Skip to main content

Advertisement

Log in

Targeting PD-L1 and TIGIT could restore intratumoral CD8 T cell function in human colorectal cancer

  • Original Article
  • Published:
Cancer Immunology, Immunotherapy Aims and scope Submit manuscript

Abstract

Microsatellite stable colorectal cancers (MSS-CRC) are resistant to anti-PD-1/PD-L1 therapy but the combination of immune checkpoints inhibitors (ICI) could be a clue to reverse resistance. Our aim was to evaluate ex vivo the capacity of the combination of atezolizumab (anti-PD-L1) and tiragolumab (anti-TIGIT) to reactivate the immune response of tumor infiltrating lymphocytes (TILs) in MSS-CRC. We analysed CRC tumor tissue and the associated blood sample in parallel. For each patient sample, extensive immunomonitoring and cytokine production were tested. We generated an ex vivo assay to study immune reactivity following immune stimulation with checkpoint inhibitors of tumor cell suspensions. Three microsatellite instable (MSI) and 13 MSS-CRC tumors were analysed. To generalize our observations, bioinformatics analyses were performed on public data of single cell RNA sequencing of CRC TILs and RNA sequencing data of TCGA. Atezolizumab alone could only reactivate T cells from MSI tumors. Atezolizumab and tiragolumab reactivated T cells in 46% of MSS-CRC samples. Reactivation by ICK was observed in patients with higher baseline frequency of Th1 and Tc1 cells, and was also associated with higher baseline T cell polyfunctionality and higher CD96 expression. We showed that a high frequency of CD96 expression on T cells could be a surrogate marker of atezolizumab and tiragolumab efficacy. Together these data suggest that the association of atezolizumab and tiragolumab could restore function of CD4 and CD8 TILs in MSS-CRC and could be tested in a clinical trial in colorectal cancer patients with MSS status.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Abbreviations

CRC:

ColoRectal Cancer

CTLA-4:

Cytotoxic T-Lymphocyte-Associated protein 4

CTV:

Cell Trace Violet

FBS:

Foetal Bovine Serum

ICK:

Immune ChecKpoint

ICI:

Immune Checkpoint Inhibitors

IFNγ:

InterFeroN gamma

IL-2:

InterLeukin-2

LAG3:

Lymphocyte-Activation Gene 3

mAbs:

Monoclonal antibodies

mCRC:

Metastatic ColoRectal Cancer

MFI:

Median Fluorescence Intensity

MSS:

MicroSatellite Stable

MSI:

MicroSatellite Instable

NK:

Natural Killer

NSCLC:

Non-Small Cell Lung Cancer

PBMC:

Peripheral Blood Mononuclear Cells

PBS:

Phosphate-Buffered Saline

PD-1:

Programmed cell Death protein 1

PD-L1:

Programmed cell Death protein Ligand 1

TIGIT:

T cell Immunoreceptor with Ig and ITIM domains

Th1:

T helper 1

TILs:

Tumor Infiltrating Lymphocytes

TNFα:

Tumor Necrosis Factor alpha

References

  1. Pagès F, Mlecnik B, Marliot F et al (2018) International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 391:2128–2139. https://doi.org/10.1016/S0140-6736(18)30789-X

    Article  PubMed  Google Scholar 

  2. Reichling C, Taieb J, Derangere V et al (2020) Artificial intelligence-guided tissue analysis combined with immune infiltrate assessment predicts stage III colon cancer outcomes in PETACC08 study. Gut 69:681–690. https://doi.org/10.1136/gutjnl-2019-319292

    Article  CAS  PubMed  Google Scholar 

  3. Bindea G, Mlecnik B, Tosolini M et al (2013) Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 39:782–795. https://doi.org/10.1016/j.immuni.2013.10.003

    Article  CAS  PubMed  Google Scholar 

  4. Turcotte S, Gros A, Hogan K et al (2013) Phenotype and function of T cells infiltrating visceral metastases from gastrointestinal cancers and melanoma: implications for adoptive cell transfer therapy. J Immunol 191:2217–2225. https://doi.org/10.4049/jimmunol.1300538

    Article  CAS  PubMed  Google Scholar 

  5. Robbins PF, Lu Y-C, El-Gamil M et al (2013) Mining exomic sequencing data to identify mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 19:747–752. https://doi.org/10.1038/nm.3161

    Article  CAS  PubMed  Google Scholar 

  6. Kong Y, Zhu L, Schell TD et al (2016) T-cell immunoglobulin and ITIM domain (TIGIT) associates with CD8+ T-cell exhaustion and poor clinical outcome in AML patients. Clin Cancer Res 22:3057–3066. https://doi.org/10.1158/1078-0432.CCR-15-2626

    Article  CAS  PubMed  Google Scholar 

  7. Johnston RJ, Comps-Agrar L, Hackney J et al (2014) The immunoreceptor TIGIT regulates antitumor and antiviral CD8+ T cell effector function. Cancer Cell 26:923–937. https://doi.org/10.1016/j.ccell.2014.10.018

    Article  CAS  PubMed  Google Scholar 

  8. Grapin M, Richard C, Limagne E et al (2019) Optimized fractionated radiotherapy with anti-PD-L1 and anti-TIGIT: a promising new combination. J Immunother Cancer. https://doi.org/10.1186/s40425-019-0634-9

    Article  PubMed  Google Scholar 

  9. Dubuisson A, Fahrner J, Goubet A et al (2021) Immunodynamics of explanted human tumors for immuno-oncology. EMBO Mol Med. https://doi.org/10.15252/emmm.202012850

    Article  PubMed  Google Scholar 

  10. Voabil P, de Bruijn M, Roelofsen LM et al (2021) An ex vivo tumor fragment platform to dissect response to PD-1 blockade in cancer. Nat Med 27:1250–1261. https://doi.org/10.1038/s41591-021-01398-3

    Article  CAS  PubMed  Google Scholar 

  11. Qian J, Olbrecht S, Boeckx B et al (2020) A pan-cancer blueprint of the heterogeneous tumor microenvironment revealed by single-cell profiling. Cell Res 30:745–762. https://doi.org/10.1038/s41422-020-0355-0

    Article  CAS  PubMed  Google Scholar 

  12. Stuart T, Butler A, Hoffman P et al (2019) Comprehensive integration of single-cell data. Cell 177:1888-1902.e21. https://doi.org/10.1016/j.cell.2019.05.031

    Article  CAS  PubMed  Google Scholar 

  13. Wan Y-W, Allen GI, Liu Z (2016) TCGA2STAT: simple TCGA data access for integrated statistical analysis in R. Bioinformatics 32:952–954. https://doi.org/10.1093/bioinformatics/btv677

    Article  CAS  PubMed  Google Scholar 

  14. Antoniotti M, Caravagna G, Sano LD et al (2021) TRONCO: TRONCO, an R package for TRanslational ONCOlogy. Bioconductor version: Release (3.12)

  15. Eide PW, Bruun J, Lothe RA, Sveen A (2017) CMScaller: an R package for consensus molecular subtyping of colorectal cancer pre-clinical models. Sci Rep. https://doi.org/10.1038/s41598-017-16747-x

    Article  PubMed  Google Scholar 

  16. Chalabi M, Fanchi LF, Dijkstra KK et al (2020) Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat Med 26:566–576. https://doi.org/10.1038/s41591-020-0805-8

    Article  CAS  PubMed  Google Scholar 

  17. Somaiah N, Conley AP, Lin HY et al (2020) A phase II multi-arm study of durvalumab and tremelimumab for advanced or metastatic sarcomas. JCO 38:11509–11509. https://doi.org/10.1200/JCO.2020.38.15_suppl.11509

    Article  Google Scholar 

  18. Yu X, Harden K, Gonzalez LC et al (2009) The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol 10:48–57. https://doi.org/10.1038/ni.1674

    Article  CAS  PubMed  Google Scholar 

  19. Manieri NA, Chiang EY, Grogan JL (2017) TIGIT: a key inhibitor of the cancer immunity cycle. Trends Immunol 38:20–28. https://doi.org/10.1016/j.it.2016.10.002

    Article  CAS  PubMed  Google Scholar 

  20. Joller N, Hafler JP, Brynedal B et al (2011) TIGIT has T cell intrinsic inhibitory functions. J Immunol 186:1338–1342. https://doi.org/10.4049/jimmunol.1003081

    Article  CAS  PubMed  Google Scholar 

  21. Stanietsky N, Simic H, Arapovic J et al (2009) The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. PNAS 106:17858–17863. https://doi.org/10.1073/pnas.0903474106

    Article  PubMed  Google Scholar 

  22. Yeo J, Ko M, Lee D-H et al (2021) TIGIT/CD226 axis regulates anti-tumor immunity. Pharmaceuticals (Basel). https://doi.org/10.3390/ph14030200

    Article  Google Scholar 

  23. Chen F, Xu Y, Chen Y, Shan S (2020) TIGIT enhances CD4+ regulatory T-cell response and mediates immune suppression in a murine ovarian cancer model. Cancer Med 9:3584–3591. https://doi.org/10.1002/cam4.2976

    Article  CAS  PubMed  Google Scholar 

  24. Guillerey C, Harjunpää H, Carrié N et al (2018) TIGIT immune checkpoint blockade restores CD8+ T-cell immunity against multiple myeloma. Blood 132:1689–1694. https://doi.org/10.1182/blood-2018-01-825265

    Article  CAS  PubMed  Google Scholar 

  25. Zhou X-M, Li W-Q, Wu Y-H et al (2018) Intrinsic expression of immune checkpoint molecule TIGIT could help tumor growth in vivo by suppressing the function of NK and CD8+ T cells. Front Immunol 9:2821. https://doi.org/10.3389/fimmu.2018.02821

    Article  CAS  PubMed  Google Scholar 

  26. Phase 2 CITYSCAPE Trial Shows Promise for Tiragolumab in NSCLC. In: Cancer network. https://www.cancernetwork.com/view/phase-2-cityscape-trial-shows-promise-for-tiragolumab-in-nsclc. Accessed 22 Mar 2021

  27. Kumar R, Yu F, Zhen Y-H et al (2017) PD-1 blockade restores impaired function of ex vivo expanded CD8+ T cells and enhances apoptosis in mismatch repair deficient EpCAM+PD-L1+ cancer cells. Onco Targets Ther 10:3453–3465. https://doi.org/10.2147/OTT.S130131

    Article  PubMed  Google Scholar 

  28. Pfannenstiel LW, Diaz-Montero CM, Tian YF et al (2019) Immune-checkpoint blockade opposes CD8+ T-cell suppression in human and murine cancer. Cancer Immunol Res 7:510–525. https://doi.org/10.1158/2326-6066.CIR-18-0054

    Article  CAS  PubMed  Google Scholar 

  29. Van den Eynde M, Mlecnik B, Bindea G et al (2018) The link between the multiverse of immune microenvironments in metastases and the survival of colorectal cancer patients. Cancer Cell 34:1012-1026.e3. https://doi.org/10.1016/j.ccell.2018.11.003

    Article  CAS  PubMed  Google Scholar 

  30. Halama N, Spille A, Lerchl T et al (2013) Hepatic metastases of colorectal cancer are rather homogeneous but differ from primary lesions in terms of immune cell infiltration. OncoImmunology 2:e24116. https://doi.org/10.4161/onci.24116

    Article  PubMed  Google Scholar 

  31. Jakubowska K, Koda M, Kisielewski W et al (2019) Tumor-infiltrating lymphocytes in primary tumors of colorectal cancer and their metastases. Exp Ther Med. https://doi.org/10.3892/etm.2019.8146

    Article  PubMed  Google Scholar 

  32. Miller BC, Sen DR, Al Abosy R et al (2019) Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol 20:326–336. https://doi.org/10.1038/s41590-019-0312-6

    Article  CAS  PubMed  Google Scholar 

  33. Blake SJ, Stannard K, Liu J et al (2016) Suppression of metastases using a new lymphocyte checkpoint target for cancer immunotherapy. Cancer Discov 6:446–459. https://doi.org/10.1158/2159-8290.CD-15-0944

    Article  CAS  PubMed  Google Scholar 

  34. Chan CJ, Martinet L, Gilfillan S et al (2014) The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions. Nat Immunol 15:431–438. https://doi.org/10.1038/ni.2850

    Article  CAS  PubMed  Google Scholar 

  35. Hung AL, Maxwell R, Theodros D et al (2018) TIGIT and PD-1 dual checkpoint blockade enhances antitumor immunity and survival in GBM. Oncoimmunology. https://doi.org/10.1080/2162402X.2018.1466769

    Article  PubMed  Google Scholar 

  36. Meyer D, Seth S, Albrecht J et al (2009) CD96 interaction with CD155 via its first Ig-like domain is modulated by alternative splicing or mutations in distal Ig-like domains. J Biol Chem 284:2235–2244. https://doi.org/10.1074/jbc.M807698200

    Article  CAS  PubMed  Google Scholar 

  37. Chiang EY, de Almeida PE, de Nagata DE et al (2020) CD96 functions as a co-stimulatory receptor to enhance CD8+ T cell activation and effector responses. Eur J Immunol 50:891–902. https://doi.org/10.1002/eji.201948405

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This study was carried out in partnership with the Roche Institute, which supplied us with atezolizumab and tiragolumab. We would like to thank Dr. Laurent Arnould and his anatomopathology laboratory for their help in the recovery of the tumor samples. Flow cytometry analyses were performed on Beckman Coulter Cytoflex. Thanks to Olivier Jaen for his help in setting up the immunomonitoring panels.

Funding

This research was supported and granted by Genentech, Inc.

Author information

Authors and Affiliations

Authors

Contributions

MT participated in the design of the study, the establishment of patient collection and draft the manuscript. LH performed patient collection, carried out the ex vivo and flow cytometry analysis and participated in analyse obtained data. EB and CT carried out all bioinformatics analysis. EL participated in the design of the study and helped to draft the manuscript. FG conceived of the study, and participated in its design and coordination and draft the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Francois Ghiringhelli.

Ethics declarations

Conflict of interest

The authors declare that there is no potential conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (XLSX 11 kb)

262_2022_3182_MOESM2_ESM.pptx

Supplementary file2Supplementary figure 1: Gating strategy for “Lymphoid cell analysis” panel. The gating strategy was done on a tumour cell suspension sample and is performed in the same way on blood samples. The expression of CD226, TIGIT and PD1 was studied in the same way as on CD8 T cells on NK cells. Expression of naive/memory subpopulations dependent on CD45RA and CCR7 expression are studied in the same way as CD8 T cells on CD4 T cells, as well as the expression of CD226, TIGIT and PD1. The expression of CD96 is positive for all CD4 and CD8 T cells, so we analyzed the median intensity fluorenscence of this marker on our populations of interest. Supplementary figure 2: Gating strategy for “Myeloid cell analysis” panel. The gating strategy was done on a blood sample and is performed in the same way on tumour samples. The expression of PD-L1, CD111, CD112 and CD155 markers was studied in each of the following populations: granulocytes (CD15+), macrophages (CD14+ CD163+), monocytic MDSCs (CD14+ CD15- CD163- HLA-DRlow) and granulocytic MDSCs (CD14+ CD15+). Supplementary figure 3: Gating strategy for “Lymphocyte function analysis” panel. The gating strategy is developed on the CD3+ CD45- blood cell population. The same gating strategy was applied to the CD3+ CD45+ population corresponding to TILs. TILs was pre-labeled with an anti-CD45 antibody so that blood and TILs could be analyzed in the same tube. For the study of cytokine expression, we first analysed the expression of TNFα and IFNγ on conventional CD4 T cells (T CD4conv), regulatory CD4 T cells (Treg), conventional CD8 T cells (T CD8conv) and regulatory CD8 T cells (T CD8reg) and then the expression of Granzyme B and IL-2 on each subpopulation. Secondly, we analysed within the CD4conv T cells each of the following subpopulations: Th1 (CD4+ Foxp3- IFNγ+ IL-17A-), Th17 (CD4+ Foxp3- IFNγ- IL-17A+), Th1/17 (CD4+ Foxp3- IFNγ+ IL-17A+) and Th2 (CD4+ Foxp3- IFNγ- IL-17A- IL-4+) cells. We performed the same analysis within CD8conv T cells: Tc1, Tc17, Tc1/17 and Tc2 cells. For each of these populations, we studied the expression of the cytokines TNFα and IL-2 as shown for Th1 in this gating strategy. Supplementary figure 4: Gating strategy for “Ex vivo restimulation analysis”. Supplementary figure 5: Double blockade of PD-L1 and TIGIT restore tumor infiltration lymphocytes function in some cancer patients with MSS CRC. Tumor cell suspensions from 16 patients with cancer colorectal cancers were thawed and after removing dead cells were labelled with 1µM of CellTrace Violet. Then, the cells were plated at 5.105 cells per well and stimulated with anti-CD3 antibody (10 ng/mL) in the presence or absence of 10 µg/mL of anti-PD-L1 (clone 6E11) and/or anti-TIGIT (clone 10A7) during 5 days. Cells were also stimulated with 5 µg/mL of anti-CD3 and anti-CD28 as positive control experiments. 20 hours before the end of the culture, Brefeldin A was added to block transport processes during cell activation and allow the staining of intracellular cytokines. After 5 days of culture, to study the effect of immune checkpoints blockade antibodies on the proliferation and cytotoxic capacities of TILs, tumor cell suspensions were stained with anti-CD3, anti-CD4, anti-CD8, anti-IFNγ and anti-TNFα and analysed by flow cytometry. A. The percentage of TNFα expression in CD4 (left) and CD8 (right) is studied for the following 3 conditions: anti-CD3, anti-CD3 plus anti-PD-L1 and anti-CD3 plus anti-PD-L1 + anti-TIGIT. Patients classified as responders to double immunotherapy are shown in colour. B. The percentage of IFNγ expression in CD4 (left) and CD8 (right) comparing the anti-CD3 plus anti-TIGIT condition to anti-CD3 alone is depicted. Supplementary figure 6: Description of the cell composition in the blood and tumour of patients. Fresh tumor tissues and associated blood samples for each patient (n=16) were stained and then analysed by flow cytometry. A. Box plots showing the frequency of CD45+ and CD45- populations in patients’ blood and tumor. B. Box plots showing the frequency of CD3+, CD4+ and CD8+ T cells, NK and NKT cells, CD11b+; CD14+, CD15+ cells and gMDSC population in patients’ blood and tumor. Supplementary figure 7: Association between baseline lymphoid cell number and response to comboimmunotherapy. Fresh tumor tissues and associated blood samples for each patient were taken on the day of surgery. After tumor dissociation, 1.106 tumor cell suspension were stained with anti-CD45, resuspended in 50 µL of whole blood sample and then transferred to a tube containing PMA, Ionomycin and Brefeldin A for 3 hours. After activation, cells were fixed, permeabilized and stained with anti-IFNγ, anti-TNFα, anti-GranzymeB, anti-IL-4, anti-Foxp3, anti-IL-17A, anti-CD3, anti-CD4, anti-CD8 and anti-IL-2 antibodies and analysed by flow cytometry. A-B. Box plots showing the different combinations of expression of the 4 cytokines IFNγ, TNFα, Granzyme B (GrB) and IL-2 in CD4 (CD45- CD3+ CD4+) (A) and CD8 (CD45- CD3+ CD8+) (B) T cells in blood patients’ depending on the responder (R) versus non responder (NR) status. C. The frequency of Granzyme B (GrzB) expression according to responder (R) or non-responder status is depicted in tumor samples. Statistical difference is determined by a Mann–Whitney test. ns = no significant, * = p< 0.05 and ** = p< 0.01. Supplementary figure 8: Association between baseline nectin expression and response to comboimmunotherapy. Fresh tumor tissues and associated blood samples for each patient were taken on the day of surgery. After tumor dissociation, 1.106 tumor cell suspension and 100 µL of whole blood sample were stained with anti-DNAM-1 (CD226), anti-CD56, anti-CD96, anti-CD45, anti-TIGIT, anti-PD-1, anti-CD3, anti-CD45RA, anti-CD8, anti-CCR7, anti-Tim-3 and anti-CD4 antibodies and with a mortality marker DRAQ7 and then analysed by flow cytometry. A-B. Box plots showing the frequency of CD226+, CD226-, TIGIT+ and TIGIT- populations in the blood (A) and tumor (B) samples by comparing expression on CD4, CD8 and NK populations. C-D. Box plots showing the different combinations of expression of the 2 nectins CD226 and TIGIT within CD4 T cells and CD8 T cells in patient blood (C) and tumor (D) according to responder (R) or non responder (NR) status. Statistical difference is determined by a Mann–Whitney test. ns = no significant and * = p< 0.05, ** = p<0.01, *** = p<0.001 and **** = p<0.0001. Supplementary figure 9: Association between baseline nectin expression and response to comboimmunotherapy. Box plots showing the median fluorescence intensity (MFI) of CD96 marker in CD4 T cells and CD8 T cells in patient blood depending on the responder (R) or non responder (NR) status. Statistical difference is determined by a Mann–Whitney test. ns = no significant and * = p< 0.05. Supplementary figure 10: Association between baseline myeloid cell number and response to comboimmunotherapy. Fresh tumor tissues and associated blood samples for each patient were taken on the day of surgery. After tumor dissociation, 1.106 tumor cell suspension and 100 µL of whole blood sample were stained with anti-CD11b, anti-HLA-DR, anti-CD111, anti-CD155, anti-CD112, anti-PD-L1, anti-CD163, anti-CD15, anti-CD14, anti-CD3, anti-CD19, anti-CD20, anti-CD56, anti-Galectin-9 and anti-CD45 antibodies and with a mortality marker DRAQ7 and then analysed by flow cytometry. The frequency of CD111, CD112 and CD155 positive populations in macrophages (CD14+ CD163+), mMDSC (CD14+ CD163- HLA-DRlow), gMDSC (CD15+ CD14+) and granulocytes (CD15+) in tumor samples according to responder (R) or non-responder (NR) status is depicted. Statistical difference is determined by a Mann–Whitney test. ns = no significant and * = p< 0.05. Supplementary figure 11: CD96 expression in lymphoid cells characterized a particular CRC subtype. A-B. Kaplan-Meier curves for overall survival with patients stratified according to the level of CD96 RNAseq expression, respectively for patients with high (A) or low (B) level of the metagene. (PPTX 3848 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Thibaudin, M., Limagne, E., Hampe, L. et al. Targeting PD-L1 and TIGIT could restore intratumoral CD8 T cell function in human colorectal cancer. Cancer Immunol Immunother 71, 2549–2563 (2022). https://doi.org/10.1007/s00262-022-03182-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00262-022-03182-9

Keywords

Navigation