J Liver Cancer.  2020 Mar;20(1):1-16. 10.17998/jlc.20.1.1.

Deciphering and Reversing Immunosuppressive Cells in the Treatment of Hepatocellular Carcinoma

Affiliations
  • 1Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
  • 2Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, USA
  • 3NCI CCR Liver Cancer Program, Bethesda, USA

Abstract

Use of immune checkpoint inhibitors (ICIs) in hepatocellular carcinoma (HCC) has been partially successful. However, most HCC patients do not respond to immunotherapy. HCC has been shown to induce several immune suppressor mechanisms in patients. These suppressor mechanisms include involvement of myeloid-derived suppressor cells, regulatory T-cells, functionally impaired dendritic cells (DCs), neutrophils, monocytes, and tumor associated macrophages. The accumulation of immunosuppressive cells may lead to an immunosuppressive tumor microenvironment as well as the dense fibrotic stroma which may contribute to immune tolerance. Our laboratory has been investigating different cellular mechanisms of immune suppression in HCC patients. In vitro as well as in vivo studies have demonstrated that abrogation of the suppressor cells enhances or unmasks tumor-specific antitumor immune responses. Two or three effective systemic therapies including ICIs and/or molecular targeted therapies and the addition of innovative combination therapies targeting immune suppressor cells may lead to increased immune recognition with a greater tumor response. We reviewed the literature for the latest research on immune suppressor cells in HCC, and here we provide a comprehensive summary of the recent studies in this field.

Keyword

Hepatocellular carcinoma; Immunotherapy; Immune checkpoint inhibitors; Immune suppression

Figure

  • Figure 1. Cross-talks among immune cells, immunosuppressive cells, stromal cells, and tumor cells in the tumor microenvironment of hepatocellular carcinoma. PDE, phosphodiesterase; STAT, signal transducer and activator of transcription; ATRA, all-trans-retinoic acid; MMP, matrix metalloproteinase; FU, fluorouracil; HSP, heat shock protein; MDSCs, myeloid-derived suppressor cells; GITR, glucocorticoid-induced TNFR family related gene; PD, programmed cell death protein; CTLA, cytotoxic T-lymphocyte-associated protein; CCR, cell cycle-related kinase; IL, interleukin; TAM, tumor associated macrophages; CCL, C-C motif chemokine ligand; CSF, stimulating factor; Treg, regulatory T-cells; PMN, polymorphonuclear; TAN, tumor-associated neutrophils; TGF, transforming growth factor; ARG, arginase; iNOS, inducible nitric oxide synthase; IDO, indoleamine 2,3-dioxygenase; DC, dendritic cells; NK, natural killer; CIK, cytokine-induced killer; TNFR, tumor necrosis factor receptor.


Reference

1. Korangy F, Ormandy LA, Bleck JS, Klempnauer J, Wilkens L, Manns MP, et al. Spontaneous tumor-specific humoral and cellular immune responses to NY-ESO-1 in hepatocellular carcinoma. Clin Cancer Res. 2004; 10:4332–4341.
2. Mizukoshi E, Nakamoto Y, Arai K, Yamashita T, Sakai A, Sakai Y, et al. Comparative analysis of various tumor-associated antigenspecific t-cell responses in patients with hepatocellular carcinoma. Hepatology. 2011; 53:1206–1216.
3. Behboudi S, Pereira SP. Alpha-fetoprotein specific CD4 and CD8 T-cell responses in patients with hepatocellular carcinoma. World J Hepatol. 2010; 2:256–260.
4. Behboudi S, Boswell S, Williams R. Cell-mediated immune responses to alpha-fetoprotein and other antigens in hepatocellular carcinoma. Liver Int. 2010; 30:521–526.
5. Zhang HG, Chen HS, Peng JR, Shang XY, Zhang J, Xing Q, et al. Specific CD8(+ )T-cell responses to HLA-A2 restricted MAGE-A3 p271-279 peptide in hepatocellular carcinoma patients without vaccination. Cancer Immunol Immunother. 2007; 56:1945–1954.
6. Arihara F, Mizukoshi E, Kitahara M, Takata Y, Arai K, Yamashita T, et al. Increase in CD14+HLA-DR -/low myeloid-derived suppressor cells in hepatocellular carcinoma patients and its impact on prognosis. Cancer Immunol Immunother. 2013; 62:1421–1430.
7. Hato T, Goyal L, Greten TF, Duda DG, Zhu AX. Immune checkpoint blockade in hepatocellular carcinoma: current progress and future directions. Hepatology. 2014; 60:1776–1782.
8. Ormandy LA, Hillemann T, Wedemeyer H, Manns MP, Greten TF, Korangy F. Increased populations of regulatory T-cells in peripheral blood of patients with hepatocellular carcinoma. Cancer Res. 2005; 65:2457–2464.
9. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018; 24:541–550.
10. Duffy AG, Ulahannan SV, Makorova-Rusher O, Rahma O, Wedemeyer H, Pratt D, et al. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J Hepatol. 2017; 66:545–551.
11. Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The immune landscape of cancer. Immunity. 2018; 48:812–830. e814.
12. Greten TF, Lai CW, Li G, Staveley-O'Carroll KF. Targeted and immune-based therapies for hepatocellular carcinoma. Gastroenterology. 2019; 156:510–524.
13. Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015; 12:453–457.
14. Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 2017; 18:220.
15. Spitzer MH, Nolan GP. Mass cytometry: single cells, many features. Cell. 2016; 165:780–791.
16. von Boehmer H. Mechanisms of suppression by suppressor T-cells. Nat Immunol. 2005; 6:338–344.
17. Gavin MA, Torgerson TR, Houston E, DeRoos P, Ho WY, Stray-Pedersen A, et al. Single-cell analysis of normal and FOXP3-mutant human T-cells: FOXP3 expression without regulatory T-cell development. Proc Natl Acad Sci U S A. 2006; 103:6659–6664.
18. Greten TF, Manns MP, Korangy F. Myeloid derived suppressor cells in human diseases. Int Immunopharmacol. 2011; 11:802–807.
19. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 2012; 12:253–268.
20. Youn JI, Collazo M, Shalova IN, Biswas SK, Gabrilovich DI. Characterization of the nature of granulocytic myeloid-derived suppressor cells in tumor-bearing mice. J Leukoc Biol. 2012; 91:167–181.
21. Gabrilovich DI. Myeloid-derived suppressor cells. Cancer Immunol Res. 2017; 5:3–8.
22. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009; 9:162–174.
23. Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Greten TF, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun. 2016; 7:12150.
24. Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008; 8:618–631.
25. Qu X, Zhuang G, Yu L, Meng G, Ferrara N. Induction of Bv8 expression by granulocyte colony-stimulating factor in CD11b+Gr1+ cells: key role of Stat3 signaling. J Biol Chem. 2012; 287:19574–19584.
26. Shojaei F, Wu X, Malik AK, Zhong C, Baldwin ME, Schanz S, et al. Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat Biotechnol. 2007; 25:911–920.
27. Shojaei F, Wu X, Qu X, Kowanetz M, Yu L, Tan M, et al. G-CSFinitiated myeloid cell mobilization and angiogenesis mediate tumor refractoriness to anti-VEGF therapy in mouse models. Proc Natl Acad Sci U S A. 2009; 106:6742–6747.
28. Ahn GO, Brown JM. Matrix metalloproteinase-9 is required for tumor vasculogenesis but not for angiogenesis: role of bone marrow-derived myelomonocytic cells. Cancer Cell. 2008; 13:193–205.
29. Yang L, DeBusk LM, Fukuda K, Fingleton B, Green-Jarvis B, Shyr Y, et al. Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell. 2004; 6:409–421.
30. Chiu DK, Tse AP, Xu IM, Di Cui J, Lai RK, Li LL, et al. Hypoxia inducible factor HIF-1 promotes myeloid-derived suppressor cells accumulation through ENTPD2/CD39L1 in hepatocellular carcinoma. Nat Commun. 2017; 8:517.
31. Zhou J, Liu M, Sun H, Feng Y, Xu L, Chan AWH, et al. Hepatomaintrinsic CCRK inhibition diminishes myeloid-derived suppressor cell immunosuppression and enhances immune-checkpoint blockade efficacy. Gut. 2018; 67:931–944.
32. Greten TF, Korangy F. CDK20 inhibition and immune checkpoint blockade: bringing cancer biology and tumour immunology together to develop novel treatment options for HCC. Gut. 2018; 67:783–784.
33. Condamine T, Dominguez GA, Youn JI, Kossenkov AV, Mony S, Alicea-Torres K, et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci Immunol. 2016; 1:aaf8943.
34. Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Krüger C, Manns MP, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+) Foxp3(+) T-cells. Gastroenterology. 2008; 135:234–243.
35. Hoechst B, Gamrekelashvili J, Manns MP, Greten TF, Korangy F. Plasticity of human Th17 cells and iTregs is orchestrated by different subsets of myeloid cells. Blood. 2011; 117:6532–6541.
36. Cai L, Zhang Z, Zhou L, Wang H, Fu J, Zhang S, et al. Functional impairment in circulating and intrahepatic NK cells and relative mechanism in hepatocellular carcinoma patients. Clin Immunol. 2008; 129:428–437.
37. Hoechst B, Voigtlaender T, Ormandy L, Gamrekelashvili J, Zhao F, Wedemeyer H, et al. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology. 2009; 50:799–807.
38. Yu SJ, Ma C, Heinrich B, Brown ZJ, Sandhu M, Zhang Q, et al. Targeting the crosstalk between cytokine-induced killer cells and myeloid-derived suppressor cells in hepatocellular carcinoma. J Hepatol. 2019; 70:449–457.
39. Califano JA, Khan Z, Noonan KA, Rudraraju L, Zhang Z, Wang H, et al. Tadalafil augments tumor specific immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res. 2015; 21:30–38.
40. Serafini P, Meckel K, Kelso M, Noonan K, Califano J, Koch W, et al. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J Exp Med. 2006; 203:2691–2702.
41. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T-cells and immune tolerance. Cell. 2008; 133:775–787.
42. Dieckmann D, Bruett CH, Ploettner H, Lutz MB, Schuler G. Human CD4(+)CD25(+) regulatory, contact-dependent T-cells induce interleukin 10-producing, contact-independent type 1-like regulatory T-cells [corrected]. J Exp Med. 2002; 196:247–253.
43. Shevach EM. CD4+ CD25+ suppressor T-cells: more questions than answers. Nat Rev Immunol. 2002; 2:389–400.
44. Chen ML, Pittet MJ, Gorelik L, Flavell RA, Weissleder R, von Boehmer H, et al. Regulatory T-cells suppress tumor-specific CD8 T-cell cytotoxicity through TGF-beta signals in vivo. Proc Natl Acad Sci U S A. 2005; 102:419–424.
45. Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T-cells: mechanisms of differentiation and function. Annu Rev Immunol. 2012; 30:531–564.
46. Sprinzl MF, Galle PR. Immune control in hepatocellular carcinoma development and progression: role of stromal cells. Semin Liver Dis. 2014; 34:376–388.
47. Chen KJ, Lin SZ, Zhou L, Xie HY, Zhou WH, Taki-Eldin A, et al. Selective recruitment of regulatory T-cell through CCR6-CCL20 in hepatocellular carcinoma fosters tumor progression and predicts poor prognosis. PLoS One. 2011; 6:e24671.
48. Wiedemann GM, Knott MM, Vetter VK, Rapp M, Haubner S, Fesseler J, et al. Cancer cell-derived IL-1α induces CCL22 and the recruitment of regulatory T-cells. Oncoimmunology. 2016; 5:e1175794.
49. Baron U, Floess S, Wieczorek G, Baumann K, Grützkau A, Dong J, et al. DNA demethylation in the human FOXP3 locus discriminates regulatory T-cells from activated FOXP3(+) conventional T-cells. Eur J Immunol. 2007; 37:2378–2389.
50. Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, et al. Specific recruitment of regulatory T-cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004; 10:942–949.
51. Hiraoka N, Onozato K, Kosuge T, Hirohashi S. Prevalence of FOXP3+ regulatory T-cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions. Clin Cancer Res. 2006; 12:5423–5434.
52. Badoual C, Hans S, Rodriguez J, Peyrard S, Klein C, Agueznay Nel H, et al. Prognostic value of tumor-infiltrating CD4+ T-cell subpopulations in head and neck cancers. Clin Cancer Res. 2006; 12:465–472.
53. Unitt E, Rushbrook SM, Marshall A, Davies S, Gibbs P, Morris LS, et al. Compromised lymphocytes infiltrate hepatocellular carcinoma: the role of T-regulatory cells. Hepatology. 2005; 41:722–730.
54. Yang XH, Yamagiwa S, Ichida T, Matsuda Y, Sugahara S, Watanabe H, et al. Increase of CD4+ CD25+ regulatory T-cells in the liver of patients with hepatocellular carcinoma. J Hepatol. 2006; 45:254–262.
55. Shen X, Li N, Li H, Zhang T, Wang F, Li Q. Increased prevalence of regulatory T-cells in the tumor microenvironment and its correlation with TNM stage of hepatocellular carcinoma. J Cancer Res Clin Oncol. 2010; 136:1745–1754.
56. Kobayashi N, Hiraoka N, Yamagami W, Ojima H, Kanai Y, Kosuge T, et al. FOXP3+ regulatory T-cells affect the development and progression of hepatocarcinogenesis. Clin Cancer Res. 2007; 13:902–911.
57. Fu J, Xu D, Liu Z, Shi M, Zhao P, Fu B, et al. Increased regulatory T-cells correlate with CD8 T-cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology. 2007; 132:2328–2339.
58. Zheng C, Zheng L, Yoo JK, Guo H, Zhang Y, Guo X, et al. Landscape of infiltrating T-cells in liver cancer revealed by single-cell sequencing. Cell. 2017; 169:1342–1356. e1316.
59. Gao Q, Qiu SJ, Fan J, Zhou J, Wang XY, Xiao YS, et al. Intratumoral balance of regulatory and cytotoxic T-cells is associated with prognosis of hepatocellular carcinoma after resection. J Clin Oncol. 2007; 25:2586–2593.
60. Zhou J, Ding T, Pan W, Zhu LY, Li L, Zheng L. Increased intratumoral regulatory T-cells are related to intratumoral macrophages and poor prognosis in hepatocellular carcinoma patients. Int J Cancer. 2009; 125:1640–1648.
61. Yang ZQ, Yang ZY, Zhang LD, Wang SG, Ma KS, et al. Increased liver-infiltrating CD8+FoxP3+ regulatory T-cells are associated with tumor stage in hepatocellular carcinoma patients. Hum Immunol. 2010; 71:1180–1186.
62. Greten TF, Ormandy LA, Fikuart A, Höchst B, Henschen S, Hörning M, et al. Low-dose cyclophosphamide treatment impairs regulatory T-cells and unmasks AFP-specific CD4+ T-cell responses in patients with advanced HCC. J Immunother. 2010; 33:211–218.
63. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014; 41:49–61.
64. Movahedi K, Van Ginderachter JA. The ontogeny and microenvironmental regulation of tumor-associated macrophages. Antioxid Redox Signal. 2016; 25:775–791.
65. Tacke F, Luedde T, Trautwein C. Inflammatory pathways in liver homeostasis and liver injury. Clin Rev Allergy Immunol. 2009; 36:4–12.
66. Scott CL, Zheng F, De Baetselier P, Martens L, Saeys Y, De Prijck S, et al. Bone marrow-derived monocytes give rise to self-renewing and fully differentiated Kupffer cells. Nat Commun. 2016; 7:10321.
67. Beattie L, Sawtell A, Mann J, Frame TCM, Teal B, de Labastida Rivera F, et al. Bone marrow-derived and resident liver macrophages display unique transcriptomic signatures but similar biological functions. J Hepatol. 2016; 65:758–768.
68. Sica A, Invernizzi P, Mantovani A. Macrophage plasticity and polarization in liver homeostasis and pathology. Hepatology. 2014; 59:2034–2042.
69. Devisscher L, Scott CL, Lefere S, Raevens S, Bogaerts E, Paridaens A, et al. Non-alcoholic steatohepatitis induces transient changes within the liver macrophage pool. Cell Immunol. 2017; 322:74–83.
70. Degroote H, Van Dierendonck A, Geerts A, Van Vlierberghe H, Devisscher L. Preclinical and clinical therapeutic strategies affecting tumor-associated macrophages in hepatocellular carcinoma. J Immunol Res. 2018; 2018:7819520.
71. Aravalli RN. Role of innate immunity in the development of hepatocellular carcinoma. World J Gastroenterol. 2013; 19:7500–7514.
72. Yang JD, Nakamura I, Roberts LR. The tumor microenvironment in hepatocellular carcinoma: current status and therapeutic targets. Semin Cancer Biol. 2011; 21:35–43.
73. Wu J, Li J, Salcedo R, Mivechi NF, Trinchieri G, Horuzsko A. The proinflammatory myeloid cell receptor TREM-1 controls Kupffer cell activation and development of hepatocellular carcinoma. Cancer Res. 2012; 72:3977–3986.
74. Wu K, Kryczek I, Chen L, Zou W, Welling TH. Kupffer cell suppression of CD8+ T-cells in human hepatocellular carcinoma is mediated by B7-H1/programmed death-1 interactions. Cancer Res. 2009; 69:8067–8075.
75. Prieto J, Melero I, Sangro B. Immunological landscape and immunotherapy of hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2015; 12:681–700.
76. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science. 2011; 331:1565–1570.
77. Laoui D, Van Overmeire E, Movahedi K, Van den Bossche J, Schouppe E, Mommer C, et al. Mononuclear phagocyte heterogeneity in cancer: different subsets and activation states reaching out at the tumor site. Immunobiology. 2011; 216:1192–1202.
78. Ostrand-Rosenberg S, Sinha P, Beury DW, Clements VK. Cross-talk between myeloid-derived suppressor cells (MDSC), macrophages, and dendritic cells enhances tumor-induced immune suppression. Semin Cancer Biol. 2012; 22:275–281.
79. Lin EY, Li JF, Gnatovskiy L, Deng Y, Zhu L, Grzesik DA, et al. Macrophages regulate the angiogenic switch in a mouse model of breast cancer. Cancer Res. 2006; 66:11238–11246.
80. Qian B, Deng Y, Im JH, Muschel RJ, Zou Y, Li J, et al. A distinct macrophage population mediates metastatic breast cancer cell extravasation, establishment and growth. PLoS One. 2009; 4:e6562.
81. De Palma M, Venneri MA, Galli R, Sergi Sergi L, Politi LS, Sampaolesi M, et al. Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell. 2005; 8:211–226.
82. Murdoch C, Tazzyman S, Webster S, Lewis CE. Expression of Tie-2 by human monocytes and their responses to angiopoietin-2. J Immunol. 2007; 178:7405–7411.
83. Venneri MA, De Palma M, Ponzoni M, Pucci F, Scielzo C, Zonari E, et al. Identification of proangiogenic TIE2-expressing monocytes (TEMs) in human peripheral blood and cancer. Blood. 2007; 109:5276–5285.
84. Zhao F, Korangy F, Greten TF. Cellular immune suppressor mechanisms in patients with hepatocellular carcinoma. Dig Dis. 2012; 30:477–482.
85. Zhou SL, Zhou ZJ, Hu ZQ, Huang XW, Wang Z, Chen EB, et al. Tumor- associated neutrophils recruit macrophages and T-regulatory cells to promote progression of hepatocellular carcinoma and resistance to sorafenib. Gastroenterology. 2016; 150:1646–1658. e1617.
86. Kuang DM, Zhao Q, Wu Y, Peng C, Wang J, Xu Z, et al. Peritumoral neutrophils link inflammatory response to disease progression by fostering angiogenesis in hepatocellular carcinoma. J Hepatol. 2011; 54:948–955.
87. Zhou SL, Dai Z, Zhou ZJ, Wang XY, Yang GH, Wang Z, et al. Overexpression of CXCL5 mediates neutrophil infiltration and indicates poor prognosis for hepatocellular carcinoma. Hepatology. 2012; 56:2242–2254.
88. Kuang DM, Zhao Q, Peng C, Xu J, Zhang JP, Wu C, et al. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med. 2009; 206:1327–1337.
89. Kuang DM, Peng C, Zhao Q, Wu Y, Chen MS, Zheng L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma promote expansion of memory T helper 17 cells. Hepatology. 2010; 51:154–164.
90. Kuang DM, Peng C, Zhao Q, Wu Y, Zhu LY, Wang J, et al. Tumoractivated monocytes promote expansion of IL-17-producing CD8+ T-cells in hepatocellular carcinoma patients. J Immunol. 2010; 185:1544–1549.
91. Zhang JP, Yan J, Xu J, Pang XH, Chen MS, Li L, et al. Increased intratumoral IL-17-producing cells correlate with poor survival in hepatocellular carcinoma patients. J Hepatol. 2009; 50:980–989.
92. Zhao F, Hoechst B, Gamrekelashvili J, Ormandy LA, Voigtländer T, Wedemeyer H, et al. Human CCR4+ CCR6+ Th17 cells suppress autologous CD8+ T-cell responses. J Immunol. 2012; 188:6055–6062.
93. Höchst B, Schildberg FA, Böttcher J, Metzger C, Huss S, Türler A, et al. Liver sinusoidal endothelial cells contribute to CD8 T-cell tolerance toward circulating carcinoembryonic antigen in mice. Hepatology. 2012; 56:1924–1933.
94. Carambia A, Freund B, Schwinge D, Heine M, Laschtowitz A, Huber S, et al. TGF-β-dependent induction of CD4+CD25+Foxp3+ Tregs by liver sinusoidal endothelial cells. J Hepatol. 2014; 61:594–599.
95. Ji J, Eggert T, Budhu A, Forgues M, Takai A, Dang H, et al. Hepatic stellate cell and monocyte interaction contributes to poor prognosis in hepatocellular carcinoma. Hepatology. 2015; 62:481–495.
96. Yu MC, Chen CH, Liang X, Wang L, Gandhi CR, Fung JJ, et al. Inhibition of T-cell responses by hepatic stellate cells via B7-H1-mediated T-cell apoptosis in mice. Hepatology. 2004; 40:1312–1321.
97. Wesolowski R, Markowitz J, Carson WE 3rd. Myeloid derived suppressor cells - a new therapeutic target in the treatment of cancer. J Immunother Cancer. 2013; 1:10.
98. Orillion A, Hashimoto A, Damayanti N, Shen L, Adelaiye-Ogala R, Arisa S, et al. Entinostat neutralizes myeloid-derived suppressor cells and enhances the antitumor effect of PD-1 inhibition in murine models of lung and renal cell carcinoma. Clin Cancer Res. 2017; 23:5187–5201.
99. Yeo W, Chung HC, Chan SL, Wang LZ, Lim R, Picus J, et al. Epigenetic therapy using belinostat for patients with unresectable hepatocellular carcinoma: a multicenter phase I/II study with biomarker and pharmacokinetic analysis of tumors from patients in the Mayo Phase II Consortium and the Cancer Therapeutics Research Group. J Clin Oncol. 2012; 30:3361–3367.
100. Bitzer M, Horger M, Giannini EG, Ganten TM, Wörns MA, Siveke JT, et al. Resminostat plus sorafenib as second-line therapy of advanced hepatocellular carcinoma - The SHELTER study. J Hepatol. 2016; 65:280–288.
101. Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J Immunol. 2001; 166:5398–5406.
102. Ugel S, Delpozzo F, Desantis G, Papalini F, Simonato F, Sonda N, et al. Therapeutic targeting of myeloid-derived suppressor cells. Curr Opin Pharmacol. 2009; 9:470–481.
103. Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, Hackett NR, et al. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell. 2002; 109:625–637.
104. Le HK, Graham L, Cha E, Morales JK, Manjili MH, Bear HD. Gemcitabine directly inhibits myeloid derived suppressor cells in BALB/c mice bearing 4T1 mammary carcinoma and augments expansion of T-cells from tumor-bearing mice. Int Immunopharmacol. 2009; 9:900–909.
105. Abou-Alfa GK, Meyer T, Cheng AL, El-Khoueiry AB, Rimassa L, Ryoo BY, et al. Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. N Engl J Med. 2018; 379:54–63.
106. Lu X, Horner JW, Paul E, Shang X, Troncoso P, Deng P, et al. Effective combinatorial immunotherapy for castration-resistant prostate cancer. Nature. 2017; 543:728–732.
107. Condamine T, Kumar V, Ramachandran IR, Youn JI, Celis E, Finnberg N, et al. ER stress regulates myeloid-derived suppressor cell fate through TRAIL-R-mediated apoptosis. J Clin Invest. 2014; 124:2626–2639.
108. Dominguez GA, Condamine T, Mony S, Hashimoto A, Wang F, Liu Q, et al. Selective targeting of myeloid-derived suppressor cells in cancer patients using DS-8273a, an agonistic TRAIL-R2 antibody. Clin Cancer Res. 2017; 23:2942–2950.
109. Ugel S, De Sanctis F, Mandruzzato S, Bronte V. Tumor-induced myeloid deviation: when myeloid-derived suppressor cells meet tumor-associated macrophages. J Clin Invest. 2015; 125:3365–3376.
110. Lesokhin AM, Hohl TM, Kitano S, Cortez C, Hirschhorn-Cymerman D, Avogadri F, et al. Monocytic CCR2(+) myeloid-derived suppressor cells promote immune escape by limiting activated CD8 T-cell infiltration into the tumor microenvironment. Cancer Res. 2012; 72:876–886.
111. Karin N, Razon H. The role of CCR5 in directing the mobilization and biological function of CD11b(+)Gr1(+)Ly6C(low) polymorphonuclear myeloid cells in cancer. Cancer Immunol Immunother. 2018; 67:1949–1953.
112. Arce Vargas F, Furness AJS, Solomon I, Joshi K, Mekkaoui L, Lesko MH, et al. Fc-Optimized Anti-CD25 depletes tumor-infiltrating regulatory T-cells and synergizes with PD-1 blockade to eradicate established tumors. Immunity. 2017; 46:577–586.
113. Zhou S, Chen L, Qin J, Li R, Tao H, Zhen Z, et al. Depletion of CD4+ CD25+ regulatory T-cells promotes CCL21-mediated antitumor immunity. PLoS One. 2013; 8:e73952.
114. Kavanagh B, O'Brien S, Lee D, Hou Y, Weinberg V, Rini B, et al. CTLA4 blockade expands FoxP3+ regulatory and activated effector CD4+ T-cells in a dose-dependent fashion. Blood. 2008; 112:1175–1183.
115. Maker AV, Attia P, Rosenberg SA. Analysis of the cellular mechanism of antitumor responses and autoimmunity in patients treated with CTLA-4 blockade. J Immunol. 2005; 175:7746–7754.
116. O'Mahony D, Morris JC, Quinn C, Gao W, Wilson WH, Gause B, et al. A pilot study of CTLA-4 blockade after cancer vaccine failure in patients with advanced malignancy. Clin Cancer Res. 2007; 13:958–964.
117. Ralph C, Elkord E, Burt DJ, O'Dwyer JF, Austin EB, Stern PL, et al. Modulation of lymphocyte regulation for cancer therapy: a phase II trial of tremelimumab in advanced gastric and esophageal adenocarcinoma. Clin Cancer Res. 2010; 16:1662–1672.
118. Weber JS, Hamid O, Chasalow SD, Wu DY, Parker SM, Galbraith S, et al. Ipilimumab increases activated T-cells and enhances humoral immunity in patients with advanced melanoma. J Immunother. 2012; 35:89–97.
119. Simpson TR, Li F, Montalvo-Ortiz W, Sepulveda MA, Bergerhoff K, Arce F, et al. Fc-dependent depletion of tumor-infiltrating regulatory T-cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med. 2013; 210:1695–1710.
120. Ko K, Yamazaki S, Nakamura K, Nishioka T, Hirota K, Yamaguchi T, et al. Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T-cells. J Exp Med. 2005; 202:885–891.
121. van Beek AA, Zhou G, Doukas M, Boor PPC, Noordam L, Mancham S, et al. GITR ligation enhances functionality of tumorinfiltrating T-cells in hepatocellular carcinoma. Int J Cancer. 2019; 145:1111–1124.
122. Xie K, Xu L, Wu H, Liao H, Luo L, Liao M, et al. OX40 expression in hepatocellular carcinoma is associated with a distinct immune microenvironment, specific mutation signature, and poor prognosis. Oncoimmunology. 2018; 7:e1404214.
123. Piconese S, Valzasina B, Colombo MP. OX40 triggering blocks suppression by regulatory T-cells and facilitates tumor rejection. J Exp Med. 2008; 205:825–839.
124. Lesokhin AM, Callahan MK, Postow MA, Wolchok JD. On being less tolerant: enhanced cancer immunosurveillance enabled by targeting checkpoints and agonists of T-cell activation. Sci Transl Med. 2015; 7:280. sr1.
125. Taylor NA, Vick SC, Iglesia MD, Brickey WJ, Midkiff BR, McKinnon KP, et al. Treg depletion potentiates checkpoint inhibition in claudin-low breast cancer. J Clin Invest. 2017; 127:3472–3483.
126. Mailloux AW, Young MR. Regulatory T-cell trafficking: from thymic development to tumor-induced immune suppression. Crit Rev Immunol. 2010; 30:435–447.
127. Ondondo B, Jones E, Godkin A, Gallimore A. Home sweet home: the tumor microenvironment as a haven for regulatory T-cells. Front Immunol. 2013; 4:197.
128. Kurose K, Ohue Y, Sato E, Yamauchi A, Eikawa S, Isobe M, et al. Increase in activated treg in TIL in lung cancer and in vitro depletion of treg by ADCC using an antihuman CCR4 mAb (KM2760). J Thorac Oncol. 2015; 10:74–83.
129. Ishida T, Ishii T, Inagaki A, Yano H, Komatsu H, Iida S, et al. Specific recruitment of CC chemokine receptor 4-positive regulatory T-cells in Hodgkin lymphoma fosters immune privilege. Cancer Res. 2006; 66:5716–5722.
130. Cheng X, Wu H, Jin ZJ, Ma D, Yuen S, Jing XQ, et al. Up-regulation of chemokine receptor CCR4 is associated with human hepatocellular carcinoma malignant behavior. Sci Rep. 2017; 7:12362.
131. Ishida T, Joh T, Uike N, Yamamoto K, Utsunomiya A, Yoshida S, et al. Defucosylated anti-CCR4 monoclonal antibody (KW-0761) for relapsed adult T-cell leukemia-lymphoma: a multicenter phase II study. J Clin Oncol. 2012; 30:837–842.
132. Redjimi N, Raffin C, Raimbaud I, Pignon P, Matsuzaki J, Odunsi K, et al. CXCR3+ T regulatory cells selectively accumulate in human ovarian carcinomas to limit type I immunity. Cancer Res. 2012; 72:4351–4360.
133. Muraoka RS, Dumont N, Ritter CA, Dugger TC, Brantley DM, Chen J, et al. Blockade of TGF-beta inhibits mammary tumor cell viability, migration, and metastases. J Clin Invest. 2002; 109:1551–1559.
134. Gorelik L, Flavell RA. Immune-mediated eradication of tumors through the blockade of transforming growth factor-beta signaling in T-cells. Nat Med. 2001; 7:1118–1122.
135. Sun Z, Fourcade J, Pagliano O, Chauvin JM, Sander C, Kirkwood JM, et al. IL10 and PD-1 Cooperate to Limit the Activity of Tumor-Specific CD8+ T Cells. Cancer Res. 2015; 75:1635–1644.
136. Vicari AP, Chiodoni C, Vaure C, Aït-Yahia S, Dercamp C, Matsos F, et al. Reversal of tumor-induced dendritic cell paralysis by CpG immunostimulatory oligonucleotide and anti-interleukin 10 receptor antibody. J Exp Med. 2002; 196:541–549.
137. Fu YP, Yi Y, Cai XY, Sun J, Ni XC, He HW, et al. Overexpression of interleukin-35 associates with hepatocellular carcinoma aggressiveness and recurrence after curative resection. Br J Cancer. 2016; 114:767–776.
138. Turnis ME, Sawant DV, Szymczak-Workman AL, Andrews LP, Delgoffe GM, Yano H, et al. Interleukin-35 limits anti-tumor immunity. Immunity. 2016; 44:316–329.
139. Wang B, Li Q, Qin L, Zhao S, Wang J, Chen X. Transition of tumorassociated macrophages from MHC class II(hi) to MHC class II(low) mediates tumor progression in mice. BMC Immunol. 2011; 12:43.
140. Li X, Yao W, Yuan Y, Chen P, Li B, Li J, et al. Targeting of tumourinfiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma. Gut. 2017; 66:157–167.
141. Yao W, Ba Q, Li X, Li H, Zhang S, Yuan Y, et al. A natural CCR2 antagonist relieves tumor-associated macrophage-mediated immunosuppression to produce a therapeutic effect for liver cancer. EBioMedicine. 2017; 22:58–67.
142. Teng KY, Han J, Zhang X, Hsu SH, He S, Wani NA, et al. Blocking the CCL2-CCR2 axis using CCL2-neutralizing antibody is an effective therapy for hepatocellular cancer in a mouse model. Mol Cancer Ther. 2017; 16:312–322.
143. Tan HY, Wang N, Man K, Tsao SW, Che CM, Feng Y. Autophagyinduced RelB/p52 activation mediates tumour-associated macrophage repolarisation and suppression of hepatocellular carcinoma by natural compound baicalin. Cell Death Dis. 2015; 6:e1942.
144. Ao JY, Zhu XD, Chai ZT, Cai H, Zhang YY, Zhang KZ, et al. Colonystimulating factor 1 receptor blockade inhibits tumor growth by altering the polarization of tumor-associated macrophages in hepatocellular carcinoma. Mol Cancer Ther. 2017; 16:1544–1554.
145. Wan S, Zhao E, Kryczek I, Vatan L, Sadovskaya A, Ludema G, et al. Tumor-associated macrophages produce interleukin 6 and signal via STAT3 to promote expansion of human hepatocellular carcinoma stem cells. Gastroenterology. 2014; 147:1393–1404.
146. Kato Y, Tabata K, Kimura T, Yachie-Kinoshita A, Ozawa Y, Yamada K, et al. Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T-cells through reduction of tumor-associated macrophage and activation of the interferon pathway. PLoS One. 2019; 14:e0212513.
147. Kudo M. Targeted and immune therapies for hepatocellular carcinoma: predictions for 2019 and beyond. World J Gastroenterol. 2019; 25:789–807.
148. Voron T, Colussi O, Marcheteau E, Pernot S, Nizard M, Pointet AL, et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T-cells in tumors. J Exp Med. 2015; 212:139–148.
149. Lapeyre-Prost A, Terme M, Pernot S, Pointet AL, Voron T, Tartour E, et al. Immunomodulatory activity of VEGF in cancer. Int Rev Cell Mol Biol. 2017; 330:295–342.
Full Text Links
  • JLC
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