Histone lactylation: A new frontier in laryngeal cancer research (Review)

  • Authors:
    • Qiaoling Tong
    • Chunsheng Huang
    • Qizhen Tong
    • Zhiyu Zhang
  • View Affiliations

  • Published online on: July 2, 2025     https://doi.org/10.3892/ol.2025.15167
  • Article Number: 421
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Laryngeal cancer is a common malignant tumor of the head and neck, and its occurrence and development are closely related to epigenetic modifications. Histone lactylation, a novel form of epigenetic modification, regulates gene expression and is involved in processes such as tumor metabolic reprogramming, shaping the tumor microenvironment, maintaining tumor stem cell characteristics and developing treatment resistance. The present review highlighted the molecular mechanisms and detection methods of histone lactylation and explores its potential role in laryngeal cancer, as it may serve as a valuable biomarker for diagnosis and prognosis. Furthermore, targeting the lactylation pathway may provide novel therapeutic strategies, with the potential to improve treatment outcomes for patients. Overall, understanding the role of histone lactylation in laryngeal cancer may potentially lead to innovative diagnostic and therapeutic approaches and improve the management of laryngeal cancer in the future.

Introduction

Laryngeal cancer is one of the common malignant tumors in the head and neck, which accounts for 20–30% of all types of head and neck tumors (1). Squamous cell carcinoma is the most common pathological type of laryngeal cancer (2). With the impact of factors such as environmental pollution, smoking, alcohol consumption and changes in lifestyle, the incidence and mortality of laryngeal cancer have been rising (3). According to the latest statistics, the incidence and mortality rates of global laryngeal cancer are 3.9 per 100,000 and 2.1 per 100,000, respectively (4). Although comprehensive treatment methods including surgery, radiotherapy and chemotherapy continue to improve, the prognosis of patients with laryngeal cancer remains unsatisfactory, particularly for advanced patients who exhibit a low 5-year survival rate (5). Thus, there is an urgent need to further explore the molecular mechanisms underlying the occurrence and development of laryngeal cancer to identify new diagnostic markers and therapeutic targets.

The importance of epigenetics in tumor research is increasingly evident, and histone modifications are considered one of the core mechanisms of epigenetic regulation (6). Among various histone post-translational modifications (PTMs), traditional modifications such as acetylation, methylation, phosphorylation and ubiquitination have been systematically studied (710). However, the emerging histone lactylation modification has recently drawn attention, especially for its potential importance in tumor metabolism and immune regulation (11). In contrast to acetylation, lactylation not only derives from the metabolic product lactate but also directly reflects changes in the cellular metabolic state, which possesses a unique dynamic and reversible nature (12).

Previous studies on lactylation modification in various squamous cell carcinomas (such as oral squamous cell carcinoma and esophageal squamous cell carcinoma (ESCC) indicated that by regulating gene expression, lactylation modification participates in tumor metabolic reprogramming, immune evasion and therapeutic resistance (Table I) (1319). According to the study reported by Wang et al (18), histone H3K9 lactylation serves a key role in immune evasion in head and neck squamous cell carcinoma (HNSCC). These studies provide an important reference for investigations on the role of lactylation modification in laryngeal cancer. The present review highlighted the molecular basis of histone lactylation modification and associated research in laryngeal cancer, which explored its potential applications in the diagnosis, prognostic evaluation and treatment of laryngeal cancer.

Table I.

Lactylation in types of squamous cell carcinoma.

Table I.

Lactylation in types of squamous cell carcinoma.

A, Oral squamous cell carcinoma

First author, yearProteins modified by lactylationAffected genes or pathwaysRole of lactic acidification(Refs.)
Jing et al, 2024DHX9 K146Not specified.Promoted the occurrence and progression of oral squamous cell carcinoma.(13)
Huang et al, 2024Histone lysineBCAMPromoted invasion, angiogenesis and chemoresistance in oral squamous cell carcinoma.(14)
Song et al, 2024SF3A1, hnRNPA1, hnRNPU and SLU7Spliceosome, ribosome and glycolysis/gluco neogenesis pathwayLactylation levels were negatively associated with the prognosis of patients with oral squamous cell carcinoma.(15)

B, Esophageal squamous cell carcinoma

First author, yearProteins modified by lactylationAffected genes or pathwaysRole of lactic acidification(Refs.)

Zang et al, 2024H3K9laLAMC2Promoted LAMC2 expression under hypoxic conditions, enhanced cell proliferation.(16)
Fu et al, 2024H3K18lac-MycPromoted the proliferation of squamous cell carcinoma.(17)

C, Head and neck squamous cell carcinoma

First author, yearProteins modified by lactylationAffected genes or pathwaysRole of lactic acidification(Refs.)

Wang et al, 2024H3K9laIL-11; JAK2/STAT3Promoted CD8+ T cell dysfunction and poor immunotherapy response.(18)

D, Cervical squamous cell carcinoma

First author, yearProteins modified by lactylationAffected genes or pathwaysRole of lactic acidification(Refs.)

Huang et al, 2024H3K18laGPD2Mediated M2 macrophage polarization and promoted the malignant progression of cervical cancer.(19)

[i] DHX9, DExH-box helicase 9. BCAM, basal cell adhesion molecule; SF3A1, splicing factor 3A subunit 1; hnRNPA1, heterogeneous nuclear ribonucleoprotein A1; hnRNPU, heterogeneous nuclear ribonucleoprotein U; SLU7, splicing factor, lethal (2) underexported in drosophila 7; LAMC2, laminin subunit γ-2; H3K, histone H3 lysine; JAK, janus kinase; GPD2, glycerol-3-phosphate dehydrogenase 2.

Epigenetic regulatory mechanisms of laryngeal cancer

Epigenetics serve a key role in the initiation and progression of laryngeal cancer and other malignant tumors in the ear, nose and throat (ENT) field. Previous studies indicated that the development of laryngeal cancer and other types of HNSCC is often accompanied by notable epigenetic abnormalities. These changes are not direct mutations of the DNA base sequence but manifest at the levels of DNA methylation, histone modification and non-coding RNA regulation, which affect cell fate by modulating gene expression (3,20,21).

As an example of DNA methylation, tumor suppressor genes such as p16INK4a, MGMT and DAPK1 frequently exhibit promoter hypermethylation in tumor tissues of patients with laryngeal cancer, which leads to transcriptional silencing (2224). This ‘epigenetic inactivation’ can inhibit apoptosis and promote aberrant proliferation, which represents a key molecular mechanism in laryngeal carcinogenesis and disease progression. Furthermore, alterations in DNA methylation status are closely associated with tumor prognosis and therapeutic response that serve as important references for molecular subtyping and prognostic assessment (25). PTMs are similarly a key mechanism of epigenetic regulation in ENT tumors (26). Dysregulated histone deacetylases and methyltransferases readily result in oncogene activation and tumor suppressor gene silencing, which thereby drive tumor development. A previous study has shown that the expression levels of H4R3me2a in laryngeal cancer cells are closely associated with their proliferative, migratory and invasive capacities (27). Non-coding RNAs, especially microRNAs (miR) such as miR-21 and miR-155 and long non-coding RNAs such as HOX transcript antisense intergenic RNA, are aberrantly expressed in laryngeal cancer tissues and can further promote tumor progression and metastasis by regulating key tumor suppressors or downstream pathway genes, which affects patient prognosis (2830). In addition, lifestyle factors such as smoking and alcohol consumption (31,32) and metabolic status, as well as exogenous factors including Epstein-Barr virus and human papillomavirus infection (33,34) may subsequently influence host epigenetics and the risk of associated malignancies, through alterations in the microbiome (35).

At the functional level, epigenetic modifications regulate gene expression by affecting chromatin structure. For example, histone acetylation generally leads to chromatin relaxation and promotes transcriptional activation; conversely, deacetylation compacts chromatin and represses gene expression (36). Histone methylation exerts distinct functions depending on the modification site; H3K4me3 is predominantly associated with gene activation (37), whereas H3K27me3 commonly mediates gene silencing (38). Other modifications, such as phosphorylation and ubiquitination, also participate in various biological processes, including the cell cycle, DNA damage repair and chromosome remodeling (39,40).

In summary, epigenetic modifications serve a key role in the initiation, progression and treatment tolerance of ENT malignancies, particularly laryngeal cancer. Multiple modifications, including PTMs and DNA methylation, together constitute a refined gene expression network. A thorough elucidation of the aberrant epigenetic mechanisms in laryngeal cancer not only contributes to the refinement of molecular tumor subtyping and the development of novel diagnostic and targeted therapeutic approaches, but also potentially provides a theoretical foundation and clinical prospects for the exploration of innovative therapies and the enhancement of precision medicine.

Molecular mechanisms of histone lactylation modification

Source and metabolism of lactate

Lactate is the end product of glycolysis, primarily generated under hypoxic conditions in normal cells (41). However, in tumor cells, even under oxygen-sufficient conditions, the glycolytic pathway remains highly active, a phenomenon known as the ‘Warburg effect’ (42). This metabolic reprogramming enables tumor cells to rapidly uptake glucose and convert it into lactate and thus meets the energy and metabolic intermediate requirements for rapid proliferation (43).

Lactic acid not only functions intracellularly but is also actively transported out of the cell, which serves as a signaling molecule involved in the regulation of various biological processes, particularly epigenetic regulation (44,45). For example, in breast cancer, lactate promoted angiogenesis by regulating the expression of hypoxia-inducible factor-1-α (46). In colorectal cancer, lactate suppressed antitumor immune responses by regulating the expression of immunosuppressive factors such as transforming growth factor-β (TGF-β) (47).

Discovery and characteristics of histone lactylation modification

Histone lactylation was first reported by Zhang et al (12) in 2019 as a novel form of epigenetic modification. The histone lactylation modification is formed through the covalent binding of lactate molecules to histone lysine residues and is one of the acylation modifications of lysine. Previous studies have reported that the histone lactylation modification mainly occurs at specific sites on histone H3, such as H3K18 (48,49). The discovery of histone lactylation expanded the current understanding of epigenetic modifications, as it suggests that lactate is not only a metabolic byproduct but can also directly participate in the regulation of gene expression.

The specificity and functions of lactylation modification may vary with cell type and environmental conditions. For example: i) in breast cancer cells, expression levels of H3K18la were markedly elevated, which promoted the expression of key transcription factors for tumor cell growth (50); ii) in ESCC, elevated H3K9la expression levels enhanced tumor cell invasion and metastasis (16); and iii) in healthy cells, lactylation modification levels were relatively low and mainly participate in the maintenance of metabolic homeostasis (51). These studies indicated that lactylation modification is dynamic and reversible, capable of quickly responding to changes in cellular metabolic status.

Regulatory mechanisms of histone lactylation modification

Regulation of histone lactylation involves multiple molecular mechanisms, but its detailed enzymatic regulatory network remains to be elucidated. Compared with classical PTMs such as acetylation, methylation and phosphorylation, lactylation exhibits a lower basal expression level and stronger tissue specificity, which indicates that under normal physiological conditions it is largely restricted to specific cell types and their metabolic activity (5255) (Table II). As a bridge between metabolic signaling and epigenetic regulation, histone lactylation can dynamically respond to changes in intracellular lactate concentration and couple metabolic state with the regulation of gene expression (56).

Table II.

Comparison of histone lactylation modifications with other modification types.

Table II.

Comparison of histone lactylation modifications with other modification types.

Type of modification

CharacteristicLactylationAcetylationMethylation Phosphorylation
Modification substrateLactateAcetyl-CoASAMATP
Target amino acidLysineLysineLysine, arginineSerine, threonine, tyrosine
Regulatory enzyme systemNot fully elucidated, mechanisms incompleteHAT/HDACHMT/HDM Kinase/phosphatase
Metabolic sensitivityVery high, closely related to glycolysisHigh, affected by energy metabolismModerate dependence on metabolismLow to moderate
Distribution characteristicsTissue-specific, predominantly in metabolically active cellsUbiquitousUbiquitousUbiquitous
Degree of variationLow, changes with metabolic adaptationModerate to high, flexible changesModerate, both activation and repressionHigh, rapid response
Physiological functionReflect cellular metabolic state, links glycolysis with gene expression, regulates inflammation and cellular adaptationRelaxes chromatin structure, promotes gene transcriptionActivates/represses gene expression, regulates gene silencing imprinting andInvolved in chromosome condensation, cell cycle regulation, stress response and DNA damage repair

[i] SAM, S-adenosylmethionine; HAT, histone acetyltransferase; HDAC, histone deacetylase; HMT, histone methyltransferase; HDM, histone demethylase.

In terms of enzyme-mediated modification, the formation of lactylation may be catalyzed by lactate dehydrogenase (LDH) or other enzymes, such as acyl-CoA synthetase short-chain family member 2 and acyl-CoA synthetase family member 2 (44,5759). LDH is a key enzyme in the glycolytic pathway, whose primary function is to catalyze the reduction of pyruvate to lactate (60). Previous studies have demonstrated that LDH, by regulation of the intracellular lactate concentration, directly or indirectly affects the occurrence of lactylation modification (12). A recent study has also reported that alanyl-tRNA synthetase 1 and alanyl-tRNA synthetases 2 have been identified as intracellular L-lactate sensors and lactate transferases, which catalyzes the ATP-dependent binding of L-lactate to lysine and forms lactylation modifications (61).

Regarding the delactylation process, specific delactylases have not yet been clearly identified. However, some studies suggested that the delactylation process may involve certain members of the deacylase family. The sirtuin (SIRT) family, which consists of nicotinamide adenine dinucleotide-dependent deacetylases, is known to serve key roles in the regulation of histone acetylation and other modifications (62). For example, SIRT3 has been found to possess delactylation activity, with its activity against histone H4K16la being notably higher compared with that of other SIRT family members (including SIRT1, SIRT2, SIRT4, SIRT5, SIRT6 and SIRT7) (63). Additionally, SIRT2 has been reported to have ‘eraser’ functions in neuroblastoma (64). Future research is warranted to further explore the specific types of delactylases and their functions in tumors.

Lactylation modification also interacts with other epigenetic modifications (such as acetylation, methylation and phosphorylation) to jointly regulate gene expression (65). For example, lactylation modification may compete with acetylation for the same lysine sites, which affects gene transcription activity (64). Furthermore, lactylation might influence chromatin openness or compaction through its interaction with methylation modifications. This complex network of modifications provides a molecular basis for the multiple roles of lactylation in tumors and its potential functions in tumors characterized by pronounced metabolic abnormalities, such as laryngeal cancer, warrant in-depth investigation in future research.

Potential roles of histone lactylation modification in laryngeal cancer

Tumor metabolic reprogramming

Similar to other tumor cells, laryngeal cancer cells exhibit markedly enhanced glycolysis; even in the presence of sufficient oxygen, tumor cells tend to rely on glycolysis for energy production, a manifestation of the Warburg effect (66). This metabolic reprogramming leads to massive lactate accumulation, which then acts as a signaling molecule to regulate gene expression through histone lactylation modification. Previous studies have demonstrated that lactylation modification can activate the expression of metabolism-related genes [such as glucose transporter type 1 (GLUT1) and lactate dehydrogenase A (LDHA)], which further enhances the activity of the glycolytic pathway (67,68). This positive feedback mechanism allows tumor cells to quickly acquire energy and metabolic intermediates to meet their rapid proliferative demands.

In addition, lactylation modification may enhance tumor metabolic reprogramming by regulating the expression of key genes involved in other metabolic pathways, such as lipid metabolism and amino acid metabolism (69,70). For example, lactylation may activate the expression of fatty acid synthase and glutamine transporter 5, thereby enhances tumor cell lipid synthesis and amino acid metabolism capacity (71,72). These metabolic adaptations not only provide the material basis for tumor cell proliferation but also enhance their tolerance to adverse environments.

Shaping the tumor microenvironment

Lactate functions not only within tumor cells but also influences the shaping of the tumor microenvironment when secreted extracellularly. A previous study has reported that in HNSCC, lactate regulates the expression of the IL-11 gene through H3K9la and subsequently activates the Janus kinase 2/STAT3 signaling pathway, which leads to CD8+ T cell exhaustion, and ultimately enables tumor cells to evade immune surveillance (18). This finding indicated that lactate can inhibit antitumor immune responses by altering the function of immune cells in the tumor microenvironment. Furthermore, lactylation modification may also promote tumor-associated inflammatory responses by regulating the expression of inflammatory cytokines (73). These cytokines can not only attract immunosuppressive cells (such as myeloid-derived suppressor cells and regulatory T-cells) into the tumor microenvironment but also supports tumor cell proliferation and metastasis by promoting angiogenesis and matrix remodeling (74).

Cancer-associated fibroblasts (CAFs) are an essential component of the tumor microenvironment; the activated state CAFs is closely associated to tumor invasiveness and metastatic capability. Lactylation modification can regulate the secretion of cytokines and growth factors [such as TGF-β and vascular endothelial growth factor (VEGF)] by CAFs, which thereby promote tumor cell invasion and angiogenesis (75). Additionally, lactylation modification may lead to acidification of the tumor microenvironment, which further enhances the invasiveness and therapeutic resistance of tumor cells (76).

Cancer stem cell characteristics

Cancer stem cells, a small population of cells within tumors that possess self-renewal capabilities and multipotent differentiation potential, are considered the primary driving force for tumor invasion, metastasis and recurrence (77). Lactylation modification may enhance the stem cell properties of laryngeal cancer cells by regulating the expression of stemness-related genes [such as the stem cell transcription factor Sox2 and Octamer-binding transcription factor 4 (OCT4)] (78,79). Sox2 and OCT4 are key transcription factors in maintaining the stem cell state; high expression levels of Sox2 and OCT4 are closely associated with tumor cell invasiveness and metastatic potential and lactylation modification may activate the expression of these genes, which endows laryngeal cancer cells with stronger migratory and invasive capacities compared to adjacent normal tissue cells or precancerous lesion cells (80,81). Furthermore, lactylation modification might further enhance cancer stem cell characteristics by regulating the key genes of signaling pathways such as Wnt/β-catenin, Notch and Hedgehog (82). These pathways serve important roles in the maintenance and function of cancer stem cells and their abnormal activation leads to increased tumor invasiveness and therapeutic resistance (83). By modulating the expression of these key genes, lactylation modification may endow laryngeal cancer cells with enhanced adaptability and survival capacity, which promotes malignant progression.

Chemotherapy and radiotherapy resistance

Lactylation modifications may enhance the tolerance of laryngeal cancer cells to chemotherapy and radiotherapy through multiple mechanisms. Firstly, lactylation modifications can regulate the expression of DNA damage repair genes, such as radiation-sensitive genes [such as RAD51 recombinase (RAD51)] (84). RAD51 is a key protein in the homologous recombination repair pathway; high expression of RAD51 effectively repairs DNA double-strand breaks induced by chemotherapeutic drugs or radiotherapy and thereby protects tumor cells from the lethality of DNA damage (85). A previous study has demonstrated that inhibiting GLUT-1 with siRNA can reduce the expression of RAD51 and DNA-dependent protein kinase catalytic subunit, which weakens the DNA repair capacity and thereby increases apoptosis to enhance the radiosensitivity of laryngeal cancer stem cells (86). Secondly, lactylation modification may enhance tumor cell tolerance to oxidative stress induced by radiotherapy by regulating the expression of antioxidant genes, which thus helps cancer cells evade treatment-induced oxidative damage (44,87). In addition, lactylation modification might further enhance chemoresistance by modulating the expression of cell cycle-related genes, which delays cell cycle progression and reduces the killing effect of chemotherapeutic drugs on rapidly dividing cells (88,89).

Clinical significance of histone lactylation modification

Diagnostic and prognostic biomarkers

Research has indicated that the level of lactylation modification is associated with tumor malignancy, invasiveness and patient survival (90). The enhanced glycolysis (Warburg effect) in laryngeal cancer cells leads to lactate accumulation, which markedly increases the level of histone lactylation modification. Therefore, detecting the levels of lactylation modification in patient tissues or blood samples potentially holds promise as an important basis for the early diagnosis and assessment of laryngeal cancer in the future. In the diagnostic context, specific sites of histone lactylation modification can serve as candidate molecular markers for laryngeal cancer. Therapeutically, the dynamic changes in lactylation modification may reflect the metabolic state and epigenetic characteristics of the tumor, which provides a reference for personalized treatment. For prognostic evaluation, high levels of histone lactylation may associate with more aggressive and metastatic tumors and lower patient survival rates, whereas lower levels may indicate improved prognosis (91,92). Furthermore, the detection of lactylation modifications can be combined with other biomarkers such as programmed cell death-ligand 1 (PD-L1) to establish a multi-biomarker diagnostic and prognostic assessment system, which thereby enhances the accuracy of cancer diagnosis and the predictive capability of prognosis (93,94).

Therapeutic targets

Given the important roles of lactylation modification in tumor metabolic reprogramming, shaping the tumor microenvironment, maintaining cancer stem cell characteristics and therapeutic resistance, targeting lactate metabolism pathways or enzymes associated with lactylation modification may potentially offer novel strategies for laryngeal cancer treatment in the future.

First, drugs targeting lactate metabolism (such as LDHA inhibitors) hold notable therapeutic potential. LDHA is a key enzyme in the glycolytic pathway that catalyzes the conversion of pyruvate to lactate, which directly influences lactate accumulation and the occurrence of lactylation modification (68). LDHA inhibitors, by reducing lactate production and lowering levels of histone lactylation, can suppress tumor metabolic reprogramming and immune evasion. For example, the LDHA inhibitor FX11 has markedly inhibited the growth of various tumors, including pancreatic cancer, breast cancer and melanoma (9597), and its application in laryngeal cancer warrants further investigation.

Second, targeting enzymes associated with lactylation modification (both the lactylation ‘writers’ and potential ‘erasers’) may provide novel intervention strategies for laryngeal cancer. Lactylation enzymes (writers) are responsible for adding lactyl groups to target proteins, while de-lactylating enzymes (erasers) can remove the lactyl groups that have already been added to the proteins (98). Although the specific enzymes catalyzing lactylation and those responsible for delactylation have not been fully elucidated, ongoing studies have suggested that the dynamic regulation of lactylation serves a key role in tumor development and progression (98). By developing inhibitors against the enzymes that catalyze lactylation, it may be possible to block the occurrence of lactylation modification, which thereby inhibits tumor cell proliferation and invasion. If specific delactylases can be identified in the future, inducing their activity could potentially reverse lactylation effects and suppress malignant tumor progression.

Finally, lactylation modification may also serve as a target for combination therapies, integrating with existing immunotherapy, chemotherapy and radiotherapy regimes (99). For example, lactylation modifications promote tumor immune evasion by regulating the expression of PD-L1 (100). Thus, targeting lactylation modification could enhance the efficacy of immune checkpoint inhibitors (such as anti-PD-1/PD-L1 antibodies) (101). Furthermore, combining LDHA inhibitors with chemotherapeutic drugs could simultaneously inhibit tumor metabolism and DNA damage repair, which thereby increases chemosensitivity (102). As research on lactylation modification deepens, its potential in the clinical application for laryngeal cancer will gradually be revealed, which offers novel prospects for precision diagnosis and treatment for patients with laryngeal cancer.

Detection methods for histone lactylation modification

Mass spectrometry analysis

Mass spectrometry is one of the core techniques for studying histone lactylation modification and is primarily used to identify specific lactylation sites and quantify their modification levels (103). Owing to its high sensitivity and resolution, mass spectrometry can precisely detect the lactylation modification on histone lysine residues. Researchers typically utilize liquid chromatography-tandem mass spectrometry, in combination with proteolytic digestion and peptide separation methods, to conduct in-depth analyses of histone samples (12). Furthermore, mass spectrometry allows for quantitative analysis of dynamic changes in lactylation under different conditions. By comparing the levels of histone lactylation modifications in normal vs. tumor cells, the potential role of lactylation modification in tumorigenesis and progression may be elucidated (92). Additionally, mass spectrometry can be combined with other omics techniques (such as metabolomics and transcriptomics) to explore the association between lactylation modification, cellular metabolism and gene expression (104). Despite its advantages, mass spectrometry faces certain technical challenges; for example, the chemical properties of lactylation are similar to those of acetylation, which may lead to ambiguity in identifying modification sites (65). Furthermore, lactylation modifications are typically of low abundance, which necessitates the development of higher-resolution mass spectrometers and optimized sample preparation protocols in the future (105).

Specific antibody detection

Specific antibody detection is another important method for the study of histone lactylation modification, primarily used to assess the levels and distribution of lactylation modifications (106,107). In recent years, specific antibodies targeting particular lactylation sites (such as H3K18la and H3K23la) have been developed, which provide key tools for the detection of lactylation modifications (65). These antibodies can recognize the unique chemical structure of lactylation, which enables highly specific detection. In practice, researchers often use western blot techniques for quantitative analysis of changes in lactylation modifications under different experimental conditions (108); immunofluorescence techniques allow visualization of the spatial distribution of lactylation modifications within the nucleus (91). Additionally, co-immunoprecipitation techniques can be employed in conjunction with specific antibodies to study the interactions between lactylation modifications and other proteins or epigenetic modifications (109). However, despite the advantages of ease of operation and high sensitivity, specific antibody detection also has limitations. For example, non-specific binding of antibodies may affect data reliability (110). Furthermore, the limited availability of commercial antibodies for lactylation modifications restricts their application in large-scale studies.

Gene editing technologies

Gene editing technologies, especially the CRISPR/Cas9 system, provide powerful tools for exploring the functions of histone lactylation modification (111). With CRISPR/Cas9, researchers can knock out or overexpress genes associated to lactylation modification (such as LDHA and LDHB) to investigate the specific role of lactylation in cellular metabolism, gene expression and disease (112). The applications of CRISPR/Cas9 in this context include several aspects. First, by knocking out enzymes associated to lactylation modification, one can study the mechanisms of lactylation formation and its impact on cellular functions (113). Second, CRISPR/Cas9-mediated site-directed mutagenesis can be used to investigate the regulatory role of specific histone lactylation sites (such as H3K18 and H3K23) on gene expression and cell behavior (114). Furthermore, CRISPR/Cas9 can be combined with other omics technologies to systematically explore the functions of lactylation modification in various biological processes (115). Although CRISPR/Cas9 technology holds broad application prospects, challenges remain in its use for lactylation research. The dynamic and complex nature of histone lactylation might complicate the interpretation of gene editing results. Additionally, the off-target effects of CRISPR/Cas9 could impact the accuracy of experimental outcomes, which necessitates the optimization of experimental designs and further validation of results (116).

Mass spectrometry analysis, specific antibody detection and gene editing technologies constitute the three core methods for the study of histone lactylation modification. The integrated application of these methods not only provides important support for the basic research of lactylation modification but also lays the foundation for its clinical translation in disease diagnosis and treatment. With ongoing technological advancements, these methods will further advance in-depth research into lactylation modification.

Conclusion and outlook

Histone lactylation modification, as a novel epigenetic regulatory mechanism, serves key roles in tumor metabolic reprogramming, shaping the tumor microenvironment, maintaining cancer stem cell characteristics and mediating therapeutic resistance. Although research on the role of histone lactylation modification in laryngeal cancer is still in its early stages, the aforementioned studies have indicated that lactylation modification regulates gene expression and is involved in the occurrence and development of laryngeal cancer. This provides novel research directions and potential targets for the diagnosis, prognostic evaluation and treatment of laryngeal cancer. Future research may focus on elucidating the molecular regulatory mechanisms of lactylation modification, its interactions with other epigenetic modifications and its specific functions in laryngeal cancer. Furthermore, the development of targeted detection tools and therapeutic agents against lactylation modification will offer technical support and pave the way for clinical translation in the precise diagnosis and treatment of laryngeal cancer. Combining basic research with clinical applications, lactylation modification could become novel avenue of research in the study of laryngeal cancer, which may potentially provide novel treatments for patients in the future.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

QiaT and CH wrote the manuscript. QiaT created the tables. QizT and ZZ revised the manuscript. CH supervised the research. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

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 

Siegel RL, Giaquinto AN and Jemal A: Cancer statistics, 2024. CA Cancer J Clin. 74:12–49. 2024. View Article : Google Scholar : PubMed/NCBI

2 

Nocini R, Molteni G, Mattiuzzi C and Lippi G: Updates on larynx cancer epidemiology. Chin J Cancer Res. 32:18–25. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Liberale C, Soloperto D, Marchioni A, Monzani D and Sacchetto L: Updates on larynx cancer: risk factors and oncogenesis. Int J Mol Sci. 24:129132023. View Article : Google Scholar : PubMed/NCBI

4 

Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI

5 

Molina-Fernández E, Palacios-García JM, Moreno-Luna R, Herrero-Salado T, Ventura-Díaz J, Sánchez-Gómez S and Vilches-Arenas Á: Survival Analysis in patients with laryngeal cancer: A retrospective cohort study. Life (Basel). 13:2952023.PubMed/NCBI

6 

Yu X, Zhao H, Wang R, Chen Y, Ouyang X, Li W, Sun Y and Peng A: Cancer epigenetics: From laboratory studies and clinical trials to precision medicine. Cell Death Discov. 10:282024. View Article : Google Scholar : PubMed/NCBI

7 

Shvedunova M and Akhtar A: Modulation of cellular processes by histone and non-histone protein acetylation. Nat Rev Mol Cell Biol. 23:329–349. 2022. View Article : Google Scholar : PubMed/NCBI

8 

Li Y, Chen X and Lu C: The interplay between DNA and histone methylation: molecular mechanisms and disease implications. EMBO Rep. 22:e518032021. View Article : Google Scholar : PubMed/NCBI

9 

Armache A, Yang S, Martínez de Paz A, Robbins LE, Durmaz C, Cheong JQ, Ravishankar A, Daman AW, Ahimovic DJ, Klevorn T, et al: Histone H3.3 phosphorylation amplifies stimulation-induced transcription. Nature. 583:852–857. 2020. View Article : Google Scholar : PubMed/NCBI

10 

Mattiroli F and Penengo L: Histone ubiquitination: An integrative signaling platform in genome stability. Trends Genet. 37:566–581. 2021. View Article : Google Scholar : PubMed/NCBI

11 

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

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 

Jing F, Zhu L, Zhang J, Zhou X, Bai J, Li X, Zhang H and Li T: Multi-omics reveals lactylation-driven regulatory mechanisms promoting tumor progression in oral squamous cell carcinoma. Genome Biol. 25:2722024. View Article : Google Scholar : PubMed/NCBI

14 

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 : PubMed/NCBI

15 

Song F, Hou C, Huang Y, Liang J, Cai H, Tian G, Jiang Y, Wang Z and Hou J: Lactylome analyses suggest systematic lysine-lactylated substrates in oral squamous cell carcinoma under normoxia and hypoxia. Cell Signal. 120:1112282024. View Article : Google Scholar : PubMed/NCBI

16 

Zang Y, Wang A, Zhang J, Xia M, Jiang Z, Jia B, Lu C, Chen C, Wang S, Zhang Y, et al: Hypoxia promotes histone H3K9 lactylation to enhance LAMC2 transcription in esophageal squamous cell carcinoma. iScience. 27:1101882024. View Article : Google Scholar : PubMed/NCBI

17 

Fu C, Jiang W, Wang C, Song SJ, Tao H, Zhang XG, Li WT, Jin X, Yu BB, Hao JJ, et al: AP001885.4 promotes the proliferation of esophageal squamous cell carcinoma cells by histone lactylation- and NF-κB (p65)-dependent transcription activation and METTL3-mediated mRNA stability of c-myc. Anim Cells Syst (Seoul). 28:536–550. 2024. View Article : Google Scholar : PubMed/NCBI

18 

Wang R, Li C, Cheng Z, Li M, Shi J, Zhang Z, Jin S and Ma H: H3K9 lactylation in malignant cells facilitates CD8(+) T cell dysfunction and poor immunotherapy response. Cell Rep. 43:1146862024. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Li Y and Lu C: Targeting epigenetic Dysregulations in head and neck squamous cell carcinoma. J Dent Res. 104:225–234. 2025. View Article : Google Scholar : PubMed/NCBI

21 

Davalos V and Esteller M: Cancer epigenetics in clinical practice. CA Cancer J Clin. 73:376–424. 2023. View Article : Google Scholar : PubMed/NCBI

22 

Pierini S, Jordanov SH, Mitkova AV, Chalakov IJ, Melnicharov MB, Kunev KV, Mitev VI, Kaneva RP and Goranova TE: Promoter hypermethylation of CDKN2A, MGMT, MLH1, and DAPK genes in laryngeal squamous cell carcinoma and their associations with clinical profiles of the patients. Head Neck. 36:1103–1108. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Gallus R, Gheit T, Holzinger D, Petrillo M, Rizzo D, Petrone G, Miccichè F, Mattiucci GC, Arciuolo D, Capobianco G, et al: Prevalence of HPV infection and p16(INK4a) overexpression in surgically treated laryngeal squamous cell carcinoma. Vaccines (Basel). 10:2042022. View Article : Google Scholar : PubMed/NCBI

24 

López F, Sampedro T, Llorente JL, Domínguez F, Hermsen M, Suárez C and Alvarez-Marcos C: Utility of MS-MLPA in DNA methylation profiling in primary laryngeal squamous cell carcinoma. Oral Oncol. 50:291–297. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Weigel C, Chaisaingmongkol J, Assenov Y, Kuhmann C, Winkler V, Santi I, Bogatyrova O, Kaucher S, Bermejo JL, Leung SY, et al: DNA methylation at an enhancer of the three prime repair exonuclease 2 gene (TREX2) is linked to gene expression and survival in laryngeal cancer. Clin Epigenetics. 11:672019. View Article : Google Scholar : PubMed/NCBI

26 

Xiao Y, Zhang Y, Hu Y, Zhang X, Tan J, Yao S, Wang X and Qin Y: Advances in the study of posttranslational modifications of histones in head and neck squamous cell carcinoma. Clin Epigenetics. 16:1652024. View Article : Google Scholar : PubMed/NCBI

27 

Li L, Cui J, Li X, Zhu Y, Wu H and Zhou L: Prmt1-mediated histone H4R3me2a methylation regulates the proliferation, migration and invasion of laryngeal cancer cells by affecting the expression level of NCOA5. Front Oncol. 14:14891642024. View Article : Google Scholar : PubMed/NCBI

28 

Wang J, Wang N, Zheng Z, Che Y, Suzuki M, Kano S, Lu J, Wang P, Sun Y and Homma A: Exosomal lncRNA HOTAIR induce macrophages to M2 polarization via PI3K/p-AKT/AKT pathway and promote EMT and metastasis in laryngeal squamous cell carcinoma. BMC Cancer. 22:12082022. View Article : Google Scholar : PubMed/NCBI

29 

Banta A, Bratosin F, Golu I, Toma AO and Domuta EM: A systematic review of circulating miRNAs by multiple independent validated studies in laryngeal cancer. Diagnostics (Basel). 15:3942025. View Article : Google Scholar : PubMed/NCBI

30 

Cui W, Meng W, Zhao L, Cao H, Chi W and Wang B: TGF-β-induced long non-coding RNA MIR155HG promotes the progression and EMT of laryngeal squamous cell carcinoma by regulating the miR-155-5p/SOX10 axis. Int J Oncol. 54:2005–2018. 2019.PubMed/NCBI

31 

Jiang R, Gao MZ, Chen M, Weatherspoon DJ, Watts TL and Osazuwa-Peters N: Genetic and molecular differences in head and neck cancer based on smoking history. JAMA Otolaryngol Head Neck Surg. 151:379–388. 2025. View Article : Google Scholar : PubMed/NCBI

32 

Ferraguti G, Terracina S, Petrella C, Greco A, Minni A, Lucarelli M, Agostinelli E, Ralli M, de Vincentiis M, Raponi G, et al: Alcohol and head and neck cancer: Updates on the role of oxidative stress, genetic, epigenetics, oral microbiota, antioxidants, and alkylating agents. Antioxidants (Basel). 11:1452022. View Article : Google Scholar : PubMed/NCBI

33 

Zhou YQ, Jiang JX, He S, Li YQ, Cheng XX, Liu SQ, Wei PP, Guan XY, Ong CK, Wang VY, et al: Epstein-Barr virus hijacks histone demethylase machinery to drive epithelial malignancy progression through KDM5B upregulation. Signal Transduct Target Ther. 10:832025. View Article : Google Scholar : PubMed/NCBI

34 

Ge J, Meng Y, Guo J, Chen P, Wang J, Shi L, Wang D, Qu H, Wu P, Fan C, et al: Human papillomavirus-encoded circular RNA circE7 promotes immune evasion in head and neck squamous cell carcinoma. Nat Commun. 15:86092024. View Article : Google Scholar : PubMed/NCBI

35 

Motoc GV, Juncar RI, Moca AE, Motoc O, Vaida LL and Juncar M: The relationship between age, gender, BMI, diet, salivary pH and periodontal pathogenic bacteria in children and adolescents: A cross-sectional study. Biomedicines. 11:23742023. View Article : Google Scholar : PubMed/NCBI

36 

Chen F, He X, Xu W, Zhou L, Liu Q, Chen W, Zhu WG and Zhang J: Chromatin lysine acylation: On the path to chromatin homeostasis and genome integrity. Cancer Sci. 115:3506–3519. 2024. View Article : Google Scholar : PubMed/NCBI

37 

Wang H and Helin K: Roles of H3K4 methylation in biology and disease. Trends Cell Biol. 35:115–128. 2025. View Article : Google Scholar : PubMed/NCBI

38 

Guo Y, Zhao S and Wang GG: Polycomb gene silencing mechanisms: PRC2 chromatin targeting, H3K27me3 ‘Readout’, and phase separation-based compaction. Trends Genet. 37:547–565. 2021. View Article : Google Scholar : PubMed/NCBI

39 

Gong P, Guo Z, Wang S, Gao S and Cao Q: Histone phosphorylation in DNA damage response. Int J Mol Sci. 26:24052025. View Article : Google Scholar : PubMed/NCBI

40 

Shu Q, Liu Y and Ai H: The emerging role of the histone H2AK13/15 ubiquitination: Mechanisms of writing, reading, and erasing in DNA damage repair and disease. Cells. 14:3072025. View Article : Google Scholar : PubMed/NCBI

41 

Rabinowitz JD and Enerbäck S: Lactate: the ugly duckling of energy metabolism. Nat Metab. 2:566–571. 2020. View Article : Google Scholar : PubMed/NCBI

42 

Fendt SM: 100 years of the Warburg effect: A cancer metabolism endeavor. Cell. 187:3824–3828. 2024. View Article : Google Scholar : PubMed/NCBI

43 

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

44 

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

45 

Zhong X, He X, Wang Y, Hu Z, Huang H, Zhao S, Wei P and Li D: Warburg effect in colorectal cancer: The emerging roles in tumor microenvironment and therapeutic implications. J Hematol Oncol. 15:1602022. View Article : Google Scholar : PubMed/NCBI

46 

Zhi S, Chen C, Huang H, Zhang Z, Zeng F and Zhang S: Hypoxia-inducible factor in breast cancer: Role and target for breast cancer treatment. Front Immunol. 15:13708002024. View Article : Google Scholar : PubMed/NCBI

47 

Zhao LP, Zheng RR, Kong RJ, Huang CY, Rao XN, Yang N, Chen AL, Yu XY, Cheng H and Li SY: Self-delivery ternary bioregulators for photodynamic amplified immunotherapy by tumor microenvironment reprogramming. ACS Nano. 16:1182–1197. 2022. View Article : Google Scholar : PubMed/NCBI

48 

Galle E, Wong CW, Ghosh A, Desgeorges T, Melrose K, Hinte LC, Castellano-Castillo D, Engl M, de Sousa JA, Ruiz-Ojeda FJ, et al: H3K18 lactylation marks tissue-specific active enhancers. Genome Biol. 23:2072022. View Article : Google Scholar : PubMed/NCBI

49 

Chu X, Di C, Chang P, Li L, Feng Z, Xiao S, Yan X, Xu X, Li H, Qi R, et al: Lactylated histone H3K18 as a potential biomarker for the diagnosis and predicting the severity of septic shock. Front Immunol. 12:7866662022. View Article : Google Scholar : PubMed/NCBI

50 

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

51 

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

52 

Duan X, Xing Z, Qiao L, Qin S, Zhao X, Gong Y and Li X: The role of histone post-translational modifications in cancer and cancer immunity: Functions, mechanisms and therapeutic implications. Front Immunol. 15:14952212024. View Article : Google Scholar : PubMed/NCBI

53 

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

54 

He W, Li Q and Li X: Acetyl-CoA regulates lipid metabolism and histone acetylation modification in cancer. Biochim Biophys Acta Rev Cancer. 1878:1888372023. View Article : Google Scholar : PubMed/NCBI

55 

Neganova ME, Klochkov SG, Aleksandrova YR and Aliev G: Histone modifications in epigenetic regulation of cancer: Perspectives and achieved progress. Semin Cancer Biol. 83:452–471. 2022. View Article : Google Scholar : PubMed/NCBI

56 

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

57 

Liu J, Zhao F and Qu Y: Lactylation: A novel post-translational modification with clinical implications in CNS diseases. Biomolecules. 14:11752024. View Article : Google Scholar : PubMed/NCBI

58 

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 : PubMed/NCBI

59 

Chen J, Feng Q, Qiao Y, Pan S, Liang L, Liu Y, Zhang X, Liu D and Liu Z and Liu Z: ACSF2 and lysine lactylation contribute to renal tubule injury in diabetes. Diabetologia. 67:1429–1443. 2024. View Article : Google Scholar : PubMed/NCBI

60 

Sun W, Jia M, Feng Y and Cheng X: Lactate is a bridge linking glycolysis and autophagy through lactylation. Autophagy. 19:3240–3241. 2023. View Article : Google Scholar : PubMed/NCBI

61 

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

62 

Wu QJ, Zhang TN, Chen HH, Yu XF, Lv JL, Liu YY, Liu YS, Zheng G, Zhao JQ, Wei YF, et al: The sirtuin family in health and disease. Signal Transduct Target Ther. 7:4022022. View Article : Google Scholar : PubMed/NCBI

63 

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

64 

Zu H, Li C, Dai C, Pan Y, Ding C, Sun H, Zhang X, Yao X, Zang J and Mo X: SIRT2 functions as a histone delactylase and inhibits the proliferation and migration of neuroblastoma cells. Cell Discov. 8:542022. View Article : Google Scholar : PubMed/NCBI

65 

Wang T, Ye Z, Li Z, Jing DS, Fan GX, Liu MQ, Zhuo QF, Ji SR, Yu XJ, Xu XW and Qin Y: Lactate-induced protein lactylation: A bridge between epigenetics and metabolic reprogramming in cancer. Cell Prolif. 56:e134782023. View Article : Google Scholar : PubMed/NCBI

66 

Liu C, Jin Y and Fan Z: The mechanism of warburg effect-induced chemoresistance in cancer. Front Oncol. 11:6980232021. View Article : Google Scholar : PubMed/NCBI

67 

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

68 

Xia J, Qiao Z, Hao X and Zhang Y: LDHA-induced histone lactylation mediates the development of osteoarthritis through regulating the transcription activity of TPI1 gene. Autoimmunity. 57:23848892024. View Article : Google Scholar : PubMed/NCBI

69 

Pucino V, Certo M, Bulusu V, Cucchi D, Goldmann K, Pontarini E, Haas R, Smith J, Headland SE, Blighe K, et al: Lactate buildup at the site of chronic inflammation promotes disease by inducing CD4(+) T cell metabolic rewiring. Cell Metab. 30:1055–1074.e8. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Xu K, Zhang K, Wang Y and Gu Y: Comprehensive review of histone lactylation: Structure, function, and therapeutic targets. Biochem Pharmacol. 225:1163312024. View Article : Google Scholar : PubMed/NCBI

71 

Chen X, Huang W and Zhang J, Li Y, Xing Z, Guo L, Jiang H and Zhang J: High-intensity interval training induces lactylation of fatty acid synthase to inhibit lipid synthesis. BMC Biol. 21:1962023. View Article : Google Scholar : PubMed/NCBI

72 

Yoo HC, Park SJ, Nam M, Kang J, Kim K, Yeo JH, Kim JK, Heo Y, Lee HS, Lee MY, et al: A variant of SLC1A5 is a mitochondrial glutamine transporter for metabolic reprogramming in cancer cells. Cell Metab. 31:267–283.e12. 2020. View Article : Google Scholar : PubMed/NCBI

73 

Luo Y, Li L, Chen X, Gou H, Yan K and Xu Y: Effects of lactate in immunosuppression and inflammation: Progress and prospects. Int Rev Immunol. 41:19–29. 2022. View Article : Google Scholar : PubMed/NCBI

74 

Gómez-Valenzuela F, Escobar E, Pérez-Tomás R and Montecinos VP: The inflammatory profile of the tumor microenvironment, orchestrated by cyclooxygenase-2, promotes epithelial-mesenchymal transition. Front Oncol. 11:6867922021. View Article : Google Scholar : PubMed/NCBI

75 

Wang X, Qu Y, Ji J, Liu H, Luo H, Li J and Han X: Colorectal cancer cells establish metabolic reprogramming with cancer-associated fibroblasts (CAFs) through lactate shuttle to enhance invasion, migration, and angiogenesis. Int Immunopharmacol 143(Pt 2). 1134702024. View Article : Google Scholar : PubMed/NCBI

76 

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

77 

Huang T, Song X, Xu D, Tiek D, Goenka A, Wu B, Sastry N, Hu B and Cheng SY: Stem cell programs in cancer initiation, progression, and therapy resistance. Theranostics. 10:8721–8743. 2020. View Article : Google Scholar : PubMed/NCBI

78 

Feng F, Wu J, Chi Q, Wang S, Liu W, Yang L, Song G, Pan L, Xu K and Wang C: Lactylome analysis unveils lactylation-dependent mechanisms of stemness remodeling in the liver cancer stem cells. Adv Sci (Weinh). 11:e24059752024. View Article : Google Scholar : PubMed/NCBI

79 

Mohiuddin IS, Wei SJ and Kang MH: Role of OCT4 in cancer stem-like cells and chemotherapy resistance. Biochim Biophys Acta Mol Basis Dis. 1866:1654322020. View Article : Google Scholar : PubMed/NCBI

80 

Montoro-Jiménez I, Granda-Díaz R, Menéndez ST, Prieto-Fernández L, Otero-Rosales M, Álvarez-González M, García-de-la-Fuente V, Rodríguez A, Rodrigo JP, Álvarez-Teijeiro S, et al: Combined PIK3CA and SOX2 gene amplification predicts laryngeal cancer risk beyond histopathological grading. Int J Mol Sci. 25:26952024. View Article : Google Scholar : PubMed/NCBI

81 

Ma Y, Chen Z and Yu G: microRNA-139-3p inhibits malignant behaviors of laryngeal cancer cells via the KDM5B/SOX2 Axis and the Wnt/β-catenin pathway. Cancer Manag Res. 12:9197–9209. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Clara JA, Monge C, Yang Y and Takebe N: Targeting signalling pathways and the immune microenvironment of cancer stem cells-a clinical update. Nat Rev Clin Oncol. 17:204–232. 2020. View Article : Google Scholar : PubMed/NCBI

83 

Wang J, Yu H, Dong W, Zhang C, Hu M, Ma W, Jiang X, Li H, Yang P and Xiang D: N6-methyladenosine-mediated up-regulation of FZD10 regulates liver cancer stem cells' properties and lenvatinib resistance through WNT/β-catenin and hippo signaling pathways. Gastroenterology. 164:990–1005. 2023. View Article : Google Scholar : PubMed/NCBI

84 

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

85 

Wang Z, Jia R, Wang L, Yang Q, Hu X, Fu Q, Zhang X, Li W and Ren Y: The emerging roles of Rad51 in cancer and its potential as a therapeutic target. Front Oncol. 12:9355932022. View Article : Google Scholar : PubMed/NCBI

86 

Zhong JT, Yu Q, Zhou SH, Yu E, Bao YY, Lu ZJ and Fan J: GLUT-1 siRNA enhances radiosensitization of laryngeal cancer stem cells via enhanced DNA damage, cell cycle redistribution, and promotion of apoptosis in vitro and in vivo. Onco Targets Ther. 12:9129–9142. 2019. View Article : Google Scholar : PubMed/NCBI

87 

Wu Y, Song Y, Wang R and Wang T: Molecular mechanisms of tumor resistance to radiotherapy. Mol Cancer. 22:962023. View Article : Google Scholar : PubMed/NCBI

88 

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

89 

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

90 

Wang G, Zou X, Chen Q, Nong W, Miao W, Luo H and Qu S: The relationship and clinical significance of lactylation modification in digestive system tumors. Cancer Cell Int. 24:2462024. View Article : Google Scholar : PubMed/NCBI

91 

Peng T, Sun F, Yang JC, Cai MH, Huai MX, Pan JX, Zhang FY and Xu LM: Novel lactylation-related signature to predict prognosis for pancreatic adenocarcinoma. World J Gastroenterol. 30:2575–2602. 2024. View Article : Google Scholar : PubMed/NCBI

92 

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

93 

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 : PubMed/NCBI

94 

Sun X, Dong H, Su R, Chen J, Li W, Yin S and Zhang C: Lactylation-related gene signature accurately predicts prognosis and immunotherapy response in gastric cancer. Front Oncol. 14:14855802024. View Article : Google Scholar : PubMed/NCBI

95 

Kitamura F, Semba T, Yasuda-Yoshihara N, Yamada K, Nishimura A, Yamasaki J, Nagano O, Yasuda T, Yonemura A, Tong Y, et al: Cancer-associated fibroblasts reuse cancer-derived lactate to maintain a fibrotic and immunosuppressive microenvironment in pancreatic cancer. JCI Insight. 8:e1630222023. View Article : Google Scholar : PubMed/NCBI

96 

Truong Hoang Q, Huynh KA, Nguyen Cao TG, Kang JH, Dang XN, Ravichandran V, Kang HC, Lee M, Kim JE, Ko YT, et al: Piezocatalytic 2D WS(2) nanosheets for ultrasound-triggered and mitochondria-targeted piezodynamic cancer therapy synergized with energy metabolism-targeted chemotherapy. Adv Mater. 35:e23004372023. View Article : Google Scholar : PubMed/NCBI

97 

Wang Z, Wu M, Jiang Y, Zhou J, Chen S, Wang Q, Sun H, Deng Y, Zhou Z and Sun M: Biomimetic calcium-chelation nanoparticles reprogram tumor metabolism to enhance antitumor immunity. J Control Release. 380:362–374. 2025. View Article : Google Scholar : PubMed/NCBI

98 

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

99 

Pai S, Yadav VK, Kuo KT, Pikatan NW, Lin CS, Chien MH, Lee WH, Hsiao M, Chiu SC, Yeh CT and Tsai JT: PDK1 Inhibitor BX795 Improves cisplatin and radio-efficacy in oral squamous cell carcinoma by downregulating the PDK1/CD47/Akt-mediated glycolysis signaling pathway. Int J Mol Sci. 22:114922021. View Article : Google Scholar : PubMed/NCBI

100 

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 : PubMed/NCBI

101 

Li T, Xu D, Ruan Z, Zhou J, Sun W, Rao B and Xu H: Metabolism/immunity dual-regulation thermogels potentiating immunotherapy of glioblastoma through lactate-excretion inhibition and PD-1/PD-L1 blockade. Adv Sci (Weinh). 11:e23101632024. View Article : Google Scholar : PubMed/NCBI

102 

Littleflower AB, Parambil ST, Antony GR and Subhadradevi L: The determinants of metabolic discrepancies in aerobic glycolysis: Providing potential targets for breast cancer treatment. Biochimie. 220:107–121. 2024. View Article : Google Scholar : PubMed/NCBI

103 

Bao C, Ma Q, Ying X, Wang F, Hou Y, Wang D, Zhu L, Huang J and He C: Histone lactylation in macrophage biology and disease: From plasticity regulation to therapeutic implications. EBioMedicine. 111:1055022025. View Article : Google Scholar : PubMed/NCBI

104 

Liu X, Wang J, Lao M, Liu F, Zhu H, Man K and Zhang J: Study on the effect of protein lysine lactylation modification in macrophages on inhibiting periodontitis in rats. J Periodontol. 95:50–63. 2024. View Article : Google Scholar : PubMed/NCBI

105 

Li H, Sun L, Gao P and Hu H: Lactylation in cancer: Current understanding and challenges. Cancer Cell. 42:1803–1807. 2024. View Article : Google Scholar : PubMed/NCBI

106 

Yang W, Wang P, Cao P, Wang S, Yang Y, Su H and Nashun B: Hypoxic in vitro culture reduces histone lactylation and impairs pre-implantation embryonic development in mice. Epigenetics Chromatin. 14:572021. View Article : Google Scholar : PubMed/NCBI

107 

Cui Z, Li Y, Lin Y, Zheng C, Luo L, Hu D, Chen Y, Xiao Z and Sun Y: Lactylproteome analysis indicates histone H4K12 lactylation as a novel biomarker in triple-negative breast cancer. Front Endocrinol (Lausanne). 15:13286792024. View Article : Google Scholar : PubMed/NCBI

108 

Dai W, Wu G, Liu K, Chen Q, Tao J, Liu H and Shen M: Lactate promotes myogenesis via activating H3K9 lactylation-dependent up-regulation of Neu2 expression. J Cachexia Sarcopenia Muscle. 14:2851–2865. 2023. View Article : Google Scholar : PubMed/NCBI

109 

Dong F, Yin H and Zheng Z: Hypoxia-inducible factor-1α 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 : PubMed/NCBI

110 

Zhao W, Xin J, Yu X, Li Z and Li N: Recent advances of lysine lactylation in prokaryotes and eukaryotes. Front Mol Biosci. 11:15109752025. View Article : Google Scholar : PubMed/NCBI

111 

Sar P and Dalai S: CRISPR/Cas9 in epigenetics studies of health and disease. Prog Mol Biol Transl Sci. 181:309–343. 2021. View Article : Google Scholar : PubMed/NCBI

112 

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

113 

Mazzio E, Badisa R, Mack N, Cassim S, Zdralevic M, Pouyssegur J and Soliman KFA: Whole-transcriptome analysis of fully viable energy efficient glycolytic-null cancer cells established by double genetic knockout of lactate dehydrogenase A/B or glucose-6-phosphate isomerase. Cancer Genomics Proteomics. 17:469–497. 2020. View Article : Google Scholar : PubMed/NCBI

114 

Miles LB, Calcinotto V, Oveissi S, Serrano RJ, Sonntag C, Mulia O, Lee C and Bryson-Richardson RJ: CRIMP: A CRISPR/Cas9 insertional mutagenesis protocol and toolkit. Nat Commun. 15:50112024. View Article : Google Scholar : PubMed/NCBI

115 

Meyers S, Demeyer S and Cools J: CRISPR screening in hematology research: From bulk to single-cell level. J Hematol Oncol. 16:1072023. View Article : Google Scholar : PubMed/NCBI

116 

Merlin JPJ and Abrahamse H: Optimizing CRISPR/Cas9 precision: Mitigating off-target effects for safe integration with photodynamic and stem cell therapies in cancer treatment. Biomed Pharmacother. 180:1175162024. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2025
Volume 30 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tong Q, Huang C, Tong Q and Zhang Z: Histone lactylation: A new frontier in laryngeal cancer research (Review). Oncol Lett 30: 421, 2025.
APA
Tong, Q., Huang, C., Tong, Q., & Zhang, Z. (2025). Histone lactylation: A new frontier in laryngeal cancer research (Review). Oncology Letters, 30, 421. https://doi.org/10.3892/ol.2025.15167
MLA
Tong, Q., Huang, C., Tong, Q., Zhang, Z."Histone lactylation: A new frontier in laryngeal cancer research (Review)". Oncology Letters 30.3 (2025): 421.
Chicago
Tong, Q., Huang, C., Tong, Q., Zhang, Z."Histone lactylation: A new frontier in laryngeal cancer research (Review)". Oncology Letters 30, no. 3 (2025): 421. https://doi.org/10.3892/ol.2025.15167