Engineering lactate-modulating nanomedicines for cancer therapy.

Lactate in tumors has long been considered "metabolic junk" derived from the glycolysis of cancer cells and utilized only as a biomarker of malignancy, but is presently believed to be a pivotal regulator of tumor development, maintenance and metastasis. Indeed, tumor lactate can be a "fuel" for energy supply and functions as a signaling molecule, which actively contributes to tumor progression, angiogenesis, immunosuppression, therapeutic resistance, etc., thus providing promising opportunities for cancer treatment. However, the current approaches for regulating lactate homeostasis with available agents are still challenging, which is mainly due to the short half-life, low bioavailability and poor specificity of these agents and their unsatisfactory therapeutic outcomes. In recent years, lactate modulation nanomedicines have emerged as a charming and efficient strategy for fighting cancer, which play important roles in optimizing the delivery of lactate-modulating agents for more precise and effective modulation and treatment. Integrating specific lactate-modulating functions in diverse therapeutic nanomedicines may overcome the intrinsic restrictions of different therapeutic modalities by remodeling the pathological microenvironment for achieving enhanced cancer therapy. In this review, the most recent advances in the engineering of functional nanomedicines that can modulate tumor lactate for cancer therapy are summarized and discussed, and the fundamental mechanisms by which lactate modulation benefits various therapeutics are elucidated. Finally, the challenges and perspectives of this emerging strategy in the anti-tumor field are highlighted.

[1]  Pan‐Chyr Yang,et al.  An acid degradable, lactate oxidizing nanoparticle formulation for non-small cell lung cancer virotherapy , 2022, Nano Today.

[2]  Yannan Yang,et al.  A Sub-6 nm MnFe2O4-dichloroacetic acid nanocomposite modulates tumor metabolism and catabolism for reversing tumor immunosuppressive microenvironment and boosting immunotherapy. , 2022, Biomaterials.

[3]  Juan Li,et al.  Localized NIR-II laser mediated chemodynamic therapy of glioblastoma , 2022, Nano Today.

[4]  M. Mazurczak,et al.  Phase II study of dichloroacetate, an inhibitor of pyruvate dehydrogenase, in combination with chemoradiotherapy for unresected, locally advanced head and neck squamous cell carcinoma , 2022, Investigational New Drugs.

[5]  Heliang Yao,et al.  Microbiotic nanomedicine for tumor-specific chemotherapy-synergized innate/adaptive antitumor immunity , 2022, Nano Today.

[6]  J. Ren,et al.  Yeast@MOF bioreactor as a tumor metabolic symbiosis disruptor for the potent inhibition of metabolically heterogeneous tumors , 2022, Nano Today.

[7]  Shiying Li,et al.  Self-Delivery Ternary Bioregulators for Photodynamic Amplified Immunotherapy by Tumor Microenvironment Reprogramming. , 2022, ACS nano.

[8]  Bingjun Sun,et al.  Synergetic lethal energy depletion initiated by cancer cell membrane camouflaged nano-inhibitor for cancer therapy , 2022, Nano Research.

[9]  He Lian,et al.  Zinc-enriched nanosystem for dual glycolysis regulation and photothermal therapy to synergistically inhibit primary melanoma and lung metastasis , 2022, Chemical Engineering Journal.

[10]  Wei-Hai Chen,et al.  A Self-Driven Bioreactor Based on Bacterium-Metal-Organic Framework Biohybrids for Boosting Chemotherapy via Cyclic Lactate Catabolism. , 2021, ACS nano.

[11]  H. Khan,et al.  Targeting lactate metabolism and glycolytic pathways in the tumor microenvironment by natural products: A promising strategy in combating cancer , 2021, BioFactors.

[12]  Mo Yang,et al.  Dual‐Depletion of Intratumoral Lactate and ATP with Radicals Generation for Cascade Metabolic‐Chemodynamic Therapy , 2021, Advanced science.

[13]  Lisi Xie,et al.  Metal-Phenolic Network-Enabled Lactic Acid Consumption Reverses Immunosuppressive Tumor Microenvironment for Sonodynamic Therapy. , 2021, ACS nano.

[14]  Zhijie Jiang,et al.  Bi-directional nanosystem breaking tumor microenvironment and vessels induced negative Möbius ring. , 2021, Journal of controlled release : official journal of the Controlled Release Society.

[15]  Pei Zhang,et al.  Lactate in the tumour microenvironment: From immune modulation to therapy , 2021, EBioMedicine.

[16]  Yan Yang,et al.  Nanofactory for metabolic and chemodynamic therapy: pro-tumor lactate trapping and anti-tumor ROS transition , 2021, Journal of Nanobiotechnology.

[17]  Yan Hu,et al.  Activatable Biomineralized Nanoplatform Remodels the Intracellular Environment of Multidrug-Resistant Tumors for Enhanced Ferroptosis/Apoptosis Therapy. , 2021, Small.

[18]  Qigang Wang,et al.  Peroxisome inspired hybrid enzyme nanogels for chemodynamic and photodynamic therapy , 2021, Nature Communications.

[19]  Leilei Tian,et al.  Advanced Cancer Starvation Therapy by Simultaneous Deprivation of Lactate and Glucose Using a MOF Nanoplatform , 2021, Advanced science.

[20]  Pan‐Chyr Yang,et al.  Potential therapeutics using tumor-secreted lactate in nonsmall cell lung cancer. , 2021, Drug discovery today.

[21]  Yu Wang,et al.  Lactate Consumption via Cascaded Enzymes Combined VEGF siRNA for Synergistic Anti‐Proliferation and Anti‐Angiogenesis Therapy of Tumors , 2021, Advanced healthcare materials.

[22]  D. Haas-Kogan,et al.  Harnessing Lactate Metabolism for Radiosensitization , 2021, Frontiers in Oncology.

[23]  Qi Wang,et al.  Zwitterion-based hydrogen sulfide nanomotors induce multiple acidosis in tumor cells by destroying tumor metabolic symbiosis. , 2021, Angewandte Chemie.

[24]  Masaki Kobayashi,et al.  Transport function, regulation, and biology of human monocarboxylate transporter 1 (hMCT1) and 4 (hMCT4). , 2021, Pharmacology & therapeutics.

[25]  Guanbin Song,et al.  Tumor-Targeted Disruption of Lactate Transport with Reactivity-Reversible Nanocatalysts to Amplify Oxidative Damage. , 2021, Small.

[26]  F. Jiang,et al.  Recent Advances in Nanomaterial‐Based Nanoplatforms for Chemodynamic Cancer Therapy , 2021, Advanced Functional Materials.

[27]  M. Bruchez,et al.  Monotherapy and Combination Therapy Using Anti‐Angiogenic Nanoagents to Fight Cancer , 2021, Advanced materials.

[28]  R. Basaraba,et al.  Lactate Metabolism and Signaling in Tuberculosis and Cancer: A Comparative Review , 2021, Frontiers in Cellular and Infection Microbiology.

[29]  Xiaobing Zhang,et al.  An Acidity-Unlocked Nanoplatform Enables Self-Boosting ROS Generation through Upregulation of Lactate for Highly Specific Chemodynamic Therapy. , 2021, Angewandte Chemie.

[30]  J. Rothstein,et al.  Metabolic support of tumor-infiltrating regulatory T cells by lactic acid , 2020, Nature.

[31]  C. Donohoe,et al.  The oncogenic and clinical implications of lactate induced immunosuppression in the tumour microenvironment. , 2020, Cancer letters.

[32]  Dongkai Wang,et al.  Multifunctional Nanoparticles Boost Cancer Immunotherapy Based on Modulating the Immunosuppressive Tumor Microenvironment. , 2020, ACS applied materials & interfaces.

[33]  A. Yamauchi,et al.  Nanoparticle-Mediated Delivery of 2-Deoxy-D-Glucose Induces Antitumor Immunity and Cytotoxicity in Liver Tumors in Mice , 2020, Cellular and molecular gastroenterology and hepatology.

[34]  K. Cai,et al.  Engineering of Cascade-Responsive Nanoplatform to Inhibit Lactate Efflux for Enhanced Tumor Chemo-Immunotherapy. , 2020, ACS nano.

[35]  A. Griffioen,et al.  Oncometabolites lactate and succinate drive pro-angiogenic macrophage response in tumors. , 2020, Biochimica et biophysica acta. Reviews on cancer.

[36]  Ping-Chih Ho,et al.  Lactate modulation of immune responses in inflammatory versus tumour microenvironments , 2020, Nature Reviews Immunology.

[37]  Si-Xue Cheng,et al.  Intervening Tumor Progression by Coupling Bacteria Respiration with Tumor Metabolism. , 2020, Angewandte Chemie.

[38]  S. Dhar,et al.  Metabolic Modulation of the Tumor Microenvironment Leads to Multiple Checkpoint Inhibition and Immune Cell Infiltration. , 2020, ACS nano.

[39]  Shumin Li,et al.  Openwork@Dendritic Mesoporous Silica Nanoparticles for Lactate Depletion and Tumor Microenvironment Regulation. , 2020, Angewandte Chemie.

[40]  Xiaoyan Zhang,et al.  Enhanced antitumor effects of follicle-stimulating hormone receptor-mediated hexokinase-2 depletion on ovarian cancer mediated by a shift in glucose metabolism , 2020, Journal of Nanobiotechnology.

[41]  Samir Mitragotri,et al.  Physical triggering strategies for drug delivery. , 2020, Advanced drug delivery reviews.

[42]  Hyesun Hyun,et al.  Harnessing nanomedicine to overcome the immunosuppressive tumor microenvironment , 2020, Acta Pharmacologica Sinica.

[43]  Zhijie Jiang,et al.  Jet‐Lagged Nanoparticles Enhanced Immunotherapy Efficiency through Synergistic Reconstruction of Tumor Microenvironment and Normalized Tumor Vasculature , 2020, Advanced healthcare materials.

[44]  V. Ganapathy,et al.  The lactate receptor GPR81 promotes breast cancer growth via a paracrine mechanism involving antigen-presenting cells in the tumor microenvironment , 2020, Oncogene.

[45]  R. Mahato,et al.  Organic Nanocarriers for Delivery and Targeting of Therapeutic Agents for Cancer Treatment , 2020 .

[46]  Yufang Zhu,et al.  Porphyrin‐Based Metal–Organic Frameworks for Biomedical Applications , 2020, Angewandte Chemie.

[47]  W. Bu,et al.  Amorphous Fe‐Based Nanoagents for Self‐Enhanced Chemodynamic Therapy by Re‐Establishing Tumor Acidosis , 2019, Advanced Functional Materials.

[48]  Yi Zhang,et al.  Tumor-intrinsic signaling pathways: key roles in the regulation of the immunosuppressive tumor microenvironment , 2019, Journal of Hematology & Oncology.

[49]  Mei‐Zhen Zou,et al.  Intra/Extracellular Lactic Acid Exhaustion for Synergistic Metabolic Therapy and Immunotherapy of Tumors , 2019, Advanced materials.

[50]  J. Manzo-Merino,et al.  Lactate in the Regulation of Tumor Microenvironment and Therapeutic Approaches , 2019, Front. Oncol..

[51]  Y. Qu,et al.  Catalytically Selective Chemotherapy from Tumor-Metabolic Generated Lactic Acid. , 2019, Small.

[52]  Xuesi Chen,et al.  Advances in nanomedicine for cancer starvation therapy , 2019, Theranostics.

[53]  Yucai Wang,et al.  Near-Infrared II Phototherapy Induces Deep Tissue Immunogenic Cell Death and Potentiates Cancer Immunotherapy. , 2019, ACS nano.

[54]  Qiu-Yun Chen,et al.  The NIR inspired nano-CuSMn(II) composites for lactate and glycolysis attenuation. , 2019, Colloids and surfaces. B, Biointerfaces.

[55]  Zhuang Liu,et al.  Hollow Cu2Se Nanozymes for Tumor Photothermal-Catalytic Therapy , 2019, Chemistry of Materials.

[56]  Pierre Sonveaux,et al.  Monocarboxylate transporters in cancer , 2019, Molecular metabolism.

[57]  Yu Chen,et al.  Nanocatalytic Medicine , 2019, Advanced materials.

[58]  K. Kataoka,et al.  Nanomedicines for Reactive Oxygen Species Mediated Approach: An Emerging Paradigm for Cancer Treatment. , 2019, Accounts of chemical research.

[59]  Kyung Soo Park,et al.  Cancer nanomedicine for combination cancer immunotherapy , 2019, Nature Reviews Materials.

[60]  R. Gillies,et al.  Causes, consequences, and therapy of tumors acidosis , 2019, Cancer and Metastasis Reviews.

[61]  Huarong Liu,et al.  Tumor Reoxygenation and Blood Perfusion Enhanced Photodynamic Therapy using Ultrathin Graphdiyne Oxide Nanosheets. , 2019, Nano letters.

[62]  Yifeng Zhang,et al.  Checkpoint blockade and nanosonosensitizer-augmented noninvasive sonodynamic therapy combination reduces tumour growth and metastases in mice , 2019, Nature Communications.

[63]  Yu Chen,et al.  Nanocatalysts-augmented Fenton chemical reaction for nanocatalytic tumor therapy. , 2019, Biomaterials.

[64]  Yu Chen,et al.  Reactive Oxygen Species (ROS)-Based Nanomedicine. , 2019, Chemical reviews.

[65]  Qin Xu,et al.  Reprogramming Tumor Immune Microenvironment (TIME) and Metabolism via Biomimetic Targeting Codelivery of Shikonin/JQ1. , 2019, Nano letters.

[66]  Yucai Wang,et al.  Nanoenabled Modulation of Acidic Tumor Microenvironment Reverses Anergy of Infiltrating T Cells and Potentiates Anti-PD-1 Therapy. , 2019, Nano letters.

[67]  Ning Wang,et al.  Tumor microenvironment-driven non-cell-autonomous resistance to antineoplastic treatment , 2019, Molecular Cancer.

[68]  E. Giannoni,et al.  Lactate: A Metabolic Driver in the Tumour Landscape. , 2019, Trends in biochemical sciences.

[69]  Heliang Yao,et al.  Nanocatalytic Tumor Therapy by Biomimetic Dual Inorganic Nanozyme‐Catalyzed Cascade Reaction , 2018, Advanced science.

[70]  M. Morris,et al.  Cellular Uptake of MCT1 Inhibitors AR-C155858 and AZD3965 and Their Effects on MCT-Mediated Transport of L-Lactate in Murine 4T1 Breast Tumor Cancer Cells , 2019, The AAPS Journal.

[71]  H. Lane,et al.  Dual Inhibition of the Lactate Transporters MCT1 and MCT4 Is Synthetic Lethal with Metformin due to NAD+ Depletion in Cancer Cells , 2018, Cell reports.

[72]  Dirk Mossmann,et al.  mTOR signalling and cellular metabolism are mutual determinants in cancer , 2018, Nature Reviews Cancer.

[73]  Xiaofei Li,et al.  Lactate dehydrogenase A: A key player in carcinogenesis and potential target in cancer therapy , 2018, Cancer medicine.

[74]  Pan‐Chyr Yang,et al.  Targeting Tumor Microenvironment by Bioreduction-Activated Nanoparticles for Light-Triggered Virotherapy. , 2018, ACS nano.

[75]  F. Alam,et al.  Lactate biosensing: The emerging point-of-care and personal health monitoring. , 2018, Biosensors & bioelectronics.

[76]  E. Giannoni,et al.  Increased Lactate Secretion by Cancer Cells Sustains Non-cell-autonomous Adaptive Resistance to MET and EGFR Targeted Therapies. , 2018, Cell metabolism.

[77]  Yu Chen,et al.  Gas‐Generating Nanoplatforms: Material Chemistry, Multifunctionality, and Gas Therapy , 2018, Advanced materials.

[78]  Shibo Wang,et al.  Interfering with Lactate‐Fueled Respiration for Enhanced Photodynamic Tumor Therapy by a Porphyrinic MOF Nanoplatform , 2018, Advanced Functional Materials.

[79]  Jian Yu,et al.  Immunogenic effects of chemotherapy-induced tumor cell death , 2018, Genes & diseases.

[80]  P. Schreiner,et al.  Computational Chemistry: The Fate of Current Methods and Future Challenges. , 2018, Angewandte Chemie.

[81]  Qiu-Yun Chen,et al.  An aptamer-Fe3+ modified nanoparticle for lactate oxidation and tumor photodynamic therapy. , 2018, Colloids and surfaces. B, Biointerfaces.

[82]  O. Feron,et al.  Interruption of lactate uptake by inhibiting mitochondrial pyruvate transport unravels direct antitumor and radiosensitizing effects , 2018, Nature Communications.

[83]  Yuzhuo Wang,et al.  Engineering Multifunctional RNAi Nanomedicine To Concurrently Target Cancer Hallmarks for Combinatorial Therapy. , 2018, Angewandte Chemie.

[84]  J. Pouysségur,et al.  Disrupting the 'Warburg effect' re-routes cancer cells to OXPHOS offering a vulnerability point via 'ferroptosis'-induced cell death. , 2017, Advances in biological regulation.

[85]  J. Guan,et al.  Suppression of FIP200 and autophagy by tumor-derived lactate promotes naïve T cell apoptosis and affects tumor immunity , 2017, Science Immunology.

[86]  Joshua D. Rabinowitz,et al.  Glucose feeds the TCA cycle via circulating lactate , 2017, Nature.

[87]  Xiaoyuan Chen,et al.  Nanotechnology for Multimodal Synergistic Cancer Therapy. , 2017, Chemical reviews.

[88]  Weijian Chen,et al.  Polymer Prodrug-Based Nanoreactors Activated by Tumor Acidity for Orchestrated Oxidation/Chemotherapy. , 2017, Nano letters.

[89]  Jianlin Shi,et al.  Tumor-selective catalytic nanomedicine by nanocatalyst delivery , 2017, Nature Communications.

[90]  D. Wallace,et al.  Foxp3 Reprograms T Cell Metabolism to Function in Low-Glucose, High-Lactate Environments. , 2017, Cell metabolism.

[91]  S. Halford,et al.  A first-in-human first-in-class (FIC) trial of the monocarboxylate transporter 1 (MCT1) inhibitor AZD3965 in patients with advanced solid tumours. , 2017 .

[92]  Liangzhu Feng,et al.  H2O2-responsive liposomal nanoprobe for photoacoustic inflammation imaging and tumor theranostics via in vivo chromogenic assay , 2017, Proceedings of the National Academy of Sciences.

[93]  W. Wagner,et al.  Stimulation of lactate receptor (HCAR1) affects cellular DNA repair capacity. , 2017, DNA repair.

[94]  P. Kantoff,et al.  Cancer nanomedicine: progress, challenges and opportunities , 2016, Nature Reviews Cancer.

[95]  Pierre Sonveaux,et al.  Monocarboxylate transporters in the brain and in cancer☆ , 2016, Biochimica et biophysica acta.

[96]  S. Horvath,et al.  MCT1 Modulates Cancer Cell Pyruvate Export and Growth of Tumors that Co-express MCT1 and MCT4. , 2016, Cell reports.

[97]  P. Zhu,et al.  A novel small-molecule compound targeting CD147 inhibits the motility and invasion of hepatocellular carcinoma cells , 2016, Oncotarget.

[98]  M. Moyer,et al.  Monocarboxylate transport inhibition potentiates the cytotoxic effect of 5-fluorouracil in colorectal cancer cells. , 2015, Cancer letters.

[99]  C. Drake,et al.  Immune checkpoint blockade: a common denominator approach to cancer therapy. , 2015, Cancer cell.

[100]  W. Zhan,et al.  Oxamate-mediated inhibition of lactate dehydrogenase induces protective autophagy in gastric cancer cells: involvement of the Akt-mTOR signaling pathway. , 2015, Cancer letters.

[101]  S. Weinberg,et al.  Targeting mitochondria metabolism for cancer therapy. , 2015, Nature chemical biology.

[102]  E. Golden,et al.  Radiotherapy and immunogenic cell death. , 2015, Seminars in radiation oncology.

[103]  Jun Hu,et al.  2-Deoxy-D-glucose targeting of glucose metabolism in cancer cells as a potential therapy. , 2014, Cancer letters.

[104]  F. Sotgia,et al.  Catabolic cancer-associated fibroblasts transfer energy and biomass to anabolic cancer cells, fueling tumor growth. , 2014, Seminars in cancer biology.

[105]  Yuanzhong Wu,et al.  Downregulation of MCT1 inhibits tumor growth, metastasis and enhances chemotherapeutic efficacy in osteosarcoma through regulation of the NF-κB pathway. , 2014, Cancer letters.

[106]  Matthew G. Vander Heiden,et al.  Metabolic targets for cancer therapy , 2013, Nature Reviews Drug Discovery.

[107]  Z. Husain,et al.  Tumor-derived lactate and myeloid-derived suppressor cells , 2013, Oncoimmunology.

[108]  Z. Huang,et al.  Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment , 2013, Cell Death and Disease.

[109]  P. Johnston,et al.  Cancer drug resistance: an evolving paradigm , 2013, Nature Reviews Cancer.

[110]  E. Rajnavölgyi,et al.  Dendritic Cell Reprogramming by Endogenously Produced Lactic Acid , 2013, The Journal of Immunology.

[111]  M. Nitti,et al.  Role of Glutathione in Cancer Progression and Chemoresistance , 2013, Oxidative medicine and cellular longevity.

[112]  E. Michelakis,et al.  Pyruvate dehydrogenase kinase as a novel therapeutic target in oncology , 2013, Front. Oncol..

[113]  J. Schlesselman,et al.  A phase I dose-escalation trial of 2-deoxy-d-glucose alone or combined with docetaxel in patients with advanced solid tumors , 2013, Cancer Chemotherapy and Pharmacology.

[114]  M. Vettraino,et al.  Galloflavin, a new lactate dehydrogenase inhibitor, induces the death of human breast cancer cells with different glycolytic attitude by affecting distinct signaling pathways. , 2012, European journal of pharmaceutical sciences : official journal of the European Federation for Pharmaceutical Sciences.

[115]  Richard A Flavell,et al.  Combination delivery of TGF-β inhibitor and IL-2 by nanoscale liposomal polymeric gels enhances tumour immunotherapy. , 2012, Nature materials.

[116]  Bin Hu,et al.  Tumor lactic acidosis suppresses CTL function by inhibition of p38 and JNK/c‐Jun activation , 2012, International journal of cancer.

[117]  E. White Deconvoluting the context-dependent role for autophagy in cancer , 2012, Nature Reviews Cancer.

[118]  R. Jain,et al.  Normalization of tumour blood vessels improves the delivery of nanomedicines in a size-dependent manner , 2012, Nature nanotechnology.

[119]  Drew M. Pardoll,et al.  The blockade of immune checkpoints in cancer immunotherapy , 2012, Nature Reviews Cancer.

[120]  R. Hamanaka,et al.  Targeting glucose metabolism for cancer therapy , 2012, The Journal of experimental medicine.

[121]  M. Recanatini,et al.  Galloflavin (CAS 568‐80‐9): A Novel Inhibitor of Lactate Dehydrogenase , 2012, ChemMedChem.

[122]  O. Feron,et al.  Lactate shuttles at a glance: from physiological paradigms to anti-cancer treatments , 2011, Disease Models & Mechanisms.

[123]  W. Wilson,et al.  Targeting hypoxia in cancer therapy , 2011, Nature Reviews Cancer.

[124]  E. Giovannetti,et al.  Discovery of N-hydroxyindole-based inhibitors of human lactate dehydrogenase isoform A (LDH-A) as starvation agents against cancer cells. , 2011, Journal of medicinal chemistry.

[125]  W. Denny Hypoxia-activated prodrugs in cancer therapy: progress to the clinic. , 2010, Future oncology.

[126]  Amanda S Barnard,et al.  One-to-one comparison of sunscreen efficacy, aesthetics and potential nanotoxicity. , 2010, Nature nanotechnology.

[127]  G. Semenza HIF-1: upstream and downstream of cancer metabolism. , 2010, Current opinion in genetics & development.

[128]  G. Semenza,et al.  Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression , 2010, Proceedings of the National Academy of Sciences.

[129]  P. Carmeliet,et al.  Mechanisms of resistance to anti-angiogenic therapy and development of third-generation anti-angiogenic drug candidates. , 2010, Genes & cancer.

[130]  A. Barnard How can ab initio simulations address risks in nanotech? , 2009, Nature nanotechnology.

[131]  L. Cantley,et al.  Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation , 2009, Science.

[132]  Anthony Mancuso,et al.  Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction , 2008, Proceedings of the National Academy of Sciences.

[133]  M.-H. Lee,et al.  Roles of p53, Myc and HIF-1 in Regulating Glycolysis — the Seventh Hallmark of Cancer , 2008, Cellular and Molecular Life Sciences.

[134]  R. Deberardinis,et al.  Beyond aerobic glycolysis: Transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis , 2007, Proceedings of the National Academy of Sciences.

[135]  J. Karp,et al.  Nanocarriers as an Emerging Platform for Cancer Therapy , 2022 .

[136]  C. Thompson,et al.  HIF and c-Myc: sibling rivals for control of cancer cell metabolism and proliferation. , 2007, Cancer cell.

[137]  R. I. Viji,et al.  Endothelial cell response to lactate: Implication of PAR modification of VEGF , 2007, Journal of cellular physiology.

[138]  R. Cardone,et al.  The role of disturbed pH dynamics and the Na+/H+ exchanger in metastasis , 2005, Nature Reviews Cancer.

[139]  A. Barclay,et al.  CD147 is tightly associated with lactate transporters MCT1 and MCT4 and facilitates their cell surface expression , 2000, The EMBO journal.

[140]  A. Halestrap,et al.  Inhibition and labelling of the erythrocyte lactate transporter by stilbene disulphonates. , 1990, Biochemical Society transactions.

[141]  R. Denton,et al.  The specificity and metabolic implications of the inhibition of pyruvate transport in isolated mitochondria and intact tissue preparations by alpha-Cyano-4-hydroxycinnamate and related compounds. , 1975, The Biochemical journal.

[142]  O. Warburg,et al.  THE METABOLISM OF TUMORS IN THE BODY , 1927, The Journal of general physiology.