Martins WK, Silva M, Pandey Ok, Maejima I, Ramalho E, Olivon VC, et al. Autophagy-targeted remedy to modulate age-related ailments: success, pitfalls, and new instructions. Curr Res Pharm Drug Discov. 2021;2:100033.
Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell. 2011;147:728–41.
Li X, He S, Ma B. Autophagy and autophagy-related proteins in most cancers. Mol Most cancers. 2020;19:12.
Liu WJ, Ye L, Huang WF, Guo LJ, Xu ZG, Wu HL, et al. p62 hyperlinks the autophagy pathway and the ubiqutin-proteasome system upon ubiquitinated protein degradation. Cell Mol Biol Let. 2016;21:29.
Boutouja F, Stiehm CM, Platta HW. mTOR: a mobile regulator interface in well being and illness. Cells. 2019;8:18.
Gupta P, Kumar N, Garg M. Rising roles of autophagy within the growth and therapy of urothelial carcinoma of the bladder. Knowledgeable Opin Ther Targets. 2021;25:787–97.
Liu RF, Fu G, Li J, Yang YF, Wang XG, Bai PD, et al. Roles of autophagy in androgen-induced benign prostatic hyperplasia in castrated rats. Knowledgeable Opin Ther Targets. 2018;15:2703–10.
Oh SH, Lee DW, Choi YB, Lee Y, Ju J. Measurement of autophagy flux in benign prostatic hyperplasia in vitro. Prostate Int. 2020;8:70–77.
Lu J, Su Y, Chen X, Chen Y, Luo P, Lin F, et al. Rapamycin‑induced autophagy attenuates hormone‑imbalance‑induced continual non‑bacterial prostatitis in rats by way of the inhibition of NLRP3 inflammasome‑mediated irritation. Mol Med Rep. 2019;19:221–30.
McIlwain DW, Zoetemelk M, Myers JD, Edwards M, Snider B, Jerde TJ. Coordinated induction of cell survival signaling within the infected microenvironment of the prostate. Prostate. 2016;76:722–34.
Zhang J, Zhang X, Cai Z, Li N, Li H. The lifetime danger and prognosis of continual prostatitis/continual pelvic ache syndrome within the middle-aged Chinese language males. Am J Mens Well being. 2019;13:1557988319865380.
Su Y, Lu J, Chen X, Liang C, Luo P, Qin C, et al. Rapamycin alleviates hormone imbalance-induced continual nonbacterial irritation in rat prostate by way of activating autophagy by way of the mTOR/ULK1/ATG13 signaling pathway. Irritation. 2018;41:1384–95.
Zhu Y, Yin Q, Wei D, Yang Z, Du Y, Ma Y. Autophagy in male copy. Syst Biol Reprod Med. 2019;65:265–72.
Kuma A, Komatsu M, Mizushima N. Autophagy-monitoring and autophagy-deficient mice. Autophagy. 2017;13:1619–28.
Deretic V, Saitoh T, Akira S. Autophagy in an infection, irritation and immunity. Nat Rev Immunol. 2013;13:722–37.
Silva JAF, Bruni-Cardoso A, Augusto TM, Damas-Souza D, Barbosa G, Felisbino SL, et al. Macrophage roles within the clearance of apoptotic cells and management of irritation within the prostate gland after castration. Prostate. 2018;78:95–103.
Clemens JQ, Meenan RT, O’Keeffe Rosetti MC, Kimes T, Calhoun E. Prevalence of and danger elements for prostatitis: inhabitants based mostly evaluation utilizing doctor assigned diagnoses. J Urol. 2007;178:1333–7.
Levine B, Mizushima N, Virgin HW. Autophagy in immunity and irritation. Nature. 2011;469:323–35.
Pei F, Wang HS, Chen Z, Zhang L. Autophagy regulates odontoblast differentiation by suppressing NF-kappaB activation in an inflammatory atmosphere. Cell Demise Dis. 2016;7:e2122.
Kashyap M, Pore S, Wang Z, Gingrich J, Yoshimura N, Tyagi P. Inflammasomes are essential mediators of prostatic irritation related to BPH. J Inflamm (Lond). 2015;12:37.
Choi S, Shin H, Tune H, Lim H. Suppression of autophagic activation within the mouse uterus by estrogen and progesterone. J Endocrinol. 2014;221:39–50.
Ng M, Baradhi KM. Benign prostatic hyperplasia. StatPearls (2023).
De Nunzio C, Giglio S, Baldassarri V, Cirombella R, Mallel G, Nacchia A, et al. Impairment of autophagy might characterize the molecular mechanism behind the connection between weight problems and irritation in sufferers with BPH and LUTS. Minerva Urol Nephrol. 2021;73:631–7.
De Nunzio C, Giglio S, Stoppacciaro A, Gacci M, Cirombella R, Luciani E, et al. Autophagy deactivation is related to extreme prostatic irritation in sufferers with decrease urinary tract signs and benign prostatic hyperplasia. Oncotarget. 2017;8:50904–10.
Lesovaya E, Kirsanov Ok, Antoshina E, Trukhanova L, Gorkova T, Shipaeva EV, et al. Rapatar, a nanoformulation of rapamycin, decreases chemically-induced benign prostate hyperplasia in rats. Oncotarget. 2015;6:9718–27.
Jiang MY, Han ZD, Li W, Yue F, Ye J, Li B, et al. Mitochondrion-associated protein peroxiredoxin 3 promotes benign prostatic hyperplasia by way of autophagy suppression and pyroptosis activation. Oncotarget. 2017;8:80295–302.
Michiels CF, Fransen P, De Munck DG, De Munck D, De Meyer G, Martinet W. Faulty autophagy in vascular easy muscle cells alters contractility and Ca(2)(+) homeostasis in mice. Am J Physiol Coronary heart Circ Physiol. 2015;308:557–67.
McCarthy CG, Wenceslau CF, Calmasini FB, Klee NS, Manufacturers MW, Joe B, et al. Reconstitution of autophagy ameliorates vascular operate and arterial stiffening in spontaneously hypertensive rats. Am J Physiol Coronary heart Circ Physiol. 2019;317:1013–27.
Li M, Yang X, Wang H, Xu E, Xi Z. Inhibition of androgen induces autophagy in benign prostate epithelial cells. Int J Urol. 2014;21:195–9.
Yang BY, Jiang CY, Dai CY, Zhao RZ, Wang XJ, Zhu YP, et al. 5-ARI induces autophagy of prostate epithelial cells by way of suppressing IGF-1 expression in prostate fibroblast. Cell Prolif. 2019;52:e12590.
Nickel JC, Gilling P, Tammela TL, Morrill B, Wilson T, Rittmaster RS. Comparability of dutasteride and finasteride for treating benign prostatic hyperplasia: the Enlarged Prostate Worldwide Comparator Examine (EPICS). BJU Int. 2011;108:388–94.
Rawla P. Epidemiology of prostate most cancers. World J Oncol. 2019;10:63–89.
Turner LS, Cheng JCM, Beckham TH, Keane TE, Norris JS, Liu X. Autophagy is elevated in prostate most cancers cells overexpressing acid ceramidase and enhances resistance to C6 ceramide. Prostate Most cancers Prostatic Dis. 2011;14:30–7.
Zhang B, Liu L. Autophagy is a double-edged sword within the remedy of colorectal most cancers. Oncol Lett. 2021;21:378.
Liu C, Xu P, Chen D, Fan X, Xu Y, Li M, et al. Roles of autophagy-related genes Beclin-1 and LC3 within the growth and development of prostate most cancers and benign prostatic hyperplasia. Biomed Rep. 2013;1:855–60.
Aita VM, Liang XH, Murty VV, Pincus D, Yu W, Cayanis E, et al. Cloning and genomic group of beclin 1, a candidate tumor suppressor gene on chromosome 17q21. Genomics. 1999;59:59–65.
White E, Di Paola RS. The double-edged sword of autophagy modulation in most cancers. Clin Most cancers Res. 2009;15:5308–16.
Ariosa AR, Lahiri V, Lei Y, Yang Y, Yin Z, Zhang Z, et al. A perspective on the function of autophagy in most cancers. Biochim Biophys Acta Mol Foundation Dis. 2021;1867:166262.
Pelekanou V, Castanas E. Androgen management in prostate most cancers. J Cell Biochem. 2016;117:2224–34.
Li M, Jiang X, Liu D, Na Y, Gao GF, Xi Z. Autophagy protects LNCaP cell beneath androgen deprivation circumstances. Autophagy. 2008;4:54–60.
Hu F, Zhao Y, Yu Y, Fang J, Cui R, Liu ZQ, et al. Docetaxel-mediated autophagy promotes chemoresistance in castration-resistant prostate most cancers cells by inhibiting STAT3. Most cancers Lett. 2018;416:24–30.
Yu Y, Yang FH, Zhang WT, Guo Y, Ye L, Yao XD. Mesenchymal stem cells desensitize castration-resistant prostate most cancers to docetaxel chemotherapy by way of inducing TGF-beta1-mediated cell autophagy. Cell Biosci. 2021;11:7.
Liu R, Zeng Y, Lei Z, Wang L, Yang H, Liu Z, et al. JAK/STAT3 signaling is required for TGF-beta-induced epithelial-mesenchymal transition in lung most cancers cells. Int J Oncol. 2014;44:1643–51.
O’Neill AJ, Prencipe M, Dowling C, Fan Y, Mulrane L, Gallagher WM, et al. Characterization and manipulation of docetaxel resistant prostate most cancers cell traces. Mol Most cancers. 2011;10:126.
Chang P, Wang T, Chang YC, Chu C, Lee C, Hsu HW, et al. Autophagy pathway is required for IL-6 induced neuroendocrine differentiation and chemoresistance of prostate most cancers LNCaP cells. PloS ONE. 2014;9:e88556.
Rodríguez-Berriguete G, Prieto A, Fraile B, Bouraoui Y, De Bethencourt FR, Martinez-Onsurbe P, et al. Relationship between IL-6/ERK and NF-kappaB: a research in regular and pathological human prostate gland. Eur Cytokine Netw. 2010;21:241–50.
Smart GJ, Marella VK, Talluri G, Shirazian D. Cytokine variations in sufferers with hormone handled prostate most cancers. J Urol. 2000;164:722–5.
Mahon KL, Henshall SM, Sutherland RL, Horvath LG. Pathways of chemotherapy resistance in castration-resistant prostate most cancers. Endocr Relat Most cancers. 2011;18:103–23.
Cristofani R, Montagnani Marelli M, Cicardi ME, Fontana F, Marzagalli M, Limonta P, et al. Twin function of autophagy on docetaxel-sensitivity in prostate most cancers cells. Cell Demise Dis. 2018;9:889.
Qiao Y, Choi JE, Tien JC, Simko SA, Rajendiran T, Vo JN, et al. Autophagy inhibition by focusing on PIKfyve potentiates response to immune checkpoint blockade in prostate most cancers. Nat Most cancers. 2021;2:978–93.
Lasorsa F, di Meo NA, Rutigliano M, Ferro M, Terracciano D, Tataru OS, et al. Rising hallmarks of metabolic reprogramming in prostate most cancers. Int J Mol Sci. 2023;24:910.
di Meo NA, Lasorsa F, Rutigliano M, Milella M, Ferro M, Battaglia M, et al. The darkish facet of lipid metabolism in prostate and renal carcinoma: novel insights into molecular diagnostic and biomarker discovery. Knowledgeable Rev Mol Diagn. 2023;23:297–313.
Loizzo D, Pandolfo SD, Rofers D, Cerrato C, di Meio NA, Autorino R, et al. Novel insights into autophagy and prostate most cancers: a complete evaluate. Int J Mol Sci. 2022;23:3826.
Strohecker AM, Guo JU, Karsli-Uzunbas G, Value SM, Chen GJ, Mathew R, et al. Autophagy sustains mitochondrial glutamine metabolism and progress of Braf600E-driven lung tumors. Most cancers Discov. 2013;3:1272–85.
Grossi V, Lucarelli G, Forte G, Peserico A, Matrone A, Germani A, et al. Lack of STK11 expression is an early occasion in prostate carginogenesis and predicts therapeutic response to focused remedy towards MAPK/p38. Autophagy. 2015;11:2102–13.
Roudsari N, Lashgari N, Momtaz S, Abaft S, Jamali F, Safaiepour P, et al. Inhibitors of the PI3K/Akt/mTOR pathway in prostate most cancers chemoprevention and intervention. Pharmaceutics. 2021;13:1195.
Wang Y, Mikhailova M, Bose S, Pan CX, deVere Whithe RW, Ghosh PM. Regulation of androgen receptor transcriptional exercise by rapamycin in prostate most cancers cell proliferation and survival. Oncogene. 2008;27:7106–17.
Ghosh PM, Malik SN, Bedolla RG, Wang Y, Mikhailova M, Prihoda TJ, et al. Sign transduction pathways in androgen-dependent and -independent prostate most cancers cell proliferation. Endocr Relat Most cancers. 2005;12:119–34.
Armstrong AJ, Netto GJ, Rudek MA, Halabi S, Wooden DP, Creel PA, et al. A pharmacodynamic research of rapamycin in males with intermediate- to high-risk localized prostate most cancers. Clin Most cancers Res. 2010;16:3057–66.
Farrow JM, Yang JC, Evans CP. Autophagy as a modulator and goal in prostate most cancers. Nat Rev Urol. 2014;11:508–16.
Shimizu S, Kanaseki T, Mizushima N, Mizuta T, Arakawa-Kobayashi S, Thompson CB, et al. Function of Bcl-2 household proteins in a non-apoptotic programmed cell demise depending on autophagy genes. Nat Cell Biol. 2004;6:1221–8.
Baspinar S, Bircan S, Orhan H, Kapucuoglu N, Bozkurt KK. The relation of beclin 1 and bcl-2 expressions in excessive grade prostatic intraepithelial neoplasia and prostate adenocarcinoma: a tissue microarray research. Pathol Res Pr. 2014;210:412–8.
Kim KY, Yun UJ, Yeom SH, Kim SC, Lee HJ, Ahn SC, et al. Inhibition of autophagy promotes hemistepsin a-induced apoptosis by way of reactive oxygen species-mediated AMPK-dependent signaling in human prostate most cancers cells. Biomolecules. 2021;11:1806.
Tan Q, Joshua AM, Wang M, Bristow RG, Wouters BG, Allen CJ, et al. Up-regulation of autophagy is a mechanism of resistance to chemotherapy and could be inhibited by pantoprazole to extend drug sensitivity. Most cancers Chemother Pharm. 2017;79:959–69.
Hansen AR, Tannock IF, Templeton A, Chen E, Evans A, Knox J, et al. Pantoprazole affecting docetaxel resistance pathways by way of autophagy (PANDORA): part II trial of excessive dose pantoprazole (autophagy inhibitor) with docetaxel in metastatic castration-resistant prostate most cancers (mCRPC). Oncologist. 2019;24:1188–94.
Chen Z, Jiang Q, Zhu P, Chen Y, Xie X, Du Z, et al. NPRL2 enhances autophagy and the resistance to Everolimus in castration-resistant prostate most cancers. Prostate. 2019;79:44–53.
Cao C, Subhawong T, Albert JM, Kim KW, Geng L, Sekhar KR, et al. Inhibition of mammalian goal of rapamycin or apoptotic pathway induces autophagy and radiosensitizes PTEN null prostate most cancers cells. Most cancers Res. 2006;66:10040–7.
Templeton AJ, Dutoit V, Cathomas R, Rothermundt C, Bärtschi D, Droge C, et al. Part 2 trial of single-agent everolimus in chemotherapy-naive sufferers with castration-resistant prostate most cancers (SAKK 08/08). Eur Urol. 2013;64:150–8.
George DJ, Halabi S, Healy P, Jonasch D, Anand M, Rasmussen J, et al. Part 2 medical trial of TORC1 inhibition with everolimus in males with metastatic castration-resistant prostate most cancers. Urol Oncol. 2020;38:e15–e22.
Rathkopf DE, Larson SM, Anand A, Morris MJ, Slovin SF, Shaffer DR, et al. Everolimus mixed with gefitinib in sufferers with metastatic castration-resistant prostate most cancers: Part 1/2 outcomes and signaling pathway implications. Most cancers. 2015;121:3853–61.
Vaishampayan U, Shevrin D, Stein M, Heilbrun L, Land S, Stark Ok, et al. Part II trial of carboplatin, everolimus, and prednisone in metastatic castration-resistant prostate most cancers pretreated with docetaxel chemotherapy: a prostate most cancers medical trial consortium research. Urology. 2015;86:1206–11.
Koshkin VS, Mir MV, Barata P, Gul A, Gupta R, Stephenson AJ, et al. Randomized part II trial of neoadjuvant everolimus in sufferers with high-risk localized prostate most cancers. Make investments N. Medicine. 2019;37:559–66.
Zedan MM, Mansour AK, Bakr AA, Sobh MA, Khodadadi H, Salles EL, et al. Impact of everolimus versus bone marrow-derived stem cells on glomerular harm in a rat mannequin of glomerulonephritis: a preventive, predictive and customized implication. Int J Mol Sci. 2021;23:344.
Armstrong AJ, Shen T, Halabi S, Kemeny G, Bitting RL, Kartcheske P, et al. A part II trial of temsirolimus in males with castration-resistant metastatic prostate most cancers. Clin Genitourin Most cancers. 2013;11:397–406.
Kruczek Ok, Ratterman M, Tolzien Ok, Sulo S, Lestingi TM, Nabhan C. A part II research evaluating the toxicity and efficacy of single-agent temsirolimus in chemotherapy-naive castration-resistant prostate most cancers. Br J Most cancers. 2013;109:1711–6.
McHugh DJ, Chudow J, DeNunzio M, Slovin SF, Danila DC, Morris MJ, et al. A part I trial of IGF-1R inhibitor cixutumumab and mTOR inhibitor temsirolimus in metastatic castration-resistant prostate most cancers. Clin Genitourin Most cancers. 2020;18:171–8.
Barata PC, Cooney M, Mendiratta P, Gupta R, Dreicer R, Garcia JA. Part I/II research evaluating the protection and medical efficacy of temsirolimus and bevacizumab in sufferers with chemotherapy refractory metastatic castration-resistant prostate most cancers. Make investments N Medicine. 2019;37:331–7.
Meulenbeld HJ, De Bono JS, Tagawa ST, Whang YE, Li X, Heath KH, et al. Tolerability, security and pharmacokinetics of ridaforolimus together with bicalutamide in sufferers with asymptomatic, metastatic castration-resistant prostate most cancers (CRPC). Most cancers Chemother Pharm. 2013;72:909–16.
Amato RJ, Wilding G, Bubley G, Loewy J, Haluska F, Gross ME. Security and preliminary efficacy evaluation of the mTOR inhibitor ridaforolimus in sufferers with taxane-treated, castration-resistant prostate most cancers. Clin Genitourin Most cancers. 2012;10:232–8.
Al-Qatati A, Aliwaini S. Mixed pitavastatin and dacarbazine therapy prompts apoptosis and autophagy leading to synergistic cytotoxicity in melanoma cells. Oncol Lett. 2017;14:7993–9.
Jo JH, Park HS, Lee DH, Han JH, Heo KS, Myung CS. Rosuvastatin inhibits the apoptosis of platelet-derived progress factor-stimulated vascular easy muscle cells by inhibiting p38 by way of autophagy. J Pharm Exp Ther. 2021;378:10–19.
Gorabi AM, Kiaie N, Aslani S, Sathyapalan T, Jamialahmadi T, Sahebkar A. Implications on the therapeutic potential of statins by way of modulation of autophagy. Longev OMAC. 2021;2021:9599608.
Toepfer N, Childress C, Parikh A, Rukstalis D, Yang W. Atorvastatin induces autophagy in prostate most cancers PC3 cells by way of activation of LC3 transcription. Most cancers Biol Ther. 2011;12:691–9.
He Z, Yuan J, Qi P, Zhang L, Wang Z. Atorvastatin induces autophagic cell demise in prostate most cancers cells in vitro. Mol Med Rep. 2015;11:4403–8.
Peltomaa AI, Raittinen P, Talala Ok, Taari Ok, Tammela TLJ, Auvinen A, et al. Prostate most cancers prognosis after initiation of androgen deprivation remedy amongst statins customers. A population-based cohort research. Prostate Most cancers Prostatic Dis. 2021;24:917–924.98.
Longo J, Freedland SJ, Penn LZ, Hamilton RJ. Statins and prostate cancer-hype or hope? The organic perspective. Prostate Most cancers Prostatic Dis. 2022;25:650–6.
Alfaqih MA, Allott EH, Hamilton RJ, Freeman MR, Freedland SJ. The present proof on statin use and prostate most cancers prevention: are we there but? Nat Rev Urol. 2017;14:107–19.
Ishola IO, Tijani HK, Dosumu OO, Anunobi CC, Oshodi TO. Atorvastatin attenuates testosterone-induced benign prostatic hyperplasia in rats: function of peroxisome proliferator-activated receptor-gamma and cyclo-oxygenase-2. Fundam Clin Pharm. 2017;31:652–62.
Allott EH, Csizmadi I, Howard LE, Muller RL, Moreira DM, Andriole GL, et al. Statin use and longitudinal modifications in prostate quantity; outcomes from the Discount by Dutasteride of prostate Most cancers Occasions (REDUCE) trial. BJU Int. 2020;125:226–33.
Hurwitz LM, Kulac I, Gumuskaya B, Valle JABD, Benedetti I, Pan F, et al. Use of aspirin and statins in relation to irritation in benign prostate tissue within the placebo arm of the prostate most cancers prevention trial. Most cancers Prev Res (Philos). 2020;13:853–62.
Zhang X, Zeng X, Dong L, Zhao X, Qu X. The results of statins on benign prostatic hyperplasia in aged sufferers with metabolic syndrome. World J Urol. 2015;33:2071–7.
Shih HJ, Huang CJ, Lin JA, Kao MC, Fan YC, Tsai PS. Hyperlipidemia is related to an elevated danger of medical benign prostatic hyperplasia. Prostate. 2018;78:113–20.
Mills IW, Crossland A, Patel A, Ramonas H. Atorvastatin therapy for males with decrease urinary tract signs and benign prostatic enlargement. Eur Urol. 2007;52:503–9.
Stamatiou KN, Zaglavira P, Skolarikos A, Sofras F. The results of lovastatin on standard medical therapy of decrease urinary tract signs with finasteride. Int Braz J Urol. 2008;34:555–61.
Tiwari R, Fleshner N. The function of metformin, statins and food regimen in males on lively surveillance for prostate most cancers. World J Urol. 2022;40:61–69.
American Diabetes Affiliation. Pharmacologic approaches to glycemic therapy: requirements of medical care in diabetes-2021. Diabetes Care. 2021;44:111–24.
Zaidi S, Gandhi J, Joshi G, Smith NL, Khan AS. The anticancer potential of metformin on prostate most cancers. Prostate Most cancers Prostatic Dis. 2023;22:351–61.
Chen C, Wang H, Geng X, Zhang D, Zhu Z, Zhang G, et al. Metformin exerts anti-AR-negative prostate most cancers exercise by way of AMPK/autophagy signaling pathway. Most cancers Cell Int. 2021;21:404.
Nguyen HG, Yang JC, Kung HJ, Shi XB, Tilki D, Lara PN Jr, et al. Concentrating on autophagy overcomes Enzalutamide resistance in castration-resistant prostate most cancers cells and improves therapeutic response in a xenograft mannequin. Oncogene. 2014;33:4521–30.
Joshua AM, Zannella VE, Downes MR, Hersey Ok, Koritzinsky M, Schwab M, et al. A pilot “window of alternative” neoadjuvant research of metformin in localized prostate most cancers. Prostate Most cancers Prostatic Dis. 2014;17:252–8.
Kuo YJ, Sung FC, Hsieh PF, Chang HP, Wu KL, Wu HC. Metformin reduces prostate most cancers danger amongst males with benign prostatic hyperplasia: A nationwide population-based cohort research. Most cancers Med. 2019;8:2514–23.
Pennanen P, Syvälä H, Bläuer M, Savinainen Ok, Ylikomi T, Tammela TLJ, et al. The results of metformin and simvastatin on the expansion of LNCaP and RWPE-1 prostate epithelial cell traces. Eur J Pharm. 2016;788:160–7.
Zhu W, Xu H, Ma J, Guo J, Xue W, Gu B, et al. An open-label pilot research of metformin as a concomitant remedy on sufferers with prostate most cancers present process androgen deprivation therapy. Urol Int. 2017;98:79–84.
Hong Y, Lee S, Received S. The preventive impact of metformin on development of benign prostate hyperplasia: a nationwide population-based cohort research in Korea. PloS ONE. 2019;14:e0219394.
Li SX, Li C, Pang XR, Zhang J, Yu GC, Yeo AJ, et al. Metformin attenuates silica-induced pulmonary fibrosis by activating autophagy by way of the AMPK-mTOR signaling pathway. Entrance Pharm. 2021;12:719589.
Eule CJ, Flaig TW, Wong Ok, Graf R, Lam ET. Effectiveness and sturdiness of advantage of mTOR inhibitors in a real-world cohort of sufferers with metastatic prostate most cancers and PI3K pathway alterations. Prostate Most cancers Prostatic Dis. 2022;26:188–93.

