ライブラリ登録: Guest
Critical Reviews™ in Eukaryotic Gene Expression

年間 6 号発行

ISSN 印刷: 1045-4403

ISSN オンライン: 2162-6502

The Impact Factor measures the average number of citations received in a particular year by papers published in the journal during the two preceding years. 2017 Journal Citation Reports (Clarivate Analytics, 2018) IF: 1.6 To calculate the five year Impact Factor, citations are counted in 2017 to the previous five years and divided by the source items published in the previous five years. 2017 Journal Citation Reports (Clarivate Analytics, 2018) 5-Year IF: 2.2 The Immediacy Index is the average number of times an article is cited in the year it is published. The journal Immediacy Index indicates how quickly articles in a journal are cited. Immediacy Index: 0.3 The Eigenfactor score, developed by Jevin West and Carl Bergstrom at the University of Washington, is a rating of the total importance of a scientific journal. Journals are rated according to the number of incoming citations, with citations from highly ranked journals weighted to make a larger contribution to the eigenfactor than those from poorly ranked journals. Eigenfactor: 0.00058 The Journal Citation Indicator (JCI) is a single measurement of the field-normalized citation impact of journals in the Web of Science Core Collection across disciplines. The key words here are that the metric is normalized and cross-disciplinary. JCI: 0.33 SJR: 0.345 SNIP: 0.46 CiteScore™:: 2.5 H-Index: 67

Indexed in

Autophagy in tumor suppression and cancer therapy

巻 21, 発行 1, 2011, pp. 71-100
DOI: 10.1615/CritRevEukarGeneExpr.v21.i1.50
Get accessGet access

要約

Autophagy is a stress-induced cell survival program whereby cells under metabolic, proteotoxic, or other stress remove dysfunctional organelles and/or misfolded/polyubiquitylated proteins by shuttling them via specialized structures called autophagosomes to the lysosome for degradation. The end result is the release of free amino acids and metabolites for use in cell survival. For tumor cells, autophagy is a double-edged sword: autophagy genes are frequently mono-allelically deleted, silenced, or mutated in human tumors, resulting in an environment of increased oxidative stress that is conducive to DNA damage, genomic instability, and tumor progression. As such, autophagy is tumor suppressive. In contrast, it is important to note that although tumor cells have reduced levels of autophagy, they do not eliminate this pathway completely. Furthermore, the exposure of tumor cells to an environment of increased metabolic and other stresses renders them reliant on basal autophagy for survival. Therefore, autophagy inhibition is an active avenue for the identification of novel anti-cancer therapies. Not surprisingly, the field of autophagy and cancer has experienced an explosion of research in the past 10 years. This review covers the basic mechanisms of autophagy, discusses its role in tumor suppression and cancer therapy, and posits emerging questions for the future.

によって引用された
  1. Yeganeh Behzad, Mukherjee Subhendu, Moir Lyn M., Kumawat Kuldeep, Kashani Hessam H., Bagchi Rushita A., Baarsma Hoeke A., Gosens Reinoud, Ghavami Saeid, Novel non-canonical TGF-β signaling networks: Emerging roles in airway smooth muscle phenotype and function, Pulmonary Pharmacology & Therapeutics, 26, 1, 2013. Crossref

  2. Giuliani Charlett M, Dass Crispin R, Autophagy and cancer: taking the ‘toxic’ out of cytotoxics, Journal of Pharmacy and Pharmacology, 65, 6, 2013. Crossref

  3. Pandey Saumya, Chandravati Chandravati, Autophagy in Cervical Cancer: An Emerging Therapeutic Target, Asian Pacific Journal of Cancer Prevention, 13, 10, 2012. Crossref

  4. Bareford M. Danielle, Hamed Hossein A., Allegood Jeremy, Cruickshanks Nichola, Poklepovic Andrew, Park Margaret A., Ogretmen Besim, Spiegel Sarah, Grant Steven, Dent Paul, Sorafenib and pemetrexed toxicity in cancer cells is mediated via SRC-ERK signaling, Cancer Biology & Therapy, 13, 9, 2012. Crossref

  5. Shi Zheng, Li Chun-yang, Zhao Si, Yu Yang, An Na, Liu Yong-xi, Wu Chuan-fang, Yue Bi-song, Bao Jin-ku, A systems biology analysis of autophagy in cancer therapy, Cancer Letters, 337, 2, 2013. Crossref

  6. East Daniel A, Campanella Michelangelo, Ca2+in quality control, Autophagy, 9, 11, 2013. Crossref

  7. Ghosh Samrat, Bishayee Kausik, Khuda-Bukhsh Anisur Rahman, Graveoline Isolated from Ethanolic Extract of Ruta graveolens Triggers Apoptosis and Autophagy in Skin Melanoma Cells: A Novel Apoptosis-Independent Autophagic Signaling Pathway, Phytotherapy Research, 28, 8, 2014. Crossref

  8. Prevarskaya Natalia, Skryma Roman, Shuba Yaroslav, Targeting Ca2+transport in cancer: close reality or long perspective?, Expert Opinion on Therapeutic Targets, 17, 3, 2013. Crossref

  9. Ma Ben, Liang Li-zhong, Liao Gui-qing, Liang Yu-jie, Liu Hai-chao, Zheng Guang-sen, Su Yu-xiong, Inhibition of autophagy enhances cisplatin cytotoxicity in human adenoid cystic carcinoma cells of salivary glands, Journal of Oral Pathology & Medicine, 42, 10, 2013. Crossref

  10. Song Hong-tao, Qin Yu, Yao Guo-dong, Tian Zhen-nan, Fu Song-bin, Geng Jing-shu, Astrocyte Elevated Gene-1 Mediates Glycolysis and Tumorigenesis in Colorectal Carcinoma CellsviaAMPK Signaling, Mediators of Inflammation, 2014, 2014. Crossref

  11. Akin Debra, Wang S Keisin, Habibzadegah-Tari Pouran, Law Brian, Ostrov David, Li Min, Yin Xiao-Ming, Kim Jae-Sung, Horenstein Nicole, Dunn William A, A novel ATG4B antagonist inhibits autophagy and has a negative impact on osteosarcoma tumors, Autophagy, 10, 11, 2014. Crossref

  12. Soares Ana S., Costa Vera M., Diniz Carmen, Fresco Paula, Combination of Cl-IB-MECA with paclitaxel is a highly effective cytotoxic therapy causing mTOR-dependent autophagy and mitotic catastrophe on human melanoma cells, Journal of Cancer Research and Clinical Oncology, 140, 6, 2014. Crossref

  13. Liu Wei Jing, Luo Mian-Na, Tan Jin, Chen Wei, Huang Lei-zhao, Yang Chen, Pan Qingjun, Li Benyi, Liu Hua-feng, Autophagy activation reduces renal tubular injury induced by urinary proteins, Autophagy, 10, 2, 2014. Crossref

  14. Winardi Daniel, Tsai Hung-Pei, Chai Chee-Yin, Chung Chia-Li, Loh Joon-Khim, Chen Yung-Hsiang, Hsieh Ching-Liang, Correlation of Altered Expression of the Autophagy Marker LC3B with Poor Prognosis in Astrocytoma, BioMed Research International, 2014, 2014. Crossref

  15. Wang Ning, Zhao Juyang, Liu Qian, Diao Xinping, Kong Baohua, Sulforaphane protects human umbilical vein cells against lipotoxicity by stimulating autophagy via an AMPK-mediated pathway, Journal of Functional Foods, 15, 2015. Crossref

  16. Park Eunjoo, Park Mi Hee, Na Hee Sam, Chung Jin, Xylitol induces cell death in lung cancer A549 cells by autophagy, Biotechnology Letters, 37, 5, 2015. Crossref

  17. Cruickshanks Nichola, Roberts Jane L, Bareford M Danielle, Tavallai Mehrad, Poklepovic Andrew, Booth Laurence, Spiegel Sarah, Dent Paul, Differential regulation of autophagy and cell viability by ceramide species, Cancer Biology & Therapy, 16, 5, 2015. Crossref

  18. Ghosh Samrat, Sikdar Sourav, Mukherjee Avinaba, Khuda-Bukhsh Anisur Rahman, Evaluation of chemopreventive potentials of ethanolic extract of Ruta graveolens against A375 skin melanoma cells in vitro and induced skin cancer in mice in vivo, Journal of Integrative Medicine, 13, 1, 2015. Crossref

  19. Watson AS, Riffelmacher T, Stranks A, Williams O, De Boer J, Cain K, MacFarlane M, McGouran J, Kessler B, Khandwala S, Chowdhury O, Puleston D, Phadwal K, Mortensen M, Ferguson D, Soilleux E, Woll P, Jacobsen SEW, Simon AK, Autophagy limits proliferation and glycolytic metabolism in acute myeloid leukemia, Cell Death Discovery, 1, 1, 2015. Crossref

  20. Chen Chia-Ling, Chiang Tzu-Hui, Tseng Po-Chun, Wang Yu-Chih, Lin Chiou-Feng, Loss of PTEN causes SHP2 activation, making lung cancer cells unresponsive to IFN-γ, Biochemical and Biophysical Research Communications, 466, 3, 2015. Crossref

  21. Lee Wen-Ying, Hsu Keng-Fu, Chiang Tai-An, Chen Chee-Jen, Phellinus LinteusExtract Induces Autophagy and Synergizes With 5-Fluorouracil to Inhibit Breast Cancer Cell Growth, Nutrition and Cancer, 67, 2, 2015. Crossref

  22. Czarny Piotr, Pawlowska Elzbieta, Bialkowska-Warzecha Jolanta, Kaarniranta Kai, Blasiak Janusz, Autophagy in DNA Damage Response, International Journal of Molecular Sciences, 16, 2, 2015. Crossref

  23. Chen Hsin-I, Tsai Hung-Pei, Chen Yi-Ting, Tsao Shu-Chuan, Chai Chee-Yin, Autophagy and Apoptosis Play Opposing Roles in Overall Survival of Esophageal Squamous Cell Carcinoma, Pathology & Oncology Research, 22, 4, 2016. Crossref

  24. Kung Che-Pei, Murphy Maureen E, The role of the p53 tumor suppressor in metabolism and diabetes, Journal of Endocrinology, 231, 2, 2016. Crossref

  25. CHUANG WAN-LING, SU CHIN-CHENG, LIN PING-YI, LIN CHI-CHEN, CHEN YAO-LI, Sann-Joong-Kuey-Jian-Tang induces autophagy in HepG2 cells via regulation of the phosphoinositide-3 kinase/Akt/mammalian target of rapamycin and p38 mitogen-activated protein kinase pathways, Molecular Medicine Reports, 12, 2, 2015. Crossref

  26. Mooneyham Ashley, Bazzaro Martina, Targeting Deubiquitinating Enzymes and Autophagy in Cancer, in Cancer Gene Networks, 1513, 2017. Crossref

  27. Law Betty, Mok Simon, Wu An, Lam Christopher, Yu Margaret, Wong Vincent, New Potential Pharmacological Functions of Chinese Herbal Medicines via Regulation of Autophagy, Molecules, 21, 3, 2016. Crossref

  28. Rahman Faiz-Ur, Ali Amjad, Khan Inam Ullah, Duong Hong-Quan, Yu Shang-Bo, Lin Yue-Jian, Wang Hui, Li Zhan-Ting, Zhang Dan-Wei, Morpholine or methylpiperazine and salicylaldimine based heteroleptic square planner platinum (II) complexes: In vitro anticancer study and growth retardation effect on E. coli, European Journal of Medicinal Chemistry, 131, 2017. Crossref

  29. Lin Chia-Hung, Chang Ching-Yao, Lee Kuan-Rong, Lin Hui-Ju, Lin Wu-Chou, Chen Ter-Hsin, Wan Lei, Cold-water extracts of Grifola frondosa and its purified active fraction inhibit hepatocellular carcinoma in vitro and in vivo, Experimental Biology and Medicine, 241, 13, 2016. Crossref

  30. Wechman Stephen L., Pradhan Anjan K., DeSalle Rob, Das Swadesh K., Emdad Luni, Sarkar Devanand, Fisher Paul B., , 137, 2018. Crossref

  31. Masuelli L., Granato M., Benvenuto M., Mattera R., Bernardini R., Mattei M., d'Amati G., D'Orazi G., Faggioni A., Bei R., Cirone M., Chloroquine supplementation increases the cytotoxic effect of curcumin against Her2/neu overexpressing breast cancer cells in vitro and in vivo in nude mice while counteracts it in immune competent mice, OncoImmunology, 6, 11, 2017. Crossref

  32. So Keum-Young, Lee Byung-Hoon, Oh Seon-Hee, The critical role of autophagy in cadmium-induced immunosuppression regulated by endoplasmic reticulum stress-mediated calpain activation in RAW264.7 mouse monocytes, Toxicology, 393, 2018. Crossref

  33. ZHANG SHI-YI, LI XUE-BO, HOU SHENG-GUANG, SUN YAO, SHI YI-RAN, LIN SONG-SEN, Cedrol induces autophagy and apoptotic cell death in A549 non-small cell lung carcinoma cells through the P13K/Akt signaling pathway, the loss of mitochondrial transmembrane potential and the generation of ROS, International Journal of Molecular Medicine, 38, 1, 2016. Crossref

  34. Mancinelli Romina, Carpino Guido, Petrungaro Simonetta, Mammola Caterina Loredana, Tomaipitinca Luana, Filippini Antonio, Facchiano Antonio, Ziparo Elio, Giampietri Claudia, Multifaceted Roles of GSK-3 in Cancer and Autophagy-Related Diseases, Oxidative Medicine and Cellular Longevity, 2017, 2017. Crossref

  35. Lund Kaja, Olsen Cathrine Elisabeth, Wong Judith Jing Wen, Olsen Petter Angell, Solberg Nina Therese, Høgset Anders, Krauss Stefan, Selbo Pål Kristian, 5-FU resistant EMT-like pancreatic cancer cells are hypersensitive to photochemical internalization of the novel endoglin-targeting immunotoxin CD105-saporin, Journal of Experimental & Clinical Cancer Research, 36, 1, 2017. Crossref

  36. Huo Ruichao, Wang Lili, Liu Peijuan, Zhao Yong, Zhang Caiqin, Bai Bing, Liu Xueying, Shi Changhong, Wei Sanhua, Zhang Hai, Cabazitaxel-induced autophagy via the PI3K/Akt/mTOR pathway contributes to A549 cell death, Molecular Medicine Reports, 14, 4, 2016. Crossref

  37. Shi Yan, Tang Bin, Yu Pei-Wu, Tang Bo, Hao Ying-Xue, Lei Xiao, Luo Hua-Xing, Zeng Dong-Zhu, Multhoff Gabriele, Autophagy Protects against Oxaliplatin-Induced Cell Death via ER Stress and ROS in Caco-2 Cells, PLoS ONE, 7, 11, 2012. Crossref

  38. Pereira Olga, Teixeira Alexandra, Sampaio‐Marques Belém, Castro Isabel, Girão Henrique, Ludovico Paula, Signalling mechanisms that regulate metabolic profile and autophagy of acute myeloid leukaemia cells, Journal of Cellular and Molecular Medicine, 22, 10, 2018. Crossref

  39. García‐Barros Mönica, Coant Nicolas, Kawamori Toshihiko, Wada Masayuki, Snider Ashley J., Truman Jean‐Philip, Wu Bill X., Furuya Hideki, Clarke Christopher J., Bialkowska Agnieszka B., Ghaleb Amr, Yang Vincent W., Obeid Lina M., Hannun Yusuf A., Role of neutral ceramidase in colon cancer, The FASEB Journal, 30, 12, 2016. Crossref

  40. Li Meng, Lee Ara, Kim Kyung Lock, Murray James, Shrinidhi Annadka, Sung Gihyun, Park Kyeng Min, Kim Kimoon, Autophagy Caught in the Act: A Supramolecular FRET Pair Based on an Ultrastable Synthetic Host-Guest Complex Visualizes Autophagosome-Lysosome Fusion, Angewandte Chemie, 130, 8, 2018. Crossref

  41. Desjarlais Michel, Annabi Borhane, Dual functions of ARP101 in targeting membrane type-1 matrix metalloproteinase: Impact on U87 glioblastoma cell invasion and autophagy signaling, Chemical Biology & Drug Design, 93, 3, 2019. Crossref

  42. Li Meng, Lee Ara, Kim Kyung Lock, Murray James, Shrinidhi Annadka, Sung Gihyun, Park Kyeng Min, Kim Kimoon, Autophagy Caught in the Act: A Supramolecular FRET Pair Based on an Ultrastable Synthetic Host-Guest Complex Visualizes Autophagosome-Lysosome Fusion, Angewandte Chemie International Edition, 57, 8, 2018. Crossref

  43. Yushan Ren, Ying Yan, Yujun Tan, Jingchun Yao, Dongguang Liu, Lihong Pan, Pingping Wang, Lili Zhao, Fanhui Zeng, Zhong Liu, Guimin Zhang, Jie Li, Isoliquiritigenin inhibits mouse S180 tumors with a new mechanism that regulates autophagy by GSK-3β/TNF-α pathway, European Journal of Pharmacology, 838, 2018. Crossref

  44. Yun Chul, Lee Sang, The Roles of Autophagy in Cancer, International Journal of Molecular Sciences, 19, 11, 2018. Crossref

  45. Goker Bagca Bakiye, Biray Avci Cigir, Cancer Stem Cells and Autophagy: Present Knowledge and Future Perspectives, in Autophagy in Health and Disease, 2018. Crossref

  46. Bala Tannan Neeta, Collu Giovanna, Humphries Ashley C., Serysheva Ekatherina, Weber Ursula, Mlodzik Marek, Perrimon Norbert, AKAP200 promotes Notch stability by protecting it from Cbl/lysosome-mediated degradation in Drosophila melanogaster, PLOS Genetics, 14, 1, 2018. Crossref

  47. Deng Shuo, Shanmugam Muthu K., Kumar Alan Prem, Yap Celestial T., Sethi Gautam, Bishayee Anupam, Targeting autophagy using natural compounds for cancer prevention and therapy, Cancer, 125, 8, 2019. Crossref

  48. Wang Ping, Du Yangyang, Wang Jingfen, Indentification of breast cancer subtypes sensitive to HCQ-induced autophagy inhibition, Pathology - Research and Practice, 215, 10, 2019. Crossref

  49. Xia Chenglai, He Zhihong, Liang Shaofen, Chen Ruihong, Xu Weikang, Yang Jie, Xiao Guohong, Jiang Shibo, Metformin combined with nelfinavir induces SIRT3/mROS-dependent autophagy in human cervical cancer cells and xenograft in nude mice, European Journal of Pharmacology, 848, 2019. Crossref

  50. Balaburski Gregor M., Budina Anna, Murphy Maureen E., Oncogenes and Tumor Suppressor Genes in Autophagy, in Autophagy and Cancer, 2013. Crossref

  51. Islam Khan Md Zahirul, Tam Shing Yau, Law Helen Ka Wai, Autophagy-Modulating Long Non-coding RNAs (LncRNAs) and Their Molecular Events in Cancer, Frontiers in Genetics, 9, 2019. Crossref

  52. Reuken Philipp A., Lutz Philipp, Casper Markus, Al‐Herwi Eihab, Stengel Sven, Spengler Ulrich, Stallmach Andreas, Lammert Frank, Nischalke Hans Dieter, Bruns Tony, The ATG16L1 gene variant rs2241880 (p.T300A) is associated with susceptibility to HCC in patients with cirrhosis , Liver International, 39, 12, 2019. Crossref

  53. Jin Juan, Tu Qiudi, Gong Jianguang, Zhao Li, Liang Shikai, He Qiang, Autophagy activity and expression pattern of autophagy-related markers in the podocytes of patients with lupus nephritis: association with pathological classification, Renal Failure, 41, 1, 2019. Crossref

  54. Liu Zhen, Jiang Xiaobing, Gao Liang, Liu Xuan, Li Jiali, Huang Xing, Zeng Tao, Synergistic Suppression of Glioblastoma Cell Growth by Combined Application of Temozolomide and Dopamine D2 Receptor Antagonists, World Neurosurgery, 128, 2019. Crossref

  55. Holczer Marianna, Hajdú Bence, Lőrincz Tamás, Szarka András, Bánhegyi Gábor, Kapuy Orsolya, A Double Negative Feedback Loop between mTORC1 and AMPK Kinases Guarantees Precise Autophagy Induction upon Cellular Stress, International Journal of Molecular Sciences, 20, 22, 2019. Crossref

  56. Castro Isabel, Sampaio-Marques Belém, Ludovico Paula, Targeting Metabolic Reprogramming in Acute Myeloid Leukemia, Cells, 8, 9, 2019. Crossref

  57. Bermúdez Mercedes, Aguilar-Medina Maribel, Lizárraga-Verdugo Erik, Avendaño-Félix Mariana, Silva-Benítez Erika, López-Camarillo Cesar, Ramos-Payán Rosalío, LncRNAs as Regulators of Autophagy and Drug Resistance in Colorectal Cancer, Frontiers in Oncology, 9, 2019. Crossref

  58. Aydinlik Seyma, Dere Egemen, Ulukaya Engin, Induction of autophagy enhances apoptotic cell death via epidermal growth factor receptor inhibition by canertinib in cervical cancer cells, Biochimica et Biophysica Acta (BBA) - General Subjects, 1863, 5, 2019. Crossref

  59. Vidoni Chiara, Ferraresi Alessandra, Secomandi Eleonora, Vallino Letizia, Dhanasekaran Danny N., Isidoro Ciro, Epigenetic targeting of autophagy for cancer prevention and treatment by natural compounds, Seminars in Cancer Biology, 66, 2020. Crossref

  60. Tracz-Gaszewska Zuzanna, Dobrzyn Pawel, Stearoyl-CoA Desaturase 1 as a Therapeutic Target for the Treatment of Cancer, Cancers, 11, 7, 2019. Crossref

  61. Wang Chung-Ching, Hueng Dueng-Yuan, Huang Ai-Fang, Chen Wei-Liang, Huang Shih-Ming, Yi-Hsin Chan James, CD164 regulates proliferation, progression, and invasion of human glioblastoma cells, Oncotarget, 10, 21, 2019. Crossref

  62. Wei Xiaolu, Yang Jian, Mao Yingqiu, Zhao Haiyu, Si Nan, Wang Hongjie, Bian Baolin, Arenobufagin Inhibits the Phosphatidylinositol 3-kinase/Protein Kinase B/Mammalian Target of Rapamycin Pathway and Induces Apoptosis and Autophagy in Pancreatic Cancer Cells, Pancreas, 49, 2, 2020. Crossref

  63. Booth Laurence, Roberts Jane L., Tavallai Mehrad, Chuckalovcak John, Stringer Daniel K., Koromilas Antonis E., Boone David L., McGuire William P., Poklepovic Andrew, Dent Paul, [Pemetrexed + Sorafenib] lethality is increased by inhibition of ERBB1/2/3-PI3K-NFκB compensatory survival signaling, Oncotarget, 7, 17, 2016. Crossref

  64. Fu Yuanyuan, Gu Qianqian, Luo Li, Xu Jiecheng, Luo Yuping, Xia Fan, Han Fanghai, Hong Liang, Yin Xiao-Ming, Huang Zhiying, Li Min, New Anti-Cancer Strategy to Suppress Colorectal Cancer Growth Through Inhibition of ATG4B and Lysosome Function, Cancers, 12, 6, 2020. Crossref

  65. Wei Xiaolu, He Jing, Gao Bo, Han Lingyu, Mao Yingqiu, Zhao Haiyu, Si Nan, Wang Hongjie, Yang Jian, Bian Baolin, Hellebrigenin anti-pancreatic cancer effects based on apoptosis and autophage, PeerJ, 8, 2020. Crossref

  66. Simioni Carolina, Cani Alice, Martelli Alberto M., Zauli Giorgio, Alameen Ayman A.M., Ultimo Simona, Tabellini Giovanna, McCubrey James A., Capitani Silvano, Neri Luca M., The novel dual PI3K/mTOR inhibitor NVP-BGT226 displays cytotoxic activity in both normoxic and hypoxic hepatocarcinoma cells, Oncotarget, 6, 19, 2015. Crossref

  67. Kulka Linda Anna Michelle, Fangmann Pia-Victoria, Panfilova Diana, Olzscha Heidi, Impact of HDAC Inhibitors on Protein Quality Control Systems: Consequences for Precision Medicine in Malignant Disease, Frontiers in Cell and Developmental Biology, 8, 2020. Crossref

  68. Zhang Hui‑Hui , Huang Zhong‑Xin , Zhong Su‑Quan , Fei Kui‑Lin , Cao You‑Han , miR‑21 inhibits autophagy and promotes malignant development in the bladder cancer T24�cell line, International Journal of Oncology, 2020. Crossref

  69. Zhang Yanyu, Huang Ning, Xu Jie, Zheng Wei, Cui Xing, <p>Homoharringtonine Exerts an Antimyeloma Effect by Promoting Excess Parkin-Dependent Mitophagy</p>, Drug Design, Development and Therapy, Volume 14, 2020. Crossref

  70. Nagini Siddavaram, Manikandan Palrasu, Malla Rama Rao, The Autophagy Conundrum in Cancer Development, Progression and Therapeutics, in Autophagy in tumor and tumor microenvironment, 2020. Crossref

  71. Al‐Bari Md. Abdul Alim, Co‐targeting of lysosome and mitophagy in cancer stem cells with chloroquine analogues and antibiotics, Journal of Cellular and Molecular Medicine, 24, 20, 2020. Crossref

  72. Han Yanyan, Fan Shujun, Qin Tao, Yang Jinfeng, Sun Yan, Lu Ying, Mao Jun, Li Lianhong, Role of autophagy in breast cancer and breast cancer stem cells (Review), International Journal of Oncology, 2018. Crossref

  73. Silva Valdenizia R., Neves Sara P., Santos Luciano de S., Dias Rosane B., Bezerra Daniel P., Challenges and Therapeutic Opportunities of Autophagy in Cancer Therapy, Cancers, 12, 11, 2020. Crossref

  74. Konac Ece, Kurman Yener, Baltaci Sümer, Contrast effects of autophagy in the treatment of bladder cancer, Experimental Biology and Medicine, 246, 3, 2021. Crossref

  75. Sarmah Dipanka Tanu, Bairagi Nandadulal, Chatterjee Samrat, Tracing the footsteps of autophagy in computational biology, Briefings in Bioinformatics, 22, 4, 2021. Crossref

  76. Wu Zhiming, Wang Huangen, Fang Sunyang, Xu Chaoyang, Roles of endoplasmic reticulum stress and autophagy on H2O2‑induced oxidative stress injury in HepG2 cells, Molecular Medicine Reports, 2018. Crossref

  77. Banikazemi Zarrin, Mirazimi Seyed Mohammad, Dashti Fatemeh, Mazandaranian Mohammad Reza, Akbari Maryam, Morshedi Korosh, Aslanbeigi Fatemeh, Rashidian Amir, Chamanara Mohsen, Hamblin Michael R., Taghizadeh Mohsen, Mirzaei Hamed, Coumarins and Gastrointestinal Cancer: A New Therapeutic Option?, Frontiers in Oncology, 11, 2021. Crossref

  78. Losmanova Tereza, Zens Philipp, Scherz Amina, Schmid Ralph A., Tschan Mario P., Berezowska Sabina, Chaperone-Mediated Autophagy Markers LAMP2A and HSPA8 in Advanced Non-Small Cell Lung Cancer after Neoadjuvant Therapy, Cells, 10, 10, 2021. Crossref

  79. Wei Renxiong, Cao Gang, Deng Zhouming, Su Jiajia, Cai Lin, miR-140-5p attenuates chemotherapeutic drug-induced cell death by regulating autophagy through inositol 1,4,5-trisphosphate kinase 2 (IP3k2) in human osteosarcoma cells, Bioscience Reports, 36, 5, 2016. Crossref

  80. Ding Guo-Bin, Sun Junqing, Wu Gengfeng, Li Binchun, Yang Peng, Li Zhuoyu, Nie Guangjun, Robust Anticancer Efficacy of a Biologically Synthesized Tumor Acidity-Responsive and Autophagy-Inducing Functional Beclin 1, ACS Applied Materials & Interfaces, 10, 6, 2018. Crossref

  81. Pandey Anchala, Yadav Pooja, Shukla Sanjeev, Unfolding the role of autophagy in the cancer metabolism, Biochemistry and Biophysics Reports, 28, 2021. Crossref

  82. Das Chandan Kanta, Banerjee Indranil, Mandal Mahitosh, Pro-survival autophagy: An emerging candidate of tumor progression through maintaining hallmarks of cancer, Seminars in Cancer Biology, 66, 2020. Crossref

  83. Lv Yuesheng, Zhang Wenjing, Zhao Jinyao, Sun Bing, Qi Yangfan, Ji Haoyu, Chen Chaoqun, Zhang Jinrui, Sheng Junxiu, Wang Taishu, Dominguez Daniel, Liu Han, Liu Quentin, Meng Songshu, Li Xiaoling, Wang Yang, SRSF1 inhibits autophagy through regulating Bcl-x splicing and interacting with PIK3C3 in lung cancer, Signal Transduction and Targeted Therapy, 6, 1, 2021. Crossref

  84. Patra Sayani, Elahi Naveed, Armorer Aaron, Arunachalam Swathi, Omala Joshua, Hamid Iman, Ashton Anthony W., Joyce David, Jiao Xuanmao, Pestell Richard G., Mechanisms Governing Metabolic Heterogeneity in Breast Cancer and Other Tumors, Frontiers in Oncology, 11, 2021. Crossref

  85. Shafabakhsh Rana, Arianfar Farzaneh, Vosough Massoud, Mirzaei Hamid Reza, Mahjoubin-Tehran Maryam, khanbabaei Hashem, Kowsari Hamed, Shojaie Layla, Azar Maryam Ebadi Fard, Hamblin Michael R., Mirzaei Hamed, Autophagy and gastrointestinal cancers: the behind the scenes role of long non-coding RNAs in initiation, progression, and treatment resistance, Cancer Gene Therapy, 28, 12, 2021. Crossref

  86. Sriram Navneeth, Sah Mahesh Kumar, Regulatory insights into progression of cancer and Alzheimer’s disorder from autophagy perspective, Molecular Biology Reports, 48, 12, 2021. Crossref

  87. Saffari-Chaleshtori Javad, Asadi-Samani Majid, Rasouli Maryam, Shafiee Sayed Mohammad, Autophagy and Ubiquitination as Two Major Players in Colorectal Cancer: A Review on Recent Patents, Recent Patents on Anti-Cancer Drug Discovery, 15, 2, 2020. Crossref

  88. Qazi Sahar, Sharma Ashok, Epigenetics in diagnosis, prognosis and therapy, in Epigenetics and Reproductive Health, 2021. Crossref

  89. Zhang Man, Bai Yang, Xu Chang, Qi Yiying, Meng Jiahong, Zhang Wenkan, Su Hang, Yan Weiqi, Blockage of Extracellular Signal-Regulated Kinase Exerts an Antitumor Effect via Regulating Energy Metabolism and Enhances the Efficacy of Autophagy Inhibitors by Regulating Transcription Factor EB Nuclear Translocation in Osteosarcoma, Frontiers in Cell and Developmental Biology, 9, 2021. Crossref

  90. Dewanjee Saikat, Das Sonjit, Joardar Swarnalata, Bhattacharjee Simanta, Chakraborty Pratik, Carotenoids as Anticancer Agents, in Carotenoids: Structure and Function in the Human Body, 2021. Crossref

  91. Mao Chenyu, Xu Xin, Ding Yongfeng, Xu Nong, Optimization of BCG Therapy Targeting Neutrophil Extracellular Traps, Autophagy, and miRNAs in Bladder Cancer: Implications for Personalized Medicine, Frontiers in Medicine, 8, 2021. Crossref

  92. Liang Xiao, Tang JiaCheng, Liang YueLong, Jin RenAn, Cai XiuJun, Suppression of autophagy by chloroquine sensitizes 5-fluorouracil-mediated cell death in gallbladder carcinoma cells, Cell & Bioscience, 4, 1, 2014. Crossref

  93. Jing Yuanming, Liang Wenqing, Liu Jian, Zhang Lin, Wei Jianguo, Yang Jianhui, Zhang Yu, Huang Zongliang, Autophagy-mediating microRNAs in cancer chemoresistance, Cell Biology and Toxicology, 36, 6, 2020. Crossref

  94. Wang Sheng, Feng Wei, Wang Wulin, Ye Xiaoman, Chen Hao, Yu Chunzhao, Girdin Knockdown Increases Gemcitabine Chemosensitivity to Pancreatic Cancer by Modulating Autophagy, Frontiers in Oncology, 11, 2021. Crossref

  95. Pal Singh Chawla Jatinder, Understanding Hidden Perspectives in Biology of Cancer - Approach to its Prevention and Management, Journal of Cancer Prevention & Current Research, 3, 5, 2015. Crossref

  96. Lee Su Hyun, Cho Won Jin, Najy Abdo J., Saliganan Allen-Dexter, Pham Tri, Rakowski Joseph, Loughery Brian, Ji Chang Hoon, Sakr Wael, Kim Seongho, Kato Ikuko, Chung Weon Kuu, Kim Harold E., Kwon Yong Tae, Kim Hyeong-Reh C., p62/SQSTM1-induced caspase-8 aggresomes are essential for ionizing radiation-mediated apoptosis, Cell Death & Disease, 12, 11, 2021. Crossref

  97. Bibyk Michael J., Campbell Melanie J., Hummon Amanda B., Mass spectrometric investigations of caloric restriction mimetics, PROTEOMICS, 21, 9, 2021. Crossref

  98. Jena Bikash Chandra, Rout Lipsa, Dey Ankita, Mandal Mahitosh, Active autophagy in cancer‐associated fibroblasts: Recent advances in understanding the novel mechanism of tumor progression and therapeutic response, Journal of Cellular Physiology, 236, 11, 2021. Crossref

  99. Ascenzi Francesca, De Vitis Claudia, Maugeri-Saccà Marcello, Napoli Christian, Ciliberto Gennaro, Mancini Rita, SCD1, autophagy and cancer: implications for therapy, Journal of Experimental & Clinical Cancer Research, 40, 1, 2021. Crossref

  100. Wang Yung-Li, Zheng Cai-Mei, Lee Yu-Hsuan, Cheng Ya-Yun, Lin Yuh-Feng, Chiu Hui-Wen, Micro- and Nanosized Substances Cause Different Autophagy-Related Responses, International Journal of Molecular Sciences, 22, 9, 2021. Crossref

  101. Park Jee Won, Kim Yesol, Lee Soo‐been, Oh Chae Won, Lee Eun Ji, Ko Je Yeong, Park Jong Hoon, Autophagy inhibits cancer stemness in triple‐negative breast cancer via miR‐181a‐mediated regulation of ATG5 and/or ATG2B, Molecular Oncology, 16, 9, 2022. Crossref

  102. Kurdi Ammar, Cleenewerck Matthias, Vangestel Christel, Lyssens Sophie, Declercq Wim, Timmermans Jean-Pierre, Stroobants Sigrid, Augustyns Koen, De Meyer Guido R.Y., Van Der Veken Pieter, Martinet Wim, ATG4B inhibitors with a benzotropolone core structure block autophagy and augment efficiency of chemotherapy in mice, Biochemical Pharmacology, 138, 2017. Crossref

  103. Bahar Entaz, Han Sun-Young, Kim Ji-Ye, Yoon Hyonok, Chemotherapy Resistance: Role of Mitochondrial and Autophagic Components, Cancers, 14, 6, 2022. Crossref

  104. Mengual Daniela, Medrano Luz Elena, Villamizar-Villamizar Wendy, Osorio-Llanes Estefanie, Mendoza-Torres Evelyn, Bolívar Samir, Novel Effects of Statins on Cancer via Autophagy, Pharmaceuticals, 15, 6, 2022. Crossref

  105. Singh Shikha Satendra, Vats Somya, Chia Amelia Yi-Qian, Tan Tuan Zea, Deng Shuo, Ong Mei Shan, Arfuso Frank, Yap Celestial T., Goh Boon Cher, Sethi Gautam, Huang Ruby Yun-Ju, Shen Han Ming, Manjithaya Ravi, Kumar Alan Prem, Dual role of autophagy in hallmarks of cancer, Oncogene, 37, 9, 2018. Crossref

  106. Kumar Prashant, Jagtap Yuvraj Anandrao, Patwa Som Mohanlal, Kinger Sumit, Dubey Ankur Rakesh, Prajapati Vijay Kumar, Dhiman Rohan, Poluri Krishna Mohan, Mishra Amit, Autophagy based cellular physiological strategies target oncogenic progression, Journal of Cellular Physiology, 237, 1, 2022. Crossref

  107. Rao Jingdong, Mei Ling, Liu Ji, Tang Xian, Yin Sheng, Xia Chunyu, Wei Jiaojie, Wan Dandan, Wang Xuhui, Wang Yashi, Li Man, Zhang Zhirong, He Qin, Size-adjustable micelles co-loaded with a chemotherapeutic agent and an autophagy inhibitor for enhancing cancer treatment via increased tumor retention, Acta Biomaterialia, 89, 2019. Crossref

  108. Kim Na-Yeon, Han Byeal-I, Lee Michael, Cytoprotective role of autophagy against BH3 mimetic gossypol in ATG5 knockout cells generated by CRISPR-Cas9 endonuclease, Cancer Letters, 370, 1, 2016. Crossref

  109. Ishihara Keiichi, Kanai Shiho, Tanaka Kikuko, Kawashita Eri, Akiba Satoshi, Group IVA phospholipase A2 deficiency prevents CCl4-induced hepatic cell death through the enhancement of autophagy, Biochemical and Biophysical Research Communications, 471, 1, 2016. Crossref

  110. Han Byeal-I, Hwang Sung-Hee, Lee Michael, A progressive reduction in autophagic capacity contributes to induction of replicative senescence in Hs68 cells, The International Journal of Biochemistry & Cell Biology, 92, 2017. Crossref

  111. Papadopoli David, Pollak Michael, Topisirovic Ivan, The role of GSK3 in metabolic pathway perturbations in cancer, Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, 1868, 8, 2021. Crossref

  112. Trybus Wojciech, Król Teodora, Trybus Ewa, Rhein induces changes in the lysosomal compartment of HeLa cells, Journal of Cellular Biochemistry, 123, 9, 2022. Crossref

Begell Digital Portal Begellデジタルライブラリー 電子書籍 ジャーナル 参考文献と会報 リサーチ集 価格及び購読のポリシー Begell House 連絡先 Language English 中文 Русский Português German French Spain