Research progress on lactatedehydrogenase and immunometabolism in cancer
Zhou Wenli1, Miao Mingyong 2
1.Department of Medical Oncology, Changzheng Hospital, Navy Medical University, Shanghai 200070, China;2.Department of Biochemistry and Molecular Biology, Navy Medical University, Shanghai 200433, China
Abstract:The metabolic characteristics are different in different immune cell subsets. The immune cells in antitumor adaptive immunity mainly rely on aerobic glycolysis and glutamine catabolism to obtain energy and metabolites. Immunosuppressive cells in promoting cancer occurrence and progression can use the metabolites from cancer cells to get energy and intermediate products through fatty acid metabolism or other manners. As the critical product of glycolysis, lactic acid directly or indirectly affects the biological behavior of cancer and immunometabolism in tumor microenvironment. Lactate dehydrogenase (LDH) is a key enzyme connecting the metabolism of immune cells by lactic acid metabolism, generally with high expression in cancers, which regulates immune response and participates in carcinogenesis and development by activating some signal transduction pathway. It is identified as an important driver of immunosuppressive microenvironment. The increase of LDH level is mainly caused by the increase of glycolysis activity and hypoxia necrosis in cancer. To some extent, the levels of LDH can reflect and be used for the assessment of glycolysis activity and immune metabolism. However, the roles of LDH in immunometabolic regulation varies in different immune cells such as T lymphocytes, macrophages and dendritic cells. This paper reviews the researches on LDH and metabolism of these immune cells to explore the possible application in evaluating prognosis, predicting and monitoring efficacy of anti-tumor treatment, especially in cancer immunotherapy.
周文丽,缪明永. 乳酸脱氢酶与肿瘤免疫代谢研究进展[J]. 肿瘤代谢与营养电子杂志, 2020, 7(4): 396-401.
Zhou Wenli, Miao Mingyong. Research progress on lactatedehydrogenase and immunometabolism in cancer. Electron J Metab Nutr Cancer, 2020, 7(4): 396-401.
1.HANAHAN D, WEINBERG R A. Hallmarks of cancer: the next generation[J]. Cell, 2011, 144(5):646-674.
2.LIBERTI M V, LOCASALE J W. The warburg effect: how does it benefit cancer cells[J]? Trends Biochem Sci, 2016, 41(3):211-218.
3.FONDY T P, KAPLAN N O. Structural and functional properties of the H and M subunits of lactic dehydrogenases[J]. Ann N Y Acad Sci, 1965, 119(3):888-904.
4.MARKERT C L, SHAKLEE J B, WHITT G S. Evolution of a gene. Multiple genes for LDH isozymes provide a model of the evolution of gene structure, function and regulation[J]. Science, 1975, 189(4197):102-114.
5.KOUKOURAKIS M I, GIATROMANOLAKI A, SIVRIDIS E, et al. Prognostic and predictive role of lactate dehydrogenase 5 expression in colorectal cancer patients treated with PTK787/ZK 222584 (vatalanib) antiangiogenic therapy[J]. Clin Cancer Res, 2011, 17(14):4892-4900.
6.JIA Z, ZHANG J, WANG Z, et al. An explorative analysis of the prognostic value of lactate dehydrogenase for survival and the chemotherapeutic response in patients with advanced triple-negative breast cancer[J]. Oncotarget, 2018, 9(12):10714-10722.
7.LIU X, MENG Q H, YE Y, et al. Prognostic significance of pretreatment serum levels of albumin, LDH and total bilirubin in patients with non-metastatic breast cancer[J]. Carcinogenesis, 2015, 36(2):243-248.
8.URBAN′SK A K, ORZECHOWSKI A. Unappreciated role of LDHA and LDHB to control apoptosis and autophagy in tumor cells[J]. Int J Mol Sci, 2019, 20(9):2085.
9.KOUKOURAKIS M I, GIATROMANOLAKI A, PANTELIADOU M, et al. Lactate dehydrogenase 5 isoenzyme overexpression defines resistance of prostate cancer to radiotherapy[J]. Br J Cancer, 2014, 110(9):2217-2223.
10.LIU J, CHEN G, LIU Z, et al. Aberrant FGFR tyrosine kinase signaling enhances the warburg effect by reprogramming LDH isoform expression and activity in prostate cancer[J]. Cancer Res, 2018, 78(16):4459.
11.MCCLELAND M L, ADLER A S, SHANG Y, et al. An integrated genomic screen identifies LDHB as an essential gene for triple-negative breast cancer[J]. Cancer Res, 2012, 72(22):5812.
12.CUI J, QUAN M, JIANG W, et al. Suppressed expression of LDHB promotes pancreatic cancer progression via inducing glycolytic phenotype[J]. Med Oncol, 2015, 32(5):143.
13.KURPIN′SK A A, SURAJ J, BONAR E, et al. Proteomic characterization of early lung response to breast cancer metastasis in mice[J]. Exp Mol Pathol, 2019, 107:129-140.
14.KIM J W, GAO P, LIU Y C, et al. Hypoxia-inducible factor 1 and dysregulated c-Myc cooperatively induce vascular endothelial growth factor and metabolic switches hexokinase 2 and pyruvate dehydrogenase kinase 1[J]. Mol Cell Biol, 2007, 27(21):7381-7393.
15.CUI X G, HAN Z T, HE S H, et al. HIF1/2α mediates hypoxia-induced LDHA expression in human pancreatic cancer cells[J]. Oncotarget, 2017, 8(15):24840-24852.
16.ZHAO Y H, ZHOU M, LIU H, et al. Upregulation of lactate dehydrogenase A by ErbB2 through heat shock factor 1 promotes breast cancer cell glycolysis and growth[J]. Oncogene, 2009, 28(42):3689-3701.
17.KIM H S, LEE H E, YANG H K, et al. High lactate dehydrogenase 5 expression correlates with high tumoral and stromal vascular endothelial growth factor expression in gastric cancer[J]. Pathobiology, 2014, 81(2):78-85.
18.KOUKOURAKIS M I, GIATROMANOLAKI A, SIVRIDIS E, et al. Lactate dehydrogenase 5 expression in operable colorectal cancer: strong association with survival and activated vascular endothelial growth factor pathway—a report of the tumour angiogenesis research group[J]. J Clin Oncol, 2006, 24(26):4301-4308.
19.SCARTOZZI M, GIAMPIERI R, MACCARONI E, et al. Pre-treatment lactate dehydrogenase levels as predictor of efficacy of first-line bevacizumab-based therapy in metastatic colorectal cancer patients[J]. Br J Cancer, 2012, 106(5):799-804.
20.BUCK M D, SOWELL R T, KAECH S M, et al. Metabolic instruction of immunity[J]. Cell, 2017, 169(4):570-586.
21.THORSSON V, GIBBS D L, BROWN S D, et al. The immune landscape of cancer[J]. Immunity, 2018, 48(4):812-830.e814.
22.PEARCE E L, PEARCE E J. Metabolic pathways in immune cell activation and quiescence[J]. Immunity, 2013, 38(4):633-643.
23.DOMBLIDES C, LARTIGUE L, FAUSTIN B. Metabolic stress in the immune function of T cells, macrophages and dendritic cells[J]. Cells, 2018, 7(7):68.
24.BUCK M D, OSULLIVAN D, PEARCE E L. T cell metabolism drives immunity[J]. J Exp Med, 2015, 212(9):1345-1360.
25.CHANG C H, CURTIS J D, MAGGI LB, J R, et al. Posttranscriptional control of T cell effector function by aerobic glycolysis[J]. Cell, 2013, 153(6):1239-1251.
26.HU Z, QU G, YU X, et al. Acylglycerol kinase maintains metabolic state and immune responses of CD8+ T cells[J]. Cell Metab, 2019, 30(2):290-302.e295.
27.GEIGER R, RIECKMANN J C, WOLF T, et al. L-Arginine modulates T cell metabolism and enhances survival and Anti-tumor activity[J]. Cell, 2016, 167(3):829-842.e813.
28.SHIME H, YABU M, AKAZAWA T, et al. Tumor-secreted lactic acid promotes IL-23/IL-17 proinflammatory pathway[J]. J Immunol, 2008, 180(11):7175-7183.
29.BRAND A, SINGER K, KOEHL G E, et al. LDHA-Associated lactic acid production blunts tumor immunosurveillance by T and NK cells[J]. Cell Metab, 2016, 24(5):657-671.
30.OHUE Y, NISHIKAWA H. Regulatory T (Treg) cells in cancer: can treg cells be a new therapeutic target[J]? Cancer Sci, 2019, 110(7):2080-2089.
31.KIM J H, KIM B S, LEE S K. Regulatory T cells in tumor microenvironment and approach for anticancer immunotherapy[J]. Immune Netw, 2020, 20(1):e4.
32.SHANG B, LIU Y, JIANG S J, et al. Prognostic value of tumor-infiltrating FoxP3+regulatory T cells in cancers: a systematic review and meta-analysis[J]. Sci Rep, 2015, 5:15179-15179.
33.ANGELIN A, GIL-DE-GóMEZ L, DAHIYA S, et al. Foxp3 reprograms T cell metabolism to function in low-glucose, high-lactate environments[J]. Cell Metab, 2017, 25(6):1282-1293.e1287.
34.CHANMEE T, ONTONG P, KONNO K, et al. Tumor-associated macrophages as major players in the tumor microenvironment[J]. Cancers (Basel), 2014, 6(3):1670-1690.
35.HUGHES R, QIAN B Z, ROWAN C, et al. Perivascular M2 macrophages stimulate tumor relapse after chemotherapy[J]. Cancer Res, 2015, 75(17):3479-3491.
36.COLEGIO O R, CHU N Q, SZABO A L, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid[J]. Nature, 2014, 513(7519):559-563.
37.ZHANG D, TANG Z, HUANG H, et al. Metabolic regulation of gene expression by histone lactylation[J]. Nature, 2019, 574(7779):575-580.
38.MARTíN P, DEL HOYO G M N, ANJURE F, et al. Concept of lymphoid versus myeloid dendritic cell lineages revisited: both CD8alpha(-) and CD8alpha(+) dendritic cells are generated from CD4(low) lymphoid-committed precursors[J]. Blood, 2000, 96(7):2511-2519.
39.PATENTE T A, PINHO M P, OLIVEIRA A A, et al. Human dendritic cells: their heterogeneity and clinical application potential in cancer immunotherapy[J]. Front Immunol, 2019, 9:3176.
40.KELLY B, ONEILL L A J. Metabolic reprogramming in macrophages and dendritic cells in innate immunity[J]. Cell Res, 2015, 25(7):771-784.
41.RYAN D G, ONEILL L A J. Krebs cycle rewired for macrophage and dendritic cell effector functions[J]. FEBS Lett, 2017, 591(19):2992-3006.
42.THWE P M, PELGROM L R, COOPER R, et al. Cell-intrinsic glycogen metabolism supports early glycolytic reprogramming required for dendritic cell immune responses[J]. Cell Metab, 2017, 26(3):558-567.e555.
43.WCULEK S K, KHOUILI S C, PRIEGO E, et al. Metabolic control of dendritic cell functions: digesting information[J]. Front Immunol, 2019, 10:775.
44.GUAK H, AL HABYAN S, MA E H, et al. Glycolytic metabolism is essential for CCR7 oligomerization and dendritic cell migration[J]. Nat Commun, 2018, 9(1):2463.
45.MALINARICH F, DUAN K, HAMID R A, et al. High mitochondrial respiration and glycolytic capacity represent a metabolic phenotype of human tolerogenic dendritic cells[J]. J Immunol, 2015, 194(11):5174-5186.
46.KRAWCZYK C M, HOLOWKA T, SUN J, et al. Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation[J]. Blood, 2010, 115(23):4742-4749.
47.CARONNI N, SIMONCELLO F, STAFETTA F, et al. Downregulation of membrane trafficking proteins and lactate conditioning determine loss of dendritic cell function in lung cancer[J]. Cancer Res, 2018, 78(7):1685.
48.LARKIN J, MINOR D, DANGELO S, et al. Overall survival in patients with advanced melanoma who received nivolumab versus investigators choice chemotherapy in checkMate 037: A randomized, controlled, open-label phase Ⅲ trial[J]. J Clin Oncol, 2018, 36(4):383-390.
49.BRAHMER J, RECKAMP K L, BAAS P, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer[J]. N Engl J Med, 2015, 373(2):123-135.
50.HODI F S, CHIARION-SILENI V, GONZALEZ R, et al. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial[J]. Lancet Oncol, 2018, 19(11):1480-1492.
51.OYA Y, YOSHIDA T, KURODA H, et al. Predictive clinical parameters for the response of nivolumab in pretreated advanced non-small-cell lung cancer[J]. Oncotarget, 2017, 8(61):103117-103128.
52.JOSEPH R W, ELASSAISS-SCHAAP J, KEFFORD R, et al. Baseline tumor size is an independent prognostic factor for overall survival in patients with melanoma treated with pembrolizumab[J]. Clin Cancer Res, 2018, 24(20):4960.
53.AGARWALA S S, KEILHOLZ U, GILLES E, et al. LDH correlation with survival in advanced melanoma from two large, randomised trials (Oblimersen GM301 and EORTC 18951)[J]. Eur J Cancer, 2009, 45(10):1807-1814.