Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Resident and circulating memory T cells persist for years in melanoma patients with durable responses to immunotherapy

Abstract

While T-cell responses to cancer immunotherapy have been avidly studied, long-lived memory has been poorly characterized. In a cohort of metastatic melanoma survivors with exceptional responses to immunotherapy, we probed memory CD8+ T-cell responses across tissues, and across several years. Single-cell RNA sequencing revealed three subsets of resident memory T (TRM) cells shared between tumors and distant vitiligo-affected skin. Paired T-cell receptor sequencing further identified clonotypes in tumors that co-existed as TRM in skin and as effector memory T (TEM) cells in blood. Clonotypes that dispersed throughout tumor, skin and blood preferentially expressed an IFNG/TNF-high signature, which had a strong prognostic value for patients with melanoma. Remarkably, clonotypes from tumors were found in patient skin and blood up to 9 years later, with skin maintaining the most focused tumor-associated clonal repertoire. These studies reveal that cancer survivors can maintain durable memory as functional, broadly distributed TRM and TEM compartments.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Overlapping transcriptional signatures of CD8+ T cells from skin and tumor of long-term melanoma survivors.
Fig. 2: TRM cells in skin and tumor are composed of three subpopulations with discrete features and prognostic signatures.
Fig. 3: Promiscuously distributed CD8+ T-cell clonotypes show a propensity to form TRM-IFNG cells in skin and tumor.
Fig. 4: Melanoma antigen-specific T cells accumulate in skin and blood and are capable of long-lived functional recall.
Fig. 5: Tumor-associated T-cell clonotypes persist for up to 9 years, with skin sustaining a focused repertoire.
Fig. 6: CD8+ T-cell clones from tumors persist as TRM cells in skin and as TEM cells in blood.

Similar content being viewed by others

Data availability

Single-cell RNA-seq and TCR-seq data that support the findings of this study have been deposited in the Database of Genotypes and Phenotypes (dbGaP) under the accession code phs002309.v1.p1. Bulk TCR-seq data can be accessed through the ImmuneACCESS database of Adaptive Biotechnologies (https://doi.org/10.21417/JH2021NC; https://clients.adaptivebiotech.com/pub/han-2021-natcancer). The published microarray datasets used to generate the comprehensive CD8+ TRM signature for the GSEA analysis were accessible at the Gene Expression Omnibus (GEO) under accession codes GSE47045, GSE15907 and GSE37448. The remaining gene sets used in the GSEA analysis were accessible through the MSigDB database (https://www.gsea-msigdb.org/gsea/msigdb). The published TCGA skin cutaneous melanoma (SKCM) RNA-seq data used to perform the survival analysis are available at Firehose (http://gdac.broadinstitute.org/). Two additional previously published stage III/IV melanoma patient RNA-seq datasets are available at the GEO database with the following accession numbers: GSE54467 and GSE19234. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

Code availability

The open-source code is available at GitHub. Codes for gene expression analyses, including single-cell RNA-seq data analysis, survival analysis and GSEA analysis, are publicly available on GitHub (https://github.com/TrmMelanoma/Gene-expression-related-analysis). Codes for TCR analyses, including single-cell TCR-seq data analysis and bulk TCR-seq data analysis, are publicly available on GitHub (https://github.com/TrmMelanoma/TCR-analysis).

References

  1. Larkin, J. et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 381, 1535–1546 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Robert, C. et al. Nivolumab in previously untreated melanoma without BRAF mutation. N. Engl. J. Med. 372, 320–330 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Weber, J. S. et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 16, 375–384 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Huang, A. C. et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 545, 60–65 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Krieg, C. et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat. Med. 24, 144–153 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Ribas, A. et al. PD-1 blockade expands intratumoral memory T cells. Cancer Immunol. Res. 4, 194–203 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013.e20 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Freeman-Keller, M. et al. Nivolumab in resected and unresectable metastatic melanoma: characteristics of immune-related adverse events and association with outcomes. Clin. Cancer Res. 22, 886–894 (2016).

    Article  CAS  PubMed  Google Scholar 

  9. Hua, C. et al. Association of vitiligo with tumor response in patients with metastatic melanoma treated with pembrolizumab. JAMA Dermatol. 152, 45–51 (2016).

    Article  PubMed  Google Scholar 

  10. Nakamura, Y. et al. Correlation between vitiligo occurrence and clinical benefit in advanced melanoma patients treated with nivolumab: a multi-institutional retrospective study. J. Dermatol. 44, 117–122 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Malik, B. T. et al. Resident memory T cells in the skin mediate durable immunity to melanoma. Sci. Immunol. 2, eaam6346 (2017).

  12. Gerlach, C. et al. The chemokine receptor CX3CR1 defines three antigen-experienced CD8 T cell subsets with distinct roles in immune surveillance and homeostasis. Immunity 45, 1270–1284 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Sallusto, F., Lenig, D., Forster, R., Lipp, M. & Lanzavecchia, A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401, 708–712 (1999).

    Article  CAS  PubMed  Google Scholar 

  14. Herndler-Brandstetter, D. et al. KLRG1+ effector CD8+ T cells lose KLRG1, differentiate into all memory T cell lineages, and convey enhanced protective immunity. Immunity 48, 716–729.e8 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Guo, X. et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat. Med. 24, 978–985 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Martin, M. D. & Badovinac, V. P. Defining memory CD8 T cell. Front. Immunol. 9, 2692 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Muri, J. et al. The thioredoxin-1 system is essential for fueling DNA synthesis during T-cell metabolic reprogramming and proliferation. Nat. Commun. 9, 1851 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Wei, H. et al. Cutting edge: Foxp1 controls naive CD8+ T cell quiescence by simultaneously repressing key pathways in cellular metabolism and cell cycle progression. J. Immunol. 196, 3537–3541 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Zheng, C. et al. Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell 169, 1342–1356 e1316 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Szabo, P. A. et al. Single-cell transcriptomics of human T cells reveals tissue and activation signatures in health and disease. Nat. Commun. 10, 4706 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Hurton, L. V. et al. Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells. Proc. Natl Acad. Sci. USA 113, E7788–E7797 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gattinoni, L. et al. A human memory T cell subset with stem cell-like properties. Nat. Med. 17, 1290–1297 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Molodtsov, A. & Turk, M. J. Tissue resident CD8 memory T cell responses in cancer and autoimmunity. Front. Immunol. 9, 2810 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Kumar, B. V. et al. Human tissue-resident memory T cells are defined by core transcriptional and functional signatures in lymphoid and mucosal sites. Cell Rep. 20, 2921–2934 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wu, T. D. et al. Peripheral T cell expansion predicts tumour infiltration and clinical response. Nature 579, 274–278 (2020).

    Article  CAS  PubMed  Google Scholar 

  26. Mackay, L. K. et al. The developmental pathway for CD103+CD8+ tissue-resident memory T cells of skin. Nat. Immunol. 14, 1294–1301 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Cheuk, S. et al. CD49a expression defines tissue-resident CD8+ T cells poised for cytotoxic function in human skin. Immunity 46, 287–300 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Li, H. et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell 176, 775–789.e18 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Savas, P. et al. Single-cell profiling of breast cancer T cells reveals a tissue-resident memory subset associated with improved prognosis. Nat. Med. 24, 986–993 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. Pasetto, A. et al. Tumor- and neoantigen-reactive T-cell receptors can be identified based on their frequency in fresh tumor. Cancer Immunol. Res. 4, 734–743 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Fairfax, B. P. et al. Peripheral CD8+ T cell characteristics associated with durable responses to immune checkpoint blockade in patients with metastatic melanoma. Nat. Med. 26, 193–199 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Haymaker, C. L. et al. Metastatic melanoma patient had a complete response with clonal expansion after whole brain radiation and PD-1 blockade. Cancer Immunol. Res. 5, 100–105 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Tarhini, A. et al. Neoadjuvant ipilimumab (3 mg/kg or 10 mg/kg) and high dose IFN-ɑ2b in locally/regionally advanced melanoma: safety, efficacy and impact on T-cell repertoire. J. Immunother. Cancer 6, 112 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Edwards, J. et al. CD103+ tumor-resident CD8+ T cells are associated with improved survival in immunotherapy-naive melanoma patients and expand significantly during anti-PD-1 treatment. Clin. Cancer Res. 24, 3036–3045 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Park, S. L. et al. Tissue-resident memory CD8+ T cells promote melanoma-immune equilibrium in skin. Nature 565, 366–371 (2019).

    Article  CAS  PubMed  Google Scholar 

  36. Shankaran, V. et al. IFNγ and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 410, 1107–1111 (2001).

    Article  CAS  PubMed  Google Scholar 

  37. Higgs, B. W. et al. Interferon gamma messenger RNA signature in tumor biopsies predicts outcomes in patients with non-small cell lung carcinoma or urothelial cancer treated with durvalumab. Clin. Cancer Res. 24, 3857–3866 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Karachaliou, N. et al. Interferon gamma, an important marker of response to immune checkpoint blockade in non-small cell lung cancer and melanoma patients. Ther. Adv. Med. Oncol. 10, 1758834017749748 (2018).

  39. Alspach, E., Lussier, D. M. & Schreiber, R. D. Interferon gamma and its important roles in promoting and inhibiting spontaneous and therapeutic cancer immunity. Cold Spring Harb. Perspect. Biol. 11, a028480 (2019).

  40. Gaide, O. et al. Common clonal origin of central and resident memory T cells following skin immunization. Nat. Med. 21, 647–653 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Slutter, B. et al. Dynamics of influenza-induced lung-resident memory T cells underlie waning heterosubtypic immunity. Sci. Immunol. 2, eaag2031 (2017).

  42. Fonseca, R. et al. Developmental plasticity allows outside-in immune responses by resident memory T cells. Nat. Immunol. 21, 412–421 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Jiang, T. et al. Tumor neoantigens: from basic research to clinical applications. J. Hematol. Oncol. 12, 93 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Schumacher, T. N. & Schreiber, R. D. Neoantigens in cancer immunotherapy. Science 348, 69–74 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Elpek, K. G. et al. The tumor microenvironment shapes lineage, transcriptional, and functional diversity of infiltrating myeloid cells. Cancer Immunol. Res. 2, 655–667 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Painter, M. W. et al. Transcriptomes of the B and T lineages compared by multiplatform microarray profiling. J. Immunol. 186, 3047–3057 (2011).

    Article  CAS  PubMed  Google Scholar 

  50. Bolstad, B. M., Irizarry, R. A., Astrand, M. & Speed, T. P. A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics 19, 185–193 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. Leek, J. T., Johnson, W. E., Parker, H. S., Jaffe, A. E. & Storey, J. D. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics 28, 882–883 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Varn, F. S., Wang, Y., Mullins, D. W., Fiering, S. & Cheng, C. Systematic pan-cancer analysis reveals immune cell interactions in the tumor microenvironment. Cancer Res. 77, 1271–1282 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhao, Y. et al. A leukocyte infiltration score defined by a gene signature predicts melanoma patient prognosis. Mol. Cancer Res. 17, 109–119 (2019).

    Article  CAS  PubMed  Google Scholar 

  56. Menares, E. et al. Tissue-resident memory CD8+ T cells amplify anti-tumor immunity by triggering antigen spreading through dendritic cells. Nat. Commun. 10, 4401 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful for the generosity of all the patients who volunteered their time and tissue for this study. We thank the nursing staff of the General Surgery clinic at Dartmouth-Hitchcock Medical Center led by L. O’Rourke, and the Norris Cotton Cancer Center melanoma research team, especially B. Highhouse and M. Stannard, for assistance in patient recruitment and coordination. We appreciate the effort from the core facilities—the Dartlab and the Single Cell Genomics Core—at Dartmouth. We thank G. Ward at the Dartlab for FACS sorting expertise. We thank M. Pasca di Magliano for discussion and advice. This work was funded by The Dartmouth CTSA (grant NIH KL2TR0010), the American Cancer Society (grant CSDG 18-167-01), the Dow-Crichlow Career Development Award in Surgery and the Society of Surgical Oncology Clinical Investigator Award to C.V.A.; grant NIH R01 CA225028 and The Knights of the York Cross of Honour Philanthropic Fund to M.J.T.; a Borroughs Welcome BDLS Training Grant to J.H.; grant NIH F31CA232554 to A.M.; and support from grant 5P30 CA023108-40 (Immune Monitoring and Genomics and Molecular Biology Shared Resources). Single-cell sequencing was conducted at the Dartmouth Center for Quantitative Biology with support from NIGMS (grant P20GM130454) and NIH S10 (grant S10OD025235) awards. The views expressed are those of the authors and not necessarily those of the NIH or the American Cancer Society.

Author information

Authors and Affiliations

Authors

Contributions

C.V.A. and M.J.T. conceived and supervised the study. J.H., M.J.T. and C.V.A. drafted the paper and figures. J.H. and Y.Z. carried out the primary analysis. C.V.A., M.S.E. and K.S. carried out patient recruitment. J.H., J.L.F., P.Z. and T.G.S. processed tissues and carried out flow cytometry and FACS. J.H. and T.G.S. extracted the DNA for bulk TCR sequencing. S.Y. provided dermatopathology expertise and assisted with IHC analysis. F.W.K. carried out scRNA and scTCR library preparation and sequencing. C.C., Y.Z. and A.M. provided bioinformatic support. C.V.A., K.S. and J.M.B. collated the clinical data. J.G. provided statistical support. M.J.T. provided scientific and infrastructural support. All authors reviewed and edited the final paper.

Corresponding authors

Correspondence to Mary Jo Turk or Christina V. Angeles.

Ethics declarations

Competing interests

All authors declare no competing interests.

Additional information

Peer review information Nature Cancer thanks Adil Daud, Ansuman Satpathy and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Summary of melanoma patient treatments and specimens.

Detailed summary of the melanoma clinical stage, types and durations of immunotherapy treatments, and specimens collected per patient. Large arrow indicates the timeline for individual patient (not to scale). The time between date of last treatment and tissue biopsy is annotated in black. Small arrows show the timepoints of each specimen collection along with the experiments done for each specimen annotated in parentheses. scRNA=scRNA/scTCR-seq; flow = flow cytometry; Adaptive = TCR Vb DNA sequencing. Different colors indicate different types of immunotherapy as depicted in the legend.

Extended Data Fig. 2 FACS gating strategy.

Pseudocolor graphs show the gating strategy for FACS of each tissue. CD45+CD8+ and CD45+CD4+ cells were sorted by FACS into the same well. Values of x and y axis represent fluorescence intensities. Percentages of cell populations were labelled in each graph.

Extended Data Fig. 3 Resident memory CD8+ T cells expressing CD69 reside in both the epidermis and dermis in long-term metastatic melanoma survivors with immunotherapy-associated vitiligo.

a, Representative pseudocolor dot plots showing the gating strategy for the live CD45+ CD3+ CD8+ population. b, Contour plots show the expression of CD69, CD103 and CD62L on pre-gated CD8+ population by flow cytometry. The number represents the proportion out of total CD8+ T cells. c, Immunohistochemistry staining for CD8+ CD69+ resident memory CD8+ T cells in the skin from patient PT628. Images are representative of multiple fields from at least three skin sections taken from each of 4 individual patients. CD8 was stained in green and CD69 was stained in red. CD8+ and CD69+ single-stain cells are indicated by green and red arrows, respectively. Black cells (indicated by black arrows) are CD8+CD69+ co-stained cells. Original magnification: 10X (left) and 40X (right).

Extended Data Fig. 4 Patient contributions to each CD8+ T-cell cluster.

Pie charts depicting the proportion of cells from each patient to the total number of cells in each cluster (C1-C10). Each color represents one individual patient, with the absolute number of cells from each patient labeled in the corresponding slice of the pie chart.

Extended Data Fig. 5 Transcriptional profiles of clusters C8-C10 are enriched in consensus TRM gene lists while the transcription profiles of clusters C1-C7 are not enriched for core TRM genes.

a, GSEA analysis showing that the upregulated genes of TRM were enriched in the downregulated genes in clusters C1-C7 demonstrating that only clusters C8-C10 have key features of TRM. NESs and FDR q-values are shown for each gene set. The statistics were performed by the two-sample Kolmogorov-Smirnov test. b, Heatmaps depict the z-transformed mean expression of published consensus skin or tumor TRM gene lists across CD8+ T cell clusters C1-C10. c, Venn diagrams show the number of genes that overlap between the marker gene list of each cluster (red circle) with the published human cancer TRM gene list (blue circle). The overlap levels between two gene lists were evaluated by two-sided fisher exact test. Enrichment scores and P-values are labeled accordingly. d, Venn diagrams show the number of genes that overlap between the marker gene list of each cluster (red circle) with the published mouse skin/gut/lung TRM gene list (blue circle). The overlap levels between two gene lists were evaluated by two-sided fisher exact test. Enrichment scores and P-values are labeled accordingly. e, Heatmap depicts the z-transformed mean expression of a published melanoma infiltrating dysfunctional CD8+ gene list across clusters C1-C10.

Extended Data Fig. 6 TRM-IFNG signature is superior in predicting the survival of stage III/IV melanoma patients.

a, Kaplan–Meier overall survival curves of melanoma patients from two different published datasets, GSE54467 (top, n=75 patients) and GSE19324 (bottom, n=44 patients), stratified by enrichment of signatures derived from each of three TRM clusters. High and low groups were separated by the median value of the Z-transformed normalized mean expression of each gene set (top: N= 37 patients high and N=38 patients low; bottom: N=22 patients high and N=22 patients low). P-values were calculated by two-sided log-rank test. OR P-values were calculated by multivariate Cox regression. b, Multivariable cox survival regression model evaluating the individual contribution of different variables to the prognosis of melanoma patients from the above datasets, GSE54467 (left) and GSE19324 (right). Forest plots show the means of hazard ratios (HRs) represented by blue squares, the 95% confidence intervals of HRs represented by horizontal bars, and p-values calculated by the two-sided Wald test for each variable.

Extended Data Fig. 7 Tumor-associated TCR clonotypes in the skin and blood of long-term metastatic melanoma survivors.

a, Gini indexes of each tissue (left) or each TRM cluster (right) calculated for n=4 individual patients, showing no significant differences in baseline clonal expansion among different tissues or different TRM clusters. The lines indicate the average Gini index across all four patients for each cluster. Two-sided Wilcoxon test. b, Venn diagrams showing the number of matched TCR clonotypes between different specimens from individual patients. Colors indicate different tissue origins. The number of TCR clonotypes belonging to each group was labeled accordingly. c, The distribution of the remaining Resident/Circulating clonotypes (11/15) to the UMAP plot. Dots indicate CD8+ T cells from the same clone. Colors designate different specimen types. d, The distribution of the remaining Resident-Only clonotypes (14/18) to the UMAP plot. Dots indicate CD8+ T cells from the same clone. Colors designate different specimen types. e, The distribution of the remaining Circulation-Capable clonotypes (7/11) to the UMAP plot. Dots indicate CD8+ T cells from the same clone. Colors designate different specimen types.

Source data

Extended Data Fig. 8 Tumor-associated clonotypes in the skin and blood of patients.

Venn diagrams showing the number of matched TCR clonotypes between skin, blood and historically banked tumors of each patient. The number of TCR clonotypes belonging to each group was labeled accordingly.

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1–7.

Source data

Source Data Fig. 1

Individual graphed data points.

Source Data Fig. 3

Individual graphed data points.

Source Data Fig. 4

Individual graphed data points.

Source Data Fig. 5

Individual graphed data points.

Source Data Fig. 6

Individual graphed data points.

Source Data Extended Data Fig. 7

Individual graphed data points.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

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

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-021-00180-1

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer