Abstract
Cancer immunotherapies continue to face numerous obstacles in the successful treatment of solid malignancies. While immunotherapy has emerged as an extremely effective treatment option for hematologic malignancies, it is largely ineffective against solid tumors due in part to metabolic challenges present in the tumor microenvironment (TME). Tumor-infiltrating CD8+ T cells face fierce competition with cancer cells for limited nutrients. The strong metabolic suppression in the TME often leads to impaired T-cell recruitment to the tumor site and hyporesponsive effector functions via T-cell exhaustion. Growing evidence suggests that mitochondria play a key role in CD8+ T-cell activation, migration, effector functions, and persistence in tumors. Therefore, targeting the mitochondrial metabolism of adoptively transferred T cells has the potential to greatly improve the effectiveness of cancer immunotherapies in treating solid malignancies.
Keywords: Metabolism, CD8 Positive T lymphocytes, Chimeric antigen receptor therapy, Mitochondria
INTRODUCTION
Despite continued advancements in adoptive T-cell therapies, success in treating cancer patients with solid malignancies remains limited (1). The immunosuppressive tumor microenvironment (TME) of solid tumors presents many obstacles that inhibit the effector functions of CD8+ T cells (2). A number of these challenges can be attributed to the high metabolic flux of cancer cells (3). The augmented proliferation rate of cancer cells requires elevated metabolism to produce enough energy and metabolic intermediates (4). Therefore, nutrients in the TME that are available to CD8+ T cells are often scarce and metabolic waste products accumulate, thus inhibiting the activity of adoptively transferred T cells (5,6).
One possible way to overcome the metabolic challenges of the TME is to selectively boost the metabolism of only CD8+ T cells. Specifically, mitochondrial metabolism has been shown by a number of groups to be essential for various T-cell effector functions (7,8,9,10,11). In this Review, we summarize the metabolic dynamics and plasticity of CD8+ T cells, describe the main types of adoptive T-cell therapies, and discuss the metabolic challenges faced by adoptively transferred T cells. Finally, we justify why targeting the mitochondrial metabolism of CD8+ T cells is a probable approach to improve the outcomes of adoptive T-cell therapies.
METABOLIC DYNAMICS OF CD8+ T CELLS
Immune cells utilize numerous forms of metabolism to obtain sufficient levels of energy and metabolites from available nutrients to support their survival, differentiation, proliferation, and other important cellular functions (12,13). Upon infection or chronic inflammation, such as in cancer, immune cells undergo dramatic metabolic reprogramming to respond optimally and execute their respective effector functions (14).
CD8+ T cells can use various forms of metabolism, including glycolysis, the tricarboxylic acid (TCA) cycle, oxidative phosphorylation (OxPhos), glutaminolysis, and fatty acid oxidation (FAO) (Fig. 1) (15). CD8+ T cells at each differentiation state have unique metabolic profiles to produce energy and metabolic precursors that are important for specific T-cell functions (12). In addition, activated CD8+ T cells are metabolically flexible; therefore, they can adjust their reliance on different metabolic pathways depending on current metabolic needs, as well as what nutrients are available (12,16). Metabolic reprogramming in CD8+ T cells is essential for the acquisition of effector functions in activated T cells and for the development of memory T cells (16).
Figure 1. CD8+ T cell metabolism. Metabolic pathways used by CD8+ T cells include glycolysis, OxPhos, glutaminolysis, and FAO. Glycolysis occurs in the cytoplasm and produces 2 molecules of ATP, 2 NADH, and 2 pyruvate for every molecule of glucose. Pyruvate can be transported into mitochondria and converted into acetyl-CoA to be used in the TCA cycle to make electron carriers NADH and FADH2. When activated T cells use aerobic glycolysis, pyruvate is converted to lactate and released by the cell, even when oxygen is present, like the Warburg effect in cancer cells. OxPhos requires the electron carriers NADH and FADH2, with oxygen as the final electron acceptor to produce up to 36 molecules of ATP. When glucose is limited, activated T cells can also use glutaminolysis to support the TCA cycle and OxPhos. Naïve and memory CD8+ T cells utilize β-oxidation of fatty acids to sustain the TCA cycle. I–IV indicate complexes of the electron transport chain. Created with BioRender.com.
CoQ, coenzyme Q; α-KG, alpha-ketoglutarate; Cyt c, cytochrome c; OAA, oxaloacetate; SucCoA, succinyl-coenzyme A; Glucose-6P, glucose 6-phosphate; 3PG, 3-phosphoglycerate; PEP, phosphoenolpyruvate; GLUT, glucose transporter.
Naïve CD8+ T cells
Naïve CD8+ T cells are relatively quiescent and therefore do not have a high demand for energy as they remain in a steady state until they recognize their target antigen (12). Catabolic metabolism utilized by naïve T cells primarily involves the complete oxidation of glucose-derived pyruvate by OxPhos to produce ATP. Given that naïve CD8+ T cells are not actively proliferating, they have low levels of nutrient uptake and relatively low rates of glycolysis.
For a cell to completely oxidize glucose-derived pyruvate and produce more ATP than that produced from glycolysis alone, OxPhos needs to occur in the mitochondria. OxPhos requires the electron carriers NADH and FADH2, which are produced during either the TCA cycle or glycolysis and donate their electrons to complex I or II of the electron transport chain (ETC) in the inner mitochondrial membrane (Fig. 1). While electrons travel through complexes of the ETC, protons are simultaneously pumped from the mitochondrial matrix to the intermembrane space. The proton gradient that is formed across the inner mitochondrial membrane contributes to the mitochondrial membrane potential (Dym), which is then used by complex V, also known as ATP synthase of the ETC to phosphorylate ADP to ATP. The production of ATP in mitochondria through OxPhos requires oxygen as the final electron acceptor and can produce up to 36 molecules of ATP for each glucose molecule. In addition to making ATP, OxPhos produces ROS, water, and the oxidized electron carriers NAD+ and FAD (17). OxPhos is an efficient way for a cell to produce ATP by completely oxidizing glucose; however, it takes more time than glycolysis alone and requires oxygen. The low demand for energy in naïve CD8+ T cells makes OxPhos a suitable form of metabolism.
Effector CD8+ T cells
During T cell activation, metabolic reprogramming dependent on CD28 co-stimulation occurs to support anabolic cell growth in proliferating effector CD8+ T cells (18). Glycolysis and glucose consumption increase immensely to allow for the rapid production of ATP, metabolic intermediates, and effector cytokines (19). Similarly to the Warburg effect in cancer cells, the glycolysis end-product in effector CD8+ T cells is lactate, even in the presence of oxygen (12,20). Glycolysis yields a net of 2 ATP, 2 pyruvate, and 2 NADH molecules for each molecule of glucose (Fig. 1). Additionally, metabolic intermediates that can be utilized for fatty acid, nucleotide and amino acid biosynthesis to support cell growth and proliferation are formed at various steps in glycolysis (12). The pentose phosphate pathway, which uses the first intermediate of glycolysis, is important for nucleotide production in proliferating cells. When effector CD8+ T cells have elevated levels of glycolysis even when oxygen is present, pyruvate is converted into lactate and then is released by the cell. However, if pyruvate is to be further oxidized, it enters mitochondria and is made into acetyl-coenzyme A (CoA), a substrate of the TCA cycle, which can also produce metabolic intermediates for protein and fatty acid biosynthesis (12) (Fig. 1).
In addition to glucose, activated CD8+ T cells consume high levels of glutamine, which can be processed in the TCA cycle and is important for T cell proliferation and cytokine production (21). When glucose is in short supply and therefore glycolytic-derived pyruvate is minimal, glutamine can enter the TCA cycle after being converted into a-ketoglutarate by glutaminolysis (Fig. 1). Similar to glycolysis, glutaminolysis is also important for producing metabolites that are necessary for biosynthesis (22). In addition to entering the TCA cycle, glutamine is also a precursor for amino acid and nucleotide biosynthesis. Furthermore, a-ketoglutarate in the TCA cycle that is derived from glutamine, or pyruvate from glycolysis, can be made into citrate to be further used in lipid synthesis.
Interestingly, mitochondria in effector CD8+ T cells become highly fragmented and contain loose cristae, which can lead to inefficient OxPhos (23). Consequently, activated CD8+ T cells rely more on glycolysis and glutaminolysis to support effector functions. However, mitochondrial activity is still important for CD8+ T-cell activation and effector function (7,24,25,26). For example, mitochondrial ROS is required for T-cell activation to induce signaling pathways that are important for T-cell proliferation and cytokine production (11). Additionally, mitochondria activity has been shown to be crucial for effector CD8+ T cell migration (9,26).
Memory CD8+ T cells
Upon resolution of an immune response, any surviving memory CD8+ T cells return to a metabolically quiescent state. Memory CD8+ T cells, similar to naïve CD8+ T cells, utilize catabolic metabolism with a strong reliance on OxPhos and FAO (16,27). When glycolysis produces pyruvate in memory CD8+ T cells, pyruvate is oxidized into acetyl-CoA in mitochondria and can then enter the TCA cycle to contribute to OxPhos or produce citrate for fatty acid synthesis. Free fatty acids can undergo β-oxidation to be converted into acetyl-CoA, enter the TCA cycle, and produce ATP through the ETC, which is known as FAO. Additionally, instead of storing long-chain fatty acids like effector CD8+ T cells, memory CD8+ T cells utilize extracellular glucose to eventually produce fatty acids for FAO (28).
To maximize the efficiency of OxPhos, memory CD8+ T cells have an abundance of elongated mitochondria with tight cristae to bring complexes of the ETC in close proximity to each other (23). Another unique characteristic of memory CD8+ T cells is that their mitochondrial spare respiratory capacity (SRC) is greater than that of both naïve and effector CD8+ T cells (29). SRC is the ability of mitochondria to increase their oxygen consumption rate above the basal oxygen consumption rate in times of increased stress (30). When a memory CD8+ T-cell becomes activated, metabolic reprogramming occurs once again, like when naïve CD8+ T cells are activated. The increased mitochondrial mass, SRC, and FAO of memory CD8+ T cells provides them the ability to rapidly respond upon TCR activation (31).
CD4+ vs. CD8+ T cell metabolism
The metabolic pathways employed at each differentiation state are largely overlapped in CD4+ and CD8+ T cells (32). However, CD4+ Tregs mainly rely on oxidative metabolism like naïve T cells, whereas both effector CD4+ and CD8+ T cells require elevated levels of glycolysis (33). In addition, polyamine metabolism has been shown to be essential in the polarization of CD4+ helper T cells (34). Furthermore, one unique characteristic of memory CD4+ T cells is that they have an increased spare glycolytic capacity in addition to an elevated mitochondrial SRC (35).
CD8+ T-cell exhaustion
The immunosuppressive TME comprises many elements that drive T-cell exhaustion (5,8,36). Exhaustion of CD8+ T cells severely impedes overall effector functions by suppressing cell proliferation and cytokine production while overexpressing inhibitory receptors (37). Chronic antigen stimulation, as seen in cancer and chronic infections, is one of the main contributors to T-cell exhaustion (38). Importantly, examination of the gene expression profiles of effector, memory, and exhausted CD8+ T cells revealed transcriptional downregulation of many genes involved in metabolism in exhausted CD8+ T cells. These include genes involved in the TCA cycle and FAO, indicating that exhausted CD8+ T cells have a metabolic defect (38). Furthermore, PD-1, a T-cell exhaustion marker, has been shown to suppress both glycolysis and mitochondrial respiration, in addition to mitochondrial biogenesis (39,40). Therefore, many current strategies to improve effector T-cell functions at the tumor site include reagents that boost T-cell metabolism (Table 1) (41).
Table 1. Current strategies to improve CD8+ T cell effector function in the TME.
| Treatment | Mechanism | Outcome |
|---|---|---|
| Deletion or inhibition of prolyl-hydroxylase | Prevents degradation of HIF & increases glycolysis | Improved tumor clearance (42) |
| Constitutively active Akt/mTORC1 pathway | Increased glucose uptake and glycolysis | Improved T cell effector functions and decreased immune checkpoint expression (43) |
| FCCP in combination with PD-1 blockade | Increased ROS production leads to activation of mTOR and increases PGC1a expression | Increased T cell accumulation at tumor site and improved anti-tumor activity (44) |
| Enforced PGC1a expression | Increased mitochondrial function | Improved anti-tumor activity (5) |
HIF, hypoxia inducible factor; FCCP, carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone.
CHALLENGES OF T-CELL IMMUNOTHERAPIES
Defining adoptive T-cell immunotherapies
Over the past decade, there has been tremendous progress in the advancement of adoptive T-cell therapies for the treatment of cancer, with higher success rates in hematological malignancies than solid malignancies. The promising outcomes of T-cell immunotherapy for cancer patients with late-stage or refractory disease have led to a rapid increase in immunotherapy research (45). There are 3 main types of adoptive T-cell immunotherapy (46). The first type of adoptive T-cell therapy requires the isolation of tumor-infiltrated lymphocytes (TILs) from a patient’s tumor. TILs that display antitumor activity are selected for and expanded with cytokine-supplemented medium before being infused back into the patient (47). Utilizing tumor-resident TILs for adoptive T-cell therapy showed initial success in the treatment of metastatic melanoma, with an even more pronounced effect in patients who also underwent lymphodepletion and received high doses of IL-2 (48). The other 2 forms of adoptive T-cell immunotherapy require the genetic modification of a cancer patient’s peripheral blood T cells to express tumor-reactive receptors (46). Circulating T cells are isolated by leukapheresis, activated with anti-CD3 and anti-CD28 antibodies, and then transduced with a viral vector to either express a tumor-specific TCR or chimeric antigen receptor (CAR). The genetically modified T cells are further expanded and cryopreserved to be administered at a later date (49). TCR-transduced T cells express the TCR a and b chains, which can recognize tumor-associated antigens in a MHC-dependent manner (50). Unlike TCR T cells, CAR-T cells recognize antigens independent of MHC. The CAR is composed of an extracellular single-chain Fv domain of a monoclonal antibody that can recognize the target antigen and an intracellular CD3z chain that acts as the TCR signaling domain. The CAR also contains a costimulatory domain, either CD28 or 4-1BB, that leads to downstream signaling cascades, stimulating the T-cell to kill the target cell (51,52). Since the first generation of CARs, there have been many improvements to the composition of the CAR to elicit stronger T-cell cytotoxicity, as well as better T-cell survival and proliferation (53).
While CAR-T-cell therapies have showed tremendous success in the treatment of hematological malignancies, there are many challenges that need to be overcome to improve the success of adoptive T-cell therapies for the treatment of solid tumors (54,55). These include improved T-cell trafficking to the tumor, persistent or increased memory T-cell formation, decreased T-cell exhaustion, and surpassed inhibitory signals in the immunosuppressive TME (Fig. 2) (1). The TME of solid cancers contains many factors that inhibit the function of TILs, such as immune checkpoint proteins, immunosuppressive cell types, accumulated immunosuppressive metabolites, and low levels of essential nutrients (6,56,57,58).
Figure 2. Challenges of adoptive T cell therapies. There are numerous obstacles to overcome to improve the efficacy of adoptive T cell therapies, some of which are depicted here. Created with BioRender.com.
T-cell trafficking
The low success rates of adoptive T-cell immunotherapies for the treatment of solid tumors can be in part attributed to impaired T-cell trafficking to the tumor (59). Patients with a higher number of TILs have a much better prognosis than patients with few infiltrated T cells (60,61). After infusion of TCR-modified or CAR-T cells, a large percentage of the transferred cells home to other nontumor-bearing organs, which may result in severe side effects through interactions of the TCR or CAR with cognate antigen expressed on healthy tissue (62).
The TME of solid tumors can prevent the infiltration of T cells into the tumor bed in numerous ways. Many tumor and stromal cells release cytokines that recruit immunosuppressive cells while also decreasing proinflammatory chemokines and cytokines (63). Tumor cells can also block T-cell extravasation into a tumor by downregulating the expression of intercellular adhesion molecule-1 (ICAM-1) while upregulating Fas ligand and inhibitory receptors on surrounding tumor endothelial cells (64). The interaction between ICAM-1 on the endothelium and LFA-1 on the surface of activated T cells is one of the main mechanisms of T-cell extravasation out of a blood vessel and into a solid tumor or the surrounding inflamed tissue (65). Consequently, when cancer cells decrease ICAM-1 expression on the endothelium and decrease proinflammatory chemokine release, it is difficult for tumor-specific CD8+ T cells to locate and infiltrate a tumor (64).
The physical barrier created by the tumor vasculature, stromal cells, and extracellular matrix also makes it extremely challenging for tumor-reactive CD8+ T cells to enter the tumor bed (66). For example, metastatic breast cancer tumors are highly fibrotic and prevent the infiltration of effector CD8+ T cells (67). Furthermore, any CD8+ T cells that enter the tumor site face even more challenges. For instance, actively migrating T cells rely on ATP and localize mitochondria to uropods to fuel cytoskeletal rearrangements; however, ATP production is significantly suppressed in the nutrient-depleted TME (9,68,69,70). Therefore, any T cells that can traffic to the tumor still have many barriers to overcome and may not be able to produce sufficient ATP to sustain cell migration within the tumor.
T-cell persistence
Patients with long-term persistence of adoptively transferred T cells are more likely to have a better prognosis (71,72). During the expansion of CAR- or TCR-modified T cells, many of these T cells become terminally differentiated effector T cells, which has a negative impact on T-cell persistence after infusion (73). Therefore, the generation of memory-like T cells to improve persistence after adoptive transfer may be very promising for future therapeutic approaches (27,74). Interestingly, activated CD8+ T cells with a low Dym have a memory-like T-cell phenotype, increased antitumor function, and increased in vivo self-renewal (10). Overall, the unique metabolic aspects of memory CD8+ T cells include decreased glycolysis, a low Dym, increased reliance on FAO, and an enhanced mitochondrial SRC (27,31,74). Currently, however, there are no clinically approved techniques to selectively manipulate the metabolism of genetically modified T cells that do not also influence other cell types.
Other potential ways to improve the persistence of adoptively transferred T cells include inhibiting Akt or glycolysis and promoting FAO (27,74,75). Inhibition of Akt was shown to result in an increase in FAO, mitochondrial SRC, and memory-like T cells with enhanced persistence (75). Reducing glycolytic metabolism led to an increase in the formation of memory CD8+ T cells with improved antitumor function (74). Additionally, using 4-1BB as the costimulation signal in CAR-T cells strengthened their preference for mitochondrial metabolism, promoting a memory T-cell phenotype and boosting T-cell persistence (76,77). Further strategies that have been identified to improve T-cell persistence in the setting of an infection or cancer are listed in Table 2 (41). Overall, there are numerous studies that suggest manipulating the metabolism of genetically modified T cells is a promising approach to improve the persistence of adoptively transferred T cells.
Table 2. Current strategies to increase CD8+ T cell persistence.
| Treatment | Mechanism | Outcome |
|---|---|---|
| Rapamycin | mTORC1 inhibitor | Increased T cell persistence and memory T cell development (27,78) |
| Mdivi | Increased mitochondrial fusion in vitro | Increased T cell anti-tumor activity in vivo (23) |
| Akt inhibitors | Decreased glycolysis & increased mitochondrial metabolism | Increased T cell anti-tumor activity in vivo (75) |
| JQ1 | Bromodomain inhibitor | Increased T cell anti-tumor activity and persistence in vivo (79) |
| Metformin | Activates AMPK signaling | Promotes T cell persistence in vivo (27) |
| 2-DG | Inhibits glycolysis | Increased T cell lifespan and memory cell induction (74) |
| L-arginine | Decreases glycolysis and promotes oxidative metabolism | Increased T cell persistence and anti-tumor activity in vivo (80) |
| IL-7/IL-15 | Promotes mitochondrial metabolism and biogenesis | Promotes T cell longevity and memory T cell formation (29,76,81) |
| Deletion of VHL | Stabilizes HIF1a & increases glycolysis | Increased long-lived effector memory CD8+ T cells (82) |
| Triiodothyronine (T3) | Improves dendritic cell mediated T cell activation | Increased number of tumor infiltrating CD8+ T cells and IFNy production (83) |
AMPK, AMP-activated protein kinase; 2-DG, 2-deoxyglucose; VHL, von Hippel-Lindau; HIF1a, hypoxia inducible factor 1a.
TARGETING T-CELL MITOCHONDRIAL METABOLISM TO IMPROVE CANCER IMMUNOTHERAPY
Activated CD8+ T cells and cancer cells utilize overlapping metabolic pathways, including OxPhos, glycolysis and glutaminolysis, which puts T cells and cancer cells in direct competition with each other for limited nutrients (6,84). The battle for glucose in the TME between proliferating TILs and cancer cells makes it difficult for T cells to sustain high levels of glycolysis to support proliferation and biosynthesis (85,86). Additionally, most solid tumors are hypoxic, preventing the use of OxPhos to produce ATP in TILs (36). The harsh, nutrient-depleted conditions of the TME prevent adequate metabolism in TILs, consequently decreasing effector functions and increasing T-cell exhaustion. Furthermore, the metabolic status of immune cells, including CD8+ T cells in the TME are likely to be different between cancer types in separate tissues, and even between patients with the same type of cancer due to diverse temporal and spatial coordination of cancer metabolism and extracellular metabolite levels within the tumor (87,88,89,90). Altogether, there is increasing interest in developing a technique to specifically boost TIL metabolism in the TME without benefitting cancer cells (91). Enhancing mitochondrial mass and overall mitochondrial health, thus improving mitochondrial respiration, has the capacity to overcome many challenges that TILs face, as well as advancing the outcome of adoptive T-cell therapies in solid malignancies (Fig. 3).
Figure 3. The role of mitochondria in CAR-T cell function. (A) CAR-T cells cultured in nutrient rich media, such as during CAR-T cell production, have healthy mitochondria that can produce ATP and metabolic intermediates that are important for T cell proliferation. Additionally, functional mitochondria can support cytokine production and effector functions. (B) CAR-T cells that infiltrate a nutrient deprived TME show decreased mitophagy and mitochondrial biogenesis, resulting in the accumulation of dysfunctional mitochondria. Reduced mitochondrial function contributes to T cell exhaustion, poor effector functions, and decreased persistence. Created with BioRender.com.
Mitochondrial health of TILs
The immunosuppressive TME causes a decrease in overall mitochondrial health in TILs, leading to a reduction in OxPhos and increased T-cell exhaustion (5). Recent studies have shown that the hypoxic TME, in addition to chronic antigen stimulation, inhibits mitochondrial ATP production as well as T-cell proliferation and effector functions, thus reducing therapy efficacy (7,36,92,93). Depolarized mitochondria accumulate in TILs due to decreased mitophagy, further contributing to T-cell exhaustion (8). T-cell exhaustion induced by dysfunctional mitochondria can be reversed by decreasing ROS levels, boosting mitophagy, or enforcing mitochondrial biogenesis in TILs (5,7,8,36). Additionally, there is evidence that chronic lymphocytic leukemia patients who receive CD19 CAR-T cells with increased mitochondrial mass are more likely to have a complete response (94). Therefore, developing a strategy to enhance mitochondrial health in adoptive T-cell therapies has tremendous promise.
PD-1 and dysfunctional T-cell mitochondria
Blocking PD-1 signaling with antibodies has shown success in treating a subset of patients with solid malignancies; however, anti-PD-1 treatment does not work well when T cells are terminally exhausted (95,96). Tumor-reactive TILs that are PD-1 positive are typically exhausted and have low antitumor function (97). Previous studies have found that TILs with high PD-1 expression accumulate dysfunctional mitochondria and have a low Dym (8,40). Many factors in the immunosuppressive TME, including PD-1 signaling, can cause a decrease in TIL metabolism, eliciting mitochondrial dysfunction. Blocking only the PD-1 pathway could partially remove the metabolic restrictions of exhausted T cells in a viral infection model, but this was not the case for intratumoral T cells (5,39). Studies that have combined anti-PD-1 therapy with techniques to increase mitochondrial biogenesis or overall mitochondrial quality have shown a synergistic effect on antitumor activity (44,77).
Mitochondrial biogenesis and mitophagy
Another way to improve the mitochondrial metabolism of adoptively transferred T cells is by boosting mitochondrial biogenesis and mitophagy (5,8,94). Treatment of CD8+ T cells with NAD precursors was shown to improve mitophagy and decrease mitochondrial dysfunction, while in vivo treatment had even more pronounced effects with increased TIL antitumor activity (8,98). Additionally, enforced expression of PPAR-g coactivator 1a (PGC1a), which controls mitochondrial biogenesis, restored mitochondrial function and effector functions in TILs (5,99). Finally, treatment of activated CD8+ T cells with 4-1BB agonist antibodies increased mitochondrial fusion and biogenesis, improving the outcome of adoptive T-cell therapy due to increased PGC1a expression (77).
CONCLUDING REMARKS
Cancer immunotherapies continue to evolve and improve; however, extensive advancements are necessary to improve treatment outcomes for a large majority of cancer patients with solid malignancies. Over the last few years, many studies have suggested that targeting CD8+ T-cell metabolism could be an ideal approach to strengthen immunotherapy efficacy. Increasing evidence has shown the importance of functional mitochondria in various avenues of T-cell activity, including their activation, proliferation, and differentiation into memory T cells. However, it is difficult to specifically target T-cell metabolism without simultaneously benefiting cancer cells or inducing systemic side effects. Current strategies utilize compounds that can affect many cell types or may need additional genetic manipulation to selectively manipulate T-cell metabolism. Therefore, novel techniques need to be further developed and tested in the clinic to control CD8+ T-cell metabolism and to improve the outcomes of adoptive T-cell therapies.
Abbreviations
- CAR
chimeric antigen receptor
- CoA
coenzyme A
- Dym
mitochondrial membrane potential
- ETC
electron transport chain
- FAO
fatty acid oxidation
- ICAM-1
intercellular adhesion molecule-1
- OxPhos
oxidative phosphorylation
- PGC1a
PPAR-g coactivator 1a
- SRC
spare respiratory capacity
- TCA
tricarboxylic acid
- TIL
tumor-infiltrated lymphocyte
- TME
tumor microenvironment
Footnotes
Conflict of Interest: The authors declare no potential conflicts of interest.
- Conceptualization: Kim M, Amitrano AM.
- Supervision: Kim M.
- Writing - original draft: Kim M, Amitrano AM.
- Writing - review & editing: Kim M, Amitrano AM.
References
- 1.Martinez M, Moon EK. CAR T cells for solid tumors: new strategies for finding, infiltrating, and surviving in the tumor microenvironment. Front Immunol. 2019;10:128. doi: 10.3389/fimmu.2019.00128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Liu G, Rui W, Zhao X, Lin X. Enhancing CAR-T cell efficacy in solid tumors by targeting the tumor microenvironment. Cell Mol Immunol. 2021;18:1085–1095. doi: 10.1038/s41423-021-00655-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Martínez-Reyes I, Chandel NS. Cancer metabolism: looking forward. Nat Rev Cancer. 2021;21:669–680. doi: 10.1038/s41568-021-00378-6. [DOI] [PubMed] [Google Scholar]
- 4.Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324:1029–1033. doi: 10.1126/science.1160809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Scharping NE, Menk AV, Moreci RS, Whetstone RD, Dadey RE, Watkins SC, Ferris RL, Delgoffe GM. The tumor microenvironment represses T Cell mitochondrial biogenesis to drive intratumoral T cell metabolic insufficiency and dysfunction. Immunity. 2016;45:374–388. doi: 10.1016/j.immuni.2016.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Chang CH, Qiu J, O’Sullivan D, Buck MD, Noguchi T, Curtis JD, Chen Q, Gindin M, Gubin MM, van der Windt GJ, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015;162:1229–1241. doi: 10.1016/j.cell.2015.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Vardhana SA, Hwee MA, Berisa M, Wells DK, Yost KE, King B, Smith M, Herrera PS, Chang HY, Satpathy AT, et al. Impaired mitochondrial oxidative phosphorylation limits the self-renewal of T cells exposed to persistent antigen. Nat Immunol. 2020;21:1022–1033. doi: 10.1038/s41590-020-0725-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Yu YR, Imrichova H, Wang H, Chao T, Xiao Z, Gao M, Rincon-Restrepo M, Franco F, Genolet R, Cheng WC, et al. Disturbed mitochondrial dynamics in CD8+ TILs reinforce T cell exhaustion. Nat Immunol. 2020;21:1540–1551. doi: 10.1038/s41590-020-0793-3. [DOI] [PubMed] [Google Scholar]
- 9.Ledderose C, Liu K, Kondo Y, Slubowski CJ, Dertnig T, Denicoló S, Arbab M, Hubner J, Konrad K, Fakhari M, et al. Purinergic P2X4 receptors and mitochondrial ATP production regulate T cell migration. J Clin Invest. 2018;128:3583–3594. doi: 10.1172/JCI120972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sukumar M, Liu J, Mehta GU, Patel SJ, Roychoudhuri R, Crompton JG, Klebanoff CA, Ji Y, Li P, Yu Z, et al. Mitochondrial membrane potential identifies cells with enhanced stemness for cellular therapy. Cell Metab. 2016;23:63–76. doi: 10.1016/j.cmet.2015.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Sena LA, Li S, Jairaman A, Prakriya M, Ezponda T, Hildeman DA, Wang CR, Schumacker PT, Licht JD, Perlman H, et al. Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. Immunity. 2013;38:225–236. doi: 10.1016/j.immuni.2012.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Pearce EL, Poffenberger MC, Chang CH, Jones RG. Fueling immunity: insights into metabolism and lymphocyte function. Science. 2013;342:1242454. doi: 10.1126/science.1242454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wang R, Green DR. Metabolic checkpoints in activated T cells. Nat Immunol. 2012;13:907–915. doi: 10.1038/ni.2386. [DOI] [PubMed] [Google Scholar]
- 14.Reina-Campos M, Scharping NE, Goldrath AW. CD8+ T cell metabolism in infection and cancer. Nat Rev Immunol. 2021;21:718–738. doi: 10.1038/s41577-021-00537-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Buck MD, O’Sullivan D, Pearce EL. T cell metabolism drives immunity. J Exp Med. 2015;212:1345–1360. doi: 10.1084/jem.20151159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.van der Windt GJ, Pearce EL. Metabolic switching and fuel choice during T-cell differentiation and memory development. Immunol Rev. 2012;249:27–42. doi: 10.1111/j.1600-065X.2012.01150.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Nolfi-Donegan D, Braganza A, Shiva S. Mitochondrial electron transport chain: Oxidative phosphorylation, oxidant production, and methods of measurement. Redox Biol. 2020;37:101674. doi: 10.1016/j.redox.2020.101674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Frauwirth KA, Riley JL, Harris MH, Parry RV, Rathmell JC, Plas DR, Elstrom RL, June CH, Thompson CB. The CD28 signaling pathway regulates glucose metabolism. Immunity. 2002;16:769–777. doi: 10.1016/s1074-7613(02)00323-0. [DOI] [PubMed] [Google Scholar]
- 19.Menk AV, Scharping NE, Moreci RS, Zeng X, Guy C, Salvatore S, Bae H, Xie J, Young HA, Wendell SG, et al. Early TCR signaling induces rapid aerobic glycolysis enabling distinct acute T cell effector functions. Cell Rep. 2018;22:1509–1521. doi: 10.1016/j.celrep.2018.01.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.MacIver NJ, Michalek RD, Rathmell JC. Metabolic regulation of T lymphocytes. Annu Rev Immunol. 2013;31:259–283. doi: 10.1146/annurev-immunol-032712-095956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Carr EL, Kelman A, Wu GS, Gopaul R, Senkevitch E, Aghvanyan A, Turay AM, Frauwirth KA. Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation. J Immunol. 2010;185:1037–1044. doi: 10.4049/jimmunol.0903586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Newsholme EA, Crabtree B, Ardawi MS. Glutamine metabolism in lymphocytes: its biochemical, physiological and clinical importance. Q J Exp Physiol. 1985;70:473–489. doi: 10.1113/expphysiol.1985.sp002935. [DOI] [PubMed] [Google Scholar]
- 23.Buck MD, O’Sullivan D, Klein Geltink RI, Curtis JD, Chang CH, Sanin DE, Qiu J, Kretz O, Braas D, van der Windt GJ, et al. Mitochondrial dynamics controls T cell fate through metabolic programming. Cell. 2016;166:63–76. doi: 10.1016/j.cell.2016.05.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ledderose C, Bao Y, Lidicky M, Zipperle J, Li L, Strasser K, Shapiro NI, Junger WG. Mitochondria are gate-keepers of T cell function by producing the ATP that drives purinergic signaling. J Biol Chem. 2014;289:25936–25945. doi: 10.1074/jbc.M114.575308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Yi JS, Holbrook BC, Michalek RD, Laniewski NG, Grayson JM. Electron transport complex I is required for CD8+ T cell function. J Immunol. 2006;177:852–862. doi: 10.4049/jimmunol.177.2.852. [DOI] [PubMed] [Google Scholar]
- 26.Amitrano AM, Berry BJ, Lim K, Kim KD, Waugh RE, Wojtovich AP, Kim M. Optical control of CD8+ T cell metabolism and effector functions. Front Immunol. 2021;12:666231. doi: 10.3389/fimmu.2021.666231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Pearce EL, Walsh MC, Cejas PJ, Harms GM, Shen H, Wang LS, Jones RG, Choi Y. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature. 2009;460:103–107. doi: 10.1038/nature08097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.O’Sullivan D, van der Windt GJ, Huang SC, Curtis JD, Chang CH, Buck MD, Qiu J, Smith AM, Lam WY, DiPlato LM, et al. Memory CD8+ T cells use cell-intrinsic lipolysis to support the metabolic programming necessary for development. Immunity. 2014;41:75–88. doi: 10.1016/j.immuni.2014.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.van der Windt GJ, Everts B, Chang CH, Curtis JD, Freitas TC, Amiel E, Pearce EJ, Pearce EL. Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity. 2012;36:68–78. doi: 10.1016/j.immuni.2011.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Marchetti P, Fovez Q, Germain N, Khamari R, Kluza J. Mitochondrial spare respiratory capacity: mechanisms, regulation, and significance in non-transformed and cancer cells. FASEB J. 2020;34:13106–13124. doi: 10.1096/fj.202000767R. [DOI] [PubMed] [Google Scholar]
- 31.van der Windt GJ, O’Sullivan D, Everts B, Huang SC, Buck MD, Curtis JD, Chang CH, Smith AM, Ai T, Faubert B, et al. CD8 memory T cells have a bioenergetic advantage that underlies their rapid recall ability. Proc Natl Acad Sci U S A. 2013;110:14336–14341. doi: 10.1073/pnas.1221740110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Almeida L, Lochner M, Berod L, Sparwasser T. Metabolic pathways in T cell activation and lineage differentiation. Semin Immunol. 2016;28:514–524. doi: 10.1016/j.smim.2016.10.009. [DOI] [PubMed] [Google Scholar]
- 33.Gerriets VA, Kishton RJ, Nichols AG, Macintyre AN, Inoue M, Ilkayeva O, Winter PS, Liu X, Priyadharshini B, Slawinska ME, et al. Metabolic programming and PDHK1 control CD4+ T cell subsets and inflammation. J Clin Invest. 2015;125:194–207. doi: 10.1172/JCI76012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Puleston DJ, Baixauli F, Sanin DE, Edwards-Hicks J, Villa M, Kabat AM, Kamiński MM, Stanckzak M, Weiss HJ, Grzes KM, et al. Polyamine metabolism is a central determinant of helper T cell lineage fidelity. Cell. 2021;184:4186–4202.e20. doi: 10.1016/j.cell.2021.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Dimeloe S, Mehling M, Frick C, Loeliger J, Bantug GR, Sauder U, Fischer M, Belle R, Develioglu L, Tay S, et al. The immune-metabolic basis of effector memory CD4+ T cell function under hypoxic conditions. J Immunol. 2016;196:106–114. doi: 10.4049/jimmunol.1501766. [DOI] [PubMed] [Google Scholar]
- 36.Scharping NE, Rivadeneira DB, Menk AV, Vignali PD, Ford BR, Rittenhouse NL, Peralta R, Wang Y, Wang Y, DePeaux K, et al. Mitochondrial stress induced by continuous stimulation under hypoxia rapidly drives T cell exhaustion. Nat Immunol. 2021;22:205–215. doi: 10.1038/s41590-020-00834-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15:486–499. doi: 10.1038/nri3862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wherry EJ, Ha SJ, Kaech SM, Haining WN, Sarkar S, Kalia V, Subramaniam S, Blattman JN, Barber DL, Ahmed R. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity. 2007;27:670–684. doi: 10.1016/j.immuni.2007.09.006. [DOI] [PubMed] [Google Scholar]
- 39.Bengsch B, Johnson AL, Kurachi M, Odorizzi PM, Pauken KE, Attanasio J, Stelekati E, McLane LM, Paley MA, Delgoffe GM, et al. Bioenergetic insufficiencies due to metabolic alterations regulated by the inhibitory receptor PD-1 are an early driver of CD8+ T cell exhaustion. Immunity. 2016;45:358–373. doi: 10.1016/j.immuni.2016.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ogando J, Sáez ME, Santos J, Nuevo-Tapioles C, Gut M, Esteve-Codina A, Heath S, González-Pérez A, Cuezva JM, Lacalle RA, et al. PD-1 signaling affects cristae morphology and leads to mitochondrial dysfunction in human CD8+ T lymphocytes. J Immunother Cancer. 2019;7:151. doi: 10.1186/s40425-019-0628-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kishton RJ, Sukumar M, Restifo NP. Metabolic regulation of T cell longevity and function in tumor immunotherapy. Cell Metab. 2017;26:94–109. doi: 10.1016/j.cmet.2017.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Clever D, Roychoudhuri R, Constantinides MG, Askenase MH, Sukumar M, Klebanoff CA, Eil RL, Hickman HD, Yu Z, Pan JH, et al. Oxygen sensing by T cells establishes an immunologically tolerant metastatic niche. Cell. 2016;166:1117–1131.e14. doi: 10.1016/j.cell.2016.07.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Siska PJ, van der Windt GJ, Kishton RJ, Cohen S, Eisner W, MacIver NJ, Kater AP, Weinberg JB, Rathmell JC. Suppression of Glut1 and Glucose metabolism by decreased Akt/mTORC1 signaling drives T cell impairment in B cell leukemia. J Immunol. 2016;197:2532–2540. doi: 10.4049/jimmunol.1502464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Chamoto K, Chowdhury PS, Kumar A, Sonomura K, Matsuda F, Fagarasan S, Honjo T. Mitochondrial activation chemicals synergize with surface receptor PD-1 blockade for T cell-dependent antitumor activity. Proc Natl Acad Sci U S A. 2017;114:E761–E770. doi: 10.1073/pnas.1620433114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat Rev Immunol. 2012;12:269–281. doi: 10.1038/nri3191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Rohaan MW, Wilgenhof S, Haanen JB. Adoptive cellular therapies: the current landscape. Virchows Arch. 2019;474:449–461. doi: 10.1007/s00428-018-2484-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Spiess PJ, Yang JC, Rosenberg SA. In vivo antitumor activity of tumor-infiltrating lymphocytes expanded in recombinant interleukin-2. J Natl Cancer Inst. 1987;79:1067–1075. [PubMed] [Google Scholar]
- 48.Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, Citrin DE, Restifo NP, Robbins PF, Wunderlich JR, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 2011;17:4550–4557. doi: 10.1158/1078-0432.CCR-11-0116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Gee AP. GMP CAR-T cell production. Best Pract Res Clin Haematol. 2018;31:126–134. doi: 10.1016/j.beha.2018.01.002. [DOI] [PubMed] [Google Scholar]
- 50.Kessels HW, Wolkers MC, van den Boom MD, van der Valk MA, Schumacher TN. Immunotherapy through TCR gene transfer. Nat Immunol. 2001;2:957–961. doi: 10.1038/ni1001-957. [DOI] [PubMed] [Google Scholar]
- 51.Eshhar Z, Waks T, Gross G, Schindler DG. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci U S A. 1993;90:720–724. doi: 10.1073/pnas.90.2.720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Maher J, Brentjens RJ, Gunset G, Rivière I, Sadelain M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRζ/CD28 receptor. Nat Biotechnol. 2002;20:70–75. doi: 10.1038/nbt0102-70. [DOI] [PubMed] [Google Scholar]
- 53.Hong M, Clubb JD, Chen YY. Engineering CAR-T cells for next-generation cancer therapy. Cancer Cell. 2020;38:473–488. doi: 10.1016/j.ccell.2020.07.005. [DOI] [PubMed] [Google Scholar]
- 54.Maus MV, Grupp SA, Porter DL, June CH. Antibody-modified T cells: CARs take the front seat for hematologic malignancies. Blood. 2014;123:2625–2635. doi: 10.1182/blood-2013-11-492231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hupperetz C, Lah S, Kim H, Kim CH. CAR T cell immunotherapy beyond haematological malignancy. Immune Netw. 2022;22:e6. doi: 10.4110/in.2022.22.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Srivastava S, Riddell SR. Chimeric antigen receptor T cell therapy: challenges to bench-to-bedside efficacy. J Immunol. 2018;200:459–468. doi: 10.4049/jimmunol.1701155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Parry RV, Chemnitz JM, Frauwirth KA, Lanfranco AR, Braunstein I, Kobayashi SV, Linsley PS, Thompson CB, Riley JL. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol Cell Biol. 2005;25:9543–9553. doi: 10.1128/MCB.25.21.9543-9553.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lee JB, Kim HR, Ha SJ. Immune checkpoint inhibitors in 10 years: contribution of basic research and clinical application in cancer immunotherapy. Immune Netw. 2022;22:e2. doi: 10.4110/in.2022.22.e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Bonaventura P, Shekarian T, Alcazer V, Valladeau-Guilemond J, Valsesia-Wittmann S, Amigorena S, Caux C, Depil S. Cold tumors: a therapeutic challenge for immunotherapy. Front Immunol. 2019;10:168. doi: 10.3389/fimmu.2019.00168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Pagès F, Kirilovsky A, Mlecnik B, Asslaber M, Tosolini M, Bindea G, Lagorce C, Wind P, Marliot F, Bruneval P, et al. In situ cytotoxic and memory T cells predict outcome in patients with early-stage colorectal cancer. J Clin Oncol. 2009;27:5944–5951. doi: 10.1200/JCO.2008.19.6147. [DOI] [PubMed] [Google Scholar]
- 61.Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, Makrigiannakis A, Gray H, Schlienger K, Liebman MN, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348:203–213. doi: 10.1056/NEJMoa020177. [DOI] [PubMed] [Google Scholar]
- 62.Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18:843–851. doi: 10.1038/mt.2010.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Allavena P, Germano G, Marchesi F, Mantovani A. Chemokines in cancer related inflammation. Exp Cell Res. 2011;317:664–673. doi: 10.1016/j.yexcr.2010.11.013. [DOI] [PubMed] [Google Scholar]
- 64.Lanitis E, Dangaj D, Irving M, Coukos G. Mechanisms regulating T-cell infiltration and activity in solid tumors. Ann Oncol. 2017;28:xii18–xii32. doi: 10.1093/annonc/mdx238. [DOI] [PubMed] [Google Scholar]
- 65.Shulman Z, Shinder V, Klein E, Grabovsky V, Yeger O, Geron E, Montresor A, Bolomini-Vittori M, Feigelson SW, Kirchhausen T, et al. Lymphocyte crawling and transendothelial migration require chemokine triggering of high-affinity LFA-1 integrin. Immunity. 2009;30:384–396. doi: 10.1016/j.immuni.2008.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Jiang H, Hegde S, Knolhoff BL, Zhu Y, Herndon JM, Meyer MA, Nywening TM, Hawkins WG, Shapiro IM, Weaver DT, et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat Med. 2016;22:851–860. doi: 10.1038/nm.4123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Chen IX, Chauhan VP, Posada J, Ng MR, Wu MW, Adstamongkonkul P, Huang P, Lindeman N, Langer R, Jain RK. Blocking CXCR4 alleviates desmoplasia, increases T-lymphocyte infiltration, and improves immunotherapy in metastatic breast cancer. Proc Natl Acad Sci U S A. 2019;116:4558–4566. doi: 10.1073/pnas.1815515116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Kishore M, Cheung KC, Fu H, Bonacina F, Wang G, Coe D, Ward EJ, Colamatteo A, Jangani M, Baragetti A, et al. Regulatory T cell migration is dependent on glucokinase-mediated glycolysis. Immunity. 2017;47:875–889.e10. doi: 10.1016/j.immuni.2017.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Marelli-Berg FM, Jangani M. Metabolic regulation of leukocyte motility and migration. J Leukoc Biol. 2018;104:285–293. doi: 10.1002/JLB.1MR1117-472R. [DOI] [PubMed] [Google Scholar]
- 70.Campello S, Lacalle RA, Bettella M, Mañes S, Scorrano L, Viola A. Orchestration of lymphocyte chemotaxis by mitochondrial dynamics. J Exp Med. 2006;203:2879–2886. doi: 10.1084/jem.20061877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, Rossig C, Russell HV, Diouf O, Liu E, et al. Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma. Blood. 2011;118:6050–6056. doi: 10.1182/blood-2011-05-354449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Robbins PF, Dudley ME, Wunderlich J, El-Gamil M, Li YF, Zhou J, Huang J, Powell DJ, Jr, Rosenberg SA. Cutting edge: persistence of transferred lymphocyte clonotypes correlates with cancer regression in patients receiving cell transfer therapy. J Immunol. 2004;173:7125–7130. doi: 10.4049/jimmunol.173.12.7125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Gattinoni L, Klebanoff CA, Palmer DC, Wrzesinski C, Kerstann K, Yu Z, Finkelstein SE, Theoret MR, Rosenberg SA, Restifo NP. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J Clin Invest. 2005;115:1616–1626. doi: 10.1172/JCI24480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Sukumar M, Liu J, Ji Y, Subramanian M, Crompton JG, Yu Z, Roychoudhuri R, Palmer DC, Muranski P, Karoly ED, et al. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J Clin Invest. 2013;123:4479–4488. doi: 10.1172/JCI69589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Crompton JG, Sukumar M, Roychoudhuri R, Clever D, Gros A, Eil RL, Tran E, Hanada K, Yu Z, Palmer DC, et al. Akt inhibition enhances expansion of potent tumor-specific lymphocytes with memory cell characteristics. Cancer Res. 2015;75:296–305. doi: 10.1158/0008-5472.CAN-14-2277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kawalekar OU, O’Connor RS, Fraietta JA, Guo L, McGettigan SE, Posey AD, Jr, Patel PR, Guedan S, Scholler J, Keith B, et al. Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity. 2016;44:380–390. doi: 10.1016/j.immuni.2016.01.021. [DOI] [PubMed] [Google Scholar]
- 77.Menk AV, Scharping NE, Rivadeneira DB, Calderon MJ, Watson MJ, Dunstane D, Watkins SC, Delgoffe GM. 4-1BB costimulation induces T cell mitochondrial function and biogenesis enabling cancer immunotherapeutic responses. J Exp Med. 2018;215:1091–1100. doi: 10.1084/jem.20171068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Araki K, Turner AP, Shaffer VO, Gangappa S, Keller SA, Bachmann MF, Larsen CP, Ahmed R. mTOR regulates memory CD8 T-cell differentiation. Nature. 2009;460:108–112. doi: 10.1038/nature08155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kagoya Y, Nakatsugawa M, Yamashita Y, Ochi T, Guo T, Anczurowski M, Saso K, Butler MO, Arrowsmith CH, Hirano N. BET bromodomain inhibition enhances T cell persistence and function in adoptive immunotherapy models. J Clin Invest. 2016;126:3479–3494. doi: 10.1172/JCI86437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Geiger R, Rieckmann JC, Wolf T, Basso C, Feng Y, Fuhrer T, Kogadeeva M, Picotti P, Meissner F, Mann M, et al. L-arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell. 2016;167:829–842.e13. doi: 10.1016/j.cell.2016.09.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Xu Y, Zhang M, Ramos CA, Durett A, Liu E, Dakhova O, Liu H, Creighton CJ, Gee AP, Heslop HE, et al. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood. 2014;123:3750–3759. doi: 10.1182/blood-2014-01-552174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Phan AT, Doedens AL, Palazon A, Tyrakis PA, Cheung KP, Johnson RS, Goldrath AW. Constitutive glycolytic metabolism supports CD8+ T cell effector memory differentiation during viral infection. Immunity. 2016;45:1024–1037. doi: 10.1016/j.immuni.2016.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Alamino VA, Mascanfroni ID, Montesinos MM, Gigena N, Donadio AC, Blidner AG, Milotich SI, Cheng SY, Masini-Repiso AM, Rabinovich GA, et al. Antitumor responses stimulated by dendritic cells are improved by triiodothyronine binding to the thyroid hormone receptor β. Cancer Res. 2015;75:1265–1274. doi: 10.1158/0008-5472.CAN-14-1875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kroemer G, Pouyssegur J. Tumor cell metabolism: cancer’s Achilles’ heel. Cancer Cell. 2008;13:472–482. doi: 10.1016/j.ccr.2008.05.005. [DOI] [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;38:2438–2450. doi: 10.1002/eji.200838289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Maciver NJ, Jacobs SR, Wieman HL, Wofford JA, Coloff JL, Rathmell JC. Glucose metabolism in lymphocytes is a regulated process with significant effects on immune cell function and survival. J Leukoc Biol. 2008;84:949–957. doi: 10.1189/jlb.0108024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Zhang Y, Kurupati R, Liu L, Zhou XY, Zhang G, Hudaihed A, Filisio F, Giles-Davis W, Xu X, Karakousis GC, 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. doi: 10.1016/j.ccell.2017.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Edwards DN, Ngwa VM, Raybuck AL, Wang S, Hwang Y, Kim LC, Cho SH, Paik Y, Wang Q, Zhang S, et al. Selective glutamine metabolism inhibition in tumor cells improves antitumor T lymphocyte activity in triple-negative breast cancer. J Clin Invest. 2021;131:e140100. doi: 10.1172/JCI140100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Best SA, Gubser PM, Sethumadhavan S, Kersbergen A, Negrón Abril YL, Goldford J, Sellers K, Abeysekera W, Garnham AL, McDonald JA, et al. Glutaminase inhibition impairs CD8 T cell activation in STK11-/Lkb1-deficient lung cancer. Cell Metab. 2022;34:874–887.e6. doi: 10.1016/j.cmet.2022.04.003. [DOI] [PubMed] [Google Scholar]
- 90.Carmona-Fontaine C, Deforet M, Akkari L, Thompson CB, Joyce JA, Xavier JB. Metabolic origins of spatial organization in the tumor microenvironment. Proc Natl Acad Sci U S A. 2017;114:2934–2939. doi: 10.1073/pnas.1700600114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Chang CH, Pearce EL. Emerging concepts of T cell metabolism as a target of immunotherapy. Nat Immunol. 2016;17:364–368. doi: 10.1038/ni.3415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Scharping NE, Menk AV, Whetstone RD, Zeng X, Delgoffe GM. Efficacy of PD-1 blockade is potentiated by metformin-induced reduction of tumor hypoxia. Cancer Immunol Res. 2017;5:9–16. doi: 10.1158/2326-6066.CIR-16-0103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Najjar YG, Menk AV, Sander C, Rao U, Karunamurthy A, Bhatia R, Zhai S, Kirkwood JM, Delgoffe GM. Tumor cell oxidative metabolism as a barrier to PD-1 blockade immunotherapy in melanoma. JCI Insight. 2019;4:e124989. doi: 10.1172/jci.insight.124989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.van Bruggen JA, Martens AW, Fraietta JA, Hofland T, Tonino SH, Eldering E, Levin MD, Siska PJ, Endstra S, Rathmell JC, et al. Chronic lymphocytic leukemia cells impair mitochondrial fitness in CD8+ T cells and impede CAR T-cell efficacy. Blood. 2019;134:44–58. doi: 10.1182/blood.2018885863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Budimir N, Thomas GD, Dolina JS, Salek-Ardakani S. Reversing T-cell exhaustion in cancer: lessons learned from PD-1/PD-L1 immune checkpoint blockade. Cancer Immunol Res. 2022;10:146–153. doi: 10.1158/2326-6066.CIR-21-0515. [DOI] [PubMed] [Google Scholar]
- 96.Pauken KE, Sammons MA, Odorizzi PM, Manne S, Godec J, Khan O, Drake AM, Chen Z, Sen DR, Kurachi M, et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science. 2016;354:1160–1165. doi: 10.1126/science.aaf2807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, Rosenberg SA. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114:1537–1544. doi: 10.1182/blood-2008-12-195792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Jang SY, Kang HT, Hwang ES. Nicotinamide-induced mitophagy: event mediated by high NAD+/NADH ratio and SIRT1 protein activation. J Biol Chem. 2012;287:19304–19314. doi: 10.1074/jbc.M112.363747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Finck BN, Kelly DP. PGC-1 coactivators: inducible regulators of energy metabolism in health and disease. J Clin Invest. 2006;116:615–622. doi: 10.1172/JCI27794. [DOI] [PMC free article] [PubMed] [Google Scholar]



