Skip to main content

The expanding Pandora’s toolbox of CD8+T cell: from transcriptional control to metabolic firing

Abstract

CD8+ T cells are the executor in adaptive immune response, especially in anti-tumor immunity. They are the subset immune cells that are of high plasticity and multifunction. Their development, differentiation, activation and metabolism are delicately regulated by multiple factors. Stimuli from the internal and external environment could remodel CD8+ T cells, and correspondingly they will also make adjustments to the microenvironmental changes. Here we describe the most updated progresses in CD8+ T biology from transcriptional regulation to metabolism mechanisms, and also their interactions with the microenvironment, especially in cancer and immunotherapy. The expanding landscape of CD8+ T cell biology and discovery of potential targets to regulate CD8+ T cells will provide new viewpoints for clinical immunotherapy.

Introduction

CD8+ T cells, the central player of the adaptive immune system in eliminating pathogens and tumor cells, exhibit functional plasticity and complexity [1,2,3]. Naive CD8+ T cells are rapidly activated and clonal expanded to produce lots of antigen-specific effector CD8+ T cells and memory T cells after receiving antigens presented by dendritic cells (DCs) in peripheral lymphoid organs. The effector CD8+ T cells then enter the blood and migrate to the primary sites of infection or tumor, secreting cytokines such as interferon, tumor necrosis factor (TNF), and cytotoxic effector molecules such as perforin, granzyme and so on, to specifically eliminate the infected target cells or tumor cells. CD8+ T cells are a loyal guardian, but when CD8+ T cells are persistently activated or metabolically disturbed, the line of defense against pathogens and tumors will be broken. Out of control of CD8+ T cells are mainly manifested as exhaustion, dysfunction and ineffective monitoring, leading to immunotherapy tolerance, especially in infectious diseases and tumors [4, 5]. There have been a number of combination therapies such as anti-PD-1/PD-L1(programmed death receptor 1/programmed cell death 1 ligand 1) combined with chemotherapy, radiotherapy, angiogenesis inhibitors, agonists of the co-stimulatory molecule, stimulator of interferon genes agonists, epigenetic modulators, or metabolic modulators and so on, showing superior efficacies and higher response rates in cancer treatment [6].

In this review, we systematically describe the updates of CD8+ T cells development, metabolism, crosstalk with tumor microenvironment in the case of tumorigenesis. And we summarize emerging evidence that how transcription regulation and T cell metabolism will affect its ability to combat cancer. In the end, we discuss unanswered questions in the field, to gain more complete understanding of T cells and provide new ideas for future CD8+ T cell-based therapies.

Transcriptional mechanisms in CD8+ T cell differentiation

CD8+ T cell activation

CD8+ T cells are important in adaptive immunity to tumor. Activation of naive CD8+ T cells trigger the change of cell cycle, protein expression, metabolism, and generation of distinct cellular phenotypes [7]. Once activation, naive T cells could differentiate into short-lived or terminal effector cells (SLECs/TEs) as well as long-lived memory precursors (MPs), and switch from quiet to active state that enable amplification up to 15–20 times approximately within one week and increase up to 50,000-fold in cell number [8,9,10]. The variety, strength and duration of antigen are key determinants of T cell differentiation [11, 12]. At the same time, APCs and/or CD4+ T cells secret co-stimulatory signals and cytokines that influence CD8+ T cells differentiation. Then CD8+ T cells undergo differentiation and expansion to generate a great numbers of effector cells which are able to migrate into the periphery. Mechanistically, naive CD8+ T cells are activated by recognition of specific peptides presented by major histocompatibility class I (MHC-I) on antigen presenting cells (APCs) in peripheral lymphatic organs (Fig. 1), however, tumor cells can significantly reduce MHC-I antigen presentation, thereby "hiding" in front of CD8+ T cells and achieving immune escape. This type of tumor with T cell infiltration and weak immune response is also known as a "cold tumor" [13]. Wang’s latest research indicate that SUSD6 and TMEM127 are two membrane molecules that simultaneously interact directly with MHC-I and jointly recruit WWP2 to form a quaternary complex. In the presence of SUSD6 and TMEM127, WWP2 mediates MHC-I ubiquitination as well as lysosomal degradation, thereby reducing MHC-I surface expression, loss of SUSD6 enhances MHC-I surface expression, which can promote the function of CD8+ T cells, thereby enhancing tumor immune surveillance [14]. In addition to MHC-I, MHC-II are also important for T cell differentiation. Research from Booki Min reported that MHC-II−/− CD8+ T cells are hyperproliferated under lymphopenic conditions, differentiated into effector cells producing proinflammatory cytokines, and mediated more severe tissue inflammation compared with wide type CD8+ T cells, the reason is that, as a MHC-II ligand, LAG3 is markedly enhanced in MHC-II−/− CD8+ T and blockade of MHC-II-LAG3 interaction further promote T cell expansion [15].

Fig. 1
figure 1

Transcriptional and epigenetic mechanisms involved in CD8+ T cell development. A APCs can recognize and present the antigens, and then present to CD8 + T cells by MHC, CD8 + T cells are activated. B When naive CD8+ T cells activated, most of them transform into effect CD8+ T cells and a small part transform into memory CD8+ T cells. Naive CD8+ T cells translate effector CD8+ T cells, this process is regulated by transcription factors, such as T-bet, and so on, at the same time, epigenetics is changed. C: memory CD8+ T cells can self-renew, and when it’s stimulated by second antigens, memory CD8+ T cells rapidly transform into effect CD8+ T cells. D: effect CD8+ T cells secrete cytokines such as TNFs in response to stimulation. APC antigen presenting cell; MHC major histocompatibility complex, TFs transcription factors, PD-1 programmed death receptor 1

CD8+ T cell differentiation

CD8+ T cell differentiation accompanies with its activation. In this part, we will discuss the role of transcription factors, T-bet, Eomesodermin (Eomes) and Runx3 in the differentiation of CD8+ T cell.

As a member of the T-box family, T-bet is the main regulator of type I differentiation in CD8+ T cells and is necessary for the expression of IFN. CD8+ T cells expressing T-bet represent short-lived effector differentiation and associates with the KLRG1hi and CD127lo phenotype [16,17,18]. Eomes is a different transcriptional factor which belongs to T-box family. Eomes  expression increase from the effector to memory phases of a CD8 T cell response while T-bet expression is observed to be maximal during effector phase [19, 20]. IL-12 upregulates T-bet expression but represses its consistent transcription, which is consistent with IL-12’s effect in regulation of robust, short-lived effector cells [21]. T-bet, also has long been known to be a key transcription factor for effector and memory CD8+ T cells (Fig. 1B) [17, 22, 23]. T-bet regulates CD8+ T cell effector and memory differentiation, enhances the expression of IL-2R and also helpful for cytotoxic CD8+ T cells to secrete IFN-γ, perforin and granzyme B [19, 20, 22]. Reiner et al. reported that T-bet and Eomes deficiency fail to respond to lymphocytic choriomeningitis virus (LCMV) infection [24]. T-bet maintains a steady state and delet of T-bet-expressing Treg cells results in severe Th1 autoimmunity in mouse [25]. IL-12 modulates T-bet in a dose-dependent manner and high amounts of T-bet induced KLRG1hi IL-7Rlo short-lived effector cells, but lower amounts upregulated the development of KLRG1lo IL-7Rhi memory precursor effector cells [18]. Meanwhile, research revealed that T-bet and Eomes regulate CD8+ T cell exhaustion which correlate with the T-bet nuclear localization, and from Chen’s research, we know that TCF-1 participates the T-bet-to-Eomes transcription factor transition in progenitor exhausted CD8+ T cells by upregulating Eomes expression and driving c-Myb expression that controlled Bcl-2 and survival [26, 27]. Unexpectedly, Iwata et al. found that T-bet also acts as a repressor of Type I interferons (IFN-I) transcription factors and IFN-I stimulated genes in Th1 cell that restrains Th1 response [28]. These findings suggests that T-bet is necessary to determine CD8+ T cell fate and its function is related to the localization of the cell.

Eomes is highly homologous to T-bet and expressed in activated CD8+ T cells and in activated NK cells. It has cooperative functions with T-bet in CD8+ T cells [29]. Andrew et al. cross-bred T-bet−/− mice with Eomes−/− mice to obtain dual gene knockout mice. The mice showed a decrease in the proportion of CD8+ T cells and the production of IFN-γ, and the cytotoxic activity under the infection with LCMV [30]. T-bet and Eomes are also involved in regulating the differentiation of CD8+ T cells into effector T cells and memory T cells. Laura et al. used flow cytometry to detect the expression levels of T-bet and Eomes on immature CD8+ T cells, central memory CD8+ T cells, effector memory T cells, and effector CD8+ T cells. The results showed that T-bet had the highest expression level on effector CD8+ T cells, Eomes has the highest expression level on effector memory T cells [31,32,33,34]. Other group also confirmed the regulatory effect of T-bet and Eomes on memory T cells [35]. Banerjee et al. found that Eomes knockout mice also had defects in the formation of long-term memory T cells, cell stability, and cell renewal ability [36]. Therefore, the differentiation direction of CD8+ T cells depends on the expression levels of T-bet and Eomes. TGF-β (transforming growth factor β) and Eomes signal coordinate to promote the homeostasis of CD8+ Treg cells. Simultaneous disruption of both TGF-β receptor and Eomes in T cells result in lethal autoimmunity [37]. Ectopic expression of Eomes is sufficient to activate effector CD8+ T cells that secrete IFN-γ, perforin and granzyme B [20]. Eomes-dependent loss of CD226 related to tumor-infiltrating lymphocytes (TILs) with reduced anti-tumor functions [38]. All in all, the regulation for CD8+ T cells is multifactors coordination in different states and periods.

In addition, Runx3 can be considered as another transcription factor. Runx3 is an important regulator of TRM cell differentiation and homeostasis through TGF-β dependent transcriptional mechanism [39]. Runx3 and T-bet colocalization with Batf that mediated effector CD8+ T cell differentiation [40]. Runx3 can be regarded as a tumor suppressor transcription factor which delays melanoma growth, mortality and enhanced tumor specific CD8+ T cell abundance [41]. Through computational biology and RNA interference screening techniques, Goldrath et al. found that Runx3 is a key regulatory factor involved in TRM differentiation and homeostasis in various tissues, and can participate in special gene expression programs in CD8+ T cells infiltrating normal tissues and tumors. It also has been confirmed through a melanoma mouse model that overexpressing Runx3 in T cells can slow down tumor growth and prolong survival, while the absence of Runx3 leads to worse outcomes [41]. Regulating the activity of Runx3 in T cells can affect the accumulation of T cells in solid tumors, which may help researchers improve current cancer immunotherapy. In the future, we can use Runx3 to reprogram CD8+ T cells, thereby driving their killing effect in tumors. Green et al. used CRISPR-based screen to identify the mammalian BRG1/BRM-associated factor (cBAF), which are positively correlated with the differentiation of activated CD8+ T into effector cells and negatively correlated with memory T cell formation [42]. Whether there is a synergistic relationship between T-bet and BRG1/BRM-associated factor has not been studied.

In conclusion, Eomes, Runx3 and T-bet are members of an interactional transcriptional network necessary for CD8+ T cell differentiation program and acquisition of effector functions. T cell receptor (TCR) signal activates T-bet that promotes IFN-γ expression. Runx3 induces the expression of IFN-γ and upregulates granzyme B, then Runx3 induces Eomes and subsequent the expression of perforin and IFN-γ expression [43]. The differential expression and function of these factors during effector and memory stages suggest an important role for them in the induction and maintenance of genetic programs that regulate effector and memory CD8+ T cell differentiation and imply that its use may greatly benefit tumor therapy. There are also other transcription factors that regulate CD8+ T cell that it is not mentioned in the text (Table 1).

Table1 Transcription factors associated with CD8+ T cells

Epigenetic regulation of CD8+ T cell

Growing studies have shown that epigenetic mechanisms cooperate with transcription factors, which is crucial for the transcriptional changes associated with CD8+ T cell differentiation (Fig. 1B). Histone post-translational modifications and DNA methylation are the main epigenetic mechanisms. DNA methylation mainly occurs on CG dinucleotide (CpG)-dense regions, namely CpG islands which are located at transcriptional start sites and associate with transcriptional repression [44, 45]. Understanding epigenetic mechanisms that regulate the differentiation of CD8+ T cell would have implications for both T cell biology and immunotherapy. Asymmetric expression and directed activity of epigenetic modifying proteins during CD8+ T cell differentiation regulate subset-specific cellular functions and may even be involved in fate decisions during the early stages of naive T cell activation. Knockout of the gene encoding methyl-CpG-binding domain protein 2 (MBD2) leads to differentiation defects in CD8+ T cells [2, 46, 47]. As a component of the H3K27me3 reader complex PRC1, the expression of BMI1 is regulated by TCR in both naive CD8+ T cells and memory precursor T cells, and BMI1 participates in cellular senescence and apoptosis through regulation of the gene expression of p16INK4A and p14ARF, however, it disappears in terminally differentiated effector T cells [48,49,50,51]. Similar results are also observed in histone-lysine N-methyltransferase, EZH2, which belongs to part of the H3K27me3 writer complex PRC2. EZH2+CD8+ T cells increased the polyfunctionality and resistance to spontaneous and induced apoptosis, which is regulated by Notch pathway [52]. At the same time, to characterize of the proteins that participate the transcriptional effects of DNA methylation in CD8+ T cells needs to be further clarified.

The ability to modulate the function of T cells through epigenetic regulation has important therapeutic implications. As a reader of acetylated lysines, BRD4, and the histone deacetylase sirtuin 1 (SIRT1), can be inhibit by JQ1 that is a pharmacological inhibitor of the BET family of bromodomain-containing proteins. Mechanistically, JQ1 reduces BATF expression, increases proliferation and cytokine production of CD8+ T [53, 54]. There are more epigenetic regulators that participat in the differentiation and function of CD8+ T cell, which need more exploration.

Mechanisms of CD8+ T cells metabolic regulation

It is universally accepted that the functions of T cells activation, differentiation and effector are basic in T cell biology, which are closely related to changes in the cellular metabolic programs. Metabolic pathways such as glycolysis, fatty acid synthesis and mitochondrial metabolism play significant roles in T cell immunometabolism [28]. In healthy persons, metabolically quiescent T cells reside in lymph nodes and peripheral tissues in order to recognize antigens. Once infection, T cells are activated in a specific manner to become effector T cells such as proliferate and/or differentiate which are accompanied by important changes in cellular metabolism, and this progress can be defined as metabolic reprogramming [5]. At the same time, the shift in energy production is accompanied by mitochondrial ultrastructural modifications that facilitate the metabolic transition [55]. Hypoxia-induced mitochondrial remodeling can also promote T cell exhaustion, reducing antitumor immunity. Up to date, metabolic pathways have been manipulated to treat immune-dysregulatory diseases. One of the therapies is rapamycin (sirolimus) that targets PI3K/Akt and glucose transporter 1(GLUT1) to regulate mTOR under the stimulation of TCR. Leucine, glutamine, and arginine also regulate mTOR expression; in patients with atopy due to CARD11 loss-of-function (LOF). Glutamine supplementation can promote Th1 differentiation through mTOR, rescuing the atopic T cell phenotype [56]. The metabolic profile of T cells is complexly linked to their differentiation state and have a considerable impact on the generation and duration of effector T cell activity [57]. Here, we discuss in detail the metabolic states of CD8+ T cells providing a guide of therapeutic basis for cancers.

CD8+ T cell metabolism from quiescence to activation

T cell metabolism is indispensable not only for priming cells for rapid activation,

but also maintaining homeostasis in naive and memory cells. Metabolic progress regulates T cell quiescence. Naive T cells have lower mitochondrial activity and glucose uptake, and produce ATP through mitochondrial OXPHOS29 to support T cell homeostasis which is different from antigen-stimulated T cells [58,59,60]. At the same time, T cell metabolism promotes and on behalf of the activation and differentiation and modifies gene transcription and post-transcriptional regulation [61, 62]. Naive T cells enter peripheral tissue from thymus and are actively maintained during cell cycle by combining TCR/CD3 and stimulated by IL-7 [63]. Activation of T cells show clonal expansion, cell growth and differentiation which is mediated by cell surface receptors, oxygen levels and nutrient availability [64]. The level of acute T cell activation relies on the co-stimulation receptors such as CD3, CD4, CD8, CTLA4 etc. and the activation of TCR [65]. TCR recognizes CD4/CD8 co-receptors and MHC-peptide complex that upregulate lymphocyte-specific protein kinase (Lck) and protein tyrosine kinase (PTK) C-terminal Src kinase (Csk), then induces the activation, recruitment and phosphorylation of the zeta-chain associated protein kinase 70 (ZAP70). ZAP70 upregulates phospholipase Cg1 (PLCg1), which stimulates calcium mobilization, activates protein kinase C (PKC) and Ras pathway [66,67,68]. When naive T cells transform to activated T cells, the metabolic activity is from low to high [69, 70]. Activated T cells rely on nutrient uptake. AMP-dependent protein kinase (AMPK), as the energy sensor in T cells can be activated by calcium calmodulin dependent protein kinase 2 (CaMKK2) and low levels of ATP and liver kinase B1 (LKB1)-dependent phosphorylation [71,72,73]. Meanwhile, TCR/CD3-CD28 signaling participated in mitochondrial biogenesis, which prepared for T cell proliferation and growth. Activated TCR/CD3-CD28 signaling phosphorylated PI3K-Akt -mTOR1/2 pathway, too, which subsequently regulated the important upstream regulator GTPase, tuberous sclerosis complex 2 (TSC2) [72, 74].

In addition to nutrient availability and cell surface receptors, oxygen tension is also necessary for T cell metabolism. T cells are mobile through obtaining a suitable aerobic environment in the body. Naive T cells are in a low oxygen environment and activated T cells are exposed in high oxygen levels in the arterial blood and lung as well as the hypoxic conditions in tumors and inflammation sites [75,76,77]. The state of T cells is influenced by exposure to hypoxia that mainly mediated by HIF-1α, which translocates into the nucleus to bind hypoxia response elements (HREs) [78, 79]. HIF-1α mediates metabolic shift by regulating the expression of genes include GLUT1, HK2, PKM2, LDHA (Fig. 2A) [79,80,81]. Different metabolic programs define different T cell subsets, and T cells state can be manipulated via modulating metabolic activity. However, the mechanism of how metabolism influences the function of T cell in response to infections and tumors are not fully understood and whether the normal functions of T cells can be restored by regulating metabolism needs to be further elucidated.

Fig. 2
figure 2

CD8+ T cell metabolism. A normal CD8+ T cells metabolism. CD8+ T cell metabolism regulated by lots of kinases, eventually producing ATP and GTP. B CD8+ T cells metabolism in an inflammatory environment (fat, diabetes…). Proinflammatory factors regulate CD8+ T cells metabolism through the PI3K/AKT/mTOR pathway and induce CD8+ T cells to produce CXCR3, then CD8 + T cells dysfunction. C CD8+ T cells metabolism in virus environment. The virus enters CD8 + T cells through the ACE/NRP receptors, alters the metabolism of CD8 + T cells, and causes apoptosis. D Tumor cells and CD8+ T cells compete for body nutrients, in addition, harmful substances are released from tumor cells, which cause dysfunction and exhaustion of CD8+ T cells. HRE hypoxia response elements, GLUT glucose transporter, mTOR mammalian target of rapamycin, TCA tricarboxylic acid cycle, 5-HTTP 5-hydroxytryptophan, TCR T cell receptor

CD8+ T cell metabolism in cancer

There is a bidirectional relationship between the occurrence and development of tumors and tumor microenvironment (TME) where include stromal, endothelial, immune, tumor cells, cytokines and chemokines (Figs. 2D and 3) [82]. Accumulating evidences clarify that the regulation of metabolism in TME are associated with the function of T cell and tumor cells, and play a main role in shaping anticancer immune responses [83]. Tumor cells compete with T cells for nutrients to meet their needs for proliferation and migration, under aerobic conditions. Tumor cells preferentially utilize glycolysis and metabolize approximately tenfold more glucose to lactate than normal tissues, this phenomenon is defined Warburg effect (Fig. 2D) [84]. We will discuss glucose and amino acids metabolism in next.

Fig. 3
figure 3

CD8+ T cell interaction with other immune cells in tumor microenvironment. A the check and balance between immune system and tumors. The three phases between immune system and tumors, elimination, equilibrium and escape, respectively. B CD8+ T cell in TME. There are a variety of cells and cytokines in TME, that crosstalk with each other to affect the function of CD8+ T cells, and M1 is tumor-suppressing, M2 is tumor-promoted. CD4 + T cell and DCs are stimulators for CD8+ T cells. C when immune system is out of control and defeated, then cancer outgrowth. M1 M1 type macrophages, M2 M2 type macrophages, DC dendritic cell, TME tumor microenvironment

Glucose participates in T cell proliferation, function and regulates cell fate. Both effector T cells and hyperactive cancer cells are heavily dependent on glucose metabolism. Glycolysis is important for sustaining effector T cell immune function such as the secretion of IFN-γ. Glucose deprivation selectively inhibited the production of IFN-γ, granzyme B protein, cyclin D2 protein, cytolytic activity [82, 85]. In some glycolytic tumors, CD8+ T cell proliferation and infiltration are very low, which associate with tumor cells that limit the energy metabolism of CD8+ T cells. As the glycolysis enzyme, GAPDH binds the AU-rich region in the 3′ untranslated regions of cytokine messenger RNAs and downregulate the expression of protein [86,87,88]. T cell function is impaired through mTOR under low levels of glucose, and the transcriptional level of IFN-γ is diminished under the background of lowed activity of mTOR and phosphorylation of the ribosomal protein S6 kinase beta-1 (p70S6K) [89].

In addition to glucose, the accumulation of the glycolytic product lactate is negative for effector T cell function and antitumor effect, and lactate impairs CD8+ T cell and NK-cell infiltration and activity in melanoma [90]. However, CD8+ T cell seems smart and flexible in such hostile environment. CD8+ T cells upregulate the catabolism of fatty acid so that provide energy for preserving effector function in TME and the activation of peroxisome proliferator receptors is positive for T cell function and delays tumor growth [91]. Additionally, acetate can rescue IFN-γ production via upregulating chromatin accessibility and histone acetylation in glucose-limited T cells [92]. Thus, some metabolic targets are potential to rescue CD8+ T cell function in a hostile environment.

Except glucose metabolism, amino acids are indispensable for T cell function and differentiation, too. Green and Frauwirth reported that glutamine as an important source for active T cells was regulated by ERK/MAPK [93, 94]. Glutaminase deficiency abolished T cell activation and Th17 differentiation, but promoted CD4+ Th1 and CD8+ CTL cells differentiation and effector function via T-bet [95]. In addition, dynamic proteomic and metabolomic analysis identified that l-arginine is a key metabolite which promotes OXPHOS, boosts T cell survival and generates antitumor memory-like T cells [96]. Under the persistent stimulation of IL-2, STAT5 is activated and then induces strong expression of tryptophan hydroxylase 1, thus catalyze the conversion to tryptophan to 5-hydroxytryptophan (5-HTP), which upregulate inhibitory receptors expression, thereby rendering CD8+. T cells dysfunctional in the TME [97]. Tumor cells methionine consumption is an immune evasion mechanism. Reducing methyl donor S-adenosylmethionine (SAM) and methionine result in loss of dimethylation at lysine 79 of histone H3 (H3K79me2), which led to decreased expression of STAT5 and impaired CD8 + T cell immunity [98].

The specific impact of glucose and amino acid on CD8+ T cells fate and function thus suggest the possibility of immunomodulation within the TME via the manipulation of glucose and amino acid levels. The balance of this competition has been linked to the activity of metabolic enzymes, and metabolic enzymes could be a potential and effective target for immune therapy. While methotrexate is the oldest but still one of the most effective available chemotherapeutic treatments in clinical, it’s necessary to explore more antitumor drugs that target metabolism so as to improve the outcome from clinical treatment [99, 100]. At the same time, metabolism treatment combined with immune treatment are expected to lead to novel and highly specific targets. As it shown in Tables 2 and 3, the clinical trials involving CD8+ T cells are concluded.

Table 2 Anti-PD-1/PD-L1 combinational therapy and applications
Table 3 Clinical trials related to CD8+ T cells

To sum up, TCR activates CD8+ T cells to induce the transfer of cellular metabolic levels to glycolysis, and the synergistic effect of CD28 leads to an upregulation of CD8+ T cell glycolysis levels, further supporting their subsequent proliferation and differentiation; The induction of high glycolytic activity in CD8+ T cells are beneficial for CD8+ T cells to differentiate into effector cells, but seriously impairs the survival of long-lived memory cells; For effector CD8+ T cells, changes in glycolysis are related in IFN- γ production, the downregulation of glycolysis levels plays an important role in the production of cytokines and immune function. Therefore, it is crucial to find a method for targeted glycolysis to restore the effector function of CD8+ T cells; The metabolic imbalance caused by the changes of glycolytic activity not only affects the function of CD8+ T cells, but also affects their effector function. The enhanced selectivity of glycolysis will further restore these functions; Glycolysis may affect partial depletion of CD8+ T cells through the mTOR pathway and affect IFN- γ, the occurrence of adverse effects. In addition, the balance between glycolysis and fatty acid oxidation (FAO) is related to the long-term survival of memory CD8 T cells.

Cross-talk between CD8+ T cells and other immune cells in tumor microenvironment

Effector CD8+ T cells are thought to be a homogenous group of cytotoxic cells that produce protease granzyme B, IFN-γ and multiple subsets of CD8+ T cells have distinct effects and cytotoxic potential [101]. CD8+ T cells can be discovered in TME, where they potentially influence the antitumor response and patient outcomes. We have described the metabolism of CD8+ T cells in TME in the previous section, and next we will discuss the crosstalk between CD8+ T cells and other immune cells.

CD4+ T cell

Tumor outgrowth is controlled by CD4 and CD8 T cells, there are three phases of tumor-immunity, namely elimination, equilibrium and escape (Fig. 3A) [102]. Studies about chronic viral infection and cancer have shown that CD4 T cells are necessary for CD8 T cell function, CD4+ T cells are dispensable for primary expansion and cytotoxic effectors of CD8+ T cells [103, 104]. From single cell RNA-seq, Zander and colleagues show that the formation of effector CD8+ T cells is critically dependent on CD4+ T cell under the function of IL-21 and the pathway could be used therapeutically to enhance the killer function of CD8+ T cells infiltrating into the tumor [105]. As for lung adenocarcinoma, the differentiation of tumor-specific CD4+ T follicular helper cells under the stimulation of B cells in a neoantigen-dependent manner, which promote CD8+ T cell effector functions and drive anti-tumor immunity [106]. In terms of the mechanism, Ahrends et al. revealed that CD4+ T cells help effector CD8+ T cells acquire their ability that involves the downregulation of PD-1 and increased motility and migration capacities. In a similar study, CD4+ T cell is beneficial for the antigen-specific CD8+ T cells clonal expansion and IFN-γ production, too [107, 108]. Microarray analysis demonstrated that without the help of CD4+ T cells, CD8+ T cells expressed elevated the levels of inhibitory receptors such as PD1, exhibited transcriptomic exhaustion and anergy profiles change [109]. CD4+ T cell help the TCR repertoire. CD27 directs the expression of the Pim1 gene and the antiapoptotic Bcl-2 that promotes the survival of CD8+ T cells and thereby increases the function of effector and memory populations, but more TCR repertoire of responder CTLs should be explored about how to prevent immune escape of tumor cells [110, 111]. In addition, the tumor-invasive capacity of CTLs can be promoted by CD4+ T cell [112]. So, we can summarize that CD4+ T cells are significant for the differentiation, effector function, antitumor of CD8+ T cells in TME, and it might be important for tumor immunotherapies (Fig. 3B).

Dendritic cell

As the most potent professional APCs, DCs play a core role in linking innate and adaptive immune responses and in the balance of CD8 T cell immunity and tolerance to tumor antigens [113]. The functions of DCs include uptake, processing and presenting antigens to activate naive antigen-specific CD4 and CD8 T cells [114]. Activated DCs can produce IL-12, which mediate Th1 differentiation and provide essential signals for the production of resident memory CD8+ T cells in human and mice [115,116,117,118]. DCs can provide a friendly extracellular microenvironment for T lymphocyte activation, and DCs-derived IL-15 can promote CTL differentiation (Fig. 3B) [119, 120]. Batf3, also known as Jun dimerization protein p21SNFT, is important for DCs. Hildner and colleagues clarified that the abilities of cross-presentation and antitumor immunity were impaired in Batf3−/− mice, which downregulated CD8 T cell-mediated anti-tumor immunity [121]. Broz et al. identified that CD103+ DCs not only induce the proliferation of naive CD8+ T cells, but also establish CTLs in the TME, and it’s the mediator that transport solid tumor antigens from TME to tumor draining lymph nodes for CD8+ T cells. The Cancer Genome Atlas (TCGA) database analysis indicated that CD103+/CD103 is strongly correlates with cancerous patients survival [122,123,124]. From the study about melanoma, the activation of β-catenin signaling reduces the numbers of intratumoral CD103+ DCs that prevent tumor-specific T cell priming and anti-tumor immunity, at the same time, CD103+ DCs are also critical target for the efficacy of immunotherapy with PD-L1 and Braf inhibition [123, 125]. The number of tumor infiltrated CD8+ CD103+ TRM cells have been identified correlating with prolonged survival and better prognosis in ovarian, endometrial, breast and lung cancer [126,127,128,129,130]. It’s worth noting that Spranger et al. have shown that vaccination with DCs improved the efficacy of anti-PD-L1 and anti-CTLA-4 immunotherapy [125]. To sum up, we need to further explore the role of DCs and subsets on other T cells, and further clarify how, when and where DCs present tumor antigens to interact with CD8+ T cells. DCs offer an opportunity to manipulate CD8+ T cells and vaccine to generate anti-tumor immunity in the TME, it will be a promising target for tumor therapy.

Macrophages

Macrophages are highly multifunctional and plastic cells that participate in tissue development, homeostasis, clearance of cellular debris, elimination of pathogens, regulation of inflammatory responses and tumors. It is generally simplified into two categories: M1 or M2 macrophages (Fig. 3B) [131, 132]. Generally speaking, M1 macrophages have the anti-tumor roles, and M2 macrophages promote the occurrence and development of tumors. Specifically, M1 macrophages have two different effects, one is directly mediate cytotoxicity to kill tumor cells and another is antibody dependent cell mediated cytotoxicity (ADCC), which is faster than cytotoxicity [133]. Tumor-associated macrophages (TAMs) participate in the regulation of the TME. TAMs are widely present in various tumors, secreting a variety of cytokines such as epithelial growth factor (EGF), platelet-derived growth factor (PDGF), TGF, hepatocyte growth factor (HGF), and epithelial growth factor receptor (EGFR) family, and correlate with tumor growth, invasion, metastasis, and treatment-ineffectiveness [134,135,136]. Studies revealed that Treg cells can depress IFN-γ secreted by CD8+ T cells to promote the polarization of M2-like TAMs [137]. Intravital imaging studies shown that antigen-specific CD8+ T cells preferentially localize in TAM-rich areas in the TME [122, 138, 139].

TAMs negatively regulate T cell activation and hinder CD8+ T cell reaching tumor cells that limit the efficacy of anti-PD-1 treatment. Combinational treatment of anti-PD-1 with PLX3397, an inhibitor of colony-stimulating factor-1 receptor (CSF-1R), increases the accumulation of CD8+ T cells in malignant cells and delays tumor progression [139]. Zhang et al. reported that IL-15Rc/HIF-1α/CX3CL1 signal pathway serves as a crosstalk between macrophages and CD8+ T cells. IL-15Rα+ TAMs reduce the levels of CX3CL1 to reduce CD8+ T recruitment through releasing the IL-15/IL-15Rα complex in the TME [140]. The expression of IL-10 by macrophages depresses IL-12, which is produced by intratumoral DCs that suppress CD8+ T cells and response to paclitaxel and carboplatin, while, IL-10R blockade increase the expression of IL-12 and improve the treatment outcomes [115]. In the same experiment, Petty and colleges reported that hedgehog signal is critical for TAM M2 polarization and tumor growth that suppresses CD8+ T cell recruitment to the TME through the inhibition of CXCL9 and CXCL10 production [141]. In contrast, FOLR2+ tissue-resident macrophages are positively correlated with tumor immunity, efficiently prime effector CD8+ T cells, and better patient survival [142]. These studies highlight specific roles for TMEs and its subsets for targeted therapeutic interventions in macrophages-based cancer therapies, and macrophages could be a promising aim for immune therapy.

The composition and function of TME are also undergoing dynamic changes during the development of cancer. Through this review, we recognize the enormous complexity and interconnectedness of TME, as well as its diversity in different organs and patients. Targeted therapy of cells, biological processes, and signaling pathways in TME are considered promising strategies that can be extended to all types of cancer. The large number of co-immune and stromal cells found in TME are genetically stable, making them easier to target compared to cancer cells with unstable genomes [143, 144]. For example, standard treatments including chemotherapy and radiation therapy can cause changes in TME, regulating its therapeutic effect in an external manner to cancer cells, enhancing or interfering with the response. It is worth noting that adaptability and intrinsic resistance may be obstacles to targeted treatment of TME. Despite these challenges, there is great hope for expanding treatment strategies targeting TME, including depletion or "reprogramming" of cancer promoting host cells in TME; Intervention measures to modify extracellular matrix (ECM), matrix components, and extracellular vesicles (EVs); Cell based therapies and vaccines; And immune checkpoint inhibitors. Moreover, integrating multiple cancer model data and advanced computational analysis, including artificial intelligence, has the potential to adopt a comprehensive system level approach about analyzing and integrating all the complexities of TME to identify key nodes [145]. In addition, significant advances in bioengineering will provide a platform for large-scale testing, such as in ex vivo organoids and tissue slices that accurately recurrent organ specific TMEs [146,147,148].

Conclusions and prospective

By RNA sequencing (RNA-seq) and transposase-accessible chromatin sequencing (ATAC-seq), Pritykin defined a dysfunction and underlying transcriptional drivers and revealed a state of functional and dysfunctional T cell across cancer and infection models [149]. Zheng et al. analyzed T cell populations from hepatocellular carcinoma (HCC), and revealed distinct subtypes and clonal expansion of infiltrating lymphocytes [150]. A better understanding of CD8+ T cell clustering, dynamic, markers and developmental trajectory will provide more therapeutic strategies for diseases. In this review, we discuss CD8+ T cells development, metabolism and interaction with tumor microenvironment.

In past few years, novel checkpoint blockades have given some attention in the treatment of multiple solid cancers. Antibodies targeting inhibitory receptors including PD-1 successfully increase T cell function and clinical efficacy in tumors. As summarized in Table 4, the treatment of PD-1 combined with chemotherapy, radiotherapy, targeted therapy and cytokines make a promising outcome [151, 152]. It is amazing that cancer vaccines have also attracted lots of interest, and Sipuleucel-T and T-VEC have been approved by FDA [153, 154]. However, cancer vaccines have not shown desired results in several clinical trials, mainly because that cancer vaccines can’t effectively active T cell and the safety of the vaccine deserves further investigation. The reasons of CD8+ T cells exhaustion have been explored, and we need to clarify more biomarkers to predict CD8+ T cells exhaustion and status, the most meaningful question is whether and how the exhaustion of CD8+ T cells can be reversed. Additional combinatorial strategies should be considered, for example, antiangiogenic inhibitors targetingvascular endothelial growth factor (VEGF), which can promote infiltration of immunostimulatory cells, block immunosuppressive effects in the TME, and improve drug delivery [155]. Recombination of clinically approved treatment methods or conversion of “cold tumor” to “hot tumor” through different drug administration is an inspiring goal. Together, existing and future approaches are helpful for understanding CD8+ T cells, and we should be optimistic about therapy that will be applied to human diseases.

Table 4 In vivo experiments related to CD8+ T cells

Availability of data and materials

All data are available in the main text.

Abbreviations

MHC-I:

Major histocompatibility complex

APCs:

Antigen presenting cells

TNF:

Tumor necrosis factor

PD-1:

Programmed death receptor 1

PD-L1:

Programmed cell death 1 ligand 1

Eomes:

Eomesodermin

mTOR:

Mammalian target of rapamycin

TCR:

T cell receptor

IFN:

Interferon

DCs:

Dendritic cells

LCMV:

Lymphocytic choriomeningitis virus

TGF-β:

Transforming growth factor β

AMPK:

AMP-dependent protein kinase

HREs:

Hypoxia response elements

GLUT1:

Glucose transporter 1

CTLs:

Cytotoxic T lymphocytes

TME:

Tumor microenvironment

LAG3:

Lymphocyte-activation gene 3

Tim-3:

T cell immunoglobulin domain and mucin domain 3

TIGIT:

Ig and ITIM domain

CTLs:

Cytotoxic T lymphocytes

TAMs:

Tumor-associated macrophages

HCC:

Hepatocellular carcinoma

NK:

Natural killer cells

SLECs:

Short-lived effector cells

TEs:

Terminal effector cells

MPs:

Memory precursors

TILs:

Tumor-infiltrating lymphocytes

FAO:

Fatty acid oxidation

EVs:

Extracellular vesicles

ECM:

Extracellular matrix

VEGF:

Vascular endothelial growth factor

References

  1. Taniuchi I. CD4 helper and CD8 cytotoxic T cell differentiation. Annu Rev Immunol. 2018;36:579–601.

    Article  CAS  PubMed  Google Scholar 

  2. Henning AN, Roychoudhuri R, Restifo NP. Epigenetic control of CD8+ T cell differentiation. Nat Rev Immunol. 2018;18:340–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Laurenti E, Göttgens B. From haematopoietic stem cells to complex differentiation landscapes. Nature. 2018;553:418–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Troha K, Ayres JS. Metabolic adaptations to infections at the organismal level. Trends Immunol. 2020;41:113–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Reina-Campos M, Scharping NE, Goldrath AW. CD8(+) T cell metabolism in infection and cancer. Nat Rev Immunol. 2021;21:718–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Yi M, et al. Combination strategies with PD-1/PD-L1 blockade: current advances and future directions. Mol Cancer. 2022;21:28.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Henning AN, Roychoudhuri R, Restifo NP. Epigenetic control of CD8(+) T cell differentiation. Nat Rev Immunol. 2018;18:340–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kaech SM, Cui W. Transcriptional control of effector and memory CD8+ T cell differentiation. Nat Rev Immunol. 2012;12:749–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Buchholz VR, Schumacher TN, Busch DH. T cell fate at the single-cell level. Annu Rev Immunol. 2016;34:65–92.

    Article  CAS  PubMed  Google Scholar 

  10. Williams MA, Bevan MJ. Effector and memory CTL differentiation. Annu Rev Immunol. 2007;25:171–92.

    Article  CAS  PubMed  Google Scholar 

  11. Restifo NP, Gattinoni L. Lineage relationship of effector and memory T cells. Curr Opin Immunol. 2013;25:556–63.

    Article  CAS  PubMed  Google Scholar 

  12. Teixeiro E, et al. Different T cell receptor signals determine CD8+ memory versus effector development. Science. 2009;323:502–5.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  14. Chen X, et al. A membrane-associated MHC-I inhibitory axis for cancer immune evasion. Cell. 2023;186:3903-3920.e21.

    Article  CAS  PubMed  Google Scholar 

  15. Do JS, Valujskikh A, Vignali DA, Fairchild RL, Min B. Unexpected role for MHC II-peptide complexes in shaping CD8 T-cell expansion and differentiation in vivo. Proc Natl Acad Sci U S A. 2012;109:12698–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Szabo SJ, et al. A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell. 2000;100:655–69.

    Article  CAS  PubMed  Google Scholar 

  17. Szabo SJ, et al. Distinct effects of T-bet in TH1 lineage commitment and IFN-gamma production in CD4 and CD8 T cells. Science. 2002;295:338–42.

    Article  CAS  PubMed  Google Scholar 

  18. Joshi NS, et al. Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity. 2007;27:281–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Intlekofer AM, et al. Effector and memory CD8+ T cell fate coupled by T-bet and eomesodermin. Nat Immunol. 2005;6:1236–44.

    Article  CAS  PubMed  Google Scholar 

  20. Pearce EL, et al. Control of effector CD8+ T cell function by the transcription factor Eomesodermin. Science. 2003;302:1041–3.

    Article  CAS  PubMed  Google Scholar 

  21. Takemoto N, Intlekofer AM, Northrup JT, Wherry EJ, Reiner SL. Cutting Edge: IL-12 inversely regulates T-bet and eomesodermin expression during pathogen-induced CD8+ T cell differentiation. J Immunol. 2006;177:7515–9.

    Article  CAS  PubMed  Google Scholar 

  22. Sullivan BM, Juedes A, Szabo SJ, von Herrath M, Glimcher LH. Antigen-driven effector CD8 T cell function regulated by T-bet. Proc Natl Acad Sci U S A. 2003;100:15818–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Juedes AE, Rodrigo E, Togher L, Glimcher LH, von Herrath MG. T-bet controls autoaggressive CD8 lymphocyte responses in type 1 diabetes. J Exp Med. 2004;199:1153–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Intlekofer AM, et al. Anomalous type 17 response to viral infection by CD8+ T cells lacking T-bet and eomesodermin. Science. 2008;321:408–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Levine AG, et al. Stability and function of regulatory T cells expressing the transcription factor T-bet. Nature. 2017;546:421–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Chen Z, et al. TCF-1-centered transcriptional network drives an effector versus exhausted CD8 T cell-fate decision. Immunity. 2019;51:840-855.e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. McLane LM, et al. Role of nuclear localization in the regulation and function of T-bet and Eomes in exhausted CD8 T cells. Cell Rep. 2021;35: 109120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Iwata S, et al. The transcription factor T-bet limits amplification of type I IFN transcriptome and circuitry in T helper 1 cells. Immunity. 2017;46:983-991.e4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Townsend MJ, et al. T-bet regulates the terminal maturation and homeostasis of NK and Valpha14i NKT cells. Immunity. 2004;20:477–94.

    Article  CAS  PubMed  Google Scholar 

  30. Knox JJ, Cosma GL, Betts MR, McLane LM. Characterization of T-bet and eomes in peripheral human immune cells. Front Immunol. 2014;5:217.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Buggert M, et al. T-bet and Eomes are differentially linked to the exhausted phenotype of CD8+ T cells in HIV infection. PLoS Pathog. 2014;10: e1004251.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Carty SA, Koretzky GA, Jordan MS. Interleukin-4 regulates eomesodermin in CD8+ T cell development and differentiation. PLoS ONE. 2014;9: e106659.

    Article  PubMed  PubMed Central  Google Scholar 

  33. McLane LM, et al. Differential localization of T-bet and Eomes in CD8 T cell memory populations. J Immunol. 2013;190:3207–15.

    Article  CAS  PubMed  Google Scholar 

  34. Rao RR, Li Q, Odunsi K, Shrikant PA. The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and Eomesodermin. Immunity. 2010;32:67–78.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Li G, et al. T-bet and eomes regulate the balance between the effector/central memory T cells versus memory stem like T cells. PLoS ONE. 2013;8: e67401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Banerjee A, et al. Cutting edge: The transcription factor eomesodermin enables CD8+ T cells to compete for the memory cell niche. J Immunol. 2010;185:4988–92.

    Article  CAS  PubMed  Google Scholar 

  37. Mishra S, et al. TGF-beta and Eomes control the homeostasis of CD8+ regulatory T cells. J Exp Med. 2021;218.

  38. Weulersse M, et al. Eomes-dependent loss of the co-activating receptor CD226 restrains CD8(+) T cell anti-tumor functions and limits the efficacy of cancer immunotherapy. Immunity. 2020;53:824-839.e10.

    Article  CAS  PubMed  Google Scholar 

  39. Fonseca R, et al. Runx3 drives a CD8(+) T cell tissue residency program that is absent in CD4(+) T cells. Nat Immunol. 2022;23:1236–45.

    Article  CAS  PubMed  Google Scholar 

  40. Tsao HW, et al. Batf-mediated epigenetic control of effector CD8+ T cell differentiation. Sci Immunol. 2022;7:eabi4919.

    Article  CAS  PubMed  Google Scholar 

  41. Milner JJ, et al. Runx3 programs CD8(+) T cell residency in non-lymphoid tissues and tumours. Nature. 2017;552:253–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Guo A, et al. cBAF complex components and MYC cooperate early in CD8(+) T cell fate. Nature. 2022;607:135–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Cruz-Guilloty F, et al. Runx3 and T-box proteins cooperate to establish the transcriptional program of effector CTLs. J Exp Med. 2009;206:51–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ball MP, et al. Targeted and genome-scale strategies reveal gene-body methylation signatures in human cells. Nat Biotechnol. 2009;27:361–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13:484–92.

    Article  CAS  PubMed  Google Scholar 

  46. Scharer CD, Barwick BG, Youngblood BA, Ahmed R, Boss JM. Global DNA methylation remodeling accompanies CD8 T cell effector function. J Immunol. 2013;191:3419–29.

    Article  CAS  PubMed  Google Scholar 

  47. Kersh EN. Impaired memory CD8 T cell development in the absence of methyl-CpG-binding domain protein 2. J Immunol. 2006;177:3821–6.

    Article  CAS  PubMed  Google Scholar 

  48. Di Croce L, Helin K. Transcriptional regulation by Polycomb group proteins. Nat Struct Mol Biol. 2013;20:1147–55.

    Article  PubMed  Google Scholar 

  49. Heffner M, Fearon DT. Loss of T cell receptor-induced Bmi-1 in the KLRG1(+) senescent CD8(+) T lymphocyte. Proc Natl Acad Sci USA. 2007;104:13414–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Jacobs JJ, Kieboom K, Marino S, DePinho RA, van Lohuizen M. The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus. Nature. 1999;397:164–8.

    Article  CAS  PubMed  Google Scholar 

  51. Sherr CJ. Principles of tumor suppression. Cell. 2004;116:235–46.

    Article  CAS  PubMed  Google Scholar 

  52. Zhao E, et al. Cancer mediates effector T cell dysfunction by targeting microRNAs and EZH2 via glycolysis restriction. Nat Immunol. 2016;17:95–103.

    Article  CAS  PubMed  Google Scholar 

  53. Kuroda S, et al. Basic leucine zipper transcription factor, ATF-like (BATF) regulates epigenetically and energetically effector CD8 T-cell differentiation via Sirt1 expression. Proc Natl Acad Sci U S A. 2011;108:14885–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kagoya Y, et al. BET bromodomain inhibition enhances T cell persistence and function in adoptive immunotherapy models. J Clin Invest. 2016;126:3479–94.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Buck MD, et al. Mitochondrial dynamics controls T cell fate through metabolic programming. Cell. 2016;166:63–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Chopp L, Redmond C, O’Shea JJ, Schwartz DM. From thymus to tissues and tumors: a review of T-cell biology. J Allergy Clin Immunol. 2023;151:81–97.

    Article  CAS  PubMed  Google Scholar 

  57. Kinnaird A, Zhao S, Wellen KE, Michelakis ED. Metabolic control of epigenetics in cancer. Nat Rev Cancer. 2016;16:694–707.

    Article  CAS  PubMed  Google Scholar 

  58. Baixauli F, et al. Mitochondrial respiration controls lysosomal function during inflammatory T cell responses. Cell Metab. 2015;22:485–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Rathmell JC, Vander Heiden MG, Harris MH, Frauwirth KA, Thompson CB. In the absence of extrinsic signals, nutrient utilization by lymphocytes is insufficient to maintain either cell size or viability. Mol Cell. 2000;6:683–92.

    Article  CAS  PubMed  Google Scholar 

  60. Tarasenko TN, et al. Cytochrome C oxidase activity is a metabolic checkpoint that regulates cell fate decisions during T cell activation and differentiation. Cell Metab. 2017;25:1254–1268.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. MacIver NJ, Michalek RD, Rathmell JC. Metabolic regulation of T lymphocytes. Annu Rev Immunol. 2013;31:259–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. O’Neill LA, Kishton RJ, Rathmell J. A guide to immunometabolism for immunologists. Nat Rev Immunol. 2016;16:553–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Chapman NM, Boothby MR, Chi H. Metabolic coordination of T cell quiescence and activation. Nat Rev Immunol. 2020;20:55–70.

    Article  CAS  PubMed  Google Scholar 

  64. Chapman NM, Chi H. Hallmarks of T-cell Exit from Quiescence. Cancer Immunol Res. 2018;6:502–8.

    Article  CAS  PubMed  Google Scholar 

  65. Smith-Garvin JE, Koretzky GA, Jordan MS. T cell activation. Annu Rev Immunol. 2009;27:591–619.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Mariuzza RA, Agnihotri P, Orban J. The structural basis of T-cell receptor (TCR) activation: an enduring enigma. J Biol Chem. 2020;295:914–25.

    Article  PubMed  Google Scholar 

  67. Williams BL, et al. Phosphorylation of Tyr319 in ZAP-70 is required for T-cell antigen receptor-dependent phospholipase C-gamma1 and Ras activation. EMBO J. 1999;18:1832–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Wang H, et al. ZAP-70: an essential kinase in T-cell signaling. Cold Spring Harb Perspect Biol. 2010;2: a002279.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Menk AV, et al. Early TCR signaling induces rapid aerobic glycolysis enabling distinct acute T cell effector functions. Cell Rep. 2018;22:1509–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Wik JA, Skalhegg BS. T cell metabolism in infection. Front Immunol. 2022;13: 840610.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Racioppi L, Means AR. Calcium/calmodulin-dependent protein kinase kinase 2: roles in signaling and pathophysiology. J Biol Chem. 2012;287:31658–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Tamas P, et al. Regulation of the energy sensor AMP-activated protein kinase by antigen receptor and Ca2+ in T lymphocytes. J Exp Med. 2006;203:1665–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hardie DG, Sakamoto K. AMPK: a key sensor of fuel and energy status in skeletal muscle. Physiology (Bethesda). 2006;21:48–60.

    CAS  PubMed  Google Scholar 

  74. Mondino A, Mueller DL. mTOR at the crossroads of T cell proliferation and tolerance. Semin Immunol. 2007;19:162–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Braun RD, Lanzen JL, Snyder SA, Dewhirst MW. Comparison of tumor and normal tissue oxygen tension measurements using OxyLite or microelectrodes in rodents. Am J Physiol Heart Circ Physiol. 2001;280:H2533–44.

    Article  CAS  PubMed  Google Scholar 

  76. Hale LP, Braun RD, Gwinn WM, Greer PK, Dewhirst MW. Hypoxia in the thymus: role of oxygen tension in thymocyte survival. Am J Physiol Heart Circ Physiol. 2002;282:1467–77.

    Article  Google Scholar 

  77. Thurnher M, Gruenbacher G. T lymphocyte regulation by mevalonate metabolism. Sci Signal. 2015;8:4.

    Article  Google Scholar 

  78. McNamee EN, Korns Johnson D, Homann D, Clambey ET. Hypoxia and hypoxia-inducible factors as regulators of T cell development, differentiation, and function. Immunol Res. 2013;55:58–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Tao JH, Barbi J, Pan F. Hypoxia-inducible factors in T lymphocyte differentiation and function. A review in the theme: cellular responses to hypoxia. Am J Physiol Cell Physiol. 2015;309:580–9.

    Article  Google Scholar 

  80. Serganova I, et al. LDH-A regulates the tumor microenvironment via HIF-signaling and modulates the immune response. PLoS ONE. 2018;13: e0203965.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Kim JW, Tchernyshyov I, Semenza GL, Dang CV. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 2006;3:177–85.

    Article  PubMed  Google Scholar 

  82. Elia I, Haigis MC. Metabolites and the tumour microenvironment: from cellular mechanisms to systemic metabolism. Nat Metab. 2021;3:21–32.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Siska PJ, Rathmell JC. T cell metabolic fitness in antitumor immunity. Trends Immunol. 2015;36:257–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Koppenol WH, Bounds PL, Dang CV. Otto Warburg’s contributions to current concepts of cancer metabolism. Nat Rev Cancer. 2011;11:325–37.

    Article  CAS  PubMed  Google Scholar 

  85. Cham CM, Driessens G, O’Keefe JP, Gajewski TF. Glucose deprivation inhibits multiple key gene expression events and effector functions in CD8+ T cells. Eur J Immunol. 2008;38:2438–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Singer K, et al. Warburg phenotype in renal cell carcinoma: high expression of glucose-transporter 1 (GLUT-1) correlates with low CD8(+) T-cell infiltration in the tumor. Int J Cancer. 2011;128:2085–95.

    Article  CAS  PubMed  Google Scholar 

  87. Cascone T, et al. Increased tumor glycolysis characterizes immune resistance to adoptive T cell therapy. Cell Metab. 2018;27:977-987.e4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Chang CH, et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell. 2013;153:1239–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Cham CM, Gajewski TF. Glucose availability regulates IFN-gamma production and p70S6 kinase activation in CD8+ effector T cells. J Immunol. 2005;174:4670–7.

    Article  CAS  PubMed  Google Scholar 

  90. Brand A, et al. LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 2016;24:657–71.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Qiu J, et al. Acetate promotes T cell effector function during glucose restriction. Cell Rep. 2019;27:2063-2074.e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Wang R, et al. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity. 2011;35:871–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Carr EL, et al. Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation. J Immunol. 2010;185:1037–44.

    Article  CAS  PubMed  Google Scholar 

  95. Johnson MO, et al. Distinct regulation of Th17 and Th1 cell differentiation by glutaminase-dependent metabolism. Cell. 2018;175:1780-1795.e19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Geiger R, et al. L-arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell. 2016;167:829-842.e13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Liu Y, et al. IL-2 regulates tumor-reactive CD8(+) T cell exhaustion by activating the aryl hydrocarbon receptor. Nat Immunol. 2021;22:358–69.

    Article  CAS  PubMed  Google Scholar 

  98. Bian Y, et al. Cancer SLC43A2 alters T cell methionine metabolism and histone methylation. Nature. 2020;585:277–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Farber S, Diamond LK. Temporary remissions in acute leukemia in children produced by folic acid antagonist, 4-aminopteroyl-glutamic acid. N Engl J Med. 1948;238:787–93.

    Article  CAS  PubMed  Google Scholar 

  100. Locasale JW. Serine, glycine and one-carbon units: cancer metabolism in full circle. Nat Rev Cancer. 2013;13:572–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. St Paul M, Ohashi PS. The roles of CD8(+) T cell subsets in antitumor immunity. Trends Cell Biol. 2020;30:695–704.

    Article  CAS  PubMed  Google Scholar 

  102. Shankaran V, et al. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature. 2001;410:1107–11.

    Article  CAS  PubMed  Google Scholar 

  103. Battegay M, et al. Enhanced establishment of a virus carrier state in adult CD4+ T-cell-deficient mice. J Virol. 1994;68:4700–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Janssen EM, et al. CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes. Nature. 2003;421:852–6.

    Article  CAS  PubMed  Google Scholar 

  105. Zander R, et al. 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;51:1028-1042.e4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Cui C, et al. Neoantigen-driven B cell and CD4 T follicular helper cell collaboration promotes anti-tumor CD8 T cell responses. Cell. 2021;184:6101-6118.e13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Ahrends T, et al. CD4+ T cell help confers a cytotoxic T cell effector program including coinhibitory receptor downregulation and increased tissue invasiveness. Immunity. 2017;47:848–861

    Article  CAS  PubMed  Google Scholar 

  108. Wang J-CE, Livingstone AM. Cutting edge: CD4+ T cell help can be essential for primary CD8+ T cell responses in vivo. J Immunol. 2003;171:6339–43.

    Article  CAS  PubMed  Google Scholar 

  109. Provine NM, et al. immediate dysfunction of vaccine-elicited CD8+ T cells primed in the absence of CD4+ T cells. J Immunol. 2016;197:1809–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Peperzak V, Veraar EA, Keller AM, Xiao Y, Borst J. The Pim kinase pathway contributes to survival signaling in primed CD8+ T cells upon CD27 costimulation. J Immunol. 2010;185:6670–8.

    Article  CAS  PubMed  Google Scholar 

  111. van Gisbergen KP, et al. The costimulatory molecule CD27 maintains clonally diverse CD8(+) T cell responses of low antigen affinity to protect against viral variants. Immunity. 2011;35:97–108.

    Article  PubMed  Google Scholar 

  112. Ahrends T, et al. CD4(+) T cell help confers a cytotoxic T cell effector program including coinhibitory receptor downregulation and increased tissue invasiveness. Immunity. 2017;47:848-861.e5.

    Article  CAS  PubMed  Google Scholar 

  113. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–52.

    Article  CAS  PubMed  Google Scholar 

  114. Marciscano AE, Anandasabapathy N. The role of dendritic cells in cancer and anti-tumor immunity. Semin Immunol. 2021;52: 101481.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Ruffell B, et al. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell. 2014;26:623–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Mashayekhi M, et al. CD8alpha(+) dendritic cells are the critical source of interleukin-12 that controls acute infection by Toxoplasma gondii tachyzoites. Immunity. 2011;35:249–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Martinez-Lopez M, Iborra S, Conde-Garrosa R, Sancho D. Batf3-dependent CD103+ dendritic cells are major producers of IL-12 that drive local Th1 immunity against Leishmania major infection in mice. Eur J Immunol. 2015;45:119–29.

    Article  CAS  PubMed  Google Scholar 

  118. Iborra S, et al. Optimal generation of tissue-resident but not circulating memory T cells during viral infection requires crosspriming by DNGR-1(+) dendritic cells. Immunity. 2016;45:847–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Angelini G, et al. Antigen-presenting dendritic cells provide the reducing extracellular microenvironment required for T lymphocyte activation. Proc Natl Acad Sci USA. 2002;99:1491–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Schluns KS, Williams K, Ma A, Zheng XX, Lefrançois L. Cutting edge: requirement for IL-15 in the generation of primary and memory antigen-specific CD8 T cells. J Immunol. 2002;168:4827–31.

    Article  CAS  PubMed  Google Scholar 

  121. Hildner K, et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science. 2008;322:1097–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Broz ML, 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  CAS  PubMed  PubMed Central  Google Scholar 

  123. Salmon H, et al. Expansion and activation of CD103(+) dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity. 2016;44:924–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Roberts EW, 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  CAS  PubMed  PubMed Central  Google Scholar 

  125. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature. 2015;523:231–5.

    Article  CAS  PubMed  Google Scholar 

  126. Webb JR, et al. Profound elevation of CD8+ T cells expressing the intraepithelial lymphocyte marker CD103 (alphaE/beta7 Integrin) in high-grade serous ovarian cancer. Gynecol Oncol. 2010;118:228–36.

    Article  CAS  PubMed  Google Scholar 

  127. 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  CAS  PubMed  Google Scholar 

  128. Webb JR, Milne K, Nelson BH. PD-1 and CD103 are widely coexpressed on prognostically favorable intraepithelial CD8 T cells in human ovarian cancer. Cancer Immunol Res. 2015;3:926–35.

    Article  CAS  PubMed  Google Scholar 

  129. de Mingo Pulido Á, et al. TIM-3 regulates CD103+ dendritic cell function and response to chemotherapy in breast cancer. Cancer Cell. 2018;33:60-74.e6.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Ganesan AP, 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  CAS  PubMed  PubMed Central  Google Scholar 

  131. Varol C, Mildner A, Jung S. Macrophages: development and tissue specialization. Annu Rev Immunol. 2015;33:643–75.

    Article  CAS  PubMed  Google Scholar 

  132. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Bruns H, et al. Vitamin D-dependent induction of cathelicidin in human macrophages results in cytotoxicity against high-grade B cell lymphoma. Sci Transl Med. 2015;7:282ra47.

    Article  PubMed  Google Scholar 

  134. Ngambenjawong C, Gustafson HH, Pun SH. Progress in tumor-associated macrophage (TAM)-targeted therapeutics. Adv Drug Deliv Rev. 2017;114:206–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. DeNardo DG, Ruffell B. Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol. 2019;19:369–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Yin M, et al. Tumor-associated macrophages drive spheroid formation during early transcoelomic metastasis of ovarian cancer. J Clin Invest. 2016;126:4157–73.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Liu C, et al. Treg cells promote the SREBP1-dependent metabolic fitness of tumor-promoting macrophages via repression of CD8(+) T Cell-derived interferon-gamma. Immunity. 2019;51:381-397.e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Boissonnas A, et al. CD8+ tumor-infiltrating T cells are trapped in the tumor-dendritic cell network. Neoplasia. 2013;15:85–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Peranzoni E, et al. Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti-PD-1 treatment. Proc Natl Acad Sci U S A. 2018;115:E4041–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Zhang W, et al. Crosstalk between IL-15Ralpha(+) tumor-associated macrophages and breast cancer cells reduces CD8(+) T cell recruitment. Cancer Commun (Lond). 2022;42:536–57.

    Article  PubMed  Google Scholar 

  141. Petty AJ, et al. Hedgehog signaling promotes tumor-associated macrophage polarization to suppress intratumoral CD8+ T cell recruitment. J Clin Invest. 2019;129:5151–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Nalio Ramos R, et al. Tissue-resident FOLR2(+) macrophages associate with CD8(+) T cell infiltration in human breast cancer. Cell. 2022;185:1189-1207.e25.

    Article  CAS  PubMed  Google Scholar 

  143. Lambrechts D, et al. Phenotype molding of stromal cells in the lung tumor microenvironment. Nat Med. 2018;24:1277–89.

    Article  CAS  PubMed  Google Scholar 

  144. de Visser KE, Joyce JA. The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell. 2023;41:374–403.

    Article  PubMed  Google Scholar 

  145. Gao Q, et al. The artificial intelligence and machine learning in lung cancer immunotherapy. J Hematol Oncol. 2023;16:55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Gurbatri CR, Arpaia N, Danino T. Engineering bacteria as interactive cancer therapies. Science. 2022;378:858–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Saglam-Metiner P, Gulce-Iz S, Biray-Avci C. Bioengineering-inspired three-dimensional culture systems: organoids to create tumor microenvironment. Gene. 2019;686:203–12.

    Article  CAS  PubMed  Google Scholar 

  148. Yuki K, Cheng N, Nakano M, Kuo CJ. Organoid models of tumor immunology. Trends Immunol. 2020;41:652–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Pritykin Y, et al. A unified atlas of CD8 T cell dysfunctional states in cancer and infection. Mol Cell. 2021;81:2477-2493.e10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Zheng C, et al. Landscape of Infiltrating T Cells in Liver Cancer Revealed by Single-Cell Sequencing. Cell. 2017;169:1342-1356.e16.

    Article  CAS  PubMed  Google Scholar 

  151. Abbas HA, et al. Single cell T cell landscape and T cell receptor repertoire profiling of AML in context of PD-1 blockade therapy. Nat Commun. 2021;12:6071.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Sun C, Mezzadra R, Schumacher TN. Regulation and Function of the PD-L1 Checkpoint. Immunity. 2018;48:434–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Kantoff PW, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–22.

    Article  CAS  PubMed  Google Scholar 

  154. Ribas A, et al. Oncolytic virotherapy promotes intratumoral t cell infiltration and improves anti-PD-1 immunotherapy. Cell. 2017;170:1109–1119.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Hegde PS, Wallin JJ, Mancao C. Predictive markers of anti-VEGF and emerging role of angiogenesis inhibitors as immunotherapeutics. Semin Cancer Biol. 2018;52:117–24.

    Article  CAS  PubMed  Google Scholar 

  156. Glimcher LH, Townsend MJ, Sullivan BM, Lord GM. Recent developments in the transcriptional regulation of cytolytic effector cells. Nat Rev Immunol. 2004;4:900–11.

    Article  CAS  PubMed  Google Scholar 

  157. Taniuchi I, et al. Differential requirements for Runx proteins in CD4 repression and epigenetic silencing during T lymphocyte development. Cell. 2002;111:621–33.

    Article  CAS  PubMed  Google Scholar 

  158. Fan Y, et al. Epithelial SOX9 drives progression and metastases of gastric adenocarcinoma by promoting immunosuppressive tumour microenvironment. Gut. 2023;72:624–37.

    Article  CAS  PubMed  Google Scholar 

  159. Agnellini P, et al. Impaired NFAT nuclear translocation results in split exhaustion of virus-specific CD8+ T cell functions during chronic viral infection. Proc Natl Acad Sci U S A. 2007;104:4565–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Pipkin ME, et al. Interleukin-2 and inflammation induce distinct transcriptional programs that promote the differentiation of effector cytolytic T cells. Immunity. 2010;32:79–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Bianchi T, Gasser S, Trumpp A, MacDonald HR. c-Myc acts downstream of IL-15 in the regulation of memory CD8 T-cell homeostasis. Blood. 2006;107:3992–9.

    Article  CAS  PubMed  Google Scholar 

  162. Intlekofer AM, et al. Requirement for T-bet in the aberrant differentiation of unhelped memory CD8+ T cells. J Exp Med. 2007;204:2015–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Kallies A, et al. Transcriptional repressor Blimp-1 is essential for T cell homeostasis and self-tolerance. Nat Immunol. 2006;7:466–74.

    Article  CAS  PubMed  Google Scholar 

  164. Ichii H, et al. Role for Bcl-6 in the generation and maintenance of memory CD8+ T cells. Nat Immunol. 2002;3:558–63.

    Article  CAS  PubMed  Google Scholar 

  165. Liou HC, et al. c-Rel is crucial for lymphocyte proliferation but dispensable for T cell effector function. Int Immunol. 1999;11:361–71.

    Article  CAS  PubMed  Google Scholar 

  166. Palaga T, Miele L, Golde TE, Osborne BA. TCR-mediated Notch signaling regulates proliferation and IFN-gamma production in peripheral T cells. J Immunol. 2003;171:3019–24.

    Article  CAS  PubMed  Google Scholar 

  167. Cho OH, et al. Notch regulates cytolytic effector function in CD8+ T cells. J Immunol. 2009;182:3380–9.

    Article  CAS  PubMed  Google Scholar 

  168. Carter LL, Murphy KM. Lineage-specific requirement for signal transducer and activator of transcription (Stat)4 in interferon gamma production from CD4(+) versus CD8(+) T cells. J Exp Med. 1999;189:1355–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Meraz MA, et al. Targeted disruption of the Stat1 gene in mice reveals unexpected physiologic specificity in the JAK-STAT signaling pathway. Cell. 1996;84:431–42.

    Article  CAS  PubMed  Google Scholar 

  170. Aguilar EJ, et al. Outcomes to first-line pembrolizumab in patients with non-small-cell lung cancer and very high PD-L1 expression. Ann Oncol. 2019;30:1653–9.

    Article  CAS  PubMed  Google Scholar 

  171. Balar AV, et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol. 2017;18:1483–92.

    Article  CAS  PubMed  Google Scholar 

  172. Shitara K, et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet (London, England). 2018;392:123–33.

    Article  CAS  PubMed  Google Scholar 

  173. Mo D-C, Luo P-H, Huang S-X, Wang H-L, Huang J-F. Safety and efficacy of pembrolizumab plus lenvatinib versus pembrolizumab and lenvatinib monotherapies in cancers: a systematic review. Int Immunopharmacol. 2021;91: 107281.

    Article  CAS  PubMed  Google Scholar 

  174. Ready NE, et al. Nivolumab monotherapy and nivolumab plus ipilimumab in recurrent small cell lung cancer: results from the checkmate 032 randomized cohort. J Thorac Oncol. 2020;15:426–35.

    Article  CAS  PubMed  Google Scholar 

  175. Paik J. Nivolumab plus relatlimab: first approval. Drugs. 2022;82:925–31.

    Article  CAS  PubMed  Google Scholar 

  176. Luo H, et al. Effect of camrelizumab vs placebo added to chemotherapy on survival and progression-free survival in patients with advanced or metastatic esophageal squamous cell carcinoma: the ESCORT-1st randomized clinical trial. JAMA. 2021;326:916–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Ren S, et al. Camrelizumab plus carboplatin and paclitaxel as first-line treatment for advanced squamous NSCLC (CameL-Sq): a phase 3 trial. J Thorac Oncol. 2022;17:544–57.

    Article  CAS  PubMed  Google Scholar 

  178. Yang Y, et al. Camrelizumab versus placebo in combination with gemcitabine and cisplatin as first-line treatment for recurrent or metastatic nasopharyngeal carcinoma (CAPTAIN-1st): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 2021;22:1162–74.

    Article  CAS  PubMed  Google Scholar 

  179. Wang J, et al. Tislelizumab plus chemotherapy vs chemotherapy alone as first-line treatment for advanced squamous non-small-cell lung cancer: a phase 3 randomized clinical trial. JAMA Oncol. 2021;7:709–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Lu S, et al. Tislelizumab plus chemotherapy as first-line treatment for locally advanced or metastatic nonsquamous NSCLC (RATIONALE 304): a randomized phase 3 trial. J Thorac Oncol. 2021;16:1512–22.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  182. Mansfield AS, et al. Safety and patient-reported outcomes of atezolizumab, carboplatin, and etoposide in extensive-stage small-cell lung cancer (IMpower133): a randomized phase I/III trial. Ann Oncol. 2020;31:310–7.

    Article  CAS  PubMed  Google Scholar 

  183. West H, et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019;20:924–37.

    Article  CAS  PubMed  Google Scholar 

  184. Paz-Ares L, et al. Durvalumab plus platinum-etoposide versus platinum-etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): a randomised, controlled, open-label, phase 3 trial. Lancet. 2019;394:1929–39.

    Article  CAS  PubMed  Google Scholar 

  185. Altorki NK, et al. Neoadjuvant durvalumab with or without stereotactic body radiotherapy in patients with early-stage non-small-cell lung cancer: a single-centre, randomised phase 2 trial. Lancet Oncol. 2021;22:824–35.

    Article  CAS  PubMed  Google Scholar 

  186. Goldman JW, et al. Durvalumab, with or without tremelimumab, plus platinum-etoposide versus platinum-etoposide alone in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): updated results from a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2021;22:51–65.

    Article  CAS  PubMed  Google Scholar 

  187. Powles T, et al. Durvalumab alone and durvalumab plus tremelimumab versus chemotherapy in previously untreated patients with unresectable, locally advanced or metastatic urothelial carcinoma (DANUBE): a randomised, open-label, multicentre, phase 3 trial. Lancet Oncol. 2020;21:1574–88.

    Article  CAS  PubMed  Google Scholar 

  188. Keskin DB, et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature. 2019;565:234–9.

    Article  CAS  PubMed  Google Scholar 

  189. Yang G, et al. Functional status and spatial architecture of tumor-infiltrating CD8+ T cells are associated with lymph node metastases in non-small cell lung cancer. J Transl Med. 2023;21:320.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Wang P, et al. CD103(+)CD8(+) T lymphocytes in non-small cell lung cancer are phenotypically and functionally primed to respond to PD-1 blockade. Cell Immunol. 2018;325:48–55.

    Article  CAS  PubMed  Google Scholar 

  191. Maniar R, et al. Self-renewing CD8+ T-cell abundance in blood associates with response to immunotherapy. Cancer Immunol Res. 2023;11:164–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Liu Y, et al. Combined application of bevacizumab and PD-1 blockade displays durable treatment effects by increasing the infiltration and cytotoxic function of CD8(+) T cells in lung cancer. Immunotherapy. 2022;14:695–708.

    Article  CAS  PubMed  Google Scholar 

  193. Qu QX, et al. 4–1BB agonism combined with PD-L1 blockade increases the number of tissue-resident CD8+ T cells and facilitates tumor abrogation. Front Immunol. 2020;11:577.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Palermo B, et al. CD28/PD1 co-expression: dual impact on CD8(+) T cells in peripheral blood and tumor tissue, and its significance in NSCLC patients’ survival and ICB response. J Exp Clin Cancer Res. 2023;42:287.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Fehlings M, et al. Late-differentiated effector neoantigen-specific CD8+ T cells are enriched in peripheral blood of non-small cell lung carcinoma patients responding to atezolizumab treatment. J Immunother Cancer. 2019;7:249.

    Article  PubMed  PubMed Central  Google Scholar 

  196. Liu Z, et al. Progenitor-like exhausted SPRY1(+)CD8(+) T cells potentiate responsiveness to neoadjuvant PD-1 blockade in esophageal squamous cell carcinoma. Cancer Cell. 2023;41:1852–1870.

    Article  CAS  PubMed  Google Scholar 

  197. Komdeur FL, et al. CD103+ tumor-infiltrating lymphocytes are tumor-reactive intraepithelial CD8+ T cells associated with prognostic benefit and therapy response in cervical cancer. Oncoimmunology. 2017;6: e1338230.

    Article  PubMed  PubMed Central  Google Scholar 

  198. Edwards J, 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  CAS  PubMed  Google Scholar 

  199. Wanhainen KM, et al. P2RX7 enhances tumor control by CD8+ T cells in adoptive cell therapy. Cancer Immunol Res. 2022;10:871–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Savas P, 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  CAS  PubMed  Google Scholar 

  201. Pinjusic K, et al. Activin-A impairs CD8 T cell-mediated immunity and immune checkpoint therapy response in melanoma. J Immunother Cancer. 2022;10.

  202. Zou Q, et al. DNA methylation-based signature of CD8+ tumor-infiltrating lymphocytes enables evaluation of immune response and prognosis in colorectal cancer. J Immunother Cancer. 2021;9.

  203. Liu Z, et al. Intratumoral TIGIT(+) CD8(+) T-cell infiltration determines poor prognosis and immune evasion in patients with muscle-invasive bladder cancer. J Immunother Cancer. 2020;8.

  204. Huang F, et al. δ-Catenin peptide vaccines repress hepatocellular carcinoma growth via CD8(+) T cell activation. Oncoimmunology. 2018;7: e1450713.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable

Funding

This study was supported by the Beijing Xisike Clinical Oncology Research Foundation (China) (Grant No. Y-HR2020MS-0156 to T.M.).

Author information

Authors and Affiliations

Authors

Contributions

Concept and design: TM and JHW. Data analysis and interpretation: JHW. Manuscript writing: all authors. Final approval of manuscript: all authors.

Corresponding authors

Correspondence to Jinghui Wang or Teng Ma.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no conflicts of interest.

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 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, J., Lu, Z., Zhao, H. et al. The expanding Pandora’s toolbox of CD8+T cell: from transcriptional control to metabolic firing. J Transl Med 21, 905 (2023). https://doi.org/10.1186/s12967-023-04775-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-023-04775-3

Keywords