Skip to main content

Advertisement

Log in

Spatiotemporal commonality of the TCR repertoire in a T-cell memory murine model and in metastatic human colorectal cancer

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

Abstract

Immune checkpoint inhibitors (ICIs) have shown superior clinical responses and significantly prolong overall survival (OS) for many types of cancer. However, some patients exhibit long-term OS, whereas others do not respond to ICI therapy at all. To develop more effective and long-lasting ICI therapy, understanding the host immune response to tumors and the development of biomarkers are imperative. In this study, we established an MC38 immunological memory mouse model by administering an anti-PD-L1 antibody and evaluating the detailed characteristics of the immune microenvironment including the T cell receptor (TCR) repertoire. In addition, we found that the memory mouse can be established by surgical resection of residual tumor following anti-PD-L1 antibody treatment with a success rate of > 40%. In this model, specific depletion of CD8 T cells revealed that they were responsible for the rejection of reinoculated MC38 cells. Analysis of the tumor microenvironment (TME) of memory mice using RNA-seq and flow cytometry revealed that memory mice had a quick and robust immune response to MC38 cells compared with naïve mice. A TCR repertoire analysis indicated that T cells with a specific TCR repertoire were expanded in the TME, systemically distributed, and preserved in the host for a long time period. We also identified shared TCR clonotypes between serially resected tumors in patients with colorectal cancer (CRC). Our results suggest that memory T cells are widely preserved in patients with CRC, and the MC38 memory model is potentially useful for the analysis of systemic memory T-cell behavior.

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
Fig. 6
Fig. 7

Similar content being viewed by others

Data availability

Bulk RNA-seq and TCR sequencing data presented in this work will be submitted through the Sequencing Read Archive. The remaining data generated and/or analyzed during the current study are available within the article and its Supplementary Data files or are available from the corresponding author upon reasonable request.

References

  1. Reck M, Rodriguez-Abreu D, Robinson AG et al (2016) Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med 375:1823–1833. https://doi.org/10.1056/NEJMoa1606774

    Article  CAS  PubMed  Google Scholar 

  2. Robert C, Schachter J, Long GV et al (2015) Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med 372:2521–2532. https://doi.org/10.1056/NEJMoa1503093

    Article  CAS  PubMed  Google Scholar 

  3. Waldman AD, Fritz JM, Lenardo MJ (2020) A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol 20:651–668. https://doi.org/10.1038/s41577-020-0306-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Larkin J, Chiarion-Sileni V, Gonzalez R et al (2019) Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 381:1535–1546. https://doi.org/10.1056/NEJMoa1910836

    Article  CAS  PubMed  Google Scholar 

  5. Reck M, Rodriguez-Abreu D, Robinson AG et al (2021) Five-year outcomes with pembrolizumab versus chemotherapy for metastatic Non-small-cell lung cancer with PD-L1 tumor proportion score >/= 50. J Clin Oncol Off J Am Soc Clin Oncol 39:2339–2349. https://doi.org/10.1200/JCO.21.00174

    Article  CAS  Google Scholar 

  6. Borcoman E, Kanjanapan Y, Champiat S, Kato S, Servois V, Kurzrock R, Goel S, Bedard P, Le Tourneau C (2019) Novel patterns of response under immunotherapy. Annals Oncol Off J European Soc Med Oncol 30:385–396. https://doi.org/10.1093/annonc/mdz003

    Article  CAS  Google Scholar 

  7. Zaretsky JM, Garcia-Diaz A, Shin DS et al (2016) Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 375:819–829. https://doi.org/10.1056/NEJMoa1604958

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ahern E, Solomon BJ, Hui R, Pavlakis N, O’Byrne K, Hughes BGM (2021) Neoadjuvant immunotherapy for non-small cell lung cancer: right drugs, right patient, right time? J Immunother Cancer. https://doi.org/10.1136/jitc-2020-002248

    Article  PubMed  PubMed Central  Google Scholar 

  9. Fridman WH, Zitvogel L, Sautes-Fridman C, Kroemer G (2017) The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol 14:717–734. https://doi.org/10.1038/nrclinonc.2017.101

    Article  CAS  PubMed  Google Scholar 

  10. Spitzer MH, Carmi Y, Reticker-Flynn NE et al (2017) Systemic immunity is required for effective cancer immunotherapy. Cell. 168:487-502 e15. https://doi.org/10.1016/j.cell.2016.12.022

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Allen BM, Hiam KJ, Burnett CE et al (2020) Systemic dysfunction and plasticity of the immune macroenvironment in cancer models. Nat Med. https://doi.org/10.1038/s41591-020-0892-6

    Article  PubMed  PubMed Central  Google Scholar 

  12. Cowell LG (2020) The diagnostic, prognostic, and therapeutic potential of adaptive immune receptor repertoire profiling in Cancer. Can Res 80:643–654. https://doi.org/10.1158/0008-5472.CAN-19-1457

    Article  CAS  Google Scholar 

  13. Li N, Yuan J, Tian W, Meng L, Liu Y (2020) T-cell receptor repertoire analysis for the diagnosis and treatment of solid tumor: a methodology and clinical applications. Cancer Commun 40:473–483. https://doi.org/10.1002/cac2.12074

    Article  Google Scholar 

  14. Han J, Zhao Y, Shirai K et al (2021) Resident and circulating memory T cells persist for years in melanoma patients with durable responses to immunotherapy. Nat Cancer 2:300–311. https://doi.org/10.1038/s43018-021-00180-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Su W, Sun J, Shimizu K, Kadota K (2019) TCC-GUI: a Shiny-based application for differential expression analysis of RNA-Seq count data. BMC Res Notes 12:133. https://doi.org/10.1186/s13104-019-4179-2

    Article  PubMed  PubMed Central  Google Scholar 

  16. Fang H, Yamaguchi R, Liu X et al (2014) Quantitative T cell repertoire analysis by deep cDNA sequencing of T cell receptor alpha and beta chains using next-generation sequencing (NGS). Oncoimmunology 3:e968467. https://doi.org/10.4161/21624011.2014.968467

    Article  CAS  PubMed  Google Scholar 

  17. Choudhury NJ, Kiyotani K, Yap KL et al (2016) Low T-cell receptor diversity, high somatic mutation burden, and high neoantigen load as predictors of clinical outcome in muscle-invasive bladder cancer. Eur Urol Focus 2:445–452. https://doi.org/10.1016/j.euf.2015.09.007

    Article  PubMed  Google Scholar 

  18. Schreiber K, Karrison TG, Wolf SP et al (2020) Impact of TCR diversity on the development of transplanted or chemically induced tumors. Cancer Immunol Res 8:192–202. https://doi.org/10.1158/2326-6066.CIR-19-0567

    Article  CAS  PubMed  Google Scholar 

  19. Bolotin DA, Poslavsky S, Davydov AN et al (2017) Antigen receptor repertoire profiling from RNA-seq data. Nat Biotechnol 35:908–911. https://doi.org/10.1038/nbt.3979

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wherry EJ (2011) T cell exhaustion. Nat Immunol 12:492–499. https://doi.org/10.1038/ni.2035

    Article  CAS  PubMed  Google Scholar 

  21. Mosely SI, Prime JE, Sainson RC et al (2017) Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery. Cancer Immunol Res 5:29–41. https://doi.org/10.1158/2326-6066.CIR-16-0114

    Article  CAS  PubMed  Google Scholar 

  22. Krishnan L, Gurnani K, Dicaire CJ, van Faassen H, Zafer A, Kirschning CJ, Sad S, Sprott GD (2007) Rapid clonal expansion and prolonged maintenance of memory CD8+ T cells of the effector (CD44highCD62Llow) and central (CD44highCD62Lhigh) phenotype by an archaeosome adjuvant independent of TLR2. J Immunol 178:2396–2406. https://doi.org/10.4049/jimmunol.178.4.2396

    Article  CAS  PubMed  Google Scholar 

  23. Spranger S, Spaapen RM, Zha Y, Williams J, Meng Y, Ha TT, Gajewski TF (2013) Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells. Sci Transl Med 5:200116. https://doi.org/10.1126/scitranslmed.3006504

    Article  CAS  Google Scholar 

  24. Matloubian M, Lo CG, Cinamon G, Lesneski MJ, Xu Y, Brinkmann V, Allende ML, Proia RL, Cyster JG (2004) Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature 427:355–360. https://doi.org/10.1038/nature02284

    Article  CAS  PubMed  Google Scholar 

  25. Forde PM, Chaft JE, Smith KN et al (2018) Neoadjuvant PD-1 Blockade in Resectable Lung Cancer. N Engl J Med 378:1976–1986. https://doi.org/10.1056/NEJMoa1716078

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Koebel CM, Vermi W, Swann JB, Zerafa N, Rodig SJ, Old LJ, Smyth MJ, Schreiber RD (2007) Adaptive immunity maintains occult cancer in an equilibrium state. Nature 450:903–907. https://doi.org/10.1038/nature06309

    Article  CAS  PubMed  Google Scholar 

  27. Schreiber RD, Old LJ, Smyth MJ (2011) Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science 331:1565–1570. https://doi.org/10.1126/science.1203486

    Article  CAS  PubMed  Google Scholar 

  28. Joshi K, de Massy MR, Ismail M et al (2019) Spatial heterogeneity of the T cell receptor repertoire reflects the mutational landscape in lung cancer. Nat Med 25:1549–1559. https://doi.org/10.1038/s41591-019-0592-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Borghaei H, Gettinger S, Vokes EE et al (2021) Five-Year Outcomes From the Randomized, Phase III Trials CheckMate 017 and 057: Nivolumab Versus Docetaxel in Previously Treated Non-Small-Cell Lung Cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 39:723–733. https://doi.org/10.1200/JCO.20.01605

    Article  CAS  PubMed  Google Scholar 

  30. Inamori K, Togashi Y, Fukuoka S et al. (2021) Importance of lymph node immune responses in MSI-H/dMMR colorectal cancer. JCI Insight. 6. doi: https://doi.org/10.1172/jci.insight.137365

  31. Gros A, Parkhurst MR, Tran E et al (2016) Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat Med 22:433–438. https://doi.org/10.1038/nm.4051

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Fairfax BP, Taylor CA, Watson RA et al (2020) Peripheral CD8(+) T cell characteristics associated with durable responses to immune checkpoint blockade in patients with metastatic melanoma. Nat Med 26:193–199. https://doi.org/10.1038/s41591-019-0734-6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Dammeijer F, van Gulijk M, Mulder EE et al (2020) The PD-1/PD-L1-checkpoint restrains T cell immunity in tumor-draining lymph nodes. Cancer Cell. https://doi.org/10.1016/j.ccell.2020.09.001

    Article  PubMed  Google Scholar 

  34. Francis DM, Manspeaker MP, Schudel A, Sestito LF, O’Melia MJ, Kissick HT, Pollack BP, Waller EK, Thomas SN (2020) Blockade of immune checkpoints in lymph nodes through locoregional delivery augments cancer immunotherapy. Sci Transl Med. https://doi.org/10.1126/scitranslmed.aay3575

    Article  PubMed  PubMed Central  Google Scholar 

  35. Forde PM, Spicer J, Lu S et al (2022) Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. N Engl J Med 386:1973–1985. https://doi.org/10.1056/NEJMoa2202170

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Felip E, Altorki N, Zhou C et al (2021) Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB–IIIA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, phase 3 trial. Lancet 398:1344–1357. https://doi.org/10.1016/s0140-6736(21)02098-5

    Article  CAS  PubMed  Google Scholar 

  37. Yost KE, Satpathy AT, Wells DK et al (2019) Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med 25:1251–1259. https://doi.org/10.1038/s41591-019-0522-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Bassez A, Vos H, Van Dyck L et al (2021) A single-cell map of intratumoral changes during anti-PD1 treatment of patients with breast cancer. Nat Med 27:820–832. https://doi.org/10.1038/s41591-021-01323-8

    Article  CAS  PubMed  Google Scholar 

  39. Nagasaki J, Inozume T, Sax N et al (2022) PD-1 blockade therapy promotes infiltration of tumor-attacking exhausted T cell clonotypes. Cell Rep. 38:110331. https://doi.org/10.1016/j.celrep.2022.110331

    Article  CAS  PubMed  Google Scholar 

  40. Okamura K, Nagayama S, Tate T, Chan HT, Kiyotani K, Nakamura Y (2022) Lymphocytes in tumor-draining lymph nodes co-cultured with autologous tumor cells for adoptive cell therapy. J Transl Med 20:241. https://doi.org/10.1186/s12967-022-03444-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kudo Y, Haymaker C, Zhang J et al (2019) Suppressed immune microenvironment and repertoire in brain metastases from patients with resected non-small-cell lung cancer. Annals Oncol Off J European Soc Med Oncol 30:1521–1530. https://doi.org/10.1093/annonc/mdz207

    Article  CAS  Google Scholar 

  42. Chen DS, Mellman I (2017) Elements of cancer immunity and the cancer-immune set point. Nature 541:321–330. https://doi.org/10.1038/nature21349

    Article  CAS  PubMed  Google Scholar 

  43. Simoni Y, Becht E, Fehlings M et al (2018) Bystander CD8(+) T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 557:575–579. https://doi.org/10.1038/s41586-018-0130-2

    Article  CAS  PubMed  Google Scholar 

  44. Schoenfeld AJ, Hellmann MD (2020) Acquired resistance to immune checkpoint inhibitors. Cancer Cell 37:443–455. https://doi.org/10.1016/j.ccell.2020.03.017

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Rosenberg SA, Restifo NP (2015) Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348:62–68. https://doi.org/10.1126/science.aaa4967

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Morgan RA, Dudley ME, Wunderlich JR et al (2006) Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 314:126–129. https://doi.org/10.1126/science.1129003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Rapoport AP, Stadtmauer EA, Binder-Scholl GK et al (2015) NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med 21:914–921. https://doi.org/10.1038/nm.3910

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Oliveira G, Stromhaug K, Klaeger S et al (2021) Phenotype, specificity and avidity of antitumour CD8(+) T cells in melanoma. Nature 596:119–125. https://doi.org/10.1038/s41586-021-03704-y

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Denis M, Mathe D, Micoud M, Choffour PA, Grasselly C, Matera EL, Dumontet C (2022) Impact of mouse model tumor implantation site on acquired resistance to anti-PD-1 immune checkpoint therapy. Front Immunol 13:1011943. https://doi.org/10.3389/fimmu.2022.1011943

    Article  CAS  PubMed  Google Scholar 

  50. Yu J, Green MD, Li S et al (2021) Liver metastasis restrains immunotherapy efficacy via macrophage-mediated T cell elimination. Nat Med 27:152–164. https://doi.org/10.1038/s41591-020-1131-x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank Dr. Ai Takemoto, Ms. Junko Sugihara, Ms. Saki Matsumoto, and Ms. Satoko Baba for their help in technical assistance for the in vitro and in vivo study.

Funding

This study was supported in part by MEXT/JSPS KAKENHI grant number JP22K18383 (to R. Katayama), JP20K07399 (to S. Sakata) and the grant from the AMED grant number JP21ck0106472h0003, JP23ama221201h0002, JP23ama221210h0002, and JP23ck0106795h0001 (to R. Katayama), JP22ck0106543h0003 (to K. Kiyotani) and the grants from the Princess Takamatsu Cancer Research Fund, the Cell Science Research Foundation, and Chugai Foundation for Innovative Drug Discovery Science (to R. Katayama), and the grants from the Nippon Foundation and Takeda Science Foundation.

Author information

Authors and Affiliations

Authors

Contributions

MH, KK, KT, and RK designed the research. MH, KK, TT, SS, RS, SN, KT, and RK conducted the experiments and collected the data. MH, KK, TT, SS, ST, SN, KT, and RK analyzed the data. MH and RK wrote the initial draft of the manuscript. All authors provided critical review of the manuscript.

Corresponding author

Correspondence to Ryohei Katayama.

Ethics declarations

Conflict of interest

R. Katayama received research funding from Chugai, TAKEDA, Toppan Printing outside the submitted work. K. Kiyotani is a scientific adviser of Cancer Precision Medicine, Inc. Other authors have declared that no conflict of interest exists.

Ethics approval and consent to participate

Informed consent was obtained from all individual participants included in the study. All experiments involving CRC patients were performed in accordance with approved protocols by the Institutional Review Board of Japanese Foundation for Cancer Research (No. 2018-GA-1021). All mice studies were conducted in line with the protocols approved by the Committee for the Use and Care of Experimental Animals of the Japanese Foundation for Cancer Research (No. 10-01-20). The authors affirm that human research participants provided informed consent for publication of this study.

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.

262_2023_3473_MOESM1_ESM.pdf

Supplementary file. Fig. S1 MC38 escape host immune attack using the PD-1/PD-L1 interaction. (A) MC38 cells were subcutaneously injected into C57BL/6 mice. The tumor volumes were measured and shown as the mean ± SEM at each time point. (B) The percentage of CD8a + T cells in CD45 + cells in MC38 tumors and the expression of PD-1 were measured using flow cytometry at each time point. Day 3, 6, 9 and 12, n = 3 per time point. (C)The expression of TNF, IFN, GzmB, and Ki67 in CD8 + TILs was measured using flow cytometry at each time point. Day 3, 6, 9 and 12, n = 3 per time point. (D) MC38 PD-L1+ / + and MC38 PD-L1− / − cells were stimulated overnight with 50 ng/ml IFN to induce PD-L1 expression and PD-L1 expression was measured using flow cytometry. (E) Genomic deletion around the PD-L1 locus was confirmed by Sanger sequencing of the PCR products around the sgRNA targeting region using genomic DNA from MC38 PD-L1− / −. (F) The changes in tumor volume are plotted individually and correspond to Fig. 1A. (G) The changes in tumor volume are plotted individually and correspond to Fig. 1B. Error bars represent the mean ± SEM (A, B, C). Fig. S2 MHC expression in MC38 B2M− / − cells. (A) Genomic deletion around the B2M locus was confirmed by Sanger sequencing of the PCR products around the sgRNA targeting region using genomic DNA from MC38 B2M− / − cells. (B)The expression of pan-MHC class I on the surface of MC38 B2M+ / + and MC38 B2M− / − cells was measured using flow cytometry. Fig. S3 Acquired immune responses are increased in TME of memory mice and FTY720 did not abrogate tumor rejection of memory mice. (A) Related GO terms of genes specifically decreased in the TME of the memory mice (upper). Related GO terms of genes specifically increased in the TME of the memory mice (lower). (B) The frequency of Treg cells in the TME was measured using flow cytometry (naïve n = 3, memory n = 3). (C) The frequency of Gr1 + CD11b + cells in the TME was measured using flow cytometry (naïve n = 3, memory n = 3). (D) The expression of PD-1 on CD8 + T cells in the TME was measured using flow cytometry. (E) The expression of PD-L1 on the CD45 − cells in the TME was measured using flow cytometry. (F) The confirmed memory mice and naïve mice were administrated 75 μg/mouse of FTY720 (n = 5) or vehicle (EtOH, n = 5) 3 times a week before MC38 reinjection. The frequency of CD8 + T cells (left) or CD4 + T cells (right) in PBMCs was evaluated using flow cytometry. (G) MC38 cells were subcutaneously injected into memory mice on day 0. FTY720 injection was started on day 1 following tumor injection. Tumor volume was measured and plotted individually (FTY720 n = 9, EtOH n = 5, naïve n = 3). (H) MC38 cells were subcutaneously injected into memory mice on days 0 and 25. FTY720 injection was started before tumor injection (FTY720 n = 5, EtOH n = 5, naïve n = 3). Tumor volume was measured and plotted individually. Error bars represent the mean ± SEM (B, C, F). Data were analyzed by a two-tailed unpaired Student’s t test (B, C, F). Fig. S4 Neoadjuvant ICI treatment induces immunological memory against tumors. (A) The average number of the clonotypes contained in the right and left side tumors (naïve n = 4, memory n = 3). (B) MC38 cells were subcutaneously injected into C57BL/6 mice. The tumor-bearing mice were administrated 100 µg/mouse anti-PD-L1 antibody intraperitoneally twice a week for a total of five times, and then, the residual tumors were surgically resected. The change in tumor volume is plotted individually. (C) The change in the proportion of the shared among the resected primary tumor as well as the first and second reinjected tumors in the other memory mice corresponding to Fig. 6D. Error bars represent the mean ± SEM (A). Data were analyzed by a two-tailed unpaired Student’s t test (A). Fig. S5 Clinical course of the CRC patient. The clinical course of the CRC patient (C053) is shown. Briefly, the patient was a 71-year-old with Stage IVa colorectal cancer. Her tumor contained a KRAS G12V mutation, BRAF WT, and MSI-L/MSS. The patient was treated with primary tumor resection followed by chemotherapy. Five months later, she had an operation to resect the liver metastasis and received additional chemotherapy; however, new metastases appeared in the liver and lung sequentially and they were resected. The change in CT imaging for each resected tumor is shown. Fig. S6 Representative IHC images of C053. Representative IHC images (CD8, PD-1, GzmB) for Tumor (A), LS1-3 (B), LS4 (C), Lung (D). B2M staining of the Lung tumor is shown (E). Scale bar: 300 μm (A–D), 200 μm (E). Fig. S7 The shared TCR repertoires are observed in other patients. The resected tumor was prepared, and TCR sequencing was performed by next-generation sequencing. Only TCRs with a proportion of ≥ 0.1% were selected and presented. The change in the proportion of the shared TCR clonotype is shown. (A) C124, diagnosed as a colorectal neuroendocrine tumor. The patient recurred with liver metastasis after resection of the primary tumor. Each tumor was analyzed using TCR repertoire sequencing. (B) C105, diagnosed as colorectal cancer with lung metastasis. Each tumor was analyzed using TCR repertoire sequencing. (C) C152, diagnosed as colorectal cancer with liver metastasis. Each tumor was analyzed using TCR repertoire sequencing. Fig. S8 TILs presented in LS4 respond against other resected tumors. TILs from each resected tumor were cocultured with individual cancer cells, and specific immune responses were evaluated using an IFNγ ELISpot assay. PMA/Ionomycin was used as a positive control. The number in the upper right corner of the well represents the number of counted spots. Fig. S9 Expression of tissue resident memory T-cell markers in the skin of memory mouse. (A) The scheme of the experiment is shown. MC38 cells were subcutaneously injected into memory and naïve mouse at right side on day 0, and skin at the left side of mouse was surgically resected on days 3 after euthanized the mice. (B,C) CD8 + T cells were stained with the fluorescent antibody to detect CD45/CD3/CD4 or CD8/CD69/CD103 with live-dead detecting reagent and analyzed with multi-color flow cytometry. CD69 expression in CD8 + T-cell in the mouse skin was shown in histogram (B), and CD69 with CD103 expression was shown in dot plot (C)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Haraguchi, M., Kiyotani, K., Tate, T. et al. Spatiotemporal commonality of the TCR repertoire in a T-cell memory murine model and in metastatic human colorectal cancer. Cancer Immunol Immunother 72, 2971–2989 (2023). https://doi.org/10.1007/s00262-023-03473-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00262-023-03473-9

Keywords

Navigation