Metabolic regulation of immune responses to cancer ================================================== * Jannis Wißfeld * Anke Werner * Xin Yan * Nora ten Bosch * Guoliang Cui ## Abstract The tumor microenvironment is an ecosystem composed of multiple types of cells, such as tumor cells, immune cells, and cancer-associated fibroblasts. Cancer cells grow faster than non-cancerous cells and consume larger amounts of nutrients. The rapid growth characteristic of cancer cells fundamentally alters nutrient availability in the tumor microenvironment and results in reprogramming of immune cell metabolic pathways. Accumulating evidence suggests that cellular metabolism of nutrients, such as lipids and amino acids, beyond being essential to meet the bioenergetic and biosynthetic demands of immune cells, also regulates a broad spectrum of cellular signal transduction, and influences immune cell survival, differentiation, and anti-tumor effector function. The cancer immunometabolism research field is rapidly evolving, and exciting new discoveries are reported in high-profile journals nearly weekly. Therefore, all new findings in this field cannot be summarized within this short review. Instead, this review is intended to provide a brief introduction to this rapidly developing research field, with a focus on the metabolism of two classes of important nutrients—lipids and amino acids—in immune cells. We highlight recent research on the roles of lipids and amino acids in regulating the metabolic fitness and immunological functions of T cells, macrophages, and natural killer cells in the tumor microenvironment. Furthermore, we discuss the possibility of “editing” metabolic pathways in immune cells to act synergistically with currently available immunotherapies in enhancing anti-tumor immune responses. * Lipids * amino acids * cancer * anti-tumor immunity * T cells * NK cells * metabolism * immunometabolism ## Introduction In recent decades, cancer research has made substantial progress in the understanding of cancer biology and the development of therapy approaches against specific cancers. Scientists from the German Cancer Research Center (DKFZ) have been major drivers in this process. In 1989, a research group from DKFZ found that monoclonal antibodies targeting APO-1 (also known as CD95 or FasR) on human lymphoma cells trigger apoptosis of lymphoma cells *in vivo* and induce regression of lymphoma in a mouse model1. A broad spectrum of tumor cells express the ligand of APO-1, APO-1L (also known as CD95L, CD178, and FasL), which induces apoptosis of APO-1-expressing lymphocytes *in vitro*, in a process called tumor counterattack. *In vivo*, the interaction between APO-1 and APO-1L delays tumor growth in a neutrophil-independent manner2. More recent approaches target mechanisms of T cell exhaustion, a term used interchangeably with T cell dysfunction herein, and enhance anti-tumor immunity by using immune checkpoint blockade-based treatments. The most frequent targets are CTLA-4, PD-1, and its ligand PD-L13,4. In addition, combinatory treatments such as anti-CD40 and an inhibitor of MAPK and ERK have been found to be promising candidates. This polytherapy synergistically suppresses Kras mutation-driven pancreatic ductal adenocarcinoma5. Moreover, inhibition of C-X-C motif chemokine 12 (CXCL12) has been found to promote T cell accumulation and to act synergistically with checkpoint inhibitors, thus providing clinical benefits to patients with advanced stage pretreated metastatic colorectal and pancreatic cancer in a phase I/II trial6. Furthermore, IL-10 prevents excessive activation-induced exhaustion of CD8+ T cells in a model of chronic lymphocytic leukemia and delays the development of leukemia7. Finally, our group has found that T cells express regulator of G-protein signaling (Rgs)-16 at high levels. *Rgs16* deficiency inhibits CD8+ T cell apoptosis and acts synergistically with PD-1 blockade in enhancing anti-tumor CD8+ T cell responses. Human *RGS16* mRNA expression levels in the CD8+ tumor-infiltrating T cells of patients with melanoma negatively correlate with the expression of genes associated with T cell stemness and are predictive of low responses to PD-1 blockade8. However, checkpoint blockade therapy approaches have been only partially successful. Consequently, the influence of the tumor microenvironment (TME) on anti-tumor immunity and the field of immunometabolism have become a focus. Tumor-derived metabolic stimuli shape the TME into an immunosuppressive region, thereby hampering therapeutic approaches. In a mouse model of non-viral hepatocellular carcinoma (HCC), nonalcoholic steatohepatitis has been found to induce HCC. Liver-resident CD8+ T cells are sensitive to metabolic stimuli and are aberrantly activated by anti-PD-1 treatment. These auto-reactive CD8+PD-1+ T cells cause tissue damage instead of leading to HCC regression. Depleting CD8+ T cells or neutralizing TNFα has been found to ameliorate HCC progression in mice receiving anti-PD1, thus suggesting a rationale for stratifying patients with HCC according to etiology before immune checkpoint-based immunotherapy9,10. Kupffer cell-derived reactive oxygen species and TNFα promote cholangiocellular proliferation and oncogenic transformation. Depletion of Kupffer cells or blocking the TNFα signaling pathway decreases cholangiocellular oncogenic transformation11. Furthermore, macrophage-derived NO is required for the expression of vessel adhesion molecules, which are required for T cell extravasation and infiltration into tumors12. Interestingly, alterations of nutrient availability in the TME have also been shown to affect anti-tumor immunity. These alterations can be triggered by genetic mutations, which reprogram tumor cell metabolic pathways13. Our laboratory has found that CD8+ T cells in tumors increase the uptake of oxidized low-density lipoprotein (oxLDL) in a CD36-dependent manner. OxLDL promotes CD8+ T cell exhaustion through lipid peroxidation14. Moreover, the amino acid tryptophan is catabolized by tryptophan-2,3-dioxygenase and indoleamine 2,3 dioxygenase (IDO) in human tumor cells into kynurenine, an endogenous ligand of the transcription factor aryl hydrocarbon receptor (AHR). Kynurenine potently suppresses antitumor immune responses15, as extensively discussed in a later section herein. A recent study has revealed that the L-amino acid oxidase interleukin-4-induced-1 (IL4I1) generates indole metabolites and kynurenic acid, which are agonists of AHR, thereby promoting cancer cell motility and suppressing adaptive immunity. Because IDO inhibitors do not block IL4I1, these findings may explain the failure of a phase III clinical trial combining immune checkpoint blockade with IDO1 inhibition for cancer treatment16. An additional important finding of DKFZ scientists was that tumor cells with mutations in the isocitrate dehydrogenase gene produce the oncometabolite (R)-2-hydroxyglutarate, which inhibits immune responses and promotes tumor immunosuppression in the TME17. These recent advances in immunometabolism research underscore the importance of understanding how the altered availability of nutrients, including glucose, lipids, and amino acids, in the TME changes anti-tumor immunity. In this review, we discuss the influence of changes in lipid and amino acid content within the TME on anti-tumor immunity. The role of glucose metabolism on immune responses to cancer has been extensively reviewed elsewhere and thus will not be discussed here18–22. ## Lipid metabolism regulates immune responses to tumors Lipids are a diverse class of biomolecules with multiple important functions, such as serving as structural components of cell membranes, producing energy, and transducing intracellular and intercellular signals. Structural lipids, including phospholipids, sphingolipids, and cholesterol, are the major components of the plasma membrane, whereas fatty acids provide a basis for cellular bioenergetics through the β-oxidation pathway. Furthermore, several lipid molecules bind intracellular receptors, including peroxisome proliferator-activated receptors (PPARs) and sterol regulatory element-binding protein (SREBP), and subsequently regulate the transcription of genes involved in energy homeostasis and inflammation23. Several lines of evidence suggest that lipids are enriched in the TME and are required for immune cells to meet energy demands. However, certain lipid species in the TME cause immunosuppression, and promote cancer survival and metastasis14,23,24. In this section, we discuss the complicated immunosupportive or immunosuppressive roles of lipid species, such as fatty acids, cholesterol, and oxLDL (**Figure 1**). ![Figure 1](http://www.cancerbiomed.org/https://www.cancerbiomed.org/content/cbm/early/2022/10/24/j.issn.2095-3941.2022.0381/F1.medium.gif) [Figure 1](http://www.cancerbiomed.org/content/early/2022/10/24/j.issn.2095-3941.2022.0381/F1) Figure 1 Lipid metabolism impairs anti-tumor immunity. In the lipid-rich TME, infiltrating CD8+ T cells upregulate CD36 or FATPs, thereby increasing lipid uptake. Lipid accumulation and subsequent storage in lipid droplets results in a metabolic switch toward fatty acid oxidation (FAO) *via* STAT3 and CPT1, thus decreasing CD8+ T cell proliferation, survival, and overall effector function, but increasing susceptibility to ferroptosis and lipotoxicity. In contrast, Treg cells use FAO and oxidative phosphorylation to sustain their immunosuppressive phenotype in a FoxP3-dependent manner through downregulation of GLUT1 and the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin complex 1 (mTORC1) pathway. Lipid accumulation in DCs is mediated by MRS1/CD204 as well as in an XBP1-dependent manner, and it interferes with TLR stimulation and proliferation of DCs. Furthermore, accumulated lipids impair antigen processing and cross-presentation by HSP70 resorption. Neutrophils accumulate lipids *via* CD36/FATPs and produce leukotrienes *via* ALOX5. Lipid droplets are then transferred to metastasis-initiating tumor cells, where they facilitate survival. Natural killer (NK) cells show an impaired metabolic profile characterized by PPAR-α/δ-driven lipid accumulation and a decrease in phosphoinositide 3-kinase-Akt-mTORC1 signaling. This metabolic shift results in decreased secretion of effector cytokines, granzymes, and perforins, as well as decreased tumor cell lysis. Tumor-associated macrophages (TAMs) increase lipid uptake and storage, as well as PPAR-γ signaling *via* FAO and STAT6, thus increasing the secretion of tumor promoting and anti-inflammatory factors, and supporting angiogenesis and metastasis. Abbreviations: Akt, Akt serine/threonine kinase 1; ALOX5, arachidonate 5-lipoxygenase; ARG1, arginase 1; CCL22, chemokine (C-C motif) ligand 22; CD, cluster of differentiation; CPT1, carnitine palmitoyltransferase 1; DC, dendritic cell; FAO, fatty acid oxidation; FATP, fatty acid transport protein; FoxP3, forkhead box P3; GLUT1, glucose transporter 1; HSP70, heat shock protein 70 kDa; IFN-γ, interferon γ; IL, interleukin; MHC, major histocompatibility complex; MMP9, matrix metallopeptidase 9; MRS1, macrophage scavenger receptor 1; mTORC1, mechanistic target of rapamycin kinase complex 1; NO, nitric oxide; PD-1, programmed cell death protein 1; PD-L1, programmed cell death 1 ligand 1; PPAR, peroxisome proliferator activated receptor; STAT, signal transducer and activator of transcription; TAM, tumor-associated macrophage; TME, tumor microenvironment; Treg cells, regulatory T cells; XBP1, X-box binding protein 1. Parts of the figure were drawn by using original or modified pictures from Servier Medical Art. Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License ([https://creativecommons.org/licenses/by/3.0/](https://creativecommons.org/licenses/by/3.0/)). ### T cells T cells are important mediators of anti-tumor immunity. CD8+ cytotoxic T lymphocytes directly or indirectly kill cancer cells through releasing effector cytokines (such as IFNγ, TNFα, granzymes, and perforin) and through death receptor-mediated contact-dependent mechanisms. After activation, T cells undergo a metabolic switch by increasing the rates of glycolysis and glutaminolysis to meet the high energy demands25. However, concentrations of glucose in the TME are very low, and T cells must compete with cancer cells for this scarce nutrient26,27. To that end, T cells increase their reliance on lipids for energy production24. Promotion of fatty acid β-oxidation through treatment with the PPARα agonist fenofibrate improves CD8+ T cell multifunctionality in a mouse melanoma model and delays tumor progression24. Furthermore, bezafibrate, a pan-agonist for all PPAR isoforms, increases fatty acid β-oxidation, oxidative phosphorylation, glycolysis, and CD8+ T cell survival, and restores CD8+ T cell effector function in combination with anti-PD-L1 therapy in a sarcoma mouse model, *via* upregulation of carnitine palmitoyltransferase I (CPT1) α and B cell lymphoma 2 (BCL2)28. Moreover, activation of PPARα and PPARβ/δ *via* the tool compound GW501516 increases CPT1α expression and fatty acid β-oxidation, which are accompanied by enhanced production of IFNγ and prolonged survival of mice in an adoptive cell therapy model29. However, excessive lipid enrichment in the TME causes CD8+ T cell exhaustion. Tumor-infiltrating CD8+ T cells adapt to the lipid-rich TME by upregulating the expression of lipid transport proteins, such as CD36, which imports fatty acids including long-chain fatty acids (LCFA)14,30. The intracellular accumulation of LCFA drives CD8+ T cell dysfunction by inducing lipotoxicity. This LCFA-induced immunosuppression is further enhanced by the downregulation of very long-chain acyl-CoA dehydrogenase, an enzyme required to metabolize fatty acids, thus further increasing LCFA accumulation and exacerbating lipotoxicity30. Beyond fatty acids, CD36 is required to import oxLDL, a lipid species abundant in the TME14. OxLDL uptake into tumor-infiltrating CD8+ T cells promotes lipid peroxidation and ferroptosis of exhausted CD8+ T cells. Overexpression of the glutathione peroxidase GPX4, an antioxidant defense enzyme, enhances anti-tumor CD8+ T cell function14. One intriguing question is which molecular pathways drive tumor-infiltrating CD8+ T cells to adapt to the glucose-poor and lipid-rich TME. The interaction between PD-1 and its ligand PD-L1 inhibits glycolysis and promotes fatty acid β-oxidation in CD8+ T cells26,31,32, and glycolysis inhibition suppresses CD8+ T cell effector function33–35. PD-1 ligation activates signal transducer and activator of transcription 3 (STAT3), which in turn mediates the metabolic switch from glycolysis to fatty acid β-oxidation in CD8+ T cells in a mouse model of spontaneously developed mammary tumors32. Another subset of immune cells, CD4+ FoxP3+ regulatory T (Treg) cells, despite being functionally distinct from CD8+ T cells, require fatty acid β-oxidation to exert their immunoregulatory function36,37. FoxP3, the key transcription factor of Treg cells, promotes the expression of genes involved in oxidative metabolism at the expense of genes associated with glycolysis38,39. FoxP3 protects Treg cells against lipid enrichment-mediated lipotoxicity by enhancing fatty acid β-oxidation and thereby decreasing fatty acid accumulation40. Beyond fatty acids, cholesterol is also enriched in the TME, and tumor-infiltrating CD8+ T cells adapt to the cholesterol enrichment by increasing their uptake of cholesterol beyond that in splenic CD8+ T cells14. An increase in cholesterol in CD8+ T cells results in endoplasmic reticulum stress and T cell dysfunction41, a phenotype similar to that of CD8+ T cells deficient in synthesizing sphingolipids42. Plasma membrane cholesterol promotes T cell receptor clustering and enhances immunological synapse formation43. Genetic deficiency or pharmacological inhibition of acyl-CoA:cholesterol acyltransferase 1 (ACAT1), a key enzyme catalyzing cholesterol esterification, enhances the effector function and proliferation of CD8+ T cells and limits melanoma growth as well as metastasis in mice, owing to an increase in plasma membrane cholesterol. ### Natural killer (NK) cells NK cells are part of the innate immune system and are potent producers of cytokines, including IFNγ and TNFα. Similarly to CD8+ cytotoxic T lymphocytes, NK cells kill malignant cells through the release of cytotoxic agents, such as perforins and granzymes, or through death receptor-ligand engagement44,45. The influence of dietary lipids on NK cell effector function has been studied intensively. Obesity is inversely correlated with NK cell proliferation and cancer cell cytolytic activity in both humans and mouse models46–48, thus implying that NK cell effector function is inhibited by lipids. Prostaglandin E is a physiologically active lipid produced by thyroid cancer cells49. Cancer cell-derived prostaglandin E inhibits NK cell effector cytokine production and suppresses the cytolytic activity of NK cells. Inhibition of the prostaglandin E2 receptor EP4 by RQ-15986 has been found to rescue the effector function of NK cells and to inhibit metastasis in a mouse model of metastatic breast cancer50. Furthermore, NK cells in diffuse large B cell lymphoma undergo metabolic and transcriptional reprogramming characteristic of an increase in lipid metabolism. Exposure of NK cells to a cocktail of lipids impairs NK cell function, similarly to the NK cell dysfunction observed in the lymphoma environment51. PPARα and PPARβ/δ increase the expression of the lipid transport protein CD36 and low-density lipoprotein receptor, thus increasing lipid uptake and accumulation in NK cells. Lipid accumulation causes a dysfunctional NK cell phenotype characterized by decreased production of effector cytokines and tumor cell lysis. The CPT1 inhibitor etomoxir restores the cytotoxicity of NK cells52. Similarly to PPAR, SREBP family transcription factors are conventionally known as master regulators of lipid homeostasis. SREBPs play essential roles in interleukin-2 (IL-2) and IL-12-induced metabolic reprogramming of NK cells, independently of their role in regulating lipid biosynthesis. NK cells require SREBP to elevate glycolysis and engage the citrate-malate shuttle, thereby producing IFNγ ### Tumor-associated macrophages (TAMs) TAMs are present in many types of tumors, and their abundance is positively correlated with cancer progression and poor clinical outcomes54–58. Macrophages are broadly classified into pro-inflammatory (M1) and anti-inflammatory (M2) macrophages, although they display a broad spectrum of intermediate phenotypes between M1 and M2 macrophages59,60. This classification echoes the early definition of “classically activated macrophages” and “alternatively activated macrophages”61–63. Metabolites in the TME, such as lactate and fatty acids, promote the differentiation of TAMs into an M2-like phenotype64,65. TAMs inhibit anti-tumor immune responses through the secretion of IL-1066,67 and TGFβ68,69, and the recruitment of Treg cells *via* the chemokine CCL2270. They also promote angiogenesis by secreting vascular endothelial growth factor71–73 and promote metastasis by digesting extracellular matrix proteins74–77. The role of fatty acid β-oxidation in IL-4-driven M2 macrophage differentiation remains debated. Some studies have suggested that TAMs increase CD36 expression to import lipids for energy production. Fatty acid β-oxidation promotes reactive oxygen species production and IL-4-STAT6 activation, and is critical for TAM polarization. In addition, CD36-dependent uptake of triacylglycerol, and subsequent lipolysis and oxidation, are required for M2 macrophage activation65. Uptake of oxLDL by CD36 results in increased PPAR-γ signaling, which in turn promotes CD36 expression and fatty acid β-oxidation78–80. Moreover, uptake of LDL and oxLDL by the scavenger receptor MARCO fosters formation of lipid-loaded TAMs, which release CCL6 and thereby promote cancer cell migration81. In contrast, deficiency in CPT1a and CPT2 proteins, which are required for transporting long-chain fatty acids into the mitochondria for β-oxidation, do not affect IL-4-driven M2 macrophage differentiation82. Regardless of the complicated roles of fatty acid β-oxidation in IL-4-driven M2 macrophage differentiation, unsaturated fatty acids such as oleate promote the immunosuppressive and pro-tumor phenotype of TAMs through lipid droplet-dependent mechanisms83. In addition, SREBP1-dependent production of anti-inflammatory fatty acids contributes to the resolution of TLR4-mediated inflammation84, and Caveolin-1 participates in LXR-dependent cholesterol efflux and mediates anti-inflammatory properties85. Similarly, ovarian cancer cells facilitate membrane cholesterol efflux in TAMs, thus resulting in lipid raft breakdown and a subsequent increase in IL-4 signaling, which fosters the immunosuppressive TAM phenotype86. ### Dendritic cells (DCs) DCs are professional antigen-presenting cells with essential roles in activating antigen-specific T cells. The immunostimulatory function of DCs has been shown to be inhibited by a high-fat diet87. Administration of polyunsaturated fatty acids attenuates DC activation and maturation88,89. Intracellular lipid accumulation affects antigen presentation and subsequent activation of tumor antigen-specific T cells by DCs. Briefly, the macrophage scavenger receptor (MRS1) increases lipid uptake by DCs. Lipid-laden DCs have a diminished ability to stimulate the proliferation of antigen-specific T cells, owing to defects in antigen processing90. *Msr1* deficiency rescues the ability of DCs to activate antigen-specific T cells91. Furthermore, oxidized lipids, but not non-oxidized lipids, impair DC antigen cross-presentation92. Lipid peroxidation byproducts activate endoplasmic reticulum stress response factor X-box binding protein 1 (XBP1), thus resulting in lipid overloading and the inhibition of DC-mediated activation of anti-tumor T cells in metastatic ovarian cancer93. Oxidatively truncated lipid bodies in DCs bind heat shock protein 70 (Hsp70), a protein required for trafficking of peptide-major histocompatibility complex (MHC) class I complexes to the DC cell surface. Thus, the binding of oxidatively truncated lipid bodies to Hsp70 affects peptide-MHC trafficking to the DC cell surface and disrupts DC-mediated antigen cross-presentation in cancer94. ### Neutrophils Neutrophils suppress anti-tumor immune responses and support metastasis by producing leukotrienes95,96. Leukotrienes are bioactive lipid species that stimulate selective expansion of a subset of cancer cells with high tumorigenic potential, thus promoting tumor cell colonization of distant tissues in a mouse model of breast cancer. The production of leukotrienes is dependent on the enzyme arachidonate 5-lipoxygenase (Alox5). Inhibition of Alox5 reverses neutrophil-dependent tumor cell metastasis. The ability of neutrophils to facilitate the initiation of metastasis is associated with the accumulation of neutral lipids. In pre-metastatic states, neutrophils decrease adipose triglyceride lipase activity, thus leading to lipid accumulation. Neutrophils subsequently transfer lipids to metastatic tumor cells through a macropinocytosis-lysosome pathway, which increases tumor cell survival and proliferative ability97. In addition, tumor-derived oxysterols have been reported to recruit pro-tumor neutrophils in a manner dependent on C-X-C motif chemokine receptor 2, which in turn suppresses anti-tumor immune responses and supports tumor growth98. Collectively, lipids mediate the bi-directional communication between tumor cells and neutrophils, and cause immunosuppression and metastasis. ## Roles of amino acids in anti-tumor immunity Amino acids serve not only as building blocks for protein synthesis but also as precursors for many metabolites and signaling molecules involved in numerous intracellular and intercellular signal transduction pathways. Therefore, amino acid metabolism is essential for cell proliferation, survival, and function. More than 30 years ago, Chuang et al.99 demonstrated that deficiency in amino acids, including arginine, glutamine, leucine, threonine, and tryptophan, inhibits T cell proliferation. This study was followed by many publications in recent decades highlighting the importance of amino acids in T cell proliferation, effector function, and differentiation100,101. The availability of amino acids in the TME is substantially different from that in non-tumor tissues102, thus prompting the question of whether amino acids and their metabolites regulate T cell exhaustion in the TME. Whereas variations in certain amino acids, e.g., glutamine, are probably based on the increased uptake by cancer cells, concentrations of these nutrients fluctuate depending on the tumor type. However, one common feature of several cancer entities is the upregulation of the arginine and tryptophan degrading enzymes arginase and IDO, respectively103. Therefore, in this part of the review, we focus on arginine and tryptophan as examples to discuss the roles of amino acids in shaping the immunosuppressive nature of the TME (**Figure 2**). ![Figure 2](http://www.cancerbiomed.org/https://www.cancerbiomed.org/content/cbm/early/2022/10/24/j.issn.2095-3941.2022.0381/F2.medium.gif) [Figure 2](http://www.cancerbiomed.org/content/early/2022/10/24/j.issn.2095-3941.2022.0381/F2) Figure 2 Amino acid deprivation impairs anti-tumor immunity. The TME is characterized by low arginine and tryptophan content. Cancer cells, TAMs, and MDSCs express ARG1 and IDO enzymes. ARG1 hydrolyzes arginine to ornithine and urea, and IDO catalyzes the rate-limiting reaction in tryptophan catabolism, thus ultimately resulting in the depletion of these two amino acids within the TME. Tryptophan is finally metabolized to kynurenine, which also accumulates in the TME. Arginine starvation in T cells decreases CCND3 and CDK4, thereby arresting cells in G/G1 phase and decreasing proliferation. A decrease in immunological synapse formation is mediated by diminished dephosphorylation of CFL1 and a subsequent failure of actin remodeling, as a result of arginine starvation. Furthermore, decreases in mTORC1 signaling, glucose metabolism, and effector function are observed in T cells. Tryptophan starvation is accompanied by decreased proliferation and effector function, as well as cytokine production in T cells. Subsequently, the integrated stress response *via* GCN2 increases, and PD-1 expression is stimulated *via* kynurenine binding aryl hydrogen receptor (AHR). In NK cells, arginine starvation results in decreased proliferation, cytotoxicity, and effector function *via* downregulation of NKp30, NKp46, INF-γ, and CD3ζ. Tryptophan starvation decreases NK cell anti-tumor function by decreasing the expression of NKp44, CD25, and CD69, whereas excessive kynurenine triggers decreased expression of NKG2D and NKp46. In addition, excessive kynurenine increases the proportion of CD4+ CD25+ FOXP3+ regulatory T cells *via* AHR binding. Abbreviations: AHR, aryl hydrocarbon receptor; ARG1, arginase 1; CAT1, high affinity cationic amino acid transporter 1; CCND3, cyclin D3; CD, cluster of differentiation; CDK4, cyclin-dependent kinase 4; CFL1, cofilin 1; GCN2, general control nonderepressible 2; IDO, indolamine-2,3-dioxygenase; IFN-γ, interferon γ; IL, interleukin; MDSC, myeloid-derived suppressor cell; mTORC1, mechanistic target of rapamycin kinase complex; NK, natural killer; NKG2D, natural killer group 2D; NKp, natural killer protein; PD-1, programmed cell death protein 1; TAM, tumor-associated macrophage; TGF, transforming growth factor; TIM-3, T-cell immunoglobulin and mucin-domain containing-3; TME, tumor microenvironment. Parts of the figure were drawn by using original or modified pictures from Servier Medical Art. Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License ([https://creativecommons.org/licenses/by/3.0/](https://creativecommons.org/licenses/by/3.0/)). ### Arginine Arginine is a basic proteinogenic amino acid involved in various metabolic pathways, including the synthesis of nitric oxide, polyamines, and collagen104. Arginine is also a key activator of mammalian target of rapamycin (mTOR), an important gatekeeper regulating cell growth, proliferation, and survival105. Arginine is defined as a semi-essential amino acid; i.e., although cells can synthesize arginine from the neutral amino acid citrulline through a two-step biochemical reaction involving the enzymes argininosuccinate synthase (ASS) and argininosuccinate lyase (ASL), it becomes limiting in certain situations with substantially elevated demand for arginine, such as cancer and chronic inflammation104,106,107. The concentrations of arginine in the plasma of healthy humans range between 70 and 120 μM108,109, and are higher than those in tumor interstitial fluids102,110 or the plasma in humans and mice with hematological malignancies111,112. Furthermore, the concentrations of arginine vary and are significantly lower in the core regions than the peripheral regions of tumors113. The mechanisms of arginine depletion in tumor tissues have been intensively investigated. One plausible explanation is that arginine is consumed at high rates by rapidly growing cancer cells. Furthermore, tumor-infiltrating myeloid cells and cancer cells express arginase 1, which hydrolyzes arginine103,112,114. The mechanisms through which immunosuppressive factors induce arginase expression have recently been reviewed and will not be discussed herein64,103,115–118. Arginase 1 breaks down arginine through several intracellular and extracellular mechanisms: (i) cells increase uptake of arginine through the cationic amino acid transporter CAT2B, and arginine is then hydrolyzed by intracellular arginase 1119, or (ii) cells secrete arginase 1 into the extracellular compartment, where it hydrolyzes free arginine120,121. Arginase 1 breaks arginine down into ornithine and urea104. Ornithine is a precursor of polyamines such as putrescine, spermine, and spermidine, which are necessary for cell proliferation104. #### T cells Arginine plays an essential role in T cell activation and proliferation. The proliferation of both murine and human T cells is completely inhibited when T cells are cultured in arginine-free medium110,122,123. Comparable results have been obtained when regular cell culture medium is supplemented with arginase, whereas T cell proliferation is restored by the addition of an arginase inhibitor110,124,125. Furthermore, murine T cell proliferation is rescued in arginase-treated medium in the presence of the neutral amino acid citrulline, which is used as a substrate for the enzymes ASS and ASL to synthesize arginine125. In contrast, human T cells are not able to synthesize arginine intracellularly, owing to insufficient ASS expression in the absence of arginine122,126. The gene encoding the ASS protein is argininosuccinate synthetase 1 (*ASS1*). *ASS1* is a direct target of the transcription factors activating transcription factor 4 (ATF4) and CCAAT/enhancer binding protein β (C/EBPβ). Despite the presence of ATF4 and C/EBPβ, *ASS1* is not expressed in human T cells stimulated in the absence of arginine. Arginine starvation induces genome-wide chromatin compaction and increases H3 lysine-9/lysine-27 trimethylation, thus decreasing DNA accessibility, and disrupting ATF4 and C/EBPβ binding at target genes122. Because of their inability to synthesize arginine intracellularly, human T cells depend on the uptake of arginine from the extracellular space, a process mediated by cationic amino acid transporter 1127. Blocking this CAT-mediated arginine transport significantly decreases T cell proliferation and survival126,128. Arginine deprivation affects T cell proliferation through multiple mechanisms, such as cell cycle arrest, decreased CD3ζ chain expression, insufficient T cell receptor signaling, decreased expression of IL-2 receptor α (CD25) and effector cytokines, and impairment of T cell metabolic fitness. Arginine increases cyclin D3 expression at the transcriptional, posttranscriptional, and translational levels129. Arginine starvation decreases the expression of cyclin D3, thus resulting in cell cycle arrest in G-G1 phase125,129. In addition, arginine deficiency decreases the expression of the CD3ζ chain on T cells123,124, thereby resulting in insufficient TCR signaling to activate T cells130. T cells with diminished expression of CD3ζ chain have been found in tumors of patients with non-small cell lung carcinoma and a murine Lewis lung carcinoma (3LL) model, and are accompanied by elevated arginase expression in tumor cells and CAT-2B-mediated arginine uptake in arginase-expressing tumor-associated myeloid cells120. Interestingly, this arginine deprivation-mediated inhibitory effect on CD3ζ chain expression appears to be specific to arginine, because the depletion of other amino acids, such as glutamine, glycine, leucine, and lysine, does not change the CD3ζ chain expression level123. The efficient formation of the immune synapse is a prerequisite for signal transmission between antigen presenting cells and T cells131. The dephosphorylation of the actin-binding protein cofilin is required to induce remodeling of the actin cytoskeleton in T cells132. Arginine deprivation increases cofilin phosphorylation in T cells, thus impairing formation of the immunological synapse133. Furthermore, phorbol 12-myristate 13-acetate (PMA) and ionomycin-induced T cell activation are impaired by arginine deprivation. Because PMA and ionomycin bypass the requirement for CD3ζ chain to activate T cells, arginine depletion appears to impair T cell activation not only by affecting the CD3ζ chain-associated T cell receptor proximal signaling components but also potentially by inhibiting the distal signaling components124. Moreover, the expression of IL-2 receptor α chain significantly decreases under arginine deprivation, thereby indicating that arginine is indispensable for IL-2 signaling—a key signaling pathway required for T cell proliferation123,124. Beyond IL-2 receptor α, cytokines including IFNγ, TNFα, IL-5, and IL-10, which play important roles in T cell differentiation and effector function, are produced at low levels by T cells cultured in the absence of arginine123,133. Arginine deprivation impairs glycolysis, the pentose phosphate pathway, the tricarboxylic acid cycle, and mTORC1 activity122,125. In line with these findings, elevated arginine concentrations significantly increase the survival of human and murine CD8+ T cells, and the anti-tumor activity of CD8+ T cells in a B16 melanoma mouse model. This enhanced survival is accompanied by a shift in metabolic status from glycolysis toward mitochondrial respiration in the presence of high concentrations of arginine. Furthermore, high concentrations of arginine promote T cell differentiation into a central memory T cell-like phenotype, thus favoring the long-term persistence of anti-tumor CD8+ T cells128. Because arginine plays essential roles in T cell activation and proliferation, inhibition of arginase has led to enthusiasm for reviving arginase-mediated dysfunctional T cells. For example, inhibition of PMN-derived arginase increases human T cell proliferation and induces higher levels of production of effector cytokines, such as IFNγ, IL-9, and IL-17. Furthermore, inhibition of arginase increases the expression of CD25 and re-expression of CD28 after the initial stimulation-induced downregulation124,134,135. Of note, the inhibition of arginase activity also reverses the anergic state of T cells in patients with multiple myeloma136. #### NK cells Similarly to T cells, NK cells require arginine for optimal proliferation. Arginine deprivation impairs the proliferation of murine and human NK cells induced by multiple stimuli, including IL-2 and PMA/ionomycin110,137,138. Compared with T cells, NK cells are less sensitive to low arginine levels. For example, T cells require arginine at a concentration of 20 μM to achieve half maximal proliferation, whereas 2 μM arginine is sufficient for NK cell half maximal proliferation138. Notably, NK cell viability is not affected by arginine starvation138. Similarly to that on T cells, ζ chain expression on NK cells is decreased by arginine starvation, thus impairing downstream signal transduction and decreasing NK cell cytotoxicity137. One report has shown that NK cell granule exocytosis and cytotoxicity are independent of extracellular arginine138. Despite the contradictory description of the influence of arginine deprivation on NK cytotoxicity, one commonality among reports is that arginine deprivation impairs IFNγ expression in NK cells through a post-transcriptional mechanism137,138. Additional evidence supporting the inhibitory role of arginine deprivation on NK cells is that arginase activity has been detected in damage-associated molecular patterns derived from mitochondrial preparations (MitoDAMPs)139. MitoDAMPs impair IFNγ secretion by NK cells and decrease the expression of the NK cell activating receptor NKG2D. The inhibitory effects of MitoDAMPs are reversed by the addition of extracellular arginine or an arginase inhibitor139. MitoDAMPs are detectable in cancers140,141, thus implying that MitoDAMPs may impair NK cell function by depleting arginine in the TME. ### Tryptophan Tryptophan is an essential amino acid. Because mammalian cells are incapable of synthesizing tryptophan, dietary intake is the major source. Tryptophan is crucial for protein synthesis, and maintaining cell growth and proliferation, and is involved in the biosynthesis of the neurotransmitter serotonin and the hormone melatonin. More than 95% of free tryptophan serves as a substrate for the kynurenine pathway, in which tryptophan is degraded to nicotinic acid, the precursor of nicotinamide adenine dinucleotide, a key coenzyme in energy metabolism and redox reactions142. The first and rate limiting step of the kynurenine pathway is the degradation of tryptophan to N-formylkynurenine, catalyzed by the enzymes indoleamine 2,3-dioxygenase 1 and 2 (IDO1 and IDO 2), or the tryptophan-2,3-dioxygenase. N-formylkynurenine is then converted by N-formylkynurenine formidase to kynurenine, which is further catabolized to 3-hydroxykynurenine, quinolinic acid, and picolinic acid142. IDO1 is expressed in tumor associated myeloid cells143–145 and many tumor cells, including melanomas, and cervix, kidney, non-small lung and colorectal carcinomas142,146–148. High expression of the tryptophan degrading enzyme IDO1 decreases the abundance of tryptophan in patients with breast cancer, colorectal cancer, head and neck cancer, prostate cancer, and lung cancer143. Reciprocally, kynurenine levels are elevated in the plasma of patients with cancer143,149. #### T cells T cells show less proliferation when cultured in tryptophan-free medium, IDO-conditioned medium, or medium supplemented with kynurenine and picolinic acid than in regular tryptophan-replete medium117,144,147,150. Of note, the effects of tryptophan deprivation on T cell proliferation vary across experimental conditions. For example, the anti-CD3 or concanavalin A-driven proliferation of murine CD8+ T cells is inhibited when cells are cultured with kynurenine and low concentrations of tryptophan, and this effect is accompanied by diminished secretion of the cytokines IL-2 and IFNγ150. In contrast, murine CD8+ T cells proliferate and produce cytokines normally when stimulated with PMA and ionomycin144,147,151, thus suggesting that tryptophan deprivation affects T cell proliferation and cytokine production by impairing T cell receptor proximal signaling components. Furthermore, the observation that tryptophan deprivation induces an integrated stress response also depends on the experimental setting. Briefly, in the absence of amino acids, the general control nonderepressible 2 (GCN2) kinase is activated by the accumulation of uncharged tRNAs and triggers the integrated stress response152. Exposure of murine CD8+ T cells to IDO-expressing DCs activates GCN2 kinase and results in complete inhibition of CD8+ T cell proliferation153. Similarly, GCN2 kinase is activated in CD4+ T cells by tryptophan deprivation117. In contrast to the two studies described above, GCN2 kinase activation is not observed in tumor-infiltrating T cells154. Beyond regulating T cell proliferation and stress responses, IDO regulates T cell differentiation. Co-culturing human CD4+ T cells with IDO-expressing DCs or cancer cells increases the differentiation of CD4+ CD25+ FoxP3+ Treg cells with potent suppressor function147,155,156. The enhanced Treg cell differentiation is accompanied by increased production of IL-10 and TGFβ, and a reciprocal decrease in IL-4 and IFNγ150. Inhibition of IDO suppresses Treg cell differentiation, which is restored by the addition of kynurenine155,156. IDO-driven Treg cell differentiation is dependent on the activation of the ligand-activated transcription factor AHR *via* kynurenine binding157,158. Kynurenine-mediated AHR activation upregulates PD-1 expression in tumor-infiltrating murine CD8+ T cells, thus conferring a CD8+ T cell exhaustion phenotype159,160. Furthermore, tryptophan-derived metabolites, such as kynurenic acid and xanthurenic acid, also activate AHR161,162 and may contribute to Treg cell differentiation and CD8+ T cell exhaustion. The mechanisms through which IDO inhibits T cell proliferation *in vivo* remain open to debate. One possibility is that IDO suppresses T cell responses simply by decreasing the availability of an important essential amino acid. The other possibility is that kynurenine, the product of an IDO-mediated enzymatic reaction, causes T cell inhibition. Mass spectroscopy analysis has demonstrated that intra-tumoral tryptophan concentrations are above the threshold triggering the integrated stress response154. Thus, the *in vivo* immunosuppressive function of tryptophan metabolism is likely not to be caused by tryptophan depletion154. #### NK cells Tryptophan metabolism also affects NK cell proliferation and effector function. Culturing NK cells in the presence of purified IDO enzyme or co-culturing NK cells with IDO-expressing DCs inhibits NK cell proliferation. This inhibitory effect is partly restored by an IDO inhibitor143,147,163. Similarly to that of T cells, NK cell cytotoxicity is inhibited by kynurenine. Kynurenine also decreases the expression of the NK cell activating receptor NKG2D and natural cytotoxicity triggering receptor 1 (NCR1, also known as NKp46)164. In contrast, NKp30-mediated cytotoxicity is unaffected by kynurenine, thus implying that the inhibitory function of kynurenine on NK cells is not dependent on the NKp30 pathway164. Finally, inhibition of IDO increases NK cell cytotoxicity and NK cell numbers in ovarian tumors163,165. ## Grant support Guoliang Cui is supported by a CRI Lloyd J. Old STAR Award (Grant No. 3914), a Helmholtz Young Investigator Award (Grant No. VH-NG-1113), an EMBO Young Investigator Award, an Exploration Grant of the Boehringer Ingelheim Foundation (BIS), the German Research Foundation (DFG, Grant Nos. CU375/5-1, CU375/5-2, CU375/7-1, CU375/9-1, and 259332240/RTG2099), the German Cancer Aid Foundation (DKH, Grant Nos. 70113343 and 70114224), the Helmholtz Zukunftsthema Ageing and Metabolic Programming (AMPro, Grant No. ZT0026), HI-TRON Kick-Start Seed Funding (Grant No. HITR-2021-08), the Hector Foundation (Grant No. M20102), and an ERC Consolidator Award (Grant No. 101045416). ## Conflict of interest statement GC receives funding from Bayer and Boehringer Ingelheim. ## Footnotes * *These authors contributed equally to this work. * Received July 5, 2022. * Accepted September 15, 2022. * Copyright: © 2022, Cancer Biology & Medicine [https://creativecommons.org/licenses/by/4.0/](https://creativecommons.org/licenses/by/4.0/) This is an open access article distributed under the terms of the [Creative Commons Attribution License (CC BY) 4.0](https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited. ## References 1. 1. Trauth BC, Klas C, Peters AMJ, Matzku S, Möller P, Falk W, et al. Monoclonal antibody-mediated tumor regression by induction of apoptosis. Science. 1989; 245: 301–5. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIyNDUvNDkxNS8zMDEiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 2. 2. Igney FH, Behrens CK, Krammer PH. CD95L mediates tumor counterattack in vitro but induces neutrophil-independent tumor rejection in vivo. Int J Cancer. 2005; 113: 78–87. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1002/ijc.20538&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15386427&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 3. 3. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018; 359: 1350–5. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzNTkvNjM4Mi8xMzUwIjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 4. 4. Liu M, Sun Q, Wei F, Ren X. Comprehensive insights into the effects and regulatory mechanisms of immune cells expressing programmed death-1/programmed death ligand 1 in solid tumors. Cancer Biol Med. 2020; 17: 626–39. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiY2JtIjtzOjU6InJlc2lkIjtzOjg6IjE3LzMvNjI2IjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 5. 5. Baumann D, Hägele T, Mochayedi J, Drebant J, Vent C, Blobner S, et al. Proimmunogenic impact of MEK inhibition synergizes with agonist anti-CD40 immunostimulatory antibodies in tumor therapy. Nat Commun. 2020; 11: 2176. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 6. 6. Suarez-Carmona M, Williams A, Schreiber J, Hohmann N, Pruefer U, Krauss J, et al. Combined inhibition of CXCL12 and PD-1 in MSS colorectal and pancreatic cancer: modulation of the microenvironment and clinical effects. J Immunother Cancer. 2021; 9. 7. 7. Hanna BS, Llaó-Cid L, Iskar M, Roessner PM, Klett LC, Wong JKL, et al. Interleukin-10 receptor signaling promotes the maintenance of a PD-1(int) TCF-1+ CD8+ T cell population that sustains anti-tumor immunity. Immunity. 2021; 54: 2825–41.e10. 8. 8. Weisshaar N, Wu J, Ming Y, Madi A, Hotz-Wagenblatt A, Ma S, et al. Rgs16 promotes antitumor CD8+ T cell exhaustion. Sci Immunol. 2022; 7: eabh1873. 9. 9. Pfister D, Núñez NG, Pinyol R, Govaere O, Pinter M, Szydlowska M, et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature. 2021; 592: 450–6. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/s41586-021-03362-0&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 10. 10. Dudek M, Pfister D, Donakonda S, Filpe P, Schneider A, Laschinger M, et al. Auto-aggressive CXCR6+ CD8 T cells cause liver immune pathology in NASH. Nature. 2021; 592: 444–9. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/s41586-021-03233-8&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 11. 11. Yuan D, Huang S, Berger E, Liu L, Gross N, Heinzmann F, et al. Kupffer cell-derived Tnf triggers cholangiocellular tumorigenesis through JNK due to chronic mitochondrial dysfunction and ROS. Cancer Cell. 2017; 31: 771–89.e6. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.ccell.2017.05.006&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=28609656&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 12. 12. Sektioglu IM, Carretero R, Bender N, Bogdan C, Garbi N, Umansky V, et al. Macrophage-derived nitric oxide initiates T-cell diapedesis and tumor rejection. Oncoimmunology. 2016; 5: e1204506. 13. 13. Madi A, Cui G. Regulation of immune cell metabolism by cancer cell oncogenic mutations. Int J Cancer. 2020; 147: 307–16. 14. 14. Xu S, Chaudhary O, Rodríguez-Morales P, Sun X, Chen D, Zappasodi R, et al. Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8+ T cells in tumors. Immunity. 2021; 54: 1561–77.e7. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.immuni.2021.05.003&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 15. 15. Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S, et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature. 2011; 478: 197–203. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nature10491&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21976023&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000295782800040&link_type=ISI) 16. 16. Sadik A, Somarribas Patterson LF, Öztürk S, Mohapatra SR, Panitz V, Secker PF, et al. IL4I1 is a metabolic immune checkpoint that activates the AHR and promotes tumor progression. Cell. 2020; 182: 1252–70.e34. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cell.2020.07.038&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 17. 17. Bunse L, Pusch S, Bunse T, Sahm F, Sanghvi K, Friedrich M, et al. Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate. Nat Med. 2018; 24: 1192–203. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/s41591-018-0095-6&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 18. 18. Liu X, Zhao Y, Wu X, Liu Z, Liu X. A novel strategy to fuel cancer immunotherapy: targeting glucose metabolism to remodel the tumor microenvironment. Front Oncol. 2022; 12: 931104. 19. 19. Rangel Rivera GO, Knochelmann HM, Dwyer CJ, Smith AS, Wyatt MM, Rivera-Reyes AM, et al. Fundamentals of T cell metabolism and strategies to enhance cancer immunotherapy. Front Immunol. 2021; 12: 645242. 20. 20. Lim AR, Rathmell WK, Rathmell JC. The tumor microenvironment as a metabolic barrier to effector T cells and immunotherapy. Elife. 2020; 9: e55185. 21. 21. Terren I, Orrantia A, Vitalle J, Zenarruzabeitia O, Borrego F. NK cell metabolism and tumor microenvironment. Front Immunol. 2019; 10: 2278. 22. 22. Wu F, Cheng Y, Wu L, Zhang W, Zheng W, Wang Q, et al. Emerging landscapes of tumor immunity and metabolism. Front Oncol. 2020; 10: 575037. 23. 23. Butler LM, Perone Y, Dehairs J, Lupien LE, de Laat V, Talebi A, et al. Lipids and cancer: emerging roles in pathogenesis, diagnosis and therapeutic intervention. Adv Drug Deliv Rev. 2020; 159: 245–93. 24. 24. Zhang Y, Kurupati R, Liu L, Zhou XY, Zhang G, Hudaihed A, 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: 377–91.e9. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.ccell.2017.08.004&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 25. 25. Wang R, Dillon CP, Shi LZ, Milasta S, Carter R, Finkelstein D, et al. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity. 2011; 35: 871–82. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.immuni.2011.09.021&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22195744&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000298777300007&link_type=ISI) 26. 26. Chang C-H, Qiu J, O’Sullivan D, Buck MD, Noguchi T, Curtis JD, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015; 162: 1229–41. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cell.2015.08.016&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=26321679&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 27. 27. Ho P-C, Bihuniak JD, Macintyre AN, Staron M, Liu X, Amezquita R, et al. Phosphoenolpyruvate is a metabolic checkpoint of anti-tumor T cell responses. Cell. 2015; 162: 1217–28. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cell.2015.08.012&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=26321681&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 28. 28. Chowdhury PS, Chamoto K, Kumar A, Honjo T. PPAR-induced fatty acid oxidation in T cells increases the number of tumor-reactive CD8+ T cells and facilitates anti-PD-1 therapy. Cancer Immunol Res. 2018; 6: 1375–87. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FuaW1tIjtzOjU6InJlc2lkIjtzOjk6IjYvMTEvMTM3NSI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 29. 29. Saibil SD, St Paul M, Laister RC, Garcia-Batres CR, Israni-Winger K, Elford AR, et al. Activation of peroxisome proliferator-activated receptors α and δ synergizes with inflammatory signals to enhance adoptive cell therapy. Cancer Res. 2019; 79: 445–51. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjg6Ijc5LzMvNDQ1IjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 30. 30. Manzo T, Prentice BM, Anderson KG, Raman A, Schalck A, Codreanu GS, et al. Accumulation of long-chain fatty acids in the tumor microenvironment drives dysfunction in intrapancreatic CD8+ T cells. J Exp Med. 2020; 217: e20191920. 31. 31. Patsoukis N, Bardhan K, Chatterjee P, Sari D, Liu B, Bell LN, et al. PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat Commun. 2015; 6: 6692. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/ncomms7692&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25809635&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 32. 32. Zhang C, Yue C, Herrmann A, Song J, Egelston C, Wang T, et al. STAT3 activation-induced fatty acid oxidation in CD8+ T effector cells is critical for obesity-promoted breast tumor growth. Cell Metab. 2020; 31: 148–61 e5. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 33. 33. Blagih J, Coulombe F, Vincent Emma E, Dupuy F, Galicia-Vázquez G, Yurchenko E, et al. The energy sensor AMPK regulates T cell metabolic adaptation and effector responses in vivo. Immunity. 2015; 42: 41–54. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.immuni.2014.12.030&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25607458&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 34. 34. 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. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1002/eji.200838289&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=18792400&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 35. 35. 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. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE3NC84LzQ2NzAiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 36. 36. Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, et al. Cutting edge: Distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol. 2011; 186: 3299–303. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE4Ni82LzMyOTkiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 37. 37. Beier UH, Angelin A, Akimova T, Wang L, Liu Y, Xiao H, et al. Essential role of mitochondrial energy metabolism in Foxp3+ T-regulatory cell function and allograft survival. FASEB J. 2015; 29: 2315–26. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1096/fj.14-268409&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25681462&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 38. 38. Gerriets VA, Kishton RJ, Johnson MO, Cohen S, Siska PJ, Nichols AG, et al. Foxp3 and Toll-like receptor signaling balance Treg cell anabolic metabolism for suppression. Nat Immunol. 2016; 17: 1459–66. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/ni.3577&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 39. 39. Angelin A, Gil-de-Gomez L, Dahiya S, Jiao J, Guo L, Levine MH, et al. Foxp3 reprograms T cell metabolism to function in low-glucose, high-lactate environments. Cell Metab. 2017; 25: 1282–93 e7. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cmet.2016.12.018&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 40. 40. Howie D, Cobbold SP, Adams E, Ten Bokum A, Necula AS, Zhang W, et al. Foxp3 drives oxidative phosphorylation and protection from lipotoxicity. JCI Insight. 2017; 2: e89160. 41. 41. Ma X, Bi E, Lu Y, Su P, Huang C, Liu L, et al. Cholesterol induces CD8+ T cell exhaustion in the tumor microenvironment. Cell Metab. 2019; 30: 143–56 e5. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cmet.2019.04.002&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 42. 42. Wu J, Ma S, Sandhoff R, Ming Y, Hotz-Wagenblatt A, Timmerman V, et al. Loss of neurological disease HSAN-I-associated gene SPTLC2 impairs CD8+ T cell responses to infection by inhibiting T cell metabolic fitness. Immunity. 2019; 50: 1218–31.e5. 43. 43. Yang W, Bai Y, Xiong Y, Zhang J, Chen S, Zheng X, et al. Potentiating the antitumour response of CD8+ T cells by modulating cholesterol metabolism. Nature. 2016; 531: 651–5. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nature17412&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=26982734&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 44. 44. Smyth MJ, Cretney E, Kelly JM, Westwood JA, Street SE, Yagita H, et al. Activation of NK cell cytotoxicity. Mol Immunol. 2005; 42: 501–10. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.molimm.2004.07.034&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15607806&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000227142200019&link_type=ISI) 45. 45. Xue W, Zhang M. Updating targets for natural killer/T-cell lymphoma immunotherapy. Cancer Biol Med. 2021; 18: 52–62. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiY2JtIjtzOjU6InJlc2lkIjtzOjc6IjE4LzEvNTIiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 46. 46. Yaqoob P, Newsholme EA, Calder PC. Inhibition of natural killer cell activity by dietary lipids. Immunol Lett. 1994; 41: 241–7. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/0165-2478(94)90140-6&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=8002045&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=A1994PF42200029&link_type=ISI) 47. 47. Tobin LM, Mavinkurve M, Carolan E, Kinlen D, O’Brien EC, Little MA, et al. NK cells in childhood obesity are activated, metabolically stressed, and functionally deficient. JCI Insight. 2017; 2: e94939. 48. 48. Viel S, Besson L, Charrier E, Marcais A, Disse E, Bienvenu J, et al. Alteration of natural killer cell phenotype and function in obese individuals. Clin Immunol. 2017; 177: 12–7. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 49. 49. Park A, Lee Y, Kim MS, Kang YJ, Park YJ, Jung H, et al. Prostaglandin E2 secreted by thyroid cancer cells contributes to immune escape through the suppression of natural killer (NK) cell cytotoxicity and NK cell differentiation. Front Immunol. 2018; 9: 1859. 50. 50. Ma X, Holt D, Kundu N, Reader J, Goloubeva O, Take Y, et al. A prostaglandin E (PGE) receptor EP4 antagonist protects natural killer cells from PGE2-mediated immunosuppression and inhibits breast cancer metastasis. Oncoimmunology. 2013; 2: e22647. 51. 51. Kobayashi T, Lam PY, Jiang H, Bednarska K, Gloury R, Murigneux V, et al. Increased lipid metabolism impairs NK cell function and mediates adaptation to the lymphoma environment. Blood. 2020; 136: 3004–17. 52. 52. Michelet X, Dyck L, Hogan A, Loftus RM, Duquette D, Wei K, et al. Metabolic reprogramming of natural killer cells in obesity limits antitumor responses. Nat Immunol. 2018; 19: 1330–40. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/s41590-018-0251-7&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 53. 53. Assmann N, O’Brien KL, Donnelly RP, Dyck L, Zaiatz-Bittencourt V, Loftus RM, et al. Srebp-controlled glucose metabolism is essential for nk cell functional responses. Nat Immunol. 2017; 18: 1197–206. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/ni.3838&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=28920951&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 54. 54. Jensen TO, Schmidt H, Moller HJ, Hoyer M, Maniecki MB, Sjoegren P, et al. Macrophage markers in serum and tumor have prognostic impact in American Joint Committee on Cancer stage I/II melanoma. J Clin Oncol. 2009; 27: 3330–7. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIyNy8yMC8zMzMwIjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 55. 55. Kamper P, Bendix K, Hamilton-Dutoit S, Honore B, Nyengaard JR, d’Amore F. Tumor-infiltrating macrophages correlate with adverse prognosis and Epstein-Barr virus status in classical Hodgkin’s lymphoma. Haematologica. 2011; 96: 269–76. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiaGFlbWF0b2wiO3M6NToicmVzaWQiO3M6ODoiOTYvMi8yNjkiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 56. 56. Lee CH, Espinosa I, Vrijaldenhoven S, Subramanian S, Montgomery KD, Zhu S, et al. Prognostic significance of macrophage infiltration in leiomyosarcomas. Clin Cancer Res. 2008; 14: 1423–30. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6OToiMTQvNS8xNDIzIjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 57. 57. Medrek C, Ponten F, Jirstrom K, Leandersson K. The presence of tumor associated macrophages in tumor stroma as a prognostic marker for breast cancer patients. BMC Cancer. 2012; 12: 306. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1186/1471-2407-12-306&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22824040&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 58. 58. Zhang BC, Gao J, Wang J, Rao ZG, Wang BC, Gao JF. Tumor-associated macrophages infiltration is associated with peritumoral lymphangiogenesis and poor prognosis in lung adenocarcinoma. Med Oncol. 2011; 28: 1447–52. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1007/s12032-010-9638-5&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=20676804&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 59. 59. Mills CD, Kincaid K, Alt JM, Heilman MJ, Hill AM. M-1/M-2 macrophages and the Th1/Th2 paradigm. J Immunol. 2000; 164: 6166–73. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTE6IjE2NC8xMi82MTY2IjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 60. 60.Webpage biorad; macrophage polarization mini-review. 2020. 61. 61. Stein M, Keshav S, Harris N, Gordon S. Interleukin 4 potently enhances murine macrophage mannose receptor activity: a marker of alternative immunologic macrophage activation. J Exp Med. 1992; 176: 287–92. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamVtIjtzOjU6InJlc2lkIjtzOjk6IjE3Ni8xLzI4NyI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 62. 62. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 2014; 6: 13. 63. 63. Nathan CF, Murray HW, Wiebe ME, Rubin BY. Identification of interferon-gamma as the lymphokine that activates human macrophage oxidative metabolism and antimicrobial activity. J Exp Med. 1983; 158: 670–89. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamVtIjtzOjU6InJlc2lkIjtzOjk6IjE1OC8zLzY3MCI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 64. 64. Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. 2014; 513: 559–63. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nature13490&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=25043024&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000342075800043&link_type=ISI) 65. 65. Su P, Wang Q, Bi E, Ma X, Liu L, Yang M, et al. Enhanced lipid accumulation and metabolism are required for the differentiation and activation of tumor-associated macrophages. Cancer Res. 2020; 80: 1438–50. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjk6IjgwLzcvMTQzOCI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 66. 66. Wang R, Lu M, Zhang J, Chen S, Luo X, Qin Y, et al. Increased IL-10 mRNA expression in tumor-associated macrophage correlated with late stage of lung cancer. J Exp Clin Cancer Res. 2011; 30: 62. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1186/1756-9966-30-62&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=21595995&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 67. 67. Sica A, Saccani A, Bottazzi B, Polentarutti N, Vecchi A, van Damme J, et al. Autocrine production of IL-10 mediates defective IL-12 production and NF-kappa B activation in tumor-associated macrophages. J Immunol. 2000; 164: 762–7. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6OToiMTY0LzIvNzYyIjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 68. 68. Li L, Yang L, Wang L, Wang F, Zhang Z, Li J, et al. Impaired T cell function in malignant pleural effusion is caused by TGF-β derived predominantly from macrophages. Int J Cancer. 2016; 139: 2261–9. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 69. 69. Peng J, Tsang JY, Li D, Niu N, Ho DH, Lau KF, et al. Inhibition of TGF-β signaling in combination with TLR7 ligation re-programs a tumoricidal phenotype in tumor-associated macrophages. Cancer Lett. 2013; 331: 239–49. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.canlet.2013.01.001&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=23318200&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 70. 70. Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004; 10: 942–9. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nm1093&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15322536&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000223658400031&link_type=ISI) 71. 71. Lin EY, Li JF, Bricard G, Wang W, Deng Y, Sellers R, et al. Vascular endothelial growth factor restores delayed tumor progression in tumors depleted of macrophages. Mol Oncol. 2007; 1: 288–302. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.molonc.2007.10.003&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=18509509&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 72. 72. Lin EY, Li JF, Gnatovskiy L, Deng Y, Zhu L, Grzesik DA, et al. Macrophages regulate the angiogenic switch in a mouse model of breast cancer. Cancer Res. 2006; 66: 11238–46. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjExOiI2Ni8yMy8xMTIzOCI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 73. 73. Yeo EJ, Cassetta L, Qian BZ, Lewkowich I, Li JF, Stefater JA 3rd, et al. Myeloid WNT7b mediates the angiogenic switch and metastasis in breast cancer. Cancer Res. 2014; 74: 2962–73. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI3NC8xMS8yOTYyIjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 74. 74. Gocheva V, Wang HW, Gadea BB, Shree T, Hunter KE, Garfall AL, et al. IL-4 induces cathepsin protease activity in tumor-associated macrophages to promote cancer growth and invasion. Genes Dev. 2010; 24: 241–55. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiZ2VuZXNkZXYiO3M6NToicmVzaWQiO3M6ODoiMjQvMy8yNDEiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 75. 75. Vasiljeva O, Papazoglou A, Kruger A, Brodoefel H, Korovin M, Deussing J, et al. Tumor cell-derived and macrophage-derived cathepsin B promotes progression and lung metastasis of mammary cancer. Cancer Res. 2006; 66: 5242–50. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI2Ni8xMC81MjQyIjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 76. 76. Sabeh F, Ota I, Holmbeck K, Birkedal-Hansen H, Soloway P, Balbin M, et al. Tumor cell traffic through the extracellular matrix is controlled by the membrane-anchored collagenase MT1-MMP. J Cell Biol. 2004; 167: 769–81. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNiIjtzOjU6InJlc2lkIjtzOjk6IjE2Ny80Lzc2OSI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 77. 77. Wolf K, Wu YI, Liu Y, Geiger J, Tam E, Overall C, et al. Multi-step pericellular proteolysis controls the transition from individual to collective cancer cell invasion. Nat Cell Biol. 2007; 9: 893–904. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/ncb1616&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=17618273&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000248486700011&link_type=ISI) 78. 78. Feng J, Han J, Pearce SF, Silverstein RL, Gotto AM Jr., Hajjar DP, et al. Induction of CD36 expression by oxidized LDL and IL-4 by a common signaling pathway dependent on protein kinase C and PPAR-gamma. J Lipid Res. 2000; 41: 688–96. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamxyIjtzOjU6InJlc2lkIjtzOjg6IjQxLzUvNjg4IjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 79. 79. Rios FJ, Koga MM, Pecenin M, Ferracini M, Gidlund M, Jancar S. Oxidized LDL induces alternative macrophage phenotype through activation of CD36 and PAFR. Mediators Inflamm. 2013; 2013: 198193. 80. 80. Nagy L, Tontonoz P, Alvarez JG, Chen H, Evans RM. Oxidized LDL regulates macrophage gene expression through ligand activation of PPARgamma. Cell. 1998; 93: 229–40. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/S0092-8674(00)81574-3&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=9568715&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000073174000009&link_type=ISI) 81. 81. Masetti M, Carriero R, Portale F, Marelli G, Morina N, Pandini M, et al. Lipid-loaded tumor-associated macrophages sustain tumor growth and invasiveness in prostate cancer. J Exp Med. 2022; 219: e20210564. 82. 82. Divakaruni AS, Hsieh WY, Minarrieta L, Duong TN, Kim KKO, Desousa BR, et al. Etomoxir inhibits macrophage polarization by disrupting CoA homeostasis. Cell Metabolism. 2018; 28: 490–503.e7. 83. 83. Wu H, Han Y, Rodriguez Sillke Y, Deng H, Siddiqui S, Treese C, et al. Lipid droplet-dependent fatty acid metabolism controls the immune suppressive phenotype of tumor-associated macrophages. EMBO Mol Med. 2019; 11: e10698. 84. 84. Oishi Y, Spann NJ, Link VM, Muse ED, Strid T, Edillor C, et al. SREBP1 contributes to resolution of pro-inflammatory TLR4 signaling by reprogramming fatty acid metabolism. Cell Metab. 2017; 25: 412–27. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cmet.2016.11.009&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=28041958&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 85. 85. Ramirez CM, Torrecilla-Parra M, Pardo-Marques V, de-Frutos MF, Perez-Garcia A, Tabraue C, et al. Crosstalk between LXR and caveolin-1 signaling supports cholesterol efflux and anti-inflammatory pathways in macrophages. Front Endocrinol (Lausanne) 2021; 12: 635923. 86. 86. Goossens P, Rodriguez-Vita J, Etzerodt A, Masse M, Rastoin O, Gouirand V, et al. Membrane cholesterol efflux drives tumor-associated macrophage reprogramming and tumor progression. Cell Metab. 2019; 29: 1376–89 e4. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cmet.2019.02.016&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 87. 87. James BR, Tomanek-Chalkley A, Askeland EJ, Kucaba T, Griffith TS, Norian LA. Diet-induced obesity alters dendritic cell function in the presence and absence of tumor growth. J Immunol. 2012; 189: 1311–21. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE4OS8zLzEzMTEiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 88. 88. Weatherill AR, Lee JY, Zhao L, Lemay DG, Youn HS, Hwang DH. Saturated and polyunsaturated fatty acids reciprocally modulate dendritic cell functions mediated through TLR4. J Immunol 2005; 174: 5390–7. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE3NC85LzUzOTAiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 89. 89. Zeyda M, Saemann MD, Stuhlmeier KM, Mascher DG, Nowotny PN, Zlabinger GJ, et al. Polyunsaturated fatty acids block dendritic cell activation and function independently of NF-kappaB activation. J Biol Chem. 2005; 280: 14293–301. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamJjIjtzOjU6InJlc2lkIjtzOjEyOiIyODAvMTQvMTQyOTMiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 90. 90. Herber DL, Cao W, Nefedova Y, Novitskiy SV, Nagaraj S, Tyurin VA, et al. Lipid accumulation and dendritic cell dysfunction in cancer. Nat Med. 2010; 16: 880–6. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nm.2172&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=20622859&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 91. 91. Yi H, Yu X, Gao P, Wang Y, Baek SH, Chen X, et al. Pattern recognition scavenger receptor SRA/CD204 down-regulates toll-like receptor 4 signaling-dependent CD8 T-cell activation. Blood. 2009; 113: 5819–28. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImJsb29kam91cm5hbCI7czo1OiJyZXNpZCI7czoxMToiMTEzLzIzLzU4MTkiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 92. 92. Ramakrishnan R, Tyurin VA, Veglia F, Condamine T, Amoscato A, Mohammadyani D, et al. Oxidized lipids block antigen cross-presentation by dendritic cells in cancer. J Immunol. 2014; 192: 2920–31. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE5Mi82LzI5MjAiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 93. 93. Cubillos-Ruiz JR, Silberman PC, Rutkowski MR, Chopra S, Perales-Puchalt A, Song M, et al. ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell. 2015; 161: 1527–38. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cell.2015.05.025&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=26073941&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 94. 94. Veglia F, Tyurin VA, Mohammadyani D, Blasi M, Duperret EK, Donthireddy L, et al. Lipid bodies containing oxidatively truncated lipids block antigen cross-presentation by dendritic cells in cancer. Nat Commun. 2017; 8: 2122. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/s41467-017-02186-9&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=29242535&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 95. 95. Wculek SK, Malanchi I. Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature. 2015; 528: 413–7. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nature16140&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=26649828&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 96. 96. Zhang Y, Guoqiang L, Sun M, Lu X. Targeting and exploitation of tumor-associated neutrophils to enhance immunotherapy and drug delivery for cancer treatment. Cancer Biol Med. 2020; 17: 32–43. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiY2JtIjtzOjU6InJlc2lkIjtzOjc6IjE3LzEvMzIiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 97. 97. Li P, Lu M, Shi J, Gong Z, Hua L, Li Q, et al. Lung mesenchymal cells elicit lipid storage in neutrophils that fuel breast cancer lung metastasis. Nat Immunol. 2020; 21: 1444–55. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/s41590-020-0783-5&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 98. 98. Raccosta L, Fontana R, Maggioni D, Lanterna C, Villablanca EJ, Paniccia A, et al. The oxysterol-CXCR2 axis plays a key role in the recruitment of tumor-promoting neutrophils. J Exp Med. 2013; 210: 1711–28. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamVtIjtzOjU6InJlc2lkIjtzOjEwOiIyMTAvOS8xNzExIjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 99. 99. Chuang JC, Yu CL, Wang SR. Modulation of human lymphocyte proliferation by amino acids. Clin Exp Immunol. 1990; 81: 173–6. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=2379319&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 100.100. Hope HC, Salmond RJ. The role of non-essential amino acids in T cell function and anti-tumour immunity. Arch Immunol Ther Exp (Warsz). 2021; 69: 29. 101.101. Han C, Ge M, Ho PC, Zhang L. Fueling T-cell antitumor immunity: amino acid metabolism revisited. Cancer Immunol Res. 2021; 9: 1373–82. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FuaW1tIjtzOjU6InJlc2lkIjtzOjk6IjkvMTIvMTM3MyI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 102.102. Sullivan MR, Danai LV, Lewis CA, Chan SH, Gui DY, Kunchok T, et al. Quantification of microenvironmental metabolites in murine cancers reveals determinants of tumor nutrient availability. Elife. 2019; 8: e44235. 103.103. Grzywa TM, Sosnowska A, Matryba P, Rydzynska Z, Jasinski M, Nowis D, et al. Myeloid cell-derived arginase in cancer immune response. Front Immunol. 2020; 11: 938. 104.104. Morris SM, Jr.. Arginine metabolism revisited. J Nutr. 2016; 146: 2579S–86S. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToibnV0cml0aW9uIjtzOjU6InJlc2lkIjtzOjEyOiIxNDYvMTIvMjU3OVMiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 105.105. Chantranupong L, Scaria SM, Saxton RA, Gygi MP, Shen K, Wyant GA, et al. The CASTOR proteins are arginine sensors for the mTORC1 pathway. Cell. 2016; 165: 153–64. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cell.2016.02.035&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=26972053&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 106.106. Morris CR, Hamilton-Reeves J, Martindale RG, Sarav M, Ochoa Gautier JB. Acquired amino acid deficiencies: a focus on arginine and glutamine. Nutr Clin Pract. 2017; 32: 30s–47s. 107.107. Husson A, Brasse-Lagnel C, Fairand A, Renouf S, Lavoinne A. Argininosuccinate synthetase from the urea cycle to the citrulline-NO cycle. Eur J Biochem. 2003; 270: 1887–99. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1046/j.1432-1033.2003.03559.x&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=12709047&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000182449700001&link_type=ISI) 108.108. Böger RH. The pharmacodynamics of L-arginine. J Nutr. 2007; 137: 1650S–5S. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToibnV0cml0aW9uIjtzOjU6InJlc2lkIjtzOjExOiIxMzcvNi8xNjUwUyI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 109.109. Lüneburg N, Xanthakis V, Schwedhelm E, Sullivan LM, Maas R, Anderssohn M, et al. Reference intervals for plasma L-arginine and the L-arginine: asymmetric dimethylarginine ratio in the framingham offspring cohort. J Nutr. 2011; 141: 2186–90. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToibnV0cml0aW9uIjtzOjU6InJlc2lkIjtzOjExOiIxNDEvMTIvMjE4NiI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 110.110. Steggerda SM, Bennett MK, Chen J, Emberley E, Huang T, Janes JR, et al. Inhibition of arginase by CB-1158 blocks myeloid cell-mediated immune suppression in the tumor microenvironment. J Immunother Cancer. 2017; 5: 101. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiaml0YyI7czo1OiJyZXNpZCI7czo3OiI1LzEvMTAxIjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 111.111. Mussai F, Wheat R, Sarrou E, Booth S, Stavrou V, Fultang L, et al. Targeting the arginine metabolic brake enhances immunotherapy for leukaemia. Int J Cancer. 2019; 145: 2201–8. 112.112. Mussai F, Egan S, Higginbotham-Jones J, Perry T, Beggs A, Odintsova E, et al. Arginine dependence of acute myeloid leukemia blast proliferation: a novel therapeutic target. Blood. 2015; 125: 2386–96. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImJsb29kam91cm5hbCI7czo1OiJyZXNpZCI7czoxMToiMTI1LzE1LzIzODYiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 113.113. Pan M, Reid MA, Lowman XH, Kulkarni RP, Tran TQ, Liu X, et al. Regional glutamine deficiency in tumours promotes dedifferentiation through inhibition of histone demethylation. Nat Cell Biol. 2016; 18: 1090–101. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/ncb3410&link_type=DOI) 114.114. Mussai F, De Santo C, Abu-Dayyeh I, Booth S, Quek L, McEwen-Smith RM, et al. Acute myeloid leukemia creates an arginase-dependent immunosuppressive microenvironment. Blood. 2013; 122: 749–58. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImJsb29kam91cm5hbCI7czo1OiJyZXNpZCI7czo5OiIxMjIvNS83NDkiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 115.115. Liu Y, Van Ginderachter JA, Brys L, De Baetselier P, Raes G, Geldhof AB. Nitric oxide-independent CTL suppression during tumor progression: association with arginase-producing (M2) myeloid cells. J Immunol. 2003; 170: 5064–74. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTE6IjE3MC8xMC81MDY0IjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 116.116. Gray MJ, Poljakovic M, Kepka-Lenhart D, Morris SM. Induction of arginase I transcription by IL-4 requires a composite DNA response element for STAT6 and C/EBPβ. Gene. 2005; 353: 98–106. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.gene.2005.04.004&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15922518&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000230249500011&link_type=ISI) 117.117. Cobbold SP, Adams E, Farquhar CA, Nolan KF, Howie D, Lui KO, et al. Infectious tolerance via the consumption of essential amino acids and mTOR signaling. Proc Natl Acad Sci U S A. 2009; 106: 12055–60. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTA2LzI5LzEyMDU1IjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 118.118. Doedens AL, Stockmann C, Rubinstein MP, Liao D, Zhang N, DeNardo DG, et al. Macrophage expression of hypoxia-inducible factor-1α suppresses T-cell function and promotes tumor progression. Cancer Res. 2010; 70: 7465–75. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI3MC8xOS83NDY1IjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 119.119. Rodriguez PC, Zea AH, DeSalvo J, Culotta KS, Zabaleta J, Quiceno DG, et al. L-arginine consumption by macrophages modulates the expression of CD3 zeta chain in T lymphocytes. J Immunol. 2003; 171: 1232–9. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE3MS8zLzEyMzIiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 120.120. Rodriguez PC, Quiceno DG, Zabaleta J, Ortiz B, Zea AH, Piazuelo MB, et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004; 64: 5839–49. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI2NC8xNi81ODM5IjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 121.121. Rodriguez PC, Ernstoff MS, Hernandez C, Atkins M, Zabaleta J, Sierra R, et al. Arginase I–producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res. 2009; 69: 1553–60. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjk6IjY5LzQvMTU1MyI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 122.122. Crump NT, Hadjinicolaou AV, Xia M, Walsby-Tickle J, Gileadi U, Chen J-L, et al. Chromatin accessibility governs the differential response of cancer and T cells to arginine starvation. Cell Rep. 2021; 35: 109101. 123.123. Zea AH, Rodriguez PC, Culotta KS, Hernandez CP, DeSalvo J, Ochoa JB, et al. L-arginine modulates CD3zeta expression and T cell function in activated human T lymphocytes. Cell Immunol. 2004; 232: 21–31. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cellimm.2005.01.004&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15922712&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000229764300003&link_type=ISI) 124.124. Munder M. Suppression of T-cell functions by human granulocyte arginase. Blood. 2006; 108: 1627–34. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImJsb29kam91cm5hbCI7czo1OiJyZXNpZCI7czoxMDoiMTA4LzUvMTYyNyI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 125.125. Fletcher M, Ramirez ME, Sierra RA, Raber P, Thevenot P, Al-Khami AA, et al. L-arginine depletion blunts antitumor T-cell responses by inducing myeloid-derived suppressor cells. Cancer Res. 2015; 75: 275–83. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjg6Ijc1LzIvMjc1IjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 126.126. Werner A, Koschke M, Leuchtner N, Luckner-Minden C, Habermeier A, Rupp J, et al. Reconstitution of T cell proliferation under arginine limitation: activated human T cells take up citrulline via l-type amino acid transporter 1 and use it to regenerate arginine after induction of argininosuccinate synthase expression. Front Immunol. 2017; 8: 864. 127.127. Werner A, Amann E, Schnitzius V, Habermeier A, Luckner-Minden C, Leuchtner N, et al. Induced arginine transport via cationic amino acid transporter-1 is necessary for human T-cell proliferation. Eur J Immunol. 2016; 46: 92–103. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1002/eji.201546047&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=26449889&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 128.128. Geiger R, Rieckmann JC, Wolf T, Basso C, Feng Y, Fuhrer T, et al. L-arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell. 2016; 167: 829–42.e13. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cell.2016.09.031&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=27745970&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 129.129. Rodriguez PC, Quiceno DG, Ochoa AC. L-arginine availability regulates T-lymphocyte cell-cycle progression. Blood. 2007; 109: 1568–73. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImJsb29kam91cm5hbCI7czo1OiJyZXNpZCI7czoxMDoiMTA5LzQvMTU2OCI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 130.130. Baniyash M. TCR ζ-chain downregulation: curtailing an excessive inflammatory immune response. Nat Rev Immunol. 2004; 4: 675–87. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nri1434&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15343367&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000223657900012&link_type=ISI) 131.131. Huppa JB, Davis MM. T-cell-antigen recognition and the immunological synapse. Nat Rev Immunol. 2003; 3: 973–83. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/nri1245&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=14647479&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000186892700015&link_type=ISI) 132.132. Eibert SM, Lee K-H, Pipkorn R, Sester U, Wabnitz GH, Giese T, et al. Cofilin peptide homologs interfere with immunological synapse formation and T cell activation. Proc Natl Acad Sci. 2004; 101: 1957–62. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiMTAxLzcvMTk1NyI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 133.133. Feldmeyer N, Wabnitz G, Leicht S, Luckner-Minden C, Schiller M, Franz T, et al. Arginine deficiency leads to impaired cofilin dephosphorylation in activated human T lymphocytes. Int Immunol. 2012; 24: 303–13. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1093/intimm/dxs004&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22345165&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 134.134. Zhang J, Xu X, Shi M, Chen Y, Yu D, Zhao C, et al. CD13(hi) neutrophil-like myeloid-derived suppressor cells exert immune suppression through arginase 1 expression in pancreatic ductal adenocarcinoma. Oncoimmunology. 2017; 6: e1258504. 135.135. Mao F-Y, Zhao Y-l, Lv Y-P, Teng Y-S, Kong H, Liu Y-G, et al. CD45+CD33lowCD11bdim myeloid-derived suppressor cells suppress CD8+ T cell activity via the IL-6/IL-8-arginase I axis in human gastric cancer. Cell Death Dis. 2018; 9: 763. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 136.136. Vonwirth V, Bulbul Y, Werner A, Echchannaoui H, Windschmitt J, Habermeier A, et al. Inhibition of arginase 1 liberates potent T cell immunostimulatory activity of human neutrophil granulocytes. Front Immunol. 2020; 11: 617699. 137.137. Lamas B, Vergnaud-Gauduchon J, Goncalves-Mendes N, Perche O, Rossary A, Vasson MP, et al. Altered functions of natural killer cells in response to L-arginine availability. Cell Immunol. 2012; 280: 182–90. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.cellimm.2012.11.018&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=23399839&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 138.138. Oberlies J, Watzl C, Giese T, Luckner C, Kropf P, Muller I, et al. Regulation of NK cell function by human granulocyte arginase. J Immunol. 2009; 182: 5259–67. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE4Mi85LzUyNTkiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 139.139. Westhaver LP, Nersesian S, Nelson A, MacLean LK, Carter EB, Rowter D, et al. Mitochondrial damage-associated molecular patterns trigger arginase-dependent lymphocyte immunoregulation. Cell Rep. 2022; 39: 110847. 140.140. Jain A, Bakhshi S, Thakkar H, Gerards M, Singh A. Elevated mitochondrial DNA copy numbers in pediatric acute lymphoblastic leukemia: a potential biomarker for predicting inferior survival. Pediatr Blood Cancer. 2018; 65: e26874. 141.141. Meng X, Schwarzenbach H, Yang Y, Müller V, Li N, Tian D, et al. Circulating mitochondrial DNA is linked to progression and prognosis of epithelial ovarian cancer. Transl Oncol. 2019; 12: 1213–20. 142.142. Comai S, Bertazzo A, Brughera M, Crotti S. Tryptophan in health and disease. Adv Clin Chem. 2020; 95: 165–218. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 143.143. Liu X, Shin N, Koblish HK, Yang G, Wang Q, Wang K, et al. Selective inhibition of IDO1 effectively regulates mediators of antitumor immunity. Blood. 2010; 115: 3520–30. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImJsb29kam91cm5hbCI7czo1OiJyZXNpZCI7czoxMToiMTE1LzE3LzM1MjAiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 144.144. Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A, Mellor AL. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J Exp Med. 1999; 189: 1363–72. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamVtIjtzOjU6InJlc2lkIjtzOjEwOiIxODkvOS8xMzYzIjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 145.145. Platten M, Nollen EAA, Rohrig UF, Fallarino F, Opitz CA. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat Rev Drug Discov. 2019; 18: 379–401. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/s41573-019-0016-5&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 146.146. Theate I, van Baren N, Pilotte L, Moulin P, Larrieu P, Renauld JC, et al. Extensive profiling of the expression of the indoleamine 2,3-dioxygenase 1 protein in normal and tumoral human tissues. Cancer Immunol Res. 2015; 3: 161–72. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FuaW1tIjtzOjU6InJlc2lkIjtzOjc6IjMvMi8xNjEiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 147.147. Frumento G, Rotondo R, Tonetti M, Damonte G, Benatti U, Ferrara GB. Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase. J Exp Med. 2002; 196: 459–68. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamVtIjtzOjU6InJlc2lkIjtzOjk6IjE5Ni80LzQ1OSI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 148.148. Peyraud F, Guegan J-P, Bodet D, Cousin S, Bessede A, Italiano A. Targeting tryptophan catabolism in cancer immunotherapy era: challenges and perspectives. Front Immunol. 2022; 13: 807271. 149.149. Puccetti P, Fallarino F, Italiano A, Soubeyran I, MacGrogan G, Debled M, et al. Accumulation of an endogenous tryptophan-derived metabolite in colorectal and breast cancers. PLoS One. 2015; 10: e0122046. 150.150. Fallarino F, Grohmann U, You S, McGrath BC, Cavener DR, Vacca C, et al. The combined effects of tryptophan starvation and tryptophan catabolites down-regulate T cell receptor zeta-chain and induce a regulatory phenotype in naive T cells. J Immunol. 2006; 176: 6752–61. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTE6IjE3Ni8xMS82NzUyIjtzOjQ6ImF0b20iO3M6NTM6Ii9jYm0vZWFybHkvMjAyMi8xMC8yNC9qLmlzc24uMjA5NS0zOTQxLjIwMjIuMDM4MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 151.151. Fallarino F, Grohmann U, Hwang KW, Orabona C, Vacca C, Bianchi R, et al. Modulation of tryptophan catabolism by regulatory T cells. Nat Immunol. 2003; 4: 1206–12. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1038/ni1003&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=14578884&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000186769500014&link_type=ISI) 152.152. Costa-Mattioli M, Walter P. The integrated stress response: from mechanism to disease. Science. 2020; 368: eaat5314. 153.153. Munn DH, Sharma MD, Baban B, Harding HP, Zhang Y, Ron D, et al. Gcn2 kinase in t cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity. 2005; 22: 633–42. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.immuni.2005.03.013&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=15894280&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000229447200012&link_type=ISI) 154.154. Sonner JK, Deumelandt K, Ott M, Thome CM, Rauschenbach KJ, Schulz S, et al. The stress kinase GCN2 does not mediate suppression of antitumor T cell responses by tryptophan catabolism in experimental melanomas. Oncoimmunology. 2016; 5: e1240858. 155.155. Chen W, Liang X, Peterson AJ, Munn DH, Blazar BR. The indoleamine 2,3-dioxygenase pathway is essential for human plasmacytoid dendritic cell-induced adaptive T regulatory cell generation. J Immunol. 2008; 181: 5396–404. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE4MS84LzUzOTYiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 156.156. Curti A, Pandolfi S, Valzasina B, Aluigi M, Isidori A, Ferri E, et al. Modulation of tryptophan catabolism by human leukemic cells results in the conversion of CD25- into CD25+ T regulatory cells. Blood. 2007; 109: 2871–7. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImJsb29kam91cm5hbCI7czo1OiJyZXNpZCI7czoxMDoiMTA5LzcvMjg3MSI7czo0OiJhdG9tIjtzOjUzOiIvY2JtL2Vhcmx5LzIwMjIvMTAvMjQvai5pc3NuLjIwOTUtMzk0MS4yMDIyLjAzODEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 157.157. Mezrich JD, Fechner JH, Zhang X, Johnson BP, Burlingham WJ, Bradfield CA. An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J Immunol. 2010; 185: 3190–8. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE4NS82LzMxOTAiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 158.158. Stockinger B, Di Meglio P, Gialitakis M, Duarte JH. The aryl hydrocarbon receptor: multitasking in the immune system. Annu Rev Immunol. 2014; 32: 403–32. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1146/annurev-immunol-032713-120245&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=24655296&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000336427400013&link_type=ISI) 159.159. Liu Y, Liang X, Dong W, Fang Y, Lv J, Zhang T, et al. Tumor-repopulating cells induce PD-1 expression in CD8+ T cells by transferring kynurenine and AhR activation. Cancer Cell. 2018; 33: 480–94.e7. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1016/j.ccell.2018.02.005&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) 160.160. Amobi-McCloud A, Muthuswamy R, Battaglia S, Yu H, Liu T, Wang J, et al. IDO1 expression in ovarian cancer induces PD-1 in T cells via aryl hydrocarbon receptor activation. Front Immunol. 2021; 12: 678999. 161.161. Novikov O, Wang Z, Stanford EA, Parks AJ, Ramirez-Cardenas A, Landesman E, et al. An aryl hydrocarbon receptor-mediated amplification loop that enforces cell migration in ER-/PR-/Her2- human breast cancer cells. Mol Pharmacol. 2016; 90: 674–88. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoibW9scGhhcm0iO3M6NToicmVzaWQiO3M6ODoiOTAvNS82NzQiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 162.162. DiNatale BC, Murray IA, Schroeder JC, Flaveny CA, Lahoti TS, Laurenzana EM, et al. Kynurenic acid is a potent endogenous aryl hydrocarbon receptor ligand that synergistically induces interleukin-6 in the presence of inflammatory signaling. Toxicol Sci. 2010; 115: 89–97. [CrossRef](http://www.cancerbiomed.org/lookup/external-ref?access_num=10.1093/toxsci/kfq024&link_type=DOI) [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=20106948&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom) [Web of Science](http://www.cancerbiomed.org/lookup/external-ref?access_num=000276742200009&link_type=ISI) 163.163. Caforio M, Sorino C, Caruana I, Weber G, Camera A, Cifaldi L, et al. GD2 redirected CAR T and activated NK-cell-mediated secretion of ifngamma overcomes MYCN-dependent IDO1 inhibition, contributing to neuroblastoma cell immune escape. J Immunother Cancer. 2021; 9: e001502. 164.164. Della Chiesa M, Carlomagno S, Frumento G, Balsamo M, Cantoni C, Conte R, et al. The tryptophan catabolite L-kynurenine inhibits the surface expression of NKP46- and NKG2D-activating receptors and regulates NK-cell function. Blood. 2006; 108: 4118–25. [Abstract/FREE Full Text](http://www.cancerbiomed.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImJsb29kam91cm5hbCI7czo1OiJyZXNpZCI7czoxMToiMTA4LzEzLzQxMTgiO3M6NDoiYXRvbSI7czo1MzoiL2NibS9lYXJseS8yMDIyLzEwLzI0L2ouaXNzbi4yMDk1LTM5NDEuMjAyMi4wMzgxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 165.165. Wang D, Saga Y, Mizukami H, Sato N, Nonaka H, Fujiwara H, et al. Indoleamine-2,3-dioxygenase, an immunosuppressive enzyme that inhibits natural killer cell function, as a useful target for ovarian cancer therapy. Int J Oncol. 2012; 40: 929–34. [PubMed](http://www.cancerbiomed.org/lookup/external-ref?access_num=22179492&link_type=MED&atom=%2Fcbm%2Fearly%2F2022%2F10%2F24%2Fj.issn.2095-3941.2022.0381.atom)