Skip to main content
BMB Reports logoLink to BMB Reports
. 2020 Oct 31;53(10):512–520. doi: 10.5483/BMBRep.2020.53.10.149

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

KyeongJin Kim 1,2, Wen-Hao Yang 3, Youn-Sang Jung 4, Jong-ho Cha 1,2,*
PMCID: PMC7607149  PMID: 32731915

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

INTRODUCTION

Metformin is the first-line oral medication for type 2 diabetes (T2D), approved by the FDA in 1994. It has been used for 1.5 million T2D patients worldwide and has been recognized as a safe and well-tolerated drug over the past several decades of clinical experience (1). Interestingly, several case-control studies for T2D patients showed a reduced incidence of various cancer types, suggesting an anti-tumor effect of metformin beyond that against diabetes (2, 3). Indeed, several studies have reported an anti-proliferative effect of metformin against various types of cancer by means of different pathways (4). Metformin–activated AMPK inhibits the mammalian target of rapamycin (mTOR) pathway, which regulates protein synthesis, cell proliferation, and cell survival (5). Metformin directly inhibits complex I of the mitochondrial electron-transport chain (ETC), leading to an overall reduction in various intracellular processes that consume ATP (6). Further, metformin treatment induces mitochondria-mediated apoptosis by increasing mitochondrion reactive oxygen species (ROS) (7). In addition, metformin can suppress the activation and expression of the signal transducer and activator of transcription 3 (STAT3) important for cancer-cell survival (8, 9). These studies provide mechanisms by which metformin directly inhibits cell growth by targeting the intrinsic pathway inside cancer cells.

Recent studies in the field of immuno-oncology reported the systemic effect of metformin associated with immunity. Interestingly, the anti-tumor effect of metformin is much higher in immunocompetent mouse models than in immunodeficient ones under the same conditions (10, 11), implying that the effect of metformin can be primarily mediated by anti-tumor immunity in physiological and clinical conditions. The immunomodulatory activity of metformin was already reported seventy years ago as an anti-viral and anti-malarial effect (12). This finding is supported by several recent findings in which met-formin increases CD8+ T-cell activity (10, 13).

Since T-cells are the main effector of anti-tumor immunity, T-cell activity has been the final target for most FDA-approved cancer immunotherapies [e.g., ICB (Ipilimumab, Nivolumab, Pembrolizumab, and Atezolizumab), dendritic-cell vaccine (Sipuleucel-T), and ACT (Axicabtageneciloleucel, and Tisagenlecleucel)] (14). However, since TIL activity is effectively inhibited by pro-tumoral TME, the efficacy of current immunotherapies is limited in most solid tumors. In previous studies on diabetes, cardiovascular disease, and autoimmunity, metformin has been shown to improve the prognosis of each disease by affecting inflammation, hypoxia, angiogenesis, and T-cell activity (3, 15), major components of TME as well. These results imply that the anti-tumor effect of metformin can occur via these TME components. Because these benefits suppress multiple TME components, metformin is worth considering as a potential adjuvant for T-cell-based cancer immunotherapy.

TUMOR DEVELOPMENT AND ANTI-TUMOR IMMUNITY

Cancer cells and our immunity are engaged in a fierce battle during tumor development, which occurs in three main stages: initiation, promotion, and progression. During the initiation stage, genomic mutations caused by several carcinogens, such as mutagenic chemicals, radiation, and viruses, can activate oncogenes and inhibit tumor suppressor genes (16). Initiated cells are transformed by the accumulation of critical mutations. When initiated cells are exposed to growth factors, the cells accumulate more mutations with high proliferation during the promotion stages (17). However, most transformed cells are removed by both innate immunity and adaptive immunity [the elimination phase of immunoediting] (18). In innate immunity, cancer cells having an abnormal expression of non-classical major histocompatibility complex (MHC) I are recognized by natural killer (NK) cells, and activated NK cells lyse cancer cells by secreting cytotoxic perforin and granzymes (19). M1 macrophages and dendritic cell (DC)s are recruited by inflammatory chemokines secreted from cancer cells. These antigen-presenting cells (APCs) do not only engulf cancer cells by means of phagocytosis but also support adaptive immunity by presenting tumor antigen to T-cells in the drain lymph node. Finally, educated CD8+ cytotoxic T lymphocytes (CTL) specifically eliminate cancer cells (20). A few cancer cells undergo repeated proliferation and elimination, resulting in their long-term survival without tumor formation [the equilibrium phase of immunoediting] (21). Cancer cells that endured harsh long-term screening obtain mutations to escape from immune surveillance [the escape phase of immunoediting]. Most survived cancer cells down-regulate the level of classic MHC I and the related-antigen processing and presentation machinery (APM), thereby preventing recognition by CD8+ CTL (22). On the other hand, cancer cells increase the level of non-classical MHC I to avoid the threat of NK cells (23, 24). After all this, cancer cells that escape from immune surveillance form tumors during the progression stage.

As tumors grow, most solid tumors establish TMEs composed of cellular and non-cellular constituents, including immune cells, fibroblasts, blood vessels, inflammatory signals, hypoxia, and glucose deprivation (25-27). The malignancy of cancer is closely related to the interaction between cancer cells and TME. In particular, pro-tumoral TME, such as hypoxia, chronic inflammation, and immune suppressors [e.g., inhibitory immune checkpoints, M2-like tumor-associated macrophages (TAMs), regulatory T-cells (T regs), and myeloid-derived suppressor cells (MDSCs)], inhibit T-cell surveillance and facilitate metastasis (28).

THE EFFECT OF METFORMIN ON ANTI-TUMOR IMMUNITY

TME components are entangled in very complex associations, and the malfunction of anti-tumor immunity in TME results from the accumulation of multiple immunosuppressive effects at different stages. Therefore, a comprehensive approach against multiple targets can be effective in improving the efficacy of cancer immunotherapy. The following are TME components targeted by metformin (Fig. 1 and Table 1). We want to understand the effect of metformin in terms of anti-tumor immunity.

Fig. 1.

Fig. 1

Several beneficial effects of metformin on anti-tumor immunity. Metformin indirectly increases T-cell activity by negatively regulating (A) chronic inflammation, (B) hypoxia, and (C) PD-L1 levels that inhibit T-cell activity. (D) Metformin directly relieves T-cell exhaustion by means of metabolic reprogramming of TIL and promotes memory T-cell differentiation. (E) Metformin shifts the profile of gut microbiota more favorably to T-cell immunity (TAM) tumor-associated macrophages; (Mj) macrophages; (MDSC) myeloid-derived suppressor cells; (T) T-cell; (DAMPs) Damage-associated molecular patterns; (APC) antigen presenting; (SCFA) short-chain fatty acid.

Table 1.

The effect of metformin and its downstream factors on TME components

TME components Key regulator / region Regulatory mechanism Disease References
Chronic inflammation TNF-γ, TNF-α, IL-6, and IL-17 Metformin down-regulates the level of chemokines and pro-inflammatory cytokines. Multiple sclerosis (41)
TNF-α and IL-6 Metformin decreases the level of inflammatory cytokines by suppressing NF-κB signaling. T2D, cardiovascular complications (42, 43)
(44, 45)
ROS and IL-1β Metformin decreases the level of inflammatory cytokines from LPS-activated macrophages by means of inhibition of mitochondrial complex I. Inflammation (46)
TAM polarization Metformin inhibit tumor growth by promoting M2-TAM polarization Cancer (47)
MDSC chemotaxis Metformin has anti-tumor effect by reducing MDSC accumulation in the TME via AMPK/DACH1/CXCL1 axis. Cancer (48)
Hypoxia Hypoxia Metformin reduces intratumoral hypoxia. Cancer (58)
HIF-1α Metformin suppresses accumulation of HIF-1α. Cancer (59, 60)
Oxygen consumption Metformin relieves intratumoral hypoxia, resulting in improving T-cell immunity in the TME. Cancer (61)
PD-L1 expression PD-L1 Metformin-activated AMPK induces ERAD of PD-L1, resulting in enhanced CD8+ TIL activity. Cancer (11)
PD-L1 Metformin improves cytotoxicity of CD8+ T-cell by reducing membrane-bound PD-L1 level. Cancer (68)
PD-L1 Metformin treatment reverses PD-L1 level, which sensitizes PARPi-resistant cells to CTL. Cancer (69)
Metabolic reprogramming of T-cell AMPK Metformin does not only increase the multi-functionality of CD8+ CTL but also improve long-term memory immunity. Cancer (10)
Oxidative metabolism AMPK maintain consistent activity of CTL under glucose depletion. Cancer (76)
Protein phosphatases Genetic ablation of AMPKα1 in T-cell cause apoptosis of CD8+ CTL. Cancer (77)
mTORC1 TSC2 KO mice generate highly glycolytic and potent effector CTL. Cancer (78)
mTOR Rapamycin treatment increases the population of CD8 + memory T-cells. Virus infection (82)

The effect of metformin in chronic inflammation

Inflammation is like a double-edged sword in cancer progression. As an integral part of innate immunity, inflammation plays important roles in removing transformed cells and subsequently presenting cancer antigens for adaptive immunity (29). Nevertheless, chronic inflammation is known to increase the incidence and malignancy in various types of cancer (30, 31). Activation of various inflammatory cells, such as macrophages, neutrophils, and mast cells, causes a cytokine storm, including tumor necrosis factor (TNF)-α, Interleukin (IL)-1β, IL-6, transforming growth factor (TGF)-β, and ROS (32). Chronic exposure to these inflammatory signals increases the population of M2-like TAM in TME (33), and not only induces metastasis by activating angiogenesis and epithelial-mesenchymal transition (EMT) (26, 34), but also suppresses T-cell immunity (35). Further, chronic inflammation can differentiate bone-marrow myeloid precursor cells into MDSCs, a major type of immune suppressive cells (36), which deplete T-cells by expressing inhibitory checkpoints and interfering with T-cell trafficking into tumors (37).

Pharmacological and genetic inhibition of these inflammatory factors has reduced tumor growth in animal models, and several control-case studies have shown that using non-steroidal anti-inflammatory drugs (NSAIDs) reduces incidence and mortality in various types of cancer (38-40). Recently, an anti-inflammatory effect of metformin has been reported in several different disease models (Fig. 1A and Table 1). Metformin attenuated the induction of multiple sclerosis, a central nervous system (CNS) autoimmune disease. In this study, metformin inhibited the infiltration of monocytes into the CNS by down-regulating the level of chemokines and pro-inflammatory cytokines like TNF-γ, TNF-α, IL-6, and IL-17 (41). Control-case studies on T2D patients indicated that metformin reduces cardiovascular complications (42, 43), and related mechanism studies suggested that metformin decreases the level of inflammatory cytokines, including TNF-α and IL-6, by inactivating NF-κB signaling via the AMPK pathway (44, 45). Further, metformin also inhibited generation of ROS and IL-1β from LPS-activated macrophages by inhibiting the mitochondrial complex I (46). Consistently, Wang et al. showed that metformin inhibits both tumor growth and angiogenesis by tilting TAM polarization from an M2- to an M1-like phenotype (47). Moreover, Qin et al. reported that metformin has an anti-tumor effect by reducing MDSC accumulation in the TME via the AMPK/DACH1/CXCL1 axis (48). Collectively, metformin has a beneficial effect of alleviating pro-tumoral inflammation, which is expected to prevent cancer malignancy and T-cell exhaustion in the TME.

The effect of metformin in intratumoral hypoxia

Hypoxia is an important malignant TME component (49). As a tumor grows, cancer cells rapidly consume oxygen. which induces hypoxia in the tumor (50). Cancer cells significantly alter their genetic profiles to overcome the harsh condition and maintain proliferation (51). Hypoxia-inducible factor (HIF-1) accumulated by hypoxia is the major transcription factor that activates genes involved in glucose absorption, glycolysis, and angiogenesis (51, 52). Hypoxia and HIF signaling also induce metastasis by increasing expression of EMT-associated genes and by increasing dissemination of cancer cells by means of angiogenesis (53). Furthermore, hypoxia and HIF signaling help cancer cells escape from immune surveillance. Hypoxia and HIF signaling are known to increase the population of immuno-suppressive cells, such as MDSCs, M2-like TAM, and Treg, in the tumor (54, 55). In addition, hypoxia may interfere with adaptive immunity by inhibiting the stimulatory capacity of DC to activate T-cells (56). Hypoxia also limits the number of TILs by reducing proliferation and viability in the TME (57). Consistently, hypoxia increases the level of PD-L1, a major inhibitory immune checkpoint molecule that induces T-cell exhaustion in the TME.

A recent study reported the anti-hypoxic effect of metformin (Fig. 1B and Table 1). In this study, metformin treatment reduced a hypoxic probe-positive area, which was accompanied by a reduced level of HIF-1α and pro-angiogenic factors (58). Consistently, metformin suppresses accumulation of HIF-1α by hypoxia in hepatocellular carcinoma (59) and oral squamouscell carcinoma (60). Importantly, Scharping et al. reported that metformin can relieve intratumoral hypoxia by reducing oxygen consumption, resulting in improving T-cell immunity in the TME. Consequently, metformin had a significant synergetic anti-tumor effect with ICB targeting PD-1 (61). Such evidence suggests that metformin treatment can improve T-cell immunity by alleviating intratumoral hypoxia.

The effect of metformin in the expression of PD-L1

Programmed death-ligand 1 (PD-L1) is a main immune check-point molecule that negatively regulates T-cell activity. In various cancer types, such as renalcell carcinoma (RCC) breast cancer, colorectal cancer, gastric cancer, non-small-cell lung cancer (NSCLC), papillary thyroid, and testicular cancers (62, 63), a high level of PD-L1 expression is observed, which is associated with poor prognosis (63-65). The binding of PD-L1 with programmed cell death protein 1 (PD-1) receptor on activated CD8+ CTL inhibits a T-cell receptor (TCR) signaling cascade and suppresses the activity of CTLs (66, 67). Therefore, there have been active attempts to restore CTL activity by blocking PD-L1/PD-1 signaling. FDA approved-ICBs, such as nivolumab, pembrolizumab, and atezolizumab, target PD-1 or PD-L1 (14).

Recently, our group reported that metformin can increase anti-tumor immunity by improving CD8+ TIL activity (Fig. 1C and Table 1) (11). As the underlining mechanism, metformin-activated AMPK directly phosphorylates at Serine 195 of PD-L1, and this phosphorylation induces abnormal glycosylation of PD-L1. Eventually, abnormal PD-L1 is degraded by means of the endoplasmic-reticulum-associated protein degradation (ERAD) pathway, which recovers CD8+ CTL activity. Furthermore, metformin-treated breast cancer patients’ data and animal work strongly support that a combination of metformin with a non-PD-1/PD-L1 targeting ICB can be effective in treating patients with PD-L1-expressing tumors. Similar to these results, Verdure et al. mentioned that metformin improves cytotoxicity of CD8+ T-cells by reducing the membrane-bound PD-L1 level increased by TNF-γ treatment (68). Consistently, Han et al. showed that poly ADP ribose polymerase inhibitor (PARPi)-resistant cells up-regulate PD-L1 expression, and metformin treatment reverses the PD-L1 level, which sensitizes PARPi-resistant cells to CTL (69). These studies all suggest that metformin can increase T-cell immunity by reducing PD-L1 level in the TME.

The effect of metformin in metabolic reprogramming of T-cells

In adaptive immunity, effector CD8+ CTLs educated by phagocytic APCs in the drain lymph nodes can specifically recognize and kill tumor cells (70). However, effector CTLs undergo metabolic exhaustion in TME, which causes their malfunction and apoptosis (71). The cancer cell relies on ineffective aerobic glycolysis as the main source of ATP, not oxidative phosphorylation, and fills the need by actively absorbing glucose (72). An unusual metabolic property called the ‘Warburg effect’ has an advantage for cancer cells in survival under hypoxia. Interestingly, the metabolism of effector CTL is converted to aerobic glycolysis after binding with a cancer antigen in the tumor (73). This metabolic similarity encourages glucose-uptake competition between cancer cells and CTL, leading to metabolic exhaustion of effector CD8 + CTL (74). In this regard, the metabolic reprogramming of exhausted T-cells can be a therapeutic target to increase anti-tumor immunity.

Since AMPK and mTOR, downstream factors of metformin, are the main regulators in energy sensing and mitochondrial function, metformin may play on the metabolic reprogramming of exhausted T-cells (Fig. 1D and Table 1) (75). Indeed, metformin treatment has been demonstrated to increase the number of CD8+ CTL and prevent their exhaustion in the tumor (10, 11). AMPK is reported to maintain consistent activity of CTL by activating oxidative metabolism under glucose depletion (76). Rao et al. also showed that genetic ablation of AMPKα1 in T-cells cause apoptosis of CD8+ CTL during in vitro activation and in vivo tumor development, and promote tumor growth in an immune-competent mouse model (77). Consistently, when tuberous sclerosis complex 2 (TSC2), an mTORC1 negative regulator, is genetically ablated, the mice showed the generation of highly glycolytic and potent effector CTL (78).

Most CD8+ T-cells that have completed their mission are removed by means of apoptosis, and some of them differentiate into CD8+ memory T-cells, which respond faster and more effectively to the same enemy, preventing recurrence (79). This transition to CD8 + memory T-cells also requires metabolic reprogramming (80). Interestingly, Elikawa et al. showed that metformin not only increases the multi-functionality of CD8+ CTL but also improves long-term memory immunity (10). In this study, metformin treatment increased the effector memory T-cell/central memory T-cell (TEM/TCM) ratio. Indeed, the AMPK-mTOR pathway is known to regulate memory CD8+ T-cell differentiation. Cantrell et al showed that AMPKα1-deficient T-cells have a defect in generation of CD8 + memory T-cells (81). Consistent with this result, mTOR inhibition with rapamycin increased the population of CD8 + memory T-cells in a virus-infected mice and primate model (82). Overall, metformin probably increases anti-tumor efficacy of CTL and its in vivo persistence by means of metabolic reprogramming via the AMPK/ mTOR pathway.

PERSPECTIVES AND FUTURE DIRECTIONS

Abnormal gut microbiota profiles have emerged in a variety of diseases related to immune disorder, such as inflammatory bowel disease, rheumatoid arthritis, myocardial metabolic disease, CNS autoimmune disease, and cancer (83, 84). Symptoms of such diseases have been improved by means of microbial modulation by dietary therapy or direct transplantation of beneficial microbes (85), suggesting that the balance between the immune system and the symbiotic microbiota is very important for maintaining our immunity. The following studies suggest that gut microbiota may be an important mediator of metformin which positively affects anti-tumor immunity.

Recently, three different groups reported at similar times that distinct gut microbiota profiles are observed between ICB responders and non-reponders, and that the therapeutic efficacy of ICB is greatly affected by the gut microbiota (86-88).The fecal microbial transplant (FMT) model showed that the efficacy of ICB is much higher in mice transplanted from IBC responders than in those from non-responders, and FMT from ICB responders highly improved T-cell immunity. In addition, microbes beneficial for ICB immunotherapy were identified as (1) Faecalibacterium prausnitzii (86), (2) Bifidobacterium longum (87), and (3) Akkermansia muciniphila (88).

Cuesta-Zuluaga et al. demonstrated the relevance of T2D, metformin, and gut microbiota (Fig. 1E and Table 1) (89). Interestingly, metformin shifted the gut microbiota profile of T2D patients, and in particular increased A. muciniphila and several Bifidobacterium species. As mentioned, A. muciniphila and Bifidobacterium are species identified as microbes beneficial for ICB immunotherapy (87, 88). Further, A. muciniphila and Bifidobacterium species are known to produce short-chain fatty acids (SCFA), like acetate, propionate, and butyrate, which have immunemodulatory and anti-inflammatory effects (90, 91). Such evidence strongly supports that metformin could affect anti-tumor immunity in a systematic manner via gut microbiota. However, it is largely undefined how metformin can shift a gut microbiota profile to be favorable to anti-tumor immunity and what is the link between metformin-increased germ species and anti-tumor immunity. To address these issues, future studies are needed, which are expected to greatly increase the usefulness of metformin and provide new insights and therapeutic targets for cancer immunity.

CONCLUDING REMARKS

Most of the current cancer immunotherapy targets TIL and works by maintaining the activity of TIL in TME. In order to ensure their effectiveness, anti-tumor immune processes, including APCs infiltration, cancer antigen presentation, T-cell priming, and TIL activity maintenance, must be adequately achieved (92). Indeed, “hot” tumors that achieve these prerequisites respond well to cancer immunotherapy, but “cold” tumors that do not meet the prerequisites have a poor response to immunotherapy (93). If cold tumors can turn into hot ones, we could greatly improve the efficacy of current cancer immunotherapy, as well as expand the beneficiaries.

In this favorable conversion, the problem is that immuno-suppressive TME in cold tumors inhibits multiple targets at different stages of anti-tumor immunity (94). Therefore, it would be difficult to expect an effective conversion with a single factor or path-adjusting approach. Although the choice of inhibitor cocktail can be considered to target multiple immuno-suppressive factors, it would probably cause excessive side effects, such as accumulated toxicity. To support the conversion of cold tumors, the repositioning of previously well-tolerated drugs that have a multi-immunomodulatory effect would be a more effective and safer approach.

As summarized in this mini-review, metformin’s safety and tolerability have been proven by decades of clinical experience. In addition, this drug has multiple beneficial effects on anti-tumor immunity, such as by havinging anti-inflammatory and anti-hypoxic effects, increasing T-cell activity, and acting favorably on the gut microbiota (Fig. 1 and Table 2). Moreover, control-case studies showed that metformin reduces the incidence of various types of cancer, and the positive effect of metformin on anti-tumor immunity has been consistently observed by means of several animal models. These findings strongly suggest that metformin has a high potential to be an adjuvant to comprehensively overwhelm immunosuppressive TMEs by supporting the stable conversion of cold tumors.

Table 2.

Current clinical trials of metformin combined with ICB

Title (NCT No.) Schedule Disease Drug Sponsor
(Collaborator)
Phases
Nivolumab and Metformin Hydrochloride in Treating Patients With Stage III-IV Non-small Cell Lung Cancer That Cannot Be Removed by Surgery (NCT03048500) Start : Jun. 2017
End : Feb. 2021
NSCLC Metformin
Nivolumab
Northwestern University (BMS, NCI) Phase 2
Nivolumab and Metformin in Patients With Treatment Refractory MSS Colorectal Cancer (NCT03800602) Start: Jan. 2019
End : Jan. 2023
Colorectal
cancer
Metformin
Nivolumab
Emory University (BMS) Phase 2
Combining Pembrolizumab and Metformin in Metastatic Head and Neck Cancer Patients (NCT04414540) Start: Jul. 2020
End : Feb. 2024
HNSCC Metformin
Pembrolizumab
UC Health (American Cancer Society) Phase 2
Anti-PD-1 mAb Plus Metabolic Modulator in Solid Tumor Malignancies (NCT04114136) Start: May 2020
End : Nov. 2023
Melanoma
RCC
NSCLC
HCC HNSCC
Metformin
Pembrolizumab
Nivolumab
University of Pittsburgh Phase 2
A Trial of Pembrolizumab and MetforminVersus
Pembrolizumab Alone in Advanced Melanoma (NCT03311308)
Start: Dec. 2017
End : Dec. 2028
Melanoma Metformin
Pembrolizumab
University of Pittsburgh (Merck) Phase 1

In current clinical trials that reflect this possibility, metformin is being co-administered with ICB targeting PD-L1/PD-1 (nivolumab and pembrolizumab) in patients with advanced melanoma, colorectal cancer, hepatocellular carcinoma (HCC), head and neck squamous-cell carcinoma (HNSCC), NSCLC, and RCC (Table 2). In anticipation of the old friend's re-evaluation, let's pay attention to the upcoming results.

ACKNOWLEDGEMENTS

This work was supported in part by the following: The National Research Foundation of Korea (NRF) grant funded by the Korea government Ministry of Science and ICT (MSIT) (2020R1C1C1005631 to J.-H. Cha and 2020R1C1C1004015 to K. Kim), INHA UNIVERSITY Research Grant [to K. Kim and J.-H. Cha], Ying Tsai Young Scholar Award (CMU108-YTY-04), the Ministry of Science and Technology (MOST; 109-2314-B-039-054) [to W.-H. Yang], and CHUNG-ANG UNIVERSITY Grant [to Y.-S. Jung].

Footnotes

CONFLICTS OF INTEREST

The authors have no conflicting interest.

REFERENCES

  • 1.Irons BK, Minze MG. Drug treatment of type 2 diabetes mellitus in patients for whom metformin is contraindicated. Diabetes Metab Syndr Obes. 2014;7:15–24. doi: 10.2147/DMSO.S38753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metformin and reduced risk of cancer in diabetic patients. BMJ. 2005;330:1304–1305. doi: 10.1136/bmj.38415.708634.F7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Viollet B, Guigas B, Sanz Garcia N, Leclerc J, Foretz M, Andreelli F. Cellular and molecular mechanisms of metformin: an overview. Clin Sci (Lond) 2012;(Lond):253–270. doi: 10.1042/CS20110386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Aljofan M, Riethmacher D. Anticancer activity of metformin: a systematic review of the literature. Future Sci OA. 2019;5:FSO410. doi: 10.2144/fsoa-2019-0053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Dowling RJ, Zakikhani M, Fantus IG, Pollak M, Sonenberg N. Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells. Cancer Res. 2007;67:10804–10812. doi: 10.1158/0008-5472.CAN-07-2310. [DOI] [PubMed] [Google Scholar]
  • 6.Andrzejewski S, Siegel PM, St-Pierre J. Metabolic Profiles Associated With Metformin Efficacy in Cancer. Front Endocrinol (Lausanne) 2018;(Lausanne):372. doi: 10.3389/fendo.2018.00372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Tseng HW, Li SC, Tsai KW. Metformin Treatment Suppresses Melanoma Cell Growth and Motility Through Modulation of microRNA Expression. Cancers (Basel) 2019;(Basel):209. doi: 10.3390/cancers11020209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Deng XS, Wang S, Deng A, et al. Metformin targets Stat3 to inhibit cell growth and induce apoptosis in triple-negative breast cancers. Cell Cycle. 2012;11:367–376. doi: 10.4161/cc.11.2.18813. [DOI] [PubMed] [Google Scholar]
  • 9.Feng Y, Ke C, Tang Q, et al. Metformin promotes autophagy and apoptosis in esophageal squamous cell carcinoma by downregulating Stat3 signaling. Cell Death Dis. 2014;5:e1088. doi: 10.1038/cddis.2014.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Eikawa S, Nishida M, Mizukami S, Yamazaki C, Nakayama E, Udono H. Immune-mediated antitumor effect by type 2 diabetes drug, metformin. Proc Natl Acad Sci U S A. 2015;112:1809–1814. doi: 10.1073/pnas.1417636112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cha JH, Yang WH, Xia W, et al. Metformin Promotes Antitumor Immunity via Endoplasmic-Reticulum-Associated Degradation of PD-L1. Mol Cell. 2018;71:606–620. doi: 10.1016/j.molcel.2018.07.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Garcia EY. Flumamine, a new synthetic analgesic and anti-flu drug. J Philipp Med Assoc. 1950;26:287–293. [PubMed] [Google Scholar]
  • 13.Zhang Z, Li F, Tian Y, et al. Metformin Enhances the Antitumor Activity of CD8(+) T Lymphocytes via the AMPK-miR-107-Eomes-PD-1 Pathway. J Immunol. 2020;204:2575–2588. doi: 10.4049/jimmunol.1901213. [DOI] [PubMed] [Google Scholar]
  • 14.Cha JH, Chan LC, Song MS, Hung MC. New Approaches on Cancer Immunotherapy. Cold Spring Harb Perspect Med. 2019;10:a036863. doi: 10.1101/cshperspect.a036863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Del Barco S, Vazquez-Martin A, Cufi S, et al. Met-formin: multi-faceted protection against cancer. Oncotarget. 2011;2:896–917. doi: 10.18632/oncotarget.387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Basu AK. DNA Damage, Mutagenesis and Cancer. Int J Mol Sci. 2018;19:970. doi: 10.3390/ijms19040970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hyndman IJ. Review: the Contribution of both Nature and Nurture to Carcinogenesis and Progression in Solid Tumours. Cancer Microenviron. 2016;9:63–69. doi: 10.1007/s12307-016-0183-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases--elimination, equilibrium and escape. Curr Opin Immunol. 2014;27:16–25. doi: 10.1016/j.coi.2014.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Paul S, Lal G. The Molecular Mechanism of Natural Killer Cells Function and Its Importance in Cancer Immunotherapy. Front Immunol. 2017;8:1124. doi: 10.3389/fimmu.2017.01124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Reeves E, James E. Antigen processing and immune regulation in the response to tumours. Immunology. 2017;150:16–24. doi: 10.1111/imm.12675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kim R, Emi M, Tanabe K. Cancer immunoediting from immune surveillance to immune escape. Immunology. 2007;121:1–14. doi: 10.1111/j.1365-2567.2007.02587.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.de Charette M, Marabelle A, Houot R. Turning tumour cells into antigen presenting cells: The next step to improve cancer immunotherapy? Eur J Cancer. 2016;68:134–147. doi: 10.1016/j.ejca.2016.09.010. [DOI] [PubMed] [Google Scholar]
  • 23.Halenius A, Gerke C, Hengel H. Classical and non-classical MHC I molecule manipulation by human cytomegalovirus: so many targets-but how many arrows in the quiver? Cell Mol Immunol. 2015;12:139–153. doi: 10.1038/cmi.2014.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kochan G, Escors D, Breckpot K, Guerrero-Setas D. Role of non-classical MHC class I molecules in cancer immunosuppression. Oncoimmunology. 2013;2:e26491. doi: 10.4161/onci.26491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Balkwill FR, Capasso M, Hagemann T. The tumor microenvironment at a glance. J Cell Sci. 2012;125:5591–5596. doi: 10.1242/jcs.116392. [DOI] [PubMed] [Google Scholar]
  • 26.Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 2013;14:1014–1022. doi: 10.1038/ni.2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wang M, Zhao J, Zhang L, et al. Role of tumor microenvironment in tumorigenesis. J Cancer. 2017;8:761–773. doi: 10.7150/jca.17648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19:1423–1437. doi: 10.1038/nm.3394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kambayashi T, Laufer TM. Atypical MHC class II-expressing antigen-presenting cells: can anything replace a dendritic cell? Nat Rev Immunol. 2014;14:719–730. doi: 10.1038/nri3754. [DOI] [PubMed] [Google Scholar]
  • 30.Hagemann T, Balkwill F, Lawrence T. Inflammation and cancer: a double-edged sword. Cancer Cell. 2007;12:300–301. doi: 10.1016/j.ccr.2007.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zitvogel L, Pietrocola F, Kroemer G. Nutrition, inflammation and cancer. Nat Immunol. 2017;18:843–850. doi: 10.1038/ni.3754. [DOI] [PubMed] [Google Scholar]
  • 32.Dranoff G. Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer. 2004;4:11–22. doi: 10.1038/nrc1252. [DOI] [PubMed] [Google Scholar]
  • 33.Poh AR, Ernst M. Targeting Macrophages in Cancer: From Bench to Bedside. Front Oncol. 2018;8:49. doi: 10.3389/fonc.2018.00049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Landskron G, De la Fuente M, Thuwajit P, Thuwajit C, Hermoso MA. Chronic inflammation and cytokines in the tumor microenvironment. J Immunol Res. 2014;2014:149185. doi: 10.1155/2014/149185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.D'Elia RV, Harrison K, Oyston PC, Lukaszewski RA, Clark GC. Targeting the "cytokine storm" for therapeutic benefit. Clin Vaccine Immunol. 2013;20:319–327. doi: 10.1128/CVI.00636-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The Nature of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Trends Immunol. 2016;37:208–220. doi: 10.1016/j.it.2016.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hanson EM, Clements VK, Sinha P, Ilkovitch D, Ostrand-Rosenberg S. Myeloid-derived suppressor cells down-regulate L-selectin expression on CD4+ and CD8+ T cells. J Immunol. 2009;183:937–944. doi: 10.4049/jimmunol.0804253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Crusz SM, Balkwill FR. Inflammation and cancer: advances and new agents. Nat Rev Clin Oncol. 2015;12:584–596. doi: 10.1038/nrclinonc.2015.105. [DOI] [PubMed] [Google Scholar]
  • 39.Brasky TM, Potter JD, Kristal AR, et al. Non-steroidal anti-inflammatory drugs and cancer incidence by sex in the VITamins And Lifestyle (VITAL) cohort. Cancer Causes Control. 2012;23:431–444. doi: 10.1007/s10552-011-9891-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Liu Y, Chen JQ, Xie L, et al. Effect of aspirin and other non-steroidal anti-inflammatory drugs on prostate cancer incidence and mortality: a systematic review and meta-analysis. BMC Med. 2014;12:55. doi: 10.1186/1741-7015-12-55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Nath N, Khan M, Paintlia MK, Singh I, Hoda MN, Giri S. Metformin attenuated the autoimmune disease of the central nervous system in animal models of multiple sclerosis. J Immunol. 2009;182:8005–8014. doi: 10.4049/jimmunol.0803563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Triggle CR, Ding H. Cardiovascular impact of drugs used in the treatment of diabetes. Ther Adv Chronic Dis. 2014;5:245–268. doi: 10.1177/2040622314546125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lund SS, Tarnow L, Stehouwer CD, et al. 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. 2008;158:631–641. doi: 10.1530/EJE-07-0815. [DOI] [PubMed] [Google Scholar]
  • 44.Huang NL, Chiang SH, Hsueh CH, Liang YJ, Chen YJ, Lai LP. Metformin inhibits TNF-alpha-induced Ikap-paB kinase phosphorylation, IkappaB-alpha degradation and IL-6 production in endothelial cells through PI3K-depen-dent AMPK phosphorylation. Int J Cardiol. 2009;134:169–175. doi: 10.1016/j.ijcard.2008.04.010. [DOI] [PubMed] [Google Scholar]
  • 45.Ersoy C, Kiyici S, Budak F, et al. The effect of metformin treatment on VEGF and PAI-1 levels in obese type 2 diabetic patients. Diabetes Res Clin Pract. 2008;81:56–60. doi: 10.1016/j.diabres.2008.02.006. [DOI] [PubMed] [Google Scholar]
  • 46.Kelly B, Tannahill GM, Murphy MP, O'Neill LA. Metformin Inhibits the Production of Reactive Oxygen Species from NADH:Ubiquinone Oxidoreductase to Limit Induction of Interleukin-1beta (IL-1beta) and Boosts Inter-leukin-10 (IL-10) in Lipopolysaccharide (LPS)-activated Macrophages. J Biol Chem. 2015;290:20348–20359. doi: 10.1074/jbc.M115.662114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wang JC, Sun X, Ma Q, et al. Metformin's anti-tumour and anti-angiogenic activities are mediated by skewing macrophage polarization. J Cell Mol Med. 2018;22:3825–3836. doi: 10.1111/jcmm.13655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Qin G, Lian J, Huang L, et al. Metformin blocks myeloid-derived suppressor cell accumulation through AMPK-DACH1-CXCL1 axis. Oncoimmunology. 2018;7:e1442167. doi: 10.1080/2162402X.2018.1442167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Semenza GL. The hypoxic tumor microenvironment: A driving force for breast cancer progression. Biochim Biophys Acta. 2016;1863:382–391. doi: 10.1016/j.bbamcr.2015.05.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Brown JM. The hypoxic cell: a target for selective cancer therapy--eighteenth Bruce F. Cain Memorial Award lecture. Cancer Res. 1999;59:5863–5870. [PubMed] [Google Scholar]
  • 51.Eales KL, Hollinshead KE, Tennant DA. Hypoxia and metabolic adaptation of cancer cells. Oncogenesis. 2016;5:e190. doi: 10.1038/oncsis.2015.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Weidemann A, Johnson RS. Biology of HIF-1alpha. Cell Death Differ. 2008;15:621–627. doi: 10.1038/cdd.2008.12. [DOI] [PubMed] [Google Scholar]
  • 53.Subarsky P, Hill RP. The hypoxic tumour microenvironment and metastatic progression. Clin Exp Metastasis. 2003;20:237–250. doi: 10.1023/A:1022939318102. [DOI] [PubMed] [Google Scholar]
  • 54.Noman MZ, Messai Y, Carre T, et al. Microenviron-mental hypoxia orchestrating the cell stroma cross talk, tumor progression and antitumor response. Crit Rev Immunol. 2011;31:357–377. doi: 10.1615/CritRevImmunol.v31.i5.10. [DOI] [PubMed] [Google Scholar]
  • 55.Clambey ET, McNamee EN, Westrich JA, et al. 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. 2012;109:E2784–2793. doi: 10.1073/pnas.1202366109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Mancino A, Schioppa T, Larghi P, et al. Divergent effects of hypoxia on dendritic cell functions. Blood. 2008;112:3723–3734. doi: 10.1182/blood-2008-02-142091. [DOI] [PubMed] [Google Scholar]
  • 57.Vuillefroy de Silly R, Dietrich PY, Walker PR. Hypoxia and antitumor CD8+ T cells: An incompatible alliance? Oncoimmunology. 2016;5:e1232236. doi: 10.1080/2162402X.2016.1232236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Wang JC, Li GY, Li PP, et al. Suppression of hypoxia-induced excessive angiogenesis by metformin via elevating tumor blood perfusion. Oncotarget. 2017;8:73892–73904. doi: 10.18632/oncotarget.18029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Zhou X, Chen J, Yi G, et al. Metformin suppresses hypoxia-induced stabilization of HIF-1alpha through reprogramming of oxygen metabolism in hepatocellular carcinoma. Oncotarget. 2016;7:873–884. doi: 10.18632/oncotarget.6418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Guimaraes TA, Farias LC, Santos ES, et al. Metformin increases PDH and suppresses HIF-1alpha under hypoxic conditions and induces cell death in oral squamous cell carcinoma. Oncotarget. 2016;7:55057–55068. doi: 10.18632/oncotarget.10842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Scharping NE, Menk AV, Whetstone RD, Zeng X, Delgoffe GM. Efficacy of PD-1 Blockade Is Potentiated by Metformin-Induced Reduction of Tumor Hypoxia. Cancer Immunol Res. 2017;5:9–16. doi: 10.1158/2326-6066.CIR-16-0103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Thompson RH, Gillett MD, Cheville JC, et al. Costimulatory B7-H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. Proc Natl Acad Sci U S A. 2004;101:17174–17179. doi: 10.1073/pnas.0406351101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Zhang M, Li G, Wang Y, et al. PD-L1 expression in lung cancer and its correlation with driver mutations: a meta-analysis. Sci Rep. 2017;7:10255. doi: 10.1038/s41598-017-10925-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Brody R, Zhang Y, Ballas M, et al. PD-L1 expression in advanced NSCLC: Insights into risk stratification and treatment selection from a systematic literature review. Lung Cancer. 2017;112:200–215. doi: 10.1016/j.lungcan.2017.08.005. [DOI] [PubMed] [Google Scholar]
  • 65.Li J, Wang P, Xu Y. Prognostic value of programmed cell death ligand 1 expression in patients with head and neck cancer: A systematic review and meta-analysis. PLoS One. 2017;12:e0179536. doi: 10.1371/journal.pone.0179536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Schildberg FA, Klein SR, Freeman GJ, Sharpe AH. Coinhibitory Pathways in the B7-CD28 Ligand-Receptor Family. Immunity. 2016;44:955–972. doi: 10.1016/j.immuni.2016.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–264. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Verdura S, Cuyas E, Martin-Castillo B, Menendez JA. Metformin as an archetype immuno-metabolic adjuvant for cancer immunotherapy. Oncoimmunology. 2019;8:e1633235. doi: 10.1080/2162402X.2019.1633235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Han Y, Li CW, Hsu JM, et al. Metformin reverses PARP inhibitors-induced epithelial-mesenchymal transition and PD-L1 upregulation in triple-negative breast cancer. Am J Cancer Res. 2019;9:800–815. doi: 10.1200/JCO.2019.37.15_suppl.1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Garner H, de Visser KE. Immune crosstalk in cancer progression and metastatic spread: a complex conversation. Nat Rev Immunol. 2020;20:483–497. doi: 10.1038/s41577-019-0271-z. [DOI] [PubMed] [Google Scholar]
  • 71.Voss K, Larsen SE, Snow AL. Metabolic reprogramming and apoptosis sensitivity: Defining the contours of a T cell response. Cancer Lett. 2017;408:190–196. doi: 10.1016/j.canlet.2017.08.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Burns JS, Manda G. Metabolic Pathways of the Warburg Effect in Health and Disease: Perspectives of Choice, Chain or Chance. Int J Mol Sci. 2017;18:2755. doi: 10.3390/ijms18122755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Almeida L, Lochner M, Berod L, Sparwasser T. Metabolic pathways in T cell activation and lineage differentiation. Semin Immunol. 2016;28:514–524. doi: 10.1016/j.smim.2016.10.009. [DOI] [PubMed] [Google Scholar]
  • 74.Afonso J, Santos LL, Longatto-Filho A, Baltazar F. Competitive glucose metabolism as a target to boost bladder cancer immunotherapy. Nat Rev Urol. 2020;17:77–106. doi: 10.1038/s41585-019-0263-6. [DOI] [PubMed] [Google Scholar]
  • 75.Garza-Lombo C, Schroder A, Reyes-Reyes EM, Franco R. mTOR/AMPK signaling in the brain: Cell metabolism, proteostasis and survival. Curr Opin Toxicol. 2018;8:102–110. doi: 10.1016/j.cotox.2018.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Blagih J, Coulombe F, Vincent EE, et al. The energy sensor AMPK regulates T cell metabolic adaptation and effector responses in vivo. Immunity. 2015;42:41–54. doi: 10.1016/j.immuni.2014.12.030. [DOI] [PubMed] [Google Scholar]
  • 77.Rao E, Zhang Y, Zhu G, et al. Deficiency of AMPK in CD8+ T cells suppresses their anti-tumor function by inducing protein phosphatase-mediated cell death. Oncotarget. 2015;6:7944–7958. doi: 10.18632/oncotarget.3501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Pollizzi KN, Patel CH, Sun IH, et al. mTORC1 and mTORC2 selectively regulate CD8(+) T cell differentiation. J Clin Invest. 2015;125:2090–2108. doi: 10.1172/JCI77746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Pennock ND, White JT, Cross EW, Cheney EE, Tamburini BA, Kedl RM. T cell responses: naive to memory and everything in between. Adv Physiol Educ. 2013;37:273–283. doi: 10.1152/advan.00066.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.van der Windt GJ, Pearce EL. Metabolic switching and fuel choice during T-cell differentiation and memory development. Immunol Rev. 2012;249:27–42. doi: 10.1111/j.1600-065X.2012.01150.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Rolf J, Zarrouk M, Finlay DK, Foretz M, Viollet B, Cantrell DA. AMPKalpha1: a glucose sensor that controls CD8 T-cell memory. Eur J Immunol. 2013;43:889–896. doi: 10.1002/eji.201243008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Araki K, Turner AP, Shaffer VO, et al. mTOR regulates memory CD8 T-cell differentiation. Nature. 2009;460:108–112. doi: 10.1038/nature08155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Durack J, Lynch SV. The gut microbiome: Relationships with disease and opportunities for therapy. J Exp Med. 2019;216:20–40. doi: 10.1084/jem.20180448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Xu H, Liu M, Cao J, et al. The Dynamic Interplay between the Gut Microbiota and Autoimmune Diseases. J Immunol Res. 2019;2019:7546047. doi: 10.1155/2019/7546047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Balakrishnan B, Taneja V. Microbial modulation of the gut microbiome for treating autoimmune diseases. Expert Rev Gastroenterol Hepatol. 2018;12:985–996. doi: 10.1080/17474124.2018.1517044. [DOI] [PubMed] [Google Scholar]
  • 86.Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome modulates response to anti-PD-1 immunothe-rapy in melanoma patients. Science. 2018;359:97–103. doi: 10.1126/science.aan4236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Matson V, Fessler J, Bao R, et al. The commensal microbiome is associated with anti-PD-1 efficacy in meta-static melanoma patients. Science. 2018;359:104–108. doi: 10.1126/science.aao3290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Routy B, Le Chatelier E, Derosa L, et al. Gut micro-biome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359:91–97. doi: 10.1126/science.aan3706. [DOI] [PubMed] [Google Scholar]
  • 89.de la Cuesta-Zuluaga J, Mueller NT, Corrales-Agudelo V, et al. 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. 2017;40:54–62. doi: 10.2337/dc16-1324. [DOI] [PubMed] [Google Scholar]
  • 90.Feng W, Ao H, Peng C. Gut Microbiota, Short-Chain Fatty Acids, and Herbal Medicines. Front Pharmacol. 2018;9:1354. doi: 10.3389/fphar.2018.01354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Parada Venegas D, De la Fuente MK, Landskron G, et al. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front Immunol. 2019;10:277. doi: 10.3389/fimmu.2019.00277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol. 2020;20:1–18. doi: 10.1038/s41577-020-0306-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Duan Q, Zhang H, Zheng J, Zhang L. Turning Cold into Hot: Firing up the Tumor Microenvironment. Trends Cancer. 2020;6:605–618. doi: 10.1016/j.trecan.2020.02.022. [DOI] [PubMed] [Google Scholar]
  • 94.Bonaventura P, Shekarian T, Alcazer V, et al. Cold Tumors: A Therapeutic Challenge for Immunotherapy. Front Immunol. 2019;10:168. doi: 10.3389/fimmu.2019.00168. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from BMB Reports are provided here courtesy of Korean Society for Biochemistry and Molecular Biology

RESOURCES