Autophagy: Shedding Light on the Mechanisms and Multifaceted Roles in Cancers
Abstract
:1. Introduction
2. Major Types of Autophagy
3. Process of Autophagy
3.1. Initiation
3.2. Nucleation
3.3. Elongation
4. Links Between Autophagy and Cancers
4.1. Lung Cancer
4.2. Breast Cancer
4.3. Esophageal Cancer
4.4. Colorectal Cancer
4.5. Prostate Cancer
4.6. Hematologic Malignancies
4.7. Other Cancers
5. The Possibility of Developing Autophagy Modulators for Cancer Treatment
6. Discussion and Future Directions
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
AMBRA1 | Autophagy and Beclin 1 regulator 1 |
AMPK | AMP-activated protein kinase |
ATG | Autophagy-related genes |
CMA | Chaperone-mediated autophagy |
CML | Chronic myeloid leukemia |
CPT | Camptothecin |
CRC | Colorectal cancer |
EAC | Esophageal adenocarcinoma |
ER | Endoplasmic reticulum |
ESCC | Esophageal squamous cell carcinoma |
HCC | Hepatocellular carcinoma |
HER2 | Human epidermal growth factor receptor 2 |
LAMP2A | Lysosomal-associated membrane protein 2A |
LC3 | Microtubule-associated protein light chain 3 |
MTORC1 | Mechanistic target of rapamycin kinase complex 1 |
NRBF2 | Nuclear receptor binding factor 2 |
NSCLC | Non-small cell lung cancer |
OXA | Oxaliplatin |
PAS | Pre-autophagosomal structure |
PIK3C3 | Phosphatidylinositol-3-kinase catalytic subunit type 3 |
PIK3R4 | Phosphoinositide-3-kinase regulatory subunit 4 |
PTC | Papillary thyroid carcinoma |
PtdIns3K | The class III phosphatidylinositol 3-kinase |
RB1CC1 | RB1-inducible coiled-coil 1 |
ROS | Reactive oxygen species |
RUBCN | Rubicon autophagy regulator |
SCLC | Small-cell lung cancer |
SH3GLB1 | SH3 domain containing GRB2-like endophilin B1 |
TKIs | Tyrosine kinase inhibitors |
ULK1/2 | Unc-51-like autophagy-activating kinase 1/2 |
UVRAG | UV radiation resistance-associated |
3MA | 3-methyladenine |
References
- De Duve, C.; Wattiaux, R. Functions of lysosomes. Annu. Rev. Physiol. 1966, 28, 435–492. [Google Scholar] [CrossRef]
- Kawamata, T.; Kamada, Y.; Kabeya, Y.; Sekito, T.; Ohsumi, Y. Organization of the pre-autophagosomal structure responsible for autophagosome formation. Mol. Biol. Cell 2008, 19, 2039–2050. [Google Scholar] [CrossRef] [PubMed]
- Xie, Z.; Klionsky, D.J. Autophagosome formation: Core machinery and adaptations. Nat. Cell Biol. 2007, 9, 1102–1109. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.; Klionsky, D.J. Mammalian autophagy: Core molecular machinery and signaling regulation. Curr. Opin. Cell Biol. 2010, 22, 124–131. [Google Scholar] [CrossRef] [PubMed]
- Glick, D.; Barth, S.; Macleod, K.F. Autophagy: Cellular and molecular mechanisms. J. Pathol. 2010, 221, 3–12. [Google Scholar] [CrossRef]
- Kocaturk, N.M.; Akkoc, Y.; Kig, C.; Bayraktar, O.; Gozuacik, D.; Kutlu, O. Autophagy as a molecular target for cancer treatment. Eur. J. Pharm. Sci. 2019, 134, 116–137. [Google Scholar] [CrossRef]
- Dikic, I.; Elazar, Z. Mechanism and medical implications of mammalian autophagy. Nat. Rev. Mol. Cell Biol. 2018, 19, 349–364. [Google Scholar] [CrossRef]
- International Agency for Research on Cancer. Global cancer burden growing, amidst mounting need for services. Saudi. Med. J. 2024, 45, 326–327. [Google Scholar]
- Poole, L.P.; Macleod, K.F. Mitophagy in tumorigenesis and metastasis. Cell. Mol. Life Sci. 2021, 78, 3817–3851. [Google Scholar] [CrossRef]
- Liu, J.; Debnath, J. The Evolving, Multifaceted Roles of Autophagy in Cancer. Adv. Cancer Res. 2016, 130, 1–53. [Google Scholar] [CrossRef]
- Bhatt, V.; Khayati, K.; Hu, Z.S.; Lee, A.; Kamran, W.; Su, X.; Guo, J.Y. Autophagy modulates lipid metabolism to maintain metabolic flexibility for Lkb1-deficient Kras-driven lung tumorigenesis. Genes Dev. 2019, 33, 150–165. [Google Scholar] [CrossRef]
- Niu, X.; You, Q.; Hou, K.; Tian, Y.; Wei, P.; Zhu, Y.; Gao, B.; Ashrafizadeh, M.; Aref, A.R.; Kalbasi, A.; et al. Autophagy in cancer development, immune evasion, and drug resistance. Drug Resist. Updat. 2025, 78, 101170. [Google Scholar] [CrossRef] [PubMed]
- Chao, X.; Qian, H.; Wang, S.; Fulte, S.; Ding, W.X. Autophagy and liver cancer. Clin. Mol. Hepatol. 2020, 26, 606–617. [Google Scholar] [CrossRef]
- Debnath, J.; Gammoh, N.; Ryan, K.M. Autophagy and autophagy-related pathways in cancer. Nat. Rev. Mol. Cell Biol. 2023, 24, 560–575. [Google Scholar] [CrossRef] [PubMed]
- Seo, W.; Silwal, P.; Song, I.C.; Jo, E.K. The dual role of autophagy in acute myeloid leukemia. J. Hematol. Oncol. 2022, 15, 51. [Google Scholar] [CrossRef]
- Barnard, R.A.; Regan, D.P.; Hansen, R.J.; Maycotte, P.; Thorburn, A.; Gustafson, D.L. Autophagy Inhibition Delays Early but Not Late-Stage Metastatic Disease. J. Pharmacol. Exp. Ther. 2016, 358, 282–293. [Google Scholar] [CrossRef] [PubMed]
- White, E. Deconvoluting the context-dependent role for autophagy in cancer. Nat. Rev. Cancer 2012, 12, 401–410. [Google Scholar] [CrossRef]
- Wang, D.; Li, Y.; Chang, W.; Feng, M.; Yang, Y.; Zhu, X.; Liu, Z.; Fu, Y. CircSEC24B activates autophagy and induces chemoresistance of colorectal cancer via OTUB1-mediated deubiquitination of SRPX2. Cell Death Dis. 2024, 15, 693. [Google Scholar] [CrossRef]
- Grandvallet, C.; Feugeas, J.P.; Monnien, F.; Despouy, G.; Valerie, P.; Michael, G.; Hervouet, E.; Peixoto, P. Autophagy is associated with a robust specific transcriptional signature in breast cancer subtypes. Genes Cancer 2020, 11, 154–168. [Google Scholar] [CrossRef]
- Qu, X.; Yu, J.; Bhagat, G.; Furuya, N.; Hibshoosh, H.; Troxel, A.; Rosen, J.; Eskelinen, E.L.; Mizushima, N.; Ohsumi, Y.; et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J. Clin. Investig. 2003, 112, 1809–1820. [Google Scholar] [CrossRef]
- Yue, Z.; Jin, S.; Yang, C.; Levine, A.J.; Heintz, N. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc. Natl. Acad. Sci. USA 2003, 100, 15077–15082. [Google Scholar] [CrossRef] [PubMed]
- Zhan, Y.; Wang, K.; Li, Q.; Zou, Y.; Chen, B.; Gong, Q.; Ho, H.I.; Yin, T.; Zhang, F.; Lu, Y.; et al. The Novel Autophagy Inhibitor Alpha-Hederin Promoted Paclitaxel Cytotoxicity by Increasing Reactive Oxygen Species Accumulation in Non-Small Cell Lung Cancer Cells. Int. J. Mol. Sci. 2018, 19, 3221. [Google Scholar] [CrossRef]
- Xu, S.W.; Law, B.Y.; Mok, S.W.; Leung, E.L.; Fan, X.X.; Coghi, P.S.; Zeng, W.; Leung, C.H.; Ma, D.L.; Liu, L.; et al. Autophagic degradation of epidermal growth factor receptor in gefitinib-resistant lung cancer by celastrol. Int. J. Oncol. 2016, 49, 1576–1588. [Google Scholar] [CrossRef]
- Li, J.; Zhan, H.; Ren, Y.; Feng, M.; Wang, Q.; Jiao, Q.; Wang, Y.; Liu, X.; Zhang, S.; Du, L.; et al. Sirtuin 4 activates autophagy and inhibits tumorigenesis by upregulating the p53 signaling pathway. Cell Death Differ. 2023, 30, 313–326. [Google Scholar] [CrossRef]
- Mei, L.; Liu, H.; Ding, Q.; Xie, Y.; Shen, X.; Chen, H.; Wang, K.; Li, M.; He, Q. Self-Propelled Smart Nanomotors for Enhanced Mild Photothermal Therapy of Tumors through Autophagy Modulation. Acta Biomater. 2025. [Google Scholar] [CrossRef] [PubMed]
- Cheng, B.; Li, M.; Zheng, J.; Liang, J.; Li, Y.; Liang, R.; Tian, H.; Zhou, Z.; Ding, L.; Ren, J.; et al. Chemically engineered antibodies for autophagy-based receptor degradation. Nat. Chem. Biol. 2025, 21, 855–866. [Google Scholar] [CrossRef]
- Zhang, G.; Chen, Y.; Huang, X.; Liang, T. Cancer immunotherapeutic challenges from autophagy-immune checkpoint reciprocal regulation. Trends Cancer 2025, 11, 169–184. [Google Scholar] [CrossRef] [PubMed]
- Mizushima, N. A brief history of autophagy from cell biology to physiology and disease. Nat. Cell Biol. 2018, 20, 521–527. [Google Scholar] [CrossRef]
- Wang, L.; Klionsky, D.J.; Shen, H.M. The emerging mechanisms and functions of microautophagy. Nat. Rev. Mol. Cell Biol. 2023, 24, 186–203. [Google Scholar] [CrossRef]
- Yamamoto, H.; Zhang, S.; Mizushima, N. Autophagy genes in biology and disease. Nat. Rev. Genet. 2023, 24, 382–400. [Google Scholar] [CrossRef]
- Marzella, L.; Ahlberg, J.; Glaumann, H. Autophagy, heterophagy, microautophagy and crinophagy as the means for intracellular degradation. Virchows Arch. B Cell Pathol. Incl. Mol. Pathol. 1981, 36, 219–234. [Google Scholar] [CrossRef]
- Levy, J.M.M.; Towers, C.G.; Thorburn, A. Targeting autophagy in cancer. Nat. Rev. Cancer 2017, 17, 528–542. [Google Scholar] [CrossRef]
- Martin-Aparicio, E.; Yamamoto, A.; Hernandez, F.; Hen, R.; Avila, J.; Lucas, J.J. Proteasomal-dependent aggregate reversal and absence of cell death in a conditional mouse model of Huntington’s disease. J. Neurosci. 2001, 21, 8772–8781. [Google Scholar] [CrossRef] [PubMed]
- Nixon, R.A. Autophagy, amyloidogenesis and Alzheimer disease. J. Cell Sci. 2007, 120, 4081–4091. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.; Klionsky, D.J. An overview of the molecular mechanism of autophagy. Curr. Top. Microbiol. Immunol. 2009, 335, 1–32. [Google Scholar] [CrossRef] [PubMed]
- Mizushima, N.; Yoshimori, T.; Ohsumi, Y. The role of Atg proteins in autophagosome formation. Annu. Rev. Cell Dev. Biol. 2011, 27, 107–132. [Google Scholar] [CrossRef]
- Anding, A.L.; Baehrecke, E.H. Cleaning House: Selective Autophagy of Organelles. Dev. Cell 2017, 41, 10–22. [Google Scholar] [CrossRef]
- Kirkin, V.; Rogov, V.V. A Diversity of Selective Autophagy Receptors Determines the Specificity of the Autophagy Pathway. Mol. Cell 2019, 76, 268–285. [Google Scholar] [CrossRef]
- Kaushik, S.; Cuervo, A.M. The coming of age of chaperone-mediated autophagy. Nat. Rev. Mol. Cell Biol. 2018, 19, 365–381. [Google Scholar] [CrossRef]
- Klionsky, D.J.; Petroni, G.; Amaravadi, R.K.; Baehrecke, E.H.; Ballabio, A.; Boya, P.; Bravo-San Pedro, J.M.; Cadwell, K.; Cecconi, F.; Choi, A.M.K.; et al. Autophagy in major human diseases. EMBO J. 2021, 40, e108863. [Google Scholar] [CrossRef]
- Nakatogawa, H.; Ichimura, Y.; Ohsumi, Y. Atg8, a ubiquitin-like protein required for autophagosome formation, mediates membrane tethering and hemifusion. Cell 2007, 130, 165–178. [Google Scholar] [CrossRef]
- Axe, E.L.; Walker, S.A.; Manifava, M.; Chandra, P.; Roderick, H.L.; Habermann, A.; Griffiths, G.; Ktistakis, N.T. Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum. J. Cell Biol. 2008, 182, 685–701. [Google Scholar] [CrossRef] [PubMed]
- Hamasaki, M.; Furuta, N.; Matsuda, A.; Nezu, A.; Yamamoto, A.; Fujita, N.; Oomori, H.; Noda, T.; Haraguchi, T.; Hiraoka, Y.; et al. Autophagosomes form at ER-mitochondria contact sites. Nature 2013, 495, 389–393. [Google Scholar] [CrossRef]
- Simonsen, A.; Tooze, S.A. Coordination of membrane events during autophagy by multiple class III PI3-kinase complexes. J. Cell Biol. 2009, 186, 773–782. [Google Scholar] [CrossRef]
- Galluzzi, L.; Baehrecke, E.H.; Ballabio, A.; Boya, P.; Bravo-San Pedro, J.M.; Cecconi, F.; Choi, A.M.; Chu, C.T.; Codogno, P.; Colombo, M.I.; et al. Molecular definitions of autophagy and related processes. EMBO J. 2017, 36, 1811–1836. [Google Scholar] [CrossRef] [PubMed]
- He, C.; Klionsky, D.J. Regulation mechanisms and signaling pathways of autophagy. Annu. Rev. Genet. 2009, 43, 67–93. [Google Scholar] [CrossRef]
- Nanji, T.; Liu, X.; Chew, L.H.; Li, F.K.; Biswas, M.; Yu, Z.Q.; Lu, S.; Dong, M.Q.; Du, L.L.; Klionsky, D.J.; et al. Conserved and unique features of the fission yeast core Atg1 complex. Autophagy 2017, 13, 2018–2027. [Google Scholar] [CrossRef] [PubMed]
- Hosokawa, N.; Hara, T.; Kaizuka, T.; Kishi, C.; Takamura, A.; Miura, Y.; Iemura, S.; Natsume, T.; Takehana, K.; Yamada, N.; et al. Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol. Biol. Cell 2009, 20, 1981–1991. [Google Scholar] [CrossRef]
- Popelka, H.; Klionsky, D.J. The RB1CC1 Claw-binding motif: A new piece in the puzzle of autophagy regulation. Autophagy 2022, 18, 237–239. [Google Scholar] [CrossRef]
- Nagy, P.; Karpati, M.; Varga, A.; Pircs, K.; Venkei, Z.; Takats, S.; Varga, K.; Erdi, B.; Hegedus, K.; Juhasz, G. Atg17/FIP200 localizes to perilysosomal Ref(2)P aggregates and promotes autophagy by activation of Atg1 in Drosophila. Autophagy 2014, 10, 453–467. [Google Scholar] [CrossRef]
- Park, S.H.; Choi, W.H.; Lee, M.J. Effects of mTORC1 inhibition on proteasome activity and levels. BMB Rep. 2022, 55, 161–165. [Google Scholar] [CrossRef] [PubMed]
- Nwadike, C.; Williamson, L.E.; Gallagher, L.E.; Guan, J.L.; Chan, E.Y.W. AMPK Inhibits ULK1-Dependent Autophagosome Formation and Lysosomal Acidification via Distinct Mechanisms. Mol. Cell Biol. 2018, 38, e00023-18. [Google Scholar] [CrossRef]
- Yuan, W.; Fang, W.; Zhang, R.; Lyu, H.; Xiao, S.; Guo, D.; Ali, D.W.; Michalak, M.; Chen, X.Z.; Zhou, C.; et al. Therapeutic strategies targeting AMPK-dependent autophagy in cancer cells. Biochim. Biophys. Acta Mol. Cell Res. 2023, 1870, 119537. [Google Scholar] [CrossRef]
- Birgisdottir, A.B.; Mouilleron, S.; Bhujabal, Z.; Wirth, M.; Sjottem, E.; Evjen, G.; Zhang, W.; Lee, R.; O’Reilly, N.; Tooze, S.A.; et al. Members of the autophagy class III phosphatidylinositol 3-kinase complex I interact with GABARAP and GABARAPL1 via LIR motifs. Autophagy 2019, 15, 1333–1355. [Google Scholar] [CrossRef]
- Takahashi, Y.; Coppola, D.; Matsushita, N.; Cualing, H.D.; Sun, M.; Sato, Y.; Liang, C.; Jung, J.U.; Cheng, J.Q.; Mule, J.J.; et al. Bif-1 interacts with Beclin 1 through UVRAG and regulates autophagy and tumorigenesis. Nat. Cell Biol. 2007, 9, 1142–1151. [Google Scholar] [CrossRef] [PubMed]
- Liang, C.; Feng, P.; Ku, B.; Oh, B.H.; Jung, J.U. UVRAG: A new player in autophagy and tumor cell growth. Autophagy 2007, 3, 69–71. [Google Scholar] [CrossRef]
- Fimia, G.M.; Stoykova, A.; Romagnoli, A.; Giunta, L.; Di Bartolomeo, S.; Nardacci, R.; Corazzari, M.; Fuoco, C.; Ucar, A.; Schwartz, P.; et al. Ambra1 regulates autophagy and development of the nervous system. Nature 2007, 447, 1121–1125. [Google Scholar] [CrossRef] [PubMed]
- Yamamuro, T.; Nakamura, S.; Yanagawa, K.; Tokumura, A.; Kawabata, T.; Fukuhara, A.; Teranishi, H.; Hamasaki, M.; Shimomura, I.; Yoshimori, T. Loss of RUBCN/rubicon in adipocytes mediates the upregulation of autophagy to promote the fasting response. Autophagy 2022, 18, 2686–2696. [Google Scholar] [CrossRef]
- Yang, C.; Tao, H.; Zhang, H.; Xia, Y.; Bai, J.; Ge, G.; Li, W.; Zhang, W.; Xiao, L.; Xu, Y.; et al. TET2 regulates osteoclastogenesis by modulating autophagy in OVX-induced bone loss. Autophagy 2022, 18, 2817–2829. [Google Scholar] [CrossRef]
- Polson, H.E.; de Lartigue, J.; Rigden, D.J.; Reedijk, M.; Urbe, S.; Clague, M.J.; Tooze, S.A. Mammalian Atg18 (WIPI2) localizes to omegasome-anchored phagophores and positively regulates LC3 lipidation. Autophagy 2010, 6, 506–522. [Google Scholar] [CrossRef]
- Stromhaug, P.E.; Reggiori, F.; Guan, J.; Wang, C.W.; Klionsky, D.J. Atg21 is a phosphoinositide binding protein required for efficient lipidation and localization of Atg8 during uptake of aminopeptidase I by selective autophagy. Mol. Biol. Cell 2004, 15, 3553–3566. [Google Scholar] [CrossRef]
- Gong, X.; Pan, L. ATG16L1 is equipped with two distinct WIPI2-binding sites to drive autophagy. Autophagy 2024, 20, 938–940. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Zheng, L.; Zheng, X.; Ge, L. Autophagosomal Membrane Origin and Formation. Adv. Exp. Med. Biol. 2021, 1208, 17–42. [Google Scholar] [CrossRef]
- Liu, G.; Pei, F.; Yang, F.; Li, L.; Amin, A.D.; Liu, S.; Buchan, J.R.; Cho, W.C. Role of Autophagy and Apoptosis in Non-Small-Cell Lung Cancer. Int. J. Mol. Sci. 2017, 18, 367. [Google Scholar] [CrossRef]
- Nazio, F.; Bordi, M.; Cianfanelli, V.; Locatelli, F.; Cecconi, F. Autophagy and cancer stem cells: Molecular mechanisms and therapeutic applications. Cell Death Differ. 2019, 26, 690–702. [Google Scholar] [CrossRef]
- Yun, C.W.; Lee, S.H. The Roles of Autophagy in Cancer. Int. J. Mol. Sci. 2018, 19, 3466. [Google Scholar] [CrossRef] [PubMed]
- Delaney, J.R.; Patel, C.B.; Bapat, J.; Jones, C.M.; Ramos-Zapatero, M.; Ortell, K.K.; Tanios, R.; Haghighiabyaneh, M.; Axelrod, J.; DeStefano, J.W.; et al. Autophagy gene haploinsufficiency drives chromosome instability, increases migration, and promotes early ovarian tumors. PLoS Genet. 2020, 16, e1008558. [Google Scholar] [CrossRef]
- Marsh, T.; Kenific, C.M.; Suresh, D.; Gonzalez, H.; Shamir, E.R.; Mei, W.; Tankka, A.; Leidal, A.M.; Kalavacherla, S.; Woo, K.; et al. Autophagic Degradation of NBR1 Restricts Metastatic Outgrowth during Mammary Tumor Progression. Dev. Cell 2020, 52, 591–604.e596. [Google Scholar] [CrossRef] [PubMed]
- Russell, R.C.; Guan, K.L. The multifaceted role of autophagy in cancer. EMBO J. 2022, 41, e110031. [Google Scholar] [CrossRef]
- Biswas, U.; Roy, R.; Ghosh, S.; Chakrabarti, G. The interplay between autophagy and apoptosis: Its implication in lung cancer and therapeutics. Cancer Lett. 2024, 585, 216662. [Google Scholar] [CrossRef]
- Liang, X.H.; Jackson, S.; Seaman, M.; Brown, K.; Kempkes, B.; Hibshoosh, H.; Levine, B. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 1999, 402, 672–676. [Google Scholar] [CrossRef] [PubMed]
- Degenhardt, K.; Mathew, R.; Beaudoin, B.; Bray, K.; Anderson, D.; Chen, G.; Mukherjee, C.; Shi, Y.; Gelinas, C.; Fan, Y.; et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 2006, 10, 51–64. [Google Scholar] [CrossRef]
- Lahiri, A.; Maji, A.; Potdar, P.D.; Singh, N.; Parikh, P.; Bisht, B.; Mukherjee, A.; Paul, M.K. Lung cancer immunotherapy: Progress, pitfalls, and promises. Mol. Cancer 2023, 22, 40. [Google Scholar] [CrossRef]
- Wang, M.; Yu, H.; Wu, R.; Chen, Z.Y.; Hu, Q.; Zhang, Y.F.; Gao, S.H.; Zhou, G.B. Autophagy inhibition enhances the inhibitory effects of ursolic acid on lung cancer cells. Int. J. Mol. Med. 2020, 46, 1816–1826. [Google Scholar] [CrossRef] [PubMed]
- Liao, S.X.; Sun, P.P.; Gu, Y.H.; Rao, X.M.; Zhang, L.Y.; Ou-Yang, Y. Autophagy and pulmonary disease. Ther. Adv. Respir. Dis. 2019, 13, 1753466619890538. [Google Scholar] [CrossRef]
- Guo, J.Y.; Teng, X.; Laddha, S.V.; Ma, S.; Van Nostrand, S.C.; Yang, Y.; Khor, S.; Chan, C.S.; Rabinowitz, J.D.; White, E. Autophagy provides metabolic substrates to maintain energy charge and nucleotide pools in Ras-driven lung cancer cells. Genes Dev. 2016, 30, 1704–1717. [Google Scholar] [CrossRef] [PubMed]
- Xue, Y.; Han, H.; Wu, L.; Pan, B.; Dong, B.; Yin, C.C.; Tian, Z.; Liu, X.; Yang, Y.; Zhang, H.; et al. iASPP facilitates tumor growth by promoting mTOR-dependent autophagy in human non-small-cell lung cancer. Cell Death Dis. 2017, 8, e3150. [Google Scholar] [CrossRef]
- Guo, J.Y.; Karsli-Uzunbas, G.; Mathew, R.; Aisner, S.C.; Kamphorst, J.J.; Strohecker, A.M.; Chen, G.; Price, S.; Lu, W.; Teng, X.; et al. Autophagy suppresses progression of K-ras-induced lung tumors to oncocytomas and maintains lipid homeostasis. Genes Dev. 2013, 27, 1447–1461. [Google Scholar] [CrossRef] [PubMed]
- Amaravadi, R.; Kimmelman, A.C.; White, E. Recent insights into the function of autophagy in cancer. Genes Dev. 2016, 30, 1913–1930. [Google Scholar] [CrossRef]
- Rao, S.; Tortola, L.; Perlot, T.; Wirnsberger, G.; Novatchkova, M.; Nitsch, R.; Sykacek, P.; Frank, L.; Schramek, D.; Komnenovic, V.; et al. A dual role for autophagy in a murine model of lung cancer. Nat. Commun. 2014, 5, 3056. [Google Scholar] [CrossRef]
- Han, T.; Guo, M.; Gan, M.; Yu, B.; Tian, X.; Wang, J.B. TRIM59 regulates autophagy through modulating both the transcription and the ubiquitination of BECN1. Autophagy 2018, 14, 2035–2048. [Google Scholar] [CrossRef] [PubMed]
- Gewirtz, D.A. The Challenge of Developing Autophagy Inhibition as a Therapeutic Strategy. Cancer Res 2016, 76, 5610–5614. [Google Scholar] [CrossRef]
- Wu, H.M.; Shao, L.J.; Jiang, Z.F.; Liu, R.Y. Gemcitabine-Induced Autophagy Protects Human Lung Cancer Cells from Apoptotic Death. Lung 2016, 194, 959–966. [Google Scholar] [CrossRef] [PubMed]
- Wu, T.; Wang, M.C.; Jing, L.; Liu, Z.Y.; Guo, H.; Liu, Y.; Bai, Y.Y.; Cheng, Y.Z.; Nan, K.J.; Liang, X. Autophagy facilitates lung adenocarcinoma resistance to cisplatin treatment by activation of AMPK/mTOR signaling pathway. Drug Des. Devel. Ther. 2015, 9, 6421–6431. [Google Scholar] [CrossRef]
- Liu, Y.; Fu, Y.; Hu, X.; Chen, S.; Miao, J.; Wang, Y.; Zhou, Y.; Zhang, Y. Caveolin-1 knockdown increases the therapeutic sensitivity of lung cancer to cisplatin-induced apoptosis by repressing Parkin-related mitophagy and activating the ROCK1 pathway. J. Cell Physiol. 2020, 235, 1197–1208. [Google Scholar] [CrossRef]
- Chiu, Y.H.; Hsu, S.H.; Hsu, H.W.; Huang, K.C.; Liu, W.; Wu, C.Y.; Huang, W.P.; Chen, J.Y.; Chen, B.H.; Chiu, C.C. Human non-small cell lung cancer cells can be sensitized to camptothecin by modulating autophagy. Int. J. Oncol. 2018, 53, 1967–1979. [Google Scholar] [CrossRef] [PubMed]
- Xie, C.; Zhou, X.; Liang, C.; Li, X.; Ge, M.; Chen, Y.; Yin, J.; Zhu, J.; Zhong, C. Apatinib triggers autophagic and apoptotic cell death via VEGFR2/STAT3/PD-L1 and ROS/Nrf2/p62 signaling in lung cancer. J. Exp. Clin. Cancer Res. 2021, 40, 266. [Google Scholar] [CrossRef]
- Fan, J.; Ren, D.; Wang, J.; Liu, X.; Zhang, H.; Wu, M.; Yang, G. Bruceine D induces lung cancer cell apoptosis and autophagy via the ROS/MAPK signaling pathway in vitro and in vivo. Cell Death Dis. 2020, 11, 126. [Google Scholar] [CrossRef]
- Liu, T.; Li, Y.; Sun, J.; Tian, G.; Shi, Z. Gitogenin suppresses lung cancer progression by inducing apoptosis and autophagy initiation through the activation of AMPK signaling. Int. Immunopharmacol. 2022, 111, 108806. [Google Scholar] [CrossRef]
- Redig, A.J.; McAllister, S.S. Breast cancer as a systemic disease: A view of metastasis. J. Intern. Med. 2013, 274, 113–126. [Google Scholar] [CrossRef]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef]
- Bowman-Colin, C.; Xia, B.; Bunting, S.; Klijn, C.; Drost, R.; Bouwman, P.; Fineman, L.; Chen, X.; Culhane, A.C.; Cai, H.; et al. Palb2 synergizes with Trp53 to suppress mammary tumor formation in a model of inherited breast cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 8632–8637. [Google Scholar] [CrossRef]
- Huo, Y.; Cai, H.; Teplova, I.; Bowman-Colin, C.; Chen, G.; Price, S.; Barnard, N.; Ganesan, S.; Karantza, V.; White, E.; et al. Autophagy opposes p53-mediated tumor barrier to facilitate tumorigenesis in a model of PALB2-associated hereditary breast cancer. Cancer Discov. 2013, 3, 894–907. [Google Scholar] [CrossRef] [PubMed]
- Ross, J.S.; Slodkowska, E.A.; Symmans, W.F.; Pusztai, L.; Ravdin, P.M.; Hortobagyi, G.N. The HER-2 receptor and breast cancer: Ten years of targeted anti-HER-2 therapy and personalized medicine. Oncologist 2009, 14, 320–368. [Google Scholar] [CrossRef] [PubMed]
- Vega-Rubin-de-Celis, S.; Zou, Z.; Fernandez, A.F.; Ci, B.; Kim, M.; Xiao, G.; Xie, Y.; Levine, B. Increased autophagy blocks HER2-mediated breast tumorigenesis. Proc. Natl. Acad. Sci. USA 2018, 115, 4176–4181. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Liu, J.; Li, S.; Feng, Y.; Yi, F.; Wang, L.; Wei, S.; Cao, L. Autophagy-related 7 modulates tumor progression in triple-negative breast cancer. Lab. Investig. 2019, 99, 1266–1274. [Google Scholar] [CrossRef]
- Claude-Taupin, A.; Fonderflick, L.; Gauthier, T.; Mansi, L.; Pallandre, J.R.; Borg, C.; Perez, V.; Monnien, F.; Algros, M.P.; Vigneron, M.; et al. ATG9A Is Overexpressed in Triple Negative Breast Cancer and Its In Vitro Extinction Leads to the Inhibition of Pro-Cancer Phenotypes. Cells 2018, 7, 248. [Google Scholar] [CrossRef]
- Hamurcu, Z.; Delibasi, N.; Gecene, S.; Sener, E.F.; Donmez-Altuntas, H.; Ozkul, Y.; Canatan, H.; Ozpolat, B. Targeting LC3 and Beclin-1 autophagy genes suppresses proliferation, survival, migration and invasion by inhibition of Cyclin-D1 and uPAR/Integrin beta1/ Src signaling in triple negative breast cancer cells. J. Cancer Res. Clin. Oncol. 2018, 144, 415–430. [Google Scholar] [CrossRef]
- Maycotte, P.; Gearheart, C.M.; Barnard, R.; Aryal, S.; Mulcahy Levy, J.M.; Fosmire, S.P.; Hansen, R.J.; Morgan, M.J.; Porter, C.C.; Gustafson, D.L.; et al. STAT3-mediated autophagy dependence identifies subtypes of breast cancer where autophagy inhibition can be efficacious. Cancer Res. 2014, 74, 2579–2590. [Google Scholar] [CrossRef]
- Adamczyk, A.; Kruczak, A.; Harazin-Lechowska, A.; Ambicka, A.; Grela-Wojewoda, A.; Domagala-Haduch, M.; Janecka-Widla, A.; Majchrzyk, K.; Cichocka, A.; Rys, J.; et al. Relationship between HER2 gene status and selected potential biological features related to trastuzumab resistance and its influence on survival of breast cancer patients undergoing trastuzumab adjuvant treatment. OncoTargets Ther. 2018, 11, 4525–4535. [Google Scholar] [CrossRef]
- White, C.; Alshaker, H.; Cooper, C.; Winkler, M.; Pchejetski, D. The emerging role of FTY720 (Fingolimod) in cancer treatment. Oncotarget 2016, 7, 23106–23127. [Google Scholar] [CrossRef]
- Chung, W.P.; Huang, W.L.; Liao, W.A.; Hung, C.H.; Chiang, C.W.; Cheung, C.H.A.; Su, W.C. FTY720 in resistant human epidermal growth factor receptor 2-positive breast cancer. Sci. Rep. 2022, 12, 241. [Google Scholar] [CrossRef]
- Bjorkoy, G.; Lamark, T.; Brech, A.; Outzen, H.; Perander, M.; Overvatn, A.; Stenmark, H.; Johansen, T. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J. Cell Biol. 2005, 171, 603–614. [Google Scholar] [CrossRef] [PubMed]
- Kataoka, Y.; Mukohara, T.; Shimada, H.; Saijo, N.; Hirai, M.; Minami, H. Association between gain-of-function mutations in PIK3CA and resistance to HER2-targeted agents in HER2-amplified breast cancer cell lines. Ann. Oncol. 2010, 21, 255–262. [Google Scholar] [CrossRef] [PubMed]
- Liu, B.; Qi, X.; Zhang, X.; Gao, D.; Fang, K.; Guo, Z.; Li, L. Med19 is involved in chemoresistance by mediating autophagy through HMGB1 in breast cancer. J. Cell. Biochem. 2019, 120, 507–518. [Google Scholar] [CrossRef]
- Kang, R.; Livesey, K.M.; Zeh, H.J.; Loze, M.T.; Tang, D. HMGB1: A novel Beclin 1-binding protein active in autophagy. Autophagy 2010, 6, 1209–1211. [Google Scholar] [CrossRef] [PubMed]
- Ke, S.; Zhou, F.; Yang, H.; Wei, Y.; Gong, J.; Mei, Z.; Wu, L.; Yu, H.; Zhou, Y. Downregulation of high mobility group box 1 modulates telomere homeostasis and increases the radiosensitivity of human breast cancer cells. Int. J. Oncol. 2015, 46, 1051–1058. [Google Scholar] [CrossRef]
- Ladoire, S.; Enot, D.; Senovilla, L.; Ghiringhelli, F.; Poirier-Colame, V.; Chaba, K.; Semeraro, M.; Chaix, M.; Penault-Llorca, F.; Arnould, L.; et al. The presence of LC3B puncta and HMGB1 expression in malignant cells correlate with the immune infiltrate in breast cancer. Autophagy 2016, 12, 864–875. [Google Scholar] [CrossRef]
- Luo, J.; Chen, J.; He, L. mir-129-5p Attenuates Irradiation-Induced Autophagy and Decreases Radioresistance of Breast Cancer Cells by Targeting HMGB1. Med. Sci. Monit. 2015, 21, 4122–4129. [Google Scholar] [CrossRef]
- Chittaranjan, S.; Bortnik, S.; Dragowska, W.H.; Xu, J.; Abeysundara, N.; Leung, A.; Go, N.E.; DeVorkin, L.; Weppler, S.A.; Gelmon, K.; et al. Autophagy inhibition augments the anticancer effects of epirubicin treatment in anthracycline-sensitive and -resistant triple-negative breast cancer. Clin. Cancer Res. 2014, 20, 3159–3173. [Google Scholar] [CrossRef]
- Wang, J.; Fan, P.; Shen, P.; Fan, C.; Zhao, P.; Yao, S.; Dong, K.; Ling, R.; Chen, S.; Zhang, J. XBP1s activates METTL3/METTL14 for ER-phagy and paclitaxel sensitivity regulation in breast cancer. Cancer Lett. 2024, 596, 216846. [Google Scholar] [CrossRef] [PubMed]
- Chipurupalli, S.; Ganesan, R.; Martini, G.; Mele, L.; Reggio, A.; Esposito, M.; Kannan, E.; Namasivayam, V.; Grumati, P.; Desiderio, V.; et al. Cancer cells adapt FAM134B/BiP mediated ER-phagy to survive hypoxic stress. Cell Death Dis. 2022, 13, 357. [Google Scholar] [CrossRef]
- Huang, P.Y.; Liang, S.Y.; Xiang, Y.; Li, M.R.; Wang, M.R.; Liu, L.H. Endoplasmic Reticulum-Targeting Self-Assembly Nanosheets Promote Autophagy and Regulate Immunosuppressive Tumor Microenvironment for Efficient Photodynamic Immunotherapy. Small 2024, 20, e2311056. [Google Scholar] [CrossRef] [PubMed]
- Chan, E.Y.; Longatti, A.; McKnight, N.C.; Tooze, S.A. Kinase-inactivated ULK proteins inhibit autophagy via their conserved C-terminal domains using an Atg13-independent mechanism. Mol. Cell Biol. 2009, 29, 157–171. [Google Scholar] [CrossRef]
- Petherick, K.J.; Conway, O.J.; Mpamhanga, C.; Osborne, S.A.; Kamal, A.; Saxty, B.; Ganley, I.G. Pharmacological inhibition of ULK1 kinase blocks mammalian target of rapamycin (mTOR)-dependent autophagy. J. Biol. Chem. 2015, 290, 11376–11383. [Google Scholar] [CrossRef]
- Zhang, L.; Ouyang, L.; Guo, Y.; Zhang, J.; Liu, B. UNC-51-like Kinase 1: From an Autophagic Initiator to Multifunctional Drug Target. J. Med. Chem. 2018, 61, 6491–6500. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.; Zhang, D.; Li, J.; Deng, X.; Liang, G.; Long, Y.; He, X.; Dai, T.; Ren, D. MACC1 induces autophagy to regulate proliferation, apoptosis, migration and invasion of squamous cell carcinoma. Oncol. Rep. 2017, 38, 2369–2377. [Google Scholar] [CrossRef] [PubMed]
- Saxena, R.; Klochkova, A.; Murray, M.G.; Kabir, M.F.; Samad, S.; Beccari, T.; Gang, J.; Patel, K.; Hamilton, K.E.; Whelan, K.A. Roles for Autophagy in Esophageal Carcinogenesis: Implications for Improving Patient Outcomes. Cancers 2019, 11, 1697. [Google Scholar] [CrossRef]
- Chen, Y.; Li, X.; Wu, X.; He, C.; Guo, L.; Zhang, S.; Xiao, Y.; Guo, W.; Tan, B. Autophagy-related proteins LC3 and Beclin-1 impact the efficacy of chemoradiation on esophageal squamous cell carcinoma. Pathol. Res. Pract. 2013, 209, 562–567. [Google Scholar] [CrossRef]
- Hao, C.L.; Li, Y.; Yang, H.X.; Luo, R.Z.; Zhang, Y.; Zhang, M.F.; Cheng, Y.F.; Wang, X. High level of microtubule-associated protein light chain 3 predicts poor prognosis in resectable esophageal squamous cell carcinoma. Int. J. Clin. Exp. Pathol. 2014, 7, 4213–4221. [Google Scholar]
- Whelan, K.A.; Chandramouleeswaran, P.M.; Tanaka, K.; Natsuizaka, M.; Guha, M.; Srinivasan, S.; Darling, D.S.; Kita, Y.; Natsugoe, S.; Winkler, J.D.; et al. Autophagy supports generation of cells with high CD44 expression via modulation of oxidative stress and Parkin-mediated mitochondrial clearance. Oncogene 2017, 36, 4843–4858. [Google Scholar] [CrossRef]
- Sakurai, T.; Okumura, H.; Matsumoto, M.; Uchikado, Y.; Setoyama, T.; Omoto, I.; Owaki, T.; Maemura, K.; Ishigami, S.; Natsugoe, S. The expression of LC-3 is related to tumor suppression through angiogenesis in esophageal cancer. Med. Oncol. 2013, 30, 701. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Yu, L.; Zhou, J.; Ren, J.; Wang, S.; Tan, Y.; Ding, Y. Inhibition of p62 and/or NFE2L2 induced autophagy impaires esophageal squamous cell cancer metastasis by reversing EMT. Gene 2023, 858, 147194. [Google Scholar] [CrossRef] [PubMed]
- Weh, K.M.; Howell, A.B.; Kresty, L.A. Expression, modulation, and clinical correlates of the autophagy protein Beclin-1 in esophageal adenocarcinoma. Mol. Carcinog. 2016, 55, 1876–1885. [Google Scholar] [CrossRef]
- Zhang, J.; Dong, W. Lentiviral-Mediated Beclin-1 Overexpression Inhibits Cell Proliferation and Induces Autophagy of Human Esophageal Carcinoma Eca109 Cell Xenograft in Nude Mice. Recent Pat. Anticancer Drug Discov. 2020, 15, 70–77. [Google Scholar] [CrossRef] [PubMed]
- Galluzzi, L.; Pietrocola, F.; Bravo-San Pedro, J.M.; Amaravadi, R.K.; Baehrecke, E.H.; Cecconi, F.; Codogno, P.; Debnath, J.; Gewirtz, D.A.; Karantza, V.; et al. Autophagy in malignant transformation and cancer progression. EMBO J. 2015, 34, 856–880. [Google Scholar] [CrossRef]
- Panda, P.K.; Mukhopadhyay, S.; Das, D.N.; Sinha, N.; Naik, P.P.; Bhutia, S.K. Mechanism of autophagic regulation in carcinogenesis and cancer therapeutics. Semin. Cell Dev. Biol. 2015, 39, 43–55. [Google Scholar] [CrossRef]
- Cho, D.H.; Jo, Y.K.; Kim, S.C.; Park, I.J.; Kim, J.C. Down-regulated expression of ATG5 in colorectal cancer. Anticancer Res. 2012, 32, 4091–4096. [Google Scholar]
- Ahn, C.H.; Jeong, E.G.; Lee, J.W.; Kim, M.S.; Kim, S.H.; Kim, S.S.; Yoo, N.J.; Lee, S.H. Expression of beclin-1, an autophagy-related protein, in gastric and colorectal cancers. APMIS 2007, 115, 1344–1349. [Google Scholar] [CrossRef]
- Zhang, M.Y.; Wang, L.Y.; Zhao, S.; Guo, X.C.; Xu, Y.Q.; Zheng, Z.H.; Lu, H.; Zheng, H.C. Effects of Beclin 1 overexpression on aggressive phenotypes of colon cancer cells. Oncol. Lett. 2019, 17, 2441–2450. [Google Scholar] [CrossRef]
- Park, J.M.; Huang, S.; Wu, T.T.; Foster, N.R.; Sinicrope, F.A. Prognostic impact of Beclin 1, p62/sequestosome 1 and LC3 protein expression in colon carcinomas from patients receiving 5-fluorouracil as adjuvant chemotherapy. Cancer Biol. Ther. 2013, 14, 100–107. [Google Scholar] [CrossRef]
- Jo, Y.K.; Kim, S.C.; Park, I.J.; Park, S.J.; Jin, D.H.; Hong, S.W.; Cho, D.H.; Kim, J.C. Increased expression of ATG10 in colorectal cancer is associated with lymphovascular invasion and lymph node metastasis. PLoS ONE 2012, 7, e52705. [Google Scholar] [CrossRef] [PubMed]
- Sakitani, K.; Hirata, Y.; Hikiba, Y.; Hayakawa, Y.; Ihara, S.; Suzuki, H.; Suzuki, N.; Serizawa, T.; Kinoshita, H.; Sakamoto, K.; et al. Inhibition of autophagy exerts anti-colon cancer effects via apoptosis induced by p53 activation and ER stress. BMC Cancer 2015, 15, 795. [Google Scholar] [CrossRef] [PubMed]
- Xiong, L.; Liu, Z.; Ouyang, G.; Lin, L.; Huang, H.; Kang, H.; Chen, W.; Miao, X.; Wen, Y. Autophagy inhibition enhances photocytotoxicity of Photosan-II in human colorectal cancer cells. Oncotarget 2017, 8, 6419–6432. [Google Scholar] [CrossRef] [PubMed]
- Fesler, A.; Liu, H.; Wu, N.; Liu, F.; Ling, P.; Ju, J. Autophagy regulated by miRNAs in colorectal cancer progression and resistance. Cancer Transl. Med. 2017, 3, 96–100. [Google Scholar] [CrossRef]
- Long, J.; He, Q.; Yin, Y.; Lei, X.; Li, Z.; Zhu, W. The effect of miRNA and autophagy on colorectal cancer. Cell Prolif. 2020, 53, e12900. [Google Scholar] [CrossRef]
- Zhang, W.; Yuan, W.; Song, J.; Wang, S.; Gu, X. LncRNA CPS1-IT1 suppresses EMT and metastasis of colorectal cancer by inhibiting hypoxia-induced autophagy through inactivation of HIF-1alpha. Biochimie 2018, 144, 21–27. [Google Scholar] [CrossRef]
- Gustavsson, B.; Carlsson, G.; Machover, D.; Petrelli, N.; Roth, A.; Schmoll, H.J.; Tveit, K.M.; Gibson, F. A review of the evolution of systemic chemotherapy in the management of colorectal cancer. Clin. Color. Cancer 2015, 14, 1–10. [Google Scholar] [CrossRef]
- Becouarn, Y.; Ychou, M.; Ducreux, M.; Borel, C.; Bertheault-Cvitkovic, F.; Seitz, J.F.; Nasca, S.; Nguyen, T.D.; Paillot, B.; Raoul, J.L.; et al. Phase II trial of oxaliplatin as first-line chemotherapy in metastatic colorectal cancer patients. Digestive Group of French Federation of Cancer Centers. J. Clin. Oncol. 1998, 16, 2739–2744. [Google Scholar] [CrossRef]
- Meyerhardt, J.A.; Mayer, R.J. Systemic therapy for colorectal cancer. N. Engl. J. Med. 2005, 352, 476–487. [Google Scholar] [CrossRef]
- Zhang, H.; Tang, J.; Li, C.; Kong, J.; Wang, J.; Wu, Y.; Xu, E.; Lai, M. MiR-22 regulates 5-FU sensitivity by inhibiting autophagy and promoting apoptosis in colorectal cancer cells. Cancer Lett. 2015, 356, 781–790. [Google Scholar] [CrossRef] [PubMed]
- Han, J.; Li, J.; Tang, K.; Zhang, H.; Guo, B.; Hou, N.; Huang, C. miR-338-3p confers 5-fluorouracil resistance in p53 mutant colon cancer cells by targeting the mammalian target of rapamycin. Exp. Cell Res. 2017, 360, 328–336. [Google Scholar] [CrossRef] [PubMed]
- Park, Y.L.; Kim, H.P.; Cho, Y.W.; Min, D.W.; Cheon, S.K.; Lim, Y.J.; Song, S.H.; Kim, S.J.; Han, S.W.; Park, K.J.; et al. Activation of WNT/beta-catenin signaling results in resistance to a dual PI3K/mTOR inhibitor in colorectal cancer cells harboring PIK3CA mutations. Int. J. Cancer 2019, 144, 389–401. [Google Scholar] [CrossRef] [PubMed]
- Yu, X.; Shi, W.; Zhang, Y.; Wang, X.; Sun, S.; Song, Z.; Liu, M.; Zeng, Q.; Cui, S.; Qu, X. CXCL12/CXCR4 axis induced miR-125b promotes invasion and confers 5-fluorouracil resistance through enhancing autophagy in colorectal cancer. Sci. Rep. 2017, 7, 42226. [Google Scholar] [CrossRef]
- Tan, S.; Shi, H.; Ba, M.; Lin, S.; Tang, H.; Zeng, X.; Zhang, X. miR-409-3p sensitizes colon cancer cells to oxaliplatin by inhibiting Beclin-1-mediated autophagy. Int. J. Mol. Med. 2016, 37, 1030–1038. [Google Scholar] [CrossRef]
- Zheng, S.; Zhong, Y.F.; Tan, D.M.; Xu, Y.; Chen, H.X.; Wang, D. miR-183-5p enhances the radioresistance of colorectal cancer by directly targeting ATG5. J. Biosci. 2019, 44, 1–11. [Google Scholar] [CrossRef]
- Kim, J.; Fiesel, F.C.; Belmonte, K.C.; Hudec, R.; Wang, W.X.; Kim, C.; Nelson, P.T.; Springer, W.; Kim, J. miR-27a and miR-27b regulate autophagic clearance of damaged mitochondria by targeting PTEN-induced putative kinase 1 (PINK1). Mol. Neurodegener. 2016, 11, 55. [Google Scholar] [CrossRef]
- Che, J.; Wang, W.; Huang, Y.; Zhang, L.; Zhao, J.; Zhang, P.; Yuan, X. miR-20a inhibits hypoxia-induced autophagy by targeting ATG5/FIP200 in colorectal cancer. Mol. Carcinog. 2019, 58, 1234–1247. [Google Scholar] [CrossRef]
- Taniguchi, K.; Sugito, N.; Kumazaki, M.; Shinohara, H.; Yamada, N.; Nakagawa, Y.; Ito, Y.; Otsuki, Y.; Uno, B.; Uchiyama, K.; et al. MicroRNA-124 inhibits cancer cell growth through PTB1/PKM1/PKM2 feedback cascade in colorectal cancer. Cancer Lett. 2015, 363, 17–27. [Google Scholar] [CrossRef]
- Shan, T.D.; Xu, J.H.; Yu, T.; Li, J.Y.; Zhao, L.N.; Ouyang, H.; Luo, S.; Lu, X.J.; Huang, C.Z.; Lan, Q.S.; et al. Knockdown of linc-POU3F3 suppresses the proliferation, apoptosis, and migration resistance of colorectal cancer. Oncotarget 2016, 7, 961–975. [Google Scholar] [CrossRef]
- Wu, J.; Meng, X.; Gao, R.; Jia, Y.; Chai, J.; Zhou, Y.; Wang, J.; Xue, X.; Dang, T. Long non-coding RNA LINC00858 inhibits colon cancer cell apoptosis, autophagy, and senescence by activating WNK2 promoter methylation. Exp. Cell Res. 2020, 396, 112214. [Google Scholar] [CrossRef] [PubMed]
- Wang, M.; Han, D.; Yuan, Z.; Hu, H.; Zhao, Z.; Yang, R.; Jin, Y.; Zou, C.; Chen, Y.; Wang, G.; et al. Long non-coding RNA H19 confers 5-Fu resistance in colorectal cancer by promoting SIRT1-mediated autophagy. Cell Death Dis. 2018, 9, 1149. [Google Scholar] [CrossRef]
- Liu, Y.; Chen, X.; Chen, X.; Liu, J.; Gu, H.; Fan, R.; Ge, H. Long non-coding RNA HOTAIR knockdown enhances radiosensitivity through regulating microRNA-93/ATG12 axis in colorectal cancer. Cell Death Dis. 2020, 11, 175. [Google Scholar] [CrossRef] [PubMed]
- Xia, C.; Li, Q.; Cheng, X.; Wu, T.; Gao, P.; Gu, Y. Insulin-like growth factor 2 mRNA-binding protein 2-stabilized long non-coding RNA Taurine up-regulated gene 1 (TUG1) promotes cisplatin-resistance of colorectal cancer via modulating autophagy. Bioengineered 2022, 13, 2450–2469. [Google Scholar] [CrossRef] [PubMed]
- Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
- Swami, U.; McFarland, T.R.; Nussenzveig, R.; Agarwal, N. Advanced Prostate Cancer: Treatment Advances and Future Directions. Trends Cancer 2020, 6, 702–715. [Google Scholar] [CrossRef]
- Ingrosso, G.; Detti, B.; Scartoni, D.; Lancia, A.; Giacomelli, I.; Baki, M.; Carta, G.; Livi, L.; Santoni, R. Current therapeutic options in metastatic castration-resistant prostate cancer. Semin. Oncol. 2018, 45, 303–315. [Google Scholar] [CrossRef]
- Lorenc, T.; Klimczyk, K.; Michalczewska, I.; Slomka, M.; Kubiak-Tomaszewska, G.; Olejarz, W. Exosomes in Prostate Cancer Diagnosis, Prognosis and Therapy. Int. J. Mol. Sci. 2020, 21, 2118. [Google Scholar] [CrossRef]
- Kretschmer, A.; Tilki, D. Biomarkers in prostate cancer—Current clinical utility and future perspectives. Crit. Rev. Oncol. Hematol. 2017, 120, 180–193. [Google Scholar] [CrossRef]
- Turner, L.S.; Cheng, J.C.; Beckham, T.H.; Keane, T.E.; Norris, J.S.; Liu, X. Autophagy is increased in prostate cancer cells overexpressing acid ceramidase and enhances resistance to C6 ceramide. Prostate Cancer Prostatic Dis. 2011, 14, 30–37. [Google Scholar] [CrossRef]
- Zhang, B.; Liu, L. Autophagy is a double-edged sword in the therapy of colorectal cancer. Oncol. Lett. 2021, 21, 378. [Google Scholar] [CrossRef] [PubMed]
- Aita, V.M.; Liang, X.H.; Murty, V.V.; Pincus, D.L.; Yu, W.; Cayanis, E.; Kalachikov, S.; Gilliam, T.C.; Levine, B. Cloning and genomic organization of beclin 1, a candidate tumor suppressor gene on chromosome 17q21. Genomics 1999, 59, 59–65. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Jiang, X.; Liu, D.; Na, Y.; Gao, G.F.; Xi, Z. Autophagy protects LNCaP cells under androgen deprivation conditions. Autophagy 2008, 4, 54–60. [Google Scholar] [CrossRef] [PubMed]
- Farrow, J.M.; Yang, J.C.; Evans, C.P. Autophagy as a modulator and target in prostate cancer. Nat. Rev. Urol. 2014, 11, 508–516. [Google Scholar] [CrossRef]
- Ziparo, E.; Petrungaro, S.; Marini, E.S.; Starace, D.; Conti, S.; Facchiano, A.; Filippini, A.; Giampietri, C. Autophagy in prostate cancer and androgen suppression therapy. Int. J. Mol. Sci. 2013, 14, 12090–12106. [Google Scholar] [CrossRef]
- Roudsari, N.M.; Lashgari, N.A.; Momtaz, S.; Abaft, S.; Jamali, F.; Safaiepour, P.; Narimisa, K.; Jackson, G.; Bishayee, A.; Rezaei, N.; et al. Inhibitors of the PI3K/Akt/mTOR Pathway in Prostate Cancer Chemoprevention and Intervention. Pharmaceutics 2021, 13, 1195. [Google Scholar] [CrossRef]
- Ghosh, P.M.; Malik, S.N.; Bedolla, R.G.; Wang, Y.; Mikhailova, M.; Prihoda, T.J.; Troyer, D.A.; Kreisberg, J.I. Signal transduction pathways in androgen-dependent and -independent prostate cancer cell proliferation. Endocr. Relat. Cancer 2005, 12, 119–134. [Google Scholar] [CrossRef]
- Wang, Y.; Mikhailova, M.; Bose, S.; Pan, C.X.; deVere White, R.W.; Ghosh, P.M. Regulation of androgen receptor transcriptional activity by rapamycin in prostate cancer cell proliferation and survival. Oncogene 2008, 27, 7106–7117. [Google Scholar] [CrossRef]
- Miyazawa, Y.; Sekine, Y.; Oka, D.; Nakazawa, S.; Tsuji, Y.; Nakayama, H.; Suzuki, K. Simvastatin Induces Autophagy and Inhibits Proliferation in Prostate Cancer Cells. Anticancer Res. 2023, 43, 5377–5386. [Google Scholar] [CrossRef]
- George, D.J.; Halabi, S.; Healy, P.; Jonasch, D.; Anand, M.; Rasmussen, J.; Wood, S.Y.; Spritzer, C.; Madden, J.F.; Armstrong, A.J. Phase 2 clinical trial of TORC1 inhibition with everolimus in men with metastatic castration-resistant prostate cancer. Urol. Oncol. 2020, 38, 79.e15–79.e22. [Google Scholar] [CrossRef]
- Templeton, A.J.; Dutoit, V.; Cathomas, R.; Rothermundt, C.; Bartschi, D.; Droge, C.; Gautschi, O.; Borner, M.; Fechter, E.; Stenner, F.; et al. Phase 2 trial of single-agent everolimus in chemotherapy-naive patients with castration-resistant prostate cancer (SAKK 08/08). Eur. Urol. 2013, 64, 150–158. [Google Scholar] [CrossRef] [PubMed]
- Rathkopf, D.E.; Larson, S.M.; Anand, A.; Morris, M.J.; Slovin, S.F.; Shaffer, D.R.; Heller, G.; Carver, B.; Rosen, N.; Scher, H.I. Everolimus combined with gefitinib in patients with metastatic castration-resistant prostate cancer: Phase 1/2 results and signaling pathway implications. Cancer 2015, 121, 3853–3861. [Google Scholar] [CrossRef]
- American Diabetes Association. 9. Pharmacologic Approaches to Glycemic Treatment: Standards of Medical Care in Diabetes-2021. Diabetes Care 2021, 44, S111–S124. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.; Wang, H.; Geng, X.; Zhang, D.; Zhu, Z.; Zhang, G.; Hou, J. Metformin exerts anti-AR-negative prostate cancer activity via AMPK/autophagy signaling pathway. Cancer Cell Int. 2021, 21, 404. [Google Scholar] [CrossRef] [PubMed]
- Xia, Q.; Sung, J.; Chowdhury, W.; Chen, C.L.; Hoti, N.; Shabbeer, S.; Carducci, M.; Rodriguez, R. Chronic administration of valproic acid inhibits prostate cancer cell growth in vitro and in vivo. Cancer Res. 2006, 66, 7237–7244. [Google Scholar] [CrossRef]
- Lin, J.F.; Tsai, T.F.; Liao, P.C.; Lin, Y.H.; Lin, Y.C.; Chen, H.E.; Chou, K.Y.; Hwang, T.I. Benzyl isothiocyanate induces protective autophagy in human prostate cancer cells via inhibition of mTOR signaling. Carcinogenesis 2013, 34, 406–414. [Google Scholar] [CrossRef]
- Ertmer, A.; Huber, V.; Gilch, S.; Yoshimori, T.; Erfle, V.; Duyster, J.; Elsasser, H.P.; Schatzl, H.M. The anticancer drug imatinib induces cellular autophagy. Leukemia 2007, 21, 936–942. [Google Scholar] [CrossRef]
- Altman, B.J.; Jacobs, S.R.; Mason, E.F.; Michalek, R.D.; MacIntyre, A.N.; Coloff, J.L.; Ilkayeva, O.; Jia, W.; He, Y.W.; Rathmell, J.C. Autophagy is essential to suppress cell stress and to allow BCR-Abl-mediated leukemogenesis. Oncogene 2011, 30, 1855–1867. [Google Scholar] [CrossRef]
- Mortensen, M.; Soilleux, E.J.; Djordjevic, G.; Tripp, R.; Lutteropp, M.; Sadighi-Akha, E.; Stranks, A.J.; Glanville, J.; Knight, S.; Jacobsen, S.E.; et al. The autophagy protein Atg7 is essential for hematopoietic stem cell maintenance. J. Exp. Med. 2011, 208, 455–467. [Google Scholar] [CrossRef]
- Bellodi, C.; Lidonnici, M.R.; Hamilton, A.; Helgason, G.V.; Soliera, A.R.; Ronchetti, M.; Galavotti, S.; Young, K.W.; Selmi, T.; Yacobi, R.; et al. Targeting autophagy potentiates tyrosine kinase inhibitor-induced cell death in Philadelphia chromosome-positive cells, including primary CML stem cells. J. Clin. Investig. 2009, 119, 1109–1123. [Google Scholar] [CrossRef]
- Liang, C.; Qiao, G.; Liu, Y.; Tian, L.; Hui, N.; Li, J.; Ma, Y.; Li, H.; Zhao, Q.; Cao, W.; et al. Overview of all-trans-retinoic acid (ATRA) and its analogues: Structures, activities, and mechanisms in acute promyelocytic leukaemia. Eur. J. Med. Chem. 2021, 220, 113451. [Google Scholar] [CrossRef]
- Pan, T.; Kondo, S.; Zhu, W.; Xie, W.; Jankovic, J.; Le, W. Neuroprotection of rapamycin in lactacystin-induced neurodegeneration via autophagy enhancement. Neurobiol. Dis. 2008, 32, 16–25. [Google Scholar] [CrossRef] [PubMed]
- Wen, W.; Ertas, Y.N.; Erdem, A.; Zhang, Y. Dysregulation of autophagy in gastric carcinoma: Pathways to tumor progression and resistance to therapy. Cancer Lett. 2024, 591, 216857. [Google Scholar] [CrossRef] [PubMed]
- Yu, X.; Feng, M.; Guo, J.; Wang, H.; Yu, J.; Zhang, A.; Wu, J.; Han, Y.; Sun, Z.; Liao, Y.; et al. MLKL promotes hepatocarcinogenesis through inhibition of AMPK-mediated autophagy. Cell Death Differ. 2024, 31, 1085–1098. [Google Scholar] [CrossRef]
- Wang, M.; Qiu, S.; Qin, J. Baicalein induced apoptosis and autophagy of undifferentiated thyroid cancer cells by the ERK/PI3K/Akt pathway. Am. J. Transl. Res. 2019, 11, 3341–3352. [Google Scholar] [PubMed]
- Huang, H.; Han, Q.; Zheng, H.; Liu, M.; Shi, S.; Zhang, T.; Yang, X.; Li, Z.; Xu, Q.; Guo, H.; et al. MAP4K4 mediates the SOX6-induced autophagy and reduces the chemosensitivity of cervical cancer. Cell Death Dis. 2021, 13, 13. [Google Scholar] [CrossRef]
- Orfanelli, T.; Jeong, J.M.; Doulaveris, G.; Holcomb, K.; Witkin, S.S. Involvement of autophagy in cervical, endometrial and ovarian cancer. Int. J. Cancer 2014, 135, 519–528. [Google Scholar] [CrossRef]
- Wu, J.; Guo, Q.; Li, J.; Yuan, H.; Xiao, C.; Qiu, J.; Wu, Q.; Wang, D. Loperamide induces protective autophagy and apoptosis through the ROS/JNK signaling pathway in bladder cancer. Biochem. Pharmacol. 2023, 218, 115870. [Google Scholar] [CrossRef]
- Bai, Y.; Liu, X.; Qi, X.; Liu, X.; Peng, F.; Li, H.; Fu, H.; Pei, S.; Chen, L.; Chi, X.; et al. PDIA6 modulates apoptosis and autophagy of non-small cell lung cancer cells via the MAP4K1/JNK signaling pathway. eBioMedicine 2019, 42, 311–325. [Google Scholar] [CrossRef]
- Baquero, P.; Dawson, A.; Helgason, G.V. Autophagy and mitochondrial metabolism: Insights into their role and therapeutic potential in chronic myeloid leukaemia. FEBS J. 2019, 286, 1271–1283. [Google Scholar] [CrossRef]
- Mao, K.; Breen, P.; Ruvkun, G. The Caenorhabditis elegans ARIP-4 DNA helicase couples mitochondrial surveillance to immune, detoxification, and antiviral pathways. Proc. Natl. Acad. Sci. USA 2022, 119, e2215966119. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Wu, W.K.K.; Gao, J.; Li, Z.; Dong, B.; Lin, X.; Li, Y.; Li, Y.; Gong, J.; Qi, C.; et al. Autophagy inhibition enhances PD-L1 expression in gastric cancer. J. Exp. Clin. Cancer Res. 2019, 38, 140. [Google Scholar] [CrossRef] [PubMed]
- Qiu, D.M.; Wang, G.L.; Chen, L.; Xu, Y.Y.; He, S.; Cao, X.L.; Qin, J.; Zhou, J.M.; Zhang, Y.X.; Qun, E. The expression of beclin-1, an autophagic gene, in hepatocellular carcinoma associated with clinical pathological and prognostic significance. BMC Cancer 2014, 14, 327. [Google Scholar] [CrossRef]
- Inami, Y.; Waguri, S.; Sakamoto, A.; Kouno, T.; Nakada, K.; Hino, O.; Watanabe, S.; Ando, J.; Iwadate, M.; Yamamoto, M.; et al. Persistent activation of Nrf2 through p62 in hepatocellular carcinoma cells. J. Cell Biol. 2011, 193, 275–284. [Google Scholar] [CrossRef]
- Ni, H.M.; Woolbright, B.L.; Williams, J.; Copple, B.; Cui, W.; Luyendyk, J.P.; Jaeschke, H.; Ding, W.X. Nrf2 promotes the development of fibrosis and tumorigenesis in mice with defective hepatic autophagy. J. Hepatol. 2014, 61, 617–625. [Google Scholar] [CrossRef] [PubMed]
- Takamura, A.; Komatsu, M.; Hara, T.; Sakamoto, A.; Kishi, C.; Waguri, S.; Eishi, Y.; Hino, O.; Tanaka, K.; Mizushima, N. Autophagy-deficient mice develop multiple liver tumors. Genes Dev. 2011, 25, 795–800. [Google Scholar] [CrossRef]
- Tian, Y.; Kuo, C.F.; Sir, D.; Wang, L.; Govindarajan, S.; Petrovic, L.M.; Ou, J.H. Autophagy inhibits oxidative stress and tumor suppressors to exert its dual effect on hepatocarcinogenesis. Cell Death Differ. 2015, 22, 1025–1034. [Google Scholar] [CrossRef]
- Song, J.; Qu, Z.; Guo, X.; Zhao, Q.; Zhao, X.; Gao, L.; Sun, K.; Shen, F.; Wu, M.; Wei, L. Hypoxia-induced autophagy contributes to the chemoresistance of hepatocellular carcinoma cells. Autophagy 2009, 5, 1131–1144. [Google Scholar] [CrossRef]
- Yang, K.; Zhang, X.; Yang, K.; Liu, S.; Zhang, J.; Fu, Y.; Liu, T.; Wu, K.; Li, J.; Liu, C.; et al. Overexpression of c-Myc triggers p62 aggregation-mediated mitochondrial mitophagy in cabozantinib resistance of hepatocellular carcinoma. Mol. Med. 2025, 31, 209. [Google Scholar] [CrossRef]
- Qin, Y.; Sun, W.; Wang, Z.; Dong, W.; He, L.; Zhang, T.; Lv, C.; Zhang, H. RBM47/SNHG5/FOXO3 axis activates autophagy and inhibits cell proliferation in papillary thyroid carcinoma. Cell Death Dis. 2022, 13, 270. [Google Scholar] [CrossRef]
- Yin, X.; Liu, X.; Gong, H.; Chu, Z. LncRNA STARD7-AS1 suppresses cervical cancer cell proliferation while promoting autophagy by regulating miR-31-5p/TXNIP axis to inactivate the mTOR signaling. J. Gynecol. Oncol. 2024, 35, e97. [Google Scholar] [CrossRef]
- Zhang, T.; Ji, T.; Duan, Z.; Xue, Y. Long non-coding RNA MLLT4 antisense RNA 1 induces autophagy to inhibit tumorigenesis of cervical cancer through modulating the myosin-9/ATG14 axis. Sci. Rep. 2024, 14, 6379. [Google Scholar] [CrossRef] [PubMed]
- Chen, P.; Zhang, X.; Fang, Q.; Zhao, Z.; Lin, C.; Zhou, Y.; Liu, F.; Zhu, C.; Wu, A. Betulinic acid induces apoptosis of HeLa cells via ROS-dependent ER stress and autophagy in vitro and in vivo. J. Nat. Med. 2024, 78, 677–692. [Google Scholar] [CrossRef]
- Shin, M.K.; Payne, S.; Bilger, A.; Matkowskyj, K.A.; Carchman, E.; Meyer, D.S.; Bentires-Alj, M.; Deming, D.A.; Lambert, P.F. Activating Mutations in Pik3ca Contribute to Anal Carcinogenesis in the Presence or Absence of HPV-16 Oncogenes. Clin. Cancer Res. 2019, 25, 1889–1900. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.; Zeng, T.; Zhang, X.; Liu, C.; Wu, Z.; Yao, L.; Xie, C.; Xia, H.; Lin, Q.; Xie, L.; et al. ATR/Chk1 signaling induces autophagy through sumoylated RhoB-mediated lysosomal translocation of TSC2 after DNA damage. Nat. Commun. 2018, 9, 4139. [Google Scholar] [CrossRef] [PubMed]
- Kayacan, S.; Yilancioglu, K.; Akdemir, A.S.; Kaya-Dagistanli, F.; Melikoglu, G.; Ozturk, M. Synergistic Effect of Apigenin and Curcumin on Apoptosis, Paraptosis and Autophagy-related Cell Death in HeLa Cells. Anticancer Res. 2021, 41, 1271–1282. [Google Scholar] [CrossRef]
- Luo, C.L.; Liu, Y.Q.; Wang, P.; Song, C.H.; Wang, K.J.; Dai, L.P.; Zhang, J.Y.; Ye, H. The effect of quercetin nanoparticle on cervical cancer progression by inducing apoptosis, autophagy and anti-proliferation via JAK2 suppression. Biomed. Pharmacother. 2016, 82, 595–605. [Google Scholar] [CrossRef]
- Liu, K.; Chen, H.; Li, Y.; Wang, B.; Li, Q.; Zhang, L.; Liu, X.; Wang, C.; Ertas, Y.N.; Shi, H. Autophagy flux in bladder cancer: Cell death crosstalk, drug and nanotherapeutics. Cancer Lett. 2024, 591, 216867. [Google Scholar] [CrossRef]
- Dong, D.; Yao, Y.; Song, J.; Sun, L.; Zhang, G. Cancer-Associated Fibroblasts Regulate Bladder Cancer Invasion and Metabolic Phenotypes through Autophagy. Dis. Markers 2021, 2021, 6645220. [Google Scholar] [CrossRef]
- Dyshlovoy, S.A.; Madanchi, R.; Hauschild, J.; Otte, K.; Alsdorf, W.H.; Schumacher, U.; Kalinin, V.I.; Silchenko, A.S.; Avilov, S.A.; Honecker, F.; et al. The marine triterpene glycoside frondoside A induces p53-independent apoptosis and inhibits autophagy in urothelial carcinoma cells. BMC Cancer 2017, 17, 93. [Google Scholar] [CrossRef]
- Fan, B.; Zhang, X.; Ma, Y.; Zhang, A. Fangchinoline Induces Apoptosis, Autophagy and Energetic Impairment in Bladder Cancer. Cell. Physiol. Biochem. 2017, 43, 1003–1011. [Google Scholar] [CrossRef] [PubMed]
- Kang, M.; Lee, K.H.; Lee, H.S.; Jeong, C.W.; Kwak, C.; Kim, H.H.; Ku, J.H. Concurrent Autophagy Inhibition Overcomes the Resistance of Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Human Bladder Cancer Cells. Int. J. Mol. Sci. 2017, 18, 321. [Google Scholar] [CrossRef]
- Jobst, M.; Hossain, M.; Kiss, E.; Bergen, J.; Marko, D.; Del Favero, G. Autophagy modulation changes mechano-chemical sensitivity of T24 bladder cancer cells. Biomed. Pharmacother. 2024, 170, 115942. [Google Scholar] [CrossRef]
- Schlutermann, D.; Skowron, M.A.; Berleth, N.; Bohler, P.; Deitersen, J.; Stuhldreier, F.; Wallot-Hieke, N.; Wu, W.; Peter, C.; Hoffmann, M.J.; et al. Targeting urothelial carcinoma cells by combining cisplatin with a specific inhibitor of the autophagy-inducing class III PtdIns3K complex. Urol. Oncol. 2018, 36, 160.e1–160.e13. [Google Scholar] [CrossRef]
- Du, H.; Tao, T.; Xu, S.; Xu, C.; Li, S.; Su, Q.; Yan, J.; Liu, B.; Li, R. 4-Methoxydalbergione Inhibits Bladder Cancer Cell Growth via Inducing Autophagy and Inhibiting Akt/ERK Signaling Pathway. Front. Mol. Biosci. 2021, 8, 789658. [Google Scholar] [CrossRef]
- Anlas, A.A.; Nelson, C.M. Soft Microenvironments Induce Chemoresistance by Increasing Autophagy Downstream of Integrin-Linked Kinase. Cancer Res. 2020, 80, 4103–4113. [Google Scholar] [CrossRef] [PubMed]
- Jin, C.; Wang, T.; Yang, Y.; Zhou, P.; Li, J.; Wu, W.; Lv, X.; Ma, G.; Wang, A. Rational targeting of autophagy in colorectal cancer therapy: From molecular interactions to pharmacological compounds. Environ. Res. 2023, 227, 115721. [Google Scholar] [CrossRef] [PubMed]
- Gong, Y.; Chen, W.; Chen, X.; He, Y.; Jiang, H.; Zhang, X.; Pan, L.; Ni, B.; Yang, F.; Xu, Y.; et al. An Injectable Epigenetic Autophagic Modulatory Hydrogel for Boosting Umbilical Cord Blood NK Cell Therapy Prevents Postsurgical Relapse of Triple-Negative Breast Cancer. Adv. Sci. 2022, 9, e2201271. [Google Scholar] [CrossRef]
- Pellegrini, F.R.; De Martino, S.; Fianco, G.; Ventura, I.; Valente, D.; Fiore, M.; Trisciuoglio, D.; Degrassi, F. Blockage of autophagosome-lysosome fusion through SNAP29 O-GlcNAcylation promotes apoptosis via ROS production. Autophagy 2023, 19, 2078–2093. [Google Scholar] [CrossRef]
- Briceno, E.; Reyes, S.; Sotelo, J. Therapy of glioblastoma multiforme improved by the antimutagenic chloroquine. Neurosurg. Focus 2003, 14, e3. [Google Scholar] [CrossRef]
- Boone, B.A.; Bahary, N.; Zureikat, A.H.; Moser, A.J.; Normolle, D.P.; Wu, W.C.; Singhi, A.D.; Bao, P.; Bartlett, D.L.; Liotta, L.A.; et al. Safety and Biologic Response of Pre-operative Autophagy Inhibition in Combination with Gemcitabine in Patients with Pancreatic Adenocarcinoma. Ann. Surg. Oncol. 2015, 22, 4402–4410. [Google Scholar] [CrossRef]
- Liu, L.Q.; Wang, S.B.; Shao, Y.F.; Shi, J.N.; Wang, W.; Chen, W.Y.; Ye, Z.Q.; Jiang, J.Y.; Fang, Q.X.; Zhang, G.B.; et al. Hydroxychloroquine potentiates the anti-cancer effect of bevacizumab on glioblastoma via the inhibition of autophagy. Biomed. Pharmacother. 2019, 118, 109339. [Google Scholar] [CrossRef] [PubMed]
- Mulcahy Levy, J.M.; Zahedi, S.; Griesinger, A.M.; Morin, A.; Davies, K.D.; Aisner, D.L.; Kleinschmidt-DeMasters, B.K.; Fitzwalter, B.E.; Goodall, M.L.; Thorburn, J.; et al. Autophagy inhibition overcomes multiple mechanisms of resistance to BRAF inhibition in brain tumors. eLife 2017, 6, e19671. [Google Scholar] [CrossRef]
- Cheng, S.M.; Lin, T.Y.; Chang, Y.C.; Lin, I.W.; Leung, E.; Cheung, C.H.A. YM155 and BIRC5 downregulation induce genomic instability via autophagy-mediated ROS production and inhibition in DNA repair. Pharmacol. Res. 2021, 166, 105474. [Google Scholar] [CrossRef] [PubMed]
- Gong, R.; Wan, X.; Jiang, S.; Guan, Y.; Li, Y.; Jiang, T.; Chen, Z.; Zhong, C.; He, L.; Xiang, Z.; et al. GPX4-AUTAC induces ferroptosis in breast cancer by promoting the selective autophagic degradation of GPX4 mediated by TRAF6-p62. Cell Death Differ. 2025. [Google Scholar] [CrossRef] [PubMed]
- Zielke, S.; Kardo, S.; Zein, L.; Mari, M.; Covarrubias-Pinto, A.; Kinzler, M.N.; Meyer, N.; Stolz, A.; Fulda, S.; Reggiori, F.; et al. ATF4 links ER stress with reticulophagy in glioblastoma cells. Autophagy 2021, 17, 2432–2448. [Google Scholar] [CrossRef]
- He, H.; Zhou, C.; Chen, X. ATNC: Versatile Nanobody Chimeras for Autophagic Degradation of Intracellular Unligandable and Undruggable Proteins. J. Am. Chem. Soc. 2023, 145, 24785–24795. [Google Scholar] [CrossRef]
- Guo, J.Y.; Xia, B.; White, E. Autophagy-mediated tumor promotion. Cell 2013, 155, 1216–1219. [Google Scholar] [CrossRef]
- Eskelinen, E.L. The dual role of autophagy in cancer. Curr. Opin. Pharmacol. 2011, 11, 294–300. [Google Scholar] [CrossRef]
- Li, X.; He, S.; Ma, B. Autophagy and autophagy-related proteins in cancer. Mol. Cancer 2020, 19, 12. [Google Scholar] [CrossRef]
- Rosenfeldt, M.T.; Ryan, K.M. The multiple roles of autophagy in cancer. Carcinogenesis 2011, 32, 955–963. [Google Scholar] [CrossRef] [PubMed]
- Singh, S.S.; Vats, S.; Chia, A.Y.; Tan, T.Z.; Deng, S.; Ong, M.S.; Arfuso, F.; Yap, C.T.; Goh, B.C.; Sethi, G.; et al. Dual role of autophagy in hallmarks of cancer. Oncogene 2018, 37, 1142–1158. [Google Scholar] [CrossRef]
- Amaravadi, R.K.; Kimmelman, A.C.; Debnath, J. Targeting Autophagy in Cancer: Recent Advances and Future Directions. Cancer Discov. 2019, 9, 1167–1181. [Google Scholar] [CrossRef] [PubMed]
- Ferreira, P.M.P.; Sousa, R.W.R.; Ferreira, J.R.O.; Militao, G.C.G.; Bezerra, D.P. Chloroquine and hydroxychloroquine in antitumor therapies based on autophagy-related mechanisms. Pharmacol. Res. 2021, 168, 105582. [Google Scholar] [CrossRef]
- Gong, C.; Lin, Q.; Qin, T.; Zeng, Y.; Xu, F.; Yang, Y.; Yin, D.; Duan, Z.; Chen, C.L.; Wing-Cheong Chow, L.; et al. Targeting autophagy plus high-dose CDK4/6 inhibitors in advanced HR+HER2- breast cancer: A phase 1b/2 trial. Med 2025, 6, 100559. [Google Scholar] [CrossRef] [PubMed]
- Xue, W.; Xu, C.; Zhang, K.; Cui, L.; Huang, X.; Nan, Y.; Ju, D.; Chang, X.; Zhang, X. Enhancing antitumor efficacy of CLDN18.2-directed antibody-drug conjugates through autophagy inhibition in gastric cancer. Cell Death Discov. 2024, 10, 393. [Google Scholar] [CrossRef] [PubMed]
- Galluzzi, L.; Bravo-San Pedro, J.M.; Demaria, S.; Formenti, S.C.; Kroemer, G. Activating autophagy to potentiate immunogenic chemotherapy and radiation therapy. Nat. Rev. Clin. Oncol. 2017, 14, 247–258. [Google Scholar] [CrossRef]
- Pua, H.H.; Dzhagalov, I.; Chuck, M.; Mizushima, N.; He, Y.W. A critical role for the autophagy gene Atg5 in T cell survival and proliferation. J. Exp. Med. 2007, 204, 25–31. [Google Scholar] [CrossRef]
- Puleston, D.J.; Zhang, H.; Powell, T.J.; Lipina, E.; Sims, S.; Panse, I.; Watson, A.S.; Cerundolo, V.; Townsend, A.R.; Klenerman, P.; et al. Autophagy is a critical regulator of memory CD8(+) T cell formation. eLife 2014, 3, e03706. [Google Scholar] [CrossRef]
- Galluzzi, L.; Bravo-San Pedro, J.M.; Levine, B.; Green, D.R.; Kroemer, G. Pharmacological modulation of autophagy: Therapeutic potential and persisting obstacles. Nat. Rev. Drug Discov. 2017, 16, 487–511. [Google Scholar] [CrossRef]
- Klionsky, D.J.; Abdelmohsen, K.; Abe, A.; Abedin, M.J.; Abeliovich, H.; Acevedo Arozena, A.; Adachi, H.; Adams, C.M.; Adams, P.D.; Adeli, K.; et al. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy 2016, 12, 1–222. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Song, Y. Proteolysis-targeting chimera (PROTAC) for targeted protein degradation and cancer therapy. J. Hematol. Oncol. 2020, 13, 50. [Google Scholar] [CrossRef] [PubMed]
- Galluzzi, L.; Bravo-San Pedro, J.M.; Blomgren, K.; Kroemer, G. Autophagy in acute brain injury. Nat. Rev. Neurosci. 2016, 17, 467–484. [Google Scholar] [CrossRef]
- Kaizuka, T.; Morishita, H.; Hama, Y.; Tsukamoto, S.; Matsui, T.; Toyota, Y.; Kodama, A.; Ishihara, T.; Mizushima, T.; Mizushima, N. An Autophagic Flux Probe that Releases an Internal Control. Mol. Cell 2016, 64, 835–849. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Wu, Q.; Zhang, L.; Wang, Q.; Yang, Z.; Liu, J.; Feng, L. Caffeic acid reduces A53T alpha-synuclein by activating JNK/Bcl-2-mediated autophagy in vitro and improves behaviour and protects dopaminergic neurons in a mouse model of Parkinson’s disease. Pharmacol. Res. 2019, 150, 104538. [Google Scholar] [CrossRef]
- Chen, M.L.; Hong, C.G.; Yue, T.; Li, H.M.; Duan, R.; Hu, W.B.; Cao, J.; Wang, Z.X.; Chen, C.Y.; Hu, X.K.; et al. Inhibition of miR-331-3p and miR-9-5p ameliorates Alzheimer’s disease by enhancing autophagy. Theranostics 2021, 11, 2395–2409. [Google Scholar] [CrossRef]
- Di Meco, A.; Curtis, M.E.; Lauretti, E.; Pratico, D. Autophagy Dysfunction in Alzheimer’s Disease: Mechanistic Insights and New Therapeutic Opportunities. Biol. Psychiatry 2020, 87, 797–807. [Google Scholar] [CrossRef]
- Zhang, Z.; Yang, X.; Song, Y.Q.; Tu, J. Autophagy in Alzheimer’s disease pathogenesis: Therapeutic potential and future perspectives. Ageing Res. Rev. 2021, 72, 101464. [Google Scholar] [CrossRef]
- Luo, C.; Yang, J. Age- and disease-related autophagy impairment in Huntington disease: New insights from direct neuronal reprogramming. Aging Cell 2024, 23, e14285. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
You, H.; Wang, L.; Meng, H.; Li, J.; Fang, G. Autophagy: Shedding Light on the Mechanisms and Multifaceted Roles in Cancers. Biomolecules 2025, 15, 915. https://doi.org/10.3390/biom15070915
You H, Wang L, Meng H, Li J, Fang G. Autophagy: Shedding Light on the Mechanisms and Multifaceted Roles in Cancers. Biomolecules. 2025; 15(7):915. https://doi.org/10.3390/biom15070915
Chicago/Turabian StyleYou, Hongmei, Ling Wang, Hongwu Meng, Jun Li, and Guoying Fang. 2025. "Autophagy: Shedding Light on the Mechanisms and Multifaceted Roles in Cancers" Biomolecules 15, no. 7: 915. https://doi.org/10.3390/biom15070915
APA StyleYou, H., Wang, L., Meng, H., Li, J., & Fang, G. (2025). Autophagy: Shedding Light on the Mechanisms and Multifaceted Roles in Cancers. Biomolecules, 15(7), 915. https://doi.org/10.3390/biom15070915