Open Access

Protein lactylation and immunotherapy in gliomas: A novel regulatory axis in tumor metabolism (Review)

  • Authors:
    • Tao Luo
    • Liang Liu
    • Hao Wang
    • Shuai Wen
  • View Affiliations

  • Published online on: June 18, 2025     https://doi.org/10.3892/ijo.2025.5764
  • Article Number: 58
  • Copyright: © Luo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gliomas are the most common primary brain tumors, and exhibit highly heterogeneous and aggressive biological behaviors. Metabolic reprogramming is a hallmark of gliomas, and lactate accumulation serves a critical role in tumor progression. In addition to its traditional role as a metabolic byproduct, lactate has been recognized as a signaling molecule that modifies proteins through lactylation, which is a novel post‑translational modification. Lactate‑induced lactylation of histone and non‑histone proteins is emerging as a key epigenetic and metabolic regulator that influences glioma development, immune evasion, angiogenesis and therapeutic resistance. The present review provides mechanistic insights into protein lactylation, its role in glioma progression and its potential therapeutic implications. Targeting lactate metabolism and lactylation‑modifying enzymes holds promise for improving glioma treatment outcomes.

Introduction

Glioma is one of the most common types of brain tumors, accounting for ~30% of all brain tumors and 80% of all malignant brain tumors (1). There are several types of gliomas, including astrocytomas, oligodendrogliomas and ependymomas. Low-grade gliomas (LGGs) grow slowly, whereas high-grade gliomas, such as glioblastoma multiforme (GBM), are highly aggressive (2,3). LGGs generally have a more favorable prognosis, whereas GBMs have a 5-year survival rate of <7% (4,5). Common symptoms vary depending on the tumor location and may include headaches, seizures, memory loss, personality changes, nausea and disrupted vision (6). Standard treatments for gliomas include surgery, radiation therapy and chemotherapy (7). Additionally, immunotherapy and targeted therapies are emerging as approaches for glioma treatment (8-10).

Metabolic adaptations, particularly the Warburg effect, facilitate rapid tumor growth by shifting cellular energy production toward glycolysis, even in the presence of oxygen (11). This metabolic shift leads to the excessive accumulation of lactate, which has traditionally been considered to be a metabolic waste product (12,13). However, recent studies have highlighted the role of lactate as a crucial signaling molecule that is capable of modifying proteins through lactylation, which is an epigenetic and metabolic regulatory mechanism that impacts tumor behavior (14,15). Lactylation, which is a type of post-translational modification (PTM) that was first identified as a histone modification, influences gene expression and immune cell differentiation (16-19). In addition to histone lactylation, non-histone protein lactylation has been implicated in various cellular processes, including tumor cell proliferation, immune suppression, angiogenesis and therapy resistance (20-22).

Lactylation has been demonstrated to serve critical roles in glioma development and progression (23). Lactylation-related genes that were identified using single-cell RNA-sequencing data revealed two distinct molecular subtypes, forming a prognostic signature that classified patients with glioma into high-score and low-score groups (24). Notably, high-score patients exhibited greater immune infiltration, elevated expression of chemokines and immune checkpoints, and poorer overall survival but an enhanced response to immunotherapy compared with low-score patients. This classification provides a valuable tool to predict survival outcomes and optimize treatment strategies (24). A pan-cancer multi-omics analysis of lactylation-associated genes has revealed that the prognostic significance of the lactylation score differs across tumor types. The lactylation score is a protective factor in LGG, kidney renal clear cell carcinoma, adrenocortical carcinoma, rectum adenocarcinoma and uveal melanoma (25). Higher lactylation scores are associated with a cold tumor microenvironment (TME), reduced immune cell infiltration and poorer responses to immunotherapy (25). The present review investigates the mechanisms and functional significance of lactate-induced protein lactylation in gliomas, emphasizing its implications for tumor progression and potential therapeutic interventions. The functions of protein lactylation in non-glioma cancer types are also described, indicating that protein lactylation occurs not only in gliomas but also serves a role in the development and progression of other tumors.

Mechanisms of protein lactylation

Lactate production and accumulation in cancer cells

Under normal conditions, cells generate ATP through glycolysis and mitochondrial oxidative phosphorylation (26,27). During these processes, glycolysis converts glucose into pyruvate, which then enters the tricarboxylic acid (TCA) cycle in mitochondria for energy production. However, a number of cancer cells preferentially rely on aerobic glycolysis, which is a phenomenon known as the Warburg effect (28,29). In cancer cells, most pyruvate is converted into lactate rather than entering the TCA cycle. This metabolic shift is influenced by multiple factors, including hypoxia, hypoxia-inducible factor 1-α (HIF-1α) activation, oncogene activation and mitochondrial dysfunction (30). HIF-1α is frequently upregulated in tumor cells and enhances the expression of key glycolytic enzymes, such as hexokinase, phosphofructokinase and pyruvate kinase M2 (PKM2), and it promotes the expression of lactate dehydrogenase A (LDHA) (31,32).

LDHA catalyzes the conversion of pyruvate into lactate (33). Monocarboxylate transporter (MCT)1 and MCT4 regulate lactate flux: MCT1 facilitates the import of lactate into cells, whereas MCT4 exports lactate from highly glycolytic cells, preventing intracellular acidification (34,35). Numerous oncoproteins such as c-Myc, Akt and KRAS promote glycolysis and lactate production, reinforcing the metabolic reprogramming of cancer cells (36,37). Additionally, mitochondrial dysfunction can shift metabolism toward glycolysis, leading to increased lactate accumulation (38,39). In recent years, lactate has been recognized not only as a metabolic byproduct but also as a key signaling molecule (40). Lactate serves as a substrate for protein lactylation, which modifies gene expression and promotes tumor progression (40-42).

Biochemical basis of protein lactylation

Protein lactylation is a type of PTM that involves the covalent attachment of lactate-derived acyl groups to lysine residues on histone or other proteins (43). This process is medicated by specific enzymes that regulate the addition and removal of lactyl groups. Several key enzymes, including histone lactyltransferases [p300 and lysine acetyltransferase 2A (KAT2A)], acyl-CoA synthetase short chain family member 2 (ACSS2), LDHs and histone deacetylases (HDACs), have been demonstrated to contribute to protein lactylation (44-46). p300, which is primarily known as a histone acetyltransferase, also mediates histone lactylation by transferring the lactyl group from lactyl-CoA to lysine residues on histones (47). KAT2A, which is also known as general control non-depressible 5, has been reported to exhibit lactyltransferase activity. ACSS2 converts lactate into lactyl-CoA, which serves as a donor molecule for protein lactylation (45). KAT2A often works in conjunction with ACSS2, which produces lactyl-CoA from lactate, enabling KAT2A to perform its lactyltransferase function (45).

LDHA and LDHB catalyze the conversion of pyruvate to lactate, increasing intracellular lactate levels, which can provide the necessary substrate for lactyl-CoA synthesis (48). Sirtuin 1 (SIRT1) has been identified as a delactylase that is responsible for removing lactylation marks from histones (49). Additionally, HDAC2 and HDAC3 have been implicated in the removal of lactylation, further regulating this modification (50-52). Although chromobox protein homolog 3 (CBX3) is not a direct lactyltransferase, it binds to lactylated histones and facilitates transcriptional regulation through interactions with p300 (53). MCT1 and MCT4 regulate lactate transport into and out of the cell, which influences the availability of lactate for lactyl-CoA synthesis (54,55). Therefore, protein lactylation is a highly dynamic process that involves a complex interplay of multiple enzymes that work together to maintain a balance between lactylation and delactylation (56) (Fig. 1).

Histone and non-histone protein lactylation
Histone lactylation

Both histone and nonhistone proteins are modified, and these modifications serve important roles in gene expression, metabolism and cell signaling (57). For example, tumor-derived lactate induces bevacizumab resistance in colorectal cancer (CRC) by enhancing rubicon-like autophagy enhancer expression through histone H3 lysine 18 lactylation (H3K18la) (58). Similarly, H3K18la promotes cisplatin resistance in bladder cancer by enhancing glycolytic metabolism and activating the key transcription factors Y-box binding protein 1 (YBX1) and YY1, while targeted inhibition of H3K18la restores cisplatin sensitivity (59). Evodiamine suppresses semaphorin 3A-mediated angiogenesis and programmed death-ligand 1 (PD-L1) expression in prostate cancer by impairing HIF-1α histone lactylation and inducing ferroptosis (60). In CRC, G protein-coupled receptor 37 promotes glycolysis and histone lactylation through the Hippo pathway, promoting liver metastasis (61). Furthermore, the m5C methyltransferase NOP2/Sun RNA methyltransferase 2 (NSUN2) promotes CRC progression by reprogramming glucose metabolism through the NSUN2/YBX1/m5C-enolase 1 axis, whereas lactate accumulation enhances NSUN2 expression and its lactylation via H3K18la (62). In pancreatic cancer, H3K18la is highly expressed in tumor tissues, and is positively associated with the serum lactate, CA19-9 and CEA levels, demonstrating its potential for use as a novel biomarker for diagnosis and prognosis (63). Elevated H3K18la levels in pancreatic ductal adenocarcinoma (PDAC) are associated with poor prognosis and promote the transcription of TTK and BUB1 mitotic checkpoint serine/threonine kinase B, which enhance p300 expression and glycolysis (64). Additionally, TTK phosphorylates LDHA, leading to increased lactate production and H3K18la levels (64).

Downregulation of aldo-keto reductase family 1 member B10 reduces cell proliferation and alters the expression of key target genes, including Bcl-2, Bax, Pan histone lysine lactylation (Kla) and H3K18la, highlighting the role of histone lactylation in the pathogenesis of hepatocellular carcinoma (HCC) (65). In HCC, protein arginine methyltransferase 3 (PRMT3) facilitates tumor immunosuppression by upregulating PD-L1 expression through the PRMT3-pyruvate dehydrogenase kinase 1-lactate-H3K18la axis (66). Targeting this pathway with JX06 or anti-PD-L1 therapy effectively suppresses tumor growth and restores CD8+ T cell infiltration, suggesting a promising immunotherapeutic approach (66). In ovarian cancer, high histone H3K18la levels are associated with poor prognosis, advanced tumor stage, shorter platinum recurrence time and increased metastasis (67). Similarly, elevated H3K18la levels in gastric cancer are associated with poor prognosis (49). SIRT1, which acts as a histone delactylase, disrupts a positive feedback loop involving H19, glycolysis and H3K18la, suggesting a novel strategy for the treatment of gastric cancer (49). In breast cancer, elevated H3K18la in tumor tissues promotes peroxisome proliferator activated receptor δ expression, which increases AKT transcription and phosphorylation to support cell survival under anaerobic glycolytic conditions (52).

Nonhistone protein lactylation

Nonhistone protein lactylation has been reported in various cancer types, including PDAC, breast cancer and HCC (16). The p53 tumor suppressor protein is a key regulator of cell cycle arrest, apoptosis, DNA repair and senescence, and it functions as a critical guardian of genomic stability to prevent tumorigenesis (68). Recently, p53 has been identified to be lactylated. Specifically, alanyl-tRNA synthetase 1 acts as a lactate sensor in tumor cells, mediating global lysine lactylation, including the lactylation of p53, which impairs its function and promotes tumorigenesis (69). Notably, β-alanine disrupts this process and reduces cancer progression (69). In pancreatic cancer, RHOF promotes tumor progression by upregulating c-Myc, enhancing PKM2-mediated glycolysis and inducing Snail1 lactylation, which drives epithelial-mesenchymal transition and promotes tumor growth (70). Elevated lactylation in PDAC reshapes the TME and promotes immunotherapy resistance by inducing K63 lactylation of endosulfine α (ENSA) and activating STAT3-C-C motif chemokine ligand 2 (CCL2) signaling. Targeting the ENSA-K63la or CCL2 pathway enhances the efficacy of immune checkpoint blockade therapy (71).

In triple-negative breast cancer (TNBC), increased levels of the m6A modification via the HDAC2-mediated delactylation of METTL3 promote cisplatin resistance. Inhibition of HDAC2 by tucidinostat enhances cisplatin sensitivity (51). In cervical cancer, lactate-induced lactylation of DCBLD1 at K172 stabilizes its expression, promoting glucose-6-phosphate dehydrogenase activity and the subsequent pentose phosphate pathway activation, thereby facilitating cancer cell proliferation and metastasis (72). In HCC, lactylation at K28 inhibits adenylate kinase 2 function, thus promoting cell proliferation and metastasis by regulating cellular metabolism (73). Additionally, a study has identified lactylation of phosphofructokinase platelet at lysine 688 and aldolase A at lysine 147 in colon cancer cells (74). These studies highlight the metabolic impact of protein lactylation on cancer progression and treatment (70-73).

Functional roles of protein lactylation in gliomas

Protein lactylation has been identified to be a pivotal regulator of gene expression by promoting the transcription of genes that are involved in cell proliferation, angiogenesis, apoptosis, the cell cycle, invasion and drug resistance (20). This section discusses the functional roles of protein lactylation in gliomas (Fig. 2).

Influence on tumor cell proliferation

Numerous studies have shown that protein lactylation regulates cell proliferation in a wide range of human cancer types, including gliomas (75,76). Batsios et al (75) revealed a crucial role of lactate in nucleotide biosynthesis in pediatric diffuse midline gliomas (DMGs). The oncogenic H3K27M mutation (a mutation in the histone H3 gene where lysine 27 is replaced with methionine) upregulated phosphoglycerate kinase 1, leading to increased lactate production from glucose in DMGs. Lactate subsequently activated the nucleoside diphosphate kinase NME/NM23 nucleoside diphosphate kinase 1 (NME1) through lactylation, promoting the synthesis of nucleoside triphosphates, which are essential for tumor cell proliferation. Their study established a novel H3K27M-lactate-NME1 axis, suggesting potential clinical strategies for tumor monitoring and therapy evaluation (75). Dong et al (76) reported that lactylation of transcription factors, such as HIF-1α, augmented hypoxia-driven tumor progression. Hypoxia-induced HIF-1α upregulation enhanced glycolysis and promoted glioma progression by promoting BCL2 interacting protein 3 (BNIP3)-dependent mitophagy. Notably, H3K18la upregulated YTH N6-methyladenosine RNA binding protein F2 (YTHDF2) to sustain mitophagy. Conversely, YTHDF2 downregulation disrupted this process and inhibited tumor progression. Therefore, HIF-1α orchestrates metabolic reprogramming through BNIP3-dependent mitophagy, which is a process that is modulated by H3K18la-induced YTHDF2 expression, ultimately contributing to glioma cell proliferation and invasion (76).

Regulation of angiogenesis and invasion

Lactylation of proteins has been implicated in the regulation of cellular invasion and angiogenesis in GBM. For example, dexamethasone (Dex) inhibits GBM cell viability, migration, invasion and glycolysis (77). The oncogene C-Myc is highly expressed in GBM cells but its expression is reduced following Dex treatment (78). Dex suppresses c-Myc lactylation by inhibiting glycolysis, leading to decreased stability of the c-Myc protein. Notably, the administration of sodium lactate reverses the inhibitory effects of Dex, restoring GBM cell proliferation and invasion (77). These findings suggest that Dex modulates GBM progression by disrupting glycolysis-dependent c-Myc stabilization via the regulation of c-Myc lactylation, highlighting potential metabolic vulnerability that can be used in the treatment of GBM (77). Additionally, MAPK6P4 promotes vasculogenic mimicry (VM) in GBM by encoding the peptide P4-135aa, which phosphorylates KLF transcription factor 15, enhancing its stability and nuclear translocation to activate LDHA. LDHA, in turn, facilitates the lactylation of VEGFR2 and vascular endothelial cadherin, leading to their increased expression. Conversely, MAPK6P4 deficiency suppresses VM development, as confirmed in xenograft mouse models (79). VEGFR2 has been shown to serve an important role in tumor metastasis and angiogenesis through the regulation of multiple signaling pathways, including the PI3K, AKT and MAPK pathways (80,81). Therefore, VEGFR2 lactylation may be linked to tumor angiogenesis in GBM.

Regulation of cancer stem cells (CSCs)

CSCs exhibit stem-like properties, including self-renewal, differentiation potential and tumorigenic capacity (82). Extensive evidence suggests that glioma stem cells (GSCs) are the primary contributors to glioma progression, recurrence and drug resistance (83-85). The common biomarkers for GSCs include CD133, nextin, CD44, integrin a6, CD15, SOX2, aldehyde dehydrogenase 1 family member A3 (ALDH1A3), L1 cell adhesion molecule and A2B5 (86,87). Protein lactylation has been demonstrated to participate in the maintenance of CSCs in glioma. For example, the ALDH1A3-PKM2 interaction enhances lactate accumulation in glioblastoma stem cells, leading to X-ray repair cross complementing 1 (XRCC1) lactylation. This modification increases the affinity of XRCC1 for importin α, promoting its nuclear translocation and affecting DNA repair, thereby influencing tumor progression (88). Another study revealed that global lactylation levels were elevated in GSCs, with polypyrimidine tract binding protein 1 (PTBP1) hyperlactylation maintaining GSC self-renewal and promoting glioma progression. Specifically, lactylation of PTBP1 prevented its degradation via tripartite motif containing 21, enhanced its RNA-binding capacity, stabilized 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 mRNA and increased glycolysis. Conversely, SIRT1 counteracted this process by inducing PTBP1 delactylation, thereby suppressing tumor growth (89). Additionally, Warburg effect-induced H3 histone lactylation drives the expression of NF-κB-related LINC01127 in GBM, promoting self-renewal by regulating MAP4K4 expression and activating the JNK/NF-κB axis. Targeting the JNK/NF-κB pathway hinders tumor growth (90). Overall, these findings underscore the critical roles of protein lactylation in sustaining cancer stemness and driving glioma development and progression.

Regulation of therapy resistance

Drug resistance, which includes intrinsic resistance and acquired resistance, is a major obstacle in cancer therapy and contributes to poor prognosis (91). Intrinsic resistance refers to a pre-existing insensitivity of cancer cells to therapy, while acquired resistance develops after initial responsiveness through adaptive changes or mutations (92,93). Gliomas are resistant to standard therapies, including radiotherapy, chemotherapy and immunotherapy (94-96). Resistance to temozolomide (TMZ) remains a major challenge in glioma treatment (97). The mechanisms of drug resistance include efflux of drugs, drug inactivation, alteration of drug targets and epigenetic alterations (98,99). For example, upregulation of efflux transporters expels chemotherapeutic agents from GBM cells, reducing intracellular drug concentrations (100). Intercellular adhesion molecule-1 reduces TMZ sensitivity in gliomas with acquired resistance by promoting the membrane localization of ATP binding cassette subfamily B member 1 transporters (101). O-6-methylguanine-DNA methyltransferase (MGMT) repairs TMZ-induced O6-methylguanine DNA lesions, with its promoter hypermethylation predicting TMZ sensitivity, while MGMT genomic rearrangements drive chemotherapy resistance in gliomas (102). Mutations in tumor-derived protein tyrosine phosphatase receptor type K impair its functional activity and modify cell responsiveness to chemotherapeutic agents in glioma (103). USP6 N-terminal like (USP6NL), in coordination with the EGFR signaling pathway, modulates ubiquitin-dependent DNA repair mechanisms and contributes to TMZ resistance in GBM (104). TNF receptor associated factor 4 (TRAF4) sustains the deubiquitination of Caveolin-1, thereby enhancing glioblastoma stem cell characteristics and resistance to TMZ (105). Additionally, TMA exposure upregulates high mobility group box 1, which facilitates GSC formation through activation of the toll like receptor 2 (TLR2)/nuclear paraspeckle assembly transcript 1/Wnt signaling cascade (106). Therefore, various factors such as USP6NL, TRAF4 and TLR2 contribute to TMZ resistance in GBM (107).

Lactylation has been revealed to participate in the regulation of drug resistance in breast cancer, HCC and GBM (108-110). For instance, cancer-associated fibroblast (CAF)-derived lactate promotes doxorubicin (DOX) resistance in TNBC by enhancing histone lactylation-mediated ZFP64 expression. ZFP64 inhibits ferroptosis through the upregulation of GTP cyclohydrolase 1 and ferritin heavy chain 1, while targeting this pathway can restore DOX sensitivity (110). Similarly, USP34 promotes HCC progression and cisplatin resistance by regulating histone lactylation levels, and USP34 knockdown enhances cisplatin sensitivity (109). In GBM, the interaction between ALDH1A3 and PKM2 promotes PKM2 tetramerization and lactate accumulation. By profiling the lactylated proteome in GSCs with elevated lactate levels, lysine 247 of XRCC1 has been identified as a key lactylation site (88). This modification enhances the binding affinity between XRCC1 and importin α, thereby facilitating XRCC1 nuclear import and promoting more efficient DNA repair (88). Using high-throughput screening of a small-molecule compound library, D34-919 has been found to effectively disrupt the interaction between ALDH1A3 and PKM2, thereby inhibiting ALDH1A3-induced PKM2 tetramer formation. Treatment with D34-919 increases the susceptibility of GBM cells to apoptosis following chemoradiotherapy. Collectively, these results highlight ALDH1A3-driven PKM2 tetramerization as a promising target for enhancing chemoradiotherapy sensitivity in GBM (88).

Yue et al (111) reported that histone H3K9 lactylation (H3K9la) was increased in TMZ-resistant cells as well as in recurrent GBM tissues. Chronic TMZ exposure upregulated lactate and H3K9la levels in GBM cells and subcutaneous xenograft mice (111). Suppression of H3K9la using short hairpin RNAs targeting LDHA and LDHB increases TMZ sensitivity in GBM. Furthermore, stiripentol, an anti-epileptic drug, reduces H3K9la levels and leads to TMZ sensitivity in TMZ-resistant cells (111). Additionally, H3K9la promotes TMZ resistance by activating LUC7L2 transcription, which induces retention of intron 7 of mutL homolog 1 (MLH1), thereby reducing MLH1 expression and impairing mismatch repair in GBM (111). Additionally, Liu et al (112) analyzed the crystallographic structure of guanosine triphosphate-specific succinyl-CoA synthetase (GTPSCS) and L-lactate, and performed mutagenesis experiments. The authors revealed that GTPSCS translocated into the nucleus, interacted with p300 and specifically enhanced H3K18la. This nuclear translocation relied on a signal within the G1 subunit and acetylation at lysine 73 of the G2 subunit, which facilitated its binding to p300. GTPSCS and p300 cooperatively increased H3K18la, leading to promotion of growth differentiation factor 15 expression, thereby driving glioma cell proliferation and resistance to radiotherapy. The study identified GTPSCS as the first enzyme to catalyze lactyl-CoA synthesis for epigenetic regulation in glioma, which supports its role in therapy resistance (112).

Regulation of immune evasion and immunotherapy

Studies have shown that protein lactylation governs immune evasion and immunotherapy in human cancer (113-116). For example, serine and arginine rich splicing factor 10 (SRSF10) promotes immune evasion and anti-programmed cell death protein 1 (PD-1) resistance in HCC by driving a positive feedback loop that involves glycolysis, lactate production, H3K18la and M2 macrophage polarization (113). Pharmacological inhibition of SRSF10 with 1C8 enhances the efficacy of anti-PD-1 therapy, indicating that SRSF10 is a potential therapeutic target for overcoming immune resistance (113). Glycolysis-driven H3K18la upregulates B7 homolog 3 protein (B7-H3) expression via the cAMP responsive element binding protein 1/E1A binding protein P300 axis, promoting tumor immune evasion by inhibiting CD8+ T cell cytotoxicity. Notably, the inhibition of glycolysis and B7-H3 enhances antitumor immunity and improves the efficacy of anti-PD-1 therapy (116). In ovarian cancer, knockdown of LDHB suppresses cancer cell proliferation, reduces lactate production, enhances T cell-mediated immune activation and inhibits PD-L1 expression by decreasing H3K18la at the PD-L1 promoter, thereby reducing immune evasion (114). In cervical cancer, lactate that is secreted by tumor cells promotes M2 macrophage polarization by upregulating glycerol-3-phosphate dehydrogenase 2 expression through H3K18la-mediated histone lactylation, contributing to tumor progression (115). In bladder cancer, single-cell analysis has identified parkin RBR E3 ubiquitin protein ligase (PRKN) as a key regulator of mitophagy in M2 macrophages, with H3K18la-enhanced PRKN expression promoting M2 polarization, immune suppression and tumor progression (117). In gastric cancer, CAFs reduce the efficacy of PD-1/PD-L1 immunotherapy by secreting lysyl oxidase, activating TGF-β signaling, promoting insulin like growth factor 1 expression, enhancing glycolysis and inducing H3K18la-mediated PD-L1 transcription, thereby reinforcing immune evasion (118).

Lactate-induced protein lactylation has been identified to regulate the tumor immune microenvironment and influence immunotherapy strategies across various cancer types, including gliomas (119,120). H3K18la and H3K9la function as transcription initiators in CD8+ T cells, modulating their activity and antitumor immunity through metabolic and epigenetic modulation (121). For example, H3K18la promotes immune evasion in non-small cell lung cancer (NSCLC) by activating the POM121/MYC/PD-L1 pathway. However, metabolic reprogramming and immunotherapy can restore CD8+ T-cell cytotoxicity and enhance antitumor efficacy (122). In glioma, high-score cases based on lactylation-related genes exhibit greater immune infiltration, elevated checkpoint expression and poorer survival (123). In addition, lactate production in GBM induces histone lactylation, leading to CD47-dependent immune evasion by promoting an immunosuppressive transcriptional program. Targeting lactate production or inhibiting CBX3 enhances anti-CD47 therapeutic efficacy and tumor cell phagocytosis, providing a promising strategy to overcome immune resistance in GBM (53).

Hypoxia enhances the malignancy of glioma cells by promoting glycolysis and lactic acid accumulation (124). This lactate is absorbed by macrophages to induce M2 polarization through the MCT-1/H3K18la signaling pathway, which subsequently upregulates tumor necrosis factor superfamily member 9 expression and facilitates glioma progression (124). Chimeric antigen receptor (CAR) T-cell therapy is an advanced immunotherapy that harnesses the power of the immune system in patients to fight cancer (125,126). CAR-T cells recognize specific antigens on cancer cells and destroy cancer cells with high specificity, and this approach has shown success in the treatment of certain hematologic malignancies, such as B-cell acute lymphoblastic leukemia and diffuse large B-cell lymphoma, as well as some solid tumors (127,128). One group has revealed that oxamate enhanced the immune activation of tumor-infiltrating CAR-T cells in a GBM model by altering immune molecule phenotypes, increasing regulatory T cell infiltration, and reducing the histone H3K18la-mediated expression of CD39, CD73 and C-C motif chemokine receptor 8 (129). Another group has revealed that EGFR activation triggered ERK-mediated phosphorylation of ACSS2, leading to its nuclear translocation and interaction with KAT2A, enabling histone H3 lactylation. This process activated Wnt/β-catenin, NF-κB and PD-L1 expression, promoting brain tumor growth and immune evasion (45).

Therapeutic implications of targeting lactylation in gliomas

Inhibition of lactate production

Evidence suggests that targeting lactate metabolism with inhibitors of LDHA [7-benzyl-2,3-dihydroxy-6-methyl-4-propylnaphthalene-1-carboxylic acid (FX11)] and MCT1 (AZD3965) reduces lactate accumulation, thereby limiting lactylation-mediated tumor progression (130,131). For example, inhibition of LDHA by FX11 has been shown to suppress tumor progression in various cancer types, including lymphoma (132), pancreatic cancer (133,134), prostate cancer (135), osteosarcoma (136), neuroblastoma (137), gallbladder carcinoma (138), papillary thyroid carcinoma (139) and breast cancer (140). One study demonstrated that inhibition of LDHA by FX11 suppressed proliferation, migration and invasion, while promoting apoptosis in prostate cancer cells by reducing the Warburg effect, glucose consumption, lactate secretion, and MMP-9, PLAU and cathepsin B expression (131). AZD3965, which is a promising MCT1 inhibitor, exerts antitumor effects by inducing metabolic reprogramming (130); this represents a novel therapeutic approach in various cancer types, including small cell lung cancer (141,142), NSCLC (143), lymphoma (144,145), breast cancer (146) and bladder cancer (147). Additionally, AR-C155858 selectively inhibits MCT1 and MCT2, which are responsible for the transport of monocarboxylates, such as lactate, pyruvate and ketone bodies, across the cell membrane (148). One study has shown that AR-C155858 inhibited the migration and invasion of pancreatic cancer cells (149). Further research is needed to explore the therapeutic potential of FX11, AZD3965 and AR-C155858 in glioma.

Modulation of protein lactylation

Given that p300 is a key regulator of lactylation, inhibitors of p300 (such as A-485) may suppress the lactylation of both histone and nonhistone proteins, thereby altering glioma epigenetics and metabolism. Several inhibitors of p300 have been observed to have anticancer effects in various malignancies (150-153). For example, in neuroblastoma, the expression of cyclooxygenase-2, which is induced by enterovirus 71, is inhibited by pretreatment with the p300 inhibitor GR343 (150). In acute myeloid leukemia, the p300 inhibitor C646 selectively enhances apoptosis and triggers cell cycle arrest (154). Similarly, C646 suppresses human papillomavirus E6-E7 transcription, leading to p53 accumulation, inhibition of cell proliferation, disruption of glucose metabolism and induction of apoptosis in cervical cancer cells (151). Additionally, C646 effectively overcomes the resistance of resistant melanoma cells to BRAF inhibitors (153). The p300 inhibitor A-485 has been demonstrated to suppress cell proliferation, growth hormone secretion and tumor growth by downregulating key oncogenes and pathways, including Pttg1, c-Myc, cAMP and PI3K/AKT/mTOR, in growth hormone pituitary adenoma (152). Furthermore, A-485 induces proteasome-mediated degradation of SOX10 in melanoma cells, effectively inhibiting tumor cell proliferation (155).

In GBM, the CREB-binding protein/p300 inhibitor CPI-1612 suppresses H3K27Ac, targets c-MYC, synergizes with TMZ and penetrates into the brain, which makes it a promising therapeutic candidate (156). Other compounds have also been implicated in modulating lactylation-driven tumor progression. Tanshinone I inhibits ovarian cancer cell proliferation by blocking glycolysis, reducing histone H3K18la, downregulating tumor-associated gene expression (such as TTK, platelet-derived growth factor receptor β, YTHDF2 and rubicon-like autophagy enhancer), and alleviating the immunosuppressive TME (157). Additionally, in lung cancer, collagen triple helix repeat containing 1 (CTHRC1)+ CAFs promote EGFR-tyrosine kinase inhibitor resistance by enhancing glycolysis and creating a histone lactylation-driven positive feedback loop. Notably, gambogenic acid disrupts this loop to improve therapeutic efficacy, suggesting that CTHRC1+ CAFs are a predictive biomarker and therapeutic target in lung cancer (158).

Combination approaches with radiotherapy, chemotherapy and immunotherapy

A study has demonstrated that structural maintenance of chromosomes 4 serves a crucial role in CRC cell dormancy, where its attenuation enhances glycolysis, increases lactate production and upregulates ABC transporters via histone lactylation, ultimately leading to chemotherapy resistance (159). In HCC, histone lactylation has been linked to tumor prognosis and may serve as an independent biomarker (160). The genes nuclear receptor subfamily 6 group A member 1, oxysterol binding protein 2 and unc-119 lipid binding chaperone B are associated with prognosis, immunotherapy response and chemotherapy resistance in HCC (160). Similarly, high nijmegen breakage syndrome protein 1 lactylation predicts chemotherapy resistance, whereas reduction of lactate levels via LDHA inhibition impairs DNA repair and sensitizes tumors to therapy (161). In oral squamous cell carcinoma (OSCC), Kla-specific genes serve key roles in tumor progression, with BCAM, cingulin like 1, diacylglycerol kinase γ and oxidized low density lipoprotein receptor 1 serving as prognostic markers (162). Notably, BCAM promotes invasion, angiogenesis and chemotherapy resistance in OSCC (162). In ovarian cancer, malate enzyme 2 (ME2) promotes lactate production and chemotherapy resistance by converting glutamine-derived malate to pyruvate (163). In addition, chemotherapy-induced glucose reduction triggers ME2 acetylation by acetyl-CoA acetyltransferase 1, enhancing lactate production and the lactylation of homologous recombination repair proteins, leading to increased DNA repair capacity and drug resistance (163).

Given the critical role of lactylation in therapy resistance, strategies that target lactylation pathways may sensitize gliomas to radiation and TMZ by impairing DNA repair mechanisms and drug efflux pathways. Lactylation-related gene expression has been used to classify patients with GBM into high-score and low-score groups, with high-score patients exhibiting an improved response to immunotherapy (123). Notably, the antiepileptic drug stiripentol, which crosses the blood-brain barrier and inhibits LDHA/B activity, inhibits lactylation, sensitizing GBM cells to TMZ in both in vitro and in vivo models (111). Furthermore, blocking the ACSS2-KAT2A interaction combined with anti-PD-1 therapy enhances tumor inhibition in brain tumors (45). Furthermore, a lactate generation inhibitor has been revealed to reprogram CSC metabolism, reduce tumor immunosuppression and decrease CAR-regulatory T cell infiltration, potentially enhancing the efficacy of CAR-T-cell therapy in GBM (129).

Conclusion and future perspectives

Lactate-induced protein lactylation represents a novel regulatory mechanism with profound implications for glioma progression, immune suppression and therapy resistance (Fig. 2). Understanding the molecular landscape of lactylation in gliomas provides novel avenues for the development of therapeutic interventions. Several reviews have addressed lactylation in the context of glioma progression (164,165). Pienkowski et al (164) primarily focused on PTMs such as phosphorylation, methylation, acetylation, ubiquitination and glycosylation, but did not discuss lactylation in detail. Han et al (165) explored the role of protein lactylation in various brain diseases, including cerebrovascular disorders, neurodegenerative diseases, neuroinflammation, mental disorders and brain tumors; however, the role of lactylation in GBM could not be discussed in detail due to space limitations. Qiu et al (23) highlighted lactylation in GBM, emphasizing its role as a link between epigenetic plasticity and metabolic reprogramming. By contrast, the present review provides a comprehensive discussion on the functional roles of protein lactylation in GBM, including its involvement in proliferation, angiogenesis, invasion, therapeutic resistance, regulation of CSCs and immune evasion. Furthermore, we hypothesized that targeting lactate metabolism, lactylation-modifying enzymes and immune evasion mechanisms holds promise for improving glioma treatment outcomes. It is important to mention that several critical challenges must be addressed to fully elucidate the molecular mechanisms underlying the role of lactylation in glioma development and treatment. First, although p300 is known to catalyze lactylation, to the best of our knowledge, the enzymatic regulation of lactylation removal remains unclear. Identifying delactylases is critical for better understanding the dynamic lactylation in glioma. Characterizing lactyltransferase and delactylases will be pivotal for developing targeted pharmacological modulators of lactylation. Furthermore, the regulation of histone vs. non-histone protein lactylation in different glioma subtypes and stages requires further investigation. Second, the integration of lactylation with glioma metabolism and the TME requires comprehensive analysis. Given the hypoxic and lactate-rich nature of the TME in glioma, lactylation could act as a critical mediator of immunosuppression. Single-cell epigenomics ad spatial transcriptomics could elucidate how lactylation shapes cellular interactions in the glioma niche. Third, current inhibitors that target p300 or lactate metabolism lack specificity for lactylation, necessitating the development of small molecules that selectively inhibit protein lactylation to design more precise therapeutic strategies. Inhibitors of lactate production (LDHA inhibitors), lactylation writers (p300) or delactylase activators could synergize with chemotherapy or immune checkpoint blockade. Using patient-derived glioma models and organoids could be essential for preclinical validation of drugs targeting lactylation. Additionally, p300 has been identified as an acetyltransferase capable of catalyzing multiple types of protein modifications, including acetylation (166,167), propionylation (168), butyrylation (169,170) and lactylation (171-173) in oncogenesis. Targeting of lactylation by p300 inhibitors could also affect other PTMs in tumorigenesis. Fourth, identifying lactylation-specific biomarkers in gliomas could aid in early diagnosis, patient stratification and treatment response monitoring. In parallel, lactylation-associated gene signatures could be integrated into existing molecular classification frameworks to enhance predictive accuracy for therapy response. To the best of our knowledge, to date, no clinical trials specifically targeting lactylation have been reported. Therefore, clinical trials are essential to determine the feasibility and efficacy of targeting lactylation in patients with glioma in the future. Continued exploration of lactylation and its role in gliomas may pave the way for the development of innovative treatment strategies, ultimately improving patient outcomes and advancing the field of glioma therapy.

Availability of data and materials

Not applicable.

Authors' contributions

TL, LL, HW and SW were involved in writing the manuscript draft. Data authentication is not applicable. All authors have read and approved the final version of the 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.

Abbreviations:

ACSS2

acyl-CoA synthetase short chain family member 2

CAF

cancer-associated fibroblast

CAR

chimeric antigen receptor

CBX3

chromobox protein homolog 3

DMG

diffuse midline glioma

FX11

7-benzyl-2,3-dihydroxy-6-methyl-4-propylnaphthalene-1-carboxylic acid

GBM

glioblastoma multiforme

GSC

glioma stem cell

GTPSCS

guanosine triphosphate-specific succinyl-CoA synthetase

H3K9la

histone H3K9 lactylation

H3K18la

histone H3 lysine 18 lactylation

HIF-1α

hypoxia-inducible factor-1α

KAT2A

lysine acetyltransferase 2A

LDH

lactate dehydrogenase

MCT

monocarboxylate transporter

NSCLC

non-small cell lung cancer

PDAC

pancreatic ductal adenocarcinoma

PTM

post-translational modification

TCA

tricarboxylic acid

TME

tumor microenvironment

TMZ

temozolomide

Acknowledgements

Not applicable.

Funding

The present study was supported by the Hunan Provincial Health and Family Planning Commission Project (grant nos. 202204043722 and 20201100), the Hunan University of Chinese Medicine-Hospital Collaborative Research Grant Program (grant nos. 2024XYLH248 and 2024XYLH252), and the Natural Science Foundation of Changsha (grant no. kq2502355).

References

1 

Siegel RL, Kratzer TB, Giaquinto AN, Sung H and Jemal A: Cancer statistics, 2025. CA Cancer J Clin. 75:10–45. 2025. View Article : Google Scholar : PubMed/NCBI

2 

Yao L, Hatami M, Ma W and Skutella T: Vaccine-based immunotherapy and related preclinical models for glioma. Trends Mol Med. 30:965–981. 2024. View Article : Google Scholar : PubMed/NCBI

3 

Tariq R, Hussain N, Bajwa MH, Aziz HF, Shamim MS and Enam SA: Multicentric low-grade glioma: A systematic review of a rare neuro-oncological disease. Clin Neurol Neurosurg. 251:1088212025. View Article : Google Scholar : PubMed/NCBI

4 

Weller M, Wen PY, Chang SM, Dirven L, Lim M, Monje M and Reifenberger G: Glioma. Nat Rev Dis Primers. 10:332024. View Article : Google Scholar : PubMed/NCBI

5 

Awuah WA, Ben-Jaafar A, Roy S, Nkrumah-Boateng PA, Tan JK, Abdul-Rahman T and Atallah O: Predicting survival in malignant glioma using artificial intelligence. Eur J Med Res. 30:612025. View Article : Google Scholar : PubMed/NCBI

6 

Schaff LR and Mellinghoff IK: Glioblastoma and other primary brain malignancies in adults: A review. JAMA. 329:574–87. 2023. View Article : Google Scholar : PubMed/NCBI

7 

Kampers LFC, Metselaar DS, Vinci M, Scirocchi F, Veldhuijzen van Zanten S, Eyrich M, Biassoni V, Hulleman E, Karremann M, Stucker W and Van Gool SW: The complexity of malignant glioma treatment. Cancers (Basel). 17:8792025. View Article : Google Scholar : PubMed/NCBI

8 

Poorva P, Mast J, Cao B, Shah MV, Pollok KE and Shen J: Killing the killers: Natural killer cell therapy targeting glioma stem cells in high-grade glioma. Mol Ther. 33:2462–2478. 2025. View Article : Google Scholar : PubMed/NCBI

9 

Ma X, Sun C, Ding X, Xu J, Zhang Y, Deng T, Wang Y, Yang H, Ding R, Li H, et al: Mechanism analysis and targeted therapy of IDH gene mutation in glioma. Am J Cancer Res. 15:248–270. 2025. View Article : Google Scholar : PubMed/NCBI

10 

Gong G, Jiang L, Zhou J and Su Y: Advancements in targeted and immunotherapy strategies for glioma: Toward precision treatment. Front Immunol. 15:15370132024. View Article : Google Scholar

11 

Llibre A, Kucuk S, Gope A, Certo M and Mauro C: Lactate: A key regulator of the immune response. Immunity. 58:535–554. 2025. View Article : Google Scholar : PubMed/NCBI

12 

Papaneophytou C: The warburg effect: Is it always an enemy? Front Biosci (Landmark Ed). 29:4022024. View Article : Google Scholar : PubMed/NCBI

13 

Gong T, Wang QD, Loughran PA, Li YH, Scott MJ, Billiar TR, Liu YT and Fan J: Mechanism of lactic acidemia-promoted pulmonary endothelial cells death in sepsis: Role for CIRP-ZBP1-PANoptosis pathway. Mil Med Res. 11:712024.PubMed/NCBI

14 

Iozzo M, Pardella E, Giannoni E and Chiarugi P: The role of protein lactylation: A kaleidoscopic post-translational modification in cancer. Mol Cell. 85:1263–1279. 2025. View Article : Google Scholar : PubMed/NCBI

15 

Lv M, Huang Y, Chen Y and Ding K: Lactylation modification in cancer: Mechanisms, functions, and therapeutic strategies. Exp Hematol Oncol. 14:322025. View Article : Google Scholar : PubMed/NCBI

16 

Chen J, Huang Z, Chen Y, Tian H, Chai P, Shen Y, Yao Y, Xu S, Ge S and Jia R: Lactate and lactylation in cancer. Signal Transduct Target Ther. 10:382025. View Article : Google Scholar : PubMed/NCBI

17 

Zhao L, Qi H, Lv H, Liu W, Zhang R and Yang A: Lactylation in health and disease: Physiological or pathological? Theranostics. 15:1787–821. 2025. View Article : Google Scholar : PubMed/NCBI

18 

Li S, Dong L and Wang K: Current and future perspectives of lysine lactylation in cancer. Trends Cell Biol. 35:190–193. 2025. View Article : Google Scholar : PubMed/NCBI

19 

Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, Liu W, Kim S, Lee S, Perez-Neut M, et al: Metabolic regulation of gene expression by histone lactylation. Nature. 574:575–580. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Luo Y, Zhang N, Ye J, Wang Z, Zhou X, Liu J, Cai J, Li C and Chen L: Unveiling lactylation modification: A new hope for cancer treatment. Biomed Pharmacother. 184:1179342025. View Article : Google Scholar : PubMed/NCBI

21 

Sui Y, Shen Z, Wang Z, Feng J and Zhou G: Lactylation in cancer: Metabolic mechanism and therapeutic strategies. Cell Death Discov. 11:682025. View Article : Google Scholar : PubMed/NCBI

22 

Al-Malsi K, Xie S, Cai Y, Mohammed N, Xie K, Lan T and Wu H: The role of lactylation in tumor growth and cancer progression. Front Oncol. 15:15167852025. View Article : Google Scholar : PubMed/NCBI

23 

Qiu Q, Deng H, Song P, Liu Y and Zhang M: Lactylation in glioblastoma: A novel epigenetic modifier bridging epigenetic plasticity and metabolic reprogramming. Int J Mol Sci. 26:33682025. View Article : Google Scholar : PubMed/NCBI

24 

Lu X, Zhou Z, Qiu P and Xin T: Integrated single-cell and bulk RNA-sequencing data reveal molecular subtypes based on lactylation-related genes and prognosis and therapeutic response in glioma. Heliyon. 10:e307262024. View Article : Google Scholar : PubMed/NCBI

25 

Wu Z, Wu H, Dai Y, Wang Z, Han H, Shen Y, Zhang R and Wang X: A pan-cancer multi-omics analysis of lactylation genes associated with tumor microenvironment and cancer development. Heliyon. 10:e274652024. View Article : Google Scholar : PubMed/NCBI

26 

Khan T, Nagarajan M, Kang I, Wu C and Wangpaichitr M: Targeting metabolic vulnerabilities to combat drug resistance in cancer therapy. J Pers Med. 15:502025. View Article : Google Scholar : PubMed/NCBI

27 

Clay R, Li K and Jin L: Metabolic signaling in the tumor microenvironment. Cancers (Basel). 17:1552025. View Article : Google Scholar : PubMed/NCBI

28 

Barba I, Carrillo-Bosch L and Seoane J: Targeting the warburg effect in cancer: Where do we stand? Int J Mol Sci. 25:31422024. View Article : Google Scholar : PubMed/NCBI

29 

Liao M, Yao D, Wu L, Luo C, Wang Z, Zhang J and Liu B: Targeting the Warburg effect: A revisited perspective from molecular mechanisms to traditional and innovative therapeutic strategies in cancer. Acta Pharm Sin B. 14:953–1008. 2024. View Article : Google Scholar : PubMed/NCBI

30 

Zhang W, Xia M, Li J, Liu G, Sun Y, Chen X and Zhong J: Warburg effect and lactylation in cancer: Mechanisms for chemoresistance. Mol Med. 31:1462025. View Article : Google Scholar : PubMed/NCBI

31 

Upadhyay S, Khan S and Hassan MI: Exploring the diverse role of pyruvate kinase M2 in cancer: Navigating beyond glycolysis and the Warburg effect. Biochim Biophys Acta Rev Cancer. 1879:1890892024. View Article : Google Scholar : PubMed/NCBI

32 

Basheeruddin M and Qausain S: Hypoxia-Inducible Factor 1-Alpha (HIF-1alpha): An essential regulator in cellular metabolic control. Cureus. 16:e638522024.

33 

Sharma D, Singh M and Rani R: Role of LDH in tumor glycolysis: Regulation of LDHA by small molecules for cancer therapeutics. Semin Cancer Biol. 87:184–95. 2022. View Article : Google Scholar : PubMed/NCBI

34 

Liu T, Han S, Yao Y and Zhang G: Role of human monocarboxylate transporter 1 (hMCT1) and 4 (hMCT4) in tumor cells and the tumor microenvironment. Cancer Manag Res. 15:957–975. 2023. View Article : Google Scholar : PubMed/NCBI

35 

Singh M, Afonso J, Sharma D, Gupta R and Kumar V, Rani R, Baltazar F and Kumar V: Targeting monocarboxylate transporters (MCTs) in cancer: How close are we to the clinics? Semin Cancer Biol. 90:1–14. 2023. View Article : Google Scholar : PubMed/NCBI

36 

Yang L, Li S, Yu L, Leng J and Li N: Targeting glycolysis: Exploring a new frontier in glioblastoma therapy. Front Immunol. 15:15223922025. View Article : Google Scholar : PubMed/NCBI

37 

Paul S, Ghosh S and Kumar S: Tumor glycolysis, an essential sweet tooth of tumor cells. Semin Cancer Biol. 86:1216–1230. 2022. View Article : Google Scholar : PubMed/NCBI

38 

Sun F, Li W, Du R, Liu M, Cheng Y, Ma J and Yan S: Impact of glycolysis enzymes and metabolites in regulating DNA damage repair in tumorigenesis and therapy. Cell Commun Signal. 23:442025. View Article : Google Scholar : PubMed/NCBI

39 

Kooshan Z, Cardenas-Piedra L, Clements J and Batra J: Glycolysis, the sweet appetite of the tumor microenvironment. Cancer Lett. 600:2171562024. View Article : Google Scholar : PubMed/NCBI

40 

Wan L, Zhang H, Liu J, He Q, Zhao J, Pan C, Zheng K and Tang Y: Lactylation and human disease. Expert Rev Mol Med. 27:e102025. View Article : Google Scholar : PubMed/NCBI

41 

Yang Y, Luo N, Gong Z, Zhou W, Ku Y and Chen Y: Lactate and lysine lactylation of histone regulate transcription in cancer. Heliyon. 10:e384262024. View Article : Google Scholar : PubMed/NCBI

42 

Zhu Y, Liu W, Luo Z, Xiao F and Sun B: New insights into the roles of lactylation in cancer. Front Pharmacol. 15:14126722024. View Article : Google Scholar : PubMed/NCBI

43 

Zhang W, Shan G, Bi G, Hu Z, Yi Y, Zeng D, Lin Z and Zhan C: Lactylation and regulated cell death. Biochim Biophys Acta Mol Cell Res. 1872:1199272025. View Article : Google Scholar : PubMed/NCBI

44 

Li H, Liu C, Li R, Zhou L, Ran Y, Yang Q, Huang H, Lu H, Song H, Yang B, et al: AARS1 and AARS2 sense L-lactate to regulate cGAS as global lysine lactyltransferases. Nature. 634:1229–1237. 2024. View Article : Google Scholar : PubMed/NCBI

45 

Zhu R, Ye X, Lu X, Xiao L, Yuan M, Zhao H, Guo D, Meng Y, Han H, Luo S, et al: ACSS2 acts as a lactyl-CoA synthetase and couples KAT2A to function as a lactyltransferase for histone lactylation and tumor immune evasion. Cell Metab. 37:361–376 e7. 2025. View Article : Google Scholar

46 

Ju J, Zhang H, Lin M, Yan Z, An L, Cao Z, Geng D, Yue J, Tang Y, Tian L, et al: The alanyl-tRNA synthetase AARS1 moonlights as a lactyltransferase to promote YAP signaling in gastric cancer. J Clin Invest. 134:e1745872024. View Article : Google Scholar : PubMed/NCBI

47 

Tsusaka T, Najar MA, Sharma I, Marcinkiewicz MM, Crispim CVDS, Snyder NW, Burslem GM and Goldberg EL: Class I histone deacetylases catalyze lysine lactylation. bioRxiv [Preprint] 2025.02.25.640220. 2025.

48 

Urbanska K and Orzechowski A: Unappreciated Role of LDHA and LDHB to control apoptosis and autophagy in tumor cells. Int J Mol Sci. 20:20852019. View Article : Google Scholar : PubMed/NCBI

49 

Tsukihara S, Akiyama Y, Shimada S, Hatano M, Igarashi Y, Taniai T, Tanji Y, Kodera K, Yasukawa K, Umeura K, et al: Delactylase effects of SIRT1 on a positive feedback loop involving the H19-glycolysis-histone lactylation in gastric cancer. Oncogene. 44:724–738. 2025. View Article : Google Scholar

50 

Zou Y, Cao M, Tai M, Zhou H, Tao L, Wu S, Yang K, Zhang Y, Ge Y, Wang H, et al: A feedback loop driven by H4K12 lactylation and HDAC3 in macrophages regulates lactate-induced collagen synthesis in fibroblasts via the TGF-beta signaling. Adv Sci (Weinh). 12:e24114082025. View Article : Google Scholar

51 

He X, Li Y, Li J, Li Y, Chen S, Yan X, Xie Z, Du J, Chen G, Song J and Mei Q: HDAC2-Mediated METTL3 delactylation promotes DNA damage repair and chemotherapy resistance in triple-negative breast cancer. Adv Sci (Weinh). 12:e24131212025. View Article : Google Scholar : PubMed/NCBI

52 

Xu Y, Meng W, Dai Y, Xu L, Ding N, Zhang J and Zhuang X: Anaerobic metabolism promotes breast cancer survival via Histone-3 Lysine-18 lactylation mediating PPARD axis. Cell Death Discov. 11:542025. View Article : Google Scholar : PubMed/NCBI

53 

Wang S, Huang T, Wu Q, Yuan H, Wu X, Yuan F, Duan T, Taori S, Zhao Y, Snyder NW, et al: Lactate reprograms glioblastoma immunity through CBX3-regulated histone lactylation. J Clin Invest. 134:e1768512024. View Article : Google Scholar : PubMed/NCBI

54 

Fan M, Yang K, Wang X, Chen L, Gill PS, Ha T, Liu L, Lewis NH, Williams DL and Li C: Lactate promotes endothelial-to-mesenchymal transition via Snail1 lactylation after myocardial infarction. Sci Adv. 9:eadc94652023. View Article : Google Scholar : PubMed/NCBI

55 

Zhang T, Chen L, Kueth G, Shao E, Wang X, Ha T, Williams DL, Li C, Fan M and Yang K: Lactate's impact on immune cells in sepsis: Unraveling the complex interplay. Front Immunol. 15:14834002024. View Article : Google Scholar : PubMed/NCBI

56 

Zhang X, Liang C, Wu C, Wan S and Xu L, Wang S, Wang J, Huang X and Xu L: A rising star involved in tumour immunity: Lactylation. J Cell Mol Med. 28:e701462024. View Article : Google Scholar : PubMed/NCBI

57 

Shi P, Ma Y and Zhang S: Non-histone lactylation: Unveiling its functional significance. Front Cell Dev Biol. 13:15356112025. View Article : Google Scholar : PubMed/NCBI

58 

Li W, Zhou C, Yu L, Hou Z, Liu H, Kong L, Xu Y, He J, Lan J, Ou Q, et al: Tumor-derived lactate promotes resistance to bevacizumab treatment by facilitating autophagy enhancer protein RUBCNL expression through histone H3 lysine 18 lactylation (H3K18la) in colorectal cancer. Autophagy. 20:114–130. 2024. View Article : Google Scholar :

59 

Li F, Zhang H, Huang Y, Li D, Zheng Z, Xie K, Cao C, Wang Q, Zhao X, Huang Z, et al: Single-cell transcriptome analysis reveals the association between histone lactylation and cisplatin resistance in bladder cancer. Drug Resist Updat. 73:1010592024. View Article : Google Scholar : PubMed/NCBI

60 

Yu Y, Huang X, Liang C and Zhang P: Evodiamine impairs HIF1A histone lactylation to inhibit Sema3A-mediated angiogenesis and PD-L1 by inducing ferroptosis in prostate cancer. Eur J Pharmacol. 957:1760072023. View Article : Google Scholar : PubMed/NCBI

61 

Zhou J, Xu W, Wu Y, Wang M, Zhang N, Wang L, Feng Y, Zhang T, Wang L and Mao A: GPR37 promotes colorectal cancer liver metastases by enhancing the glycolysis and histone lactylation via Hippo pathway. Oncogene. 42:3319–30. 2023. View Article : Google Scholar : PubMed/NCBI

62 

Chen B, Deng Y, Hong Y, Fan L, Zhai X, Hu H, Yin S, Chen Q, Xie X, Ren X, et al: Metabolic recoding of NSUN2-mediated m(5)C modification promotes the progression of colorectal cancer via the NSUN2/YBX1/m(5)C-ENO1 positive feedback loop. Adv Sci (Weinh). 11:e23098402024. View Article : Google Scholar : PubMed/NCBI

63 

Hou J, Guo M, Li Y and Liao Y: Lactylated histone H3K18 as a potential biomarker for the diagnosis and prediction of the severity of pancreatic cancer. Clinics (Sao Paulo). 80:1005442024. View Article : Google Scholar : PubMed/NCBI

64 

Li F, Si W, Xia L, Yin D, Wei T, Tao M, Cui X, Yang J, Hong T and Wei R: Positive feedback regulation between glycolysis and histone lactylation drives oncogenesis in pancreatic ductal adenocarcinoma. Mol Cancer. 23:902024. View Article : Google Scholar : PubMed/NCBI

65 

Yu Y, Li Y, Zhou L, Cheng X and Gong Z: Hepatic stellate cells promote hepatocellular carcinoma development by regulating histone lactylation: Novel insights from single-cell RNA sequencing and spatial transcriptomics analyses. Cancer Lett. 604:2172432024. View Article : Google Scholar : PubMed/NCBI

66 

Ding CH, Yan FZ, Xu BN, Qian H, Hong XL, Liu SQ, Luo YY, Wu SH, Cai LY, Zhang X, et al: PRMT3 drives PD-L1-mediated immune escape through activating PDHK1-regulated glycolysis in hepatocellular carcinoma. Cell Death Dis. 16:1582025. View Article : Google Scholar : PubMed/NCBI

67 

Chao J, Chen GD, Huang ST, Gu H, Liu YY, Luo Y, Lin Z, Chen ZZ, Li X, Zhang B, et al: High histone H3K18 lactylation level is correlated with poor prognosis in epithelial ovarian cancer. Neoplasma. 71:319–332. 2024. View Article : Google Scholar : PubMed/NCBI

68 

Huang Y, Che X, Wang PW and Qu X: p53/MDM2 signaling pathway in aging, senescence and tumorigenesis. Semin Cancer Biol. 101:44–57. 2024. View Article : Google Scholar : PubMed/NCBI

69 

Zong Z, Xie F, Wang S, Wu X, Zhang Z, Yang B and Zhou F: Alanyl-tRNA synthetase, AARS1, is a lactate sensor and lactyltransferase that lactylates p53 and contributes to tumorigenesis. Cell. 187:2375–2392 e33. 2024. View Article : Google Scholar : PubMed/NCBI

70 

Zhao R, Yi Y, Liu H, Xu J, Chen S, Wu D, Wang L and Li F: RHOF promotes Snail1 lactylation by enhancing PKM2-mediated glycolysis to induce pancreatic cancer cell endothelial-mesenchymal transition. Cancer Metab. 12:322024. View Article : Google Scholar : PubMed/NCBI

71 

Sun K, Zhang X, Shi J, Huang J, Wang S, Li X, Lin H, Zhao D, Ye M, Zhang S, et al: Elevated protein lactylation promotes immunosuppressive microenvironment and therapeutic resistance in pancreatic ductal adenocarcinoma. J Clin Invest. 135:e1870242025. View Article : Google Scholar : PubMed/NCBI

72 

Meng Q, Sun H, Zhang Y, Yang X, Hao S, Liu B, Zhou H, Xu ZX and Wang Y: Lactylation stabilizes DCBLD1 activating the pentose phosphate pathway to promote cervical cancer progression. J Exp Clin Cancer Res. 43:362024. View Article : Google Scholar : PubMed/NCBI

73 

Yang Z, Yan C, Ma J, Peng P, Ren X, Cai S, Shen X, Wu Y, Zhang S, Wang X, et al: Lactylome analysis suggests lactylation-dependent mechanisms of metabolic adaptation in hepatocellular carcinoma. Nat Metab. 5:61–79. 2023. View Article : Google Scholar : PubMed/NCBI

74 

Cheng Z, Huang H, Li M and Chen Y: Proteomic analysis identifies PFKP lactylation in SW480 colon cancer cells. iScience. 27:1086452023. View Article : Google Scholar : PubMed/NCBI

75 

Batsios G, Taglang C, Udutha S, Gillespie AM, Robinson SP, Phoenix T, Mueller S, Venneti S, Koschmann C and Viswanath P: Lactylation fuels nucleotide biosynthesis and facilitates deuterium metabolic imaging of tumor proliferation in H3K27M-mutant gliomas. bioRxiv. Jan 3–2025.Epub ahead of print.

76 

Dong F, Yin H and Zheng Z: Hypoxia-inducible factor-1alpha regulates BNIP3-dependent mitophagy and mediates metabolic reprogramming through histone lysine lactylation modification to affect glioma proliferation and invasion. J Biochem Mol Toxicol. 39:e700692025. View Article : Google Scholar

77 

Zhu J and Zhang Y: Dexmedetomidine inhibits the migration, invasion, and glycolysis of glioblastoma cells by lactylation of c-myc. Neurol Res. 46:1105–1112. 2024. View Article : Google Scholar : PubMed/NCBI

78 

Trejo-Solis C, Castillo-Rodriguez RA, Serrano-Garcia N, Silva-Adaya D, Vargas-Cruz S, Chavez-Cortez EG, Gallardo-Perez JC, Zavala-Vega S, Cruz-Salgado A and Magana-Maldonado R: Metabolic roles of HIF1, c-Myc, and p53 in glioma cells. Metabolites. 14:2492024. View Article : Google Scholar : PubMed/NCBI

79 

Zhang M, Zhao Y, Liu X, Ruan X, Wang P, Liu L, Wang D, Dong W, Yang C and Xue Y: Pseudogene MAPK6P4-encoded functional peptide promotes glioblastoma vasculogenic mimicry development. Commun Biol. 6:10592023. View Article : Google Scholar : PubMed/NCBI

80 

Uba AI: Computer-aided design of VEGFR-2 inhibitors as anticancer agents: A review. J Mol Recognit. 38:e31042025. View Article : Google Scholar

81 

Mehta K, Hegde M, Girisa S, Vishwa R, Alqahtani MS, Abbas M, Shakibaei M, Sethi G and Kunnumakkara AB: Targeting RTKs/nRTKs as promising therapeutic strategies for the treatment of triple-negative breast cancer: Evidence from clinical trials. Mil Med Res. 11:762024.PubMed/NCBI

82 

Batlle E and Clevers H: Cancer stem cells revisited. Nat Med. 23:1124–1134. 2017. View Article : Google Scholar : PubMed/NCBI

83 

Agosti E, Zeppieri M, Ghidoni M, Ius T, Tel A, Fontanella MM and Panciani PP: Role of glioma stem cells in promoting tumor chemo- and radioresistance: A systematic review of potential targeted treatments. World J Stem Cells. 16:604–614. 2024. View Article : Google Scholar : PubMed/NCBI

84 

Friess D, Brauer S, Poysti A, Choudhury C and Harris L: Tools to study neural and glioma stem cell quiescence. Trends Neurosci. 47:736–748. 2024. View Article : Google Scholar : PubMed/NCBI

85 

Ramar V, Guo S, Hudson B and Liu M: Progress in glioma stem cell research. Cancers (Basel). 16:1022023. View Article : Google Scholar

86 

Agosti E, Antonietti S, Ius T, Fontanella MM, Zeppieri M and Panciani PP: Glioma stem cells as promoter of glioma progression: A systematic review of molecular pathways and targeted therapies. Int J Mol Sci. 25:79792024. View Article : Google Scholar : PubMed/NCBI

87 

Tang J, Amin MA and Campian JL: Glioblastoma stem cells at the nexus of tumor heterogeneity, immune evasion, and therapeutic resistance. Cells. 14:5622025. View Article : Google Scholar : PubMed/NCBI

88 

Li G, Wang D, Zhai Y, Pan C, Zhang J, Wang C, Huang R, Yu M, Li Y, Liu X, et al: Glycometabolic reprogramming-induced XRCC1 lactylation confers therapeutic resistance in ALDH1A3-overexpressing glioblastoma. Cell Metab. 36:1696–1710 e10. 2024. View Article : Google Scholar : PubMed/NCBI

89 

Zhou Z, Yin X, Sun H, Lu J, Li Y, Fan Y, Lv P, Han M, Wu J, Li S, et al: PTBP1 lactylation promotes glioma stem cell maintenance through PFKFB4-Driven glycolysis. Cancer Res. 85:739–757. 2025. View Article : Google Scholar

90 

Li L, Li Z, Meng X, Wang X, Song D, Liu Y, Xu T, Qin J, Sun N, Tian K, et al: Histone lactylation-derived LINC01127 promotes the self-renewal of glioblastoma stem cells via the cis-regulating the MAP4K4 to activate JNK pathway. Cancer Lett. 579:2164672023. View Article : Google Scholar : PubMed/NCBI

91 

Holohan C, Van Schaeybroeck S, Longley DB and Johnston PG: Cancer drug resistance: An evolving paradigm. Nat Rev Cancer. 13:714–726. 2013. View Article : Google Scholar : PubMed/NCBI

92 

Russo M, Chen M, Mariella E, Peng H, Rehman SK, Sancho E, Sogari A, Toh TS, Balaban NQ, Batlle E, et al: Cancer drug-tolerant persister cells: From biological questions to clinical opportunities. Nat Rev Cancer. 24:694–717. 2024. View Article : Google Scholar : PubMed/NCBI

93 

Kuczynski EA, Sargent DJ, Grothey A and Kerbel RS: Drug rechallenge and treatment beyond progression-implications for drug resistance. Nat Rev Clin Oncol. 10:571–587. 2013. View Article : Google Scholar : PubMed/NCBI

94 

Liu J, Yang F, Hu J and Zhang X: Nanoparticles for efficient drug delivery and drug resistance in glioma: New perspectives. CNS Neurosci Ther. 30:e147152024. View Article : Google Scholar : PubMed/NCBI

95 

Huo X, Li H, Xing Y, Liu W, Chen P, Du F, Song L, Yu Z, Cao X and Tian J: Two decades of progress in glioma methylation research: The rise of temozolomide resistance and immunotherapy insights. Front Neurosci. 18:14407562024. View Article : Google Scholar : PubMed/NCBI

96 

Chen T, Ma W, Wang X, Ye Q, Hou X, Wang Y, Jiang C, Meng X, Sun Y and Cai J: Insights of immune cell heterogeneity, tumor-initiated subtype transformation, drug resistance, treatment and detecting technologies in glioma microenvironment. J Adv Res. 72:527–554. 2025. View Article : Google Scholar :

97 

Davidson CL, Vengoji R, Jain M, Batra SK and Shonka N: Biological, diagnostic and therapeutic implications of exosomes in glioma. Cancer Lett. 582:2165922024. View Article : Google Scholar :

98 

Brown R, Curry E, Magnani L, Wilhelm-Benartzi CS and Borley J: Poised epigenetic states and acquired drug resistance in cancer. Nat Rev Cancer. 14:747–753. 2014. View Article : Google Scholar : PubMed/NCBI

99 

Shibue T and Weinberg RA: EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nat Rev Clin Oncol. 14:611–629. 2017. View Article : Google Scholar : PubMed/NCBI

100 

Munoz JL, Walker ND, Scotto KW and Rameshwar P: Temozolomide competes for P-glycoprotein and contributes to chemoresistance in glioblastoma cells. Cancer Lett. 367:69–75. 2015. View Article : Google Scholar : PubMed/NCBI

101 

Zhang X, Tan Y, Li T, Tan D, Fu B, Yang M, Chen Y, Cao M, Xuan C, Du Q, et al: Intercellular adhesion molecule-1 suppresses TMZ chemosensitivity in acquired TMZ-resistant gliomas by increasing assembly of ABCB1 on the membrane. Drug Resist Updat. 76:1011122024. View Article : Google Scholar : PubMed/NCBI

102 

Oldrini B, Vaquero-Siguero N, Mu Q, Kroon P, Zhang Y, Galan-Ganga M, Bao Z, Wang Z, Liu H, Sa JK, et al: MGMT genomic rearrangements contribute to chemotherapy resistance in gliomas. Nat Commun. 11:38832020. View Article : Google Scholar : PubMed/NCBI

103 

Agarwal S, Al-Keilani MS, Alqudah MA, Sibenaller ZA, Ryken TC and Assem M: Tumor derived mutations of protein tyrosine phosphatase receptor type K affect its function and alter sensitivity to chemotherapeutics in glioma. PLoS One. 8:e628522013. View Article : Google Scholar : PubMed/NCBI

104 

Su IC, Su YK, Chuang HY, Yadav VK, Setiawan SA, Fong IH, Yeh CT, Huang HC and Lin CM: Ubiquitin-specific protease 6 n-terminal-like protein (USP6NL) and the epidermal growth factor receptor (EGFR) signaling axis regulates ubiquitin-mediated DNA repair and temozolomide-resistance in glioblastoma. Biomedicines. 10:15312022. View Article : Google Scholar : PubMed/NCBI

105 

Li Y, Wang T, Wan Q, Wang Q, Chen Z, Gao Y, Ye Y, Lin J, Zhao B, Wang H, et al: TRAF4 maintains deubiquitination of caveolin-1 to drive glioblastoma stemness and temozolomide resistance. Cancer Res. 82:3573–3587. 2022. View Article : Google Scholar : PubMed/NCBI

106 

Gao XY, Zang J, Zheng MH, Zhang YF, Yue KY, Cao XL, Cao Y, Li XX, Han H, Jiang XF and Liang L: Temozolomide treatment induces HMGB1 to promote the formation of glioma stem cells via the TLR2/NEAT1/Wnt pathway in glioblastoma. Front Cell Dev Biol. 9:6208832021. View Article : Google Scholar : PubMed/NCBI

107 

Pu J, Yuan K, Tao J, Qin Y, Li Y, Fu J, Li Z, Zhou H, Tang Z, Li L, et al: Glioblastoma multiforme: An updated overview of temozolomide resistance mechanisms and strategies to overcome resistance. Discov Oncol. 16:7312025. View Article : Google Scholar : PubMed/NCBI

108 

Yu D, Zhong Q, Wang Y, Yin C, Bai M, Zhu J, Chen J, Li H and Hong W: Lactylation: The metabolic accomplice shaping cancer's response to radiotherapy and immunotherapy. Ageing Res Rev. 104:1026702025. View Article : Google Scholar : PubMed/NCBI

109 

Fan M, Liu JS, Wei XL, Nie Y and Liu HL: Histone lactylation-driven ubiquitin-specific protease 34 promotes cisplatin resistance in hepatocellular carcinoma. Gastroenterology Res. 18:23–30. 2025. View Article : Google Scholar : PubMed/NCBI

110 

Zhang K, Guo L, Li X, Hu Y and Luo N: Cancer-associated fibroblasts promote doxorubicin resistance in triple-negative breast cancer through enhancing ZFP64 histone lactylation to regulate ferroptosis. J Transl Med. 23:2472025. View Article : Google Scholar : PubMed/NCBI

111 

Yue Q, Wang Z, Shen Y, Lan Y, Zhong X, Luo X, Yang T, Zhang M, Zuo B, Zeng T, et al: Histone H3K9 lactylation confers temozolomide resistance in glioblastoma via LUC7L2-Mediated MLH1 intron retention. Adv Sci (Weinh). 11:e23092902024. View Article : Google Scholar : PubMed/NCBI

112 

Liu R, Ren X, Park YE, Feng H, Sheng X, Song X, AminiTabrizi R, Shah H, Li L, Zhang Y, et al: Nuclear GTPSCS functions as a lactyl-CoA synthetase to promote histone lactylation and gliomagenesis. Cell Metab. 37:377–394 e9. 2025. View Article : Google Scholar

113 

Cai J, Song L, Zhang F, Wu S, Zhu G, Zhang P, Chen S, Du J, Wang B, Cai Y, et al: Targeting SRSF10 might inhibit M2 macrophage polarization and potentiate anti-PD-1 therapy in hepatocellular carcinoma. Cancer Commun (Lond). 44:1231–1260. 2024. View Article : Google Scholar : PubMed/NCBI

114 

Hu X, Huang Z and Li L: LDHB mediates histone lactylation to activate PD-L1 and promote ovarian cancer immune escape. Cancer Invest. 43:70–79. 2025. View Article : Google Scholar

115 

Huang C, Xue L, Lin X, Shen Y and Wang X: Histone lactylation-driven GPD2 mediates M2 macrophage polarization to promote malignant transformation of cervical cancer progression. DNA Cell Biol. 43:605–618. 2024. View Article : Google Scholar : PubMed/NCBI

116 

Ma Z, Yang J, Jia W, Li L, Li Y, Hu J, Luo W, Li R, Ye D and Lan P: Histone lactylation-driven B7-H3 expression promotes tumor immune evasion. Theranostics. 15:2338–2359. 2025. View Article : Google Scholar : PubMed/NCBI

117 

Deng X, Huang Y, Zhang J, Chen Y, Jiang F, Zhang Z, Li T, Hou L, Tan W and Li F: Histone lactylation regulates PRKN-Mediated mitophagy to promote M2 macrophage polarization in bladder cancer. Int Immunopharmacol. 148:1141192025. View Article : Google Scholar : PubMed/NCBI

118 

Li Z, Liang P, Chen Z, Chen Z, Jin T, He F, Chen X and Yang K: CAF-secreted LOX promotes PD-L1 expression via histone lactylation and regulates tumor EMT through TGFβ/IGF1 signaling in gastric cancer. Cell Signal. 124:1114622024. View Article : Google Scholar

119 

Wang W, Wang H, Wang Q, Yu X and Ouyang L: Lactate-induced protein lactylation in cancer: Functions, biomarkers and immunotherapy strategies. Front Immunol. 15:15130472025. View Article : Google Scholar : PubMed/NCBI

120 

Hao ZN, Tan XP, Zhang Q, Li J, Xia R and Ma Z: Lactate and lactylation: Dual regulators of T-cell-mediated tumor immunity and immunotherapy. Biomolecules. 14:16462024. View Article : Google Scholar :

121 

Raychaudhuri D, Singh P, Chakraborty B, Hennessey M, Tannir AJ, Byregowda S, Natarajan SM, Trujillo-Ocampo A, Im JS and Goswami S: Histone lactylation drives CD8(+) T cell metabolism and function. Nat Immunol. 25:2140–2151. 2024. View Article : Google Scholar : PubMed/NCBI

122 

Zhang C, Zhou L, Zhang M, Du Y, Li C, Ren H and Zheng L: H3K18 lactylation potentiates immune escape of non-small cell lung cancer. Cancer Res. 84:3589–3601. 2024. View Article : Google Scholar : PubMed/NCBI

123 

De Leo A, Ugolini A, Yu X, Scirocchi F, Scocozza D, Peixoto B, Pace A, D'Angelo L, Liu JKC, Etame AB, et al: Glucose-driven histone lactylation promotes the immunosuppressive activity of monocyte-derived macrophages in glioblastoma. Immunity. 57:1105–1123 e8. 2024. View Article : Google Scholar : PubMed/NCBI

124 

Li M, Sun P, Tu B, Deng G, Li D and He W: Hypoxia conduces the glioma progression by inducing M2 macrophage polarization via elevating TNFSF9 level in a histone-lactylation-dependent manner. Am J Physiol Cell Physiol. 327:C487–C504. 2024. View Article : Google Scholar : PubMed/NCBI

125 

Mulvey A, Trueb L, Coukos G and Arber C: Novel strategies to manage CAR-T cell toxicity. Nat Rev Drug Discov. 24:379–397. 2025. View Article : Google Scholar : PubMed/NCBI

126 

Diorio C, Teachey DT and Grupp SA: Allogeneic chimeric antigen receptor cell therapies for cancer: Progress made and remaining roadblocks. Nat Rev Clin Oncol. 22:10–27. 2025. View Article : Google Scholar

127 

Tang L, Huang ZP, Mei H and Hu Y: Insights gained from single-cell analysis of chimeric antigen receptor T-cell immunotherapy in cancer. Mil Med Res. 10:522023.PubMed/NCBI

128 

Uslu U and June CH: Beyond the blood: Expanding CAR T cell therapy to solid tumors. Nat Biotechnol. 43:506–515. 2025. View Article : Google Scholar

129 

Sun T, Liu B, Li Y, Wu J, Cao Y, Yang S, Tan H, Cai L, Zhang S, Qi X, et al: Oxamate enhances the efficacy of CAR-T therapy against glioblastoma via suppressing ectonucleotidases and CCR8 lactylation. J Exp Clin Cancer Res. 42:2532023. View Article : Google Scholar : PubMed/NCBI

130 

Silva A, Antunes B, Batista A, Pinto-Ribeiro F, Baltazar F and Afonso J: In vivo anticancer activity of AZD3965: A systematic review. Molecules. 27:1812021. View Article : Google Scholar

131 

Xian ZY, Liu JM, Chen QK, Chen HZ, Ye CJ, Xue J, Yang HQ, Li JL, Liu XF and Kuang SJ: Inhibition of LDHA suppresses tumor progression in prostate cancer. Tumour Biol. 36:8093–8100. 2015. View Article : Google Scholar : PubMed/NCBI

132 

Le A, Cooper CR, Gouw AM, Dinavahi R, Maitra A, Deck LM, Royer RE, Vander Jagt DL, Semenza GL and Dang CV: Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc Natl Acad Sci USA. 107:2037–2042. 2010. View Article : Google Scholar : PubMed/NCBI

133 

Rajeshkumar NV, Dutta P, Yabuuchi S, de Wilde RF, Martinez GV, Le A, Kamphorst JJ, Rabinowitz JD, Jain SK, Hidalgo M, et al: Therapeutic targeting of the warburg effect in pancreatic cancer relies on an absence of p53 function. Cancer Res. 75:3355–3364. 2015. View Article : Google Scholar : PubMed/NCBI

134 

Mohammad GH, Vassileva V, Acedo P, Olde Damink SWM, Malago M, Dhar DK and Pereira SP: Targeting pyruvate kinase M2 and lactate dehydrogenase a is an effective combination strategy for the treatment of pancreatic cancer. Cancers (Basel). 11:13722019. View Article : Google Scholar : PubMed/NCBI

135 

Hao J, Graham P, Chang L, Ni J, Wasinger V, Beretov J, Deng J, Duan W, Bucci J, Malouf D, et al: Proteomic identification of the lactate dehydrogenase A in a radioresistant prostate cancer xenograft mouse model for improving radiotherapy. Oncotarget. 7:74269–74285. 2016. View Article : Google Scholar : PubMed/NCBI

136 

Gao S, Tu DN, Li H, Jiang JX, Cao X, You JB and Zhou XQ: Pharmacological or genetic inhibition of LDHA reverses tumor progression of pediatric osteosarcoma. Biomed Pharmacother. 81:388–393. 2016. View Article : Google Scholar : PubMed/NCBI

137 

Rellinger EJ, Craig BT, Alvarez AL, Dusek HL, Kim KW, Qiao J and Chung DH: FX11 inhibits aerobic glycolysis and growth of neuroblastoma cells. Surgery. 161:747–752. 2017. View Article : Google Scholar

138 

He Y, Chen X, Yu Y, Li J, Hu Q, Xue C, Chen J, Shen S, Luo Y, Ren F, et al: LDHA is a direct target of miR-30d-5p and contributes to aggressive progression of gallbladder carcinoma. Mol Carcinog. 57:772–783. 2018. View Article : Google Scholar : PubMed/NCBI

139 

Hou X, Shi X, Zhang W, Li D, Hu L, Yang J, Zhao J, Wei S, Wei X, Ruan X, et al: LDHA induces EMT gene transcription and regulates autophagy to promote the metastasis and tumorigenesis of papillary thyroid carcinoma. Cell Death Dis. 12:3472021. View Article : Google Scholar : PubMed/NCBI

140 

Jiang J, Roman J, Xu HN and Li LZ: An observation on enhanced extracellular acidification and lactate production induced by inhibition of lactate dehydrogenase A. Adv Exp Med Biol. 1269:163–167. 2021. View Article : Google Scholar : PubMed/NCBI

141 

Polanski R, Hodgkinson CL, Fusi A, Nonaka D, Priest L, Kelly P, Trapani F, Bishop PW, White A, Critchlow SE, et al: Activity of the monocarboxylate transporter 1 inhibitor AZD3965 in small cell lung cancer. Clin Cancer Res. 20:926–937. 2014. View Article : Google Scholar :

142 

Bola BM, Chadwick AL, Michopoulos F, Blount KG, Telfer BA, Williams KJ, Smith PD, Critchlow SE and Stratford IJ: Inhibition of monocarboxylate transporter-1 (MCT1) by AZD3965 enhances radiosensitivity by reducing lactate transport. Mol Cancer Ther. 13:2805–2816. 2014. View Article : Google Scholar : PubMed/NCBI

143 

Huang CY, Hsu LH, Chen CY, Chang GC, Chang HW, Hung YM, Liu KJ and Kao SH: Inhibition of alternative cancer cell metabolism of EGFR mutated non-small cell lung cancer serves as a potential therapeutic strategy. Cancers (Basel). 12:1812020. View Article : Google Scholar : PubMed/NCBI

144 

Beloueche-Babari M, Wantuch S, Casals Galobart T, Koniordou M, Parkes HG, Arunan V, Chung YL, Eykyn TR, Smith PD and Leach MO: MCT1 INHIBITOR AZD3965 increases mitochondrial metabolism, facilitating combination therapy and noninvasive magnetic resonance spectroscopy. Cancer Res. 77:5913–5924. 2017. View Article : Google Scholar : PubMed/NCBI

145 

Beloueche-Babari M, Casals Galobart T, Delgado-Goni T, Wantuch S, Parkes HG, Tandy D, Harker JA and Leach MO: Monocarboxylate transporter 1 blockade with AZD3965 inhibits lipid biosynthesis and increases tumour immune cell infiltration. Br J Cancer. 122:895–903. 2020. View Article : Google Scholar : PubMed/NCBI

146 

Benyahia Z, Blackman MCNM, Hamelin L, Zampieri LX, Capeloa T, Bedin ML, Vazeille T, Schakman O and Sonveaux P: In vitro and in vivo characterization of MCT1 inhibitor AZD3965 confirms preclinical safety compatible with breast cancer treatment. Cancers (Basel). 13:5692021. View Article : Google Scholar : PubMed/NCBI

147 

Silva A, Felix A, Cerqueira M, Goncalves CS, Sampaio-Marques B, Longatto-Filho A, Baltazar F and Afonso J: Effects of lactate transport inhibition by AZD3965 in muscle-invasive urothelial bladder cancer. Pharmaceutics. 15:26882023. View Article : Google Scholar : PubMed/NCBI

148 

Puri S and Juvale K: Monocarboxylate transporter 1 and 4 inhibitors as potential therapeutics for treating solid tumours: A review with structure-activity relationship insights. Eur J Med Chem. 199:1123932020. View Article : Google Scholar : PubMed/NCBI

149 

Kong SC, Nohr-Nielsen A, Zeeberg K, Reshkin SJ, Hoffmann EK, Novak I and Pedersen SF: Monocarboxylate transporters MCT1 and MCT4 regulate migration and invasion of pancreatic ductal adenocarcinoma cells. Pancreas. 45:1036–1047. 2016. View Article : Google Scholar : PubMed/NCBI

150 

Tung WH, Hsieh HL, Lee IT and Yang CM: Enterovirus 71 modulates a COX-2/PGE2/cAMP-dependent viral replication in human neuroblastoma cells: Role of the c-Src/EGFR/p42/p44 MAPK/CREB signaling pathway. J Cell Biochem. 112:559–570. 2011. View Article : Google Scholar : PubMed/NCBI

151 

He H, Lai Y, Hao Y, Liu Y, Zhang Z, Liu X, Guo C, Zhang M, Zhou H, Wang N, et al: Selective p300 inhibitor C646 inhibited HPV E6-E7 genes, altered glucose metabolism and induced apoptosis in cervical cancer cells. Eur J Pharmacol. 812:206–215. 2017. View Article : Google Scholar : PubMed/NCBI

152 

Ji C, Xu W, Ding H, Chen Z, Shi C, Han J, Yu L, Qiao N, Zhang Y, Cao X, et al: The p300 inhibitor A-485 exerts antitumor activity in growth hormone pituitary adenoma. J Clin Endocrinol Metab. 107:e2291–e2300. 2022. View Article : Google Scholar : PubMed/NCBI

153 

Zhang F, Tang X, Fan S, Liu X, Sun J, Ju C, Liang Y, Liu R, Zhou R, Yu B, et al: Targeting the p300/NONO axis sensitizes melanoma cells to BRAF inhibitors. Oncogene. 40:4137–4150. 2021. View Article : Google Scholar : PubMed/NCBI

154 

Gao XN, Lin J, Ning QY, Gao L, Yao YS, Zhou JH, Li YH, Wang LL and Yu L: A histone acetyltransferase p300 inhibitor C646 induces cell cycle arrest and apoptosis selectively in AML1-ETO-positive AML cells. PLoS One. 8:e554812013. View Article : Google Scholar : PubMed/NCBI

155 

Waddell A, Grbic N, Leibowitz K, Wyant WA, Choudhury S, Park K, Collard M, Cole PA and Alani RM: p300 KAT regulates SOX10 stability and function in human melanoma. Cancer Res Commun. 4:1894–1907. 2024. View Article : Google Scholar : PubMed/NCBI

156 

Mladek AC, Yan H, Tian S, Decker PA, Burgenske DM, Bakken K, Hu Z, He L, Connors MA, Carlson BL, et al: RBBP4-p300 axis modulates expression of genes essential for cell survival and is a potential target for therapy in glioblastoma. Neuro Oncol. 24:1261–1272. 2022. View Article : Google Scholar : PubMed/NCBI

157 

Jin Z, Yun L and Cheng P: Tanshinone I reprograms glycolysis metabolism to regulate histone H3 lysine 18 lactylation (H3K18la) and inhibits cancer cell growth in ovarian cancer. Int J Biol Macromol. 291:1390722025. View Article : Google Scholar

158 

Zhang C, Zhou W, Xu H, Xu J, Li J, Liu X, Lu X, Dai J, Jiang Y, Wang W, et al: Cancer-associated fibroblasts promote EGFR-TKI resistance via the CTHRC1/glycolysis/H3K18la positive feedback loop. Oncogene. 44:1400–1414. 2025. View Article : Google Scholar : PubMed/NCBI

159 

Sun X, He L, Liu H, Thorne RF, Zeng T, Liu L, Zhang B, He M, Huang Y, Li M, et al: The diapause-like colorectal cancer cells induced by SMC4 attenuation are characterized by low proliferation and chemotherapy insensitivity. Cell Metab. 35:1563–1579 e8. 2023. View Article : Google Scholar : PubMed/NCBI

160 

Wu Q, Li X, Long M, Xie X and Liu Q: Integrated analysis of histone lysine lactylation (Kla)-specific genes suggests that NR6A1, OSBP2 and UNC119B are novel therapeutic targets for hepatocellular carcinoma. Sci Rep. 13:186422023. View Article : Google Scholar : PubMed/NCBI

161 

Chen H, Li Y, Li H, Chen X, Fu H, Mao D, Chen W, Lan L, Wang C, Hu K, et al: NBS1 lactylation is required for efficient DNA repair and chemotherapy resistance. Nature. 631:663–669. 2024. View Article : Google Scholar : PubMed/NCBI

162 

Huang G, Chen S, He J, Li H, Ma Z, Lubamba GP, Wang L, Guo Z and Li C: Histone lysine lactylation (Kla)-induced BCAM promotes OSCC progression and Cis-platinum resistance. Oral Dis. 31:1116–1132. 2025. View Article : Google Scholar

163 

Zheng C, Tan H, Niu G, Huang X, Lu J, Chen S, Li H, Zhu J, Zhou Z, Xu M, et al: ACAT1-Mediated ME2 acetylation drives chemoresistance in ovarian cancer by linking glutaminolysis to lactate production. Adv Sci (Weinh). 12:e24164672025. View Article : Google Scholar : PubMed/NCBI

164 

Pienkowski T, Kowalczyk T, Cysewski D, Kretowski A and Ciborowski M: Glioma and post-translational modifications: A complex relationship. Biochim Biophys Acta Rev Cancer. 1878:1890092023. View Article : Google Scholar : PubMed/NCBI

165 

Han M, He W, Zhu W and Guo L: The role of protein lactylation in brain health and disease: Current advances and future directions. Cell Death Discov. 11:2132025. View Article : Google Scholar : PubMed/NCBI

166 

Wu X, Zhang X, Tang S and Wang Y: The important role of the histone acetyltransferases p300/CBP in cancer and the promising anticancer effects of p300/CBP inhibitors. Cell Biol Toxicol. 41:322025. View Article : Google Scholar : PubMed/NCBI

167 

Chen Q, Yang B, Liu X, Zhang XD, Zhang L and Liu T: Histone acetyltransferases CBP/p300 in tumorigenesis and CBP/p300 inhibitors as promising novel anticancer agents. Theranostics. 12:4935–4948. 2022. View Article : Google Scholar : PubMed/NCBI

168 

Liu B, Lin Y, Darwanto A, Song X, Xu G and Zhang K: Identification and characterization of propionylation at histone H3 lysine 23 in mammalian cells. J Biol Chem. 284:32288–32295. 2009. View Article : Google Scholar : PubMed/NCBI

169 

Chen Y, Sprung R, Tang Y, Ball H, Sangras B, Kim SC, Falck JR, Peng J, Gu W and Zhao Y: Lysine propionylation and butyrylation are novel post-translational modifications in histones. Mol Cell Proteomics. 6:812–819. 2007. View Article : Google Scholar : PubMed/NCBI

170 

Bhattacharya A, Chatterjee S, Bhaduri U, Singh AK, Vasudevan M, Sashidhara KV, Guha R, Nazir A, Rath SK, Natesh N and Kundu TK: Butyrylation meets adipogenesis-probed by a p300-catalyzed acylation-specific small molecule inhibitor: Implication in anti-obesity therapy. J Med Chem. 65:12273–12291. 2022. View Article : Google Scholar : PubMed/NCBI

171 

Jiang C, He X, Chen X, Huang J, Liu Y, Zhang J, Chen H, Sui X, Lv X, Zhao X, et al: Lactate accumulation drives hepatocellular carcinoma metastasis through facilitating tumor-derived exosome biogenesis by Rab7A lactylation. Cancer Lett. 627:2176362025. View Article : Google Scholar : PubMed/NCBI

172 

Deng J, Li Y, Yin L, Liu S, Li Y, Liao W, Mu L, Luo X and Qin J: Histone lactylation enhances GCLC expression and thus promotes chemoresistance of colorectal cancer stem cells through inhibiting ferroptosis. Cell Death Dis. 16:1932025. View Article : Google Scholar : PubMed/NCBI

173 

Dai J, Lu X, Zhang C, Qu T, Li W, Su J, Guo R, Yin D, Wu P, Han L and Zhang E: NNMT promotes acquired EGFR-TKI resistance by forming EGR1 and lactate-mediated double positive feedback loops in non-small cell lung cancer. Mol Cancer. 24:792025. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2025
Volume 67 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Luo T, Liu L, Wang H and Wen S: Protein lactylation and immunotherapy in gliomas: A novel regulatory axis in tumor metabolism (Review). Int J Oncol 67: 58, 2025.
APA
Luo, T., Liu, L., Wang, H., & Wen, S. (2025). Protein lactylation and immunotherapy in gliomas: A novel regulatory axis in tumor metabolism (Review). International Journal of Oncology, 67, 58. https://doi.org/10.3892/ijo.2025.5764
MLA
Luo, T., Liu, L., Wang, H., Wen, S."Protein lactylation and immunotherapy in gliomas: A novel regulatory axis in tumor metabolism (Review)". International Journal of Oncology 67.1 (2025): 58.
Chicago
Luo, T., Liu, L., Wang, H., Wen, S."Protein lactylation and immunotherapy in gliomas: A novel regulatory axis in tumor metabolism (Review)". International Journal of Oncology 67, no. 1 (2025): 58. https://doi.org/10.3892/ijo.2025.5764