1
|
Portillo EGD, Olivares-Hernández A, Gudino LC, Félix LC, Hernández LB, Domínguez LP, Jiménez DL, Sarmiento RG, Morillo EDB, Sánchez EF, Miramontes-Gonzáleze JP. Evaluation of the effect of metformin as a radiosensitiser in solid tumours: A systematic review. Clin Transl Radiat Oncol 2025; 52:100930. [PMID: 40028423 PMCID: PMC11871473 DOI: 10.1016/j.ctro.2025.100930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/13/2025] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Background Metformin is an antidiabetic drug that has shown its benefit in increasing the effect of radiotherapy in the treatment of solid tumors in preclinical studies. The objective of this systematic review is to study the effect of metformin as a radiosensitizer in studies carried out in clinical practice. Methods Systematic review carried out according to PRISMA criteria of clinical trials, systematic reviews and observational studies focused on the influence of metformin as a radiosensitizer in solid tumors. The studies were published between the years 2010 and 2022. The results of the studies have been analyzed in terms of survival (OS, PFS, DFS, DMFS) and response (ORR) between patients treated with metformin and without it. Results A total of 16 studies have been found in the literature (the most frequent tumor was prostate cancer, 5 studies). External radiotherapy was administered in all the studies and in two of them to greater brachytherapy. The use of metformin with radiotherapy showed a consistent benefit in terms of survival and response in tumors of prostate, hepatic and gynecological origin. The benefit in the rest of the tumors analyzed (lung, rectal, and head and neck cancer) is doubtful, and the results are contradictory. The greatest benefits were observed in prostate tumors both in OS and SLE. Conclusions The use of metformin in combination with radiotherapy in solid tumors is one of the most promising treatments under development in oncology. The benefit observed in real-life studies makes it necessary to develop clinical trials that specifically evaluate its use in clinical practice in the future.
Collapse
Affiliation(s)
| | - Alejandro Olivares-Hernández
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Medical Oncology, University Hospital of Salamanca, Salamanca, Spain
| | - Luis Corral Gudino
- Internal Medicine Unit, Río Hortega Hospital, Valladolid, Spain
- Medicine Department, University of Valladolid Medical School, Spain
| | - Laura Corvo Félix
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Medical Oncology, University Hospital of Salamanca, Salamanca, Spain
| | - Lorena Bellido Hernández
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Medical Oncology, University Hospital of Salamanca, Salamanca, Spain
- Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Luis Posado Domínguez
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Medical Oncology, University Hospital of Salamanca, Salamanca, Spain
| | | | - Rogelio González Sarmiento
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Internal Medicine Unit, Río Hortega Hospital, Valladolid, Spain
| | - Edel del Barco Morillo
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Medical Oncology, University Hospital of Salamanca, Salamanca, Spain
- Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Emilio Fonseca Sánchez
- Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
- Department of Medical Oncology, University Hospital of Salamanca, Salamanca, Spain
- Internal Medicine Unit, Río Hortega Hospital, Valladolid, Spain
| | - José Pablo Miramontes-Gonzáleze
- Internal Medicine Unit, Río Hortega Hospital, Valladolid, Spain
- Medicine Department, University of Valladolid Medical School, Spain
| |
Collapse
|
2
|
Yu R, Ji X, Zhang P, Zhang H, Qu H, Dong W. The potential of chimeric antigen receptor -T cell therapy for endocrine cancer. World J Surg Oncol 2025; 23:153. [PMID: 40264184 PMCID: PMC12012980 DOI: 10.1186/s12957-025-03745-x] [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: 12/20/2024] [Accepted: 03/07/2025] [Indexed: 04/24/2025] Open
Abstract
Endocrine cancer, a relatively rare and heterogeneous tumor with diverse clinical features. The facile synthesis of hormones further complicates endocrine cancer treatment. Thus, the development of safe and effective systemic treatment approaches, such as chimeric antigen receptor (CAR) T cell therapy, is imperative to enhance the prognosis of patients with endocrine cancer. Although this therapy has achieved good results in the treatment of hematological malignancies, it encounters diverse complications and challenges in the context of endocrine cancer. This review delineates the generation of CAR-T cells, examines the potential of CAR-T cell therapy for endocrine cancer, enumerates pivotal antigens linked to endocrine cancer, encapsulates the challenges confronted with CAR-T cell therapy for endocrine cancer, and expounds upon strategies to overcome these limitations. The primary objective is to provide insightful perspectives that can contribute to the advancement of CAR-T cell therapy in the field of endocrine cancer.
Collapse
Affiliation(s)
- Ruonan Yu
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Xiaoyu Ji
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Ping Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Huiling Qu
- Department of Neurology, The General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, Liaoning, 110840, China.
| | - Wenwu Dong
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
3
|
Meng HJ, Mao Y, Zhao DQ, Shi SG. Prognostic value of the triglyceride-glucose index in advanced gastric cancer: A call for further exploration. World J Gastroenterol 2025; 31:104574. [PMID: 40309236 PMCID: PMC12038551 DOI: 10.3748/wjg.v31.i15.104574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/09/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Gastric cancer (GC) remains a leading cause of cancer-related mortality worldwide, necessitating the identification of reliable prognostic indicators to enhance treatment outcomes. Recent research has highlighted the triglyceride-glucose (TyG) index as a potential surrogate marker for insulin resistance, which may significantly influence the prognosis of patients undergoing immunotherapy combined with chemotherapy. In this context, the study by Yao et al demonstrates that a high TyG index correlates with improved overall survival and progression-free survival in advanced GC patients receiving sintilimab and chemotherapy. Specifically, patients in the high TyG group had a significantly longer median progression-free survival of 9.8 months [95% confidence interval (CI): 9.2-10.9] compared to 8.0 months (95%CI: 7.5-8.5) in the low TyG group (hazard ratio = 0.58, 95%CI: 0.43-0.79, P < 0.001). Similarly, the median overall survival was significantly longer in the high TyG group at 23.1 months (95%CI: 21.2-NA) vs 16.5 months (95%CI: 13.9-18.3) in the low TyG group (hazard ratio = 0.30, 95%CI: 0.21-0.42, P < 0.001). These findings underscore the strong prognostic potential of the TyG index in guiding treatment strategies for advanced GC. These findings underscore the need for further investigation into the TyG index's role as a prognostic tool and its underlying mechanisms in influencing treatment efficacy. We advocate for additional multicenter studies to validate these results and explore the TyG index's applicability across diverse patient populations, ultimately aiming to refine treatment strategies and improve patient outcomes in advanced GC.
Collapse
Affiliation(s)
- Hong-Jie Meng
- Department of Gastrointestinal Surgery Ward, Zhuji People’s Hospital, Zhuji 311800, Zhejiang Province, China
| | - Yi Mao
- Department of Gastrointestinal Surgery Ward, Zhuji People’s Hospital, Zhuji 311800, Zhejiang Province, China
| | - De-Qing Zhao
- Department of Gastrointestinal Surgery Ward, Zhuji People’s Hospital, Zhuji 311800, Zhejiang Province, China
| | - Sheng-Guang Shi
- Department of Gastrointestinal Surgery Ward, Zhuji People’s Hospital, Zhuji 311800, Zhejiang Province, China
| |
Collapse
|
4
|
Wang X, Liao Y, Liu D, Zheng J, Shi M. Presetting CAR-T cells during ex vivo biomanufacturing. Mol Ther 2025; 33:1380-1406. [PMID: 39988874 PMCID: PMC11997485 DOI: 10.1016/j.ymthe.2025.02.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/21/2024] [Accepted: 02/19/2025] [Indexed: 02/25/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has revolutionized the treatment of hematologic malignancies. However, it continues to encounter significant obstacles, including treatment relapse and limited efficacy in solid tumors. While effector T cells exhibit robust cytotoxicity, central memory T cells and stem cell-like T cells are essential for in vivo expansion, long-term survival, and persistence. Strategies such as genetic engineering to enhance CAR-T cell efficacy and durability are often accompanied by increased safety risks, which not only raise regulatory approval thresholds but also escalate CAR-T production costs. In contrast, optimizing ex vivo manufacturing conditions represents a more straightforward and practical approach, offering the potential for rapid application to commercially approved CAR-T products and enhancement of their clinical outcomes. This review examines several factors that have been shown to improve T cell memory phenotype and in vivo cytotoxic activity, including cytokines, electrolytes, signaling pathway inhibitors, metabolic modulators, and epigenetic agents. The insights provided will guide the optimization of CAR-T cell industrial production. Furthermore, considerations for selecting appropriate conditions are discussed, balancing effectiveness, cost-efficiency, safety, and regulatory compliance while addressing current challenges in the field.
Collapse
Affiliation(s)
- Xu Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Ying Liao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Dan Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Ming Shi
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
5
|
Li W, Liu N, Chen M, Liu D, Liu S. Metformin as an immunomodulatory agent in enhancing head and neck squamous cell carcinoma therapies. Biochim Biophys Acta Rev Cancer 2025; 1880:189262. [PMID: 39827973 DOI: 10.1016/j.bbcan.2025.189262] [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: 08/05/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains a significant clinical challenge due to its aggressive behavior and poor prognosis, making the development of novel therapeutics with enhanced efficacy and minimal side effects critical. Metformin, a widely used antidiabetic agent, has recently emerged as a potential adjunctive therapy for HNSCC, exhibiting both direct anti-tumor and immunomodulatory effects. This review comprehensively explores the multifaceted role of metformin in shaping the tumor immune microenvironment within HNSCC. We emphasize its pivotal role in modulating immune cell populations and its potential for synergistic action with immunotherapeutic strategies. Furthermore, we address the current challenges associated with optimizing dosing regimens, identifying predictive biomarkers, and integrating metformin with immunotherapy. By dissecting these aspects, this review aims to pave the way for the development of personalized HNSCC treatment strategies that fully exploit the therapeutic potential of metformin.
Collapse
Affiliation(s)
- Wenting Li
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang 110002, Liaoning, China
| | - Nanshu Liu
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang 110002, Liaoning, China
| | - Mingwei Chen
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang 110002, Liaoning, China
| | - Dongjuan Liu
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang 110002, Liaoning, China.
| | - Sai Liu
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang 110002, Liaoning, China.
| |
Collapse
|
6
|
Sato K, Ogasawara H, Ikeda Y, Kumagai H, Inoue R, Tsuno T, Matsunaga K, Ishida E, Shirakawa J. The antitumor effects of metformin are potentially mediated through LPA receptor inhibition. Diabetes Res Clin Pract 2025; 222:112094. [PMID: 40073948 DOI: 10.1016/j.diabres.2025.112094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/20/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
AIMS Although metformin has antitumor effects, the detailed mechanism of action, particularly with respect to the cellular responses mediated through G protein-coupled receptors (GPCRs), remains unclear. METHODS AND RESULTS Here, we assayed a panel of 200 GPCRs in cells treated with metformin and reported that signaling through several receptors, including lysophosphatidic acid (LPA) receptors, was suppressed. Metformin significantly attenuated LPA-induced intracellular Ca2+ mobilization in LPA receptor 1 (LPAR1)-, 2 (LPAR2)-, and 3 (LPAR3)-transfected rat hepatoma RH7777 cells. LPA treatment increased LPAR3-transfected RH7777 cell adhesion and migration. This response to LPA was attenuated by treatment with the Gq/11 inhibitor YM-254890 and metformin. In contrast, these inhibitors had minimal effects on the cell migration induced by epidermal growth factor. CONCLUSIONS These results indicate that the inhibition of LPA receptor signaling by metformin, especially the consequent suppression of LPAR3-mediated cell migration, may contribute to the antitumor effects of metformin.
Collapse
Affiliation(s)
- Koichi Sato
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
| | | | - Yuichi Ikeda
- Tanso Biosciences, Inc., Bunkyo-ku, Tokyo 1130023, Japan
| | | | - Ryota Inoue
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan; Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Takahiro Tsuno
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan; Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan
| | - Koichi Matsunaga
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
| | - Emi Ishida
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan
| | - Jun Shirakawa
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi 371-8512, Japan; Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama 236-0004, Japan.
| |
Collapse
|
7
|
Smith RJ, Zollo R, Kalvapudi S, Vedire Y, Pachimatla AG, Petrucci C, Shaller G, Washington D, Rr V, Sass SN, Srinivasan A, Kannisto E, Bawek S, Jain P, Rosario S, Barbi J, Yendamuri S. Obesity-specific improvement of lung cancer outcomes and immunotherapy efficacy with metformin. J Natl Cancer Inst 2025; 117:673-684. [PMID: 39560490 PMCID: PMC11972684 DOI: 10.1093/jnci/djae295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/16/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Preclinical cancer studies ascribe promising anticancer properties to metformin. Yet, clinical findings vary, casting uncertainty on its therapeutic value for non-small cell lung cancer (NSCLC) patients. We hypothesized that metformin could benefit obese and overweight patients with NSCLC. METHODS We retrospectively analyzed 2 clinical cohorts and employed complementary mouse models to test our hypothesis. One cohort included NSCLC patients with overweight body mass index (≥25 kg/m2, n = 511) and nonoverweight body mass index (<25 kg/m2, n = 232) who underwent lobectomy, evaluating metformin's impact on clinical outcomes. Another cohort examined metformin's effect on progression-free survival after immune checkpoint inhibitors in overweight (n = 284) vs nonoverweight (n = 184) NSCLC patients. Metformin's effects on tumor progression, antitumor immunity, and immune checkpoint inhibitor response in obese and normal-weight mice were assessed with lung cancer models. RESULTS Metformin is associated with increased recurrence-free survival in overweight patients (hazard ratio [HR] = 0.47, 95% confidence interval [CI] = 0.24 to 0.94; P = .035) after lobectomy. It also corrected accelerated tumor growth in diet-induced obese mouse models in a lymphocyte-specific manner while reversing several mechanisms of immune suppression potentiated by obesity. Programmed cell death 1 blockade coupled with metformin was more effective at limiting tumor burden in obese mice and correlated with progression-free survival only in overweight patients on immunotherapy (HR = 0.60, 95% CI = 0.39 to 0.93; P = .024). CONCLUSIONS Metformin may improve lung cancer-specific clinical outcomes in obese and overweight lung cancer patients and enhance immunotherapy efficacy in this growing population. This work identifies obesity as a potential predictive biomarker of metformin's anticancer and immunotherapy-enhancing properties in lung cancer while shedding light on the underlying immunological phenomena.
Collapse
Affiliation(s)
- Randall J Smith
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Robert Zollo
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Sukumar Kalvapudi
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Yeshwanth Vedire
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Akhil Goud Pachimatla
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Cara Petrucci
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Garrison Shaller
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Deschana Washington
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Vethanayagam Rr
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Stephanie N Sass
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Aravind Srinivasan
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Eric Kannisto
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Sawyer Bawek
- Department of Medicine, University at Buffalo, State University of New York, Buffalo, NY 14263, United States
| | - Prantesh Jain
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Spencer Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Joseph Barbi
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Sai Yendamuri
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| |
Collapse
|
8
|
Suzuki H, Hasegawa S, Fushimi S, Tagami K, Nishikawa M, Kondo Y, Yasuda H. Metformin prevents diabetes development in type 1 diabetes models via suppression of mTOR and STAT3 signaling in immune cells. Sci Rep 2025; 15:10641. [PMID: 40148472 PMCID: PMC11950226 DOI: 10.1038/s41598-025-93647-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Type 1 diabetes (T1D) is an organ-specific autoimmune disease caused by T cell-mediated pancreatic β cell destruction. To evaluate the effects of metformin on immune cells in autoimmune diabetes, we administered metformin intraperitoneally to two T1D mouse models and analyzed autoimmune diabetes progression. In a cyclophosphamide (CY)-induced T1D model in male non-obese diabetic (NOD) mice, intraperitoneal administration of metformin significantly prevented autoimmune diabetes. Treatment with metformin showed a decrease in activated T cells, CD44hiCD62Llo effector memory cells, macrophages, and dendritic cells (DCs), and an increase in CD44hiCD62Lhi central memory cells, B cells, and regulatory T cells (Tregs) in splenocytes. Interestingly, metformin treatment showed a decrease in activated T cells, CD4+ effector memory T cells and Th1-type antigen-specific cells in PLN cells. IL-17 production was significantly suppressed in metformin-treated mice. TNF-α production from DCs in vitro was dose-dependently suppressed by metformin. Activity of mTOR signaling was significantly reduced in CD4+ T cells, CD8+ T cells, and B220+ B cells. In addition, activities of mTOR and STAT3 signaling in DCs were also reduced significantly. Furthermore, metformin treatment in female NOD mice, a spontaneous T1D model, significantly suppressed autoimmune diabetes onset as well and an increase in Tregs was observed. Our results suggest that metformin may suppress autoimmunity and have therapeutic potential in T1D progression as an immunomodulator.
Collapse
MESH Headings
- Animals
- Metformin/pharmacology
- Diabetes Mellitus, Type 1/prevention & control
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/drug therapy
- STAT3 Transcription Factor/metabolism
- TOR Serine-Threonine Kinases/metabolism
- Signal Transduction/drug effects
- Mice
- Mice, Inbred NOD
- Male
- Disease Models, Animal
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Hypoglycemic Agents/pharmacology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/drug effects
Collapse
Affiliation(s)
- Haruka Suzuki
- Division of Health Sciences, Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Shuji Hasegawa
- Division of Health Sciences, Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Sae Fushimi
- Division of Health Sciences, Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Kanako Tagami
- Division of Health Sciences, Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Minaho Nishikawa
- Division of Health Sciences, Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Yuichi Kondo
- Division of Health Sciences, Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan
| | - Hisafumi Yasuda
- Division of Health Sciences, Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, 654-0142, Japan.
- Department of General Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| |
Collapse
|
9
|
Dong J, Su T, Wu J, Xiang Y, Song M, He C, Shao L, Yang Y, Chen S. Drug functional remapping: a new promise for tumor immunotherapy. Front Oncol 2025; 15:1519355. [PMID: 40161377 PMCID: PMC11949826 DOI: 10.3389/fonc.2025.1519355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
The research and development of new anti-cancer drugs face challenges such as high costs, lengthy development cycles, and limited data on side effects. In contrast, the clinical safety and side effects of traditional drugs have been well established through long-term use. The development or repurposing of traditional drugs with potential applications in cancer treatment offers an economical, feasible, and promising strategy for new drug development. This article reviews the novel applications of traditional drugs in tumor immunotherapy, discussing how they can enhance tumor treatment efficacy through functional repositioning, while also reducing development time and costs. Recent advancements in cancer immunotherapy have revolutionized treatment options, but resistance to ICIs remains a significant challenge. Drug repurposing has emerged as a promising strategy to identify novel agents that can enhance the efficacy of immunotherapies by overcoming ICI resistance. A study suggests that drug repositioning has the potential to modulate immune cell activity or alter the tumor microenvironment, thereby circumventing the resistance mechanisms associated with immune checkpoint blockade. This approach provides a rapid and cost-effective pathway for identifying therapeutic candidates that can be quickly transitioned into clinical trials. To improve the effectiveness of tumor immunotherapy, it is crucial to explore systematic methods for identifying repurposed drug candidates. Methods such as high-throughput screening, computational drug repositioning, and bioinformatic analysis have been employed to efficiently identify potential candidates for cancer treatment. Furthermore, leveraging databases related to immunotherapy and drug repurposing can provide valuable resources for drug discovery and facilitate the identification of promising compounds. It focuses on the latest advancements in the use of antidiabetic drugs, antihypertensive agents, weight-loss medications, antifungal agents, and antiviral drugs in tumor immunotherapy, examining their mechanisms of action, clinical application prospects, and associated challenges. In this context, our aim is to explore these strategies and highlight their potential for expanding the therapeutic options available for cancer immunotherapy, providing valuable references for cancer research and treatment.
Collapse
Affiliation(s)
- Jiayi Dong
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ting Su
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiexiong Wu
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yu Xiang
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Minghan Song
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
| | - Canfeng He
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lijuan Shao
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yubin Yang
- Traditional Chinese Medicine Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Size Chen
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
10
|
Cote AL, Munger CJ, Ringel AE. Emerging insights into the impact of systemic metabolic changes on tumor-immune interactions. Cell Rep 2025; 44:115234. [PMID: 39862435 DOI: 10.1016/j.celrep.2025.115234] [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: 09/17/2024] [Revised: 11/24/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Tumors are inherently embedded in systemic physiology, which contributes metabolites, signaling molecules, and immune cells to the tumor microenvironment. As a result, any systemic change to host metabolism can impact tumor progression and response to therapy. In this review, we explore how factors that affect metabolic health, such as diet, obesity, and exercise, influence the interplay between cancer and immune cells that reside within tumors. We also examine how metabolic diseases influence cancer progression, metastasis, and treatment. Finally, we consider how metabolic interventions can be deployed to improve immunotherapy. The overall goal is to highlight how metabolic heterogeneity in the human population shapes the immune response to cancer.
Collapse
Affiliation(s)
- Andrea L Cote
- Ragon Institute of Mass General, MIT, and Harvard, 600 Main Street, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, 31 Ames Street, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Chad J Munger
- Ragon Institute of Mass General, MIT, and Harvard, 600 Main Street, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, 31 Ames Street, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Alison E Ringel
- Ragon Institute of Mass General, MIT, and Harvard, 600 Main Street, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, 31 Ames Street, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA.
| |
Collapse
|
11
|
Wang Y, Sun Y, Hu J, Ma H. Clinical Effect of Treatment with Metformin for Type 2 Diabetes on Non-Small Cell Lung Cancer Patients Undergoing Immunotherapy: A Retrospective Study. Int J Gen Med 2024; 17:6595-6604. [PMID: 39759894 PMCID: PMC11699848 DOI: 10.2147/ijgm.s495449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/24/2024] [Indexed: 01/07/2025] Open
Abstract
Purpose To further identify the clinical impact of metformin on the prognosis of non-small cell lung cancer (NSCLC) with type 2 diabetes who received immunotherapy. Methods Stage IV NSCLC patients with type 2 diabetes receiving the immunotherapy from 2017 to 2021 were retrospectively enrolled and divided into the metformin group or non-metformin group according to the treatment strategy for type 2 diabetes (metformin vs other hypoglycemic medicines). The overall response rate (ORR) was primary endpoint, and overall survival (OS), progression-free survival (PFS) and disease control rate (DCR) were secondary endpoints. These outcomes were compared between two groups. Results A total of 34 patients were eventually enrolled, including 18 patients in the metformin group. No significant differences in the basic characteristics and incidence of adverse events were observed between two groups. In addition, there was no significant difference in ORR (44.4%, 8/18 vs 25.0%, 4/16, P = 0.236) and DCR (77.8%, 14/18 vs 75.0%, 12/16, P > 0.999) between the metformin and non-metformin groups. Kaplan-Meier survival curve (P = 0.039) and Cox regression analysis indicated that the use of metformin was an independent factor for OS (HR: 0.310, 95% CI: 0.113-0.845, P = 0.022), but not for PFS (Cox regression analysis: P = 0.145). Conclusion For NSCLC patients with type 2 diabetes, the combination of metformin and immunotherapy may contribute to OS benefits. However, more high-quality prospective studies with big sample sizes are needed to further clarify the effect of metformin use on the efficacy of immunotherapy in advanced NSCLC patients with diabetes.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
- Department of Thoracic Surgery, Affiliated Hospital of Chengdu University, Chengdu, 610081, People’s Republic of China
| | - Yu Sun
- Department of Thoracic Surgery, Affiliated Hospital of Chengdu University, Chengdu, 610081, People’s Republic of China
| | - Jingguo Hu
- Department of Thoracic Surgery, Affiliated Hospital of Chengdu University, Chengdu, 610081, People’s Republic of China
| | - Haitao Ma
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| |
Collapse
|
12
|
Capelletti MM, Montini O, Ruini E, Tettamanti S, Savino AM, Sarno J. Unlocking the Heterogeneity in Acute Leukaemia: Dissection of Clonal Architecture and Metabolic Properties for Clinical Interventions. Int J Mol Sci 2024; 26:45. [PMID: 39795903 PMCID: PMC11719665 DOI: 10.3390/ijms26010045] [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: 11/21/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Genetic studies of haematological cancers have pointed out the heterogeneity of leukaemia in its different subpopulations, with distinct mutations and characteristics, impacting the treatment response. Next-generation sequencing (NGS) and genome-wide analyses, as well as single-cell technologies, have offered unprecedented insights into the clonal heterogeneity within the same tumour. A key component of this heterogeneity that remains unexplored is the intracellular metabolome, a dynamic network that determines cell functions, signalling, epigenome regulation, immunity and inflammation. Understanding the metabolic diversities among cancer cells and their surrounding environments is therefore essential in unravelling the complexities of leukaemia and improving therapeutic strategies. Here, we describe the currently available methodologies and approaches to addressing the dynamic heterogeneity of leukaemia progression. In the second section, we focus on metabolic leukaemic vulnerabilities in acute myeloid leukaemia (AML) and acute lymphoblastic leukaemia (ALL). Lastly, we provide a comprehensive overview of the most interesting clinical trials designed to target these metabolic dependencies, highlighting their potential to advance therapeutic strategies in leukaemia treatment. The integration of multi-omics data for cancer identification with the metabolic states of tumour cells will enable a comprehensive "micro-to-macro" approach for the refinement of clinical practices and delivery of personalised therapies.
Collapse
Affiliation(s)
- Martina Maria Capelletti
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Orsola Montini
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Emilio Ruini
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
| | - Sarah Tettamanti
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Angela Maria Savino
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Jolanda Sarno
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| |
Collapse
|
13
|
Anand S, Patel TN. Integrating the metabolic and molecular circuits in diabetes, obesity and cancer: a comprehensive review. Discov Oncol 2024; 15:779. [PMID: 39692821 DOI: 10.1007/s12672-024-01662-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/02/2024] [Indexed: 12/19/2024] Open
Abstract
The progressive globalization of sedentary lifestyles and diets rich in lipids and processed foods has caused two major public health hazards-diabetes and obesity. The strong interlink between obesity and type 2 diabetes mellitus and their combined burden encompass them into a single term 'Diabesity'. They have also been tagged as the drivers for the onset of cancer. The clinical association between diabetes, obesity, and several types of human cancer demands an assessment of vital junctions correlating the three. This review focuses on revisiting the molecular axis linking diabetes and obesity to cancer through pathways that get imbalanced owing to metabolic upheaval. We also attempt to describe the functional disruptions of DNA repair mechanisms due to overwhelming oxidative DNA damage caused by diabesity. Genomic instability, a known cancer hallmark results when DNA repair does not work optimally, and as will be inferred from this review the obtruded metabolic homeostasis in diabetes and obesity creates a favorable microenvironment supporting metabolic reprogramming and enabling malignancies. Altered molecular and hormonal landscapes in these two morbidities provide a novel connection between metabolomics and oncogenesis. Understanding various aspects of the tumorigenic process in diabesity-induced cancers might help in the discovery of new biomarkers and prompt targeted therapeutic interventions.
Collapse
Affiliation(s)
- Shrikirti Anand
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Trupti N Patel
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
14
|
Abdallah FM, Ghoneim AI, Abd-Alhaseeb MM, Abdel-Raheem IT, Helmy MW. Unveiling the antitumor synergy between pazopanib and metformin on lung cancer through suppressing p-Akt/ NF-κB/ STAT3/ PD-L1 signal pathway. Biomed Pharmacother 2024; 180:117468. [PMID: 39332188 DOI: 10.1016/j.biopha.2024.117468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/01/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024] Open
Abstract
Pazopanib, an inhibitor of the VEGF receptor tyrosine kinase, has demonstrated significant antitumor effects in lung cancer. However, its application as a standard treatment for this type of cancer is limited by its drug resistance and toxicity. Metformin has the potential to combat lung cancer by modifying the tumor's immune microenvironment. In this study, we investigated the potential antitumor effects and the associated underlying molecular mechanisms of the combination of pazopanib and metformin in lung cancer. In vitro studies were conducted using the A549 and H460 lung cancer cell lines, whereas urethane-induced lung cancer-bearing mice were used for in vivo assessments. The urethane-induced mice received oral administration of pazopanib (50 mg/kg) and/or metformin (250 mg/kg) for a duration of 21 days. The results indicated that the MTT assay demonstrated a combined cytotoxic effect of the pazopanib/metformin combination in H460 and A549 cells, as evidenced by CI and DRI analyses. The observed increase in annexin V levels and the corresponding increase in Caspase-3 activity strongly suggest that this combination induced apoptosis. Furthermore, the pazopanib/metformin combination significantly inhibited the p-Akt/NF-κB/IL-6/STAT3, HIF1α/VEGF, and TLR2/TGF-β/PD-L1 pathways while also increasing CD8 expression in vivo. Immunohistochemical analysis revealed that these antitumor mechanisms were manifested by the suppression of the proliferation marker Ki67. In conclusion, these findings revealed that metformin augments the antitumor efficacy of pazopanib in lung cancer by simultaneously targeting proliferative, angiogenic, and immunogenic signaling pathways, metformin enhances the antitumor effectiveness of pazopanib in lung cancer, making it a promising therapeutic option for lung cancer.
Collapse
Affiliation(s)
- Fatma M Abdallah
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, 22511, Egypt; Faculty of Health Sciences Technology, Borg Al Arab Technological University, New Borg El Arab, Egypt.
| | - Asser I Ghoneim
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, 22511, Egypt.
| | - Mohammad M Abd-Alhaseeb
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, 22511, Egypt; Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.
| | - Ihab T Abdel-Raheem
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, 22511, Egypt.
| | - Maged W Helmy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, 22511, Egypt.
| |
Collapse
|
15
|
Taranto D, Kloosterman DJ, Akkari L. Macrophages and T cells in metabolic disorder-associated cancers. Nat Rev Cancer 2024; 24:744-767. [PMID: 39354070 DOI: 10.1038/s41568-024-00743-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2024] [Indexed: 10/03/2024]
Abstract
Cancer and metabolic disorders have emerged as major global health challenges, reaching epidemic levels in recent decades. Often viewed as separate issues, metabolic disorders are shown by mounting evidence to heighten cancer risk and incidence. The intricacies underlying this connection are still being unraveled and encompass a complex interplay between metabolites, cancer cells and immune cells within the tumour microenvironment (TME). Here, we outline the interplay between metabolic and immune cell dysfunction in the context of three highly prevalent metabolic disorders, namely obesity; two associated liver diseases, metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH); and type 2 diabetes. We focus primarily on macrophages and T cells, the critical roles of which in dictating inflammatory response and immune surveillance in metabolic disorder-associated cancers are widely reported. Moreover, considering the ever-increasing number of patients prescribed with metabolism disorder-altering drugs and diets in recent years, we discuss how these therapies modulate systemic and local immune phenotypes, consequently impacting cancer malignancy. Collectively, unraveling the determinants of metabolic disorder-associated immune landscape and their role in fuelling cancer malignancy will provide a framework essential to therapeutically address these highly prevalent diseases.
Collapse
Affiliation(s)
- Daniel Taranto
- Division of Tumour Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daan J Kloosterman
- Division of Tumour Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Leila Akkari
- Division of Tumour Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
16
|
Wissbroecker KB, Zmuda AJ, Karumanchi H, Niehaus TD. Biochemical and genomic evidence for converging metabolic routes of metformin and biguanide breakdown in environmental Pseudomonads. J Biol Chem 2024; 300:107935. [PMID: 39476966 DOI: 10.1016/j.jbc.2024.107935] [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: 09/02/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 12/01/2024] Open
Abstract
Metformin is commonly used to lower blood glucose levels and is one of the most widely used pharmaceuticals worldwide. Typical doses are high (0.5-2.0 g day-1) and the majority travels through the digestive system unabsorbed and enters the wastewater system. Metformin is not removed by standard wastewater treatments and eventually enters freshwater systems, where it can form N-chloro-derivatives that are toxic to fish and human cells. Thus, metformin is one of the most prevalent anthropogenic pollutants worldwide and there has been considerable interest in finding ways to remove it. We recently isolated Pseudomonads capable of growing on metformin as the sole nitrogen source. We identified candidate genes involved in metformin breakdown through genomic analyses informed by feeding studies. One candidate, a pair of genes that are located on ∼80kb extra-genomic plasmids, was shown to encode a heteromeric Ni-dependent hydrolase that converts metformin to guanylurea and dimethylamine. Metforminase activity of these gene products is now well established as our results confirm three recently published independent studies. Our isolated Pseudomonads also grow on biguanide, suggesting the existence of an additional breakdown enzyme. Another candidate gene located on the ∼80kb plasmids was shown to encode an aminohydrolase that converts biguanide to guanylurea. Biguanide may arise through successive N-demethylations of metformin or come from other sources. Our results suggest that the recent evolution of metforminase and biguanide hydrolase enzymes allow Pseudomonads to convert either metformin or biguanide to guanylurea, which can be assimilated by existing pathways.
Collapse
Affiliation(s)
- Katie B Wissbroecker
- The Department of Plant and Microbial Biology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anthony J Zmuda
- The Department of Plant and Microbial Biology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Harsheeth Karumanchi
- The Department of Plant and Microbial Biology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Thomas D Niehaus
- The Department of Plant and Microbial Biology, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
17
|
Han S, Bommireddy R, Kim P, Selvaraj P, Shin DM. Chemoprevention of Head and Neck Cancer: A Review of Current Approaches and Future Perspectives. Cancer Prev Res (Phila) 2024; 17:443-455. [PMID: 38978394 PMCID: PMC11844769 DOI: 10.1158/1940-6207.capr-24-0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/07/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a spectrum of heterogeneous malignancies. A variety of genetic, environmental, and lifestyle factors contribute to the development of HNSCC. Carcinogenesis is a multistep process in which cell proliferation-associated oncogenes and cell-cycle regulation-associated tumor suppressor genes are dysregulated, resulting in premalignant lesions. Immune evasion is a critical step in the progression of benign lesions to advanced cancer. This review discusses the advances that have been made in chemoprevention strategies for HNSCC. The rationale for the use of chemopreventive agents to inhibit head and neck cancer development is highlighted by the positive outcomes of several clinical trials. We discuss the potential of some of the commonly studied agents including vitamin A analogs, EGFR inhibitors, COX-2 inhibitors, metabolic modulators, and natural compounds such as green tea, as well as immunotherapy and photodynamic therapy to prevent HNSCC. Our review provides insight into the potential benefits of these agents and the gaps that remain to be addressed. The published results reaffirm the promise of chemoprevention in head and neck cancer and suggest that continued exploration is needed to overcome the limitations. Because the current focus on chemopreventive agents is limited, major efforts in precision oncology approaches and substantial increase in funding will promote research into chemoprevention, which will eventually decrease the incidence of HNSCC.
Collapse
Affiliation(s)
- Sanghoon Han
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Ramireddy Bommireddy
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Pauline Kim
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
- Department of Pharmaceutical Services, Emory University Hospital Midtown, Atlanta, GA
| | - Periasamy Selvaraj
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA
| | - Dong M. Shin
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
18
|
Kang DH, Kim M, Lew BL, Kwon SH. Metformin is not associated with decreased risk of non-melanoma skin cancer: A distributed network analysis of 14 real-world databases in Korea. J Eur Acad Dermatol Venereol 2024; 38:e880-e882. [PMID: 38545875 DOI: 10.1111/jdv.19977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 03/11/2024] [Indexed: 09/26/2024]
Affiliation(s)
- Da-Hyun Kang
- Department of Dermatology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Korea
| | - Myoungsuk Kim
- Healthcare Big-Data Center, Research Institute of Clinical Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Bark-Lynn Lew
- Department of Dermatology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Korea
| | - Soon-Hyo Kwon
- Department of Dermatology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Korea
| |
Collapse
|
19
|
Park SH, Lee J, Yun HJ, Kim SH, Lee JH. Metformin Suppresses Both PD-L1 Expression in Cancer Cells and Cancer-Induced PD-1 Expression in Immune Cells to Promote Antitumor Immunity. Ann Lab Med 2024; 44:426-436. [PMID: 38529546 PMCID: PMC11169777 DOI: 10.3343/alm.2023.0443] [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: 11/09/2023] [Revised: 01/03/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024] Open
Abstract
Background Metformin, a drug prescribed for patients with type 2 diabetes, has potential efficacy in enhancing antitumor immunity; however, the detailed underlying mechanisms remain to be elucidated. Therefore, we aimed to identify the inhibitory molecular mechanisms of metformin on programmed death ligand 1 (PD-L1) expression in cancer cells and programmed death 1 (PD-1) expression in immune cells. Methods We employed a luciferase reporter assay, quantitative real-time PCR, immunoblotting analysis, immunoprecipitation and ubiquitylation assays, and a natural killer (NK) cell-mediated tumor cell cytotoxicity assay. A mouse xenograft tumor model was used to evaluate the effect of metformin on tumor growth, followed by flow-cytometric analysis using tumor-derived single-cell suspensions. Results Metformin decreased AKT-mediated β-catenin S552 phosphorylation and subsequent β-catenin transactivation in an adenosine monophosphate-activated protein kinase (AMPK) activation-dependent manner, resulting in reduced CD274 (encoding PD-L1) transcription in cancer cells. Tumor-derived soluble factors enhanced PD-1 protein stability in NK and T cells via dissociation of PD-1 from ubiquitin E3 ligases and reducing PD-1 polyubiquitylation. Metformin inhibited the tumor-derived soluble factor-reduced binding of PD-1 to E3 ligases and PD-1 polyubiquitylation, resulting in PD-1 protein downregulation in an AMPK activation-dependent manner. These inhibitory effects of metformin on both PD-L1 and PD-1 expression ameliorated cancer-reduced cytotoxic activity of immune cells in vitro and decreased tumor immune evasion and growth in vivo. Conclusions Metformin blocks both PD-L1 and PD-1 within the tumor microenvironment. This study provided a mechanistic insight into the efficacy of metformin in improving immunotherapy in human cancer.
Collapse
Affiliation(s)
- Su Hwan Park
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, Korea
| | - Juheon Lee
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, Korea
| | - Hye Jin Yun
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, Korea
| | - Seok-Ho Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, Korea
- Department of Medicinal Biotechnology, College of Health Science, Dong-A University, Busan, Korea
| | - Jong-Ho Lee
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, Korea
- Department of Biomedical Sciences, Dong-A University, Busan, Korea
| |
Collapse
|
20
|
Rimini M, Montes M, Amadeo E, Vitiello F, Kudo M, Tada T, Suda G, Shimose S, Lonardi S, Finkelmeier F, Salani F, Antonuzzo L, Marra F, Iavarone M, Cabibbo G, Foschi FG, Silletta M, Sacco R, Rapposelli IG, Scartozzi M, Nicoletta P, Aldrighetti L, Persano M, Camera S, Rossari F, Foti S, Kumada T, Hiraoka A, Iwamoto H, Rizzato MD, Himmelsbach V, Masi G, Corradi M, Celsa C, Fabio C, Frassineti GL, Cascinu S, Casadei-Gardini A, Presa J. Impact of metformin, statin, aspirin and insulin on the prognosis of uHCC patients receiving first line Lenvatinib or Atezolizumab plus Bevacizumab. Sci Rep 2024; 14:20200. [PMID: 39215078 PMCID: PMC11364777 DOI: 10.1038/s41598-024-70928-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Recently, in Hepatocellular carcinoma (HCC) setting, the use of metformin has been associated to a trend toward worse response rate, overall survival and progression free survival in patients who received immunotherapy. The study population included individuals from both Eastern and Western regions with a confirmed diagnosis of HCC and receiving first line treatment with Atezolizumab plus bevacizumab or Lenvatinib. Univariate and multivariate analyses were performed by Cox proportional. For the analysis, patients were stratified based on their use of concomitant medication or not. At the time of database lock, 319 deaths were observed: 209 in the Lenvatinib cohort, 110 in the Atezolizumab plus bevacizumab cohort. In the Atezolizumab plus Bevacizumab arm, 50 (16.5%) patients were on chronic metformin use. At the univariate analysis for OS, patients who used metformin showed significantly shorter OS compared to patients who did not use metformin (HR 1.9, 95% CI 1.1-3.2). Multivariate analysis confirmed that patients in metformin group had significantly shorter OS compared to patients in no-metformin group (HR 1.9; 95% CI 1.1-3.1). At the univariate analysis for PFS, patients in metformin group had significantly shorter PFS compared to patients in no-metformin group (HR 1.6, 95% CI 1.0-2.6). Multivariate analysis confirmed that patients in metformin group had significantly shorter PFS compared to patients in no-metformin group (HR 1.7; 95% CI 1.1-2.7; p = 0.0147). No differences were reported in terms of ORR and DCR between patients in metformin group and those in no-metformin group. In the Lenvatinib cohort, 65 (15%) patients were recorded to chronically use metformin. No statistically significant differences in terms of both OS and PFS were found between patients in metformin group and patients in no-metformin group. This analysis unveils a negative prognostic role associated with metformin use specifically within the Atezolizumab plus Bevacizumab group.
Collapse
Affiliation(s)
- Margherita Rimini
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Vita-Salute San Raffaele University, Milan, Italy
- Vita-Salute University San Raffaele, Milan, Italy
| | | | - Elisabeth Amadeo
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Vitiello
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, Higashiosaka, Japan
| | - Toshifumi Tada
- Department of Internal Medicine, Japanese Red Cross Himeji Hospital, Himeji, Japan
| | - Goki Suda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, North 15, West 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Shigeo Shimose
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, 830-0011, Japan
| | - Sara Lonardi
- Oncology Unit 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Fabian Finkelmeier
- Department of Internal Medicine 1, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Francesca Salani
- Unit of Medical Oncology 2, University Hospital of Pisa, Pisa, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Fabio Marra
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Massimo Iavarone
- Division of Gastroenterology and Hepatology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giuseppe Cabibbo
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, PROMISE, University of Palermo, Palermo, Italy
| | | | - Marianna Silletta
- Department of Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Rodolfo Sacco
- Gastroenterology and Digestive Endoscopy Unit, Foggia University Hospital, Foggia, Puglia, Italy
| | | | - Mario Scartozzi
- Medical Oncology, University and University Hospital of Cagliari, Cagliari, Italy
| | - Pella Nicoletta
- Department of Oncology, ASUFC University Hospital, Udine, Italy
| | - Luca Aldrighetti
- Hepato-Biliary Surgery Department, IRCCS San Raffaele Hospital, Milan, Italy
| | - Mara Persano
- Medical Oncology, University and University Hospital of Cagliari, Cagliari, Italy
| | - Silvia Camera
- Medical Oncology, University and University Hospital of Cagliari, Cagliari, Italy
| | - Federico Rossari
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Silvia Foti
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Takashi Kumada
- Department of Internal Medicine, Japanese Red Cross Himeji Hospital, Himeji, Japan
| | - Atsushi Hiraoka
- Department of Internal Medicine, Japanese Red Cross Himeji Hospital, Himeji, Japan
| | - Hideki Iwamoto
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka, 830-0011, Japan
| | | | - Vera Himmelsbach
- Department of Internal Medicine 1, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Gianluca Masi
- Unit of Medical Oncology 2, University Hospital of Pisa, Pisa, Italy
| | - Mattia Corradi
- Division of Gastroenterology and Hepatology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Ciro Celsa
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, PROMISE, University of Palermo, Palermo, Italy
| | - Conti Fabio
- Department of Internal Medicine, Ospedale degli Infermi di Faenza, Faenza, Emilia Romagna, Italy
| | | | - Stefano Cascinu
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Vita-Salute San Raffaele University, Milan, Italy
- Vita-Salute University San Raffaele, Milan, Italy
| | - Andrea Casadei-Gardini
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Vita-Salute San Raffaele University, Milan, Italy.
- Vita-Salute University San Raffaele, Milan, Italy.
| | | |
Collapse
|
21
|
Li JH, Hsin PY, Hsiao YC, Chen BJ, Zhuang ZY, Lee CW, Lee WJ, Vo TTT, Tseng CF, Tseng SF, Lee IT. A Narrative Review: Repurposing Metformin as a Potential Therapeutic Agent for Oral Cancer. Cancers (Basel) 2024; 16:3017. [PMID: 39272875 PMCID: PMC11394296 DOI: 10.3390/cancers16173017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Oral cancer, particularly oral squamous cell carcinoma (OSCC), is a significant global health challenge because of its high incidence and limited treatment options. Major risk factors, including tobacco use, alcohol consumption, and specific microbiota, contribute to the disease's prevalence. Recently, a compelling association between diabetes mellitus (DM) and oral cancer has been identified, with metformin, a widely used antidiabetic drug, emerging as a potential therapeutic agent across various cancers, including OSCC. This review explores both preclinical and clinical studies to understand the mechanisms by which metformin may exert its anticancer effects, such as inhibiting cancer cell proliferation, inducing apoptosis, and enhancing the efficacy of existing treatments. Preclinical studies demonstrate that metformin modulates crucial metabolic pathways, reduces inflammation, and impacts cellular proliferation, thereby potentially lowering cancer risk and improving patient outcomes. Additionally, metformin's ability to reverse epithelial-to-mesenchymal transition (EMT), regulate the LIN28/let-7 axis, and its therapeutic role in head and neck squamous cell carcinoma (HNSCC) are examined through experimental models. In clinical contexts, metformin shows promise in enhancing therapeutic outcomes and reducing recurrence rates, although challenges such as drug interactions, complex dosing regimens, and risks such as vitamin B12 deficiency remain. Future research should focus on optimizing metformin's application, investigating its synergistic effects with other therapies, and conducting rigorous clinical trials to validate its efficacy in OSCC treatment. This dual exploration underscores metformin's potential to play a transformative role in both diabetes management and cancer care, potentially revolutionizing oral cancer treatment strategies.
Collapse
Affiliation(s)
- Jui-Hsiang Li
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 33004, Taiwan
| | - Pei-Yi Hsin
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yung-Chia Hsiao
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Bo-Jun Chen
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Zhi-Yun Zhuang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chiang-Wen Lee
- Department of Nursing, Division of Basic Medical Sciences, Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chiayi 61363, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi 61363, Taiwan
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| | - Wei-Ju Lee
- School of Food Safety, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
| | - Thi Thuy Tien Vo
- Faculty of Dentistry, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Vietnam
| | - Chien-Fu Tseng
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei 10048, Taiwan
- Department of Dentistry, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 33004, Taiwan
| | - Shih-Fen Tseng
- Department of Emergency Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 33004, Taiwan
| | - I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
22
|
Sirtori CR, Castiglione S, Pavanello C. METFORMIN: FROM DIABETES TO CANCER TO PROLONGATION OF LIFE. Pharmacol Res 2024; 208:107367. [PMID: 39191336 DOI: 10.1016/j.phrs.2024.107367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
The metformin molecule dates back to over a century, but its clinical use started in the '50s. Since then, its use in diabetics has grown constantly, with over 150 million users today. The therapeutic profile also expanded, with improved understanding of novel mechanisms. Metformin has a major activity on insulin resistance, by acting on the insulin receptors and mitochondria, most likely by activation of the adenosine monophosphate-activated kinase. These and associated mechanisms lead to significant lipid lowering and body weight loss. An anti-cancer action has come up in recent years, with mechanisms partly dependent on the mitochondrial activity and also on phosphatidylinositol 3-kinase resistance occurring in some malignant tumors. The potential of metformin to raise life-length is the object of large ongoing studies and of several basic and clinical investigations. The present review article will attempt to investigate the basic mechanisms behind these diverse activities and the potential clinical benefits. Metformin may act on transcriptional activity by histone modification, DNA methylation and miRNAs. An activity on age-associated inflammation (inflammaging) may occur via activation of the nuclear factor erythroid 2 related factor and changes in gut microbiota. A senolytic activity, leading to reduction of cells with the senescent associated secretory phenotype, may be crucial in lifespan prolongation as well as in ancillary properties in age-associated diseases, such as Parkinson's disease. Telomere prolongation may be related to the activity on mitochondrial respiratory factor 1 and on peroxisome gamma proliferator coactivator 1-alpha. Very recent observations on the potential to act on the most severe neurological disorders, such as amyotrophic lateral sclerosis and frontotemporal dementia, have raised considerable hope.
Collapse
Affiliation(s)
- Cesare R Sirtori
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - Sofia Castiglione
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Chiara Pavanello
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
23
|
van Eijck CWF, Vadgama D, van Eijck CHJ, Wilmink JW. Metformin boosts antitumor immunity and improves prognosis in upfront resected pancreatic cancer: an observational study. J Natl Cancer Inst 2024; 116:1374-1383. [PMID: 38530777 PMCID: PMC11308183 DOI: 10.1093/jnci/djae070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/12/2024] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Beyond demographic and immune factors, metabolic considerations, particularly metformin's recognized impact in oncology, warrant exploration in treating pancreatic cancer. This study aimed to investigate the influence of metformin on patient survival and its potential correlation with distinct immune profiles in pancreatic ductal adenocarcinoma (PDAC) tumors. METHODS We included 82 upfront resected and 66 gemcitabine-based neoadjuvant chemoradiotherapy (nCRT)-treated patients from the PREOPANC randomized controlled trial (RCT). Transcriptomic NanoString immunoprofiling was performed for a subset of 96 available resected specimens. RESULTS Disparities in survival outcomes and immune profiles were apparent between metformin and non-metformin users in upfront resected patients but lacking in nCRT-treated patients. Compared to non-metformin users, upfront resected metformin users showed a higher median overall survival (OS) of 29 vs 14 months and a better 5-year OS rate of 19% vs 5%. Furthermore, metformin use was a favorable prognostic factor for OS in the upfront surgery group (HR = 0.56; 95% CI = 0.32 to 0.99). Transcriptomic data revealed that metformin users significantly underexpressed genes related to pro-tumoral immunity, including monocyte to M2 macrophage polarization and activation. Furthermore, the relative abundance of anti-inflammatory CD163+ MRC1+ M2 macrophages in non-metformin users and immune-activating CD1A+ CD1C+ dendritic cells in metformin users was heightened (P < .001). CONCLUSION This study unveils immune profile changes resulting from metformin use in upfront resected pancreatic cancer patients, possibly contributing to prolonged survival outcomes. Specifically, metformin use may decrease the abundance and activity of pro-tumoral M2 macrophages and increase the recruitment and function of tumor-resolving DCs, favoring antitumor immunity.[PREOPANC trial EudraCT: 2012-003181-40].
Collapse
Affiliation(s)
- Casper W F van Eijck
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Disha Vadgama
- Erasmus MC Cancer Institute, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Casper H J van Eijck
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Johanna W Wilmink
- Department of Medical Oncology, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| |
Collapse
|
24
|
Thomas P, Paris P, Pecqueur C. Arming Vδ2 T Cells with Chimeric Antigen Receptors to Combat Cancer. Clin Cancer Res 2024; 30:3105-3116. [PMID: 38747974 PMCID: PMC11292201 DOI: 10.1158/1078-0432.ccr-23-3495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/19/2024] [Accepted: 04/18/2024] [Indexed: 08/02/2024]
Abstract
Immunotherapy has emerged as a promising approach in the field of cancer treatment, with chimeric antigen receptor (CAR) T-cell therapy demonstrating remarkable success. However, challenges such as tumor antigen heterogeneity, immune evasion, and the limited persistence of CAR-T cells have prompted the exploration of alternative cell types for CAR-based strategies. Gamma delta T cells, a unique subset of lymphocytes with inherent tumor recognition capabilities and versatile immune functions, have garnered increasing attention in recent years. In this review, we present how arming Vδ2-T cells might be the basis for next-generation immunotherapies against solid tumors. Following a comprehensive overview of γδ T-cell biology and innovative CAR engineering strategies, we discuss the clinical potential of Vδ2 CAR-T cells in overcoming the current limitations of immunotherapy in solid tumors. Although the applications of Vδ2 CAR-T cells in cancer research are relatively in their infancy and many challenges are yet to be identified, Vδ2 CAR-T cells represent a promising breakthrough in cancer immunotherapy.
Collapse
Affiliation(s)
- Pauline Thomas
- Nantes Université, CRCI2NA, INSERM, CNRS, Nantes, France
| | - Pierre Paris
- Nantes Université, CRCI2NA, INSERM, CNRS, Nantes, France
| | | |
Collapse
|
25
|
Duan Z, Shi R, Gao B, Cai J. N-linked glycosylation of PD-L1/PD-1: an emerging target for cancer diagnosis and treatment. J Transl Med 2024; 22:705. [PMID: 39080767 PMCID: PMC11290144 DOI: 10.1186/s12967-024-05502-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
During tumorigenesis and progression, the immune checkpoint programmed death-1 (PD-1) and its ligand programmed death ligand-1 (PD-L1) play critical roles in suppressing T cell-mediated anticancer immune responses, leading to T-cell exhaustion and subsequent tumor evasion. Therefore, anti-PD-L1/PD-1 therapy has been an attractive strategy for treating cancer over the past decade. However, the overall efficacy of this approach remains suboptimal, revealing an urgent need for novel insights. Interestingly, increasing evidence indicates that both PD-L1 on tumor cells and PD-1 on tumor-specific T cells undergo extensive N-linked glycosylation, which is essential for the stability and interaction of these proteins, and this modification promotes tumor evasion. In various preclinical models, targeting the N-linked glycosylation of PD-L1/PD-1 was shown to significantly increase the efficacy of PD-L1/PD-1 blockade therapy. Furthermore, deglycosylation of PD-L1 strengthens the signal intensity in PD-L1 immunohistochemistry (IHC) assays, improving the diagnostic and therapeutic relevance of this protein. In this review, we provide an overview of the regulatory mechanisms underlying the N-linked glycosylation of PD-L1/PD-1 as well as the crucial role of N-linked glycosylation in PD-L1/PD-1-mediated immune evasion. In addition, we highlight the promising implications of targeting the N-linked glycosylation of PD-L1/PD-1 in the clinical diagnosis and treatment of cancer. Our review identifies knowledge gaps and sheds new light on the cancer research field.
Collapse
Affiliation(s)
- Zhiyun Duan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, P.R. China
| | - Runhan Shi
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, P.R. China
- Department of Ophthalmology and Vision Science, Shanghai Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, 200031, P.R. China
| | - Bo Gao
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, P.R. China
| | - Jiabin Cai
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Liver Cancer Institute, Fudan University, Shanghai, 200032, P.R. China.
- Department of Liver Surgery, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, 361015, P.R. China.
| |
Collapse
|
26
|
Tokumasu M, Nishida M, Zhao W, Chao R, Imano N, Yamashita N, Hida K, Naito H, Udono H. Metformin synergizes with PD-1 blockade to promote normalization of tumor vessels via CD8T cells and IFNγ. Proc Natl Acad Sci U S A 2024; 121:e2404778121. [PMID: 39018197 PMCID: PMC11287262 DOI: 10.1073/pnas.2404778121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/21/2024] [Indexed: 07/19/2024] Open
Abstract
Tumor blood vessels are highly leaky in structure and have poor blood perfusion, which hampers infiltration and function of CD8T cells within tumor. Normalizing tumor vessels is thus thought to be important in promoting the flux of immune T cells and enhancing ant-tumor immunity. However, how tumor vasculature is normalized is poorly understood. Metformin (Met) combined with ant-PD-1 therapy is known to stimulate proliferation of and to produce large amounts of IFNγ from tumor-infiltrating CD8T lymphocytes (CD8TILs). We found that the combination therapy promotes the pericyte coverage of tumor vascular endothelial cells (ECs) to improve blood perfusion and that it suppresses the hyperpermeability through the increase of VE-cadherin. Peripheral node addressin(PNAd) and vascular cell adhesion molecule (VCAM)-1, both implicated to promote tumor infiltration of CD8T cells, were also increased. Importantly, tumor vessel normalization, characterized as the reduced 70-kDa dextran leakage and the enhancement of VE-cadherin and VCAM-1, were canceled by anti-CD8 Ab or anti-IFNγ Ab injection to mice. The increased CD8TILs were also abrogated by anti-IFNγ Ab injection. In vascular ECs, flow cytometry analysis revealed that pSTAT1 expression was found to be associated with VE-cadherin expression. Moreover, in vitro treatment with Met and IFNγ enhanced VE-cadherin and VCAM-1 on human umbilical vein endothelial cells (HUVECs). The Kaplan-Meier method revealed a correlation of VE-cadherin or VCAM-1 levels with overall survival in patients treated with immune checkpoint inhibitors. These data indicate that IFNγ-mediated cross talk of CD8TILs with tumor vessels is important for creating a better tumor microenvironment and maintaining sustained antitumor immunity.
Collapse
Affiliation(s)
- Miho Tokumasu
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama700-8558, Japan
| | - Mikako Nishida
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama700-8558, Japan
| | - Weiyang Zhao
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama700-8558, Japan
| | - Ruoyu Chao
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama700-8558, Japan
| | - Natsumi Imano
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama700-8558, Japan
| | - Nahoko Yamashita
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama700-8558, Japan
| | - Kyoko Hida
- Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo060-8586, Japan
| | - Hisamichi Naito
- Department of Vascular Physiology, Kanazawa University Graduate School of Medical Sciences, Kanazawa920-8640, Ishikawa, Japan
| | - Heiichiro Udono
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama700-8558, Japan
| |
Collapse
|
27
|
Dou Y, Zheng J, Kang J, Wang L, Huang D, Liu Y, He C, Lin C, Lu C, Wu D, Han R, Li L, Tang L, He Y. Mesoporous manganese nanocarrier target delivery metformin for the co-activation STING pathway to overcome immunotherapy resistance. iScience 2024; 27:110150. [PMID: 39040065 PMCID: PMC11261061 DOI: 10.1016/j.isci.2024.110150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/24/2024] [Accepted: 05/28/2024] [Indexed: 07/24/2024] Open
Abstract
Targeting the stimulator of interferon genes (STING) pathway is a promising strategy to overcome primary resistance to immune checkpoint inhibitors in non-small cell lung cancer with the STK11 mutation. We previously found metformin enhances the STING pathway and thus promotes immune response. However, its low concentration in tumors limits its clinical use. Here, we constructed high-mesoporous Mn-based nanocarrier loading metformin nanoparticles (Mn-MSN@Met-M NPs) that actively target tumors and respond to release higher concentration of Mn2+ ions and metformin. The NPs significantly enhanced the T cells to kill lung cancer cells with the STK11 mutant. The mechanism shows that enhanced STING pathway activation promotes STING, TBKI, and IRF3 phosphorylation through Mn2+ ions and metformin release from NPs, thus boosting type I interferon production. In vivo, NPs in combination with a PD-1 inhibitor effectively decreased tumor growth. Collectively, we developed a Mn-MSN@Met-M nanoactivator to intensify immune activation for potential cancer immunotherapy.
Collapse
Affiliation(s)
- Yuanyao Dou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jie Zheng
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing 400042, China
- School of Medicine, Chongqing University, Chongqing 400044, China
| | - Jun Kang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Liping Wang
- Department of pain treatment, the seventh people’s Hospital of Chongqing, Chongqing 401320, China
| | - Daijuan Huang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing 400042, China
- School of Medicine, Chongqing University, Chongqing 400044, China
| | - Yihui Liu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Chao He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Caiyu Lin
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Conghua Lu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Di Wu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Rui Han
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Li Li
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yong He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing 400042, China
- School of Medicine, Chongqing University, Chongqing 400044, China
| |
Collapse
|
28
|
Pujalte‐Martin M, Belaïd A, Bost S, Kahi M, Peraldi P, Rouleau M, Mazure NM, Bost F. Targeting cancer and immune cell metabolism with the complex I inhibitors metformin and IACS-010759. Mol Oncol 2024; 18:1719-1738. [PMID: 38214418 PMCID: PMC11223609 DOI: 10.1002/1878-0261.13583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/15/2023] [Accepted: 12/29/2023] [Indexed: 01/13/2024] Open
Abstract
Metformin and IACS-010759 are two distinct antimetabolic agents. Metformin, an established antidiabetic drug, mildly inhibits mitochondrial complex I, while IACS-010759 is a new potent mitochondrial complex I inhibitor. Mitochondria is pivotal in the energy metabolism of cells by providing adenosine triphosphate through oxidative phosphorylation (OXPHOS). Hence, mitochondrial metabolism and OXPHOS become a vulnerability when targeted in cancer cells. Both drugs have promising antitumoral effects in diverse cancers, supported by preclinical in vitro and in vivo studies. We present evidence of their direct impact on cancer cells and their immunomodulatory effects. In clinical studies, while observational epidemiologic studies on metformin were encouraging, actual trial results were not as expected. However, IACS-01075 exhibited major adverse effects, thereby causing a metabolic shift to glycolysis and elevated lactic acid concentrations. Therefore, the future outlook for these two drugs depends on preventive clinical trials for metformin and investigations into the plausible toxic effects on normal cells for IACS-01075.
Collapse
Affiliation(s)
- Marc Pujalte‐Martin
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M)NiceFrance
- Equipe Labellisée Ligue Nationale Contre le Cancer
- Faculté de MédecineUniversité Côte d'AzurNiceFrance
| | - Amine Belaïd
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M)NiceFrance
- Equipe Labellisée Ligue Nationale Contre le Cancer
- Faculté de MédecineUniversité Côte d'AzurNiceFrance
| | - Simon Bost
- Equipe Labellisée Ligue Nationale Contre le Cancer
- Faculté de MédecineUniversité Côte d'AzurNiceFrance
| | - Michel Kahi
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M)NiceFrance
- Equipe Labellisée Ligue Nationale Contre le Cancer
- Faculté de MédecineUniversité Côte d'AzurNiceFrance
| | - Pascal Peraldi
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M)NiceFrance
- Equipe Labellisée Ligue Nationale Contre le Cancer
- Faculté de MédecineUniversité Côte d'AzurNiceFrance
| | - Matthieu Rouleau
- Equipe Labellisée Ligue Nationale Contre le Cancer
- Faculté de MédecineUniversité Côte d'AzurNiceFrance
- CNRS UMR7370, LP2MNiceFrance
| | - Nathalie M. Mazure
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M)NiceFrance
- Equipe Labellisée Ligue Nationale Contre le Cancer
- Faculté de MédecineUniversité Côte d'AzurNiceFrance
| | - Frédéric Bost
- Inserm U1065, Centre Méditerranéen de Médecine Moléculaire (C3M)NiceFrance
- Equipe Labellisée Ligue Nationale Contre le Cancer
- Faculté de MédecineUniversité Côte d'AzurNiceFrance
| |
Collapse
|
29
|
Liu J, Jiao X, Ma D, Fang Y, Gao Q. CAR-T therapy and targeted treatments: Emerging combination strategies in solid tumors. MED 2024; 5:530-549. [PMID: 38547867 DOI: 10.1016/j.medj.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/20/2023] [Accepted: 03/01/2024] [Indexed: 06/17/2024]
Abstract
CAR-T cell therapies hold great potential in achieving long-term remission in patients suffering from malignancies. However, their efficacy in treating solid tumors is impeded by challenges such as limited infiltration, compromised cancer recognition, decreased cytotoxicity, heightened exhaustion, absence of memory phenotypes, and inevitable toxicity. To surmount these obstacles, researchers are exploring innovative strategies, including the integration of CAR-T cells with targeted inhibitors. The combination of CAR-T therapies with specific targeted drugs has shown promise in enhancing CAR-T cell infiltration into tumor sites, boosting their tumor recognition capabilities, strengthening their cytotoxicity, alleviating exhaustion, promoting the development of a memory phenotype, and reducing toxicity. By harnessing the synergistic potential, a wider range of patients with solid tumors may potentially experience favorable outcomes. To summarize the current combined strategies of CAR-T therapies and targeted therapies, outline the potential mechanisms, and provide insights for future studies, we conducted this review by collecting existing experimental and clinical evidence.
Collapse
Affiliation(s)
- Jiahao Liu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofei Jiao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Ma
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Fang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qinglei Gao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
30
|
Chaudhary S, Kulkarni A. Metformin: Past, Present, and Future. Curr Diab Rep 2024; 24:119-130. [PMID: 38568468 DOI: 10.1007/s11892-024-01539-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 05/12/2024]
Abstract
PURPOSE OF REVIEW This review provides the most recent update of metformin, a biguanide oral antihyperglycemic drug used as a first-line treatment in type 2 diabetes mellitus. RECENT FINDINGS Metformin continues to dominate in the world of antidiabetics, and its use will continue to rise because of its high efficiency and easy availability. Apart from type 2 diabetes, research is exploring its potential in other conditions such as cancer, memory loss, bone disorders, immunological diseases, and aging. Metformin is the most prescribed oral antidiabetic worldwide. It has been in practical use for the last six decades and continues to be the preferred drug for newly diagnosed type 2 diabetes mellitus. It reduces glucose levels by decreasing hepatic glucose production, reducing intestinal glucose absorption, and increasing insulin sensitivity. It can be used as monotherapy or combined with other antidiabetics like sulfonylureas, DPP-4 inhibitors, SGLT-2 inhibitors, or insulin, improving its efficacy. Metformin can be used once or twice daily, depending on requirements. Prolonged usage of metformin may lead to abdominal discomfort, deficiency of Vitamin B12, or lactic acidosis. It should be used carefully in patients with renal impairment. Recent studies have explored additional benefits of metformin in polycystic ovarian disease, gestational diabetes mellitus, cognitive disorders, and immunological diseases. However, more extensive studies are needed to confirm these additional benefits.
Collapse
|
31
|
Li F, Ouyang J, Chen Z, Zhou Z, Milon Essola J, Ali B, Wu X, Zhu M, Guo W, Liang XJ. Nanomedicine for T-Cell Mediated Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2301770. [PMID: 36964936 DOI: 10.1002/adma.202301770] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/14/2023] [Indexed: 06/18/2023]
Abstract
T-cell immunotherapy offers outstanding advantages in the treatment of various diseases, and with the selection of appropriate targets, efficient disease treatment can be achieved. T-cell immunotherapy has made great progress, but clinical results show that only a small proportion of patients can benefit from T-cell immunotherapy. The extensive mechanistic work outlines a blueprint for using T cells as a new option for immunotherapy, but also presents new challenges, including the balance between different fractions of T cells, the inherent T-cell suppression patterns in the disease microenvironment, the acquired loss of targets, and the decline of T-cell viability. The diversity, flexibility, and intelligence of nanomedicines give them great potential for enhancing T-cell immunotherapy. Here, how T-cell immunotherapy strategies can be adapted with different nanomaterials to enhance therapeutic efficacy is discussed. For two different pathological states, immunosuppression and immune activation, recent advances in nanomedicines for T-cell immunotherapy in diseases such as cancers, rheumatoid arthritis, systemic lupus erythematosus, ulcerative colitis, and diabetes are summarized. With a focus on T-cell immunotherapy, this review highlights the outstanding advantages of nanomedicines in disease treatment, and helps advance one's understanding of the use of nanotechnology to enhance T-cell immunotherapy.
Collapse
Affiliation(s)
- Fangzhou Li
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, School of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
| | - Jiang Ouyang
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, School of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, P. R. China
| | - Zuqin Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
| | - Ziran Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Julien Milon Essola
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Barkat Ali
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, School of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, P. R. China
- Food Sciences Research Institute, Pakistan Agricultural Research Council, 44000, Islamabad, Pakistan
| | - Xinyue Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Mengliang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
| | - Weisheng Guo
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, School of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, P. R. China
| | - Xing-Jie Liang
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, School of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
32
|
Tone M, Iwahori K, Hirata M, Ueyama A, Tani A, Haruta JI, Takeda Y, Shintani Y, Kumanogoh A, Wada H. Tetracyclines enhance antitumor T-cell immunity via the Zap70 signaling pathway. J Immunother Cancer 2024; 12:e008334. [PMID: 38621815 PMCID: PMC11328671 DOI: 10.1136/jitc-2023-008334] [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] [Accepted: 03/18/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Cancer immunotherapy including immune checkpoint inhibitors is only effective for a limited population of patients with cancer. Therefore, the development of novel cancer immunotherapy is anticipated. In preliminary studies, we demonstrated that tetracyclines enhanced T-cell responses. Therefore, we herein investigated the efficacy of tetracyclines on antitumor T-cell responses by human peripheral T cells, murine models, and the lung tumor tissues of patients with non-small cell lung cancer (NSCLC), with a focus on signaling pathways in T cells. METHODS The cytotoxicity of peripheral and lung tumor-infiltrated human T cells against tumor cells was assessed by using bispecific T-cell engager (BiTE) technology (BiTE-assay system). The effects of tetracyclines on T cells in the peripheral blood of healthy donors and the tumor tissues of patients with NSCLC were examined using the BiTE-assay system in comparison with anti-programmed cell death-1 (PD-1) antibody, nivolumab. T-cell signaling molecules were analyzed by flow cytometry, ELISA, and qRT-PCR. To investigate the in vivo antitumor effects of tetracyclines, tetracyclines were administered orally to BALB/c mice engrafted with murine tumor cell lines, either in the presence or absence of anti-mouse CD8 inhibitors. RESULTS The results obtained revealed that tetracyclines enhanced antitumor T-cell cytotoxicity with the upregulation of granzyme B and increased secretion of interferon-γ in human peripheral T cells and the lung tumor tissues of patients with NSCLC. The analysis of T-cell signaling showed that CD69 in both CD4+ and CD8+ T cells was upregulated by minocycline. Downstream of T-cell receptor signaling, Zap70 phosphorylation and Nur77 were also upregulated by minocycline in the early phase after T-cell activation. These changes were not observed in T cells treated with anti-PD-1 antibodies under the same conditions. The administration of tetracyclines exhibited antitumor efficacy with the upregulation of CD69 and increases in tumor antigen-specific T cells in murine tumor models. These changes were canceled by the administration of anti-mouse CD8 inhibitors. CONCLUSIONS In conclusion, tetracyclines enhanced antitumor T-cell immunity via Zap70 signaling. These results will contribute to the development of novel cancer immunotherapy.
Collapse
Affiliation(s)
- Mari Tone
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kota Iwahori
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Michinari Hirata
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Biopharmaceutical Research Division, Shionogi & Co., Ltd, Osaka, Japan
| | - Azumi Ueyama
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Biopharmaceutical Research Division, Shionogi & Co., Ltd, Osaka, Japan
| | - Akiyoshi Tani
- Compound Library Screening Center, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Jun-Ichi Haruta
- Lead Explorating Units, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yasushi Shintani
- Department of General Thoracic Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Osaka, Japan
- Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Osaka, Japan
| | - Hisashi Wada
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
33
|
Turpin R, Liu R, Munne PM, Peura A, Rannikko JH, Philips G, Boeckx B, Salmelin N, Hurskainen E, Suleymanova I, Aung J, Vuorinen EM, Lehtinen L, Mutka M, Kovanen PE, Niinikoski L, Meretoja TJ, Mattson J, Mustjoki S, Saavalainen P, Goga A, Lambrechts D, Pouwels J, Hollmén M, Klefström J. Respiratory complex I regulates dendritic cell maturation in explant model of human tumor immune microenvironment. J Immunother Cancer 2024; 12:e008053. [PMID: 38604809 PMCID: PMC11015234 DOI: 10.1136/jitc-2023-008053] [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] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Combining cytotoxic chemotherapy or novel anticancer drugs with T-cell modulators holds great promise in treating advanced cancers. However, the response varies depending on the tumor immune microenvironment (TIME). Therefore, there is a clear need for pharmacologically tractable models of the TIME to dissect its influence on mono- and combination treatment response at the individual level. METHODS Here we establish a patient-derived explant culture (PDEC) model of breast cancer, which retains the immune contexture of the primary tumor, recapitulating cytokine profiles and CD8+T cell cytotoxic activity. RESULTS We explored the immunomodulatory action of a synthetic lethal BCL2 inhibitor venetoclax+metformin drug combination ex vivo, discovering metformin cannot overcome the lymphocyte-depleting action of venetoclax. Instead, metformin promotes dendritic cell maturation through inhibition of mitochondrial complex I, increasing their capacity to co-stimulate CD4+T cells and thus facilitating antitumor immunity. CONCLUSIONS Our results establish PDECs as a feasible model to identify immunomodulatory functions of anticancer drugs in the context of patient-specific TIME.
Collapse
Affiliation(s)
- Rita Turpin
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - Ruixian Liu
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - Pauliina M Munne
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - Aino Peura
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | | | | | - Bram Boeckx
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Natasha Salmelin
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - Elina Hurskainen
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - Ilida Suleymanova
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - July Aung
- University of Helsinki Faculty of Medicine, Helsinki, Finland
| | | | | | - Minna Mutka
- Department of Pathology, Helsinki University Central Hospital, Helsinki, Finland
| | - Panu E Kovanen
- Department of Pathology, HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - Laura Niinikoski
- Breast Surgery Unit, Helsinki University Central Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Tuomo J Meretoja
- Breast Surgery Unit, Helsinki University Central Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Johanna Mattson
- Department of oncology, Helsinki University Central Hospital, Helsinki, Finland
| | - Satu Mustjoki
- TRIMM, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- University of Helsinki Helsinki Institute of Life Sciences, Helsinki, Finland
| | | | - Andrei Goga
- Department of Cell & Tissue Biology, UCSF, San Francisco, California, USA
| | | | - Jeroen Pouwels
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | | | - Juha Klefström
- Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
- Finnish Cancer Institute, Helsinki, Finland
| |
Collapse
|
34
|
Pan X, Wang J, Zhang L, Li G, Huang B. Metabolic plasticity of T cell fate decision. Chin Med J (Engl) 2024; 137:762-775. [PMID: 38086394 PMCID: PMC10997312 DOI: 10.1097/cm9.0000000000002989] [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: 08/08/2023] [Indexed: 04/06/2024] Open
Abstract
ABSTRACT The efficacy of adaptive immune responses in cancer treatment relies heavily on the state of the T cells. Upon antigen exposure, T cells undergo metabolic reprogramming, leading to the development of functional effectors or memory populations. However, within the tumor microenvironment (TME), metabolic stress impairs CD8 + T cell anti-tumor immunity, resulting in exhausted differentiation. Recent studies suggested that targeting T cell metabolism could offer promising therapeutic opportunities to enhance T cell immunotherapy. In this review, we provide a comprehensive summary of the intrinsic and extrinsic factors necessary for metabolic reprogramming during the development of effector and memory T cells in response to acute and chronic inflammatory conditions. Furthermore, we delved into the different metabolic switches that occur during T cell exhaustion, exploring how prolonged metabolic stress within the TME triggers alterations in cellular metabolism and the epigenetic landscape that contribute to T cell exhaustion, ultimately leading to a persistently exhausted state. Understanding the intricate relationship between T cell metabolism and cancer immunotherapy can lead to the development of novel approaches to improve the efficacy of T cell-based treatments against cancer.
Collapse
Affiliation(s)
- Xiaoli Pan
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China
| | - Jiajia Wang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China
| | - Lianjun Zhang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China
| | - Guideng Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China
- Key Laboratory of Synthetic Biology Regulatory Element, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China
| | - Bo Huang
- Department of Immunology & National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 100005, China
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
35
|
Shao W, Yao Y, Yang L, Li X, Ge T, Zheng Y, Zhu Q, Ge S, Gu X, Jia R, Song X, Zhuang A. Novel insights into TCR-T cell therapy in solid neoplasms: optimizing adoptive immunotherapy. Exp Hematol Oncol 2024; 13:37. [PMID: 38570883 PMCID: PMC10988985 DOI: 10.1186/s40164-024-00504-8] [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: 12/08/2023] [Accepted: 03/21/2024] [Indexed: 04/05/2024] Open
Abstract
Adoptive immunotherapy in the T cell landscape exhibits efficacy in cancer treatment. Over the past few decades, genetically modified T cells, particularly chimeric antigen receptor T cells, have enabled remarkable strides in the treatment of hematological malignancies. Besides, extensive exploration of multiple antigens for the treatment of solid tumors has led to clinical interest in the potential of T cells expressing the engineered T cell receptor (TCR). TCR-T cells possess the capacity to recognize intracellular antigen families and maintain the intrinsic properties of TCRs in terms of affinity to target epitopes and signal transduction. Recent research has provided critical insight into their capability and therapeutic targets for multiple refractory solid tumors, but also exposes some challenges for durable efficacy. In this review, we describe the screening and identification of available tumor antigens, and the acquisition and optimization of TCRs for TCR-T cell therapy. Furthermore, we summarize the complete flow from laboratory to clinical applications of TCR-T cells. Last, we emerge future prospects for improving therapeutic efficacy in cancer world with combination therapies or TCR-T derived products. In conclusion, this review depicts our current understanding of TCR-T cell therapy in solid neoplasms, and provides new perspectives for expanding its clinical applications and improving therapeutic efficacy.
Collapse
Affiliation(s)
- Weihuan Shao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Ludi Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Xiaoran Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Tongxin Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Yue Zheng
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Qiuyi Zhu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Xiang Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| | - Xin Song
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| | - Ai Zhuang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
36
|
Shiba S, Harao M, Saito A, Sakuragi M, Kitayama J, Sata N. Metformin Alters Tumor Immune Microenvironment, Improving the Outcomes of Breast Cancer Patients With Type 2 Diabetes Mellitus. J Breast Cancer 2024; 27:121-129. [PMID: 38529589 PMCID: PMC11065500 DOI: 10.4048/jbc.2023.0285] [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: 12/15/2023] [Revised: 01/25/2024] [Accepted: 02/06/2024] [Indexed: 03/27/2024] Open
Abstract
This study investigated the clinical effect of metformin on breast cancer patients with preexisting type 2 diabetes mellitus (T2DM). We analyzed 177 patients with T2DM who underwent breast cancer surgery and assessed tumor-associated macrophages (TAMs) and tumor-infiltrating lymphocytes (TILs) in patients who underwent tumor resection with or without metformin treatment using multiplex immunohistochemistry (IHC). Patients who received metformin either pre- or postoperatively exhibited reduced distant organ recurrence and improved postoperative recurrence-free survival compared to those of patients who did not. Additionally, in a subgroup of 40 patients receiving preoperative systemic therapy, metformin treatment was associated with increased rates of pathological complete response. IHC analysis revealed significantly lower levels of cluster of differentiation (CD) 68(+) CD163(+) M2-type TAMs (p < 0.01) but higher CD3(+) and CD8(+) TIL densities in the metformin-treated group compared with the same parameters in those without metformin treatment, with a significant difference in the CD8(+)/CD3(+) TIL ratio (p < 0.01). Despite the constraints posed by our small sample size, our findings suggest a potential role for metformin in modulating the immunological microenvironment, which may contribute to improved outcomes in diabetes patients with breast cancer.
Collapse
Affiliation(s)
- Satomi Shiba
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
- Department of Breast Oncology, Jichi Medical University Hospital, Shimotsuke, Japan
| | - Michiko Harao
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
- Department of Breast Oncology, Jichi Medical University Hospital, Shimotsuke, Japan
| | - Akira Saito
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Masako Sakuragi
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
- Department of Breast Oncology, Jichi Medical University Hospital, Shimotsuke, Japan
| | - Joji Kitayama
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
- Department of Breast Oncology, Jichi Medical University Hospital, Shimotsuke, Japan.
| | - Naohiro Sata
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
| |
Collapse
|
37
|
Tu SM, Trikannad AK, Vellanki S, Hussain M, Malik N, Singh SR, Jillella A, Obulareddy S, Malapati S, Bhatti SA, Arnaoutakis K, Atiq OT. Stem Cell Origin of Cancer: Clinical Implications beyond Immunotherapy for Drug versus Therapy Development in Cancer Care. Cancers (Basel) 2024; 16:1151. [PMID: 38539487 PMCID: PMC10969562 DOI: 10.3390/cancers16061151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 11/11/2024] Open
Abstract
Although immunotherapy has revolutionized cancer care, there is still an urgent need to enhance its efficacy and ensure its safety. A correct cancer theory and proper scientific method empower pertinent cancer research and enable effective and efficient drug versus therapy development for patient care. In this perspective, we revisit the concept of immune privilege in a cancer cell versus normal cell, as well as in a cancer stem cell versus normal stem cell. We re-examine whether effective immunotherapies are efficacious due to their anti-cancer and/or immune modulatory mechanisms. We reassess why checkpoint inhibitors (CPIs) are not equal. We reconsider whether one can attribute the utility of immunotherapy to specific cancer subtypes and its futility to certain tumor/immune compartments, components, and microenvironments. We propose ways and means to advance immunotherapy beyond CPIs by combining anti-PD1/L1 with various other treatment modalities according to an appropriate scientific theory, e.g., stem cell origin of cancer, and based on available clinical evidence, e.g., randomized clinical trials. We predict that a stem cell theory of cancer will facilitate the design of better and safer immunotherapy with improved selection of its use for the right patient with the right cancer type at the right time to optimize clinical benefits and minimize potential toxic effects and complications.
Collapse
Affiliation(s)
- Shi-Ming Tu
- Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA (M.H.); (N.M.); (S.R.S.); (A.J.); (S.O.); (S.M.); (S.A.B.); (K.A.); (O.T.A.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wang Y, Tang Q, Wu R, Yang S, Geng Z, He P, Li X, Chen Q, Liang X. Metformin-Mediated Fast Charge-Reversal Nanohybrid for Deep Penetration Piezocatalysis-Augmented Chemodynamic Immunotherapy of Cancer. ACS NANO 2024; 18:6314-6332. [PMID: 38345595 DOI: 10.1021/acsnano.3c11174] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Immune checkpoint blockade (ICB) therapy still suffers from insufficient immune response and adverse effect of ICB antibodies. Chemodynamic therapy (CDT) has been demonstrated to be an effective way to synergize with ICB therapy. However, a low generation rate of reactive oxygen species and poor tumor penetration of CDT platforms still decline the immune effects. Herein, a charge-reversal nanohybrid Met@BF containing both Fe3O4 and BaTiO3 nanoparticles in the core and Metformin (Met) on the surface was fabricated for tumor microenvironment (TME)- and ultrasound (US)-activated piezocatalysis-chemodynamic immunotherapy of cancer. Interestingly, Met@BF had a negative charge in blood circulation, which was rapidly changed into positive when exposed to acidic TME attributed to quaternization of tertiary amine in Met, facilitating deep tumor penetration. Subsequently, with US irradiation, Met@BF produced H2O2 based on piezocatalysis of BaTiO3, which greatly enhanced the Fenton reaction of Fe3O4, thus boosting robust antitumor immune response. Furthermore, PD-L1 expression was inhibited by the local released Met to further augment the antitumor immune effect, achieving effective inhibitions for both primary and metastatic tumors. Such a combination of piezocatalysis-enhanced chemodynamic therapy and Met-mediated deep tumor penetration and downregulation of PD-L1 provides a promising strategy to augment cancer immunotherapy.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Qingshuang Tang
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Ruiqi Wu
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Shiyuan Yang
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Zhishuai Geng
- National Engineering Research Center of Flame Retardant Materials, School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Ping He
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Xiaoda Li
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
39
|
Guo Q, Wang L, Wuriqimuge, Dong L, Feng M, Bao X, Zhang K, Cai Z, Qu X, Zhang S, Wu J, Wu H, Wang C, Yu X, Kong W, Zhang H. Metformin improved a heterologous prime-boost of dual-targeting cancer vaccines to inhibit tumor growth in a melanoma mouse model. Int Immunopharmacol 2024; 128:111431. [PMID: 38244520 DOI: 10.1016/j.intimp.2023.111431] [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: 09/30/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/22/2024]
Abstract
Therapeutic cancer vaccines, which induce anti-tumor immunity by targeting specific antigens, constitute a promising approach to cancer therapy. Our previous work proposed an optimized heterologous immunization strategy using cancer gene vaccines co-targeting MUC1 and survivin. Administration of a DNA vaccine three times within a week followed by a single recombinant MVA (rMVA) boost was able to efficiently induce anti-tumor immunity and inhibit tumor growth in tumor-bearing mouse models However, the complex immunosuppressive tumor microenvironment always limits infiltration by vaccine-induced T cells. Modifying the immunosuppressive microenvironment of tumors would be a breakthrough in enhancing the therapeutic effects of a cancer vaccine. Recent studies have reported that metformin, a type 2 diabetes drug, may ameliorate the tumor microenvironment, thereby enhancing anti-tumor immunity. Here, we tested whether the combinational therapeutic strategy of cancer vaccines administered with a heterologous prime-boost strategy with metformin enhanced anti-tumor effects in a melanoma mouse model. The results showed that metformin promoted the transition of M2-tumor-associated macrophages (M2-TAM) to M1-TAM, induced more tumor-infiltrating proliferative CD4 and CD8 T cells, and decreased exhausted T cells. This combinational treatment induced anti-tumor immunity from cancer vaccines, ameliorating the tumor microenvironment, showing improved tumor inhibition, and prolonging survival in tumor-bearing mice compared with either a cancer vaccine or metformin alone.
Collapse
Affiliation(s)
- Qianqian Guo
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Lizheng Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wuriqimuge
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Ling Dong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Mengfan Feng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xin Bao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Ke Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Zongyu Cai
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xueli Qu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Shiqi Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jiaxin Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Hui Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Chu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Haihong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
40
|
Li Y, Liu B, Cao Y, Cai L, Zhou Y, Yang W, Sun T. Metformin-induced reduction of CCR8 enhances the anti-tumor immune response of PD-1 immunotherapy in glioblastoma. Eur J Pharmacol 2024; 964:176274. [PMID: 38142852 DOI: 10.1016/j.ejphar.2023.176274] [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: 09/08/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/26/2023]
Abstract
Immunotherapy strategies targeting the programmed cell death 1 (PD-1) in clinical treatments have shown limited success in controlling glioblastoma malignancies. Metformin exserts antitumor function, yet the underlying mechanisms remain unclear. Here, we investigated whether metformin could enhance the effectiveness of anti-PD-1 therapy by activating the immune system. Whether combination of an anti-PD-1 antibody or not, metformin significantly increased tumor-infiltrating CD4+ T cells while decreased regulatory T (Treg) cells in a mouse GBM model. Additionally, metformin reduced CC motif chemokine receptor CCR8 and elevated Interleukin 17 A (IL-17 A) expressions. Mechanistically, metformin reduces histone acetylation at the CCR8 promotor and inhibits CCR8 expression by upregulating AMP-activated protein kinase (AMPK)-activated sirtuin 2 (SIRT2). Metformin enhances the effectiveness of anti-PD-1 immunotherapy by reducing CCR8 expression on tumor-infiltrating Treg cells, suggesting that metformin has an antitumor effect by alleviating immunosuppression and promoting T cell-mediated immune response.
Collapse
Affiliation(s)
- Yanyan Li
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bin Liu
- Department of Neurosurgery, Qinghai Provincial People's Hospital, Xining, Qinghai, China
| | - Yufei Cao
- Department of Critical Care Medicine, Affiliated First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lize Cai
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Youxin Zhou
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wei Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, China.
| | - Ting Sun
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
41
|
Cheng H, Zheng Y. Advances in macrophage and T cell metabolic reprogramming and immunotherapy in the tumor microenvironment. PeerJ 2024; 12:e16825. [PMID: 38239299 PMCID: PMC10795528 DOI: 10.7717/peerj.16825] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/02/2024] [Indexed: 01/22/2024] Open
Abstract
Macrophages and T cells in the tumor microenvironment (TME) play an important role in tumorigenesis and progression. However, TME is also characterized by metabolic reprogramming, which may affect macrophage and metabolic activity of T cells and promote tumor escape. Immunotherapy is an approach to fight tumors by stimulating the immune system in the host, but requires support and modulation of cellular metabolism. In this process, the metabolic roles of macrophages and T cells become increasingly important, and their metabolic status and interactions play a critical role in the success of immunotherapy. Therefore, understanding the metabolic state of T cells and macrophages in the TME and the impact of metabolic reprogramming on tumor therapy will help optimize subsequent immunotherapy strategies.
Collapse
Affiliation(s)
- Hua Cheng
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yongbin Zheng
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
42
|
Zaki HM, Ali KM, Abd Allah MYY, Abouelnaga AM, Abdraboh ME, Hussein O. Metronomic cyclophosphamide and metformin inhibited tumor growth and repopulated tumor-infiltrating lymphocytes in an experimental carcinoma model. BMC Res Notes 2024; 17:4. [PMID: 38167322 PMCID: PMC10759693 DOI: 10.1186/s13104-023-06651-1] [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: 04/09/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Metformin is a widely used antidiabetic biguanide. Retrospective data demonstrated the association of metformin use with survival benefit in multiple tumor types. Interest in repurposing metformin to treat cancer has not been translated into encouraging clinical benefit. In animal models, metformin activated cytotoxic T cells and exerted an immune-mediated anticancer effect. The current research was conducted to investigate the possible therapeutic benefit of metformin in combination with metronomic cyclophosphamide in an experimental cancer model. Ehrlich ascites carcinoma was injected into the subcutaneous tissue to induce solid tumors in syngeneic mice. Exponential solid tumor growth ensued and was effectively arrested with the administration of a cytotoxic dose of parenteral cyclophosphamide. Alternatively, oral metformin and continuous, low-dose cyclophosphamide significantly inhibited tumor growth relative to untreated mice. The drug combination was well tolerated. Histopathological examination of the tumor showed an increased number of tumor-infiltrating lymphocytes and enhanced expression of granzyme B by this drug combination. The current data suggests a potential role of metformin and metronomic chemotherapy that warrants further investigation.
Collapse
Affiliation(s)
- Heba Mohamed Zaki
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Khadiga Mohamed Ali
- Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | | | | | | | - Osama Hussein
- Surgical Oncology Department, Faculty of Medicine, Mansoura University Oncology Center, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
43
|
Jain H, Marsool MDM, Goyal A, Sulaiman SA, Fatima L, Idrees M, Sharma B, Borra V, Gupta P, Nadeem A, Jain J, Ali H, Sohail AH. Unveiling the relationship between gut microbiota and heart failure: Recent understandings and insights. Curr Probl Cardiol 2024; 49:102179. [PMID: 37923029 DOI: 10.1016/j.cpcardiol.2023.102179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 11/07/2023]
Abstract
Gut microbiota, which comprises a broad range of bacteria inhabiting the human intestines, plays a crucial role in establishing a mutually beneficial relationship with the host body. Dysbiosis refers to the perturbations in the composition or functioning of the microbial community, which can result in a shift from a balanced microbiota to an impaired state. This alteration has the potential to contribute to the development of chronic systemic inflammation. Heart failure (HF) is a largely prevalent clinical condition that has been demonstrated to have variations in the gut microbiome, indicating a potential active involvement in the pathogenesis and advancement of the disease. The exploration of the complex interplay between the gut microbiome and HF presents a potential avenue for the discovery of innovative biomarkers, preventive measures, and therapeutic targets. This review aims to investigate the impact of gut bacteria on HF.
Collapse
Affiliation(s)
- Hritvik Jain
- Department of Internal Medicine, All India Institute of Medical Sciences (AIIMS), Jodhpur, India.
| | | | - Aman Goyal
- Department of Internal Medicine, Seth GS Medical College and KEM Hospital, Mumbai, India
| | | | | | | | - Bhavya Sharma
- Department of Internal Medicine, Baroda Medical College and SSG Hospital, Vadodara, India
| | - Vamsikalyan Borra
- Department of Internal Medicine, University of Texas Rio Grande Valley, TX, United States
| | - Prakash Gupta
- Virgen Milagrosa University Foundation College of Medicine, San Carlos City, Philippines
| | - Abdullah Nadeem
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Jyoti Jain
- Department of Internal Medicine, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Hassam Ali
- Department of Gastroenterology, East Carolina University, North Carolina, United States
| | - Amir H Sohail
- Department of Surgery, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
44
|
Saito A, Koinuma K, Kawashima R, Miyato H, Ohzawa H, Horie H, Yamaguchi H, Kawahira H, Mimura T, Kitayama J, Sata N. Metformin may improve the outcome of patients with colorectal cancer and type 2 diabetes mellitus partly through effects on neutrophil extracellular traps. BJC REPORTS 2023; 1:20. [PMID: 39516686 PMCID: PMC11524073 DOI: 10.1038/s44276-023-00022-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/16/2024]
Abstract
BACKGROUND Although metformin reduces the risk of cancer-related mortality in patents with type 2 diabetes, the mechanism of its anti-cancer effects has not been fully understood. METHOD Impact of metformin on survival was examined in patients who underwent curative colectomy for colorectal cancer (CRC). The effects of metformin in neutrophil extracellular traps (NETs) were examined with in-vitro experiments and multiplex immunohistochemistry of surgically resected CRC specimens. RESULTS Prior intake of metformin prolonged relapse-free (P = 0.036) and overall survival (P = 0.041) in 289 patients with T2DM to the comparable levels to those of 1576 non-diabetic patients. Metformin reduced the production of NETs stimulated with lipopolysaccharide or HT-29 colon cancer cells to 60% of control. Neutrophils markedly suppressed the chemotactic migration of activated T cells in an NET-dependent manner, which was reversed by metformin treatment up to approximately half of the migration without neutrophils. Immunohistochemical analysis revealed a significant association between metformin intake and a reduction in the numbers of tumor-associated neutrophils (TANs) and NETs. Simultaneously, metformin intake was found to increase the presence of CD3(+) and CD8(+) tumor-infiltrating T cells (TILs), particularly at the tumor-invasion front, especially in areas with fewer TANs and NETs. CONCLUSION Metformin suppresses the diabetes-associated enhancement of NET formation, which can augment the infiltration of TILs in CRC tissues. The anti-tumor effect of metformin in patients with T2DM may be, at least partly, attributable to the inhibition of NETs.
Collapse
Affiliation(s)
- Akira Saito
- Department of Gastrointestinal Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Koji Koinuma
- Department of Gastrointestinal Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Rie Kawashima
- Department of Oral and Maxillofacial Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Hideyo Miyato
- Department of Gastrointestinal Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Hideyuki Ohzawa
- Department of Clinical Oncology, Jichi Medical University, Shimotsuke, Japan
| | - Hisanaga Horie
- Department of Gastrointestinal Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Hironori Yamaguchi
- Department of Clinical Oncology, Jichi Medical University, Shimotsuke, Japan
| | - Hiroshi Kawahira
- Department of Gastrointestinal Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Toshiki Mimura
- Department of Gastrointestinal Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Joji Kitayama
- Department of Gastrointestinal Surgery, Jichi Medical University, Shimotsuke, Japan.
| | - Naohiro Sata
- Department of Gastrointestinal Surgery, Jichi Medical University, Shimotsuke, Japan
| |
Collapse
|
45
|
Noch EK, Palma LN, Yim I, Bullen N, Qiu Y, Ravichandran H, Kim J, Rendeiro A, Davis MB, Elemento O, Pisapia DJ, Zhai K, LeKaye HC, Koutcher JA, Wen PY, Ligon KL, Cantley LC. Insulin feedback is a targetable resistance mechanism of PI3K inhibition in glioblastoma. Neuro Oncol 2023; 25:2165-2176. [PMID: 37399061 PMCID: PMC10708938 DOI: 10.1093/neuonc/noad117] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Indexed: 07/04/2023] Open
Abstract
BACKGROUND Insulin feedback is a critical mechanism responsible for the poor clinical efficacy of phosphatidylinositol 3-kinase (PI3K) inhibition in cancer, and hyperglycemia is an independent factor associated with poor prognosis in glioblastoma (GBM). We investigated combination anti-hyperglycemic therapy in a mouse model of GBM and evaluated the association of glycemic control in clinical trial data from patients with GBM. METHODS The effect of the anti-hyperglycemic regimens, metformin and the ketogenic diet, was evaluated in combination with PI3K inhibition in patient-derived GBM cells and in an orthotopic GBM mouse model. Insulin feedback and the immune microenvironment were retrospectively evaluated in blood and tumor tissue from a Phase 2 clinical trial of buparlisib in patients with recurrent GBM. RESULTS We found that PI3K inhibition induces hyperglycemia and hyperinsulinemia in mice and that combining metformin with PI3K inhibition improves the treatment efficacy in an orthotopic GBM xenograft model. Through examination of clinical trial data, we found that hyperglycemia was an independent factor associated with poor progression-free survival in patients with GBM. We also found that PI3K inhibition increased insulin receptor activation and T-cell and microglia abundance in tumor tissue from these patients. CONCLUSION Reducing insulin feedback improves the efficacy of PI3K inhibition in GBM in mice, and hyperglycemia worsens progression-free survival in patients with GBM treated with PI3K inhibition. These findings indicate that hyperglycemia is a critical resistance mechanism associated with PI3K inhibition in GBM and that anti-hyperglycemic therapy may enhance PI3K inhibitor efficacy in GBM patients.
Collapse
Affiliation(s)
- Evan K Noch
- Division of Neuro-oncology, Department of Neurology, Weill Cornell Medicine, New York, New York, USA
- Sandra and Edward Meyer Cancer Center, Weill Department of Medicine, New York, New York, USA
| | - Laura N Palma
- Sandra and Edward Meyer Cancer Center, Weill Department of Medicine, New York, New York, USA
| | - Isaiah Yim
- Sandra and Edward Meyer Cancer Center, Weill Department of Medicine, New York, New York, USA
| | - Nayah Bullen
- Sandra and Edward Meyer Cancer Center, Weill Department of Medicine, New York, New York, USA
| | - Yuqing Qiu
- Department of Population Health Sciences, Division of Biostatistics and Epidemiology, Weill Cornell Medicine, New York, New York, USA
| | - Hiranmayi Ravichandran
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Junbum Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Andre Rendeiro
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Medical University of Vienna, Vienna, Austria
| | - Melissa B Davis
- Department of Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, USA
| | - David J Pisapia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Kevin Zhai
- Division of Neuro-oncology, Department of Neurology, Weill Cornell Medicine, New York, New York, USA
| | - Hongbiao Carl LeKaye
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jason A Koutcher
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Patrick Y Wen
- Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Center for Neuro-oncology, Boston, Massachusetts, USA
| | - Keith L Ligon
- Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Pathology, Boston, Massachusetts, USA
| | - Lewis C Cantley
- Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
46
|
Repas J, Peternel L, Sourij H, Pavlin M. Low glucose availability potentiates the effects of metformin on model T cell activation and exhaustion markers in vitro. Front Endocrinol (Lausanne) 2023; 14:1216193. [PMID: 38116319 PMCID: PMC10728603 DOI: 10.3389/fendo.2023.1216193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/07/2023] [Indexed: 12/21/2023] Open
Abstract
Modulation of immune cell metabolism is one of promising strategies to improve cancer immunotherapies. Metformin is an anti-diabetic drug with potential anti-cancer effects, ranging from normalization of blood glucose and insulin levels, direct anti-proliferative effects on cancer cells to emerging immunomodulatory effects on anti-tumor immunity. Metformin can reduce tumor hypoxia and PD-L1 expression, as well as normalize or improve T cell function and potentiate the effect of immune checkpoint inhibitors, making it a promising adjuvant to immunotherapy of tumors with poor response such as triple negative breast cancer (TNBC). However, although the effects of metformin on cancer cells are glucose-dependent, the role of glucose in modulating its effect on T cells has not been systematically studied. We thus investigated the effect of metformin as a function of glucose level on Jurkat cell and PBMC T cell models in vitro. While low metformin concentrations had little effect on T cell function, high concentration reduced proliferation and IFN-γ secretion in both models and induced a shift in T cell populations from memory to effector subsets. The PD-1/CD69 ratio was improved by high metformin in T cells from PBMC. Low glucose and metformin synergistically reduced PD-1 and CD69 expression and IFN-γ secretion in T cells from PBMC. Low glucose level itself suppressed Jurkat cell function due to their limited metabolic plasticity, but had limited effects on T cells from PBMC apart from reduced proliferation. Conversely, high glucose did not strongly affect either T cell model. Metformin in combination with glycolysis inhibitor 2-deoxy-D-glucose (2DG) reduced PD-1 in Jurkat cells, but also strongly suppressed their function. However, low, physiologically achievable 2DG concentration itself reduced PD-1 while mostly maintaining IL-2 secretion and, interestingly, even strongly increased IFN-γ secretion regardless of glucose level. Overall, glucose metabolism can importantly influence some of the effects of metformin on T cell functionality in the tumor microenvironment. Additionally, we show that 2DG could potentially improve the anti-tumor T cell response.
Collapse
Affiliation(s)
- Jernej Repas
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Lea Peternel
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Harald Sourij
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetology, Medical University Graz, Graz, Austria
| | - Mojca Pavlin
- Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Group for Nano- and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
47
|
He W, Wang X, Chen M, Li C, Chen W, Pan L, Cui Y, Yu Z, Wu G, Yang Y, Xu M, Dong Z, Ma K, Wang J, He Z. Metformin reduces hepatocarcinogenesis by inducing downregulation of Cyp26a1 and CD8 + T cells. Clin Transl Med 2023; 13:e1465. [PMID: 37997519 PMCID: PMC10668005 DOI: 10.1002/ctm2.1465] [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: 04/14/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a highly heterogeneous cancer with major challenges in both prevention and therapy. Metformin, adenosine monophosphate-activated protein kinase (AMPK) activator, has been suggested to reduce the incidence of HCC when used for patients with diabetes in preclinical and clinical studies. However, the possible effects of metformin and their mechanisms of action in non-diabetic HCC have not been adequately investigated. METHODS Fah-/- mice were used to construct a liver-injury-induced non-diabetic HCC model for exploring hepatocarcinogenesis and therapeutic potential of metformin. Changes in relevant tumour and biochemical indicators were measured. Bulk and single-cell RNA-sequencing analyses were performed to validate the crucial role of proinflammatory/pro-tumour CD8+ T cells. In vitro and in vivo experiments were performed to confirm Cyp26a1-related antitumour mechanisms of metformin. RESULTS RNA-sequencing analysis showed that chronic liver injury led to significant changes in AMPK-, glucose- and retinol metabolism-related pathways in Fah-/- mice. Metformin prevented the formation of non-diabetic HCC in Fah-/- mice with chronic liver injury. Cyp26a1 ddexpression in hepatocytes was significantly suppressed after metformin treatment. Moreover, downregulation of Cyp26a1 occurred in conjunction with increased levels of all-trans-retinoic acid (atRA), which is involved in the activation of metformin-suppressed hepatocarcinogenesis in Fah-/- mice. In contrast, both CD8+ T-cell infiltration and proinflammatory/pro-tumour cytokines in the liver were significantly upregulated in Fah-/- mice during chronic liver injury, which was notably reversed by either metformin or atRA treatment. Regarding mechanisms, metformin regulated the decrease in Cyp26a1 enzyme expression and increased atRA expression via the AMPK/STAT3/Gadd45β/JNK/c-Jun pathway. CONCLUSIONS Metformin inhibits non-diabetic HCC by upregulating atRA levels and downregulating CD8+ T cells. This is the first reporting that the traditional drug metformin regulates the metabolite atRA via the Cyp26a1-involved pathway. The present study provides a potential application of metformin and atRA in non-diabetic HCC.
Collapse
Affiliation(s)
- Weizhi He
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
- Fudan University Shanghai Cancer Center, International Co‐Laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Shanghai Medical College of Fudan University, Institutes of Biomedical SciencesShanghai Key Laboratory of Medical EpigeneticsShanghaiChina
| | - Xicheng Wang
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Miaomiao Chen
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Chong Li
- Zhoupu Community Health Service Center of Pudong New AreaShanghaiChina
| | - Wenjian Chen
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Lili Pan
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Yangyang Cui
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Postgraduate Training Base of Shanghai East HospitalJinzhou Medical UniversityJinzhouLiaoningChina
| | - Zhao Yu
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Guoxiu Wu
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Yang Yang
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Mingyang Xu
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Zhaoxuan Dong
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Keming Ma
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Jinghan Wang
- Department of Hepatobiliary and Pancreatic SurgeryShanghai East Hospital, Tongji UniversityShanghaiChina
| | - Zhiying He
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| |
Collapse
|
48
|
Zhu H, Jia Z, Li YR, Danelisen I. Molecular mechanisms of action of metformin: latest advances and therapeutic implications. Clin Exp Med 2023; 23:2941-2951. [PMID: 37016064 PMCID: PMC10072049 DOI: 10.1007/s10238-023-01051-y] [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: 02/26/2023] [Accepted: 03/17/2023] [Indexed: 04/06/2023]
Abstract
Metformin is among the most widely used antidiabetic drugs. Studies over the past few years have identified multiple novel molecular targets and pathways that metformin acts on to exert its beneficial effects in treating type 2 diabetes as well as other disorders involving dysregulated inflammation and redox homeostasis. In this mini-review, we discuss the latest cutting-edge research discoveries on novel molecular targets of metformin in glycemic control, cardiovascular protection, cancer intervention, anti-inflammation, antiaging, and weight control. Identification of these novel targets and pathways not only deepens our understanding of the molecular mechanisms by which metformin exerts diverse beneficial biological effects, but also provides opportunities for developing new mechanistically based drugs for human diseases.
Collapse
Affiliation(s)
- Hong Zhu
- Department of Physiology and Pathophysiology, Jerry M. Wallace School of Osteopathic Medicine, Campbell University SOM, Buies Creek, NC, USA.
| | - Zhenquan Jia
- Department of Biology, College of Arts and Sciences, University of North Carolina, Greensboro, NC, USA
| | - Yunbo Robert Li
- Department of Pharmacology, Jerry M. Wallace School of Osteopathic Medicine, Campbell University, Buies Creek, NC, USA
| | - Igor Danelisen
- Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| |
Collapse
|
49
|
Hahn AW, Venkatesh N, Msaouel P, McQuade JL. The Influence of Obesity on Outcomes with Immune Checkpoint Blockade: Clinical Evidence and Potential Biological Mechanisms. Cells 2023; 12:2551. [PMID: 37947629 PMCID: PMC10649394 DOI: 10.3390/cells12212551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Immune checkpoint blockade (ICB) is a mainstay of treatment for advanced cancer, yet tumor response and host toxicity are heterogenous in those patients who receive ICB. There is growing interest in understanding how host factors interact with tumor intrinsic properties and the tumor microenvironment to influence the therapeutic index with ICB. Obesity, defined by body mass index, is a host factor associated with improved outcomes in select cancers when treated with ICB. While the biological mechanism for this obesity paradox is not fully understood, pre-clinical and translational studies suggest obesity may potentially impact tumor metabolism, inflammation, and angiogenesis. Herein, we summarize clinical studies that support an obesity paradox with ICB, explore potential biological mechanisms that may account for the obesity paradox, and address methodological challenges to consider when studying obesity and treatment outcomes.
Collapse
Affiliation(s)
- Andrew W. Hahn
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Neha Venkatesh
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer L. McQuade
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
50
|
Akce M, Farran B, Switchenko JM, Rupji M, Kang S, Khalil L, Ruggieri-Joyce A, Olson B, Shaib WL, Wu C, Alese OB, Diab M, Lesinski GB, El-Rayes BF. Phase II trial of nivolumab and metformin in patients with treatment-refractory microsatellite stable metastatic colorectal cancer. J Immunother Cancer 2023; 11:e007235. [PMID: 37852737 PMCID: PMC10603338 DOI: 10.1136/jitc-2023-007235] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Preclinical studies showed metformin reduces exhaustion of tumor-infiltrating lymphocytes and potentiates programmed cell death protein-1 (PD-1) blockade. We hypothesized that metformin with nivolumab would elicit potent antitumor and immune modulatory activity in metastatic microsatellite stable (MSS) colorectal cancer (CRC). We evaluated this hypothesis in a phase II study. METHODS Nivolumab (480 mg) was administered intravenously every 4 weeks while metformin (1000 mg) was given orally, two times per day following a 14-day metformin only lead-in phase. Patients ≥18 years of age, with previously treated, stage IV MSS CRC, and Eastern Cooperative Oncology Group 0-1, having received no prior anti-PD-1 agent were eligible. The primary endpoint was overall response rate with secondary endpoints of overall survival (OS) and progression-free survival (PFS). Correlative studies using paired pretreatment/on-treatment biopsies and peripheral blood evaluated a series of immune biomarkers in the tumor microenvironment and systemic circulation using ChipCytometry and flow cytometry. RESULTS A total of 24 patients were enrolled, 6 patients were replaced per protocol, 18 patients had evaluable disease. Of the 18 evaluable patients, 11/18 (61%) were women and the median age was 58 (IQR 50-67). Two patients had stable disease, but no patients had objective response, hence the study was stopped for futility. Median OS and PFS was 5.2 months (95% CI (3.2 to 11.7)) and 2.3 months (95% CI (1.7 to 2.3)). Most common grade 3/4 toxicities: Anemia (n=2), diarrhea (n=2), and fever (n=2). Metformin alone failed to increase the infiltration of T-cell subsets in the tumor, but combined metformin and nivolumab increased percentages of tumor-infiltrating leukocytes (p=0.031). Dual treatment also increased Tim3+ levels in patient tissues and decreased naïve CD8+T cells (p=0.0475). CONCLUSIONS Nivolumab and metformin were well tolerated in patients with MSS CRC but had no evidence of efficacy. Correlative studies did not reveal an appreciable degree of immune modulation from metformin alone, but showed trends in tumorous T-cell infiltration as a result of dual metformin and PD-1 blockade despite progression in a majority of patients.
Collapse
Affiliation(s)
- Mehmet Akce
- Division of Hematology and Oncology, Department of Medicine, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| | - Batoul Farran
- Department of Oncology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jeffrey M Switchenko
- Biostatistics Shared Resource, Emory University Winship Cancer institute, Atlanta, Georgia, USA
- Department of Biostsatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Manali Rupji
- Biostatistics Shared Resource, Emory University Winship Cancer institute, Atlanta, Georgia, USA
| | - Sandra Kang
- Department of Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Lana Khalil
- Department of Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Amanda Ruggieri-Joyce
- Department of Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Brian Olson
- Department of Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Walid L Shaib
- Northwest Georgia Oncology Centers Wellstar, Marietta, Georgia, USA
| | - Christina Wu
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Olatunji B Alese
- Department of Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Maria Diab
- Department of Internal Medicine, Michigan State University, East Lansing, Michigan, USA
- Department of Internal Medicine, Henry Ford Health, Detroit, Michigan, USA
| | - Gregory B Lesinski
- Department of Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Bassel F El-Rayes
- Division of Hematology and Oncology, Department of Medicine, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| |
Collapse
|