1932

Abstract

In order to fuel their relentless expansion, cancers must expand their vasculature to augment delivery of oxygen and essential nutrients. The disordered web of irregular vessels that results, however, leaves gaps in oxygen delivery that foster tumor hypoxia. At the same time, tumor cells increase their oxidative metabolism to cope with the energetic demands of proliferation, which further worsens hypoxia due to heightened oxygen consumption. In these hypoxic, nutrient-deprived environments, tumors and suppressive stroma evolve to flourish while antitumor immunity collapses due to a combination of energetic deprivation, toxic metabolites, acidification, and other suppressive signals. Reversal of cancer hypoxia thus has the potential to increase the survival and effector function of tumor-infiltrating T cells, as well as to resensitize tumors to immunotherapy. Early clinical trials combining hypoxia reduction with immune checkpoint blockade have shown promising results in treating patients with advanced, metastatic, and therapeutically refractory cancers.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-060619-022830
2022-01-27
2024-05-14
Loading full text...

Full text loading...

/deliver/fulltext/med/73/1/annurev-med-060619-022830.html?itemId=/content/journals/10.1146/annurev-med-060619-022830&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Saggar JK, Yu M, Tan Q, Tannock IF 2013. The tumor microenvironment and strategies to improve drug distribution. Front. Oncol. 3:154
    [Google Scholar]
  2. 2. 
    Wilson W, Hay M 2011. Targeting hypoxia in cancer therapy. Nat. Rev. Cancer 11:393–410
    [Google Scholar]
  3. 3. 
    Vaupel P, Harrison L. 2004. Tumor hypoxia: causative factors, compensatory mechanisms, and cellular response. Oncologist 9:Suppl. 54–9
    [Google Scholar]
  4. 4. 
    Hensley CT, Faubert B, Yuan Q et al. 2016. Metabolic heterogeneity in human lung tumors. Cell 164:4681–94
    [Google Scholar]
  5. 5. 
    Noman MZ, Hasmim M, Messai Y et al. 2015. Hypoxia: a key player in antitumor immune response. A review in the theme: Cellular Responses to Hypoxia. Am. J. Physiol. Cell Physiol. 309:9C569–79
    [Google Scholar]
  6. 6. 
    Ryan HE, Lo J, Johnson RS. 1998. HIF-1 alpha is required for solid tumor formation and embryonic vascularization. EMBO J 17:113005–15
    [Google Scholar]
  7. 7. 
    Hielscher A, Qiu C, Porterfield J et al. 2013. Hypoxia affects the structure of breast cancer cell-derived matrix to support angiogenic responses of endothelial cells. J. Carcinog. Mutagen. Suppl 13:005
    [Google Scholar]
  8. 8. 
    Folkman J. 1971. Tumor angiogenesis: therapeutic implications. N. Engl. J. Med. 285:211182–86
    [Google Scholar]
  9. 9. 
    Weis SM, Cheresh DA. 2011. Tumor angiogenesis: molecular pathways and therapeutic targets. Nat. Med. 17:111359–70
    [Google Scholar]
  10. 10. 
    Tang N, Wang L, Esko J et al. 2004. Loss of HIF-1α in endothelial cells disrupts a hypoxia-driven VEGF autocrine loop necessary for tumorigenesis. Cancer Cell 6:5485–95
    [Google Scholar]
  11. 11. 
    Skuli N, Liu L, Runge A et al. 2009. Endothelial deletion of hypoxia-inducible factor-2α (HIF-2α) alters vascular function and tumor angiogenesis. Blood 114:2469–77
    [Google Scholar]
  12. 12. 
    Skuli N, Majmundar AJ, Krock BL et al. 2012. Endothelial HIF-2α regulates murine pathological angiogenesis and revascularization processes. J. Clin. Investig. 122:41427–43
    [Google Scholar]
  13. 13. 
    Erkan M, Reiser-Erkan C, Michalski CW et al. 2009. Cancer-stellate cell interactions perpetuate the hypoxia-fibrosis cycle in pancreatic ductal adenocarcinoma. Neoplasia 11:5497–508
    [Google Scholar]
  14. 14. 
    Olumi AF, Grossfeld GD, Hayward SW et al. 1999. Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res 59:195002–11
    [Google Scholar]
  15. 15. 
    Petrova V, Annicchiarico-Petruzzelli M, Melino et al. 2018. The hypoxic tumour microenvironment. Oncogenesis 7:10
    [Google Scholar]
  16. 16. 
    Yen TW, Aardal NP, Bronner MP et al. 2002. Myofibroblasts are responsible for the desmoplastic reaction surrounding human pancreatic carcinomas. Surgery 131:2129–34
    [Google Scholar]
  17. 17. 
    Spivak-Kroizman TR, Hostetter G, Posner R et al. 2013. Hypoxia triggers hedgehog-mediated tumor-stromal interactions in pancreatic cancer. Cancer Res 73:113235–47
    [Google Scholar]
  18. 18. 
    Chau KY, Lily MA, Wu PC et al. 1992. Myofibroblasts in hepatitis B related cirrhosis and hepatocellular carcinoma. J. Clin. Pathol. 45:5446–48
    [Google Scholar]
  19. 19. 
    Kawashiri S, Tanaka A, Noguchi N et al. 2009. Significance of stromal desmoplasia and myofibroblast appearance at the invasive front in squamous cell carcinoma of the oral cavity. Head Neck 31:101346–53
    [Google Scholar]
  20. 20. 
    Najjar YG, Menk AV, Sander C et al. 2019. Tumor cell oxidative metabolism as a barrier to PD-1 blockade immunotherapy in melanoma. JCI Insight 4:5e124989
    [Google Scholar]
  21. 21. 
    Scharping NE, Menk AV, Whetstone RD et al. 2017. Efficacy of PD-1 blockade is potentiated by metformin-induced reduction of tumor hypoxia. Cancer Immunol. Res. 5:19–16
    [Google Scholar]
  22. 22. 
    Scharping NE, Rivadeneira DB, Menk AV et al. 2021. Mitochondrial stress induced by continuous stimulation under hypoxia rapidly drives T cell exhaustion. Nat. Immunol. 22:205–15
    [Google Scholar]
  23. 23. 
    Doedens AL, Phan AT, Stradner MH et al. 2013. Hypoxia-inducible factors enhance the effector responses of CD8+ T cells to persistent antigen. Nat. Immunol. 14:111173–82
    [Google Scholar]
  24. 24. 
    Palazon A, Tyrakis PA, Macias D et al. 2017. An HIF-1α/VEGF-A axis in cytotoxic T cells regulates tumor progression. Cancer Cell 32:5669–683.e5
    [Google Scholar]
  25. 25. 
    Gropper Y, Feferman T, Shalit T et al. 2017. Culturing CTLs under hypoxic conditions enhances their cytolysis and improves their anti-tumor function. Cell Rep 20:112547–55
    [Google Scholar]
  26. 26. 
    Veliça P, Cunha PP, Vojnovic N et al. 2021. Modified hypoxia-inducible factor expression in CD8+ T cells increases antitumor efficacy. Cancer Immunol. Res. 9:4401–14
    [Google Scholar]
  27. 27. 
    Li Y, Patel SP, Roszik J et al. 2018. Hypoxia-driven immunosuppressive metabolites in the tumor microenvironment: new approaches for combinational immunotherapy. Front. Immunol. 9:1591
    [Google Scholar]
  28. 28. 
    Caldwell CC, Kojima H, Lukashev D et al. 2001. Differential effects of physiologically relevant hypoxic conditions on T lymphocyte development and effector functions. J. Immunol. 167:116140–49
    [Google Scholar]
  29. 29. 
    Lukashev D, Klebanov B, Kojima H et al. 2006. Cutting edge: hypoxia-inducible factor 1α and its activation-inducible short isoform I.1 negatively regulate functions of CD4+ and CD8+ T lymphocytes. J. Immunol. 177:84962–65
    [Google Scholar]
  30. 30. 
    Zuckerberg AL, Goldberg LI, Lederman HM. 1994. Effects of hypoxia on interleukin-2 mRNA expression by T lymphocytes. Crit. Care Med. 22:2197–203
    [Google Scholar]
  31. 31. 
    Sun J, Zhang Y, Yang M et al. 2010. Hypoxia induces T-cell apoptosis by inhibiting chemokine C receptor 7 expression: the role of adenosine receptor A(2). Cell Mol. Immunol. 7:177–82
    [Google Scholar]
  32. 32. 
    Motz GT, Santoro SP, Wang LP et al. 2014. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat. Med. 20:6607–15
    [Google Scholar]
  33. 33. 
    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. PNAS 109:E2784–93
    [Google Scholar]
  34. 34. 
    Facciabene A, Peng X, Hagemann IS et al. 2011. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and Treg cells. Nature 475:226–30
    [Google Scholar]
  35. 35. 
    Nakagawa Y, Negishi Y, Shimizu M et al. 2015. Effects of extracellular pH and hypoxia on the function and development of antigen-specific cytotoxic T lymphocytes. Immunol. Lett. 167:272–86
    [Google Scholar]
  36. 36. 
    Lugini L, Matarrese P, Tinari A et al. 2006. Cannibalism of live lymphocytes by human metastatic but not primary melanoma cells. Cancer Res 66:73629–38
    [Google Scholar]
  37. 37. 
    Fischer K, Hoffmann P, Voelkl S et al. 2007. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 109:93812–19
    [Google Scholar]
  38. 38. 
    Mendler AN, Hu B, Prinz PU et al. 2012. Tumor lactic acidosis suppresses CTL function by inhibition of p38 and JNK/c-Jun activation. Int. J. Cancer 131:3633–40
    [Google Scholar]
  39. 39. 
    Haas R, Smith J, Rocher-Ros V et al. 2015. Lactate regulates metabolic and pro-inflammatory circuits in control of T cell migration and effector functions. PLOS Biol 13:7e1002202
    [Google Scholar]
  40. 40. 
    Calcinotto A, Filipazzi P, Grioni M et al. 2012. Modulation of microenvironment acidity reverses anergy in human and murine tumor-infiltrating T lymphocytes. Cancer Res 72:112746–56
    [Google Scholar]
  41. 41. 
    Angelin A, Gil-de-Gómez L, Dahiya S et al. 2017. Foxp3 reprograms T cell metabolism to function in low-glucose, high-lactate environments. Cell Metab 25:61282–93.e7
    [Google Scholar]
  42. 42. 
    Watson MJ, Vignali PDA, Mullett SJ et al. 2021. Metabolic support of tumour-infiltrating regulatory T cells by lactic acid. Nature 591:645–51
    [Google Scholar]
  43. 43. 
    Deaglio S, Dwyer KM, Gao W et al. 2007. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J. Exp. Med. 204:1257–65
    [Google Scholar]
  44. 44. 
    Kobie JJ, Shah PR, Yang L et al. 2006. T regulatory and primed uncommitted CD4 T cells express CD73, which suppresses effector CD4 T cells by converting 5′-adenosine monophosphate to adenosine. J. Immunol. 177:6780–86
    [Google Scholar]
  45. 45. 
    Lukashev D, Ohta A, Sitkovsky M. 2007. Hypoxia-dependent anti-inflammatory pathways in protection of cancerous tissues. Cancer Metastasis Rev 26:273–79
    [Google Scholar]
  46. 46. 
    Grassi F. 2020. The P2X7 receptor as regulator of T cell development and function. Front. Immunol. 11:1179
    [Google Scholar]
  47. 47. 
    Hatfield SM, Kjaergaard J, Lukashev D et al. 2015. Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci. Transl. Med. 7:277-ra230
    [Google Scholar]
  48. 48. 
    Nagaraj S, Gupta K, Pisarev V et al. 2007. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat. Med. 13:828–35
    [Google Scholar]
  49. 49. 
    Molon B, Ugel S, Del Pozzo F et al. 2011. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J. Exp. Med. 208:1949–62
    [Google Scholar]
  50. 50. 
    Scharping NE, Menk AV, Moreci RS et al. 2016. The tumor microenvironment represses T cell mitochondrial biogenesis to drive intratumoral T cell metabolic insufficiency and dysfunction. Immunity 45:2374–88 Erratum. 2016 Immunity 45:3701–3
    [Google Scholar]
  51. 51. 
    Jiang Y, Li Y, Zhu B 2015. T-cell exhaustion in the tumor microenvironment. Cell Death Dis 6:e1792
    [Google Scholar]
  52. 52. 
    Miller BC, Sen DR, Al Abosy R et al. 2019. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 20:326–36
    [Google Scholar]
  53. 53. 
    Paik ES, Kim TH, Cho YJ et al. 2020. Preclinical assessment of the VEGFR inhibitor axitinib as a therapeutic agent for epithelial ovarian cancer. Sci. Rep. 10:4904
    [Google Scholar]
  54. 54. 
    Du Four S, Maenhout SK, De Pierre K et al. 2015. Axitinib increases the infiltration of immune cells and reduces the suppressive capacity of monocytic MDSCs in an intracranial mouse melanoma model. Oncoimmunology 4:4e998107
    [Google Scholar]
  55. 55. 
    Saha D, Wakimoto H, Peters CW et al. 2018. Combinatorial effects of VEGFR kinase inhibitor axitinib and oncolytic virotherapy in mouse and human glioblastoma stem-like cell models. Clin. Cancer Res. 24:143409–22
    [Google Scholar]
  56. 56. 
    Hillman GG, Lonardo F, Hoogstra DJ et al. 2014. Axitinib improves radiotherapy in murine xenograft lung tumors. Transl. Oncol. 7:3400–9
    [Google Scholar]
  57. 57. 
    Qian BZ, Li J, Zhang H et al. 2011. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 475:7355222–25
    [Google Scholar]
  58. 58. 
    Chun E, Lavoie S, Michaud M et al. 2015. CCL2 promotes colorectal carcinogenesis by enhancing polymorphonuclear myeloid-derived suppressor cell population and function. Cell Rep 12:2244–57
    [Google Scholar]
  59. 59. 
    Inamoto S, Itatani Y, Yamamoto T et al. 2016. Loss of SMAD4 promotes colorectal cancer progression by accumulation of myeloid-derived suppressor cells through the CCL15-CCR1 chemokine axis. Clin. Cancer Res. 22:2492–501
    [Google Scholar]
  60. 60. 
    Sceneay J, Chow MT, Chen A et al. 2012. Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res 72:163906–11
    [Google Scholar]
  61. 61. 
    Chafe SC, Lou Y, Sceneay J et al. 2015. Carbonic anhydrase IX promotes myeloid-derived suppressor cell mobilization and establishment of a metastatic niche by stimulating G-CSF production. Cancer Res 75:6996–1008
    [Google Scholar]
  62. 62. 
    Sica A, Saccani A, Bottazzi B et al. 2000. Defective expression of the monocyte chemotactic protein-1 receptor CCR2 in macrophages associated with human ovarian carcinoma. J. Immunol. 164:2733–38
    [Google Scholar]
  63. 63. 
    Grimshaw MJ, Balkwill FR. 2001. Inhibition of monocyte and macrophage chemotaxis by hypoxia and inflammation—a potential mechanism. Eur. J. Immunol. 31:2480–89
    [Google Scholar]
  64. 64. 
    Casazza A, Laoui D, Wenes M et al. 2013. Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity. Cancer Cell 24:6695–709
    [Google Scholar]
  65. 65. 
    Kumar V, Cheng P, Condamine T et al. 2016. CD45 phosphatase inhibits STAT3 transcription factor activity in myeloid cells and promotes tumor-associated macrophage differentiation. Immunity 44:2303–15
    [Google Scholar]
  66. 66. 
    Colegio OR, Chu NQ, Szabo AL et al. 2014. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 513:7519559–63
    [Google Scholar]
  67. 67. 
    Movahedi K, Laoui D, Gysemans C et al. 2010. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res 70:145728–39
    [Google Scholar]
  68. 68. 
    Corzo CA, Condamine T, Lu L et al. 2010. HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J. Exp. Med. 207:112439–53
    [Google Scholar]
  69. 69. 
    Maenhout SK, Van Lint S, Emeagi PU et al. 2014. Enhanced suppressive capacity of tumor-infiltrating myeloid-derived suppressor cells compared with their peripheral counterparts. Int. J. Cancer 134:51077–90
    [Google Scholar]
  70. 70. 
    Noman MZ, Desantis G, Janji B et al. 2014. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 211:5781–90
    [Google Scholar]
  71. 71. 
    Geiger R, Rieckmann JC, Wolf T et al. 2016. L-Arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell 167:3829–42.e13
    [Google Scholar]
  72. 72. 
    Doedens AL, Stockmann C, Rubinstein MP et al. 2010. Macrophage expression of hypoxia-inducible factor-1 alpha suppresses T-cell function and promotes tumor progression. Cancer Res 70:197465–75
    [Google Scholar]
  73. 73. 
    Noman MZ, Janji B, Hu S et al. 2015. Tumor-promoting effects of myeloid-derived suppressor cells are potentiated by hypoxia-induced expression of miR-210. Cancer Res 75:183771–87
    [Google Scholar]
  74. 74. 
    Hunter FW, Wouters BG, Wilson WR. 2016. Hypoxia-activated prodrugs: paths forward in the era of personalised medicine. Br. J. Cancer 114:101071–77
    [Google Scholar]
  75. 75. 
    Meng F, Evans JW, Bhupathi D et al. 2012. Molecular and cellular pharmacology of the hypoxia-activated prodrug TH-302. Mol. Cancer Ther. 11:3740–51
    [Google Scholar]
  76. 76. 
    Jamieson SM, Tsai P, Kondratyev MK et al. 2018. Evofosfamide for the treatment of human papillomavirus-negative head and neck squamous cell carcinoma. JCI Insight 3:16e122204
    [Google Scholar]
  77. 77. 
    Jayaprakash P, Ai M, Liu A 2018. Targeted hypoxia reduction restores T cell infiltration and sensitizes prostate cancer to immunotherapy. J. Clin. Investig. 128:115137–49
    [Google Scholar]
  78. 78. 
    Hunter FW, Devaux JBL, Meng F et al. 2019. Functional CRISPR and shRNA screens identify involvement of mitochondrial electron transport in the activation of evofosfamide. Mol. Pharmacol. 95:6638–51
    [Google Scholar]
  79. 79. 
    Kaanders JH, Bussink J, van der Kogel AJ. 2002. ARCON: a novel biology-based approach in radiotherapy. Lancet Oncol 3:12728–37
    [Google Scholar]
  80. 80. 
    Hatfield SM, Kjaergaard J, Lukashev D et al. 2014. Systemic oxygenation weakens the hypoxia and hypoxia inducible factor 1α-dependent and extracellular adenosine-mediated tumor protection. J. Mol. Med. 92:121283–92
    [Google Scholar]
  81. 81. 
    Moan NL, Leung P, Ng S et al. 2018. The oxygen carrier OMX restores antitumor immunity and cures tumors as a single agent or in combination with checkpoint inhibitors in an intracranial glioblastoma mouse model. Cancer Res 78:13 Suppl.4726A Abstr. )
    [Google Scholar]
  82. 82. 
    Li L, Wang L, Li J et al. 2018. Metformin-induced reduction of CD39 and CD73 blocks myeloid-derived suppressor cell activity in patients with ovarian cancer. Cancer Res 78:71779–91
    [Google Scholar]
  83. 83. 
    Molina JR, Sun Y, Protopopova M et al. 2018. An inhibitor of oxidative phosphorylation exploits cancer vulnerability. Nat. Med. 24:71036–46
    [Google Scholar]
  84. 84. 
    Chen D, Barsoumian HB, Fischer G et al. 2020. Combination treatment with radiotherapy and a novel oxidative phosphorylation inhibitor overcomes PD-1 resistance and enhances antitumor immunity. JITC 8:e000289
    [Google Scholar]
  85. 85. 
    Yap TA, Ahnert JR, Piha-Paul SA. 2019. Phase I trial of IACS-010759 (IACS), a potent, selective inhibitor of complex I of the mitochondrial electron transport chain, in patients (pts) with advanced solid tumors. J. Clin. Oncol. 37:15 Suppl.3014
    [Google Scholar]
  86. 86. 
    Rha SY, Beom SH, Shin YG 2020. Phase I study of IM156, a novel potent biguanide oxidative phosphorylation (OXPHOS) inhibitor, in patients with advanced solid tumors. J. Clin. Oncol. 38:15 Suppl.3590
    [Google Scholar]
  87. 87. 
    Brenner A, Zuniga R, Sun JD et al. 2018. Hypoxia-activated evofosfamide for treatment of recurrent bevacizumab-refractory glioblastoma: a phase I surgical study. Neuro Oncol 20:91231–39
    [Google Scholar]
  88. 88. 
    Hegde A, Jayaprakash P, Couillault CA et al. 2021. A phase I dose-escalation study to evaluate the safety and tolerability of evofosfamide in combination with ipilimumab in advanced solid malignancies. Clin. Cancer Res. 27:113069–78
    [Google Scholar]
  89. 89. 
    Owen MR, Doran E, Halestrap AP. 2000. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem. J. 348:Pt. 3607–14
    [Google Scholar]
  90. 90. 
    Wheaton WW, Weinberg SE, Hamanaka RB et al. 2014. Metformin inhibits mitochondrial complex I of cancer cells to reduce tumorigenesis. eLife 3:e02242
    [Google Scholar]
  91. 91. 
    Madiraju AK, Erion DM, Rahimi Y et al. 2014. Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Nature 510:7506542–46
    [Google Scholar]
  92. 92. 
    Lee MS, Hsu CC, Wahlqvist ML et al. 2011. Type 2 diabetes increases and metformin reduces total, colorectal, liver and pancreatic cancer incidences in Taiwanese: a representative population prospective cohort study of 800,000 individuals. BMC Cancer 11:20
    [Google Scholar]
  93. 93. 
    Decensi A, Puntoni M, Goodwin P et al. 2010. Metformin and cancer risk in diabetic patients: a systematic review and meta-analysis. Cancer Prev. Res. 3:111451–61
    [Google Scholar]
  94. 94. 
    Saraei P, Asadi I, Kakar MA et al. 2019. The beneficial effects of metformin on cancer prevention and therapy: a comprehensive review of recent advances. Cancer Manag. Res. 11:3295–313
    [Google Scholar]
  95. 95. 
    Coyle C, Cafferty FH, Vale C et al. 2016. Metformin as an adjuvant treatment for cancer: a systematic review and meta-analysis. Ann. Oncol. 27:122184–95
    [Google Scholar]
  96. 96. 
    Afzal MZ, Dragnev K, Sarwar T et al. 2019. Clinical outcomes in non-small-cell lung cancer patients receiving concurrent metformin and immune checkpoint inhibitors. Lung Cancer Manag 8:2LMT11
    [Google Scholar]
  97. 97. 
    Afzal MZ, Mercado RR, Shirai K. 2018. Efficacy of metformin in combination with immune checkpoint inhibitors (anti-PD-1/anti-CTLA-4) in metastatic malignant melanoma. J. Immunother. Cancer 6:164
    [Google Scholar]
  98. 98. 
    Yendamuri S, Barbi J, Pabla S et al. 2019. Body mass index influences the salutary effects of metformin on survival after lobectomy for stage I NSCLC. J. Thorac. Oncol. 14:122181–87
    [Google Scholar]
  99. 99. 
    Richtig G, Hoeller C, Wolf M et al. 2018. Body mass index may predict the response to ipilimumab in metastatic melanoma: an observational multi-centre study. PLOS ONE 13:10e0204729
    [Google Scholar]
  100. 100. 
    Wang Z, Aguilar EG, Luna JI et al. 2019. Paradoxical effects of obesity on T cell function during tumor progression and PD-1 checkpoint blockade. Nat. Med. 25:1141–51
    [Google Scholar]
/content/journals/10.1146/annurev-med-060619-022830
Loading
/content/journals/10.1146/annurev-med-060619-022830
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error