Tumour-reactive T cell subsets in the microenvironment of ovarian cancer

Marie Christine Wulff Westergaard, Rikke Andersen, Chloé Chong, Julie Westerlin Kjeldsen, Magnus Pedersen, Christina Friese, Thomas Hasselager, Henrik Lajer, George Coukos, Michal Bassani-Sternberg, Marco Donia, Inge Marie Svane, Marie Christine Wulff Westergaard, Rikke Andersen, Chloé Chong, Julie Westerlin Kjeldsen, Magnus Pedersen, Christina Friese, Thomas Hasselager, Henrik Lajer, George Coukos, Michal Bassani-Sternberg, Marco Donia, Inge Marie Svane

Abstract

Background: Solid malignancies are frequently infiltrated with T cells. The success of adoptive cell transfer (ACT) with expanded tumour-infiltrating lymphocytes (TILs) in melanoma warrants its testing in other cancer types. In this preclinical study, we investigated whether clinical-grade TILs could be manufactured from ovarian cancer (OC) tumour specimens.

Methods: Thirty-four tumour specimens were obtained from 33 individual patients with OC. TILs were analysed for phenotype, antigen specificity and functionality.

Results: Minimally expanded TILs (Young TILs) were successfully established from all patients. Young TILs contained a high frequency of CD3+ cells with a variable CD4/CD8 ratio. TILs could be expanded to clinical numbers. Importantly, recognition of autologous tumour cells was demonstrated in TILs in >50% of the patients. We confirmed with mass spectrometry the presentation of multiple tumour antigens, including peptides derived from the cancer-testis antigen GAGE, which could be recognised by antigen-specific TILs. Antigen-specific TILs could be isolated and further expanded in vitro.

Conclusion: These findings support the hypothesis that patients with OC can benefit from ACT with TILs and led to the initiation of a pilot clinical trial at our institution .

Trial registration: clinicaltrials.gov: NCT02482090.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Ovarian tumour-infiltrating lymphocytes (TILs). Young TILs were further expanded using a small scale Rapid Expansion Protocol (REP). a Fold expansion of TILs from 24 ovarian cancer (OC) patients (purple lines) and from 11 metastatic melanoma (MM) patients (blue lines) following a 14 days REP performed in parallel were compared. Median fold expansion for all OC patients (n = 34) was 1660 (range [440–5544]). Median fold expansion for MM (n = 11) was 2842 (range [816–6900]). b Scatterplot showing the final fold expansion. No significant difference was seen when comparing the final fold expansion of OC-TILs (n = 34) and MM-TILs (n = 11). The two groups were compared using the Mann–Whitney test. Data are presented with the median
Fig. 2
Fig. 2
Phenotypic characterisation of ovarian cancer TILs. Young TILs and REP-TILs were analysed with flow cytometry for phenotypic markers. a Scatterplot showing percentages of CD3+ T cells in Young TIL (n = 34) and REP TIL (n = 33) populations. Data are presented with the median. b Scatterplot showing the percentage of NK cells in the Young TIL population (n = 34). Data are presented with the median. c Scatterplot showing percentages of γδ T cells in Young TIL (n = 34) and REP TIL (n = 33) populations. Data are presented with the median. d Scatterplot showing the CD4/CD8 ratios in Young TILs (n = 34) and REP-TILs (n = 33). The median CD4/CD8 ratio in Young TILs was 7.2 (range [0.01–306]) and 10.0 (range [0.02–1067]) in REP TIL population. Data were log-transformed before the two groups were compared. A significant increase in the ratio was observed (p = 0.0253). Data are presented with the median. e The pie charts show the phenotypic distribution of CD8+, CD4+, CD4+CD8+ and CD4−CD8− of CD3+ TILs in the Young TIL (n = 34) and REP TIL (n = 33) populations. Data are presented with mean values. f, g Scatterplots illustrating the percentage of naïve T cells, central memory T cells and effector memory T cells and h, i Scatterplots illustrating the percentage of Exhaustion markers: LAG-3, BTLA, PD-1 and TIM-3 in f, h the CD8+ T cell population and g, i the CD4+ T cell population in Young TILs (n = 34) and REP-TILs (n = 33). Data are presented with the median with interquartile range. Panel f shows a significant increase of CD8+ Tem cells p = 0.0004 during the REP and a significant decrease in the Naïve T cell subpopulation, p < 0.0001. g The CD4-expressing population had a significant increase in CD4+ Tem cells during the REP, p = 0.0003, and a significant decrease in the Naïve T cell subpopulation during the REP, p < 0.0001. Panel h shows significant increase of CD8+ LAG-3+ T cells p = 0.0263 during the REP and a significant decrease in the PD-1+ T cells, p = 0.0002, and a significant increase in the TIM-3+ T cells, p = 0.0379. i The CD4-expressing population had a significant decrease in CD4+ BTLA+ cells during the REP, p = 0.013, and a significant decrease in the PD-1+ T cell subpopulation during the REP, p < 0.0001. Young TILs and REP-TILs were compared using the Wilcoxon signed rank test. Statistical significant differences is indicated with *, **, *** or **** for p values less than 0.05, 0.01, 0.001, or 0.0001, respectively
Fig. 3
Fig. 3
In vitro antitumour activity of CD8+ Young TILs. The antitumour activity of the in vitro-expanded TILs was evaluated by defining the frequency of T cells expressing at least one of the following T cell functions: TNF-α, IFN-γ or CD107a, upon stimulation with autologous Fresh tumour digest (FTD) or tumour cell line (TCL) treated with low-dose IFN-γ (100 IU/ml) or left untreated. A specific antitumour response was defined as the presence of minimum 0.5% responding cells, with a minimum number of 50 positive events. The frequency of tumour-reactive cells in stimulated samples was subtracted from the unstimulated samples. In all, 0.5% was used as a threshold for detection of tumour reactivity. a Antitumour responses of CD8+ T cells were detected in 13 of 31 patients analysed. *: OC.TIL.03 is not tested with FTD. ϕ: TILs generated from OC.TIL.04 2nd was tested for reactivity against FTD from OC.TIL.04. b FACS plot showing cytokine production from TIL alone (unstimulated, serving as a negative control) and TIL stimulated with autologous TCL, from a representative patient (OC.TIL.11). c FACS plot showing CD107a mobilisation of TIL upon co-culture with an autologous TCL. Unstimulated TIL (TIL alone) serves as a negative control. An example of the gating strategy is showed in Supplementary Figure 7A
Fig. 4
Fig. 4
In vitro antitumour activity of CD4+ Young TILs. The antitumour activity of the in vitro-expanded TILs was evaluated by defining the frequency of T cells expressing at least one of the following T cell functions: TNF-α, IFN-γ or CD107a, upon stimulation with autologous fresh tumour digest (FTD) or tumour cell line (TCL) treated with low-dose IFN-γ (100 IU/ml) or left untreated. A specific antitumour response was defined as the presence of minimum 0.5% responding cells, with a minimum number of 50 positive events. The frequency of tumour-reactive cells in stimulated samples was subtracted from unstimulated samples; 0.5% was used as a threshold for detection of tumour reactivity. a Antitumour responses of CD4+ T cells were detected in 16 of 31 patients. *OC.TIL.03 is not tested with FTD. ϕ: TILs generated from OC.TIL.04 2nd was tested for reactivity against FTD from OC.TIL.04. b FACS plot showing cytokine production from TIL alone (unstimulated, serving as a negative control) and TIL stimulated with autologous TCL, from a representative patient (OC.TIL.15). An example of the gating strategy is showed in Supplementary Figure 7A
Fig. 5
Fig. 5
IFN-γ ELISPOT assay: Selected peptides identified by MS-based immunopeptidomics tested for the ability to induce an IFN-γ response in REP-TILs from patient OC.TIL.11. Background (spots in wells without added peptides) was subtracted. Example of IFN-γ ELISPOT response against GAGE peptides
Fig. 6
Fig. 6
GAGE-specific TILs. a 1600 GAGE12-specific Young TILs from patient OC.TIL.11 were sorted using tetramers HLA-A3/STYYWPRPR APC/PE. b The sorted culture was tested for specificity with tetramers HLA-A3/STYYWPRPR APC/PE after two times REP. c FACS plot illustrating cytokine production (upper panel) and CD107a mobilisation (lower panel) in TIL alone (unstimulated) as a negative control, TILs stimulated with autologous tumour cell line (TCL), TILs stimulated with autologous TCL pre-stimulated with IFN-γ for 3 days prior to experiment and TILs stimulated with the GAGE peptide. An example of the gating strategy is showed in Supplementary Figure 7A. d xCELLigence killing assay showing the killing of autologous tumour cells with GAGE-specific TILs from patients OC.TIL.11 (ratio 0.2:1). HLA class I antibody, HLA class II antibody and a combination were added as controls

References

    1. Gooden MJM, de Bock GH, Leffers N, Daemen T, Nijman HW. The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis. Br. J. Cancer. 2011;105:93–103. doi: 10.1038/bjc.2011.189.
    1. Adams SF, et al. Intraepithelial T cells and tumour proliferation: impact on the benefit from surgical cytoreduction in advanced serous ovarian cancer. Cancer. 2009;115:2891–2902. doi: 10.1002/cncr.24317.
    1. Zhang L, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N. Engl. J. Med. 2003;348:203–213. doi: 10.1056/NEJMoa020177.
    1. Sato E, et al. Intraepithelial CD8+tumour-infiltrating lymphocytes and a hiThe Danish National Committee on Health Research Ethics approved the scientific use of the patient material (protocol number H-2–2014–055).gh CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc. Natl. Acad. Sci. USA. 2005;102:18538–18543. doi: 10.1073/pnas.0509182102.
    1. Kvistborg P, van Buuren MM, Schumacher TN. Human cancer regression antigens. Curr. Opin. Immunol. 2013;25:284–290. doi: 10.1016/j.coi.2013.03.005.
    1. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74. doi: 10.1126/science.aaa4971.
    1. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348:62–68. doi: 10.1126/science.aaa4967.
    1. Wick Da, et al. Surveillance of the tumour mutanome by T cells during progression from primary to recurrent ovarian cancer. Clin. Cancer Res. 2014;20:1125–1134. doi: 10.1158/1078-0432.CCR-13-2147.
    1. Ye Q, et al. CD137 accurately identifies and enriches for naturally occurring tumour-reactive T cells in tumour. Clin. Cancer Res. 2014;20:44–55. doi: 10.1158/1078-0432.CCR-13-0945.
    1. Attig S, et al. Simultaneous infiltration of polyfunctional effector and suppressor T cells into renal cell carcinomas. Cancer Res. 2009;69:8412–8419. doi: 10.1158/0008-5472.CAN-09-0852.
    1. Alexandrov LB, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421. doi: 10.1038/nature12477.
    1. Coukos G, Tanyi J, Kandalaft LE. Opportunities in immunotherapy of ovarian cancer. Ann. Oncol. 2016;27:i11–i15. doi: 10.1093/annonc/mdw084.
    1. Armstrong DK, et al. Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N. Engl. J. Med. 2016;354:34–43. doi: 10.1056/NEJMoa052985.
    1. . (2016).
    1. Brahmer JR, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012;188:2148–2149.
    1. Hamanishi J, et al. Safety and antitumor activity of anti-PD-1 antibody, Nivolumab, in patients with platinum-resistant ovarian cancer. J. Clin. Oncol. 2015;33:4015–4022. doi: 10.1200/JCO.2015.62.3397.
    1. Varga A., et al. Antitumor activity and safety of pembrolizumab in patients (pts) with PD-L1 positive advanced ovarian cancer: interim results from a phase Ib study. J. Clin. Oncol. 33(Suppl.): abstr 5510 (2015).
    1. Disis ML, et al. 2749 Avelumab (MSB0010718C), an anti-PD-L1 antibody, in patients with recurrent or refractory ovarian cancer: a phase Ib trial reporting safety and clinical activity. Eur. J. Cancer. 2015;51:S546–S547. doi: 10.1016/S0959-8049(16)31515-5.
    1. Donia M, et al. Characterization and comparison of ‘standard’ and ‘young’ tumour-infiltrating lymphocytes for adoptive cell therapy at a Danish translational research institution. Scand. J. Immunol. 2011;75:157–167. doi: 10.1111/j.1365-3083.2011.02640.x.
    1. Donia M, et al. Aberrant expression of MHC Class II in melanoma attracts inflammatory tumour specific CD4+T cells which dampen CD8+T cell antitumor reactivity. Cancer Res. 2015;75:3747–3760. doi: 10.1158/0008-5472.CAN-14-2956.
    1. Peper JK, et al. An impedance-based cytotoxicity assay for real-time and label-free assessment of T-cell-mediated killing of adherent cells. J. Immunol. Methods. 2014;405:192–198. doi: 10.1016/j.jim.2014.01.012.
    1. Chong C, et al. High-throughput and sensitive immunopeptidomics platform reveals profound interferonγ-mediated remodeling of the human leukocyte antigen (HLA) ligandome. Mol. Cell. Proteomics. 2018;17:533–548. doi: 10.1074/mcp.TIR117.000383.
    1. Toebes M, et al. Design and use of conditional MHC class I ligands. Nat. Med. 2006;12:246–251. doi: 10.1038/nm1360.
    1. Andersen R, et al. Long-lasting complete responses in patients with metastatic melanoma after adoptive cell therapy with tumour-infiltrating lymphocytes and an attenuated IL2 regimen. Clin. Cancer Res. 2016;22:3734–3745. doi: 10.1158/1078-0432.CCR-15-1879.
    1. Junker N, et al. Bimodal ex vivo expansion of T cells from patients with head and neck squamous cell carcinoma: a prerequisite for adoptive cell transfer. Cytotherapy. 2011;13:822–834. doi: 10.3109/14653249.2011.563291.
    1. Andersen R, et al. T-cell responses in the microenvironment of primary renal cell carcinoma-implications for adoptive cell therapy. Cancer Immunol. Res. 2018;6:222–235. doi: 10.1158/2326-6066.CIR-17-0467.
    1. Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu. Rev. Immunol. 2004;22:745–763. doi: 10.1146/annurev.immunol.22.012703.104702.
    1. Donia M, Larsen SM, Met O, Svane IM. Simplified protocol for clinical-grade tumour-infiltrating lymphocyte manufacturing with use of the wave bioreactor. Cytotherapy. 2014;0:1–4.
    1. Besser MJ, et al. Clinical responses in a phase II study using adoptive transfer of short-term cultured tumour infiltration lymphocytes in metastatic melanoma patients. Clin. Cancer Res. 2010;16:2646–2655. doi: 10.1158/1078-0432.CCR-10-0041.
    1. Lo CS, et al. Neoadjuvant chemotherapy of ovarian cancer results in three patterns of tumour-infiltrating lymphocyte response with distinct implications for immunotherapy. Am. Assoc. Cancer Res. 2017;23:925–934.
    1. Dudley ME, Wunderlich JR, Shelton TE, Even J, Rosenberg SA. Generation of tumour-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J. Immunother. 2003;26:332–342. doi: 10.1097/00002371-200307000-00005.
    1. Itzhaki O, et al. Establishment and large-scale expansion of minimally cultured ‘young’ tumour infiltrating lymphocytes for adoptive transfer therapy. J. Immunother. 2011;34:212–220. doi: 10.1097/CJI.0b013e318209c94c.
    1. Turcotte S, et al. Tumor-reactive cd8+ tcells in metastatic gastrointestinal cancer refractory to chemotherapy. Clin. Cancer Res. 2014;20:331–343. doi: 10.1158/1078-0432.CCR-13-1736.
    1. Markel GAL, et al. Preclinical evaluation of adoptive cell therapy for patients with metastatic renal cell carcinoma. Anticancer Res. 2009;154:145–154.
    1. Baldan V, Griffiths R, Hawkins RE, Gilham DE. Efficient and reproducible generation of tumour-infiltrating lymphocytes for renal cell carcinoma. Br. J. Cancer. 2015;112:1510–1518. doi: 10.1038/bjc.2015.96.
    1. Owens GL, et al. Ex vivo expanded tumour-infiltrating lymphocytes from ovarian cancer patients release anti-tumour cytokines in response to autologous primary ovarian cancer cells. Cancer Immunol. Immunother. 2018;67:1519–1531. doi: 10.1007/s00262-018-2211-3.
    1. Linnemann C, et al. High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+T cells in human melanoma. Nat. Med. 2014;21:1–7.
    1. Friedman KM, et al. Tumor-specific CD4+melanoma tumour-infiltrating lymphocytes. J. Immunother. 2012;35:400–408. doi: 10.1097/CJI.0b013e31825898c5.
    1. Backer ODe, et al. Characterization of the GAGE genes that are expressed in various human cancers and in normal testis characterization of the GAGE genes that are expressed in various human cancers and in normal testis 1. Cancer Res. 1999;59:3157–3165.
    1. Bobisse B, et al. Sensitive and frequent identification of high avidity neo-epitope specific CD8+T cells in immunotherapy-naive ovarian cancer. Nat. Commun. 2018;9:1–10. doi: 10.1038/s41467-018-03301-0.
    1. Martin SD, et al. Low mutation burden in ovarian cancer may limit the utility of neoantigen-targeted vaccines. PLoS ONE. 2016;11:1–22.
    1. Bassani-Sternberg M, Coukos G. Mass spectrometry-based antigen discovery for cancer immunotherapy. Curr. Opin. Immunol. 2016;41:9–17. doi: 10.1016/j.coi.2016.04.005.
    1. Kelderman S, et al. Antigen-specific TIL therapy for melanoma: a flexible platform for personalized cancer immunotherapy. Eur. J. Immunol. 2016;46:1351–1360. doi: 10.1002/eji.201545849.

Source: PubMed

3
Abonner