Abstract
Tissue-resident memory (TRM) T cells are distinct population of non-circulating lymphocytes that play an important role in mediating regional immunity. TRM- like cells have now been identified as a component of tumor-infiltrating lymphocytes in several human tumors and correlate with outcome and response to immunotherapy. TRM cells have also been shown to mediate anti-tumor immunity in murine models. Biology of TRM cells has several implications for clinical cancer immunotherapy. Here we discuss newer insights into the biology of TRM T cells and discuss their implications for understanding the heterogeneity of immune microenvironment in tumors as well as improving the efficacy of cancer vaccines, immune-checkpoint blockade and adoptive cellular therapies in the clinic.
Immune system consists of both innate as well as adaptive components with potent capacity to control tumors. Success of therapies that block inhibitory immune checkpoints on T cells, as well as therapies that redirect T cells (such as via chimeric-antigen receptors (CAR), or bispecific antibodies), have transformed clinical oncology and placed T cells at the center of immune-mediated approaches to battle cancer1. An important aspect of T cell immunity is the property of immunologic memory, which is essential for long term protection2. Tissue-resident memory (TRM) T cells are a phenotypically and transcriptionally distinct population of non-recirculating memory T cells that reside within non-lymphoid tissue and provide regional protection against pathogens and tumors in several models3,4. In this review, we discuss emerging insights about the biology of these cells in the setting of cancer with a particular focus on human TRM cells and the implications of recent studies on TRMs for harnessing tumor immunity in the clinic.
Lessons from Mice:
TRM cells have been classically identified as non-recirculating T cells that reside and renew within tissues and do not equilibrate with circulating T cell pool. In mice, TRM compartment has been studied using various approaches including parabiosis5,6, transplantation7,8, intravascular labeling9,10 and targeted antibody-mediated T cell depletion4. While the majority of studies have focused on CD8+ TRM cells, the presence of CD4+ TRM cells has also been characterized in both humans11,12 and mice13. In contrast to circulating T cells (including effector memory and central memory T cells) that may transit through tissue but return to blood or secondary lymphoid organs, TRM T cells do not circulate through these compartments4. CD8+ TRM cells in several epithelial tissues express CD69 and CD103. CD69 is expressed early during TRM development and serves to maintain tissue retention via downregulating egress signals such as S1PR1(sphingosine 1-phosphate receptor-1)14. CD103 is regulated by cytokine signals such as TGF-beta and helps maintain interactions with epithelial markers such as E-cadherin15. TRM cells lacking CD103 can however be detected in some tissues and phenotypic identification of these cells based on limited markers can therefore be challenging. Transcriptome signatures that distinguish TRM cells from other counterparts have been published, but many of the genes in these signatures are not specific to TRM cells and are also expressed in T cell subsets16–19. It is increasingly apparent particularly with new single cell tools that TRM cells exhibit functional and phenotypic heterogeneity, which may depend in part on the specific tissue studied. CD8+ TRM cells develop early after activation from effector-like precursors wherein local tissue microenvironment drives resident fate. In some settings, cytokines such as IL15 and TGF-beta are important for TRM generation20. Expression of certain transcription factors such as Hobit16, Runx321 and Nur-77 / NR4A118 has been shown to be important for generation of TRM cells. Antigen recognition within tissues was shown to be important for TRM cells in neural tissue and models of HSV-1 infection in the lung, but less relevant in other settings22,23. In the steady state, TRM cells in many tissues can be maintained without ongoing replenishment from circulating precursors, although this may be required for TRM cells in murine lung8,24. In mouse skin and female reproductive tract, TRM cells are able to proliferate locally upon activation, without being displaced by other cell types, but can also leave tissues to contribute to systemic immunity25–27. It is increasingly appreciated that TRM cells are indeed capable of contributing to the circulating memory pool and thereby providing an “outside-in” mechanism for protective immunity28. TRM cells may also be capable of retrograde migration to lymph nodes where they may contribute to long lived immune cells29. These considerations are leading to proposals for new models for integrating TRM cells into traditional T cell differentiation models, understanding the plasticity and interconversion between these compartments30,31. It is notable that even in the studies wherein ex-TRM cells reach lymphoid tissues or systemic circulation, they seem to maintain a stable epigenome and seem poised to preferentially home back to tissue of origin28,29. Small proportions of T cells with TRM signatures and phenotypes have been detected among circulating T cells in humans18,32. A small subset of TRM cells with dye efflux properties seem to be a quiescent subset of T cells in both humans and mice18,33, with “label retention” properties similar to tissue stem cells, which may allow them to persist long term. Until recently, most of the data relating to protective effects of TRM cells in mice has been limited to pathogens8,22. In several studies, TRM cells provide superior protection against viral or bacterial pathogens in peripheral tissues6,8. The underlying mechanisms involve direct effects as well as recruitment of other immune cells, although TRM cells are able to mediate protection without the requirement of circulating effectors27,34. As discussed below, TRM cells are now also emerging as important players in mediating tumor immunity35.
Evidence for human TRM T cells
As experimental parabiosis is not feasible in the human setting, evidence for equivalent tissue-localized T cells in humans has been based on finding T cells that persist in tissues following organ transplantation or antibody-mediated depletion of circulating T cells. HLA-mismatched TRM cells can persist long term in patients following lung and bowel transplants36–38. Administration of anti-CD52 antibody (alemtuzumab) depletes circulating T cells without concurrent depletion of skin-resident T cells, illustrating their disequilibrium with circulating compartment39,40. T cells with TRM phenotype have been shown to be enriched in barrier tissues studied over several decades of life41. In humans, as in mice, TRM cells have been characterized by the expression of markers such as CD69, CD103 (in some tissues), and downregulation of tissue egress or migration molecules such as S1PR1, KLF2, and L-selection4. The phenotype is often tissue-specific and also heterogenous within tissues, leading to functional differences with some subsets such as CD49a+ TRM cells shown to express more interferon-γ42,43. Transcriptional profiles of human TRM cells seem to resemble murine counterparts, but there are some differences. For example, Hobit is commonly expressed in murine TRM cells wherein it plays an important functional role16, but its expression in human TRM cells has been harder to detect11.
Preclinical studies for anti-tumor potential of TRM cells
Potential role for TRM cells in tumor immunity first emerged from studies showing TRM phenotype of T cells infiltrating human tumors44,45, but their anti-tumor properties and role in immune surveillance has now been demonstrated in the context of murine models as well46,47. Mice lacking molecules expressed on TRM cells such as CD69 or CD103 are more susceptible to transplantable melanoma than wild-type mice46,47. TRM cells generated by vaccination mediate protection against tumor challenge48–50. In a melanoma model, TRM cells generated during tumorigenesis and prior to tumor challenge were able to inhibit tumor growth, supporting a role for TRM cells in surveillance and equilibrium phase of cancer control by maintaining occult melanoma in check47.
Mechanisms of anti-tumor effects of TRM cells
The mechanisms by which TRM cells mediate anti-tumor effects remain to be fully clarified and may differ in specific settings and tumor type. CD103 expressed on some TRM cells may promote immunologic synapse by binding to E-cadherin on tumor cells51,52. A major mechanism of TRM-mediated control may be via non-cytotoxic mechanisms and mediated in part by cytokines such as TNF47. In this regard, TRM cells may act more as controllers than as killers, similar to their role in control of latent / persistent viruses such as HSV1, where TRM-mediated immunity does not lead to complete virus eradication but can maintain viral control for the life of the host. TRM cells may also engage other immune cells and enhance tumor immunity via antigen spreading by activating DCs53. CD8+ TRM-like cells may also mediate or amplify tumor immunity both by secreting cytokines (e.g. interferon-γ) or chemokines (e.g. CCL3, XCL1) to enhance recruitment of monocytes, NK cells and XCR1+ cDC1 to tumor site54.
Generation and maintenance of TRM T cells within tumors:
Information about the generation of intra-tumoral TRM-like T cells is more limited compared to that for virus-specific T cells. Subsets of intra-tumoral T cells in human tumors express Hobit, but its role in biology of human TRM-like cells is not clear. Lineage tracing and single cell transcriptomic studies have identified subsets of circulating effector cells as precursors with greater potential for TRM generation in tumor models55,56. TGFβ is abundant in many tumors and could regulate TRM generation, consistent with its role in non-tumor setting. Studies have also suggested a role for other pathways such as IL15 in intestinal cancer57 or Notch signaling in head/neck cancer58. Other mechanisms that may be operative include TRM associated metabolic pathways such as exogenous fatty acid uptake59 in tumors such as melanoma and breast cancer known to be enriched for free fatty acids, and hypoxic environments common in several tumors. It has also been suggested that local activation by distinct subsets of dendritic cells, such as the newly described cDC3 cells may play an important role in stimulation of TRM cells60. Taken together, these data, derived mostly from mouse models, suggest that diverse tumor and tissue-dependent pathways may be involved in the generation and maintenance of TRM-like T cells within tumors.
TRM-like T cells in human tumors
It is now well recognized that tumors are commonly infiltrated by heterogeneous populations of lymphocytes that include both innate and adaptive immune cells. Several studies have now demonstrated that a proportion of tumor-infiltrating lymphocytes (TILs) in human tumors express surface markers (e.g. CD69) or molecular signatures consistent with tissue resident memory T cells44,45,58,61,62. CD69 expression can in principle also be induced by cognate antigen stimulation, although the genomic signatures of TRM cells in tumor tissues, when studied, is different from that in recently activated T cells. In this regard, at least some of the TRM cells within tumor tissues may be similar to those in the setting of persistent viruses wherein the local antigenic stimulus is persistent. Whether the expression of TRM markers by human T cells in tumor tissue is analogous to those in non-malignant tissue is an area of active investigation. Although tissue residence of human tumor-infiltrating TRM cells can not be directly tested by classical methods such as parabiosis, studies in patients with metastatic cancer have shown that dominant T cell receptors of TRM cells in individual metastatic lesions show very little overlap in spite of the fact that all metastatic lesions share a systemic circulation as with parabiotic mice45. Nonetheless, due to these experimental limitations, we have referred to these cells as TRM-like TILs (TRM TILs).
While detection of TRM-like TILs cells based on single markers such as CD69 is challenging, we suggest that concurrent evaluation of tissue-retention signatures (such as downregulation of S1PR1) provides more robust evaluation of these cells. It is notable, that while CD103 is commonly utilized to detect TRM cells in epithelial tumors, the expression of CD103 is tissue-dependent, and lacking for examples in TRM cells in the bone marrow.
TRM TILs have now been described in several human tumors including non-small cell lung cancer44,58, breast63,64, colorectal65, melanoma45,66, genitourinary67, gynecologic cancers68, glioma61 and myeloma62 and constitute nearly 25–75% of all TILs45,69. The presence of TILs with TRM phenotype may not be restricted to solid tumors, and T cells with such phenotype can also be detected within tumor tissue in hematologic tumors such as myeloma and its precursor states62,70,71. Infiltration of tumors by TILs has been associated with improved survival in several tumor types72. However recent data suggest that the TRM component of the TILs may be particularly relevant for predicting outcome58,66. TRM TILs have also been shown to exhibit strong effector and cytolytic function compared to non-TRM TILs44. As an example, CD39+ TRM-TILs were shown to be enriched for tumor reactivity in several tumor types73. It is important to note that while tumor-antigen-specific T cells have been claimed to be enriched within subsets of TRM cells, not all of the TRM cells within tumor tissue are tumor-specific. Both self-reactive as well as pathogen-reactive TRM cells have been detected within tumors74,75. Interestingly, studies have also suggested that activation of these “by-stander” T cells may mediate anti-tumor activity and therefore could in principle be repurposed for tumor therapy75. Together, these studies have not only documented the presence of TRM- like TILs in several tumors, but their strong correlation with clinical outcome also suggest that these are important components of protective tumor immunity. Recent application of single cell tools has characterized the heterogeneity of tumor-infiltrating lymphocytes including TRM cells, that may impact their biology62,76. Below, we discuss some aspects of TRM biology that may impact clinical application of immunotherapy.
Non-uniform regional immunity and inter-lesional heterogeneity
It is increasingly appreciated that degree of T cell infiltration within tumors is associated with improved outcome, occasionally in the absence of detectable circulating tumor-specific T cells, which points to the importance of regional immunity in cancer. However, the degree to which this regional immunity remains localized to the tumor lesion or whether it has considerable systemic component has broad implications not just for measuring and harnessing immune responses in the clinic but also cancer biology in general. Dominant T cell receptors infiltrating individual metastatic lesions can differ considerably45,77,78. In a recent study, it was shown that this inter-lesional heterogeneity in dominant T cell receptors (TCRs) is predominantly derived from the subset of T cells that express markers of TRM cells45. Importantly, this heterogeneity of T cell response exceeds the differences in mutations or predicted neoantigens in these lesions. As all metastatic lesions in any individual patient share the same systemic circulation, the concept that the T cells do not “equilibrate” in vivo in patients in spite of antigenic similarity between individual metastatic lesions is highly reminiscent of regional nature of immune responses due to TRM cells in parabiotic mice. This concept in turn suggests that it is important to pay attention to the specific lesion biopsied in order to track changes in immune response following immune therapies. Inter-lesional differences in immune response may also impact genomic evolution of tumors at individual sites, as it may impact immune pressure that each lesion experiences, particularly in view of emerging role of TRM cells in the equilibrium phase of immune editing47,77,79. This heterogeneity may also provide a potential explanation for several observations following immune therapies in the clinic. For example, it is well known that some patients achieve “mixed responses” following checkpoint blockade, with varying degree of regression in individual lesions80. Along the same lines, regional nature of immune response may also provide an explanation for the observations in the clinic that disease progression at a single site may not equate to global loss of systemic tumor immunity and some of these patients have achieved durable responses following local control of the progressing lesion81,82. It should be noted that while TRM cells do not recirculate in steady state, it is possible that they are capable of in situ proliferation following activation and this may then primes them towards exiting tumor tissue and contribute to systemic immunity as has been shown in mouse models25. Whether such biology occurs in clinically relevant settings needs to be studied.
TRM cells as targets to improve cancer vaccines
The field of cancer vaccines is replete with examples wherein vaccine-induced systemic or circulating T cell responses do not correlate with protective immunity or tumor regressions in the clinic. Based on the considerations discussed earlier, it is tempting to speculate that an important missing link may be the inability of current vaccines to effectively enhance TRM cells within tumors. Mucosal vaccination protocols that lead to greater generation of TRM cells mediate greater protection than systemic vaccines against head and neck cancer or melanoma in murine models48,83,84. Vaccine-induced TRM cells have been shown to mediate anti-tumor effects in mice48. At present, the degree to which current vaccines effectively induce tumor-infiltrating TRM cells and how to optimize it requires further study. Regional administration of vaccine constructs or immune stimulatory agents to mediate in situ vaccination may be one way to achieve these goals and is undergoing evaluation.
Repeated boosting with heterologous viruses carrying cognate antigen can enhance TRM cells within tissues and such approach could lead to enhanced tumor immunity85. Another strategy that has been attempted involves a push and pull approach that combines initial priming to generate TRM precursors and then pulling them into tumors via chemokine gradients86. The nature of tumor-infiltrating TRM cells induced by vaccines do not have to be tumor-specific in order to mediate tumor immunity. In a preclinical study, peptide injection targeting virus-specific TRM cells led to effective tumor regression, particularly in combination with checkpoint blockade75. While the importance of dendritic cells in generation of T cell responses in well appreciated, the nature of specific antigen-presenting cells or DC subsets that need to be targeted to induce TRM T cells in situ needs greater clarity. In one study, engagement of CD1c+ DCs was shown to lead to induction of skin-homing TRM cells87. Induction of immune response following activation of TRM cells may depend on engagement of Batf3-expressing CD141+ subset of DCs that are specialized for cross-presentation88. Strategies to enhance targeting of DCs via DC-specific antibodies or nanoparticles are under active evaluation but will need to consider their capacity to enhance TRM T cells within tumors89–91.
TRM cells as targets of immune checkpoint blockade
Blockade of immune checkpoints such as PD-1 has transformed immune therapy of human cancer. However, these checkpoints are expressed on only a subset of T cells within human tumors. Several studies have shown that T cells with TRM phenotype and genomic signatures are the subset of tumor-infiltrating T cells with highest level of expression of inhibitory immune checkpoints within human tumors45,61, raising the possibility that TRM cells are an important target of immune checkpoint blockade, particularly those targeting PD-1. Blockade of PD-1 in vitro can enhance cytokine production by these T cells, supporting the possibility that these immune checkpoints serve to provide a restraint on TRM function in situ92. Tumor immunity in melanoma can be associated with the development of vitiligo in some patients, which in turn is now recognized as associated with activation of skin-resident TRM T cells42,93. Presence of T cells with TRM phenotype or TRM signatures within human tumors has been correlated with improved survival following IC blockade58,66. At present, it remains unclear whether the TRM compartment within tumors undergoes proliferation within tumors situ to mediate anti-tumor effects, or whether recruitment of new T cells into tumors is needed to mediate effects of IC blockade (or combination thereof) and findings supportive of both possibilities have been described in preclinical models94–96. Recent studies have also identified a subset of TCF1+ T cells termed as precursor exhausted or stem-like memory T cells that undergo proliferative boost following PD-1 blockade in mouse models of chronic LCMV infection97. Such T cells have also been identified in human tumors, and in some small cohorts linked to outcome following checkpoint blockade as well98. While TCF1+ cells within tumors do commonly express CD69 (a marker of TRM cells), they are distinct from CD103+ TRM cells which do not express TCF1. In preclinical models TCF1 was shown to inhibit CD103 and tissue residence99. In a recent study of T cells infiltrating pediatric gliomas, TCF1+ T cells were found to be mostly perivascular in location, while CD103+ T cells were further away from the vessels and infiltrating the tumor tissue.100 Further studies are needed to better understand the functional and spatial heterogeneity within the TRM-like compartments and their role/contributions to effects of IC blockade.
TRM cells as targets of adoptive cell therapies
The ability of adoptively transferred T cells to infiltrate into tumors and persist long term have emerged as essential characteristics for effective therapy. These features are also imprinted into the biology of TRM cells. Adoptive transfer of tumor infiltrating lymphocytes, which contain a variable but high proportion of TRM cells, has been shown to mediate tumor regression in patients with melanoma and renal cell cancer101. Indeed, among TILs, tumor-specific T cells were shown to express higher levels of PD-1, which is also a characteristic of TRM-like TILs102. Activated T cells infiltrating murine tumors rapidly acquire large component of transcriptional program of TRM cells103,104. T cells with Hoechst dye exclusion phenotype, which also exhibit TRM-like signatures, lead to greater tissue infiltration upon adoptive transfer18. Overexpression of residency promoting transcription factor Runx3 in T cells led to greater tissue infiltration of adoptive transferred T cells and enhanced anti-tumor effects of adoptive cell therapy21. Enhancing tumor or tissue infiltration may be particularly important in the setting of chimeric-antigen receptor (CAR)-T cell therapy for solid tumors, which have this far been relatively unresponsive to such therapies105. Several strategies are being considered to enhance the effectiveness of adoptive cell therapies by engaging TRM biology. This includes genetically reprogramming T cells to enhance TRM signatures, or preselecting T cell substrates with greater potential for differentiation to TRM cells, either isolating precursor T cells from tissues themselves or mobilizing T cells from tissues into circulation. One example of the latter strategy is the use of CXCR4/SDF1 antagonists to mobilize T cells from the bone marrow, which is analogous to stem cell mobilization currently used in stem cell transplantation18. Further studies are needed to better understand whether enhancing TRM-like features in adoptively transferred T cells will lead to improved efficacy, particularly against solid tumors.
Summary:
Cells with phenotype and genomic signatures of tissue-resident memory T cells have been identified in several human tumors, wherein they correlate with outcome and response to immune therapy35,69. Anti-tumor effects of these T cells have also been demonstrated in preclinical models. TRM biology has profound implications for how to optimally engage and enhance tumor immunity in the clinic. However, several questions relating to their generation, heterogeneity, tissue residence, maintenance and anti-tumor effects remain to be fully characterized. Further studies should help to clarify the biology of these T cells, particularly in human tumors, and may lead to novel approaches for cancer immunotherapy.
Acknowledgements:
KMD is supported in part by funds from the NIH (RO1CA238471and RO1AR077926) and SCOR award from the Leukemia and Lymphoma Society. MVD is supported in part by funds from the NIH (R35CA197603) and SCOR award from the Leukemia and Lymphoma Society.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References:
- 1.Zou W, Wolchok JD & Chen L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci Transl Med 8, 328rv324 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Farber DL, Netea MG, Radbruch A, Rajewsky K. & Zinkernagel RM Immunological memory: lessons from the past and a look to the future. Nat Rev Immunol 16, 124–128 (2016). [DOI] [PubMed] [Google Scholar]
- 3.Mueller SN, Gebhardt T, Carbone FR & Heath WR Memory T cell subsets, migration patterns, and tissue residence. Annu Rev Immunol 31, 137–161 (2013). [DOI] [PubMed] [Google Scholar]
- 4.Masopust D. & Soerens AG Tissue-Resident T Cells and Other Resident Leukocytes. Annu Rev Immunol 37, 521–546 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Klonowski KD, et al. Dynamics of blood-borne CD8 memory T cell migration in vivo. Immunity 20, 551–562 (2004). [DOI] [PubMed] [Google Scholar]
- 6.Jiang X, et al. Skin infection generates non-migratory memory CD8+ T(RM) cells providing global skin immunity. Nature 483, 227–231 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Masopust D, et al. Dynamic T cell migration program provides resident memory within intestinal epithelium. J Exp Med 207, 553–564 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.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). [DOI] [PubMed] [Google Scholar]
- 9.Steinert EM, et al. Quantifying Memory CD8 T Cells Reveals Regionalization of Immunosurveillance. Cell 161, 737–749 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Anderson KG, et al. Cutting edge: intravascular staining redefines lung CD8 T cell responses. J Immunol 189, 2702–2706 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kumar BV, 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Oja AE, et al. Trigger-happy resident memory CD4(+) T cells inhabit the human lungs. Mucosal Immunol 11, 654–667 (2018). [DOI] [PubMed] [Google Scholar]
- 13.Iijima N. & Iwasaki A. T cell memory. A local macrophage chemokine network sustains protective tissue-resident memory CD4 T cells. Science 346, 93–98 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Mackay LK, et al. Cutting edge: CD69 interference with sphingosine-1-phosphate receptor function regulates peripheral T cell retention. J Immunol 194, 2059–2063 (2015). [DOI] [PubMed] [Google Scholar]
- 15.Mohammed J, et al. Stromal cells control the epithelial residence of DCs and memory T cells by regulated activation of TGF-beta. Nat Immunol 17, 414–421 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Mackay LK, et al. Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science 352, 459–463 (2016). [DOI] [PubMed] [Google Scholar]
- 17.Wakim LM, et al. The molecular signature of tissue resident memory CD8 T cells isolated from the brain. J Immunol 189, 3462–3471 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Boddupalli CS, et al. ABC transporters and NR4A1 identify a quiescent subset of tissue-resident memory T cells. J Clin Invest 126, 3905–3916 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Mackay LK, et al. The developmental pathway for CD103(+)CD8+ tissue-resident memory T cells of skin. Nat Immunol 14, 1294–1301 (2013). [DOI] [PubMed] [Google Scholar]
- 20.Mackay LK, et al. T-box Transcription Factors Combine with the Cytokines TGF-beta and IL-15 to Control Tissue-Resident Memory T Cell Fate. Immunity 43, 1101–1111 (2015). [DOI] [PubMed] [Google Scholar]
- 21.Milner JJ, et al. Runx3 programs CD8(+) T cell residency in non-lymphoid tissues and tumours. Nature 552, 253–257 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Mackay LK, et al. Long-lived epithelial immunity by tissue-resident memory T (TRM) cells in the absence of persisting local antigen presentation. Proc Natl Acad Sci U S A 109, 7037–7042 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Casey KA, et al. Antigen-independent differentiation and maintenance of effector-like resident memory T cells in tissues. J Immunol 188, 4866–4875 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Takamura S. & Kohlmeier JE Establishment and Maintenance of Conventional and Circulation-Driven Lung-Resident Memory CD8(+) T Cells Following Respiratory Virus Infections. Front Immunol 10, 733 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Beura LK, et al. T Cells in Nonlymphoid Tissues Give Rise to Lymph-Node-Resident Memory T Cells. Immunity 48, 327–338 e325 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Beura LK, 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Park SL, et al. Local proliferation maintains a stable pool of tissue-resident memory T cells after antiviral recall responses. Nat Immunol 19, 183–191 (2018). [DOI] [PubMed] [Google Scholar]
- 28.Fonseca R, et al. Developmental plasticity allows outside-in immune responses by resident memory T cells. Nat Immunol 21, 412–421 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Stolley JM, et al. Retrograde migration supplies resident memory T cells to lung-draining LN after influenza infection. J Exp Med 217(2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Rosato PC, Wijeyesinghe S, Stolley JM & Masopust D. Integrating resident memory into T cell differentiation models. Curr Opin Immunol 63, 35–42 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Carbone FR & Gebhardt T. Should I stay or should I go-Reconciling clashing perspectives on CD4(+) tissue-resident memory T cells. Sci Immunol 4(2019). [DOI] [PubMed] [Google Scholar]
- 32.Klicznik MM, et al. Human CD4(+)CD103(+) cutaneous resident memory T cells are found in the circulation of healthy individuals. Sci Immunol 4(2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kumar BV, et al. Functional heterogeneity of human tissue-resident memory T cells based on dye efflux capacities. JCI Insight 3(2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ariotti S, et al. T cell memory. Skin-resident memory CD8(+) T cells trigger a state of tissue-wide pathogen alert. Science 346, 101–105 (2014). [DOI] [PubMed] [Google Scholar]
- 35.Amsen D, van Gisbergen KPJM, Hombrink P. & van Lier RAW Tissue-resident memory T cells at the center of immunity to solid tumors. Nature Immunology 19, 538–546 (2018). [DOI] [PubMed] [Google Scholar]
- 36.Snyder ME, et al. Generation and persistence of human tissue-resident memory T cells in lung transplantation. Sci Immunol 4(2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zuber J, et al. Bidirectional intragraft alloreactivity drives the repopulation of human intestinal allografts and correlates with clinical outcome. Sci Immunol 1(2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bartolome-Casado R, et al. Resident memory CD8 T cells persist for years in human small intestine. J Exp Med 216, 2412–2426 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Clark RA, et al. Skin effector memory T cells do not recirculate and provide immune protection in alemtuzumab-treated CTCL patients. Sci Transl Med 4, 117ra117 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Watanabe R, et al. Human skin is protected by four functionally and phenotypically discrete populations of resident and recirculating memory T cells. Sci Transl Med 7, 279ra239 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Thome JJ, et al. Spatial map of human T cell compartmentalization and maintenance over decades of life. Cell 159, 814–828 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Cheuk S, et al. CD49a Expression Defines Tissue-Resident CD8(+) T Cells Poised for Cytotoxic Function in Human Skin. Immunity 46, 287–300 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hombrink P, et al. Programs for the persistence, vigilance and control of human CD8(+) lung-resident memory T cells. Nat Immunol 17, 1467–1478 (2016). [DOI] [PubMed] [Google Scholar]
- 44.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). [DOI] [PubMed] [Google Scholar]
- 45.Boddupalli CS, 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Malik BT, et al. Resident memory T cells in the skin mediate durable immunity to melanoma. Sci Immunol 2(2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Park SL, et al. Tissue-resident memory CD8(+) T cells promote melanoma-immune equilibrium in skin. Nature 565, 366–371 (2019). [DOI] [PubMed] [Google Scholar]
- 48.Nizard M, et al. Induction of resident memory T cells enhances the efficacy of cancer vaccine. Nat Commun 8, 15221 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Galvez-Cancino F, et al. Vaccination-induced skin-resident memory CD8(+) T cells mediate strong protection against cutaneous melanoma. Oncoimmunology 7, e1442163 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Enamorado M, et al. Enhanced anti-tumour immunity requires the interplay between resident and circulating memory CD8(+) T cells. Nat Commun 8, 16073 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Franciszkiewicz K, et al. CD103 or LFA-1 engagement at the immune synapse between cytotoxic T cells and tumor cells promotes maturation and regulates T-cell effector functions. Cancer Res 73, 617–628 (2013). [DOI] [PubMed] [Google Scholar]
- 52.Franciszkiewicz K, et al. Intratumoral induction of CD103 triggers tumor-specific CTL function and CCR5-dependent T-cell retention. Cancer Res 69, 6249–6255 (2009). [DOI] [PubMed] [Google Scholar]
- 53.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Brewitz A, et al. CD8(+) T Cells Orchestrate pDC-XCR1(+) Dendritic Cell Spatial and Functional Cooperativity to Optimize Priming. Immunity 46, 205–219 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Milner JJ, et al. Heterogenous Populations of Tissue-Resident CD8(+) T Cells Are Generated in Response to Infection and Malignancy. Immunity 52, 808–824 e807 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kok L, et al. A committed tissue-resident memory T cell precursor within the circulating CD8+ effector T cell pool. J Exp Med 217(2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Mlecnik B, et al. Functional network pipeline reveals genetic determinants associated with in situ lymphocyte proliferation and survival of cancer patients. Sci Transl Med 6, 228ra237 (2014). [DOI] [PubMed] [Google Scholar]
- 58.Ganesan AP, 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Pan Y, et al. Survival of tissue-resident memory T cells requires exogenous lipid uptake and metabolism. Nature 543, 252–256 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Bourdely P, et al. Transcriptional and Functional Analysis of CD1c(+) Human Dendritic Cells Identifies a CD163(+) Subset Priming CD8(+)CD103(+) T Cells. Immunity 53, 335–352 e338 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Vasquez JC, et al. SOX2 immunity and tissue resident memory in children and young adults with glioma. J Neurooncol 134, 41–53 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Bailur JK, et al. Early alterations in stem-like/resident T cells, innate and myeloid cells in the bone marrow in preneoplastic gammopathy. JCI Insight 5(2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Egelston CA, et al. Resident memory CD8+ T cells within cancer islands mediate survival in breast cancer patients. JCI Insight 4(2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wang ZQ, et al. CD103 and Intratumoral Immune Response in Breast Cancer. Clin Cancer Res 22, 6290–6297 (2016). [DOI] [PubMed] [Google Scholar]
- 65.de Vries NL, et al. High-dimensional cytometric analysis of colorectal cancer reveals novel mediators of antitumour immunity. Gut 69, 691–703 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.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). [DOI] [PubMed] [Google Scholar]
- 67.Hartana CA, 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Webb JR, Milne K, Watson P, Deleeuw RJ & Nelson BH 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). [DOI] [PubMed] [Google Scholar]
- 69.Dhodapkar KM Role of Tissue-Resident Memory in Intra-Tumor Heterogeneity and Response to Immune Checkpoint Blockade. Front Immunol 9, 1655 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Bar N, et al. Differential effects of PD-L1 versus PD-1 blockade on myeloid inflammation in human cancer. JCI Insight (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Dhodapkar MV & Dhodapkar KM Moving Immunoprevention Beyond Virally Mediated Malignancies: Do We Need to Link It to Early Detection? Front Immunol 10, 2385 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Bindea G, Mlecnik B, Fridman WH, Pages F. & Galon J. Natural immunity to cancer in humans. Curr Opin Immunol 22, 215–222 (2010). [DOI] [PubMed] [Google Scholar]
- 73.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Nelson CE, et al. Robust Iterative Stimulation with Self-Antigens Overcomes CD8(+) T Cell Tolerance to Self- and Tumor Antigens. Cell Rep 28, 3092–3104 e3095 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Rosato PC, et al. Virus-specific memory T cells populate tumors and can be repurposed for tumor immunotherapy. Nat Commun 10, 567 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Jimenez-Sanchez A, et al. Heterogeneous Tumor-Immune Microenvironments among Differentially Growing Metastases in an Ovarian Cancer Patient. Cell 170, 927–938 e920 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Zhao F, et al. Melanoma Lesions Independently Acquire T-cell Resistance during Metastatic Latency. Cancer Res 76, 4347–4358 (2016). [DOI] [PubMed] [Google Scholar]
- 79.McGranahan N. & Swanton C. Cancer Evolution Constrained by the Immune Microenvironment. Cell 170, 825–827 (2017). [DOI] [PubMed] [Google Scholar]
- 80.Hodi FS, et al. Immune-Modified Response Evaluation Criteria In Solid Tumors (imRECIST): Refining Guidelines to Assess the Clinical Benefit of Cancer Immunotherapy. J Clin Oncol 36, 850–858 (2018). [DOI] [PubMed] [Google Scholar]
- 81.Puza CJ, et al. The Emerging Role of Surgery for Patients With Advanced Melanoma Treated With Immunotherapy. J Surg Res 236, 209–215 (2019). [DOI] [PubMed] [Google Scholar]
- 82.Klemen ND, et al. Patterns of failure after immunotherapy with checkpoint inhibitors predict durable progression-free survival after local therapy for metastatic melanoma. Journal for immunotherapy of cancer 7, 196 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Sandoval F, et al. Mucosal imprinting of vaccine-induced CD8(+) T cells is crucial to inhibit the growth of mucosal tumors. Sci Transl Med 5, 172ra120 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Liu L, et al. Epidermal injury and infection during poxvirus immunization is crucial for the generation of highly protective T cell-mediated immunity. Nat Med 16, 224–227 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Schenkel JM, et al. IL-15-Independent Maintenance of Tissue-Resident and Boosted Effector Memory CD8 T Cells. J Immunol 196, 3920–3926 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Shin H. & Iwasaki A. A vaccine strategy that protects against genital herpes by establishing local memory T cells. Nature 491, 463–467 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Yu CI, et al. Human CD1c+ dendritic cells drive the differentiation of CD103+ CD8+ mucosal effector T cells via the cytokine TGF-beta. Immunity 38, 818–830 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Ali N, et al. Skin-Resident T Cells Drive Dermal Dendritic Cell Migration in Response to Tissue Self-Antigen. J Immunol 200, 3100–3108 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Wculek SK, et al. Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol 20, 7–24 (2020). [DOI] [PubMed] [Google Scholar]
- 90.Dhodapkar MV, et al. Induction of Antigen-Specific Immunity with a Vaccine Targeting NY-ESO-1 to the Dendritic Cell Receptor DEC-205. Sci Transl Med 6, 232ra251 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sehgal K, Ragheb R, Fahmy TM, Dhodapkar MV & Dhodapkar KM Nanoparticle-mediated combinatorial targeting of multiple human dendritic cell (DC) subsets leads to enhanced T cell activation via IL-15-dependent DC crosstalk. J Immunol 193, 2297–2305 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Shwetank, et al. Maintenance of PD-1 on brain-resident memory CD8 T cells is antigen independent. Immunol Cell Biol 95, 953–959 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Richmond JM, et al. Antibody blockade of IL-15 signaling has the potential to durably reverse vitiligo. Sci Transl Med 10(2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Spitzer MH, et al. Systemic Immunity Is Required for Effective Cancer Immunotherapy. Cell 168, 487–502 e415 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Crittenden MR, et al. Tumor cure by radiation therapy and checkpoint inhibitors depends on pre-existing immunity. Sci Rep 8, 7012 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Williams JB, et al. The EGR2 targets LAG-3 and 4–1BB describe and regulate dysfunctional antigen-specific CD8+ T cells in the tumor microenvironment. J Exp Med 214, 381–400 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Im SJ, et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Sade-Feldman M, et al. Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma. Cell 175, 998–1013 e1020 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wu J, et al. T Cell Factor 1 Suppresses CD103+ Lung Tissue-Resident Memory T Cell Development. Cell Rep 31, 107484 (2020). [DOI] [PubMed] [Google Scholar]
- 100.Robinson MH, et al. Subtype and grade-dependent spatial heterogeneity of T cell infiltration in pediatric glioma. Journal for immunotherapy of cancer in press.(2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Dudley ME, Wunderlich JR, Shelton TE, Even J. & Rosenberg SA Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J Immunother 26, 332–342 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Gros A, et al. PD-1 identifies the patient-specific CD8(+) tumor-reactive repertoire infiltrating human tumors. J Clin Invest 124, 2246–2259 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Milner JJ & Goldrath AW Transcriptional programming of tissue-resident memory CD8(+) T cells. Curr Opin Immunol 51, 162–169 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Doedens AL, et al. Molecular Programming of Tumor-Infiltrating CD8+ T Cells and IL15 Resistance. Cancer Immunol Res 4, 799–811 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Sadelain M, Riviere I. & Riddell S. Therapeutic T cell engineering. Nature 545, 423–431 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]