1,522
Views
27
CrossRef citations to date
0
Altmetric
Research Paper

The novel autophagy inhibitor elaiophylin exerts antitumor activity against multiple myeloma with mutant TP53 in part through endoplasmic reticulum stress-induced apoptosis

, , , , , , & show all
Pages 584-595 | Received 09 Jan 2017, Accepted 17 Jun 2017, Published online: 11 Aug 2017

References

  • Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin 2013; 63:11-30; PMID:23335087; https://doi.org/10.3322/caac.21166
  • Usmani SZ, Crowley J, Hoering A, Mitchell A, Waheed S, Nair B, AlSayed Y, Vanrhee F, Barlogie B. Improvement in long-term outcomes with successive Total Therapy trials for multiple myeloma: are patients now being cured? Leukemia 2013; 27(1):226-32; PMID:22705990; https://doi.org/10.1038/leu.2012.160
  • Adams J. Proteasome inhibition in cancer: Development of PS-341. Semin Oncol 2001; 28(6):613-9; PMID:11740819
  • Jung L, Holle L, Dalton WS. Discovery, Development, and clinical applications of bortezomib. Oncology 2004; 18(14 Suppl 11):4-13; PMID:15688597
  • Moreau P, Richardson PG, Cavo M, Orlowski RZ, San Miguel JF, Palumbo A, Harousseau JL. Proteasome inhibitors in multiple myeloma: 10 years later. Blood 2012; 120(5):947-59; PMID:22645181; https://doi.org/10.1182/blood-2012-04-403733
  • Mateos MV. Management of treatment-related adverse events in patients with multiple myeloma. Cancer Treat Rev 2010; 36(Suppl 2):S24-32; PMID:20472185; https://doi.org/10.1016/S0305-7372(10)70009-8
  • Vogelstein B, Lane D, Levine AJ. Surfing the p53 network. Nature 2000; 408:307-10; PMID:11099028; https://doi.org/10.1038/35042675
  • Saha MN, Jiang H, Jayakar J, Reece D, Branch DR, Chang H. MDM2 antagonist nutlin plus proteasome inhibitor velcade combination displays a synergistic anti-myeloma activity. Cancer Biol Ther 2010; 9:936-44; PMID:20418664; https://doi.org/10.4161/cbt.9.11.11882
  • Drach J, Ackermann J, Fritz E, Kromer E, Schuster R, Gisslinger H, DeSantis M, Zojer N, Fiegl M, Roka S, et al. Presence of a p53 gene deletion in patients with multiple myeloma predicts for short survival after conventional-dose chemotherapy. Blood 1998; 92:802-9; PMID:9680348
  • Vousden KH, Lu X. Live or let die: the cell's response to p53. Nat Rev Cancer 2002; 2:594-604; PMID:12154352; https://doi.org/10.1038/nrc864
  • Chng WJ, Price-Troska T, Gonzalez-Paz N, Van Wier S, Jacobus S, Blood E, Henderson K, Oken M, Van Ness B, Greipp P, et al. Clinical significance of TP53 mutation in myeloma. Leukemia 2007; 21(3):582-4; PMID:17215851; https://doi.org/10.1038/sj.leu.2404524
  • Tiedemann RE, Gonzalez-Paz N, Kyle RA, Santana-Davila R, Price-Troska T, Van Wier SA, Chng WJ, Ketterling RP, Gertz MA, Henderson K, et al. Genetic aberrations and survival in plasma cell leukemia. Leukemia 2008; 22(5):1044-52; PMID:18216867; https://doi.org/10.1038/leu.2008.4
  • Teoh PJ, Chung TH, Sebastian S, Choo SN, Yan J, Ng SB, Fonseca R, Chng WJ. p53 haploinsufficiency and functional abnormalities in multiple myeloma. Leukemia 2014; 28(10):2066-74; PMID:24625551; https://doi.org/10.1038/leu.2014.102
  • Auner HW, Cenci S. Recent advances and future directions in targeting the secretory apparatus in multiple myeloma. Br J Haematol 2015; 168:14-25; PMID:25296649; https://doi.org/10.1111/bjh.13172
  • Mizushima N, Levine B, Cuervo AM. Klionsky DJ Autophagy fights disease through cellular self-digestion. Nature 2008; 451(7182):1069-75; PMID:18305538; https://doi.org/10.1038/nature06639
  • Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell 2008; 132:27-42; PMID:18191218; https://doi.org/10.1016/j.cell.2007.12.018
  • Aronson LI, Davenport EL, Giuntoli SG, Srikanth M, Smith E, Morgan GJ, Davies F. Autophagy is a key myeloma survival pathway that can be manipulated therapeutically to enhance apoptosis. Blood 2010;116(21):4083; https://www.bloodjournal.org/content/116/21/4083
  • Hoang B, Benavides A, Shi Y, Frost P, Lichtenstein A. Effect of autophagy on multiple myeloma cell viability. Mol Cancer Ther 2009; 8(7):1974-84; PMID:19509276; https://doi.org/10.1158/1535-7163.MCT-08-1177
  • Carroll RG, Martin SJ. Autophagy in multiple myeloma: what makes you stronger can also kill you. Cancer cell 2013; 23(4):425-6; PMID:23597558; https://doi.org/10.1016/j.ccr.2013.04.001
  • Gorlach A, Klappa P, Kietzmann T. The endoplasmic reticulum: folding, calcium homeostasis, signaling, and redox control. Antiox Redox Signal 2006; 8:1391-418; PMID:16986999; https://doi.org/10.1089/ars.2006.8.1391
  • Verfaillie T, Salazar M, Velasco G, Agostinis P. Linking ER stress to autophagy: Potential implications for cancer therapy. Int J Cell Biol 2010; 2010:930509; PMID:20145727; https://doi.org/10.1155/2010/930509
  • Zhao X, Fang Y, Yang Y, Qin Y, Wu P, Wang T, Lai H, Meng L, Wang D, Zheng Z, et al. Elaiophylin, a novel autophagy inhibitor, exerts antitumor activity as a single agent in ovarian cancer cells. Autophagy 2015; 11(10):1849-63; PMID:25893854; https://doi.org/10.1080/15548627.2015.1017185
  • Arai M. Azalomycins B and F, two new antibiotics. I. Production and isolation. J Antibiot 1960; 13: 46-50; PMID:13848830
  • Bianchi G, Oliva L, Cascio P, Pengo N, Fontana F, Cerruti F, Orsi A, Pasqualetto E, Mezghrani A, Calbi V, et al. The proteasome load versus capacity balance determines apoptotic sensitivity of multiple myeloma cells to proteasome inhibition. Blood 2009; 113:3040-9; PMID:19164601; https://doi.org/10.1182/blood-2008-08-172734
  • Mizushima N, Klionsky DJ. Protein turnover via autophagy: Implications for metabolism. Ann Rev Nutr 2007; 27:19-40; PMID:17311494; https://doi.org/10.1146/annurev.nutr.27.061406.093749
  • Deretic V, Saitoh T, Akira S. Autophagy in infection, inflammation and immunity. Nat Rev Immunol 2013; 13:722-37; PMID:24064518; https://doi.org/10.1038/nri3532
  • Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-Arozena A, Adeli K, Agholme L, Agnello M, Agostinis P, Aguirre-Ghiso JA, et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 2012; 8:445-544; PMID:22966490; https://doi.org/10.4161/auto.19496
  • Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, Øvervatn A, Bjørkøy G, Johansen T. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 2007; 282:24131-45; PMID:17580304; https://doi.org/10.1074/jbc.M702824200
  • Moretti L, Cha YI, Niermann KJ, Lu B. Switch between apoptosis and autophagy: radiation-induced endoplasmic reticulum stress? Cell cycle 2007; 6:793-8; PMID:17377498; https://doi.org/10.4161/cc.6.7.4036
  • Codogno P, Meijer AJ. Autophagy and signaling: their role in cell survival and cell death. Cell Death Differ 2005; 12(Suppl 2):1509-18; PMID:16247498; https://doi.org/10.1038/sj.cdd.4401751
  • Bernales S, McDonald KL, Walter P. Autophagy counterbalances endoplasmic reticulum expansion during the unfolded protein response. PLoS Biol 2006; 4:e423; PMID:17132049; https://doi.org/10.1371/journal.pbio.0040423
  • Ozcan U, Yilmaz E, Ozcan L, Furuhashi M, Vaillancourt E, Smith RO, Görgün CZ, Hotamisligil GS. Chemical chaperones reduce ER stress and restore glucose homeostasis in a mouse model of type 2 diabetes. Science 2006; 313:1137-40; PMID:16931765; https://doi.org/10.1126/science.1128294
  • Vandewynckel YP, Laukens D, Devisscher L, Paridaens A, Bogaerts E, Verhelst X, Van den Bussche A, Raevens S, Van Steenkiste C, Van Troys M, et al. Tauroursodeoxycholic acid dampens oncogenic apoptosis induced by endoplasmic reticulum stress during hepatocarcinogen exposure. Oncotarget 2015; 6:28011-25; PMID:26293671; https://doi.org/10.18632/oncotarget.4377
  • Lin J, Zhang W, Zhao JJ, Kwart AH, Yang C, Ma D, Ren X, Tai YT, Anderson KC, Handin RI, et al. A clinically relevant in vivo zebrafish model of human multiple myeloma to study preclinical therapeutic efficacy. Blood 2016; 128:249-52; PMID:27207793; https://doi.org/10.1182/blood-2016-03-704460
  • Clarke HJ, Chambers JE, Liniker E, Marciniak SJ. Endoplasmic reticulum stress in malignancy. Cancer cell 2014; 25:563-73; PMID:24823636; https://doi.org/10.1016/j.ccr.2014.03.015
  • Obeng EA, Carlson LM, Gutman DM, Harrington WJ, Jr., Lee KP, Boise LH. Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells. Blood 2006; 107:4907-16; PMID:16507771; https://doi.org/10.1182/blood-2005-08-3531
  • Hiramatsu N, Chiang WC, Kurt TD, Sigurdson CJ, Lin JH. Multiple mechanisms of unfolded protein response-induced cell death. Am J Pathol 2015; 185:1800-8; PMID:25956028; https://doi.org/10.1016/j.ajpath.2015.03.009
  • Walter P, Ron D. The unfolded protein response: From stress pathway to homeostatic regulation. Science 2011; 334:1081-6; PMID:22116877; https://doi.org/10.1126/science.1209038
  • Xie Q, Khaoustov VI, Chung CC, Sohn J, Krishnan B, Lewis DE, Yoffe B. Effect of tauroursodeoxycholic acid on endoplasmic reticulum stress-induced caspase-12 activation. Hepatology 2002; 36:592-601; PMID:12198651; https://doi.org/10.1053/jhep.2002.35441
  • Aronson LI, Davies FE. DangER: Protein ovERload. Targeting protein degradation to treat myeloma. Haematologica 2012; 97:1119-30; PMID:22580998; https://doi.org/10.3324/haematol.2012.064923
  • Kawaguchi T, Miyazawa K, Moriya S, Ohtomo T, Che XF, Naito M, Itoh M, Tomoda A. Combined treatment with bortezomib plus bafilomycin A1 enhances the cytocidal effect and induces endoplasmic reticulum stress in U266 myeloma cells: crosstalk among proteasome, autophagy-lysosome and ER stress. Int J Oncol 2011; 38:643-54; PMID:21174067; https://doi.org/10.3892/ijo.2010.882
  • Davenport EL, Moore HE, Dunlop AS, Sharp SY, Workman P, Morgan GJ, Davies FE. Heat shock protein inhibition is associated with activation of the unfolded protein response pathway in myeloma plasma cells. Blood 2007; 110:2641-9; PMID:17525289; https://doi.org/10.1182/blood-2006-11-053728
  • Mitsiades CS, Mitsiades NS, McMullan CJ, Poulaki V, Kung AL, Davies FE, Morgan G, Akiyama M, Shringarpure R, Munshi NC, et al. Antimyeloma activity of heat shock protein-90 inhibition. Blood 2006; 107:1092-100; PMID:16234364; https://doi.org/10.1182/blood-2006-11-053728
  • Davenport EL, Zeisig A, Aronson LI, Moore HE, Hockley S, Gonzalez D, Smith EM, Powers MV, Sharp SY, Workman P, et al. Targeting heat shock protein 72 enhances Hsp90 inhibitor-induced apoptosis in myeloma. Leukemia 2010; 24:1804-7; PMID:20703255; https://doi.org/10.1038/leu.2010.168
  • Richardson PG, Mitsiades CS, Laubach JP, Lonial S, Chanan-Khan AA, Anderson KC. Inhibition of heat shock protein 90 (HSP90) as a therapeutic strategy for the treatment of myeloma and other cancers. Br J Haematol 2011; 152:367-79; PMID:21219297; https://doi.org/10.1111/j.1365-2141.2010.08360.x
  • Kouroku Y, Fujita E, Tanida I, Ueno T, Isoai A, Kumagai H, Ogawa S, Kaufman RJ, Kominami E, Momoi T. ER stress (PERK/eIF2alpha phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ 2007; 14:230-9; PMID:16794605; https://doi.org/10.1038/sj.cdd.4401984
  • Ogata M, Hino S, Saito A, Morikawa K, Kondo S, Kanemoto S, Murakami T, Taniguchi M, Tanii I, Yoshinaga K, et al. Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol 2006; 26:9220-31; PMID:17030611; https://doi.org/10.1128/MCB.01453-06
  • Michallet AS, Mondiere P, Taillardet M, Leverrier Y, Genestier L, Defrance T. Compromising the unfolded protein response induces autophagy-mediated cell death in multiple myeloma cells. PloS one 2011; 6:e25820; PMID:22028791; https://doi.org/10.1371/journal.pone.0025820
  • Lwin ST, Edwards CM, Silbermann R. Preclinical animal models of multiple myeloma. BoneKEy Rep 2016; 5:772; PMID:26909147; https://doi.org/10.1038/bonekey.2015.142
  • Vincenz L, Jager R, O'Dwyer M, Samali A. Endoplasmic reticulum stress and the unfolded protein response: targeting the Achilles heel of multiple myeloma. Mol Cancer Ther 2013; 12:831-43; PMID:23729400; https://doi.org/10.1158/1535-7163.MCT-12-0782
  • Eisenberg-Lerner A, Bialik S, Simon HU, Kimchi A. Life and death partners: apoptosis, autophagy and the cross-talk between them. Cell Death Differ 2009; 16:966-75; PMID:19325568; https://doi.org/10.1038/cdd.2009.33
  • Maiuri MC, Zalckvar E, Kimchi A, Kroemer G. Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol 2007; 8:741-52; PMID:17717517; https://doi.org/10.1038/nrm2239

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.