The Paradigm of T Cells in Shaping Tumor Microenvironment

Dia Roy ( Bose Institute )

Sayantan Bose ( Bose Institute )

Saikat Dutta ( Bose Institute )

Gaurisankar Sa ( Bose Institute )

https://doi.org/10.37155/2717-5278-0202-2

Abstract

The infiltration of immune cells in the tumor micro-environment is well-documented in cancer patients and the resultant complex interactions are determinants of disease prognosis. Consequently, a proper understanding of this interplay is essential for the development and advancement of therapeutic strategies as well as novel prognostic markers. The co-existence of immune cells with the tumor is often accompanied by an impaired immune response that creates a tumor-promoting micro-environment. T-cell mediated immunity forms the major branch of the immune system that is required to mount an effective response against nascent tumors. Major research in the last few decades indicates that a potent source of immunosuppressive cellular and molecular networks prevailing at the site of tumor is mediated by dysfunctional and defective responses mediated by T cell thereby redirecting and reshaping the destiny of T cell and promoting tumor progression. In this review, we aim to summarize the breakthrough advances in recent years that help us gain a better understanding of the immunosuppressive networks resulting from T-cell anergy, exhaustion, senescence and presence of Treg cells in the tumor micro-environment. We also focus on recent discoveries regarding advance in the Th17 balance, polyfunctionality of T cells as well as T cell stemness that improve our perception of the tumor-immunity interactions. We try to emphasize how such information has an impact on therapeutic development and the clinical outcome of the patients.

Keywords

T cell;; Tumor infiltrating lymphocytes;; Tumor microenvironment;; Treg;; CART cell

Full Text

PDF

References

[1] Roma-Rodrigues, C., Mendes, R., Baptista, P. V. & Fernandes, A. R. Targeting Tumor Microenvironment for Cancer Therapy. Int. J. Mol. Sci, 2019, 20(4).
[2] Cooper, G. M. The Development and Causes of Cancer. Cell Mol. Approach 2nd Ed, 2000.
[3] Dvorak, H. F. Tumors: Wounds that do not heal--Redux. Cancer Immunol. Res, 2015, 3. 1-11.
[4] Whiteside, T. The tumor microenvironment and its role in promoting tumor growth. Oncogene,2008, 27, 5904–5912.
[5] Whiteside, T. L. Immune responses to malignancies. J. Allergy Clin. Immunol. 2010, 125, S272–S283.
[6] Dunn, G. P., Old, L. J. & Schreiber, R. D. The three Es of cancer immunoediting. Annu. Rev. Immunol. 2004, 22, 329–360.
[7] Roles of the immune system in cancer: from tumor initiation to metastatic progression. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6169832/.
[8] Seung, L. P., Seung, S. K. & Schreiber, H. Antigenic cancer cells that escape immune destruction are stimulated by host cells. Cancer Res. 1995, 55, 5094–5100.
[9] Fouad, Y. A. & Aanei, C. Revisiting the hallmarks of cancer. Am. J. Cancer Res. 2017,7, 1016–1036.
[10] Greten, T. F., Mauda-Havakuk, M., Heinrich, B., Korangy, F. & Wood, B. J. Combined locoregional-immunotherapy for liver cancer. J. Hepatol. 2019,70, 999–1007 .
[11] Thommen, D. S. & Schumacher, T. N. T Cell Dysfunction in Cancer. Cancer Cell 2018, 33, 547–562.
[12] Liechtenstein, T., Dufait, I., Lanna, A., Breckpot, K. & Escors, D. MODULATING CO-STIMULATION DURING ANTIGEN PRESENTATION TO ENHANCE CANCER IMMUNOTHERAPY. Immunol. Endocr. Metab. Agents Med. Chem.2012, 12, 224–235.
[13] Okoye, I. S., Houghton, M., Tyrrell, L., Barakat, K. & Elahi, S. Coinhibitory Receptor Expression and Immune Checkpoint Blockade: Maintaining a Balance in CD8+ T Cell Responses to Chronic Viral Infections and Cancer. Front. Immunol. 2017,8.
[14] Teague, R. M. & Kline, J. Immune evasion in acute myeloid leukemia: current concepts and future directions. J. Immunother. Cancer 2013,1, 13.
[15] Bercovici, N. & Trautmann, A. Revisiting the role of T cells in tumor regression. Oncoimmunology , 2012,1, 346–350.
[16] Zamarron, B. F. & Chen, W. Dual Roles of Immune Cells and Their Factors in Cancer Development and Progression. Int. J. Biol. Sci. 2011,7, 651–658.
[17] Muz, B., de la Puente, P., Azab, F., & Azab, A. K.. The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy. Hypoxia (Auckland, N.Z.), 2015,3, 83–92. https://doi.org/10.2147/HP.S93413
[18] Meng, S., Li, L., Zhou, M., Jiang, W., Niu, H., & Yang, K.. Distribution and prognostic value of tumorinfiltrating T cells in breast cancer. Molecular medicine reports, 2018, 18(5), 4247–4258. https://doi.org/10.3892/mmr.2018.9460
[19] Direct activation of antigen-presenting cells is required for CD8+ T-cell priming and tumor vaccination. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3198339/.
[20] Kaiko, G. E., Horvat, J. C., Beagley, K. W. & Hansbro, P. M. Immunological decision-making: how does the immune system decide to mount a helper T-cell response? Immunology 2008, 123, 326–338.
[21] Xia, A., Zhang, Y., Xu, J., Yin, T. & Lu, X.-J. T Cell Dysfunction in Cancer Immunity and Immunotherapy. Front. Immunol. 2019,10.
[22] Murray, T. et al. Very Late Antigen-1 Marks Functional Tumor-Resident CD8 T Cells and Correlates with Survival of Melanoma Patients. Front. Immunol. 2016,7.
[23] Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4009604/.
[24] Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases - PubMed. https://pubmed.ncbi.nlm.nih.gov/7636184/.
[25] Baecher-Allan, C., Viglietta, V. & Hafler, D. A. Human CD4+CD25+ regulatory T cells. Semin. Immunol. 2004,16, 89–98.
[26] Devaud, C., Darcy, P. K. & Kershaw, M. H. Foxp3 expression in T regulatory cells and other cell lineages. Cancer Immunol. Immunother. CII 2014, 63, 869–876.
[27] Plitas, G. & Rudensky, A. Y. Regulatory T Cells: Differentiation and Function. Cancer Immunol. Res. 2016, 4, 721–725.
[28] Nair, V. S., Song, M. H., Ko, M. & Oh, K. I. DNA Demethylation of the Foxp3 Enhancer Is Maintained through Modulation of Ten-Eleven-Translocation and DNA Methyltransferases. Mol. Cells 2016, 39, 888–897.
[29] Liu, W., Putnam, A. L., Xu-Yu, Z., Szot, G. L., Lee, M. R., Zhu, S., Gottlieb, P. A., Kapranov, P., Gingeras, T. R., Fazekas de St Groth, B., Clayberger, C., Soper, D. M., Ziegler, S. F., & Bluestone, J. A. . CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. The Journal of experimental medicine, 2006,203(7), 1701–1711. https://doi.org/10.1084/jem.20060772
[30] Bilate, A. M. & Lafaille, J. J. Induced CD4+Foxp3+ Regulatory T Cells in Immune Tolerance. Annu. Rev. Immunol. 2012, 30, 733–758.
[31] Povoleri, G. A., Scottà, C., Nova-Lamperti, E. A., John, S., Lombardi, G., & Afzali, B.. Thymic versus induced regulatory T cells - who regulates the regulators?. Frontiers in immunology, 4, 169. https://doi.org/10.3389/fimmu.2013.00169
[32] Cheng, G., Yu, A. & Malek, T. R. T cell tolerance and the multi-functional role of IL-2R signaling in T regulatory cells. Immunol. Rev. 2011,241, 63–76.
[33] Gol-Ara, M., Jadidi-Niaragh, F., Sadria, R., Azizi, G. & Mirshafiey, A. The Role of Different Subsets of Regulatory T Cells in Immunopathogenesis of Rheumatoid Arthritis. Arthritis 2012.
[34] Thornton, A. M., & Shevach, E. M. (1998). CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. The Journal of experimental medicine, 188(2), 287–296. https://doi.org/10.1084/jem.188.2.287
[35] Paluskievicz, C. M. et al. T Regulatory Cells and Priming the Suppressive Tumor Microenvironment. Front. Immunol. 2019, 10.
[36] Sojka, D. K., Huang, Y.-H. & Fowell, D. J. Mechanisms of regulatory T-cell suppression – a diverse arsenal for a moving target. Immunology 2008, 124, 13–22.
[37] Sansom, D. M. CD28, CTLA-4 and their ligands: who does what and to whom? Immunology 2000, 101, 169–177.
[38] Zhai, L., Bell, A., Ladomersky, E., Lauing, K. L., Bollu, L., Sosman, J. A., Zhang, B., Wu, J. D., Miller, S. D., Meeks, J. J., Lukas, R. V., Wyatt, E., Doglio, L., Schiltz, G. E., McCusker, R. H., & Wainwright, D. A.. Immunosuppressive IDO in Cancer: Mechanisms of Action, Animal Models, and Targeting Strategies. Frontiers in immunology, 11, 1185. https://doi.org/10.3389/fimmu.2020.01185
[39] Corthay A.. How do regulatory T cells work?. Scandinavian journal of immunology, 2009, 70(4), 326–336. https://doi.org/10.1111/j.1365-3083.2009.02308.x
[40] Schmidt, A., Oberle, N., & Krammer, P. H. (2012). Molecular mechanisms of treg-mediated T cell suppression. Frontiers in immunology, 3, 51. https://doi.org/10.3389/fimmu.2012.00051
[41] Sawant, D. V., Hamilton, K., & Vignali, D. A. (2015). Interleukin-35: Expanding Its Job Profile. Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research, 35(7), 499–512. https://doi.org/10.1089/jir.2015.0015
[42] Cedeno-Laurent, F., Opperman, M., Barthel, S. R., Kuchroo, V. K., & Dimitroff, C. J. (2012). Galectin-1 triggers an immunoregulatory signature in Th cells functionally defined by IL-10 expression. Journal of immunology (Baltimore, Md. : 1950), 188(7), 3127–3137. https://doi.org/10.4049/jimmunol.1103433
[43] Cedeno-Laurent, F., Opperman, M., Barthel, S. R., Kuchroo, V. K. & Dimitroff, C. J. Galectin-1 triggers an immunoregulatory signature in T helper cells functionally defined by IL-10 expression. J. Immunol. Baltim. Md 1950 ,2012, 188, 3127–3137.
[44] Whiteside, T. L. & Jackson, E. K. Adenosine and Prostaglandin E2 Production by Human Inducible Regulatory T Cells in Health and Disease. Front. Immunol. 2013, 4.
[45] Mandapathil, M. et al. Increased ectonucleotidase expression and activity in Treg of patients with head and neck cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2009, 15, 6348–6357.
[46] Levring, T. B. et al. Activated human CD4+ T cells express transporters for both cysteine and cystine. Sci. Rep. 2012,2.
[47] Yan, Z., Garg, S. K. & Banerjee, R. Regulatory T Cells Interfere with Glutathione Metabolism in Dendritic Cells and T Cells. J. Biol. Chem. 2010,285, 41525–41532.
[48] Liu, X., Mo, W., Ye, J., Li, L., Zhang, Y., Hsueh, E. C., Hoft, D. F., & Peng, G.. Regulatory T cells trigger effector T cell DNA damage and senescence caused by metabolic competition. Nature communications, 2018, 9(1), 249. https://doi.org/10.1038/s41467-017-02689-5
[49] McGeachy, M. J., & Cua, D. J.. Th17 cell differentiation: the long and winding road. Immunity, 28(4), 445–453. https://doi.org/10.1016/j.immuni.2008.03.001
[50] Brembilla, N. C., Senra, L. & Boehncke, W.-H. The IL-17 Family of Cytokines in Psoriasis: IL-17A and Beyond. Front. Immunol. 2018,9.
[51] Zhu, J. & Paul, W. E. Peripheral CD4 T cell differentiation regulated by networks of cytokines and transcription factors. Immunol. Rev. 2010, 238, 247–262.
[52] Tsai, H.-C., Velichko, S., Hung, L.-Y. & Wu, R. IL-17A and Th17 Cells in Lung Inflammation: An Update on the Role of Th17 Cell Differentiation and IL-17R Signaling in Host Defense against Infection. Clinical and Developmental Immunology vol. 2013 e267971 https://www.hindawi.com/journals/jir/2013/267971/ (2013).
[53] Waite, J. C. & Skokos, D. Th17 Response and Inflammatory Autoimmune Diseases. International Journal of Inflammation vol. 2012 e819467 https://www.hindawi.com/journals/iji/2012/819467/ (2011).
[54] Chen, X. & Oppenheim, J. J. Th17 cells and Tregs: unlikely allies. J. Leukoc. Biol. 2014,95, 723–731.
[55] Du, R., Zhao, H., Yan, F. & Li, H. IL-17+Foxp3+ T cells: an intermediate differentiation stage between Th17 cells and regulatory T cells. J. Leukoc. Biol. 2014,96, 39–48.
[56] Ye, J., Livergood, R. S. & Peng, G. The Role and Regulation of Human Th17 Cells in Tumor Immunity. Am. J. Pathol. 2013, 182, 10–20.
[57] Stadhouders, R., Lubberts, E. & Hendriks, R. W. A cellular and molecular view of T helper 17 cell plasticity in autoimmunity. J. Autoimmun. 2018, 87, 1–15.
[58] Martin, F., Apetoh, L. & Ghiringhelli, F. Controversies on the role of Th17 in cancer: a TGF-β-dependent immunosuppressive activity? Trends Mol. Med. 2012, 18, 742–749.
[59] Lee, G. R. The Balance of Th17 versus Treg Cells in Autoimmunity. Int. J. Mol. Sci. 2018, 19.
[60] Knochelmann, H. M. et al. When worlds collide: Th17 and Treg cells in cancer and autoimmunity. Cell. Mol. Immunol. 2018, 15, 458–469.
[61] Duan, M. C., Zhong, X. N., Liu, G. N., & Wei, J. R. (2014). The Treg/Th17 paradigm in lung cancer. Journal of immunology research, 2014, 730380. https://doi.org/10.1155/2014/730380
[62] De Simone, V., Pallone, F., Monteleone, G. & Stolfi, C. Role of TH17 cytokines in the control of colorectal cancer. Oncoimmunology 2013, 2.
[63] Martiniuk, F. et al. TH17 is involved in the remarkable regression of metastatic malignant melanoma to topical diphencyprone. J. Drugs Dermatol. JDD 2010, 9, 1368–1372.
[64] Lippens, C., Duraes, F. V., Dubrot, J., Brighouse, D., Lacroix, M., Irla, M., Aubry-Lachainaye, J. P., Reith, W., Mandl, J. N., & Hugues, S. (2016). IDO-orchestrated crosstalk between pDCs and Tregs inhibits autoimmunity. Journal of autoimmunity, 75, 39–49. https://doi.org/10.1016/j.jaut.2016.07.004
[65] Guéry, L. & Hugues, S. Th17 Cell Plasticity and Functions in Cancer Immunity. BioMed Research International vol. 2015 e314620 https://www.hindawi.com/journals/bmri/2015/314620/ (2015).
[66] Chatterjee, S., Thyagarajan, K., Kesarwani, P., Song, J. H., Soloshchenko, M., Fu, J., Bailey, S. R., Vasu, C., Kraft, A. S., Paulos, C. M., Yu, X. Z., & Mehrotra, S. (2014). Reducing CD73 expression by IL1β-Programmed Th17 cells improves immunotherapeutic control of tumors. Cancer research, 74(21), 6048–6059. https://doi.org/10.1158/0008-5472.CAN-14-1450
[67] Hayata, K. et al. Inhibition of IL-17A in Tumor Microenvironment Augments Cytotoxicity of Tumor-Infiltrating Lymphocytes in Tumor-Bearing Mice. PLoS ONE ,2013, 8.
[68] Alizadeh, D., Katsanis, E. & Larmonier, N. The Multifaceted Role of Th17 Lymphocytes and Their Associated Cytokines in Cancer. Clin. Dev. Immunol. 2013.10. 1-11.
[69] Wilke, C. M., Kryczek, I., Wei, S., Zhao, E., Wu, K., Wang, G., & Zou, W. (2011). Th17 cells in cancer: help or hindrance? Carcinogenesis, 32(5), 643–649. https://doi.org/10.1093/carcin/bgr019
[70] Karin, N. CXCR3 Ligands in Cancer and Autoimmunity, Chemoattraction of Effector T Cells, and Beyond. Front. Immunol. 2020,11.
[71] Guéry, L. & Hugues, S. Th17 Cell Plasticity and Functions in Cancer Immunity. BioMed Research International vol. 2015 e314620 https://www.hindawi.com/journals/bmri/2015/314620/ (2015).
[72] Schwartz, R. H. T cell anergy. Annu. Rev. Immunol. 2003, 21, 305–334.
[73] Lechler, R., Chai, J.-G., Marelli-Berg, F. & Lombardi, G. The contributions of T-cell anergy to peripheral T-cell tolerance. Immunology 2001, 103, 262–269.
[74] Nurieva, R., Wang, J., & Sahoo, A. (2013). T-cell tolerance in cancer. Immunotherapy, 5(5), 513–531. https://doi.org/10.2217/imt.13.33
[75] Jung, K. & Choi, I. Emerging Co-signaling Networks in T Cell Immune Regulation. Immune Netw. 2013,13, 184–193.
[76] Wherry, E. J. & Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499.
[77] Riley, J. L. PD-1 signaling in primary T cells. Immunol. Rev. 2009, 229, 114–125.
[78] Zhao, Y., Shao, Q. & Peng, G. Exhaustion and senescence: two crucial dysfunctional states of T cells in the tumor microenvironment. Cell. Mol. Immunol. 2020, 17, 27–35.
[79] Li, L., Wan, S., Tao, K., Wang, G. & Zhao, E. KLRG1 restricts memory T cell antitumor immunity. Oncotarget 2016, 7, 61670–61678.
[80] Li, H. et al. Tim-3/galectin-9 signaling pathway mediates T-cell dysfunction and predicts poor prognosis in patients with hepatitis B virus-associated hepatocellular carcinoma. Hepatology 2012, 56, 1342–1351.
[81] Gattinoni L.. Memory T cells officially join the stem cell club. Immunity, 2014, 41(1), 7–9. https://doi.org/10.1016/j.immuni.2014.07.003
[82] Pennock, N. D. et al. T cell responses: naïve to memory and everything in between. Adv. Physiol. Educ. 2013, 37, 273–283.
[83] Steinbach, K., Vincenti, I. & Merkler, D. Resident-Memory T Cells in Tissue-Restricted Immune Responses: For Better or Worse? Front. Immunol. 2018, 9.
[84] Samji, T. & Khanna, K. M. Understanding Memory CD8+ T cells. Immunol. Lett. 2017, 185, 32–39.
[85] Kagoya, Y., Nakatsugawa, M., Ochi, T., Cen, Y., Guo, T., Anczurowski, M., Saso, K., Butler, M. O., & Hirano, N. (2017). Transient stimulation expands superior antitumor T cells for adoptive therapy. JCI insight, 2(2), e89580. https://doi.org/10.1172/jci.insight.89580
[86] Tanel, A., Fonseca, S. G., Yassine-Diab, B., Bordi, R., Zeidan, J., Shi, Y., Benne, C., & Sékaly, R. P. (2009). Cellular and molecular mechanisms of memory T-cell survival. Expert review of vaccines, 8(3), 299–312. https://doi.org/10.1586/14760584.8.3.299
[87] Muranski, P., Borman, Z. A., Kerkar, S. P., Klebanoff, C. A., Ji, Y., Sanchez-Perez, L., Sukumar, M., Reger, R. N., Yu, Z., Kern, S. J., Roychoudhuri, R., Ferreyra, G. A., Shen, W., Durum, S. K., Feigenbaum, L., Palmer, D. C., Antony, P. A., Chan, C. C., Laurence, A., Danner, R. L., … Restifo, N. P. . Th17 cells are long lived and retain a stem cell-like molecular signature. Immunity, 2011, 35(6), 972–985. https://doi.org/10.1016/j.immuni.2011.09.019
[88] Wei, S., Zhao, E., Kryczek, I. & Zou, W. Th17 cells have stem cell-like features and promote long-term immunity. Oncoimmunology 2012, 1, 516–519.
[89] Zhu, J., Yamane, H., & Paul, W. E.. Differentiation of effector CD4 T cell populations (*). Annual review of immunology, 2010, 28, 445–489. https://doi.org/10.1146/annurev-immunol-030409-101212
[90] De Groot, R., Van Loenen, M. M., Guislain, A., Nicolet, B. P., Freen-Van Heeren, J. J., Verhagen, O., Van Den Heuvel, M. M., De Jong, J., Burger, P., Van Der Schoot, C. E., Spaapen, R. M., Amsen, D., Haanen, J., Monkhorst, K., Hartemink, K. J., & Wolkers, M. C. (2019). Polyfunctional tumor-reactive T cells are effectively expanded from non-small cell lung cancers, and correlate with an immune-engaged T cell profile. Oncoimmunology, 8(11), e1648170. https://doi.org/10.1080/2162402X.2019.1648170
[91] Burel, J. G., Apte, S. H., Groves, P. L., McCarthy, J. S., & Doolan, D. L. (2017). Polyfunctional and IFN-γ monofunctional human CD4+ T cell populations are molecularly distinct. JCI insight, 2(3), e87499. https://doi.org/10.1172/jci.insight.87499
[92] Soghoian, D. Z., & Streeck, H. (2010). Cytolytic CD4(+) T cells in viral immunity. Expert review of vaccines, 9(12), 1453–1463. https://doi.org/10.1586/erv.10.132
[93] Salido, J., Ruiz, M. J., Trifone, C., Figueroa, M. I., Caruso, M. P., Gherardi, M. M., Sued, O., Salomón, H., Laufer, N., Ghiglione, Y., & Turk, G. (2018). Phenotype, Polyfunctionality, and Antiviral Activity of in vitro Stimulated CD8+ T-Cells From HIV+ Subjects Who Initiated cART at Different Time-Points After Acute Infection. Frontiers in immunology, 9, 2443. https://doi.org/10.3389/fimmu.2018.02443
[94] Bhat, P., Leggatt, G., Waterhouse, N., & Frazer, I. H. (2017). Interferon-γ derived from cytotoxic lymphocytes directly enhances their motility and cytotoxicity. Cell death & disease, 8(6), e2836. https://doi.org/10.1038/cddis.2017.67
[95] Ding, Z. C., Huang, L., Blazar, B. R., Yagita, H., Mellor, A. L., Munn, D. H., & Zhou, G. (2012). Polyfunctional CD4⁺ T cells are essential for eradicating advanced B-cell lymphoma after chemotherapy. Blood, 120(11), 2229–2239. https://doi.org/10.1182/blood-2011-12-398321
[96] Seidel, J. A., Otsuka, A., & Kabashima, K. (2018). Anti-PD-1 and Anti-CTLA-4 Therapies in Cancer: Mechanisms of Action, Efficacy, and Limitations. Frontiers in oncology, 8, 86. https://doi.org/10.3389/fonc.2018.00086
[97] Gallimore, A., & Godkin, A. (2008). Regulatory T cells and tumour immunity - observations in mice and men. Immunology, 123(2), 157–163. https://doi.org/10.1111/j.1365-2567.2007.02748.
[98] Kim, J. H., Kim, B. S., & Lee, S. K. (2020). Regulatory T Cells in Tumor Microenvironment and Approach for Anticancer Immunotherapy. Immune network, 20(1), e4. https://doi.org/10.4110/in.2020.20.e4
[99] Toor, S. M., Murshed, K., Al-Dhaheri, M., Khawar, M., Abu Nada, M., & Elkord, E. (2019). Immune Checkpoints in Circulating and Tumor-Infiltrating CD4+ T Cell Subsets in Colorectal Cancer Patients. Frontiers in immunology, 10, 2936. https://doi.org/10.3389/fimmu.2019.02936
[100] Ohue, Y. & Nishikawa, H. Regulatory T (Treg) cells in cancer: Can Treg cells be a new therapeutic target? Cancer Sci. 2019, 110, 2080–2089.
[101] Han, S., Toker, A., Liu, Z. Q. & Ohashi, P. S. Turning the Tide Against Regulatory T Cells. Front. Oncol. 2019, 9.
[102] Dai, H., Wang, Y., Lu, X. & Han, W. Chimeric Antigen Receptors Modified T-Cells for Cancer Therapy. JNCI J. Natl. Cancer Inst. 2016, 108.

Copyright © 2020 Dia Roy, Sayantan Bose, Saikat Dutta, Gaurisankar Sa Creative Commons License Publishing time:2020-12-10
This work is licensed under a Creative Commons Attribution 4.0 International License