
Multifaceted roles of lactate dehydrogenase in liver cancer (Review)
- Authors:
- Published online on: May 20, 2025 https://doi.org/10.3892/ijo.2025.5756
- Article Number: 50
-
Copyright: © Jin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
As an important metabolic organ in the human body, the liver serves a crucial role in maintaining the homeostasis of the internal environment and the normal function of the organism. As a malignant tumor with high morbidity and mortality worldwide, liver cancer has become one of the leading causes of cancer-related deaths. According to the latest statistics, the incidence of liver cancer in 2021 was 6.71/100,000 individuals, and the crude mortality rate of liver cancer was 6.13/100,000 individuals (1,2). Metabolic reprogramming of hepatocellular carcinoma (HCC) cells is one of the central mechanisms of tumor cell proliferation and development (3). In contrast to normal cells, which rely primarily on oxidative phosphorylation (OXPHOS) for energy, HCC cells tend to obtain energy through the glycolytic pathway even under aerobic conditions, a phenomenon known as the 'Warburg effect' (4). Although each glucose molecule produces only two adenosine triphosphate (ATP) molecules through aerobic glycolysis, while 36 ATP molecules can be produced through OXPHOS, the faster rate of aerobic glycolysis better meets the requirements for the rapid proliferation of tumor cells (5). In addition, the glycolytic product lactate can provide fuel for neighboring oxygenated tumor cells, allowing the cooccurrence of glycolytic and oxidative metabolism in the tumor microenvironment (TME) (6). Lactate dehydrogenase (LDH) is an enzyme found in a wide range of tissues (such as heart muscle, liver, kidney, skeletal muscle and lung) and is involved in the final step of the glycolytic reaction, catalyzing the reversible conversion between lactate and pyruvate (5). In tumor cells, LDH not only is closely related to the rate of glycolysis and cofactor ratios but also significantly affects the acidic environment of tumor cells through the regulation of lactate production (7). LDH acts as a key regulator of tumor development through the regulation of the biochemical environment of the tumor, which has an important impact on processes such as tumor cell invasion, immunosuppression, angiogenesis and metastasis (7).
LDH serves an important role in the development, diagnosis, treatment and prognosis of cardiovascular diseases, infectious diseases, cancer and other diseases (8). LDH is not only an important marker of myocardial injury but also serves an important role in the development, progression and treatment of cardiovascular diseases (9). Lactate dehydrogenase A (LDHA) promotes cardiac remodeling by attenuating reactive oxygen species (ROS) levels and promoting M2 macrophage polarization and has the potential to be a target for myocardial infarction repair (10,11). LDHA deficiency may lead to cardiac hypertrophy and heart failure, and serum LDH levels are significantly correlated with mortality in acute heart failure (12,13). In addition, under high-glucose conditions, LDHA may have some therapeutic potential, as it can slow the progression of aortic coarctation (14). In hematologic diseases, elevation of LDH levels is closely associated with a larger tumor load and cell proliferation rate and can be used as an adjunctive diagnostic and monitoring indicator for diseases such as leukemia and pernicious anemia (15-18). Elevation of LDH levels in patients with COVID-19 reflects cytokine-mediated tissue damage, multiorgan dysfunction and hypercoagulability and significantly increases the risk of severe illness and death, suggesting that elevated LDH expression is closely related to poor prognosis (19-23). In scientific research, LDH is often used in combination with other tumor markers as an important marker of malignant tumors, and studies have shown that the degree to which LDH levels are elevated is closely related to tumor load and aggressiveness (8,24).
Although LDH serves an important role in disease development, tumor metabolic reprogramming, regulation of the TME, and has diagnostic and therapeutic potential, a systematic summary and description of the specific role of LDH in liver cancer are currently lacking. By reviewing the biological properties of LDH and its role in the development of HCC, the present study aimed to explore research progress related to the value of LDH in the diagnosis, treatment and prognostic evaluation of HCC and to provide novel targets and concepts for the diagnosis and treatment of HCC.
Biological properties of LDH
LDH is an oxidoreductase consisting of four peptide chains with a molecular weight of 134 kDa (25). LDH is a tetrameric enzyme consisting of M (LDHA) and/or H [lactate dehydrogenase B (LDHB)] subunits (7). LDHA and LDHB are encoded by the LDHA gene located on chromosome 11 (11p15.1) and the LDHB gene located on chromosome 12 (12p12.1), respectively (8). Five isoforms of LDH, namely, LDH-1 (H4), LDH-2 (H3M), LDH-3 (H2M2), LDH-4 (HM3) and LDH-5 (M4), can be produced by combining the A and B subunits in different ways (7). LDH isoforms are distributed in different tissues, with LDH-1 and LDH-2 being detected predominantly in the heart and erythrocytes, LDH-3 being detected predominantly in the brain, and LDH-5 and LDH-4 being detected predominantly in the liver and skeletal muscle (26). There is also a sperm-specific isozyme, LDH6, encoded by the LDHC gene (located on chromosome 11) (5).
Of the LDH isoforms, LDH-5 (LDHA) has the strongest ability to convert pyruvate to lactate because subunit M has a strong ability to catalyze this reaction (27). By contrast, subunit H has the ability to convert lactate back to pyruvate through the reverse reaction (28). LDH is active mainly in the cytoplasm and its main primary function is to catalyze the reversible redox reaction between lactate and pyruvate. LDH serves an important role in the balanced regulation of the glycolytic and gluconeogenic pathways and participates in cellular energy metabolism (29). Under aerobic conditions, pyruvate enters mitochondria and the tricarboxylic acid (TCA) cycle to produce large amounts of ATP (30). During hypoxia or high metabolic demand, LDH facilitates the reversible conversion of pyruvate to lactate while oxidizing NADH to NAD+ (26). Catalysis of the conversion of pyruvate to lactate by LDH helps to regulate the intracellular NADH/NAD+ ratio, and a normal intracellular NADH/NAD+ ratio is essential for various cellular processes, including the oxidative stress response, DNA repair and cell signaling (8). Thus, LDH not only serves a key role in energy metabolism but is also involved in maintaining cellular redox homeostasis (27). LDH, an important catalytic enzyme in glycolysis, can further support the metabolic demands of tumor cells by converting lactate to pyruvate and promoting metabolic synergy between glycolysis and OXPHOS (Fig. 1) (31).
LDH is a non-secreted enzyme and serum LDH activity mainly reflects overall enzyme activity. According to standard assays, the baseline activity of serum LDH is usually <247-248 U/l in adults (8,32). LDH activity in organs is closely related to its metabolic activity and the physiological activity of LDH in organs can reach as high as 9,000-25,000 U/g (26,33). There is a significant difference in the expression levels of LDH in different organs and tissues; LDH1 and LDH2 are more abundant in myocardium, while LDH5 and LDH4 are mainly in skeletal muscle and liver (26). LDH expression varies significantly among different organs and is finely regulated by numerous metabolic regulators, including the AMP-activated protein kinase (AMPK) and mTOR signaling pathways (34). These key pathways directly influence cellular metabolic adaptations by regulating LDH expression and enzymatic activity. Under pathological conditions, aberrant expression or activity of LDH is often closely associated with the onset and progression of cardiovascular disease, liver disease and cancer (8). In addition, the metabolic activity of LDH is closely associated with inflammatory responses and immunoregulatory networks, further highlighting its complex role in pathophysiological processes (35). LDH not only is an important metabolic enzyme that serves a key pivotal role in glycolysis and the TCA cycle but also performs multidimensional functions in the regulation of cellular metabolism, tissue-specific gene and protein expression and disease development. Elucidating the biological properties and mechanism of action of LDH in metabolic and pathological processes will provide an important theoretical basis for understanding the pathogenesis of metabolic diseases and tumors and LDH has potential value for the clinical diagnosis and monitoring of diseases, in addition to serving as a potential target for the development of novel therapeutic strategies against these diseases.
Roles of LDH in metabolic reprogramming in HCC
As the metabolic center of the body, the liver not only maintains glucose homeostasis by controlling multiple glucose metabolic pathways (36) but also participates in various lipid metabolism processes (37). HCC cells are metabolically reprogrammed to meet the rapid proliferation needs of tumors (34,38). HCC cells often exhibit enhanced aerobic glycolysis, known as the 'Warburg effect' (34). Tumor cells usually produce lactic acid through glycolysis in the presence of LDH (38). The study by Yang et al (39) has shown that lactate can promote the lactylation of adenylate kinase 2 at the K28 site, which affects its enzyme activity, leading to an imbalance in energy homeostasis in HCC cells and promoting tumor proliferation, invasion and metastasis. Lactic acid produced by glycolysis can also serve as a nutrient for cancer cells, which is closely related to its ability to alleviate oxidative stress and facilitate lipid synthesis, promoting tumor growth (34,40). Aerobic glycolysis in HCC cells also provides important synthetic raw materials for macromolecular synthesis, such as ribose 5-phosphate, a substrate of the pentose-phosphate pathway, for nucleotide synthesis and the glycolytic intermediates glycerol 3-phosphate and acetyl-coenzyme A for lipid and protein synthesis (41,42). Abnormal lipid metabolism in HCC cells can also drive the development of HCC (43). The increased ab initio synthesis of lipids in HCC cells meets the energy and raw material requirements for cell membrane synthesis and cell proliferation (44). However, when the fatty acid oxidation pathway is inhibited, the resulting lipid accumulation enhances immunosuppression and promotes tumor growth and metastasis; thus, it is often suggested that decreased fatty acid metabolism is closely related to the aggressiveness of HCC (45,46). In addition, the loss of branched-chain amino acid catabolism has been shown to promote the development of HCC (47). Metabolic reprogramming in HCC is regulated by oncogene activation or inactivation, signaling pathway abnormalities and the TME (48-50). The activation of oncogenes such as c-Myc, KRAS and PI3K/AKT can promote metabolic reprogramming in HCC cells by increasing the expression of genes related to glycolysis and lipid metabolism (51-53). Hypoxia, a low pH and a tumor environment containing inflammatory cytokines can induce glycolytic reprogramming of HCC cells by activating related signaling pathways, such as the hypoxia-inducible factor (HIF) pathway (54). HIF-1α, which is stably expressed under hypoxic conditions, upregulates the expression of various glycolysis-related genes, such as LDH and promotes glycolysis and lactic acid production (55).
High expression and activity of LDH are key factors for maintaining continuous glycolysis in HCC cells (56). Owing to the rapid proliferation of HCC cells, the local oxygen supply is relatively insufficient and these cells rely on glycolysis for energy (57). LDH catalyzes the conversion of pyruvate to lactate and ensures the smooth progression to glycolysis through the regeneration of NAD+ (58,59). The study by Manerba et al (60) found that the inhibition of LDH activity or expression significantly reduces the rate of glycolysis in HCC cells, leading to a decrease in intracellular ATP levels, the inhibition of cell proliferation and the induction of apoptosis. LDH deficiency also leads to an intracellular redox imbalance, which affects the normal metabolism and function of cells (61). The catalysis of lactate production by LDH affects the proliferation and invasion of HCC cells through the regulation of the glutathione system and the production of ROS, which subsequently affects the proliferation, invasion and metastasis of HCC cells (38,62,63). LDH also promotes tumor growth by promoting lactate production and enhancing ferroptosis resistance in HCC cells (40). LDH-catalyzed lactate metabolism serves a key role in HCC development, not only providing energy support to tumor cells but also promoting invasion and metastasis through metabolic regulation.
Although LDH is often associated with the glucose metabolism pathway, LDH interacts also closely with other metabolic pathways through glucose metabolism (64). For example, lactate can enter mitochondria through the lactate shuttle and be converted to pyruvate through reverse catalysis by LDH, which participates in the TCA and provides additional energy to cells (65). Lipid metabolism serves a key role in the development and progression of HCC and increased lipid synthesis promotes the progression of HCC (46,66). LDHA indirectly affects the activity of lipid oxidases and reduces the precursors required for lipid synthesis by regulating NAD+/NADH homeostasis (67,68). LDHA also upregulates sterol regulatory factor binding protein 1 through activation of the HIF-1α/mTOR pathway, which in turn promotes the expression of lipid synthases, such as fatty acid synthase and acetyl-CoA carboxylase (69). Inhibition of LDHA may result in the reduction of precursors required for lipid synthesis by restoring TCA activity (42). Thus, LDH maintains the homeostasis of glucose metabolism while indirectly influencing the balance of other metabolic pathways, thus resulting in the rapid proliferation and survival of HCC cells (Fig. 2) (7).
Roles of LDH in the TME in HCC
The TME in HCC is a complex multicomponent environment composed of immune cell populations (including T lymphocytes, macrophages, neutrophils and dendritic cells) as well as non-immune components [such as fibroblasts, vascular and lymphatic endothelial cells and the extracellular matrix (ECM)] (70). The TME not only exerts tumor suppressive effects through immunosurveillance and immunomodulation but also promotes tumor cell invasion and metastasis as well as immune escape to achieve tumor progression (71,72).
Synergistically, LDH and monocarboxylic acid transporter protein coordinate lactate production and transport and serve an important role in the regulation of the tumor immune microenvironment (73-75). LDHA is expressed mainly in bone and liver tissues, but numerous studies have shown that it is also commonly expressed in malignant tumors (73-76,78). The stability of tumor-infiltrating T regulatory cells (Treg cells) is affected by LDHA knockdown or LDH inhibitors (77,78). In a previous study of immune cells in tumors, LDHA levels were found to be significantly elevated in activated lymphocytes, which can exert tumor suppressive effects by upregulating LDHA and engaging in glycolysis (79). LDH can also be involved in modulating the function of suppressor immune cells, such as T lymphocytes, which help tumor cells evade immune surveillance and promote the progression of HCC (80). The important role of LDH in the TME was pointed out in a study by Verma et al (78), which reported that the inhibition of LDH not only regulates the glycolytic balance between tumor cells and Tregs, but also alters the functional state of antitumor T cells (78). In HCC, LDH regulates tumor-associated macrophages (TAMs) and tumor-associated fibroblasts (TAFs) by modulating lactate production (81). Studies have shown that lactate can contribute to the polarization of TAMs to an M2-like phenotype, causing them to secrete cytokines and chemokines that promote tumor cell growth, angiogenesis and metastasis and facilitate tumor progression (82,83). In TAFs, lactate can stimulate the production of ECM and growth factors to form a supportive stromal environment for HCC cells (84). LDH-mediated production of lactate can also affect endothelial cell function in the tumor vasculature, promoting angiogenesis and increasing nutrient and oxygen supply to the tumor (85). Therefore, LDH can influence tumor development by affecting the function of immune cells and non-immune components in the immune microenvironment of HCC tumors (Fig. 3).
Roles of LDH in the diagnosis and prognostic evaluation of HCC
Owing to its wide distribution in tissues and its important role in metabolic reprogramming, LDH serves an important role in the development of different diseases. Numerous studies have suggested that LDH has an important role in the development of liver diseases such as liver injury, liver fibrosis and HCC (34,55,86). In a study of liver disease development, LDHA was found to affect the interaction between LDHA and HIF-1α in the classical Wnt signaling pathway, thereby regulating glycolysis and liver fibrosis in hepatic stellate cells (55). Salvianolic acid B can inhibit M1 macrophage polarization by downregulating the expression levels of LDHA, thus exerting a therapeutic effect on liver injury (87). A recent study has shown that the upregulation of LDHA expression in liver injury contributes to pericentral regeneration of the liver (86). Protein arginine methyltransferase 3 promotes HCC growth by enhancing arginine methylation of LDHA (88).
Since different organs contain different proportions of LDH isoforms, the proportion of LDH isoforms in the blood can help determine the source of released LDH (8,26). By measuring the activity of different LDH isoforms, it is possible to distinguish between different diseases, such as infections, hemolytic anemia, liver disease, kidney disease, heart disease and cancer (8,89). In studies of melanoma and non-Hodgkin's lymphoma, elevated serum total LDH levels and the levels of different subtypes of LDH were found to be useful not only for diagnosing tumors but also for the prognostic assessment of tumors (90,91). Serum LDH levels are often elevated in patients with HCC (92). Elevation of the serum LDH level is thought to be due to the release of LDH from tumor cells into the bloodstream, which may be due to cell necrosis or active secretion (27). Serum LDH levels have been shown to be correlated with tumor size, stage and metastasis in HCC (27,93). Measuring serum LDH activity can aid the early screening and diagnosis of HCC (94,95). Changes in the proportions of different LDH isoforms can also reflect the pathological type and stage of HCC to a certain extent, which can improve the accuracy of diagnosis (96,97). Combining serum LDH with other traditional biomarkers, such as a-fetoprotein (AFP), can improve diagnostic accuracy for HCC (98). In certain cases, patients with normal AFP levels but elevated serum LDH levels may still be at increased risk for HCC (99), suggesting that LDH can be used as an adjunctive diagnostic indicator, especially for AFP-negative HCC (100).
The expression patterns of LDH isoforms in tumor tissues can provide valuable information for tumor diagnosis and prognostic evaluation (101,102). LDH5 is usually upregulated in HCC samples compared with that of normal liver tissue, and the expression ratio of LDH5 to other LDH isoforms may be altered (103,104). Increased levels of LDH5 in tumor tissues are associated with more aggressive tumor behavior, including increased rates of proliferation and greater invasion and metastasis abilities (7,105). The expression levels of LDH isozymes can be used to predict the response to therapy in patients with HCC (101). Patients with high LDH5-expressing tumors may have a poorer response to conventional chemotherapy or targeted therapies, suggesting that LDH isoform analysis can aid in selecting personalized treatment regimens for patients with HCC (7). High serum LDH levels are associated with poor prognosis in patients with HCC (106). Patients with HCC with elevated serum LDH levels at multiple time points have shorter overall survival and higher recurrence rates after treatment, including surgical resection, liver transplantation and ablative therapy (2). Previous studies have shown that high preoperative serum LDH levels in patients with HCC tend to portend a poorer prognosis, including shorter survival and higher recurrence rates (2,106,107). The serum LDH level can be an independent prognostic factor, even after adjusting for other clinicopathological factors (108). LDH expression in tumor tissues also has important prognostic significance (101). Patients with high levels of LDH expression have a worse prognosis compared with that of patients with low levels of LDH expression (7,56,101). Monitoring LDH expression in tumor tissue may help clinicians stratify patients according to outcomes and determine more appropriate treatment strategies (109). However, the specificity of serum LDH as a diagnostic biomarker is limited because elevated LDH levels may be observed in other diseases, such as cirrhosis, hepatitis and other malignancies (99,100,110). Therefore, serum LDH has some applications in research as a tumor marker, especially in exploring energy metabolism in and the microenvironment of tumor cells, but it is not a commonly used tumor marker in clinical practice. Owing to its lack of specificity and insufficient sensitivity, serum LDH is usually not used as a preferred indicator for diagnosing or monitoring tumors. In practice, physicians usually use LDH in combination with other more specific tumor markers, imaging techniques and pathology to comprehensively determine a patient's condition (98-100).
LDH-based therapeutic strategies for HCC
Numerous small-molecule compounds are currently being developed to treat HCC, with the aim of inhibiting the activity of LDH and blocking abnormal glycolysis in cancer cells to suppress the proliferation of HCC cells and induce their apoptosis (111-114). Previous studies have suggested that the regulation of angiogenesis through the VEGF/VEGF-receptor signaling pathway, which is the target of tyrosine kinase inhibitors such as sorafenib, serves a key role in the progression of HCC (111,112). A previous study reported that knockdown of LDHA significantly inhibits the proliferation, migration and invasion of HCC cells and increases sensitivity to sorafenib (113). Polyadenylation-specific factor 6 causes changes in LDH to achieve Warburg effect-mediated immune escape and angiogenesis; contributes to cancer progression through c-Myc via the hexokinase, programmed death-ligand 1 and VEGF pathways, and has the potential to synergize with sorafenib to treat HCC (114). Given the key role of LDH in HCC metabolism and progression, LDH inhibitors have emerged as promising therapeutic agents for HCC (115,116). Several small-molecule inhibitors of LDH, such as FX11 and GNE-140 (117,118), have been developed. These inhibitors target the active site of LDH, block its enzymatic activity and disrupt the glycolytic pathway in HCC cells (116).
In preclinical studies, the targeted inhibition of LDH using drugs and other means has been shown to have significant antitumor effects in HCC cell lines and animal models (119). Treatment with LDH inhibitors results in decreased glycolytic flux, reduced ATP production and induction of HCC cell apoptosis (7,120). In addition, combining LDH inhibitors with other anticancer drugs, such as sorafenib, can, in some cases, enhance antitumor efficacy and overcome drug resistance (121). However, the translation of LDH inhibitors from preclinical to clinical use is hampered by issues such as poor solubility, off-target effects and limited bioavailability (122). The ability of gene therapy approaches such as RNA interference (RNAi) and CRISPR/Cas9-mediated gene editing have also been explored (123). RNAi-based strategies can specifically reduce the expression of the LDH gene, which results in reduced LDH protein levels and enzymatic activity (124). However, there are challenges related to RNAi and CRISPR/Cas9-based, such as achieving effective delivery to tumor cells, potential immune responses and off-target effects (125-127). Overcoming these challenges is critical for the successful development of gene therapy strategies targeting LDH for the treatment of HCC. Combining LDH-targeted therapies with traditional surgery, chemotherapy, radiotherapy and emerging immunotherapies to achieve synergistic effects may further increase the efficacy of HCC treatment and improve the quality of life and survival of patients (Table I).
Conclusion
LDH serves a key multidimensional role in the complex pathology of HCC and influences the developmental trajectory of HCC in numerous aspects. From the perspective of metabolic regulation, LDH actively maintains glycolysis to ensure that tumor cells can efficiently produce energy in an aerobic environment, providing a sufficient energy supply for the rapid proliferation of cancer cells (128). Moreover, LDH also finely regulates the redox balance, maintains the stability of the intracellular environment, and creates favorable conditions for the survival and proliferation of cancer cells (129). LDH also closely interacts with other metabolic pathways, further strengthening the metabolic adaptability and viability of tumor cells and promoting HCC occurrence, development, and metastasis (64). In the TME, LDH regulates the secretion of cytokines and chemokines to remodel the TME and promote the invasion and metastasis of tumor cells (31).
LDH has also shown value in the diagnosis and treatment of liver cancer, as LDH serves a central role in the disease (8). As a potential diagnostic marker for HCC, LDH can act as a sensitive indicator in the early stages of the disease, although it is not commonly used in clinical practice because of its lack of specificity. The combination of LDH with commonly used HCC markers can improve the accuracy of HCC diagnosis and can assist in the early screening and diagnosis of HCC (100). In addition, the expression patterns of different isoforms of LDH are important in the pathological typing and staging of HCC (8). Regarding the prognostic assessment of HCC, serum LDH levels are correlated with survival and recurrence rates and high LDH5 expression in tumor tissues is correlated with aggressive tumor behavior as well as the therapeutic response; thus, by using LDH, clinicians may accurately assess disease progression and therapeutic effects and formulate personalized therapeutic plans (7). Given the important role of LDH in the metabolic reprogramming of HCC as well as in the TME, the use of LDH as a therapeutic target is a key direction for the development of novel anticancer drugs. Despite the potential of LDH as a target for liver cancer therapy, numerous challenges to fully utilizing the therapeutic potential of targeting LDH remain. Currently, more specific and potent LDH inhibitors are needed; specifically, the ability of inhibitors to target LDH in tumor cells, their inhibitory effect and pharmacokinetic properties need to be improved to ensure that the drugs can exert their effects stably and efficiently in vivo and to reduce toxicity and side effects on normal tissues. In addition, inefficiency in delivering gene therapy components targeting LDH to cells as well as the risk of off-target effects are key obstacles to clinical application (122). Furthermore, the mechanisms underlying the complex interactions between LDH and other metabolic pathways and signaling pathways are not fully understood and an in-depth analysis of these mechanisms will provide strong support for the development of more precise and effective therapeutic strategies for HCC. In the future, LDH-related biomarkers have potential for use in clinical practice for early diagnosis, precise prognostic prediction and the selection of personalized therapeutic regimens for liver cancer. With the continuous advances in precision medicine and tumor metabolism research, targeting LDH may become an important approach for the comprehensive treatment of HCC, bringing new treatment opportunities for patients with liver cancer in the future.
Availability of data and materials
Not applicable.
Authors' contributions
HJ wrote the original draft. QL wrote the original draft and created the figures and tables. JL participated in the literature search and analysis of the data to be included in the review. SZ was assisted in the preparation of the figures and table. BT acquired funding acquisition and helped with review writing and editing. Data authentication is not applicable. All authors read and approved the final manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Acknowledgments
Not applicable.
Funding
The present study was supported by grants from The National Natural Science Foundation of China (grant no. 82460466), the Science and Technology Plan Project of Guizhou Province [grant nos. QIAN KE HE JI CHU-ZK (2024) YI BAN 323, QIAN KE HE JI CHU-ZK (2023) ZHONG DIAN 058, and QIAN KE HE JI CHU-ZK (2022) YI BAN 646].
References
Satriano L, Lewinska M, Rodrigues PM, Banales JM and Andersen JB: Metabolic rearrangements in primary liver cancers: Cause and consequences. Nat Rev Gastroenterol Hepatol. 16:748–766. 2019. View Article : Google Scholar : PubMed/NCBI | |
Tan EY, Danpanichkul P, Yong JN, Yu Z, Tan DJH, Lim WH, Koh B, Lim RYZ, Tham EKJ, Mitra K, et al: Liver cancer in 2021: Global burden of disease study. J Hepatol. 82:851–860. 2025. View Article : Google Scholar | |
Schmidt DR, Patel R, Kirsch DG, Lewis CA, Vander Heiden MG and Locasale JW: Metabolomics in cancer research and emerging applications in clinical oncology. CA Cancer J Clin. 71:333–358. 2021. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Comandatore A, Franczak M, Smolenski RT, Morelli L, Peters GJ and Giovannetti E: Lactate dehydrogenase and its clinical significance in pancreatic and thoracic cancers. Semin Cancer Biol. 86:93–100. 2022. View Article : Google Scholar : PubMed/NCBI | |
Sonveaux P, Végran F, Schroeder T, Wergin MC, Verrax J, Rabbani ZN, De Saedeleer CJ, Kennedy KM, Diepart C, Jordan BF, et al: Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice. J Clin Invest. 118:3930–3942. 2008.PubMed/NCBI | |
Sharma D, Singh M and Rani R: Role of LDH in tumor glycolysis: Regulation of LDHA by small molecules for cancer therapeutics. Semin Cancer Biol. 87:184–195. 2022. View Article : Google Scholar : PubMed/NCBI | |
Claps G, Faouzi S, Quidville V, Chehade F, Shen S, Vagner S and Robert C: The multiple roles of LDH in cancer. Nat Rev Clin Oncol. 19:749–762. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zhang H, Kang K, Chen S, Su Q, Zhang W, Zeng L, Lin X, Peng F, Lin J and Chai D: High serum lactate dehydrogenase as a predictor of cardiac insufficiency at follow-up in elderly patients with acute myocardial infarction. Arch Gerontol Geriatr. 117:1052532024. View Article : Google Scholar | |
Feng H, Wu J, Chen P, Wang J, Deng Y, Zhu G, Xian J, Huang L and Ouyang W: MicroRNA-375-3p inhibitor suppresses angiotensin II-induced cardiomyocyte hypertrophy by promoting lactate dehydrogenase B expression. J Cell Physiol. 234:14198–14209. 2019. View Article : Google Scholar : PubMed/NCBI | |
Dai C, Li Q, May HI, Li C, Zhang G, Sharma G, Sherry AD, Malloy CR, Khemtong C, Zhang Y, et al: Lactate dehydrogenase A governs cardiac hypertrophic growth in response to hemodynamic stress. Cell Rep. 32:1080872020. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Wu G, Li M, Hesse M, Ma Y, Chen W, Huang H, Liu Y, Xu W, Tang Y, et al: LDHA-mediated metabolic reprogramming promoted cardiomyocyte proliferation by alleviating ROS and inducing M2 macrophage polarization. Redox Biol. 56:1024462022. View Article : Google Scholar : PubMed/NCBI | |
Yamaguchi S, Abe M, Arakaki T, Arasaki O and Shimabukuro M: Prognostic value of lactate dehydrogenase for mid-term mortality in acute decompensated heart failure: A comparison to established biomarkers and brain natriuretic peptide. Heart Lung Circ. 29:1318–1327. 2020. View Article : Google Scholar | |
Wu X, Ye J, Cai W, Yang X, Zou Q, Lin J, Zheng H, Wang C, Chen L and Li Y: LDHA mediated degradation of extracellular matrix is a potential target for the treatment of aortic dissection. Pharmacol Res. 176:1060512022. View Article : Google Scholar : PubMed/NCBI | |
Ibraheem A, Nashwan AJ and Yassin MA: Elderly patient with hematological and neurological manifestations of undetermined origin: A diagnostic dilemma of pernicious anemia. Cureus. 15:e430452023.PubMed/NCBI | |
Wahhab Ali KA, Ahmed AA and Mohammed ST: Determination of serum myeloperoxidase (MPO) and lactate dehydrogenase (LDH) as a tumour marker in chronic myeloid leukaemia (CML). J Pak Med Assoc. 74(10 (Supple-8)): S283–S286. 2024. View Article : Google Scholar : PubMed/NCBI | |
Oriaifo IA, Gerard JM and Thomas SM: Diagnostic value of lactate dehydrogenase and uric acid as screening tools for malignancies in children. Pediatr Emerg Care. 38:e1327–e1331. 2022. View Article : Google Scholar : PubMed/NCBI | |
Labban H, Begum F, Paracha A, John V and Islam M: Hemolytic anemia and pancytopenia secondary to vitamin B12 deficiency: Evaluation and clinical significance. Cureus. 16:e572862024.PubMed/NCBI | |
Henry BM, Aggarwal G, Wong J, Benoit S, Vikse J, Plebani M and Lippi G: Lactate dehydrogenase levels predict coronavirus disease 2019 (COVID-19) severity and mortality: A pooled analysis. Am J Emerg Med. 38:1722–1726. 2020. View Article : Google Scholar : PubMed/NCBI | |
Assiri A, Al-Tawfiq JA, Al-Rabeeah AA, Al-Rabiah FA, Al-Hajjar S, Al-Barrak A, Flemban H, Al-Nassir WN, Balkhy HH, Al-Hakeem RF, et al: Epidemiological, demographic, and clinical characteristics of 47 cases of Middle East respiratory syndrome coronavirus disease from Saudi Arabia: A descriptive study. Lancet Infect Dis. 13:752–761. 2013. View Article : Google Scholar : PubMed/NCBI | |
Martinez-Outschoorn UE, Prisco M, Ertel A, Tsirigos A, Lin Z, Pavlides S, Wang C, Flomenberg N, Knudsen ES, Howell A, et al: Ketones and lactate increase cancer cell 'stemness', driving recurrence, metastasis and poor clinical outcome in breast cancer: Achieving personalized medicine via metabolo-genomics. Cell Cycle. 10:1271–1286. 2011. View Article : Google Scholar : PubMed/NCBI | |
Lippi G and Favaloro EJ: D-dimer is associated with severity of coronavirus disease 2019: A pooled analysis. Thromb Haemost. 120:876–878. 2020. View Article : Google Scholar : PubMed/NCBI | |
Serrano-Lorenzo P, Coya ON, López-Jimenez A, Blázquez A, Delmiro A, Lucia A, Arenas J and Martín MA; COVID-19 '12 Octubre' Hospital Clinical Biochemistry Study Group: Plasma LDH: A specific biomarker for lung affectation in COVID-19? Pract Lab Med. 25:e002262021. View Article : Google Scholar : PubMed/NCBI | |
Zhou Y, Qi M and Yang M: Current status and future perspectives of lactate dehydrogenase detection and medical implications: A review. Biosensors (Basel). 12:11452022. View Article : Google Scholar : PubMed/NCBI | |
Panteghini M: Lactate dehydrogenase: an old enzyme reborn as a COVID-19 marker (and not only). Clin Chem Lab Med. 58:1979–1981. 2020. View Article : Google Scholar : PubMed/NCBI | |
Khan AA, Allemailem KS, Alhumaydhi FA, Gowder SJT and Rahmani AH: The biochemical and clinical perspectives of lactate dehydrogenase: An enzyme of active metabolism. Endocr Metab Immune Disord Drug Targets. 20:855–868. 2020. View Article : Google Scholar | |
Forkasiewicz A, Dorociak M, Stach K, Szelachowski P, Tabola R and Augoff K: The usefulness of lactate dehydrogenase measurements in current oncological practice. Cell Mol Biol Lett. 25:352020. View Article : Google Scholar : PubMed/NCBI | |
Fondy TP and Kaplan NO: Structural and functional properties of the H and M subunits of lactic dehydrogenases. Ann N Y Acad Sci. 119:888–904. 1965. View Article : Google Scholar : PubMed/NCBI | |
Hicks KG, Cluntun AA, Schubert HL, Hackett SR, Berg JA, Leonard PG, Ajalla Aleixo MA, Zhou Y, Bott AJ, Salvatore SR, et al: Protein-metabolite interactomics of carbohydrate metabolism reveal regulation of lactate dehydrogenase. Science. 379:996–1003. 2023. View Article : Google Scholar : PubMed/NCBI | |
Eniafe J and Jiang S: The functional roles of TCA cycle metabolites in cancer. Oncogene. 40:3351–3363. 2021. View Article : Google Scholar : PubMed/NCBI | |
Tufail M, Jiang CH and Li N: Altered metabolism in cancer: Insights into energy pathways and therapeutic targets. Mol Cancer. 23:2032024. View Article : Google Scholar : PubMed/NCBI | |
Schumann G and Klauke R: New IFCC reference procedures for the determination of catalytic activity concentrations of five enzymes in serum: Preliminary upper reference limits obtained in hospitalized subjects. Clin Chim Acta. 327:69–79. 2003. View Article : Google Scholar | |
Roman W: Quantitative estimation of lactate dehydrogenase isoenzymes in serum. I. Review of methods and distribution in human tissues. Enzymologia. 36:189–219. 1969.PubMed/NCBI | |
Du D, Liu C, Qin M, Zhang X, Xi T, Yuan S, Hao H and Xiong J: Metabolic dysregulation and emerging therapeutical targets for hepatocellular carcinoma. Acta Pharm Sin B. 12:558–580. 2022. View Article : Google Scholar : PubMed/NCBI | |
Gupta GS: The lactate and the lactate dehydrogenase in inflammatory diseases and major risk factors in COVID-19 patients. Inflammation. 45:2091–2123. 2022. View Article : Google Scholar : PubMed/NCBI | |
Han HS, Kang G, Kim JS, Choi BH and Koo SH: Regulation of glucose metabolism from a liver-centric perspective. Exp Mol Med. 48:e2182016. View Article : Google Scholar : PubMed/NCBI | |
Piccinin E, Villani G and Moschetta A: Metabolic aspects in NAFLD, NASH and hepatocellular carcinoma: The role of PGC1 coactivators. Nat Rev Gastroenterol Hepatol. 16:160–174. 2019. View Article : Google Scholar | |
Yang F, Hilakivi-Clarke L, Shaha A, Wang Y, Wang X, Deng Y, Lai J and Kang N: Metabolic reprogramming and its clinical implication for liver cancer. Hepatology. 78:1602–1624. 2023. 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 | |
Zhao Y, Li M, Yao X, Fei Y, Lin Z, Li Z, Cai K, Zhao Y and Luo Z: HCAR1/MCT1 regulates tumor ferroptosis through the lactate-mediated AMPK-SCD1 activity and its therapeutic implications. Cell Rep. 33:1084872020. View Article : Google Scholar : PubMed/NCBI | |
Vander Heiden MG, Cantley LC and Thompson CB: Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science. 324:1029–1033. 2009. View Article : Google Scholar : PubMed/NCBI | |
Ganapathy-Kanniappan S: Molecular intricacies of aerobic glycolysis in cancer: Current insights into the classic metabolic phenotype. Crit Rev Biochem Mol Biol. 53:667–682. 2018. View Article : Google Scholar | |
Borrelli A, Bonelli P, Tuccillo FM, Goldfine ID, Evans JL, Buonaguro FM and Mancini A: Role of gut microbiota and oxidative stress in the progression of non-alcoholic fatty liver disease to hepatocarcinoma: Current and innovative therapeutic approaches. Redox Biol. 15:467–479. 2018. View Article : Google Scholar : PubMed/NCBI | |
Cheng Y, He J, Zuo B and He Y: Role of lipid metabolism in hepatocellular carcinoma. Discov Oncol. 15:2062024. View Article : Google Scholar : PubMed/NCBI | |
Guo D, Zhang X, Cui H, Yu D, Zhang H, Shi X, Pang C, Li J, Guo W and Zhang S: ACADL functions as a tumor suppressor in hepatocellular carcinoma metastasis by inhibiting matrix metalloproteinase 14. Front Oncol. 12:8214842022. View Article : Google Scholar : PubMed/NCBI | |
Cai J, Chen T, Jiang Z, Yan J, Ye Z, Ruan Y, Tao L, Shen Z, Liang X, Wang Y, et al: Bulk and single-cell transcriptome profiling reveal extracellular matrix mechanical regulation of lipid metabolism reprograming through YAP/TEAD4/ACADL axis in hepatocellular carcinoma. Int J Biol Sci. 19:2114–2131. 2023. View Article : Google Scholar : PubMed/NCBI | |
Ericksen RE, Lim SL, McDonnell E, Shuen WH, Vadiveloo M, White PJ, Ding Z, Kwok R, Lee P, Radda GK, et al: Loss of BCAA catabolism during carcinogenesis enhances mTORC1 activity and promotes tumor development and progression. Cell Metab. 29:1151–1165.e6. 2019. View Article : Google Scholar : PubMed/NCBI | |
Tian LY, Smit DJ and Jücker M: The Role of PI3K/AKT/mTOR signaling in hepatocellular carcinoma metabolism. Int J Mol Sci. 24:26522023. View Article : Google Scholar : PubMed/NCBI | |
Bao MHR and Wong CCL: Hypoxia, metabolic reprogramming, and drug resistance in liver cancer. Cells. 10:17152021. View Article : Google Scholar : PubMed/NCBI | |
Ng CKY, Dazert E, Boldanova T, Coto-Llerena M, Nuciforo S, Ercan C, Suslov A, Meier MA, Bock T, Schmidt A, et al: Integrative proteogenomic characterization of hepatocellular carcinoma across etiologies and stages. Nat Commun. 13:24362022. View Article : Google Scholar : PubMed/NCBI | |
An J, Oh M, Kim SY, Oh YJ, Oh B, Oh JH, Kim W, Jung JH, Kim HI, Kim JS, et al: PET-based radiogenomics supports mTOR pathway targeting for hepatocellular carcinoma. Clin Cancer Res. 28:1821–1831. 2022. View Article : Google Scholar : PubMed/NCBI | |
Chen J, Ding C, Chen Y, Hu W, Yu C, Peng C, Feng X, Cheng Q, Wu W, Lu Y, et al: ACSL4 reprograms fatty acid metabolism in hepatocellular carcinoma via c-Myc/SREBP1 pathway. Cancer Lett. 502:154–165. 2021. View Article : Google Scholar | |
Luo YD, Liu XY, Fang L, Yu HQ, Zhang YJ, Chen M, Zhang LD and Xie CM: Mutant Kras and mTOR crosstalk drives hepatocellular carcinoma development via PEG3/STAT3/BEX2 signaling. Theranostics. 12:7903–7919. 2022. View Article : Google Scholar : PubMed/NCBI | |
Lin D and Wu J: Hypoxia inducible factor in hepatocellular carcinoma: A therapeutic target. World J Gastroenterol. 21:12171–12178. 2015. View Article : Google Scholar : PubMed/NCBI | |
Wang F, Chen L, Kong D, Zhang X, Xia S, Liang B, Li Y, Zhou Y, Zhang Z, Shao J, et al: Canonical Wnt signaling promotes HSC glycolysis and liver fibrosis through an LDH-A/HIF-1α transcriptional complex. Hepatology. 79:606–623. 2024. View Article : Google Scholar | |
Faloppi L, Bianconi M, Memeo R, Casadei Gardini A, Giampieri R, Bittoni A, Andrikou K, Del Prete M, Cascinu S and Scartozzi M: Lactate dehydrogenase in hepatocellular carcinoma: something old, something new. Biomed Res Int. 2016:71962802016. View Article : Google Scholar : PubMed/NCBI | |
Feng J, Li J, Wu L, Yu Q, Ji J, Wu J, Dai W and Guo C: Emerging roles and the regulation of aerobic glycolysis in hepatocellular carcinoma. J Exp Clin Cancer Res. 39:1262020. View Article : Google Scholar : PubMed/NCBI | |
Vaupel P, Schmidberger H and Mayer A: The Warburg effect: Essential part of metabolic reprogramming and central contributor to cancer progression. Int J Radiat Biol. 95:912–919. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Li W, Bian Y, Li Y and Cong L: Multifaceted roles of aerobic glycolysis and oxidative phosphorylation in hepatocellular carcinoma. PeerJ. 11:e147972023. View Article : Google Scholar : PubMed/NCBI | |
Manerba M, Di Ianni L, Govoni M, Roberti M, Recanatini M and Di Stefano G: LDH inhibition impacts on heat shock response and induces senescence of hepatocellular carcinoma cells. Eur J Pharm Sci. 105:91–8. 2017. View Article : Google Scholar : PubMed/NCBI | |
Sun Z, Liu L, Liang H and Zhang L: Nicotinamide mononucleotide induces autophagy and ferroptosis via AMPK/mTOR pathway in hepatocellular carcinoma. Mol Carcinog. 63:577–588. 2024. View Article : Google Scholar : PubMed/NCBI | |
Certo M, Tsai CH, Pucino V, Ho PC and Mauro C: Lactate modulation of immune responses in inflammatory versus tumour microenvironments. Nat Rev Immunol. 21:151–161. 2021. View Article : Google Scholar | |
Jeong DW, Cho IT, Kim TS, Bae GW, Kim IH and Kim IY: Effects of lactate dehydrogenase suppression and glycerol-3-phosphate dehydrogenase overexpression on cellular metabolism. Mol Cell Biochem. 284:1–8. 2006. 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 | |
Lin J, Rao D, Zhang M and Gao Q: Metabolic reprogramming in the tumor microenvironment of liver cancer. J Hematol Oncol. 17:62024. View Article : Google Scholar : PubMed/NCBI | |
Zhang RN and Fan JG: Lipid metabolism-related long noncoding RNAs: A potential prognostic biomarker for hepatocellular carcinoma. World J Gastroenterol. 30:3799–3802. 2024. View Article : Google Scholar : PubMed/NCBI | |
Koukourakis MI, Kakouratos C, Kalamida D, Bampali Z, Mavropoulou S, Sivridis E and Giatromanolaki A: Hypoxia-inducible proteins HIF1α and lactate dehydrogenase LDH5, key markers of anaerobic metabolism, relate with stem cell markers and poor post-radiotherapy outcome in bladder cancer. Int J Radiat Biol. 92:353–363. 2016. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Zhao X, Jiang P, Deng X, Li Z, Tian F, Guo F, Li X and Wang S: Inhibition of mTORC1 signaling sensitizes hepatocellular carcinoma cells to glycolytic stress. Am J Cancer Res. 6:2289–2298. 2016.PubMed/NCBI | |
Sas Z, Cendrowicz E, Weinhäuser I and Rygiel TP: Tumor microenvironment of hepatocellular carcinoma: Challenges and opportunities for new treatment options. Int J Mol Sci. 23:37782022. View Article : Google Scholar : PubMed/NCBI | |
Fares J, Fares MY, Khachfe HH, Salhab HA and Fares Y: Molecular principles of metastasis: A hallmark of cancer revisited. Signal Transduct Target Ther. 5:282020. View Article : Google Scholar : PubMed/NCBI | |
Neophytou CM, Panagi M, Stylianopoulos T and Papageorgis P: The role of tumor microenvironment in cancer metastasis: Molecular mechanisms and therapeutic opportunities. Cancers (Basel). 13:20532021. View Article : Google Scholar : PubMed/NCBI | |
Peng X, He Z, Yuan D, Liu Z and Rong P: Lactic acid: The culprit behind the immunosuppressive microenvironment in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer. 1879:1891642024. 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 | |
Zhang Y, Zhai Z, Duan J, Wang X, Zhong J, Wu L, Li A, Cao M, Wu Y, Shi H, et al: Lactate: The mediator of metabolism and immunosuppression. Front Endocrinol (Lausanne). 13:9014952022. View Article : Google Scholar : PubMed/NCBI | |
Maeda M, Ko M, Mane MM, Cohen IJ, Shindo M, Vemuri K, Serganova I and Blasberg R: Genetic and drug inhibition of LDH-A: Effects on murine gliomas. Cancers (Basel). 14:23062022. View Article : Google Scholar : PubMed/NCBI | |
Sarkar T, Dhar S and Sa G: Tumor-infiltrating T-regulatory cells adapt to altered metabolism to promote tumor-immune escape. Curr Res Immunol. 2:132–141. 2021. View Article : Google Scholar | |
Verma S, Budhu S, Serganova I, Dong L, Mangarin LM, Khan JF, Bah MA, Assouvie A, Marouf Y, Schulze I, et al: Pharmacologic LDH inhibition redirects intratumoral glucose uptake and improves antitumor immunity in solid tumor models. J Clin Invest. 134:e1776062024. View Article : Google Scholar : PubMed/NCBI | |
O'Neill LA, Kishton RJ and Rathmell J: A guide to immunometabolism for immunologists. Nat Rev Immunol. 16:553–565. 2016. View Article : Google Scholar : PubMed/NCBI | |
Niu D, Luo T, Wang H, Xia Y and Xie Z: Lactic acid in tumor invasion. Clin Chim Acta. 522:61–69. 2021. View Article : Google Scholar : PubMed/NCBI | |
Han S, Bao X, Zou Y, Wang L, Li Y, Yang L, Liao A, Zhang X, Jiang X, Liang D, et al: d-lactate modulates M2 tumor-associated macrophages and remodels immunosuppressive tumor microenvironment for hepatocellular carcinoma. Sci Adv. 9:eadg26972023. View Article : Google Scholar : PubMed/NCBI | |
Jiang Y, Han Q, Zhao H and Zhang J: Promotion of epithelial-mesenchymal transformation by hepatocellular carcinoma-educated macrophages through Wnt2b/β-catenin/c-Myc signaling and reprogramming glycolysis. J Exp Clin Cancer Res. 40:132021. View Article : Google Scholar | |
Li D, Zhang T, Guo Y, Bi C, Liu M and Wang G: Biological impact and therapeutic implication of tumor-associated macrophages in hepatocellular carcinoma. Cell Death Dis. 15:4982024. View Article : Google Scholar : PubMed/NCBI | |
Jin M, Cao W, Chen B, Xiong M and Cao G: Tumor-derived lactate creates a favorable niche for tumor via supplying energy source for tumor and modulating the tumor microenvironment. Front Cell Dev Biol. 10:8088592022. View Article : Google Scholar : PubMed/NCBI | |
Ye J, Gao X, Huang X, Huang S, Zeng D, Luo W, Zeng C, Lu C, Lu L, Huang H, et al: Integrating single-cell and spatial transcriptomics to uncover and elucidate GP73-mediated pro-angiogenic regulatory networks in hepatocellular carcinoma. Research (Wash D C). 7:03872024.PubMed/NCBI | |
Wang S, Wang X, Shan Y, Tan Z, Su Y, Cao Y, Wang S, Dong J, Gu J and Wang Y: Region-specific cellular and molecular basis of liver regeneration after acute pericentral injury. Cell Stem Cell. 31:341–358.e7. 2024. View Article : Google Scholar : PubMed/NCBI | |
Hu S, Yang Z, Li L, Yan Q, Hu Y, Zhou F, Tan Y and Pei G: Salvianolic acid B alleviates liver injury by regulating lactate-mediated histone lactylation in macrophages. Molecules. 29:2362024. View Article : Google Scholar : PubMed/NCBI | |
Lei Y, Han P, Chen Y, Wang H, Wang S, Wang M, Liu J, Yan W, Tian D and Liu M: Protein arginine methyltransferase 3 promotes glycolysis and hepatocellular carcinoma growth by enhancing arginine methylation of lactate dehydrogenase A. Clin Transl Med. 12:e6862022. View Article : Google Scholar : PubMed/NCBI | |
Agarwala SS, Keilholz U, Gilles E, Bedikian AY, Wu J, Kay R, Stein CA, Itri LM, Suciu S and Eggermont AM: LDH correlation with survival in advanced melanoma from two large, randomised trials (Oblimersen GM301 and EORTC 18951). Eur J Cancer. 45:1807–1814. 2009. View Article : Google Scholar : PubMed/NCBI | |
Bouafia F, Drai J, Bienvenu J, Thieblemont C, Espinouse D, Salles G and Coiffier B: Profiles and prognostic values of serum LDH isoenzymes in patients with haematopoietic malignancies. Bull Cancer. 91:E229–E240. 2004.PubMed/NCBI | |
Ho J, de Moura MB, Lin Y, Vincent G, Thorne S, Duncan LM, Hui-Min L, Kirkwood JM, Becker D, Van Houten B and Moschos SJ: Importance of glycolysis and oxidative phosphorylation in advanced melanoma. Mol Cancer. 11:762012. View Article : Google Scholar : PubMed/NCBI | |
Rafaqat S, Sattar A, Khalid A and Rafaqat S: Role of liver parameters in diabetes mellitus-a narrative review. Endocr Regul. 57:200–220. 2023. View Article : Google Scholar : PubMed/NCBI | |
Dercle L, Ammari S, Roblin E, Bigorgne A, Champiat S, Taihi L, Plaian A, Hans S, Lakiss S, Tselikas L, et al: High serum LDH and liver metastases are the dominant predictors of primary cancer resistance to anti-PD(L)1 immunotherapy. Eur J Cancer. 177:80–93. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zhu W and Huang Y: Research progress of serum biomarkers for early screening of hepatocellular carcinoma. Zhonghua Gan Zang Bing Za Zhi. 29:308–312. 2021.In Chinese. PubMed/NCBI | |
Liao XM, Zhao SR, Dai WC and Fan R: Research advances of metabolomics in early diagnosis of hepatocellular carcinoma. Zhonghua Gan Zang Bing Za Zhi. 30:803–808. 2022.In Chinese. PubMed/NCBI | |
Sevinc A, Sari R and Fadillioglu E: The utility of lactate dehydrogenase isoenzyme pattern in the diagnostic evaluation of malignant and nonmalignant ascites. J Natl Med Assoc. 97:79–84. 2005.PubMed/NCBI | |
Nagasue N: Changes in lactic dehydrogenase isoenzymes after hepatic artery ligation in patients with hepatic carcinoma. Gastroenterol Jpn. 10:150–156. 1975. View Article : Google Scholar : PubMed/NCBI | |
Yan Q, Sun YS, An R, Liu F, Fang Q, Wang Z, Xu T, Chen L and Du J: Application and progress of the detection technologies in hepatocellular carcinoma. Genes Dis. 10:1857–1869. 2022. View Article : Google Scholar : PubMed/NCBI | |
Faloppi L, Scartozzi M, Bianconi M, Svegliati Baroni G, Toniutto P, Giampieri R, Del Prete M, De Minicis S, Bitetto D, Loretelli C, et al: The role of LDH serum levels in predicting global outcome in HCC patients treated with sorafenib: Implications for clinical management. BMC Cancer. 14:1102014. View Article : Google Scholar : PubMed/NCBI | |
Wang T and Zhang KH: New blood biomarkers for the diagnosis of AFP-negative hepatocellular carcinoma. Front Oncol. 10:13162020. View Article : Google Scholar : PubMed/NCBI | |
Kong W, Zuo X, Liang H, Hu J, Zhang H, Wang X and Chen W: Prognostic value of lactate dehydrogenase in patients with hepatocellular carcinoma: A meta-analysis. Biomed Res Int. 2018:17231842018. View Article : Google Scholar | |
Augoff K and Grabowski K: Significance of lactate dehydrogenase measurements in diagnosis of malignancies. Pol Merkur Lekarski. 17:644–647. 2004.In Polish. | |
Lee SC, Kao MC, Yin SJ and Lin CY: Serum lactate dehydrogenase isoenzymes in patients with hepatocellular carcinoma. Taiwan Yi Xue Hui Za Zhi. 81:218–223. 1982.PubMed/NCBI | |
Urbańska K and Orzechowski A: Unappreciated role of LDHA and LDHB to control apoptosis and autophagy in tumor cells. Int J Mol Sci. 20:20852019. View Article : Google Scholar | |
Yuan C, Li Z, Wang Y, Qi B, Zhang W, Ye J, Wu H, Jiang H, Song LN, Yang J and Cheng J: Overexpression of metabolic markers PKM2 and LDH5 correlates with aggressive clinicopathological features and adverse patient prognosis in tongue cancer. Histopathology. 65:595–605. 2014. View Article : Google Scholar : PubMed/NCBI | |
Su K, Huang W, Li X, Xu K, Gu T, Liu Y, Song J, Qian K, Xu Y, Zeng H, et al: Evaluation of lactate dehydrogenase and alkaline phosphatase as predictive biomarkers in the prognosis of hepatocellular carcinoma and development of a new nomogram. J Hepatocell Carcinoma. 10:69–79. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wu SJ, Lin YX, Ye H, Xiong XZ, Li FY and Cheng NS: Prognostic value of alkaline phosphatase, gamma-glutamyl transpeptidase and lactate dehydrogenase in hepatocellular carcinoma patients treated with liver resection. Int J Surg. 36:143–151. 2016. View Article : Google Scholar : PubMed/NCBI | |
Li J, Wu MF, Lu HW, Chen Q, Lin ZQ and Wang LJ: Pretreatment serum lactate dehydrogenase is an independent prognostic factor for patients receiving neoadjuvant chemotherapy for locally advanced cervical cancer. Cancer Med. 5:1863–1872. 2016. View Article : Google Scholar : PubMed/NCBI | |
Hu Z, Yuan Y, Hu Z, Liu Q, Fu Y, Hou J, Sun X, Li S, Duan W and Chen M: Development and validation of prognostic nomograms for hepatocellular carcinoma after hepatectomy based on inflammatory markers. J Hepatocell Carcinoma. 9:1403–1413. 2022. View Article : Google Scholar | |
Krishnamurthy K, Medina AM and Howard L: The utility of elevated serum lactate dehydrogenase in current clinical practice. Lab Med. 52:e17–e22. 2021. View Article : Google Scholar | |
Han L, Lin X, Yan Q, Gu C, Li M, Pan L, Meng Y, Zhao X, Liu S and Li A: PBLD inhibits angiogenesis via impeding VEGF/VEGFR2-mediated microenvironmental cross-talk between HCC cells and endothelial cells. Oncogene. 41:1851–1865. 2022. View Article : Google Scholar : PubMed/NCBI | |
Liu L, Cao Y, Chen C, Zhang X, McNabola A, Wilkie D, Wilhelm S, Lynch M and Carter C: Sorafenib blocks the RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in hepatocellular carcinoma model PLC/PRF/5. Cancer Res. 66:11851–11858. 2006. View Article : Google Scholar : PubMed/NCBI | |
Man S, Yao J, Lv P, Liu Y, Yang L and Ma L: Curcumin-enhanced antitumor effects of sorafenib via regulating the metabolism and tumor microenvironment. Food Funct. 11:6422–6432. 2020. View Article : Google Scholar : PubMed/NCBI | |
Sim DY, Lee HJ, Ahn CH, Park J, Park SY, Kil BJ, Shim BS, Kim B and Kim SH: Negative regulation of CPSF6 suppresses the warburg effect and angiogenesis leading to tumor progression via c-Myc signaling network: Potential therapeutic target for liver cancer therapy. Int J Biol Sci. 20:3442–3460. 2024. View Article : Google Scholar : PubMed/NCBI | |
Fiume L, Manerba M, Vettraino M and Di Stefano G: Inhibition of lactate dehydrogenase activity as an approach to cancer therapy. Future Med Chem. 6:429–445. 2014. View Article : Google Scholar : PubMed/NCBI | |
Miao P, Sheng S, Sun X, Liu J and Huang G: Lactate dehydrogenase A in cancer: A promising target for diagnosis and therapy. IUBMB Life. 65:904–910. 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 | |
Alobaidi B, Hashimi SM, Alqosaibi AI, AlQurashi N and Alhazmi S: Targeting the monocarboxylate transporter MCT2 and lactate dehydrogenase A LDHA in cancer cells with FX-11 and AR-C155858 inhibitors. Eur Rev Med Pharmacol Sci. 27:6605–6617. 2023.PubMed/NCBI | |
Li X, Lu P, Li B, Yang R, Chu Y, Zhang Z, Wan H, Niu C, Wang C and Luo K: Sensitization of hepatocellular carcinoma cells to irradiation by miR-34a through targeting lactate dehydrogenase-A. Mol Med Rep. 13:3661–3667. 2016. View Article : Google Scholar : PubMed/NCBI | |
Paul SK, Dutta Chowdhury K, Dey SR, Paul A and Haldar R: Exploring the possibility of drug repurposing for cancer therapy targeting human lactate dehydrogenase A: A computational approach. J Biomol Struct Dyn. 41:9967–9976. 2023. View Article : Google Scholar | |
Brower V: Sorafenib plus cisplatin for hepatocellular carcinoma. Lancet Oncol. 17:e4242016. View Article : Google Scholar : PubMed/NCBI | |
Han JH, Lee EJ, Park W, Ha KT and Chung HS: Natural compounds as lactate dehydrogenase inhibitors: Potential therapeutics for lactate dehydrogenase inhibitors-related diseases. Front Pharmacol. 14:12750002023. View Article : Google Scholar : PubMed/NCBI | |
Zheng R, Fang X, Chen X, Huang Y, Xu G, He L, Li Y, Niu X, Yang L, Wang L, et al: Knockdown of lactate dehydrogenase by adeno-associated virus-delivered CRISPR/Cas9 system alleviates primary hyperoxaluria type 1. Clin Transl Med. 10:e2612020. View Article : Google Scholar : PubMed/NCBI | |
Ariceta G, Barrios K, Brown BD, Hoppe B, Rosskamp R and Langman CB: Hepatic lactate dehydrogenase A: An RNA interference target for the treatment of all known types of primary hyperoxaluria. Kidney Int Rep. 6:1088–1098. 2021. View Article : Google Scholar : PubMed/NCBI | |
Tang Q and Khvorova A: RNAi-based drug design: Considerations and future directions. Nat Rev Drug Discov. 23:341–364. 2024. View Article : Google Scholar : PubMed/NCBI | |
Alshaer W, Zureigat H, Al Karaki A, Al-Kadash A, Gharaibeh L, Hatmal MM, Aljabali AAA and Awidi A: siRNA: Mechanism of action, challenges, and therapeutic approaches. Eur J Pharmacol. 905:1741782021. View Article : Google Scholar : PubMed/NCBI | |
Cheng X, Fan S, Wen C and Du X: CRISPR/Cas9 for cancer treatment: Technology, clinical applications and challenges. Brief Funct Genomics. 19:209–214. 2020. View Article : Google Scholar : PubMed/NCBI | |
Van Wilpe S, Koornstra R, Den Brok M, De Groot JW, Blank C, De Vries J, Gerritsen W and Mehra N: Lactate dehydrogenase: A marker of diminished antitumor immunity. Oncoimmunology. 9:17319422020. View Article : Google Scholar : PubMed/NCBI | |
Lin Y, Wang Y and Li PF: Mutual regulation of lactate dehydrogenase and redox robustness. Front Physiol. 13:10384212022. View Article : Google Scholar : PubMed/NCBI |