1,597
Views
0
CrossRef citations to date
0
Altmetric
Research Article

Identification and study of cuproptosis-related genes in prognostic model of multiple myeloma

, , , , , , & show all
Article: 2249217 | Received 31 Mar 2023, Accepted 11 Aug 2023, Published online: 23 Aug 2023

References

  • Tai YT, Cho SF, Anderson KC. Osteoclast immunosuppressive effects in multiple myeloma: role of programmed cell death ligand 1. Front Immunol. 2018;9:1822. doi:10.3389/fimmu.2018.01822
  • Rajkumar SV, Blood E, Vesole D, et al. Phase Iii clinical trial of thalidomide plus dexamethasone compared with dexamethasone alone in newly diagnosed multiple myeloma: A clinical trial coordinated by the eastern cooperative oncology group. J Clin Oncol. 2006;24(3):431–436. doi:10.1200/JCO.2005.03.0221
  • Laubach JP, Voorhees PM, Hassoun H, et al. Current strategies for treatment of relapsed/refractory multiple myeloma. Expert Rev Hematol. 2014;7(1):97–111. doi:10.1586/17474086.2014.882764
  • Sonneveld P, Avet-Loiseau H, Lonial S, et al. Treatment of multiple myeloma with high-risk cytogenetics: A consensus of the international myeloma working group. Blood. 2016;127(24):2955–2962. doi:10.1182/blood-2016-01-631200
  • Ruiz LM, Libedinsky A, Elorza AA. Role of copper on mitochondrial function and metabolism. Front Mol Biosci. 2021;8:711227. doi:10.3389/fmolb.2021.711227
  • Oliveri V. Selective targeting of cancer cells by copper ionophores: An overview. Front Mol Biosci. 2022;9:841814. doi:10.3389/fmolb.2022.841814
  • Gupte A, Mumper RJ. Elevated copper and oxidative stress in cancer cells as a target for cancer treatment. Cancer Treat Rev. 2009;35(1):32–46. doi:10.1016/j.ctrv.2008.07.004
  • Rezaei A, Khanamani Falahati-Pour S, Mohammadizadeh F, et al. Effect of a copper (Ii) complex on the induction of apoptosis in human hepatocellular carcinoma cells. Asian Pac J Cancer Prev. 2018;19(10):2877–2884. doi:10.22034/apjcp.2018.19.10.2877
  • Li H, Wang J, Wu C, et al. The combination of disulfiram and copper for cancer treatment. Drug Discov Today. 2020;25(6):1099–1108. doi:10.1016/j.drudis.2020.04.003
  • Xu Y, Zhou Q, Feng X, et al. Disulfiram/copper markedly induced myeloma cell apoptosis through activation of Jnk and intrinsic and extrinsic apoptosis pathways. Biomed Pharmacother Biomed Pharmacotherapie. 2020;126:110048. doi:10.1016/j.biopha.2020.110048
  • Agarwal A, Khandelwal A, Pal K, et al. A novel Pro-oxidant combination of resveratrol and copper reduces transplant related toxicities in patients receiving high dose melphalan for multiple myeloma (Rescu 001). PloS one. 2022;17(2):e0262212. doi:10.1371/journal.pone.0262212
  • Tsvetkov P, Coy S, Petrova B, et al. Copper induces cell death by targeting lipoylated Tca cycle proteins. Science (New York, NY). 2022;375(6586):1254–1261. doi:10.1126/science.abf0529
  • Gaggelli E, Kozlowski H, Valensin D, et al. Copper homeostasis and neurodegenerative disorders (Alzheimer's, prion, and Parkinson's diseases and amyotrophic lateral sclerosis). Chem Rev. 2006;106:1995–2044. doi:10.1021/cr040410w
  • Kahlson MA, Dixon SJ. Copper-induced cell death. Science (New York, NY). 2022;375(6586):1231–1232. doi:10.1126/science.abo3959
  • Dong C, Dang D, Zhao X, et al. Integrative characterization of the role of Il27 in melanoma using bioinformatics analysis. Front Immunol. 2021;12:713001. doi:10.3389/fimmu.2021.713001
  • Wang Y, Wang Z, Sun J, et al. Identification of Hcc subtypes with different prognosis and metabolic patterns based on mitophagy. Front Cell Dev Biol. 2021;9:799507. doi:10.3389/fcell.2021.799507
  • Shi H, Hou Y, Su X, et al. Mechanism of action of Tripterygium wilfordii for treatment of idiopathic membranous nephropathy based on network pharmacology. Renal Fail. 2022;44(1):116–125. doi:10.1080/0886022X.2021.2024850
  • Zhang M, Zhu K, Pu H, et al. An immune-related signature predicts survival in patients with lung adenocarcinoma. Front Oncol. 2019;9:1314. doi:10.3389/fonc.2019.01314
  • Wang S, Su W, Zhong C, et al. An eight-circrna assessment model for predicting biochemical recurrence in prostate cancer. Front Cell Dev Biol. 2020;8:599494. doi:10.3389/fcell.2020.599494
  • Liu TT, Li R, Huo C, et al. Identification of Cdk2-related immune forecast model and cerna in lung adenocarcinoma, a Pan-cancer analysis. Front Cell Dev Biol. 2021;9:682002. doi:10.3389/fcell.2021.682002
  • Fang Y, Huang S, Han L, et al. Comprehensive analysis of peritoneal metastasis sequencing data to identify Linc00924 as a prognostic biomarker in gastric cancer. Cancer Manag Res. 2021;13:5599–5611. doi:10.2147/CMAR.S318704
  • Ding C, Shan Z, Li M, et al. Characterization of the fatty acid metabolism in colorectal cancer to guide clinical therapy. Mol Ther Oncolytics. 2021;20:532–544. doi:10.1016/j.omto.2021.02.010
  • Leleu X, Fouquet G, Richez V, et al. Carfilzomib weekly plus melphalan and prednisone in newly diagnosed transplant-ineligible multiple myeloma (Ifm 2012-03): A phase I trial. Clin Cancer Res Official J Am Assoc Cancer Res. 2019;25(14):4224–4230. doi:10.1158/1078-0432.CCR-18-3642
  • Späth F, Wibom C, Krop EJM, et al. Immune marker changes and risk of multiple myeloma: A nested case-control study using repeated Pre-diagnostic blood samples. Haematologica. 2019;104(12):2456–2464. doi:10.3324/haematol.2019.216895
  • Liu W, Liu J, Song Y, et al. Mortality of lymphoma and myeloma in China, 2004-2017: An observational study. J Hematol Oncol. 2019;12(1):22. doi:10.1186/s13045-019-0706-9
  • Kyle RA, Durie BG, Rajkumar SV, et al. Monoclonal gammopathy of undetermined significance (mgus) and smoldering (asymptomatic) multiple myeloma: imwg consensus perspectives risk factors for progression and guidelines for monitoring and management. Leukemia. 2010;24(6):1121–1127. doi:10.1038/leu.2010.60
  • Hayes KE, Batsomboon P, Chen WC, et al. Inhibition of the Fad containing Er oxidoreductin 1 (Ero1) protein by En-460 as a strategy for treatment of multiple myeloma. Bioorg Med Chem. 2019;27(8):1479–1488. doi:10.1016/j.bmc.2019.02.016
  • Szalat R, Munshi NC. Novel agents in multiple myeloma. Cancer J (Sudbury, Mass). 2019;25(1):45–53. doi:10.1097/PPO.0000000000000355
  • Rayess H, Wang MB, Srivatsan ES. Cellular senescence and tumor suppressor gene P16. Int J Cancer. 2012;130(8):1715–1725. doi:10.1002/ijc.27316
  • Kimura H, Klein AP, Hruban RH, et al. The role of inherited pathogenic Cdkn2a variants in susceptibility to pancreatic cancer. Pancreas. 2021;50(8):1123–1130. doi:10.1097/MPA.0000000000001888
  • Śledzińska P, Bebyn MG, Furtak J, et al. Prognostic and predictive biomarkers in gliomas. Int J Mol Sci. 2021;22(19):10373. doi:10.3390/ijms221910373
  • Bui TO, Dao VT, Nguyen VT, et al. Genomics of clear-cell renal cell carcinoma: A systematic review and meta-analysis. Eur Urol. 2022;81(4):349–361. doi:10.1016/j.eururo.2021.12.010
  • Smith EM, Boyd K, Davies FE. The potential role of epigenetic therapy in multiple myeloma. Br J Haematol. 2010;148(5):702–713. doi:10.1111/j.1365-2141.2009.07976.x
  • Maldonado V, Melendez-Zajgla J. Role of Bcl-3 in solid tumors. Mol Cancer. 2011;10:152. doi:10.1186/1476-4598-10-152
  • Wulczyn FG, Naumann M, Scheidereit C. Candidate proto-oncogene Bcl-3 encodes a subunit-specific inhibitor of transcription factor Nf-kappa B. Nature. 1992;358(6387):597–599. doi:10.1038/358597a0
  • Brocke-Heidrich K, Ge B, Cvijic H, et al. Bcl3 Is induced by Il-6 Via Stat3 binding to intronic enhancer Hs4 and represses its own transcription. Oncogene. 2006;25(55):7297–7304. doi:10.1038/sj.onc.1209711
  • Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science (New York, NY). 2011;331(6024):1565–1570. doi:10.1126/science.1203486
  • Guillerey C, Nakamura K, Vuckovic S, et al. Immune responses in multiple myeloma: role of the natural immune surveillance and potential of immunotherapies. Cell Mol Life Sci: CMLS. 2016;73(8):1569–1589. doi:10.1007/s00018-016-2135-z
  • Leone P, Berardi S, Frassanito MA, et al. Dendritic cells accumulate in the bone marrow of myeloma patients where they protect tumor plasma cells from Cd8+ T-cell killing. Blood. 2015;126(12):1443–1451. doi:10.1182/blood-2015-01-623975
  • Leone P, Di Lernia G, Solimando AG, et al. Bone marrow endothelial cells sustain a tumor-specific Cd8(+) T cell subset with suppressive function in myeloma patients. Oncoimmunology. 2019;8(1):e1486949. doi:10.1080/2162402X.2018.1486949
  • Chae HD, Dutta R, Tiu B, et al. RSK inhibitor BI-D1870 inhibits acute myeloid leukemia cell proliferation by targeting mitotic exit. Oncotarget. 2020;11(25):2387–2403. doi:10.18632/oncotarget.27630
  • Shimura Y, Kuroda J, Ri M, et al. RSK2(Ser227) at N-terminal kinase domain is a potential therapeutic target for multiple myeloma. Mol Cancer Ther. 2012;11(12):2600–2609. doi:10.1158/1535-7163.MCT-12-0605
  • Chaudhary RK, Patil P, Ananthesh L, et al. Identification of signature genes and drug candidates for primary plasma cell leukemia: An integrated system biology approach. Comput Biol Med. 2023;8(162):107090. doi:10.1016/j.compbiomed.2023.107090
  • Heo JY, Kim HJ, Kim SM, et al. Embelin suppresses STAT3 signaling, proliferation, and survival of multiple myeloma via the protein tyrosine phosphatase PTEN. Cancer Lett. 2011;308(1):71–80. doi:10.1016/j.canlet.2011.04.015
  • Kelly RJ, Rixe O. Axitinib. (AG-013736). Recent Results Cancer Res. 2010;184:33–44. doi:10.1007/978-3-642-01222-8_3
  • Motzer RJ, Robbins PB, Powles T, et al. Avelumab plus axitinib versus sunitinib in advanced renal cell carcinoma: biomarker analysis of the phase 3 JAVELIN renal 101 trial. Nat Med. 2020;26(11):1733–1741. doi:10.1038/s41591-020-1044-8
  • Motzer RJ, Penkov K, Haanen J, et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380(12):1103–1115. doi:10.1056/NEJMoa1816047
  • Xiang RF, Wang Y, Zhang N, et al. MK2206 enhances the cytocidal effects of bufalin in multiple myeloma by inhibiting the AKT/mTOR pathway. Cell Death Dis. 2017;8(5):e2776. doi:10.1038/cddis.2017.188
  • Ramakrishnan V, Kimlinger T, Haug J, et al. Anti-myeloma activity of Akt inhibition is linked to the activation status of PI3 K/Akt and MEK/ERK pathway. PLoS One. 2012;7(11):e50005. doi:10.1371/journal.pone.0050005
  • Dahl ES, Aird KM. Ataxia-Telangiectasia mutated modulation of carbon metabolism in cancer. Front Oncol. 2017;29(7):291. doi:10.3389/fonc.2017.00291
  • Jin MH, Oh DY. ATM in DNA repair in cancer. Pharmacol Therap. 2019;203:107391. doi:10.1016/j.pharmthera.2019.07.002
  • Harris BRE, Zhang Y, Tao J, et al. ATM inhibitor KU-55933 induces apoptosis and inhibits motility by blocking GLUT1-mediated glucose uptake in aggressive cancer cells with sustained activation of Akt. FASEB J. 2021;35(4):e21264. doi:10.1096/fj.202001415RR
  • Shi Y, Ma IT, Patel RH, et al. NSC-87877 inhibits DUSP26 function in neuroblastoma resulting in p53-mediated apoptosis. Cell Death Dis. 2015;6(8):e1841. doi:10.1038/cddis.2015.207