Ann Lab Med.  2024 Sep;44(5):426-436. 10.3343/alm.2023.0443.

Metformin Suppresses Both PD-L1 Expression in Cancer Cells and Cancer-Induced PD-1 Expression in Immune Cells to Promote Antitumor Immunity

Affiliations
  • 1Department of Health Sciences, The Graduate School of Dong-A University, Busan, Korea
  • 2Department of Medicinal Biotechnology, College of Health Science, Dong-A University, Busan, Korea
  • 3Department of Biomedical Sciences, Dong-A University, Busan, Korea

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.

Keyword

E3 ligase; Immunotherapy; Metformin; PD-L1; PD-1; Tumor immune evasion

Figure

  • Fig. 1 Metformin inhibits both PD-L1 expression on cancer cells and PD-1 expression on infiltrating immune cells in the TME. (A) Schematic diagram of the experimental procedures. B16F10 murine melanoma cells were subcutaneously injected into C57BL/6 mice. From day 3 after tumor implantation, the mice received an intraperitoneal injection of metformin or PBS daily. On day 20, after tumor implantation, the mice were sacrificed. (B) Representative images of B16F10 tumors. Scale bar, 1 cm. (C) Growth curve of subcutaneous B16F10 tumors derived from metformin-treated (red line; N=5) and PBS-treated (blue line; N=5) mice. (D) Tumor weight of subcutaneous B16F10 tumors on day 20 derived from metformin-treated (red line; N=5) and PBS-treated (blue line; N=5) mice. (E) Cell-surface analysis of PD-L1 protein expression in B16F10 single-cell suspensions derived from B16F10 tumors using flow cytometry. A representative histogram (left panel) and graph (right panel) are shown. (F) Cell-surface analysis of PD-1 protein expression in NK or CD8+ T single-cell suspensions derived from B16F10 tumors using flow cytometry. A representative histogram (left panel) and graph (right panel) are shown. *P<0.05; **P<0.01; ***P<0.001, Student’s t-test. Abbreviations: PD-L1, programmed death ligand 1; PD-1, programmed death 1; TME, tumor microenvironment; PBS, phosphate-buffered saline; NK, natural killer.

  • Fig. 2 Metformin inhibits transcriptional expression of PD-L1 in an AMPK-downregulated AKT/β-catenin signaling-dependent manner. (A, B) The indicated cells were treated or not with metformin for 12 hrs. Then, mRNA and protein expression levels of PD-L1 were determined using quantitative real-time PCR (A) and immunoblotting (B) with the indicated primers and antibodies, respectively. (C) A431 cells were treated or not with metformin for 12 hrs and subjected to immunoblotting analysis using the indicated antibodies. (D) A431 cells were transfected with TOP-FLASH or FOP-FLASH, followed by metformin (10 mM) treatment for 12 hrs. Luciferase activity was measured. Relative luciferase activity levels were normalized to levels in untreated cells and to Renilla luciferase activity levels. (E–G) A431 cells stably expressing control vector or CA-β-catenin were transfected with TOP-FLASH or FOP-FLASH, followed by metformin (10 mM) treatment for 12 hrs. Luciferase activity was measured. Relative luciferase activity levels were normalized to levels in untreated cells and to Renilla luciferase activity levels (E). mRNA and protein expression levels of PD-L1 were determined using quantitative real-time PCR (F) and immunoblotting (G) with the indicated primers and antibodies, respectively. (H–J) A431 cells stably expressing the control vector or HA-myr-AKT1 were transfected with TOP-FLASH or FOP-FLASH, followed by metformin (10 mM) treatment for 12 hrs. Luciferase activity was measured. Relative levels of luciferase activity were normalized to levels in untreated cells and to Renilla luciferase activity levels (H). The mRNA and protein expression levels of PD-L1 were determined using quantitative real-time PCR (I) and immunoblotting (J) with the indicated primers and antibodies, respectively. (K, L) A431 cells were pretreated or not pretreated with compound C (5 μM) for 1 hr and then treated or not pretreated with metformin (10 mM) for 12 hrs. Protein and mRNA expression levels of PD-L1 were determined using immunoblotting (K and bottom panel of L) and quantitative real-time PCR (L, top panel) with the indicated antibodies and primers, respectively. Data are presented as mean±SD of three independent experiments (A, D, E, F, H, I, L). *P<0.05; **P<0.01; ***P<0.001, Student’s t-test or one-way ANOVA followed by Tukey post-hoc tests. Abbreviations: PD-L1, programmed death ligand 1; AMPK, adenosine monophosphate-activated protein kinase; PCR, polymerase chain reaction; CA, constitutively active; HA, hemagglutinin.

  • Fig. 3 Cancer cells enhance PD-1 protein stability in NK and T cells. (A) NK-92 and Jurkat cells were treated or not treated with CM (50%) for the indicated periods. Protein and mRNA expression levels of PD-1 were determined using immunoblotting (upper panel) and quantitative real-time PCR (bottom panel) with the indicated antibodies and primers, respectively. Data are presented as mean±SD of three independent experiments. (B) NK-92 and Jurkat cells were pretreated with DMSO or CHX (100 μg/mL) for 1 hr and then treated with CM (50%) for 6 hrs. Immunoblotting analyses were performed using the indicated antibodies. (C) NK-92 and Jurkat cells were pretreated with CM (50%) for 12 hrs and then treated with CHX (100 μg/mL) for the indicated periods. Immunoblotting analyses were performed using the indicated antibodies (left panel). Quantification of PD-1 levels relative to tubulin levels is shown (right panel). Band intensities were quantified using the ImageJ software. Data are presented as mean±SD of three independent experiments. **P<0.01, Student’s t-test. (D) NK-92 and Jurkat cells were treated or not treated with CM (50%) for 12 hrs. Immunoprecipitation analyses were performed using an anti-PD-1 antibody, followed by immunoblotting analyses using the indicated antibodies. (E) NK-92 and Jurkat cells were treated with CM (50%) for the indicated periods. The cells were incubated with MG132 (10 μM) for 6 hrs before they were harvested using a guanidine-HCl-containing buffer. Immunoprecipitation was performed using an anti-PD-1 antibody, followed by immunoblotting analyses using the indicated antibodies. Abbreviations: PD-1, programmed death 1; NK, natural killer; CM, conditioned medium; DMSO, dimethyl sulfoxide; CHX, cycloheximide.

  • Fig. 4 Metformin inhibits cancer-enhanced PD-1 expression via AMPK-dependent deregulation of PD-1 protein stability. (A) NK-92 and Jurkat cells were treated or not with CM (50%) or metformin (10 mM) for 12 hrs. Immunoblotting analyses were performed using the indicated antibodies. (B) NK-92 and Jurkat cells were pretreated with DMSO or MG132 (10 μM) for 1 hr and then treated or not with CM (50%) or metformin (10 mM) for 12 hrs. Immunoblotting analyses were performed using the indicated antibodies. (C) NK-92 and Jurkat cells were pretreated or not with CM (50%) or metformin (10 mM) for 12 hrs and then treated with CHX (100 μg/mL) for the indicated periods. Immunoblotting analyses were performed using the indicated antibodies (left panel). Quantification of PD-1 levels relative to tubulin levels is shown (right panel). Band intensity was quantified using the ImageJ software. Data are presented as mean±SD of three independent experiments. *P<0.05 and **P<0.01, Student’s t-test. (D) NK-92 and Jurkat cells were treated or not with CM (50%) and metformin (10 mM) for 12 hrs. Immunoprecipitation was performed using an anti-PD-1 antibody, followed by immunoblotting analyses using the indicated antibodies. (E) NK-92 and Jurkat cells were treated or not with CM (50%) or metformin (10 mM) for 12 hrs. The cells were incubated with MG132 (10 μM) for 6 hrs before they were harvested using a guanidine-HCl-containing buffer. Immunoprecipitation was performed using an anti-PD-1 antibody, followed by immunoblotting analyses using the indicated antibodies. (F) 293T cells stably expressing V5-PD-1 and Flag-vector or Flag-CA-AMPK were treated or not with CM (50%) for 12 hrs. The cells were incubated with MG132 (10 μM) for 6 hrs before they were harvested using a guanidine-HCl-containing buffer. Immunoprecipitation was performed using an anti-V5 antibody, followed by immunoblotting analyses using the indicated antibodies. (G) AMPK WT and AMPK DKO MEFs were transfected or not with V5-PD-1. These cells were incubated with MG132 (10 μM) for 6 hrs before they were harvested using a guanidine-HCl-containing buffer. Immunoprecipitation was performed using an anti-V5 antibody, followed by immunoblotting analyses using the indicated antibodies. (H) NK-92 and Jurkat cells were pretreated with DMSO or compound C (5 μM) for 1 hr and then treated or not with CM (50%) and metformin (10 mM) for 12 hrs. Immunoblotting analyses were performed using the indicated antibodies. (I) 293T cells stably expressing V5-PD-1 were pretreated with DMSO or compound C (5 μM) for 1 hr and then treated or not with CM (50%) or metformin (10 mM) for 12 hrs. The cells were incubated with MG132 (10 μM) for 6 hrs before they were harvested using a guanidine-HCl-containing buffer. Immunoprecipitation was performed using an anti-V5 antibody, followed by immunoblotting analyses using the indicated antibodies. Abbreviations: PD-1, programmed death 1; AMPK, adenosine monophosphate-activated protein kinase; CM, conditioned medium; DMSO, dimethyl sulfoxide; CHX, cycloheximide; CA, constitutively active; WT, wild-type; DKO, double knockout.

  • Fig. 5 Schematic representation of the proposed mechanisms.


Reference

References

1. Boussiotis VA. 2016; Molecular and biochemical aspects of the PD-1 checkpoint pathway. N Engl J Med. 375:1767–78. DOI: 10.1056/NEJMra1514296. PMID: 27806234. PMCID: PMC5575761.
Article
2. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. 2017; Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 168:707–23. DOI: 10.1016/j.cell.2017.01.017. PMID: 28187290. PMCID: PMC5391692.
Article
3. Jiang X, Wang J, Deng X, Xiong F, Ge J, Xiang B, et al. 2019; Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol Cancer. 18:10. DOI: 10.1186/s12943-018-0928-4. PMID: 30646912. PMCID: PMC6332843. PMID: 70238db4d68c4e4bb58930633279c9c3.
Article
4. Du L, Lee JH, Jiang H, Wang C, Wang S, Zheng Z, et al. 2020; β-Catenin induces transcriptional expression of PD-L1 to promote glioblastoma immune evasion. J Exp Med. 217:e20191115. DOI: 10.1084/jem.20191115. PMID: 32860047. PMCID: PMC7596815.
Article
5. Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, et al. 2007; Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci U S A. 104:3360–5. DOI: 10.1073/pnas.0611533104. PMID: 17360651. PMCID: PMC1805580.
6. Thompson RH, Gillett MD, Cheville JC, Lohse CM, Dong H, Webster WS, 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. 101:17174–9. DOI: 10.1073/pnas.0406351101. PMID: 15569934. PMCID: PMC534606.
Article
7. Nusse R, Clevers H. 2017; Wnt/β-catenin signaling, disease, and emerging therapeutic modalities. Cell. 169:985–99. DOI: 10.1016/j.cell.2017.05.016. PMID: 28575679.
Article
8. Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, et al. 2009; Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 114:1537–44. DOI: 10.1182/blood-2008-12-195792. PMID: 19423728. PMCID: PMC2927090.
Article
9. Pesce S, Greppi M, Tabellini G, Rampinelli F, Parolini S, Olive D, et al. 2017; Identification of a subset of human natural killer cells expressing high levels of programmed death 1: a phenotypic and functional characterization. J Allergy Clin Immunol. 139:335–46.e3. DOI: 10.1016/j.jaci.2016.04.025. PMID: 27372564.
10. Hsu J, Hodgins JJ, Marathe M, Nicolai CJ, Bourgeois-Daigneault MC, Trevino TN, et al. 2018; Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J Clin Invest. 128:4654–68. DOI: 10.1172/JCI99317. PMID: 30198904. PMCID: PMC6159991.
Article
11. Muti P, Berrino F, Krogh V, Villarini A, Barba M, Strano S, et al. 2009; Metformin, diet and breast cancer: an avenue for chemoprevention. Cell Cycle. 8:2661. DOI: 10.4161/cc.8.16.9226. PMID: 19571669.
Article
12. Vancura A, Bu P, Bhagwat M, Zeng J, Vancurova I. 2018; Metformin as an anticancer agent. Trends Pharmacol Sci. 39:867–78. DOI: 10.1016/j.tips.2018.07.006. PMID: 30150001. PMCID: PMC6153060.
Article
13. Wink KCJ, Belderbos JSA, Dieleman EMT, Rossi M, Rasch CRN, Damhuis RAM, et al. 2016; Improved progression free survival for patients with diabetes and locally advanced non-small cell lung cancer (NSCLC) using metformin during concurrent chemoradiotherapy. Radiother Oncol. 118:453–9. DOI: 10.1016/j.radonc.2016.01.012. PMID: 26861738.
Article
14. Saif MW, Rajagopal S, Caplain J, Grimm E, Serebrennikova O, Das M, et al. 2019; A phase I delayed-start, randomized and pharmacodynamic study of metformin and chemotherapy in patients with solid tumors. Cancer Chemother Pharmacol. 84:1323–31. DOI: 10.1007/s00280-019-03967-3. PMID: 31583436.
Article
15. Iliopoulos D, Hirsch HA, Struhl K. 2011; Metformin decreases the dose of chemotherapy for prolonging tumor remission in mouse xenografts involving multiple cancer cell types. Cancer Res. 71:3196–201. DOI: 10.1158/0008-5472.CAN-10-3471. PMID: 21415163. PMCID: PMC3085572.
Article
16. Zhang CS, Li M, Ma T, Zong Y, Cui J, Feng JW, et al. 2016; Metformin activates AMPK through the lysosomal pathway. Cell Metab. 24:521–2. DOI: 10.1016/j.cmet.2016.09.003. PMID: 27732831.
Article
17. Dowling RJ, Zakikhani M, Fantus IG, Pollak M, Sonenberg N. 2007; Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells. Cancer Res. 67:10804–12. DOI: 10.1158/0008-5472.CAN-07-2310. PMID: 18006825.
Article
18. Blandino G, Valerio M, Cioce M, Mori F, Casadei L, Pulito C, et al. 2012; Metformin elicits anticancer effects through the sequential modulation of DICER and c-MYC. Nat Commun. 3:865. DOI: 10.1038/ncomms1859. PMID: 22643892.
Article
19. Ben Sahra I, Regazzetti C, Robert G, Laurent K, Le Marchand-Brustel Y, Auberger P, et al. 2011; Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1. Cancer Res. 71:4366–72. DOI: 10.1158/0008-5472.CAN-10-1769. PMID: 21540236.
Article
20. Cha JH, Yang WH, Xia W, Wei Y, Chan LC, Lim SO, et al. 2018; Metformin promotes antitumor immunity via endoplasmic-reticulum-associated degradation of PD-L1. Mol Cell. 71:606–20.e7. DOI: 10.1016/j.molcel.2018.07.030. PMID: 30118680. PMCID: PMC6786495.
Article
21. Lu Y, Xin D, Guan L, Xu M, Yang Y, Chen Y, et al. 2021; Metformin downregulates PD-L1 expression in esophageal squamous cell carcinoma by inhibiting IL-6 signaling pathway. Front Oncol. 11:762523. DOI: 10.3389/fonc.2021.762523. PMID: 34881181. PMCID: PMC8645640. PMID: c861b8c7f13c4c35a4f101a9ac29c246.
Article
22. Zhang JJ, Zhang QS, Li ZQ, Zhou JW, Du J. 2019; Metformin attenuates PD-L1 expression through activating Hippo signaling pathway in colorectal cancer cells. Am J Transl Res. 11:6965–76. PMID: 31814900. PMCID: PMC6895520.
23. Yang W, Xia Y, Ji H, Zheng Y, Liang J, Huang W, et al. 2011; Nuclear PKM2 regulates beta-catenin transactivation upon EGFR activation. Nature. 480:118–22. DOI: 10.1038/nature10598. PMID: 22056988. PMCID: PMC3235705.
Article
24. Rattan R, Giri S, Singh AK, Singh I. 2005; 5-Aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside inhibits cancer cell proliferation in vitro and in vivo via AMP-activated protein kinase. J Biol Chem. 280:39582–93. DOI: 10.1074/jbc.M507443200. PMID: 16176927.
25. Russo V, Protti MP. 2017; Tumor-derived factors affecting immune cells. Cytokine Growth Factor Rev. 36:79–87. DOI: 10.1016/j.cytogfr.2017.06.005. PMID: 28606733.
Article
26. Meng X, Liu X, Guo X, Jiang S, Chen T, Hu Z, et al. 2018; FBXO38 mediates PD-1 ubiquitination and regulates anti-tumour immunity of T cells. Nature. 564:130–5. DOI: 10.1038/s41586-018-0756-0. PMID: 30487606.
Article
27. Lyle C, Richards S, Yasuda K, Napoleon MA, Walker J, Arinze N, et al. 2019; c-Cbl targets PD-1 in immune cells for proteasomal degradation and modulates colorectal tumor growth. Sci Rep. 9:20257. DOI: 10.1038/s41598-019-56208-1. PMID: 31882749. PMCID: PMC6934810.
Article
28. Zhou XA, Zhou J, Zhao L, Yu G, Zhan J, Shi C, et al. 2020; KLHL22 maintains PD-1 homeostasis and prevents excessive T cell suppression. Proc Natl Acad Sci U S A. 117:28239–50. DOI: 10.1073/pnas.2004570117. PMID: 33109719. PMCID: PMC7668036.
Article
29. Kubo T, Ninomiya T, Hotta K, Kozuki T, Toyooka S, Okada H, et al. 2018; Study protocol: phase-Ib trial of nivolumab combined with metformin for refractory/recurrent solid tumors. Clin Lung Cancer. 19:e861–4. DOI: 10.1016/j.cllc.2018.07.010. PMID: 30172698.
Article
30. Oestreich KJ, Yoon H, Ahmed R, Boss JM. 2008; NFATc1 regulates PD-1 expression upon T cell activation. J Immunol. 181:4832–9. DOI: 10.4049/jimmunol.181.7.4832. PMID: 18802087. PMCID: PMC2645436.
Article
31. Austin JW, Lu P, Majumder P, Ahmed R, Boss JM. 2014; STAT3, STAT4, NFATc1, and CTCF regulate PD-1 through multiple novel regulatory regions in murine T cells. J Immunol. 192:4876–86. DOI: 10.4049/jimmunol.1302750. PMID: 24711622. PMCID: PMC4011967.
Article
32. Cho HY, Lee SW, Seo SK, Choi IW, Choi I, Lee SW. 2008; Interferon-sensitive response element (ISRE) is mainly responsible for IFN-alpha-induced upregulation of programmed death-1 (PD-1) in macrophages. Biochim Biophys Acta. 1779:811–9. DOI: 10.1016/j.bbagrm.2008.08.003. PMID: 18771758.
33. Terawaki S, Chikuma S, Shibayama S, Hayashi T, Yoshida T, Okazaki T, et al. 2011; IFN-α directly promotes programmed cell death-1 transcription and limits the duration of T cell-mediated immunity. J Immunol. 186:2772–9. DOI: 10.4049/jimmunol.1003208. PMID: 21263073.
Article
34. Tie Y, Tang F, Wei YQ, Wei XW. 2022; Immunosuppressive cells in cancer: mechanisms and potential therapeutic targets. J Hematol Oncol. 15:61. DOI: 10.1186/s13045-022-01282-8. PMID: 35585567. PMCID: PMC9118588. PMID: 274a447483bf40e79ac6103100cdf0e6.
Article
35. Wang S, Lin Y, Xiong X, Wang L, Guo Y, Chen Y, et al. 2020; Low-dose metformin reprograms the tumor immune microenvironment in human esophageal cancer: results of a phase II clinical trial. Clin Cancer Res. 26:4921–32. DOI: 10.1158/1078-0432.CCR-20-0113. PMID: 32646922.
Article
36. Eikawa S, Nishida M, Mizukami S, Yamazaki C, Nakayama E, Udono H. 2015; Immune-mediated antitumor effect by type 2 diabetes drug, metformin. Proc Natl Acad Sci U S A. 112:1809–14. DOI: 10.1073/pnas.1417636112. PMID: 25624476. PMCID: PMC4330733.
Article
37. Crist M, Yaniv B, Palackdharry S, Lehn MA, Medvedovic M, Stone T, et al. 2022; Metformin increases natural killer cell functions in head and neck squamous cell carcinoma through CXCL1 inhibition. J Immunother Cancer. 10:e005632. DOI: 10.1136/jitc-2022-005632. PMID: 36328378. PMCID: PMC9639146. PMID: d5e39b30e67c484baead80341faf7c76.
Article
Full Text Links
  • ALM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr