Immune Netw.  2020 Feb;20(1):e6. 10.4110/in.2020.20.e6.

IL-17-Producing Cells in Tumor Immunity: Friends or Foes?

Affiliations
  • 1Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea. yeonseok@snu.ac.kr
  • 2BK21 Plus Program, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea.

Abstract

IL-17 is produced by RAR-related orphan receptor gamma t (RORγt)-expressing cells including Th17 cells, subsets of γδT cells and innate lymphoid cells (ILCs). The biological significance of IL-17-producing cells is well-studied in contexts of inflammation, autoimmunity and host defense against infection. While most of available studies in tumor immunity mainly focused on the role of T-bet-expressing cells, including cytotoxic CD8⁺ T cells and NK cells, and their exhaustion status, the role of IL-17-producing cells remains poorly understood. While IL-17-producing T-cells were shown to be anti-tumorigenic in adoptive T-cell therapy settings, mice deficient in type 17 genes suggest a protumorigenic potential of IL-17-producing cells. This review discusses the features of IL-17-producing cells, of both lymphocytic and myeloid origins, as well as their suggested pro- and/or anti-tumorigenic functions in an organ-dependent context. Potential therapeutic approaches targeting these cells in the tumor microenvironment will also be discussed.

Keyword

Tumor microenvironment; Th17 cells; Interleukin-17; T-lymphocytes

MeSH Terms

Animals
Autoimmunity
Child
Child, Orphaned
Friends*
Humans
Inflammation
Interleukin-17
Killer Cells, Natural
Lymphocytes
Mice
T-Lymphocytes
Th17 Cells
Tumor Microenvironment
Interleukin-17

Figure

  • Figure 1. Type17 compartments in the tumor microenvironment. The tissue-specific niche, and the stage of cancer progression dictate the heterogeneous composition of type 17 compartment in the tumor microenvironment. Both innate and adaptive arms of the immune system are capable of producing IL-17. Studies involving both CD4+ Th17 and CD8+ Tc17 T-cells reported protumorigenic or anti-tumorigenic roles. Anti-tumorigenic Th/c17 cells co-secrete IFN-γ, and exhibit type 1–17 hybrid phenotypes that lead to enhanced dendritic cell infiltration and tumor-Ag presentation, type 1-helper function or Tc1 conversion in the case of Tc17 cells under the appropriate cytokine stimuli. However, an increasing amount of evidence illustrate the protumorigenic mechanisms (highlighted in blue) of type 17 cells within the immunosuppressive endogenous microenvironment within the growing tumor. Largely, they work by two broad mechanisms: 1) the IL-17 mediated recruitment of immunosuppressive myeloid compartment, either directly by type 17 cells or indirectly by cancer cells in a chemokine dependent manner, 2) cancer intrinsic IL-17 signaling, leading to enhanced cancer cell survival, EMT and angiogenesis promotion. EMT, epithelial–mesenchymal transition; TAM, tumor-associated macrophage.

  • Figure 2. Developing strategies to utilize type 17 axis for anti-cancer therapy. Currently developing strategies to transform the type 17 landscape within the tumor microenvironment for successful cancer elimination are illustrated. TAM, tumor-associated macrophage.


Cited by  1 articles

Coalition Forces of Immunologists and Oncologists for Defeating Cancer
Eui-Cheol Shin
Immune Netw. 2020;20(1):.    doi: 10.4110/in.2020.20.e1.


Reference

References

1. Pelletier M, Maggi L, Micheletti A, Lazzeri E, Tamassia N, Costantini C, Cosmi L, Lunardi C, Annunziato F, Romagnani S, et al. Evidence for a cross-talk between human neutrophils and Th17 cells. Blood. 2010; 115:335–343.
Article
2. Mangan PR, Harrington LE, O'Quinn DB, Helms WS, Bullard DC, Elson CO, Hatton RD, Wahl SM, Schoeb TR, Weaver CT. Transforming growth factor-β induces development of the T H17 lineage. Nature. 2006; 441:231–234.
3. Chung Y, Chang SH, Martinez GJ, Yang XO, Nurieva R, Kang HS, Ma L, Watowich SS, Jetten AM, Tian Q, et al. Critical regulation of early Th17 cell differentiation by interleukin-1 signaling. Immunity. 2009; 30:576–587.
Article
4. McGeachy MJ, Chen Y, Tato CM, Laurence A, Joyce-Shaikh B, Blumenschein WM, McClanahan TK, O'Shea JJ, Cua DJ. The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo. Nat Immunol. 2009; 10:314–324.
5. Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ, Littman DR. The orphan nuclear receptor RORγ t directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006; 126:1121–1133.
6. Chung Y, Yang X, Chang SH, Ma L, Tian Q, Dong C. Expression and regulation of IL-22 in the IL-17-producing CD4+ T lymphocytes. Cell Res. 2006; 16:902–907.
7. Yang XO, Chang SH, Park H, Nurieva R, Shah B, Acero L, Wang YH, Schluns KS, Broaddus RR, Zhu Z, et al. Regulation of inflammatory responses by IL-17F. J Exp Med. 2008; 205:1063–1075.
Article
8. El-Behi M, Ciric B, Dai H, Yan Y, Cullimore M, Safavi F, Zhang GX, Dittel BN, Rostami A. The encephalitogenicity of T H17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF. Nat Immunol. 2011; 12:568–575.
9. Codarri L, Gyülvészi G, Tosevski V, Hesske L, Fontana A, Magnenat L, Suter T, Becher B. RORγ t drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat Immunol. 2011; 12:560–567.
Article
10. Kim BS, Park YJ, Chung Y. Targeting IL-17 in autoimmunity and inflammation. Arch Pharm Res. 2016; 39:1537–1547.
Article
11. Zhong W, Li Q. Rituximab or irradiation promotes IL-17 secretion and thereby induces resistance to rituximab or irradiation. Cell Mol Immunol. 2017; 14:1020–1022.
Article
12. Muranski P, Boni A, Antony PA, Cassard L, Irvine KR, Kaiser A, Paulos CM, Palmer DC, Touloukian CE, Ptak K, et al. Tumor-specific Th17-polarized cells eradicate large established melanoma. Blood. 2008; 112:362–373.
Article
13. Kryczek I, Wei S, Szeliga W, Vatan L, Zou W. Endogenous IL-17 contributes to reduced tumor growth and metastasis. Blood. 2009; 114:357–359.
Article
14. Sethi V, Kurtom S, Tarique M, Lavania S, Malchiodi Z, Hellmund L, Zhang L, Sharma U, Giri B, Garg B, et al. Gut microbiota promotes tumor growth in mice by modulating immune response. Gastroenterology. 2018; 155:33–37. e6.
Article
15. Kuang DM, Peng C, Zhao Q, Wu Y, Zhu LY, Wang J, Yin XY, Li L, Zheng L. Tumor-activated monocytes promote expansion of IL-17-producing CD8+ T cells in hepatocellular carcinoma patients. J Immunol. 2010; 185:1544–1549.
16. Du JW, Xu KY, Fang LY, Qi XL. Interleukin-17, produced by lymphocytes, promotes tumor growth and angiogenesis in a mouse model of breast cancer. Mol Med Rep. 2012; 6:1099–1102.
Article
17. Irshad S, Flores-Borja F, Lawler K, Monypenny J, Evans R, Male V, Gordon P, Cheung A, Gazinska P, Noor F, et al. RORγ t+ Innate Lymphoid Cells Promote Lymph Node Metastasis of Breast Cancers. Cancer Res. 2017; 77:1083–1096.
18. Rei M, Gonçalves-Sousa N, Lança T, Thompson RG, Mensurado S, Balkwill FR, Kulbe H, Pennington DJ, Silva-Santos B. Murine CD27(−) Vγ6(+) γδ T cells producing IL-17A promote ovarian cancer growth via mobilization of protumor small peritoneal macrophages. Proc Natl Acad Sci U S A. 2014; 111:E3562–E3570.
19. Charles KA, Kulbe H, Soper R, Escorcio-Correia M, Lawrence T, Schultheis A, Chakravarty P, Thompson RG, Kollias G, Smyth JF, et al. The tumor-promoting actions of TNF-α involve TNFR1 and IL-17 in ovarian cancer in mice and humans. J Clin Invest. 2009; 119:3011–3023.
Article
20. Cheng M, Qian L, Shen G, Bian G, Xu T, Xu W, Shen G, Hu S. Microbiota modulate tumoral immune surveillance in lung through a γδ T17 immune cell-dependent mechanism. Cancer Res. 2014; 74:4030–4041.
Article
21. Calcinotto A, Brevi A, Chesi M, Ferrarese R, Garcia Perez L, Grioni M, Kumar S, Garbitt VM, Sharik ME, Henderson KJ, et al. Microbiota-driven interleukin-17-producing cells and eosinophils synergize to accelerate multiple myeloma progression. Nat Commun. 2018; 9:4832.
Article
22. Coffelt SB, Kersten K, Doornebal CW, Weiden J, Vrijland K, Hau CS, Verstegen NJ, Ciampricotti M, Hawinkels LJ, Jonkers J, et al. IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature. 2015; 522:345–348.
Article
23. Chang SH, Mirabolfathinejad SG, Katta H, Cumpian AM, Gong L, Caetano MS, Moghaddam SJ, Dong C. T helper 17 cells play a critical pathogenic role in lung cancer. Proc Natl Acad Sci U S A. 2014; 111:5664–5669.
Article
24. Jin C, Lagoudas GK, Zhao C, Bullman S, Bhutkar A, Hu B, Ameh S, Sandel D, Liang XS, Mazzilli S, et al. Commensal microbiota promote lung cancer development via γδ T cells. Cell. 2019; 176:998–1013. e16.
Article
25. You R, DeMayo FJ, Liu J, Cho SN, Burt BM, Creighton CJ, Casal RF, Lazarus DR, Lu W, Tung HY, et al. IL17A regulates tumor latency and metastasis in lung adeno and squamous SQ.2b and AD.1 cancer. Cancer Immunol Res. 2018; 6:645–657.
Article
26. Wang L, Yi T, Zhang W, Pardoll DM, Yu H. IL-17 enhances tumor development in carcinogen-induced skin cancer. Cancer Res. 2010; 70:10112–10120.
Article
27. Chan KS, Sano S, Kiguchi K, Anders J, Komazawa N, Takeda J, DiGiovanni J. Disruption of Stat3 reveals a critical role in both the initiation and the promotion stages of epithelial carcinogenesis. J Clin Invest. 2004; 114:720–728.
Article
28. Housseau F, Wu S, Wick EC, Fan H, Wu X, Llosa NJ, Smith KN, Tam A, Ganguly S, Wanyiri JW, et al. Redundant innate and adaptive sources of IL17 production drive colon tumorigenesis. Cancer Res. 2016; 76:2115–2124.
Article
29. Kryczek I, Lin Y, Nagarsheth N, Peng D, Zhao L, Zhao E, Vatan L, Szeliga W, Dou Y, Owens S, et al. IL-22+ CD4+ T cells promote colorectal cancer stemness via STAT3 transcription factor activation and induction of the methyltransferase DOT1L. Immunity. 2014; 40:772–784.
30. Wang K, Kim MK, Di Caro G, Wong J, Shalapour S, Wan J, Zhang W, Zhong Z, Sanchez-Lopez E, Wu LW, et al. Interleukin-17 receptor a signaling in transformed enterocytes promotes early colorectal tumorigenesis. Immunity. 2014; 41:1052–1063.
Article
31. Kirchberger S, Royston DJ, Boulard O, Thornton E, Franchini F, Szabady RL, Harrison O, Powrie F. Innate lymphoid cells sustain colon cancer through production of interleukin-22 in a mouse model. J Exp Med. 2013; 210:917–931.
Article
32. Abousamra NK, Salah El-Din M, Helal R. Prognostic value of Th17 cells in acute leukemia. Med Oncol. 2013; 30:732.
Article
33. Han Y, Ye A, Bi L, Wu J, Yu K, Zhang S. Th17 cells and interleukin-17 increase with poor prognosis in patients with acute myeloid leukemia. Cancer Sci. 2014; 105:933–942.
Article
34. Chen WC, Lai YH, Chen HY, Guo HR, Su IJ, Chen HH. Interleukin-17-producing cell infiltration in the breast cancer tumour microenvironment is a poor prognostic factor. Histopathology. 2013; 63:225–233.
Article
35. Faucheux L, Grandclaudon M, Perrot-Dockès M, Sirven P, Berger F, Hamy AS, Fourchotte V, Vincent-Salomon A, Mechta-Grigoriou F, Reyal F, et al. A multivariate Th17 metagene for prognostic stratification in T cell non-inflamed triple negative breast cancer. OncoImmunology. 2019; 8:e1624130.
Article
36. Liu J, Duan Y, Cheng X, Chen X, Xie W, Long H, Lin Z, Zhu B. IL-17 is associated with poor prognosis and promotes angiogenesis via stimulating VEGF production of cancer cells in colorectal carcinoma. Biochem Biophys Res Commun. 2011; 407:348–354.
Article
37. Tosolini M, Kirilovsky A, Mlecnik B, Fredriksen T, Mauger S, Bindea G, Berger A, Bruneval P, Fridman WH, Pagès F, et al. Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, th2, treg, th17) in patients with colorectal cancer. Cancer Res. 2011; 71:1263–1271.
Article
38. Chen J, Chen Z. The effect of immune microenvironment on the progression and prognosis of colorectal cancer. Med Oncol. 2014; 31:82.
Article
39. Alves JJ, De Medeiros Fernandes TA, De Araújo JM, Cobucci RN, Lanza DC, Bezerra FL, Andrade VS, Fernandes JV. Th17 response in patients with cervical cancer. Oncol Lett. 2018; 16:6215–6227.
40. Punt S, Fleuren GJ, Kritikou E, Lubberts E, Trimbos JB, Jordanova ES, Gorter A. Angels and demons: Th17 cells represent a beneficial response, while neutrophil IL-17 is associated with poor prognosis in squamous cervical cancer. Oncoimmunology. 2015; 4:e984539.
Article
41. Wang B, Li L, Liao Y, Li J, Yu X, Zhang Y, Xu J, Rao H, Chen S, Zhang L, et al. Mast cells expressing interleukin 17 in the muscularis propria predict a favorable prognosis in esophageal squamous cell carcinoma. Cancer Immunol Immunother. 2013; 62:1575–1585.
Article
42. Lv L, Pan K, Li XD, She KL, Zhao JJ, Wang W, Chen JG, Chen YB, Yun JP, Xia JC. The accumulation and prognosis value of tumor infiltrating IL-17 producing cells in esophageal squamous cell carcinoma. PLoS One. 2011; 6:e18219.
Article
43. Zhang B, Rong G, Wei H, Zhang M, Bi J, Ma L, Xue X, Wei G, Liu X, Fang G. The prevalence of Th17 cells in patients with gastric cancer. Biochem Biophys Res Commun. 2008; 374:533–537.
Article
44. Yamada Y, Saito H, Ikeguchi M. Prevalence and clinical relevance of Th17 cells in patients with gastric cancer. J Surg Res. 2012; 178:685–691.
Article
45. Zhang JP, Yan J, Xu J, Pang XH, Chen MS, Li L, Wu C, Li SP, Zheng L. Increased intratumoral IL-17-producing cells correlate with poor survival in hepatocellular carcinoma patients. J Hepatol. 2009; 50:980–989.
Article
46. Liao R, Sun J, Wu H, Yi Y, Wang JX, He HW, Cai XY, Zhou J, Cheng YF, Fan J, et al. High expression of IL-17 and IL-17RE associate with poor prognosis of hepatocellular carcinoma. J Exp Clin Cancer Res. 2013; 32:3.
Article
47. Yan J, Liu XL, Xiao G, Li NL, Deng YN, Han LZ, Yin LC, Ling LJ, Liu LX. Prevalence and clinical relevance of T-helper cells, Th17 and Th1, in hepatitis B virus-related hepatocellular carcinoma. PLoS One. 2014; 9:e96080.
Article
48. Ye ZJ, Zhou Q, Gu YY, Qin SM, Ma WL, Xin JB, Tao XN, Shi HZ. Generation and differentiation of IL-17-producing CD4+ T cells in malignant pleural effusion. J Immunol. 2010; 185:6348–6354.
49. Gong Y, Chen SX, Gao BA, Yao RC, Guan L. Cell origins and significance of IL-17 in malignant pleural effusion. Clin Transl Oncol. 2014; 16:807–813.
Article
50. Bao Z, Lu G, Cui D, Yao Y, Yang G, Zhou J. IL-17A-producing T cells are associated with the progression of lung adenocarcinoma. Oncol Rep. 2016; 36:641–650.
Article
51. Zelba H, Weide B, Martens A, Derhovanessian E, Bailur JK, Kyzirakos C, Pflugfelder A, Eigentler TK, Di Giacomo AM, Maio M, et al. Circulating CD4+ T cells that produce IL4 or IL17 when stimulated by Melan-A but not by NY-ESO-1 have negative impacts on survival of patients with stage IV melanoma. Clin Cancer Res. 2014; 20:4390–4399.
52. Dhodapkar KM, Barbuto S, Matthews P, Kukreja A, Mazumder A, Vesole D, Jagannath S, Dhodapkar MV. Dendritic cells mediate the induction of polyfunctional human IL17-producing cells (Th17–1 cells) enriched in the bone marrow of patients with myeloma. Blood. 2008; 112:2878–2885.
Article
53. Shen CJ, Yuan ZH, Liu YX, Hu GY. Increased numbers of T helper 17 cells and the correlation with clinicopathological characteristics in multiple myeloma. J Int Med Res. 2012; 40:556–564.
Article
54. Zhang YL, Li J, Mo HY, Qiu F, Zheng LM, Qian CN, Zeng YX. Different subsets of tumor infiltrating lymphocytes correlate with NPC progression in different ways. Mol Cancer. 2010; 9:4.
Article
55. Li J, Mo HY, Xiong G, Zhang L, He J, Huang ZF, Liu ZW, Chen QY, Du ZM, Zheng LM, et al. Tumor microenvironment macrophage inhibitory factor directs the accumulation of interleukin-17-producing tumor-infiltrating lymphocytes and predicts favorable survival in nasopharyngeal carcinoma patients. J Biol Chem. 2012; 287:35484–35495.
Article
56. Xu C, Yu L, Zhan P, Zhang Y. Elevated pleural effusion IL-17 is a diagnostic marker and outcome predictor in lung cancer patients. Eur J Med Res. 2014; 19:23.
Article
57. Chen X, Wan J, Liu J, Xie W, Diao X, Xu J, Zhu B, Chen Z. Increased IL-17-producing cells correlate with poor survival and lymphangiogenesis in NSCLC patients. Lung Cancer. 2010; 69:348–354.
Article
58. Miyahara Y, Odunsi K, Chen W, Peng G, Matsuzaki J, Wang RF. Generation and regulation of human CD4+ IL-17-producing T cells in ovarian cancer. Proc Natl Acad Sci U S A. 2008; 105:15505–15510.
59. Kryczek I, Banerjee M, Cheng P, Vatan L, Szeliga W, Wei S, Huang E, Finlayson E, Simeone D, Welling TH, et al. Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments. Blood. 2009; 114:1141–1149.
Article
60. He S, Fei M, Wu Y, Zheng D, Wan D, Wang L, Li D. Distribution and clinical significance of Th17 cells in the tumor microenvironment and peripheral blood of pancreatic cancer patients. Int J Mol Sci. 2011; 12:7424–7437.
Article
61. Sfanos KS, Bruno TC, Maris CH, Xu L, Thoburn CJ, DeMarzo AM, Meeker AK, Isaacs WB, Drake CG. Phenotypic analysis of prostate-infiltrating lymphocytes reveals T H17 and Treg skewing. Clin Cancer Res. 2008; 14:3254–3261.
62. Derhovanessian E, Adams V, Hähnel K, Groeger A, Pandha H, Ward S, Pawelec G. Pretreatment frequency of circulating IL-17+ CD4+ T-cells, but not Tregs, correlates with clinical response to whole-cell vaccination in prostate cancer patients. Int J Cancer. 2009; 125:1372–1379.
63. Huang Y, Wang J, Jia P, Li X, Pei G, Wang C, Fang X, Zhao Z, Cai Z, Yi X, et al. Clonal architectures predict clinical outcome in clear cell renal cell carcinoma. Nat Commun. 2019; 10:1245.
Article
64. Koyama K, Kagamu H, Miura S, Hiura T, Miyabayashi T, Itoh R, Kuriyama H, Tanaka H, Tanaka J, Yoshizawa H, et al. Reciprocal CD4+ T-cell balance of effector CD62L low CD4+ and CD62L high CD25+ CD4+ regulatory T cells in small cell lung cancer reflects disease stage. Clin Cancer Res. 2008; 14:6770–6779.
65. Su X, Ye J, Hsueh EC, Zhang Y, Hoft DF, Peng G. Tumor microenvironments direct the recruitment and expansion of human Th17 cells. J Immunol. 2010; 184:1630–1641.
Article
66. Chen D, Jiang R, Mao C, Shi L, Wang S, Yu L, Hu Q, Dai D, Xu H. Chemokine/chemokine receptor interactions contribute to the accumulation of Th17 cells in patients with esophageal squamous cell carcinoma. Hum Immunol. 2012; 73:1068–1072.
Article
67. Yu Q, Lou XM, He Y. Preferential recruitment of Th17 cells to cervical cancer via CCR6-CCL20 pathway. PLoS One. 2015; 10:e0120855.
Article
68. Pan B, Shen J, Cao J, Zhou Y, Shang L, Jin S, Cao S, Che D, Liu F, Yu Y. Interleukin-17 promotes angiogenesis by stimulating VEGF production of cancer cells via the STAT3/GIV signaling pathway in non-small-cell lung cancer. Sci Rep. 2015; 5:16053.
Article
69. Li J, Huang ZF, Xiong G, Mo HY, Qiu F, Mai HQ, Chen QY, He J, Chen SP, Zheng LM, et al. Distribution, characterization, and induction of CD8+ regulatory T cells and IL-17-producing CD8+ T cells in nasopharyngeal carcinoma. J Transl Med. 2011; 9:189.
Article
70. Zhuang Y, Peng LS, Zhao YL, Shi Y, Mao XH, Chen W, Pang KC, Liu XF, Liu T, Zhang JY, et al. CD8+ T cells that produce interleukin-17 regulate myeloid-derived suppressor cells and are associated with survival time of patients with gastric cancer. Gastroenterology. 2012; 143:951–62. e8.
71. Lee MH, Tung-Chieh Chang J, Liao CT, Chen YS, Kuo ML, Shen CR. Interleukin 17 and peripheral IL-17-expressing T cells are negatively correlated with the overall survival of head and neck cancer patients. Oncotarget. 2018; 9:9825–9837.
Article
72. Hinrichs CS, Kaiser A, Paulos CM, Cassard L, Sanchez-Perez L, Heemskerk B, Wrzesinski C, Borman ZA, Muranski P, Restifo NP. Type 17 CD8+ T cells display enhanced antitumor immunity. Blood. 2009; 114:596–599.
73. Tajima M, Wakita D, Satoh T, Kitamura H, Nishimura T. IL-17/IFN-γ double producing CD8+ T (Tc17/IFN-γ) cells: a novel cytotoxic T-cell subset converted from Tc17 cells by IL-12. Int Immunol. 2011; 23:751–759.
74. Muranski P, Borman ZA, Kerkar SP, Klebanoff CA, Ji Y, Sanchez-Perez L, Sukumar M, Reger RN, Yu Z, Kern SJ, et al. Th17 cells are long lived and retain a stem cell-like molecular signature. Immunity. 2011; 35:972–985.
Article
75. Yu Y, Cho HI, Wang D, Kaosaard K, Anasetti C, Celis E, Yu XZ. Adoptive transfer of Tc1 or Tc17 cells elicits antitumor immunity against established melanoma through distinct mechanisms. J Immunol. 2013; 190:1873–1881.
Article
76. Bowers JS, Nelson MH, Majchrzak K, Bailey SR, Rohrer B, Kaiser AD, Atkinson C, Gattinoni L, Paulos CM. Th17 cells are refractory to senescence and retain robust antitumor activity after long-term ex vivo expansion. JCI Insight. 2017; 2:e90772.
Article
77. Hu X, Majchrzak K, Liu X, Wyatt MM, Spooner CJ, Moisan J, Zou W, Carter LL, Paulos CM. In vitro priming of adoptively transferred T cells with a RORγ agonist confers durable memory and stemness in vivo. Cancer Res. 2018; 78:3888–3898.
78. Wu P, Wu D, Ni C, Ye J, Chen W, Hu G, Wang Z, Wang C, Zhang Z, Xia W, et al. γδ T17 cells promote the accumulation and expansion of myeloid-derived suppressor cells in human colorectal cancer. Immunity. 2014; 40:785–800.
Article
79. Daley D, Zambirinis CP, Seifert L, Akkad N, Mohan N, Werba G, Barilla R, Torres-Hernandez A, Hundeyin M, Mani VR, et al. γδ T cells support pancreatic oncogenesis by restraining αβ T cell activation. Cell. 2016; 166:1485–1499. e15.
Article
80. Kwon DI, Lee YJ. Lineage differentiation program of invariant natural killer T cells. Immune Netw. 2017; 17:365–377.
Article
81. Crosby CM, Kronenberg M. Tissue-specific functions of invariant natural killer T cells. Nat Rev Immunol. 2018; 18:559–574.
Article
82. Kim CH. Control of innate and adaptive lymphocytes by the RAR-retinoic acid axis. Immune Netw. 2018; 18:e1.
Article
83. Bruchard M, Ghiringhelli F. Deciphering the roles of innate lymphoid cells in cancer. Front Immunol. 2019; 10:656.
Article
84. Koh J, Kim HY, Lee Y, Park IK, Kang CH, Kim YT, Kim JE, Choi M, Lee WW, Jeon YK, et al. IL23-producing human lung cancer cells promote tumor growth via conversion of innate lymphoid cell 1 (ILC1) into ILC3. Clin Cancer Res. 2019; 25:4026–4037.
Article
85. Zhu X, Mulcahy LA, Mohammed RA, Lee AH, Franks HA, Kilpatrick L, Yilmazer A, Paish EC, Ellis IO, Patel PM, et al. IL-17 expression by breast-cancer-associated macrophages: IL-17 promotes invasiveness of breast cancer cell lines. Breast Cancer Res. 2008; 10:R95.
Article
86. Vykhovanets EV, Maclennan GT, Vykhovanets OV, Gupta S. IL-17 Expression by macrophages is associated with proliferative inflammatory atrophy lesions in prostate cancer patients. Int J Clin Exp Pathol. 2011; 4:552–565.
87. Chen X, Churchill MJ, Nagar KK, Tailor YH, Chu T, Rush BS, Jiang Z, Wang EB, Renz BW, Wang H, et al. IL-17 producing mast cells promote the expansion of myeloid-derived suppressor cells in a mouse allergy model of colorectal cancer. Oncotarget. 2015; 6:32966–32979.
Article
88. Tu JF, Pan HY, Ying XH, Lou J, Ji JS, Zou H. Mast cells comprise the major of interleukin 17-producing cells and predict a poor prognosis in hepatocellular carcinoma. Medicine (Baltimore). 2016; 95:e3220.
Article
89. Liu X, Jin H, Zhang G, Lin X, Chen C, Sun J, Zhang Y, Zhang Q, Yu J. Intratumor IL-17-positive mast cells are the major source of the IL-17 that is predictive of survival in gastric cancer patients. PLoS One. 2014; 9:e106834.
Article
90. Spolski R, Leonard WJ. Interleukin-21: basic biology and implications for cancer and autoimmunity. Annu Rev Immunol. 2008; 26:57–79.
Article
91. Petrella TM, Tozer R, Belanger K, Savage KJ, Wong R, Smylie M, Kamel-Reid S, Tron V, Chen BE, Hunder NN, et al. Interleukin-21 has activity in patients with metastatic melanoma: a phase II study. J Clin Oncol. 2012; 30:3396–3401.
Article
92. Petrella TM, Mihalcioiu CL, McWhirter E, Belanger K, Savage KJ, Song X, Hamid O, Cheng T, Davis ML, Lee CW, et al. Final efficacy results of NCIC CTG IND.202: a randomized phase II study of recombinant interleukin-21 (rIL21) in patients with recurrent or metastatic melanoma (MM). J Clin Oncol. 2013; 31:9032.
Article
93. Chapuis AG, Lee SM, Thompson JA, Roberts IM, Margolin KA, Bhatia S, Sloan HL, Lai I, Wagener F, Shibuya K, et al. Combined IL-21-primed polyclonal CTL plus CTLA4 blockade controls refractory metastatic melanoma in a patient. J Exp Med. 2016; 213:1133–1139.
Article
94. Duhen T, Geiger R, Jarrossay D, Lanzavecchia A, Sallusto F. Production of interleukin 22 but not interleukin 17 by a subset of human skin-homing memory T cells. Nat Immunol. 2009; 10:857–863.
Article
95. Jin M, Yoon J. From bench to clinic: the potential of therapeutic targeting of the IL-22 signaling pathway in atopic dermatitis. Immune Netw. 2018; 18:e42.
Article
96. Nardinocchi L, Sonego G, Passarelli F, Avitabile S, Scarponi C, Failla CM, Simoni S, Albanesi C, Cavani A. Interleukin-17 and interleukin-22 promote tumor progression in human nonmelanoma skin cancer. Eur J Immunol. 2015; 45:922–931.
Article
97. Eyerich K, Dimartino V, Cavani A. IL-17 and IL-22 in immunity: driving protection and pathology. Eur J Immunol. 2017; 47:607–614.
Article
98. Jiang R, Wang H, Deng L, Hou J, Shi R, Yao M, Gao Y, Yao A, Wang X, Yu L, et al. IL-22 is related to development of human colon cancer by activation of STAT3. BMC Cancer. 2013; 13:59.
Article
99. Lim C, Savan R. The role of the IL-22/IL-22R1 axis in cancer. Cytokine Growth Factor Rev. 2014; 25:257–271.
Article
100. Wen Z, Liao Q, Zhao J, Hu Y, You L, Lu Z, Jia C, Wei Y, Zhao Y. High expression of interleukin-22 and its receptor predicts poor prognosis in pancreatic ductal adenocarcinoma. Ann Surg Oncol. 2014; 21:125–132.
Article
101. Zhang W, Chen Y, Wei H, Zheng C, Sun R, Zhang J, Tian Z. Antiapoptotic activity of autocrine interleukin-22 and therapeutic effects of interleukin-22-small interfering RNA on human lung cancer xenografts. Clin Cancer Res. 2008; 14:6432–6439.
Article
102. Pylayeva-Gupta Y, Lee KE, Hajdu CH, Miller G, Bar-Sagi D. Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell. 2012; 21:836–847.
Article
103. Bayne LJ, Beatty GL, Jhala N, Clark CE, Rhim AD, Stanger BZ, Vonderheide RH. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell. 2012; 21:822–835.
Article
104. Su S, Liu Q, Chen J, Chen J, Chen F, He C, Huang D, Wu W, Lin L, Huang W, et al. A positive feedback loop between mesenchymal-like cancer cells and macrophages is essential to breast cancer metastasis. Cancer Cell. 2014; 25:605–620.
Article
105. Chen Y, Zhao Z, Chen Y, Lv Z, Ding X, Wang R, Xiao H, Hou C, Shen B, Feng J, et al. An epithelial-to-mesenchymal transition-inducing potential of granulocyte macrophage colony-stimulating factor in colon cancer. Sci Rep. 2017; 7:8265.
Article
106. Martin-Orozco N, Muranski P, Chung Y, Yang XO, Yamazaki T, Lu S, Hwu P, Restifo NP, Overwijk WW, Dong C. T helper 17 cells promote cytotoxic T cell activation in tumor immunity. Immunity. 2009; 31:787–798.
Article
107. Ankathatti Munegowda M, Deng Y, Mulligan SJ, Xiang J. Th17 and Th17-stimulated CD8+ T cells play a distinct role in Th17-induced preventive and therapeutic antitumor immunity. Cancer Immunol Immunother. 2011; 60:1473–1484.
108. Qian X, Gu L, Ning H, Zhang Y, Hsueh EC, Fu M, Hu X, Wei L, Hoft DF, Liu J. Increased Th17 cells in the tumor microenvironment is mediated by IL-23 via tumor-secreted prostaglandin E2. J Immunol. 2013; 190:5894–5902.
Article
109. Numasaki M, Watanabe M, Suzuki T, Takahashi H, Nakamura A, McAllister F, Hishinuma T, Goto J, Lotze MT, Kolls JK, et al. IL-17 enhances the net angiogenic activity and in vivo growth of human non-small cell lung cancer in SCID mice through promoting CXCR-2-dependent angiogenesis. J Immunol. 2005; 175:6177–6189.
110. Lee JW, Wang P, Kattah MG, Youssef S, Steinman L, DeFea K, Straus DS. Differential regulation of chemokines by IL-17 in colonic epithelial cells. J Immunol. 2008; 181:6536–6545.
Article
111. Kim CH, Park J, Kim M. Gut microbiota-derived short-chain fatty acids, T cells, and inflammation. Immune Netw. 2014; 14:277–288.
Article
112. Wu S, Rhee KJ, Albesiano E, Rabizadeh S, Wu X, Yen HR, Huso DL, Brancati FL, Wick E, McAllister F, et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat Med. 2009; 15:1016–1022.
Article
113. Grivennikov SI, Wang K, Mucida D, Stewart CA, Schnabl B, Jauch D, Taniguchi K, Yu GY, Osterreicher CH, Hung KE, et al. Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature. 2012; 491:254–258.
Article
114. Chan IH, Jain R, Tessmer MS, Gorman D, Mangadu R, Sathe M, Vives F, Moon C, Penaflor E, Turner S, et al. Interleukin-23 is sufficient to induce rapid de novo gut tumorigenesis, independent of carcinogens, through activation of innate lymphoid cells. Mucosal Immunol. 2014; 7:842–856.
115. Bagheri N, Azadegan-Dehkordi F, Shirzad H, Rafieian-Kopaei M, Rahimian G, Razavi A. The biological functions of IL-17 in different clinical expressions of Helicobacter pylori-infection. Microb Pathog. 2015; 81:33–38.
116. McAllister F, Bailey JM, Alsina J, Nirschl CJ, Sharma R, Fan H, Rattigan Y, Roeser JC, Lankapalli RH, Zhang H, et al. Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 signaling axis in preinvasive pancreatic neoplasia. Cancer Cell. 2014; 25:621–637.
Article
117. Li Q, Anderson CD, Egilmez NK. Inhaled IL-10 suppresses lung tumorigenesis via abrogation of inflammatory macrophage-Th17 cell axis. J Immunol. 2018; 201:2842–2850.
Article
118. Wang L, Yi T, Kortylewski M, Pardoll DM, Zeng D, Yu H. IL-17 can promote tumor growth through an IL-6-Stat3 signaling pathway. J Exp Med. 2009; 206:1457–1464.
Article
119. Chen X, Cai G, Liu C, Zhao J, Gu C, Wu L, Hamilton TA, Zhang CJ, Ko J, Zhu L, et al. IL-17R-EGFR axis links wound healing to tumorigenesis in Lrig1+ stem cells. J Exp Med. 2019; 216:195–214.
120. Fanok MH, Sun A, Fogli LK, Narendran V, Eckstein M, Kannan K, Dolgalev I, Lazaris C, Heguy A, Laird ME, et al. Role of dysregulated cytokine signaling and bacterial triggers in the pathogenesis of cutaneous T-cell lymphoma. J Invest Dermatol. 2018; 138:1116–1125.
121. Haudenschild D, Moseley T, Rose L, Reddi AH. Soluble and transmembrane isoforms of novel interleukin-17 receptor-like protein by RNA splicing and expression in prostate cancer. J Biol Chem. 2002; 277:4309–4316.
Article
122. Steiner GE, Newman ME, Paikl D, Stix U, Memaran-Dagda N, Lee C, Marberger MJ. Expression and function of pro-inflammatory interleukin IL-17 and IL-17 receptor in normal, benign hyperplastic, and malignant prostate. Prostate. 2003; 56:171–182.
Article
123. Yu H, Kortylewski M, Pardoll D. Crosstalk between cancer and immune cells: role of STAT3 in the tumour microenvironment. Nat Rev Immunol. 2007; 7:41–51.
Article
124. Li J, Lau GK, Chen L, Dong SS, Lan HY, Huang XR, Li Y, Luk JM, Yuan YF, Guan XY. Interleukin 17A promotes hepatocellular carcinoma metastasis via NF-kB induced matrix metalloproteinases 2 and 9 expression. PLoS One. 2011; 6:e21816.
Article
125. Yang B, Kang H, Fung A, Zhao H, Wang T, Ma D. The role of interleukin 17 in tumour proliferation, angiogenesis, and metastasis. Mediators Inflamm. 2014; 2014:623759.
Article
126. Xiang T, Long H, He L, Han X, Lin K, Liang Z, Zhuo W, Xie R, Zhu B. Interleukin-17 produced by tumor microenvironment promotes self-renewal of CD133+ cancer stem-like cells in ovarian cancer. Oncogene. 2015; 34:165–176.
Article
127. Zhang Q, Liu S, Parajuli KR, Zhang W, Zhang K, Mo Z, Liu J, Chen Z, Yang S, Wang AR, et al. Interleukin-17 promotes prostate cancer via MMP7-induced epithelial-to-mesenchymal transition. Oncogene. 2017; 36:687–699.
Article
128. Corcoran RB, Contino G, Deshpande V, Tzatsos A, Conrad C, Benes CH, Levy DE, Settleman J, Engelman JA, Bardeesy N. STAT3 plays a critical role in KRAS-induced pancreatic tumorigenesis. Cancer Res. 2011; 71:5020–5029.
129. Ho PL, Lay EJ, Jian W, Parra D, Chan KS. Stat3 activation in urothelial stem cells leads to direct progression to invasive bladder cancer. Cancer Res. 2012; 72:3135–3142.
Article
130. Punt S, Langenhoff JM, Putter H, Fleuren GJ, Gorter A, Jordanova ES. The correlations between IL-17 vs. Th17 cells and cancer patient survival: a systematic review. OncoImmunology. 2015; 4:e984547.
131. Park YJ, Kuen DS, Chung Y. Future prospects of immune checkpoint blockade in cancer: from response prediction to overcoming resistance. Exp Mol Med. 2018; 50:109.
Article
132. Cochaud S, Giustiniani J, Thomas C, Laprevotte E, Garbar C, Savoye AM, Curé H, Mascaux C, Alberici G, Bonnefoy N, et al. IL-17A is produced by breast cancer TILs and promotes chemoresistance and proliferation through ERK1/2. Sci Rep. 2013; 3:3456.
Article
133. Sui G, Qiu Y, Yu H, Kong Q, Zhen B. Interleukin-17 promotes the development of cisplatin resistance in colorectal cancer. Oncol Lett. 2019; 17:944–950.
Article
134. Chung AS, Wu X, Zhuang G, Ngu H, Kasman I, Zhang J, Vernes JM, Jiang Z, Meng YG, Peale FV, et al. An interleukin-17-mediated paracrine network promotes tumor resistance to anti-angiogenic therapy. Nat Med. 2013; 19:1114–1123.
Article
135. Nuñez S, Saez JJ, Fernandez D, Flores-Santibañez F, Alvarez K, Tejon G, Ruiz P, Maldonado P, Hidalgo Y, Manriquez V, et al. T helper type 17 cells contribute to anti-tumour immunity and promote the recruitment of T helper type 1 cells to the tumour. Immunology. 2013; 139:61–71.
Article
136. Majchrzak K, Nelson MH, Bailey SR, Bowers JS, Yu XZ, Rubinstein MP, Himes RA, Paulos CM. Exploiting IL-17-producing CD4+ and CD8+ T cells to improve cancer immunotherapy in the clinic. Cancer Immunol Immunother. 2016; 65:247–259.
137. Liu J, Zhou G, Zhang L, Zhao Q. Building Potent Chimeric Antigen Receptor T Cells With CRISPR Genome Editing. Front Immunol. 2019; 10:456.
Article
138. Cheng J, Zhao L, Zhang Y, Qin Y, Guan Y, Zhang T, Liu C, Zhou J. Understanding the mechanisms of resistance to CAR T-cell therapy in malignancies. Front Oncol. 2019; 9:1237.
Article
139. Chung Y, Qin H, Kang CY, Kim S, Kwak LW, Dong C. An NKT-mediated autologous vaccine generates CD4 T-cell dependent potent antilymphoma immunity. Blood. 2007; 110:2013–2019.
140. Min B, Choi H, Her JH, Jung MY, Kim HJ, Jung MY, Lee EK, Cho SY, Hwang YK, Shin EC. Optimization of Large-Scale Expansion and Cryopreservation of Human Natural Killer Cells for Anti-Tumor Therapy. Immune Netw. 2018; 18:e31.
Article
141. Mahalingam D, Wang JS, Hamilton EP, Sarantopoulos J, Nemunaitis J, Weems G, Carter L, Hu X, Schreeder M, Wilkins HJ. Phase 1 open-label, multicenter study of first-in-class RORγ agonist LYC-55716 (cintirorgon): safety, tolerability, and preliminary evidence of antitumor activity. Clin Cancer Res. 2019; 25:3508–3516.
Article
142. Bie Q, Sun C, Gong A, Li C, Su Z, Zheng D, Ji X, Wu Y, Guo Q, Wang S, et al. Non-tumor tissue derived interleukin-17B activates IL-17RB/AKT/β-catenin pathway to enhance the stemness of gastric cancer. Sci Rep. 2016; 6:25447.
Article
143. Majchrzak K, Nelson MH, Bowers JS, Bailey SR, Wyatt MM, Wrangle JM, Rubinstein MP, Varela JC, Li Z, Himes RA, et al. β-catenin and PI3Kδ inhibition expands precursor Th17 cells with heightened stemness and antitumor activity. JCI Insight. 2017; 2:90547.
Article
144. Guedan S, Chen X, Madar A, Carpenito C, McGettigan SE, Frigault MJ, Lee J, Posey AD Jr, Scholler J, Scholler N, et al. ICOS-based chimeric antigen receptors program bipolar TH17/TH1 cells. Blood. 2014; 124:1070–1080.
Article
145. Lee M, Rhee I. Cytokine Signaling in Tumor Progression. Immune Netw. 2017; 17:214–227.
Article
Full Text Links
  • IN
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