Skip to main content

Phenotypic and spatial heterogeneity of CD8+ tumour infiltrating lymphocytes

Abstract

CD8+ T cells are the workhorses executing adaptive anti-tumour response, and targets of various cancer immunotherapies. Latest advances have unearthed the sheer heterogeneity of CD8+ tumour infiltrating lymphocytes, and made it increasingly clear that the bulk of the endogenous and therapeutically induced tumour-suppressive momentum hinges on a particular selection of CD8+ T cells with advantageous attributes, namely the memory and stem-like exhausted subsets. A scrutiny of the contemporary perception of CD8+ T cells in cancer and the subgroups of interest along with the factors arbitrating their infiltration contextures, presented herein, may serve as the groundwork for future endeavours to probe further into the regulatory networks underlying their differentiation and migration, and optimise T cell-based immunotherapies accordingly.

Introduction

Tumour infiltrating lymphocytes (TILs) are the frontliner immune cells that populate the tumour microenvironment (TME), the very interface of cancer-immunity confrontation, in solid tumours. CD8+ T lymphocytes are considered one of if not the most important constituents of TILs with quasi-universal prognostic benefits owing to their capability to unleash direct cytotoxicity upon encounter with tumour cells expressing the cognate antigen [1, 2]. In reality, however, the presence or even abundance of CD8+ TILs per se does not consistently translate into tumour control or regression, favourable survival outcomes, or response to T-cell-based immunotherapy to the same effect size [3], suggesting failure to divulge relevant modifying factors through undifferentiated evaluation of all CD8+ T cells as a congruent whole. Recent advances in high-throughput, high-definition cellular characterisation modalities such as single-cell RNA sequencing (scRNA-seq), T cell receptor sequencing (TCR-seq), and Assay for Transposase Accessible Chromatin with high-throughput sequencing (ATAC-seq) have uncovered an unprecedented level of CD8+ TIL heterogeneity [4], while spatial transcriptomic (ST) technologies have enabled faithful reconstructions of the positional details inevitably lost during the tissue dissociation process required for scRNA-seq, permitting in-depth analysis of the contact-mediated and paracrine actions between cell populations that are intrinsically location-dependent [5]. Accumulating data on the immensely complex multi-omic landscape of CD8+ TILs suggest that only subpopulations with certain functional and differentiation statuses are empowered by the pre-requisite molecular machinery and stand at the immunological vantage points to act as the principal mediators of anti-tumour immunity and response to immunotherapy [6].

The blowout of high-dimensional data has nonetheless stirred up a taxonomical kerfuffle as it inundates the conventional T-cell classification systems. Before advancing the discussion, it is therefore crucial to reiterate the inherent limitations of the current T cell nomenclature and the caveats when making inferences based on it. Any cell clustering or annotation attempts are more or less arbitrary in nature and at risk of misinterpreting or under-representing the true biological significance of cells. Functional overlaps between pre-defined cell populations frequently exist, and misclassification is common. Besides, the transition from one distinguishable cell phenotype to another routinely involves a gradual, uninterrupted spectrum, but overly refined discretisation to meticulously mirror the continuum may produce unjustified sophistication and compromise generalisability, thus calling for an optimal resolution to meaningfully group cells.

A reference cell tree brilliantly named ‘consensus ontology’ has been envisaged, where the collection of all given cells in both physiological and pathological states over the lifespan of an organism can be moulded into a three-dimensional tree-like architecture with lineage, differentiation, and elapsed time being the localising axial parameters, decorated with the key transcriptional programmes, molecular features, signalling pathways, and, where relevant, spatial information of the cells [7]. Such an epochal idea has provided much inspiration for this review, where we aim to present a lineage-orientated, function-centred panorama of the CD8+ TILs based on prevailing immunological knowledge, zoom in on the subpopulations of interest with regard to anti-cancer immunity and therapeutic benefits, and complement the discussion with pivotal cell-extrinsic and spatial determinants of their successful tumour infiltration.

CD8+ T cell evolution in cancer

Early T cell activation

Naïve CD8+ T cells (TN) are the positively and negatively selected, heterodimeric αβTCR-bearing, and lineage-committed single positive (CD4CD8+) T cells that have not encountered the cognate antigen. Cytometric definition of TN entails exclusive expression of the CD45RA isoform of leukocyte common antigen (CD45RA+CD45RO) along with auxiliary naïve T cell markers, many of which are homing receptors such as C-C motif chemokine receptor 7 (CCR7) and CD62L (also known as L-selectin) that allows homing to lymphoid organs and tissues via blood circulation and then manoeuvring through the white pulp of the spleen or the T cell zone of lymph nodes to survey antigens. They are the last common precursors that beget all CD8+ T cell subtypes found in the secondary lymphoid organs (SLOs) and tumours [1, 8]. There exist numerous obstacles that act as physiological deterrents to autoimmunity and need to be surmounted before TN can be activated to effectors (TEFF) with the full tumoricidal potential. In nascent tumorigenesis, immunologic ignorance can occur when pathological expression of tumour-associated self-antigens is below threshold, not displaying a recognisable differential pattern, or restricted to the immune-privileged sites; or when transformed cells expressing tumour-specific neoantigens are shielded by surrounding tissues from detection and translocation to tumour-draining lymph nodes (TDLNs) for T cell priming. On the other hand, antigen recognition in the absence of a co-stimulus or signal transduction and augmentation by dedicated antigen-presenting cells (APCs) in an appropriately conditioned inflammatory milieu results in a hyporesponsive ‘anergic’ state of CD8+ T cells with limited proliferative and effector capacities [9]. Meanwhile, over-exposure to self-antigens can induce a ‘tolerised’ state that is deficient in cytotoxicity and prone to apoptosis, a phenomenon known as ‘peripheral deletion’ [10, 11].

In the meantime, the effector-memory equilibrium is known to be under tight regulation of orchestrated, potentially antigen-independent, and frequently counterbalancing determinants in relation to cytokines (e.g., effector-predisposing interleukin-12 [IL-12], and IFN-γ versus memory-favouring IL-7, IL-15, IL-21, and transforming growth factor [TGF]-β), TCR signal strength and antigen affinity, antagonising transcription factor duos (e.g., pro-effector T-bet versus pro-memory Eomesodermin, BLIMP-1 versus BCL-6, ID2 versus ID3, and STAT4 versus STAT3), epigenetic remodelling, and metabolic machineries [12]. Resolution of an acute infection customarily leads to a clear-cut inclination towards memory differentiation, survival and self-renewal under the composite force of the regulatory network [13]. In contrast, with an unremitting threat such as a developing tumour, differing chronology of antigen exposure and stimulation precludes fate unambiguity and accentuates the relevance of the variant, intermediate, and hybrid states of memory, effector, and exhausted CD8+ T cells. An entr’acte from constant antigen stimulation, most commonly in the refuge of TDLNs [14, 15] and the blood circulation, is of utmost importance in initiating the memory-associated programmes while postponing the path to exhaustion, a fate ultimately manifested in most tumour-reactive CD8+ T cells [4, 10, 14, 16,17,18,19].

The transmuted memory lane in cancer

Immunological memory denotes the ability to stage a more expeditious and pronounced response upon re-encounter with an earlier insult. Stemness and migrating capacity are the two pillars that uphold the depth (that is, the stretches of the possible effector progenies) and breadth (the tissue versatility and specificity) of the immune protection memory CD8+ T cells (TMEM) are capable of evoking. A substantial proportion of TMEM continually traverse the blood and/or the lymphatics to maximise the antigen screening range. The circulating TMEM were initially divided into central memory cells (TCM) and effector memory cells (TEM) based on expression of CCR7 and CD62L (thus having access to lymphoid tissue), or the lack thereof, respectively [20]. Subsequent investigation has revealed the extraordinary longevity and tremendous developmental and proliferative potentials of TCM [21,22,23,24,25]. Additionally, stem cell memory T cells (TSCM) have been identified as a body of CD45RA+IL-7Rα+ cells sharing overlapping cardinal memory and recirculation features with TCM but are even less differentiated and demonstrate enhanced self-renewal, plasticity, and multipotency [26,27,28,29,30], though Galletti et al. have demonstrated that the apparent gene expression profile differences could very well be accounted for by differential inclusion of PD-1+TIGIT+ progenitors committed to exhausted-like progenies [31]. TEM, in comparison, are characterised by non-lymphoid tissue (NLT) tropism, more immediate effector functions upon restimulation, but less secondary expansion capacity [25].

Although TCM and TSCM are abundant in the peripheral blood of cancer patients [17] and theoretically constitute a systemic reserve of effector CD8+ TILs, such untainted memory phenotypes are difficult to maintain with the systemic perturbations and local TME of tumour-bearing individuals. Tumour-specific bona fide memory cells have been identified in TDLNs [14], but a plethora of single-cell and TCR-profiling studies on melanoma [18], hepatocellular carcinoma [32], colorectal carcinoma [33], and pan-cancer types [4] have unanimously indicated that infiltrating CD8+ T cells that are tumour-reactive almost exclusively reside in an exhausted state of various degrees, and that the heavily expanded, cancer antigen-recognising intra-tumoural TCR clonotypes are rarely detected in the memory compartment of the peripheral circulation [18]. This could suggest inadequate development of a circulating memory repertoire specifically targeting tumours across different cancer types, and even if spawned, these memory cells may be rapidly depleted after mobilisation. Therefore, the precise size of the circulating tumour-reactive memory compartment and magnitude of contribution from these cells to naturally occurring anti-cancer immunity prior to acquiring exhaustion features are to be further assessed in human cancers.

That being said, the superior proliferative potential of TCM and TSCM can be leveraged in various forms of cancer immunotherapies. TSCM can be generated ex vivo from naïve precursors using clinical-grade culture protocols utilising IL7 and IL15 [27], or a combination of IL-7, IL-21 and the WNT agonist TWS119 [34]. Recently, MEK inhibition has also been shown to be capable of inducing TSCM and functionally superior TCM [30]. Compared to chimeric antigen receptor (CAR)-T cell therapy derived from unselected T cells, adoptive transfer of CAR-TSCM and TCM have engendered expanded, persistent, and intensified anti-tumour responses against haematologic malignancy xenografts [34,35,36] with reduced risk of cytokine release syndrome [35], and, in combination with PD-1 blockade, achieved tumour control in OVCAR-3-inoculated mice [37]. Likewise, engineered expression of IL-7 along with CCL19 on CAR-T cells boosted memory responses to solid tumours and prolonged survival in mice [38].

Tissue-resident memory

The observation of ample long-lived, TEM-like CD8+ T cells in NLT heralded the revelation of tissue-resident memory T cells (TRM) [39], which have been formally recognised with skin [40] and intestinal [41] transplantation in murine models and subsequently found prevalent in SLOs and both barrier and non-barrier tissues in mice and humans [42, 43]. The non-migratory nature of TRM stems from the expression of integrins CD49a and CD103, the latter of which is the integrin subunit αE (ITGAE) that serves as a canonical TRM marker, and CD69, an early activation marker and down-regulator of relocation-promoting sphingosine 1-phosphate receptor-1 (S1PR1), in addition to a concomitant lack of tissue egress mediators [42, 43]. An array of universal (e.g., CXCR6 and VIM) and site-specific (e.g., CCR4, CCR10, and CXCR4 for skin versus CCR9 for small intestine predilection) cell matrix and adhesion molecules also reliably delineate TRM [44, 45]. Additionally, upregulation of transcription factors (TFs) such as BLIMP-1, HOBIT, RUNX3 and NOTCH is pivotal to TRM formation and has further instituted TRM as separate entities to the circulating memory T cells in mouse models [46,47,48]. Suppression of KLF2 is another TRM hallmark that provides an additional binding force for TRM to the surrounding tissue via resultant down-regulation of S1PR1 [49]. Many of these TRM-enriched TFs, including Runx3, Notch, and Hif1a, are known harbingers of effector T cell functions, which underlie the phenotypic preparedness of TRM in repelling invading pathogens and rejecting transformed cells [47, 48, 50]. However, it remains uncertain the extent to which the core transcriptional signature is shared with human TRM. Notwithstanding the discrepancies in Hobit expression pattern [48], recent studies have corroborated perpetuation of some of these TRM TFs in humans, and identified other ones of interest such as AHR, KLF4, and Hic1 [44, 51, 52].

Mounting evidence has brought TRM to the centre of attention in quelling numerous cancers, particularly those of epithelial origins. It has been recognised that TRM cells are central to immunosurveillance, especially in tissue compartments not readily accessible by circulating memory, as well as curtailment and, in an ideal situation, prompt elimination of transformed cells and subclinical tumours, analogous to their presumed contribution in local containment of latent viruses [53,54,55,56]. Park et al. have notably shown in their seminal work that tumour-specific TRM unfailingly prevented melanoma development upon rechallenge independent of circulating memory depletion, and upon TRM knockdown, tumour outgrowth manifested in approximately 20% of mice with occult melanomas [54]. This is echoed by TRM-mediated protection against secondary breast tumour rechallenge in the ipsi- but not contralateral mammary fat pad [55]. TRM can also derive from intratumoural TMEM/TEM [57] and reside in the lymph nodes [58], offering protection against metastases at distant sites. Furthermore, tumour-associated clones of TRM generated post-immune checkpoint blockade (ICB) were able to persist for up to 9 years [59], illustrating the long-lasting protection that TRM confer. The consequent selective pressure from TRM immunosurveillance, however, may paradoxically favour the clonal growth of cancer cells that are adept at immune evasive and suppressive manoeuvres, such as the cases in lung cancers where tumour cells escape TRM hammering by down-regulation of MHC-I molecules [60] and engaging epithelial-to-mesenchymal transition [61], which epitomises the phenomenon of immunoediting that signifies the complex dynamic of the cancer-immunity equilibrium [62].

In established solid tumours of many types, enrichment of TRM transcriptomic signatures in tumours is also found invariably correlated with more intense anti-tumour activity with respect to cytolytic molecule expression and pro-inflammatory cytokine release, and better prognoses, irrespective of the overall amount of CD8+ T cell infiltrates [59, 63,64,65,66,67,68,69,70,71]. As known expressors of immune checkpoint molecules [63, 66, 71, 72], infiltrating TRM represent one of the prime salvageable targets of ICB therapy [55, 67, 71, 73, 74], where reinvigoration and expansion of TRM have been consistently observed following ICB, and pre-treatment TRM signature prognosticated response to ICB-based regimens [71, 73]. However, accurate measurement of the anti-tumour momentum and therapeutic efficacy attributable to conventional TRM and their progenies is challenging due to the entangled CD8+ T cell fates within the TME, as TILs with TRM-like phenotypes could very well develop from activated T cells of distinct lineages secondarily acquiring TRM features [55, 70, 75, 76]. Gavil et al. have demonstrated that, in the context of a tumour, the common TRM markers (CD69 and CD103) failed to reflect tumour residence, nor did CD62L exclude the same [75]. Instead, chronic antigen stimulation endowed incoming tumour-specific CD8+ T cells with a distinct resident programme that matured in concert with exhaustion [75]. TME factors such as TGF-β, which is known to induce CD103 [52, 61, 68, 77], could also be contributing. Furthermore, recently conducted in-depth TIL characterisation studies have resolved TRM subpopulations of extended significance. In tumours and vitiligo-affected skins of metastatic melanoma patients who responded well to immunotherapy, subcluster of TRM distinguished by abundant expression of certain pro-inflammatory cytokines and chemokines predicted better patient overall survival among other clinical variables, whereas the TRM cluster defined by TCR signalling transcripts did not [59]. In ovarian cancers, a group of progenitor TRM descended from a recently activated CD103CD69+ re-circulating population was pinpointed as the reservoir of tumour-reactive effectors and associated with improved outcomes [70]. Therefore, additional investigations are warranted for more rigorous ways to distinguish bona fide TRM from TRM-like cells in the TME and resolve the subsets with the most tumoricidal capacity and the manipulable therapy targets.

The transcriptomic and epigenetic stigmata of exhaustion

Exhaustion entails the compromise in anti-tumour immunity due to prolonged stimulation of immune cells, which ubiquitously occurs in tumour-reactive CD8+ TILs and leads to dwindling overall magnitude of T cell anti-tumour response in the long term [78]. TCR activation is naturally an antecedent to and propagator of T cell exhaustion. Low-affinity interactions usually lead to functional inertness or failure to instigate a self-sustaining tumour-reactive T cell population [79], whereas exceedingly strong TCR signals may predispose tumour-reactive CD8+ T cells to accelerated exhaustion [80,81,82]. A minimum length of time is also required for initiation towards exhaustion differentiation [15, 83, 84], as evidenced by conserved lineage flexibility of virus-specific CD8+ T cells when removed from chronic infection in the first weeks [85], and yet the time point at which CD8+ T cells commit to the exhaustion fate in cancer remains a subject of active investigation. Recently, an interrogation of chromatin accessibility and transcriptomic alterations of early activated CD8+ T cells in tumour-bearing mice has revealed that acquisition of exhaustion hallmarks commenced as early as within 6 h of tumour antigen exposure, well in advance of the previously thought timeframe of days to short weeks [86]. Also, elements of exhaustion (loss of effector function, upregulation of immune checkpoint molecules, and contracted proliferative capacity) are regulated independently and ripen asynchronously [86,87,88], where, for instance, IFN-γ and TNF loss preceded that of proliferative capacity by a large margin [86]. These findings suggest that extrapolation of CD8+ T cell exhaustion state from isolated parameters is an error-prone and misleading practice, and instead an all-inclusive, weighted scoring fabric may be necessitated.

Liberal usage of the term ‘exhaustion’ frequently constitutes a misnomer in that cells commonly designated as exhausted CD8+ T cells (TEX) encompass a wide spectrum that spans across near-complete preservation of function to absolute indolence [4, 89], and that the degree of reprogrammability by ICB is not indicated by the somewhat deceptive terminology [90]. Despite these variations, once lineage committed, all TEX are, without exception, constrained by the transcriptional rewiring and epigenetic scarring that persisted in progenies and precluded complete reversal to the pristine naïve or memory-level of functional reserve even after antigen withdrawal [31, 91, 92]. TCR signalling-mediated upregulation of nuclear factor of activated T-cells (NFAT) family of TFs lays the foundation for the extensive and incompletely understood regulatory network of interdependent TFs that perpetuate the exhaustion programme [84]. NFAT holds critical roles in both T cell activation/effector function and tolerance/exhaustion, the former of which typically requires partnering of NFAT with AP-1 proteins [93]. With relative scarcity of AP-1 secondary to, for instance, insufficient MAPK signalling from inadequate co-stimulation, uncomplexed NFAT is diverted to monomeric binding motifs unaccompanied by an adjacent AP-1 site, inducing transcription of exhaustion-related genes encoding inhibitory molecules (e.g., Pdcd1 [encoding PD-1], Lag3, Havcr2 [TIM3], Tigit, Ctla4) and repressive TFs (e.g., Tox, Egr1, Egr2, and Nr4a) [86,87,88, 93,94,95,96,97]. In conformation with the dynamics of the down-stream transcripts, NFAT motifs are enriched in the differentially accessible chromatin regions of TEX [84, 86, 97], and the predicted NFAT binding sites in the exhaustion-related genes also increased in accessibility [87, 88]. The partner-dependent functional bivalency of NFAT provides an opening for manipulating the exhaustion-versus-effector differentiation, where overexpressing c-Jun, a canonical AP-1 factor, granted the CAR T cells exhaustion resistance [98]. Akin to NFAT, BATF has bipartite roles after TCR signalling induction, and overexpressing BATF promoted BATF-IRF4 cooperation, thereby skewing CAR TILs towards an effector phenotype [99].

Among other NFAT-induced TFs, TOX has gained recognition in recent years as an essential potentiator of exhaustion [87, 92, 100,101,102], without which differentiation into TEX is averted altogether [100, 103]. Although acute infections saw temporary presence of TOX at low levels, sustained high-intensity TOX expression in chronic infections and tumours upregulates inhibitory receptors, further modulates the TF network, and eventually engraves modification of the corresponding exhaustion-specific chromatin landscape in an NFAT-independent, self-propagating manner [84, 87, 100], thereby inflicting the indelible epigenetic scars on TEX that forbid them from full recovery of memory/effector functions after antigen withdrawal [92] or ICB rescue [104]. With that said, TOX-knockout tumour-specific T cells failed to persevere in tumours and did not exhibit superior effector functions compared to the wild-type counterparts [87], which shows that TOX is necessary for TEX survival [102] and exemplifies the diverging regulatory networks underlying different exhaustion modules.

Stemness sustains response in the face of exhaustion

Founding and upkeep of tissues and cellular cohorts are routinely overseen by precursors with regenerative and stem-like attributes, and TEX are no exception. Despite the distinct exhaustion-associated epigenetic scars, an extensive and still expanding body of studies has firmly established a distinguishable subgroup of TEX with reserves for longevity, self-renewal, and proliferation transcending those of the rest, which is correspondingly named ‘precursor exhausted’ or ‘progenitor exhausted’ T cells (TPEX) but also sometimes referred to as ‘stem-like’ and ‘memory-like’ cells [16, 76, 89, 103, 105,106,107,108,109,110]. The relevance of TPEX in cancer and immunotherapy was presaged by cornerstone works in chronic viral infections [105, 111, 112], and notably a PD-1+CXCR5+CD8+ T cell population possessed a gene signature resembling memory precursor cells and haematopoietic stem cell progenitors, and proliferative bursts after PD-1 blockade while differentiating into more terminally differentiated effector-like progenies [105], although TPEX are typically lacking in cytotoxicity themselves [17]. The existence and behaviour of such cells have been then substantiated in lung cancer [110] and melanomas [76, 90], and ablation of these cells dampened efficacy of checkpoint blockade. Further studies have validated significant post-treatment intra-tumoural accumulation of TPEX exclusively in ICB responders, where lineage tracking analyses credited the cell population increase to the composite effect of local expansion of pre-existing tumour-specific clonotypes and likely an influx of extratumoural T cells, in light of the novel TCR clonotypes not present within the tumour prior to therapy [74, 107, 113].

Unsurprisingly, TPEX may co-express exhaustion (e.g., TOX and PD-1), memory (e.g., EOMES, SLAMF6 [Ly108 in mice], BCL6, ID3, and IL7R[CD127]), and naïve-like cell markers (e.g., CCR7 and CD62L) [84, 105, 114]. TPEX differentiation is governed by an expansive set of TFs such as FOXO1 [115, 116], BCL6 [114], MYB [117], BACH2 [118], and HMGB2 [119]. Notably, T cell factor 1 (TCF1, encoded by Tcf7) has emerged as a key TF that dependably defines TPEX [76, 84, 108, 109]. TCF1 is a well-known enactor of Wnt-β-catenin signals and assumes a myriad of roles in T cell development and maturation pending the precise circumstance [120, 121]. At the post-antigen encounter bifurcating point of CD8+ T cell fate, TCF1 conspicuously steer differentiation of effector-phase lymphocytes towards the stem-like endpoints as opposed to short-lived effectors, giving rise to TCM in acute infection [22, 122] and TPEX in chronic infection and tumour models [29, 92, 114, 123,124,125], suggesting the transcriptional schemes that instil stemness in these two cell populations are, at the very least, partially overlapping [14, 31, 113, 126]. On the contrary, as TCF-1 suppresses TGF-β-induced CD103 expression [77], TRM are known to be TCF1 [70, 76, 77], despite displaying a stem-like phenotype comparable to that observed in TCM and TPEX. Moreover, involvement of TCF1 in TPEX generation is likely redundant and relies on coordinated changes in a set of additional factors [125], where Tcf7 knockout did not significantly alter the abundance of TPEX TIL [76]. On the other hand, TCF1 has been demonstrated to be indispensable for the maintenance of the TPEX phenotype and arbitrates tumour control and response to ICB as well as cancer vaccination, with clear superiority of the TCF1+CD8+ T cell population over the TCF1 counterparts in these regards [76, 90, 109, 127]. In fact, a recent study has shown that TCF1 is required for optimal priming of tumour-specific CD8+ T cells and ICB efficacy in the context of poorly but not highly immunogenic tumours [124], resembling the critical role of TCF1 in ‘pre-programming’ the chromatin landscape of TCM for proper upregulation of the necessary cell cycle, DNA replication and glycolysis-related genes during a recall response [122].

Nevertheless, closer examination of TPEX has shown that TCF1 alone is insufficient to demarcate the subsets of biological significance contained within the heterogenous cell population [103]. A recent study conducted in the murine LCMV infection model unveiled that CD62L+ TPEX, whose development relies on MYB, are transcriptionally distinct and functionally superior to the CD62L ones, and bring forth the proliferative burst following PD-1 blockade[117]. Cells with a comparable transcriptomic profile have been identified in CD8+ CAR-T cells responding to B-cell acute lymphoblastic leukaemia, with FOXP1 being a newly identified hub TF that orchestrates stemness and anti-tumour response, although the equivalent to this subpopulation fortified with stemness was not readily evident in the cells infiltrating solid tumours in the same study[149]. Similarly, TCF1+ TPEX with circulating capacities and greater proliferative potentials are found to be associated with an SLAMF6+ as opposed to SLAMF6phenotype in a murine lung adenocarcinoma model[163].

En route to terminal exhaustion

Tumour-specific TPEX on the ground, along with other stem-like populations, eventually embark on the forced march in the direction of terminal exhaustion (Fig. 1A), at which point irrecoverable loss of intrinsic and therapeutically inducible anti-tumour activity manifests. Although terminally exhausted CD8+ T cells (TTEX) exuberantly express IFNG, PRF1 and GZMB transcripts [4, 18], diminished polyfunctionality, survival, and expanding capacities fundamentally restrict the cumulative anti-tumoural potential of individual TTEX, which are not amenable to ICB reinvigoration despite plentiful expression of checkpoint molecules such as PD-1 and TIM-3 [88, 109, 113]. Markers for TTEX also notably include CD39 (ENTPD1) [4, 90, 113, 126], an ectonucleotidase that, in conjunction with CD73, potently generates extracellular adenosine, which is known to inhibit T cell signalling cascades [128,129,130]. Accordingly, Vignali et al. have recently shown that not only do TTEX take on a powerless stance against tumour cells, but they also exert immunosuppressive effects comparable to those of Foxp3+ regulatory T cells (TREG) isolated from the same tumour environment, primarily through hypoxia-driven enhancement of CD39 density and activity [131], in accordance with the benefits observed with exploratory usage of CD39/CD73 monoclonal antibodies [132] and dual inhibition of CD39 and TIM3 [90] in mouse tumour models. Contrarily, the authors did not observe significant alterations in tumour growths or TTEX-mediated immune suppression with germline deletion of or neutralising antibody administration against IL-10 [131], a pleiotropic cytokine with well-recognised anti-inflammatory and immunomodulatory functions whose involvement in cancers is obscured by the discordant findings in available studies [133]. Interestingly, an engineered IL-10-Fc fusion protein actualised TPEX-independent re-expansion and cytotoxic function revitalisation of TTEX through fostering mitochondrial pyruvate carrier-dependent oxidative phosphorylation [134], echoing other pre-clinical [135] and clinical [136, 137] studies. However, the following phase II trials combining PEGylated IL-10 and anti-PD-1 antibodies led to doubling of treatment discontinuation rate due to adverse events while bringing no added survival benefits in metastatic non-small cell lung cancer patients [138]. Taken together, these findings highlight the fact that, in addition to being functionally subpar, TTEX can potentially exert suppressive actions owing to susceptibility to the various TME elements, and that therapeutic strategies alternative to ICB, especially those pertaining to reprogramming T cell metabolism, could potentially exploit the remaining, albeit very limited, plasticity of TTEX, though how they may best synergize with other modalities of immunotherapy and accurately target the intended cells is to be explored.

Fig. 1
figure 1

Outlining CD8+ T cell evolution in cancer. (A) On the way to terminally differentiated states, naïve CD8+ T cells in cancer move through a meshwork of transcriptomically distinct phenotypes. The cells may stroll down the exhausted (top) or memory (bottom) thoroughfare, though memory cells can give rise to exhausted cells after acquiring the epigenetic imprints and the TRM branch has clear transcriptional idiosyncrasies. In general terms, progression through either lane sees the waning of stem-like features and reprogrammability by immunotherapy and eventual fate convergence at the TTEX terminus. TEXEFF and TEMRA are lineage-wise juxtaposed with TPEX but infused with heightened effector capacities. (B) Subsets of TEX are identifiable by the expression of an assortment of TFs and surface proteins. TPEX are vital to maintaining the exhausted T cell populations due to their stem-like features and capabilities to self-renew, as indicated by TCF1 expression, whereas TOX marks the inability of exhausted cells to fully reconstitute the memory or effector functions. The quiescent TPEX1 subset is distinguished from the mobilised ones by SLAMF6 and CD69 expression profiles. The activated state of TPEX2 is illustrated by the acquisition of the proliferative marker MKI67 and effector molecules IFN-γ and TNF-α, whose expression plateaus in TINT before returning to low levels in TTEX. In addition to escalation of immune checkpoint molecules (e.g., PD-1), TTEX also express terminal exhaustion makers (e.g., CD39 and TIM-3). TEXEFF may form in place of TTEX only under certain circumstances such as TH-derived IL-21 induction. TEX differentiation is under the oversight of an extensive, tightly regulated TF network, as shown in the figure. Notably, many of the TFs maintaining the TPEX population are implicated in memory formation, and those propelling their differentiation support the effector cells during acute infection.

Meanwhile, progression from TPEX to TTEX entails a sophisticated succession of events subject to an array of cell-intrinsic and extrinsic modifiers and thus markedly variable in pace, but generally aligns with the alterations in expression levels of the master regulators, namely a hierarchical loss of TCF1-instituted stemness and culmination of TOX-associated epigenetic imprinting, which foreshadow the downstream transcriptional and functional changes [88, 102]. Though direct transitioning from TPEX to TTEX has been observed [139], TPEX differentiation into a metabolically rewired and actively proliferating intermediate-exhausted phenotype (TINT), marked by a Tim3+CD101 phenotype or CX3CR1 expression in chronic infections [17, 139, 140], proves to be a pre-requisite step to liberate the full anti-tumour potential and ICB responsiveness of TPEX [103, 140,141,142,143], since TPEX are mostly quiescent by default, as attested by low expression of cell cycling genes such as MKI67 [17]. This is validated by the latest advances, where toning down Ikzf1 (encoding IKAROS, which suppresses TCF1) has successfully retarded TPEX-to-TINT differentiation, but failed to evoke added antitumour effects [142]. Admittedly, the transitory TEX populations were evident in earlier TME profiling studies [19, 32, 89, 90], but their structured categorization was pioneered by Beltra et al., where, using Ly108 (a proxy marker for TCF1) and CD69, they have distinguished the interconverting quiescent (TPEX1, Ly108+CD69+) and proliferating (TPEX2, Ly108+CD69) subsets of TPEX, and established the developmental trajectory from TPEX2 to Ly108CD69 TINT and ultimately Ly108CD69+ TTEX [103], thus completing the TPEX1 ➜ TPEX2 ➜ TINT ➜ TTEX paradigm that was consolidated by succeeding studies [142, 144,145,146] (Fig. 1B). Increasing evidence suggests that the TPEX2-to-TINT junction harbours a critical epigenetic, transcriptional and metabolic checkpoint, whose regulation incorporates inputs from activation of the mTOR pathway [142], which serves as a control panel integrating cellular metabolism sensor signals [147], and a range of TFs, including T-bet [103, 142, 148], BATF [148], KLF2 [149], ETS1 [142], and STAT5 [150]. In fact, augmentation of Stat5 activity by employing an orthogonal IL-2:IL2Rβ pair system in established TEX cells not only stimulated TINT formation, especially when combined with PD-L1 blockade, but also improved their durability and effector functions in the face of persistent antigen stimulation, and suppressed differentiation to TTEX [150].

Indeed, TINT may still bear therapeutically accessible developmental pliability and effector potential. A growing body of studies has shed light onto an alternative differentiation path from the TINT waypoint to an effector-like exhausted (TEXEFF) population that is inducible by IL-21-producing CD4+ helper T cells (TH) [139], TF ZEB2 [144], and lower TCR avidity [81, 146], and lineage-wise juxtaposed with TTEX but possesses strong effector or NK cell characteristics [81, 139, 146]. Multiple lines of evidence derived from murine chronic infection models are in support of this bifurcating model placing TINT at the branch point, including the in silico predictions based on pseudotime and RNA velocity trajectory analyses, retained phenotypic plasticity in TINT but not TEXEFF or TTEX upon adoptive transfer, and shared open chromatin regions between TINT with either TEXEFF or TTEX and the paucity thereof between TEXEFF and TTEX [81, 139, 146]. This novel model is of great therapeutic relevance, as it provides the theoretical foundation for a window of opportunity, perhaps the last one before the end-stage epigenetic ossification takes place, to expand the aggregate effector activity by diverting TINT towards the TEXEFF fate in lieu of TTEX [151].

To complicate matters even further, concurrent ‘non-TPEX-origin’ trajectories to TTEX progressing through memory subsets have been described [4, 70, 72, 74, 144, 152], which are generally organised into two interlaced paths marked by the resident-versus-circulating duality of memory (Fig. 1A). In a pan-cancer TIL atlas constructed from scRNA-seq data from 316 patients across 21 cancer types, Zheng et al. projected that CD8+ TN could ultimately develop into either TEX or TEMRA [4], concordant with recent identification of a non-TEX ZNF683high TEFF/TEM endpoint that was accentuated in marrow-infiltrating CD8+ T cells in Richter syndrome responding to PD-1 blockade [152]. Along the path to TEX, TN first progressed to IL7R+ TMEM (TCM/TSCM) and then split into the GZMK+ TEM and ZNF683+CXCR6+ TRM branches before uniting at the GZMK+ TEX and, finally, TTEX fate [4]. A similar scheme was outlined in an NSCLC dataset, involving progression to TTEX (CD8-LAYN) through a transitional population (CD8-GZMH) from circulating (CD8-KFL2/CD8-GZMK) and tissue-resident (CD8-XCL1) precursors, where the TRM population is deemed the main source of the more differentiated tumour antigen-specific T cells based on TCR sharing [72] – a clonal predilection that was also seen in ovarian [70] and neoadjuvant ICB-treated oral cancers [74]. Taken together, suffice it to say, notwithstanding the concrete accomplishments in the nuanced dissection of the phenotypes, current models may still merit refinement and reconciliation for an unequivocal representation of CD8+ TIL development and evolution, which is made particularly challenging by the confounding qualities unique to chronic antigen stimulation and TME, the logistical and technical constraints, and the taxonomical dilemmas for the betwixt and between cell populations.

Navigating the haemolymphatics and TME: a spatial odyssey

The relevance of tumour-specific CD8+ T cell phenotypes is nullified if detached from the position coordinates, given the contact-dependent nature of their tumoricidal weaponries. Indeed, the quantity and spatial distribution of CD8+ TILs demonstrated drastic variations across the cancer types, anatomical sites, and lesion classifications (i.e., primary, recurrent, or metastatic) [2, 4]. This observation has prompted development of Immunoscore, a histological type-agnostic grading system quantifying CD3+ and CD8+ lymphocyte densities in the tumour core and the invasive margin that has shown prognostic values in colorectal and other cancers [2, 153,154,155,156], effectively categorising tumours into immune infiltration-based ‘immunotypes’, namely the heavily infiltrated ‘hot’, the infiltration-devoid ‘cold’, and, between these two extremes, the uniformly poorly infiltrated ‘immunosuppressed’, and the ‘excluded’ tumours where infiltrates are limited to the invasive margin [155]. Importantly, the immunotypes cannot be explicated by tumour cell antigenicity alone and are in fact telltale snapshots that mark the culmination of the long march of CD8+ T cells as they migrate through different circulatory compartments, penetrate multiple barriers, and persevere in spite of suppressive environments, to complete the larger cancer-immunity cycle [157]. Along the journey, CD8+ T cells adjust the balance of tissue anchoring and egress factors to facilitate or restrain translocation, perform tissue-specific adaptive manoeuvres, and pass through several key anatomical and microenvironmental locales that profoundly shape their dynamics (Fig. 2), leading to reverberations in the intratumoural CD8+ T cell landscape.

Fig. 2
figure 2

Critical anatomical and microenvironmental waypoints dictating success of CD8+ T cell infiltration. (A) TDLNs are the ideal venue for tumour antigen-bearing migratory DCs from the TME to condition the TH and CD8+ T cells. The migratory DCs also pass on the antigens and TME cues to their resident counterparts. With provision of the primary, costimulatory, and cytokine signals by the DCs and TH sans the inhibitory TME elements, CD8+ T cells have the privilege of remaining in the stem-like states and serving as a reservoir of tumour-repressive cells. Establishment of metastases in the LNs demolishes the supportive context for these population-sustaining CD8+ T cells. (B) Tumour-associated vascular endothelium is a major barrier against T cell infiltration. Endothelial expression of FasL can precipitate direct CD8+ T cell death. Aberrant adhesion molecule expression secondary to VEGF-induced endothelial cell anergy renders it impregnable to CD8+ T cells. Tumour-associated high endothelial venules are important sites for CD8+ T cell extravasation and aggregation. (C) CAFs orchestrate the tumour-associated stroma and mediate T cell exclusion in a multitude of ways, as illustrated. The APC niches and TLSs are on-site powerhouses that harbour stem-like CD8+ T cells and, by safeguarding their survival, greatly amplifies anti-tumour immunity. Conversion to the more terminally differentiated phenotypes ensues the longer the dwell time and the closer the distance to the tumour cells, which are known to foment an immunosuppressive aura.

Tumour-specific CD8+ T cells gear up in TDLNs

The conventional school of oncology often view TDLNs as receptables forming the catchment for metastatic seeding, which is predisposed by the architectural and functional deviations from the non-draining nodes provoked by tumour-derived lymph-borne factors [158]. Removal of uninvolved TDLNs, however, failed to improve outcomes in preclinical models or cancer patients [159,160,161]. In this day and age, the significance of TDLNs as a nexus between the cancer and immune cell ends of the tug-of-war has been brought back to attention and reinforced, and their role in facilitating generation of CD8+ TILs reaffirmed, for the key window for antigen presentation and T cell priming arises as tumour antigens, T lymphocytes, and dendritic cells (DCs) converge in the lymphatic endothelial cell (LEC)-lined and macrophage-interspersed subscapular sinuses (SCS) of TDLNs [158, 162]. Current evidence suggests that CCR7-expressing type I conventional DCs (cDC1s) migrated from the TME are the main DC subset that internalise and transport tumour antigens in large quantities and potently prime CD8+ T cells through MHC-I-mediated cross-presentation or cross-dressing (direct transfer of peptide-MHC-I complex) [162,163,164,165,166,167,168] and CD4+ T cells through MHC-II-mediated direct presentation, which reciprocally augment the immunostimulatory proficiency of DCs [169]. Meanwhile, the TME IFN-I-induced ISG+ cDC2s also demonstrated capability to activate CD8+ T cells by cross-dressing in the absence of cDC1s [170]. Once arrived at the TDLN, the migratory DCs may in turn transfer tumour antigen to their resident counterparts to prepare the inbuilt DC terrain for CD8+ T priming [168, 171], where the DC status across the tissue origin and the LN may also be synchronised with co-transfer of contextual cues [172].

Given their nurturing multilayered cellular infrastructure and strategic anatomical locations, TDLNs are naturally the reservoirs of tumour-specific CD8+ TILs. An abundance of studies have provided evidence of CD8+ T cell transmigration from TDLNs to the tumour [14, 15, 141, 159, 163, 173,174,175,176] and vice versa [15, 177], as well as enrichment of tumour-reactive CD8+ clonotypes in TDLNs [175, 176, 178]. Unencumbered by the stress from persistent antigen stimulation and TME factors, activated tumour-specific CD8+ T cells in TDLNs are typically found in the minimally exhausted TPEX or TPEX-like states [79, 117, 141, 163, 173, 174, 178, 179], whereas those TILs clonally related to them predominantly display the TTEX phenotypes [141, 173, 178]. In fact, expansion of pre-existing or de novo LN-dwelling stem-like populations is vital to ICB response [14, 105, 175, 176, 180] (Fig. 3A) and potentially accountable for a greater share of efficacy than those circulating or residing in the TME, as surgical removal of TDLNs [14, 181], co-administration of FTY720, an inhibitor of T cell egress from lymphoid organs [175, 176, 181], was sufficient to negate the antitumour effects of ICB, and targeted delivery of ICB monoclonal antibodies to the TDLNs eclipsed systemic therapy in terms of efficacy and permitted dose sparing [179]. Furthermore, studies have consistently shown that the responses of TDLNs to ICB and the integrity of TLDNs as an immune reservoir and line of defence in general can be profoundly undermined by presence of metastases [141, 182,183,184,185]. In both melanoma murine models and human patients, LN metastases precipitated an altered immune contexture involving IFN-II-associated accumulation of regulatory T cells (TREGs) and PD-L1+ DCs that suppressed CD8+ T cells [183, 184]. These tolerogenic cells were also observed in LNs with HNSCC metastasis and, along with naïve/quiescent (CD45ROloPD-1loTCF-1hi) CD4+ T cells, formed the immunosuppressive niches encircling TPEX and TINT post-ICB [141]. Overall, these evidences offered an additional explanation to the prognostic values of TDLN statuses from the perspective of CD8+ T cell anti-tumour immunity, revealed the underappreciated role of TDLN statuses in predicting the response to ICB, and conceptualised the combination of ICB with agents targeting LN metastases, the latter of which, however, are yet to be developed.

Fig. 3
figure 3

Modifying CD8+ T response in cancer. (A) Immune checkpoint blockade radically changes the CD8+ T landscape via mobilisation of the stem-like populations, which primarily occurs in the company of supportive cells in the TDLNs and microenvironmental niches. Cancer cell death as a result of amplified response fosters a self-amplifying cycle owing to tumour antigen release and subsequent increased uptake and priming by APCs. ICBs can simultaneously stimulate TRM, which contributes to increased tumour killing and long-lasting immunosurveillance that deters recurrence and metastasis. (B) In addition to the conventional steps implicated in adoptive cell therapies (lymphocyte procurement, isolation, activation and expansion, pre-infusion lymphodepletion and post-infusion IL-2 administration), there have been multiple strategies in the phases of ex vivo manipulation and in vivo augmentation (marked with plus signs) to maximise the utility of transferred cells. A deeper understanding of the transcriptional, epigenetic, and metabolic controls of CD8+ T phenotypes will shed light on novel ways to engineer cells for the desirable attributes.

Overcoming the vascular endothelial barrier

Following potentiation of tumour-specific CD8+ T cells in the TDLNs, an endurance run through systemic circulation, tissue environment, and microvasculature awaits before they are able to reach the designated destination in the centre of tumour. Execution of such an extended process that requires extreme precision relies on coordination and fine-tuning of various homeostatic control systems. After priming, exit from the LNs is classically conducted with upregulation of S1PR1 in the context of high concentration of S1P in efferent lymphatics and synchronous curtailment of CCR7 and CD69 [6, 186], whereas sympathetic neural inputs typically promote retention of T cells in the LNs [187, 188]. Intact chemotactic systems are then a prerequisite for tumour homing, which include the CCR5-CCL5, CXCR3-CXCL9/CXCL10/CXCL11, CXCR5-CXCL13, CX3CR1-CX3CL1, and CXCR6-CXCL16 axes for CD8+ T cells [105, 109, 139, 189,190,191,192,193,194], where the transition from TPEX to TINT/TTEX may be accompanied by specific changes in expression of chemokine receptors [139, 177, 191]. Conversely, the elaborate interactions can be sabotaged or manipulated by solid tumours to diminish the driving force for cytotoxic T cell migration while fostering preferential recruitment of suppressive immune cells including TREGs, myeloid-derived suppressor cells (MDSCs), and tumour-associated macrophages (TAMs) and neutrophils (TANs) [189, 192]. The directionality of chemoattractant concentration gradient, which steers the CD8+ T cell traffic, also bears obvious significance as, for instance, the interplay between CXCR4+ TEFF and tumour-associated lymphatic vessel-derived CXCL12 resulted in marginalisation and egress of TILs, which undermines tumour control and therapeutic response [177].

Before CD8+ T cells could set foot on the core of the tumour, they can be repulsed by multiple chemical, physical, and cellular ramparts, and the tumour vasculature represents the first tumour-orchestrated line of defence that must be surmounted. In acute inflammation, leukocyte extravasation typically follows a multi-step progression, where friction for arresting cell locomotion is jointly supplied by selectins and adhesion molecules such as ICAMs and VCAM-1. Tumours resort to formation of blood vessels (termed ‘neo-angiogenesis’), which is stimulated by mechanical strain, hypoxia, and angiogenic molecules, with the VEGF family (VEGFA - F) being the most prominent, to fulfill the metabolic demands for neoplastic growths [195,196,197]. VEGF has the ability to directly influence immune cells, promoting development of TREGs, M2-macrophags, and MDSCs, impairing DC maturation and T cell development, and inducing CD8+ TIL exhaustion [195, 198,199,200,201]. When faced with relentless malignancy-driven bombardment by VEGF and other angiogenic factors, endothelial cells lining the tumour-associated blood vessels are prevented from activation by pro-inflammatory cytokines and conspicuously deviate from those in inflammation in terms of surface protein expression. Differentially expressed by neoangiogenic endothelium are immune checkpoint molecules [202, 203], FasL that selectively mediate CD8+ T cell demise [204], and, importantly, fewer adhesion molecules [205, 206], referred to as ‘endothelial cell anergy’, thereby veiling the portal of entry for tumour-specific CD8+ T cells. These observations serve as the mechanistic underpinning for combining anti-angiogenic agents with ICBs [194, 199, 201, 204, 207], where clinical trials have verified additive if not synergistic effects across several cancer types [208,209,210,211,212,213], which may be seen regardless of the intra-tumoural immune checkpoint molecule expression status [208, 211]. Notably, recent advances have demonstrated endothelial cells comprising the post-capillary venule (PCV)-derived MECA-79+ tumour-associated high endothelial venules (TA-HEV) as major breach sites for CD8+ T cells extravasation at baseline and in anti-CTLA-4 (+/- anti-PD-1) therapy, and increased number and maturation of these endothelial cells ushered a higher percentage of stem-like (SMALF6+) CD8+ TILs [214]. In preclinical models, anti-VEGFR2, anti-PD-L1 and LTβRAg triple therapy were sufficient to induce differentiation of PCVs into TA-HEVs that indeed facilitated lymphocyte infiltration and TPEX accumulation [215, 216].

Cancer-associated fibroblasts at the crux of CD8+ T-excluding and suppressing stroma

The tumour-associated stroma, apart from requitting the cancer-derived factors with direct facilitative and prohibitive influences on neoplastic cell-led tumour progression and metastatic transition, and thus holding cardinal non-immune-related biological and prognostic implications [196, 217,218,219], lies in between the extravasated CD8+ T cells and the nest of tumour cells, and contains various cellular and inanimate components with shielding effects. Being one of the most abundant and functionally significant constituents of tumour stroma, cancer-associated fibroblasts (CAFs) are evidently discernible from normal quiescent fibroblasts [218, 220, 221], commonly arise from co-option of resident fibroblasts by mechanical stress and cancer cell-secrete chemokines and growth factors (e.g., TGF-β, IL-1, IL-6, and PDGF), but are otherwise of remarkable plasticity (i.e., reactivity to external cues) and heterogeneity in origins and phenotypes and across tumour/tissue types, for which a unanimous classification system is lacking despite tentative, crude categorisation into myofibroblastic (myoCAFs, typically dependent on TGF-β for differentiation and ECM-associated genes/αSMAhigh), inflammatory (iCAFs, IL-1-dependent and IL-6/CXCL12high), and antigen-presenting CAFs (apCAFs, HLA-DR/MHC-IIhigh) [219,220,221,222,223,224,225,226].

Transcriptomic analyses have established the inverse correlations between myoCAF-associated signatures and abundance of CD8+ T cells as well as immunotherapy efficacy [219, 227,228,229]. Integration of spatial information allowed pinpointing of several myoCAF subsets in particular that mediate T cell repression and exclusion predominantly by deposition of extracellular matrix (ECM, a known trigger of CD8+ T cell exhaustion in cancer [230] and impediment to lymphocyte locomotion when densely packed or mal-aligned [231, 232]), as well as TGF-β secretion and interacting with other suppressive cell populations [221, 222, 227, 228, 233,234,235,236]. Costa et al. are among the first groups to portray myoCAFs at higher resolutions, where, using six fibroblast markers, they ascertained one particular myoCAF subset that tends to cluster near epithelial tumour cells in TNBCs and attracted, maintained, and differentiation into CD25highFOXP3high TREGs with augmented suppressive effects on CD8+ T cells [234]. Further deciphering endeavours followed thereafter. For example, in KPR mouse pancreatic adenocarcinomas, TGF-β-signalling-dependent LRRC15+ myoCAFs are shown to be in close proximity of and capable of suppressing CD8+ T cells [222]. In early- and advanced-stage lung cancers, MYH11+αSMA+ and FAP+αSMA+ myoCAFs have been respectively identified to obstruct T cell contact with tumour cells, albeit in different ways, where the former form a monolayer of elongated cells enveloping the tumour parenchyma and deposit type IV collagen fibres, and the latter are organised into patches or multiple layers and collagen XI and XII-producing instead [221].

In contrast, CAFs exhibiting an inflammatory or immune modulatory phenotype sculpt the immune cell landscape via extensive ligand-receptor interactions [224, 237]. CAF-emanated IL-6 recruits FOXP3+ TILs [235], invokes CD73 expression in γδTREGs [130], and induces differentiation of TME monocytes into MDSCs through STAT3 signalling [238]. Even though CAF production of CXCL12 can attract CD8+ T cells in infiltrated tumours [193], it oftentimes culminates in T cell exclusion [239, 240]. This could be the combinatory result of CD8+ T cells being misdirected towards the tumour margin [239,240,241], the paradoxical repelling effects of CXCL12 at high concentrations [242], and the CXCR4-CXCL12 apparatus-orchestrated CAF crosstalk with TREGs [234], SPP1+ TAMs [243], and other myeloid populations [244]. Moreover, Ma et al. have computationally derived from harmonised scRNA and ST data a multitude of ligand-receptor interactions through which iCAF can directly corrupt CD8+ T cell robustness [224]. Notably, the LGALS1-PTPRC interaction is among the most frequently seen on ST slices, which triggers NFAT signalling and thus CD8+ T cell exhaustion [224]. On the other hand, apCAFs may render anergic or convert naïve CD4+ T cells to TREGs with antigen-specific MHC-II-TCR crosslinking void of backing from co-stimulatory molecules [226, 245], and direct antigen-dependent interaction with CAFs can precipitate PD-L2 and FasL-mediated lethality to the antigen-specific CD8+ T cells [246]. That being said, in lung cancers, apCAFs are found to activate and protect CD4+ T cells from apoptosis and consequently tumour-suppressive rather than permissive [247]. Additionally, non-canonical markers have also proven to be useful for setting apart non-ternary CAF subsets with immunomodulatory effects. A distinct cluster of CAFs has been discovered in HNSCC that sequester TPEX (TCF1+GZMK+) by enhanced MHC-I and CXCLs expressions and impede their effector transformation through leveraging galectin-9 [240]. In pancreatic ductal adenomas (PDACs), CD105 effectively partitioned CAFs into two fractions displaying opposite correlations with immune cell subpopulations, with the CD105 subsets being positively associated with proliferation of the CD8+ subsets, whilst the CD105+ counterparts had a negative association [248]. Specifically, the CD105MHCII+CD74+ CAFs were the sole mesenchymal subset securing a positive correlation with antigen-experienced but non-terminally exhausted CD8+ TILs [248].

Considering the myriad of ways CD8+ T cells are obstructed and subjugated, therapies targeting CAFs have been extensively investigated, and combination with therapeutic strategies carrying direct empowering effects on CD8+ T cells (e.g., ICBs and CAR-T therapy) attempted as a natural course of action for the prospect of “infiltrate and eliminate“ [233, 239, 242]. As an embodiment of this approach, antecedent treatment with CAR-T targeting FAP+ CAFs in a recent study on murine PDAC models dismantled the desmoplastic stroma and dispersed the associated immunosuppressive environment, allowing full engagement of ICB-reprogrammable endogenous CD8+ T cells and CAR-T cells targeting tumour-associated antigen (Meso-CAR-T) with tumour cells [249]. Nonetheless, given the phenotypic multiplicity of CAFs and their restraining effects on tumours [226, 247, 250, 251], indiscriminate ablation of CAFs may accelerate cancer progression even with increased CD8+ T cell infiltration [250], hence the imperative to selectively deplete the reprehensible CAF subsets for clear-cut clinical benefits.

Seeking sanctuary in the immune-permissive microenvironmental enclaves

Once advanced to the immediate vicinity of tumour cells, CD8+ TILs plunge into the hypoxic, acidified, adenosine and potassium ion-imbued, and nutrient-deficient or perturbed quagmire, which overwhelmingly favours CD8+ T dysfunction and sealing of the exhaustion fate [252, 253]. In accordance with this, intra- or peri-tumoural TPEX are rarely found flanked by tumour cells unbuffered, but tend to congregate in spatial niches conducive for population maintenance and effector differentiation [76, 106, 143, 174, 190, 191, 216, 254,255,256], and in some cancer types, the density and quality of these structures may set patients with favourable outcomes apart from those with poor prognosis [106, 257], and ICB responders from non-responders in the scenario of intact T cell priming in TDLNs and unhindered infiltration [143]. Cells with both antigen-presenting and immunomodulatory capabilities, especially DCs, are almost invariably implicated in the organisation of these CD8+ T-accommodating alcoves [174, 190, 191, 257,258,259]. DCs close the distance with the inbound CD8+ T cells through specific interactions between chemokines and their receptors (e.g., CXCL16-CXCR6 [191] and CXCL9/CXCL10-CXCR3 [190, 194, 257]), granting the primary and co-stimulatory licences [169, 174, 255, 260], and unleash cytokines crucial for survival and induction of effector functions, such as IL-12 [261] and IL-15 [143, 191]. Prostaglandin E2 [262] and immune checkpoint molecules like TIM-3 [258] and PD-L1 [259] are notable for interfering with the DC-CD8+ TIL communication, thus explaining at least part of the benefits seen in respective therapies. Spatial profiling studies have also substantiated that DCs can complex with the CD4+ TH subsets they primed to form dyadic powerhouses that exert strengthened chemoattractant and invigorating influences on CD8+ T cells [143, 169, 263].

Tertiary lymphoid structures (TLSs) are the most sophisticated form of ectopic lymphoid tissues that can be found in tumours. Representing the pinnacle of immune cell reciprocity within the TME, TLSs are structurally less organised (i.e., unencapsulated and variable in T and B cell compartmentalisation) but cellular composition-wise reminiscent of SLOs, where each type of cells undertake corresponding roles that ultimately contribute to a brisk and self-replenishing local adaptive response [264], as tumours with TLSs had denser and less dysfunctional intra-tumoural T cell infiltrates [254, 265,266,267]. Like in SLOs, presence of HEVs [216, 268] and a stromal network primarily consisting of follicular reticular cells (FRCs) [264] in TLSs secures the microanatomical space for lymphocyte passage and residence. The TLS-defining CD20+ B cells can be intermingled with T cells or gather in follicles with or without germinal centres, and nurtured by the CD4+ follicular helper or CD21+ follicular dendritic cells [264]. In addition to the canonical role of producing antibodies, TLS B cell subsets have been reported to be capable of energizing T lymphocytes by antigen presentation and cytokine secretion [266, 269,270,271]. In the meantime, the CD4+ TH and DC duo have been found enriched in TLSs, predominantly in the T cell zones [254, 263], and so have tumour-reactive and stem-like CD8+ T cells [76, 106, 256, 265, 272], which integrate the inputs from the neighbouring immune cells and proceed to tackle the tumour cells.

TLSs are generally associated with improved survival outcomes and immunotherapy responses [265, 266, 269, 270], and yet, on scrutinisation, the localisation and maturity level of TLSs wield pivotal influences [267, 273], where extra-tumoural positioning and immaturity may underpin the negative prognostic impacts of TLSs in clear cell renal cell carcinoma [273, 274]. TLS formation beholds a choreography of lymphocytes, myeloid cells and mesenchymal cells under the guidance of molecular factors including lymphoid chemokines (e.g., CXCL13, CCL19, and CCL21), LTs, IFN-Is, TNFs, IL-17, many of which have been appropriated to experimentally induce TLSs in murine models [264]. CXCL13+CD8+ T cells have been described, were determined to be tumour-reactive and predictive of ICB response [275], and indeed correlated with TLS presence [272, 276], though it was reported that their dysfunctional states and contribution to an immunosuppressive milieu might ultimately lead to unfavourable outcomes [276].

Perspectives

The notion that not all CD8+ TILs are created equal stands truer than ever. An outpouring of multi-dimensional and high-resolution data over the past few years spotlighted the stem-like exhausted and memory cells as the subsets of the highest tumour-suppressive yield at baseline and upon therapeutic intervention. Their transcriptomic and epigenetic properties are being characterised and phylogenetics elucidated, to a level of certainty, precision, and scale previously unfathomable, revealing invaluable insights into the promising biomarkers for prognostication, subpopulations for successful adoptive transfer, and strategies for engineering CAR-T cells (Fig. 3B).

Innovative and painstaking iterative processes with state-of-the-art technologies paved the way for these ground-breaking discoveries. Recent implementation of in vivo sc-CRISPR screening to TFs concerning CD8+ T cell differentiation and exhaustion in cancer may have just constructed the most comprehensive TF directory for T cell fate to date [142]. Synergising available omics modalities represents an alternative tactic for new discoveries. Mapping and deconvolution algorithms are routinely applied to scRNA-seq and ST data to confer spatial coordinates to individual cells and deduce the cellular composition of each ST spot [5, 224, 236, 240]. Building on the previously known markers for tumour specificity, which are frequently also tokens of exhaustion (e.g., CD39, PD-1, TIM-3 and TOX) [4, 18, 68, 277], coupling of scRNA-seq, sc-TCR-seq, and TCR specificity testing data helped to fish out the tumour-reactive stem-like CD8+ T cells from the swarm of bystanders, which are otherwise difficult to tell apart due to transcriptomic similarity [18, 56, 127, 275, 278]. Disciplined application of these investigative tools to the matched longitudinal samples enabled a chronicle of the tumour-reactive clonotype dynamics in naturally occurring anti-tumour immunity and immunotherapies [18, 56, 275], laying down the ground truth against which computational inferences can be compared, and the methods refined. Furthermore, the recent invention of Zman-seq, which combines scRNA-seq with time-stamping by sequential injections of fluorescent anti-CD45 antibodies that selectively tag circulating immune cells but not those already infiltrated the tumour, allows cataloguing of transcriptomic states with different durations of TME residence and consequently deciphering of the temporal dynamics [279]. Much to the excitement of the academia and the medical community, the methodological revolution seems to have provided the necessary means to materialise the envisioned all-encompassing cell tree for CD8+ T cells in health and disease [7]. The much-anticipated nomenclatural reform, however, has lagged behind, and work still needs to be done to mend the gap between the newly acquired high-dimensional information and legacy observations from the pre-omics era. Besides, caution should be exercised when interpreting data from the viral models, due to the overlapping but nuanced phenotypes [56, 81, 84] and asynchronous pace of exhaustion [86] between the exhausted CD8+ T subsets in chronic viral infections and those in tumours.

That key steps of CD8+ T cell priming and maturation take place across anatomical locations outside of the tumour mandates a cross-the-body relay in parallel with the phenotypic evolution of tumour-specific CD8+ T cells, which is an extremely error-prone process. Any misstep could end up with flawed immunotypes that render tumour cells inaccessible to the tumouricidal arsenal of CD8+ T cells, which operate exclusively in close quarters. Conversely, drawing on the continually expanding mechanistic insights, each nucleating point of CD8+ T anti-tumour immunity, be it the TDLNs, APC-rich niches, or TLSs, may represent an opportunity for therapeutic amplification, and the vascular and stromal barriers for ablation, especially in the ‘excluded’ immunotype. Similarly, the phenotypic pliability of CD8+ T cells, whilst hijacked by TME factors for exhaustion induction, is a double-edged sword malleable to therapeutic honing in recognised and less obvious ways. In broader terms, CD8+ T cells are inscribed with marks of systemic immunological wellbeing, which is dictated by age, sex, hormone levels, antigen exposure history, and comorbidities of individuals, but also newly established determinants such as commensal microbiota. An elegant testament bolstering the linkage is the recent discovery of PD-1 blockade efficacy enhancement by gut L. johnsonii and C. sporogenes, which collaborate to convert tryptophan to IPA, a metabolite that promotes H3K27 acetylation of the super-enhancer region of Tcf7, thereby diverting CD8+ T cells towards differentiation into TPEX [280]. The impacts of systemic immune health factors on CD8+ T-mediated anti-tumour immunity are therefore also worthy of further investigation, from which individualised and easy-to-implement immune-boosting diet and lifestyle interventions can be devised.

In conclusion, CD8+ T cells are indispensable for executing adaptive antineoplastic response, as they are positioned at the converging point downstream of miscellaneous immunomodulatory pathways to administer the coup de grâce to tumorous cells by unleashing direct cytotoxicity. The vast phenotypic and spatial heterogeneity of CD8+ TILs are being unveiled as we venture forth into the age of omics. Expanding on the preceding efforts to activate, reprogramme, and facilitate the infiltration of tumour-specific CD8+ T cells, the first-of-its-kind FDA approval for TIL infusion therapy in melanoma has officially advanced the forefront of immeasurable possibilities into clinical practice [281]. Working towards a consensus ontogeny to synthesise the multimodal data and delving into the mechanistic underpinnings will help to maximise the yield of CD8+ TIL-based cancer therapies, ultimately translating into better outcomes for patients.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

αSMA:

alpha smooth muscle actin

ACAT-seq:

Assay for Transposase Accessible Chromatin with high-throughput sequencing

AHR:

aryl hydrocarbon receptor

AP-1:

activator protein 1

APCs:

antigen-presenting cells

BACH2:

BTB and CNC homologue 2

BATF:

Basic Leucine Zipper ATF-Like Transcription Factor

BCL-6:

B-Cell Lymphoma 6

BLIMP-1:

B-lymphocyte-induced maturation protein 1

CAFs:

cancer-associated fibroblasts (myoCAFs – myofibroblastic CAFs; iCAFs – inflammatory CAFs; apCAFs – antigen-presenting CAFs)

CAR:

chimeric antigen receptor

CCL/R:

C-C motif chemokine ligand/receptor

cDC1/2s:

type I/II conventional dendritic cells

CRISPR:

clustered regularly interspaced short palindromic repeats

C. sporogenes:

Clostridium sporogenes

CTLA4:

Cytotoxic T-Lymphocyte Associated Protein 4

CX3CL/R:

C-X3-C motif chemokine ligand/receptor

CXCL/R:

C-X-C motif ligand/receptor

DCs:

dendritic cells

ECM:

extracellular matrix

Egr1/2:

early growth response 1/2

ENTPD1:

ectonucleoside triphosphate diphosphohydrolase-1

EOMES:

Eomesodermin

FAP:

fibroblast activation protein

FasL:

Fas ligand

FOXO1:

forkhead box protein O1

FOXP1/3:

forkhead box protein P1/3

FRCs:

follicular reticular cells

GZMB/H/K:

granzyme B/H/K

H3K27:

histone H3 lysine 27

Havcr2:

Hepatitis A Virus Cellular Receptor 2

Hic1:

hypermethylated in cancer 1

Hif1a:

hypoxia inducible factor 1 subunit alpha

HLA-DR:

human leukocyte antigen, DR isotype

HMGB2:

high mobility group box 2

HNSCC:

head and neck squamous cell carcinoma

HOBIT:

homolog of Blimp-1 in T cells

ICAM:

intracellular adhesion molecule 1

ICB:

immune checkpoint blockade

ID2/3:

inhibitor of DNA binding 2/3

IFN-I/II:

type I/II interferons

IFN-γ/IFNG:

interferon gamma

Ikzf1:

IKAROS family zinc finger 1

IL:

interleukin

IL7R:

interleukin 7 receptor

IRF4:

interferon regulatory factor 4

IPA:

indole-3-propionic acid

ISG:

interferon-stimulated genes

ITGAE:

integrin subunit alpha E

KLF:

Kruppel-like factors

Lag3:

lymphocyte-activation gene 3

LAYN:

Layilin

LEC:

lymphatic endothelial cell

LGALS1:

galectin 1

L. johnsonii:

Lactobacillus johnsonii

LRRC15:

leucine rich repeat containing 15

LTβRAg:

lymphotoxin beta receptor agonist

LTs:

leukotrienes

MAPK:

mitogen-Activated Protein Kinase 1

MDSCs:

myeloid-derived suppressor cells

MEK:

also known as MAPKK (mitogen-activated protein kinase kinase)

MHC-I/II:

major histocompatibility complex I/II

MKI67:

marker of proliferation Ki-67

mTOR:

mammalian target of rapamycin kinase

MYH11:

myosin heavy chain 11

NFAT:

nuclear factor of activated T-cells

NK:

natural killer (cells)

NLT:

non-lymphoid tissue

NOTCH:

neurogenic locus notch homolog protein 1

Nr4a:

nuclear receptor subfamily 4 A

OVCAR-3:

(NIH) ovarian carcinoma cell line 3

PCV:

post-capillary venule

PD-1:

programmed death-1

PDACs:

pancreatic ductal adenomas

Pdcd1:

programmed cell death 1

PDGF:

platelet-derived growth factor

PD-L1/2:

programmed death-ligand 1

PRF1:

perforin 1

PTPRC:

protein tyrosine phosphatase receptor type C

RUNX3:

runt-related transcription factor 3

S1PR1:

sphingosine 1-phosphate receptor-1

scRNA-seq:

single-cell RNA sequencing

SCS:

subscapular sinuses

SLAMF6:

SLAM family member 6

SLOs:

secondary lymphoid organs

SPP1:

secreted phosphoprotein 1

ST:

spatial transcriptomic

STAT:

signal transducer and activator of transcription

TA-HEV:

tumour-associated high endothelial venules

TAM/Ns:

tumour-associated macrophages/neutrophils

TCF1:

transcription factor 1

TCM :

central memory cells

TCR-seq:

T cell receptor sequencing

TDLNs:

tumour-draining lymph nodes

TEFF :

effector CD8+ T cells

TEM :

effector memory cells

TEMRA :

effector memory re-expressing CD45RA (terminally differentiated effector memory) CD8+ T cells

TEX :

exhausted CD8+ T cells

TEX EFF :

effector-like exhausted

TFs:

transcription factors

TGF-β:

transforming growth factor beta

TH :

CD4+ helper T cells

TIGIT:

T cell immunoreceptor with Ig snd ITIM domains

TILs:

tumour infiltrating lymphocytes

TIM3:

T cell immunoglobulin mucin 3

TINT :

intermediate-exhausted phenotype

TLSs:

tertiary lymphoid structures

TME:

tumour microenvironment

TMEM :

memory CD8+ T cells

TN :

naïve CD8+ T cells

TNF-α:

tumour necrosis factor alpha

TOX:

thymocyte selection-associated HMG box

TPEX :

progenitor exhausted T cells

TREG :

(CD4+) regulatory T cells

TRM :

tissue-resident memory T cells

TSCM :

stem cell memory T cells

TTEX :

terminally exhausted CD8+ T cells

WNT:

wingless-related integration site (signalling pathway)

VCAM:

vascular cell adhesion molecule

VEGF(R):

vascular endothelial growth factor (receptor)

VIM:

vimentin

XCL1:

X-C motif chemokine ligand 1

ZEB2:

zinc finger E-box binding homeobox 2

ZNF683:

zinc finger protein 683

References

  1. Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther. 2023;8:235.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Bruni D, Angell HK, Galon J. The immune contexture and immunoscore in cancer prognosis and therapeutic efficacy. Nat Rev Cancer. 2020;20:662–80.

    Article  PubMed  CAS  Google Scholar 

  3. Brummel K, Eerkens AL, de Bruyn M, Nijman HW. Tumour-infiltrating lymphocytes: from prognosis to treatment selection. Br J Cancer. 2023;128:451–8.

    Article  PubMed  CAS  Google Scholar 

  4. Zheng L, Qin S, Si W, Wang A, Xing B, Gao R, et al. Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science. 2021;374:abe6474.

    Article  PubMed  Google Scholar 

  5. Rao A, Barkley D, França GS, Yanai I. Exploring tissue architecture using spatial transcriptomics. Nature. 2021;596:211–20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Schenkel JM, Pauken KE. Localization, tissue biology and T cell state — implications for cancer immunotherapy. Nat Rev Immunol. 2023;23:807–23.

    Article  PubMed  CAS  Google Scholar 

  7. Domcke S, Shendure J. A reference cell tree will serve science better than a reference cell atlas. Cell. 2023;186:1103–14.

    Article  PubMed  CAS  Google Scholar 

  8. van den Broek T, Borghans JAM, van Wijk F. The full spectrum of human naive T cells. Nat Rev Immunol. 2018;18:363–73.

    Article  PubMed  Google Scholar 

  9. Philip M, Schietinger A. CD8+ T cell differentiation and dysfunction in cancer. Nat Rev Immunol. 2022;22:209–23.

    Article  PubMed  CAS  Google Scholar 

  10. Naulaerts S, Datsi A, Borras DM, Antoranz Martinez A, Messiaen J, Vanmeerbeek I, et al. Multiomics and spatial mapping characterizes human CD8+ T cell states in cancer. Sci Transl Med. 2023;15:eadd1016.

    Article  PubMed  CAS  Google Scholar 

  11. Van Der Byl W, Nüssing S, Peters TJ, Ahn A, Li H, Ledergor G, et al. The CD8+ T cell tolerance checkpoint triggers a distinct differentiation state defined by protein translation defects. Immunity. 2024;57:1324–44. e8.

    Article  PubMed  CAS  Google Scholar 

  12. Chang JT, Wherry EJ, Goldrath AW. Molecular regulation of effector and memory T cell differentiation. Nat Immunol. 2014;15:1104–15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Zhang N, Hartig H, Dzhagalov I, Draper D, He YW. The role of apoptosis in the development and function of T lymphocytes. Cell Res. 2005;15:749–69.

    Article  PubMed  CAS  Google Scholar 

  14. Huang Q, Wu X, Wang Z, Chen X, Wang L, Lu Y, et al. The primordial differentiation of tumor-specific memory CD8+ T cells as bona fide responders to PD-1/PD-L1 blockade in draining lymph nodes. Cell. 2022;185:4049–66. e25.

    Article  PubMed  CAS  Google Scholar 

  15. Li Z, Tuong ZK, Dean I, Willis C, Gaspal F, Fiancette R et al. In vivo labeling reveals continuous trafficking of TCF-1+ T cells between tumor and lymphoid tissue. J Exp Med. 2022;219.

  16. Gebhardt T, Park SL, Parish IA. Stem-like exhausted and memory CD8 + T cells in cancer. Nat Rev Cancer. 2023.

  17. Andreatta M, Corria-Osorio J, Müller S, Cubas R, Coukos G, Carmona SJ. Interpretation of T cell states from single-cell transcriptomics data using reference atlases. Nat Commun. 2021;12:2965.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Oliveira G, Stromhaug K, Klaeger S, Kula T, Frederick DT, Le PM, et al. Phenotype, specificity and avidity of antitumour CD8+ T cells in melanoma. Nature. 2021;596:119–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Guo X, Zhang Y, Zheng L, Zheng C, Song J, Zhang Q, et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat Med. 2018;24:978–85.

    Article  PubMed  CAS  Google Scholar 

  20. Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401:708–12.

    Article  PubMed  CAS  Google Scholar 

  21. Lin WW, Nish SA, Yen B, Chen YH, Adams WC, Kratchmarov R, et al. CD8+ T lymphocyte Self-Renewal during Effector Cell determination. Cell Rep. 2016;17:1773–82.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Pais Ferreira D, Silva JG, Wyss T, Fuertes Marraco SA, Scarpellino L, Charmoy M, et al. Central memory CD8+ T cells derive from stem-like Tcf7hi effector cells in the absence of cytotoxic differentiation. Immunity. 2020;53:985–e100011.

    Article  PubMed  CAS  Google Scholar 

  23. Bresser K, Kok L, Swain AC, King LA, Jacobs L, Weber TS, et al. Replicative history marks transcriptional and functional disparity in the CD8+ T cell memory pool. Nat Immunol. 2022;23:791–801.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Graef P, Buchholz VR, Stemberger C, Flossdorf M, Henkel L, Schiemann M, et al. Serial transfer of single-cell-derived immunocompetence reveals stemness of CD8+ central memory T cells. Immunity. 2014;41:116–26.

    Article  PubMed  CAS  Google Scholar 

  25. Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol. 2004;22:745–63.

    Article  PubMed  CAS  Google Scholar 

  26. Gattinoni L, Lugli E, Ji Y, Pos Z, Paulos CM, Quigley MF, et al. A human memory T cell subset with stem cell-like properties. Nat Med. 2011;17:1290–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Cieri N, Camisa B, Cocchiarella F, Forcato M, Oliveira G, Provasi E, et al. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood. 2013;121:573–84.

    Article  PubMed  CAS  Google Scholar 

  28. Roberto A, Castagna L, Zanon V, Bramanti S, Crocchiolo R, McLaren JE, et al. Role of naive-derived T memory stem cells in T-cell reconstitution following allogeneic transplantation. Blood. 2015;125:2855–64.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Gattinoni L, Zhong XS, Palmer DC, Ji Y, Hinrichs CS, Yu Z, et al. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat Med. 2009;15:808–13.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Verma V, Jafarzadeh N, Boi S, Kundu S, Jiang Z, Fan Y, et al. MEK inhibition reprograms CD8+ T lymphocytes into memory stem cells with potent antitumor effects. Nat Immunol. 2021;22:53–66.

    Article  PubMed  CAS  Google Scholar 

  31. Galletti G, De Simone G, Mazza EMC, Puccio S, Mezzanotte C, Bi TM, et al. Two subsets of stem-like CD8+ memory T cell progenitors with distinct fate commitments in humans. Nat Immunol. 2020;21:1552–62.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Zheng C, Zheng L, Yoo J-K, Guo H, Zhang Y, Guo X, et al. Landscape of infiltrating T cells in Liver Cancer revealed by single-cell sequencing. Cell. 2017;169:1342–e5616.

    Article  PubMed  CAS  Google Scholar 

  33. Zhang L, Yu X, Zheng L, Zhang Y, Li Y, Fang Q, et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature. 2018;564:268–72.

    Article  PubMed  CAS  Google Scholar 

  34. Sabatino M, Hu J, Sommariva M, Gautam S, Fellowes V, Hocker JD, et al. Generation of clinical-grade CD19-specific CAR-modified CD8 + memory stem cells for the treatment of human B-cell malignancies. Blood. 2016;128:519–28.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Arcangeli S, Bove C, Mezzanotte C, Camisa B, Falcone L, Manfredi F et al. CAR T cell manufacturing from naive/stem memory T lymphocytes enhances antitumor responses while curtailing cytokine release syndrome. J Clin Invest. 2022;132.

  36. Hurton LV, Singh H, Najjar AM, Switzer KC, Mi T, Maiti S, et al. Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells. Proc Natl Acad Sci U S A. 2016;113:E7788–97.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Meyran D, Zhu JJ, Butler J, Tantalo D, MacDonald S, Nguyen TN, et al. TSTEM-like CAR-T cells exhibit improved persistence and tumor control compared with conventional CAR-T cells in preclinical models. Sci Transl Med. 2023;15:eabk1900.

    Article  PubMed  CAS  Google Scholar 

  38. Adachi K, Kano Y, Nagai T, Okuyama N, Sakoda Y, Tamada K. IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor. Nat Biotechnol. 2018;36:346–51.

    Article  PubMed  CAS  Google Scholar 

  39. Masopust D, Vezys V, Marzo AL, Lefrançois L. Preferential localization of effector memory cells in nonlymphoid tissue. Science. 2001;291:2413–7.

    Article  PubMed  CAS  Google Scholar 

  40. Gebhardt T, Wakim LM, Eidsmo L, Reading PC, Heath WR, Carbone FR. Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat Immunol. 2009;10:524–30.

    Article  PubMed  CAS  Google Scholar 

  41. Masopust D, Choo D, Vezys V, Wherry EJ, Duraiswamy J, Akondy R, et al. Dynamic T cell migration program provides resident memory within intestinal epithelium. J Exp Med. 2010;207:553–64.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Yenyuwadee S, Sanchez-Trincado Lopez JL, Shah R, Rosato PC, Boussiotis VA. The evolving role of tissue-resident memory T cells in infections and cancer. Sci Adv. 2022;8:eabo5871.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Buggert M, Price DA, Mackay LK, Betts MR. Human circulating and tissue-resident memory CD8 + T cells. Nat Immunol. 2023;24:1076–86.

    Article  PubMed  CAS  Google Scholar 

  44. Poon MML, Caron DP, Wang Z, Wells SB, Chen D, Meng W, et al. Tissue adaptation and clonal segregation of human memory T cells in barrier sites. Nat Immunol. 2023;24:309–19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Szabo PA, Levitin HM, Miron M, Snyder ME, Senda T, Yuan J, et al. Single-cell transcriptomics of human T cells reveals tissue and activation signatures in health and disease. Nat Commun. 2019;10:4706.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Mackay LK, Minnich M, Kragten NA, Liao Y, Nota B, Seillet C, et al. Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science. 2016;352:459–63.

    Article  PubMed  CAS  Google Scholar 

  47. Milner JJ, Toma C, Yu B, Zhang K, Omilusik K, Phan AT, et al. Runx3 programs CD8+ T cell residency in non-lymphoid tissues and tumours. Nature. 2017;552:253–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Hombrink P, Helbig C, Backer RA, Piet B, Oja AE, Stark R, et al. Programs for the persistence, vigilance and control of human CD8 + lung-resident memory T cells. Nat Immunol. 2016;17:1467–78.

    Article  PubMed  CAS  Google Scholar 

  49. Skon CN, Lee JY, Anderson KG, Masopust D, Hogquist KA, Jameson SC. Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat Immunol. 2013;14:1285–93.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Liikanen I, Lauhan C, Quon S, Omilusik K, Phan AT, Bartrolí LB et al. Hypoxia-inducible factor activity promotes antitumor effector function and tissue residency by CD8+ T cells. J Clin Invest. 2021;131.

  51. Corgnac S, Malenica I, Mezquita L, Auclin E, Voilin E, Kacher J, et al. CD103+CD8+ TRM cells accumulate in tumors of Anti-PD-1-Responder Lung Cancer patients and are tumor-reactive lymphocytes enriched with Tc17. Cell Rep Med. 2020;1:100127.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Crowl JT, Heeg M, Ferry A, Milner JJ, Omilusik KD, Toma C, et al. Tissue-resident memory CD8+ T cells possess unique transcriptional, epigenetic and functional adaptations to different tissue environments. Nat Immunol. 2022;23:1121–31.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Malik BT, Byrne KT, Vella JL, Zhang P, Shabaneh TB, Steinberg SM et al. Resident memory T cells in the skin mediate durable immunity to melanoma. Sci Immunol. 2017;2.

  54. Park SL, Buzzai A, Rautela J, Hor JL, Hochheiser K, Effern M, et al. Tissue-resident memory CD8+ T cells promote melanoma–immune equilibrium in skin. Nature. 2019;565:366–71.

    Article  PubMed  CAS  Google Scholar 

  55. Virassamy B, Caramia F, Savas P, Sant S, Wang J, Christo SN, 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–e6018.

    Article  PubMed  CAS  Google Scholar 

  56. Caushi JX, Zhang J, Ji Z, Vaghasia A, Zhang B, Hsiue EH, et al. Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature. 2021;596:126–32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Christian LS, Wang L, Lim B, Deng D, Wu H, Wang XF, Li QJ. Resident memory T cells in tumor-distant tissues fortify against metastasis formation. Cell Rep. 2021;35:109118.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Molodtsov AK, Khatwani N, Vella JL, Lewis KA, Zhao Y, Han J et al. Resident memory CD8+ T cells in regional lymph nodes mediate immunity to metastatic melanoma. Immunity. 2021;54:2117-32.e7.

  59. Han J, Zhao Y, Shirai K, Molodtsov A, Kolling FW, Fisher JL, et al. Resident and circulating memory T cells persist for years in melanoma patients with durable responses to immunotherapy. Nat Cancer. 2021;2:300–11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Weeden CE, Gayevskiy V, Marceaux C, Batey D, Tan T, Yokote K et al. Early immune pressure initiated by tissue-resident memory T cells sculpts tumor evolution in non-small cell lung cancer. Cancer Cell. 2023;41:837 – 52.e6.

  61. Corgnac S, Damei I, Gros G, Caidi A, Terry S, Chouaib S et al. Cancer stem-like cells evade CD8+CD103+ tumor-resident memory T (TRM) lymphocytes by initiating an epithelial-to-mesenchymal transition program in a human lung tumor model. J Immunother Cancer. 2022;10.

  62. O’Donnell JS, Teng MWL, Smyth MJ. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat Reviews Clin Oncol. 2019;16:151–67.

    Article  Google Scholar 

  63. Savas P, Virassamy B, Ye C, Salim A, Mintoff CP, Caramia F, et al. Single-cell profiling of breast cancer T cells reveals a tissue-resident memory subset associated with improved prognosis. Nat Med. 2018;24:986–93.

    Article  PubMed  CAS  Google Scholar 

  64. 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. 2014;20:434–44.

    Article  PubMed  CAS  Google Scholar 

  65. Djenidi F, Adam J, Goubar A, Durgeau A, Meurice G, de Montpréville V, 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. 2015;194:3475–86.

    Article  PubMed  CAS  Google Scholar 

  66. Jaiswal A, Verma A, Dannenfelser R, Melssen M, Tirosh I, Izar B, et al. An activation to memory differentiation trajectory of tumor-infiltrating lymphocytes informs metastatic melanoma outcomes. Cancer Cell. 2022;40:524–e445.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Edwards J, Wilmott JS, Madore J, Gide TN, Quek C, Tasker A, et al. CD103+ tumor-resident CD8+ T cells are Associated with Improved Survival in Immunotherapy-Naïve Melanoma patients and Expand significantly during Anti-PD-1 treatment. Clin Cancer Res. 2018;24:3036–45.

    Article  PubMed  CAS  Google Scholar 

  68. Duhen T, Duhen R, Montler R, Moses J, Moudgil T, de Miranda NF, et al. Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat Commun. 2018;9:2724.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Ganesan AP, Clarke J, Wood O, Garrido-Martin EM, Chee SJ, Mellows T, et al. Tissue-resident memory features are linked to the magnitude of cytotoxic T cell responses in human lung cancer. Nat Immunol. 2017;18:940–50.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Anadon CM, Yu X, Hänggi K, Biswas S, Chaurio RA, Martin A et al. Ovarian cancer immunogenicity is governed by a narrow subset of progenitor tissue-resident memory T cells. Cancer Cell. 2022;40:545 – 57.e13.

  71. Barsch M, Salié H, Schlaak AE, Zhang Z, Hess M, Mayer LS, et al. T-cell exhaustion and residency dynamics inform clinical outcomes in hepatocellular carcinoma. J Hepatol. 2022;77:397–409.

    Article  PubMed  CAS  Google Scholar 

  72. Gueguen P, Metoikidou C, Dupic T, Lawand M, Goudot C, Baulande S et al. Contribution of resident and circulating precursors to tumor-infiltrating CD8+ T cell populations in lung cancer. Sci Immunol. 2021;6.

  73. Krishna C, DiNatale RG, Kuo F, Srivastava RM, Vuong L, Chowell D et al. Single-cell sequencing links multiregional immune landscapes and tissue-resident T cells in ccRCC to tumor topology and therapy efficacy. Cancer Cell. 2021;39:662 – 77.e6.

  74. Luoma AM, Suo S, Wang Y, Gunasti L, Porter CBM, Nabilsi N et al. Tissue-resident memory and circulating T cells are early responders to pre-surgical cancer immunotherapy. Cell. 2022;185:2918-35.e29.

  75. Gavil NV, Scott MC, Weyu E, Smith OC, O’Flanagan SD, Wijeyesinghe S, et al. Chronic antigen in solid tumors drives a distinct program of T cell residence. Sci Immunol. 2023;8:eadd5976.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Siddiqui I, Schaeuble K, Chennupati V, Fuertes Marraco SA, Calderon-Copete S, Pais Ferreira D, et al. Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like Properties promote Tumor Control in Response to Vaccination and Checkpoint Blockade Immunotherapy. Immunity. 2019;50:195–e21110.

    Article  PubMed  CAS  Google Scholar 

  77. Wu J, Madi A, Mieg A, Hotz-Wagenblatt A, Weisshaar N, Ma S, et al. T cell factor 1 suppresses CD103+ lung tissue-Resident memory T cell development. Cell Rep. 2020;31:107484.

    Article  PubMed  CAS  Google Scholar 

  78. Zehn D, Thimme R, Lugli E, de Almeida GP, Oxenius A. Stem-like’ precursors are the fount to sustain persistent CD8+ T cell responses. Nat Immunol. 2022;23:836–47.

    Article  PubMed  CAS  Google Scholar 

  79. Lan X, Mi T, Alli S, Guy C, Djekidel MN, Liu X, et al. Antitumor progenitor exhausted CD8+ T cells are sustained by TCR engagement. Nat Immunol. 2024;25:1046–58.

    Article  PubMed  CAS  Google Scholar 

  80. Shakiba M, Zumbo P, Espinosa-Carrasco G, Menocal L, Dündar F, Carson SE et al. TCR signal strength defines distinct mechanisms of T cell dysfunction and cancer evasion. J Exp Med. 2022;219.

  81. Daniel B, Yost KE, Hsiung S, Sandor K, Xia Y, Qi Y, et al. Divergent clonal differentiation trajectories of T cell exhaustion. Nat Immunol. 2022;23:1614–27.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. + progenitor CD8 T cell phenotypes in tumors. Cell. 2021;184:4996–5014.e26.

  83. Schietinger A, Philip M, Krisnawan VE, Chiu EY, Delrow JJ, Basom RS, et al. Tumor-specific T cell dysfunction is a dynamic Antigen-Driven differentiation program initiated early during Tumorigenesis. Immunity. 2016;45:389–401.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Pritykin Y, van der Veeken J, Pine AR, Zhong Y, Sahin M, Mazutis L, et al. A unified atlas of CD8 T cell dysfunctional states in cancer and infection. Mol Cell. 2021;81:2477–e9310.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Angelosanto JM, Wherry EJ. Transcription factor regulation of CD8+ T-cell memory and exhaustion. Immunol Rev. 2010;236:167–75.

    Article  PubMed  CAS  Google Scholar 

  86. Rudloff MW, Zumbo P, Favret NR, Roetman JJ, Detrés Román CR, Erwin MM, et al. Hallmarks of CD8+ T cell dysfunction are established within hours of tumor antigen encounter before cell division. Nat Immunol. 2023;24:1527–39.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Scott AC, Dündar F, Zumbo P, Chandran SS, Klebanoff CA, Shakiba M, et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature. 2019;571:270–4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Philip M, Fairchild L, Sun L, Horste EL, Camara S, Shakiba M, et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature. 2017;545:452–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Li H, van der Leun AM, Yofe I, Lubling Y, Gelbard-Solodkin D, van Akkooi ACJ et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell. 2019;176:775 – 89.e18.

  90. Sade-Feldman M, Yizhak K, Bjorgaard SL, Ray JP, de Boer CG, Jenkins RW, et al. Defining T Cell States Associated with response to Checkpoint Immunotherapy in Melanoma. Cell. 2018;175:998–e101320.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Utzschneider DT, Legat A, Fuertes Marraco SA, Carrié L, Luescher I, Speiser DE, Zehn D. T cells maintain an exhausted phenotype after antigen withdrawal and population reexpansion. Nat Immunol. 2013;14:603–10.

    Article  PubMed  CAS  Google Scholar 

  92. Abdel-Hakeem MS, Manne S, Beltra J-C, Stelekati E, Chen Z, Nzingha K, et al. Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation. Nat Immunol. 2021;22:1008–19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Martinez GJ, Pereira RM, Äijö T, Kim EY, Marangoni F, Pipkin ME, et al. The transcription factor NFAT promotes exhaustion of activated CD8⁺ T cells. Immunity. 2015;42:265–78.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Dash B, Hogan PG. The road not taken en route to T cell exhaustion. Nat Immunol. 2023;24:1402–4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Mognol GP, Spreafico R, Wong V, Scott-Browne JP, Togher S, Hoffmann A, et al. Exhaustion-associated regulatory regions in CD8+ tumor-infiltrating T cells. Proc Natl Acad Sci U S A. 2017;114:E2776–85.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Tillé L, Cropp D, Charmoy M, Reichenbach P, Andreatta M, Wyss T, et al. Activation of the transcription factor NFAT5 in the tumor microenvironment enforces CD8+ T cell exhaustion. Nat Immunol. 2023;24:1645–53.

    Article  PubMed  Google Scholar 

  97. Chen J, López-Moyado IF, Seo H, Lio C-WJ, Hempleman LJ, Sekiya T, et al. NR4A transcription factors limit CAR T cell function in solid tumours. Nature. 2019;567:530–4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Lynn RC, Weber EW, Sotillo E, Gennert D, Xu P, Good Z, et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature. 2019;576:293–300.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Seo H, González-Avalos E, Zhang W, Ramchandani P, Yang C, Lio CJ, et al. BATF and IRF4 cooperate to counter exhaustion in tumor-infiltrating CAR T cells. Nat Immunol. 2021;22:983–95.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Khan O, Giles JR, McDonald S, Manne S, Ngiow SF, Patel KP, et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature. 2019;571:211–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Sekine T, Perez-Potti A, Nguyen S, Gorin J-B, Wu VH, Gostick E, et al. TOX is expressed by exhausted and polyfunctional human effector memory CD8+ T cells. Sci Immunol. 2020;5:eaba7918.

    Article  PubMed  CAS  Google Scholar 

  102. Alfei F, Kanev K, Hofmann M, Wu M, Ghoneim HE, Roelli P, et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature. 2019;571:265–9.

    Article  PubMed  CAS  Google Scholar 

  103. Beltra JC, Manne S, Abdel-Hakeem MS, Kurachi M, Giles JR, Chen Z et al. Developmental relationships of four exhausted CD8+ T cell subsets reveals underlying transcriptional and epigenetic Landscape Control mechanisms. Immunity. 2020;52:825 – 41.e8.

  104. Pauken KE, Sammons MA, Odorizzi PM, Manne S, Godec J, Khan O, et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science. 2016;354:1160–5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Im SJ, Hashimoto M, Gerner MY, Lee J, Kissick HT, Burger MC, et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature. 2016;537:417–21.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Jansen CS, Prokhnevska N, Master VA, Sanda MG, Carlisle JW, Bilen MA, et al. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature. 2019;576:465–70.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Liu B, Hu X, Feng K, Gao R, Xue Z, Zhang S, et al. Temporal single-cell tracing reveals clonal revival and expansion of precursor exhausted T cells during anti-PD-1 therapy in lung cancer. Nat Cancer. 2022;3:108–21.

    Article  PubMed  CAS  Google Scholar 

  108. Kurtulus S, Madi A, Escobar G, Klapholz M, Nyman J, Christian E et al. Checkpoint blockade immunotherapy induces dynamic changes in PD-1-CD8+ tumor-infiltrating T cells. Immunity. 2019;50:181 – 94.e6.

  109. Miller BC, Sen DR, Al Abosy R, Bi K, Virkud YV, LaFleur MW, et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol. 2019;20:326–36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Brummelman J, Mazza EMC, Alvisi G, Colombo FS, Grilli A, Mikulak J, et al. High-dimensional single cell analysis identifies stem-like cytotoxic CD8+ T cells infiltrating human tumors. J Exp Med. 2018;215:2520–35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  111. Leong YA, Chen Y, Ong HS, Wu D, Man K, Deleage C, et al. CXCR5+ follicular cytotoxic T cells control viral infection in B cell follicles. Nat Immunol. 2016;17:1187–96.

    Article  PubMed  CAS  Google Scholar 

  112. He R, Hou S, Liu C, Zhang A, Bai Q, Han M, et al. Follicular CXCR5- expressing CD8+ T cells curtail chronic viral infection. Nature. 2016;537:412–28.

    Article  PubMed  CAS  Google Scholar 

  113. Yost KE, Satpathy AT, Wells DK, Qi Y, Wang C, Kageyama R, et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med. 2019;25:1251–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Wu T, Ji Y, Moseman EA, Xu HC, Manglani M, Kirby M et al. The TCF1-Bcl6 axis counteracts type I interferon to repress exhaustion and maintain T cell stemness. Sci Immunol. 2016;1.

  115. Staron MM, Gray SM, Marshall HD, Parish IA, Chen JH, Perry CJ, et al. The transcription factor FoxO1 sustains expression of the inhibitory receptor PD-1 and survival of antiviral CD8+ T cells during chronic infection. Immunity. 2014;41:802–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Utzschneider DT, Delpoux A, Wieland D, Huang X, Lai CY, Hofmann M, et al. Active maintenance of T cell memory in Acute and chronic viral infection depends on continuous expression of FOXO1. Cell Rep. 2018;22:3454–67.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  117. Tsui C, Kretschmer L, Rapelius S, Gabriel SS, Chisanga D, Knöpper K, et al. MYB orchestrates T cell exhaustion and response to checkpoint inhibition. Nature. 2022;609:354–60.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  118. Yao C, Lou G, Sun HW, Zhu Z, Sun Y, Chen Z, et al. BACH2 enforces the transcriptional and epigenetic programs of stem-like CD8+ T cells. Nat Immunol. 2021;22:370–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Neubert EN, DeRogatis JM, Lewis SA, Viramontes KM, Ortega P, Henriquez ML, et al. HMGB2 regulates the differentiation and stemness of exhausted CD8+ T cells during chronic viral infection and cancer. Nat Commun. 2023;14:5631.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Zhao X, Shan Q, Xue H-H. TCF1 in T cell immunity: a broadened frontier. Nat Rev Immunol. 2022;22:147–57.

    Article  PubMed  CAS  Google Scholar 

  121. Escobar G, Mangani D, Anderson AC. T cell factor 1: a master regulator of the T cell response in disease. Sci Immunol. 2020;5.

  122. Shan Q, Hu SS, Zhu S, Chen X, Badovinac VP, Peng W, et al. Tcf1 preprograms the mobilization of glycolysis in central memory CD8 + T cells during recall responses. Nat Immunol. 2022;23:386–98.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Shan Q, Hu S, Chen X, Danahy DB, Badovinac VP, Zang C, Xue HH. Ectopic Tcf1 expression instills a stem-like program in exhausted CD8+ T cells to enhance viral and tumor immunity. Cell Mol Immunol. 2021;18:1262–77.

    Article  PubMed  CAS  Google Scholar 

  124. Escobar G, Tooley K, Oliveras JP, Huang L, Cheng H, Bookstaver ML et al. Tumor immunogenicity dictates reliance on TCF1 in CD8+ T cells for response to immunotherapy. Cancer Cell. 2023;41:1662-79.e7.

  125. Chen Z, Ji Z, Ngiow SF, Manne S, Cai Z, Huang AC et al. TCF-1-Centered Transcriptional Network Drives an effector versus exhausted CD8 T cell-fate decision. Immunity. 2019;51:840 – 55.e5.

  126. Krishna S, Lowery FJ, Copeland AR, Bahadiroglu E, Mukherjee R, Jia L, et al. Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer. Science. 2020;370:1328–34.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Eberhardt CS, Kissick HT, Patel MR, Cardenas MA, Prokhnevska N, Obeng RC, et al. Functional HPV-specific PD-1+ stem-like CD8 T cells in head and neck cancer. Nature. 2021;597:279–84.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Nagate Y, Ezoe S, Fujita J, Okuzaki D, Motooka D, Ishibashi T, et al. Ectonucleotidase CD39 is highly expressed on ATLL cells and is responsible for their immunosuppressive function. Leukemia. 2021;35:107–18.

    Article  PubMed  CAS  Google Scholar 

  129. Canale FP, Ramello MC, Núñez N, Araujo Furlan CL, Bossio SN, Gorosito Serrán M, et al. CD39 expression defines cell exhaustion in Tumor-infiltrating CD8+ T cells. Cancer Res. 2018;78:115–28.

    Article  PubMed  CAS  Google Scholar 

  130. Hu G, Cheng P, Pan J, Wang S, Ding Q, Jiang Z, et al. An IL6–Adenosine positive Feedback Loop between CD73+ γδTregs and CAFs promotes Tumor Progression in human breast Cancer. Cancer Immunol Res. 2020;8:1273–86.

    Article  PubMed  CAS  Google Scholar 

  131. Vignali PDA, DePeaux K, Watson MJ, Ye C, Ford BR, Lontos K, et al. Hypoxia drives CD39-dependent suppressor function in exhausted T cells to limit antitumor immunity. Nat Immunol. 2023;24:267–79.

    Article  PubMed  CAS  Google Scholar 

  132. Perrot I, Michaud HA, Giraudon-Paoli M, Augier S, Docquier A, Gros L et al. Blocking Antibodies Targeting the CD39/CD73 Immunosuppressive Pathway Unleash Immune Responses in Combination Cancer Therapies. Cell Rep. 2019;27:2411-25.e9.

  133. Carlini V, Noonan DM, Abdalalem E, Goletti D, Sansone C, Calabrone L, Albini A. The multifaceted nature of IL-10: regulation, role in immunological homeostasis and its relevance to cancer, COVID-19 and post-COVID conditions. Front Immunol. 2023;14.

  134. Guo Y, Xie Y-Q, Gao M, Zhao Y, Franco F, Wenes M, et al. Metabolic reprogramming of terminally exhausted CD8+ T cells by IL-10 enhances anti-tumor immunity. Nat Immunol. 2021;22:746–56.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Qiao J, Liu Z, Dong C, Luan Y, Zhang A, Moore C, et al. Targeting tumors with IL-10 prevents dendritic cell-mediated CD8+ T cell apoptosis. Cancer Cell. 2019;35:901–e154.

    Article  PubMed  CAS  Google Scholar 

  136. Naing A, Infante JR, Papadopoulos KP, Chan IH, Shen C, Ratti NP et al. PEGylated IL-10 (Pegilodecakin) induces systemic Immune activation, CD8+ T cell invigoration and polyclonal T cell expansion in Cancer patients. Cancer Cell. 2018;34:775 – 91.e3.

  137. Naing A, Wong DJ, Infante JR, Korn WM, Aljumaily R, Papadopoulos KP, et al. Pegilodecakin combined with pembrolizumab or nivolumab for patients with advanced solid tumours (IVY): a multicentre, multicohort, open-label, phase 1b trial. Lancet Oncol. 2019;20:1544–55.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  138. Spigel D, Jotte R, Nemunaitis J, Shum M, Schneider J, Goldschmidt J, et al. Randomized phase 2 studies of checkpoint inhibitors alone or in Combination with Pegilodecakin in patients with metastatic NSCLC (CYPRESS 1 and CYPRESS 2). J Thorac Oncol. 2021;16:327–33.

    Article  PubMed  CAS  Google Scholar 

  139. Zander R, Schauder D, Xin G, Nguyen C, Wu X, Zajac A, Cui W. CD4+ T Cell Help Is Required for the Formation of a Cytolytic CD8+ T Cell Subset that Protects against Chronic Infection and Cancer. Immunity. 2019;51:1028-42.e4.

  140. Hudson WH, Gensheimer J, Hashimoto M, Wieland A, Valanparambil RM, Li P et al. Proliferating Transitory T Cells with an Effector-like Transcriptional Signature Emerge from PD-1+ Stem-like CD8+ T Cells during Chronic Infection. Immunity. 2019;51:1043-58.e4.

  141. Rahim MK, Okholm TLH, Jones KB, McCarthy EE, Liu CC, Yee JL, et al. Dynamic CD8+ T cell responses to cancer immunotherapy in human regional lymph nodes are disrupted in metastatic lymph nodes. Cell. 2023;186:1127–e4318.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  142. Zhou P, Shi H, Huang H, Sun X, Yuan S, Chapman NM, et al. Single-cell CRISPR screens in vivo map T cell fate regulomes in cancer. Nature. 2023;624:154–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  143. Magen A, Hamon P, Fiaschi N, Soong BY, Park MD, Mattiuz R, et al. Intratumoral dendritic cell-CD4+ T helper cell niches enable CD8+ T cell differentiation following PD-1 blockade in hepatocellular carcinoma. Nat Med. 2023;29:1389–99.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  144. Giles JR, Ngiow SF, Manne S, Baxter AE, Khan O, Wang P, et al. Shared and distinct biological circuits in effector, memory and exhausted CD8+ T cells revealed by temporal single-cell transcriptomics and epigenetics. Nat Immunol. 2022;23:1600–13.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  145. Wilson TL, Kim H, Chou CH, Langfitt D, Mettelman RC, Minervina AA, et al. Common trajectories of highly effective CD19-Specific CAR T cells identified by endogenous T-cell receptor lineages. Cancer Discov. 2022;12:2098–119.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. Kasmani MY, Zander R, Chung HK, Chen Y, Khatun A, Damo M et al. Clonal lineage tracing reveals mechanisms skewing CD8 + T cell fate decisions in chronic infection. J Exp Med. 2023;220.

  147. Chen Y, Xu Z, Sun H, Ouyang X, Han Y, Yu H, et al. Regulation of CD8+ T memory and exhaustion by the mTOR signals. Cell Mol Immunol. 2023;20:1023–39.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  148. Chen Y, Zander RA, Wu X, Schauder DM, Kasmani MY, Shen J, et al. BATF regulates progenitor to cytolytic effector CD8+ T cell transition during chronic viral infection. Nat Immunol. 2021;22:996–1007.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  149. Zhu Z, Lou G, Teng X-L, Wang H, Luo Y, Shi W, et al. FOXP1 and KLF2 reciprocally regulate checkpoints of stem-like to effector transition in CAR T cells. Nat Immunol. 2024;25:117–28.

    Article  PubMed  Google Scholar 

  150. Beltra J-C, Abdel-Hakeem MS, Manne S, Zhang Z, Huang H, Kurachi M et al. Stat5 opposes the transcription factor Tox and rewires exhausted CD8+ T cells toward durable effector-like states during chronic antigen exposure. Immunity. 2023;56:2699 – 718.e11.

  151. Zander R, Cui W. Exhausted CD8+ T cells face a developmental fork in the road. Trends Immunol. 2023;44:276–86.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  152. Parry EM, Lemvigh CK, Deng S, Dangle N, Ruthen N, Knisbacher BA et al. ZNF683 marks a CD8+ T cell population associated with anti-tumor immunity following anti-PD-1 therapy for Richter syndrome. Cancer Cell. 2023;41:1803-16.e8.

  153. Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pagès C, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313:1960–4.

    Article  PubMed  CAS  Google Scholar 

  154. Pagès F, Mlecnik B, Marliot F, Bindea G, Ou FS, Bifulco C, et al. International validation of the consensus immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet. 2018;391:2128–39.

    Article  PubMed  Google Scholar 

  155. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discovery. 2019;18:197–218.

    Article  PubMed  CAS  Google Scholar 

  156. Angell HK, Bruni D, Barrett JC, Herbst R, Galon J. The immunoscore: Colon Cancer and Beyond. Clin Cancer Res. 2020;26:332–9.

    Article  PubMed  CAS  Google Scholar 

  157. Chen Daniel S, Mellman I. Oncology meets immunology: the Cancer-Immunity cycle. Immunity. 2013;39:1–10.

    Article  PubMed  CAS  Google Scholar 

  158. du Bois H, Heim TA, Lund AW. Tumor-draining lymph nodes: at the crossroads of metastasis and immunity. Sci Immunol. 2021;6:eabg3551.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Inamori K, Togashi Y, Fukuoka S, Akagi K, Ogasawara K, Irie T et al. Importance of lymph node immune responses in MSI-H/dMMR colorectal cancer. JCI Insight. 2021;6.

  160. Harter P, Sehouli J, Lorusso D, Reuss A, Vergote I, Marth C, et al. A Randomized Trial of Lymphadenectomy in patients with Advanced Ovarian neoplasms. N Engl J Med. 2019;380:822–32.

    Article  PubMed  Google Scholar 

  161. Fear VS, Forbes CA, Neeve SA, Fisher SA, Chee J, Waithman J, et al. Tumour draining lymph node-generated CD8 T cells play a role in controlling lung metastases after a primary tumour is removed but not when adjuvant immunotherapy is used. Cancer Immunol Immunother. 2021;70:3249–58.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Delclaux I, Ventre KS, Jones D, Lund AW. The tumor-draining lymph node as a reservoir for systemic immune surveillance. Trends Cancer. 2024;10:28–37.

    Article  PubMed  CAS  Google Scholar 

  163. Schenkel JM, Herbst RH, Canner D, Li A, Hillman M, Shanahan SL, et al. Conventional type I dendritic cells maintain a reservoir of proliferative tumor-antigen specific TCF-1+ CD8+ T cells in tumor-draining lymph nodes. Immunity. 2021;54:2338–53. e6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  164. Roberts EW, Broz ML, Binnewies M, Headley MB, Nelson AE, Wolf DM, et al. Critical role for CD103+/CD141+ dendritic cells bearing CCR7 for Tumor Antigen Trafficking and priming of T cell immunity in Melanoma. Cancer Cell. 2016;30:324–36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  165. Broz ML, Binnewies M, Boldajipour B, Nelson AE, Pollack JL, Erle DJ, et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell. 2014;26:638–52.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  166. Heras-Murillo I, Adán-Barrientos I, Galán M, Wculek SK, Sancho D. Dendritic cells as orchestrators of anticancer immunity and immunotherapy. Nat Reviews Clin Oncol. 2024.

  167. MacNabb BW, Tumuluru S, Chen X, Godfrey J, Kasal DN, Yu J et al. Dendritic cells can prime anti-tumor CD8+ T cell responses through major histocompatibility complex cross-dressing. Immunity. 2022;55:982 – 97.e8.

  168. Ruhland MK, Roberts EW, Cai E, Mujal AM, Marchuk K, Beppler C et al. Visualizing synaptic transfer of Tumor antigens among dendritic cells. Cancer Cell. 2020;37:786 – 99.e5.

  169. Ferris ST, Durai V, Wu R, Theisen DJ, Ward JP, Bern MD, et al. cDC1 prime and are licensed by CD4+ T cells to induce anti-tumour immunity. Nature. 2020;584:624–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  170. Duong E, Fessenden TB, Lutz E, Dinter T, Yim L, Blatt S et al. Type I interferon activates MHC class I-dressed CD11b+ conventional dendritic cells to promote protective anti-tumor CD8+ T cell immunity. Immunity. 2022;55:308 – 23.e9.

  171. Gurevich I, Feferman T, Milo I, Tal O, Golani O, Drexler I, Shakhar G. Active dissemination of cellular antigens by DCs facilitates CD8+ T-cell priming in lymph nodes. Eur J Immunol. 2017;47:1802–18.

    Article  PubMed  CAS  Google Scholar 

  172. Pirillo C, Al Khalidi S, Sims A, Devlin R, Zhao H, Pinto R, et al. Cotransfer of antigen and contextual information harmonizes peripheral and lymph node conventional dendritic cell activation. Sci Immunol. 2023;8:eadg8249.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  173. Connolly KA, Kuchroo M, Venkat A, Khatun A, Wang J, William I, et al. A reservoir of stem-like CD8+ T cells in the tumor-draining lymph node preserves the ongoing antitumor immune response. Sci Immunol. 2021;6:eabg7836.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  174. Prokhnevska N, Cardenas MA, Valanparambil RM, Sobierajska E, Barwick BG, Jansen C et al. CD8+ T cell activation in cancer comprises an initial activation phase in lymph nodes followed by effector differentiation within the tumor. Immunity. 2023;56:107 – 24.e5.

  175. Nagasaki J, Inozume T, Sax N, Ariyasu R, Ishikawa M, Yamashita K, et al. PD-1 blockade therapy promotes infiltration of tumor-attacking exhausted T cell clonotypes. Cell Rep. 2022;38:110331.

    Article  PubMed  CAS  Google Scholar 

  176. Dammeijer F, van Gulijk M, Mulder EE, Lukkes M, Klaase L, van den Bosch T, et al. The PD-1/PD-L1-Checkpoint restrains T cell immunity in Tumor-Draining Lymph Nodes. Cancer Cell. 2020;38:685–e7008.

    Article  PubMed  CAS  Google Scholar 

  177. Steele MM, Jaiswal A, Delclaux I, Dryg ID, Murugan D, Femel J, et al. T cell egress via lymphatic vessels is tuned by antigen encounter and limits tumor control. Nat Immunol. 2023;24:664–75.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  178. Pai JA, Hellmann MD, Sauter JL, Mattar M, Rizvi H, Woo HJ et al. Lineage tracing reveals clonal progenitors and long-term persistence of tumor-specific T cells during immune checkpoint blockade. Cancer Cell. 2023;41:776 – 90.e7.

  179. Francis DM, Manspeaker MP, Schudel A, Sestito LF, O’Melia MJ, Kissick HT et al. Blockade of immune checkpoints in lymph nodes through locoregional delivery augments cancer immunotherapy. Sci Transl Med. 2020;12.

  180. Manspeaker MP, O’Melia MJ, Thomas SN. Elicitation of stem-like CD8+ T cell responses via lymph node-targeted chemoimmunotherapy evokes systemic tumor control. J Immunother Cancer. 2022;10.

  181. Fransen MF, Schoonderwoerd M, Knopf P, Camps MG, Hawinkels LJ, Kneilling M et al. Tumor-draining lymph nodes are pivotal in PD-1/PD-L1 checkpoint therapy. JCI Insight. 2018;3.

  182. Yang H, Sun B, Ma W, Fan L, Xu K, Jia Y, et al. Multi-scale characterization of tumor-draining lymph nodes in resectable lung cancer treated with neoadjuvant immune checkpoint inhibitors. EBioMedicine. 2022;84:104265.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  183. Reticker-Flynn NE, Zhang W, Belk JA, Basto PA, Escalante NK, Pilarowski GOW, et al. Lymph node colonization induces tumor-immune tolerance to promote distant metastasis. Cell. 2022;185:1924–42. .e23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  184. van Krimpen A, Gerretsen VIV, Mulder EEAP, van Gulijk M, van den Bosch TPP, von der Thüsen J, et al. Immune suppression in the tumor-draining lymph node corresponds with distant disease recurrence in patients with melanoma. Cancer Cell. 2022;40:798–9.

    Article  PubMed  Google Scholar 

  185. Núñez NG, Tosello Boari J, Ramos RN, Richer W, Cagnard N, Anderfuhren CD, et al. Tumor invasion in draining lymph nodes is associated with Treg accumulation in breast cancer patients. Nat Commun. 2020;11:3272.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Baeyens A, Fang V, Chen C, Schwab SR. Exit strategies: S1P Signaling and T Cell Migration. Trends Immunol. 2015;36:778–87.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  187. Nakai A, Hayano Y, Furuta F, Noda M, Suzuki K. Control of lymphocyte egress from lymph nodes through β2-adrenergic receptors. J Exp Med. 2014;211:2583–98.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  188. Globig AM, Zhao S, Roginsky J, Maltez VI, Guiza J, Avina-Ochoa N, et al. The β1-adrenergic receptor links sympathetic nerves to T cell exhaustion. Nature. 2023;622:383–92.

    Article  PubMed  CAS  Google Scholar 

  189. Ozga AJ, Chow MT, Luster AD. Chemokines and the immune response to cancer. Immunity. 2021;54:859–74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  190. Chow MT, Ozga AJ, Servis RL, Frederick DT, Lo JA, Fisher DE et al. Intratumoral Activity of the CXCR3 chemokine system is required for the efficacy of Anti-PD-1 therapy. Immunity. 2019;50:1498 – 512.e5.

  191. Di Pilato M, Kfuri-Rubens R, Pruessmann JN, Ozga AJ, Messemaker M, Cadilha BL, et al. CXCR6 positions cytotoxic T cells to receive critical survival signals in the tumor microenvironment. Cell. 2021;184:4512–e3022.

    Article  PubMed  PubMed Central  Google Scholar 

  192. Dangaj D, Bruand M, Grimm AJ, Ronet C, Barras D, Duttagupta PA, et al. Cooperation between constitutive and Inducible Chemokines Enables T Cell Engraftment and Immune Attack in Solid tumors. Cancer Cell. 2019;35:885–e90010.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  193. Hornburg M, Desbois M, Lu S, Guan Y, Lo AA, Kaufman S et al. Single-cell dissection of cellular components and interactions shaping the tumor immune phenotypes in ovarian cancer. Cancer Cell. 2021;39:928 – 44.e6.

  194. Li S, Li K, Wang K, Yu H, Wang X, Shi M, et al. Low-dose radiotherapy combined with dual PD-L1 and VEGFA blockade elicits antitumor response in hepatocellular carcinoma mediated by activated intratumoral CD8+ exhausted-like T cells. Nat Commun. 2023;14:7709.

    Article  PubMed  PubMed Central  Google Scholar 

  195. Huijbers EJM, Khan KA, Kerbel RS, Griffioen AW. Tumors resurrect an embryonic vascular program to escape immunity. Sci Immunol. 2022;7:eabm6388.

    Article  PubMed  CAS  Google Scholar 

  196. Xu M, Zhang T, Xia R, Wei Y, Wei X. Targeting the tumor stroma for cancer therapy. Mol Cancer. 2022;21:208.

    Article  PubMed  PubMed Central  Google Scholar 

  197. Holash J, Maisonpierre PC, Compton D, Boland P, Alexander CR, Zagzag D, et al. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science. 1999;284:1994–8.

    Article  PubMed  CAS  Google Scholar 

  198. Ohm JE, Gabrilovich DI, Sempowski GD, Kisseleva E, Parman KS, Nadaf S, Carbone DP. VEGF inhibits T-cell development and may contribute to tumor-induced immune suppression. Blood. 2003;101:4878–86.

    Article  PubMed  CAS  Google Scholar 

  199. Kim CG, Jang M, Kim Y, Leem G, Kim KH, Lee H et al. VEGF-A drives TOX-dependent T cell exhaustion in anti-PD-1-resistant microsatellite stable colorectal cancers. Sci Immunol. 2019;4.

  200. Voron T, Colussi O, Marcheteau E, Pernot S, Nizard M, Pointet AL, et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med. 2015;212:139–48.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  201. Deng H, Kan A, Lyu N, Mu L, Han Y, Liu L, et al. Dual vascular endothelial growth factor receptor and fibroblast growth factor receptor inhibition elicits Antitumor Immunity and enhances programmed cell Death-1 checkpoint blockade in Hepatocellular Carcinoma. Liver Cancer. 2020;9:338–57.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  202. Liu S, Qin T, Liu Z, Wang J, Jia Y, Feng Y, et al. Anlotinib alters tumor immune microenvironment by downregulating PD-L1 expression on vascular endothelial cells. Cell Death Dis. 2020;11:309.

    Article  PubMed  PubMed Central  Google Scholar 

  203. Crispen PL, Sheinin Y, Roth TJ, Lohse CM, Kuntz SM, Frigola X, et al. Tumor cell and tumor vasculature expression of B7-H3 predict survival in clear cell renal cell carcinoma. Clin Cancer Res. 2008;14:5150–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  204. Motz GT, Santoro SP, Wang L-P, Garrabrant T, Lastra RR, Hagemann IS, et al. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat Med. 2014;20:607–15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  205. Dirkx AE, Oude Egbrink MG, Kuijpers MJ, van der Niet ST, Heijnen VV, Bouma-ter Steege JC, et al. Tumor angiogenesis modulates leukocyte-vessel wall interactions in vivo by reducing endothelial adhesion molecule expression. Cancer Res. 2003;63:2322–9.

    PubMed  CAS  Google Scholar 

  206. Griffioen AW, Damen CA, Blijham GH, Groenewegen G. Tumor angiogenesis is accompanied by a decreased inflammatory response of tumor-associated endothelium. Blood. 1996;88:667–73.

    Article  PubMed  CAS  Google Scholar 

  207. Huinen ZR, Huijbers EJM, van Beijnum JR, Nowak-Sliwinska P, Griffioen AW. Anti-angiogenic agents — overcoming tumour endothelial cell anergy and improving immunotherapy outcomes. Nat Reviews Clin Oncol. 2021;18:527–40.

    Article  Google Scholar 

  208. Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, et al. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med. 2019;380:1116–27.

    Article  PubMed  CAS  Google Scholar 

  209. Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, et al. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med. 2019;380:1103–15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  210. Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim T-Y, et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N Engl J Med. 2020;382:1894–905.

    Article  PubMed  CAS  Google Scholar 

  211. Socinski MA, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, et al. Atezolizumab for First-Line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378:2288–301.

    Article  PubMed  CAS  Google Scholar 

  212. Makker V, Colombo N, Casado Herráez A, Santin AD, Colomba E, Miller DS, et al. Lenvatinib plus Pembrolizumab for Advanced Endometrial Cancer. N Engl J Med. 2022;386:437–48.

    Article  PubMed  CAS  Google Scholar 

  213. Meng X, Wu T, Hong Y, Fan Q, Ren Z, Guo Y, et al. Camrelizumab plus Apatinib as second-line treatment for advanced oesophageal squamous cell carcinoma (CAP 02): a single-arm, open-label, phase 2 trial. Lancet Gastroenterol Hepatol. 2022;7:245–53.

    Article  PubMed  Google Scholar 

  214. Asrir A, Tardiveau C, Coudert J, Laffont R, Blanchard L, Bellard E et al. Tumor-associated high endothelial venules mediate lymphocyte entry into tumors and predict response to PD-1 plus CTLA-4 combination immunotherapy. Cancer Cell. 2022;40:318 – 34.e9.

  215. Allen E, Jabouille A, Rivera LB, Lodewijckx I, Missiaen R, Steri V et al. Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Sci Transl Med. 2017;9.

  216. Hua Y, Vella G, Rambow F, Allen E, Antoranz Martinez A, Duhamel M et al. Cancer immunotherapies transition endothelial cells into HEVs that generate TCF1+ T lymphocyte niches through a feed-forward loop. Cancer Cell. 2022;40:1600-18.e10.

  217. Frankenstein Z, Basanta D, Franco OE, Gao Y, Javier RA, Strand DW, et al. Stromal reactivity differentially drives tumour cell evolution and prostate cancer progression. Nat Ecol Evol. 2020;4:870–84.

    Article  PubMed  PubMed Central  Google Scholar 

  218. Zhao Y, Shen M, Wu L, Yang H, Yao Y, Yang Q, et al. Stromal cells in the tumor microenvironment: accomplices of tumor progression? Cell Death Dis. 2023;14:587.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  219. Cords L, Engler S, Haberecker M, Rüschoff JH, Moch H, de Souza N, Bodenmiller B. Cancer-associated fibroblast phenotypes are associated with patient outcome in non-small cell lung cancer. Cancer Cell. 2024.

  220. Buechler MB, Pradhan RN, Krishnamurty AT, Cox C, Calviello AK, Wang AW, et al. Cross-tissue organization of the fibroblast lineage. Nature. 2021;593:575–9.

    Article  PubMed  CAS  Google Scholar 

  221. Grout JA, Sirven P, Leader AM, Maskey S, Hector E, Puisieux I, et al. Spatial positioning and Matrix Programs of Cancer-Associated fibroblasts promote T-cell exclusion in human lung tumors. Cancer Discov. 2022;12:2606–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  222. Krishnamurty AT, Shyer JA, Thai M, Gandham V, Buechler MB, Yang YA, et al. LRRC15+ myofibroblasts dictate the stromal setpoint to suppress tumour immunity. Nature. 2022;611:148–54.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  223. Caligiuri G, Tuveson DA. Activated fibroblasts in cancer: perspectives and challenges. Cancer Cell. 2023;41:434–49.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  224. Ma C, Yang C, Peng A, Sun T, Ji X, Mi J, et al. Pan-cancer spatially resolved single-cell analysis reveals the crosstalk between cancer-associated fibroblasts and tumor microenvironment. Mol Cancer. 2023;22:170.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  225. Luo H, Xia X, Huang L-B, An H, Cao M, Kim GD, et al. Pan-cancer single-cell analysis reveals the heterogeneity and plasticity of cancer-associated fibroblasts in the tumor microenvironment. Nat Commun. 2022;13:6619.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  226. Elyada E, Bolisetty M, Laise P, Flynn WF, Courtois ET, Burkhart RA, et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals Antigen-Presenting Cancer-Associated fibroblasts. Cancer Discov. 2019;9:1102–23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  227. Jenkins L, Jungwirth U, Avgustinova A, Iravani M, Mills A, Haider S, et al. Cancer-Associated fibroblasts suppress CD8+ T-cell infiltration and Confer Resistance to Immune-Checkpoint Blockade. Cancer Res. 2022;82:2904–17.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  228. Kieffer Y, Hocine HR, Gentric G, Pelon F, Bernard C, Bourachot B, et al. Single-cell analysis reveals fibroblast clusters linked to Immunotherapy Resistance in Cancer. Cancer Discov. 2020;10:1330–51.

    Article  PubMed  CAS  Google Scholar 

  229. Chakravarthy A, Khan L, Bensler NP, Bose P, De Carvalho DD. TGF-β-associated extracellular matrix genes link cancer-associated fibroblasts to immune evasion and immunotherapy failure. Nat Commun. 2018;9:4692.

    Article  PubMed  PubMed Central  Google Scholar 

  230. Peng DH, Rodriguez BL, Diao L, Chen L, Wang J, Byers LA, et al. Collagen promotes anti-PD-1/PD-L1 resistance in cancer through LAIR1-dependent CD8+ T cell exhaustion. Nat Commun. 2020;11:4520.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  231. Pruitt HC, Lewis D, Ciccaglione M, Connor S, Smith Q, Hickey JW et al. Collagen fiber structure guides 3D motility of cytotoxic T lymphocytes. Matrix Biol. 2020;85–6:147–59.

  232. Salmon H, Franciszkiewicz K, Damotte D, Dieu-Nosjean MC, Validire P, Trautmann A, et al. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J Clin Invest. 2012;122:899–910.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  233. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018;554:544–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  234. Costa A, Kieffer Y, Scholer-Dahirel A, Pelon F, Bourachot B, Cardon M, et al. Fibroblast heterogeneity and immunosuppressive environment in human breast Cancer. Cancer Cell. 2018;33:463–. – 79.e10.

    Article  PubMed  CAS  Google Scholar 

  235. Kato T, Noma K, Ohara T, Kashima H, Katsura Y, Sato H, et al. Cancer-Associated fibroblasts affect Intratumoral CD8+ and FoxP3+ T cells Via IL6 in the Tumor Microenvironment. Clin Cancer Res. 2018;24:4820–33.

    Article  PubMed  CAS  Google Scholar 

  236. Qi J, Sun H, Zhang Y, Wang Z, Xun Z, Li Z, et al. Single-cell and spatial analysis reveal interaction of FAP+ fibroblasts and SPP1+ macrophages in colorectal cancer. Nat Commun. 2022;13:1742.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  237. Wu SZ, Roden DL, Wang C, Holliday H, Harvey K, Cazet AS, et al. Stromal cell diversity associated with immune evasion in human triple-negative breast cancer. Embo j. 2020;39:e104063.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  238. Zhao Q, Huang L, Qin G, Qiao Y, Ren F, Shen C, et al. Cancer-associated fibroblasts induce monocytic myeloid-derived suppressor cell generation via IL-6/exosomal miR-21-activated STAT3 signaling to promote cisplatin resistance in esophageal squamous cell carcinoma. Cancer Lett. 2021;518:35–48.

    Article  PubMed  CAS  Google Scholar 

  239. Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci U S A. 2013;110:20212–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  240. Li C, Guo H, Zhai P, Yan M, Liu C, Wang X, et al. Spatial and single-cell transcriptomics reveal a Cancer-Associated Fibroblast Subset in HNSCC that restricts infiltration and antitumor activity of CD8+ T cells. Cancer Res. 2024;84:258–75.

    Article  PubMed  CAS  Google Scholar 

  241. Yang W, Liu S, Mao M, Gong Y, Li X, Lei T, et al. T-cell infiltration and its regulatory mechanisms in cancers: insights at single-cell resolution. J Exp Clin Cancer Res. 2024;43:38.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  242. Zboralski D, Hoehlig K, Eulberg D, Frömming A, Vater A. Increasing tumor-infiltrating T cells through inhibition of CXCL12 with NOX-A12 synergizes with PD-1 blockade. Cancer Immunol Res. 2017;5:950–6.

    Article  PubMed  CAS  Google Scholar 

  243. Sathe A, Mason K, Grimes SM, Zhou Z, Lau BT, Bai X, et al. Colorectal Cancer metastases in the liver establish immunosuppressive spatial networking between Tumor-Associated SPP1+ macrophages and fibroblasts. Clin Cancer Res. 2023;29:244–60.

    Article  PubMed  CAS  Google Scholar 

  244. Timperi E, Gueguen P, Molgora M, Magagna I, Kieffer Y, Lopez-Lastra S, et al. Lipid-Associated macrophages Are Induced by Cancer-Associated fibroblasts and mediate Immune suppression in breast Cancer. Cancer Res. 2022;82:3291–306.

    Article  PubMed  CAS  Google Scholar 

  245. Huang H, Wang Z, Zhang Y, Pradhan RN, Ganguly D, Chandra R et al. Mesothelial cell-derived antigen-presenting cancer-associated fibroblasts induce expansion of regulatory T cells in pancreatic cancer. Cancer Cell. 2022;40:656 – 73.e7.

  246. Lakins MA, Ghorani E, Munir H, Martins CP, Shields JD. Cancer-associated fibroblasts induce antigen-specific deletion of CD8+ T cells to protect tumour cells. Nat Commun. 2018;9:948.

    Article  PubMed  PubMed Central  Google Scholar 

  247. Kerdidani D, Aerakis E, Verrou KM, Angelidis I, Douka K, Maniou MA et al. Lung tumor MHCII immunity depends on in situ antigen presentation by fibroblasts. J Exp Med. 2022;219.

  248. Hutton C, Heider F, Blanco-Gomez A, Banyard A, Kononov A, Zhang X, et al. Single-cell analysis defines a pancreatic fibroblast lineage that supports anti-tumor immunity. Cancer Cell. 2021;39:1227–e4420.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  249. Xiao Z, Todd L, Huang L, Noguera-Ortega E, Lu Z, Huang L, et al. Desmoplastic stroma restricts T cell extravasation and mediates immune exclusion and immunosuppression in solid tumors. Nat Commun. 2023;14:5110.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  250. Chen Y, Kim J, Yang S, Wang H, Wu CJ, Sugimoto H et al. Type I collagen deletion in αSMA+ myofibroblasts augments immune suppression and accelerates progression of pancreatic cancer. Cancer Cell. 2021;39:548 – 65.e6.

  251. Bhattacharjee S, Hamberger F, Ravichandra A, Miller M, Nair A, Affo S et al. Tumor restriction by type I collagen opposes tumor-promoting effects of cancer-associated fibroblasts. J Clin Invest. 2021;131.

  252. Verma NK, Wong BHS, Poh ZS, Udayakumar A, Verma R, Goh RKJ et al. Obstacles for T-lymphocytes in the tumour microenvironment: therapeutic challenges, advances and opportunities beyond immune checkpoint. eBioMedicine. 2022;83.

  253. Liu X, Hoft DF, Peng G. Tumor microenvironment metabolites directing T cell differentiation and function. Trends Immunol. 2022;43:132–47.

    Article  PubMed  CAS  Google Scholar 

  254. Goc J, Germain C, Vo-Bourgais TKD, Lupo A, Klein C, Knockaert S, et al. Dendritic cells in Tumor-Associated Tertiary lymphoid structures signal a Th1 cytotoxic Immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res. 2014;74:705–15.

    Article  PubMed  CAS  Google Scholar 

  255. Humblin E, Korpas I, Lu J, Filipescu D, van der Heide V, Goldstein S, et al. Sustained CD28 costimulation is required for self-renewal and differentiation of TCF-1+ PD-1+ CD8 T cells. Sci Immunol. 2023;8:eadg0878.

    Article  PubMed  PubMed Central  Google Scholar 

  256. Im SJ, Obeng RC, Nasti TH, McManus D, Kamphorst AO, Gunisetty S, et al. Characteristics and anatomic location of PD-1+TCF1+ stem-like CD8 T cells in chronic viral infection and cancer. Proc Natl Acad Sci U S A. 2023;120:e2221985120.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  257. Meiser P, Knolle MA, Hirschberger A, de Almeida GP, Bayerl F, Lacher S et al. A distinct stimulatory cDC1 subpopulation amplifies CD8+ T cell responses in tumors for protective anti-cancer immunity. Cancer Cell. 2023;41:1498 – 515.e10.

  258. Dixon KO, Tabaka M, Schramm MA, Xiao S, Tang R, Dionne D, et al. TIM-3 restrains anti-tumour immunity by regulating inflammasome activation. Nature. 2021;595:101–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  259. Lee CYC, Kennedy BC, Richoz N, Dean I, Tuong ZK, Gaspal F, et al. Tumour-retained activated CCR7+ dendritic cells are heterogeneous and regulate local anti-tumour cytolytic activity. Nat Commun. 2024;15:682.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  260. Duraiswamy J, Turrini R, Minasyan A, Barras D, Crespo I, Grimm AJ, et al. Myeloid antigen-presenting cell niches sustain antitumor T cells and license PD-1 blockade via CD28 costimulation. Cancer Cell. 2021;39:1623–e4220.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  261. Maier B, Leader AM, Chen ST, Tung N, Chang C, LeBerichel J, et al. A conserved dendritic-cell regulatory program limits antitumour immunity. Nature. 2020;580:257–62.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  262. Bayerl F, Meiser P, Donakonda S, Hirschberger A, Lacher SB, Pedde AM, et al. Tumor-derived prostaglandin E2 programs cDC1 dysfunction to impair intratumoral orchestration of anti-cancer T cell responses. Immunity. 2023;56:1341–e5811.

    Article  PubMed  CAS  Google Scholar 

  263. Cohen M, Giladi A, Barboy O, Hamon P, Li B, Zada M, et al. The interaction of CD4+ helper T cells with dendritic cells shapes the tumor microenvironment and immune checkpoint blockade response. Nat Cancer. 2022;3:303–17.

    Article  PubMed  CAS  Google Scholar 

  264. Sato Y, Silina K, van den Broek M, Hirahara K, Yanagita M. The roles of tertiary lymphoid structures in chronic diseases. Nat Rev Nephrol. 2023;19:525–37.

    Article  PubMed  Google Scholar 

  265. Cabrita R, Lauss M, Sanna A, Donia M, Skaarup Larsen M, Mitra S, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577:561–5.

    Article  PubMed  CAS  Google Scholar 

  266. Petitprez F, de Reyniès A, Keung EZ, Chen TW-W, Sun C-M, Calderaro J, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature. 2020;577:556–60.

    Article  PubMed  CAS  Google Scholar 

  267. Vanhersecke L, Brunet M, Guégan JP, Rey C, Bougouin A, Cousin S, et al. Mature tertiary lymphoid structures predict immune checkpoint inhibitor efficacy in solid tumors independently of PD-L1 expression. Nat Cancer. 2021;2:794–802.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  268. Sawada J, Hiraoka N, Qi R, Jiang L, Fournier-Goss AE, Yoshida M, et al. Molecular signature of Tumor-Associated High Endothelial venules that can predict breast Cancer survival. Cancer Immunol Res. 2022;10:468–81.

    Article  PubMed  PubMed Central  Google Scholar 

  269. Patil NS, Nabet BY, Müller S, Koeppen H, Zou W, Giltnane J, et al. Intratumoral plasma cells predict outcomes to PD-L1 blockade in non-small cell lung cancer. Cancer Cell. 2022;40:289–e3004.

    Article  PubMed  CAS  Google Scholar 

  270. Helmink BA, Reddy SM, Gao J, Zhang S, Basar R, Thakur R, et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature. 2020;577:549–55.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  271. Bruno TC. New predictors for immunotherapy responses sharpen our view of the tumour microenvironment. Nature. 2020;577:474–6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  272. Thommen DS, Koelzer VH, Herzig P, Roller A, Trefny M, Dimeloe S, et al. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat Med. 2018;24:994–1004.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  273. Xu W, Lu J, Liu WR, Anwaier A, Wu Y, Tian X et al. Heterogeneity in tertiary lymphoid structures predicts distinct prognosis and immune microenvironment characterizations of clear cell renal cell carcinoma. J Immunother Cancer. 2023;11.

  274. Masuda T, Tanaka N, Takamatsu K, Hakozaki K, Takahashi R, Anno T et al. Unique characteristics of tertiary lymphoid structures in kidney clear cell carcinoma: prognostic outcome and comparison with bladder cancer. J Immunother Cancer. 2022;10.

  275. Liu B, Zhang Y, Wang D, Hu X, Zhang Z. Single-cell meta-analyses reveal responses of tumor-reactive CXCL13+ T cells to immune-checkpoint blockade. Nat Cancer. 2022;3:1123–36.

    Article  PubMed  CAS  Google Scholar 

  276. Dai S, Zeng H, Liu Z, Jin K, Jiang W, Wang Z, et al. Intratumoral CXCL13+CD8+T cell infiltration determines poor clinical outcomes and immunoevasive contexture in patients with clear cell renal cell carcinoma. J Immunother Cancer. 2021;9:e001823.

    Article  PubMed  PubMed Central  Google Scholar 

  277. Simoni Y, Becht E, Fehlings M, Loh CY, Koo S-L, Teng KWW, et al. Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature. 2018;557:575–9.

    Article  PubMed  CAS  Google Scholar 

  278. Borràs DM, Verbandt S, Ausserhofer M, Sturm G, Lim J, Verge GA, et al. Single cell dynamics of tumor specificity vs bystander activity in CD8+ T cells define the diverse immune landscapes in colorectal cancer. Cell Discov. 2023;9:114.

    Article  PubMed  PubMed Central  Google Scholar 

  279. Kirschenbaum D, Xie K, Ingelfinger F, Katzenelenbogen Y, Abadie K, Look T et al. Time-resolved single-cell transcriptomics defines immune trajectories in glioblastoma. Cell. 2024;187:149 – 65.e23.

  280. Jia D, Wang Q, Qi Y, Jiang Y, He J, Lin Y, et al. Microbial metabolite enhances immunotherapy efficacy by modulating T cell stemness in pan-cancer. Cell. 2024;187:1651–e6521.

    Article  PubMed  CAS  Google Scholar 

  281. Mullard A. FDA approves first tumour-infiltrating lymphocyte (TIL) therapy, bolstering hopes for cell therapies in solid cancers 2024 [cited 2024 March 30]. https://www.nature.com/articles/d41573-024-00035-1

Download references

Acknowledgements

We appreciate all the valuable inputs from the colleagues in our laboratory. We also would like to thank Zhou He (College of Computer Science and Technology, Zhejiang University), Xuan Li (University of Bristol), Dr Chang Ge (School of Medicine, Zhejiang University), and Dr Jieqiong Chen (Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University) for their contribution to this work. The figures in this manuscript were created with BioRender.com.

Funding

This work was supported by National Natural Science Foundation of China (82273178) and Zhejiang Province Traditional Chinese Medicine Science and Technology Plan (2023ZR108).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, Y.S., E.Y., S.W., Z.W., B.L., and Z.Y.; Writing – Original Draft, Y.S., E.Y., F.W., Y.X., and W.Z.; Writing – Review & Editing, Y.S., E.Y., S.W., Z.W., F.W., Y.X., W.Z., S.Z., H.M., S.C., and L.J.; Visualisation, Y.S, E.Y., S.W., F.W. and Y.X.; Supervision, E.Y., S.W., Z.W., B.L., and Z.Y.

Corresponding authors

Correspondence to Binghao Li or Zhaoming Ye.

Ethics declarations

Ethical approval

This study did not involve the participation of human or animal subjects, and, therefore, was exempt from formal assessment by the ethics committee for clinical research of our centre.

Competing interests

The 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.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sun, Y., Yinwang, E., Wang, S. et al. Phenotypic and spatial heterogeneity of CD8+ tumour infiltrating lymphocytes. Mol Cancer 23, 193 (2024). https://doi.org/10.1186/s12943-024-02104-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12943-024-02104-w

Keywords