Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Aug 1.
Published in final edited form as: Curr Opin Immunol. 2023 May 23;83:102338. doi: 10.1016/j.coi.2023.102338

In the Right Place at the Right Time: Trm Cells in Immunity to Cancer

Delaney E Ramirez 1, Asmaa Mohamed 1, Yina H Huang 1, Mary Jo Turk 1,*
PMCID: PMC10631801  NIHMSID: NIHMS1904217  PMID: 37229984

Abstract

Tissue resident memory (Trm) cells have recently emerged as essential components of the immune response to cancer. Here we highlight new studies that demonstrate how CD8+ Trm cells are ideally suited to accumulate in tumors and associated tissues, to recognize a wide range of tumor antigens, and to persist as durable memory. We discuss compelling evidence that Trm cells maintain potent recall function and serve as principal mediators of immune checkpoint blockade therapeutic efficacy in patients. Finally, we propose that Trm and circulating memory T cell compartments together form a formidable barrier against metastatic cancer. These studies affirm Trm cells as potent, durable, and necessary mediators of cancer immunity.

Keywords: resident memory T cells, Trm, cancer, CD103, tumor antigen, CXCR6, stemness, immune checkpoint blockade, recall

Introduction

Trm cells are a non-recirculating memory T cell lineage that resides durably in tissues [1,2]. Originally identified for their potency against viral reinfection [3], Trm cells have more recently become recognized for their role in opposing cancer [47]. Trm cells are classically defined by their expression of tissue retention markers CD103 and CD69 [1,8] together with a lack of egress-associated molecules including CD62L, CCR7, S1PR1 [2]. Expression of CXCR6, FABP, PD-1, CTLA-4 and CD39 also identifies Trm populations in normal tissues and in tumors, although expression of these molecules and their isoforms can vary in different tissues [912]. Transcriptionally, Trm cells in cancer are also defined by high expression of ZFP683 (HOBIT), RUNX3, FOS, BHLHE40 and NR4A-family members, as well as low KLF2 and EOMES expression [2,5,1316]. Over the past several years, Trm cells with these features have been identified infiltrating a broad range of solid tumors [11,17].

Here we highlight recent studies that demonstrate how CD8+ Trm cells are ideally suited to form and persist in tumors and associated tissues, to maintain effector function, and to respond to immune checkpoint blockade (ICB) therapy. These studies elevate Trm cells as critical players in the anti-tumor response, revealing their role as broadly distributed mediators of cancer immunity.

The right place: Trm cells in tissues and tumors

Recent studies have shown that tumor growth alone is sufficient for tumor Ag-specific Trm generation in tumors and across peripheral tissues [18]. However, robust protective Trm populations can also be induced therapeutically. Tissue localized [19] and systemic vaccinations [20] elicit Trm responses, which can be further augmented by adjuvant treatments such as anti-CTLA-4 [18,20]. Our studies showed that depletion of regulatory T cells is a key driver of melanoma-specific Trm generation in tumor-bearing hosts, inducing transcriptionally distinct Trm populations across skin, lungs, liver, and lymph nodes (LNs) [21]. Thus, Trm cells can be generated within tumors and across tissues in response to tumor growth and therapeutic interventions (Fig. 1A).

Figure 1. Overview of Trm cell persistence and function in immunity to cancer.

Figure 1.

(A) Tumor-Ag-specific Trm populations form across tissues where tumors grow and metastasize, with clonal counterparts in the blood; “?” denotes the unknown relationship between these compartments. (B) Multiple mechanisms enforce Trm cell persistence, retention, and fitness in the tumor microenvironment (TME). (C) Immune checkpoint blockade (ICB) therapy promotes clonal expansion and effector molecule production by Trm cells in tumors.

In contrast to pre-malignant lesions [22], a wide range of human solid tumors contain tumor infiltrating lymphocyte (TIL) populations with pronounced Trm characteristics [11]. This is consistent with the findings that features inherent to the TME fortuitously promote Trm generation. TGF-β can be produced and used in an autocrine or paracrine fashion by Trm cells, with integrin αv-expressing cells affording TGF-β transactivation in skin [23]. Tumor cell-expressed αv integrin similarly supports Trm differentiation [24]. Accordingly, alterations in the microbiome that impair TGF-β production in tumors were shown to reduce Trm formation [25]. Effects of TGF-β are highly tissue dependent [26] and alone insufficient for the full Trm transcriptional program [27], although human T cells cultured with TGF-β under hypoxic conditions expressed prominent Trm characteristics [28]. This is congruent with findings that T cell-intrinsic hypoxia-inducible factor (HIF) activity promotes the generation of cytolytic Trm-like (CD103+CD69+) TILs in mouse melanoma [29]. These results have important therapeutic implications, as knockout of a HIF negative regulator improved anti-tumor function and Trm formation by chimeric antigen receptor (CAR)-T cells [29].

CXCR6 expression has also emerged as a cell-intrinsic mediator of Trm retention and function in the TME. In human ovarian cancer, CD8+CD103+ Trm-like TILs express high levels of CXCR6 [30,31], and CXCR6 deficiency impairs intratumoral T cell retention, anti-tumor function, and responsiveness to anti-PD-1 therapy [30,32]. Vaccine-induced CXCR6-deficient T cells are similarly impaired in their recruitment to tumors [33]. Mechanistically, we showed that CXCR6 mediates melanoma-specific Trm interactions with CXCL16-expressing dendritic cells (DCs) and is required for Trm maintenance and associated melanoma protection in the skin [34]. CXCR6-CXCL16 interactions also enable IL-15 trans-presentation by DCs to CD8 T cells in tumors [35] allowing delivery of survival signals for T cell maintenance. The importance of Trm-DC interactions is underscored by findings that DCs are associated with elevated Trm proportions in human cancers [3638]. Thus, numerous features of the TME are conducive to Trm cell occupancy (Fig. 1B).

Tumor antigen specificity

Tumor-infiltrating Trm cells recognize a wide range of tumor antigens (Ags). In oral cancer and breast cancer, CD103+CD69+ TIL populations were found to recognize cancer testes Ags MAGE-1 and NY-ESO-1, respectively [39,40]. In melanoma survivors with vitiligo, Trm cells in tumors were clonally matched to those in skin, inferring specificity for melanoma/melanocyte shared Ags [15]. Using neo-Ag prediction for human non-small cell lung cancer (NSCLC), studies recently revealed that Trm populations recognize a range of mutation-associated neoantigens [14]. Moreover, Trm cells in tumors can recognize viral Ags, including tumor-associated viral Ags, such as Hepatitis B in hepatocellular carcinoma (HCC) [41]. However, TILs also recognize solid tumor-irrelevant viruses, such as influenza and Epstein bar virus [9,14,40,42], and such cells can be repurposed for immunotherapy [42]. The T cell exhaustion marker CD39 is useful for distinguishing tumor Ag-specific Trm cells from such bystander cells in tumors [9,14,40]. Compared with tumor specific T cells, bystanders also have lower expression of Trm markers [14,40], suggesting that the Trm phenotype and transcriptional state is enforced by Ag recognition in the TME. Whereas persistent TCR signaling is dispensable for Trm maintenance in viral infection models [43,44], its role in maintaining Trm responses to cancer remains unknown.

Transcriptional heterogeneity and markers of exhaustion

In accordance with ongoing TCR engagement, CD8 Trm cells in tumors often express markers of exhaustion/activation. In NSCLC, CD103+CD8+ TILs express more PD-1, TIM-3, and CD39 than their CD103-negative counterparts [45]. However, Trm cells are transcriptionally heterogeneous within tumors and across human cancer types. Single cell RNA sequencing of CD8 T cells in HCC revealed five separate Trm-like clusters, with only a minor subset expressing exhaustion markers PDCD1, HAVCR2 (TIM-3), and ENTPD1 (CD39) [41]. Similarly, Trm in melanoma formed three Trm-like sub-clusters one of which expressed negative checkpoints (PDCD1, CTLA4, and HAVCR2), and another of which was enriched for effector markers (GZMA, TNFA, and IFNG) [15]. A similar Trm cluster expressing IFNG and other effector cytokine transcripts was also identified within HBV Ag-specific T cells from HCC [41]. In oral head and neck squamous cell carcinoma (HNSCC), two major ITGAE (CD103) and ZFP638 (HOBIT) expressing Trm clusters were identified, only one of which expressed PDCD1 [39].

While it is tempting to use the above markers to infer cellular function or exhaustion, caution is needed. Indeed, studies using gene signatures derived from viral infection models revealed that TILs with features of exhaustion also co-express programs of activation [36]. While some studies show that Trm-like TILs can express the canonical exhaustion marker TOX [15,40], others show that TOX is lower on Trm cells than their terminally exhausted (Tex) counterparts [46]. TIL clonotype and differentiation trajectory analyses show that Trm cells exist in a functional effector state but can also differentiate into TOX+ Tex populations [31]. This is consistent with a model in which Trm cells can adopt either functional or exhausted states in tumors.

Cytokine production, stemness, and persistence: hallmarks of resident memory in cancer

Recent ex vivo and prognostic studies have provided compelling evidence that CD8 Trm cells can mount functional recall responses against cancer. Luminal breast cancer-derived CD39+ Trm cells produced robust IFN-γ, TNF-α, and IL-2 upon restimulation [47]. Accordingly, restimulated CD103+ CD8 T cells from gastric cancer produced more IFN-γ and TNF-α than their CD103-negative counterparts [48]. In HCC, Trm-like cells were also clearly distinct from more exhausted TILs that produced lower levels of effector cytokines [49]. In line with superior cytokine production, Trm-associated phenotypes and gene signatures are associated with improved prognosis across a wide range of human cancers including bladder [50], liver [41], ovarian [31], melanoma [15,21,36], and breast [40,47]. In HNSCC, the presence of Trm in HPV-negative tumors rescues prognosis to levels observed for HPV-positive patients [51]. In melanoma, an IFNG-expressing Trm subset was more prognostic than a TOX-expressing Trm subcluster [15], consistent with findings that IFN-related gene signatures associate with improved prognosis and the presence of Trm cells [36,50]. Combined with prior studies in mouse models [52], these newer studies provide convincing evidence that Trm cells can serve as functional mediators of immunity in humans.

Stemness and durable persistence of the anti-tumor Trm compartment has now also been demonstrated. Whereas the stemness marker TCF-1 has not generally been considered a marker of Trm cells, a subset of sorted CD8+CD103+CD69+ TILs from ovarian cancer were found to express TCF-1/TCF7 as well as IL7R, and LEF1 [31]. Trajectory analysis suggested that these stem-like Trm cells give rise to effector-like Trm cells, with a stem-like Trm signature optimally predicting improved survival in ovarian cancer patients [31]. Such stemness may also explain how Trm clonotypes can persist. In melanoma long-term survivors, we observed that clonotypes identified in early tumor biopsies persisted for up to 9 years in skin, where they exhibited Trm transcriptional properties [15]. Indeed, the presence of “late memory” and “Trm” transcriptional signatures in melanoma patient specimens were most strongly associated with patient survival [36]. Collectively, these studies provide compelling evidence that Trm cells are a functional memory compartment that can confer durable immunity in cancer patients.

The right time: Trm cell function during immune checkpoint blockade (ICB) therapy

Despite their prognostic value, the presence of Trm cells in progressively growing tumors infers their susceptibility to immunosuppressive factors in the TME. Indeed, PD-1/PD-L1 blocking antibodies can act both directly and indirectly to augment Trm cell function. Proliferation and cytokine production by triple negative breast cancer (TNBC)-derived CD39+CD8 Trm cells was rescued by the addition of anti-PD1 during ex vivo restimulation [40]. Cytokine production from gastric cancer-derived CD103+CD8+ T cells was also greatly augmented by anti-PD-L1 treatment in vivo. Interestingly, anti-PD-L1 decreased FABP4/5 expression by gastric tumor cells while simultaneously increasing its expression on Trm cells, thus favoring fatty acid uptake by Trm cells in the TME [48]. Recently it was shown that anti-PD-1/CTLA-4 dual checkpoint blockade preferentially enhances the expansion and killing capacity of CD8+ Trm-like cells, but not their exhausted counterparts [46]. Thus negative checkpoint blockade appears to preferentially rescue Trm function in tumors.

These studies are consistent with findings that Trm-like TIL presence is predictive of patient response to immunotherapy. A large study of NSCLC and bladder cancer patients showed that CD103+ Trm cells in tumors predicted anti-PD-L1 treatment outcomes [53]. Similarly, high CD39+CD103+PD1+ Trm-like TILs at baseline predicted significantly lower risk of recurrence in the adjuvant immunotherapy setting for melanoma [54]. Even more compelling are findings of preferential Trm cell expansion in patients who respond to anti-PD-1 therapy. In melanoma and oral cancer patients receiving anti-PD-1, Trm-like TILs expanded during treatment [36,39]. In contrast, ICB treatment did not expand non-Trm TILs, suggesting that Trm cells are uniquely poised to respond to ICB therapy [39]. These studies convincingly demonstrate that Trm cells mount recall responses during ICB therapy (Fig. 1C).

Trm cells as a component of systemic memory to cancer

While Trm cell localization across tumors and tissues positions them to respond to disseminated cancers, circulating memory (Tcirm) cells also play a key role in this process. In contrast to melanoma protection in the skin and lymph nodes, we found that protection against metastatic-like melanoma in the lungs is afforded by Tcirm cells [21]. Interestingly, studies in patients indicate a link between Trm and Tcirm populations, with numerous clonotypes shared between these compartments [15,39]. Although the relationship between these populations is not yet completely understood, it is known that circulating memory T cells can differentiate into Trm cells in mouse models [6,55]. In patients, neoadjuvant ICB treatment resulted in the expansion of tumor Ag-specific clonotypes in blood [14,39], and these cells shared transcriptional features of TILs (e.g. CXCR6) [39]. Moreover, in gastric cancer patients, ICB treatment induced expression of CD103 on Tcirm cells in blood [56]. It is intriguing to speculate that tumor-specific Trm cells from tumors or tissues might replenish the circulating memory response, particularly considering the recent discovery of such “outside-in” Trm cell responses to peripheral viral reinfection [44,57].

Importantly, the identification of Trm clonal matches across tumor, lymphoid tissues, blood and normal tissues, has revealed that Trm populations form truly systemic memory against cancer. Human melanoma and breast cancer-involved LNs were recently shown to contain Trm populations [21,36,40]. In mice, we demonstrated that Trm cells are protective against melanoma rechallenge in tumor-draining LNs [21]. Trm clonotypes from tumors have similarly been identified across patient-matched normal lung [14,45], healthy liver [41,49], and skin [15]. Together with Tcirm, such broadly disseminated and functional Trm responses constitute a formidable barrier against metastatic cancer.

Conclusions

Trm cells thrive in tissues and tumors as functional memory T cells that are poised for response to ICB therapy (Fig. 1). Additional work is needed to better understand Trm recall capacity, locational dynamics, and plasticity in relation to Tcirm populations. Further study of Trm features within CD4 T cell compartments is also warranted. Therapeutic generation of Trm populations for adoptive therapy is currently in its infancy. However, based on this compelling evidence of Trm function against cancer, future advances must focus on generating these potent, tissue-wide memory T cell compartments.

HIGHLIGHTS.

  • Trm cells are ideally suited to persist in solid tumors and associated tissues

  • Trm cells recognize a wide range of tumor antigens

  • Heterogenous subpopulations of Trm cells occupy human tumors

  • Tumor-specific Trm cells are functional and long-lived

  • Trm cells are uniquely poised to respond to immune checkpoint blockade therapy

ACKNOWLEDGEMENTS

Support was provided by NIH T32 AI00763 to DER and AM, NIH R01CA254042 to YHH and MJT, NIH R01CA22502 to MJT, the Knights of the York Cross of Honour and O. Ross McIntyre, M.D. Endowment to MJT, and a Prouty grant from the Dartmouth Cancer Center to YHH. Figure illustrations were created on BioRender.com

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.Mackay LK, Rahimpour A, Ma JZ, Collins N, Stock AT, Hafon ML, Vega-Ramos J, Lauzurica P, Mueller SN, Stefanovic T, et al. : The developmental pathway for CD103(+)CD8+ tissue-resident memory T cells of skin. Nat Immunol 2013, 14:1294–1301. [DOI] [PubMed] [Google Scholar]
  • 2.Gebhardt T, Palendira U, Tscharke DC, Bedoui S: Tissue-resident memory T cells in tissue homeostasis, persistent infection, and cancer surveillance. Immunol Rev 2018, 283:54–76. [DOI] [PubMed] [Google Scholar]
  • 3.Masopust D, Vezys V, Marzo AL, Lefrancois L: Preferential localization of effector memory cells in nonlymphoid tissue. Science 2001, 291:2413–2417. [DOI] [PubMed] [Google Scholar]
  • 4.Malik BT, Byrne KT, Vella JL, Zhang P, Shabaneh TB, Steinberg SM, Molodtsov AK, Bowers JS, Angeles CV, Paulos CM, et al. : Resident memory T cells in the skin mediate durable immunity to melanoma. Sci Immunol 2017, 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Milner JJ, Toma C, Yu B, Zhang K, Omilusik K, Phan AT, Wang D, Getzler AJ, Nguyen T, Crotty S, et al. : Runx3 programs CD8(+) T cell residency in non-lymphoid tissues and tumours. Nature 2017, 552:253–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Enamorado M, Iborra S, Priego E, Cueto FJ, Quintana JA, Martinez-Cano S, Mejias-Perez E, Esteban M, Melero I, Hidalgo A, et al. : Enhanced anti-tumour immunity requires the interplay between resident and circulating memory CD8(+) T cells. Nat Commun 2017, 8:16073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Park SL, Buzzai A, Rautela J, Hor JL, Hochheiser K, Effern M, McBain N, Wagner T, Edwards J, McConville R, et al. : Tissue-resident memory CD8(+) T cells promote melanoma-immune equilibrium in skin. Nature 2019, 565:366–371. [DOI] [PubMed] [Google Scholar]
  • 8.Walsh DA, Borges da Silva H, Beura LK, Peng C, Hamilton SE, Masopust D, Jameson SC: The Functional Requirement for CD69 in Establishment of Resident Memory CD8(+) T Cells Varies with Tissue Location. J Immunol 2019, 203:946–955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Simoni Y, Becht E, Fehlings M, Loh CY, Koo SL, Teng KWW, Yeong JPS, Nahar R, Zhang T, Kared H, et al. : Bystander CD8(+) T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 2018, 557:575–579. [DOI] [PubMed] [Google Scholar]
  • 10.Pan Y, Tian T, Park CO, Lofftus SY, Mei S, Liu X, Luo C, O’Malley JT, Gehad A, Teague JE, et al. : Survival of tissue-resident memory T cells requires exogenous lipid uptake and metabolism. Nature 2017, 543:252–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Han J, Khatwani N, Searles TG, Turk MJ, Angeles CV: Memory CD8(+) T cell responses to cancer. Semin Immunol 2020, 49:101435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Frizzell H, Fonseca R, Christo SN, Evrard M, Cruz-Gomez S, Zanluqui NG, von Scheidt B, Freestone D, Park SL, McWilliam HEG, et al. : Organ-specific isoform selection of fatty acid-binding proteins in tissue-resident lymphocytes. Sci Immunol 2020, 5. [DOI] [PubMed] [Google Scholar]
  • 13.Li C, Zhu B, Son YM, Wang Z, Jiang L, Xiang M, Ye Z, Beckermann KE, Wu Y, Jenkins JW, et al. : The Transcription Factor Bhlhe40 Programs Mitochondrial Regulation of Resident CD8(+) T Cell Fitness and Functionality. Immunity 2019, 51:491–507 e497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Caushi JX, Zhang J, Ji Z, Vaghasia A, Zhang B, Hsiue EH, Mog BJ, Hou W, Justesen S, Blosser R, et al. : Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature 2021, 596:126–132. *In neoadjvant immunotherapy treated NSCLC tumors, mutational associated neoantigen (MANA)-specific T cells were shown to be enriched in Trm transcriptional state based on expression of HOBIT and CD103. MANA-specific T cells in tumors also had more pronounced Trm transcriptional properties than viral Ag-specific bystander cells. MANA-specific T cell clonotypes appeared in blood following ICB therapy, suggesting a link between Trm and Tcirm compartments. These studies provide novel insights into Ag specificity and behavior of Trm cells in cancer.
  • 15. Han J, Zhao Y, Shirai K, Molodtsov A, Kolling FW, Fisher JL, Zhang P, Yan S, Searles TG, Bader JM, et al. : Resident and circulating memory T cells persist for years in melanoma patients with durable responses to immunotherapy. Nat Cancer 2021, 2:300–311. **By analyzing blood, tumor, and vitiligo-affected skin samples from melanoma patients who were long-term survivors, these studies demonstrated that tumor-matched T cell clonotypes persist in skin as Trm, and in blood as Tcirm for several years. Trm cells in skin and Tcirm cells in blood recognized the melanoma/melanocyte antigen MART-1. Persisting T cell clones preferentially adopted a TRM-IFNG gene signature, which was strongly predictive of patient survival. This study demonstrates the durability of circulating and resident memory T cells in cancer survivors.
  • 16.Sun R, Wu Y, Zhou H, Wu Y, Yang Z, Gu Y, Jiang J, Lu B, Zhu Y: Eomes Impedes Durable Response to Tumor Immunotherapy by Inhibiting Stemness, Tissue Residency, and Promoting the Dysfunctional State of Intratumoral CD8(+) T Cells. Front Cell Dev Biol 2021, 9:640224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Amsen D, van Gisbergen K, Hombrink P, van Lier RAW: Tissue-resident memory T cells at the center of immunity to solid tumors. Nat Immunol 2018, 19:538–546. [DOI] [PubMed] [Google Scholar]
  • 18.Burbach BJ, O’Flanagan SD, Shao Q, Young KM, Slaughter JR, Rollins MR, Street TJL, Granger VE, Beura LK, Azarin SM, et al. : Irreversible electroporation augments checkpoint immunotherapy in prostate cancer and promotes tumor antigen-specific tissue-resident memory CD8+ T cells. Nat Commun 2021, 12:3862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.MacKerracher A, Sommershof A, Groettrup M: PLGA particle vaccination elicits resident memory CD8 T cells protecting from tumors and infection. Eur J Pharm Sci 2022, 175:106209. [DOI] [PubMed] [Google Scholar]
  • 20.van der Gracht ET, Schoonderwoerd MJ, van Duikeren S, Yilmaz AN, Behr FM, Colston JM, Lee LN, Yagita H, van Gisbergen KP, Hawinkels LJ, et al. : Adenoviral vaccines promote protective tissue-resident memory T cell populations against cancer. J Immunother Cancer 2020, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Molodtsov AK, Khatwani N, Vella JL, Lewis KA, Zhao Y, Han J, Sullivan DE, Searles TG, Preiss NK, Shabaneh TB, et al. : Resident memory CD8(+) T cells in regional lymph nodes mediate immunity to metastatic melanoma. Immunity 2021, 54:2117–2132 e2117. *This study uses a mouse melanoma model to show that Tissue localization defines the transcriptional profiles of tumor specific Trm cells. They showed that melanoma specific CD8 Trm cells in LNs prevented metastatic melanoma growth in LNs, whereas circulating memory T cells were sufficient for protection in the lungs. A LN Trm transcriptional profile was also most strongly predictive of prognosis in melanoma patients with LN metastases. These studies reveal the importance of broadly-distributed memory compartments for immunity to metastatic cancer.
  • 22.Willemsen M, Tio D, Krebbers G, Kasiem FR, Jaspars EH, Matos TR, Bekkenk MW, Bakker WJ, Luiten RM: Presence of Skin Tissue-Resident Memory T Cells in Human Nonmalignant and Premalignant Melanocytic Skin Lesions and in Melanoma. Am J Dermatopathol 2022, 44:416–423. [DOI] [PubMed] [Google Scholar]
  • 23.Hirai T, Yang Y, Zenke Y, Li H, Chaudhri VK, De La Cruz Diaz JS, Zhou PY, Nguyen BA, Bartholin L, Workman CJ, et al. : Competition for Active TGFbeta Cytokine Allows for Selective Retention of Antigen-Specific Tissue- Resident Memory T Cells in the Epidermal Niche. Immunity 2021, 54:84–98 e85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Malenica I, Adam J, Corgnac S, Mezquita L, Auclin E, Damei I, Grynszpan L, Gros G, de Montpreville V, Planchard D, et al. : Integrin-alphaV-mediated activation of TGF-beta regulates anti-tumour CD8 T cell immunity and response to PD-1 blockade. Nat Commun 2021, 12:5209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Peng R, Liu S, You W, Huang Y, Hu C, Gao Y, Jia X, Li G, Xu Z, Chen Y: Gastric Microbiome Alterations Are Associated with Decreased CD8+ Tissue-Resident Memory T Cells in the Tumor Microenvironment of Gastric Cancer. Cancer Immunol Res 2022, 10:1224–1240. [DOI] [PubMed] [Google Scholar]
  • 26.Crowl JT, Heeg M, Ferry A, Milner JJ, Omilusik KD, Toma C, He Z, Chang JT, Goldrath AW: Tissue-resident memory CD8(+) T cells possess unique transcriptional, epigenetic and functional adaptations to different tissue environments. Nat Immunol 2022, 23:1121–1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Nath AP, Braun A, Ritchie SC, Carbone FR, Mackay LK, Gebhardt T, Inouye M: Comparative analysis reveals a role for TGF-beta in shaping the residency-related transcriptional signature in tissue-resident memory CD8+ T cells. PLoS One 2019, 14:e0210495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hasan F, Chiu Y, Shaw RM, Wang J, Yee C: Hypoxia acts as an environmental cue for the human tissue-resident memory T cell differentiation program. JCI Insight 2021, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Liikanen I, Lauhan C, Quon S, Omilusik K, Phan AT, Bartroli LB, Ferry A, Goulding J, Chen J, Scott-Browne JP, et al. : Hypoxia-inducible factor activity promotes antitumor effector function and tissue residency by CD8+ T cells. J Clin Invest 2021, 131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Muthuswamy R, McGray AR, Battaglia S, He W, Miliotto A, Eppolito C, Matsuzaki J, Takemasa T, Koya R, Chodon T, et al. : CXCR6 by increasing retention of memory CD8(+) T cells in the ovarian tumor microenvironment promotes immunosurveillance and control of ovarian cancer. J Immunother Cancer 2021, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Anadon CM, Yu X, Hanggi K, Biswas S, Chaurio RA, Martin A, Payne KK, Mandal G, Innamarato P, Harro CM, et al. : Ovarian cancer immunogenicity is governed by a narrow subset of progenitor tissue-resident memory T cells. Cancer Cell 2022, 40:545–557 e513. **Using partition-based graph abstraction trajectory analysis, this study reports that circulating stem-like TCF1-expressing CD8+ T cells give rise to Trm cells, and eventually exhausted Trm cells, in human ovarian cancer. These stem-like Trm cells replenish exhausted Trm and predict improved survival in ovarian cancer patients. This study provides crucial insights into the maintenance of Trm populations in cancer.
  • 32.Wang B, Wang Y, Sun X, Deng G, Huang W, Wu X, Gu Y, Tian Z, Fan Z, Xu Q, et al. : CXCR6 is required for antitumor efficacy of intratumoral CD8(+) T cell. J Immunother Cancer 2021, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Karaki S, Blanc C, Tran T, Galy-Fauroux I, Mougel A, Dransart E, Anson M, Tanchot C, Paolini L, Gruel N, et al. : CXCR6 deficiency impairs cancer vaccine efficacy and CD8(+) resident memory T-cell recruitment in head and neck and lung tumors. J Immunother Cancer 2021, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Vella JL, Molodtsov A, Angeles CV, Branchini BR, Turk MJ, Huang YH: Dendritic cells maintain anti-tumor immunity by positioning CD8 skin-resident memory T cells. Life Sci Alliance 2021, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Di Pilato M, Kfuri-Rubens R, Pruessmann JN, Ozga AJ, Messemaker M, Cadilha BL, Sivakumar R, Cianciaruso C, Warner RD, Marangoni F, et al. : CXCR6 positions cytotoxic T cells to receive critical survival signals in the tumor microenvironment. Cell 2021, 184:4512–4530 e4522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Jaiswal A, Verma A, Dannenfelser R, Melssen M, Tirosh I, Izar B, Kim TG, Nirschl CJ, Devi KSP, Olson WC Jr., et al. : An activation to memory differentiation trajectory of tumor-infiltrating lymphocytes informs metastatic melanoma outcomes. Cancer Cell 2022, 40:524–544 e525. *This study takes a systems biology approach using to characterize T cell populations in human cancer. They report that T cell transcriptional states previously derived from study of viral infection do not translate well to TILs, which generally scored for enrichment of multiple transcriptional states. They showed that the presence of late memory and Trm states in melanoma patient tumors are most strongly associated with survival and responsiveness to ICI therapy. This study provides compelling evidence that memory T cells are required for immunity to cancer in patients.
  • 37.Huang Y, Zhou L, Zhang H, Zhang L, Xi X, Sun Y: BMDCs induce the generation of the CD103(+)CD8(+) tissue-resident memory T cell subtype, which amplifies local tumor control in the genital tract. Cell Immunol 2022, 374:104502. [DOI] [PubMed] [Google Scholar]
  • 38.Zhao H, Wang H, Zhao Y, Sun Q, Ren X: Tumor-Resident T Cells, Associated With Tertiary Lymphoid Structure Maturity, Improve Survival in Patients With Stage III Lung Adenocarcinoma. Front Immunol 2022, 13:877689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Luoma AM, Suo S, Wang Y, Gunasti L, Porter CBM, Nabilsi N, Tadros J, Ferretti AP, Liao S, Gurer C, et al. : Tissue-resident memory and circulating T cells are early responders to pre-surgical cancer immunotherapy. Cell 2022, 185:2918–2935 e2929. **This study demonstrates the presence of two transcriptionally distinct Trm subsets in oral head and neck squamous cell carcinoma. They show that Trm clonotypes in tumors are uniquely poised to expand following anti-PD1 or anti-PD1/anti-CTLA4 neoadjuvant treatment. The study also reports an expansion of matched clonotypes in blood two weeks after treatment, in association with improved patient responses. This study convincingly illustrates a role for both resident and circulating compartments in response to immunotherapy.
  • 40. Lee YJ, Kim JY, Jeon SH, Nam H, Jung JH, Jeon M, Kim ES, Bae SJ, Ahn J, Yoo TK, et al. : CD39(+) tissue-resident memory CD8(+) T cells with a clonal overlap across compartments mediate antitumor immunity in breast cancer. Sci Immunol 2022, 7:eabn8390. **This study identified cancer testes Ag-specific CD39+ Trm cells in triple negative breast cancer tumors and metastatic lymph nodes. They found clonal overlap between Trm and non-Trm cells in tumors, as well as effector memory T cells in blood. Although, CD39+ Trm from tumors were functionally exhausted, ex vivo anti-PD1 treatment rescued cytokine production and proliferation, providing compelling evidence that immune checkpoint blockade can directly reinvigorate exhausted Trm cells.
  • 41.Cheng Y, Gunasegaran B, Singh HD, Dutertre CA, Loh CY, Lim JQ, Crawford JC, Lee HK, Zhang X, Lee B, et al. : Non-terminally exhausted tumor-resident memory HBV-specific T cell responses correlate with relapse-free survival in hepatocellular carcinoma. Immunity 2021, 54:1825–1840 e1827. [DOI] [PubMed] [Google Scholar]
  • 42.Rosato PC, Wijeyesinghe S, Stolley JM, Nelson CE, Davis RL, Manlove LS, Pennell CA, Blazar BR, Chen CC, Geller MA, et al. : Virus-specific memory T cells populate tumors and can be repurposed for tumor immunotherapy. Nat Commun 2019, 10:567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Stolley JM, Johnston TS, Soerens AG, Beura LK, Rosato PC, Joag V, Wijeyesinghe SP, Langlois RA, Osum KC, Mitchell JS, et al. : Retrograde migration supplies resident memory T cells to lung-draining LN after influenza infection. J Exp Med 2020, 217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Wijeyesinghe S, Beura LK, Pierson MJ, Stolley JM, Adam OA, Ruscher R, Steinert EM, Rosato PC, Vezys V, Masopust D: Expansible residence decentralizes immune homeostasis. Nature 2021, 592:457–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gueguen P, Metoikidou C, Dupic T, Lawand M, Goudot C, Baulande S, Lameiras S, Lantz O, Girard N, Seguin-Givelet A, et al. : Contribution of resident and circulating precursors to tumor-infiltrating CD8(+) T cell populations in lung cancer. Sci Immunol 2021, 6. [DOI] [PubMed] [Google Scholar]
  • 46.Virassamy B, Caramia F, Savas P, Sant S, Wang J, Christo SN, Byrne A, Clarke K, Brown E, Teo ZL, et al. : Intratumoral CD8(+) T cells with a tissue-resident memory phenotype mediate local immunity and immune checkpoint responses in breast cancer. Cancer Cell 2023, 41:585–601 e588. [DOI] [PubMed] [Google Scholar]
  • 47.Losurdo A, Scirgolea C, Alvisi G, Brummelman J, Errico V, Di Tommaso L, Pilipow K, Colombo FS, Fernandes B, Peano C, et al. : Single-cell profiling defines the prognostic benefit of CD39(high) tissue resident memory CD8+ T cells in luminal-like breast cancer. Commun Biol 2021, 4:1117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lin R, Zhang H, Yuan Y, He Q, Zhou J, Li S, Sun Y, Li DY, Qiu HB, Wang W, et al. : Fatty Acid Oxidation Controls CD8(+) Tissue-Resident Memory T-cell Survival in Gastric Adenocarcinoma. Cancer Immunol Res 2020, 8:479–492. [DOI] [PubMed] [Google Scholar]
  • 49.Barsch M, Salie H, Schlaak AE, Zhang Z, Hess M, Mayer LS, Tauber C, Otto-Mora P, Ohtani T, Nilsson T, et al. : T-cell exhaustion and residency dynamics inform clinical outcomes in hepatocellular carcinoma. J Hepatol 2022, 77:397–409. [DOI] [PubMed] [Google Scholar]
  • 50.Jin K, Yu Y, Zeng H, Liu Z, You R, Zhang H, Liu C, Su X, Yan S, Chang Y, et al. : CD103(+)CD8(+) tissue-resident memory T cell infiltration predicts clinical outcome and adjuvant therapeutic benefit in muscle-invasive bladder cancer. Br J Cancer 2022, 126:1581–1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Hewavisenti R, Ferguson A, Wang K, Jones D, Gebhardt T, Edwards J, Zhang M, Britton W, Yang J, Hong A, et al. : CD103+ tumor-resident CD8+ T cell numbers underlie improved patient survival in oropharyngeal squamous cell carcinoma. J Immunother Cancer 2020, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Molodtsov A, Turk MJ: Tissue Resident CD8 Memory T Cell Responses in Cancer and Autoimmunity. Front Immunol 2018, 9:2810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Banchereau R, Chitre AS, Scherl A, Wu TD, Patil NS, de Almeida P, Kadel Iii EE, Madireddi S, Au-Yeung A, Takahashi C, et al. : Intratumoral CD103+ CD8+ T cells predict response to PD-L1 blockade. J Immunother Cancer 2021, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Attrill GH, Owen CN, Ahmed T, Vergara IA, Colebatch AJ, Conway JW, Nahar KJ, Thompson JF, Pires da Silva I, Carlino MS, et al. : Higher proportions of CD39+ tumor-resident cytotoxic T cells predict recurrence-free survival in patients with stage III melanoma treated with adjuvant immunotherapy. J Immunother Cancer 2022, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Beura LK, Mitchell JS, Thompson EA, Schenkel JM, Mohammed J, Wijeyesinghe S, Fonseca R, Burbach BJ, Hickman HD, Vezys V, et al. : Intravital mucosal imaging of CD8(+) resident memory T cells shows tissue-autonomous recall responses that amplify secondary memory. Nat Immunol 2018, 19:173–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Nose Y, Saito T, Yamamoto K, Yamashita K, Tanaka K, Yamamoto K, Makino T, Takahashi T, Kawashima A, Haruna M, et al. : The tissue-resident marker CD103 on peripheral blood T cells predicts responses to anti-PD-1 therapy in gastric cancer. Cancer Immunol Immunother 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Fonseca R, Beura LK, Quarnstrom CF, Ghoneim HE, Fan Y, Zebley CC, Scott MC, Fares-Frederickson NJ, Wijeyesinghe S, Thompson EA, et al. : Developmental plasticity allows outside-in immune responses by resident memory T cells. Nat Immunol 2020, 21:412–421. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES