Browse > Article
http://dx.doi.org/10.5483/BMBRep.2020.53.10.149

A new aspect of an old friend: the beneficial effect of metformin on anti-tumor immunity  

Kim, KyeongJin (Department of Biomedical Sciences, College of Medicine, Inha University)
Yang, Wen-Hao (Graduate Institute of Biomedical Sciences, China Medical University)
Jung, Youn-Sang (Department of Life Science, Chung-Ang University)
Cha, Jong-ho (Department of Biomedical Sciences, College of Medicine, Inha University)
Publication Information
BMB Reports / v.53, no.10, 2020 , pp. 512-520 More about this Journal
Abstract
T-cell-based cancer immunotherapies, such as immune checkpoint blockers (ICBs) and chimeric antigen receptor (CAR)-T-cells, have significant anti-tumor effects against certain types of cancer, providing a new paradigm for cancer treatment. However, the activity of tumor infiltrating T-cells (TILs) can be effectively neutralized in the tumor microenvironment (TME) of most solid tumors, rich in various immunosuppressive factors and cells. Therefore, to improve the clinical outcomes of established T-cell-based immunotherapy, adjuvants that can comprehensively relieve multiple immunosuppressive mechanisms of TME are needed. In this regard, recent studies have revealed that metformin has several beneficial effects on anti-tumor immunity. In this mini-review, we understand the immunosuppressive properties of TME and how metformin comprehensively enhances anti-tumor immunity. Finally, we will discuss this old friend's potential as an adjuvant for cancer immunotherapy.
Keywords
Adjuvant; Cancer immunotherapy; Cold tumor and tumor microenvironment; Metformin;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Gopalakrishnan V, Spencer CN, Nezi L et al (2018) Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359, 97-103   DOI
2 Matson V, Fessler J, Bao R et al (2018) The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 359, 104-108   DOI
3 Routy B, Le Chatelier E, Derosa L et al (2018) Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359, 91-97   DOI
4 de la Cuesta-Zuluaga J, Mueller NT, Corrales-Agudelo V et al (2017) Metformin Is Associated With Higher Relative Abundance of Mucin-Degrading Akkermansia muciniphila and Several Short-Chain Fatty Acid-Producing Microbiota in the Gut. Diabetes Care 40, 54-62   DOI
5 Feng W, Ao H and Peng C (2018) Gut Microbiota, Short-Chain Fatty Acids, and Herbal Medicines. Front Pharmacol 9, 1354   DOI
6 Parada Venegas D, De la Fuente MK, Landskron G et al (2019) Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front Immunol 10, 277   DOI
7 Waldman AD, Fritz JM and Lenardo MJ (2020) A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol 20, 1-18   DOI
8 Duan Q, Zhang H, Zheng J and Zhang L (2020) Turning Cold into Hot: Firing up the Tumor Microenvironment. Trends Cancer 6, 605-618   DOI
9 Bonaventura P, Shekarian T, Alcazer V et al (2019) Cold Tumors: A Therapeutic Challenge for Immunotherapy. Front Immunol 10, 168   DOI
10 Quail DF and Joyce JA (2013) Microenvironmental regulation of tumor progression and metastasis. Nat Med 19, 1423-1437   DOI
11 Kambayashi T and Laufer TM (2014) Atypical MHC class II-expressing antigen-presenting cells: can anything replace a dendritic cell? Nat Rev Immunol 14, 719-730   DOI
12 Hagemann T, Balkwill F and Lawrence T (2007) Inflammation and cancer: a double-edged sword. Cancer Cell 12, 300-301   DOI
13 Zitvogel L, Pietrocola F and Kroemer G (2017) Nutrition, inflammation and cancer. Nat Immunol 18, 843-850   DOI
14 Dranoff G (2004) Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer 4, 11-22   DOI
15 Poh AR and Ernst M (2018) Targeting Macrophages in Cancer: From Bench to Bedside. Front Oncol 8, 49   DOI
16 Landskron G, De la Fuente M, Thuwajit P, Thuwajit C and Hermoso MA (2014) Chronic inflammation and cytokines in the tumor microenvironment. J Immunol Res 2014, 149185   DOI
17 Gajewski TF, Schreiber H and Fu YX (2013) Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 14, 1014-1022   DOI
18 Wang M, Zhao J, Zhang L et al (2017) Role of tumor microenvironment in tumorigenesis. J Cancer 8, 761-773   DOI
19 D'Elia RV, Harrison K, Oyston PC, Lukaszewski RA and Clark GC (2013) Targeting the "cytokine storm" for therapeutic benefit. Clin Vaccine Immunol 20, 319-327   DOI
20 Kumar V, Patel S, Tcyganov E and Gabrilovich DI (2016) The Nature of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Trends Immunol 37, 208-220   DOI
21 Triggle CR and Ding H (2014) Cardiovascular impact of drugs used in the treatment of diabetes. Ther Adv Chronic Dis 5, 245-268   DOI
22 Hanson EM, Clements VK, Sinha P, Ilkovitch D and Ostrand-Rosenberg S (2009) Myeloid-derived suppressor cells down-regulate L-selectin expression on CD4+ and CD8+ T cells. J Immunol 183, 937-944   DOI
23 Crusz SM and Balkwill FR (2015) Inflammation and cancer: advances and new agents. Nat Rev Clin Oncol 12, 584-596   DOI
24 Brasky TM, Potter JD, Kristal AR et al (2012) Non-steroidal anti-inflammatory drugs and cancer incidence by sex in the VITamins And Lifestyle (VITAL) cohort. Cancer Causes Control 23, 431-444   DOI
25 Liu Y, Chen JQ, Xie L et al (2014) Effect of aspirin and other non-steroidal anti-inflammatory drugs on prostate cancer incidence and mortality: a systematic review and meta-analysis. BMC Med 12, 55   DOI
26 Nath N, Khan M, Paintlia MK, Singh I, Hoda MN and Giri S (2009) Metformin attenuated the autoimmune disease of the central nervous system in animal models of multiple sclerosis. J Immunol 182, 8005-8014   DOI
27 Lund SS, Tarnow L, Stehouwer CD et al (2008) Impact of metformin versus repaglinide on non-glycaemic cardiovascular risk markers related to inflammation and endothelial dysfunction in non-obese patients with type 2 diabetes. Eur J Endocrinol 158, 631-641   DOI
28 Kelly B, Tannahill GM, Murphy MP and O'Neill LA (2015) Metformin Inhibits the Production of Reactive Oxygen Species from NADH:Ubiquinone Oxidoreductase to Limit Induction of Interleukin-1beta (IL-1beta) and Boosts Interleukin- 10 (IL-10) in Lipopolysaccharide (LPS)-activated Macrophages. J Biol Chem 290, 20348-20359   DOI
29 Huang NL, Chiang SH, Hsueh CH, Liang YJ, Chen YJ and Lai LP (2009) Metformin inhibits TNF-alpha-induced IkappaB kinase phosphorylation, IkappaB-alpha degradation and IL-6 production in endothelial cells through PI3K-dependent AMPK phosphorylation. Int J Cardiol 134, 169-175   DOI
30 Ersoy C, Kiyici S, Budak F et al (2008) The effect of metformin treatment on VEGF and PAI-1 levels in obese type 2 diabetic patients. Diabetes Res Clin Pract 81, 56-60   DOI
31 Andrzejewski S, Siegel PM and St-Pierre J (2018) Metabolic Profiles Associated With Metformin Efficacy in Cancer. Front Endocrinol (Lausanne) 9, 372   DOI
32 Viollet B, Guigas B, Sanz Garcia N, Leclerc J, Foretz M and Andreelli F (2012) Cellular and molecular mechanisms of metformin: an overview. Clin Sci (Lond) 122, 253-270   DOI
33 Aljofan M and Riethmacher D (2019) Anticancer activity of metformin: a systematic review of the literature. Future Sci OA 5, FSO410
34 Dowling RJ, Zakikhani M, Fantus IG, Pollak M and Sonenberg N (2007) Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells. Cancer Res 67, 10804-10812   DOI
35 Tseng HW, Li SC and Tsai KW (2019) Metformin Treatment Suppresses Melanoma Cell Growth and Motility Through Modulation of microRNA Expression. Cancers (Basel) 11, 209   DOI
36 Cha JH, Yang WH, Xia W et al (2018) Metformin Promotes Antitumor Immunity via Endoplasmic-Reticulum-Associated Degradation of PD-L1. Mol Cell 71, 606-620 e607   DOI
37 Deng XS, Wang S, Deng A et al (2012) Metformin targets Stat3 to inhibit cell growth and induce apoptosis in triple-negative breast cancers. Cell Cycle 11, 367-376   DOI
38 Feng Y, Ke C, Tang Q et al (2014) Metformin promotes autophagy and apoptosis in esophageal squamous cell carcinoma by downregulating Stat3 signaling. Cell Death Dis 5, e1088   DOI
39 Eikawa S, Nishida M, Mizukami S, Yamazaki C, Nakayama E and Udono H (2015) Immune-mediated antitumor effect by type 2 diabetes drug, metformin. Proc Natl Acad Sci U S A 112, 1809-1814   DOI
40 Garcia EY (1950) Flumamine, a new synthetic analgesic and anti-flu drug. J Philipp Med Assoc 26, 287-293
41 Zhang Z, Li F, Tian Y et al (2020) Metformin Enhances the Antitumor Activity of CD8(+) T Lymphocytes via the AMPKmiR-107-Eomes-PD-1 Pathway. J Immunol 204, 2575-2588   DOI
42 Paul S and Lal G (2017) The Molecular Mechanism of Natural Killer Cells Function and Its Importance in Cancer Immunotherapy. Front Immunol 8, 1124   DOI
43 Irons BK and Minze MG (2014) Drug treatment of type 2 diabetes mellitus in patients for whom metformin is contraindicated. Diabetes Metab Syndr Obes 7, 15-24   DOI
44 Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR and Morris AD (2005) Metformin and reduced risk of cancer in diabetic patients. BMJ 330, 1304-1305   DOI
45 Cha JH, Chan LC, Song MS and Hung MC (2019) New Approaches on Cancer Immunotherapy. Cold Spring Harb Perspect Med 10, a036863
46 Del Barco S, Vazquez-Martin A, Cufi S et al (2011) Metformin: multi-faceted protection against cancer. Oncotarget 2, 896-917   DOI
47 Basu AK (2018) DNA Damage, Mutagenesis and Cancer. Int J Mol Sci 19, 970   DOI
48 Hyndman IJ (2016) Review: the Contribution of both Nature and Nurture to Carcinogenesis and Progression in Solid Tumours. Cancer Microenviron 9, 63-69   DOI
49 Mittal D, Gubin MM, Schreiber RD and Smyth MJ (2014) New insights into cancer immunoediting and its three component phases--elimination, equilibrium and escape. Curr Opin Immunol 27, 16-25   DOI
50 Reeves E and James E (2017) Antigen processing and immune regulation in the response to tumours. Immunology 150, 16-24   DOI
51 Kim R, Emi M and Tanabe K (2007) Cancer immunoediting from immune surveillance to immune escape. Immunology 121, 1-14   DOI
52 de Charette M, Marabelle A and Houot R (2016) Turning tumour cells into antigen presenting cells: The next step to improve cancer immunotherapy? Eur J Cancer 68, 134-147   DOI
53 Halenius A, Gerke C and Hengel H (2015) Classical and non-classical MHC I molecule manipulation by human cytomegalovirus: so many targets-but how many arrows in the quiver? Cell Mol Immunol 12, 139-153   DOI
54 Kochan G, Escors D, Breckpot K and Guerrero-Setas D (2013) Role of non-classical MHC class I molecules in cancer immunosuppression. Oncoimmunology 2, e26491   DOI
55 Balkwill FR, Capasso M and Hagemann T (2012) The tumor microenvironment at a glance. J Cell Sci 125, 5591-5596   DOI
56 Brown JM (1999) The hypoxic cell: a target for selective cancer therapy--eighteenth Bruce F. Cain Memorial Award lecture. Cancer Res 59, 5863-5870
57 Wang JC, Sun X, Ma Q et al (2018) Metformin's antitumour and anti-angiogenic activities are mediated by skewing macrophage polarization. J Cell Mol Med 22, 3825-3836   DOI
58 Qin G, Lian J, Huang L et al (2018) Metformin blocks myeloid-derived suppressor cell accumulation through AMPK-DACH1-CXCL1 axis. Oncoimmunology 7, e1442167   DOI
59 Semenza GL (2016) The hypoxic tumor microenvironment: A driving force for breast cancer progression. Biochim Biophys Acta 1863, 382-391   DOI
60 Eales KL, Hollinshead KE and Tennant DA (2016) Hypoxia and metabolic adaptation of cancer cells. Oncogenesis 5, e190   DOI
61 Weidemann A and Johnson RS (2008) Biology of HIF-1alpha. Cell Death Differ 15, 621-627   DOI
62 Subarsky P and Hill RP (2003) The hypoxic tumour microenvironment and metastatic progression. Clin Exp Metastasis 20, 237-250   DOI
63 Noman MZ, Messai Y, Carre T et al (2011) Microenvironmental hypoxia orchestrating the cell stroma cross talk, tumor progression and antitumor response. Crit Rev Immunol 31, 357-377   DOI
64 Clambey ET, McNamee EN, Westrich JA et al (2012) Hypoxia-inducible factor-1 alpha-dependent induction of FoxP3 drives regulatory T-cell abundance and function during inflammatory hypoxia of the mucosa. Proc Natl Acad Sci U S A 109, E2784-2793   DOI
65 Mancino A, Schioppa T, Larghi P et al (2008) Divergent effects of hypoxia on dendritic cell functions. Blood 112, 3723-3734   DOI
66 Guimaraes TA, Farias LC, Santos ES et al (2016) Metformin increases PDH and suppresses HIF-1alpha under hypoxic conditions and induces cell death in oral squamous cell carcinoma. Oncotarget 7, 55057-55068   DOI
67 Vuillefroy de Silly R, Dietrich PY and Walker PR (2016) Hypoxia and antitumor CD8+ T cells: An incompatible alliance? Oncoimmunology 5, e1232236   DOI
68 Wang JC, Li GY, Li PP et al (2017) Suppression of hypoxia-induced excessive angiogenesis by metformin via elevating tumor blood perfusion. Oncotarget 8, 73892-73904   DOI
69 Zhou X, Chen J, Yi G et al (2016) Metformin suppresses hypoxia-induced stabilization of HIF-1alpha through reprogramming of oxygen metabolism in hepatocellular carcinoma. Oncotarget 7, 873-884   DOI
70 Scharping NE, Menk AV, Whetstone RD, Zeng X and Delgoffe GM (2017) Efficacy of PD-1 Blockade Is Potentiated by Metformin-Induced Reduction of Tumor Hypoxia. Cancer Immunol Res 5, 9-16   DOI
71 Thompson RH, Gillett MD, Cheville JC et al (2004) Costimulatory B7-H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. Proc Natl Acad Sci U S A 101, 17174-17179   DOI
72 Zhang M, Li G, Wang Y et al (2017) PD-L1 expression in lung cancer and its correlation with driver mutations: a meta-analysis. Sci Rep 7, 10255   DOI
73 Brody R, Zhang Y, Ballas M et al (2017) PD-L1 expression in advanced NSCLC: Insights into risk stratification and treatment selection from a systematic literature review. Lung Cancer 112, 200-215   DOI
74 Li J, Wang P and Xu Y (2017) Prognostic value of programmed cell death ligand 1 expression in patients with head and neck cancer: A systematic review and metaanalysis. PLoS One 12, e0179536   DOI
75 Han Y, Li CW, Hsu JM et al (2019) Metformin reverses PARP inhibitors-induced epithelial-mesenchymal transition and PD-L1 upregulation in triple-negative breast cancer. Am J Cancer Res 9, 800-815
76 Schildberg FA, Klein SR, Freeman GJ and Sharpe AH (2016) Coinhibitory Pathways in the B7-CD28 Ligand-Receptor Family. Immunity 44, 955-972   DOI
77 Pardoll DM (2012) The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 12, 252-264   DOI
78 Verdura S, Cuyas E, Martin-Castillo B and Menendez JA (2019) Metformin as an archetype immuno-metabolic adjuvant for cancer immunotherapy. Oncoimmunology 8, e1633235   DOI
79 Garner H and de Visser KE (2020) Immune crosstalk in cancer progression and metastatic spread: a complex conversation. Nat Rev Immunol 20, 483-497   DOI
80 Voss K, Larsen SE and Snow AL (2017) Metabolic reprogramming and apoptosis sensitivity: Defining the contours of a T cell response. Cancer Lett 408, 190-196   DOI
81 Burns JS and Manda G (2017) Metabolic Pathways of the Warburg Effect in Health and Disease: Perspectives of Choice, Chain or Chance. Int J Mol Sci 18, 2755   DOI
82 Almeida L, Lochner M, Berod L and Sparwasser T (2016) Metabolic pathways in T cell activation and lineage differentiation. Semin Immunol 28, 514-524   DOI
83 Afonso J, Santos LL, Longatto-Filho A and Baltazar F (2020) Competitive glucose metabolism as a target to boost bladder cancer immunotherapy. Nat Rev Urol 17, 77-106   DOI
84 Garza-Lombo C, Schroder A, Reyes-Reyes EM and Franco R (2018) mTOR/AMPK signaling in the brain: Cell metabolism, proteostasis and survival. Curr Opin Toxicol 8, 102-110   DOI
85 Pennock ND, White JT, Cross EW, Cheney EE, Tamburini BA and Kedl RM (2013) T cell responses: naive to memory and everything in between. Adv Physiol Educ 37, 273-283   DOI
86 Blagih J, Coulombe F, Vincent EE et al (2015) The energy sensor AMPK regulates T cell metabolic adaptation and effector responses in vivo. Immunity 42, 41-54   DOI
87 Rao E, Zhang Y, Zhu G et al (2015) Deficiency of AMPK in CD8+ T cells suppresses their anti-tumor function by inducing protein phosphatase-mediated cell death. Oncotarget 6, 7944-7958   DOI
88 Pollizzi KN, Patel CH, Sun IH et al (2015) mTORC1 and mTORC2 selectively regulate CD8(+) T cell differentiation. J Clin Invest 125, 2090-2108   DOI
89 van der Windt GJ and Pearce EL (2012) Metabolic switching and fuel choice during T-cell differentiation and memory development. Immunol Rev 249, 27-42   DOI
90 Rolf J, Zarrouk M, Finlay DK, Foretz M, Viollet B and Cantrell DA (2013) AMPKalpha1: a glucose sensor that controls CD8 T-cell memory. Eur J Immunol 43, 889-896   DOI
91 Araki K, Turner AP, Shaffer VO et al (2009) mTOR regulates memory CD8 T-cell differentiation. Nature 460, 108-112   DOI
92 Durack J and Lynch SV (2019) The gut microbiome Relationships with disease and opportunities for therapy. J Exp Med 216, 20-40   DOI
93 Xu H, Liu M, Cao J et al (2019) The Dynamic Interplay between the Gut Microbiota and Autoimmune Diseases. J Immunol Res 2019, 7546047
94 Balakrishnan B and Taneja V (2018) Microbial modulation of the gut microbiome for treating autoimmune diseases. Expert Rev Gastroenterol Hepatol 12, 985-996   DOI