Lactylation in cancer: Advances and opportunities for treatment resistance

Keke Xu , Yiyi Shou , Ruiqi Liu , Hao Xiong , Xiaomeng Dai , Xuanwen Bao , Xiaoyan Chen , Luanluan Huang , Hailong Sheng , Haibo Zhang , Yanwei Lu

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (10) : e70478

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (10) : e70478 DOI: 10.1002/ctm2.70478
REVIEW

Lactylation in cancer: Advances and opportunities for treatment resistance

Author information +
History +
PDF

Abstract

Background: Lactylation, a recently identified post-translational modification that utilizes lactic acidas a substrate, has emerged as an important regulator of gene expression andprotein function. Since its discovery in 2019, lactylation has beenincreasingly recognized for its roles in cancer biology and treatment response.

Main text: Lactylationis strongly associated with tumor progression and malignancy, underscoring itspotential as a therapeutic target. Recent studies also link lactylation tocancer treatment resistance, suggesting that modulating this modification couldenhance therapeutic efficacy. As treatment resistance remains a major clinicalchallenge in oncology, accumulating evidence indicates that dysregulatedlactylation contributes to resistance across chemotherapy, immunotherapy, targeted therapy, and radiotherapy. Preclinical and clinical research has begunto delineate the molecular pathways through which lactylation shapes theseresistance processes, and experimental approaches targeting lactylation arebeing explored to restore therapeutic sensitivity.

Conclusion: This review systematically summarizes the mechanisms of lactylation and its roles intreatment resistance, highlighting the interplay between lactylation andtherapeutic response. We discuss current and emerging strategies that targetlactylation, providing a foundation for future therapeutic development aimed atovercoming resistance and improving cancer treatment outcomes.

Keywords

chemotherapy / immunotherapy / lactylation / targeted therapy / treatment resistance

Cite this article

Download citation ▾
Keke Xu, Yiyi Shou, Ruiqi Liu, Hao Xiong, Xiaomeng Dai, Xuanwen Bao, Xiaoyan Chen, Luanluan Huang, Hailong Sheng, Haibo Zhang, Yanwei Lu. Lactylation in cancer: Advances and opportunities for treatment resistance. Clinical and Translational Medicine, 2025, 15(10): e70478 DOI:10.1002/ctm2.70478

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Warburg O. On the origin of cancer cells. Science. 1956; 123(3191): 309-14.

[2]

Brooks GA. Lactate as a fulcrum of metabolism. Redox Biol. 2020; 35: 101454.

[3]

Brown TP, Bhattacharjee P, Ramachandran S, et al. The lactate receptor GPR81 promotes breast cancer growth via a paracrine mechanism involving antigen-presenting cells in the tumor microenvironment. Oncogene. 2020; 39(16): 3292-3304.

[4]

Nii T, Prabhu VV, Ruvolo V, et al. Imipridone ONC212 activates orphan G protein-coupled receptor GPR132 and integrated stress response in acute myeloid leukemia. Leukemia. 2019; 33(12): 2805-2816.

[5]

Chen P, Zuo H, Xiong H, et al. Gpr132 sensing of lactate mediates tumor-macrophage interplay to promote breast cancer metastasis. Proc Natl Acad Sci USA. 2017; 114(3): 580-585.

[6]

Wang R, Li C, Cheng Z, Li M, Shi J, Zhang Z, et al. H3K9 lactylation in malignant cells facilitates CD8+ T cell dysfunction and poor immunotherapy response. Cell Rep. 2024; 43(9): 114686.

[7]

Ippolito L, Morandi A, Giannoni E, Lactate CP: A metabolic driver in the tumour landscape. Trends Biochem Sci. 2019; 44(2): 153-166.

[8]

Suhail Y, Cain MP, Vanaja K, Kurywchak PA, Levchenko A, Kalluri R, et al. Systems biology of cancer metastasis. Cell Syst. 2019; 9(2): 109-127.

[9]

Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019; 574(7779): 575-580.

[10]

Faubert B, Li KY, Cai L, et al. Lactate metabolism in human lung tumors. Cell. 2017; 171(2): 358-371.e9.

[11]

Zhu R, Ye X, Lu X, et al. “ACSS2 acts as a lactyl-CoA synthetase and couples KAT2A to function as a lactyltransferase for histone lactylation and tumor immune evasion.” Cell Metab. 2025; 37(2): 361–376.e7.

[12]

Liu R, Ren X, Park YE, et al. “Nuclear GTPSCS functions as a lactyl-CoA synthetase to promote histone lactylation and gliomagenesis.” Cell Metab. 2025; 37(2): 377–394.e9.

[13]

Wang Y, Wang Y, Patel H, et al. Epigenetic modification of m6A regulator proteins in cancer. Mol Cancer. 2023; 22(1): 102.

[14]

Gao R, Li Y, Xu Z, et al. Mitochondrial pyruvate carrier 1 regulates fatty acid synthase lactylation and mediates treatment of nonalcoholic fatty liver disease. Hepatology. 2023; 78(6): 1800-1815.

[15]

Xie B, Lin J, Chen X, et al. CircXRN2 suppresses tumor progression driven by histone lactylation through activating the Hippo pathway in human bladder cancer. Mol Cancer. 2023; 22(1): 151.

[16]

Yang K, Fan M, Wang X, et al. Lactate promotes macrophage HMGB1 lactylation, acetylation, and exosomal release in polymicrobial sepsis. Cell Death Differ. 2022; 29(1): 133-146.

[17]

Yano T, Muto M, Minashi K, et al. Photodynamic therapy as salvage treatment for local failure after chemoradiotherapy in patients with esophageal squamous cell carcinoma: a phase II study. Int J Cancer. 2012; 131(5): 1228-34.

[18]

Yu L, Gu C, Zhong D, et al. Induction of autophagy counteracts the anticancer effect of cisplatin in human esophageal cancer cells with acquired drug resistance. Cancer Lett. 2014; 355(1): 34-45.

[19]

Enzinger PC, Mayer RJ. Esophageal cancer. N Engl J Med. 2003; 349(23): 2241-52.

[20]

Kelland L. The resurgence of platinum-based cancer chemotherapy. Nat Rev Cancer. 2007; 7(8): 573-84.

[21]

Chen H, Li Y, Li H, et al. NBS1 lactylation is required for efficient DNA repair and chemotherapy resistance. Nature. 2024; 631(8021): 663-669.

[22]

De Leo A, Ugolini A, Yu X, et al. Glucose-driven histone lactylation promotes the immunosuppressive activity of monocyte-derived macrophages in glioblastoma. Immunity. 2024; 57(5): 1105-1123.e8.

[23]

Chen Y, Wu J, Zhai L, et al. Metabolic regulation of homologous recombination repair by MRE11 lactylation. Cell. 2024; 187(2): 294-311.e21.

[24]

Williams RS, Williams JS, Tainer JA. Mre11-Rad50-Nbs1 is a keystone complex connecting DNA repair machinery, double-strand break signaling, and the chromatin template. Biochem Cell Biol. 2007; 85(4): 509-20.

[25]

Sajid A, Rahman H, Ambudkar SV. Advances in the structure, mechanism and targeting of chemoresistance-linked ABC transporters. Nat Rev Cancer. 2023; 23(11): 762-779.

[26]

Duan W, Liu W, Xia S, et al. Warburg effect enhanced by AKR1B10 promotes acquired resistance to pemetrexed in lung cancer-derived brain metastasis. J Transl Med. 2023; 21(1): 547.

[27]

Sun X, He L, Liu H, et al. The diapause-like colorectal cancer cells induced by SMC4 attenuation are characterized by low proliferation and chemotherapy insensitivity. Cell Metab. 2023; 35(9): 1563-1579.e8.

[28]

Chen X, Yang M, Yin J, et al. Tumor-associated macrophages promote epithelial-mesenchymal transition and the cancer stem cell properties in triple-negative breast cancer through CCL2/AKT/β-catenin signaling. Cell Commun Signal. 2022; 20(1): 92.

[29]

Li F, Zhang H, Huang Y, et al. Single-cell transcriptome analysis reveals the association between histone lactylation and cisplatin resistance in bladder cancer. Drug Resist Updat. 2024; 73: 101059.

[30]

Sun Y, Shen W, Hu S, et al. METTL3 promotes chemoresistance in small cell lung cancer by inducing mitophagy. J Exp Clin Cancer Res. 2023; 42(1): 65.

[31]

Liu ZD, Shi YH, Xu QC, et al. CSNK2A1 confers gemcitabine resistance to pancreatic ductal adenocarcinoma via inducing autophagy. Cancer Lett. 2024; 585: 216640.

[32]

Young MJ, Wang SA, Chen YC, et al. USP24-i-101 targeting of USP24 activates autophagy to inhibit drug resistance acquired during cancer therapy. Cell Death Differ. 2024; 31(5): 574-591.

[33]

Cao LL, Wu YK, Lin TX, et al. CDK5 promotes apoptosis and attenuates chemoresistance in gastric cancer via E2F1 signaling. Cancer Cell Int. 2023; 23(1): 286.

[34]

Zhu H, Chen K, Chen Y, et al. RNA-binding protein ZCCHC4 promotes human cancer chemoresistance by disrupting DNA-damage-induced apoptosis. Signal Transduct Target Ther. 2022; 7(1): 240.

[35]

Huang Y, Luo G, Peng K, et al. Lactylation stabilizes TFEB to elevate autophagy and lysosomal activity. J Cell Biol. 2024; 223(11): e202308099.

[36]

Jia M, Yue X, Sun W, et al. ULK1-mediated metabolic reprogramming regulates Vps34 lipid kinase activity by its lactylation. Sci Adv. 2023; 9(22): eadg4993.

[37]

Jiang Y, Zhang H, Wang J, Targeting extracellular matrix stiffness and mechanotransducers to improve cancer therapy. J Hematol Oncol. 2022; 15(1): 34.

[38]

Tang Y, Zang H, Wen Q, Fan S. AXL in cancer: a modulator of drug resistance and therapeutic target. J Exp Clin Cancer Res. 2023; 42(1): 148.

[39]

Liu J, Cao X. Glucose metabolism of TAMs in tumor chemoresistance and metastasis. Trends Cell Biol. 2023; 33(11): 967-978.

[40]

Allis CD, Berger SL, Cote J, et al. “New nomenclature for chromatin-modifying enzymes.” Cell 2007; 131(4): 633–6.

[41]

Sabari BR, Zhang D, Allis CD, et al. “Metabolic regulation of gene expression through histone acylations.” Nat Rev Mol Cell Biol. 2017; 18(2): 90–101.

[42]

Deng J, Li Y, Yin L, et al. Histone lactylation enhances GCLC expression and thus promotes chemoresistance of colorectal cancer stem cells through inhibiting ferroptosis. Cell Death Dis. 2025; 16(1): 193.

[43]

Liu R, Wu J, Guo H, et al. Post-translational modifications of histones: mechanisms, biological functions, and therapeutic targets. MedComm (2020). 2023; 4(3): e292.

[44]

Rho H, Terry AR, Chronis C, et al. “Hexokinase 2-mediated gene expression via histone lactylation is required for hepatic stellate cell activation and liver fibrosis.” Cell Metab. vol. 35,8 (2023): 1406–1423.e8.

[45]

Johnson DB, Nebhan CA, Moslehi JJ, Balko JM. Immune-checkpoint inhibitors: long-term implications of toxicity. Nat Rev Clin Oncol. 2022; 19(4): 254-267.

[46]

Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011; 144(5): 646-74.

[47]

Vesely MD, Zhang T, Chen L. Resistance mechanisms to anti-PD cancer immunotherapy. Annu Rev Immunol. 2022; 40: 45-74.

[48]

Chen L, Han X. Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J Clin Invest. 2015; 125(9): 3384-91.

[49]

Vesely MD, Chen L. Normalization cancer immunotherapy for melanoma. J Invest Dermatol. 2020; 140(6): 1134-1142.

[50]

Sanmamed MF, Chen L. A paradigm shift in cancer immunotherapy: from enhancement to normalization. Cell. 2018; 175(2): 313-326.

[51]

Park I, Larson PE, Zierhut ML, et al. Hyperpolarized 13C magnetic resonance metabolic imaging: application to brain tumors. Neuro Oncol. 2010; 12(2): 133-44.

[52]

Deng H, Kan A, Lyu N, He M, Huang X, Qiao S, et al. Tumor-derived lactate inhibit the efficacy of lenvatinib through regulating PD-L1 expression on neutrophil in hepatocellular carcinoma. J Immunother Cancer. 2021; 9(6): e002305.

[53]

Chaudagar K, Hieromnimon HM, Kelley A, et al. Suppression of tumor cell lactate-generating signaling pathways eradicates murine PTEN/p53-deficient aggressive-variant prostate cancer via macrophage phagocytosis. Clin Cancer Res. 2023; 29(23): 4930-4940.

[54]

Chaudagar K, Hieromnimon HM, Khurana R, et al. Reversal of lactate and PD-1-mediated macrophage immunosuppression controls growth of PTEN/p53-deficient prostate cancer. Clin Cancer Res. 2023; 29(10): 1952-1968.

[55]

Wang J, Yang P, Yu T, et al. Lactylation of PKM2 suppresses inflammatory metabolic adaptation in pro-inflammatory macrophages. Int J Biol Sci. 2022; 18(16): 6210-6225.

[56]

Colegio OR, Chu NQ, Szabo AL, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. 2014; 513(7519): 559-63.

[57]

Zong Z, Xie F, Wang S, et al. Alanyl-tRNA synthetase, AARS1, is a lactate sensor and lactyltransferase that lactylates p53 and contributes to tumorigenesis. Cell. 2024; 187(10): 2375-2392.e33.

[58]

Li H, Liu C, Li R, et al. AARS1 and AARS2 sense L-lactate to regulate cGAS as global lysine lactyltransferases. Nature. 2024.

[59]

Xiong J, He J, Zhu J, et al. Lactylation-driven METTL3-mediated RNA m6A modification promotes immunosuppression of tumor-infiltrating myeloid cells. Mol Cell. 2022; 82(9): 1660-1677.e10.

[60]

Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017; 27(1): 109-118.

[61]

Ghiringhelli F, Puig PE, Roux S, et al. Tumor cells convert immature myeloid dendritic cells into TGF-beta-secreting cells inducing CD4+CD25+ regulatory T cell proliferation. J Exp Med. 2005; 202(7): 919-29.

[62]

Wang W, Wang X, Lu S, et al. Metabolic disturbance and Th17/Treg imbalance are associated with progression of gingivitis. Front Immunol. 2021; 12: 670178.

[63]

Rastogi S, Mishra SS, Arora MK, et al. Lactate acidosis and simultaneous recruitment of TGF-β leads to alter plasticity of hypoxic cancer cells in tumor microenvironment. Pharmacol Ther. 2023; 250: 108519.

[64]

Takahashi H, Alves CRR, Stanford KI, et al. TGF-β2 is an exercise-induced adipokine that regulates glucose and fatty acid metabolism. Nat Metab. 2019; 1(2): 291-303.

[65]

Gu J, Zhou J, Chen Q, Xu X, et al. Tumor metabolite lactate promotes tumorigenesis by modulating MOESIN lactylation and enhancing TGF-β signaling in regulatory T cells. Cell Rep. 2022; 40(3): 111122.

[66]

Chen J, Zhao D, Wang Y, et al. Lactylated apolipoprotein C-II induces immunotherapy resistance by promoting extracellular lipolysis. Adv Sci (Weinh). 2024; 11(38): e2406333.

[67]

Ansa-Addo EA, Zhang Y, Yang Y, et al. Membrane-organizing protein moesin controls Treg differentiation and antitumor immunity via TGF-β signaling. J Clin Invest. 2017; 127(4): 1321-1337.

[68]

Wang S, Huang T, Wu Q, et al. Lactate reprograms glioblastoma immunity through CBX3-regulated histone lactylation. J Clin Invest. 2024; 134(22): e176851.

[69]

Ye D, Zhou S, Dai X, et al. Targeting the MHC-I endosomal-lysosomal trafficking pathway in cancer: from mechanism to immunotherapy. Biochim Biophys Acta Rev Cancer. 2024; 1879(5): 189161.

[70]

Zhang C, Zhou L, Zhang M, et al. H3K18 lactylation potentiates immune escape of non-small cell lung cancer. Cancer Res. 2024.

[71]

Zhou C, Li W, Liang Z, et al. Mutant KRAS-activated circATXN7 fosters tumor immunoescape by sensitizing tumor-specific T cells to activation-induced cell death. Nat Commun. 2024; 15(1): 499.

[72]

Tyl MD, Merengwa VU, Cristea IM. Infection-induced lysine lactylation enables herpesvirus immune evasion. Sci Adv. 2025; 11(2): eads6215.

[73]

Liu R, Wu J, Guo H, et al. Post-translational modifications of histones: mechanisms, biological functions, and therapeutic targets. MedComm (2020). 2023; 4(3): e292.

[74]

Ganesh K, Massagué J. Targeting metastatic cancer. Nat Med. 2021; 27(1): 34-44.

[75]

Pajak B, Siwiak E, Sołtyka M, et al. 2-Deoxy-D-glucose and its analogs: from diagnostic to therapeutic agents. Int J Mol Sci. 2019; 21(1): 234.

[76]

Geschwind JF, Georgiades CS, Ko YH, Pedersen PL. Recently elucidated energy catabolism pathways provide opportunities for novel treatments in hepatocellular carcinoma. Expert Rev Anticancer Ther. 2004; 4(3): 449-57.

[77]

Sheng SL, Liu JJ, Dai YH, Sun XG, Xiong XP, Huang G. Knockdown of lactate dehydrogenase A suppresses tumor growth and metastasis of human hepatocellular carcinoma. FEBS J. 2012; 279(20): 3898-910.

[78]

Yao J, Liu J, Zhao W. By blocking hexokinase-2 phosphorylation, limonin suppresses tumor glycolysis and induces cell apoptosis in hepatocellular carcinoma. Onco Targets Ther. 2018; 11: 3793-3803.

[79]

Kirkland JL, Tchkonia T. Senolytic drugs: from discovery to translation. J Intern Med. 2020; 288(5): 518-536.

[80]

Wang Y, Hao F, Nan Y, et al. PKM2 inhibitor shikonin overcomes the cisplatin resistance in bladder cancer by inducing necroptosis. Int J Biol Sci. 2018; 14(13): 1883-1891.

[81]

Martin SP, Fako V, Dang H, et al. PKM2 inhibition may reverse therapeutic resistance to transarterial chemoembolization in hepatocellular carcinoma. J Exp Clin Cancer Res. 2020; 39(1): 99.

[82]

Amorim R, Pinheiro C, Miranda-Gonçalves V, et al. Monocarboxylate transport inhibition potentiates the cytotoxic effect of 5-fluorouracil in colorectal cancer cells. Cancer Lett. 2015; 365(1): 68-78.

[83]

Zhao Z, Han F, Yang S, Wu J, Zhan W. Oxamate-mediated inhibition of lactate dehydrogenase induces protective autophagy in gastric cancer cells: involvement of the Akt-mTOR signaling pathway. Cancer Lett. 2015; 358(1): 17-26.

[84]

Shelley MD, Hartley L, Fish RG, et al. Stereo-specific cytotoxic effects of gossypol enantiomers and gossypolone in tumour cell lines. Cancer Lett. 1999; 135(2): 171-80.

[85]

Kim EY, Chung TW, Han CW, et al. A novel lactate dehydrogenase inhibitor, 1-(phenylseleno)-4-(trifluoromethyl) benzene, suppresses tumor growth through apoptotic cell death. Sci Rep. 2019; 9(1): 3969.

[86]

Park JH, Kundu A, Lee SH, et al. Specific pyruvate kinase M2 inhibitor, compound 3K, induces autophagic cell death through disruption of the glycolysis pathway in ovarian cancer cells. Int J Biol Sci. 2021; 17(8): 1895-1908.

[87]

George K, Thomas NS, Malathi R. 4,4'-Diisothiocyanatostilbene-2,2'-disulfonate modulates voltage-gated K+ current and influences cell cycle arrest in androgen sensitive and insensitive human prostate cancer cell lines. Toxicol Mech Methods. 2020; 30(5): 358-369.

[88]

Liu Y, Guo JZ, Liu Y, et al. Nuclear lactate dehydrogenase A senses ROS to produce α-hydroxybutyrate for HPV-induced cervical tumor growth. Nat Commun. 2018; 9(1): 4429.

[89]

Liu X, Zou X, Zhou Y, Chen R, Peng Y, Qu M. LDHA and LDHB overexpression promoted the Warburg effect in malignantly transformed GES-1 cells induced by N-nitroso compounds. Food Chem Toxicol. 2023; 180: 114007.

[90]

Qing Y, Dong L, Gao L, et al. R-2-hydroxyglutarate attenuates aerobic glycolysis in leukemia by targeting the FTO/m6A/PFKP/LDHB axis. Mol Cell. 2021; 81(5): 922-939.e9.

[91]

Dorneburg C, Fischer M, Barth TFE, et al. LDHA in neuroblastoma is associated with poor outcome and its depletion decreases neuroblastoma growth independent of aerobic glycolysis. Clin Cancer Res. 2018; 24(22): 5772-5783.

[92]

Huo N, Cong R, Sun ZJ, et al. STAT3/LINC00671 axis regulates papillary thyroid tumor growth and metastasis via LDHA-mediated glycolysis. Cell Death Dis. 2021; 12(9): 799.

[93]

Pouysségur J, Marchiq I, Parks SK, Durivault J, Ždralević M, Vucetic M. ‘Warburg effect’ controls tumor growth, bacterial, viral infections and immunity—genetic deconstruction and therapeutic perspectives. Semin Cancer Biol. 2022; 86(Pt 2): 334-346.

[94]

Cheng A, Zhang P, Wang B, et al. Aurora-A mediated phosphorylation of LDHB promotes glycolysis and tumor progression by relieving the substrate-inhibition effect. Nat Commun. 2019; 10(1): 5566.

[95]

Tian LR, Lin MZ, Zhong HH, et al. Nanodrug regulates lactic acid metabolism to reprogram the immunosuppressive tumor microenvironment for enhanced cancer immunotherapy. Biomater Sci. 2022; 10(14): 3892-3900.

[96]

Ma J, Tang L, Tan Y, et al. Lithium carbonate revitalizes tumor-reactive CD8+ T cells by shunting lactic acid into mitochondria. Nat Immunol. 2024; 25(3): 552-561.

[97]

Brandstetter G, Blatt S, Goldschmitt J, Taylor L, Heymann P, Al-Nawas B, Ziebart T. Targeted sensitization of tumor cells for radiation through monocarboxylate transporters 1 and 4 inhibition in vitro. Clin Oral Investig. 2021; 25(1): 295-310.

[98]

Harris T, Eliyahu G, Frydman L, Degani H. Kinetics of hyperpolarized 13C1-pyruvate transport and metabolism in living human breast cancer cells. Proc Natl Acad Sci USA. 2009; 106(43): 18131-6.

[99]

Wang ZH, Zhang P, Peng WB, et al. Altered phenotypic and metabolic characteristics of FOXP3+CD3+CD56+ natural killer T (NKT)-like cells in human malignant pleural effusion. Oncoimmunology. 2022; 12(1): 2160558.

[100]

Yu Y, Huang X, Liang C, Zhang P. Evodiamine impairs HIF1A histone lactylation to inhibit Sema3A-mediated angiogenesis and PD-L1 by inducing ferroptosis in prostate cancer. Eur J Pharmacol. 2023; 957: 176007.

[101]

Pan L, Feng F, Wu J, et al. Demethylzeylasteral targets lactate by inhibiting histone lactylation to suppress the tumorigenicity of liver cancer stem cells. Pharmacol Res. 2022; 181: 106270.

[102]

Benson AB, Venook AP, Adam M, et al. NCCN Guidelines® Insights: rectal cancer, Version 3.2024. J Natl Compr Canc Netw. 2024; 22(6): 366-375.

[103]

Li W, Zhou C, Yu L, 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. 2024; 20(1): 114-130.

[104]

Azzam EI, Jay-Gerin JP, Pain D. Ionizing radiation-induced metabolic oxidative stress and prolonged cell injury. Cancer Lett. 2012; 327(1-2): 48-60.

[105]

Huang RX, Zhou PK. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduct Target Ther. 2020; 5(1): 60.

[106]

Xu J, Zhu W, Xu W, et al. Silencing of MBD1 reverses pancreatic cancer therapy resistance through inhibition of DNA damage repair. Int J Oncol. 2013; 42(6): 2046-52.

[107]

Watanabe S, Watanabe K, Akimov V, et al. JMJD1C demethylates MDC1 to regulate the RNF8 and BRCA1-mediated chromatin response to DNA breaks. Nat Struct Mol Biol. 2013; 20(12): 1425-33.

[108]

Maréchal A, Zou L. DNA damage sensing by the ATM and ATR kinases. Cold Spring Harb Perspect Biol. 2013; 5(9): a012716.

[109]

Li G, Wang D, Zhai Y, Pan C, Zhang J, Wang C, et al. Glycometabolic reprogramming-induced XRCC1 lactylation confers therapeutic resistance in ALDH1A3-overexpressing glioblastoma. Cell Metab. 2024; 36(8): 1696-1710.e10.

[110]

Yang Y, Chong Y, Chen M, et al. Targeting lactate dehydrogenase A improves radiotherapy efficacy in non-small cell lung cancer: from bedside to bench. J Transl Med. 2021; 19: 170.

[111]

Chaube B, Malv P, Singh SV, et al. Targeting metabolic flexibility by simultaneously inhibiting respiratory complex I and lactate generation retards melanoma progression. Oncotarget. 2015; 6: 37281-37299.

[112]

Rai G, Brimacombe KR, et al. Discovery and optimization of potent, cell-active pyrazole-based inhibitors of lactate dehydrogenase (LDH). J Med Chem. 2017; 60: 9184-9204.

[113]

Le A, Cooper CR, Gouw AM, et al. Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc Natl Acad Sci USA. 2010; 107: 2037-2042.

[114]

Halford S, Veal GJ, Wedge SR, et al. A Phase I dose-escalation study of AZD3965, an oral monocarboxylate transporter 1 inhibitor, in patients with advanced cancer. Clin Cancer Res. 2023; 29: 1429-1439.

[115]

Noble RA, Bell N, Blair H, et al. Inhibition of monocarboxyate transporter 1 by AZD3965 as a novel therapeutic approach for diffuse large B-cell lymphoma and Burkitt lymphoma. Haematologica. 2017; 102: 1247-1257.

[116]

Barros LF. Metabolic signaling by lactate in the brain. Trends Neurosci. 2013; 36: 396-404.

[117]

Lai FL. & Gao F. Auto-Kla: a novel web server to discriminate lysine lactylation sites using automated machine learning. Brief. Bioinform. 2023; 24: bbad070.

[118]

Levitin HM, Yuan J. & Sims PA. Single-cell transcriptomic analysis of tumor heterogeneity. Trends Cancer 2018; 4: 264-268.

[119]

Khoo BL, Grenci G, Lim Y, et al. Expansion of patient-derived circulating tumor cells from liquid biopsies using a CTC microfluidic culture device. Nat Protoc. 2018; 13: 34-58.

[120]

Meng Z, Zhang X, Tan H. & Lian H. Zinc-enriched nanosystem for dual glycolysis regulation and photothermal therapy to synergistically inhibit primary melanoma and lung metastasis. Chem Eng J. 2022; 435: 134781.

[121]

Karlsson MJ, Svedman FC, Tebani A, et al. Inflammation and apolipoproteins are potential biomarkers for stratification of cutaneous melanoma patients for immunotherapy and targeted therapy. Cancer Res. 2021; 81: 2545-2555.

[122]

Qiao T, Xiong Y, Feng Y, et al. Inhibition of LDH-A by oxamate enhances the efficacy of anti-PD-1 treatment in an NSCLC humanized mouse model. Front Oncol. 2021; 11: 632364.

[123]

Bonnet S, Archer SL, Turner JA, et al. A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth. Cancer Cell. 2007; 11: 37-51.

[124]

Hong H, Han H, Wang L, et al. ABCF1-K430-Lactylation promotes HCC malignant progression via transcriptional activation of HIF1 signaling pathway. Cell Death Differ. Published online 2025.

[125]

Tian Z, Yang K, Yao Y, et al. Catalytically selective chemotherapy from tumor-metabolic generated lactic acid. Small. 2019; 15: e1903746.

[126]

Sun K, Shi Y, Yan C, et al. Glycolysis-derived lactate induces ACSL4 expression and lactylation to activate ferroptosis during intervertebral disc degeneration. Adv Sci (Weinh). 2025; 12(21): e2416149.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

6

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/