Abonnement à la biblothèque: Guest
Critical Reviews™ in Oncogenesis

Publication de 4  numéros par an

ISSN Imprimer: 0893-9675

ISSN En ligne: 2162-6448

SJR: 0.395 SNIP: 0.322 CiteScore™:: 2.5 H-Index: 54

Indexed in

Autophagy and Hallmarks of Cancer

Volume 23, Numéro 5-6, 2018, pp. 247-267
DOI: 10.1615/CritRevOncog.2018027913
Get accessGet access

RÉSUMÉ

Autophagy is a catabolic program that is responsible for the degradation of dysfunctional or unnecessary proteins and organelles to maintain cellular homeostasis. Mechanistically, it involves the formation of double-membrane autophagosomes that sequester cytoplasmic material and deliver it to lysosomes for degradation. Eventually, the material is recycled back to the cytoplasm. Abnormalities of autophagy often lead to human diseases, such as neurodegeneration and cancer. In the case of cancer, increasing evidence has revealed the paradoxical roles of autophagy in both tumor inhibition and tumor promotion. Here, we summarize the context-dependent role of autophagy and its complicated molecular mechanisms in the hallmarks of cancer. Moreover, we discuss how therapeutics targeting autophagy can counter malignant transformation and tumor progression. Overall, the findings of studies discussed here shed new light on exploiting the complicated mechanisms of the autophagic machinery and relevant small-molecule modulators as potential antitumor agents to improve therapeutic outcomes.

CITÉ PAR
  1. Delou , Souza , Souza , Borges , Highlights in Resistance Mechanism Pathways for Combination Therapy, Cells, 8, 9, 2019. Crossref

  2. Rein Theo, Is Autophagy Involved in the Diverse Effects of Antidepressants?, Cells, 8, 1, 2019. Crossref

  3. Wang Hairui, Huang Yongzhuo, Combination therapy based on nano codelivery for overcoming cancer drug resistance, Medicine in Drug Discovery, 6, 2020. Crossref

  4. Yun Chul Won, Jeon Juhee, Go Gyeongyun, Lee Jun Hee, Lee Sang Hun, The Dual Role of Autophagy in Cancer Development and a Therapeutic Strategy for Cancer by Targeting Autophagy, International Journal of Molecular Sciences, 22, 1, 2020. Crossref

  5. da Silva Filho Antônio Felix, de Sousa Lizandra Maia, Consonni Silvio Roberto, da Rocha Pitta Maira Galdino, Carvalho Hernandes Faustino, de Melo Rêgo Moacyr Jesus Barreto, Galectin-3 Expression in Pancreatic Cell Lines Under Distinct Autophagy-Inducing Stimulus, Microscopy and Microanalysis, 26, 6, 2020. Crossref

  6. Alvarez-Meythaler Jose G., Garcia-Mayea Yoelsis, Mir Cristina, Kondoh Hiroshi, LLeonart Matilde E., Autophagy Takes Center Stage as a Possible Cancer Hallmark, Frontiers in Oncology, 10, 2020. Crossref

  7. Basak Debasish, Arrighi Scott, Darwiche Yasenya, Deb Subrata, Comparison of Anticancer Drug Toxicities: Paradigm Shift in Adverse Effect Profile, Life, 12, 1, 2021. Crossref

  8. Song Yan, Zhang Hongxing, Wang Xia, Geng Xin, Sun Yuanqiang, Liu Jing, Li Zhaohui, One Stone, Three Birds: pH Triggered Transformation of Aminopyronine and Iminopyronine Based Lysosome Targeting Viscosity Probe for Cancer Visualization, Analytical Chemistry, 93, 3, 2021. Crossref

  9. Gupta Shruti, Kumar Atul, Tejavath Kiran Kumar, A pharmacognostic approach for mitigating pancreatic cancer: emphasis on herbal extracts and phytoconstituents, Future Journal of Pharmaceutical Sciences, 7, 1, 2021. Crossref

  10. Carvajal Lorena, Gutiérrez Jaime, Morselli Eugenia, Leiva Andrea, Autophagy Process in Trophoblast Cells Invasion and Differentiation: Similitude and Differences With Cancer Cells, Frontiers in Oncology, 11, 2021. Crossref

  11. He Bin, Zhang Wenkan, He Jiaming, Fraxinellone Has Anticancer Activity by Inducing Osteosarcoma Cell Apoptosis via Promoting Excessive Autophagy Flux, Frontiers in Pharmacology, 12, 2021. Crossref

  12. Auger Clément, Christou Niki, Brunel Aude, Perraud Aurélie, Verdier Mireille, Autophagy and Extracellular Vesicles in Colorectal Cancer: Interactions and Common Actors?, Cancers, 13, 5, 2021. Crossref

  13. Wang Ying, Wei Zihao, Pan Keran, Li Jing, Chen Qianming, The function and mechanism of ferroptosis in cancer, Apoptosis, 25, 11-12, 2020. Crossref

  14. Kabel Ahmed M., Arab Hany H., Abd Elmaaboud Maaly A., Effect of dapagliflozin and/or L‐arginine on solid tumor model in mice: The interaction between nitric oxide, transforming growth factor‐beta 1, autophagy, and apoptosis, Fundamental & Clinical Pharmacology, 35, 6, 2021. Crossref

  15. Verma Elika, Kumar Aviral, Devi Daimary Uzini, Parama Dey, Girisa Sosmitha, Sethi Gautam, Kunnumakkara Ajaikumar B., Potential of baicalein in the prevention and treatment of cancer: A scientometric analyses based review, Journal of Functional Foods, 86, 2021. Crossref

  16. Ponte Luis Gustavo Saboia, Pavan Isadora Carolina Betim, Mancini Mariana Camargo Silva, da Silva Luiz Guilherme Salvino, Morelli Ana Paula, Severino Matheus Brandemarte, Bezerra Rosangela Maria Neves, Simabuco Fernando Moreira, The Hallmarks of Flavonoids in Cancer, Molecules, 26, 7, 2021. Crossref

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

  18. Martins Waleska Kerllen, Silva Maryana do Nascimento da, Pandey Kiran, Maejima Ikuko, Ramalho Ercília, Olivon Vania Claudia, Diniz Susana Nogueira, Grasso Daniel, Autophagy-targeted therapy to modulate age-related diseases: Success, pitfalls, and new directions, Current Research in Pharmacology and Drug Discovery, 2, 2021. Crossref

  19. Xu Chunxiao, Zang Yusheng, Zhao Yuxiang, Cui Weiqiang, Zhang Hong, Zhu Yingcui, Xu Man, Comprehensive Pan-Cancer Analysis Confirmed That ATG5 Promoted the Maintenance of Tumor Metabolism and the Occurrence of Tumor Immune Escape, Frontiers in Oncology, 11, 2021. Crossref

  20. Sediqi Nelofar, Jahidin Aisyah Hasyila, Hasan Mizaton Hazizul, Zakaria Yuslina, Two-Pore Channels in Cancer Hallmarks: An Update Review, Biomedical and Pharmacology Journal, 14, 3, 2021. Crossref

  21. Guo Jin-Cheng, Wei Qing-Shuang, Dong Lei, Fang Shuang-Sang, Li Feng, Zhao Yi, Prognostic Value of an Autophagy-Related Five-Gene Signature for Lower-Grade Glioma Patients, Frontiers in Oncology, 11, 2021. Crossref

  22. Song Kun, Li Bin, Chen Ying-Ying, Wang Hua, Liu Kang-Cheng, Tan Wei, Zou Jing, LRPPRC regulates metastasis and glycolysis by modulating autophagy and the ROS/HIF1-α pathway in retinoblastoma, Molecular Therapy - Oncolytics, 22, 2021. Crossref

  23. Zhou Hong, Xie Xin, Chen Ying, Lin Yi, Cai Zhaogen, Ding Li, Wu Yijie, Peng Yongde, Tang Shanshan, Xu Huanbai, Chaperone-mediated Autophagy Governs Progression of Papillary Thyroid Carcinoma via PPARγ-SDF1/CXCR4 Signaling, The Journal of Clinical Endocrinology & Metabolism, 105, 10, 2020. Crossref

  24. Ding Yi, Li Tian, Li Min, Tayier Tuersong, Zhang MeiLin, Chen Long, Feng ShuMei, Xu Yan-Ming, A Novel Autophagy-Related lncRNA Gene Signature to Improve the Prognosis of Patients with Melanoma, BioMed Research International, 2021, 2021. Crossref

  25. Wan Teng, Fu Mingyuan, Wu Zhuan, Gao Xue, Zhou Shouhong, Advances in the role of autophagy in the development of retinoblastoma (Review), Oncology Letters, 22, 2, 2021. Crossref

  26. Papaefthymiou Apostolis, Christodoulidis Gregory, Koffas Apostolos, Doulberis Michael, Polyzos Stergios A, Manolakis Anastasios, Potamianos Spyros, Kapsoritakis Andreas, Kountouras Jannis, Role of autophagy in gastric carcinogenesis, World Journal of Gastrointestinal Oncology, 13, 10, 2021. Crossref

  27. 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

  28. Barca Ida, Mignogna Chiara, Novembre Daniela, Ferragina Francesco, Cristofaro Maria Giulia, Immunohistochemical Analysis of the Beclin-1 Expression Predicts the Progression of Oral Squamous Cell Carcinoma, International Journal of Environmental Research and Public Health, 18, 21, 2021. Crossref

  29. Kim So Young, Hwangbo Hyun, Kim Min Yeong, Ji Seon Yeong, Lee Hyesook, Kim Gi-Young, Kwon Chan-Young, Leem Sun-Hee, Hong Su Hyun, Cheong JaeHun, Choi Yung Hyun, Coptisine induces autophagic cell death through down-regulation of PI3K/Akt/mTOR signaling pathway and up-regulation of ROS-mediated mitochondrial dysfunction in hepatocellular carcinoma Hep3B cells, Archives of Biochemistry and Biophysics, 697, 2021. Crossref

  30. Li Juan, Zhang Zhi-chao, Yuan Xiang-shi, Tang Shan-shan, Wang Tao, Liu Hong-fu, Cao Yu, Shi Zhongjie, TOPK Affects Autophagy of Skin Squamous Cell Carcinoma by Regulating NF-KB Pathway through HDAC1, Disease Markers, 2022, 2022. Crossref

  31. Hirata Amanda S., La Clair James J., Jimenez Paula C., Costa-Lotufo Leticia Veras, Fenical William, Preclinical Development of Seriniquinones as Selective Dermcidin Modulators for the Treatment of Melanoma, Marine Drugs, 20, 5, 2022. Crossref

  32. Zhou Huimin, Wang Kexin, Wang Mengyan, Zhao Wenxia, Zhang Conghui, Cai Meilian, Qiu Yuhan, Zhang Tianshu, Shao Rongguang, Zhao Wuli, ER-phagy in the Occurrence and Development of Cancer, Biomedicines, 10, 3, 2022. Crossref

  33. Ravi Swapna, Alencar Antonio M, Arakelyan Jemma, Xu Weihao, Stauber Roberta, Wang Cheng-Chi I, Papyan Ruzanna, Ghazaryan Narine, Pereira Rosalina M, An Update to Hallmarks of Cancer, Cureus, 2022. Crossref

  34. Huang Wei, Huang Hongyan, Xiao Yuzhen, Wang Lei, Zhang Tingting, Fang Xiaoling, Xia Xiaomeng, UBE2T is upregulated, predicts poor prognosis, and promotes cell proliferation and invasion by promoting epithelial-mesenchymal transition via inhibiting autophagy in an AKT/mTOR dependent manner in ovarian cancer, Cell Cycle, 21, 8, 2022. Crossref

  35. Jia Li, Chen Yan, Chen Fukun, Lv Juan, Li Yanling, Hou Fei, Yang Zhixian, Deng Zhiyong, Small activating RNA-activated NIS gene promotes 131I uptake and inhibits thyroid cancer via AMPK/mTOR pathway, Pathology - Research and Practice, 229, 2022. Crossref

  36. Spano Daniela, Colanzi Antonino, Golgi Complex: A Signaling Hub in Cancer, Cells, 11, 13, 2022. Crossref

  37. Kurhaluk Natalia, Tkachenko Halyna, Effects of melatonin and metformin in preventing lysosome-induced autophagy and oxidative stress in rat models of carcinogenesis and the impact of high-fat diet, Scientific Reports, 12, 1, 2022. Crossref

  38. Chen Zheng, Liu Shuang, Xie Peiyi, Zhang Bo, Yu Mincheng, Yan Jiuliang, Jin Lei, Zhang Wentao, Zhou Binghai, Li Xiaoqiang, Xiao Yongsheng, Xu Yongfeng, Ye Qinghai, Li Hui, Guo Lei, Tumor-derived PD1 and PD-L1 could promote hepatocellular carcinoma growth through autophagy induction in vitro, Biochemical and Biophysical Research Communications, 605, 2022. Crossref

  39. Zhao Weiwei, Wang Qing, Li Le, Xie Chengshen, Wu Yequn, Gautam Mayank, Li Lijia, SIRT1 regulates mitotic catastrophe via autophagy and BubR1 signaling, Molecular and Cellular Biochemistry, 2022. Crossref

  40. Ferrari Elena, Bettuzzi Saverio, Naponelli Valeria, The Potential of Epigallocatechin Gallate (EGCG) in Targeting Autophagy for Cancer Treatment: A Narrative Review, International Journal of Molecular Sciences, 23, 11, 2022. Crossref

  41. Chaeichi-Tehrani Negin, Ferns Gordon A., Hassanian Seyed Mahdi, Khazaei Majid, Avan Amir, The Therapeutic Potential of Targeting Autophagy in the Treatment of Cancer, Current Cancer Drug Targets, 21, 9, 2021. Crossref

  42. Skelding Kathryn A., Barry Daniel L., Theron Danielle Z., Lincz Lisa F., Targeting the two-pore channel 2 in cancer progression and metastasis, Exploration of Targeted Anti-tumor Therapy, 2022. Crossref

  43. Wang Wenwen, Pei Qingshan, Wang Lifen, Mu Tong, Feng Hua, Construction of a Prognostic Signature of 10 Autophagy-Related lncRNAs in Gastric Cancer, International Journal of General Medicine, Volume 15, 2022. Crossref

  44. 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

  45. Mirzaei Sepideh, Gholami Mohammad Hossein, Zabolian Amirhossein, Saleki Hossein, Farahani Mahdi Vasheghani, Hamzehlou Soodeh, Far Fatemeh Bakhtiari, Sharifzadeh Seyed Omid, Samarghandian Saeed, Khan Haroon, Aref Amir Reza, Ashrafizadeh Milad, Zarrabi Ali, Sethi Gautam, Caffeic acid and its derivatives as potential modulators of oncogenic molecular pathways: New hope in the fight against cancer, Pharmacological Research, 171, 2021. Crossref

  46. Mudaliar Priyanka, Nalawade Apoorva, Devarajan Shine, Aich Jyotirmoi, Therapeutic potential of autophagy activators and inhibitors in lung and breast cancer- a review, Molecular Biology Reports, 2022. Crossref

  47. Yao Yinan, Shi Yuxin, Gao Zizhe, Sun Yutong, Yao Fan, Ma Li, Ferroptosis at the crossroads of tumor-host interactions, metastasis, and therapy response, American Journal of Physiology-Cell Physiology, 323, 1, 2022. Crossref

  48. Wei Xianzhe, Li Juan, Yang Xiaofeng, Dong Baoli, Geng Bing, Li Zengjun, Hu Xiaoxiao, Ding Biyan, Zhang Jing, Yan Mei, An enzyme-activated two-photon ratiometric fluorescent probe with lysosome targetability for imaging β-glucuronidase in colon cancer cells and tissue, Analytica Chimica Acta, 1192, 2022. Crossref

  49. Wu Dan, Ding Yi, Fan JunBai, Bioinformatics Analysis of Autophagy-related lncRNAs in Esophageal Carcinoma, Combinatorial Chemistry & High Throughput Screening, 25, 8, 2022. Crossref

  50. Wu Guangzhen, Xu Yingkun, Zhang Huayu, Ruan Zihao, Zhang Peizhi, Wang Zicheng, Gao Han, Che Xiangyu, Xia Qinghua, Chen Feng, A new prognostic risk model based on autophagy-related genes in kidney renal clear cell carcinoma, Bioengineered, 12, 1, 2021. Crossref

  51. Qin Yan-qiu, Liu Si-yu, Lv Mei-ling, Sun Wei-liang, Ambra1 in cancer: implications for clinical oncology, Apoptosis, 27, 9-10, 2022. Crossref

  52. Li Hanxue, Li Meng, Chen Kuichi, Li Yueheng, Yang Zhengyan, Zhou Zhi, The circadian clock gene ARNTL overexpression suppresses oral cancer progression by inducing apoptosis via activating autophagy, Medical Oncology, 39, 12, 2022. Crossref

  53. De Lorenzo Stefania, Tovoli Francesco, Trevisani Franco, Mechanisms of Primary and Acquired Resistance to Immune Checkpoint Inhibitors in Patients with Hepatocellular Carcinoma, Cancers, 14, 19, 2022. Crossref

  54. Gao Xiaozhuo, Gao Bo, Li Shenglong, Extracellular vesicles: A new diagnostic biomarker and targeted drug in osteosarcoma, Frontiers in Immunology, 13, 2022. Crossref

  55. Warzecha Karolina W., Pudełek Maciej, Catapano Jessica, Madeja Zbigniew, Czyż Jarosław, Long-Term Fenofibrate Treatment Stimulates the Phenotypic Microevolution of Prostate Cancer Cells In Vitro, Pharmaceuticals, 15, 11, 2022. Crossref

  56. Xu Haifeng, Xu Bing, Hu Jiayu, Xia Jun, Tong Le, Zhang Ping, Yang Lei, Tang Lusheng, Chen Sufeng, Du Jing, Wang Ying, Li Yanchun, Development of a novel autophagy-related gene model for gastric cancer prognostic prediction, Frontiers in Oncology, 12, 2022. Crossref

  57. Smith Fraser, States of Ill Health: The Ground of Clinical Presentations, in Naturopathic Medicine, 2022. Crossref

  58. Catapano Jessica, Luty Marcin, Wróbel Tomasz, Pudełek Maciej, Piwowarczyk Katarzyna, Kędracka-Krok Sylwia, Siedlar Maciej, Madeja Zbigniew, Czyż Jarosław, Acquired drug resistance interferes with the susceptibility of prostate cancer cells to metabolic stress, Cellular & Molecular Biology Letters, 27, 1, 2022. Crossref

Portail numérique Bibliothèque numérique eBooks Revues Références et comptes rendus Collections Prix et politiques d'abonnement Begell House Contactez-nous Language English 中文 Русский Português German French Spain