Intratumoral FoxP3+Helios+ Regulatory T Cells Upregulating Immunosuppressive Molecules Are Expanded in Human Colorectal Cancer

Azharuddin Sajid Syed Khaja, Salman M Toor, Haytham El Salhat, Bassam R Ali, Eyad Elkord, Azharuddin Sajid Syed Khaja, Salman M Toor, Haytham El Salhat, Bassam R Ali, Eyad Elkord

Abstract

Regulatory T cells (Tregs) can be antitumorigenic or pro-tumorigenic in colorectal cancer (CRC) depending on the presence of different Treg subsets with various immunosuppressive molecules. Some studies reported the phenotypic characteristics of tumor-infiltrating immune cells in CRC, but limited studies have focused on the co-expression of suppressive molecules on immune cells. The aim of this study was to characterize immune cells in the tumor microenvironment (TME), compared to paired adjacent non-tumor colon tissue of CRC patients. Additionally, we investigated co-expression of immunosuppressive molecules on different Treg subsets in the TME, normal colon tissue, and peripheral blood of CRC patients and healthy donors. In this preliminary study, we report that the majority of CD3+ T cells in the TME are CD4+ T cells with high co-expression of programmed death 1 (PD-1)/cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and PD-1/CD39 molecules. Levels of CD4+FoxP3+Helios+ Tregs were significantly increased in the TME. Furthermore, we observed increased levels of PD-1/CTLA-4 and PD-1/CD39 co-expressing cells within FoxP3+Helios+ and FoxP3+Helios- Treg subsets, indicative of their potent immunosuppressive potential. These results suggest synergistic associations between PD-1/CTLA-4 and PD-1/CD39 in dampening T-cell activation and function along with suppressing tumor-specific immune responses, suggesting that dual blockade of these molecules could be a more effective strategy for inducing antitumor immune responses in CRC.

Keywords: Forkhead box protein 3; Helios; colorectal cancer; immune checkpoint receptors; regulatory T cells; tumor microenvironment.

Figures

Figure 1
Figure 1
Phenotypic characterization of CD4+ T cells in non-tumor-infiltrating lymphocytes (NILs) and tumor-infiltrating lymphocytes (TILs). Freshly isolated NILs and TILs were stained for CD25, LAP, CD39, and PD-1 surface markers and their frequencies were calculated in CD4+ T cells. (A) Representative flow cytometric plots for these markers in NILs and TILs from a cancer patient. (B) Scatter plots showing the differences between NILs and TILs.
Figure 2
Figure 2
Co-expression of PD-1/CTLA-4 and PD-1/CD39 in CD4+ T cells in healthy donor (HD) peripheral blood mononuclear cells (PBMCs), colorectal cancer (CRC) patients PBMCs, non-tumor-infiltrating lymphocytes (NILs), and tumor-infiltrating lymphocytes (TILs). PBMCs from HD and CRC patients, NILs, and TILs were stained for CD3, CD4, PD-1, and CD39. After fixation and permeabilization, cells were stained for CTLA-4 expression. Live cells were gated using Fixable Viability Dye 660. Representative flow cytometric plots showing PD-1 and CTLA-4 in CD4+ T cells (A) and whisker plots (B) showing differences in their expression in HD-PBMC, CRC-PBMC, NILs, and TILs. (C) Pie charts show the relative percentages of PD-1 and CTLA-4 in CD4+ T cells. Representative flow cytometric plots showing different subpopulations of PD-1 and CD39 in CD4+ T cells (D) and whisker plots (E) showing differences in their expression in different samples.
Figure 3
Figure 3
FoxP3 and Helios expression in CD4+ T cells. Peripheral blood mononuclear cells (PBMCs) from healthy donor and colorectal cancer (CRC) patients, non-tumor-infiltrating lymphocytes (NILs) and tumor-infiltrating lymphocytes (TILs) were stained for CD3 and CD4 followed by FoxP3 and Helios intracellular staining. Live cells were first gated using Fixable Viability Dye 660. Representative flow cytometric plots of FoxP3 staining are shown in (A). Adjacent Scatter plot shows the differences between frequencies of regulatory T cells (Tregs) based on FoxP3 expression between different samples. (B) Non-parametric Spearman’s test was performed to investigate correlations between FoxP3, Helios, and PD-1 expressions in CRC-PBMCs and TILs. (C) Flow cytometric plots of FoxP3 and Helios co-expression in Tregs from different samples, and the adjacent whisker plot showing differences in various subsets of FoxP3 and Helios between them. (D) Pie charts show the relative percentages of three different FoxP3 and Helios Treg subsets.
Figure 4
Figure 4
PD-1 and CTLA-4 co-expression in different FoxP3 and Helios regulatory T cell (Treg) subsets. (A) Representative flow cytometric plots showing PD-1 and CTLA-4 co-expression in different FoxP3 and Helios Treg subsets from healthy donor (HD)-peripheral blood mononuclear cell (PBMC), colorectal cancer (CRC)-PBMC, non-tumor-infiltrating lymphocytes (NILs), and tumor-infiltrating lymphocytes (TILs). Whisker plots comparing the relative levels of PD-1+CTLA-4+ cells (B) and PD-1+CTLA-4− cells (C) in FoxP3−Helios+, FoxP3+Helios+, and FoxP3+Helios− Treg subsets. (D) Absolute percentages of PD-1+CTLA-4+ and PD-1+CTLA-4− in different FoxP3 (F) and Helios (H) Treg subsets in TILs.
Figure 5
Figure 5
PD-1 and CD39 co-expression in different FoxP3 and Helios regulatory T cell (Treg) subsets. Representative flow cytometric plots showing PD-1 and CD39 expression in different FoxP3 and Helios Treg subsets from healthy donor (HD)-peripheral blood mononuclear cell (PBMC), colorectal cancer (CRC)-PBMC, non-tumor-infiltrating lymphocytes (NILs), and tumor-infiltrating lymphocytes (TILs) are shown in panel (A). Whisker plots comparing the levels of PD-1+CD39+ cells (B) and PD-1−CD39+ cells (C) in FoxP3−Helios+, FoxP3+Helios+, and FoxP3+Helios− Treg subsets.

References

    1. Chaudhary B, Elkord E. Regulatory T cells in the tumor microenvironment and cancer progression: role and therapeutic targeting. Vaccines (Basel) (2016) 4(3):E28.10.3390/vaccines4030028
    1. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. 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. J Immunol (1995) 155(3):1151–64.
    1. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science (2003) 299(5609):1057–61.10.1126/science.1079490
    1. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol (2003) 4(4):330–6.10.1038/ni904
    1. Thornton AM, Korty PE, Tran DQ, Wohlfert EA, Murray PE, Belkaid Y, et al. Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells. J Immunol (2010) 184(7):3433–41.10.4049/jimmunol.0904028
    1. Elkord E. Helios should not be cited as a marker of human thymus-derived Tregs. Commentary: Helios(+) and Helios(-) cells coexist within the natural FOXP3(+) T regulatory cell subset in humans. Front Immunol (2016) 7:276.10.3389/fimmu.2016.00276
    1. Verhagen J, Wraith DC. Comment on “expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells”. J Immunol (2010) 185(12):7129; author reply 30.10.4049/jimmunol.1090105
    1. Elkord E. Comment on “expression of Helios in peripherally induced Foxp3+ regulatory T cells”. J Immunol (2012) 189(2):500; author reply 1.10.4049/jimmunol.1290033
    1. Akimova T, Beier UH, Wang L, Levine MH, Hancock WW. Helios expression is a marker of T cell activation and proliferation. PLoS One (2011) 6(8):e24226.10.1371/journal.pone.0024226
    1. Kim HJ, Barnitz RA, Kreslavsky T, Brown FD, Moffett H, Lemieux ME, et al. Stable inhibitory activity of regulatory T cells requires the transcription factor Helios. Science (2015) 350(6258):334–9.10.1126/science.aad0616
    1. Elkord E, Abd Al Samid M, Chaudhary B. Helios, and not FoxP3, is the marker of activated Tregs expressing GARP/LAP. Oncotarget (2015) 6(24):20026–36.10.18632/oncotarget.4771
    1. Abd Al Samid M, Chaudhary B, Khaled YS, Ammori BJ, Elkord E. Combining FoxP3 and Helios with GARP/LAP markers can identify expanded Treg subsets in cancer patients. Oncotarget (2016) 7(12):14083–94.10.18632/oncotarget.7334
    1. Mandapathil M, Hilldorfer B, Szczepanski MJ, Czystowska M, Szajnik M, Ren J, et al. Generation and accumulation of immunosuppressive adenosine by human CD4+CD25highFOXP3+ regulatory T cells. J Biol Chem (2010) 285(10):7176–86.10.1074/jbc.M109.047423
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer (2012) 12(4):252–64.10.1038/nrc3239
    1. Read S, Greenwald R, Izcue A, Robinson N, Mandelbrot D, Francisco L, et al. Blockade of CTLA-4 on CD4+CD25+ regulatory T cells abrogates their function in vivo. J Immunol (2006) 177(7):4376–83.10.4049/jimmunol.177.7.4376
    1. Khan S, Burt DJ, Ralph C, Thistlethwaite FC, Hawkins RE, Elkord E. Tremelimumab (anti-CTLA4) mediates immune responses mainly by direct activation of T effector cells rather than by affecting T regulatory cells. Clin Immunol (2011) 138(1):85–96.10.1016/j.clim.2010.09.011
    1. Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev (2010) 236:219–42.10.1111/j.1600-065X.2010.00923.x
    1. Wang W, Lau R, Yu D, Zhu W, Korman A, Weber J. PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4+ CD25(Hi) regulatory T cells. Int Immunol (2009) 21(9):1065–77.10.1093/intimm/dxp072
    1. Borsellino G, Kleinewietfeld M, Di Mitri D, Sternjak A, Diamantini A, Giometto R, et al. Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: hydrolysis of extracellular ATP and immune suppression. Blood (2007) 110(4):1225–32.10.1182/blood-2006-12-064527
    1. Jie HB, Gildener-Leapman N, Li J, Srivastava RM, Gibson SP, Whiteside TL, et al. Intratumoral regulatory T cells upregulate immunosuppressive molecules in head and neck cancer patients. Br J Cancer (2013) 109(10):2629–35.10.1038/bjc.2013.645
    1. Bastid J, Cottalorda-Regairaz A, Alberici G, Bonnefoy N, Eliaou JF, Bensussan A. ENTPD1/CD39 is a promising therapeutic target in oncology. Oncogene (2013) 32(14):1743–51.10.1038/onc.2012.269
    1. Schuler PJ, Schilling B, Harasymczuk M, Hoffmann TK, Johnson J, Lang S, et al. Phenotypic and functional characteristics of CD4+ CD39+ FOXP3+ and CD4+ CD39+ FOXP3neg T-cell subsets in cancer patients. Eur J Immunol (2012) 42(7):1876–85.10.1002/eji.201142347
    1. Dunne MR, Ryan C, Nolan B, Tosetto M, Geraghty R, Winter DC, et al. Enrichment of inflammatory IL-17 and TNF-alpha secreting CD4(+) T cells within colorectal tumors despite the presence of elevated CD39(+) T regulatory cells and increased expression of the immune checkpoint molecule, PD-1. Front Oncol (2016) 6:50.10.3389/fonc.2016.00050
    1. Scurr M, Ladell K, Besneux M, Christian A, Hockey T, Smart K, et al. Highly prevalent colorectal cancer-infiltrating LAP(+) Foxp3(-) T cells exhibit more potent immunosuppressive activity than Foxp3(+) regulatory T cells. Mucosal Immunol (2014) 7(2):428–39.10.1038/mi.2013.62
    1. Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global patterns and trends in colorectal cancer incidence and mortality. Gut (2017) 66(4):683–91.10.1136/gutjnl-2015-310912
    1. Ling KL, Pratap SE, Bates GJ, Singh B, Mortensen NJ, George BD, et al. Increased frequency of regulatory T cells in peripheral blood and tumour infiltrating lymphocytes in colorectal cancer patients. Cancer Immun (2007) 7:7.
    1. Ladoire S, Martin F, Ghiringhelli F. Prognostic role of FOXP3+ regulatory T cells infiltrating human carcinomas: the paradox of colorectal cancer. Cancer Immunol Immunother (2011) 60(7):909–18.10.1007/s00262-011-1046-y
    1. Betts G, Jones E, Junaid S, El-Shanawany T, Scurr M, Mizen P, et al. Suppression of tumour-specific CD4(+) T cells by regulatory T cells is associated with progression of human colorectal cancer. Gut (2012) 61(8):1163–71.10.1136/gutjnl-2011-300970
    1. Clarke SL, Betts GJ, Plant A, Wright KL, El-Shanawany TM, Harrop R, et al. CD4+CD25+FOXP3+ regulatory T cells suppress anti-tumor immune responses in patients with colorectal cancer. PLoS One (2006) 1:e129.10.1371/journal.pone.0000129
    1. Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity (2009) 30(6):899–911.10.1016/j.immuni.2009.03.019
    1. Saito T, Nishikawa H, Wada H, Nagano Y, Sugiyama D, Atarashi K, et al. Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med (2016) 22(6):679–84.10.1038/nm.4086
    1. Toor S, Syed Khaja AS, El Salhat H, Bekdache O, Kanbar J, Jaloudi M, et al. Increased levels of circulating and tumor-infiltrating granulocytic myeloid cells in colorectal cancer patients. Front Immunol (2016) 7:560.10.3389/fimmu.2016.00560
    1. Timperi E, Pacella I, Schinzari V, Focaccetti C, Sacco L, Farelli F, et al. Regulatory T cells with multiple suppressive and potentially pro-tumor activities accumulate in human colorectal cancer. Oncoimmunology (2016) 5(7):e1175800.10.1080/2162402X.2016.1175800
    1. Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol (2016) 39(1):98–106.10.1097/COC.0000000000000239
    1. Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pages C, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science (2006) 313(5795):1960–4.10.1126/science.1129139
    1. Bevan MJ. Helping the CD8(+) T-cell response. Nat Rev Immunol (2004) 4(8):595–602.10.1038/nri1413
    1. Liu VC, Wong LY, Jang T, Shah AH, Park I, Yang X, et al. Tumor evasion of the immune system by converting CD4+CD25-T cells into CD4+CD25+ T regulatory cells: role of tumor-derived TGF-beta. J Immunol (2007) 178(5):2883–92.10.4049/jimmunol.178.5.2883
    1. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature (2006) 439(7077):682–7.10.1038/nature04444
    1. Zhang Y, Huang S, Gong D, Qin Y, Shen Q. Programmed death-1 upregulation is correlated with dysfunction of tumor-infiltrating CD8+ T lymphocytes in human non-small cell lung cancer. Cell Mol Immunol (2010) 7(5):389–95.10.1038/cmi.2010.28
    1. Duraiswamy J, Kaluza KM, Freeman GJ, Coukos G. Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Res (2013) 73(12):3591–603.10.1158/0008-5472.CAN-12-4100
    1. Getnet D, Grosso JF, Goldberg MV, Harris TJ, Yen HR, Bruno TC, et al. A role for the transcription factor Helios in human CD4(+)CD25(+) regulatory T cells. Mol Immunol (2010) 47(7–8):1595–600.10.1016/j.molimm.2010.02.001
    1. Ondondo B, Jones E, Godkin A, Gallimore A. Home sweet home: the tumor microenvironment as a haven for regulatory T cells. Front Immunol (2013) 4:197.10.3389/fimmu.2013.00197
    1. Nikolova M, Carriere M, Jenabian MA, Limou S, Younas M, Kok A, et al. CD39/adenosine pathway is involved in AIDS progression. PLoS Pathog (2011) 7(7):e1002110.10.1371/journal.ppat.1002110
    1. Callahan MK, Wolchok JD. At the bedside: CTLA-4- and PD-1-blocking antibodies in cancer immunotherapy. J Leukoc Biol (2013) 94(1):41–53.10.1189/jlb.1212631
    1. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med (2012) 366(26):2443–54.10.1056/NEJMoa1200690
    1. Grosso JF, Jure-Kunkel MN. CTLA-4 blockade in tumor models: an overview of preclinical and translational research. Cancer Immun (2013) 13:5.
    1. Callahan MK, Postow MA, Wolchok JD. CTLA-4 and PD-1 pathway blockade: combinations in the clinic. Front Oncol (2014) 4:385.10.3389/fonc.2014.00385
    1. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med (2013) 369(2):122–33.10.1056/NEJMoa1302369
    1. Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A (2010) 107(9):4275–80.10.1073/pnas.0915174107

Source: PubMed

3
Suscribir