Investigation of the prognostic value of CD4 T cell subsets expanded from tumor-infiltrating lymphocytes of colorectal cancer liver metastases

Marie Kroemer, Celia Turco, Laurie Spehner, Julien Viot, Idir Idirène, Adeline Bouard, Elodie Renaude, Marina Deschamps, Yann Godet, Olivier Adotévi, Samuel Limat, Bruno Heyd, Marine Jary, Romain Loyon, Christophe Borg, Marie Kroemer, Celia Turco, Laurie Spehner, Julien Viot, Idir Idirène, Adeline Bouard, Elodie Renaude, Marina Deschamps, Yann Godet, Olivier Adotévi, Samuel Limat, Bruno Heyd, Marine Jary, Romain Loyon, Christophe Borg

Abstract

Background: The positive role of CD8+ tumor-infiltrating lymphocytes (TIL) in patients with colorectal cancer (CRC) has been well described but the prognostic value of CD4 T cell subsets remained to be investigated. In this study, we expanded TIL from surgically resected liver metastases of patients with CRC and characterized the phenotype and the prognostic value of expanded-CD4 T cells.

Methods: Liver metastases were surgically resected from 23 patients with CRC. Tumors were enzymatically digested and cultured in high dose of interleukin-2 for up to 5 weeks. T cell phenotype and reactivity of cultured-T cells were measured by flow cytometry and correlated with patients' clinical outcomes.

Results: We successfully expanded 21 over 23 TIL from liver metastases of patients with CRC. Interestingly, we distinguished two subsets of expanded T cells based on T cell immunoglobulin mucin domain-containing protein 3 (TIM-3) expression. Medians fold expansion of expanded T cells after rapid expansion protocol was higher in CD3+TIM-3low cultures. In an attempt to investigate the correlation between the phenotype of expanded CD4 T cells and clinical outcomes, we observed on one hand that the level of Tregs in culture as well as the expression of both PD1 and TIM-3 by expanded T cells was not correlated to the clinical outcomes. Interestingly, on the other hand, cultures containing high levels of Th17 cells were associated with a poor prognosis (p=0.0007).

Conclusions: Our data confirmed the presence of Th17 cells in expanded T cells from liver metastases. Among CD4 T cell characteristics investigated, TIM-3 but not programmed cell death protein 1 predicted the expansion capacity of TIL while only the Th17 phenotype showed correlation with patients' survival, suggesting a particular role of this T cell subset in CRC immune contexture.

Trial registration number: NCT02817178.

Keywords: CD4-positive t-lymphocytes; adaptive immunity; biomarkers; gastrointestinal neoplasms; lymphocytes; tumor; tumor-infiltrating.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Flow chart of available material and analysis plan. mCRC, metastatic colorectal cancer; REP, rapid expansion protocol; TIL, tumor-infiltrating lymphocytes.
Figure 2
Figure 2
Fold expansion and characteristics of expanded tumor-infiltrating lymphocytes (TIL). (A) Scheme of isolation and expansion of liver metastases-derived TIL. (B) Median fold expansion after rapid expansion protocol (REP) (3438 (451–11 118)). (C, D) Frequencies of CD4 and CD8 T cells at three major steps : isolation day (n=10), Pre-REP (n=21) and Post-REP (n=21). (E, F) Median frequencies of CD28, respectively, on CD4 and CD8 T cells before REP (94.9 (31.6–99.4), 93.7 (19.0–99.0)) and after (58.1 (5.4–98.3), 63.3 (7.3–87.5)). (G) Gating strategy of REP T-cells memory profile. (H, I) CD8 and CD4 T cells memory profile after REP. (J) Unsupervised hierarchical clustering of the percentages of CD4 and CD8 T cells expressing T cell immunoglobulin mucin domain-containing protein 3 and programmed cell death protein 1, CD4 T cells polarization and Treg level (CD4+CD25HighFoxP3+CD127−) from pre-REP T cells. Statistical differences were calculated using Mann-Whitney test. Median (min−max). ns, not significant.
Figure 3
Figure 3
T cell immunoglobulin mucin domain-containing protein 3 (TIM-3) expression discriminates T-cell cultures fold expansion. (A) Gating strategy of inhibitory receptor expression profile on pre-rapid expansion protocol (REP) T-cells. (B, C) CTLA4, TIM-3 and programmed cell death protein 1 (PD1) expression (%) on CD4 and CD8 T cells were, respectively, 1.7% (0.1–7.2) and 1.3% (0.0–13.7), 56.6% (3.2–96.2) and 45.8% (6.7–99.5), 47.8% (21.7–86.4) and 57.1% (11.2–87.0). (D, E) Medians for PD1 expression (%) before and after REP, respectively, on CD4 (47.3 (21.7–86.4) and 33.4 (10.7–63.8) (p=0.0026)) and CD8 T cells (56.6 (11.2–87.0) and 21.2 (4.6–44.7) (p

Figure 4

Functional potential of rapid expansion…

Figure 4

Functional potential of rapid expansion protocol (REP) T-cells. REP T cells were subjected…

Figure 4
Functional potential of rapid expansion protocol (REP) T-cells. REP T cells were subjected to a 16 hours CD3/28 bead stimulation (1:1) and analyzed by flow cytometry using intracellular staining for Th1 cytokines (IFN-γ, TNF-α and interleukin-2 (IL-2)) gating on CD4 and CD8 T cell populations (n=11). (A, B) IFN-γ, TNF-α and IL-2 expression by (A) CD4 and CD8 REP T cells and (B) according to TIM-3 expression. (C) CD107a production by previously stimulated CD8 REP T cells (n=8). Median (%) with 95 CI or median (%) (min–max). Mann-Whitney tests: *p

Figure 5

Characterization of Treg within T-cell…

Figure 5

Characterization of Treg within T-cell cultures. (A) Regulatory T cells (Treg) was investigated…

Figure 5
Characterization of Treg within T-cell cultures. (A) Regulatory T cells (Treg) was investigated by intracellular staining for FoxP3 on the CD4+CD25HighCD127− population before and after the rapid expansion protocol (REP) (p=0.2921). (B) Median fold expansion in Treg low and in Treg high culture. There was no significant difference (p=0.5920) (C) Percentage of Treg before and after REP according to T cell immunoglobulin mucin domain-containing protein 3 (TIM-3) expression on CD8 T cells. (D) Glycoprotein-A repetitions predominant (GARP) expression (%) on Treg within peripheral blood mononuclear cells isolated from healthy donors, patients with metastatic colorectal cancer (mCRC) and match Pre-REP T cells. Plots are representative of at least five experiments. (E, F) Kaplan-Meier curve for relapse free survival () according to the percentage of (E) Treg and (F) CD8+TIM-3+ T cells (low vs high) in Pre-REP T cell cultures following resection of liver metastases from patients with mCRC. Log-rank test was used for the comparison of survival curves. Mann-Whitney tests: *p<0.05, ns, not significant; PBMC, peripheral blood mononuclear cells; TIL, tumor-infiltrating lymphocytes.

Figure 6

Characterization and prognostic value of…

Figure 6

Characterization and prognostic value of expanded Th17 lymphocytes. (A) Gating strategy of CD4…

Figure 6
Characterization and prognostic value of expanded Th17 lymphocytes. (A) Gating strategy of CD4 T cells Th1, Th17 and Th1* subsets relied on cytokine production of interleukin (IL)-17A and IFN-γ in regard of their chemokine receptors expression. (B, C) Levels of each Th cells within pre-rapid expansion protocol (REP) culture were analyzed according to (B) CD4+TIM-3low/high populations and (C) CD8+TIM-3low/high population.(D) Median frequencies of Th1, Th17, Th1* CD4 T cells and double negative subsets were, respectively, 34.0% (0.2–81.5), 50.7% (10.5–99.4), 5.9% (0.2–33.0) and 4.2% (0.0–29.5).(E–G) Kaplan-Meier curve for rapid expansion protocol (RFS) according to the percentage of Th1, Th1* and Th17 T cell (low vs high) in Pre-REP T cell cultures following resection of liver metastases from patients with metastatic colorectal cancer. Log-rank test was used for the comparison of survival curves. Median (%). *P<0.05, **p<0.01; ***p<0.001, ****, p<0.0001; ns, not significant; TIM-3, T cell immunoglobulin mucin domain-containing protein 3.
Similar articles
Cited by
References
    1. Galon J, Costes A, Sanchez-Cabo F, et al. . Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006;313:1960–4. 10.1126/science.1129139 - DOI - PubMed
    1. Pagès F, Berger A, Camus M, et al. . Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med 2005;353:2654–66. 10.1056/NEJMoa051424 - DOI - PubMed
    1. Mlecnik B, Tosolini M, Kirilovsky A, et al. . Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction. J Clin Oncol 2011;29:610–8. 10.1200/JCO.2010.30.5425 - DOI - PubMed
    1. Fridman WH, Zitvogel L, Sautès-Fridman C, et al. . The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol 2017;14:717–34. 10.1038/nrclinonc.2017.101 - DOI - PubMed
    1. Pagès F, Mlecnik B, Marliot F, et al. . International validation of the consensus immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 2018;391:2128–39. 10.1016/S0140-6736(18)30789-X - DOI - PubMed
Show all 52 references
Publication types
MeSH terms
Associated data
Related information
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4
Figure 4
Functional potential of rapid expansion protocol (REP) T-cells. REP T cells were subjected to a 16 hours CD3/28 bead stimulation (1:1) and analyzed by flow cytometry using intracellular staining for Th1 cytokines (IFN-γ, TNF-α and interleukin-2 (IL-2)) gating on CD4 and CD8 T cell populations (n=11). (A, B) IFN-γ, TNF-α and IL-2 expression by (A) CD4 and CD8 REP T cells and (B) according to TIM-3 expression. (C) CD107a production by previously stimulated CD8 REP T cells (n=8). Median (%) with 95 CI or median (%) (min–max). Mann-Whitney tests: *p

Figure 5

Characterization of Treg within T-cell…

Figure 5

Characterization of Treg within T-cell cultures. (A) Regulatory T cells (Treg) was investigated…

Figure 5
Characterization of Treg within T-cell cultures. (A) Regulatory T cells (Treg) was investigated by intracellular staining for FoxP3 on the CD4+CD25HighCD127− population before and after the rapid expansion protocol (REP) (p=0.2921). (B) Median fold expansion in Treg low and in Treg high culture. There was no significant difference (p=0.5920) (C) Percentage of Treg before and after REP according to T cell immunoglobulin mucin domain-containing protein 3 (TIM-3) expression on CD8 T cells. (D) Glycoprotein-A repetitions predominant (GARP) expression (%) on Treg within peripheral blood mononuclear cells isolated from healthy donors, patients with metastatic colorectal cancer (mCRC) and match Pre-REP T cells. Plots are representative of at least five experiments. (E, F) Kaplan-Meier curve for relapse free survival () according to the percentage of (E) Treg and (F) CD8+TIM-3+ T cells (low vs high) in Pre-REP T cell cultures following resection of liver metastases from patients with mCRC. Log-rank test was used for the comparison of survival curves. Mann-Whitney tests: *p<0.05, ns, not significant; PBMC, peripheral blood mononuclear cells; TIL, tumor-infiltrating lymphocytes.

Figure 6

Characterization and prognostic value of…

Figure 6

Characterization and prognostic value of expanded Th17 lymphocytes. (A) Gating strategy of CD4…

Figure 6
Characterization and prognostic value of expanded Th17 lymphocytes. (A) Gating strategy of CD4 T cells Th1, Th17 and Th1* subsets relied on cytokine production of interleukin (IL)-17A and IFN-γ in regard of their chemokine receptors expression. (B, C) Levels of each Th cells within pre-rapid expansion protocol (REP) culture were analyzed according to (B) CD4+TIM-3low/high populations and (C) CD8+TIM-3low/high population.(D) Median frequencies of Th1, Th17, Th1* CD4 T cells and double negative subsets were, respectively, 34.0% (0.2–81.5), 50.7% (10.5–99.4), 5.9% (0.2–33.0) and 4.2% (0.0–29.5).(E–G) Kaplan-Meier curve for rapid expansion protocol (RFS) according to the percentage of Th1, Th1* and Th17 T cell (low vs high) in Pre-REP T cell cultures following resection of liver metastases from patients with metastatic colorectal cancer. Log-rank test was used for the comparison of survival curves. Median (%). *P<0.05, **p<0.01; ***p<0.001, ****, p<0.0001; ns, not significant; TIM-3, T cell immunoglobulin mucin domain-containing protein 3.
Figure 5
Figure 5
Characterization of Treg within T-cell cultures. (A) Regulatory T cells (Treg) was investigated by intracellular staining for FoxP3 on the CD4+CD25HighCD127− population before and after the rapid expansion protocol (REP) (p=0.2921). (B) Median fold expansion in Treg low and in Treg high culture. There was no significant difference (p=0.5920) (C) Percentage of Treg before and after REP according to T cell immunoglobulin mucin domain-containing protein 3 (TIM-3) expression on CD8 T cells. (D) Glycoprotein-A repetitions predominant (GARP) expression (%) on Treg within peripheral blood mononuclear cells isolated from healthy donors, patients with metastatic colorectal cancer (mCRC) and match Pre-REP T cells. Plots are representative of at least five experiments. (E, F) Kaplan-Meier curve for relapse free survival () according to the percentage of (E) Treg and (F) CD8+TIM-3+ T cells (low vs high) in Pre-REP T cell cultures following resection of liver metastases from patients with mCRC. Log-rank test was used for the comparison of survival curves. Mann-Whitney tests: *p<0.05, ns, not significant; PBMC, peripheral blood mononuclear cells; TIL, tumor-infiltrating lymphocytes.
Figure 6
Figure 6
Characterization and prognostic value of expanded Th17 lymphocytes. (A) Gating strategy of CD4 T cells Th1, Th17 and Th1* subsets relied on cytokine production of interleukin (IL)-17A and IFN-γ in regard of their chemokine receptors expression. (B, C) Levels of each Th cells within pre-rapid expansion protocol (REP) culture were analyzed according to (B) CD4+TIM-3low/high populations and (C) CD8+TIM-3low/high population.(D) Median frequencies of Th1, Th17, Th1* CD4 T cells and double negative subsets were, respectively, 34.0% (0.2–81.5), 50.7% (10.5–99.4), 5.9% (0.2–33.0) and 4.2% (0.0–29.5).(E–G) Kaplan-Meier curve for rapid expansion protocol (RFS) according to the percentage of Th1, Th1* and Th17 T cell (low vs high) in Pre-REP T cell cultures following resection of liver metastases from patients with metastatic colorectal cancer. Log-rank test was used for the comparison of survival curves. Median (%). *P<0.05, **p<0.01; ***p<0.001, ****, p<0.0001; ns, not significant; TIM-3, T cell immunoglobulin mucin domain-containing protein 3.

References

    1. Galon J, Costes A, Sanchez-Cabo F, et al. . Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006;313:1960–4. 10.1126/science.1129139
    1. Pagès F, Berger A, Camus M, et al. . Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med 2005;353:2654–66. 10.1056/NEJMoa051424
    1. Mlecnik B, Tosolini M, Kirilovsky A, et al. . Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction. J Clin Oncol 2011;29:610–8. 10.1200/JCO.2010.30.5425
    1. Fridman WH, Zitvogel L, Sautès-Fridman C, et al. . The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol 2017;14:717–34. 10.1038/nrclinonc.2017.101
    1. Pagès F, Mlecnik B, Marliot F, et al. . International validation of the consensus immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 2018;391:2128–39. 10.1016/S0140-6736(18)30789-X
    1. Mlecnik B, Bindea G, Angell HK, et al. . Integrative analyses of colorectal cancer show immunoscore is a stronger predictor of patient survival than microsatellite instability. Immunity 2016;44:698–711. 10.1016/j.immuni.2016.02.025
    1. Valderrama-Treviño AI, Barrera-Mera B, Ceballos-Villalva JC, et al. . Hepatic metastasis from colorectal cancer. Euroasian J Hepatogastroenterol 2017;7:166–75. 10.5005/jp-journals-10018-1241
    1. Turcotte S, Gros A, Hogan K, et al. . Phenotype and function of T cells infiltrating visceral metastases from gastrointestinal cancers and melanoma: implications for adoptive cell transfer therapy. J Immunol 2013;191:2217–25. 10.4049/jimmunol.1300538
    1. Turcotte S, Gros A, Tran E, et al. . Tumor-reactive CD8+ T cells in metastatic gastrointestinal cancer refractory to chemotherapy. Clin Cancer Res 2014;20:331–43. 10.1158/1078-0432.CCR-13-1736
    1. Robbins PF, Lu Y-C, El-Gamil M, et al. . Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 2013;19:747–52. 10.1038/nm.3161
    1. Scheper W, Kelderman S, Fanchi LF, et al. . Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers. Nat Med 2019;25:89–94. 10.1038/s41591-018-0266-5
    1. Stevanović S, Helman SR, Wunderlich JR, et al. . A phase II study of tumor-infiltrating lymphocyte therapy for human papillomavirus-associated epithelial cancers. Clin Cancer Res 2019;25:1486–93. 10.1158/1078-0432.CCR-18-2722
    1. Rosenberg SA, Yang JC, Sherry RM, et al. . Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 2011;17:4550–7. 10.1158/1078-0432.CCR-11-0116
    1. Tran KQ, Zhou J, Durflinger KH, et al. . Minimally cultured tumor-infiltrating lymphocytes display optimal characteristics for adoptive cell therapy. J Immunother 2008;31:742–51. 10.1097/CJI.0b013e31818403d5
    1. Dudley ME, Gross CA, Langhan MM, et al. . CD8+ enriched "young" tumor infiltrating lymphocytes can mediate regression of metastatic melanoma. Clin Cancer Res 2010;16:6122–31. 10.1158/1078-0432.CCR-10-1297
    1. Muranski P, Restifo NP. Adoptive immunotherapy of cancer using CD4(+) T cells. Curr Opin Immunol 2009;21:200–8. 10.1016/j.coi.2009.02.004
    1. Tran E, Turcotte S, Gros A, et al. . Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 2014;344:641–5. 10.1126/science.1251102
    1. Linnemann C, van Buuren MM, Bies L, et al. . High-Throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma. Nat Med 2015;21:81–5. 10.1038/nm.3773
    1. Borst J, Ahrends T, Bąbała N, et al. . CD4+ T cell help in cancer immunology and immunotherapy. Nat Rev Immunol 2018;18:635–47. 10.1038/s41577-018-0044-0
    1. Ahrends T, Spanjaard A, Pilzecker B, et al. . CD4+ T Cell Help Confers a Cytotoxic T Cell Effector Program Including Coinhibitory Receptor Downregulation and Increased Tissue Invasiveness. Immunity 2017;47:848–61. 10.1016/j.immuni.2017.10.009
    1. Donia M, Andersen R, Kjeldsen JW, et al. . Aberrant Expression of MHC Class II in Melanoma Attracts Inflammatory Tumor-Specific CD4 + T- Cells, Which Dampen CD8 + T-cell Antitumor Reactivity. Cancer Res 2015;75:3747–59. 10.1158/0008-5472.CAN-14-2956
    1. Tran E, Ahmadzadeh M, Lu Y-C, et al. . Immunogenicity of somatic mutations in human gastrointestinal cancers. Science 2015;350:1387–90. 10.1126/science.aad1253
    1. Asadzadeh Z, Mohammadi H, Safarzadeh E, et al. . The paradox of Th17 cell functions in tumor immunity. Cell Immunol 2017;322:15–25. 10.1016/j.cellimm.2017.10.015
    1. Wu D, Wu P, Huang Q, et al. . Interleukin-17: a promoter in colorectal cancer progression. Clin Dev Immunol 2013;2013:1–7. 10.1155/2013/436307
    1. Biragyn A, Ferrucci L. Gut dysbiosis: a potential link between increased cancer risk in ageing and inflammaging. Lancet Oncol 2018;19:e295–304. 10.1016/S1470-2045(18)30095-0
    1. Tosolini M, Kirilovsky A, Mlecnik B, 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;71:1263–71. 10.1158/0008-5472.CAN-10-2907
    1. Dudley ME, Wunderlich JR, Shelton TE, et al. . Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J Immunother 2003;26:332–42. 10.1097/00002371-200307000-00005
    1. Li Y, Liu S, Hernandez J, et al. . MART-1-specific melanoma tumor-infiltrating lymphocytes maintaining CD28 expression have improved survival and expansion capability following antigenic restimulation in vitro. J Immunol 2010;184:452–65. 10.4049/jimmunol.0901101
    1. Klebanoff CA, Scott CD, Leonardi AJ, et al. . Memory T cell-driven differentiation of naive cells impairs adoptive immunotherapy. J Clin Invest 2016;126:318–34. 10.1172/JCI81217
    1. Blank CU, Haining WN, Held W, et al. . Defining 'T cell exhaustion'. Nat Rev Immunol 2019;19:665–74. 10.1038/s41577-019-0221-9
    1. Becattini S, Latorre D, Mele F, et al. . T cell immunity. functional heterogeneity of human memory CD4⁺ T cell clones primed by pathogens or vaccines. Science 2015;347:400–6. 10.1126/science.1260668
    1. Wherry EJ. T cell exhaustion. Nat Immunol 2011;12:492–9. 10.1038/ni.2035
    1. Gauthy E, Cuende J, Stockis J, et al. . Garp is regulated by miRNAs and controls latent TGF-β1 production by human regulatory T cells. PLoS One 2013;8:e76186. 10.1371/journal.pone.0076186
    1. Chung AS, Wu X, Zhuang G, et al. . An interleukin-17-mediated paracrine network promotes tumor resistance to anti-angiogenic therapy. Nat Med 2013;19:1114–23. 10.1038/nm.3291
    1. Limagne E, Euvrard R, Thibaudin M, et al. . Accumulation of MDSC and Th17 cells in patients with metastatic colorectal cancer predicts the efficacy of a FOLFOX-Bevacizumab drug treatment regimen. Cancer Res 2016;76:5241–52. 10.1158/0008-5472.CAN-15-3164
    1. Tran E, Robbins PF, Lu Y-C, et al. . T-Cell transfer therapy targeting mutant KRAS in cancer. N Engl J Med 2016;375:2255–62. 10.1056/NEJMoa1609279
    1. Cavnar MJ, Turcotte S, Katz SC, et al. . Tumor-Associated macrophage infiltration in colorectal cancer liver metastases is associated with better outcome. Ann Surg Oncol 2017;24:1835–42. 10.1245/s10434-017-5812-8
    1. Katz SC, Pillarisetty V, Bamboat ZM, et al. . T cell infiltrate predicts long-term survival following resection of colorectal cancer liver metastases. Ann Surg Oncol 2009;16:2524–30. 10.1245/s10434-009-0585-3
    1. Katz SC, Bamboat ZM, Maker AV, et al. . Regulatory T cell infiltration predicts outcome following resection of colorectal cancer liver metastases. Ann Surg Oncol 2013;20:946–55. 10.1245/s10434-012-2668-9
    1. Goff SL, Dudley ME, Citrin DE, et al. . Randomized, prospective evaluation comparing intensity of Lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes for patients with metastatic melanoma. J Clin Oncol 2016;34:2389–97. 10.1200/JCO.2016.66.7220
    1. Beltra J-C, Manne S, Abdel-Hakeem MS, et al. . Developmental Relationships of Four Exhausted CD8+ T Cell Subsets Reveals Underlying Transcriptional and Epigenetic Landscape Control Mechanisms. Immunity 2020;52:825–41. 10.1016/j.immuni.2020.04.014
    1. Saleh R, Taha RZ, Toor SM, et al. . Expression of immune checkpoints and T cell exhaustion markers in early and advanced stages of colorectal cancer. Cancer Immunol Immunother 2020;69:1989–99. 10.1007/s00262-020-02593-w
    1. Ben-Avi R, Farhi R, Ben-Nun A, et al. . Establishment of adoptive cell therapy with tumor infiltrating lymphocytes for non-small cell lung cancer patients. Cancer Immunol Immunother 2018;67:1221–30. 10.1007/s00262-018-2174-4
    1. Yan W, Liu X, Ma H, et al. . Tim-3 fosters HCC development by enhancing TGF-β-mediated alternative activation of macrophages. Gut 2015;64:1593–604. 10.1136/gutjnl-2014-307671
    1. Saito T, Nishikawa H, Wada H, et al. . Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med 2016;22:679–84. 10.1038/nm.4086
    1. Wing JB, Tanaka A, Sakaguchi S. Human FOXP3+ Regulatory T Cell Heterogeneity and Function in Autoimmunity and Cancer. Immunity 2019;50:302–16. 10.1016/j.immuni.2019.01.020
    1. Liénart S, Merceron R, Vanderaa C, et al. . Structural basis of latent TGF-β1 presentation and activation by GARP on human regulatory T cells. Science 2018;362:952–6. 10.1126/science.aau2909
    1. Gaublomme JT, Yosef N, Lee Y, et al. . Single-Cell genomics unveils critical regulators of Th17 cell pathogenicity. Cell 2015;163:1400–12. 10.1016/j.cell.2015.11.009
    1. Omenetti S, Bussi C, Metidji A, et al. . The intestine harbors functionally distinct homeostatic tissue-resident and inflammatory Th17 cells. Immunity 2019;51:77–89. 10.1016/j.immuni.2019.05.004
    1. Ye J, Livergood RS, Peng G. The role and regulation of human Th17 cells in tumor immunity. Am J Pathol 2013;182:10–20. 10.1016/j.ajpath.2012.08.041
    1. Kryczek I, Banerjee M, Cheng P, et al. . Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments. Blood 2009;114:1141–9. 10.1182/blood-2009-03-208249
    1. Renaude E, Kroemer M, Loyon R, et al. . The fate of Th17 cells is shaped by epigenetic modifications and remodeled by the tumor microenvironment. Int J Mol Sci 2020;21. 10.3390/ijms21051673. [Epub ahead of print: 29 Feb 2020].

Source: PubMed

3
Tilaa