Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Oncology Letters
Join Editorial Board Propose a Special Issue
Print ISSN: 1792-1074 Online ISSN: 1792-1082
Journal Cover
October-2025 Volume 30 Issue 4

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
October-2025 Volume 30 Issue 4

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Mechanisms of T‑cell metabolic reprogramming in the microenvironment of acute myeloid leukemia and its therapeutic potential (Review)

  • Authors:
    • Yanhong Luo
    • Jie Luo
    • Min Yang
    • Xueya Zhao
  • View Affiliations / Copyright

    Affiliations: Department of Hematology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China, Bishan Hospital of Chongqing Medical University, Chongqing 402760, P.R. China, Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, P.R. China
    Copyright: © Luo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 455
    |
    Published online on: July 22, 2025
       https://doi.org/10.3892/ol.2025.15201
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Acute myeloid leukemia (AML) is an aggressive hematological malignancy that is often resistant to conventional therapies. The present narrative review discusses on the role of T cell metabolic reprogramming in the AML tumor microenvironment (TME), which markedly impacts the effectiveness of immunotherapy. The TME of AML, influenced by factors such as high lactic acid (LA) levels, hypoxia and nutrient competition, hampers T cell functions such as glycolysis, lipid metabolism and amino acid metabolism, leading to impaired T cell proliferation and antitumor response. Metabolic waste products, including LA and adenosine, further contribute to the immunosuppressive environment. T cell exhaustion, induced by nutrient deprivation and metabolic dysregulation, serves a key role in the failure of immune responses. Moreover, strategies to modulate T cell metabolism, such as targeting glycolysis and fatty acid oxidation, show promise in enhancing immunotherapy outcomes. The current review also highlights emerging technologies, such as single‑cell metabolomics and CRISPR screening, which are critical for identifying metabolic targets and advancing personalized therapies. Despite challenges in translating these findings to clinical settings, understanding T cell metabolism in the AML TME offers new therapeutic avenues for improving patient outcomes.

Introduction

Acute myeloid leukemia (AML) is an aggressive cancer marked by the rapid growth of immature myeloid cells, which disrupts normal blood cell production (1). Despite advancements in treatments such as chemotherapy and stem cell transplantation (2), the lack of understanding of the tumor microenvironment (TME) has hindered the effectiveness of immunotherapies. The TME includes tumor cells, immune cells, fibroblasts, blood vessels and cytokines, all interacting to influence tumor progression. Immunotherapy aims to boost antitumor responses whilst reducing immunosuppressive effects (3). In leukemia, the bone marrow is the primary site for leukemia stem cells, with secondary lymphoid organs also part of the TME. Current therapies such as checkpoint inhibitors combined with chemotherapy and hypomethylating agents, show promise. Further research into the immune microenvironment of AML is essential to develop more effective immunotherapies (4).

Unlike in solid tumors, the AML TME is primarily in the bone marrow, with unique anatomical, cellular and metabolic features. It includes hematopoietic stem/progenitor, stromal, endothelial and immune regulatory cells, but lacks tumor-associated fibroblasts and persistent antigenic stimulation typical of solid tumors (5). The AML TME is highly immunosuppressive, marked by increased regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs) and exhausted T cells, which limit immune responses (6). Leukemia cells remodel the TME by altering C-X-C motif chemokine ligand 12 (CXCL12) expression in stromal cells, suppressing normal hematopoiesis and promoting their survival (7). The metabolic profile, hypoxia and immune composition of AML differ from solid tumors, further reducing immunotherapy efficacy (8). Recent studies have highlighted that AML TME characteristics are associated with disease progression and serve as a key factor for patient risk stratification (9,10). Therefore, understanding AML-specific TME structure and function is crucial for optimizing immunotherapy strategies.

Furthermore, T cells are crucial in antitumor immunity, especially CD8+ cytotoxic T lymphocytes (CTLs), which kill tumor cells via granzyme B, perforin and interferon-γ (IFN-γ), improving prognosis (6). Helper T cells (Th) 1, 2 and 17 also serve roles: Th1 enhances CTL activity through IFN-γ and IL-2; Th2 recruits eosinophils and macrophages; and Th17 can have pro or antitumor effects (11). Conversely, Tregs suppress effector T cells (Teffs), often associated with poor prognosis (12). In numerous cancers, Tregs are more abundant in tumor tissues than in adjacent healthy tissues. Their presence in the TME inhibits effector T cell function, promoting disease progression and poor outcomes, as observed in colorectal cancer (11).

Cells require nutrients for normal function, and immune cells rely on nutrient uptake to regulate their activities (13). T cell activation drives metabolic shifts, boosting glycolysis to meet energy needs for proliferation. Unlike cancer cells, which experience dysregulated metabolism, T cells respond normally to these changes (14). Lipid and amino acid metabolism are also crucial; lipid biosynthesis affects mTOR, a key regulator of metabolic reprogramming (15), whilst amino acid availability, particularly L-arginine, influences T cell survival and adaptability (16). In the TME, tumor cells compete with T cells for essential nutrients such as glucose, glutamine and arginine, impairing T cell function and promoting tumor progression. Through the Warburg effect, tumor cells outcompete T cells for glucose, whilst programmed death-1 (PD-1) expression further inhibits glycolysis. Fatty acid accumulation disrupts mitochondrial function, driving T cell exhaustion. Tumor cells also deplete arginine and glutamine, reducing their availability for T cells. Other immune cells, such as dendritic cells, MDSCs and tumor-associated macrophages, overexpress enzymes such as arginase and indoleamine 2,3-dioxygenase (IDO), depleting essential amino acids and altering T cell activation and differentiation. The accumulation of immunosuppressive Tregs further exacerbates T cell exhaustion (17).

The AML TME is marked by high glucose metabolism and lactic acid (LA) accumulation, creating an acidic environment that suppresses T cell function (18). Neutralizing acidity with sodium bicarbonate (NaBi) has been reported to enhance CD8+ T cell immunotherapy efficacy (19). The fms-related tyrosine kinase 3-internal tandem duplication mutation increases glycolytic activity, contributing to chemotherapy resistance and impaired T cell function (20). Elevated glucose metabolism in AML cells further drives chemoresistance. Excessive glucose consumption and high LA levels in AML cells inhibit T cell function, weakening antitumor immunity (21,22). Lipid metabolism also serves a key role in tumor progression (23), immune evasion and drug resistance (24). Dysregulated lipid metabolism alters the balance between Tregs and Teffs in the TME (13).

Additionally, recent studies have highlighted the critical role of T-cell metabolic reprogramming in the AML TME. The abnormal metabolic features of the AML microenvironment, such as high LA levels, hypoxia and nutrient competition, directly suppress T cell effector functions and drive exhaustion by altering metabolic pathways such as glycolysis and fatty acid oxidation (FAO) (25–27). These factors notably contribute to immunotherapy failure. The present review assesses the molecular mechanisms of T cell metabolic reprogramming in the AML TME, focusing on the way metabolic imbalances impair T cell functionality. It also explores potential strategies for combining metabolic interventions with immunotherapy. By synthesizing current research, the current review aims to provide a theoretical foundation for developing novel AML therapies based on metabolic regulation.

Factors in the TME that influence T cell metabolism in AML

High levels of LA

A defining feature of the AML TME is the production of excessive LA, creating a highly acidic environment (18). A study reported that LA inhibits T cell growth and proliferation, and its accumulation in the TME disrupts LA efflux from T cells, impairing their metabolism, function and antitumor immunity (28). Additionally, LA downregulates perforin and granzyme B, which are essential for T cell-mediated tumor cell killing and proliferation (29,30). Dichloroacetic acid (DCA) targets pyruvate dehydrogenase kinase (PDK), an enzyme that drives glucose metabolism toward glycolysis instead of oxidative phosphorylation (OXPHOS) in tumor cells. By inhibiting PDK, DCA offers a potential strategy to modulate glucose metabolism in the AML TME (31). DCA can inhibit the shift toward glycolysis, reducing LA levels and notably enhancing T cell proliferation, and cytokine production and function whilst decreasing apoptosis (32). Tregs use monocarboxylate transporter 1 to uptake LA, which activates signaling pathways that promote nuclear factor of activated T cells 1 translocation to the nucleus, upregulating PD-1 expression (Fig. 1) (33). Therefore, high LA levels increase PD-1 expression in Tregs, amplifying their immunosuppressive effects. Additionally, LA accumulation and the resulting acidic pH in the TME inhibit CD4+ T cell activity, particularly the secretion of key cytokines such as IL-2 and IFN-γ. These cytokines are crucial for immune responses, and their reduction weakens CD4+ T cell proliferation and helper function, further promoting immunosuppression (34). In summary, high LA levels in the AML TME are a major contributor to T cell dysfunction, impairing antitumor immunity through metabolic disruption, cytotoxic inhibition and induction of exhaustion.

Acute myeloid leukemia tumor
microenvironment is characterized by high lactic acid levels, which
create an acidic milieu that impairs T cell function. Lactic acid
inhibits perforin and granzyme B, essential for T cell-mediated
tumor cell killing, and reduces the secretion of cytokines such as
IL-2 and IFN-γ, critical for CD4+ T cell proliferation.
Lactic acid accumulation in Tregs is facilitated by MCT1,
activating NFAT1 signaling and upregulating PD-1, enhancing
Treg-mediated immunosuppression. IFN-γ, interferon-γ; Treg,
regulatory T cell; MCT1, monocarboxylate transporter 1; NFAT1,
nuclear factor of activated T cells 1; PD-1, programmed
death-1.

Figure 1.

Acute myeloid leukemia tumor microenvironment is characterized by high lactic acid levels, which create an acidic milieu that impairs T cell function. Lactic acid inhibits perforin and granzyme B, essential for T cell-mediated tumor cell killing, and reduces the secretion of cytokines such as IL-2 and IFN-γ, critical for CD4+ T cell proliferation. Lactic acid accumulation in Tregs is facilitated by MCT1, activating NFAT1 signaling and upregulating PD-1, enhancing Treg-mediated immunosuppression. IFN-γ, interferon-γ; Treg, regulatory T cell; MCT1, monocarboxylate transporter 1; NFAT1, nuclear factor of activated T cells 1; PD-1, programmed death-1.

Hypoxia

Hypoxia is a defining feature of the bone marrow TME, markedly impacting AML cell growth, metabolic reprogramming and immune interactions. Studies have reported that patients with higher hypoxia risk scores tend to have shorter overall survival rates, linking hypoxia to poor prognosis in AML. Elevated hypoxia risk scores are also strongly associated with disease progression and the immunosuppressive TME (35,36). Under hypoxic conditions, hypoxia-inducible factor 1α (HIF-1α) is activated and directly upregulates programmed death-ligand 1 (PD-L1) expression. High PD-L1 levels bind to PD-1 on T cells, inhibiting their activation and signaling, further suppressing antitumor immunity (37).

Nutrition competition

AML tumor cells and immune cells compete for essential nutrients such as glucose and amino acids, which are vital for both rapid tumor growth and normal immune function. Tumor cells, with their high metabolic demands, prioritize the uptake of glucose, glutamine and other substrates for energy and biosynthesis, depriving immune cells of these resources. For instance, AML cells depend on glutamine for OXPHOS, and the inhibition of glutaminase-1 has been reported to suppress AML development in mouse models (38). Glutamine is also critical for T cell activation, proliferation and cytokine production (39). Glutamine deficiency in the TME promotes the generation of Tregs, further exacerbating immunosuppression (40). Glutamine deficiency not only restricts effector T cell proliferation and function, but also supports the metabolic adaptation of Tregs, enhancing their immunosuppressive activity. Studies have highlighted the antisense non-coding RNA at the INK4 locus (ANRIL) as a key regulator of glucose metabolism in AML, where it is notably upregulated. ANRIL modulates glucose metabolism via the AMP-activated protein kinase (AMPK)/ sirtuin 1 pathway, promoting AML cell survival. Its knockdown reduces glucose uptake and inhibits AML cell maintenance (41,42). Under oxygen-sufficient conditions, AML cells rely heavily on glucose as their primary metabolic substrate, rapidly converting it to LA through glycolysis to meet energy demands (43). A study by Cunningham and Kohno (44) using 18FDG labeling in 124 patients reported consistently high glucose uptake in AML bone marrow, highlighting the glucose dependence of AML cells. This excessive glucose consumption by AML cells suppresses T cell activation, induces exhaustion and drives leukemia progression.

Chemokines and cytokines

The AML TME shapes T cell metabolism and function by secreting chemokines and cytokines, influencing tumor progression (45,46). Studies have reported that chemokines such as chemokine CCL3 and CXCL12 in the AML microenvironment promote Treg accumulation, which competitively inhibits Teffs and indirectly affects their metabolic activity (47,48). Whilst research does not directly address whether chemokines regulate T cell metabolic pathways, it highlights the potential of blocking Treg migration to delay disease progression. Inhibiting these chemokines has been reported to slow AML progression in mouse models (45).

Accumulation of other metabolic waste products

Potassium ions (K+), abundant in intracellular fluid, are essential electrolytes that regulate immune cell function and several cellular processes (49,50). Tumor cell necrosis releases large amounts of K+ into the extracellular fluid of the TME. Elevated extracellular K+ concentrations impair T cell receptor (TCR)-mediated Akt-mTOR phosphorylation, hindering effector T cell activation and function (51). Conversely, higher K+ levels can enhance T cell stemness, maintaining their undifferentiated state (52). This indicates that K+ dynamics in the TME influence both immediate T cell effector functions and their long-term survival and potential. However, research on the role of K+ in the AML TME remains limited.

Changes in T cell metabolic pathways in the AML microenvironment

Glycolysis and OXPHOS

Before antigen exposure, naive T cells remain in a quiescent state maintained by IL-7. As they do not require clonal expansion or high cytokine production, their reliance on anabolic pathways for DNA, protein and molecule synthesis is minimal. Instead, they generate ATP primarily through mitochondrial OXPHOS (53). By contrast, tumor cells undergo a notable metabolic shift, favoring glycolysis over OXPHOS despite its lower ATP yield. This adaptation supports their rapid proliferation and survival by quickly meeting energy and metabolic intermediate demands (54). Tregs derived from CD4+ T cells serve a dual role: They maintain immune homeostasis and prevent autoimmune diseases; however, during AML progression, they suppress CTL activity, weakening antitumor immunity and promoting immune evasion (55). Studies have reported that AML blasts promote T cell differentiation into a Treg phenotype by expressing inducible T cell co-stimulator ligand, markedly expanding the Treg population (56,57). At the AML disease site, Tregs suppress CTL activity, limiting their proliferation and hindering the expansion of adoptively transferred CTLs in vivo. This suppression weakens CTL-mediated antitumor effects, further compromising immune responses against AML (58). Research indicates that mTOR and glucose transporter-1 (GLUT-1) regulate CD4+ T cell activation by influencing glycolysis. Increased glycolysis in CD4+ T cells enhances their activation, proliferation and survival whilst promoting effector T cell differentiation and inhibiting Treg development, which suppresses immune responses (59). Similarly, the differentiation of CD8+ T cells from naive to effector states requires upregulated glucose metabolism as glycolysis provides the energy needed for their immune effector functions (60). In mouse CD8+ T cells, branched-chain amino acid (BCAA) accumulation boosts glucose transporter 1 (GLUT1) levels via a forkhead box protein O1-dependent mechanism, enhancing glycolysis and OXPHOS to strengthen antitumor immunity. BCAA supplementation also improves the efficacy of PD-1 immunotherapy in tumors (61). Thus, increased glycolysis serves a crucial role in enhancing antitumor immune responses.

In AML, leukemia cells rely heavily on glycolysis (Warburg effect) for energy, resulting in the accumulation of glycolytic byproducts such as methylglyoxal. These reactive compounds react non-enzymatically with proteins, lipids and DNA, forming advanced glycosylation end products (AGEs) (62). AGEs bind to the receptor for AGEs (RAGE), activating signaling pathways such as NF-κB, MAPK and phosphoinositide 3-kinase (PI3K)/Akt, which drive pro-inflammatory and pro-survival responses, influencing cellular functions and disease progression (63,64). AGE-RAGE signaling promotes the proliferation of AML cell lines, such as HL60 and HEL, by inhibiting apoptosis and autophagy, enhancing cancer cell survival and invasiveness. This mechanism contributes to the progression of several tumors, including AML (64). Consequently, the AGE-RAGE axis represents both a hallmark of AML metabolic reprogramming and a potential diagnostic and therapeutic target.

FAO

Fatty acid uptake and metabolism are essential for AML cell proliferation, providing energy and metabolic intermediates whilst inhibiting apoptosis and conferring resistance to cytotoxic drugs. AML cells thus depend on fatty acid metabolism for survival (65). Lipid metabolism also serves a key role in T cell metabolic reprogramming, supporting membrane expansion through the production of phospholipids and cholesterol. Naive CD8+ T cells in the lymphatic system primarily rely on FAO for energy (66). The differentiation of Teffs compared with memory T cells depends on the strength of signals from co-stimulatory molecules, cytokines and antigen presentation (67). Strong stimulation leads to the generation of short-lived terminally differentiated effector cells, whilst weaker stimulation promotes the differentiation of memory precursor cells, which further transform into long-lived memory cells, providing protection against re-infection (68). Certain memory T cells also arise from Teffs that survive apoptosis at the end of an immune response (69). In the AML TME, memory T cells exhibit markedly reduced metabolic adaptability and persistence. Research indicates that these cells fail to sustain critical metabolic pathways such as FAO and mitochondrial OXPHOS resulting in energy deficiency that compromises their survival and long-term effector function (6,70). AML cells exacerbate this dysfunction by competing for essential nutrients such as glucose and glutamine, further suppressing T cell metabolism. Prolonged nutrient deprivation not only impairs memory T cell function, but also promotes their exhaustion, characterized by upregulated expression of exhaustion markers such as PD-1 and thymocyte selection-associated high mobility group box protein, and diminished cytotoxic capacity (56,71). Notably, memory T cells also serve a role in modulating the response of hematological malignancies to PD-1 blockade therapy (72). Studies have reported that TNF receptor-associated factor 6 (TRAF6) influences CD8+ memory T cells through lipid metabolism regulation. Mice with T cell-specific TRAF6 deficiency exhibit strong effector T cell responses but fail to form memory T cells effectively (73,74). IL-15 regulates mitochondrial spare respiratory capacity and oxidative metabolism by modulating mitochondrial biogenesis and the key enzyme carnitine O-palmitoyl transferase 1 (CPT1)a. CPT1a is critical for the rate-limiting step in mitochondrial FAO (75). Enhancing FAO through AMPK activation or mTOR inhibition can notably increase memory T cell numbers (73,76). These findings emphasize the pivotal role of lipid metabolism in T cell metabolic reprogramming.

Amino acid metabolism

Studies have reported that amino acids regulate immune responses by influencing T cell activation, cytokine production and other immune functions (77). Glutamine, the most abundant amino acid in serum, is vital for maintaining metabolic balance and cell function. Its absence in culture medium markedly impairs naive T cell activation, proliferation and cytokine production (39). In the hypoxic TME, glutamine acts as a primary carbon source, supporting the energy and metabolic needs of tumor cells (78). Amino acids and their metabolites are essential in regulating both tumor and immune cell proliferation within the TME. For instance, glutamine and its metabolic pathways are crucial for tumor cell glycolysis. Using glutamine antagonists can effectively inhibit glycolysis in tumor cells, suppressing their growth and enhancing the antitumor immune response by altering the immunosuppressive TME, thereby overcoming immune evasion (79). Besides glutamine, the metabolism of arginine and tryptophan also serves a pivotal role in immune regulation within the TME. Studies have reported that monocytes, under macrophage-stimulating factor influence, rapidly degrade tryptophan through increased IDO activity, thereby suppressing T cell proliferation. This mechanism aids tumor cells and macrophages in immune evasion. Furthermore, extracellular arginine availability in the TME directly influences T cell function and antitumor responses (80,81). Arginine deprivation leads T cells to initiate autophagy, downregulate the CD3ζ chain and ultimately undergo apoptosis. In AML, this effect is more pronounced, as AML blasts express and secrete arginase II, a key enzyme for arginine metabolism, whilst arginase I is typically low and only detectable under specific conditions. This metabolic regulation further impairs T cell function, aiding AML cells in evading immune surveillance (70). These findings highlight the crucial association between amino acid metabolism in the TME and T cell-mediated antitumor immunity.

Nucleotide metabolism

Nucleotides, as essential components of genetic material, are critical for highly proliferating cells, particularly purine and pyrimidine nucleotides. Consequently, nucleotide metabolism presents a potential target for cancer therapy (82,83). Drugs such as methotrexate, which target nucleotide metabolism, have been reported to be effective in treating acute lymphoblastic leukemia (ALL). However, non-specific targeting of nucleotide metabolism can inhibit normal cell processes, leading to severe side effects (84,85). In the AML TME, high concentrations of adenosine act as an immunosuppressive metabolite. Elevated adenosine levels suppress T cell activity by inhibiting activation, proliferation and cytokine secretion through adenosine receptor binding such as A2A receptors (56). The A2A adenosine receptor signaling pathway markedly inhibits T lymphocyte proliferation, activation and cytokine production. Additionally, this pathway activates immunosuppressive cells, such as Tregs and MDSCs, further impairing effector immune cell function (86). Activation of A2A receptors not only suppresses effector T cell activity, but also enhances Treg cell immunoregulatory function by upregulating key molecules and metabolic pathways, thus promoting immune suppression (Fig. 2) (87). Furthermore, studies have reported that LA treatment reduces nucleotide abundance in T cells, impairing proliferation and cell cycle activity. LA also disrupts several metabolic pathways, including amino acid biosynthesis and pyrimidine metabolism. NaBi itself can serve as a substrate for multiple carboxylase reactions such as pyrimidine metabolism (88,89). The application of NaBi can reverse these changes, pyrimidine metabolism increased in T cells rescued with NaBi (19).

Adenosine binds to receptors on Teff,
Treg, and APCs, suppressing Teff activation, proliferation and
cytokine secretion whilst enhancing Treg-mediated immunoregulation.
This signaling pathway also decreases the production of IL-12 and
increases IL-10, further weakening the antitumor immune response.
Treg, regulatory T cell; Teff, Tregs suppress effector T cell; APC,
adenomatous polyposis coli.

Figure 2.

Adenosine binds to receptors on Teff, Treg, and APCs, suppressing Teff activation, proliferation and cytokine secretion whilst enhancing Treg-mediated immunoregulation. This signaling pathway also decreases the production of IL-12 and increases IL-10, further weakening the antitumor immune response. Treg, regulatory T cell; Teff, Tregs suppress effector T cell; APC, adenomatous polyposis coli.

Signaling pathways of T cell metabolic remodeling in AML TME

mTOR signaling pathway

mTOR, a serine/threonine kinase, serves a pivotal role in several cellular processes, including metabolism regulation. It is a key regulator of cell metabolism and serves as the catalytic subunit of both mTORC1 and mTORC2 complexes. mTORC1 supports effector T cell function by promoting glycolysis and protein synthesis, whilst mTORC2 regulates T cell differentiation and survival through the cytoskeleton and lipid metabolism (90). mTOR enhances glucose uptake and glycogen synthesis by modulating the insulin receptor substrate 1 (IRS1)/PI3K/Akt pathway, thereby boosting glycolysis. Inhibition of mTOR activation or its downregulation in CD4+ T cells reduce glycolysis, impairing their activation (31). mTOR activation also increases GLUT1 expression, promoting T cell proliferation and cytokine production (Fig. 3) (91). Additionally, mTOR is a crucial regulator of memory CD8+ T cell differentiation, with the mTOR-specific inhibitor rapamycin, an immunosuppressive drug, demonstrating an immunostimulatory effect on memory CD8+ T cells (87).

mTOR enhances glucose uptake and
glycolysis in CD4+ T cells by increasing GLUT1
expression and regulating the IRS1/PI3K/AKT pathway. This promotes
glycogen synthesis, T cell proliferation and activation, favoring
Th1/Th17 differentiation whilst reducing Treg generation.
Inhibition of mTOR impairs glycolysis and CD4+ T cell
activation. GLUT1, glucose transporter 1; IRS1, insulin receptor
substrate 1; Treg, regulatory T cell; AMPK, AMP-activated protein
kinase; Th, Helper T cell.

Figure 3.

mTOR enhances glucose uptake and glycolysis in CD4+ T cells by increasing GLUT1 expression and regulating the IRS1/PI3K/AKT pathway. This promotes glycogen synthesis, T cell proliferation and activation, favoring Th1/Th17 differentiation whilst reducing Treg generation. Inhibition of mTOR impairs glycolysis and CD4+ T cell activation. GLUT1, glucose transporter 1; IRS1, insulin receptor substrate 1; Treg, regulatory T cell; AMPK, AMP-activated protein kinase; Th, Helper T cell.

AMPK signaling pathway

In the AML TME, AMPK, as a key energy sensor, serves a crucial role in regulating the metabolic state of immune cells, enabling them to effectively maintain their activity and function. It also serves an important role in T cell metabolic reprogramming. A previous study reported that AMPK signaling promotes lipid metabolism to generate functional memory CD8+ T cells (92). Furthermore, as an upstream inhibitor of mTOR activity, AMPK can inhibit mTOR through the AMPK activator metformin, which helps reduce glycolysis in T cells (Fig. 4). This, in turn, promotes the generation of Tregs by suppressing Th1 and Th17 cells (93). Metformin also activates AMPK and inhibits the proliferation of AML cell lines and primary AML cells (94). However, future research needs to further explore the specific molecular mechanisms of AMPK activation and its potential for clinical translation.

As an mTOR inhibitor, AMPK suppresses
mTOR activity via metformin, reducing glycolysis and promoting Treg
generation whilst suppressing Th1/Th17 differentiation. AMPK also
upregulates CPT1, enhancing FAO and supporting T cell metabolic
reprogramming. Additionally, HIF-1α promotes Treg migration by
upregulating glycolysis and FAO. AMPK, AMP-activated protein
kinase; Th, Helper T cell; Treg, regulatory T cell; CPT1, carnitine
O-palmitoyl transferase 1; HIF-1α, hypoxia-inducible factor 1α;
LDHA, lactate dehydrogenase A; FAO, fatty acid oxidation; Tm,
memory T cell.

Figure 4.

As an mTOR inhibitor, AMPK suppresses mTOR activity via metformin, reducing glycolysis and promoting Treg generation whilst suppressing Th1/Th17 differentiation. AMPK also upregulates CPT1, enhancing FAO and supporting T cell metabolic reprogramming. Additionally, HIF-1α promotes Treg migration by upregulating glycolysis and FAO. AMPK, AMP-activated protein kinase; Th, Helper T cell; Treg, regulatory T cell; CPT1, carnitine O-palmitoyl transferase 1; HIF-1α, hypoxia-inducible factor 1α; LDHA, lactate dehydrogenase A; FAO, fatty acid oxidation; Tm, memory T cell.

Peroxisome proliferator-activated receptor (PPAR) family of transcription factors

The PPAR family includes PPARα, PPARδ and PPARγ. The nuclear receptor PPARγ serves an essential role in adipogenesis, immune responses and the metabolism of lipids and carbohydrates. Fatty acids can also act as ligands for PPARγ (95,96). Study have reported that in chronic lymphocytic leukemia (CLL), high doses of glucocorticoids induce the activation of PPARα and downstream FAO, leading to drug resistance (97). Moreover, it has been reported that activation of PPAR promotes the proliferation of CD8+ T cells, increasing the number of functional Teffs. The activation of the PPAR pathway can also rescue PD-1 blockade-induced T cell apoptosis by upregulating anti-apoptotic proteins such as Bcl2, baculoviral IAP repeat containing 3 and apoptosis inhibitor 5. Additionally, PPAR activation can reprogram CTL energy metabolism and overcome the reduction in the number of functional Teffs associated with PD-1 blockade by reducing apoptosis or increasing proliferation (98). Therefore, targeting the PPAR signaling pathway, such as by using PPAR agonists, may serve as a potential therapeutic target for AML.

HIF-1α and hypoxic response

HIF-1α is a central transcription factor in hypoxic cells and a hallmark of TME. It is also a downstream target of GLUT-1 (99). It facilitates Treg migration by promoting glycolysis and FAO. Elevated glucose uptake by cancer cells stabilizes HIF-1α, thereby suppressing antitumor immunity (100,101). Moreover, HIF-1α-driven transcription enhances glycolysis in T cells, supporting Th17 differentiation whilst inhibiting Tregs (91). Mitochondrial dysfunction and HIF-1α-mediated metabolic reprogramming contribute to T cell exhaustion, a process reversible through glycolysis inhibition (102). Treatment with digoxin or acriflavine, both inhibitors of HIF-1 expression and function, in subcutaneous tumor mice has been reported to limit tumor growth (103). Therefore, targeting HIF-1 in the TME may be an effective therapeutic strategy for AML.

Metabolic features of the AML microenvironment and their impact on immunotherapy

Immune checkpoint (IC) inhibitors

ICs are molecular mechanisms that regulate immune system activity, comprising co-stimulatory receptors such as CD40 and CD80 and inhibitory receptors such as cytotoxic T-lymphocyte-associated protein 4 and PD-1. These checkpoints maintain immune tolerance, protect normal tissues from excessive immune responses, or, in certain cases, enable cancer cells to evade immune surveillance (34). The advent of IC inhibitors has improved the prognosis for numerous solid tumors and certain lymphomas by blocking inhibitory signals such as the PD-1/PD-L1 pathway, thereby enhancing antitumor immunity (104). However, their efficacy in AML remains limited, particularly with PD-1/PD-L1 inhibitors (105), for reasons that are not yet fully understood. In the AML TME, competition for nutrients between AML cells and T cells restricts T cell access to glucose and glutamine, impairing their metabolic function and antitumor response. Consequently, IC inhibitors fail to enhance T cell-mediated antitumor effects (22). Furthermore, the PD-1/PD-L1 interaction serves a critical immunosuppressive role in the TME, promoting regulatory T cell function and inhibiting the activation and proliferation of Teffs, thereby further dampening the antitumor immune response (106).

Limitations of adoptive T cell therapy (ACT)

ACT enhances immune responses against tumors or infections by modifying or expanding autologous or donor-derived T cells ex vivo. This includes engineering T cells with chimeric antigen receptors (CARs) or TCRs to recognize specific tumor antigens. Following expansion, these Teffs are re-infused to mediate targeted immune responses (107,108). However, unlike in ALL, CAR-T cell therapy shows limited efficacy in AML, largely due to the immunosuppressive TME (29). Lymphodepleting chemotherapy prior to CAR-T cell infusion can enhance therapeutic outcomes by reducing Tregs in the TME and alleviating their suppressive effects, thereby improving CAR-T cell proliferation and persistence in vivo (109). In AML, expanded Tregs secrete immunosuppressive cytokines such as IL-10 and TGF-β, which impair CAR-T cell function (57). Additionally, AML cells secrete arginase II, disrupting T cell metabolism and promoting immune evasion (41). Inhibition of arginine metabolism has been reported to enhance the efficacy of CD33-CAR T cells in preclinical AML models (110). Similarly, blocking the adenosine A2A receptor (A2AR), a downstream mediator of adenosine signaling, improves CAR-T cell efficacy in solid tumors (111). Adenosine suppresses T cell proliferation, activation and effector function via A2AR, whilst promoting Treg expansion, thereby dampening antitumor immunity (112). However, whether this mechanism extends to hematologic malignancies such as AML and CLL remains unclear.

Potential therapeutic strategies for metabolic regulation

In patients with AML, elevated intracellular and plasma arginase activity markedly inhibits T cell proliferation, contributing to immune dysfunction. This effect is largely mediated by increased expression and activity of arginase II in AML cells, identifying it as a potential biomarker for immune status and disease progression. Inhibition of arginase II has been reported to restore T cell proliferation and enhance antitumor immunity (110,113). When PD-1 binds to its ligand PD-L1, activated T cells are unable to continue glycolysis and normal amino acid metabolism, which results in insufficient energy production to support their effector functions. In addition to inhibiting glycolysis and amino acid metabolism, PD-1 may also impair T cell oxidative detoxification capacity, reducing their ability to cope with oxidative stress (114). Thus, elucidating the PD-1 signaling axis is crucial for understanding T cell dysfunction and identifying novel therapeutic targets.

Future research directions and challenges

Key areas for further exploration in T cell metabolism in AML TME

Although the role of T cell metabolism in the AML TME has been preliminarily elucidated, further in-depth exploration is needed in the following key areas.

Metabolic reprogramming and personalized therapy

In future research, individualized treatments targeting T cell metabolic reprogramming in the TME of patients with AML should focus on the impact of metabolic heterogeneity on treatment responses. Differences in metabolic characteristics between patients with AML may profoundly influence the metabolic state of T cells and their antitumor capabilities. For example, variations in glycolysis, FAO or amino acid metabolism across different patients could lead to notable differences in therapeutic efficacy. Utilizing metabolomics and single-cell analysis techniques could uncover individual differences in T cell metabolic reprogramming and provide a basis for precise metabolic interventions. However, this approach faces several challenges, such as the ways to integrate multidimensional data to accurately identify key metabolic nodes, implement personalized metabolic regulation of targets in clinical applications and minimize potential side effects of metabolic interventions on systemic metabolic homeostasis. In the future, combining advanced technological methods and large-scale clinical studies will be necessary to explore the feasibility of personalized metabolic interventions, with the goal of achieving precision treatment for patients with AML.

Application of emerging technologies

In future research on T cell metabolic reprogramming within the AML TME, emerging technologies will provide crucial support for uncovering metabolic regulatory mechanisms and therapeutic potential. The application of single-cell metabolomics and spatial metabolomics can capture the dynamic changes and spatial heterogeneity of T cell metabolism in the AML microenvironment with high resolution, offering a new perspective on the role of metabolic reprogramming in tumor immune evasion. Moreover, metabolic flux analysis can track the dynamic changes in key metabolic pathways within T cells in real-time, revealing the flow and regulation patterns of metabolites under different conditions. By contrast, CRISPR screening technology can precisely identify key genes and metabolic nodes involved in T cell metabolic reprogramming, providing specific targets for developing intervention strategies. The combination of these technologies will not only deepen the understanding of T cell metabolic regulation mechanisms but also advance the design of personalized metabolic treatment plans. However, the application of the aforementioned technologies in AML still faces challenges, such as integrating multidimensional data, high costs and unclear clinical translation pathways. Future multi-disciplinary collaborations will be required to further optimize their application.

Challenge of clinical translation

There are still notable obstacles and challenges in translating the research on T cell metabolic reprogramming within the AML TME into clinical application. A key obstacle from basic research to clinical use is the way to simplify complex metabolic mechanism studies into clear clinical targets, whilst ensuring these targets have broad applicability across heterogeneous patient populations. Moreover, although certain preliminary progress has been made in clinical trials of metabolic interventions in AML, such as improving the immune microenvironment through the regulation of glycolysis, FAO or amino acid metabolism, issues such as individual variability in efficacy, long-term treatment side effects and resistance remain prominent. Metabolic interventions may affect systemic metabolic homeostasis leading to unpredictable toxic reactions, and the adaptive metabolic mechanisms of tumor cells may induce resistance. Therefore, future research needs more precise targeting strategies to achieve efficient regulation within specific metabolic pathways, whilst minimizing systemic effects. The combination of advanced technologies, such as metabolomics and single-cell analysis for personalized treatment design, along with the development of combination therapies to mitigate resistance, may be key approaches to overcoming these challenges.

Conclusions

The present review explored the key mechanisms of T cell metabolic reprogramming in the TME of AML and its notable impact on antitumor immune responses. The AML microenvironment, through the synergistic effects of high LA levels, hypoxia, nutrient competition and chemokines, markedly suppresses critical metabolic pathways in T cells, such as glycolysis, lipid metabolism and amino acid metabolism, weakening their proliferation, effector functions and antitumor capabilities. Additionally, the accumulation of metabolic waste products from AML cells, as well as abnormalities in adenosine and potassium ion metabolism, further promotes the establishment of an immunosuppressive state. Furthermore, although the mechanisms of T cell metabolic reprogramming are preliminarily understood, designing personalized treatment strategies based on the metabolic characteristics of patients remains a major challenge. Emerging technologies, such as single-cell metabolomics and metabolic flux analysis, provide new research directions for uncovering metabolic mechanisms and developing metabolic-targeted therapies. At the same time, future clinical translation needs to balance efficacy with side effects, optimizing metabolic intervention strategies to enhance the effectiveness of immunotherapy.

In summary, in-depth research on T cell metabolic reprogramming in the AML microenvironment will provide important theoretical support for improving AML immunotherapy strategies, whilst also offering new insights for the clinical application of metabolic intervention therapies.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

XYZ designed and conceived the study. YHL wrote the manuscript. JL and MY revised the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Döhner H, Weisdorf DJ and Bloomfield CD: Acute myeloid leukemia. N Engl J Med. 373:1136–1152. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Liu H: Emerging agents and regimens for AML. J Hematol Oncol. 14:492021. View Article : Google Scholar : PubMed/NCBI

3 

Peng C, Xu Y, Wu J, Wu D, Zhou L and Xia X: TME-related biomimetic strategies against cancer. Int J Nanomedicine. 19:109–135. 2024. View Article : Google Scholar : PubMed/NCBI

4 

Bawek S, Gurusinghe S, Burwinkel M and Przespolewski A: Updates in novel immunotherapeutic strategies for relapsed/refractory AML. Front Oncol. 14:13749632024. View Article : Google Scholar : PubMed/NCBI

5 

Menter T and Tzankov A: Tumor microenvironment in acute myeloid leukemia: Adjusting niches. Front Immunol. 13:8111442022. View Article : Google Scholar : PubMed/NCBI

6 

Lamble AJ and Lind EF: Targeting the immune microenvironment in acute myeloid leukemia: A focus on T cell immunity. Front Oncol. 8:2132018. View Article : Google Scholar : PubMed/NCBI

7 

Korn C and Méndez-Ferrer S: Myeloid malignancies and the microenvironment. Blood. 129:811–822. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Rieger CT and Fiegl M: Microenvironmental oxygen partial pressure in acute myeloid leukemia: Is there really a role for hypoxia? Exp Hematol. 44:578–582. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Yu S and Jiang J: Immune infiltration-related genes regulate the progression of AML by invading the bone marrow microenvironment. Front Immunol. 15:14099452024. View Article : Google Scholar : PubMed/NCBI

10 

Zeng T, Cui L, Huang W, Liu Y, Si C, Qian T, Deng C and Fu L: The establishment of a prognostic scoring model based on the new tumor immune microenvironment classification in acute myeloid leukemia. BMC Med. 19:1762021. View Article : Google Scholar : PubMed/NCBI

11 

Chraa D, Naim A, Olive D and Badou A: T lymphocyte subsets in cancer immunity: Friends or foes. J Leukoc Biol. 105:243–255. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Plitas G and Rudensky AY: Regulatory T cells: Differentiation and function. Cancer Immunol Res. 4:721–725. 2016. View Article : Google Scholar : PubMed/NCBI

13 

MacIver NJ, Michalek RD and Rathmell JC: Metabolic regulation of T lymphocytes. Annu Rev Immunol. 31:259–283. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Lochner M, Berod L and Sparwasser T: Fatty acid metabolism in the regulation of T cell function. Trends Immunol. 36:81–91. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Endo Y, Kanno T and Nakajima T: Fatty acid metabolism in T-cell function and differentiation. Int Immunol. 34:579–587. 2022. View Article : Google Scholar : PubMed/NCBI

16 

Geiger R, Rieckmann JC, Wolf T, Basso C, Feng Y, Fuhrer T, Kogadeeva M, Picotti P, Meissner F, Mann M, et al: L-arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell. 167:829–842.e13. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Wang J, He Y, Hu F, Hu C, Sun Y, Yang K and Yang S: Metabolic reprogramming of immune cells in the tumor microenvironment. Int J Mol Sci. 25:122232024. View Article : Google Scholar : PubMed/NCBI

18 

Halestrap AP: The monocarboxylate transporter family-structure and functional characterization. IUBMB Life. 64:1–9. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Uhl FM, Chen S, O'Sullivan D, Edwards-Hicks J, Richter G, Haring E, Andrieux G, Halbach S, Apostolova P, Büscher J, et al: Metabolic reprogramming of donor T cells enhances graft-versus-leukemia effects in mice and humans. Sci Transl Med. 12:eabb89692020. View Article : Google Scholar : PubMed/NCBI

20 

Ju HQ, Zhan G, Huang A, Sun Y, Wen S, Yang J, Lu WH, Xu RH, Li J, Li Y, et al: ITD mutation in FLT3 tyrosine kinase promotes Warburg effect and renders therapeutic sensitivity to glycolytic inhibition. Leukemia. 31:2143–2150. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Herst PM, Howman RA, Neeson PJ, Berridge MV and Ritchie DS: The level of glycolytic metabolism in acute myeloid leukemia blasts at diagnosis is prognostic for clinical outcome. J Leukoc Biol. 89:51–55. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Herst PM, Hesketh EL, Ritchie DS and Berridge MV: Glycolytic metabolism confers resistance to combined all-trans retinoic acid and arsenic trioxide-induced apoptosis in HL60rho0 cells. Leuk Res. 32:327–333. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Jones RG and Thompson CB: Tumor suppressors and cell metabolism: A recipe for cancer growth. Genes Dev. 23:537–548. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Röhrig F and Schulze A: The multifaceted roles of fatty acid synthesis in cancer. Nat Rev Cancer. 16:732–749. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Heintzman DR, Fisher EL and Rathmell JC: Microenvironmental influences on T cell immunity in cancer and inflammation. Cell Mol Immunol. 19:316–326. 2022. View Article : Google Scholar : PubMed/NCBI

26 

Zha C, Yang X, Yang J, Zhang Y and Huang R: Immunosuppressive microenvironment in acute myeloid leukemia: Overview, therapeutic targets and corresponding strategies. Ann Hematol. 103:4883–4899. 2024. View Article : Google Scholar : PubMed/NCBI

27 

Böttcher M, Baur R, Stoll A, Mackensen A and Mougiakakos D: Linking immunoevasion and metabolic reprogramming in B-cell-derived lymphomas. Front Oncol. 10:5947822020. View Article : Google Scholar : PubMed/NCBI

28 

Fischer K, Hoffmann P, Voelkl S, Meidenbauer N, Ammer J, Edinger M, Gottfried E, Schwarz S, Rothe G, Hoves S, et al: Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood. 109:3812–3819. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Chen Y, Feng Z, Kuang X, Zhao P, Chen B, Fang Q, Cheng W and Wang J: Increased lactate in AML blasts upregulates TOX expression, leading to exhaustion of CD8+ cytolytic T cells. Am J Cancer Res. 11:5726–6742. 2021.PubMed/NCBI

30 

Voskoboinik I, Whisstock JC and Trapani JA: Perforin and granzymes: Function, dysfunction and human pathology. Nat Rev Immunol. 15:388–400. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Sradhanjali S and Reddy MM: Inhibition of pyruvate dehydrogenase kinase as a therapeutic strategy against cancer. Curr Top Med Chem. 18:444–453. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Rostamian H, Khakpoor-Koosheh M, Jafarzadeh L, Masoumi E, Fallah-Mehrjardi K, Tavassolifar MJ, M Pawelek J, Mirzaei HR and Hadjati J: Restricting tumor lactic acid metabolism using dichloroacetate improves T cell functions. BMC Cancer. 22:392022. View Article : Google Scholar : PubMed/NCBI

33 

Kumagai S, Koyama S, Itahashi K, Tanegashima T, Lin YT, Togashi Y, Kamada T, Irie T, Okumura G, Kono H, et al: Lactic acid promotes PD-1 expression in regulatory T cells in highly glycolytic tumor microenvironments. Cancer Cell. 40:201–218.e9. 2022. View Article : Google Scholar : PubMed/NCBI

34 

Yang P, Sun Y, Zhang M, Hu L, Wang X, Luo L, Qiao C, Wang J, Xiao H, Li X, et al: The inhibition of CD4+ T cell proinflammatory response by lactic acid is independent of monocarboxylate transporter 1. Scand J Immunol. 94:e131032021. View Article : Google Scholar

35 

Jiang F, Mao Y, Lu B, Zhou G and Wang J: A hypoxia risk signature for the tumor immune microenvironment evaluation and prognosis prediction in acute myeloid leukemia. Sci Rep. 11:146572021. View Article : Google Scholar : PubMed/NCBI

36 

Liu X, Wang L, Kang Q, Feng C and Wang J: A hypoxia-related genes prognostic risk model, and mechanisms of hypoxia contributing to poor prognosis through immune microenvironment and drug resistance in acute myeloid leukemia. Front Pharmacol. 15:13394652024. View Article : Google Scholar : PubMed/NCBI

37 

Augustin RC, Delgoffe GM and Najjar YG: Characteristics of the tumor microenvironment that influence immune cell functions: Hypoxia, oxidative stress, metabolic alterations. Cancers (Basel). 12:38022020. View Article : Google Scholar : PubMed/NCBI

38 

Jacque N, Ronchetti AM, Larrue C, Meunier G, Birsen R, Willems L, Saland E, Decroocq J, Maciel TT, Lambert M, et al: Targeting glutaminolysis has antileukemic activity in acute myeloid leukemia and synergizes with BCL-2 inhibition. Blood. 126:1346–1356. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Carr EL, Kelman A, Wu GS, Gopaul R, Senkevitch E, Aghvanyan A, Turay AM and Frauwirth KA: Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation. J Immunol. 185:1037–1044. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Klysz D, Tai XG, Robert PA, Craveiro M, Cretenet G, Oburoglu L, Mongellaz C, Floess S, Fritz V, Matias MI, et al: Glutamine-dependent α-ketoglutarate production regulates the balance between T helper 1 cell and regulatory T cell generation. Sci Signal. 8:ra972015. View Article : Google Scholar : PubMed/NCBI

41 

Sun LY, Li XJ, Sun YM, Huang W, Fang K, Han C, Chen ZH, Luo XQ, Chen YQ and Wang WT: LncRNA ANRIL regulates AML development through modulating the glucose metabolism pathway of AdipoR1/AMPK/SIRT1. Mol Cancer. 17:1272018. View Article : Google Scholar : PubMed/NCBI

42 

Balihodzic A, Barth DA, Prinz F and Pichler M: Involvement of long non-coding RNAs in glucose metabolism in cancer. Cancers (Basel). 13:9772021. View Article : Google Scholar : PubMed/NCBI

43 

Pavlova NN, Zhu J and Thompson CB: The hallmarks of cancer metabolism: Still emerging. Cell Metab. 34:355–377. 2022. View Article : Google Scholar : PubMed/NCBI

44 

Cunningham I and Kohno B: 18 FDG-PET/CT: 21st century approach to leukemic tumors in 124 cases. Am J Hematol. 91:379–384. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Wang R, Feng W, Wang H, Wang L, Yang X, Yang F, Zhang Y, Liu X, Zhang D, Ren Q, et al: Blocking migration of regulatory T cells to leukemic hematopoietic microenvironment delays disease progression in mouse leukemia model. Cancer Lett. 469:151–161. 2020. View Article : Google Scholar : PubMed/NCBI

46 

Bakker E, Qattan M, Mutti L, Demonacos C and Krstic-Demonacos M: The role of microenvironment and immunity in drug response in leukemia. Biochim Biophys Acta. 1863:414–426. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Zhang M, Yang Y, Liu J, Guo L, Guo Q and Liu W: Bone marrow immune cells and drug resistance in acute myeloid leukemia. Exp Biol Med (Maywood). 250:102352025. View Article : Google Scholar : PubMed/NCBI

48 

Ciciarello M, Corradi G, Forte D, Cavo M and Curti A: Emerging bone marrow microenvironment-driven mechanisms of drug resistance in acute myeloid leukemia: Tangle or chance? Cancers (Basel). 13:53192021. View Article : Google Scholar : PubMed/NCBI

49 

Feske S, Colucci F and Coetzee WA: Do KATP channels have a role in immunity? Front Immunol. 15:14849712024. View Article : Google Scholar : PubMed/NCBI

50 

Feske S, Wulff H and Skolnik EY: Ion channels in innate and adaptive immunity. Annu Rev Immunol. 33:291–353. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Eil R, Vodnala SK, Clever D, Klebanoff CA, Sukumar M, Pan JH, Palmer DC, Gros A, Yamamoto TN, Patel SJ, et al: Ionic immune suppression within the tumour microenvironment limits T cell effector function. Nature. 537:539–543. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Vodnala SK, Eil R, Kishton RJ, Sukumar M, Yamamoto TN, Ha NH, Lee PH, Shin M, Patel SJ, Yu Z, et al: T cell stemness and dysfunction in tumors are triggered by a common mechanism. Science. 363:eaau01352019. View Article : Google Scholar : PubMed/NCBI

53 

Almeida L, Lochner M, Berod L and Sparwasser T: Metabolic pathways in T cell activation and lineage differentiation. Semin Immunol. 28:514–524. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Fukushi A, Kim HD, Chang YC and Kim CH: Revisited metabolic control and reprogramming cancers by means of the warburg effect in tumor cells. Int J Mol Sci. 23:100372022. View Article : Google Scholar : PubMed/NCBI

55 

Riether C: Regulation of hematopoietic and leukemia stem cells by regulatory T cells. Front Immunol. 13:10493012022. View Article : Google Scholar : PubMed/NCBI

56 

Epperly R, Gottschalk S and Velasquez MP: A bump in the road: how the hostile AML microenvironment affects CAR T cell therapy. Front Oncol. 10:2622020. View Article : Google Scholar : PubMed/NCBI

57 

Han Y, Dong Y, Yang Q, Xu W, Jiang S, Yu Z, Yu K and Zhang S: Acute myeloid leukemia cells express ICOS ligand to promote the expansion of regulatory T cells. Front Immunol. 9:22272018. View Article : Google Scholar : PubMed/NCBI

58 

Zhou Q, Bucher C, Munger ME, Highfill SL, Tolar J, Munn DH, Levine BL, Riddle M, June CH, Vallera DA, et al: Depletion of endogenous tumor-associated regulatory T cells improves the efficacy of adoptive cytotoxic T-cell immunotherapy in murine acute myeloid leukemia. Blood. 114:3793–3802. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Liu SY, Liao S, Liang L, Deng J and Zhou Y: The relationship between CD4+ T cell glycolysis and their functions. Trends Endocrinol Metab. 34:345–360. 2023. View Article : Google Scholar : PubMed/NCBI

60 

Cao J, Liao S, Zeng F, Liao Q, Luo G and Zhou Y: Effects of altered glycolysis levels on CD8+ T cell activation and function. Cell Death Dis. 14:4072023. View Article : Google Scholar : PubMed/NCBI

61 

Yao CC, Sun RM, Yang Y, Zhou HY, Meng ZW, Chi R, Xia LL, Ji P, Chen YY, Zhang GQ, et al: Accumulation of branched-chain amino acids reprograms glucose metabolism in CD8+ T cells with enhanced effector function and anti-tumor response. Cell Rep. 42:1121862023. View Article : Google Scholar : PubMed/NCBI

62 

Rabbani N and Thornalley PJ: Methylglyoxal, glyoxalase 1 and the dicarbonyl proteome. Amino Acids. 42:1133–1142. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Palanissami G and Paul SFD: AGEs and RAGE: Metabolic and molecular signatures of the glycation-inflammation axis in malignant or metastatic cancers. Explor Target Antitumor Ther. 4:812–849. 2023. View Article : Google Scholar : PubMed/NCBI

64 

Waghela BN, Vaidya FU, Ranjan K, Chhipa AS, Tiwari BS and Pathak C: AGE-RAGE synergy influences programmed cell death signaling to promote cancer. Mol Cell Biochem. 476:585–598. 2021. View Article : Google Scholar : PubMed/NCBI

65 

Bakhtiyari M, Liaghat M, Aziziyan F, Shapourian H, Yahyazadeh S, Alipour M, Shahveh S, Maleki-Sheikhabadi F, Halimi H, Forghaniesfidvajani R, et al: The role of bone marrow microenvironment (BMM) cells in acute myeloid leukemia (AML) progression: Immune checkpoints, metabolic checkpoints, and signaling pathways. Cell Commun Signal. 21:2522023. View Article : Google Scholar : PubMed/NCBI

66 

Wang R, Liu Z, Fan Z and Zhan H: Lipid metabolism reprogramming of CD8+ T cell and therapeutic implications in cancer. Cancer Lett. 567:2162672023. View Article : Google Scholar : PubMed/NCBI

67 

Jameson SC and Masopust D: Understanding subset diversity in T cell memory. Immunity. 48:214–226. 2018. View Article : Google Scholar : PubMed/NCBI

68 

Kaech SM and Cui W: Transcriptional control of effector and memory CD8+ T cell differentiation. Nat Rev Immunol. 12:749–761. 2012. View Article : Google Scholar : PubMed/NCBI

69 

D'Cruz LM, Rubinstein MP and Goldrath AW: Surviving the crash: Transitioning from effector to memory CD8+ T cell. Semin Immunol. 21:92–98. 2009. View Article : Google Scholar : PubMed/NCBI

70 

Mougiakakos D: The induction of a permissive environment to promote T cell immune evasion in acute myeloid leukemia: The metabolic perspective. Front Oncol. 9:11662019. View Article : Google Scholar : PubMed/NCBI

71 

Noviello M, Manfredi F, Ruggiero E, Perini T, Oliveira G, Cortesi F, De Simone P, Toffalori C, Gambacorta V, Greco R, et al: Bone marrow central memory and memory stem T-cell exhaustion in AML patients relapsing after HSCT. Nat Commun. 10:10652019. View Article : Google Scholar : PubMed/NCBI

72 

Abbas HA, Hao D, Tomczak K, Barrodia P, Im JS, Reville PK, Alaniz Z, Wang W, Wang R, Wang F, et al: Single cell T cell landscape and T cell receptor repertoire profiling of AML in context of PD-1 blockade therapy. Nat Commun. 12:60712021. View Article : Google Scholar : PubMed/NCBI

73 

Pearce EL, Walsh MC, Cejas PJ, Harms GM, Shen H, Wang LS, Jones RG and Choi Y: Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature. 460:103–107. 2009. View Article : Google Scholar : PubMed/NCBI

74 

Raud B, McGuire PJ, Jones RG, Sparwasser T and Berod L: Fatty acid metabolism in CD8+ T cell memory: Challenging current concepts. Immunol Rev. 283:213–231. 2018. View Article : Google Scholar : PubMed/NCBI

75 

van der Windt GJW, Everts B, Chang CH, Curtis JD, Freitas TC, Amiel E, Pearce EJ and Pearce EL: Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity. 36:68–78. 2012. View Article : Google Scholar : PubMed/NCBI

76 

Araki K, Turner AP, Shaffer VO, Gangappa S, Keller SA, Bachmann MF, Larsen CP and Ahmed R: mTOR regulates memory CD8 T-cell differentiation. Nature. 460:108–112. 2009. View Article : Google Scholar : PubMed/NCBI

77 

Li P, Yin YL, Li D, Woo Kim S and Wu G: Amino acids and immune function. Br J Nutr. 98:237–252. 2007. View Article : Google Scholar : PubMed/NCBI

78 

Wang Y, Bai C, Ruan Y, Liu M, Chu Q, Qiu L, Yang C and Li B: Coordinative metabolism of glutamine carbon and nitrogen in proliferating cancer cells under hypoxia. Nat Commun. 10:2012019. View Article : Google Scholar : PubMed/NCBI

79 

Leone RD, Zhao L, Englert JM, Sun IM, Oh MH, Sun IH, Arwood ML, Bettencourt IA, Patel CH, Wen J, et al: Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science. 366:1013–1021. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A and Mellor AL: Inhibition of T cell proliferation by macrophage tryptophan catabolism. J Exp Med. 189:1363–1372. 1999. View Article : Google Scholar : PubMed/NCBI

81 

Murray PJ: Amino acid auxotrophy as a system of immunological control nodes. Nat Immunol. 17:132–139. 2016. View Article : Google Scholar : PubMed/NCBI

82 

Di Marcantonio D, Martinez E, Kanefsky JS, Huhn JM, Gabbasov R, Gupta A, Krais JJ, Peri S, Tan Y, Skorski T, et al: ATF3 coordinates serine and nucleotide metabolism to drive cell cycle progression in acute myeloid leukemia. Mol Cell. 81:2752–2764.e6. 2021. View Article : Google Scholar : PubMed/NCBI

83 

Yabushita T and Goyama S: Nucleic acid metabolism: The key therapeutic target for myeloid tumors. Exp Hematol. 142:1046932025. View Article : Google Scholar : PubMed/NCBI

84 

Capelletti MM, Montini O, Ruini E, Tettamanti S, Savino AM and Sarno J: Unlocking the heterogeneity in acute leukaemia: Dissection of clonal architecture and metabolic properties for clinical interventions. Int J Mol Sci. 26:452024. View Article : Google Scholar : PubMed/NCBI

85 

Wu HL, Gong Y, Ji P, Xie YF, Jiang YZ and Liu GY: Targeting nucleotide metabolism: A promising approach to enhance cancer immunotherapy. J Hematol Oncol. 15:452022. View Article : Google Scholar : PubMed/NCBI

86 

Wang H, Wei Y and Wang N: Purinergic pathways and their clinical use in the treatment of acute myeloid leukemia. Purinergic Signal. Mar 6–2024.(Epub ahead of print). View Article : Google Scholar

87 

Ohta A: A metabolic immune checkpoint: Adenosine in tumor microenvironment. Front Immunol. 7:1092016. View Article : Google Scholar : PubMed/NCBI

88 

Evans DR and Guy HI: Mammalian pyrimidine biosynthesis: Fresh insights into an ancient pathway. J Biol Chem. 279:33035–33038. 2004. View Article : Google Scholar : PubMed/NCBI

89 

Santi A, Caselli A, Paoli P, Corti D, Camici G, Pieraccini G, Taddei ML, Serni S, Chiarugi P and Cirri P: The effects of CA IX catalysis products within tumor microenvironment. Cell Commun Signal. 11:812013. View Article : Google Scholar : PubMed/NCBI

90 

Pollizzi KN, Patel CH, Sun IH, Oh MH, Waickman AT, Wen J, Delgoffe GM and Powell JD: mTORC1 and mTORC2 selectively regulate CD8+ T cell differentiation. J Clin Invest. 125:2090–2108. 2015. View Article : Google Scholar : PubMed/NCBI

91 

Dabi YT, Andualem H, Degechisa ST and Gizaw ST: Targeting metabolic reprogramming of T-cells for enhanced anti-tumor response. Biologics. 16:35–45. 2022.PubMed/NCBI

92 

Saravia J, Raynor JL, Chapman NM, Lim SA and Chi H: Signaling networks in immunometabolism. Cell Res. 30:328–342. 2020. View Article : Google Scholar : PubMed/NCBI

93 

Yan Y, Huang L, Liu Y, Yi M, Chu Q, Jiao D and Wu K: Metabolic profiles of regulatory T cells and their adaptations to the tumor microenvironment: Implications for antitumor immunity. J Hematol Oncol. 15:1042022. View Article : Google Scholar : PubMed/NCBI

94 

Castro I, Sampaio-Marques B and Ludovico P: Targeting metabolic reprogramming in acute myeloid leukemia. Cells. 8:9672019. View Article : Google Scholar : PubMed/NCBI

95 

Ahmadian M, Suh JM, Hah N, Liddle C, Atkins AR, Downes M and Evans RM: PPARγ signaling and metabolism: The good, the bad and the future. Nat Med. 19:557–566. 2013. View Article : Google Scholar : PubMed/NCBI

96 

Angela M, Endo Y, Asou HK, Yamamoto T, Tumes DJ, Tokuyama H, Yokote K and Nakayama T: Fatty acid metabolic reprogramming via mTOR-mediated inductions of PPARγ directs early activation of T cells. Nat Commun. 7:136832016. View Article : Google Scholar : PubMed/NCBI

97 

Tabe Y, Konopleva M and Andreeff M: Fatty acid metabolism, bone marrow adipocytes, and AML. Front Oncol. 10:1552020. View Article : Google Scholar : PubMed/NCBI

98 

Chowdhury PS, Chamoto K, Kumar A and 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. 6:1375–1387. 2018. View Article : Google Scholar : PubMed/NCBI

99 

Zhang JZ, Behrooz A and Ismail-Beigi F: Regulation of glucose transport by hypoxia. Am J Kidney Dis. 34:189–202. 1999. View Article : Google Scholar : PubMed/NCBI

100 

Miska J, Lee-Chang C, Rashidi A, Muroski ME, Chang AL, Lopez-Rosas A, Zhang P, Panek WK, Cordero A, Han Y, et al: HIF-1α is a metabolic switch between glycolytic-driven migration and oxidative phosphorylation-driven immunosuppression of tregs in glioblastoma. Cell Rep. 27:226–237.e4. 2019. View Article : Google Scholar : PubMed/NCBI

101 

Nagao A, Kobayashi M, Koyasu S, Chow CCT and Harada H: HIF-1-dependent reprogramming of glucose metabolic pathway of cancer cells and its therapeutic significance. Int J Mol Sci. 20:2382019. View Article : Google Scholar : PubMed/NCBI

102 

Wu H, Zhao X, Hochrein SM, Eckstein M, Gubert GF, Knöpper K, Mansilla AM, Öner A, Doucet-Ladevèze R, Schmitz W, et al: Mitochondrial dysfunction promotes the transition of precursor to terminally exhausted T cells through HIF-1α-mediated glycolytic reprogramming. Nat Commun. 14:68582023. View Article : Google Scholar : PubMed/NCBI

103 

Pan F, Barbi J and Pardoll DM: Hypoxia-inducible factor 1: A link between metabolism and T cell differentiation and a potential therapeutic target. Oncoimmunology. 1:510–515. 2012. View Article : Google Scholar : PubMed/NCBI

104 

Alatrash G, Daver N and Mittendorf EA: Targeting immune checkpoints in hematologic malignancies. Pharmacol Rev. 68:1014–1025. 2016. View Article : Google Scholar : PubMed/NCBI

105 

Stahl M and Goldberg AD: Immune checkpoint inhibitors in acute myeloid leukemia: Novel combinations and therapeutic targets. Curr Oncol Rep. 21:372019. View Article : Google Scholar : PubMed/NCBI

106 

Zhou Q, Munger ME, Highfill SL, Tolar J, Weigel BJ, Riddle M, Sharpe AH, Vallera DA, Azuma M, Levine BL, et al: Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia. Blood. 116:2484–2493. 2010. View Article : Google Scholar : PubMed/NCBI

107 

Sadelain M, Brentjens R and Rivière I: The basic principles of chimeric antigen receptor design. Cancer Discov. 3:388–398. 2013. View Article : Google Scholar : PubMed/NCBI

108 

Riddell SR, Jensen MC and June CH: Chimeric antigen receptor-modified T cells: Clinical translation in stem cell transplantation and beyond. Biol Blood Marrow Transplant. 19 (1 Suppl):S2–S5. 2013. View Article : Google Scholar : PubMed/NCBI

109 

Suryadevara CM, Desai R, Farber SH, Choi BD, Swartz AM, Shen SH, Gedeon PC, Snyder DJ, Herndon JE II, Healy P, et al: Preventing Lck activation in CAR T cells confers treg resistance but requires 4-1BB signaling for them to persist and treat solid tumors in nonlymphodepleted hosts. Clin Cancer Res. 25:358–368. 2019. View Article : Google Scholar : PubMed/NCBI

110 

Mussai F, Wheat R, Sarrou E, Booth S, Stavrou V, Fultang L, Perry T, Kearns P, Cheng P, Keeshan K, et al: Targeting the arginine metabolic brake enhances immunotherapy for leukaemia. Int J Cancer. 145:2201–2208. 2019. View Article : Google Scholar : PubMed/NCBI

111 

Beavis PA, Henderson MA, Giuffrida L, Mills JK, Sek K, Cross RS, Davenport AJ, John LB, Mardiana S, Slaney CY, et al: Targeting the adenosine 2A receptor enhances chimeric antigen receptor T cell efficacy. J Clin Invest. 127:929–941. 2017. View Article : Google Scholar : PubMed/NCBI

112 

Leone RD, Sun IM, Oh MH, Sun IH, Wen J, Englert J and Powell JD: Inhibition of the adenosine A2a receptor modulates expression of T cell coinhibitory receptors and improves effector function for enhanced checkpoint blockade and ACT in murine cancer models. Cancer Immunol Immunother. 67:1271–1284. 2018. View Article : Google Scholar : PubMed/NCBI

113 

Mussai F, De Santo C, Abu-Dayyeh I, Booth S, Quek L, McEwen-Smith RM, Qureshi A, Dazzi F, Vyas P and Cerundolo V: Acute myeloid leukemia creates an arginase-dependent immunosuppressive microenvironment. Blood. 122:749–758. 2013. View Article : Google Scholar : PubMed/NCBI

114 

Patsoukis N, Bardhan K, Chatterjee P, Sari D, Liu B, Bell LN, Karoly ED, Freeman GJ, Petkova V, Seth P, et al: PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat Commun. 6:66922015. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Luo Y, Luo J, Yang M and Zhao X: Mechanisms of T‑cell metabolic reprogramming in the microenvironment of acute myeloid leukemia and its therapeutic potential (Review). Oncol Lett 30: 455, 2025.
APA
Luo, Y., Luo, J., Yang, M., & Zhao, X. (2025). Mechanisms of T‑cell metabolic reprogramming in the microenvironment of acute myeloid leukemia and its therapeutic potential (Review). Oncology Letters, 30, 455. https://doi.org/10.3892/ol.2025.15201
MLA
Luo, Y., Luo, J., Yang, M., Zhao, X."Mechanisms of T‑cell metabolic reprogramming in the microenvironment of acute myeloid leukemia and its therapeutic potential (Review)". Oncology Letters 30.4 (2025): 455.
Chicago
Luo, Y., Luo, J., Yang, M., Zhao, X."Mechanisms of T‑cell metabolic reprogramming in the microenvironment of acute myeloid leukemia and its therapeutic potential (Review)". Oncology Letters 30, no. 4 (2025): 455. https://doi.org/10.3892/ol.2025.15201
Copy and paste a formatted citation
x
Spandidos Publications style
Luo Y, Luo J, Yang M and Zhao X: Mechanisms of T‑cell metabolic reprogramming in the microenvironment of acute myeloid leukemia and its therapeutic potential (Review). Oncol Lett 30: 455, 2025.
APA
Luo, Y., Luo, J., Yang, M., & Zhao, X. (2025). Mechanisms of T‑cell metabolic reprogramming in the microenvironment of acute myeloid leukemia and its therapeutic potential (Review). Oncology Letters, 30, 455. https://doi.org/10.3892/ol.2025.15201
MLA
Luo, Y., Luo, J., Yang, M., Zhao, X."Mechanisms of T‑cell metabolic reprogramming in the microenvironment of acute myeloid leukemia and its therapeutic potential (Review)". Oncology Letters 30.4 (2025): 455.
Chicago
Luo, Y., Luo, J., Yang, M., Zhao, X."Mechanisms of T‑cell metabolic reprogramming in the microenvironment of acute myeloid leukemia and its therapeutic potential (Review)". Oncology Letters 30, no. 4 (2025): 455. https://doi.org/10.3892/ol.2025.15201
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team