Effector and Regulatory CD4+ T helper lineages in cancer

Estefanía Nova-Lamperti1,2*, Marco Fraga1, Marco Romano3, Giovanna Lombardi3#

1Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, 4070386. Concepcion, Chile

2PreveGen Laboratory, Chacabuco 556, Concepcion, Chile

3MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, SE1 9RT. London, United Kingdom


Regulatory CD4+T-cells have been recently classified as regulatory T helper (Th)-like cells according to the expression of specific transcription factors, cytokines and chemokine receptors that mirror effector Th lineages. In our report: “An Atlas of Human Regulatory T Helper-like Cells Reveals Features of Th2-like Tregs that Support a Tumorigenic Environment” we found that Th2-like Tregs were increased in the tumour microenvironment in comparison with Th1-like, Th17-like and Th1/Th17-like Tregs. In addition, despite similar expression of CCR4 between all Tregs subtypes, Th2-like Tregs migrated more toward CCL17 and CCL22, and expressed higher levels of CCR8 than the other Th-like Tregs. Other studies have characterised human tissue-infiltrating Tregs and demonstrated that CCR8 is the main chemokine receptor differentially expressed in Tregs isolated from malignant tissues in comparison with healthy tissues. Given that CCR8 is a marker of Th2 lineages it is possible that Th2-like Tregs and CCR8+ Tregs are the same population of cells involved in tumour progression. However, whether they are recruited or differentiated in situ by the tumour microenvironment is still unknown. In this mini review, we describe the role of the different Th subsets in health and cancer and we present the different hypotheses about the presence of Th2-like Tregs in the tumorigenic environments.


CD4+T-cells have been classified by several research groups into different sub-types based on their genetic profile, phenotype, maturation state or function. Naïve CD4+T-cells undergo differentiation into specific T helper (Th) subsets via specific cytokine signals1,2. The main effector CD4+Th lineages are the Th1, Th2 and Th17 subset, whereas the main immuno-modulatory population is called regulatory T-cells (Tregs). These CD4+T-cell subsets play different roles in the immunological response: Th1 cells produce high levels of IFN-γ and are protective against intracellular pathogens. The key factor necessary for their differentiation is IL-12, however a role for IFN-γ and at the late stage IL-183 has been shown. IL-12, through STAT-4, and IFN-γ through STAT-1, promote activation of T-bet (Th1 master transcription factor), while IL-18 bolster the IFN-γ production. Th2 are induced by and confer protection against helminths, but they can also promote acute and chronic inflammatory responses against a myriad of allergens. IL-4 signalling through STAT6, is essential for Th2 activation and differentiation. Th17 cells are characterized by the capacity to secrete IL-17 and its differentiation is induced with several cytokines such as IL1-b, IL-6 and TGF-b, in humans. They have an important role in the clearance of extracellular bacteria and fungi, due to their capability to recruit and activate neutrophils. In addition, they have been associated with the pathogenesis of several autoimmune diseases. The expression of chemokine receptors by of these subsets mediates their chemotaxis towards different tissues4-6. Clarifying how chemokines and their cognate receptor orchestrate T-cell trafficking and activity is essential in gaining a better understanding of their role and distribution in health and disease. Imbalance between Th-like effector cells and Tregs has been observed in several cancers7-9 however how Tregs, but not pro-inflammatory effectors, preferentially migrate to these malignant tissues and which subset is the main responsible for tumour maintenance remains to be fully elucidated.

Th1 cells are the most abundant Th subset in peripheral blood and have been involved in pro- inflammatory immune responses. Th1 responses have been associated with the production of opsonising and complement-fixing antibodies by B cells, activation of macrophages, cell cytotoxicity and induction of cell-mediated immunity10. This subset is characterised by the expression of the transcription factor T-bet11, which promotes the secretion of IFN-γ12 and the expression of the chemokine receptor CXCR35. CXCR3 mediates migration to its ligand CXCL10, which is highly expressed in a diverse range of human diseases such as infectious diseases, inflammatory and autoimmune diseases13.

Th2 cells have been associated with allergy14 and asthma15. These cells are characterised by the transcription factor GATA316, they produce IL-4, IL-5 and IL-1310 and express the chemokine receptors CCR417 and CCR818. CCR4 mediates the migration to its ligands CCL17 and CCL22 produced by DCs upon maturation thereby playing a key role in recruiting cells into lymphoid tissues19,20.

Th17 cells mediate host defensive mechanisms to various infections, especially extracellular bacteria infections and are involved in the pathogenesis of many autoimmune diseases. The main transcription factor expressed by this subset is RORγτ21. These cells produce IL-1721 and express the chemokine receptor CCR622 and CCR423. The expression of CCL20, the ligand for CCR6, is induced by IL-17 and secreted by Th17 cells during inflammation and co-ordinates the migration of Th17 and Tregs to inflammatory sites22.

s JCTD-17-1120-Fig1

Figure 1: A model for transcriptional activation by enhancer.

TF, transcription factor. Med, Mediator complex. Pol II, RNA polymerase II. Transcription factors often recruit chromatin-modifying coactivators, transcription elongation complex, and architectural proteins in interaction between enhancer and promoter.

Tregs are a subset of T-cells that maintain immunological tolerance and regulate immune homeostasis24,25. Human Tregs are characterised by the high expression of the IL-2 receptor α-chain (CD25), the transcription factor FoxP3 and the low expression of the IL-7 receptor ?-chain (CD127). In 2012, Duhen et al., described new subpopulations of memory Tregs mirroring the classical CD4+Th cells. These new subpopulations, named Th-like Tregs, express chemokine receptors CXCR3, CCR6 and CCR4, typically expressed by T-bet+-Th1, RORγt+-Th17 and GATA3+-Th2, respectively23. The high levels of expression of CCR4 by memory Tregs, and concomitantly the presence of CCL22 in the tumour environment has been shown to induce chemotaxis of Tregs in melanoma26, gastric cancer27 and breast cancer19. In addition to effector cells, CXCL10 also mediates the recruitment of Tregs into chronically inflamed liver, as liver-infiltrating Tregs expressed higher levels of CXCR3 when compared to peripheral blood Tregs28. Th17 cells are classically pro-inflammatory, however studies have shown that Foxp3+IL17+T-cells detected in colorectal cancer have the ability to suppress tumour-specific CD8+T-cells29 and promote the development of cancer-initiating cells30. Recently, we performed a phenotypic and functional characterization of these Th-like Tregs in periphery and we observed a similar cytokine distribution pattern than effector Th subsets (Figure 1)31.

Effector Th1 lineages evolved to eradicate intracellular pathogens, such as intracellular bacteria and viruses, through activation of cytotoxic T-cell (CTL) responses, as well as immunoglobulin (Ig) G2a and IgG3 class switch32,33. Thus, Th1 responses are primarily responsible for activating and regulating the development and persistence of CTL, which can directly kill tumours. In addition, IFN-g mediated Th1 responses have been shown to trigger tumouricidal activity of tumour-infiltrating macrophages and promote the secretion of CXCL9 and CXCL10, supporting the recruitment of new Th1 cells and the prevention of cancer progression34. Furthermore, Th1 cytokines can promote anti-tumour responses and utilise a granzyme-perforin-dependent pathway for killing cancer cells35. Th2 responses have been associated with several cancers such as pancreatic cancer36,37, oesophageal and gastric cancer38, melanoma39 and ovarian cancer40. The role of Th17 responses in cancer is context-dependent41. IL-17 plays a dual role in the antitumor response through a cytotoxic T-cell action and by promoting angiogenesis. In addition, they display a great degree of context-dependent plasticity making them able to acquire functional characteristics of Tregs and effector T helper cells.

Several research groups have recently described the genetic profile of tumour-resident Tregs42. In 2016, Tirosh et al., analysed single-cell expression profile of immune cell isolated from melanoma samples, including effector CD8+ T-cells, CD4+T-cells and Tregs. The main gene associated with migratory functions expressed by infiltrating-tumour Tregs in this study was CCR843. The classical ligand for CCR8 are CCL1 and CCL18, however it has been shown that CCL17 also induces migration of CCR8-expressing cells44, which is relevant in the cancer field as CCL17 has been strongly associated with tumour progression27. Same year, two independent research groups also described CCR8 as one of the main chemokine receptor in Tregs present in breast45, colorectal and non-small-cell lung cancer46. Whereas Plitas et al., mentioned that tumour environment has the ability to potentiate CCR8 expression in Tregs, De Simone et al., discussed that the presence of CCL18 in different tumours might be associated with the migration of CCR8+ cells45,46. In 2017, our research group characterized the presence of Th1, Th2, Th17 and Th1/17 Tregs in peripheral blood and several tissues, such as skin, colon, thymus, spleen and liver perfusates31. We observed that different Treg subsets exhibited different location, cytokine profile, viability and migratory capacity. Interestingly, the presence of these subsets was significantly different between healthy and cancer tissues, obtained from patients with melanoma and colorectal cancer. Our data showed an increment of Th2-like Tregs in malignant tissues compared to the healthy counterpart; however, colorectal cancer was enriched in Th2 effector cells as well, suggesting that tumour microenvironment may preferentially select this lineage. Before the identification of Th-like Tregs, CCR8 expression was described in CD4+ memory T-cells enriched for Tregs and Th2 effector cells, characterised by the production of Th2-type cytokines18. However, whether Th2-like Tregs and CCR8+ Tregs are the same population needs further investigation.

Altogether the evidence that Th2-like Tregs are increased in the tumour environment raised the question which is the main mechanism used by the tumour to promote the presence of Th2-like Tregs. The secretion of specific chemokines at the tumour site stimulating the migration of Th2-like cells is one possible scenario. Another possible explanation is the specific expansion of Th2-like subsets at the cancer site to reduce the anti-tumour immunological response. For example, the expression of TIGIT (T-Cell Immunoreceptor With Ig And ITIM Domains) has been found in tumour-infiltrating conventional CD4+T-cells compared to circulating CD4+T-cells45. It has been demonstrated that TIGIT contributes to the induction of a selective Treg-cell-mediated suppression of pro-inflammatory Th1 and Th17 cells but not Th2 cell responses47, thus it is possible that Th2 cells are not only recruited to the tumour site, but also promoted by evading TIGIT-mediated inhibition. The final explanation is that the production of IL-4, a classical Th2 cytokine, promote the differentiation of Th2 cells and inhibit IFN-g production by CD4+T-cells48, therefore the secretion of this cytokine could also promote an enriched Th2 environment.

The increased presence of Tregs in malignant tissues plays a detrimental role in the battle between the immune system against the tumour. More recently, the presence of a specific type of Tregs (Th2-like Tregs) identified by us in melanoma and colorectal cancer could indicate that the malignant tissue is able to modify the immunological microenvironment surrounding the tumour in order to promote its development and avoid an anti-cancer response. Future studies about tumour-infiltrating Tregs or other regulatory population like tumour-associated macrophages can help us to clarify the mechanisms involved in tumour progression and to develop new alternative anti-tumour strategies.

Acknowledgement: CONICYT - Fondecyt de Iniciación (Grant Number: 11170610) and Programa de Atracción e Inserción (Grant Number: 79170073) supported E.N-L and M.F. The British Heart Foundation supported M.R (Grant Number: TG/16/2/32657).

s Estefanía Nova-Lamperti, Marco Fraga, Marco Romano and Giovanna Lombardi declare that they have no conflict of interest.

  1. Zhu J, Yamane H, Paul WE. Differentiation of effector CD4+ T cell populations. Annual Review of Immunology. 2010; 28: 445–489.
  2. Zhu J, Paul WE. Peripheral CD4+ T-cell differentiation regulated by networks of cytokines and transcription factors. Immunol Rev. 2010; 238: 247–262.
  3. Tominaga K, Yoshimoto T, Torigoe K, et al. IL-12 synergizes with IL-18 or IL-1β for IFN-γ production from human T cells. Int Immunol. 2000; 12: 151–160.
  4. Yamazaki T, Yang XO, Chung Y, et al. CCR6 regulates the migration of inflammatory and regulatory T cells. J Immunol. 2008; 181: 8391–8401.
  5. Groom JR, Luster A D. CXCR3 in T cell function. Exp Cell Res. 2011; 317: 620–631.
  6. Liu W, Wei X, Li L, et al. CCR4 mediated chemotaxis of regulatory T cells suppress the activation of T cells and NK cells via TGF-β pathway in human non-small cell lung cancer. Biochem Biophys Res Commun. 2017; 488: 196–203.
  7. Hung K, Hayashi R, Lafond-Walker A, et al. The central role of CD4+ T cells in the antitumor immune response. J Exp Med. 1998; 188: 2357–2368.
  8. Giuntoli RL, Webb TJ, Zoso A, et al. Ovarian cancer-associated ascites demonstrates altered immune environment: implications for antitumor immunity. Anticancer Res. 2009; 29: 2875–2884.
  9. Knutson KL, Disis ML. Tumor antigen-specific T helper cells in cancer immunity and immunotherapy. Cancer Immunol Immunother. 2005; 54: 721–728.
  10. Romagnani S. T-cell subsets (Th1 versus Th2). Annals of Allergy, Asthma & Immunology. 2000; 85: 9–21.
  11. Finotto S, Neurath MF, Glickman JN, et al. Development of spontaneous airway changes consistent with human asthma in mice lacking T-bet. Science. 2002; 295: 336–338.
  12. Lazarevic V, Glimcher LH, Lord GM. T-bet: A bridge between innate and adaptive immunity. Nat Rev Immunol. 2013; 13: 777–789.
  13. Lee EY, Lee ZH, Song YW. CXCL10 and autoimmune diseases. Autoimmun Rev. 2009; 8: 379–383.
  14. Licona-Limón P, Kim LK, Palm NW, et al. TH2, allergy and group 2 innate lymphoid cells. Nat Immunol. 2013; 14: 536–542.
  15. Coyle AJ, Tsuyuki S. Th2 cells and cytokine networks in allergic inflammation of the lung. Mediators Inflamm. 1995; 4: 239–247.
  16. Lee HJ, Takemoto N, Kurata H, et al. GATA-3 induces T helper cell type 2 (Th2) cytokine expression and chromatin remodeling in committed Th1 cells. J Exp Med. 2000; 192: 105–115.
  17. Yamamoto J, Adachi Y, Onoue Y, et al. Differential expression of the chemokine receptors by the Th1-and Th2-type effector populations within circulating CD4+T cells. Journal of Leukocyte Biology. 2000; 68: 568–574.
  18. Soler D, Chapman TR, Poisson LR, et al. CCR8 expression identifies CD4 memory T cells enriched for FOXP3+ regulatory and Th2 effector lymphocytes. The Journal of Immunology. 2006; 177: 6940–6951.
  19. Gobert M, Treilleux I, Bendriss-Vermare N, et al. Regulatory T cells recruited through CCL22/CCR4 are selectively activated in lymphoid infiltrates surrounding primary breast tumors and lead to an adverse clinical outcome. Cancer Research. 2009; 69: 2000–2009.
  20. Perros F, Hoogsteden HC, Coyle AJ, et al. Blockade of CCR4 in a humanized model of asthma reveals a critical role for DC-derived CCL17 and CCL22 in attracting Th2 cells and inducing airway inflammation. Allergy. 2009; 64: 995–1002.
  21. Ivanov II, McKenzie BS, Zhou L, et al. The Orphan Nuclear Receptor RORγt Directs the Differentiation Program of Proinflammatory IL-17+T Helper Cells. Cell. 2006; 126: 1121–1133.
  22. Hirota K, Yoshitomi H, Hashimoto M, et al. Preferential recruitment of CCR6-expressing Th17 cells to inflamed joints via CCL20 in rheumatoid arthritis and its animal model. J Exp Med. 2007; 204: 2803–2812.
  23. Duhen T, Duhen R, Lanzavecchia A, et al. Functionally distinct subsets of human FOXP3+ Treg cells that phenotypically mirror effector Th cells. Blood. 2012; 119: 4430–4440.
  24. Sakaguchi S, Yamaguchi T, Nomura T, et al. Regulatory T cells and immune tolerance. Cell. 2008; 133: 775–787.
  25. Rosenblum MD, Way SS, Abbas AK. Regulatory T cell memory. Nat Rev Immunol. 2016; 16: 90–101.
  26. Klarquist J, Tobin K, Farhangi Oskuei P, et al. Ccl22 Diverts T Regulatory Cells and Controls the Growth of Melanoma. Cancer Research. 2016; 76: 6230–6240.
  27. Mizukami Y, Kono K, Kawaguchi Y, et al. CCL17 and CCL22 chemokines within tumor microenvironment are related to accumulation of Foxp3+ regulatory T cells in gastric cancer. Int J Cancer. 2008; 122: 2286–2293.
  28. Oo YH, Weston CJ, Lalor PF, et al. Distinct roles for CCR4 and CXCR3 in the recruitment and positioning of regulatory T cells in the inflamed human liver. J Immunol. 2010; 184: 2886–2898.
  29. Ma C, Dong X. Colorectal cancer-derived Foxp3(+) IL-17(+) T cells suppress tumour-specific CD8+ T cells. Scand. J. Immunol. 2011; 74: 47–51.
  30. Yang S, Wang B, Guan C, et al. Foxp3+IL-17+ T cells promote development of cancer-initiating cells in colorectal cancer. Journal of Leukocyte Biology. 2011; 89: 85–91.
  31. Halim L, Romano M, McGregor R, et al. An Atlas of Human Regulatory T Helper-like Cells Reveals Features of Th2-like Tregs that Support a Tumorigenic Environment. Cell Rep. 2017; 20: 757–770.
  32. Stavnezer J. Immunoglobulin class switching. Current Opinion in Immunology. 1996; 8: 199–205.
  33. Weaver CT, Harrington LE, Mangan PR, et al. Th17: an effector CD4 T cell lineage with regulatory T cell ties. Immunity. 2006; 24: 677–688.
  34. Haabeth OAW, Lorvik KB, Hammarström C, et al. Inflammation driven by tumour-specific Th1 cells protects against B-cell cancer. Nature Communications. 2011; 2: 240.
  35. Echchakir H, Bagot M, Dorothée G, et al. Cutaneous T cell lymphoma reactive CD4+cytotoxic T lymphocyte clones display a Th1 cytokine profile and use a Fas-independent pathway for specific tumor cell lysis. J Invest Dermatol. 2000; 115: 74–80.
  36. De Monte L, Reni M, Tassi E, et al. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J Exp Med. 2011; 208: 469–478.
  37. Ochi A, Nguyen AH, Bedrosian AS, et al. MyD88 inhibition amplifies dendritic cell capacity to promote pancreatic carcinogenesis via Th2 cells. J Exp Med. 2012; 209: 1671–1687.
  38. Gabitass RF, Annels NE, Stocken DD, et al. Elevated myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin-13. Cancer Immunol. Immunother. 2011; 60: 1419–1430.
  39. Enninga EAL, Nevala WK, Holtan SG, et al. Galectin-9 modulates immunity by promoting Th2/M2 differentiation and impacts survival in patients with metastatic melanoma. Melanoma Res. 2016; 26: 429–441.
  40. Lutgendorf SK, Lamkin DM, DeGeest K, et al. Depressed and anxious mood and T-cell cytokine expressing populations in ovarian cancer patients. Brain Behav Immun. 2008; 22: 890–900.
  41. Bailey SR. Nelson MH, Himes RA, et al. Th17 cells in cancer: The ultimate identity crisis. Front Immunol. 2014; 5: 276.
  42. Ward-Hartstonge KA, Kemp RA. Regulatory T-cell heterogeneity and the cancer immune response. Nature Publishing Group. 2017; 6: e154.
  43. Tirosh I, Izar B, Prakadan SM, et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science. 2016; 352: 189–196.
  44. Bernardini G, Hedrick J, Sozzani S, et al. Identification of the CC chemokines TARC and macrophage inflammatory protein-1 beta as novel functional ligands for the CCR8 receptor. Eur J Immunol. 1998; 28, 582–588.
  45. Plitas G, Konopacki C, Wu K, et al. Regulatory T Cells Exhibit Distinct Features in Human Breast Cancer. Immunity. 2016; 45: 1122–1134.
  46. De Simone M, Arrigoni A, Rossetti G, et al. Transcriptional Landscape of Human Tissue Lymphocytes Unveils Uniqueness of Tumor-Infiltrating T Regulatory Cells. Immunity. 2016; 45: 1135–1147.
  47. Joller N, Lozano E, Burkett PR, et al. Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses. Immunity. 2014; 40: 569–581.
  48. Wurtz O, Bajénoff M, Guerder S. IL-4-mediated inhibition of IFN-gamma production by CD4+ T cells proceeds by several developmentally regulated mechanisms. Int Immunol. 2004; 16: 501–508.
 

Article Info

Article Notes

  • Published on: January 18, 2018

Keywords

  • T helper-like regulatory cells

  • Immunoregulation
  • Tumor immunology

*Correspondence:

#Dr. Giovanna Lombardi
MRC Centre for Transplantation, School of Immunology & Microbial Sciences
King’s College London, Guy’s Hospital, SE1 9RT. London, United Kingdom
Telephone: (+44) 0207 1887674
Email: giovanna.lombardi@kcl.ac.uk
*Dr. Estefania Nova-Lampertis
Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology
Faculty of Pharmacy, University of Concepcion, 4070386. Concepcion, Chile
Telephone: (+56) 0412203167
Email: enova@udec.cls