pathways. In this review, we discuss the possible role of selenoproteins as chemopreventive and an anticancer agent in obese breast cancer.
Collapse
Affiliation(s)
- Supriya Bevinakoppamath
- Center of Excellence in Molecular Biology and Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, India
| | - Adel Mohammed Saleh Ahmed
- Center of Excellence in Molecular Biology and Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, India
| | - Shobha Chikkavaddaraguddi Ramachandra
- Center of Excellence in Molecular Biology and Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, India
| | - Prashant Vishwanath
- Center of Excellence in Molecular Biology and Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, India
| | - Akila Prashant
- Center of Excellence in Molecular Biology and Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, India
| |
Collapse
|
30
|
Bustamante-Marin XM, Merlino JL, Devericks E, Carson MS, Hursting SD, Stewart DA. Mechanistic Targets and Nutritionally Relevant Intervention Strategies to Break Obesity-Breast Cancer Links. Front Endocrinol (Lausanne) 2021; 12:632284. [PMID: 33815289 PMCID: PMC8011316 DOI: 10.3389/fendo.2021.632284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/17/2021] [Indexed: 12/29/2022] Open
Abstract
The worldwide prevalence of overweight and obesity has tripled since 1975. In the United States, the percentage of adults who are obese exceeds 42.5%. Individuals with obesity often display multiple metabolic perturbations, such as insulin resistance and persistent inflammation, which can suppress the immune system. These alterations in homeostatic mechanisms underlie the clinical parameters of metabolic syndrome, an established risk factor for many cancers, including breast cancer. Within the growth-promoting, proinflammatory milieu of the obese state, crosstalk between adipocytes, immune cells and breast epithelial cells occurs via obesity-associated hormones, angiogenic factors, cytokines, and other mediators that can enhance breast cancer risk and/or progression. This review synthesizes evidence on the biological mechanisms underlying obesity-breast cancer links, with emphasis on emerging mechanism-based interventions in the context of nutrition, using modifiable elements of diet alone or paired with physical activity, to reduce the burden of obesity on breast cancer.
Collapse
Affiliation(s)
| | - Jenna L. Merlino
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
| | - Emily Devericks
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
| | - Meredith S. Carson
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
| | - Stephen D. Hursting
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina, Kannapolis, NC, United States
| | - Delisha A. Stewart
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina, Kannapolis, NC, United States
| |
Collapse
|
31
|
Hillers-Ziemer LE, Williams AE, Janquart A, Grogan C, Thompson V, Sanchez A, Arendt LM. Obesity-Activated Lung Stromal Cells Promote Myeloid Lineage Cell Accumulation and Breast Cancer Metastasis. Cancers (Basel) 2021; 13:1005. [PMID: 33670906 PMCID: PMC7957630 DOI: 10.3390/cancers13051005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/13/2021] [Accepted: 02/23/2021] [Indexed: 12/25/2022] Open
Abstract
Obesity is correlated with increased incidence of breast cancer metastasis; however, the mechanisms underlying how obesity promotes metastasis are unclear. In a diet-induced obese mouse model, obesity enhanced lung metastasis in both the presence and absence of primary mammary tumors and increased recruitment of myeloid lineage cells into the lungs. In the absence of tumors, obese mice demonstrated increased numbers of myeloid lineage cells and elevated collagen fibers within the lung stroma, reminiscent of premetastatic niches formed by primary tumors. Lung stromal cells isolated from obese tumor-naïve mice showed increased proliferation, contractility, and expression of extracellular matrix, inflammatory markers and transforming growth factor beta-1 (TGFβ1). Conditioned media from lung stromal cells from obese mice promoted myeloid lineage cell migration in vitro in response to colony-stimulating factor 2 (CSF2) expression and enhanced invasion of tumor cells. Together, these results suggest that prior to tumor formation, obesity alters the lung microenvironment, creating niches conducive to metastatic growth.
Collapse
Affiliation(s)
- Lauren E. Hillers-Ziemer
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Abbey E. Williams
- Program in Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Amanda Janquart
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| | - Caitlin Grogan
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| | - Victoria Thompson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| | - Adriana Sanchez
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| | - Lisa M. Arendt
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA;
- Program in Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| |
Collapse
|
32
|
Soni S, Torvund M, Mandal CC. Molecular insights into the interplay between adiposity, breast cancer and bone metastasis. Clin Exp Metastasis 2021; 38:119-138. [PMID: 33591548 DOI: 10.1007/s10585-021-10076-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 02/03/2021] [Indexed: 01/20/2023]
Abstract
Cancer is a complex disease, with various pre-existing health ailments enhancing its pathology. In cancer, the extracellular environment contains various intrinsic physiological factors whose levels are altered with aging and pre-existing conditions. In obesity, the tumor microenvironment and metastases are enriched with factors that are both derived locally, and from other physiological compartments. Similarly, in obesity, the cancer cell environment both at the site of origin and at the secondary site i.e., metastatic niche, contains significantly more phenotypically-altered adipocytes than that of un-obese cancer patients. Indeed, obesity has been linked with cancer progression, metastasis, and therapy resistance. Adipocytes not only interact with tumor cells, but also with adjacent stromal cells at primary and metastatic sites. This review emphasizes the importance of bidirectional interactions between adipocytes and breast tumor cells in breast cancer progression and its bone metastases. This paper not only chronicles the role of various adipocyte-derived factors in tumor growth, but also describes the significance of adipocyte-derived bone metastatic factors in the development of bone metastasis of breast cancer. It provides a molecular view of the interplay between the adipocytes and tumor cells involved in breast cancer bone metastasis. However, more research is needed to determine if targeting cancer-associated adipocytes holds promise as a potential therapeutic approach for breast cancer bone metastasis treatment. Interplay between adipocytes and breast cancer cells at primary cancer site and metastatic bone microenvironment. AMSC Adipose-derived mesenchymal stem cell, CAA Cancer associated adipocytes, CAF Cancer associated fibroblast, BMSC Bone marrow derived mesenchymal stem cell, BMA Bone marrow adipocyte.
Collapse
Affiliation(s)
- Sneha Soni
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan, 305817, India
| | - Meaghan Torvund
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan, 305817, India.
| |
Collapse
|
33
|
Sauer S, Beinart D, Finn SMB, Kumar SL, Cheng Q, Hwang SE, Parker W, Devi GR. Hymenolepis diminuta-based helminth therapy in C3(1)-TAg mice does not alter breast tumor onset or progression. Evol Med Public Health 2021; 9:131-138. [PMID: 33738103 PMCID: PMC7953836 DOI: 10.1093/emph/eoab007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 02/07/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND OBJECTIVES An individual's risk of breast cancer is profoundly affected by evolutionary mismatch. Mismatches in Western society known to increase the risk of breast cancer include a sedentary lifestyle and reproductive factors. Biota alteration, characterized by a loss of biodiversity from the ecosystem of the human body as a result of Western society, is a mismatch known to increase the risk of a variety of inflammation-related diseases, including colitis-associated colon cancer. However, the effect of biota alteration on breast cancer has not been evaluated. METHODOLOGY In this study, we utilized the C3(1)-TAg mouse model of breast cancer to evaluate the role of biota alteration in the development of breast cancer. This model has been used to recapitulate the role of exercise and pregnancy in reducing the risk of breast cancer. C3(1)-TAg mice were treated with Hymenolepis diminuta, a benign helminth that has been shown to reverse the effects of biota alteration in animal models. RESULTS No effect of the helminth H. diminuta was observed. Neither the latency nor tumor growth was affected by the therapy, and no significant effects on tumor transcriptome were observed based on RNAseq analysis. CONCLUSIONS AND IMPLICATIONS These findings suggest that biota alteration, although known to affect a variety of Western-associated diseases, might not be a significant factor in the high rate of breast cancer observed in Western societies. LAY SUMMARY An almost complete loss of intestinal worms in high-income countries has led to increases in allergic disorders, autoimmune conditions, and perhaps colon cancer. However, in this study, results using laboratory mice suggest that loss of intestinal worms might not be associated with breast cancer.
Collapse
Affiliation(s)
- Scott Sauer
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Dylan Beinart
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Sade M B Finn
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Sereena L Kumar
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Qing Cheng
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Shelley E Hwang
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - William Parker
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Gayathri R Devi
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
34
|
Acheva A, Kärki T, Schaible N, Krishnan R, Tojkander S. Adipokine Leptin Co-operates With Mechanosensitive Ca 2 +-Channels and Triggers Actomyosin-Mediated Motility of Breast Epithelial Cells. Front Cell Dev Biol 2021; 8:607038. [PMID: 33490070 PMCID: PMC7815691 DOI: 10.3389/fcell.2020.607038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022] Open
Abstract
In postmenopausal women, a major risk factor for the development of breast cancer is obesity. In particular, the adipose tissue-derived adipokine leptin has been strongly linked to tumor cell proliferation, migration, and metastasis, but the underlying mechanisms remain unclear. Here we show that treatment of normal mammary epithelial cells with leptin induces EMT-like features characterized by higher cellular migration speeds, loss of structural ordering of 3D-mammo spheres, and enhancement of epithelial traction forces. Mechanistically, leptin triggers the phosphorylation of myosin light chain kinase-2 (MLC-2) through the interdependent activity of leptin receptor and Ca2+ channels. These data provide evidence that leptin-activated leptin receptors, in co-operation with mechanosensitive Ca2+ channels, play a role in the development of breast carcinomas through the regulation of actomyosin dynamics.
Collapse
Affiliation(s)
- Anna Acheva
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Tytti Kärki
- Department of Applied Physics, School of Science, Aalto University, Espoo, Finland
| | - Niccole Schaible
- Beth Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Ramaswamy Krishnan
- Beth Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Sari Tojkander
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
35
|
Wishart AL, Conner SJ, Guarin JR, Fatherree JP, Peng Y, McGinn RA, Crews R, Naber SP, Hunter M, Greenberg AS, Oudin MJ. Decellularized extracellular matrix scaffolds identify full-length collagen VI as a driver of breast cancer cell invasion in obesity and metastasis. SCIENCE ADVANCES 2020; 6:6/43/eabc3175. [PMID: 33087348 PMCID: PMC7577726 DOI: 10.1126/sciadv.abc3175] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/08/2020] [Indexed: 05/05/2023]
Abstract
The extracellular matrix (ECM), a major component of the tumor microenvironment, promotes local invasion to drive metastasis. Here, we describe a method to study whole-tissue ECM effects from disease states associated with metastasis on tumor cell phenotypes and identify the individual ECM proteins and signaling pathways that are driving these effects. We show that decellularized ECM from tumor-bearing and obese mammary glands drives TNBC cell invasion. Proteomics of the ECM from the obese mammary gland led us to identify full-length collagen VI as a novel driver of TNBC cell invasion whose abundance in tumor stroma increases with body mass index in human TNBC patients. Last, we describe the mechanism by which collagen VI contributes to TNBC cell invasion via NG2-EGFR cross-talk and MAPK signaling. Overall, these studies demonstrate the value of decellularized ECM scaffolds obtained from tissues to identify novel functions of the ECM.
Collapse
Affiliation(s)
- Andrew L Wishart
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Sydney J Conner
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Justinne R Guarin
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Jackson P Fatherree
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Yifan Peng
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Rachel A McGinn
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Rebecca Crews
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Stephen P Naber
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Martin Hunter
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Andrew S Greenberg
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
- Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA 02111, USA
- Tufts University School of Medicine, Boston, MA 02111, USA
| | - Madeleine J Oudin
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
36
|
Blücher C, Iberl S, Schwagarus N, Müller S, Liebisch G, Höring M, Hidrobo MS, Ecker J, Spindler N, Dietrich A, Burkhardt R, Stadler SC. Secreted Factors from Adipose Tissue Reprogram Tumor Lipid Metabolism and Induce Motility by Modulating PPARα/ANGPTL4 and FAK. Mol Cancer Res 2020; 18:1849-1862. [DOI: 10.1158/1541-7786.mcr-19-1223] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 07/16/2020] [Accepted: 08/24/2020] [Indexed: 11/16/2022]
|
37
|
Kolb R, Zhang W. Obesity and Breast Cancer: A Case of Inflamed Adipose Tissue. Cancers (Basel) 2020; 12:E1686. [PMID: 32630445 PMCID: PMC7352736 DOI: 10.3390/cancers12061686] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/09/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
Obesity is associated with an increased risk of estrogen receptor-positive breast cancer in postmenopausal women and a worse prognosis for all major breast cancer subtypes regardless of menopausal status. While the link between obesity and the pathogenesis of breast cancer is clear, the molecular mechanism of this association is not completely understood due to the complexity of both obesity and breast cancer. The aim of this review is to highlight the association between obesity and breast cancer and discuss the literature, which indicates that this association is due to chronic adipose tissue inflammation. We will discuss the epidemiological data for the association between breast cancer incidence and progression as well as the potential molecular mechanisms for this association. We will focus on the role of inflammation within the adipose tissue during the pathogenesis of breast cancer. A better understanding of how obesity and adipose tissue inflammation affects the pathogenesis of breast cancer will lead to new strategies to reduce breast cancer risk and improve patient outcomes for obese patients.
Collapse
Affiliation(s)
- Ryan Kolb
- Department of Pathology, Immunology and Laboratory Medicine, Gainesville, FL 32610, USA;
- University of Florida Health Cancer Center, Gainesville, FL 32610, USA
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, Gainesville, FL 32610, USA;
- University of Florida Health Cancer Center, Gainesville, FL 32610, USA
| |
Collapse
|
38
|
Sipe LM, Chaib M, Pingili AK, Pierre JF, Makowski L. Microbiome, bile acids, and obesity: How microbially modified metabolites shape anti-tumor immunity. Immunol Rev 2020; 295:220-239. [PMID: 32320071 PMCID: PMC7841960 DOI: 10.1111/imr.12856] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023]
Abstract
Bile acids (BAs) are known facilitators of nutrient absorption but recent paradigm shifts now recognize BAs as signaling molecules regulating both innate and adaptive immunity. Bile acids are synthesized from cholesterol in the liver with subsequent microbial modification and fermentation adding complexity to pool composition. Bile acids act on several receptors such as Farnesoid X Receptor and the G protein-coupled BA receptor 1 (TGR5). Interestingly, BA receptors (BARs) are expressed on immune cells and activation either by BAs or BAR agonists modulates innate and adaptive immune cell populations skewing their polarization toward a more tolerogenic anti-inflammatory phenotype. Intriguingly, recent evidence also suggests that BAs promote anti-tumor immune response through activation and recruitment of tumoricidal immune cells such as natural killer T cells. These exciting findings have redefined BA signaling in health and disease wherein they may suppress inflammation on the one hand, yet promote anti-tumor immunity on the other hand. In this review, we provide our readers with the most recent understanding of the interaction of BAs with the host microbiome, their effect on innate and adaptive immunity in health and disease with a special focus on obesity, bariatric surgery-induced weight loss, and immune checkpoint blockade in cancer.
Collapse
Affiliation(s)
- Laura M. Sipe
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mehdi Chaib
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ajeeth K. Pingili
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Joseph F. Pierre
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Liza Makowski
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
39
|
Santoni M, Occhipinti G, Romagnoli E, Miccini F, Scoccia L, Giulietti M, Principato G, Saladino T, Piva F, Battelli N. Different Cardiotoxicity of Palbociclib and Ribociclib in Breast Cancer: Gene Expression and Pharmacological Data Analyses, Biological Basis, and Therapeutic Implications. BioDrugs 2020; 33:613-620. [PMID: 31529317 DOI: 10.1007/s40259-019-00382-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breast cancer is the most frequent tumor in women. The recent advent of cyclin-dependent kinase (CDK) 4/6 inhibitors palbociclib and ribociclib has represented a major step forward for patients with hormone receptor-positive breast cancer. These two agents have showed similar efficacy in terms of breast cancer outcome but different cardiotoxic effects. In particular, ribociclib, but not palbociclib, has been associated with QT interval prolongation, and the mechanisms underlying this event are still unclear. In order to clarify such difference, we matched the candidate genes associated with QT interval prolongation with genes whose expression is altered following palbociclib or ribociclib treatment. We also investigated whether pharmacokinetic and pharmacodynamic characteristics, such as IC50 (hERG) [concentration of drug producing 50% inhibition (human ether-à-go-go related gene)] and maximum concentration (Cmax), could justify the different effects on QT interval prolongation. Our results show that ribociclib, but not palbociclib, could act by down-regulating the expression of KCNH2 (encoding for potassium channel hERG) and up-regulating SCN5A and SNTA1 (encoding for sodium channels Nav1.5 and syntrophin-α1, respectively), three genes associated with long QT syndrome. Consistent with the cardiotoxicity induced by ribociclib, its IC50 (hERG)/free concentration (Cmax free) ratio is closer to the safety threshold than that of palbociclib. In summary, we hypothesize that the different cardiotoxicity associated with ribociclib and palbociclib could be due to the alteration of potassium and sodium channels induced by ribociclib. A better comprehension of the mechanisms of cardiac channelopathies and drug-induced QT interval prolongation will be fundamental to avoid serious and potentially lethal adverse events and, as a consequence, optimize the management of breast cancer patients.
Collapse
Affiliation(s)
- Matteo Santoni
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, Macerata, Italy
| | - Giulia Occhipinti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Monte d'Ago, 60131, Ancona, Italy
| | | | - Francesca Miccini
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, Macerata, Italy
| | | | - Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Monte d'Ago, 60131, Ancona, Italy
| | - Giovanni Principato
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Monte d'Ago, 60131, Ancona, Italy
| | - Tiziana Saladino
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, Macerata, Italy
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Monte d'Ago, 60131, Ancona, Italy.
| | - Nicola Battelli
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, Macerata, Italy
| |
Collapse
|
40
|
Ma B, Wells A, Clark AM. The pan-therapeutic resistance of disseminated tumor cells: Role of phenotypic plasticity and the metastatic microenvironment. Semin Cancer Biol 2020; 60:138-147. [PMID: 31376430 PMCID: PMC6992520 DOI: 10.1016/j.semcancer.2019.07.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 02/07/2023]
Abstract
Cancer metastasis is the leading cause of mortality in patients with solid tumors. The majority of these deaths are associated with metastatic disease that occurs after a period of clinical remission, anywhere from months to decades following removal of the primary mass. This dormancy is prominent in cancers of the breast and prostate among others, leaving the survivors uncertain about their longer-term prognosis. The most daunting aspect of this dormancy and re-emergence is that the micrometastases in particular, and even large lethal outgrowths are often show resistance to agents to which they have not been exposed. This suggests that in addition to specific mutations that target single agents, there also exist adaptive mechanisms that provide this pan-resistance. Potential molecular underpinnings of which are the topic of this review.
Collapse
Affiliation(s)
- Bo Ma
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA; VA Pittsburgh Healthcare System, Pittsburgh, PA 15213, USA
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA; VA Pittsburgh Healthcare System, Pittsburgh, PA 15213, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA; UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Amanda M Clark
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA; VA Pittsburgh Healthcare System, Pittsburgh, PA 15213, USA.
| |
Collapse
|
41
|
Abstract
Adipose tissue contribution to body mass ranges from 6% in male athletes to over 25% in obese men and over 30% in obese women. Crosstalk between adipocytes and cancer cells that exist in close proximity can lead to changes in the function and phenotype of both cell types. These interactions actively alter the tumour microenvironment (TME). Obesity is one of the major risk factors for multiple types of cancer, including breast cancer. In obesity, the increase in both size and number of adipocytes leads to instability of the TME, as well as increased hypoxia within the TME, which further enhances tumour invasion and metastasis. In this chapter, we will discuss the diverse aspects of adipocytes and adipocyte-derived factors that affect the TME as well as tumour progression and metastasis. In addition, we discuss how obesity affects the TME. We focus primarily on breast cancer but discuss what is known in other cancer types when relevant. We finish by discussing the studies needed to further understand these complex interactions.
Collapse
|
42
|
Breast Cancer Stem Cells as Drivers of Tumor Chemoresistance, Dormancy and Relapse: New Challenges and Therapeutic Opportunities. Cancers (Basel) 2019; 11:cancers11101569. [PMID: 31619007 PMCID: PMC6826533 DOI: 10.3390/cancers11101569] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most frequent cancer among women worldwide. Therapeutic strategies to prevent or treat metastatic disease are still inadequate although great progress has been made in treating early-stage breast cancer. Cancer stem-like cells (CSCs) that are endowed with high plasticity and self-renewal properties have been shown to play a key role in breast cancer development, progression, and metastasis. A subpopulation of CSCs that combines tumor-initiating capacity and a dormant/quiescent/slow cycling status is present throughout the clinical history of breast cancer patients. Dormant/quiescent/slow cycling CSCs are a key component of tumor heterogeneity and they are responsible for chemoresistance, tumor migration, and metastatic dormancy, defined as the ability of CSCs to survive in target organs and generate metastasis up to two decades after diagnosis. Understanding the strategies that are used by CSCs to resist conventional and targeted therapies, to interact with their niche, to escape immune surveillance, and finally to awaken from dormancy is of key importance to prevent and treat metastatic cancer. This review summarizes the current understanding of mechanisms involved in CSCs chemoresistance, dissemination, and metastasis in breast cancer, with a particular focus on dormant cells. Finally, we discuss how advancements in the detection, molecular understanding, and targeting of dormant CSCs will likely open new therapeutic avenues for breast cancer treatment.
Collapse
|
43
|
Cancer and Body Composition: An Association of Global Relevance. WOMEN’S HEALTH BULLETIN 2019. [DOI: 10.5812/whb.65315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
44
|
Barone I, Giordano C, Bonofiglio D, Andò S, Catalano S. The weight of obesity in breast cancer progression and metastasis: Clinical and molecular perspectives. Semin Cancer Biol 2019; 60:274-284. [PMID: 31491560 DOI: 10.1016/j.semcancer.2019.09.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/01/2019] [Indexed: 02/06/2023]
Abstract
The escalating epidemic of overweight and obesity is currently recognized as one of the most significant health and economic concern worldwide. At the present time, over 1.9 billion adults and more than 600 million people can be, respectively, classified as overweight or obese, and numbers will continue to increase in the coming decades. This alarming scenario implies important clinical implications since excessive adiposity can progressively cause and/or exacerbate a wide spectrum of co-morbidities, including type 2 diabetes mellitus, hypertension, cardiovascular disease, and even certain types of cancer, including breast cancer. Indeed, pathological remodelling of white adipose tissue and increased levels of fat-specific cytokines (mainly leptin), as a consequence of the obesity condition, have been associated with several hallmarks of breast cancer, such as sustained proliferative signaling, cellular energetics, inflammation, angiogenesis, activating invasion and metastasis. Different preclinical and clinical data have provided evidence indicating that obesity may worsen the incidence, the severity, and the mortality of breast cancer. In the present review, we will discuss the epidemiological connection between obesity and breast cancer progression and metastasis and we will highlight the candidate players involved in this dangerous relationship. Since the major cause of death from cancer is due to widespread metastases, understanding these complex mechanisms will provide insights for establishing new therapeutic interventions to prevent/blunt the effects of obesity and thwart breast tumor progression and metastatic growth.
Collapse
Affiliation(s)
- Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, 87036, Rende, CS, Italy.
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, 87036, Rende, CS, Italy; Centro Sanitario, University of Calabria, Via P Bucci, 87036, Rende, CS, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, 87036, Rende, CS, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, 87036, Rende, CS, Italy; Centro Sanitario, University of Calabria, Via P Bucci, 87036, Rende, CS, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, 87036, Rende, CS, Italy.
| |
Collapse
|
45
|
Alečković M, McAllister SS, Polyak K. Metastasis as a systemic disease: molecular insights and clinical implications. Biochim Biophys Acta Rev Cancer 2019; 1872:89-102. [PMID: 31202687 PMCID: PMC6692219 DOI: 10.1016/j.bbcan.2019.06.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/06/2019] [Accepted: 06/09/2019] [Indexed: 12/18/2022]
Abstract
Metastasis is a complex systemic disease that develops as a result of interactions between tumor cells and their local and distant microenvironments. Local and systemic immune-related changes play especially critical roles in limiting or enabling the development of metastatic disease. Although anti-tumor immune responses likely eliminate most early primary and metastatic lesions, factors secreted by cancer or stromal cells in the primary tumor can mobilize and activate cells in distant organs in a way that promotes the outgrowth of disseminated cancer cells into macrometastatic lesions. Therefore, the prevention, detection, and effective treatment of metastatic disease require a deeper understanding of the systemic effects of primary tumors as well as predisposing hereditary and acquired host factors including chronic inflammatory conditions. The success of immunotherapy in a subset of cancer patients is an example of how modulating the microenvironment and tumor-immune cell interactions can be exploited for the effective eradiation of even advanced-stage tumors. Here, we highlight emerging insights and clinical implications of cancer as a systemic disease.
Collapse
Affiliation(s)
- Maša Alečković
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America; Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Department of Medicine, Harvard Medical School, Boston, MA, United States of America
| | - Sandra S McAllister
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Department of Medicine, Harvard Medical School, Boston, MA, United States of America
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America; Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Department of Medicine, Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
46
|
Abstract
Development of novel and effective therapeutics for treating various cancers is probably the most congested and challenging enterprise of pharmaceutical companies. Diverse drugs targeting malignant and nonmalignant cells receive clinical approval each year from the FDA. Targeting cancer cells and nonmalignant cells unavoidably changes the tumor microenvironment, and cellular and molecular components relentlessly alter in response to drugs. Cancer cells often reprogram their metabolic pathways to adapt to environmental challenges and facilitate survival, proliferation, and metastasis. While cancer cells' dependence on glycolysis for energy production is well studied, the roles of adipocytes and lipid metabolic reprogramming in supporting cancer growth, metastasis, and drug responses are less understood. This Review focuses on emerging mechanisms involving adipocytes and lipid metabolism in altering the response to cancer treatment. In particular, we discuss mechanisms underlying cancer-associated adipocytes and lipid metabolic reprogramming in cancer drug resistance.
Collapse
|
47
|
Wang R, Tao B, Fan Q, Wang S, Chen L, Zhang J, Hao Y, Dong S, Wang Z, Wang W, Cai Y, Li X, Bao T, Wang X, Qiu X, Wang K, Mo X, Kang Y, Wang Z. Fatty-acid receptor CD36 functions as a hydrogen sulfide-targeted receptor with its Cys333-Cys272 disulfide bond serving as a specific molecular switch to accelerate gastric cancer metastasis. EBioMedicine 2019; 45:108-123. [PMID: 31262715 PMCID: PMC6642364 DOI: 10.1016/j.ebiom.2019.06.037] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/12/2019] [Accepted: 06/19/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Hydrogen Sulfide (H2S), a third member of gasotransmitter family along with nitric oxide (NO) and carbon monoxide (CO), exerts a wide range of cellular and molecular actions in our body. There is a large body of evidence suggesting that H2S plays an important role in cancer metastasis; however, the molecular mechanisms of H2S-mediated acceleration of cancer metastasis remain unknown. METHODS We examined the promote effects of H2S on phenotype of gastric cancer (GC) cells (including those of express wild type CD36 and mutant CD36) in vitro and in vivo. GC patients' samples were used for clinical translational significance evaluation. FINDINGS H2S triggered lipid metabolism reprogramming by significantly up-regulating the expression of the fatty-acid receptor CD36 (CD36) and directly activating CD36 in GC cells. Mechanistically, a disulfide bond located between cysteine (Cys)333 and Cys272 within the CD36 protein structure that was labile to H2S-mediated modification. The long chain-fatty acid (LC-FA) binding pocket was capped by a turn in the CD36 protein, located between helical and sheet structures that were stabilized by the Cys333-Cys272. This limited the secondary binding between LC-FAs and lysine (Lys)334. Breaking the Cys333-Cys272 disulfide bond restored the second LC-FA binding conformation of CD36. Targeting CD36 in vivo blocked H2S-promoted metastasis and improved animal survival. INTERPRETATION These findings identify that the Cys333-Cys272 disulfide bond disrupted the integrity of the second LC-FA binding conformation of CD36. Therefore, CD36 can directly activate LC-FA access to the cytoplasm by acting as a direct target molecule for H2S.
Collapse
Affiliation(s)
- Rui Wang
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Beibei Tao
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fudan University, Shanghai 200032, China
| | - Qilin Fan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shengyue Wang
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Li Chen
- Department of Gastroenterology, Baoshan Branch, Renji Hospital, Affiliated to Shanghai Jiaotong University, Shanghai 200444, China
| | - Junjie Zhang
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Yinfang Hao
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Shuang Dong
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Zhe Wang
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Wei Wang
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China
| | - Yixi Cai
- Department of Pediatrics, First People's Hospital of Liangjiang New District, Chongqing 401121, China
| | - Xutong Li
- Department of Neurology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Tuvshin Bao
- Department of Anesthesia, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaohui Wang
- Department of Pancreatic Surgery, State Key Laboratory of Oncology in South China, Zhongshan University, Guangzhou 510001, China
| | - Xiaoming Qiu
- Department of Orthopedics, Provincial Hospital of Gansu Province, Lanzhou 730001, China
| | - Kekun Wang
- School of Health Science, Wuhan University, Wuhan 430030, China
| | - Xinyu Mo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300001, China
| | - Yuqi Kang
- Department of Oncology, Oncology Hospital of Guizhou Province, Guiyang 550001, China
| | - Zhirong Wang
- Department of Gastroenterology, Tongji Hospital, Affiliated to Tongji University, Shanghai 200065, China.
| |
Collapse
|
48
|
Sudhakaran M, Sardesai S, Doseff AI. Flavonoids: New Frontier for Immuno-Regulation and Breast Cancer Control. Antioxidants (Basel) 2019; 8:E103. [PMID: 30995775 PMCID: PMC6523469 DOI: 10.3390/antiox8040103] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/01/2019] [Accepted: 04/05/2019] [Indexed: 12/15/2022] Open
Abstract
Breast cancer (BC) remains the second most common cause of cancer-related deaths in women in the US, despite advances in detection and treatment. In addition, breast cancer survivors often struggle with long-term treatment related comorbidities. Identifying novel therapies that are effective while minimizing toxicity is critical in curtailing this disease. Flavonoids, a subclass of plant polyphenols, are emerging as promising treatment options for the prevention and treatment of breast cancer. Recent evidence suggests that in addition to anti-oxidant properties, flavonoids can directly interact with proteins, making them ideal small molecules for the modulation of enzymes, transcription factors and cell surface receptors. Of particular interest is the ability of flavonoids to modulate the tumor associated macrophage function. However, clinical applications of flavonoids in cancer trials are limited. Epidemiological and smaller clinical studies have been largely hypothesis generating. Future research should aim at addressing known challenges with a broader use of preclinical models and investigating enhanced dose-delivery systems that can overcome limited bioavailability of dietary flavonoids. In this review, we discuss the structure-functional impact of flavonoids and their action on breast tumor cells and the tumor microenvironment, with an emphasis on their clinical role in the prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Meenakshi Sudhakaran
- Department Physiology, Michigan State University, East Lansing, MI 48824, USA.
- Physiology Graduate Program, Michigan State University, East Lansing, MI 48824, USA.
| | - Sagar Sardesai
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Andrea I Doseff
- Department Physiology, Michigan State University, East Lansing, MI 48824, USA.
- Department Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
49
|
Using Mouse and Drosophila Models to Investigate the Mechanistic Links between Diet, Obesity, Type II Diabetes, and Cancer. Int J Mol Sci 2018; 19:ijms19124110. [PMID: 30567377 PMCID: PMC6320797 DOI: 10.3390/ijms19124110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 02/06/2023] Open
Abstract
Many of the links between diet and cancer are controversial and over simplified. To date, human epidemiological studies consistently reveal that patients who suffer diet-related obesity and/or type II diabetes have an increased risk of cancer, suffer more aggressive cancers, and respond poorly to current therapies. However, the underlying molecular mechanisms that increase cancer risk and decrease the response to cancer therapies in these patients remain largely unknown. Here, we review studies in mouse cancer models in which either dietary or genetic manipulation has been used to model obesity and/or type II diabetes. These studies demonstrate an emerging role for the conserved insulin and insulin-like growth factor signaling pathways as links between diet and cancer progression. However, these models are time consuming to develop and expensive to maintain. As the world faces an epidemic of obesity and type II diabetes we argue that the development of novel animal models is urgently required. We make the case for Drosophila as providing an unparalleled opportunity to combine dietary manipulation with models of human metabolic disease and cancer. Thus, combining diet and cancer models in Drosophila can rapidly and significantly advance our understanding of the conserved molecular mechanisms that link diet and diet-related metabolic disorders to poor cancer patient prognosis.
Collapse
|
50
|
Breast Cancer: Metastasis, Molecular Subtypes, and Overweight and Obesity in Veracruz, Mexico. Clin Breast Cancer 2018; 19:e166-e171. [PMID: 30236925 DOI: 10.1016/j.clbc.2018.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/11/2018] [Indexed: 01/08/2023]
Abstract
INTRODUCTION We assessed the association between overweight, obesity, and morbid obesity with the incidence of the most aggressive breast cancer subtypes in women. METHODS AND MATERIALS A cross-sectional study was performed. We conducted a record review to identify the following aspects: body mass index, sociodemographic features, tumor characteristics, and reproductive and molecular aspects. Descriptive statistics and univariate analysis were performed to identify the association between the molecular subtypes and the study variables. In addition, we used multivariate analysis to identify the association between obesity and the presence of metastatic lymph nodes. RESULTS We included 1446 women with an average age of 52.5 ± 12.1 years. Of the 1446 patients, 47% were premenopausal and 75% were overweight. Univariate analysis indicated a statistically significant association between obesity and advanced disease stage, as well as nulliparity and multiparity. Similar results were found for women with morbid obesity. Model 1 of the multivariate analysis showed an association between the presence of metastatic lymph nodes and obesity (odds ratio [OR], 1.6; P = .008) and histologic grade 2 or 3 (OR, 2.4; P = .003). Using model 2, an association was identified between an advanced disease stage and 2 factors: morbid obesity (OR, 1.9; P = .02) and positive human epidermal growth factor receptor 2 (OR, 1.8; P = .045). CONCLUSION We found that obesity is associated with the more advanced stages of breast cancer. Further studies are needed to evaluate the role of obesity in breast cancer progression in women.
Collapse
|