Recently Used Drug Targets and Autophagy: Comparison
Please note this is a comparison between Version 2 by Catherine Yang and Version 1 by Abu Saim Mohammad Saikat.

Autophagy is a governed catabolic framework enabling the recycling of nutrients from injured organelles and other cellular constituents via a lysosomal breakdown. This mechanism has been associated with the development of various pathologic conditions, including cancer and neurological disorders; however, recently updated studies have indicated that autophagy plays a dual role in cancer, acting as a cytoprotective or cytotoxic mechanism. It has recently been found that autophagy regulation is an intriguing and potentially useful strategy to improve cancer treatments. Numerous studies have indicated autophagy’s dual roles for several drugs that might be modulated to suppress or promote tumor growth, and the following sections describe autophagy’s dual function in recent drug treatments.

  • autophagy
  • cancer
  • autophagy inhibitors
  • autophagy activators

1. Targeted Drug for Autophagy Activation in Cancer Therapy

In cancer, autophagy helps promote and inhibit tumors by helping or hindering cancer cells from growing and dividing, while some drugs that fight cancer control autophagy; therefore, chemotherapy controlled by autophagy can cause cancer cells to live or die [30][1]. Autophagy regulation also affects the expression of proteins that promote or inhibit tumor growth [31][2], and the mechanism of action associated with autophagy stimulation is presented in Table 1. Additionally, the stimulation of autophagy by various chemicals and medications in cancer treatments is presented in Figure 31.
Figure 31. Autophagy stimulation by several compounds and drugs used in cancer therapy.
Table 1. Recently used drugs for autophagy stimulation and their mechanisms of action.
Autophagy may be tumor-suppressive, neutral, and tumor-promoting in cancer, depending on the microenvironment stress, nutrient availability, immune system, and pathogens [21][15]. The mTOR protein kinase is related to various biological functions, such as cell survival, growth, immunity, and metabolism. Consequently, mTOR is implicated in supervising numerous biological processes, notably autophagy, apoptosis, and the cell cycle, thus preventing the commencement of the concluding process [44][16]. A P75NTR activation by rapamycin induces autophagy, leading to Kaposi’s sarcoma cell death [43][14]. Additionally, rapamycin (sirolimus), a secondary metabolite derived from Streptomyces hygroscopicus, has significant anticancer and immunosuppressive activities [45][17]. Rapamycin and its semi-synthetic counterparts (rapalogs) are allosterically-precise and potent mammalian targets of rapamycin complex 1 (mTORC1) blockers that influence downstream targets, such as autophagy stimulation [46][18]. Other mTOR blockers interfere with adenosine triphosphate (ATP), preventing the phosphorylation of its corresponding proteins and leading to more effective mTOR suppression [47][19]. Altogether, these observations show that mTOR blockers may be used to promote cell death through several pathways. Their mechanisms of action rely on the tumor microenvironment, overcoming cancer cell resistance in combination therapy [48][20].
While cannabinoids have shown strong anticancer properties associated with autophagy, they have also shown cytoprotective properties, depending on the cell type and cannabinoid used [49][21]. Active cell death (ACD) in melanoma and glioma cells occurs via mTORC1 inhibition and autolysosome permeabilization, resulting in cathepsin secretion and apoptosis activation [50][22]. JWH-015, for example, is a synthetic cannabinoid specific to the CB2 receptor that suppressed tumor development in hepatocellular carcinoma (HCC) cells in vivo via inhibiting the Akt/mTORC1 cascade through an AMP-activated protein kinase (AMPK) stimulation [9][23]. Moreover, obatoclax (GX15-070) is another BH3 mimic that induced necroptosis in oral squamous cell carcinoma, acute lymphoblastic leukemia, and rhabdomyosarcoma via autophagic signaling [51][24]. Additionally, obatoclax promoted autophagy in adenoid cystic carcinoma cells and inhibited autophagy in colorectal cancer cells in the absence of Bcl-1 [52][25]. Subsequently, ABT-737 was found to be effective in vitro against HCC cells via an autophagy-dependent mechanism involving Bcl-1 [53][26].
Furthermore, APO866 inhibited nicotinamide adenine dinucleotide biosynthesis and induced apoptotic cell death in hematological cancer cells [54][27]. Elsewhere, histone deacetylase (HDAC) inhibitors were shown to induce autophagy. A recent study used the HDAC inhibitor, suberoylanilide hydroxamic acid (SAHA), to treat cutaneous T-cell lymphoma and glioblastoma [55][28]. While apoptosis had been identified as the primary mechanism through which the HDAC inhibitors promoted cancer cell death, autophagy promotion was also associated with a PI3K/Akt/mTOR pathway inhibition [56][29]. In addition, SAHA inhibited breast cancer cell proliferation in vitro by stimulating autophagy and activating cathepsin B [57,58][30][31]. Moreover, in vivo and in vitro studies have shown that MHY2256 (a synthetic class III HDAC inhibitor) stimulates CCA, ACD, and apoptosis in endocervical curettage [59][32]. SAHA activates autophagy by inhibiting mTOR and upregulating the LC3 expression. Autophagy mitigates a SAHA-induced apoptotic and nonapoptotic cell death, suggesting that targeting autophagy may improve SAHA’s therapeutic effects [60][33].

2. Targeted Drug for the Inhibition of Autophagy in Cancer Therapy

It has been shown that inhibiting autophagy makes an anticancer therapy more effective, suggesting that it is a potentially valuable approach that could be combined with other anticancer therapeutic approaches to improve cancer treatments [61][34]. Numerous autophagy inhibitors impede the autophagy processes at various stages, as discussed in Table 2 and Figure 42.
Figure 42.
Autophagy inhibition by several compounds and drugs used in cancer therapy.
Table 2.
Recently used autophagy inhibitors and their mechanisms of action in different cancers.
Nigrosporins B, a potential anti-cervical cancer agent, induced apoptosis and protective autophagy in human cervical cancer Ca Ski cells. These effects were mediated via the PI3K/AKT/mTOR signaling pathway [62][35]. Additionally, autophagy is affected by antimetabolites, antiangiogenic agents, DNA-damaging agents, proteasome inhibitors, microtubule-targeted drugs, death receptor agonists, hormonal agents, HDAC inhibitors, and kinase inhibitors [74][47]. ATG proteins and other factors form complexes that lengthen phagophores. ATG7 is involved in creating the ATG5-ATG12 complex and coupling PE to LC3 and GABARAP [75][48]. Consequently, many ATG7 suppressors (e.g., WO2018/089786) have been developed, extending the use of micro RNAs (miRNAs) targeting the ATG7 gene, including miR-154, which suppresses blade cancer growth [76][49]. Conversely, ATG4B cleaves LC3, stimulating it in preparation for coupling with PE, which is required for autophagosome development and activation [77][50].
Verteporfin is a photodynamic therapeutic agent derived from benzoporphyrin. It also suppresses autophagosome production induced by serum and glucose deprivation but not mTOR suppression [78][51]. Verteporfin is suspected of suppressing p62 oligomerization, a protein crucial for ubiquitinated targets to be sequestered into autophagosomes [78][51]. Inhibiting ULK1 slowed the tumor development and activated apoptosis. This finding has resulted in the discovery of many chemicals that compete for ATP-binding sites, including compound-6, MRT67307, and MRT68921 [9][23]. Apart from these, the most investigated compound is SBI-0206965, which has been shown to suppress autophagy and promotes apoptosis in neuronal ceroid lipofuscinoses, clear-cell renal cell carcinoma cells, and non-small-cell lung cancer (NSCLC) cells [79][52]. SF1126, a conjugated LY294002 derivative, was engineered to assemble in integrin-expressing tissues, increasing LY294002′s solubility and pharmacokinetics, promoting tumor site retention, and showing anticancer and antiangiogenic activity in mouse models [80][53].
3-methyladenine (3-MA) was among the earliest autophagy inhibitors. It inhibits autophagy in starvation situations by inhibiting PI3KC3 [81][54]. Conversely, when nutrients are present, they induce autophagy by blocking PI3KC1. The VPS34 inhibitor, SAR405, is a (2S)-tetrahydropyrimido-pyrimidinone class member that inhibits kinases via an extreme competition for their ATP binding site, thus suppressing autophagy produced by an mTOR inhibition or malnutrition [82][55]. VPS34-IN1 is a bipyrimidinamine that preferentially suppresses PI3KC3 [9][23]. Esomeprazole’s antiproliferative action, for example, was enhanced when combined with the PI3K inhibitor, 3-MA, in gastric cancer cells through EGFR downregulation via the PI3K/FOXO3a pathway [63][36]. In gemcitabine-resistant pancreatic cancer cells, however, 4’-acetylantroquinonol B promoted cell death and inhibited autophagy by downregulating the PI3K/Akt/MDR1 pathway, increasing cell death [64][37].
Autophagy’s late phase involves autophagosomes fusing with lysosomes, which contain hydrolases that destroy the autophagosomes’ contents. During this stage, autophagy is suppressed by the lysosomal blockers, bafilomycin A1 (Baf-A1), chloroquine (CQ), and its derivatives Lys05 and hydroxychloroquine (HCQ), which are antimalarial medicines used for treating malaria and, more recently, cancer [50][22].
Only CQ and HCQ are authorized for therapeutic use as autophagy inhibitors [42][13]. Consequently, CQ derivatives with increased inhibitory-efficacy against autophagy have been synthesized. Lys05, for example, is a dimeric CQ analog that aggregates better than HCQ within acidic organelles, particularly lysosomes [83][56]. Duroquinone DQ661 disrupts lysosomal degradation, notably autophagy, and suppresses PPT-1, inhibiting the mTORC1 cascade. Furthermore, DQ661 has shown independent efficacy in tumor mouse models and the ability to overcome gemcitabine resistance [9][23]. VATG-027 is another antimalaria drug that suppresses autophagy with antitumoral effects [84][57]. Meanwhile, mefloquine accumulates in lysosomes, impairing autophagy, inducing apoptosis, and inhibiting MDRP1, making it potent against MDRT cells. It sensitizes chronic myeloid leukemia cells created from chronic-stage patients to tyrosine kinase blockers, with a preference for stem/progenitor tumor cells over normal cells [85][58].
CQ and its analogs, however, are not the only medications that suppress autophagy by affecting lysosomes [86][59]. Baf A is a vacuolar-H+ ATPase blocker that prevents H+ from entering lysosomes, vacuoles, and vesicles. Baf A also suppresses autophagosome-lysosome fusion by disrupting the Ca2+ gradients required for this pathway [87][60]. Additionally, ionophores can alter the pH of the lysosome, affecting the autophagy mechanism. Tambjamine derivatives are ASI synthesized from naturally produced tambjamines [88][61]. Importantly, because lysosomes are involved in tumor invasion, these blockers are efficacious against metastasis by addressing cancer stem cells and promoting tumor vessel normalization.

References

  1. Bahar, E.; Han, S.Y.; Kim, J.Y.; Yoon, H. Chemotherapy Resistance: Role of Mitochondrial and Autophagic Components. Cancers 2022, 14, 1462.
  2. Peng, F.; Liao, M.; Qin, R.; Zhu, S.; Peng, C.; Fu, L.; Chen, Y.; Han, B. Regulated cell death (RCD) in cancer: Key pathways and targeted therapies. Signal Transduct. Target. Ther. 2022, 7, 286.
  3. Toriyama, K.; Takano, N.; Kokuba, H.; Kazama, H.; Moriya, S.; Hiramoto, M.; Abe, S.; Miyazawa, K. Azithromycin enhances the cytotoxicity of DNA-damaging drugs via lysosomal membrane permeabilization in lung cancer cells. Cancer Sci. 2021, 112, 3324–3337.
  4. Tung, M.-C.; Lin, Y.-W.; Lee, W.-J.; Wen, Y.-C.; Liu, Y.-C.; Chen, J.-Q.; Hsiao, M.; Yang, Y.-C.; Chien, M.-H. Targeting DRD2 by the antipsychotic drug, penfluridol, retards growth of renal cell carcinoma via inducing stemness inhibition and autophagy-mediated apoptosis. Cell Death Dis. 2022, 13, 400.
  5. Park, C.H.; Bin Lee, B.; Han, M.; Rhee, W.J.; Kwak, M.S.; Yoo, T.-H.; Shin, J.-S. Canagliflozin protects against cisplatin-induced acute kidney injury by AMPK-mediated autophagy in renal proximal tubular cells. Cell Death Discov. 2022, 8, 12.
  6. Tang, Z.Y.; Li, Y.; Tang, Y.T.; Ma, X.D.; Tang, Z.Y. Anticancer activity of oleanolic acid and its derivatives: Recent advances in evidence, target profiling and mechanisms of action. Biomed. Pharm. 2022, 145, 112397.
  7. Chang, S.N.; Haroon, M.; Dey, D.K.; Kang, S.C. Rhabdomyolysis-induced acute kidney injury and concomitant apoptosis induction via ROS-mediated ER stress is efficaciously counteracted by epigallocatechin gallate. J. Nutr. Biochem. 2022, 110, 109134.
  8. Kang, K.A.; Yao, C.W.; Piao, M.J.; Zhen, A.X.; Fernando, P.D.S.M.; Herath, H.M.U.L.; Song, S.E.; Cho, S.J.; Hyun, J.W. Anticolon Cancer Effect of Korean Red Ginseng via Autophagy- and Apoptosis-Mediated Cell Death. Nutrients 2022, 14, 3558.
  9. Wróblewska-Łuczka, P.; Cabaj, J.; Bąk, W.; Bargieł, J.; Grabarska, A.; Góralczyk, A.; Łuszczki, J.J. Additive Interactions between Betulinic Acid and Two Taxanes in In Vitro Tests against Four Human Malignant Melanoma Cell Lines. Int. J. Mol. Sci. 2022, 23, 9641.
  10. Zhang, L.; Xu, S.; Cheng, X.; Wu, J.; Wu, L.; Wang, Y.; Wang, X.; Bao, J.; Yu, H. Curcumin induces autophagic cell death in human thyroid cancer cells. Toxicol. Vitr. 2022, 78, 105254.
  11. Wang, Z.; Zhang, P.; Jiang, H.; Sun, B.; Luo, H.; Jia, A. Ursolic Acid Enhances the Sensitivity of MCF-7 and MDA-MB-231 Cells to Epirubicin by Modulating the Autophagy Pathway. Molecules 2022, 27, 3399.
  12. Parkhurst, A.; Wang, S.Z.; Findlay, T.R.; Malebranche, K.J.; Odabas, A.; Alt, J.; Maxwell, M.J.; Kaur, H.; Peer, C.J.; Figg, W.D.; et al. Dual mTORC1/2 inhibition compromises cell defenses against exogenous stress potentiating Obatoclax-induced cytotoxicity in atypical teratoid/rhabdoid tumors. Cell Death Dis. 2022, 13, 410.
  13. Cocco, S.; Leone, A.; Roca, M.S.; Lombardi, R.; Piezzo, M.; Caputo, R.; Ciardiello, C.; Costantini, S.; Bruzzese, F.; Sisalli, M.J.; et al. Inhibition of autophagy by chloroquine prevents resistance to PI3K/AKT inhibitors and potentiates their antitumor effect in combination with paclitaxel in triple negative breast cancer models. J. Transl. Med. 2022, 20, 290.
  14. Lupinacci, S.; Perri, A.; Toteda, G.; Vizza, D.; Lofaro, D.; Pontrelli, P.; Stallone, G.; Divella, C.; Tessari, G.; La Russa, A.; et al. Rapamycin promotes autophagy cell death of Kaposi’s sarcoma cells through P75NTR activation. Exp. Dermatol. 2022, 31, 143–153.
  15. Lei, Y.; Zhang, E.; Bai, L.; Li, Y. Autophagy in Cancer Immunotherapy. Cells 2022, 11, 2996.
  16. Vakrakou, A.G.; Alexaki, A.; Brinia, M.-E.; Anagnostouli, M.; Stefanis, L.; Stathopoulos, P. The mTOR signaling pathway in multiple sclerosis; from animal models to human data. Int. J. Mol. Sci. 2022, 23, 8077.
  17. Khushboo; Kumar, P.; Dubey, K.K.; Usmani, Z.; Sharma, M.; Gupta, V.K. Biotechnological and industrial applications of Streptomyces metabolites. Biofuels Bioprod. Biorefining 2022, 16, 244–264.
  18. Movahedpour, A.; Vakili, O.; Khalifeh, M.; Mousavi, P.; Mahmoodzadeh, A.; Taheri-Anganeh, M.; Razmeh, S.; Shabaninejad, Z.; Yousefi, F.; Behrouj, H.; et al. Mammalian target of rapamycin (mTOR) signaling pathway and traumatic brain injury: A novel insight into targeted therapy. Cell Biochem. Funct. 2022, 40, 232–247.
  19. Morales-Jiménez, C.; Balanta-Melo, J.; Arias-Calderón, M.; Hernández, N.; Gómez-Valenzuela, F.; Escobar, A.; Jaimovich, E.; Buvinic, S. Mechanical Disturbance of Osteoclasts Induces ATP Release That Leads to Protein Synthesis in Skeletal Muscle through an Akt-mTOR Signaling Pathway. Int. J. Mol. Sci. 2022, 23, 9444.
  20. Quan, Z.; Yang, Y.; Zheng, H.; Zhan, Y.; Luo, J.; Ning, Y.; Fan, S. Clinical implications of the interaction between PD-1/PD-L1 and PI3K/AKT/mTOR pathway in progression and treatment of non-small cell lung cancer. J. Cancer 2022, 13, 3434–3443.
  21. Loubaki, L.; Rouabhia, M.; Al Zahrani, M.; Al Amri, A.; Semlali, A. Oxidative Stress and Autophagy Mediate Anti-Cancer Properties of Cannabis Derivatives in Human Oral Cancer Cells. Cancers 2022, 14, 4924.
  22. Al-Bari, A.A. Inhibition of autolysosomes by repurposing drugs as a promising therapeutic strategy for the treatment of cancers. All Life 2022, 15, 568–601.
  23. Emon, K.H. Recent Advances in Cancer Treatment Targeting Autophagy and Future Aspects; Brac University: Dhaka, Bangladesh, 2022.
  24. Spiesschaert, B.; Angerer, K.; Park, J.; Wollmann, G. Combining Oncolytic Viruses and Small Molecule Therapeutics: Mutual Benefits. Cancers 2021, 13, 3386.
  25. Bata, N.; Cosford, N.D.P. Cell survival and cell death at the intersection of autophagy and apoptosis: Implications for current and future cancer therapeutics. ACS Pharmacol. Transl. Sci. 2021, 4, 1728–1746.
  26. Yao, X.; Li, X.; Zhang, D.; Xie, Y.; Sun, B.; Li, H.; Sun, L.; Zhang, X. B-cell lymphoma 2 inhibitor ABT-737 induces Beclin1- and reactive oxygen species-dependent autophagy in Adriamycin-resistant human hepatocellular carcinoma cells. Tumour. Biol. 2017, 39, 1010428317695965.
  27. Zerp, S.F. Modulating Tumor Cell Death to Enhance Radiation Response: A Focus on Apoptosis. Master’s Thesis, Radboud University, Nijmegen, The Netherlands, 2022.
  28. Gammoh, N.; Lam, D.; Puente, C.; Ganley, I.; Marks, P.A.; Jiang, X. Role of autophagy in histone deacetylase inhibitor-induced apoptotic and nonapoptotic cell death. Proc. Natl. Acad. Sci. USA 2012, 109, 6561–6565.
  29. Mrakovcic, M.; Bohner, L.; Hanisch, M.; Fröhlich, L.F. Epigenetic Targeting of Autophagy via HDAC Inhibition in Tumor Cells: Role of p53. Int. J. Mol. Sci. 2018, 19, 3952.
  30. Mann, B.S.; Johnson, J.R.; Cohen, M.H.; Justice, R.; Pazdur, R. FDA approval summary: Vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist 2007, 12, 1247–1252.
  31. Han, H.; Li, J.; Feng, X.; Zhou, H.; Guo, S.; Zhou, W. Autophagy-related genes are induced by histone deacetylase inhibitor suberoylanilide hydroxamic acid via the activation of cathepsin B in human breast cancer cells. Oncotarget 2017, 8, 53352–53365.
  32. De, U.; Son, J.Y.; Sachan, R.; Park, Y.J.; Kang, D.; Yoon, K.; Lee, B.M.; Kim, I.S.; Moon, H.R.; Kim, H.S. A New Synthetic Histone Deacetylase Inhibitor, MHY2256, Induces Apoptosis and Autophagy Cell Death in Endometrial Cancer Cells via p53 Acetylation. Int. J. Mol. Sci. 2018, 19, 2743.
  33. Garmpis, N.; Damaskos, C.; Garmpi, A.; Kalampokas, E.; Kalampokas, T.; Spartalis, E.; Daskalopoulou, A.; Valsami, S.; Kontos, M.; Nonni, A.; et al. Histone Deacetylases as New Therapeutic Targets in Triple-negative Breast Cancer: Progress and Promises. Cancer Genom. Proteom. 2017, 14, 299–313.
  34. Gillson, J.; El-Aziz, Y.S.A.; Leck, L.Y.W.; Jansson, P.J.; Pavlakis, N.; Samra, J.S.; Mittal, A.; Sahni, S. Autophagy: A Key Player in Pancreatic Cancer Progression and a Potential Drug Target. Cancers 2022, 14, 3528.
  35. Zhang, J.; Guo, Z.-Y.; Shao, C.-L.; Zhang, X.-Q.; Cheng, F.; Zou, K.; Chen, J.-F.; Nigrosporins, B. A Potential Anti-Cervical Cancer Agent, Induces Apoptosis and Protective Autophagy in Human Cervical Cancer Ca Ski Cells Mediated by PI3K/AKT/mTOR Signaling Pathway. Molecules 2022, 27, 2431.
  36. Du, J.; Xu, Q.; Zhao, H.; Jia, X.; Ba, N.; Peng, F.; Zhang, Z. PI3K inhibitor 3-MA promotes the antiproliferative activity of esomeprazole in gastric cancer cells by downregulating EGFR via the PI3K/FOXO3a pathway. Biomed. Pharmacother. 2022, 148, 112665.
  37. Chen, Y.-Y.; Chen, S.-Y.; Li, T.-J.; Lin, T.-W.; Chen, C.-C.; Yen, G.-C. 4-Acetylantroquinonol B enhances cell death and inhibits autophagy by downregulating the PI3K/Akt/MDR1 pathway in gemcitabine-resistant pancreatic cancer cells. Oncol. Lett. 2022, 23, 128.
  38. Huang, H.-Y.; Li, K.-N.; Lau, H.-C.; Hsueh, C.-Y.; Cong, N.; Zhang, M. Dual inhibition of autophagy and PI3K/mTOR pathway as a potential therapeutic strategy against laryngeal squamous cell carcinoma. Transl. Cancer Res. 2022, 11, 1076.
  39. Park, Y.; Jang, J. Prospect of ULK1 modulators in targeting regulatory T cells. Bioorganic. Chem. 2022, 129, 106141.
  40. Li, S.; Chen, J.; Fan, Y.; Wang, C.; Wang, C.; Zheng, X.; Chen, F.; Li, W. Liposomal Honokiol induces ROS-mediated apoptosis via regulation of ERK/p38-MAPK signaling and autophagic inhibition in human medulloblastoma. Signal Transduct. Target. Ther. 2022, 7, 49.
  41. Wang, X.; Li, Y.; Jia, F.; Cui, X.; Pan, Z.; Wu, Y. Boosting nutrient starvation-dominated cancer therapy through curcumin-augmented mitochondrial Ca2+ overload and obatoclax-mediated autophagy inhibition as supported by a novel nano-modulator CaP/CO. J. Nanobiotechnol. 2022, 20, 225.
  42. Kang, Y.J.; Jang, J.Y.; Kwon, Y.H.; Lee, J.H.; Lee, S.; Park, Y.; Jung, Y.-S.; Im, E.; Moon, H.R.; Chung, H.Y.; et al. MHY2245, a Sirtuin Inhibitor, Induces Cell Cycle Arrest and Apoptosis in HCT116 Human Colorectal Cancer Cells. Int. J. Mol. Sci. 2022, 23, 1590.
  43. Hoque, M.M.; Iida, Y.; Kotani, H.; Kartika, I.D.; Harada, M. Hydroxychloroquine Promotes Bcl-xL Inhibition-induced Apoptosis in BxPC-3 Human Pancreatic Cancer Cells. Anticancer Res. 2022, 42, 3495–3506.
  44. Zhou, R.; Kusaka, E.; Wang, Y.; Zhang, J.; Webb, A.; Carrico-Moniz, D. Isoprenylated Coumarin Exhibits Anti-proliferative Effects in Pancreatic Cancer Cells Under Nutrient Starvation by Inhibiting Autophagy. Anticancer Res. 2022, 42, 2835–2845.
  45. Holm, T.M.; Bian, Z.C.; Manupati, K.; Guan, J.-L. Inhibition of autophagy mitigates cell migration and invasion in thyroid cancer. Surgery 2022, 171, 235–244.
  46. Liu, P.-F.; Tsai, K.-L.; Hsu, C.-J.; Tsai, W.-L.; Cheng, J.-S.; Chang, H.-W.; Shiau, C.-W.; Goan, Y.-G.; Tseng, H.-H.; Wu, C.-H.; et al. Drug Repurposing Screening Identifies Tioconazole as an ATG4 Inhibitor that Suppresses Autophagy and Sensitizes Cancer Cells to Chemotherapy. Theranostics 2018, 8, 830–845.
  47. Hu, C.; Gu, F.; Gong, C.; Xia, Q.; Gao, Y.; Gao, S. Co-delivery of the autophagy inhibitor si-Beclin1 and the doxorubicin nano-delivery system for advanced prostate cancer treatment. J. Biomater. Appl. 2022, 36, 1317–1331.
  48. Majeed, S.T.; Majeed, R.; Andrabi, K.I. Expanding the view of the molecular mechanisms of autophagy pathway. J. Cell. Physiol. 2022, 237, 3257–3277.
  49. Zhang, J.; Mao, S.; Wang, L.; Zhang, W.; Zhang, Z.; Guo, Y.; Wu, Y.; Yi, F.; Yao, X. MicroRNA-154 functions as a tumor suppressor in bladder cancer by directly targeting ATG7. Oncol. Rep. 2019, 41, 819–828.
  50. Huang, X.; Yao, J.; Liu, L.; Luo, Y.; Yang, A. Atg8–PE protein-based in vitro biochemical approaches to autophagy studies. Autophagy 2022, 18, 2020–2035.
  51. Ye, J.; Yu, B.; Hu, H.; Zhou, D.; Jin, Q.; Ji, J.; Tang, Z. Verteporfin-loaded supramolecular micelles for enhanced cisplatin-based chemotherapy via autophagy inhibition. J. Mater. Chem. B 2022, 10, 2670–2679.
  52. Desai, J.M.; Karve, A.S.; Gudelsky, G.A.; Gawali, M.V.; Seibel, W.; Sallans, L.; DasGupta, B.; Desai, P.B. Brain pharmacokinetics and metabolism of the AMP-activated protein kinase selective inhibitor SBI-0206965, an investigational agent for the treatment of glioblastoma. Investig. New Drugs 2022, 40, 944–952.
  53. Chen, J.; Hu, Q.; Luo, Y.; Luo, L.; Lin, H.; Chen, D.; Xu, Y.; Liu, B.; He, Y.; Liang, C.; et al. Salvianolic acid B attenuates membranous nephropathy by activating renal autophagy via microRNA-145-5p/phosphatidylinositol 3-kinase/AKT pathway. Bioengineered 2022, 13, 13956–13969.
  54. Kwon, Y.; Haam, C.E.; Byeon, S.; Choi, S.K.; Lee, Y.H. Effects of 3-methyladenine, an autophagy inhibitor, on the elevated blood pressure and arterial dysfunction of angiotensin II-induced hypertensive mice. Biomed. Pharmacother. 2022, 154, 113588.
  55. Stempels, F.C.; Janssens, M.H.; Beest, M.; Mesman, R.J.; Revelo, N.H.; Ioannidis, M.; Bogaart, G.D. Novel and conventional inhibitors of canonical autophagy differently affect LC3-associated phagocytosis. FEBS Lett. 2022, 596, 491–509.
  56. Lu, G.; Wang, Y.; Shi, Y.; Zhang, Z.; Huang, C.; He, W.; Wang, C.; Shen, H. Autophagy in health and disease: From molecular mechanisms to therapeutic target. MedComm 2022, 3, e150.
  57. Kumar, M.; Sarkar, A. Repurposing of Anti-Malarial Drug Quinacrine for Cancer Treatment: A Review. Sci. Pharm. 2022, 90, 12.
  58. Shin, J.H.; Park, S.J.; Jo, Y.K.; Kim, E.S.; Kang, H.; Park, J.-H.; Lee, E.H.; Cho, D.-H. Suppression of autophagy exacerbates Mefloquine-mediated cell death. Neurosci. Lett. 2012, 515, 162–167.
  59. Mauthe, M.; Orhon, I.; Rocchi, C.; Zhou, X.; Luhr, M.; Hijlkema, K.J.; Coppes, R.P.; Engedal, N.; Mari, M.; Reggiori, F. Chloroquine inhibits autophagic flux by decreasing autophagosome-lysosome fusion. Autophagy 2018, 14, 1435–1455.
  60. Tong, B.C.; Wu, A.J.; Huang, A.S.; Dong, R.; Malampati, S.; Iyaswamy, A.; Krishnamoorthi, S.; Sreenivasmurthy, S.G.; Zhu, Z.; Su, C.; et al. Lysosomal TPCN (two pore segment channel) inhibition ameliorates beta-amyloid pathology and mitigates memory impairment in Alzheimer disease. Autophagy 2022, 18, 624–642.
  61. Rodilla Martín, A.M. Anticancer Effect and Molecular Target Identification of Novel Anionophores in Lung Cancer; University of Barcelona: Barcelona, Spain, 2018.
More
Video Production Service