Lactate and lactylation in gastrointestinal cancer: Current progress and perspectives (Review)
- Authors:
- Published online on: November 8, 2024 https://doi.org/10.3892/or.2024.8839
- Article Number: 6
-
Copyright: © He et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Gastrointestinal (GI) cancers represent more than 25% of all cancer incidence and 35% of cancer-related mortality globally (1). These malignancies include colorectal cancer (CRC) (2), gastric (3), esophageal (4) and pancreatic cancer (5), each contributing notably to morbidity and mortality worldwide. Despite advancements in diagnosis and treatment, prognosis for patients with GI cancer remains poor, primarily due to late-stage detection and the aggressive nature of these tumors. Understanding the molecular mechanisms underlying GI cancer progression is key for developing more effective therapeutic strategies.
A hallmark of cancer metabolism is the Warburg effect, characterized by increased glycolysis and lactate production even in the presence of oxygen (6,7). This metabolic reprogramming supports rapid cell proliferation and creates an acidic tumor microenvironment (TME) that promotes invasion, metastasis and immune evasion. Lactate, traditionally viewed as a metabolic waste product, has emerged as a key player in cancer biology, serving as both a signaling molecule and an energy source (7). Lactate influences various aspects of tumor progression by modifying the TME, promoting immune evasion and enhancing cancer cell survival and metastasis (8–10).
Recent advances have identified lactylation, a post-translational modification driven by lactate, as a key regulatory mechanism linking cellular metabolism with gene expression (8,11). Histone lactylation, in particular, has emerged as a key process influencing chromatin structure and function, thereby affecting gene transcription (8). This modification underscores the intricate interplay between metabolism and epigenetics, highlighting the potential for novel therapeutic targets in cancer treatment (11).
Lactate and lactylation modulate various cellular processes, including inflammation, immune response and proliferation (12). In GI cancers, these modifications are hypothesized to contribute to tumor development and progression (13). Elucidating the mechanisms underlying lactate production and lactylation may identify new biomarkers for early detection and prognosis, as well as novel therapeutic strategies to disrupt these metabolic and epigenetic pathways.
The present review aimed to provide a comprehensive overview of current progress in understanding lactate and lactylation in GI cancers, the biochemical basis of these processes, impact on the TME and their clinical relevance. The present review highlights the potential of targeting lactate metabolism and lactylation as innovative approaches for treating GI cancer.
Lactate metabolism in GI cancer
Glycolysis and the Warburg effect
Cancer cells possess unique metabolic traits distinct from normal cells, notably the Warburg effect (14,15). This phenomenon involves cancer cells preferring energy production via glycolysis followed by lactate fermentation in cytosol, despite sufficient oxygen for oxidative phosphorylation. This metabolic shift facilitates rapid proliferation of tumor cells and notably alters the TME. The excess lactate produced is exported, leading to acidification of the TME, which degrades the extracellular matrix and promotes tumor invasion and metastasis. Additionally, the acidic environment suppresses immune cell function, helping cancer cells evade the immune system. This shift also supports biosynthetic processes essential for rapid cancer cell proliferation (7). Typically, cells generate ATP through oxidative phosphorylation in mitochondria, producing up to 36 ATP/glucose molecule. Conversely, glycolysis in cytoplasm yields only 2 ATP/glucose molecule but occurs at a much faster rate, enabling rapid ATP production. Cancer cells enhance glycolysis through various mechanisms. Tumor cells often show elevated levels of glycolytic enzymes such as hexokinase, phosphofructokinase and pyruvate kinase, facilitating glucose to pyruvate conversion and promoting glycolytic flux (8). Oncogenes such as MYC and RAS upregulate glycolysis, while tumor suppressors such as p53, which typically inhibit glycolysis, are frequently mutated or inactivated in cancer. Hypoxic conditions within tumors stabilize hypoxia-inducible factor (HIF)-1, which activates genes involved in glycolysis, including glucose transporter 1 and glycolytic enzymes. HIF-1 also induces lactate dehydrogenase (LDH) expression, converting pyruvate to lactate (12). The Warburg effect notably influences tumor progression. Elevated glycolytic activity leads to high lactate production, exported via monocarboxylate transporters (MCTs), contributing to acidification of the TME (10). The acidic environment promotes tumor invasion and metastasis by degrading extracellular matrix and inhibiting immune cell function while selecting aggressive, apoptosis-resistant cancer cells. Additionally, glycolysis intermediaries support biosynthetic pathways essential for the synthesis of nucleotides, amino acids and lipids required by rapidly proliferating cancer cells (16). Non-malignant cells, such as tumor-associated macrophages (TAMs) and cancer-associated fibroblasts, also contribute to lactate accumulation in the TME through the reverse Warburg effect (16). Hypoxic tumor cells secrete lactate, which normoxic tumor cells take up, facilitating glucose diffusion towards hypoxic cells. This lactate-based metabolic symbiosis supports both hypoxic and normoxic cancer cells (11). Studies indicate lactate concentration is associated with cancer grade and prognosis (17,18). Isotope tracer measurements reveal rapid lactate exchange between tumors and circulation, with tumors converting pyruvate to lactate faster than adjacent benign tissue (17,18).
In GI cancers, including CRC, gastric, esophageal and pancreatic cancers, the Warburg effect is prominent. These tumors exhibit high glycolytic rates and elevated lactate levels, which are associated with poor prognosis and therapy resistance (6). Targeting glycolysis and its regulatory pathways offers a promising therapeutic strategy for GI cancer. Inhibitors of key glycolytic enzymes, lactate transporters and HIF-1 are under investigation for their potential to disrupt metabolic adaptability of cancer cells and enhance treatment outcomes (8). Recent studies have also underscored the role of glycolysis in modulating the immune response within the TME (12,19). For example, lactate inhibits cytolytic functions of cytotoxic T cells and natural killer (NK) cells by reducing the production of key molecules such as perforin, granzyme and IFN-γ, and by downregulating signaling pathways such as nuclear factor of activated T cells and Peroxisome proliferator-activated receptor (PPARγ), ultimately facilitating tumor immune evasion (19). Understanding the interaction between cancer metabolism and immune regulation is key for developing effective immunotherapies. In summary, the Warburg effect is a key aspect of cancer metabolism that promotes tumor cell proliferation, survival and invasiveness. Elucidating the molecular mechanisms of this metabolic reprogramming may identify novel therapeutic targets for combating GI cancers.
Lactate transport and accumulation
Lactagenesis, or augmented lactate production, has been proposed as an explanation for the Warburg effect that drives carcinogenesis. This process encompasses five steps: Enhanced glucose uptake, increased expression and activity of glycolytic enzymes, decreased mitochondrial function, elevated lactate generation, accumulation and release and upregulation of MCT1/4 for accelerated lactate shuttling (Fig. 1) (20).
Lactate, a byproduct of the Warburg effect, serves a pivotal role in the metabolic reprogramming of cancer cells. Its accumulation in the TME, facilitated by specialized transporters, notably impacts tumor progression, immune evasion and metastasis. Understanding lactate transport and accumulation mechanisms may provide potential therapeutic targets for GI cancer (19). Lactate crosses the cell membrane via MCTs, a family of proton-linked transporters mediating bidirectional transfer of lactate and other MCs, such as pyruvate and ketone bodies. The primary MCTs involved in cancer cell lactate transport are MCT1 and MCT4. MCT1 is widely expressed by various tissues, including skeletal muscle, heart, brain, liver, and endothelial cells (21). It plays a crucial role in facilitating lactate uptake and export depending on the metabolic needs of the cell and the tissue environment. In tumor cells, MCT1 imports lactate to support oxidative metabolism and prevent toxic intracellular lactate accumulation (21). MCT4 primarily exports lactate and is upregulated in highly glycolytic cells, such as those in the hypoxic tumor core (21). This transporter maintains intracellular pH by removing excess lactate, thereby supporting continuous glycolytic activity. Several factors, including HIF-1 and c-Myc, regulate MCT1 and MCT4 expression. HIF-1, stabilized under hypoxic conditions, upregulates glycolytic enzymes and lactate transporters to adapt to low-oxygen environments (22).
Elevated glycolytic activity in cancer cells leads to excessive lactate production, which is exported into the TME via MCTs. This lactate export, coupled with proton co-transport, acidifies the extracellular space, creating a low-pH environment that promotes tumor invasion and metastasis (21). Acidification activates proteolytic enzymes that degrade the extracellular matrix and select for more aggressive cancer cell phenotypes (23). Lactate accumulation in the TME also inhibits immune cells, including cytotoxic T and NK cells, by suppressing cytokine production and decreasing their cytolytic activity, thus enabling tumor cells to evade immune surveillance (24). Additionally, lactate acts as a signaling molecule promoting angiogenesis by inducing vascular endothelial growth factor (VEGF) expression, facilitating novel blood vessel formation and providing the tumor with increased oxygen and nutrient supply, thereby supporting continued tumor growth (8).
Mechanisms and roles of lactylation in cancer
Discovery and mechanism of lactylation
Lactylation, first identified as a post-translational modification by Zhang et al (25), involves addition of lactate molecules to lysine residues on histones, termed histone lactylation. This followed an investigation into the non-metabolic functions of lactate, revealing its role beyond traditional metabolic contexts (25). Advanced mass spectrometry techniques have facilitated the precise detection and characterization of histone lactylation, showing its occurrence under both normoxic and hypoxic conditions, indicating its potential relevance in numerous physiological and pathological contexts, including cancer (26). Protein lactylation, especially histone lactylation, involves covalent attachment of lactate to the ε-amino group of lysine residues. This process is similar to other acylation modifications such as acetylation and methylation, but uniquely uses lactate as the modifying group. The lactylation begins with lactate production, where the Warburg effect in cancer cells increases lactate production via glycolysis, with pyruvate reduced to lactate by LDH (27). For lactylation to occur, lactate must be activated, potentially due to the formation of lactyl-CoA, a high-energy intermediary that donates the lactate moiety to target proteins. The exact enzymatic pathways and cofactors involved in this activation remain under investigation (8). The transfer of lactate to lysine residues on histones and other proteins is hypothesized to be facilitated by specific enzymes, though definitive lactyltransferases have not yet been identified. It is hypothesized that enzymes with acyltransferase activity may catalyze this process (8). Potential candidates include histone acetyltransferases (HATs), which catalyze addition of acetyl groups to histones, although direct evidence linking HATs to lactylation is lacking. Sirtuins, a family of NAD+-dependent deacetylases, may serve a role in reversing lactylation, similar to their role in deacetylation; understanding the interplay between lactylation and deacetylation may reveal more about the regulatory mechanisms underlying lactylation (28,29). The involvement of cofactors such as NAD+ and acetyl-CoA analogs such as lactyl-CoA suggests a complex regulatory network, potentially influencing the availability and activity of lactylation enzymes. In GI cancers, lactylation represents a crucial regulatory mechanism linking metabolic reprogramming to gene expression (30,31).
Similar to other epigenetic modifications such as methylation and acetylation, lactylation is regulated by writers (enzymes that add lactyl groups), erasers (enzymes that remove lactyl groups) and readers (proteins that recognize lactylation and mediate its effects) (32). Lactyl-CoA, detected in mammalian cells by liquid chromatography mass spectrometry, serves as the substrate for enzymatic lactylation, while lactyl-glutathione participates in non-enzymatic lactylation (33). The enzyme p300 has been identified as a lactylation writer and may share functions with other epigenetic modifiers, indicating potential overlaps in enzymatic regulation of different epigenetic markers (34).
Histone lactylation and gene expression
Histone lactylation, a key post-translational modification, affects chromatin structure and function by loosening chromatin, thereby facilitating transcription of genes involved in tumor progression, invasion, and immune evasion (35). This modification links cellular metabolism to gene regulation by adding lactate molecules to lysine residues on histones, influencing chromatin dynamics and gene expression (36). Metabolic shifts in cancer cells directly impact the epigenetic landscape and drive tumor progression. By altering the chemical properties of histone proteins, histone lactylation modulates the interactions between histones and DNA, as well as the binding of regulatory proteins to chromatin (37). Understanding the precise molecular mechanisms of lactylation is key for developing targeted therapies aimed at disrupting this modification to hinder cancer progression. Neutralizing the positive charge of histones via lactylation reduces their affinity for negatively charged DNA, resulting in a more relaxed chromatin structure that facilitates transcriptional activation (25).
Histone lactylation activates transcription of genes involved in metabolic processes, immune responses and cell stress pathways (38–40). Research has shown that lactylation induces transcriptional activation in response to metabolic changes within cells (41). In cancer cells, histone lactylation upregulates genes associated with glycolysis and other metabolic pathways, ensuring metabolic needs of rapidly proliferating tumor cells are met, thereby promoting sustained proliferation and survival. Additionally, lactylation regulates immune response genes (42,43). For example, in macrophages, histone lactylation activates genes involved in the anti-inflammatory response, highlighting its potential role in modulating the tumor immune landscape (40).
Under cellular stress conditions such as hypoxia, lactylation activates stress response genes, aiding cells in adapting to adverse conditions, which is key in the TME where cancer cells are subject to fluctuating oxygen and nutrient levels. Identifying specific genes and pathways regulated by histone lactylation is an area of active research, such as mapping lactylation sites across the genome and associating them with changes in gene expression (44,45). Techniques such as chromatin immunoprecipitation followed by sequencing have identified that lactylation is enriched at promoters and enhancers of actively transcribed genes (46,47).
Functional studies have demonstrated that genes regulated by lactylation participate in key cellular processes, including cell cycle regulation, apoptosis and DNA repair (48,49). For example, lactylation of histone H3 at lysine 18 (H3K18la) activates genes associated with cell proliferation and survival (48,49). Understanding the mechanisms and consequences of histone lactylation provides insights into the metabolic-epigenetic interplay in cancer and may reveal novel therapeutic targets. By modulating chromatin structure and function, lactylation activates genes that drive tumor progression, immune evasion and adaptation to stress.
Impact on the TME
Cancer cells export lactate and protons via MCTs, mainly MCT1 and MCT4, thereby lowering the extracellular pH and creating an acidic environment (50–52). This acidification activates proteolytic enzymes such as MMPs, which degrade the extracellular matrix and facilitate tumor cell invasion. Additionally, acidic pH enhances cancer cell migration and invasion by inducing epithelial-mesenchymal transition (EMT), increasing cell motility (53,54).
Reprogrammed metabolism, a hallmark of cancer, links lactylation and cell metabolism (55,56). Histone lactylation connects metabolic reprogramming with abnormal gene expression in cancer cells. Metabolic changes alter lactate levels, reshaping the histone lactylation landscape and transcriptomic profile to adapt to metabolic reprogramming (57). In non-small cell lung cancer, lactate modulates metabolism by altering the expression of glycolytic and tricarboxylic acid cycle enzymes through promoter histone lactylation. Histone lactylation is associated with arginase (Arg)1 expression in TAMs and enhances VEGFA transcription during TAM polarization (55).
Immunosuppressive effects of lactylation
In addition to serving as an intermediate metabolite in energy and biosynthetic pathways, lactate accumulates during local inflammation, linking it to tumor-associated inflammation, a hallmark of cancer (19). Macrophages, key for regulating immune response and maintaining tissue homeostasis, exhibit plasticity modulated by epigenetic dynamics during inflammation (58). Macrophages exist in two types: Proinflammatory M1 and immune regulatory M2. The transition from M1 to M2 is vital for restoring immune homeostasis. B cell adapter for PI3K facilitates this transition by elevating lactate production, enhancing histone lactylation and upregulating genes such as Forkhead box protein O1 and glycogen synthase kinase-3β (GSK3β) (59). Lactate-derived histone lactylation induces expression of homeostatic genes such as Arg1, which is highly expressed by immunosuppressive myeloid-derived suppressor cells and TAMs during the macrophage transition from M1 to M2 (60). Lysine lactylation is considered a consequence rather than a cause of macrophage activation, consistent with Arg1-dependent metabolic rewiring during inflammation (60). TAMs exhibit an M1 phenotype with anti-tumor activity at tumor initiation but shift to an M2 phenotype during cancer progression (61). Lactate mediates immunosuppressive effects of efferocytosis under hypoxia by inducing anti-inflammatory genes (62). While high lactate and low pH in inflamed tissues under hypoxia can aid pathogen clearance by confining T cells to the inflammatory site, this is detrimental during tumor-associated inflammation, suppressing cytolytic function of CD8+ T cells and inducing the T helper 17 phenotype in CD4+ T cells (63). Under hypoxia, lactate mediates immunosuppressive effects by inducing expression of anti-inflammatory genes (58,63).
Lactate accumulation in the TME impairs the immune response, facilitating tumor immune evasion. Elevated lactate levels inhibit cytotoxic T and NK cells (36,64) by impairing cytokine production, such as IFN-γ, essential for anti-tumor activity. Additionally, lactate decreases the proliferation and activity of these immune cells, weakening their ability to target and eliminate cancer cells. The acidic environment created by lactate further suppresses immune function by inducing mitochondrial stress and apoptosis in NK cells, particularly in liver-resident NK cells in colorectal liver metastasis (65,66).
Lactate promotes differentiation and activity of regulatory T cells (Tregs), which suppress the immune response against tumors (67,68). Tregs in the TME inhibit cytotoxic immune cells, aiding tumor immune evasion. Lactate-induced Tregs produce anti-inflammatory cytokines and inhibit effector T and NK cells, creating an immunosuppressive environment that fosters tumor growth and progression (69).
In summary, lactate in the TME modulates the immune response by inhibiting key immune cells and promoting immunosuppressive Tregs. These effects enhance the immune evasion of tumors, underscoring the potential of targeting lactate metabolism as a therapeutic strategy in GI cancers.
Lactate and lactylation in GI cancer
Lactate accumulation in GI tumors
The accumulation of lactate in GI tumors is a hallmark of the altered metabolic state of cancer cells. There are elevated lactate levels in various types of GI cancer, including CRC (70), gastric (71–73) and pancreatic (74) cancers. This elevation is primarily due to the Warburg effect (13).
Advanced techniques have quantified lactate levels in tumors, enhancing the understanding of lactate metabolism in cancer. Proton magnetic resonance spectroscopy is a non-invasive imaging technique widely used to measure lactate concentration in vivo (16,75), allowing real-time monitoring of metabolic changes in tumors. LC-MS (76) precisely quantifies lactate levels in tumor tissue and blood samples with high sensitivity and specificity.
Clinical studies consistently show significantly elevated lactate levels in GI tumors compared with normal tissues (70,71,71). For example, patients with CRC have lactate concentrations in tumor tissue several fold higher than in adjacent non-tumorous tissues. Similar findings in gastric and pancreatic cancer show an association between high lactate levels with advanced disease stage and poor prognosis (71,74), underscoring lactate as a biomarker for tumor aggressiveness and potential therapeutic target.
Lactate accumulation in GI tumors influences tumor progression, invasion and therapy resistance. Further research into the mechanisms of lactate accumulation and its effects on the TME may advance the understanding of GI cancer biology and improve clinical outcomes.
Lactylation in GI cancer progression
Lactylation connects cellular metabolism to gene expression, affecting tumor development, invasion and metastasis.
Lactylation notably impacts tumor growth by activating genes involved in cell proliferation and survival. For example, H3K18la upregulates genes that enhance glycolysis and other metabolic pathways essential for rapidly dividing cancer cells. By boosting expression of these genes, lactylation meets the metabolic demands of tumor cells, supporting sustained tumor growth and expansion. Aberrant lactate production facilitates cancer hallmarks such as uncontrolled growth and resistance to cell death. For example, eliminating tumor-produced lactate by deleting MCT1 in lung cancer cells halts tumor growth (77). Similarly, inhibiting MCT1 or MCT4 stops proliferation of leukemia cells, and selective inhibition of MCT4 impedes invasive bladder cancer cell proliferation (78–80). Additionally, cancer-produced lactate activates G-protein-coupled receptor 81, with its deletion arresting breast cancer tumor growth in vitro and in vivo, underscoring the role of lactate in cancer proliferation (81,82).
Lactylation also enhances the invasive and metastatic potential of GI cancers. Histone lactylation activates genes encoding MMPs and other enzymes that degrade the extracellular matrix, facilitating tumor cell invasion into surrounding tissue and the establishment of metastases in distant organs (83). Additionally, lactylation-induced gene expression changes promote EMT, enabling cancer cells to acquire migratory and invasive capability (84–86).
By modulating the expression of genes involved in immune regulation, lactylation helps cancer cells evade immune surveillance (36). For example, lactylation suppresses pro-inflammatory cytokines and enhances anti-inflammatory factors, creating an immunosuppressive TME (84). This environment inhibits the activity of immune cells, such as cytotoxic T and NK cells, allowing tumor cells to proliferate unchecked (86).
Key proteins and pathways modified by lactylation in gastrointestinal GI cancers impact tumor behavior and progression (87,88). Lactylation enhances HIF-1 stability and activity, promoting angiogenesis, glycolysis and cell survival under hypoxic conditions common in tumors (89). This stabilization activates genes that support tumor growth and adaptation to low-oxygen environments (90). Similarly, lactylation modifies NF-κB, a transcription factor key for in inflammation and cell survival pathways. Lactylation increases NF-κB transcriptional activity, upregulating genes involved in inflammation, cell proliferation and resistance to apoptosis, thus aiding tumor progression (91,92). Histone lactylation, particularly at markers such as H3K18la, influences cancer progression by activating genes that drive EMT, immune evasion and tumor aggressiveness (92). By modulating these critical pathways, lactylation facilitates the complex network of gene regulation enabling GI tumors to grow, invade and resist therapeutic intervention.
Understanding lactylation mechanisms and effects in GI cancer is essential for developing targeted therapy to disrupt this modification and improve patient outcomes (Table I).
Differential mechanisms of lactylation in GI tumor subtypes
Lactylation serves distinct regulatory roles in various types of GI cancer, including gastric, CRC and pancreatic cancers, through different molecular pathways. In gastric cancer, lactylation primarily affects glycolysis and mitochondrial function by enhancing activity of key metabolic enzymes such as LDH-A, which increases glycolytic flux. This metabolic reprogramming leads to rapid cellular proliferation and migration, as well as upregulation of oncogenic and cell cycle-related genes, contributing to tumor aggressiveness and resistance to apoptosis (83). Additionally, lactylation can regulate mitochondrial dynamics and energy production, supporting the highly proliferative nature of gastric cancer cells (93). In CRC, lactylation plays a critical role in shaping the TME by modulating immune cell function. High levels of lactylation are associated with increased recruitment of Tregs and suppression of cytotoxic T cells, leading to immune evasion and poor prognosis (94,95). Furthermore, lactylation has been shown to promote chemoresistance in CRC by regulating genes involved in drug metabolism and efflux pathways (93). Recent studies have identified a panel of lactylation-associated genes associated with poor survival outcomes that may serve as potential prognostic markers (95,96). Targeting these lactylation-related pathways may overcome therapeutic resistance and improve patient outcomes. In pancreatic cancer, lactylation impacts stromal interactions and extracellular matrix remodeling. By activating cancer-associated fibroblasts, lactylation enhances production of extracellular matrix proteins, leading to a dense, fibrotic stroma that is characteristic of pancreatic tumors. This stromal barrier not only impedes drug delivery but also creates an immunosuppressive environment that shields the tumor from immune surveillance (36,96). Additionally, lactylation modulates key signaling pathways such as HIF-1α and TGF-β, further promoting fibrosis, tumor growth and metastasis (96).
Therapeutic implications
Targeting lactate metabolism
Targeting lactate metabolism offers a promising therapeutic strategy for GI cancer due to its role in tumor progression and immune evasion. Various approaches aim to disrupt lactate production and transport, thereby altering the TME and enhancing the efficacy of existing treatments.
Inhibitors of glycolytic enzymes
One approach involves inhibiting key glycolytic enzymes to reduce lactate production. Agents targeting enzymes such as hexokinase, phosphofructokinase and LDH have shown potential in preclinical studies (97–100). For example, LDH inhibitors decrease lactate levels, leading to reduced tumor growth and improved chemotherapy response. Inhibitors such as oxamate (97,98), which targets LDH-A, have demonstrated effectiveness in preclinical models by decreasing lactate production and promoting immune activation within the TME (99). Additionally, targeting pyruvate dehydrogenase kinase, which regulates conversion of pyruvate to lactate, has shown efficacy in reducing lactate production and tumor cell proliferation (100).
MCT inhibitors
Another strategy involves blocking lactate export from cancer cells by inhibiting MCTs, particularly MCT1 and MCT4 (101). MCT inhibitors prevent the acidification of the TME, enhancing immune cell function and reducing tumor invasion. AZD3965, an MCT1 inhibitor, has shown promise in clinical trials, demonstrating potential to disrupt lactate transport and improve treatment outcomes in various types of cancer (102–104). MCT4 facilitates efflux of lactic acid from glycolytic cancer cells (105). Knocking down or silencing MCT4 leads to cytoplasmic acidification and subsequent tumor cell death. MCT4 is predominantly expressed in hypoxic regions of rapidly proliferating tumors, making it a promising therapeutic target. Although existing drugs aimed at MCT4 lack specificity, further investigation into their potential effectiveness in cancer therapy is needed (106,107). Additionally, localization and stability of MCT1 and MCT4 at the plasma membrane are regulated by CD147. Targeting CD147 could be a novel strategy to inhibit both transporters. AC-73, a dimerized and humanized anti-CD147 antibody, has demonstrated antitumor activity in preclinical study (108). In summary, inhibiting lactic acid transporters MCT1 and MCT4, along with their chaperones, may have antitumor potential. However, clinical data supporting these findings are currently lacking (108). Blocking MCT1 and MCT4 normalizes the pH of the TME, thereby enhancing immune responses and reducing metastatic potential.
Combination therapy
Combining lactate metabolism inhibitors with other therapeutic modalities, such as immunotherapy and conventional chemotherapy, has been researched (109–111). For example, combining LDH inhibitors with immune checkpoint inhibitors has shown synergistic effects, enhancing anti-tumor immune responses by reversing lactate-induced immunosuppression. This multi-faceted approach targets various aspects of tumor biology, potentially improving patient outcomes (112–114). Combination therapy can enhance the efficacy of therapies such as chimeric antigen receptor T cell therapy by decreasing lactate-induced immunosuppression (108,115). In summary, targeting lactate metabolism using glycolytic enzyme and MCT inhibitors and combination therapies shows promise for treating GI cancers. These strategies aim to disrupt the lactate-driven metabolic reprogramming and immune evasion mechanisms that tumors exploit, enhancing the overall effectiveness of cancer therapy. Further research and clinical trials are essential to validate these approaches and translate them into effective treatments. Inhibitors of LDH and MCTs are still in early-phase clinical trials, with mixed results regarding efficacy and safety (108,109). Challenges include potential off-target effects, limited tumor specificity and TME complexity, which may reduce treatment effectiveness. Furthermore, the metabolic flexibility of cancer cells may enable resistance to these therapies. Addressing these issues requires comprehensive clinical studies, better biomarkers for patient selection and combination therapies to overcome resistance and enhance therapeutic outcomes.
Inhibiting lactylation
Inhibiting lactylation is a therapeutic approach in GI cancers due to its role in regulating gene expression and promoting tumor progression (116–120).
Histone deacetylase (HDAC) inhibitors
HDAC inhibitors, which can also affect histone lactylation, have been explored for their potential to modulate epigenetic markers in cancer cells (116). By decreasing lactylation levels, these inhibitors alter gene expression involved in tumor growth and immune evasion. For example, vorinostat and panobinostat, two prominent HDAC inhibitors, have efficacy in various types of cancers by disrupting epigenetic modifications and enhancing tumor suppressor gene expression (116,117). Vorinostat and panobinostat prevent removal of acetyl and, potentially, lactyl groups from histones, maintaining a more open chromatin structure conducive to gene expression that counteracts tumor growth (116,117).
Direct inhibitors of lactylation enzymes
Identifying and targeting specific enzymes responsible for lactylation, such as potential lactyltransferases, represents a promising research direction. Although these enzymes have not been definitively identified, studies have investigated small molecules that specifically inhibit lactylation, thereby disrupting its oncogenic effects (118,119). General control non-depressible 5 is a possible lactyltransferase for ERK in the MAPK signaling pathway (118,119). Inhibitors targeting such lactyltransferases could reduce tumor progression and metastasis by altering key signaling pathways involved in cancer cell survival and proliferation.
Combining lactylation inhibitors with other therapy
Combining lactylation inhibitors with other treatment modalities may enhance therapeutic efficacy (41). Integrating lactylation inhibition with immunotherapy or targeted therapy may provide a comprehensive approach to disrupting cancer progression and overcoming resistance to single-agent treatments. Studies have shown that combining lactylation inhibitors with immune checkpoint inhibitors or chemotherapeutic agents produces synergistic effects, enhancing overall anti-tumor response by reversing immunosuppression and decreasing tumor growth (41,120).
Targeting lactylation presents a potential therapeutic approach for GI cancer. HDAC and direct lactylation enzyme inhibitors and combination therapies offer multiple avenues to disrupt lactylation-driven oncogenic processes. Further research and clinical trials are key to translate these strategies into effective treatments for patients with cancer.
Future research directions
Identifying and characterizing enzymes directly responsible for protein lactylation is essential. Understanding specific mechanisms and regulatory pathways of lactylation will aid in developing targeted inhibitors. Therefore, research should prioritize discovering and validating lactyl transferases, which serve a key role in catalyzing lactylation. Identifying these enzymes may offer novel therapeutic targets to inhibit lactylation oncogenic effects and improve cancer treatment outcomes.
Developing reliable biomarkers for lactate metabolism and lactylation may enhance patient stratification and treatment monitoring. Biomarkers can identify patients likely to benefit from therapy targeting these pathways, thereby improving personalized treatment approaches. Efforts should prioritize discovering biomarkers that reflect lactate metabolism and lactylation activity, such as specific lactylation markers on histones or lactate levels in the TME.
Expanding clinical trials to assess the safety and efficacy of lactate metabolism and lactylation inhibitors in GI cancer is crucial. These trials should explore various combinations of inhibitors with existing therapy to identify the most effective regimens. Clinical research should also investigate potential side effects and resistance mechanisms, ensuring safe and effective integration into cancer treatment protocols.
Elucidating the complex interactions between lactate, lactylation, and other metabolic and epigenetic processes in cancer cells requires in-depth mechanistic studies to reveal the broader implications of targeting these pathways and potential resistance mechanisms. Research should focus on understanding how lactate and lactylation affect gene expression, tumor metabolism and the immune microenvironment to identify new therapeutic targets and strategies, ultimately improving clinical outcomes for patients with GI cancer and potentially leading to more personalized and effective cancer treatment.
Conclusion
Lactate and lactylation serve a key role in the progression of GI cancers by modulating the TME, gene expression and immune response. Elevated lactate levels promote tumor growth, invasion and immune evasion through acidification and immune suppression. Lactylation regulates gene transcription and further supports tumor progression. Targeting lactate metabolism and lactylation presents potential therapeutic strategies, including glycolytic enzyme and MCT inhibitors and combination therapies. Future research should identify specific lactylation enzymes, develop reliable biomarkers, expand clinical trials and elucidate the underlying mechanisms to enhance personalized treatments and improve clinical outcomes for patients with GI cancer. Additionally, exploring the interplay between metabolic reprogramming and epigenetic modifications may reveal novel therapeutic targets and strategies, ultimately increasing the effectiveness of cancer therapy.
Acknowledgements
Not applicable.
Funding
Funding: No funding was received.
Availability of data and materials
Not applicable.
Authors' contributions
YH, YH, LR and QY wrote the manuscript. YH and YH created figures and tables. PP, LR and QY revised the manuscript. LR and QY supervised the research. All authors have read and approved the final 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
Huang J, Lucero-Prisno DE III, Zhang L, Xu W, Wong SH, Ng SC and Wong MCS: Updated epidemiology of gastrointestinal cancers in East Asia. Nat Rev Gastroenterol Hepatol. 20:271–287. 2023. View Article : Google Scholar : PubMed/NCBI | |
Shah SC and Itzkowitz SH: Colorectal cancer in inflammatory bowel disease: Mechanisms and management. Gastroenterology. 162:715–730.e3. 2022. View Article : Google Scholar : PubMed/NCBI | |
Joshi SS and Badgwell BD: Current treatment and recent progress in gastric cancer. CA Cancer J Clin. 71:264–279. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhu H, Ma X, Ye T, Wang H, Wang Z, Liu Q and Zhao K: Esophageal cancer in China: Practice and research in the new era. Int J Cancer. 152:1741–1751. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wood LD, Canto MI, Jaffee EM and Simeone DM: Pancreatic cancer: Pathogenesis, screening, diagnosis, and treatment. Gastroenterology. 163:386–402.e1. 2022. View Article : Google Scholar : PubMed/NCBI | |
Liberti MV and Locasale JW: The Warburg effect: How does it benefit cancer cells? Trends Biochem Sci. 41:211–218. 2016. View Article : Google Scholar : PubMed/NCBI | |
Wang Y and Patti GJ: The Warburg effect: A signature of mitochondrial overload. Trends Cell Biol. 33:1014–1020. 2023. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Brooks GA: The science and translation of lactate shuttle theory. Cell Metab. 27:757–785. 2018. View Article : Google Scholar : PubMed/NCBI | |
Rabinowitz JD and Enerbäck S: Lactate: The ugly duckling of energy metabolism. Nat Metab. 2:566–571. 2020. View Article : Google Scholar : PubMed/NCBI | |
Lv X, Lv Y and Dai X: Lactate, histone lactylation and cancer hallmarks. Expert Rev Mol Med. 25:e72023. View Article : Google Scholar : PubMed/NCBI | |
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 | |
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 | |
Chen J, Cao X, Li B, Zhao Z, Chen S, Lai SWT, Muend SA, Nossa GK, Wang L, Guo W, et al: Warburg effect is a cancer immune evasion mechanism against macrophage immunosurveillance. Front Immunol. 11:6217572021. View Article : Google Scholar : PubMed/NCBI | |
Kes MMG, Van den Bossche J, Griffioen AW and Huijbers EJM: Oncometabolites lactate and succinate drive pro-angiogenic macrophage response in tumors. Biochim Biophys Acta Rev Cancer. 1874:1884272020. View Article : Google Scholar : PubMed/NCBI | |
Cheung SM, Husain E, Masannat Y, Miller ID, Wahle K, Heys SD and He J: Lactate concentration in breast cancer using advanced magnetic resonance spectroscopy. Br J Cancer. 123:261–267. 2020. View Article : Google Scholar : PubMed/NCBI | |
Hui S, Ghergurovich JM, Morscher RJ, Jang C, Teng X, Lu W, Esparza LA, Reya T, Zhan L, Yanxiang Guo J, et al: Glucose feeds the TCA cycle via circulating lactate. Nature. 551:115–118. 2017. View Article : Google Scholar : PubMed/NCBI | |
Nelson SJ, Kurhanewicz J, Vigneron DB, Larson PE, Harzstark AL, Ferrone M, van Criekinge M, Chang JW, Bok R, Park I, et al: Metabolic imaging of patients with prostate cancer using hyperpolarized [1-¹3C]pyruvate. Sci Transl Med. 5:198ra082013. View Article : Google Scholar : PubMed/NCBI | |
Mortazavi Farsani SS and Verma V: Lactate mediated metabolic crosstalk between cancer and immune cells and its therapeutic implications. Front Oncol. 13:11755322023. View Article : Google Scholar : PubMed/NCBI | |
San-Millán I and Brooks GA: Reexamining cancer metabolism: Lactate production for carcinogenesis could be the purpose and explanation of the Warburg effect. Carcinogenesis. 38:119–133. 2017.PubMed/NCBI | |
Petersen C, Nielsen MD, Andersen ES, Basse AL, Isidor MS, Markussen LK, Viuff BM, Lambert IH, Hansen JB and Pedersen SF: MCT1 and MCT4 expression and lactate flux activity increase during white and brown adipogenesis and impact adipocyte metabolism. Sci Rep. 7:131012017. View Article : Google Scholar : PubMed/NCBI | |
Zhang L, Xin C, Wang S, Zhuo S, Zhu J, Li Z, Liu Y, Yang L and Chen Y: Lactate transported by MCT1 plays an active role in promoting mitochondrial biogenesis and enhancing TCA flux in skeletal muscle. Sci Adv. 10:eadn45082024. View Article : Google Scholar : PubMed/NCBI | |
Mai Z, Lin Y, Lin P, Zhao X and Cui L: Modulating extracellular matrix stiffness: A strategic approach to boost cancer immunotherapy. Cell Death Dis. 15:3072024. View Article : Google Scholar : PubMed/NCBI | |
Caslin HL, Abebayehu D, Pinette JA and Ryan JJ: Lactate is a metabolic mediator that shapes immune cell fate and function. Front Physiol. 12:6884852021. View Article : Google Scholar : PubMed/NCBI | |
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 | |
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 | |
Sun P, Ma L and Lu Z: Lactylation: Linking the Warburg effect to DNA damage repair. Cell Metab. 36:1637–1639. 2024. View Article : Google Scholar : PubMed/NCBI | |
Liebner T, Kilic S, Walter J, Aibara H, Narita T and Choudhary C: Acetylation of histones and non-histone proteins is not a mere consequence of ongoing transcription. Nat Commun. 15:49622024. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Xie N, Zhang L, Gao W, Huang C, Huber PE, Zhou X, Li C, Shen G and Zou B: NAD+ metabolism: Pathophysiologic mechanisms and therapeutic potential. Signal Transduct Target Ther. 5:2272020. View Article : Google Scholar : PubMed/NCBI | |
Navas LE and Carnero A: NAD+ metabolism, stemness, the immune response, and cancer. Signal Transduct Target Ther. 6:22021. View Article : Google Scholar : PubMed/NCBI | |
Chen AN, Luo Y, Yang YH, Fu JT, Geng XM, Shi JP and Yang J: Lactylation, a novel metabolic reprogramming code: Current status and prospects. Front Immunol. 12:6889102021. View Article : Google Scholar : PubMed/NCBI | |
Varner EL, Trefely S, Bartee D, von Krusenstiern E, Izzo L, Bekeova C, O'Connor RS, Seifert EL, Wellen KE, Meier JL and Snyder NW: Quantification of lactoyl-CoA (lactyl-CoA) by liquid chromatography mass spectrometry in mammalian cells and tissues. Open Biol. 10:2001872020. View Article : Google Scholar : PubMed/NCBI | |
Wang T, Chen K, Yao W, Zheng R, He Q, Xia J, Li J, Shao Y, Zhang L, Huang L, et al: Acetylation of lactate dehydrogenase B drives NAFLD progression by impairing lactate clearance. J Hepatol. 74:1038–1052. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang N, Wang W, Wang X, Mang G, Chen J, Yan X, Tong Z, Yang Q, Wang M, Chen L, et al: Histone lactylation boosts reparative gene activation post-myocardial infarction. Circ Res. 131:893–908. 2022. View Article : Google Scholar : PubMed/NCBI | |
Chen L, Huang L, Gu Y, Cang W, Sun P and Xiang Y: Lactate-lactylation hands between metabolic reprogramming and immunosuppression. Int J Mol Sci. 23:119432022. View Article : Google Scholar : PubMed/NCBI | |
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 | |
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 | |
Zhang N, Zhang Y, Xu J, Wang P, Wu B, Lu S, Lu X, You S, Huang X, Li M, et al: α-myosin heavy chain lactylation maintains sarcomeric structure and function and alleviates the development of heart failure. Cell Res. 33:679–698. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wang J, Yang P, Yu T, Gao M, Liu D, Zhang J, Lu C, Chen X, Zhang X and Liu Y: Lactylation of PKM2 suppresses inflammatory metabolic adaptation in pro-inflammatory macrophages. Int J Biol Sci. 18:6210–6225. 2022. View Article : Google Scholar : PubMed/NCBI | |
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 | |
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 | |
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 | |
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 | |
Wang X, Fan W, Li N, Ma Y, Yao M, Wang G, He S, Li W, Tan J, Lu Q and Hou S: YY1 lactylation in microglia promotes angiogenesis through transcription activation-mediated upregulation of FGF2. Genome Biol. 24:872023. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Chen J, Zhang M, Liu Y, Zhao S, Wang Y, Wang M, Niu W, Jin F and Li Z: Histone lactylation driven by mROS-mediated glycolytic shift promotes hypoxic pulmonary hypertension. J Mol Cell Biol. 14:mjac0732023. View Article : Google Scholar : PubMed/NCBI | |
Wang P, Xie D, Xiao T, Cheng C, Wang D, Sun J, Wu M, Yang Y, Zhang A and Liu Q: H3K18 lactylation promotes the progression of arsenite-related idiopathic pulmonary fibrosis via YTHDF1/m6A/NREP. J Hazard Mater. 461:1325822024. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Wang Y, Qin L, Chen W, Chen Q, Sun J and Wang G: Novel strategies to improve tumour therapy by targeting the proteins MCT1, MCT4 and LAT1. Eur J Med Chem. 226:1138062021. View Article : Google Scholar : PubMed/NCBI | |
She X, Wu Q, Rao Z, Song D, Huang C, Feng S, Liu A, Liu L, Wan K, Li X, et al: SETDB1 methylates MCT1 promoting tumor progression by enhancing the lactate shuttle. Adv Sci (Weinh). 10:e23018712023. View Article : Google Scholar : PubMed/NCBI | |
Hadjihambi A, Konstantinou C, Klohs J, Monsorno K, Le Guennec A, Donnelly C, Cox IJ, Kusumbe A, Hosford PS, Soffientini U, et al: Partial MCT1 invalidation protects against diet-induced non-alcoholic fatty liver disease and the associated brain dysfunction. J Hepatol. 78:180–190. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wang F, Yi J, Chen Y, Bai X, Lu C, Feng S and Zhou X: PRSS2 regulates EMT and metastasis via MMP-9 in gastric cancer. Acta Histochem. 125:1520712023. View Article : Google Scholar : PubMed/NCBI | |
Hwang KE, Kim HJ, Song IS, Park C, Jung JW, Park DS, Oh SH, Kim YS and Kim HR: Salinomycin suppresses TGF-β1-induced EMT by down-regulating MMP-2 and MMP-9 via the AMPK/SIRT1 pathway in non-small cell lung cancer. Int J Med Sci. 18:715–726. 2021. View Article : Google Scholar : PubMed/NCBI | |
Jiang J, Huang D, Jiang Y, Hou J, Tian M, Li J, Sun L, Zhang Y, Zhang T, Li Z, et al: Lactate modulates cellular metabolism through histone lactylation-mediated gene expression in non-small cell lung cancer. Front Oncol. 11:6475592021. View Article : Google Scholar : PubMed/NCBI | |
Izzo LT and Wellen KE: Histone lactylation links metabolism and gene regulation. Nature. 574:492–493. 2019. View Article : Google Scholar : PubMed/NCBI | |
Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, Cyrus N, Brokowski CE, Eisenbarth SC, Phillips GM, et al: Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. 513:559–563. 2014. View Article : Google Scholar : PubMed/NCBI | |
Wynn TA, Chawla A and Pollard JW: Macrophage biology in development, homeostasis and disease. Nature. 496:445–455. 2013. View Article : Google Scholar : PubMed/NCBI | |
Irizarry-Caro RA, Mcdaniel MM, Overcast GR, Jain VG, Troutman TD and Pasare C: TLR signaling adapter BCAP regulates inflammatory to reparatory macrophage transition by promoting histone lactylation. Proc Natl Acad Sci USA. 117:30628–30638. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ivashkiv LB: The hypoxia-lactate axis tempers inflammation. Nat Rev Immunol. 20:85–86. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zhang F, Parayath NN, Ene CI, Stephan SB, Koehne AL, Coon ME, Holland EC and Stephan MT: Genetic programming of macrophages to perform anti-tumor functions using targeted mRNA nanocarriers. Nat Commun. 10:39742019. View Article : Google Scholar : PubMed/NCBI | |
Dichtl S, Lindenthal L, Zeitler L, Behnke K, Schlösser D, Strobl B, Scheller J, El Kasmi KC and Murray PJ: Lactate and IL6 define separable paths of inflammatory metabolic adaptation. Sci Adv. 7:eabg35052021. View Article : Google Scholar : PubMed/NCBI | |
Siska PJ, Singer K, Evert K, Renner K and Kreutz M: The immunological Warburg effect: Can a metabolic-tumor-stroma score (MeTS) guide cancer immunotherapy? Immunol Rev. 295:187–202. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wang ZH, Peng WB, Zhang P, Yang XP and Zhou Q: Lactate in the tumour microenvironment: From immune modulation to therapy. EBioMedicine. 73:1036272021. View Article : Google Scholar : PubMed/NCBI | |
Xia L, Oyang L, Lin J, Tan S, Han Y, Wu N, Yi P, Tang L, Pan Q, Rao S, et al: The cancer metabolic reprogramming and immune response. Mol Cancer. 20:282021. View Article : Google Scholar : PubMed/NCBI | |
de la Cruz-López KG, Castro-Muñoz LJ, Reyes-Hernández DO, García-Carrancá A and Manzo-Merino J: Lactate in the regulation of tumor microenvironment and therapeutic approaches. Front Oncol. 9:11432019. View Article : Google Scholar : PubMed/NCBI | |
Tay C, Tanaka A and Sakaguchi S: Tumor-infiltrating regulatory T cells as targets of cancer immunotherapy. Cancer Cell. 41:450–465. 2023. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Lopez Krol A, Nehring HP, Krause FF, Wempe A, Raifer H, Nist A, Stiewe T, Bertrams W, Schmeck B, Luu M, et al: Lactate induces metabolic and epigenetic reprogramming of pro-inflammatory Th17 cells. EMBO Rep. 23:e546852022. View Article : Google Scholar : PubMed/NCBI | |
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 | |
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 | |
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 | |
Yang H, Zou X, Yang S, Zhang A, Li N and Ma Z: Identification of lactylation related model to predict prognostic, tumor infiltrating immunocytes and response of immunotherapy in gastric cancer. Front Immunol. 14:11499892023. View Article : Google Scholar : PubMed/NCBI | |
Yang J, Ren B, Yang G, Wang H, Chen G, You L, Zhang T and Zhao Y: The enhancement of glycolysis regulates pancreatic cancer metastasis. Cell Mol Life Sci. 77:305–321. 2020. View Article : Google Scholar : PubMed/NCBI | |
Shirbandi K, Rikhtegar R, Khalafi M, Mirza Aghazadeh Attari M, Rahmani F, Javanmardi P, Iraji S, Babaei Aghdam Z and Rezaei Rashnoudi AM: Functional magnetic resonance spectroscopy of lactate in Alzheimer disease: A comprehensive review of Alzheimer disease pathology and the role of lactate. Top Magn Reson Imaging. 32:15–26. 2023.PubMed/NCBI | |
Afshar M and van Hall G: LC-MS/MS method for quantitative profiling of ketone bodies, α-keto acids, lactate, pyruvate and their stable isotopically labelled tracers in human plasma: An analytical panel for clinical metabolic kinetics and interactions. J Chromatogr B Analyt Technol Biomed Life Sci. 1230:1239062023. View Article : Google Scholar : PubMed/NCBI | |
Faubert B, Li KY, Cai L, Hensley CT, Kim J, Zacharias LG, Yang C, Do QN, Doucette S, Burguete D, et al: Lactate metabolism in human lung tumors. Cell. 171:358–371.e9. 2017. View Article : Google Scholar : PubMed/NCBI | |
Pivovarova AI and Macgregor GG: Glucose-dependent growth arrest of leukemia cells by MCT1 inhibition: Feeding Warburg's sweet tooth and blocking acid export as an anticancer strategy. Biomed Pharmacother. 98:173–179. 2018. View Article : Google Scholar : PubMed/NCBI | |
Saulle E, Spinello I, Quaranta MT, Pasquini L, Pelosi E, Iorio E, Castelli G, Chirico M, Pisanu ME, Ottone T, et al: Targeting lactate metabolism by inhibiting MCT1 or MCT4 impairs leukemic cell proliferation, induces two different related death-pathways and increases chemotherapeutic sensitivity of acute myeloid leukemia cells. Front Oncol. 10:6214582021. View Article : Google Scholar : PubMed/NCBI | |
Todenhöfer T, Seiler R, Stewart C, Moskalev I, Gao J, Ladhar S, Kamjabi A, Al Nakouzi N, Hayashi T, Choi S, et al: Selective inhibition of the lactate transporter MCT4 reduces growth of invasive bladder cancer. Mol Cancer Ther. 17:2746–2755. 2018. View Article : Google Scholar : PubMed/NCBI | |
Xie Q, Zhu Z, He Y, Zhang Z, Zhang Y, Wang Y, Luo J, Peng T, Cheng F, Gao J, et al: A lactate-induced Snail/STAT3 pathway drives GPR81 expression in lung cancer cells. Biochim Biophys Acta Mol Basis Dis. 1866:1655762020. View Article : Google Scholar : PubMed/NCBI | |
Brown TP, Bhattacharjee P, Ramachandran S, Sivaprakasam S, Ristic B, Sikder MOF and Ganapathy V: The lactate receptor GPR81 promotes breast cancer growth via a paracrine mechanism involving antigen-presenting cells in the tumor microenvironment. Oncogene. 39:3292–3304. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yang H, Yang S, He J, Li W, Zhang A, Li N, Zhou G and Sun B: Glucose transporter 3 (GLUT3) promotes lactylation modifications by regulating lactate dehydrogenase A (LDHA) in gastric cancer. Cancer Cell Int. 23:3032023. View Article : Google Scholar : PubMed/NCBI | |
Huang YF, Wang G, Ding L, Bai ZR, Leng Y, Tian JW, Zhang JZ, Li YQ, Ahmad Qin YH, et al: Lactate-upregulated NADPH-dependent NOX4 expression via HCAR1/PI3K pathway contributes to ROS-induced osteoarthritis chondrocyte damage. Redox Biol. 67:1028672023. View Article : Google Scholar : PubMed/NCBI | |
Nareika A, He L, Game BA, Slate EH, Sanders JJ, London SD, Lopes-Virella MF and Huang Y: Sodium lactate increases LPS-stimulated MMP and cytokine expression in U937 histiocytes by enhancing AP-1 and NF-kappaB transcriptional activities. Am J Physiol Endocrinol Metab. 289:E534–E542. 2005. View Article : Google Scholar : PubMed/NCBI | |
Long L, Xiong W, Lin F, Hou J, Chen G, Peng T, He Y, Wang R, Xu Q and Huang Y: Regulating lactate-related immunometabolism and EMT reversal for colorectal cancer liver metastases using shikonin targeted delivery. J Exp Clin Cancer Res. 42:1172023. View Article : Google Scholar : PubMed/NCBI | |
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 | |
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. 39:1109862022. View Article : Google Scholar : PubMed/NCBI | |
Albogami SM, Al-Kuraishy HM, Al-Maiahy TJ, Al-Buhadily AK, Al-Gareeb AI, Alorabi M, Alotaibi SS, De Waard M, Sabatier JM, Saad HM and Batiha GE: Hypoxia-inducible factor 1 and preeclampsia: A new perspective. Curr Hypertens Rep. 24:687–692. 2022. View Article : Google Scholar : PubMed/NCBI | |
You L, Wu W, Wang X, Fang L, Adam V, Nepovimova E, Wu Q and Kuca K: The role of hypoxia-inducible factor 1 in tumor immune evasion. Med Res Rev. 41:1622–1643. 2021. View Article : Google Scholar : PubMed/NCBI | |
Yang K, Xu J, Fan M, Tu F, Wang X, Ha T, Williams DL and Li C: Lactate suppresses macrophage pro-inflammatory response to LPS stimulation by inhibition of YAP and NF-κB activation via GPR81-mediated signaling. Front Immunol. 11:5879132020. View Article : Google Scholar : PubMed/NCBI | |
Wei L, Yang X, Wang J, Wang Z, Wang Q, Ding Y and Yu A: H3K18 lactylation of senescent microglia potentiates brain aging and Alzheimer's disease through the NFκB signaling pathway. J Neuroinflammation. 20:2082023. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Huang H, Chen K, Zhu Y, Hu Z, Wang Y, Chen J, Li Y, Li D and Wei P: A multi-dimensional approach to unravel the intricacies of lactylation related signature for prognostic and therapeutic insight in colorectal cancer. J Transl Med. 22:2112024. View Article : Google Scholar : PubMed/NCBI | |
Zha J, Zhang J, Lu J, Zhang G, Hua M, Guo W, Yang J and Fan G: A review of lactate-lactylation in malignancy: Its potential in immunotherapy. Front Immunol. 15:13849482024. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Li X, Lu W, Hu Y, Wen S, Qian C, Wu W and Huang P: Effective inhibition of nasopharyngeal carcinoma in vitro and in vivo by targeting glycolysis with oxamate. Int J Oncol. 43:1710–1718. 2013. View Article : Google Scholar : PubMed/NCBI | |
Manerba M, Di Ianni L, Govoni M, Roberti M, Recanatini M and Di Stefano G: Lactate dehydrogenase inhibitors can reverse inflammation induced changes in colon cancer cells. Eur J Pharm Sci. 96:37–44. 2017. View Article : Google Scholar : PubMed/NCBI | |
Moreno-Sánchez R, Marín-Hernández Á, Del Mazo-Monsalvo I, Saavedra E and Rodríguez-Enríquez S: Assessment of the low inhibitory specificity of oxamate, aminooxyacetate and dichloroacetate on cancer energy metabolism. Biochim Biophys Acta Gen Subj. 1861:3221–3236. 2017. View Article : Google Scholar : PubMed/NCBI | |
Wigfield SM, Winter SC, Giatromanolaki A, Taylor J, Koukourakis ML and Harris AL: PDK-1 regulates lactate production in hypoxia and is associated with poor prognosis in head and neck squamous cancer. Br J Cancer. 98:1975–1984. 2008. View Article : Google Scholar : PubMed/NCBI | |
Quanz M, Bender E, Kopitz C, Grünewald S, Schlicker A, Schwede W, Eheim A, Toschi L, Neuhaus R, Richter C, et al: Preclinical efficacy of the novel monocarboxylate transporter 1 inhibitor BAY-8002 and associated markers of resistance. Mol Cancer Ther. 17:2285–2296. 2018. View Article : Google Scholar : PubMed/NCBI | |
Curtis NJ, Mooney L, Hopcroft L, Michopoulos F, Whalley N, Zhong H, Murray C, Logie A, Revill M, Byth KF, et al: Pre-clinical pharmacology of AZD3965, a selective inhibitor of MCT1: DLBCL, NHL and Burkitt's lymphoma anti-tumor activity. Oncotarget. 8:69219–69236. 2017. View Article : Google Scholar : PubMed/NCBI | |
Polański R, Hodgkinson CL, Fusi A, Nonaka D, Priest L, Kelly P, Trapani F, Bishop PW, White A, Critchlow SE, et al: Activity of the monocarboxylate transporter 1 inhibitor AZD3965 in small cell lung cancer. Clin Cancer Res. 20:926–937. 2014. View Article : Google Scholar : PubMed/NCBI | |
Colen CB, Shen Y, Ghoddoussi F, Yu P, Francis TB, Koch BJ, Monterey MD, Galloway MP, Sloan AE and Mathupala SP: Metabolic targeting of lactate efflux by malignant glioma inhibits invasiveness and induces necrosis: An in vivo study. Neoplasia. 13:620–632. 2011. View Article : Google Scholar : PubMed/NCBI | |
Ullah MS, Davies AJ and Halestrap AP: The plasma membrane lactate transporter MCT4, but not MCT1, is up-regulated by hypoxia through a HIF-1alpha-dependent mechanism. J Biol Chem. 281:9030–9037. 2006. View Article : Google Scholar : PubMed/NCBI | |
Grasa L, Chueca E, Arechavaleta S, García-González MA, Sáenz MÁ, Valero A, Hördnler C, Lanas Á and Piazuelo E: Antitumor effects of lactate transport inhibition on esophageal adenocarcinoma cells. J Physiol Biochem. 79:147–161. 2023. View Article : Google Scholar : PubMed/NCBI | |
Payen VL, Mina E, Van Hée VF, Porporato PE and Sonveaux P: Monocarboxylate transporters in cancer. Mol Metab. 33:48–66. 2020. View Article : Google Scholar : PubMed/NCBI | |
Spinello I, Saulle E, Quaranta MT, Pasquini L, Pelosi E, Castelli G, Ottone T, Voso MT, Testa U and Labbaye C: The small-molecule compound AC-73 targeting CD147 inhibits leukemic cell proliferation, induces autophagy and increases the chemotherapeutic sensitivity of acute myeloid leukemia cells. Haematologica. 104:973–985. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhai X, Yang Y, Wan J, Zhu R and Wu Y: Inhibition of LDH-A by oxamate induces G2/M arrest, apoptosis and increases radiosensitivity in nasopharyngeal carcinoma cells. Oncol Rep. 30:2983–2991. 2013. View Article : Google Scholar : PubMed/NCBI | |
Schwab M, Thunborg K, Azimzadeh O, von Toerne C, Werner C, Shevtsov M, Di Genio T, Zdralevic M, Pouyssegur J, Renner K, et al: Targeting cancer metabolism breaks radioresistance by impairing the stress response. Cancers (Basel). 13:37622021. View Article : Google Scholar : PubMed/NCBI | |
Lin YL, Yuksel Durmaz Y, Nör JE and ElSayed MEH: Synergistic combination of small molecule inhibitor and RNA interference against antiapoptotic Bcl-2 protein in head and neck cancer cells. Mol Pharm. 10:2730–2738. 2013. View Article : Google Scholar : PubMed/NCBI | |
García-Castillo V, López-Urrutia E, Villanueva-Sánchez O, Ávila-Rodríguez MÁ, Zentella-Dehesa A, Cortés-González C, López-Camarillo C, Jacobo-Herrera NJ and Pérez-Plasencia C: Targeting metabolic remodeling in triple negative breast cancer in a murine model. J Cancer. 8:178–189. 2017. View Article : Google Scholar : PubMed/NCBI | |
Muramatsu H, Sumitomo M, Morinaga S, Kajikawa K, Kobayashi I, Nishikawa G, Kato Y, Watanabe M, Zennami K, Kanao K, et al: Targeting lactate dehydrogenase-A promotes docetaxel-induced cytotoxicity predominantly in castration-resistant prostate cancer cells. Oncol Rep. 42:224–230. 2019.PubMed/NCBI | |
Manerba M, Di Ianni L, Fiume L, Roberti M, Recanatini M and Di Stefano G: Lactate dehydrogenase inhibitors sensitize lymphoma cells to cisplatin without enhancing the drug effects on immortalized normal lymphocytes. Eur J Pharm Sci. 74:95–102. 2015. View Article : Google Scholar : PubMed/NCBI | |
Daei Sorkhabi A, Mohamed Khosroshahi L, Sarkesh A, Mardi A, Aghebati-Maleki A, Aghebati-Maleki L and Baradaran B: The current landscape of CAR T-cell therapy for solid tumors: Mechanisms, research progress, challenges, and counterstrategies. Front Immunol. 14:11138822023. View Article : Google Scholar : PubMed/NCBI | |
Ogura M, Ando K, Suzuki T, Ishizawa K, Oh SY, Itoh K, Yamamoto K, Au WY, Tien HF, Matsuno Y, et al: A multicentre phase II study of vorinostat in patients with relapsed or refractory indolent B-cell non-Hodgkin lymphoma and mantle cell lymphoma. Br J Haematol. 165:768–776. 2014. View Article : Google Scholar : PubMed/NCBI | |
Kaufman JL, Mina R, Jakubowiak AJ, Zimmerman TL, Wolf JJ, Lewis C, Gleason C, Sharp C, Martin T, Heffner LT, et al: Combining carfilzomib and panobinostat to treat relapsed/refractory multiple myeloma: Results of a multiple myeloma research consortium phase I study. Blood Cancer J. 9:32019. View Article : Google Scholar : PubMed/NCBI | |
Lu W, Zhang L, Ji K, Ding L and Wu G: Regulatory mechanisms of GCN5 in osteogenic differentiation of MSCs in periodontitis. Clin Exp Dent Res. 9:464–471. 2023. View Article : Google Scholar : PubMed/NCBI | |
Shao G, Liu Y, Ma T, Zhang L, Yuan M and Zhao S: GCN5 inhibition prevents IL-6-induced prostate cancer metastases through PI3K/PTEN/Akt signaling by inactivating Egr-1. Biosci Rep. 38:BSR201808162018. View Article : Google Scholar : PubMed/NCBI | |
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 | |
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 | |
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 |