1
|
Solsona-Vilarrasa E, Vousden KH. Obesity, white adipose tissue and cancer. FEBS J 2024. [PMID: 39496581 DOI: 10.1111/febs.17312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024]
Abstract
White adipose tissue (WAT) is crucial for whole-body energy homeostasis and plays an important role in metabolic and hormonal regulation. While healthy WAT undergoes controlled expansion and contraction to meet the body's requirements, dysfunctional WAT in conditions like obesity is characterized by excessive tissue expansion, alterations in lipid homeostasis, inflammation, hypoxia, and fibrosis. Obesity is strongly associated with an increased risk of numerous cancers, with obesity-induced WAT dysfunction influencing cancer development through various mechanisms involving both systemic and local interactions between adipose tissue and tumors. Unhealthy obese WAT affects circulating levels of free fatty acids and factors like leptin, adiponectin, and insulin, altering systemic lipid metabolism and inducing inflammation that supports tumor growth. Similar mechanisms are observed locally in an adipose-rich tumor microenvironment (TME), where WAT cells can also trigger extracellular matrix remodeling, thereby enhancing the TME's ability to promote tumor growth. Moreover, tumors reciprocally interact with WAT, creating a bidirectional communication that further enhances tumorigenesis. This review focuses on the complex interplay between obesity, WAT dysfunction, and primary tumor growth, highlighting potential targets for therapeutic intervention.
Collapse
|
2
|
Tang Y, Chen Z, Zuo Q, Kang Y. Regulation of CD8+ T cells by lipid metabolism in cancer progression. Cell Mol Immunol 2024; 21:1215-1230. [PMID: 39402302 PMCID: PMC11527989 DOI: 10.1038/s41423-024-01224-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/22/2024] [Indexed: 11/02/2024] Open
Abstract
Dysregulation of lipid metabolism is a key characteristic of the tumor microenvironment, where tumor cells utilize lipids for proliferation, survival, metastasis, and evasion of immune surveillance. Lipid metabolism has become a critical regulator of CD8+ T-cell-mediated antitumor immunity, with excess lipids in the tumor microenvironment impeding CD8+ T-cell activities. Considering the limited efficacy of immunotherapy in many solid tumors, targeting lipid metabolism to enhance CD8+ T-cell effector functions could significantly improve immunotherapy outcomes. In this review, we examine recent findings on how lipid metabolic processes, including lipid uptake, synthesis, and oxidation, regulate CD8+ T cells within tumors. We also assessed the impact of different lipids on CD8+ T-cell-mediated antitumor immunity, with a particular focus on how lipid metabolism affects mitochondrial function in tumor-infiltrating CD8+ T cells. Furthermore, as cancer is a systemic disease, we examined systemic factors linking lipid metabolism to CD8+ T-cell effector function. Finally, we summarize current therapeutic approaches that target lipid metabolism to increase antitumor immunity and enhance immunotherapy. Understanding the molecular and functional interplay between lipid metabolism and CD8+ T cells offers promising therapeutic opportunities for cancer treatment.
Collapse
Affiliation(s)
- Yong Tang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Ziqing Chen
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Qianying Zuo
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA.
- Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA.
| |
Collapse
|
3
|
Collins TJC, Morgan PK, Man K, Lancaster GI, Murphy AJ. The influence of metabolic disorders on adaptive immunity. Cell Mol Immunol 2024; 21:1109-1119. [PMID: 39134802 PMCID: PMC11442657 DOI: 10.1038/s41423-024-01206-1] [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: 04/07/2024] [Accepted: 07/19/2024] [Indexed: 10/02/2024] Open
Abstract
The immune system plays a crucial role in protecting the body from invading pathogens and maintaining tissue homoeostasis. Maintaining homoeostatic lipid metabolism is an important aspect of efficient immune cell function and when disrupted immune cell function is impaired. There are numerous metabolic diseases whereby systemic lipid metabolism and cellular function is impaired. In the context of metabolic disorders, chronic inflammation is suggested to be a major contributor to disease progression. A major contributor to tissue dysfunction in metabolic disease is ectopic lipid deposition, which is generally caused by diet and genetic factors. Thus, we propose the idea, that similar to tissue and organ damage in metabolic disorders, excessive accumulation of lipid in immune cells promotes a dysfunctional immune system (beyond the classical foam cell) and contributes to disease pathology. Herein, we review the evidence that lipid accumulation through diet can modulate the production and function of immune cells by altering cellular lipid content. This can impact immune cell signalling, activation, migration, and death, ultimately affecting key aspects of the immune system such as neutralising pathogens, antigen presentation, effector cell activation and resolving inflammation.
Collapse
Affiliation(s)
- Thomas J C Collins
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Pooranee K Morgan
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Kevin Man
- Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
- Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Graeme I Lancaster
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
4
|
Brummer C, Singer K, Brand A, Bruss C, Renner K, Herr W, Pukrop T, Dorn C, Hellerbrand C, Matos C, Kreutz M. Sex-Dependent T Cell Dysregulation in Mice with Diet-Induced Obesity. Int J Mol Sci 2024; 25:8234. [PMID: 39125804 PMCID: PMC11311663 DOI: 10.3390/ijms25158234] [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: 06/16/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Obesity is an emerging public health problem. Chronic low-grade inflammation is considered a major promotor of obesity-induced secondary diseases such as cardiovascular and fatty liver disease, type 2 diabetes mellitus, and several cancer entities. Most preliminary studies on obesity-induced immune responses have been conducted in male rodents. Sex-specific differences between men and women in obesity-induced immune dysregulation have not yet been fully outlined but are highly relevant to optimizing prevention strategies for overweight-associated complications. In this study, we fed C57BL/6 female vs. male mice with either standard chow or an obesity-inducing diet (OD). Blood and spleen immune cells were isolated and analyzed by flow cytometry. Lean control mice showed no sex bias in systemic and splenic immune cell composition, whereas the immune responses to obesity were significantly distinct between female and male mice. While immune cell alterations in male OD mice were characterized by a significant reduction in T cells and an increase in myeloid-derived suppressor cells (MDSC), female OD mice displayed preserved T cell numbers. The sex-dependent differences in obesity-induced T cell dysregulation were associated with varying susceptibility to body weight gain and fatty liver disease: Male mice showed significantly more hepatic inflammation and histopathological stigmata of fatty liver in comparison to female OD mice. Our findings indicate that sex impacts susceptibility to obesity-induced T cell dysregulation, which might explain sex-dependent different incidences in the development of obesity-associated secondary diseases. These results provide novel insights into the understanding of obesity-induced chronic inflammation from a sex-specific perspective. Given that most nutrition, exercise, and therapeutic recommendations for the prevention of obesity-associated comorbidities do not differentiate between men and women, the data of this study are clinically relevant and should be taken into consideration in future trials and treatment strategies.
Collapse
Affiliation(s)
- Christina Brummer
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
- Bavarian Cancer Research Centre (BZKF), 93053 Regensburg, Germany
| | - Katrin Singer
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
- Bavarian Cancer Research Centre (BZKF), 93053 Regensburg, Germany
| | - Almut Brand
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
- Bavarian Cancer Research Centre (BZKF), 93053 Regensburg, Germany
| | - Christina Bruss
- Bavarian Cancer Research Centre (BZKF), 93053 Regensburg, Germany
- Department of Gynecology and Obstetrics, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
| | - Kathrin Renner
- Bavarian Cancer Research Centre (BZKF), 93053 Regensburg, Germany
- Department of Otorhinolaryngology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
- Bavarian Cancer Research Centre (BZKF), 93053 Regensburg, Germany
| | - Tobias Pukrop
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
- Bavarian Cancer Research Centre (BZKF), 93053 Regensburg, Germany
- Comprehensive Cancer Center Eastern Bavaria (CCCO), 93053 Regensburg, Germany
- Center of Translational Oncology (CTO), 93053 Regensburg, Germany
| | - Christoph Dorn
- Institute of Pharmacy, University of Regensburg, 93053 Regensburg, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, University of Erlangen, 91054 Erlangen, Germany
| | - Carina Matos
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
- Bavarian Cancer Research Centre (BZKF), 93053 Regensburg, Germany
| | - Marina Kreutz
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
- Bavarian Cancer Research Centre (BZKF), 93053 Regensburg, Germany
| |
Collapse
|
5
|
Kong YH, Huang JY, Ding Y, Chen SH, Li QS, Xiong Y. The effect of BMI on survival outcome of breast cancer patients: a systematic review and meta-analysis. Clin Transl Oncol 2024:10.1007/s12094-024-03563-9. [PMID: 39012453 DOI: 10.1007/s12094-024-03563-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/07/2024] [Indexed: 07/17/2024]
Abstract
OBJECTIVE The main goal of the present research is to explore the potential link of body mass index (BMI) with different survival metrics in breast cancer patients. Our aim is to offer the latest and most thorough meta-analysis, assessing the strength and reliability of the connection that BMI has with prognostic indicators in this disease. PATIENTS AND METHODS As of January 2024, we conducted a systematic literature search across PubMed, Embase, Web of Science, and the Cochrane Library databases. Our search aimed to identify studies examining BMI as an exposure factor, with breast cancer patients constituting the study population, and utilizing adjusted hazard ratio (HR) as the data type of interest. RESULTS The evidence synthesis incorporated a total of 61 eligible articles involving 201,006 patients. Being underweight posed a risk factor for overall survival (OS) in breast cancer patients compared to normal weight (HR 1.15, 95% CI 0.98-1.35; P = 0.08). Overweight or obesity, in comparison to normal weight, was a risk factor for OS (HR 1.18, 95% CI 1.14-1.23; P < 0.00001), disease-free survival (DFS) (HR 1.11, 95% CI 1.08-1.13; P < 0.00001), relapse-free survival (RFS) (HR 1.14, 95% CI 1.06-1.22; P = 0.03), and breast cancer-specific survival (BCSS) (HR 1.18, 95% CI 1.11-1.26; P < 0.00001), but not for progression-free survival (PFS) (HR 0.91, 95% CI 0.76-1.10; P = 0.33). Notably, in subgroup analyses, overweight patients achieved prolonged PFS (HR 0.80, 95% CI 0.64-0.99; P = 0.04), and compared to the obese population, the overweight cohort exhibited a significant difference in OS (HR 1.11, 95% CI 1.05-1.16; P < 0.00001) and DFS (HR 1.06, 95% CI 1.03-1.10; P = 0.0004), with a considerably stronger association. Furthermore, compared to HER- patients, HER + patients exhibited a greater predictive value for OS (HR 1.23, 95% CI 1.10-1.37; P = 0.0004), RFS (HR 1.30, 95% CI 1.03-1.64; P < 0.00001), and DFS (HR 1.10, 95% CI 1.03-1.17; P = 0.003). CONCLUSIONS The results of our meta-analysis reveal a notable association between BMI and various survival measures in breast cancer prognosis. These findings provide a solid basis for predicting breast cancer outcomes and implementing more effective therapeutic approaches.
Collapse
Affiliation(s)
- Yu-Huan Kong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Jing-Yi Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Ye Ding
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Shu-Hua Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Qiu-Shuang Li
- Center of Clinical Evaluation and Analysis, Zhejiang Provincial Hospital of Chinese Medicine), The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Yang Xiong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
6
|
Zhu G, Cao L, Wu J, Xu M, Zhang Y, Wu M, Li J. Co-morbid intersections of cancer and cardiovascular disease and targets for natural drug action: Reprogramming of lipid metabolism. Biomed Pharmacother 2024; 176:116875. [PMID: 38850662 DOI: 10.1016/j.biopha.2024.116875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/24/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Cancer and cardiovascular diseases are major contributors to global morbidity and mortality, and their seemingly separate pathologies are intricately intertwined. In the context of cancer, the cardiovascular disease encompasses not only the side effects arising from anti-tumor treatments but also the metabolic shifts induced by oncological conditions. A growing body of research indicates that lipid metabolic reprogramming serves as a distinctive hallmark of tumors. Furthermore, anomalies in lipid metabolism play a significant role in the development of cardiovascular disease. This study delves into the cardiac implications of lipid metabolic reprogramming within the cancer context, closely examining abnormalities in lipid metabolism present in tumors, cardiac tissue, and immune cells within the microenvironment. Additionally, we examined risk factors such as obesity and anti-tumor therapy. Despite progress, a gap remains in the availability of drugs targeting lipid metabolism modulation for treating tumors and mitigating cardiac risk, with limited advancement seen in prior studies. Here, we present a review of previous research on natural drugs that exhibit both shared and distinct therapeutic effects on tumors and cardiac health by modulating lipid metabolism. Our aim is to provide insights for potential drug development.
Collapse
Affiliation(s)
- Guanghui Zhu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Luchang Cao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jingyuan Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Manman Xu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ying Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Min Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Jie Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
7
|
Son DS, Done KA, Son J, Izban MG, Virgous C, Lee ES, Adunyah SE. Intermittent Fasting Attenuates Obesity-Induced Triple-Negative Breast Cancer Progression by Disrupting Cell Cycle, Epithelial-Mesenchymal Transition, Immune Contexture, and Proinflammatory Signature. Nutrients 2024; 16:2101. [PMID: 38999849 PMCID: PMC11243652 DOI: 10.3390/nu16132101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
Obesity is associated with one-fifth of cancer deaths, and breast cancer is one of the obesity-related cancers. Triple-negative breast cancer (TNBC) lacks estrogen and progesterone receptors and human epidermal growth factor receptor 2, leading to the absence of these therapeutic targets, followed by poor overall survival. We investigated if obesity could hasten TNBC progression and intermittent fasting (IF) could attenuate the progression of obesity-related TNBC. Our meta-analysis of the TNBC outcomes literature showed that obesity led to poorer overall survival in TNBC patients. Fasting-mimicking media reduced cell proliferation disrupted the cell cycle, and decreased cell migration and invasion. IF decreased body weight in obese mice but no change in normal mice. Obese mice exhibited elevated plasma glucose and cholesterol levels, increased tumor volume and weight, and enhanced macrophage accumulation in tumors. The obesity-exacerbated TNBC progression was attenuated after IF, which decreased cyclin B1 and vimentin levels and reduced the proinflammatory signature in the obesity-associated tumor microenvironment. IF attenuated obesity-induced TNBC progression through reduced obesity and tumor burdens in cell and animal experiments, supporting the potential of a cost-effective adjuvant IF therapy for TNBC through lifestyle change. Further evidence is needed of these IF benefits in TNBC, including from human clinical trials.
Collapse
Affiliation(s)
- Deok-Soo Son
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA;
| | - Kaitlyn A. Done
- Biochemistry Program, College of Arts and Sciences, Spelman College, Atlanta, GA 30314, USA
| | - Jubin Son
- Neuroscience Program, College of Arts and Sciences, The University of Tennessee, Knoxville, TN 37996, USA
| | - Michael G. Izban
- Pathology, Anatomy and Cell Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Carlos Virgous
- Animal Core Facility, Meharry Medical College, Nashville, TN 37208, USA
| | - Eun-Sook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL 32301, USA;
| | - Samuel E. Adunyah
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
8
|
Cunha A, Perazzio S. Effects of immune exhaustion and senescence of innate immunity in autoimmune disorders. Braz J Med Biol Res 2024; 57:e13225. [PMID: 38896644 PMCID: PMC11186593 DOI: 10.1590/1414-431x2024e13225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/22/2024] [Indexed: 06/21/2024] Open
Abstract
Innate immune system activation is crucial in the inflammatory response, but uncontrolled activation can lead to autoimmune diseases. Cellular exhaustion and senescence are two processes that contribute to innate immune tolerance breakdown. Exhausted immune cells are unable to respond adequately to specific antigens or stimuli, while senescent cells have impaired DNA replication and metabolic changes. These processes can impair immune system function and disrupt homeostasis, leading to the emergence of autoimmunity. However, the influence of innate immune exhaustion and senescence on autoimmune disorders is not well understood. This review aims to describe the current findings on the role of innate immune exhaustion and senescence in autoimmunity, focusing on the cellular and molecular changes involved in each process. Specifically, the article explores the markers and pathways associated with immune exhaustion, such as PD-1 and TIM-3, and senescence, including Β-galactosidase (β-GAL), lamin B1, and p16ink4a, and their impact on autoimmune diseases, namely type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus, and immune-mediated myopathies. Understanding the mechanisms underlying innate immune exhaustion and senescence in autoimmunity may provide insights for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- A.L.S. Cunha
- Divisão de Reumatologia, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - S.F. Perazzio
- Divisão de Reumatologia, Universidade Federal de São Paulo, São Paulo, SP, Brasil
- Divisão de Imunologia, Laboratório Fleury, São Paulo, SP, Brasil
- Laboratório Central, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
9
|
Nedal TMV, Moen SH, Roseth IA, Tryggestad SS, Aass KR, Hov GG, Hella H, Sponaas AM, Standal T. Diet-induced obesity reduces bone marrow T and B cells and promotes tumor progression in a transplantable Vk*MYC model of multiple myeloma. Sci Rep 2024; 14:3643. [PMID: 38351079 PMCID: PMC10864380 DOI: 10.1038/s41598-024-54193-8] [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: 11/20/2023] [Accepted: 02/09/2024] [Indexed: 02/16/2024] Open
Abstract
Obesity is associated with an increased risk of developing multiple myeloma (MM). The molecular mechanisms causing this association is complex and incompletely understood. Whether obesity affects bone marrow immune cell composition in multiple myeloma is not characterized. Here, we examined the effect of diet-induced obesity on bone marrow immune cell composition and tumor growth in a Vk*MYC (Vk12653) transplant model of multiple myeloma. We find that diet-induced obesity promoted tumor growth in the bone marrow and spleen and reduced the relative number of T and B cells in the bone marrow. Our results suggest that obesity may reduce MM immune surveillance and thus may contribute to increased risk of developing MM.
Collapse
Affiliation(s)
- Tonje Marie Vikene Nedal
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Siv Helen Moen
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Research, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Ingrid Aass Roseth
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Synne Stokke Tryggestad
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Kristin Roseth Aass
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Gunhild Garmo Hov
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Clinical Chemistry, St. Olavs Hospital, Trondheim, Norway
| | - Hanne Hella
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Anne-Marit Sponaas
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Therese Standal
- Department of Clinical and Molecular Medicine, Centre of Molecular Inflammation Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- Department of Hematology, St. Olavs Hospital, Trondheim, Norway.
| |
Collapse
|
10
|
Asadipour M, Ataollahi M, Shams K, Ali-Hassanzadeh M, Martinuzzi E, Kalantar K. ADIPOPHILIN PEPTIDE (ADPH 129-137) IS NOT A TARGET ANTIGEN FOR CD8 + T-CELLS IN PATIENTS WITH OBESITY. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2024; 20:21-26. [PMID: 39372296 PMCID: PMC11449246 DOI: 10.4183/aeb.2024.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Context In obesity, the infiltration of leukocytes into adipose tissue seems to play a key role in the development of inflammation and insulin resistance. Over-expression of adipophilin (ADPH) in adipose tissue, a protein which regulates lipid droplet structure and formation, has been reported in some studies. Objective To investigate the role of ADPH 129-137 as a target for CD8+ T-cells in PBMCs of patients with obesity. Subjects and Methods PBMCs were obtained from 9 non-diabetic obese patients and 11 healthy subjects expressing the HLA-A0201 molecule. The ELISPOT assay used to monitor the presence of IFN-γ producing CD8+ T-cells against a HLA class I-restricted epitope derived from Adipophilin (ADPH 129-137) and two control peptides: Flu MP58-66 and Melan-A27-35. Results The outcomes showed no significant difference between patient group and healthy donors in response to ADPH 129-137. Conclusion These results demonstrated that ADPH 129-137 peptide possibly does not act as an autoantigen in patients with obesity.
Collapse
Affiliation(s)
- M. Asadipour
- Shiraz University of Medical Sciences, School of Medicine, Department of Immunology, Shiraz
| | - M.R. Ataollahi
- Fasa University of Medical Sciences, School of Medicine, Department of Immunology, Fasa
| | - K. Shams
- Shiraz University of Medical Sciences, School of Medicine, Department of Immunology, Shiraz
| | - M. Ali-Hassanzadeh
- Jiroft University of Medical Sciences, School of Medicine, Department of Immunology, Jiroft, Iran
| | - E. Martinuzzi
- Système Immunitaire, Cerveau et Nerfs Périphériques, Institute de Pharmacologie Moléculaire et Cellulaire (IPMC) CNRS, Valbonne, France
| | - K. Kalantar
- Shiraz University of Medical Sciences, School of Medicine, Department of Immunology, Shiraz
- Shiraz University of Medical Sciences, Autoimmune Diseases Research Center, Shiraz, Iran
| |
Collapse
|
11
|
Araújo M, Moreira D, Mesquita I, Ferreira C, Mendes-Frias A, Barros-Carvalho S, Dinis-Oliveira RJ, Duarte-Oliveira C, Cunha C, Carvalho A, Saha B, Cordeiro-da-Silva A, Estaquier J, Silvestre R. Intramacrophage lipid accumulation compromises T cell responses and is associated with impaired drug therapy against visceral leishmaniasis. Immunology 2023; 170:510-526. [PMID: 37635289 DOI: 10.1111/imm.13686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/14/2023] [Indexed: 08/29/2023] Open
Abstract
Under perturbing conditions such as infection with Leishmania, a protozoan parasite living within the phagosomes in mammalian macrophages, cellular and organellar structures, and metabolism are dynamically regulated for neutralizing the pressure of parasitism. However, how modulations of the host cell metabolic pathways support Leishmania infection remains unknown. Herein, we report that lipid accumulation heightens the susceptibility of mice to L. donovani infection and promotes resistance to first-line anti-leishmanial drugs. Despite being pro-inflammatory, the in vitro generated uninfected lipid-laden macrophages (LLMs) or adipose-tissue macrophages (ATMs) display lower levels of reactive oxygen and nitrogen species. Upon infection, LLMs secrete higher IL-10 and lower IL-12p70 cytokines, inhibiting CD4+ T cell activation and Th1 response suggesting a key modulatory role for intramacrophage lipid accumulation in anti-leishmanial host defence. We, therefore, examined this causal relationship between lipids and immunomodulation using an in vivo high-fat diet (HFD) mouse model. HFD increased the susceptibility to L. donovani infection accompanied by a defective CD4+ Th1 and CD8+ T cell response. The white adipose tissue of HFD mice displays increased susceptibility to L. donovani infection with the preferential infection of F4/80+ CD11b+ CD11c+ macrophages with higher levels of neutral lipids reserve. The HFD increased resistance to a first-line anti-leishmanial drug associated with a defective adaptive immune response. These data demonstrate that the accumulation of neutral lipids contributes to susceptibility to visceral leishmaniasis hindering host-protective immune response and reducing the efficacy of antiparasitic drug therapies.
Collapse
Affiliation(s)
- Marta Araújo
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Diana Moreira
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal
| | - Inês Mesquita
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Carolina Ferreira
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Mendes-Frias
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sónia Barros-Carvalho
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- TOXRUN - Toxicology Research Unit, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal
- MTG Research and Development Lab, Porto, Portugal
| | - Cláudio Duarte-Oliveira
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina Cunha
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Agostinho Carvalho
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | - Anabela Cordeiro-da-Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal
| | - Jérôme Estaquier
- INSERM U1124, Université Paris Cité, Paris, France
- Pathophysiology of Cell Death in Host-Pathogen Interactions, CHU de Québec - Université Laval Research Center, Québec City, Québec, Canada
| | - Ricardo Silvestre
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
12
|
Avtanski D, Reddy V, Stojchevski R, Hadzi-Petrushev N, Mladenov M. The Microbiome in the Obesity-Breast Cancer Axis: Diagnostic and Therapeutic Potential. Pathogens 2023; 12:1402. [PMID: 38133287 PMCID: PMC10747404 DOI: 10.3390/pathogens12121402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
A growing body of evidence has demonstrated a relationship between the microbiome, adiposity, and cancer development. The microbiome is emerging as an important factor in metabolic disease and cancer pathogenesis. This review aimed to highlight the role of the microbiome in obesity and its association with cancer, with a particular focus on breast cancer. This review discusses how microbiota dysbiosis may contribute to obesity and obesity-related diseases, which are linked to breast cancer. It also explores the potential of the gut microbiome to influence systemic immunity, leading to carcinogenesis via the modulation of immune function. This review underscores the potential use of the microbiome profile as a diagnostic tool and treatment target, with strategies including probiotics, fecal microbiota transplantation, and dietary interventions. However, this emphasizes the need for more research to fully understand the complex relationship between the microbiome, metabolic disorders, and breast cancer. Future studies should focus on elucidating the mechanisms underlying the impact of the microbiome on breast cancer and exploring the potential of the microbiota profile as a biomarker and treatment target.
Collapse
Affiliation(s)
- Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Varun Reddy
- New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 11545, USA;
| | - Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.); (M.M.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.); (M.M.)
| |
Collapse
|
13
|
Yang R, Yang M, Wu Z, Liu B, Zheng M, Lu L, Wu S. Tespa1 deficiency reduces the antitumour immune response by decreasing CD8 +T cell activity in a mouse Lewis lung cancer model. Int Immunopharmacol 2023; 124:110865. [PMID: 37660596 DOI: 10.1016/j.intimp.2023.110865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023]
Abstract
Thymocyte-expressed, positive selection-associated 1 (Tespa1) is a key molecule in T-cell development and has been linked to immune diseases. However, its role in antitumour CD8+T cell immunity remains unclear. Here, we demonstrated that Tespa1 plays an important role in antitumour CD8+T cell immunity. First, compared with wild-type (WT) mice, Lewis lung cancer cells grew faster in Tespa1 knockout (Tespa1-/-) mice, with reduced apoptosis, and decreased CD8+T cells in peripheral blood and tumor tissues. Second, the proportion of CD8+T and Th1 cells in the splenocytes of Tespa1-/- mice was lower than that in WT mice. Third, Tespa1-/- CD8+ tumor-infiltrating lymphocytes (TILs) showed weakened proliferation, invasion, cytotoxicity, and protein expression of IL-2 signalling pathway components compared to WT CD8+TILs. Furthermore, PD-1 expression in CD8+TILs was higher in Tespa1-/- than in WT mice. Lastly, CD8+TILs in WT mice improved the antitumour ability of Tespa1-/- mice. In conclusion, these findings suggest that Tespa1 plays a critical role in the tumor immune system by regulating CD8+T cells.
Collapse
Affiliation(s)
- Ruhui Yang
- School of Medicine and Pharmaceutical Engineering, Taizhou Vocational and Technical College, Taizhou 318000, China; Department of Pharmacology, Lishui University School of Medicine, Lishui 323000, China
| | - Mingyue Yang
- The First Clinical Department, China Medical University, Shenyang 110122, China
| | - Zehua Wu
- Faculty of Science and Engineering, University of Nottingham, Ningbo, 315000, China
| | - Bingjin Liu
- School of Medicine and Pharmaceutical Engineering, Taizhou Vocational and Technical College, Taizhou, 318000, China
| | - Mingzhu Zheng
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, 210003, China
| | - Linrong Lu
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Songquan Wu
- Department of Immunology, Lishui University School of Medicine, Lishui 323000, China.
| |
Collapse
|
14
|
Vick LV, Canter RJ, Monjazeb AM, Murphy WJ. Multifaceted effects of obesity on cancer immunotherapies: Bridging preclinical models and clinical data. Semin Cancer Biol 2023; 95:88-102. [PMID: 37499846 PMCID: PMC10836337 DOI: 10.1016/j.semcancer.2023.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/04/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Obesity, defined by excessive body fat, is a highly complex condition affecting numerous physiological processes, such as metabolism, proliferation, and cellular homeostasis. These multifaceted effects impact cells and tissues throughout the host, including immune cells as well as cancer biology. Because of the multifaceted nature of obesity, common parameters used to define it (such as body mass index in humans) can be problematic, and more nuanced methods are needed to characterize the pleiotropic metabolic effects of obesity. Obesity is well-accepted as an overall negative prognostic factor for cancer incidence, progression, and outcome. This is in part due to the meta-inflammatory and immunosuppressive effects of obesity. Immunotherapy is increasingly used in cancer therapy, and there are many different types of immunotherapy approaches. The effects of obesity on immunotherapy have only recently been studied with the demonstration of an "obesity paradox", in which some immune therapies have been demonstrated to result in greater efficacy in obese subjects despite the direct adverse effects of obesity and excess body fat acting on the cancer itself. The multifactorial characteristics that influence the effects of obesity (age, sex, lean muscle mass, underlying metabolic conditions and drugs) further confound interpretation of clinical data and necessitate the use of more relevant preclinical models mirroring these variables in the human scenario. Such models will allow for more nuanced mechanistic assessment of how obesity can impact, both positively and negatively, cancer biology, host metabolism, immune regulation, and how these intersecting processes impact the delivery and outcome of cancer immunotherapy.
Collapse
Affiliation(s)
- Logan V Vick
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Robert J Canter
- Department of Surgery, Division of Surgical Oncology, University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Arta M Monjazeb
- Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, University of California School of Medicine, Sacramento, CA, USA
| | - William J Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA; Department of Internal Medicine, Division of Malignant Hematology, Cellular Therapy and Transplantation, University of California Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
15
|
Jin HR, Wang J, Wang ZJ, Xi MJ, Xia BH, Deng K, Yang JL. Lipid metabolic reprogramming in tumor microenvironment: from mechanisms to therapeutics. J Hematol Oncol 2023; 16:103. [PMID: 37700339 PMCID: PMC10498649 DOI: 10.1186/s13045-023-01498-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
Lipid metabolic reprogramming is an emerging hallmark of cancer. In order to sustain uncontrolled proliferation and survive in unfavorable environments that lack oxygen and nutrients, tumor cells undergo metabolic transformations to exploit various ways of acquiring lipid and increasing lipid oxidation. In addition, stromal cells and immune cells in the tumor microenvironment also undergo lipid metabolic reprogramming, which further affects tumor functional phenotypes and immune responses. Given that lipid metabolism plays a critical role in supporting cancer progression and remodeling the tumor microenvironment, targeting the lipid metabolism pathway could provide a novel approach to cancer treatment. This review seeks to: (1) clarify the overall landscape and mechanisms of lipid metabolic reprogramming in cancer, (2) summarize the lipid metabolic landscapes within stromal cells and immune cells in the tumor microenvironment, and clarify their roles in tumor progression, and (3) summarize potential therapeutic targets for lipid metabolism, and highlight the potential for combining such approaches with other anti-tumor therapies to provide new therapeutic opportunities for cancer patients.
Collapse
Affiliation(s)
- Hao-Ran Jin
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Wang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zi-Jing Wang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Ming-Jia Xi
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Bi-Han Xia
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Deng
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China.
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Jin-Lin Yang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China.
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
16
|
McIntyre CL, Temesgen A, Lynch L. Diet, nutrient supply, and tumor immune responses. Trends Cancer 2023; 9:752-763. [PMID: 37400315 DOI: 10.1016/j.trecan.2023.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 07/05/2023]
Abstract
Nutrients are essential for cell function. Immune cells operating in the complex tumor microenvironment (TME), which has a unique nutrient composition, face challenges of adapting their metabolism to support effector functions. We discuss the impact of nutrient availability on immune function in the tumor, competition between immune cells and tumor cells for nutrients, and how this is altered by diet. Understanding which diets can promote antitumor immune responses could open a new era of treatment, where dietary modifications can be used as an adjunct to boost the success of existing cancer therapies.
Collapse
Affiliation(s)
- Claire L McIntyre
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ayantu Temesgen
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lydia Lynch
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
17
|
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: 2.0] [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
|
18
|
Wei W, Wei S, Huang Z, Zhang Q, Liu F, Xie Y, Wei J, Mo C, Zhou Y, Qin S, Zou Q, Yang J. The relationship between women's body mass index and breast cancer outcomes was U-shaped. Front Oncol 2023; 13:1191093. [PMID: 37305586 PMCID: PMC10248463 DOI: 10.3389/fonc.2023.1191093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Background Several studies have analyzed the relationship between body mass index (BMI) and the prognosis of breast cancer (BC). However, whether their relationship is linear or curvilinear remains unclear. This cohort study examined the specific relationship between BMI and BC outcomes. Methods This retrospective cohort study included 1049 BC patients from March 7, 2013 through December 31, 2019 in a hospital. Kaplan-Meier curves, multivariate Cox proportional models, and restricted cubic spline (RCS) was used to analysis the relationship between BMI and overall survival (OS) and breast cancer-specific survival (BCSS) was analyzed. Results During a median of 4.87 (IQR:3.26-6.84) years of follow-up period, 71 patients (6.77%) died, of which 50 (70.42%) were attributed to BC. RCS analysis revealed a U- shaped relationship between BMI levels and OS and BCSS after adjusting for other variables. The turning points of the U-shaped curves were 23 kg/m2. On the left side of the turning point, the risk of OS (HR, 0.83; 95% CI, 0.70, 0.98) and BCSS (HR, 0.80; 95% CI, 0.65, 0.98) were adversely correlated with BMI. In contrast, to the right of the turning point, the risk of OS (HR, 1.22; 95% CI, 1.10, 1.37) and BCSS (HR, 1.28; 95% CI, 1.13, 1.46) was positively related to BMI. Kaplan-Meier curves and multivariate Cox regression analyses shown consistent results with RCS analyses. Conclusion BMI was an independent prognostic factor for BC, and had a U-shaped relationship with OS and BCSS. Interventions should be designed to improve patient outcomes based on BMI.
Collapse
Affiliation(s)
- Wensong Wei
- Department of Breast and Thyroid Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Suosu Wei
- Department of Scientific Cooperation of Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Zhen Huang
- Department of Breast and Thyroid Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Qiuhuan Zhang
- Department of Colorectal and Anal Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Fei Liu
- Research Center of Medical Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guang-xi Academy of Medical Sciences, Nanning, Guangxi, China
| | - Yujie Xie
- Department of Breast and Thyroid Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Jing Wei
- Department of Breast and Thyroid Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Chongde Mo
- Department of Breast and Thyroid Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Ying Zhou
- Department of Breast and Thyroid Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Shengpeng Qin
- Department of Breast and Thyroid Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Quanqing Zou
- Department of Breast and Thyroid Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Jianrong Yang
- Department of Breast and Thyroid Surgery, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| |
Collapse
|
19
|
Ayed K, Nabi L, Akrout R, Mrizak H, Gorrab A, Bacha D, Boussen H, Gati A. Obesity and cancer: focus on leptin. Mol Biol Rep 2023:10.1007/s11033-023-08525-y. [PMID: 37227675 DOI: 10.1007/s11033-023-08525-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023]
Abstract
Over the past decades, obesity has grown to epidemic proportions worldwide. It has been associated with an increased risk for different types of cancer. In addition, obesity has been associated with a poor prognosis, an increased risk of metastasis and mortality, and resistance to anti-cancer therapies. The pathophysiological mechanisms underlying the obesity-cancer connection have not yet been fully elucidated. However, this connection could result, at least in part, from the action of adipokines, whose levels are increased in obesity. Among these adipokines, evidence suggests leptin's critical role in linking obesity to cancer. In this review, we first summarize the current state of the literature regarding the implication of leptin in tumorigenic processes. Next, we focus on the effects of leptin on the anti-tumor immune response. Then, we discuss the influence of leptin on the efficiency of antineoplastic treatments and the development of tumor resistance. Finally, we highlight the use of leptin as a potential target for the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Khouloud Ayed
- Laboratory of Genetics, Immunology, and Human Pathology, Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Lamis Nabi
- Laboratory of Genetics, Immunology, and Human Pathology, Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Rym Akrout
- Laboratory of Genetics, Immunology, and Human Pathology, Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Hela Mrizak
- Laboratory of Genetics, Immunology, and Human Pathology, Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Amal Gorrab
- Laboratory of Genetics, Immunology, and Human Pathology, Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Dhouha Bacha
- Anatomopathology Department, Mongi Slim Hospital, Tunis, Tunisia
| | - Hamouda Boussen
- Medical Oncology Department, Salah Azaiez Institute, Faculty of Medicine of Tunis, University of Tunis El Manar, Ariana, Tunisia
| | - Asma Gati
- Laboratory of Genetics, Immunology, and Human Pathology, Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia.
| |
Collapse
|
20
|
Diverse effects of obesity on antitumor immunity and immunotherapy. Trends Mol Med 2023; 29:112-123. [PMID: 36473793 DOI: 10.1016/j.molmed.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 12/07/2022]
Abstract
Currently, obesity is one of the biggest health burdens facing society because it causes several comorbidities, such as type 2 diabetes, atherosclerosis, and heart disease. Obesity is also linked to multiple types of cancer. Obesity is the second most common preventable cause of cancer after smoking; the rates of obesity are increasing worldwide, as are the rates of obesity-associated cancer. Multiple factors link obesity to cancer, such as increased levels of growth hormones and adipokines, gut dysbiosis, altered tumor metabolism, and chronic low-grade inflammation. More recently, obesity has been shown to also affect the immune response against cancer. In this review we discuss the interplay between obesity, the immune system, and cancer.
Collapse
|
21
|
Vick LV, Collins CP, Khuat LT, Wang Z, Dunai C, Aguilar EG, Stoffel K, Yendamuri S, Smith R, Mukherjee S, Barbi J, Canter RJ, Monjazeb AM, Murphy WJ. Aging augments obesity-induced thymic involution and peripheral T cell exhaustion altering the "obesity paradox". Front Immunol 2023; 13:1012016. [PMID: 36776393 PMCID: PMC9910174 DOI: 10.3389/fimmu.2022.1012016] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/07/2022] [Indexed: 01/28/2023] Open
Abstract
Introduction The incidence of obesity, a condition characterized by systemic chronic inflammation, has reached pandemic proportions and is a poor prognostic factor in many pathologic states. However, its role on immune parameters has been diverse and at times contradictory. We have previously demonstrated that obesity can result in what has been called the "obesity paradox" which results in increased T cell exhaustion, but also greater efficacy of immune checkpoint blockade in cancer treatment. Methods The role of obesity, particularly in the context of aging, has not been robustly explored using preclinical models. We therefore evaluated how age impacts the immune environment on T cell development and function using diet-induced obese (DIO) mice. Results We observed that DIO mice initially displayed greater thymopoiesis but then developed greater thymic involution over time compared to their lean counterparts. Both aging and obesity resulted in increased T cell memory conversion combined with increased expression of T cell exhaustion markers and Treg expansion. This increased T cell immunosuppression with age then resulted in a loss of anti-tumor efficacy by immune checkpoint inhibitors (ICIs) in older DIO mice compared to the younger DIO counterparts. Discussion These results suggest that both aging and obesity contribute to T cell dysfunction resulting in increased thymic involution. This combined with increased T cell exhaustion and immunosuppressive parameters affects immunotherapy efficacy reducing the advantage of obesity in cancer immunotherapy responses.
Collapse
Affiliation(s)
- Logan V. Vick
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Craig P. Collins
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Lam T. Khuat
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Ziming Wang
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Cordelia Dunai
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Ethan G. Aguilar
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Kevin Stoffel
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Sai Yendamuri
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Randall Smith
- Department of Immunology Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Sarbajit Mukherjee
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Joseph Barbi
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- Department of Immunology Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Robert J. Canter
- Division of Surgical Oncology, Department of Surgery, University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Arta M. Monjazeb
- Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, University of California School of Medicine, Sacramento, CA, United States
| | - William J. Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
- Department of Internal Medicine, Division of Hematology and Oncology, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
22
|
Park J, Hsueh PC, Li Z, Ho PC. Microenvironment-driven metabolic adaptations guiding CD8 + T cell anti-tumor immunity. Immunity 2023; 56:32-42. [PMID: 36630916 DOI: 10.1016/j.immuni.2022.12.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/04/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023]
Abstract
The metabolic stress occurring in the tumor microenvironment (TME) hampers T cell anti-tumor immunity by disturbing T cell metabolic and epigenetic programs. Recent studies are making headway toward identifying strategies to unleash T cell activities by targeting T cell metabolism. Furthermore, efforts have been made to improve the efficacy of immune checkpoint blockade and adoptive cell transfer therapies. However, distinct treatment outcomes across different cancers raise the question of whether our understanding of the features of CD8+ T cells within the TME are universal, regardless of their tissue of origin. Here, we review the common and distinct environmental factors affecting CD8+ T cells across tumors. Moreover, we discuss how distinct tissue-specific niches are interpreted by CD8+ T cells based on studies on tissue-resident memory T (Trm) cells and how these insights can pave the way for a better understanding of the metabolic regulation of CD8+ T cell differentiation and anti-tumor immunity.
Collapse
Affiliation(s)
- Jaeoh Park
- Department of Fundamental Oncology, University of Lausanne, 1066 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland.
| | - Pei-Chun Hsueh
- Department of Fundamental Oncology, University of Lausanne, 1066 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland.
| | - Zhiyu Li
- Department of Fundamental Oncology, University of Lausanne, 1066 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland; Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, 1066 Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland.
| |
Collapse
|
23
|
Smith LA, Craven DM, Rainey MA, Cozzo AJ, Carson MS, Glenny EM, Sheth N, McDonell SB, Rezeli ET, Montgomery SA, Bowers LW, Coleman MF, Hursting SD. Separate and combined effects of advanced age and obesity on mammary adipose inflammation, immunosuppression and tumor progression in mouse models of triple negative breast cancer. Front Oncol 2023; 12:1031174. [PMID: 36686775 PMCID: PMC9846347 DOI: 10.3389/fonc.2022.1031174] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/31/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction Advanced age and obesity are independent risk and progression factors for triple negative breast cancer (TNBC), which presents significant public health concerns for the aging population and its increasing burden of obesity. Due to parallels between advanced age- and obesityrelated biology, particularly adipose inflammation, we hypothesized that advanced age and obesity each accelerate mammary tumor growth through convergent, and likely interactive, mechanisms. Methods To test this hypothesis, we orthotopically transplanted murine syngeneic TNBC cells into the mammary glands of young normoweight control (7 months), young diet-induced obese (DIO), aged normoweight control (17 months), and aged DIO female C57BL/6J mice. Results Here we report accelerated tumor growth in aged control and young DIO mice, compared with young controls. Transcriptional analyses revealed, with a few exceptions, overlapping patterns of mammary tumor inflammation and tumor immunosuppression in aged control mice and young DIO mice, relative to young controls. Moreover, aged control and young DIO tumors, compared with young controls, had reduced abundance ofcytotoxic CD8 T cells. Finally, DIO in advanced age exacerbated mammary tumor growth, inflammation and tumor immunosuppression. Discussion These findings demonstrate commonalities in the mechanisms driving TNBC in aged and obese mice, relative to young normoweight controls. Moreover, we found that advanced age and DIO interact to accelerate mammary tumor progression. Given the US population is getting older and more obese, age- and obesity-related biological differences will need to be considered when developing mechanism-based strategies for preventing or controlling breast cancer.
Collapse
Affiliation(s)
- Laura A. Smith
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Dalton M. Craven
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Magdalena A. Rainey
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Alyssa J. Cozzo
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Meredith S. Carson
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Elaine M. Glenny
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Nishita Sheth
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Shannon B. McDonell
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Erika T. Rezeli
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Stephanie A. Montgomery
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Laura W. Bowers
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michael F. Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Stephen D. Hursting
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
24
|
Yamada K, Saito M, Ando M, Abe T, Mukoyama T, Agawa K, Watanabe A, Takamura S, Fujita M, Urakawa N, Hasegawa H, Kanaji S, Matsuda T, Oshikiri T, Kakeji Y, Yamashita K. Reduced Number and Immune Dysfunction of CD4+ T Cells in Obesity Accelerate Colorectal Cancer Progression. Cells 2022; 12:cells12010086. [PMID: 36611881 PMCID: PMC9818365 DOI: 10.3390/cells12010086] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Obesity, a known risk factor for various types of cancer, reduces the number and function of cytotoxic immune cells in the tumor immune microenvironment (TIME). However, the impact of obesity on CD4+ T cells remains unclear. Therefore, this study aimed to clarify the impact of obesity on CD4+ T cells in the TIME. A tumor-bearing obese mouse model was established by feeding with 45% high-fat diet (HFD), followed by inoculation with a colon cancer cell line MC38. Tumor growth was significantly accelerated compared to that in mice fed a control diet. Tumor CD4+ T cells showed a significant reduction in number and an increased expression of programmed death-1 (PD-1), and decreased CD107a expression and cytokine such as IFN-γ and TNF-α production, indicating dysfunction. We further established CD4+ T cell-depleted HFD-fed model mice, which showed reduced tumor infiltration, increased PD-1 expression in CD8+ T cells, and obesity-induced acceleration of tumor growth in a CD4+ T cell-dependent manner. These findings suggest that the reduced number and dysfunction of CD4+ T cells due to obesity led to a decreased anti-tumor response of both CD4+ and CD8+ T cells to ultimately accelerate the progression of colorectal cancer. Our findings may elucidate the pathogenesis for poor outcomes of colorectal cancer associated with obesity.
Collapse
Affiliation(s)
- Kota Yamada
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Masafumi Saito
- Department of Disaster and Emergency and Critical Care Medicine, Graduate School of Medicine, Kobe University, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Masayuki Ando
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Tomoki Abe
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Tomosuke Mukoyama
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Kyosuke Agawa
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Akihiro Watanabe
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Shiki Takamura
- Department of Immunology, Kindai University Faculty of Medicine, 377-2 Ono-higashi, Osakasayama 589-0014, Japan
| | - Mitsugu Fujita
- Center for Medical Education and Clinical Training, Kindai University Faculty of Medicine, 377-2 Onohigashi, Osaka 589-0014, Japan
| | - Naoki Urakawa
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Hiroshi Hasegawa
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Shingo Kanaji
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Takeru Matsuda
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Taro Oshikiri
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Yoshihiro Kakeji
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Kimihiro Yamashita
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
- Correspondence: ; Tel.: +81-78-382-5925; Fax: +81-78-382-5939
| |
Collapse
|
25
|
Tewari S, Vargas R, Reizes O. The impact of obesity and adipokines on breast and gynecologic malignancies. Ann N Y Acad Sci 2022; 1518:131-150. [PMID: 36302117 PMCID: PMC10092047 DOI: 10.1111/nyas.14916] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The link between obesity and multiple disease comorbidities is well established. In 2003, Calle and colleagues presented the relationship between obesity and several cancer types, including breast, ovarian, and endometrial malignancies. Nearly, 20% of cancer-related deaths in females can be accounted for by obesity. Identifying obesity as a risk factor for cancer led to a focus on the role of fat-secreted cytokines, known as adipokines, on carcinogenesis and tumor progression. Early studies indicated that the adipokine leptin increases cell proliferation, invasion, and inhibition of apoptosis in multiple cancer types. As a greater appreciation of the obesity-cancer link has amassed, we now know that additional adipokines can impact tumorigenesis. A deeper understanding of the adipokine-activated signaling in cancer may identify new treatment strategies irrespective of obesity. Moreover, adipokines may serve as disease biomarkers, harnessing the potential of obesity-associated factors to serve as indicators of treatment response and disease prognosis. As studies investigating obesity and women's cancers continue to expand, it has become evident that breast, ovarian, and uterine cancers are distinctly impacted by adipokines. While complex, these distinct interactions may provide insight into cancer progression in these organs and new opportunities for targeted therapies. This review aims to organize and present the literature from the last 5 years investigating the mechanisms and implications of adipokine signaling in breast, endometrial, and ovarian cancers with a special focus on leptin and adiponectin.
Collapse
Affiliation(s)
- Surabhi Tewari
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Roberto Vargas
- Department of Gynecologic Oncology, Women's Health Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Ofer Reizes
- Department of Gynecologic Oncology, Women's Health Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Case Comprehensive Cancer Center, Cleveland, Ohio, USA.,Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
26
|
Garcia JN, Wanjalla CN, Mashayekhi M, Hasty AH. Immune Cell Activation in Obesity and Cardiovascular Disease. Curr Hypertens Rep 2022; 24:627-637. [PMID: 36136214 PMCID: PMC9510332 DOI: 10.1007/s11906-022-01222-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW In this review, we focus on immune cell activation in obesity and cardiovascular disease, highlighting specific immune cell microenvironments present in individuals with atherosclerosis, non-ischemic heart disease, hypertension, and infectious diseases. RECENT FINDINGS Obesity and cardiovascular disease are intimately linked and often characterized by inflammation and a cluster of metabolic complications. Compelling evidence from single-cell analysis suggests that obese adipose tissue is inflammatory and infiltrated by almost all immune cell populations. How this inflammatory tissue state contributes to more systemic conditions such as cardiovascular and infectious disease is less well understood. However, current research suggests that changes in the adipose tissue immune environment impact an individual's ability to combat illnesses such as influenza and SARS-CoV2. Obesity is becoming increasingly prevalent globally and is often associated with type 2 diabetes and heart disease. An increased inflammatory state is a major contributor to this association. Widespread chronic inflammation in these disease states is accompanied by an increase in both innate and adaptive immune cell activation. Acutely, these immune cell changes are beneficial as they sustain homeostasis as inflammation increases. However, persistent inflammation subsequently damages tissues and organs throughout the body. Future studies aimed at understanding the unique immune cell populations in each tissue compartment impacted by obesity may hold potential for therapeutic applications.
Collapse
Affiliation(s)
- Jamie N Garcia
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 702 Light Hall, Nashville, TN, 37232, USA
| | - Celestine N Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mona Mashayekhi
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 702 Light Hall, Nashville, TN, 37232, USA.
- VA Tennessee Valley Healthcare System, Nashville, TN, USA.
| |
Collapse
|
27
|
Martin-Perez M, Urdiroz-Urricelqui U, Bigas C, Benitah SA. The role of lipids in cancer progression and metastasis. Cell Metab 2022; 34:1675-1699. [PMID: 36261043 DOI: 10.1016/j.cmet.2022.09.023] [Citation(s) in RCA: 134] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Lipids have essential biological functions in the body (e.g., providing energy storage, acting as a signaling molecule, and being a structural component of membranes); however, an excess of lipids can promote tumorigenesis, colonization, and metastatic capacity of tumor cells. To metastasize, a tumor cell goes through different stages that require lipid-related metabolic and structural adaptations. These adaptations include altering the lipid membrane composition for invading other niches and overcoming cell death mechanisms and promoting lipid catabolism and anabolism for energy and oxidative stress protective purposes. Cancer cells also harness lipid metabolism to modulate the activity of stromal and immune cells to their advantage and to resist therapy and promote relapse. All this is especially worrying given the high fat intake in Western diets. Thus, metabolic interventions aiming to reduce lipid availability to cancer cells or to exacerbate their metabolic vulnerabilities provide promising therapeutic opportunities to prevent cancer progression and treat metastasis.
Collapse
Affiliation(s)
- Miguel Martin-Perez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, University of Barcelona, 08028 Barcelona, Spain.
| | - Uxue Urdiroz-Urricelqui
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Claudia Bigas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain.
| |
Collapse
|
28
|
Kawaguchi K, Maeshima Y, Toi M. Tumor immune microenvironment and systemic response in breast cancer. Med Oncol 2022; 39:208. [PMID: 36175677 DOI: 10.1007/s12032-022-01782-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/24/2022] [Indexed: 06/16/2023]
Abstract
Cancer immunotherapies, particularly immune checkpoint inhibitors (ICIs) that target programmed cell death protein 1 (PD-1) or programmed cell death ligand 1 (PD-L1), have revolutionized cancer treatment. ICIs are effective against breast cancer, and their efficacy against triple-negative breast cancer (TNBC) has been reported. The efficacy of immunotherapy is related to the tumor immune microenvironment. In particular, tumor-infiltrating immune cells, hypoxia, and mitochondria in the tumor microenvironment are closely associated with anti-tumor immunity. Moreover, breast cancer (BC) tumors exhibit high heterogeneity; however, identification of effective biomarkers, via tissue biopsies, is limited owing to the invasiveness of the procedure. Therefore, it is crucial to develop non-invasive protocols (e.g., blood and fecal sampling) to identify components of the tumor immune microenvironment that reflect the systemic immune response, for the characterization of immunotherapy biomarkers. Herein, we review the relationship among systemic immune responses-via liquid biopsy analysis-the microbiome, and the tumor immune microenvironment in BC, while characterizing prospective biomarkers. Relationship between TIME and systemic response in breast cancer.
Collapse
Affiliation(s)
- Kosuke Kawaguchi
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-kawaharacho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yurina Maeshima
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-kawaharacho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Masakazu Toi
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-kawaharacho, Sakyo-Ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
29
|
Hillers-Ziemer LE, Kuziel G, Williams AE, Moore BN, Arendt LM. Breast cancer microenvironment and obesity: challenges for therapy. Cancer Metastasis Rev 2022; 41:627-647. [PMID: 35435599 PMCID: PMC9470689 DOI: 10.1007/s10555-022-10031-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023]
Abstract
Women with obesity who develop breast cancer have a worsened prognosis with diminished survival rates and increased rates of metastasis. Obesity is also associated with decreased breast cancer response to endocrine and chemotherapeutic treatments. Studies utilizing multiple in vivo models of obesity as well as human breast tumors have enhanced our understanding of how obesity alters the breast tumor microenvironment. Changes in the complement and function of adipocytes, adipose-derived stromal cells, immune cells, and endothelial cells and remodeling of the extracellular matrix all contribute to the rapid growth of breast tumors in the context of obesity. Interactions of these cells enhance secretion of cytokines and adipokines as well as local levels of estrogen within the breast tumor microenvironment that promote resistance to multiple therapies. In this review, we will discuss our current understanding of the impact of obesity on the breast tumor microenvironment, how obesity-induced changes in cellular interactions promote resistance to breast cancer treatments, and areas for development of treatment interventions for breast cancer patients with obesity.
Collapse
Affiliation(s)
- Lauren E Hillers-Ziemer
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Genevra Kuziel
- Program in Cancer Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Abbey E Williams
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Brittney N Moore
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Lisa M Arendt
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Program in Cancer Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Dr. Rm 4354A, Madison, WI, 53706, USA.
| |
Collapse
|
30
|
Böttcher-Loschinski R, Rial Saborido J, Böttcher M, Kahlfuss S, Mougiakakos D. Lipotoxicity as a Barrier for T Cell-Based Therapies. Biomolecules 2022; 12:biom12091182. [PMID: 36139021 PMCID: PMC9496045 DOI: 10.3390/biom12091182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/19/2022] Open
Abstract
Nowadays, T-cell-based approaches play an increasing role in cancer treatment. In particular, the use of (genetically engineered) T-cells has heralded a novel era for various diseases with previously poor outcomes. Concurrently, the relationship between the functional behavior of immune cells and their metabolic state, known as immunometabolism, has been found to be an important determinant for the success of immunotherapy. In this context, immune cell metabolism is not only controlled by the expression of transcription factors, enzymes and transport proteins but also by nutrient availability and the presence of intermediate metabolites. The lack of as well as an oversupply of nutrients can be detrimental and lead to cellular dysfunction and damage, potentially resulting in reduced metabolic fitness and/or cell death. This review focusses on the detrimental effects of excessive exposure of T cells to fatty acids, known as lipotoxicity, in the context of an altered lipid tumor microenvironment. Furthermore, implications of T cell-related lipotoxicity for immunotherapy will be discussed, as well as potential therapeutic approaches.
Collapse
Affiliation(s)
- Romy Böttcher-Loschinski
- Department of Hematology and Oncology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Correspondence:
| | - Judit Rial Saborido
- Medical Department 5–Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Martin Böttcher
- Department of Hematology and Oncology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health Campus Immunology, Infectiology, and Inflammation (GCI3), Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Sascha Kahlfuss
- Health Campus Immunology, Infectiology, and Inflammation (GCI3), Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Institute of Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- CHaMP, Center for Health and Medical Prevention, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
| | - Dimitrios Mougiakakos
- Department of Hematology and Oncology, University Hospital Magdeburg, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Medical Department 5–Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, 91054 Erlangen, Germany
- Health Campus Immunology, Infectiology, and Inflammation (GCI3), Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
31
|
Marelli G, Morina N, Portale F, Pandini M, Iovino M, Di Conza G, Ho PC, Di Mitri D. Lipid-loaded macrophages as new therapeutic target in cancer. J Immunother Cancer 2022; 10:jitc-2022-004584. [PMID: 35798535 PMCID: PMC9263925 DOI: 10.1136/jitc-2022-004584] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 11/04/2022] Open
Abstract
Macrophages are main players of the innate immune system. They show great heterogeneity and play diverse functions that include support to development, sustenance of tissue homeostasis and defense against infections. Dysfunctional macrophages have been described in multiple pathologies including cancer. Indeed tumor-associated macrophages (TAMs) are abundant in most tumors and sustain cancer growth, promote invasion and mediate immune evasion. Importantly, lipid metabolism influences macrophage activation and lipid accumulation confers pathogenic features on macrophages. Notably, a subset of lipid-loaded macrophages has been recently identified in many tumor types. Lipid-loaded TAMs support tumor growth and progression and exert immune-suppressive activities. In this review, we describe the role of lipid metabolism in macrophage activation in physiology and pathology and we discuss the impact of lipid accumulation in macrophages in the context of cancer.
Collapse
Affiliation(s)
- Giulia Marelli
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy
| | - Nicolò Morina
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy.,Department of Biomedical Sciences, Humanitas University, Lombardia, Italy
| | - Federica Portale
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy
| | - Marta Pandini
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy.,Department of Biomedical Sciences, Humanitas University, Lombardia, Italy
| | - Marta Iovino
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy
| | - Giusy Di Conza
- Department of Oncology, University of Lausanne, Lausanne, Switzerland.,Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Ping-Chih Ho
- Department of Oncology, University of Lausanne, Lausanne, Switzerland.,Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Diletta Di Mitri
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy .,Department of Biomedical Sciences, Humanitas University, Lombardia, Italy
| |
Collapse
|
32
|
Izquierdo-Torres E, Hernández-Oliveras A, Lozano-Arriaga D, Zarain-Herzberg Á. Obesity, the other pandemic: linking diet and carcinogenesis by epigenetic mechanisms. J Nutr Biochem 2022; 108:109092. [PMID: 35718098 DOI: 10.1016/j.jnutbio.2022.109092] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 03/19/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022]
Abstract
Both obesity and cancer are complex medical conditions that are considered public health problems. The influence of obesity on the predisposition to develop various types of cancer has been observed in a wide variety of studies. Due to their importance as public health problems, and the close relationship between both conditions, it is important to be able to understand and associate them mechanistically. In this review article, we intend to go a little further, by finding relationships between lifestyle, which can lead a person to develop obesity, and how it influences at the cellular and molecular level, affecting gene expression to favor signaling pathways or transcriptional programs involved in cancer. We describe how products of metabolism and intermediate metabolism can affect chromatin structure, participating in the regulation (or dysregulation) of gene expression, and we show an analysis of genes that are responsive to diets high in sugar and fat, and how their epigenetic landscape is altered.
Collapse
Affiliation(s)
- Eduardo Izquierdo-Torres
- Departamento de Bioquímica, Facultad de Medicina. Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Andrés Hernández-Oliveras
- Departamento de Bioquímica, Facultad de Medicina. Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Dalia Lozano-Arriaga
- Departamento de Bioquímica, Facultad de Medicina. Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Ángel Zarain-Herzberg
- Departamento de Bioquímica, Facultad de Medicina. Universidad Nacional Autónoma de México, Ciudad de México, México.
| |
Collapse
|
33
|
Yadav AK, Lee MC, Lucero MY, Su S, Reinhardt CJ, Chan J. Activity-Based NIR Bioluminescence Probe Enables Discovery of Diet-Induced Modulation of the Tumor Microenvironment via Nitric Oxide. ACS CENTRAL SCIENCE 2022; 8:461-472. [PMID: 35505872 PMCID: PMC9052803 DOI: 10.1021/acscentsci.1c00317] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Indexed: 05/15/2023]
Abstract
Nitric oxide (NO) plays a critical role in acute and chronic inflammation. NO's contributions to cancer are of particular interest due to its context-dependent bioactivities. For example, immune cells initially produce cytotoxic quantities of NO in response to the nascent tumor. However, it is believed that this fades over time and reaches a concentration that supports the tumor microenvironment (TME). These complex dynamics are further complicated by other factors, such as diet and oxygenation, making it challenging to establish a complete picture of NO's impact on tumor progression. Although many activity-based sensing (ABS) probes for NO have been developed, only a small fraction have been employed in vivo, and fewer yet are practical in cancer models where the NO concentration is <200 nM. To overcome this outstanding challenge, we have developed BL660-NO, the first ABS probe for NIR bioluminescence imaging of NO in cancer. Owing to the low intrinsic background, high sensitivity, and deep tissue imaging capabilities of our design, BL660-NO was successfully employed to visualize endogenous NO in cellular systems, a human liver metastasis model, and a murine breast cancer model. Importantly, its exceptional performance facilitated two dietary studies which examine the impact of fat intake on NO and the TME. BL660-NO provides the first direct molecular evidence that intratumoral NO becomes elevated in mice fed a high-fat diet, which became obese with larger tumors, compared to control animals on a low-fat diet. These results indicate that an inflammatory diet can increase NO production via recruitment of macrophages and overexpression of inducible nitric oxide synthase which in turn can drive tumor progression.
Collapse
|
34
|
Núñez-Ruiz A, Sánchez-Brena F, López-Pacheco C, Acevedo-Domínguez NA, Soldevila G. Obesity modulates the immune macroenvironment associated with breast cancer development. PLoS One 2022; 17:e0266827. [PMID: 35472214 PMCID: PMC9041840 DOI: 10.1371/journal.pone.0266827] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/28/2022] [Indexed: 11/19/2022] Open
Abstract
Growing evidence demonstrates a strong correlation between obesity and an increased risk of breast cancer, although the mechanisms involved have not been completely elucidated. Some reports have described a crosstalk between adipocytes, cancer cells, and immune cells within the tumor microenvironment, however, it is currently unknown whether obesity can promote tumor growth by inducing systemic alterations of the immune cell homeostasis in peripheral lymphoid organs and adipose tissue. Here, we used the E0771 breast cancer cell line in a mouse model of diet-induced obesity to analyze the immune subpopulations present in the tumors, visceral adipose tissue (VAT), and spleen of lean and obese mice. Our results showed a significant reduction in the frequency of infiltrating CD8+ T cells and a decreased M1/M2 macrophage ratio, indicative of the compromised anti-tumoral immune response reported in obesity. Despite not finding differences in the percentage or numbers of intratumoral Tregs, phenotypic analysis showed that they were enriched in CD39+, PD-1+ and CCR8+ cells, compared to the draining lymph nodes, confirming the highly immunosuppressive profile of infiltrating Tregs reported in established tumors. Analysis of peripheral T lymphocytes showed that tumor development in obese mice was associated to a significant increase in the percentage of peripheral Tregs, which supports the systemic immunosuppressive effect caused by the tumor. Interestingly, evaluation of immune subpopulations in the VAT showed that the characteristic increase in the M1/M2 macrophage ratio reported in obesity, was completely reversed in tumor-bearing mice, resembling the M2-polarized profile found in the microenvironment of the growing tumor. Importantly, VAT Tregs, which are commonly decreased in obese mice, were significantly increased in the presence of breast tumors and displayed significantly higher levels of Foxp3, indicating a regulatory feedback mechanism triggered by tumor growth. Altogether, our results identify a complex reciprocal relationship between adipocytes, immune cells, and the tumor, which may modulate the immune macroenvironment that promotes breast cancer development in obesity.
Collapse
Affiliation(s)
- Aleida Núñez-Ruiz
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, México City, México
| | - Flor Sánchez-Brena
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, México City, México
| | - Cynthia López-Pacheco
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, México City, México
- Laboratorio Nacional de Citometría de Flujo, Instituto de Investigaciones Biomédicas, UNAM, México City, México
| | | | - Gloria Soldevila
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, UNAM, México City, México
- * E-mail:
| |
Collapse
|
35
|
Mortezaee K, Majidpoor J. CD8 + T Cells in SARS-CoV-2 Induced Disease and Cancer-Clinical Perspectives. Front Immunol 2022; 13:864298. [PMID: 35432340 PMCID: PMC9010719 DOI: 10.3389/fimmu.2022.864298] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022] Open
Abstract
Dysregulated innate and adaptive immunity is a sign of SARS-CoV-2-induced disease and cancer. CD8+ T cells are important cells of the immune system. The cells belong to the adaptive immunity and take a front-line defense against viral infections and cancer. Extreme CD8+ T-cell activities in the lung of patients with a SARS-CoV-2-induced disease and within the tumor microenvironment (TME) will change their functionality into exhausted state and undergo apoptosis. Such diminished immunity will put cancer cases at a high-risk group for SARS-CoV-2-induced disease, rendering viral sepsis and a more severe condition which will finally cause a higher rate of mortality. Recovering responses from CD8+ T cells is a purpose of vaccination against SARS-CoV-2. The aim of this review is to discuss the CD8+ T cellular state in SARS-CoV-2-induced disease and in cancer and to present some strategies for recovering the functionality of these critical cells.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Jamal Majidpoor
- Department of Anatomy, Faculty of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
36
|
Thakkar N, Shin YB, Sung HK. Nutritional Regulation of Mammary Tumor Microenvironment. Front Cell Dev Biol 2022; 10:803280. [PMID: 35186923 PMCID: PMC8847692 DOI: 10.3389/fcell.2022.803280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
The mammary gland is a heterogeneous organ comprising of immune cells, surrounding adipose stromal cells, vascular cells, mammary epithelial, and cancer stem cells. In response to nutritional stimuli, dynamic interactions amongst these cell populations can be modulated, consequently leading to an alteration of the glandular function, physiology, and ultimately disease pathogenesis. For example, obesity, a chronic over-nutritional condition, is known to disrupt homeostasis within the mammary gland and increase risk of breast cancer development. In contrast, emerging evidence has demonstrated that fasting or caloric restriction can negatively impact mammary tumorigenesis. However, how fasting induces phenotypic and functional population differences in the mammary microenvironment is not well understood. In this review, we will provide a detailed overview on the effect of nutritional conditions (i.e., overnutrition or fasting) on the mammary gland microenvironment and its impact on mammary tumor progression.
Collapse
Affiliation(s)
- Nikita Thakkar
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Ye Bin Shin
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- *Correspondence: Hoon-Ki Sung,
| |
Collapse
|
37
|
Alarcon PC, Damen MSMA, Madan R, Deepe GS, Spearman P, Way SS, Divanovic S. Adipocyte inflammation and pathogenesis of viral pneumonias: an overlooked contribution. Mucosal Immunol 2021; 14:1224-1234. [PMID: 33958704 PMCID: PMC8100369 DOI: 10.1038/s41385-021-00404-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/18/2021] [Accepted: 03/27/2021] [Indexed: 02/06/2023]
Abstract
Epidemiological evidence establishes obesity as an independent risk factor for increased susceptibility and severity to viral respiratory pneumonias associated with H1N1 influenza and SARS-CoV-2 pandemics. Given the global obesity prevalence, a better understanding of the mechanisms behind obese susceptibility to infection is imperative. Altered immune cell metabolism and function are often perceived as a key causative factor of dysregulated inflammation. However, the contribution of adipocytes, the dominantly altered cell type in obesity with broad inflammatory properties, to infectious disease pathogenesis remains largely ignored. Thus, skewing of adipocyte-intrinsic cellular metabolism may lead to the development of pathogenic inflammatory adipocytes, which shape the overall immune responses by contributing to either premature immunosenescence, delayed hyperinflammation, or cytokine storm in infections. In this review, we discuss the underappreciated contribution of adipocyte cellular metabolism and adipocyte-produced mediators on immune system modulation and how such interplay may modify disease susceptibility and pathogenesis of influenza and SARS-CoV-2 infections in obese individuals.
Collapse
Affiliation(s)
- Pablo C Alarcon
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Divisions of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Medical Scientist Training Program, Cincinnati, OH, USA
- Immunology Graduate Program Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michelle S M A Damen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Divisions of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rajat Madan
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Veterans Affairs Medical Center, Cincinnati, OH, USA
| | - George S Deepe
- Division of Infectious Diseases, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paul Spearman
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Divisions of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sing Sing Way
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Divisions of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Divisions of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Medical Scientist Training Program, Cincinnati, OH, USA.
- Immunology Graduate Program Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
38
|
Daily caloric restriction limits tumor growth more effectively than caloric cycling regardless of dietary composition. Nat Commun 2021; 12:6201. [PMID: 34707136 PMCID: PMC8551193 DOI: 10.1038/s41467-021-26431-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022] Open
Abstract
Cancer incidence increases with age and is a leading cause of death. Caloric restriction (CR) confers benefits on health and survival and delays cancer. However, due to CR's stringency, dietary alternatives offering the same cancer protection have become increasingly attractive. Short cycles of a plant-based diet designed to mimic fasting (FMD) are protective against tumorigenesis without the chronic restriction of calories. Yet, it is unclear whether the fasting time, level of dietary restriction, or nutrient composition is the primary driver behind cancer protection. Using a breast cancer model in mice, we compare the potency of daily CR to that of periodic caloric cycling on FMD or an isocaloric standard laboratory chow against primary tumor growth and metastatic burden. Here, we report that daily CR provides greater protection against tumor growth and metastasis to the lung, which may be in part due to the unique immune signature observed with daily CR.
Collapse
|
39
|
Looman KIM, Santos S, Moll HA, Leijten CWE, Grosserichter-Wagener C, Voortman T, Jaddoe VVW, van Zelm MC, Kiefte-de Jong JC. Childhood Adiposity Associated With Expanded Effector Memory CD8+ and Vδ2+Vγ9+ T Cells. J Clin Endocrinol Metab 2021; 106:e3923-e3935. [PMID: 34128988 DOI: 10.1210/clinem/dgab433] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Adult obesity is associated with chronic low-grade inflammation and may give rise to future chronic disease. However, it is unclear whether adiposity-related inflammation is already apparent in childhood. OBJECTIVE To study associations between child adiposity measures with circulating monocytes and naive and memory subsets in CD4, CD8, and γδ T cell lineages. METHODS Ten-year-old children (n = 890) from the Generation R Cohort underwent dual-energy x-ray absorptiometry and magnetic resonance imaging for body composition (body mass index [BMI], fat mass index [FMI], android-to-gynoid fat mass ratio, visceral fat index, liver fat fraction). Blood samples were taken for detailed immunophenotyping of leukocytes by 11-color flow cytometry. RESULTS Several statistically significant associations were observed. A 1-SD increase in total FMI was associated with +8.4% (95% CI 2.0, 15.2) Vδ2+Vγ9+ and +7.4% (95% CI 2.4, 12.5) CD8+TEMRO cell numbers. A 1-SD increase in visceral fat index was associated with +10.7% (95% CI 3.3, 18.7) Vδ2+Vγ9+ and +8.3% (95% CI 2.6, 14.4) CD8+TEMRO cell numbers. Higher android-to-gynoid fat mass ratio was only associated with higher Vδ2+Vγ9+ T cells. Liver fat was associated with higher CD8+TEMRO cells but not with Vδ2+Vγ9+ T cells. Only liver fat was associated with lower Th17 cell numbers: a 1-SD increase was associated with -8.9% (95% CI -13.7, -3.7) Th17 cells. No associations for total CD8+, CD4+ T cells, or monocytes were observed. BMI was not associated with immune cells. CONCLUSION Higher Vδ2+Vγ9+ and CD8+TEMRO cell numbers in children with higher visceral fat index could reflect presence of adiposity-related inflammation in children with adiposity of a general population.
Collapse
Affiliation(s)
- Kirsten I M Looman
- Generation R Study Group, Erasmus MC, University Medical Center, GD, Rotterdam,the Netherlands
- Department of Pediatrics, Sophia Children's Hospital, Erasmus MC, University Medical Center, GD, Rotterdam, the Netherlands
| | - Susana Santos
- Generation R Study Group, Erasmus MC, University Medical Center, GD, Rotterdam,the Netherlands
- Department of Pediatrics, Sophia Children's Hospital, Erasmus MC, University Medical Center, GD, Rotterdam, the Netherlands
| | - Henriette A Moll
- Department of Pediatrics, Sophia Children's Hospital, Erasmus MC, University Medical Center, GD, Rotterdam, the Netherlands
| | - Charlotte W E Leijten
- Generation R Study Group, Erasmus MC, University Medical Center, GD, Rotterdam,the Netherlands
- Department of Pediatrics, Sophia Children's Hospital, Erasmus MC, University Medical Center, GD, Rotterdam, the Netherlands
| | | | - Trudy Voortman
- Generation R Study Group, Erasmus MC, University Medical Center, GD, Rotterdam,the Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center, GD, Rotterdam, the Netherlands
| | - Vincent V W Jaddoe
- Generation R Study Group, Erasmus MC, University Medical Center, GD, Rotterdam,the Netherlands
- Department of Pediatrics, Sophia Children's Hospital, Erasmus MC, University Medical Center, GD, Rotterdam, the Netherlands
| | - Menno C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Victoria, Australia
| | - Jessica C Kiefte-de Jong
- Department of Pediatrics, Sophia Children's Hospital, Erasmus MC, University Medical Center, GD, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center, GD, Rotterdam, the Netherlands
- Department of Public Health and Primary Care/LUMC Campus The Hague, Leiden University Medical Center, RC, Leiden,The Netherlands
| |
Collapse
|
40
|
Thomas AL, Alarcon PC, Divanovic S, Chougnet CA, Hildeman DA, Moreno-Fernandez ME. Implications of Inflammatory States on Dysfunctional Immune Responses in Aging and Obesity. FRONTIERS IN AGING 2021; 2:732414. [PMID: 35822048 PMCID: PMC9261339 DOI: 10.3389/fragi.2021.732414] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/25/2021] [Indexed: 12/16/2022]
Abstract
Aging and obesity are two conditions characterized by chronic, low-grade inflammation. While both conditions are also associated with dysfunctional immune responses, the shared and distinct underlying mechanisms are just starting to be uncovered. In fact, recent findings have suggested that the effects of obesity on the immune system can be thought of as a state of accelerated aging. Here we propose that chronic, low-grade inflammation seen in obesity and aging is complex, affects multiple cell types, and results in an altered basal immune state. In aging, part of this altered state is the emergence of regulatory immune populations that lead to further immune dysfunction in an attempt to reduce chronic inflammation. While in obesity, part of the altered state is the effect of expanding adipose tissue on immune cell function. Thus, in this review, we compare, and contrast altered immune states in aging and obesity and discuss their potential contribution to a shared clinical problem- decreased vaccine responsiveness.
Collapse
Affiliation(s)
- Alyssa L. Thomas
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program and Medical Scientist Training Program, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Pablo C. Alarcon
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program and Medical Scientist Training Program, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Medical Scientist Training Program, Cincinnati Children’s Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program and Medical Scientist Training Program, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Medical Scientist Training Program, Cincinnati Children’s Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Claire A. Chougnet
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program and Medical Scientist Training Program, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - David A. Hildeman
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program and Medical Scientist Training Program, Cincinnati Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Medical Scientist Training Program, Cincinnati Children’s Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Center for Transplant Immunology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Maria E. Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
41
|
Desharnais L, Walsh LA, Quail DF. Exploiting the obesity-associated immune microenvironment for cancer therapeutics. Pharmacol Ther 2021; 229:107923. [PMID: 34171329 DOI: 10.1016/j.pharmthera.2021.107923] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/11/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022]
Abstract
Obesity causes chronic low-grade inflammation and leads to changes in the immune landscape of multiple organ systems. Given the link between chronic inflammatory conditions and cancer, it is not surprising that obesity is associated with increased risk and worse outcomes in many malignancies. Paradoxically, recent epidemiological studies have shown that high BMI is associated with increased efficacy of immune checkpoint inhibitors (ICI), and a causal relationship has been demonstrated in the preclinical setting. It has been proposed that obesity-associated immune dysregulation underlies this observation by inadvertently creating a favourable microenvironment for increased ICI efficacy. The recent success of ICIs in obese cancer patients raises the possibility that additional immune-targeted therapies may hold therapeutic value in this context. Here we review how obesity affects the immunological composition of the tumor microenvironment in ways that can be exploited for cancer immunotherapies. We discuss existing literature supporting a beneficial role for obesity during ICI therapy in cancer patients, potential opportunities for targeting the innate immune system to mitigate chronic inflammatory processes, and how to pinpoint obese patients who are most likely to benefit from immune interventions without relying solely on body mass index. Given that the incidence of obesity is expanding on an international scale, we propose that understanding obesity-associated inflammation is necessary to reduce cancer mortalities and capitalize on novel therapeutic opportunities in the era of cancer immunotherapy.
Collapse
Affiliation(s)
- Lysanne Desharnais
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada.
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada; Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
42
|
Visceral Obesity Promotes Lung Cancer Progression-Toward Resolution of the Obesity Paradox in Lung Cancer. J Thorac Oncol 2021; 16:1333-1348. [PMID: 34144926 DOI: 10.1016/j.jtho.2021.04.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/12/2021] [Accepted: 04/26/2021] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Although obesity is associated with adverse cancer outcomes in general, most retrospective clinical studies suggest a beneficial effect of obesity in NSCLC. METHODS Hypothesizing that this "obesity paradox" arises partly from the limitations of using body mass index (BMI) to measure obesity, we quantified adiposity using preoperative computed tomography images. This allowed the specific determination of central obesity as abdominal visceral fat area normalized to total fat area (visceral fat index [VFI]). In addition, owing to the previously reported salutary effect of metformin on high-BMI patients with lung cancer, metformin users were excluded. We then explored associations between visceral obesity and outcomes after surgical resection of stage I and II NSCLC. We also explored potential immunologic underpinnings of such association using complimentary analyses of tumor gene expression data from NSCLC tumors and the tumor transcriptome and immune microenvironment in an immunocompetent model of lung cancer with diet-induced obesity. RESULTS We found that in 513 patients with stage I and II NSCLC undergoing lobectomy, a high VFI is associated with decreased recurrence-free and overall survival. VFI was also inversely related to an inflammatory transcriptomic signature in NSCLC tumors, consistent with observations made in immunocompetent murine models wherein diet-induced obesity promoted cancer progression while exacerbating elements of immune suppression in the tumor niche. CONCLUSIONS In all, this study uses multiple lines of evidence to reveal the adverse effects of visceral obesity in patients with NSCLC, which align with those found in animal models. Thus, the obesity paradox may, at least in part, be secondary to the use of BMI as a measure of obesity and the confounding effects of metformin use.
Collapse
|
43
|
Skeletal Muscle-Adipose Tissue-Tumor Axis: Molecular Mechanisms Linking Exercise Training in Prostate Cancer. Int J Mol Sci 2021; 22:ijms22094469. [PMID: 33922898 PMCID: PMC8123194 DOI: 10.3390/ijms22094469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Increased visceral adiposity may influence the development of prostate cancer (PCa) aggressive tumors and cancer mortality. White adipose tissue (WAT), usually referred to as periprostatic adipose tissue (PPAT), surrounds the prostatic gland and has emerged as a potential mediator of the tumor microenvironment. Exercise training (ET) induces several adaptations in both skeletal muscle and WAT. Some of these effects are mediated by ET-induced synthesis and secretion of several proteins, known as myo- and adipokines. Together, myokines and adipokines may act in an endocrine-like manner to favor communication between skeletal muscle and WAT, as they may work together to improve whole-body metabolic health. This crosstalk may constitute a potential mechanism by which ET exerts its beneficial role in the prevention and treatment of PCa-related disorders; however, this has not yet been explored. Therefore, we reviewed the current evidence on the effects of skeletal muscle–WAT–tumor crosstalk in PCa, and the potential mediators of this process to provide a better understanding of underlying ET-related mechanisms in cancer.
Collapse
|
44
|
Asiry S, Kim G, Filippou PS, Sanchez LR, Entenberg D, Marks DK, Oktay MH, Karagiannis GS. The Cancer Cell Dissemination Machinery as an Immunosuppressive Niche: A New Obstacle Towards the Era of Cancer Immunotherapy. Front Immunol 2021; 12:654877. [PMID: 33927723 PMCID: PMC8076861 DOI: 10.3389/fimmu.2021.654877] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
Although cancer immunotherapy has resulted in unpreceded survival benefits to subsets of oncology patients, accumulating evidence from preclinical animal models suggests that the immunosuppressive tumor microenvironment remains a detrimental factor limiting benefit for many patient subgroups. Recent efforts on lymphocyte-mediated immunotherapies are primarily focused on eliminating cancer foci at primary and metastatic sites, but few studies have investigated the impact of these therapies on the highly complex process of cancer cell dissemination. The metastatic cascade involves the directional streaming of invasive/migratory tumor cells toward specialized blood vessel intravasation gateways, called TMEM doorways, to the peripheral circulation. Importantly, this process occurs under the auspices of a specialized tumor microenvironment, herewith referred to as "Dissemination Trajectory", which is supported by an ample array of tumor-associated macrophages (TAMs), skewed towards an M2-like polarization spectrum, and which is also vital for providing microenvironmental cues for cancer cell invasion, migration and stemness. Based on pre-existing evidence from preclinical animal models, this article outlines the hypothesis that dissemination trajectories do not only support the metastatic cascade, but also embody immunosuppressive niches, capable of providing transient and localized immunosubversion cues to the migratory/invasive cancer cell subpopulation while in the act of departing from a primary tumor. So long as these dissemination trajectories function as "immune deserts", the migratory tumor cell subpopulation remains efficient in evading immunological destruction and seeding metastatic sites, despite administration of cancer immunotherapy and/or other cytotoxic treatments. A deeper understanding of the molecular and cellular composition, as well as the signaling circuitries governing the function of these dissemination trajectories will further our overall understanding on TAM-mediated immunosuppression and will be paramount for the development of new therapeutic strategies for the advancement of optimal cancer chemotherapies, immunotherapies, and targeted therapies.
Collapse
Affiliation(s)
- Saeed Asiry
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
| | - Gina Kim
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
| | - Panagiota S. Filippou
- School of Health and Life Sciences, Teesside University, Middlesbrough, United Kingdom
- National Horizons Centre, Teesside University, Darlington, United Kingdom
| | - Luis Rivera Sanchez
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
| | - David Entenberg
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| | - Douglas K. Marks
- Department of Medicine, NYU Long Island School of Medicine, Mineola, NY, United States
| | - Maja H. Oktay
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| | - George S. Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| |
Collapse
|
45
|
Reactive oxygen species (ROS): Critical roles in breast tumor microenvironment. Crit Rev Oncol Hematol 2021; 160:103285. [DOI: 10.1016/j.critrevonc.2021.103285] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/18/2021] [Accepted: 02/27/2021] [Indexed: 02/06/2023] Open
|
46
|
Kulkarni A, Bowers LW. The role of immune dysfunction in obesity-associated cancer risk, progression, and metastasis. Cell Mol Life Sci 2021; 78:3423-3442. [PMID: 33464384 PMCID: PMC11073382 DOI: 10.1007/s00018-020-03752-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/10/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
Obesity has been linked to an increased risk of and a worse prognosis for several types of cancer. A number of interrelated mediators contribute to obesity's pro-tumor effects, including chronic adipose inflammation and other perturbations of immune cell development and function. Here, we review studies examining the impact of obesity-induced immune dysfunction on cancer risk and progression. While the role of adipose tissue inflammation in obesity-associated cancer risk has been well characterized, the effects of obesity on immune cell infiltration and activity within the tumor microenvironment are not well studied. In this review, we aim to highlight the impact of both adipose-mediated inflammatory signaling and intratumoral immunosuppressive signaling in obesity-induced cancer risk, progression, and metastasis.
Collapse
Affiliation(s)
- Aneesha Kulkarni
- Department of Nutrition Science, College of Health and Human Sciences, Purdue University, West Lafayette, IN, 47906, USA
| | - Laura W Bowers
- Department of Nutrition Science, College of Health and Human Sciences, Purdue University, West Lafayette, IN, 47906, USA.
| |
Collapse
|
47
|
Kaul K, Misri S, Ramaswamy B, Ganju RK. Contribution of the tumor and obese microenvironment to triple negative breast cancer. Cancer Lett 2021; 509:115-120. [PMID: 33798632 DOI: 10.1016/j.canlet.2021.03.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/01/2021] [Accepted: 03/24/2021] [Indexed: 01/01/2023]
Abstract
The growing burden of obesity and incidence of the aggressive triple negative breast cancer (TNBC) is a challenge, especially amongst vulnerable populations with unmet medical needs and higher mortality from breast cancer. While some mechanisms linking obesity and TNBC have been identified, the complex nature of pathogenesis, in both obesity as well as TNBC poses a real challenge in establishing a causative role of obesity in risk of TNBC. In this review article, we discuss pathological mechanisms identified in the tumor microenvironment (TME) as well as the obese microenvironment (OME), such as inflammation, insulin resistance and survival pathways that contribute to the development and progression of TNBC. Insights into the cross-talk between TME and OME, and their contribution to TNBC development and progression, may pave the way for personalized therapies against TNBC progression, relapse and metastasis.
Collapse
Affiliation(s)
- Kirti Kaul
- Comprehensive Cancer Center, USA; Department of Pathology, USA
| | | | | | - Ramesh K Ganju
- Comprehensive Cancer Center, USA; Department of Pathology, USA.
| |
Collapse
|
48
|
Kuwano T, Izumi H, Aslam MR, Igarashi Y, Bilal M, Nishimura A, Watanabe Y, Nawaz A, Kado T, Ikuta K, Yamamoto S, Sasahara M, Fujisaka S, Yagi K, Mori H, Tobe K. Generation and characterization of a Meflin-CreERT2 transgenic line for lineage tracing in white adipose tissue. PLoS One 2021; 16:e0248267. [PMID: 33760832 PMCID: PMC7990287 DOI: 10.1371/journal.pone.0248267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/24/2021] [Indexed: 12/22/2022] Open
Abstract
Meflin (Islr) expression has gained attention as a marker for mesenchymal stem cells, but its function remains largely unexplored. Here, we report the generation of Meflin-CreERT2 mice with CreERT2 inserted under the Meflin gene promoter to label Meflin-expressing cells genetically, thereby enabling their lineages to be traced. We found that in adult mice, Meflin-expressing lineage cells were present in adipose tissue stroma and had differentiated into mature adipocytes. These cells constituted Crown-like structures in the adipose tissue of mice after high-fat diet loading. Cold stimulation led to the differentiation of Meflin-expressing lineage cells into beige adipocytes. Thus, the Meflin-CreERT2 mouse line is a useful new tool for visualizing and tracking the lineage of Meflin-expressing cells.
Collapse
Affiliation(s)
- Takahide Kuwano
- First Department of Internal Medicine, University of Toyama, Toyama-shi, Toyama, Japan
| | - Hironori Izumi
- Department of Molecular Neuroscience, University of Toyama, Toyama-shi, Toyama, Japan
| | - Muhammad Rahil Aslam
- First Department of Internal Medicine, University of Toyama, Toyama-shi, Toyama, Japan
| | - Yoshiko Igarashi
- First Department of Internal Medicine, University of Toyama, Toyama-shi, Toyama, Japan
| | - Muhammad Bilal
- First Department of Internal Medicine, University of Toyama, Toyama-shi, Toyama, Japan
| | - Ayumi Nishimura
- First Department of Internal Medicine, University of Toyama, Toyama-shi, Toyama, Japan
| | - Yoshiyuki Watanabe
- First Department of Internal Medicine, University of Toyama, Toyama-shi, Toyama, Japan
| | - Allah Nawaz
- First Department of Internal Medicine, University of Toyama, Toyama-shi, Toyama, Japan
| | - Tomonobu Kado
- First Department of Internal Medicine, University of Toyama, Toyama-shi, Toyama, Japan
| | - Koichi Ikuta
- Department of Virus Research, Laboratory of Immune Regulation, Institute of Frontier Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Seiji Yamamoto
- Department of Pathology, University of Toyama, Toyama-shi, Toyama, Japan
| | - Masakiyo Sasahara
- Department of Pathology, University of Toyama, Toyama-shi, Toyama, Japan
| | - Shiho Fujisaka
- First Department of Internal Medicine, University of Toyama, Toyama-shi, Toyama, Japan
| | - Kunimasa Yagi
- First Department of Internal Medicine, University of Toyama, Toyama-shi, Toyama, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, University of Toyama, Toyama-shi, Toyama, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, University of Toyama, Toyama-shi, Toyama, Japan
| |
Collapse
|
49
|
Zhao S, Peralta RM, Avina-Ochoa N, Delgoffe GM, Kaech SM. Metabolic regulation of T cells in the tumor microenvironment by nutrient availability and diet. Semin Immunol 2021; 52:101485. [PMID: 34462190 PMCID: PMC8545851 DOI: 10.1016/j.smim.2021.101485] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 08/12/2021] [Indexed: 12/11/2022]
Abstract
Recent advances in immunotherapies such as immune checkpoint blockade (ICB) and chimeric antigen receptor T cells (CAR-T) for the treatment of cancer have generated excitement over their ability to yield durable, and potentially curative, responses in a multitude of cancers. These findings have established that the immune system is capable of eliminating tumors and led us to a better, albeit still incomplete, understanding of the mechanisms by which tumors interact with and evade destruction by the immune system. Given the central role of T cells in immunotherapy, elucidating the cell intrinsic and extrinsic factors that govern T cell function in tumors will facilitate the development of immunotherapies that establish durable responses in a greater number of patients. One such factor is metabolism, a set of fundamental cellular processes that not only sustains cell survival and proliferation, but also serves as a means for cells to interpret their local environment. Nutrient sensing is critical for T cells that must infiltrate into a metabolically challenging tumor microenvironment and expand under these harsh conditions to eliminate cancerous cells. Here we introduce T cell exhaustion with respect to cellular metabolism, followed by a discussion of nutrient availability at the tumor and organismal level in relation to T cell metabolism and function to provide rationale for the study and targeting of metabolism in anti-tumor immune responses.
Collapse
Affiliation(s)
- Steven Zhao
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ronal M Peralta
- Tumor Microenvironment Center, Department of Immunology, UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, USA
| | - Natalia Avina-Ochoa
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Greg M Delgoffe
- Tumor Microenvironment Center, Department of Immunology, UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, USA.
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
50
|
Deshpande RP, Sharma S, Watabe K. The Confounders of Cancer Immunotherapy: Roles of Lifestyle, Metabolic Disorders and Sociological Factors. Cancers (Basel) 2020; 12:E2983. [PMID: 33076303 PMCID: PMC7602474 DOI: 10.3390/cancers12102983] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/06/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Checkpoint blockade immunotherapy (CPI) is an effective treatment option for many types of cancers. Irrespective of its wide clinical implications, the overall efficacy remains unpredictable and even poor in certain pathologies such as breast cancer. Thus, it is imperative to understand the role of factors affecting its responsiveness. In this review, we provide an overview on the involvement of sociological factors, lifestyles and metabolic disorders in modulating the CPI response in patients from multiple malignancies. Lifestyle habits including exercise, and diet promoted therapeutic responsiveness while alcohol consumption mitigated the CPI effect by decreasing mutational burden and hampering antigen presentation by dendritic cells. Metabolic disorder such as obesity was recognized to enhance the PD-1 expression while diabetes and hypertension were consequences of CPI therapy rather than causes. Among the sociologic factors, sex and race positively influenced the CPI effectiveness on account of increased effector T cell activity and increased PD-1 expression while ageing impaired CPI responsiveness by decreasing functional T cell and increased toxicity. The combined effect of these factors was observed for obesity and gender, in which obese males had the most significant effect of CPI. Therefore these variables should be carefully considered before treating patients with CPI for optimal treatment outcome.
Collapse
Affiliation(s)
| | | | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA; (R.P.D.); (S.S.)
| |
Collapse
|