Enzyme-based autophagy in anti-neoplastic management: From molecular mechanisms to clinical therapeutics
Tài liệu tham khảo
Ren, 2018, Targeting autophagy in aging and aging-related cardiovascular diseases, Trends Pharmacol. Sci., 39, 1064, 10.1016/j.tips.2018.10.005
Ren, 2018, Metabolic stress, autophagy, and cardiovascular aging: from pathophysiology to therapeutics, Trends Endocrinol. Metab., 29, 699, 10.1016/j.tem.2018.08.001
Barton, 2018, Pancreatic neoplasms and autophagy, Curr. Drug Targets, 19, 1018, 10.2174/1389450117666160622220915
Tan, 2019, Role of autophagy in inherited metabolic and endocrine myopathies, Biochimica et Biophysica Acta (BBA), 1865, 48, 10.1016/j.bbadis.2018.10.023
Velasco, 2018, The role of autophagy and cancer, Free Radic. Biol. Med., 120, S20, 10.1016/j.freeradbiomed.2018.04.085
Liu, 2015, Autosis and autophagic cell death: the dark side of autophagy, Cell Death Differ., 22, 367, 10.1038/cdd.2014.143
Li, 2020, Autophagy and cancer therapy cardiotoxicity: from molecular mechanisms to therapeutic opportunities, Biochimica et Biophysica Acta (BBA), 1867, 118493, 10.1016/j.bbamcr.2019.06.007
Doherty, 2018, Life, death and autophagy, Nat. Cell Biol., 20, 1110, 10.1038/s41556-018-0201-5
Onorati, 2018, Targeting autophagy in cancer, Cancer, 124, 3307, 10.1002/cncr.31335
Hu, 2017, Blocking autophagy improves the anti-tumor activity of afatinib in lung adenocarcinoma with activating EGFR mutations in vitro and in vivo, Sci. Rep., 7, 4559, 10.1038/s41598-017-04258-8
Fulda, 2017, Autophagy in cancer therapy, Front. Oncol., 7, 128, 10.3389/fonc.2017.00128
Bagca, 2016, Ruxolitinib induces autophagy in chronic myeloid leukemia cells, Tumor Biol., 37, 1573, 10.1007/s13277-015-3947-4
Paquette, 2018, mTOR pathways in cancer and autophagy, Cancers, 10, 18, 10.3390/cancers10010018
Towers, 2020, Autophagy and cancer: modulation of cell death pathways and cancer cell adaptations autophagy and cancer, J. Cell Biol., 219
Ishaq, 2020, Autophagy in Cancer: Recent advances and future directions
Zhou, 2015, A novel autophagy/mitophagy inhibitor liensinine sensitizes breast cancer cells to chemotherapy through DNM1L-mediated mitochondrial fission, Autophagy, 11, 1259, 10.1080/15548627.2015.1056970
Lu, 2019, The nanoparticle-facilitated autophagy inhibition of cancer stem cells for improved chemotherapeutic effects on glioblastomas, J. Mater. Chem. B, 7, 2054, 10.1039/C8TB03165G
Amaravadi, 2019, Targeting autophagy in cancer: recent advances and future directions, Cancer Dis., 9, 1167, 10.1158/2159-8290.CD-19-0292
Das, 2019, A novel triazole, NMK-T-057, induces autophagic cell death in breast cancer cells by inhibiting γ-secretase–mediated activation of Notch signaling, J. Biol. Chem., 294, 6733, 10.1074/jbc.RA119.007671
Mortensen, 2011, Lack of autophagy in the hematopoietic system leads to loss of hematopoietic stem cell function and dysregulated myeloid proliferation, Autophagy, 7, 1069, 10.4161/auto.7.9.15886
Nazio, 2019, Autophagy and cancer stem cells: molecular mechanisms and therapeutic applications, Cell Death Different., 26, 690, 10.1038/s41418-019-0292-y
Takeda, 2019, Disruption of endolysosomal rab5/7 efficiently eliminates colorectal cancer stem cells, Cancer Res., 79, 1426, 10.1158/0008-5472.CAN-18-2192
Huang, 2018, Reciprocal network between cancer stem-like cells and macrophages facilitates the progression and androgen deprivation therapy resistance of prostate cancer, Clin. Cancer Res., 24, 4612, 10.1158/1078-0432.CCR-18-0461
Alissafi, 2018, Autophagy orchestrates the regulatory program of tumor-associated myeloid-derived suppressor cells, J. Clin. Invest., 128, 3840, 10.1172/JCI120888
Ghislat, 2018, Autophagy in dendritic cells, Cell. Mol. Immunol., 15, 944, 10.1038/cmi.2018.2
Noman, 2018, Targeting autophagy blocks melanoma growth by bringing natural killer cells to the tumor battlefield, Autophagy, 14, 730, 10.1080/15548627.2018.1427398
Cheong, 2016, Role of autophagy in cancer metabolism
Ferro, 2019, Autophagy and mitophagy in cancer metabolic remodelling
Amaravadi, 2016, Recent insights into the function of autophagy in cancer, Genes Dev., 30, 1913, 10.1101/gad.287524.116
Yang, 2014, Autophagy is critical for pancreatic tumor growth and progression in tumors with p53 alterations, Cancer Dis., 4, 905, 10.1158/2159-8290.CD-14-0362
Vettore, 2019, New aspects of amino acid metabolism in cancer, Br. J. Cancer, 1
Rabanal-Ruiz, 2017, mTORC1 as the main gateway to autophagy, Essays Biochem., 61, 565, 10.1042/EBC20170027
Shimobayashi, 2016, Multiple amino acid sensing inputs to mTORC1, Cell Res., 26, 7, 10.1038/cr.2015.146
Filomeni, 2015, Oxidative stress and autophagy: the clash between damage and metabolic needs, Cell Death Differ., 22, 377, 10.1038/cdd.2014.150
Kim, 2019, mTOR as a central hub of nutrient signalling and cell growth, Nat. Cell Biol., 21, 63, 10.1038/s41556-018-0205-1
Wolfson, 2016, Sestrin2 is a leucine sensor for the mTORC1 pathway, Science, 351, 43, 10.1126/science.aab2674
Yoon, 2016, The role of amino acid-induced mammalian target of rapamycin complex 1 (mTORC1) signaling in insulin resistance, Exp. Mol. Med., 48, 10.1038/emm.2015.93
Karim, 2017, Effect and proposed mechanism of vitamin C modulating amino acid regulation of autophagic proteolysis, Biochimie, 142, 51, 10.1016/j.biochi.2017.08.004
Rebsamen, 2016, SLC38A9: a lysosomal amino acid transporter at the core of the amino acid-sensing machinery that controls MTORC1, Autophagy, 12, 1061, 10.1080/15548627.2015.1091143
Chantranupong, 2016, The CASTOR proteins are arginine sensors for the mTORC1 pathway, Cell, 165, 153, 10.1016/j.cell.2016.02.035
Carroll, 2016, Control of TSC2-Rheb signaling axis by arginine regulates mTORC1 activity, Elife, 5, 10.7554/eLife.11058
Rabanal-Ruiz, 2018, mTORC1 and nutrient homeostasis: the central role of the lysosome, Int. J. Mol. Sci., 19, 818, 10.3390/ijms19030818
Bröer, 2017, Amino acid homeostasis and signalling in mammalian cells and organisms, Biochem. J., 474, 1935, 10.1042/BCJ20160822
Stransky, 2016
Xia, 2018, GCN2 controls the cellular checkpoint: potential target for regulating inflammation, Cell Death Dis., 4, 20, 10.1038/s41420-017-0022-5
Ghislat, 2015, 69
Jin, 2016, Activation of autophagy through calcium-dependent AMPK/mTOR and PKC θ pathway causes activation of rat hepatic stellate cells under hypoxic stress, FEBS Lett., 590, 672, 10.1002/1873-3468.12090
Wayman, 2008, Calmodulin-kinases: modulators of neuronal development and plasticity, Neuron, 59, 914, 10.1016/j.neuron.2008.08.021
Simon, 2017, Retrograde signaling from autophagy modulates stress responses, Sci. Signal., 10, eaag2791, 10.1126/scisignal.aag2791
Villanueva-Paz, 2016, AMPK regulation of cell growth, apoptosis, autophagy, and bioenergetics, 45
Jang, 2017, AMPK–ULK1-mediated autophagy confers resistance to BET inhibitor JQ1 in acute myeloid leukemia stem cells, Clin. Cancer Res., 23, 2781, 10.1158/1078-0432.CCR-16-1903
Mrakovcic, 2018, p53-mediated molecular control of autophagy in tumor cells, Biomolecules, 8, 14, 10.3390/biom8020014
Batool, 2016, A comprehensive review on L-asparaginase and its applications, Appl. Biochem. Biotechnol., 178, 900, 10.1007/s12010-015-1917-3
Takahashi, 2017, Autophagy is required for cell survival under L-asparaginase-induced metabolic stress in acute lymphoblastic leukemia cells, Oncogene, 36, 4267, 10.1038/onc.2017.59
Takahashi, 2015, Autophagy inhibition sensitizes acute lymphoblastic leukemia cells to L-asparaginase, Am. Soc. Hematol.
Chen, 2017, Autophagy suppression potentiates the anti-glioblastoma effect of asparaginase in vitro and in vivo, Oncotarget, 8, 91052, 10.18632/oncotarget.19409
Ji, 2017, Deprivation of asparagine triggers cytoprotective autophagy in laryngeal squamous cell carcinoma, Appl. Microbiol. Biotechnol., 101, 4951, 10.1007/s00253-017-8221-9
Zhang, 2016, Targeting asparagine and autophagy for pulmonary adenocarcinoma therapy, Appl. Microbiol. Biotechnol., 100, 9145, 10.1007/s00253-016-7640-3
Song, 2015, Asparaginase induces apoptosis and cytoprotective autophagy in chronic myeloid leukemia cells, Oncotarget, 6, 3861, 10.18632/oncotarget.2869
Yu, 2012, L-asparaginase inhibits invasive and angiogenic activity and induces autophagy in ovarian cancer, J. Cell. Mol. Med., 16, 2369, 10.1111/j.1582-4934.2012.01547.x
Xu, 2018, Recombinant human arginase induces apoptosis through oxidative stress and cell cycle arrest in small cell lung cancer, Cancer Sci., 109, 3471, 10.1111/cas.13782
Shen, 2017, A novel and promising therapeutic approach for NSCLC: recombinant human arginase alone or combined with autophagy inhibitor, Cell Death Dis., 8, 10.1038/cddis.2017.137
Lin, 2015, The role of autophagy in the cytotoxicity induced by recombinant human arginase in laryngeal squamous cell carcinoma, Appl. Microbiol. Biotechnol., 99, 8487, 10.1007/s00253-015-6565-6
Khoury, 2015, Human recombinant arginase I (Co)-PEG5000 [HuArgI (Co)-PEG5000]-induced arginine depletion is selectively cytotoxic to human glioblastoma cells, J. Neuro-Oncol., 122, 75, 10.1007/s11060-014-1698-5
Abi-Habib, 2018
Maletzki, 2017, Deciphering molecular mechanisms of arginine deiminase-based therapy–comparative response analysis in paired human primary and recurrent glioblastomas, Chem. Biol. Interact., 278, 179, 10.1016/j.cbi.2017.10.007
Przystal, 2018, Efficacy of arginine depletion by ADI-PEG20 in an intracranial model of GBM, Cell Death Dis., 9, 1192, 10.1038/s41419-018-1195-4
Sahu, 2017, Argininosuccinate Synthetase 1 loss in invasive bladder Cancer regulates survival through general control Nonderepressible 2 kinase–mediated eukaryotic initiation factor 2α activity and is targetable by Pegylated arginine deiminase, Am. J. Pathol., 187, 200, 10.1016/j.ajpath.2016.09.004
Bean, 2016, A metabolic synthetic lethal strategy with arginine deprivation and chloroquine leads to cell death in ASS1-deficient sarcomas, Cell Death Dis., 7, 10.1038/cddis.2016.232
Kim, 2009, ADI, autophagy and apoptosis: metabolic stress as a therapeutic option for prostate cancer, Autophagy, 5, 567, 10.4161/auto.5.4.8252
Castro, 2016, Approaches to endow ribonucleases with antitumor activity: lessons learned from the native cytotoxic ribonucleases, Anti-Cancer Drugs, 135
Fiorini, 2014, Bovine seminal ribonuclease triggers Beclin1-mediated autophagic cell death in pancreatic cancer cells, Biochimica et Biophysica Acta (BBA), 1843, 976, 10.1016/j.bbamcr.2014.01.025
Siddiqui, 2015, RNase L cleavage products promote switch from autophagy to apoptosis by caspase-mediated cleavage of beclin-1, Int. J. Mol. Sci., 16, 17611, 10.3390/ijms160817611
Fiorini, 2015, Onconase induces autophagy sensitizing pancreatic cancer cells to gemcitabine and activates Akt/mTOR pathway in a ROS-dependent manner, Biochimica et Biophysica Acta (BBA), 1853, 549, 10.1016/j.bbamcr.2014.12.016
Das, 2018, Pro-survival autophagy and cancer cell resistance to therapy, Cancer Metastasis Rev., 37, 749, 10.1007/s10555-018-9727-z
Kang, 2020, Dietary restriction of amino acids for Cancer therapy, Nutr. Metabol., 17, 1, 10.1186/s12986-020-00439-x
Bhutia, 2015, Amino acid transporters in cancer and their relevance to “glutamine addiction”: novel targets for the design of a new class of anticancer drugs, Cancer Res., 75, 1782, 10.1158/0008-5472.CAN-14-3745
Fung, 2017, Drug-induced amino acid deprivation as strategy for cancer therapy, J. Hematol. Oncol., 10, 144, 10.1186/s13045-017-0509-9
Fung, 2017, Drug-induced amino acid deprivation as strategy for cancer therapy, J. Hematol. Oncol., 10, 144, 10.1186/s13045-017-0509-9
Kremer, 2017, Arginine deprivation inhibits the Warburg effect and upregulates glutamine Anaplerosis and serine biosynthesis in ASS1-deficient cancers, Cell Rep., 18, 991, 10.1016/j.celrep.2016.12.077
Boone, 2015, Safety and biologic response of pre-operative autophagy inhibition in combination with gemcitabine in patients with pancreatic adenocarcinoma, Ann. Surg. Oncol., 22, 4402, 10.1245/s10434-015-4566-4
Martin, 2018, A potent and selective ULK1 inhibitor suppresses autophagy and sensitizes cancer cells to nutrient stress, Iscience, 8, 74, 10.1016/j.isci.2018.09.012
Dyczynski, 2018, Targeting autophagy by small molecule inhibitors of vacuolar protein sorting 34 (Vps34) improves the sensitivity of breast cancer cells to Sunitinib, Cancer Lett., 435, 32, 10.1016/j.canlet.2018.07.028
Liu, 2011, Beclin1 controls the levels of p53 by regulating the deubiquitination activity of USP10 and USP13, Cell, 147, 223, 10.1016/j.cell.2011.08.037
Sutton, 2019, Amino acid deprivation-induced autophagy requires upregulation of DIRAS3 through reduction of E2F1 and E2F4 transcriptional repression, Cancers, 11, 603, 10.3390/cancers11050603
Zhang, 2018, Increased amino acid uptake supports autophagy-deficient cell survival upon glutamine deprivation, Cell Rep., 23, 3006, 10.1016/j.celrep.2018.05.006
Still, 2017, Hopefully devoted to Q: targeting glutamine addiction in cancer, Br. J. Cancer, 116, 1375, 10.1038/bjc.2017.113
Zhang, 2017, Cancer cell metabolism: the essential role of the nonessential amino acid, glutamine, EMBO J., 36, 1302, 10.15252/embj.201696151
Choi, 2018, Targeting glutamine metabolism for cancer treatment, Biomol. Ther., 26, 19, 10.4062/biomolther.2017.178
Elgogary, 2016, Combination therapy with BPTES nanoparticles and metformin targets the metabolic heterogeneity of pancreatic cancer, Proc. Natl. Acad. Sci., 113, E5328, 10.1073/pnas.1611406113
Altman, 2016, From Krebs to clinic: glutamine metabolism to cancer therapy, Nat. Rev. Cancer, 16, 619, 10.1038/nrc.2016.71
Nair, 2012, Autophagy and cardiovascular aging: lesson learned from rapamycin, Cell Cycle, 11, 2092, 10.4161/cc.20317
Asgari, 2011, An improved convergent approach for synthesis of erlotinib, a tyrosine kinase inhibitor, via a ring closure reaction of phenyl benzamidine intermediate, Bull. Kor. Chem. Soc., 32, 909, 10.5012/bkcs.2011.32.3.909
Barghi, 2012, Modified synthesis of erlotinib hydrochloride, Adv. Pharma. Bull., 2, 119
Janmaat, 2003
Khamisipour, 2016, Mechanisms of tumor cell resistance to the current targeted-therapy agents, Tumor Biol., 37, 10021, 10.1007/s13277-016-5059-1
Liu, 2017, Autophagy inhibitor facilitates gefitinib sensitivity in vitro and in vivo by activating mitochondrial apoptosis in triple negative breast cancer, PLoS One, 12, 10.1371/journal.pone.0177694
Bartholomeusz, 2012, High ERK protein expression levels correlate with shorter survival in triple-negative breast cancer patients, Oncologist, 17, 766, 10.1634/theoncologist.2011-0377
Chang, 2014, Autophagy contributes to gefitinib-induced glioma cell growth inhibition, Exp. Cell Res., 327, 102, 10.1016/j.yexcr.2014.05.011
Zhao, 2016, Gefitinib induces lung cancer cell autophagy and apoptosis via blockade of the PI3K/AKT/mTOR pathway, Oncol. Lett., 12, 63, 10.3892/ol.2016.4606
Eimer, 2011, Autophagy inhibition cooperates with erlotinib to induce glioblastoma cell death, Cancer Biol. Ther., 11, 1017, 10.4161/cbt.11.12.15693
Maddocks, 2013, Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells, Nature, 493, 542, 10.1038/nature11743
Akbarzadeh, 2019, Multifunctional nanomedicines for targeting epidermal growth factor receptor in colorectal cancer
Basha, 2018, Nanotechnology as a promising strategy for anticancer drug delivery, Curr. Drug Deliv., 15, 497, 10.2174/1567201814666170516114411
Alizadeh, 2019, Chitosan-based nanotherapeutics for ovarian cancer treatment, J. Drug Target., 27, 839, 10.1080/1061186X.2018.1564923
Patil, 2014, Novel methods for liposome preparation, Chem. Phys. Lipids, 177, 8, 10.1016/j.chemphyslip.2013.10.011
Zalba, 2015, Cetuximab-oxaliplatin-liposomes for epidermal growth factor receptor targeted chemotherapy of colorectal cancer, J. Control. Release, 210, 26, 10.1016/j.jconrel.2015.05.271
Mato, 2015, Selective antitumoral effect of sorafenib loaded PLGA nanoparticles conjugated with cetuximab on undifferentiated/anaplastic thyroid carcinoma cells, J. Nanomed. Nanotechnol., 6, 2
Donahue, 2019, Concepts of nanoparticle cellular uptake, intracellular trafficking, and kinetics in nanomedicine, Adv. Drug Deliv. Rev., 143, 68, 10.1016/j.addr.2019.04.008
Yin, 2020, Nanoformulations of small molecule protein tyrosine kinases inhibitors potentiate targeted cancer therapy, Int. J. Pharm., 573, 118785, 10.1016/j.ijpharm.2019.118785
Ou, 2018, Regulatory T cell-targeted hybrid nanoparticles combined with immuno-checkpoint blockage for cancer immunotherapy, J. Control. Release, 281, 84, 10.1016/j.jconrel.2018.05.018
Correia, 2015, Cyclodextrin-modified porous silicon nanoparticles for efficient sustained drug delivery and proliferation inhibition of breast cancer cells, ACS Appl. Mater. Interfaces, 7, 23197, 10.1021/acsami.5b07033
Do, 2019
Sharma, 2019, Improved biological activity and stability of enzyme L-Asparaginase in solid lipid nanoparticles formulation, J. Drug Deliv. Ther., 9, 325
Dhanya, 2018
Baskar, 2018, Synthesis, characterization and synergistic activity of cerium-selenium nanobiocomposite of fungal l-asparaginase against lung cancer, Mater. Sci. Eng. C, 93, 809, 10.1016/j.msec.2018.08.051
Position, 2016, Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for practice guidelines, Eur. Heart J., 37, 2768
Resteghini, 2017, Management of tyrosine kinase inhibitors (TKI) side effects in differentiated and medullary thyroid cancer patients, Best Pract. Res. Clin. Endocrinol. Metab., 31, 349, 10.1016/j.beem.2017.04.012
Costa, 2015
Tarrado-Castellarnau, 2016, Oncogenic regulation of tumor metabolic reprogramming, Oncotarget, 7, 62726, 10.18632/oncotarget.10911
Wang, 2016, Pharmacological inhibitors of autophagy as novel cancer therapeutic agents, Pharmacol. Res., 105, 164, 10.1016/j.phrs.2016.01.028
Cheong, 2012, Therapeutic targets in cancer cell metabolism and autophagy, Nat. Biotechnol., 30, 671, 10.1038/nbt.2285
Nakaya, 2014, Inflammatory T cell responses rely on amino acid transporter ASCT2 facilitation of glutamine uptake and mTORC1 kinase activation, Immunity, 40, 692, 10.1016/j.immuni.2014.04.007
Ellappan, 2014
Poncet, 2014, The catalytic subunit of the system L1 amino acid transporter (slc7a5) facilitates nutrient signalling in mouse skeletal muscle, PLoS One, 9, 10.1371/journal.pone.0089547
Lumeng, 2017, Metabotropic glutamate receptors in cancer, Neuropharmacology, 115, 193, 10.1016/j.neuropharm.2016.02.011
Lukey, 2017, Targeting amino acid metabolism for cancer therapy, Drug Discov. Today, 22, 796, 10.1016/j.drudis.2016.12.003
Linares, 2015, Amino acid activation of mTORC1 by a PB1-domain-driven kinase complex cascade, Cell Rep., 12, 1339, 10.1016/j.celrep.2015.07.045
Meijer, 2009, Autophagy: regulation and role in disease, Crit. Rev. Clin. Lab. Sci., 46, 210, 10.1080/10408360903044068
Xia, 2018
Pierotti, 2013, Targeting metabolism for cancer treatment and prevention: metformin, an old drug with multi-faceted effects, Oncogene, 32, 1475, 10.1038/onc.2012.181
Yao, 2016, 5’-Monophosphate-activated protein kinase (AMPK) improves autophagic activity in diabetes and diabetic complications, Acta Pharm. Sin. B, 6, 20, 10.1016/j.apsb.2015.07.009
Zhang, 2017, Hormetic effect of panaxatriol saponins confers neuroprotection in PC12 cells and zebrafish through PI3K/AKT/mTOR and AMPK/SIRT1/FOXO3 pathways, Sci. Rep., 7, 41082, 10.1038/srep41082
Zhou, 2015, MAPK/JNK signalling: a potential autophagy regulation pathway, Biosci. Rep., 35, 10.1042/BSR20140141