Open Access

Potential of lactylation as a therapeutic target in cancer treatment (Review)

  • Authors:
    • Zhengfeng Zhu
    • Xinzhe Zheng
    • Pengfei Zhao
    • Cheng Chen
    • Gang Xu
    • Xixian Ke
  • View Affiliations

  • Published online on: February 7, 2025     https://doi.org/10.3892/mmr.2025.13456
  • Article Number: 91
  • Copyright: © Zhu 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

Post‑translational modifications (PTMs) of proteins influence their functionality by altering the structure of precursor proteins. These modifications are closely linked to tumor progression through the regulation of processes such as cell proliferation, apoptosis, angiogenesis and invasion. Tumors produce large amounts of lactic acid through aerobic glycolysis. Lactic acid not only serves an important role in cell metabolism, but also serves an important role in cell communication. Lactylation, a PTM involving lactate and lysine residues as substrates, serves as an epigenetic regulator that modulates intracellular signaling, gene expression and protein function, thereby serving a crucial role in tumorigenesis and progression. The identification of lactylation provides a key breakthrough in elucidating the interaction between tumor metabolic reprogramming and epigenetic modification. The present review primarily summarizes the occurrence of lactylation, its effect on tumor progression, drug resistance, the tumor microenvironment and gut microbiota, and its potential as a therapeutic target for cancer. The aim of the present review was to provide novel strategies for potential cancer therapies.

Introduction

It has been demonstrated that a number of proteins in the human body undergo post-translational modifications (PTMs) (1). With the advance of mass spectrometry techniques, an increasing array of PTMs has been identified, which can be classified into phosphorylation, glycosylation, SUMOylation, ubiquitination, acetylation and methylation, based on the covalent attachment of different small molecular groups to amino acid residues (2,3). These modifications induce alterations in the structure and function of proteins, enhancing the flexibility and diversity of protein functionalities through different types of PTMs (4). By regulating protein stability, localization and conformation, PTMs are involved in various biological processes, including signal transduction and gene expression regulation (5). Aberrant levels of PTMs have been closely linked to numerous diseases, including cancer (6). As such, PTMs hold considerable potential as biomarkers for disease and therapeutic targets (7).

Under normal physiological conditions, glucose within cells undergoes glycolysis and oxidative phosphorylation to produce substantial amounts of ATP. However, in contrast to normal cells, tumor cells preferentially generate energy through glycolysis, even in the presence of oxygen, to adapt to the hypoxic, acidic and nutrient-deprived tumor microenvironment (TME); a phenomenon known as the Warburg effect (8). This aerobic glycolysis in tumor cells results in the accumulation of lactate, which exacerbates the hypoxic conditions that fuel tumor growth, further intensifying lactate production and facilitating processes such as angiogenesis, invasion, metastasis and treatment resistance (9,10). Traditionally regarded merely as an energy substrate and metabolic byproduct, lactate is now recognized as a pivotal molecule linking cellular metabolism to the regulation of cellular activities (11). Furthermore, lactate serves as a metabolic substrate that drives PTMs of proteins, with lactylation, a lactate-induced modification, previously identified in histones (12). The excessive production of lactate sustains the hypoxic and acidic TME, promoting tumor angiogenesis, invasion, metastasis and resistance to therapies (13). On the other hand, lactate can influence tumorigenesis through epigenetic modifications, acting as a bridge between tumor metabolism and epigenetic regulation (14). This novel understanding of the role of lactate broadens potential avenues for cancer therapy.

Cancer-related diseases, as a leading global health burden, have sparked widespread interest in understanding the molecular mechanisms underlying tumorigenesis, progression and treatment (15). Epigenetic modifications have emerged as critical regulatory factors in cancer development (16). Lactylation, a novel form of epigenetic modification, serves a pivotal role in regulating various biological processes and influencing disease onset (17). For instance, Kla of Sox10 leads to macrophage-like vascular smooth muscle cell accumulation and increases vascular inflammation (18). Recent studies have identified a growing prevalence of lactylation modifications across multiple cancer types (19,20), positioning it as a key molecular mechanism in cancer progression (2123) Furthermore, research has shown that protein lactylation is a dynamic and reversible process (24), suggesting its potential as a promising therapeutic target for cancer treatment (25).

Lactylation, as an emerging form of modification, offers a promising therapeutic target for cancer treatment. A comprehensive understanding of the role lactylation serves in tumorigenesis and progression will provide profound insights for the development of novel cancer treatment strategies. This not only enhances the understanding of lactate metabolism but also paves the way for future drug identification by offering novel perspectives and insights into molecular mechanisms. The present review summarizes the research progress on lactylation modification in tumor development, examines the underlying mechanisms linking lactylation with cancer therapy resistance and the TME, and describes the potential crosstalk between lactylation and gut microbiota. Furthermore, it explores the therapeutic potential of targeting lactylation as a novel approach for cancer treatment, offering valuable insights for the development of innovative therapeutic strategies.

Regulation of lactylation modifications

The formation of lactylation is closely linked to the metabolic processes of tumors. Lactylation primarily arises from lactate produced during metabolism (26), with endogenous lactate generation serving as a critical determinant of lactylation levels. Under the influence of specific enzymes, lactate molecules covalently bind to lysine residues on proteins. Lactylation can be categorized into histone lactylation and non-histone lactylation, with histone lactylation serving a role in regulating gene expression, cellular signaling and metabolism (27) (Fig. 1). Currently, lactylation modifications have been observed in various proteins located in the nucleus, cytoplasm and mitochondria (28). The identification of lactylation has broadened the understanding of tumor metabolic reprogramming and epigenetic modifications.

The levels of protein lactylation are regulated by various factors, primarily by lactoyltransferases and de-lactoyltransferases. Lactoyltransferases and de-lactoyltransferases are responsible for the addition and removal of lactoyl groups, respectively, thereby influencing lactylation modifications. For instance, under the action of the acetyltransferase p300, lactoyl groups are transferred to lysine residues on histones, leading to covalent binding of lysine residues with lactate and resulting in lactylation modifications (29). Lactate transferase p300/cAMP-response element binding protein (CREB) binding protein (CBP) includes the cysteine-histidine rich 1, CREB binding kinase-inducible domain interacting, bromodomain, histone acetyltransferase, CH3 and steroid receptor coactivator interaction domains (30). Overexpression or depletion of the acetyltransferase p300 can increase or decrease the levels of histone Kla in cells (12).

Alanyl-transfer RNA (tRNA) synthetase (AARS) retains a conserved prototype structure throughout the entire biological process, including catalytic, tRNA recognition, editing and C-Ala domains (31). AARS1 acts as a lactoyltransferase by recognizing and catalyzing the activation of lactate molecules, subsequently facilitating the binding of lactate to lysine residues on target proteins to form lactylation modifications (32). AARS1 can also translocate into the nucleus, directly utilizing lactate as a lactoyl donor and ATP as an energy source to catalyze the lactylation of histones K90 of yes-associated protein and K108 of TEA domain transcription factor 1 (33).

As research progresses, more lactoyltransferases have been identified. For example, lysine acetyltransferase 8 (KAT8) mainly contains an acetyl-CoA binding site and a C2HC zinc finger motif. Through the KAT8-dependent pathway, it can catalyze the lactylation of non-histone proteins (34). KAT8 has also been found to serve as a lactoyltransferase catalyzing the lactylation of non-histone proteins (35). Researchers have employed anti-Kla antibodies to confirm the occurrence of lysine lactylation in Escherichia coli and identified YiaC as a potential lactoyltransferase (36). YiaC belongs to the GCN5-related N-acetyltransferase family (37) and it possesses lysine acetyltransferase activity while also catalyzing lysine lactylation. Structurally, it has a specific N-terminal domain, a conserved acetyltransferase domain, conserved histone acetyltransferase structural domains and a C-terminal bromodomain at the C-terminus (38). Furthermore, a study has revealed alternative pathways regulating lactylation modifications, such as macrophages absorbing extracellular lactate through monocarboxylate transporters, which promote the lactylation of high mobility group box 1 via a p300/CBP-dependent mechanism (39).

In the process of lactylation formation, lactoyltransferases serve a crucial role, while de-lactoyltransferases can reverse the lactylation modifications. For instance, histone deacetylases (HDACs) 1–3 have been found to dynamically and reversibly regulate histone L-lactylation (40). HDAC1-3 contain an N-terminal catalytic domain (41). A study has indicated that sirtuin 3 (SIRT3) can de-lactylate the lactylation at the H4K16la site (42). SIRT3 has a catalytic core region with a large and structurally homologous Rossmann folding domain characteristic of NAD-binding proteins and a zinc finger structure (43). Identifying the key regulatory enzymes involved in lactylation pathways and their substrate proteins will lay the groundwork for functional studies of lactylation modifications and their associated regulatory pathways.

However, little is currently known regarding how de-lactoyltransferases exert their reversible effects and transitions. Furthermore, the precise enzymes and mechanisms involved in the regulation of lactylation within cells remain largely unknown, highlighting the need for further research in the field of de-lactoyltransferases.

It is worth noting that histone acetylation and lactylation exhibit a high degree of distribution similarity in the genome (12) and acetyltransferases p300 and HDAC1-3 may regulate the acetylation levels of histones while modifying them with lactylation. On the one hand, it is possible to simultaneously increase the modification levels of both to regulate gene expression levels (39). On the other hand, influenced by the level of lactate metabolism, lactylation modification may competitively inhibit acetylation modification (44,45). This crosstalk mechanism between lactylation and acetylation is not yet fully understood and requires further in-depth exploration. In addition to enzyme-catalyzed lactylation, lactoyl glutathione, an intermediate metabolic product of glyceraldehyde during glycolysis, can directly modify lysine residues through a non-enzymatic reaction, resulting in lactylation (46). This non-enzymatic pathway for lactylation expands the routes through which lactylation can occur, enriching the understanding of cellular metabolism and further underscoring the significance of lactylation modifications in biological processes.

Influence of lactylation on tumor biological behavior

The hallmark features of tumors involve the dysregulation of critical pathways that maintain the dynamic balance of normal cells. These pathways include sustaining proliferative signaling, evading growth suppression, resisting cell death, achieving replicative immortality, inducing angiogenesis, and activating invasion and metastasis (47). Lactylation modifications exhibit differential expression across various tumors and serve a pivotal role in regulating intracellular signal transduction, thereby impacting tumor initiation and progression (48).

In non-small cell lung cancer (NSCLC), the lactylation of SOX9 has been reported to promote stemness, migration and invasion of NSCLC by enhancing glycolysis (49). Furthermore, a study has demonstrated that histone H3K18la enhanced immune evasion in NSCLC cells by activating the POM121 transmembrane nucleoporin/MYC/programmed death-ligand 1 axis (50). Hypoxia-induced lactylation of the serine hydroxymethyltransferase 2 protein stimulates glycolysis and stemness in esophageal cancer cells (51). In colorectal cancer, lactylation of β-catenin facilitates cancer cell proliferation via the Wnt signaling pathway (52).

Histone lactylation has also been implicated in ocular melanoma, where it enhances the recognition of RNA N6-methyladenosine modifications by YTH N6-methyladenosine RNA binding protein F2, promoting the degradation of period circadian regulator 1 and TP53 mRNA, ultimately fostering proliferation and migration of melanoma cells (53). In pancreatic cancer, lactylation of transcription factor EB enhances autophagy and lysosomal activity (54). Elevated lactylation of adenylate kinase 2 (AK2) in hepatocellular carcinoma has been linked to metabolic alterations, with lactylation at K28 inhibiting AK2 function, thereby promoting hepatocellular carcinoma cell proliferation and metastasis (55).

In clear cell renal cell carcinoma, histone lactylation stimulates tumor progression by activating the transcription of platelet-derived growth factor receptor β (56). In breast cancer, lactylation of histone lysines induces lactate dehydrogenase A expression, increasing glycolysis and lactate production, which drives tumor proliferation, invasion and metastasis (57). Furthermore, non-histone lactylation of METTL16 at K229 promotes copper ion deposition in gastric cancer (58). In bladder cancer, histone H3K18 lactylation serves a pro-oncogenic role by upregulating lipocalin 2 expression (59). Additionally, the Warburg effect, mediated by metabolite-driven histone lactylation, promotes the proliferation of anaplastic thyroid cancer cells harboring the BRAFV600E mutation (60). Lactylation offers novel insights for both the diagnosis and treatment of tumors. However, it is important to note that the role of lactylation may vary across different types of cancer.

Currently, lactylation sites detected in a number of tumors are primarily located on histone H3K18 (61,62), e.g., renal clear cell carcinoma (56) and bladder cancer (59), which may be attributed to the limitations of existing lactylation detection technologies (63). To broaden the scope of lactylation research across various modification sites, it is imperative to develop more diverse lactylation antibodies or advanced detection methods. Furthermore, research on non-histone protein lactylation is still in its infancy, warranting greater attention to the identification of lactylation sites in non-histone proteins. Histone and non-histone lactylation are both closely associated with tumor initiation and progression (Table I).

Table I.

Lactate modification for tumor progression.

Table I.

Lactate modification for tumor progression.

First author/s, yearCancer typeTarget componentBiological function(Refs.)
Yan, 2024NSCLCSOX9Facilitating migration and invasion(49)
Zhang, 2024NSCLCPOM121Facilitating immune escape(50)
Qiao, 2024Esophageal cancerSHMT2Facilitating glycolysis(51)
Miao, 2023Colorectal cancerβ-cateninPromoting the cell proliferation and stemness of colorectal cancer(52)
Yu, 2021Ocular melanomaYTHDF2Promoting cell proliferation(53)
Huang, 2024Pancreatic cancerTFEBElevating autophagy and lysosomal activity(54)
Yang, 2023HCCAK2Facilitating migration and invasion(55)
Yang, 2022ccRCCPDGFRβFacilitating migration and invasion(56)
Hou, 2024Breast cancerLDHAFacilitating proliferation, migration and invasion(57)
Sun, 2023Gastric cancerMETTL16Promoting cuproptosis(58)
Xie, 2023Bladder cancerLCN2Promoting tumor progression(59)
Wang, 2023Anaplastic thyroid cancerH4k12laPromoting tumor progression(60)

[i] ccRCC, clear cell renal cell carcinoma; HCC, hepatocellular carcinoma; NSCLC, non-small cell lung cancer.

As scientific advancements continue, it is expected that more lactylation modification sites will be identified, and targeting lactylation may provide novel biomarkers and therapeutic targets for clinical cancer diagnosis and treatment. Further investigation into lactylation will deepen the understanding of its role in cancer, offering a promising strategy for cancer therapy.

Effect of lactylation on tumor therapy resistance

Chemotherapy remains one of the primary treatments for human malignancies, yet chemotherapy resistance in cancer cells is a factor contributing to poor prognosis in a number of patients (64). Tumor metabolism, particularly the production of lactate, can influence chemotherapy efficacy and DNA repair mechanisms (65). Lactate-induced lactylation modifications intricately link tumor metabolism with epigenetic regulation. Investigating the mechanisms by which lactylation contributes to therapy resistance in tumors will provide deeper insights into treatment outcomes and prognosis, potentially offering novel strategies to overcome tumor resistance and improve therapeutic efficacy.

Lactylation serves a crucial role in tumor resistance to therapy (66,67). A study has shown that elevated levels of NBS1 K388 lactylation promoted homologous recombination-mediated DNA repair, leading to chemotherapy resistance (65). Additionally, histone H3K9 lactylation activates the transcription of LUC7L2, thereby inhibiting mismatch repair and ultimately contributing to temozolomide resistance in glioblastoma (68). H3K18la drives key transcription factors Y-box binding protein 1 and YY1, which facilitate cisplatin resistance in bladder cancer (69). In colorectal cancer, lactylation of histones, particularly H3K18la, enhances the expression of rubicon like autophagy enhancer, a protein involved in autophagy, promoting resistance to bevacizumab treatment (70). These findings underscore the significance of lactylation in regulating tumor drug resistance and highlight novel therapeutic targets for cancer treatment. Developing combined cancer therapy strategies may help overcome drug resistance, improve treatment efficacy and enhance patient prognosis (71).

Current research has identified lactate as serving a critical role in tumor resistance; however, traditional lactate-targeting therapies have not achieved the desired clinical outcomes (72). Consequently, the development of specific inhibitors targeting histone lactylation may offer more effective therapeutic strategies. While lactylation has provided a theoretical basis for understanding tumor resistance, the knowledge regarding the modification sites, processes and related proteins remains limited. As technologies for studying lactylation, such as lactylation proteomics, continue to advance, it is probable that more lactylation-modified proteins contributing to tumor resistance will be identified.

Role of lactylation in remodeling the TME

The TME is a highly complex and heterogeneous ecosystem, comprising tumor cells, extracellular matrix, immune cells, stromal cells, endothelial cells and an array of signaling molecules (73). The survival and growth of tumors are intricately linked to the support provided by the TME (74). Through the Warburg effect and glutamine hydrolysis, tumors produce substantial amounts of lactate, which activates transport receptors on the surface of tumor cells, facilitating the rapid export of intracellular lactate to prevent excessive intracellular acidification (75). The accumulation of extracellular lactate and the resulting acidification of the TME are pivotal processes that drive tumor progression (76). The extensive buildup of extracellular lactate increases protein lactylation levels, which can recruit and regulate cancer-associated fibroblasts, tumor-infiltrating myeloid cells, including macrophages, dendritic cells and regulatory T cells (Treg cells), and cancer stem cells. These processes collectively remodel the TME and promote tumor proliferation and progression (77).

In the TME, lactylation of tumor cells, cancer stem cells and tumor-infiltrating immune cells can actively promote cancer progression by modulating the expression of downstream genes (78). Immune cells are critical components of the TME and their infiltration influences tumor progression. For instance, in colorectal cancer, METTL3 enhances its binding affinity to RNA m6A modification targets through lactylation, mediating the immunosuppressive function of tumor-infiltrating myeloid cells and facilitating tumor immune evasion (79). Additionally, the relationship between T-cell lactylation and tumor progression has attracted widespread attention. It has been shown that the lactylation of MOESIN enhances TGF-β signaling, promoting tumorigenesis by inducing the efficient generation of Treg cells (80). Treg cells, in turn, impede the activity of cytotoxic T cells, further driving tumor progression (81). Additionally, the lactylation of histone H3 at the K18la site enhances the activity of pro-tumor macrophages, which suppresses the enrichment of CD8+ T cells and the proportion of interferon-γ+CD8+ T cells within the TME, resulting in an immunosuppressive environment (82). However, inhibiting histone lactylation in tumor-associated macrophages has been found to activate macrophage phagocytosis and M2 polarization (12,83). These studies collectively suggest that lactylation may reshape the TME and immune evasion, ultimately promoting tumor progression.

Lactic acid produced by tumor metabolism remodels the TME through lactylation, thereby contributing to tumor progression (Fig. 2). This marks a new era in the study of tumor metabolism and the TME, as protein lactylation not only expands the field of PTMs but also provides novel directions for research into the role of lactate in tumor immunity and other areas. By reshaping the TME, lactylation modulates cancer progression, offering a theoretical foundation for targeting lactylation as an antitumor strategy. Understanding the interplay between lactylation and the TME will be key to developing novel cancer therapies. Furthermore, the current research on the role and mechanisms of lactylation in relation to other key cellular components within the TME remains limited and warrants further investigation.

Crosstalk between lactylation and gut microbiota

Epigenetic modifications and metabolic reprogramming are two fundamental biological processes closely associated with tumor progression (84). While current clinical treatments for tumors primarily include targeted therapies, immunotherapy and surgery, limited research has explored the therapeutic potential of targeting gut microbiota at the level of epigenetic and metabolic reprogramming. The gut microbiota serves a crucial role in regulating the epigenetic modifications of the host, influencing early development, homeostasis and disease progression (85). Increasing evidence suggests that the gut microbiota contributes to the initiation and progression of malignant tumors (86), influencing tumor development through various mechanisms (87).

A substantial number of microbial-derived molecules are absorbed by the host, exerting profound effects on epigenetic modifications (88). On the one hand, the gut microbiota synthesizes numerous bioactive compounds, which serve as substrates, cofactors or regulators of epigenetic enzymes (89), inducing histone modifications that result in chromatin alterations (90). On the other hand, the gut microbiota can suppress aerobic glycolysis in tumors via signaling molecules, leading to a reduction in lactate production (91), thereby lowering the levels of lactylation. Furthermore, lactylation can reciprocally affect tumor metabolic reprogramming, consequently influencing the gut microbiota composition (92).

The gut microbiota and its metabolites modulate gene expression through epigenetic regulation, affecting tumor cell metabolism and thereby affecting lactylation levels. For instance, a study has shown that lipopolysaccharide induces histone lactylation, upregulating LINC00152 expression and promoting tumor invasion, highlighting the potential connection between gut microbiota and lactylation (93). The gut microbiota is diverse and different gut microbiota and their metabolic derivatives have varying effects on lactylation levels. On one hand, gut microbes can generate lactate through metabolism (94), which will increase the lactate-dependent lactylation levels of the host. On the other hand, the gut microbiota can also reduce the production of lactate and exert HDAC inhibitory activity by converting lactate into short-chain fatty acids (95), thereby reducing the overall lactylation modification levels in the host. Currently, there is limited research on the effect of lactylation on the gut microbiota and a study has shown that lactylation modification regulates the metabolic pathways of the microbiota (96), indirectly affecting the microbial ecology. Furthermore, lactylation of proteins can enhance the virulence mediated by pathogen toxins (97). Additionally, lactylation may co-regulate cellular activities via the crosstalk between tumor metabolic reprogramming and gut microbiota.

While bioactive metabolites produced by the microbiota promote epigenetic modifications, the specific mechanisms by which lactylation and gut microbiota crosstalk influence tumor progression require further investigation. Future research on the intricate crosstalk between lactylation and gut microbiota may pave the way for more targeted and effective combination therapies for cancer treatment.

Lactylation as a potential target for disease treatment

The regulation of lactate production and transport is a crucial strategy for improving tumor prognosis (98). The identification of lactylation further suggests that targeting lactylation may offer novel options to inhibit cancer progression and enhance antitumor therapies (99). Based on the mechanisms of lactylation and its carcinogenic processes, potential therapeutic approaches can target lactate metabolism, transport or lactylation generation (100).

Lactylation is primarily influenced by lactate levels (48). Inhibiting key enzymes involved in lactate production in tumor cells can effectively reduce lactylation by lowering lactate levels. For instance, reducing lactate dehydrogenase activity during glycolysis can inhibit the conversion of pyruvate to lactate, thereby lowering lactylation levels (12). Additionally, some drugs targeting lactate metabolism and transport are currently in clinical trials and combined treatment strategies may provide improved therapeutic outcomes (101). However, it is important to note that inhibitors targeting lactate metabolism may not only affect lactylation but also disrupt normal metabolic processes in cells (102). Future research should focus on developing specific metabolic inhibitors to minimize side effects and improve efficacy. Researchers have reported that demethylated zeylastal could inhibit liver cancer stem cell tumorigenesis both in vitro and in vivo (103). Besides lactate metabolism, enzymes related to lactylation also affect lactylation levels and the identification of such enzymes may provide more therapeutic targets for cancer (104). These studies demonstrate the potential of lactylation as a therapeutic target for cancer, highlighting the importance of exploring the mechanisms and regulatory sites of lactylation to identify effective cancer treatment targets. Targeting lactylation, along with combination therapies such as chemotherapy, radiotherapy and immunotherapy, could provide additional options for cancer treatment in the future.

Summary and outlook

PTMs are an indispensable component in the regulation of protein functionality, occurring both prior to and subsequent to protein biosynthesis, thereby enhancing the functional diversity of the proteome (105). PTMs modulate cellular signaling, protein localization and the maintenance of cellular functions by altering protein structure and activity, serving a pivotal role in cellular development (106). Lactylation, as a novel type of PTM, is critical for elucidating the pathological processes associated with tumors.

The present review summarizes the mechanisms underlying the generation of lactylation and its biological effects on tumor progression. Additionally, it elucidates the relationship among lactylation, tumor drug resistance and the remodeling of the TME. Furthermore, it explores the intricate crosstalk between lactylation and gut microbiota, and examines the potential of targeting lactylation as a novel therapeutic strategy for cancer. By providing a comprehensive overview of the regulatory role of lactylation in tumor development, the present review aimed to clarify its scientific significance, highlight critical unresolved questions and pave the way for novel avenues in targeted cancer therapy.

It is well established that cancer is a multifactorial disease arising from a myriad of mechanisms and the precise mechanisms driving tumorigenesis remain incompletely understood. Lactylation closely links tumor metabolism with tumor development (107), providing a novel perspective on the role of lactate and presenting a promising potential therapeutic approach for cancer treatment.

However, there is currently a lack of specific inhibitors targeting lactylation modifications. Furthermore, during tumorigenesis, lactylation may be activated in tandem with other oncogenic signals, further complicating targeted therapies. Additionally, since lactylation shares common writers or erasers with other PTMs, targeting lactylation may inadvertently affect other protein modifications. This interdependence poses challenges to the development of lactylation-specific therapeutic agents (108).

Lactylation modifications can promote tumor metabolic reprogramming by regulating gene expression, particularly through their impact on enzymes involved in glucose metabolism. However, there is limited research on the role of lactylation in other fundamental metabolic pathways. Further exploration of the relationship between lactylation modifications and tumor metabolism will enhance the comprehensive understanding of tumor epigenetics and metabolism, laying the groundwork for the development of novel therapeutic targets in cancer treatment. Additionally, the potential interplay between lactylation and gut microbiota may offer novel insights for cancer therapy. Dysbiosis of the gut microbiota not only promotes tumor progression but also affects the efficacy of antitumor drugs (109). Combining treatments that target both the gut microbiota and lactylation may enhance the effectiveness of cancer therapies (110).

Lactylation, along with a spectrum of other PTMs, adds a layer of complexity to the epigenetic regulation of tumors, making the development of sensitive and specific PTM detection methods a crucial direction for research on tumor modifications. Furthermore, the interactions between different PTMs are not mutually independent; rather, their crosstalk influences disease onset and progression (111). The interplay between these modifications serves a pivotal role in tumorigenesis (112). However, the mechanisms through which lactylation interacts with other PTMs to affect tumor development require further investigation. In summary, understanding the crosstalk between lactylation and various modifications will provide valuable insights into the intricate relationship between tumor metabolism and epigenetic processes.

Future research on lactylation could prioritize the following areas: Investigating the effect of lactylation-related enzymes on tumor progression; unveiling the complex molecular mechanisms by which different lactylation modification sites influence cancer development; and developing specific lactylation inhibitors for cancer treatment.

Conclusion

In summary, lactylation, as a novel PTM, serves a role in the pathogenesis of tumors and holds substantial potential for novel diagnostic and therapeutic approaches in cancer. Currently, research targeting lactylation remains largely theoretical, with a number of mechanisms of non-histone lactylation yet to be elucidated. Future investigations should focus on exploring the specific mechanisms of lactylation and its intricate relationships with other PTMs. A deeper understanding of the regulatory roles of lactylation in various pathological processes may pave the way for the development of innovative therapeutic strategies, offering more options and hope in clinical settings.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Natural Science and Technology Fund of Guizhou Province [grant no. Qiankehe Basic-ZK (2022) General 644].

Availability of data and materials

Not applicable.

Authors' contributions

ZZ, XZ and PZ conceived and designed the review. ZZ and XZ wrote the manuscript. GX, CC and XK critically revised and polished 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 

Doyle HA and Mamula MJ: Post-translational protein modifications in antigen recognition and autoimmunity. Trends Immunol. 22:443–449. 2001. View Article : Google Scholar : PubMed/NCBI

2 

Ramazi S and Zahiri J: Posttranslational modifications in proteins: Resources, tools and prediction methods. Database (Oxford). 2021:baab0122021. View Article : Google Scholar : PubMed/NCBI

3 

Wang H, Yang L, Liu M and Luo J: Protein post-translational modifications in the regulation of cancer hallmarks. Cancer Gene Ther. 30:529–547. 2023. View Article : Google Scholar : PubMed/NCBI

4 

Zhang H and Han W: Protein post-translational modifications in head and neck cancer. Front Oncol. 10:5719442020. View Article : Google Scholar : PubMed/NCBI

5 

Visconti A and Qiu H: Recent advances in serum response factor posttranslational modifications and their therapeutic potential in cardiovascular and neurological diseases. Vascul Pharmacol. 156:1074212024. View Article : Google Scholar : PubMed/NCBI

6 

Wang Z, Li M, Jiang H, Luo S, Shao F, Xia Y, Yang M, Ren X, Liu T, Yan M, et al: Fructose-1,6-bisphosphatase 1 functions as a protein phosphatase to dephosphorylate histone H3 and suppresses PPARα-regulated gene transcription and tumour growth. Nat Cell Biol. 24:1655–1665. 2020. View Article : Google Scholar : PubMed/NCBI

7 

Zhong Q, Xiao X, Qiu Y, Xu Z, Chen C, Chong B, Zhao X, Hai S, Li S, An Z and Dai L: Protein posttranslational modifications in health and diseases: Functions, regulatory mechanisms, and therapeutic implications. MedComm (2020). 4:e2612023. View Article : Google Scholar : PubMed/NCBI

8 

Yu T, Wang Y, Fan Y, Fang N, Wang T, Xu T and Shu Y: CircRNAs in cancer metabolism: A review. J Hematol Oncol. 12:902019. View Article : Google Scholar : PubMed/NCBI

9 

San-Millan I, Sparagna GC, Chapman HL, Warkins VL, Chatfield KC, Shuff SR, Martinez JL and Brooks GA: Chronic lactate exposure decreases mitochondrial function by inhibition of fatty acid uptake and cardiolipin alterations in neonatal rat cardiomyocytes. Front Nutr. 9:8094852022. View Article : Google Scholar : PubMed/NCBI

10 

Brooks GA: Lactate as a fulcrum of metabolism. Redox Biol. 35:1014542020. View Article : Google Scholar : PubMed/NCBI

11 

Zhu W, Guo S, Sun J, Zhao Y and Liu C: Lactate and lactylation in cardiovascular diseases: Current progress and future perspectives. Metabolism. 158:1559572024. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Pérez-Tomás R and Pérez-Guillén I: Lactate in the tumor microenvironment: An essential molecule in cancer progression and treatment. Cancers (Basel). 12:32442020. View Article : Google Scholar : PubMed/NCBI

14 

Yu X, Yang J, Xu J, Pan H, Wang W, Yu X and Shi S: Histone lactylation: From tumor lactate metabolism to epigenetic regulation. Int J Biol Sci. 20:1833–1854. 2024. View Article : Google Scholar : PubMed/NCBI

15 

Heydari Z, Moeinvaziri F, Mirazimi SMA, Dashti F, Smirnova O, Shpichka A, Mirzaei H, Timashev P and Vosough M: Alteration in DNA methylation patterns: Epigenetic signatures in gastrointestinal cancers. Eur J Pharmacol. 973:1765632024. View Article : Google Scholar : PubMed/NCBI

16 

Rungratanawanich W, Ballway JW, Wang X, Won KJ, Hardwick JP and Song BJ: Post-translational modifications of histone and non-histone proteins in epigenetic regulation and translational applications in alcohol-associated liver disease: Challenges and research opportunities. Pharmacol Ther. 251:1085472023. View Article : Google Scholar : PubMed/NCBI

17 

Wang J, Wang Z, Wang Q, Li X and Guo Y: Ubiquitous protein lactylation in health and diseases. Cell Mol Biol Lett. 29:232024. View Article : Google Scholar : PubMed/NCBI

18 

Xu X, Zhang DD, Kong P, Gao YK, Huang XF, Song Y, Zhang WD, Guo RJ, Li CL, Chen BW, et al: Sox10 escalates vascular inflammation by mediating vascular smooth muscle cell transdifferentiation and pyroptosis in neointimal hyperplasia. Cell Rep. 42:1128692023. View Article : Google Scholar : PubMed/NCBI

19 

Yang D, Yin J, Shan L, Yi X, Zhang W and Ding Y: Identification of lysine-lactylated substrates in gastric cancer cells. iScience. 25:1046302022. View Article : Google Scholar : PubMed/NCBI

20 

Chen M, Cen K, Song Y, Zhang X, Liou YC, Liu P, Huang J, Ruan J, He J, Ye W, et al: NUSAP1-LDHA-glycolysis-lactate feedforward loop promotes Warburg effect and metastasis in pancreatic ductal adenocarcinoma. Cancer Lett. 567:2162852023. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Chu YD, Cheng LC, Lim SN, Lai MW, Yeh CT and Lin WR: Aldolase B-driven lactagenesis and CEACAM6 activation promote cell renewal and chemoresistance in colorectal cancer through the Warburg effect. Cell Death Dis. 14:6602023. View Article : Google Scholar : PubMed/NCBI

23 

Gu X, Zhuang A, Yu J, Yang L, Ge S, Ruan J, Jia R, Fan X and Chai P: Histone lactylation-boosted ALKBH3 potentiates tumor progression and diminished promyelocytic leukemia protein nuclear condensates by m1A demethylation of SP100A. Nucleic Acids Res. 52:2273–2289. 2024. View Article : Google Scholar : PubMed/NCBI

24 

Liu R, Wu J, Guo H, Yao W, Li S, Lu Y, Jia Y, Liang X, Tang J and Zhang H: Post-translational modifications of histones: Mechanisms, biological functions, and therapeutic targets. MedComm (2020). 4:e2922023. View Article : Google Scholar : PubMed/NCBI

25 

Pan RY, He L, Zhang J, Liu X, Liao Y, Gao J, Liao Y, Yan Y, Li Q, Zhou X, et al: Positive feedback regulation of microglial glucose metabolism by histone H4 lysine 12 lactylation in Alzheimer's disease. Cell Metab. 34:634–648.e6. 2022. View Article : Google Scholar : PubMed/NCBI

26 

Levine AJ and Puzio-Kuter AM: The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes. Science. 330:1340–1344. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Zhang Y, Song H, Li M and Lu P: Histone lactylation bridges metabolic reprogramming and epigenetic rewiring in driving carcinogenesis: Oncometabolite fuels oncogenic transcription. Clin Transl Med. 14:e16142024. View Article : Google Scholar : PubMed/NCBI

28 

Hu Y, He Z, Li Z, Wang Y, Wu N, Sun H, Zhou Z, Hu Q and Cong X: Lactylation: The novel histone modification influence on gene expression, protein function, and disease. Clin Epigenetics. 16:722024. View Article : Google Scholar : PubMed/NCBI

29 

Wu D, Spencer CB, Ortoga L, Zhang H and Miao C: Histone lactylation-regulated METTL3 promotes ferroptosis via m6A-modification on ACSL4 in sepsis-associated lung injury. Redox Biol. 74:1031942024. View Article : Google Scholar : PubMed/NCBI

30 

Wu F, Hua Y, Kaochar S, Nie S, Lin YL, Yao Y, Wu J, Wu X, Fu X, Schiff R, et al: Discovery, structure-activity relationship, and biological activity of histone-competitive inhibitors of histone acetyltransferases P300/CBP. J Med Chem. 63:4716–4731. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Antika TR, Chrestella DJ, Tseng YK, Yeh YH, Hsiao CD and Wang CC: A naturally occurring mini-alanyl-tRNA synthetase. Commun Biol. 6:3142023. View Article : Google Scholar : PubMed/NCBI

32 

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

33 

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

34 

Yoo L, Mendoza D, Richard AJ and Stephens JM: KAT8 beyond acetylation: A survey of its epigenetic regulation, genetic variability, and implications for human health. Genes (Basel). 15:6392024. View Article : Google Scholar : PubMed/NCBI

35 

Xie B, Zhang M, Li J, Cui J, Zhang P, Liu F, Wu Y, Deng W, Ma J, Li X, et al: KAT8-catalyzed lactylation promotes eEF1A2-mediated protein synthesis and colorectal carcinogenesis. Proc Natl Acad Sci USA. 121:e23141281212024. View Article : Google Scholar : PubMed/NCBI

36 

Dong H, Zhang J, Zhang H, Han Y, Lu C, Chen C, Tan X, Wang S, Bai X, Zhai G, et al: YiaC and CobB regulate lysine lactylation in Escherichia coli. Nat Commun. 13:66282022. View Article : Google Scholar : PubMed/NCBI

37 

Parks AR and Escalante-Semerena JC: Modulation of the bacterial CobB sirtuin deacylase activity by N-terminal acetylation. Proc Natl Acad Sci USA. 117:15895–15901. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Mutlu B and Puigserver P: GCN5 acetyltransferase in cellular energetic and metabolic processes. Biochim Biophys Acta Gene Regul Mech. 1864:1946262021. View Article : Google Scholar : PubMed/NCBI

39 

Yang K, Fan M, Wang X, Xu J, Wang Y, Tu F, Gill PS, Ha T, Liu L, Williams DL and Li C: Lactate promotes macrophage HMGB1 lactylation, acetylation, and exosomal release in polymicrobial sepsis. Cell Death Differ. 29:133–146. 2022. View Article : Google Scholar : PubMed/NCBI

40 

Moreno-Yruela C, Zhang D, Wei W, Bæk M, Liu W, Gao J, Danková D, Nielsen AL, Bolding JE, Yang L, et al: Class I histone deacetylases (HDAC1-3) are histone lysine delactylases. Sci Adv. 8:eabi66962022. View Article : Google Scholar : PubMed/NCBI

41 

Micelli C and Rastelli G: Histone deacetylases: Structural determinants of inhibitor selectivity. Drug Discov Today. 20:718–735. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Fan Z, Liu Z, Zhang N, Wei W, Cheng K, Sun H and Hao Q: Identification of SIRT3 as an eraser of H4K16la. iScience. 26:1077572023. View Article : Google Scholar : PubMed/NCBI

43 

Tao Z, Jin Z, Wu J, Cai G and Yu X: Sirtuin family in autoimmune diseases. Front Immunol. 14:11862312023. View Article : Google Scholar : PubMed/NCBI

44 

Hagihara H, Shoji H, Otabi H, Toyoda A, Katoh K, Namihira M and Miyakawa T: Protein lactylation induced by neural excitation. Cell Rep. 37:1098202021. View Article : Google Scholar : PubMed/NCBI

45 

Rho H, Terry AR, Chronis C and Hay N: Hexokinase 2-mediated gene expression via histone lactylation is required for hepatic stellate cell activation and liver fibrosis. Cell Metab. 35:1406–1423.e8. 2023. View Article : Google Scholar : PubMed/NCBI

46 

Gaffney DO, Jennings EQ, Anderson CC, Marentette JO, Shi T, Schou Oxvig AM, Streeter MD, Johannsen M, Spiegel DA, Chapman E, et al: Non-enzymatic lysine lactoylation of glycolytic enzymes. Cell Chem Biol. 27:206–213.e6. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Kiri S and Ryba T: Cancer, metastasis, and the epigenome. Mol Cancer. 23:1542024. View Article : Google Scholar : PubMed/NCBI

48 

Lv X, Lv Y and Dai X: Lactate, histone lactylation and cancer hallmarks. Expert Rev Mol. 25:e72023. View Article : Google Scholar : PubMed/NCBI

49 

Yan F, Teng Y, Li X, Zhong Y, Li C, Yan F and He X: Hypoxia promotes non-small cell lung cancer cell stemness, migration, and invasion via promoting glycolysis by lactylation of SOX9. Cancer Biol Ther. 25:23041612024. View Article : Google Scholar : PubMed/NCBI

50 

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

51 

Qiao Z, Li Y, Li S, Liu S and Cheng Y: Hypoxia-induced SHMT2 protein lactylation facilitates glycolysis and stemness of esophageal cancer cells. Mol Cell Biochem. 479:3063–3076. 2024. View Article : Google Scholar : PubMed/NCBI

52 

Miao Z, Zhao X and Liu X: Hypoxia induced β-catenin lactylation promotes the cell proliferation and stemness of colorectal cancer through the wnt signaling pathway. Exp Cell Res. 422:1134392023. View Article : Google Scholar : PubMed/NCBI

53 

Yu J, Chai P, Xie M, Ge S, Ruan J, Fan X and Jia R: Histone lactylation drives oncogenesis by facilitating m6A reader protein YTHDF2 expression in ocular melanoma. Genome Biol. 22:852021. View Article : Google Scholar : PubMed/NCBI

54 

Huang Y, Luo G, Peng K, Song Y, Wang Y, Zhang H, Li J, Qiu X, Pu M, Liu X, et al: Lactylation stabilizes TFEB to elevate autophagy and lysosomal activity. J Cell Biol. 223:e2023080992024. View Article : Google Scholar : PubMed/NCBI

55 

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

56 

Yang J, Luo L, Zhao C, Li X, Wang Z, Zeng Z, Yang X, Zheng X, Jie H, Kang L, et al: A positive feedback loop between inactive VHL-triggered histone lactylation and PDGFRβ signaling drives clear cell renal cell carcinoma progression. Int J Biol Sci. 18:3470–3483. 2022. View Article : Google Scholar : PubMed/NCBI

57 

Hou X, Ouyang J, Tang L, Wu P, Deng X, Yan Q, Shi L, Fan S, Fan C, Guo C, et al: KCNK1 promotes proliferation and metastasis of breast cancer cells by activating lactate dehydrogenase A (LDHA) and up-regulating H3K18 lactylation. PLoS Biol. 22:e30026662024. View Article : Google Scholar : PubMed/NCBI

58 

Sun L, Zhang Y, Yang B, Sun S, Zhang P, Luo Z, Feng T, Cui Z, Zhu T, Li Y, et al: Lactylation of METTL16 promotes cuproptosis via m6A-modification on FDX1 mRNA in gastric cancer. Nat Commun. 14:65232023. View Article : Google Scholar : PubMed/NCBI

59 

Xie B, Lin J, Chen X, Zhou X, Zhang Y, Fan M, Xiang J, He N, Hu Z and Wang F: CircXRN2 suppresses tumor progression driven by histone lactylation through activating the Hippo pathway in human bladder cancer. Mol Cancer. 22:1512023. View Article : Google Scholar : PubMed/NCBI

60 

Wang X, Ying T, Yuan J, Wang Y, Su X, Chen S, Zhao Y, Zhao Y, Sheng J, Teng L, et al: BRAFV600E restructures cellular lactylation to promote anaplastic thyroid cancer proliferation. Endocr Relat Cancer. 30:e2203442023. View Article : Google Scholar : PubMed/NCBI

61 

Dai E, Wang W and Li Y, Ye D and Li Y: Lactate and lactylation: Behind the development of tumors. Cancer Lett. 591:2168962024. View Article : Google Scholar : PubMed/NCBI

62 

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

63 

Jing F, Zhang J, Zhang H and Li T: Unlocking the multifaceted molecular functions and diverse disease implications of lactylation. Biol Rev Camb Philos Soc. 100:172–189. 2025. View Article : Google Scholar : PubMed/NCBI

64 

Xia Y, Sun M, Huang H and Jin WL: Drug repurposing for cancer therapy. Signal Transduct Target Ther. 9:922024. View Article : Google Scholar : PubMed/NCBI

65 

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

66 

Chen Y, Wu J, Zhai L, Zhang T, Yin H, Gao H, Zhao F, Wang Z, Yang X, Jin M, et al: Metabolic regulation of homologous recombination repair by MRE11 lactylation. Cell. 187:294–311.e21. 2024. View Article : Google Scholar : PubMed/NCBI

67 

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

68 

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

69 

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

70 

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 : PubMed/NCBI

71 

Komedchikova EN, Kolesnikova OA, Syuy AV, Volkov VS, Deyev SM, Nikitin MP and Shipunova VO: Targosomes: Anti-HER2 PLGA nanocarriers for bioimaging, chemotherapy and local photothermal treatment of tumors and remote metastases. J Control Release. 365:317–330. 2024. View Article : Google Scholar : PubMed/NCBI

72 

Chen S, Xu Y, Zhuo W and Zhang L: The emerging role of lactate in tumor microenvironment and its clinical relevance. Cancer Lett. 590:2168372024. View Article : Google Scholar : PubMed/NCBI

73 

Wang S, Qi X, Liu D, Xie D, Jiang B, Wang J, Wang X and Wu G: The implications for urological malignancies of non-coding RNAs in the the tumor microenvironment. Comput Struct Biotechnol J. 23:491–505. 2023. View Article : Google Scholar : PubMed/NCBI

74 

Li Y, Cao Q, Hu Y, He B, Cao T, Tang Y, Zhou XP, Lan XP and Liu SQ: Advances in the interaction of glycolytic reprogramming with lactylation. Biomed Pharmacother. 177:1169822024. View Article : Google Scholar : PubMed/NCBI

75 

Li X, Yang Y, Zhang B, Lin X, Fu X, An Y, Zou Y, Wang JX, Wang Z and Yu T: Lactate metabolism in human health and disease. Signal Transduct Target Ther. 7:3052022. View Article : Google Scholar : PubMed/NCBI

76 

Wang JX, Choi SYC, Niu X, Kang N, Xue H, Killam J and Wang Y: Lactic acid and an acidic tumor microenvironment suppress anticancer immunity. Int J Mol Sci. 21:83632020. View Article : Google Scholar : PubMed/NCBI

77 

Huber V, Camisaschi C, Berzi A, Ferro S, Lugini L, Triulzi T, Tuccitto A, Tagliabue E, Castelli C and Rivoltini L: Cancer acidity: An ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin Cancer Biol. 43:74–89. 2017. View Article : Google Scholar : PubMed/NCBI

78 

Qu J, Li P and Sun Z: Histone lactylation regulates cancer progression by reshaping the tumor microenvironment. Front Immunol. 14:12843442023. View Article : Google Scholar : PubMed/NCBI

79 

Xiong J, He J, Zhu J, Pan J, Liao W, Ye H, Wang H, Song Y, Du Y, Cui B, et al: Lactylation-driven METTL3-mediated RNA m6A modification promotes immunosuppression of tumor-infiltrating myeloid cells. Mol Cell. 82:1660–1677.e10. 2022. View Article : Google Scholar : PubMed/NCBI

80 

Gu J, Zhou J, Chen Q, Xu X, Gao J, Li X, Shao Q, Zhou B, Zhou H, Wei S, et al: Tumor metabolite lactate promotes tumorigenesis by modulating MOESIN lactylation and enhancing TGF-β signaling in regulatory T cells. Cell Rep. 40:1111222022. View Article : Google Scholar : PubMed/NCBI

81 

Chevrier S, Levine JH, Zanotelli VRT, Silina K, Schulz D, Bacac M, Ries CH, Ailles L, Jewett MAS, Moch H, et al: An immune atlas of clear cell renal cell carcinoma. Cell. 169:736–749.e18. 2017. View Article : Google Scholar : PubMed/NCBI

82 

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

83 

Chaudagar K, Hieromnimon HM, Khurana R, Labadie B, Hirz T, Mei S, Hasan R, Shafran J, Kelley A, Apostolov E, et al: Reversal of lactate and PD-1-mediated macrophage immunosuppression controls growth of PTEN/p53-deficient prostate cancer. Clin Cancer Res. 29:1952–1968. 2023. View Article : Google Scholar : PubMed/NCBI

84 

Sun L, Zhang H and Gao P: Metabolic reprogramming and epigenetic modifications on the path to cancer. Protein Cell. 13:877–919. 2022. View Article : Google Scholar : PubMed/NCBI

85 

Jiang SS, Kang ZR, Chen YX and Fang JY: The gut microbiome modulate response to immunotherapy in cancer. Sci China Life Sci. 68:381–396. 2025. View Article : Google Scholar : PubMed/NCBI

86 

Xie Y, Xie F, Zhou X, Zhang L, Yang B, Huang J, Wang F, Yan H, Zeng L, Zhang L and Zhou F: Microbiota in tumors: From understanding to application. Adv Sci (Weinh). 9:e22004702022. View Article : Google Scholar : PubMed/NCBI

87 

Sepich-Poore GD, Zitvogel L, Straussman R, Hasty J, Wargo JA and Knight R: The microbiome and human cancer. Science. 371:eabc45522021. View Article : Google Scholar : PubMed/NCBI

88 

Mischke M and Plösch T: The gut microbiota and their metabolites: Potential implications for the host epigenome. Adv Exp Med Biol. 902:33–44. 2016. View Article : Google Scholar : PubMed/NCBI

89 

Woo V and Alenghat T: Epigenetic regulation by gut microbiota. Gut Microbes. 14:20224072022. View Article : Google Scholar : PubMed/NCBI

90 

Shock T, Badang L, Ferguson B and Martinez-Guryn K: The interplay between diet, gut microbes, and host epigenetics in health and disease. J Nutr Biochem. 95:1086312021. View Article : Google Scholar : PubMed/NCBI

91 

Zhang Z, Chen Y, Zheng Y, Wang L, Shen S, Yang G, Yang Y and Wang T: Quxie capsule alleviates colitis-associated colorectal cancer through modulating the gut microbiota and suppressing A. fumigatus-induced aerobic glycolysis. Integr Cancer Ther. 21:153473542211385342022. View Article : Google Scholar : PubMed/NCBI

92 

Sun S, Xu X, Liang L, Wang X, Bai X, Zhu L, He Q, Liang H, Xin X, Wang L, et al: Lactic acid-producing probiotic saccharomyces cerevisiae attenuates ulcerative colitis via suppressing macrophage pyroptosis and modulating gut microbiota. Front Immunol. 12:7776652021. View Article : Google Scholar : PubMed/NCBI

93 

Wang J, Liu Z, Xu Y, Wang Y, Wang F, Zhang Q, Ni C, Zhen Y, Xu R, Liu Q, et al: Enterobacterial LPS-inducible LINC00152 is regulated by histone lactylation and promotes cancer cells invasion and migration. Front Cell Infect Microbiol. 12:9138152022. View Article : Google Scholar : PubMed/NCBI

94 

Wang SP, Rubio LA, Duncan SH, Donachie GE, Holtrop G, Lo G, Farquharson FM, Wagner J, Parkhill J, Louis P, et al: Pivotal roles for pH, lactate, and lactate-utilizing bacteria in the stability of a human colonic microbial ecosystem. mSystems. 5:e00645–20. 2020. View Article : Google Scholar : PubMed/NCBI

95 

Koh A, De Vadder F, Kovatcheva-Datchary P and Bäckhed F: From dietary fiber to host physiology: Short-chain fatty acids as key bacterial metabolites. Cell. 165:1332–1345. 2016. View Article : Google Scholar : PubMed/NCBI

96 

Li Z, Gong T, Wu Q, Zhang Y, Zheng X, Li Y, Ren B, Peng X and Zhou X: Lysine lactylation regulates metabolic pathways and biofilm formation in streptococcus mutans. Sci Signal. 16:eadg18492023. View Article : Google Scholar : PubMed/NCBI

97 

Wang Y, Liu Y, Xiang G, Jian Y, Yang Z, Chen T, Ma X, Zhao N, Dai Y, Lv Y, et al: Post-translational toxin modification by lactate controls staphylococcus aureus virulence. Nat Commun. 15:98352024. View Article : Google Scholar : PubMed/NCBI

98 

Lin J, Liu G, Chen L, Kwok HF and Lin Y: Targeting lactate-related cell cycle activities for cancer therapy. Semin Cancer Biol. 86:1231–1243. 2022. View Article : Google Scholar : PubMed/NCBI

99 

Fan H, Yang F, Xiao Z, Luo H, Chen H, Chen Z, Liu Q and Xiao Y: Lactylation: Novel epigenetic regulatory and therapeutic opportunities. Am J Physiol Endocrinol Metab. 324:E330–E338. 2023. View Article : Google Scholar : PubMed/NCBI

100 

Zhang Q, Cao L and Xu K: Role and mechanism of lactylation in cancer. Zhongguo Fei Ai Za Zhi. 27:471–479. 2024.(In Chinese). PubMed/NCBI

101 

De Cesare M, Pratesi G, Giusti A, Polizzi D and Zunino F: Stimulation of the apoptotic response as a basis for the therapeutic synergism of lonidamine and cisplatin in combination in human tumour xenografts. Br J Cancer. 77:434–439. 1998. View Article : Google Scholar : PubMed/NCBI

102 

Shu Y, Yue J, Li Y, Yin Y, Wang J, Li T, He X, Liang S, Zhang G, Liu Z and Wang Y: Development of human lactate dehydrogenase a inhibitors: High-throughput screening, molecular dynamics simulation and enzyme activity assay. J Comput Aided Mol Des. 38:282024. View Article : Google Scholar : PubMed/NCBI

103 

Pan L, Feng F, Wu J, Fan S, Han J, Wang S, Yang L, Liu W, Wang C and Xu K: Demethylzeylasteral targets lactate by inhibiting histone lactylation to suppress the tumorigenicity of liver cancer stem cells. Pharmacol Res. 181:1062702022. View Article : Google Scholar : PubMed/NCBI

104 

Su J, Zheng Z, Bian C, Chang S, Bao J, Yu H, Xin Y and Jiang X: Functions and mechanisms of lactylation in carcinogenesis and immunosuppression. Front Immunol. 14:12530642023. View Article : Google Scholar : PubMed/NCBI

105 

Smith LE and Rogowska-Wrzesinska A: The challenge of detecting modifications on proteins. Essays Biochem. 64:135–153. 2020. View Article : Google Scholar : PubMed/NCBI

106 

Hao Y, Gu C, Luo W, Shen J, Xie F, Zhao Y, Song X, Han Z and He J: The role of protein post-translational modifications in prostate cancer. PeerJ. 12:e177682024. View Article : Google Scholar : PubMed/NCBI

107 

Xin Q, Wang H, Li Q, Liu S, Qu K, Liu C and Zhang J: Lactylation: A passing fad or the future of posttranslational modification. Inflammation. 45:1419–1429. 2022. View Article : Google Scholar : PubMed/NCBI

108 

Gao X, Pang C, Fan Z, Wang Y, Duan Y and Zhan H: Regulation of newly identified lysine lactylation in cancer. Cancer Lett. 587:2166802024. View Article : Google Scholar : PubMed/NCBI

109 

Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CPM, et al: Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 350:1079–1084. 2015. View Article : Google Scholar : PubMed/NCBI

110 

Gori S, Inno A, Belluomini L, Bocus P, Bisoffi Z, Russo A and Arcaro G: Gut microbiota and cancer: How gut microbiota modulates activity, efficacy and toxicity of antitumoral therapy. Crit Rev Oncol Hematol. 143:139–147. 2019. View Article : Google Scholar : PubMed/NCBI

111 

Wu Z, Huang R and Yuan L: Crosstalk of intracellular post-translational modifications in cancer. Arch Biochem Biophys. 676:1081382019. View Article : Google Scholar : PubMed/NCBI

112 

Tomasi ML and Ramani K: SUMOylation and phosphorylation cross-talk in hepatocellular carcinoma. Transl Gastroenterol Hepatol. 3:202018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2025
Volume 31 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhu Z, Zheng X, Zhao P, Chen C, Xu G and Ke X: Potential of lactylation as a therapeutic target in cancer treatment (Review). Mol Med Rep 31: 91, 2025.
APA
Zhu, Z., Zheng, X., Zhao, P., Chen, C., Xu, G., & Ke, X. (2025). Potential of lactylation as a therapeutic target in cancer treatment (Review). Molecular Medicine Reports, 31, 91. https://doi.org/10.3892/mmr.2025.13456
MLA
Zhu, Z., Zheng, X., Zhao, P., Chen, C., Xu, G., Ke, X."Potential of lactylation as a therapeutic target in cancer treatment (Review)". Molecular Medicine Reports 31.4 (2025): 91.
Chicago
Zhu, Z., Zheng, X., Zhao, P., Chen, C., Xu, G., Ke, X."Potential of lactylation as a therapeutic target in cancer treatment (Review)". Molecular Medicine Reports 31, no. 4 (2025): 91. https://doi.org/10.3892/mmr.2025.13456