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.

  • Review Article
  • Published:

Tissue-resident memory T cells in breast cancer control and immunotherapy responses

Abstract

The presence of tumour-infiltrating lymphocytes (TILs) is associated with favourable outcomes in patients with breast cancer as well as in those with other solid tumours. T cells make up a considerable proportion of TILs and current evidence suggests that CD8+ T cells are a crucial determinant of favourable clinical outcomes. Studies involving tumour material from numerous solid tumour types, including breast cancer, demonstrate that the CD8+ TILs include a subpopulation of tissue-resident memory T (TRM) cells. This subpopulation has features consistent with those of TRM cells, which have been described as having a role in peripheral immune surveillance and viral immunity in both humans and mice. Patients with early-stage triple-negative breast cancers harbouring greater numbers of TRM cells have a substantially improved prognosis and longer overall survival. Furthermore, patients with advanced-stage breast cancers with higher levels of TRM cells have increased response rates to anti-PD-1 antibodies. These findings have motivated efforts to explore whether CD8+ TRM cells include tumour-specific T cells, their functional responses to cognate antigens and their role in responses to immune checkpoint inhibition. In this Review, we focus on the clinical significance of CD8+ TRM cells and the potential ways that these cells can be targeted to improve the success of immunotherapeutic approaches in patients with breast cancer, as well as in those with other solid tumour types.

Key points

  • The clinical and biological importance of qualitative differences in tumour-infiltrating lymphocyte (TIL) populations in patients with solid tumours, including breast cancer, is an area of intensive research.

  • Quantification of TILs is a reliable and robust prognostic biomarker in the management of patients with breast cancer, particularly in those with the triple-negative or HER2-overexpressing disease subtypes.

  • CD8+ tissue-resident memory T (TRM) cells have been identified as TILs in patients with various solid tumours, including those with breast cancer, and are a distinct subpopulation of CD8+ TILs.

  • High levels of expression of a TRM cell signature derived from single-cell RNA sequencing are associated with a favourable prognosis and an increased likelihood of a response to the anti-PD-1 antibody pembrolizumab in patients with triple-negative breast cancers, thus supporting the importance of this immune subset.

  • TRM cells express high levels of cytotoxic molecules, such as granzymes, and immune-checkpoint proteins and might be a key TIL subset targeted by several immunotherapies.

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: The potential role of TRM cells in antitumour immunity.

Similar content being viewed by others

References

  1. Savas, P. et al. Clinical relevance of host immunity in breast cancer: from TILs to the clinic. Nat. Rev. Clin. Oncol. 13, 228–241 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Salgado, R. et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann. Oncol. 26, 259–271 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Loi, S. et al. Tumor-infiltrating lymphocytes and prognosis: a pooled individual patient analysis of early-stage triple-negative breast cancers. J. Clin. Oncol. 37, 559–569 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Loi, S. et al. Relationship between tumor infiltrating lymphocyte (TIL) levels and response to pembrolizumab (pembro) in metastatic triple-negative breast cancer (mTNBC): results from KEYNOTE-086 [abstract LBA13]. Ann. Oncol. 28 (Suppl. 5), mdx440.005 (2017).

    Google Scholar 

  5. Loi, S. et al. Pembrolizumab plus trastuzumab in trastuzumab-resistant, advanced, HER2-positive breast cancer (PANACEA): a single-arm, multicentre, phase 1b-2 trial. Lancet Oncol. 20, 371–382 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Wein, L., Luen, S. J., Savas, P., Salgado, R. & Loi, S. Checkpoint blockade in the treatment of breast cancer: current status and future directions. Br. J. Cancer 119, 4–11 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cardoso, F. et al. Early breast cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 30, 1194–1220 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Ali, H. R. et al. Association between CD8+ T-cell infiltration and breast cancer survival in 12,439 patients. Ann. Oncol. 25, 1536–1543 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. 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 

  10. Masopust, D. & Soerens, A. G. Tissue-resident T cells and other resident leukocytes. Annu. Rev. Immunol. 37, 521–546 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yau, C. et al. Expression-based immune signatures as predictors of neoadjuvant targeted-/chemo-therapy response: Experience from the I-SPY 2 TRIAL of ~1000 patients across 10 therapies [abstract]. Cancer Res. 79 (Suppl. 4), P3-10-06 (2019).

    Google Scholar 

  12. Adams, S. et al. Pembrolizumab monotherapy for previously untreated, PD-L1-positive, metastatic triple-negative breast cancer: cohort B of the phase II KEYNOTE-086 study. Ann. Oncol. 30, 405–411 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Adams, S. et al. Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: cohort A of the phase II KEYNOTE-086 study. Ann. Oncol. 30, 397–404 (2019).

    Article  CAS  PubMed  Google Scholar 

  14. Loi, S. et al. RNA molecular signatures as predictive biomarkers of response to monotherapy pembrolizumab in patients with metastatic triple-negative breast cancer: KEYNOTE-086 [abstract]. Cancer Res. 79 (Suppl. 13), LB-225 (2019).

    Google Scholar 

  15. Wang, Z. Q. et al. CD103 and intratumoral immune response in breast cancer. Clin. Cancer Res. 22, 6290–6297 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. 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 

  17. Murray, T. et al. Very late antigen-1 marks functional tumor-resident CD8 T cells and correlates with survival of melanoma patients. Front. Immunol. 7, 573 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Boddupalli, C. S. et al. Interlesional diversity of T cell receptors in melanoma with immune checkpoints enriched in tissue-resident memory T cells. JCI Insight 1, e88955 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Djenidi, F. et al. CD8+CD103+ tumor-infiltrating lymphocytes are tumor-specific tissue-resident memory T cells and a prognostic factor for survival in lung cancer patients. J. Immunol. 194, 3475–3486 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. Ganesan, A. P. et al. Tissue-resident memory features are linked to the magnitude of cytotoxic T cell responses in human lung cancer. Nat. Immunol. 18, 940–950 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Koh, J. et al. Prognostic implications of intratumoral CD103+ tumor-infiltrating lymphocytes in pulmonary squamous cell carcinoma. Oncotarget 8, 13762–13769 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. 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 

  23. Komdeur, F. L. et al. CD103+ intraepithelial T cells in high-grade serous ovarian cancer are phenotypically diverse TCRαβ+ CD8αβ+ T cells that can be targeted for cancer immunotherapy. Oncotarget 7, 75130–75144 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Webb, J. R., Milne, K. & Nelson, B. H. PD-1 and CD103 are widely coexpressed on prognostically favorable intraepithelial CD8 T cells in human ovarian cancer. Cancer Immunol. Res. 3, 926–935 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Webb, J. R., Milne, K., Watson, P., Deleeuw, R. J. & Nelson, B. H. Tumor-infiltrating lymphocytes expressing the tissue resident memory marker CD103 are associated with increased survival in high-grade serous ovarian cancer. Clin. Cancer Res. 20, 434–444 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Duhen, T. et al. Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat. Commun. 9, 2724 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Komdeur, F. L. et al. CD103+ tumor-infiltrating lymphocytes are tumor-reactive intraepithelial CD8+ T cells associated with prognostic benefit and therapy response in cervical cancer. Oncoimmunology 6, e1338230 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Workel, H. H. et al. CD103 defines intraepithelial CD8+ PD1+ tumour-infiltrating lymphocytes of prognostic significance in endometrial adenocarcinoma. Eur. J. Cancer 60, 1–11 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Ling, K. L. et al. Modulation of CD103 expression on human colon carcinoma-specific CTL. J. Immunol. 178, 2908–2915 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Wang, B. et al. CD103+ tumor infiltrating lymphocytes predict a favorable prognosis in urothelial cell carcinoma of the bladder. J. Urol. 194, 556–562 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Lohneis, P. et al. Cytotoxic tumour-infiltrating T lymphocytes influence outcome in resected pancreatic ductal adenocarcinoma. Eur. J. Cancer 83, 290–301 (2017).

    Article  CAS  PubMed  Google Scholar 

  32. Gebhardt, T. et al. Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat. Immunol. 10, 524–530 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Masopust, D. et al. Dynamic T cell migration program provides resident memory within intestinal epithelium. J. Exp. Med. 207, 553–564 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cyster, J. G. & Schwab, S. R. Sphingosine-1-phosphate and lymphocyte egress from lymphoid organs. Annu. Rev. Immunol. 30, 69–94 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Schon, M. P. et al. Mucosal T lymphocyte numbers are selectively reduced in integrin alpha E (CD103)-deficient mice. J. Immunol. 162, 6641–6649 (1999).

    CAS  PubMed  Google Scholar 

  36. Gorfu, G., Rivera-Nieves, J. & Ley, K. Role of β7 integrins in intestinal lymphocyte homing and retention. Curr. Mol. Med. 9, 836–850 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Allakhverdi, Z. et al. Expression of CD103 identifies human regulatory T-cell subsets. J. Allergy Clin. Immunol. 118, 1342–1349 (2006).

    Article  CAS  PubMed  Google Scholar 

  38. Shiow, L. R. et al. CD69 acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature 440, 540–544 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Steinert, E. M. et al. Quantifying memory CD8 T cells reveals regionalization of immunosurveillance. Cell 161, 737–749 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Fernandez-Ruiz, D. et al. Liver-resident memory CD8+ T cells form a front-line defense against malaria liver-stage infection. Immunity 45, 889–902 (2016).

    Article  CAS  PubMed  Google Scholar 

  41. Park, S. L., Gebhardt, T. & Mackay, L. K. Tissue-resident memory T cells in cancer immunosurveillance. Trends Immunol. 40, 735–747 (2019).

    Article  CAS  PubMed  Google Scholar 

  42. Wherry, E. J. & Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 15, 486–499 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Blank, C. U. et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol. 19, 665–674 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kallies, A., Zehn, D. & Utzschneider, D. T. Precursor exhausted T cells: key to successful immunotherapy? Nat. Rev. Immunol. 20, 128–136 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Beura, L. K. et al. Intravital mucosal imaging of CD8(+) resident memory T cells shows tissue-autonomous recall responses that amplify secondary memory. Nat. Immunol. 19, 173–182 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Park, S. L. et al. Local proliferation maintains a stable pool of tissue-resident memory T cells after antiviral recall responses. Nat. Immunol. 19, 183–191 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Clarke, J. et al. Single-cell transcriptomic analysis of tissue-resident memory T cells in human lung cancer. J. Exp. Med. 216, 2128–2149 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Simoni, Y. et al. Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 557, 575–579 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. 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 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Schenkel, J. M. et al. IL-15-independent maintenance of tissue-resident and boosted effector memory CD8 T cells. J. Immunol. 196, 3920–3926 (2016).

    Article  CAS  PubMed  Google Scholar 

  51. Boutet, M. et al. TGFβ signaling intersects with CD103 integrin signaling to promote T-lymphocyte accumulation and antitumor activity in the lung tumor microenvironment. Cancer Res. 76, 1757–1769 (2016).

    Article  CAS  PubMed  Google Scholar 

  52. Mackay, L. K. et al. T-box transcription factors combine with the cytokines TGF-β and IL-15 to control tissue-resident memory T cell fate. Immunity 43, 1101–1111 (2015).

    Article  CAS  PubMed  Google Scholar 

  53. El-Asady, R. et al. TGF-β-dependent CD103 expression by CD8(+) T cells promotes selective destruction of the host intestinal epithelium during graft-versus-host disease. J. Exp. Med. 201, 1647–1657 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Mami-Chouaib, F. et al. Resident memory T cells, critical components in tumor immunology. J. Immunother. Cancer 6, 87 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Goldberg, J. E. & Schwertfeger, K. L. Proinflammatory cytokines in breast cancer: mechanisms of action and potential targets for therapeutics. Curr. Drug Targets 11, 1133–1146 (2010).

    Article  CAS  PubMed  Google Scholar 

  56. Leek, R. D. et al. Association of tumour necrosis factor alpha and its receptors with thymidine phosphorylase expression in invasive breast carcinoma. Br. J. Cancer 77, 2246–2251 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Le Bourgeois, T. et al. Targeting T cell metabolism for improvement of cancer immunotherapy. Front. Oncol. 8, 237 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Zhang, Y. et al. Enhancing CD8(+) T cell fatty acid catabolism within a metabolically challenging tumor microenvironment increases the efficacy of melanoma immunotherapy. Cancer Cell 32, 377–391.e9 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Xu, Y. et al. Glycolysis determines dichotomous regulation of T cell subsets in hypoxia. J. Clin. Invest. 126, 2678–2688 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Pan, Y. et al. Survival of tissue-resident memory T cells requires exogenous lipid uptake and metabolism. Nature 543, 252–256 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Pan, Y. & Kupper, T. S. Metabolic reprogramming and longevity of tissue-resident memory T cells. Front. Immunol. 9, 1347 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. 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  CAS  Google Scholar 

  63. 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 

  64. Park, J. H. et al. Prognostic value of tumor-infiltrating lymphocytes in patients with early-stage triple-negative breast cancers (TNBC) who did not receive adjuvant chemotherapy. Ann. Oncol. 30, 1941–1949 (2019).

    Article  CAS  PubMed  Google Scholar 

  65. Auslander, N. et al. Robust prediction of response to immune checkpoint blockade therapy in metastatic melanoma. Nat. Med. 24, 1545–1549 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Schmid, P. et al. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N. Engl. J. Med. 379, 2108–2121 (2018).

    Article  CAS  PubMed  Google Scholar 

  67. Schmid, P. et al. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 21, 44–59 (2020).

    Article  CAS  PubMed  Google Scholar 

  68. 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 

  69. Borsellino, G. et al. Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: hydrolysis of extracellular ATP and immune suppression. Blood 110, 1225–1232 (2007).

    Article  CAS  PubMed  Google Scholar 

  70. Tang, L. et al. Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery. Nat. Biotechnol. 36, 707–716 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Smith, T. T. et al. In situ programming of leukaemia-specific T cells using synthetic DNA nanocarriers. Nat. Nanotechnol. 12, 813–820 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Hartana, C. A. et al. Tissue-resident memory T cells are epigenetically cytotoxic with signs of exhaustion in human urinary bladder cancer. Clin. Exp. Immunol. 194, 39–53 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Salerno, E. P., Olson, W. C., McSkimming, C., Shea, S. & Slingluff, C. L. Jr. T cells in the human metastatic melanoma microenvironment express site-specific homing receptors and retention integrins. Int. J. Cancer 134, 563–574 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Webb, J. R. et al. Profound elevation of CD8+ T cells expressing the intraepithelial lymphocyte marker CD103 (αE/β7 integrin) in high-grade serous ovarian cancer. Gynecol. Oncol. 118, 228–236 (2010).

    Article  CAS  PubMed  Google Scholar 

  75. Quinn, E., Hawkins, N., Yip, Y. L., Suter, C. & Ward, R. CD103+ intraepithelial lymphocytes–a unique population in microsatellite unstable sporadic colorectal cancer. Eur. J. Cancer 39, 469–475 (2003).

    Article  CAS  PubMed  Google Scholar 

  76. Zhang, L. et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 564, 268–272 (2018).

    Article  CAS  PubMed  Google Scholar 

  77. Hu, W., Sun, R., Chen, L., Zheng, X. & Jiang, J. Prognostic significance of resident CD103+CD8+T cells in human colorectal cancer tissues. Acta Histochem. 121, 657–663 (2019).

    Article  CAS  PubMed  Google Scholar 

  78. Li, B. et al. The landscape of antigen-specific T cells in human cancers. bioRxiv https://doi.org/10.1101/459842 (2018).

    Article  Google Scholar 

Download references

Acknowledgements

The work of S.L. is supported by the National Breast Cancer Foundation of Australia and the Breast Cancer Research Foundation (New York, NY, USA). The work of P.A.B. is supported by a National Breast Cancer Foundation of Australia Fellowship (ECF-17-005).

Author information

Authors and Affiliations

Authors

Contributions

A.B., P.S., S.S., R.L. and B.V. researched data for the article. A.B., L.K.M., P.J.N. and S.L. made a substantial contribution to discussions of the content. A.B., P.S., S.S., R.L., B.V. and S.L. wrote the manuscript. A.B., S.J.L., P.A.B., L.K.M., P.J.N. and S.L. reviewed and/or edited the manuscript prior to submission.

Corresponding author

Correspondence to Sherene Loi.

Ethics declarations

Competing interests

S.L.’s institution receives research funding from Bristol-Myers Squibb, Eli Lilly, Genentech, Merck, Novartis, Pfizer, Puma Biotechnology and Roche. S.L. has acted as a non-compensated consultant of AstraZeneca, Bristol-Meyers Squibb, Merck, Novartis, Pfizer, Roche-Genentech and Seattle Genetics. P.J.N. receives research funding from Advaxis, Allergan, Bristol-Myers Squibb, Compugen, Juno-Celgene and Roche-Genentech. The other authors declare no competing interests.

Additional information

Publisher’s note

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

RELATED LINKS

WHO classification of tumours online: https://tumourclassification.iarc.who.int/welcome/

Glossary

Haemotoxylin and eosin (H&E) staining

A commonly used histopathological staining technique that enables the visualization of the cellular features of a clinical specimen.

Biomarker

A naturally occurring feature of a tumour that can be measured and has prognostic and/or predictive value.

Single-cell RNA sequencing

High-resolution sequencing of the RNA transcripts of individual cells using optimized next-generation sequencing technologies that confer a better understanding of cellular function.

Gene signature

A group of genes expressed by a cell with a defined, unique and characteristic pattern of expression that reflects a specific genotype and/or phenotype.

Multiplex immunohistochemistry

Labelling of protein structures on histological slides using antibodies conjugated to different fluorescent reporters that are able to render the multiple features visible.

Flow cytometry

Analysis of single cells using a variety of cell-surface and intracellular fluorescent markers.

CyTOF

High-throughput analysis of single cells using heavy metal tags followed by mass spectrometry.

Cytokines

Small proteins secreted by immune cells that result in the paracrine stimulation of other cells.

Chemokines

A family of cytokines that act as chemoattractants and promote the migration of responsive cells towards the site of an immune response.

Immune checkpoint proteins

Protein markers expressed on the surface of immune cells that regulate their effector function and proliferation.

Adoptive cell therapy

Transfer of immune cells, usually the patient’s own T cells following ex vivo modification and/or expansion, into a patient for therapeutic purposes.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Byrne, A., Savas, P., Sant, S. et al. Tissue-resident memory T cells in breast cancer control and immunotherapy responses. Nat Rev Clin Oncol 17, 341–348 (2020). https://doi.org/10.1038/s41571-020-0333-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41571-020-0333-y

This article is cited by

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