Autophagy and cancer metastasis: a Trojan horse =============================================== * Javad Alizadeh * Shahrokh Lorzadeh * Saeid Ghavami * cancer * carcinoma * cell death * cell physiological phenomena Metastasis is one of the most important challenges in cancer therapy strategies.1 Therefore, understanding the mechanisms of metastasis is a powerful weapon to increase the survival of patients with cancer and improve their quality of life. For the first time, Jean Claude used the term ‘metastasis’ as one of the most important hallmarks of cancer in 1829.2 Metastasis, a Greek word, means ‘displacement’ (meta meaning ‘next’ and stasis, ‘placement’).3 This term refers to a general description of migration and invasion of tumor cells from the primary tumor site to secondary sites. Metastasis is considered as one of the key etiologies of cancer-related death; therefore, understanding its mechanism in depth has been always on demand in basic and clinical sciences.4 Epithelial to mesenchymal transition (EMT) is one of the several processes, which is involved in metastasis, and development of drug resistance in cancer.5 During EMT, cells gradually convert from epithelial to a mesenchymal phenotype. This enables cancer cells to be more motile, have less extracellular matrix adhesion and be prone to detachment and moving toward distant organs. Beside metastasis, EMT is involved in embryonic development, wound healing, tissue fibrosis and scar formation.6–8 Macroautophagy (hereafter termed autophagy) is an essential physiologic pathway that is responsible for degradation of damaged organelles, misfolded proteins and pathogens.9 The formation of double membrane vesicles (autophagosomes) to engulf the cytosolic material is the key step in autophagy pathway and is tightly regulated by autophagy-related genes (*ATG*).10 Autophagosomes are fused to lysosomes for digestion via lysosomal degradation enzymes. Autophagy is a double-edged sword in tumor initiation and progression. Loss of autophagy machinery increases the rate of tumor initiation because of the accumulation of damaged mitochondria and accumulation of reactive oxygen species, leading to genomic damage and instability.11 Interestingly, higher activation of autophagy in established tumors helps tumor cells to adapt to metabolic stress and lack of nutrients and support further tumor growth.12 13 There is a complex relation between EMT and autophagy in tumor cells. Several recent investigations have showed that inhibition of EMT in cancer cells induces autophagy, while inhibition of mTOR (autophagy induction) slows down the metastasis of cancer cells. For example, alteronol (protein kinase B (PKB, also known as Akt)/the mechanistic target of rapamycin (mTOR) inhibitor) induces autophagy in melanoma cancer cells and inhibits their metastasis and migration via inhibition of EMT.14 On the other hand, our recent investigation showed that *ATG7* knock down and chemical inhibition of autophagy inhibit EMT and invasiveness of non-small cell lung cancer cells.7 8 Alisertib (inhibitor of phosphatidylinositol 3-kinase (PI3K)/Akt/mTOR) induces autophagy and inhibits metastasis in ovarian cancers cells via inhibition of EMT.15 16 Activation of PI3K/Akt/mTOR pathway induces EMT in several cancer models via activation of EMT transcription factors.17 PI3K/Akt/mTOR pathway is also activated via different growth factors.4 As an example, transforming growth factor beta (TGF-β) is one of the most important factors which activates EMT and is involved in the induction of the PI3K/Akt/mTOR pathway.14 18 19 Inhibition of PI3K/Akt/mTOR pathway can be reversed using specific inhibitors of the pathway and decrease tumor metastasis.15 On the other hand, TGF-β can induce simultaneous autophagy and EMT in several cancer models, while TGF-β-induced autophagy is necessary for its EMT induction.7 8 NF-κB is another factor that is involved in simultaneous regulation of EMT and autophagy.14 20 It has been shown that it induces EMT and increases metastasis via activating EMT-related transcription factors (SNAIL, SLUG, SIP1, and TWIST).20 On the other hand, NF-κB has a dual role in autophagy (inhibition and induction) by different mechanisms14 which can negatively or positively regulate EMT and tumor invasion. Besides NF-κB, p53 is also involved in the regulation of EMT and autophagy. It is a tumor-suppressing factor and plays a crucial role in the regulation of autophagy. Nuclear p53 induces autophagy via increasing the expression of *ATGs* and inhibition of PI3K/Akt/mTOR,21–23 while cytosolic p53 inhibits autophagy via regulation of mTOR activation.24 25 Under normal condition, p53 is localized in the cytoplasm and inhibits autophagy, while in the stress condition, p53 translocates to the nucleus and induces autophagy.26 On the other hand, wild-type p53 inhibits EMT and metastasis via inhibition of transcription factors that are involved in the regulation of EMT.27 Overall, we can conclude that autophagy and EMT crosstalk is a complex mechanism (figure 1), is highly context dependent and can be different in different types of cancers or different stages and grades of cancers. Therefore, it can be concluded that there is a complex crosstalk between autophagy and metastasis via regulation of EMT and targeting autophagy could be a potential mechanism to control cancer metastasis. In the meantime, extensive investigation on the regulation of metastasis via autophagy is needed to develop efficient inhibitors and inducers of autophagy to overcome cancer metastasis via autophagy pathway. ![Figure 1](/https://d3hme472k3gd2d.cloudfront.net/content/jim/early/2021/07/05/jim-2021-002016/F1.medium.gif) [Figure 1](/content/early/2021/07/05/jim-2021-002016/F1) Figure 1 Complex crosstalk between autophagy and EMT during metastasis. Autophagy regulates cancer metastasis via EMT with different mechanisms. Several different regulators may target autophagy and indirectly affect EMT. These include p53 (cytosolic and nuclear), growth factors and cytokines (like TGF-β) and PI3K/AKT/mTOR pathway. Nuclear p53 increases autophagy, while cytosolic p53 inhibits this pathway. Regulation of autophagy pathway via Atg proteins (Atg7) also inhibits autophagy and decreases EMT and potentially inhibits cancer metastasis. Akt, protein kinase B; EMT, epithelial to mesenchymal transition; mTOR, the mechanistic target of rapamycin; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; PI3K, phosphatidylinositol 3-kinase; TGF-β, transforming growth factor beta. ## Ethics statements ### Patient consent for publication Not required. ## Acknowledgments JA acknowledges CIHR Vanier PhD scholarship for the support. ## Footnotes * Contributors JA and SL did literature search and prepared the first draft. SL designed and prepared the figure. SG finalized and proofread the paper and did the revision. * Funding The authors have not declared a specific grant for this research from any funding agency in the public, commercial or not-for-profit sectors. * Competing interests None declared. * Provenance and peer review Commissioned; internally peer reviewed. ## References 1. Weber GF. Why does cancer therapy lack effective anti-metastasis drugs? Cancer Lett 2013;328:207–11.[doi:10.1016/j.canlet.2012.09.025](http://dx.doi.org/10.1016/j.canlet.2012.09.025)pmid:http://www.ncbi.nlm.nih.gov/pubmed/23059758 [CrossRef](/lookup/external-ref?access_num=10.1016/j.canlet.2012.09.025&link_type=DOI) [PubMed](/lookup/external-ref?access_num=23059758&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 2. Talmadge JE, Fidler IJ. Aacr centennial series: the biology of cancer metastasis: historical perspective. Cancer Res 2010;70:5649–69.[doi:10.1158/0008-5472.CAN-10-1040](http://dx.doi.org/10.1158/0008-5472.CAN-10-1040)pmid:http://www.ncbi.nlm.nih.gov/pubmed/20610625 [Abstract/FREE Full Text](/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FucmVzIjtzOjU6InJlc2lkIjtzOjEwOiI3MC8xNC81NjQ5IjtzOjQ6ImF0b20iO3M6NDI6Ii9qaW0vZWFybHkvMjAyMS8wNy8wNS9qaW0tMjAyMS0wMDIwMTYuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 3. Baldawa P, Shirol P, Alur J, et al. Metastasis: to and fro. J Oral Maxillofac Pathol 2017;21:463–4.[doi:10.4103/jomfp.JOMFP\_158\_17](http://dx.doi.org/10.4103/jomfp.JOMFP_158_17)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29391739 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 4. Steeg PS. Targeting metastasis. Nat Rev Cancer 2016;16:201–18.[doi:10.1038/nrc.2016.25](http://dx.doi.org/10.1038/nrc.2016.25)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27009393 [CrossRef](/lookup/external-ref?access_num=10.1038/nrc.2016.25&link_type=DOI) [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 5. Montanari M, Rossetti S, Cavaliere C, et al. Epithelial-Mesenchymal transition in prostate cancer: an overview. Oncotarget 2017;8:35376–89.[doi:10.18632/oncotarget.15686](http://dx.doi.org/10.18632/oncotarget.15686)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28430640 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 6. Lo U-G, Lee C-F, Lee M-S, et al. The role and mechanism of epithelial-to-mesenchymal transition in prostate cancer progression. Int J Mol Sci 2017;18. doi:[doi:10.3390/ijms18102079](http://dx.doi.org/10.3390/ijms18102079). [Epub ahead of print: 30 Sep 2017].pmid:http://www.ncbi.nlm.nih.gov/pubmed/28973968 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 7. Alizadeh J, Shojaei S, Sepanjnia A, et al. Simultaneous detection of autophagy and epithelial to mesenchymal transition in the non-small cell lung cancer cells. Methods Mol Biol 2019;1854:87–103.[doi:10.1007/7651\_2017\_84](http://dx.doi.org/10.1007/7651_2017_84)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29101677 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 8. Alizadeh J, Glogowska A, Thliveris J, et al. Autophagy modulates transforming growth factor beta 1 induced epithelial to mesenchymal transition in non-small cell lung cancer cells. Biochim Biophys Acta Mol Cell Res 2018;1865:749–68.[doi:10.1016/j.bbamcr.2018.02.007](http://dx.doi.org/10.1016/j.bbamcr.2018.02.007)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29481833 [CrossRef](/lookup/external-ref?access_num=10.1016/j.bbamcr.2018.02.007&link_type=DOI) [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 9. Eshraghi M, Adlimoghaddam A, Mahmoodzadeh A, et al. Alzheimer's disease pathogenesis: role of autophagy and mitophagy focusing in microglia. Int J Mol Sci 2021;22:3330. [doi:10.3390/ijms22073330](http://dx.doi.org/10.3390/ijms22073330)pmid:http://www.ncbi.nlm.nih.gov/pubmed/33805142 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 10. Shojaei S, Suresh M, Klionsky DJ, et al. Autophagy and SARS-CoV-2 infection: Apossible smart targeting of the autophagy pathway. Virulence 2020;11:805–10.[doi:10.1080/21505594.2020.1780088](http://dx.doi.org/10.1080/21505594.2020.1780088)pmid:http://www.ncbi.nlm.nih.gov/pubmed/32567972 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 11. Shojaei S, Koleini N, Samiei E, et al. Simvastatin increases temozolomide-induced cell death by targeting the fusion of autophagosomes and lysosomes. Febs J 2020;287:1005–34.[doi:10.1111/febs.15069](http://dx.doi.org/10.1111/febs.15069)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31545550 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 12. Yang A, Rajeshkumar NV, Wang X, et al. Autophagy is critical for pancreatic tumor growth and progression in tumors with p53 alterations. Cancer Discov 2014;4:905–13.[doi:10.1158/2159-8290.CD-14-0362](http://dx.doi.org/10.1158/2159-8290.CD-14-0362)pmid:http://www.ncbi.nlm.nih.gov/pubmed/24875860 [Abstract/FREE Full Text](/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiY2FuZGlzYyI7czo1OiJyZXNpZCI7czo3OiI0LzgvOTA1IjtzOjQ6ImF0b20iO3M6NDI6Ii9qaW0vZWFybHkvMjAyMS8wNy8wNS9qaW0tMjAyMS0wMDIwMTYuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 13. Rao S, Tortola L, Perlot T, et al. A dual role for autophagy in a murine model of lung cancer. Nat Commun 2014;5:3056. [doi:10.1038/ncomms4056](http://dx.doi.org/10.1038/ncomms4056)pmid:http://www.ncbi.nlm.nih.gov/pubmed/24445999 [CrossRef](/lookup/external-ref?access_num=10.1038/ncomms4056&link_type=DOI) [PubMed](/lookup/external-ref?access_num=24445999&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 14. Bao Y, Ding Z, Zhao P, et al. Autophagy inhibition potentiates the anti-EMT effects of alteronol through TGF-β/Smad3 signaling in melanoma cells. Cell Death Dis 2020;11:223.[doi:10.1038/s41419-020-2419-y](http://dx.doi.org/10.1038/s41419-020-2419-y)pmid:http://www.ncbi.nlm.nih.gov/pubmed/32265437 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 15. Ding Y-H, Zhou Z-W, Ha C-F, et al. Alisertib, an Aurora kinase A inhibitor, induces apoptosis and autophagy but inhibits epithelial to mesenchymal transition in human epithelial ovarian cancer cells. Drug Des Devel Ther 2015;9:425–64.[doi:10.2147/DDDT.S74062](http://dx.doi.org/10.2147/DDDT.S74062)pmid:http://www.ncbi.nlm.nih.gov/pubmed/25624750 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 16. Wang F, Li H, Yan X-G, et al. Alisertib induces cell cycle arrest and autophagy and suppresses epithelial-to-mesenchymal transition involving PI3K/Akt/mTOR and sirtuin 1-mediated signaling pathways in human pancreatic cancer cells. Drug Des Devel Ther 2015;9:575–601.[doi:10.2147/DDDT.S75221](http://dx.doi.org/10.2147/DDDT.S75221)pmid:http://www.ncbi.nlm.nih.gov/pubmed/25632225 [CrossRef](/lookup/external-ref?access_num=10.2147/DDDT.S75221&link_type=DOI) [PubMed](/lookup/external-ref?access_num=25632225&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 17. Karimi Roshan M, Soltani A, Soleimani A, et al. Role of Akt and mTOR signaling pathways in the induction of epithelial-mesenchymal transition (EMT) process. Biochimie 2019;165:229–34.[doi:10.1016/j.biochi.2019.08.003](http://dx.doi.org/10.1016/j.biochi.2019.08.003)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31401189 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 18. Guo R, Meng Q, Guo H, et al. TGF-β2 induces epithelial-mesenchymal transition in cultured human lens epithelial cells through activation of the PI3K/Akt/mTOR signaling pathway. Mol Med Rep 2016;13:1105–10.[doi:10.3892/mmr.2015.4645](http://dx.doi.org/10.3892/mmr.2015.4645)pmid:http://www.ncbi.nlm.nih.gov/pubmed/26647778 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 19. Lu Q, Wang W-W, Zhang M-Z, et al. Ros induces epithelial-mesenchymal transition via the TGF-β1/PI3K/Akt/mTOR pathway in diabetic nephropathy. Exp Ther Med 2019;17:835–46.[doi:10.3892/etm.2018.7014](http://dx.doi.org/10.3892/etm.2018.7014)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30651870 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 20. Pires BRB, Mencalha AL, Ferreira GM, et al. Nf-kappaB is involved in the regulation of EMT genes in breast cancer cells. PLoS One 2017;12:e0169622. [doi:10.1371/journal.pone.0169622](http://dx.doi.org/10.1371/journal.pone.0169622)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28107418 [CrossRef](/lookup/external-ref?access_num=10.1371/journal.pone.0169622&link_type=DOI) [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 21. Chen L-M, Song T-J, Xiao J-H, et al. Tripchlorolide induces autophagy in lung cancer cells by inhibiting the PI3K/Akt/mTOR pathway and improves cisplatin sensitivity in A549/DDP cells. Oncotarget 2017;8:63911–22.[doi:10.18632/oncotarget.19201](http://dx.doi.org/10.18632/oncotarget.19201)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28969040 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 22. Lin M-C, Lee Y-W, Tseng Y-Y, et al. Honokiol induces autophagic apoptosis in neuroblastoma cells through a p53-dependent pathway. Am J Chin Med 2019;47:895–912.[doi:10.1142/S0192415X19500472](http://dx.doi.org/10.1142/S0192415X19500472)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31091975 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 23. Lin C-J, Chen T-L, Tseng Y-Y, et al. Honokiol induces autophagic cell death in malignant glioma through reactive oxygen species-mediated regulation of the p53/PI3K/Akt/mTOR signaling pathway. Toxicol Appl Pharmacol 2016;304:59–69.[doi:10.1016/j.taap.2016.05.018](http://dx.doi.org/10.1016/j.taap.2016.05.018)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27236003 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 24. Babaei G, Aziz SG-G, Jaghi NZZ. Emt, cancer stem cells and autophagy; the three main axes of metastasis. Biomed Pharmacother 2021;133:110909.[doi:10.1016/j.biopha.2020.110909](http://dx.doi.org/10.1016/j.biopha.2020.110909)pmid:http://www.ncbi.nlm.nih.gov/pubmed/33227701 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 25. Zhu H, Wang D, Zhang L, et al. Upregulation of autophagy by hypoxia-inducible factor-1α promotes EMT and metastatic ability of CD133+ pancreatic cancer stem-like cells during intermittent hypoxia. Oncol Rep 2014;32:935–42.[doi:10.3892/or.2014.3298](http://dx.doi.org/10.3892/or.2014.3298)pmid:http://www.ncbi.nlm.nih.gov/pubmed/24994549 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 26. Mrakovcic M, Fröhlich LF. P53-Mediated molecular control of autophagy in tumor cells. Biomolecules 2018;8:14. [doi:10.3390/biom8020014](http://dx.doi.org/10.3390/biom8020014)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29561758 [PubMed](/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom) 27. Ren D, Wang M, Guo W, et al. Wild-Type p53 suppresses the epithelial-mesenchymal transition and stemness in PC-3 prostate cancer cells by modulating miR‑145. Int J Oncol 2013;42:1473–81.[doi:10.3892/ijo.2013.1825](http://dx.doi.org/10.3892/ijo.2013.1825)pmid:http://www.ncbi.nlm.nih.gov/pubmed/23404342 [PubMed](/lookup/external-ref?access_num=23404342&link_type=MED&atom=%2Fjim%2Fearly%2F2021%2F07%2F05%2Fjim-2021-002016.atom)