학술논문

Obesity Is Associated with Immunometabolic Changes in Adipose Tissue That May Drive Treatment Resistance in Breast Cancer: Immune-Metabolic Reprogramming and Novel Therapeutic Strategies.
Document Type
Article
Source
Cancers. May2023, Vol. 15 Issue 9, p2440. 20p.
Subject
*OBESITY complications
*DISEASE progression
*CYTOKINES
*ADIPOKINES
*IMMUNE checkpoint inhibitors
*TRASTUZUMAB
*PHOSPHOTRANSFERASES
*INFLAMMATION
*MACROPHAGES
*METABOLIC reprogramming
*CELLULAR signal transduction
*METFORMIN
*AROMATASE
*ADIPOSE tissues
*DRUG resistance in cancer cells
*BREAST tumors
*T-cell lymphoma
Language
ISSN
2072-6694
Abstract
Simple Summary: Obesity is associated with metabolic changes in the immunological tumour infiltrate in breast cancer. The crosstalk between adipocytes, macrophages and proinflammatory cytokines may promote immunometabolic dysregulation and immunosuppressive phenotypes in breast tumours which may be correlated to treatment resistance. In this review, we provide a comprehensive overview of emerging evidence describing the association between obesity and immunometabolic dysfunction in breast cancer and discuss novel therapeutic strategies to overcome immunosuppressive phenotypes. White adipose tissue (WAT) represents an endocrinologically and immunologically active tissue whose primary role is energy storage and homeostasis. Breast WAT is involved in the secretion of hormones and proinflammatory molecules that are associated with breast cancer development and progression. The role of adiposity and systemic inflammation in immune responses and resistance to anti-cancer treatment in breast cancer (BC) patients is still not clear. Metformin has demonstrated antitumorigenic properties both in pre-clinical and clinical studies. Nevertheless, its immunomodulating properties in BC are largely unknown. This review aims to evaluate the emerging evidence on the crosstalk between adiposity and the immune-tumour microenvironment in BC, its progression and treatment resistance, and the immunometabolic role of metformin in BC. Adiposity, and by extension subclinical inflammation, are associated with metabolic dysfunction and changes in the immune-tumour microenvironment in BC. In oestrogen receptor positive (ER+) breast tumours, it is proposed that these changes are mediated via a paracrine interaction between macrophages and preadipocytes, leading to elevated aromatase expression and secretion of pro-inflammatory cytokines and adipokines in the breast tissue in patients who are obese or overweight. In HER2+ breast tumours, WAT inflammation has been shown to be associated with resistance to trastuzumab mediated via MAPK or PI3K pathways. Furthermore, adipose tissue in patients with obesity is associated with upregulation of immune checkpoints on T-cells that is partially mediated via immunomodulatory effects of leptin and has been paradoxically associated with improved responses to immunotherapy in several cancers. Metformin may play a role in the metabolic reprogramming of tumour-infiltrating immune cells that are dysregulated by systemic inflammation. In conclusion, evidence suggests that body composition and metabolic status are associated with patient outcomes. To optimise patient stratification and personalisation of treatment, prospective studies are required to evaluate the role of body composition and metabolic parameters in metabolic immune reprogramming with and without immunotherapy in patients with BC. [ABSTRACT FROM AUTHOR]