22
Views
0
CrossRef citations to date
0
Altmetric
Research Article

S-(N,N-diethyldithiocarbamoyl)-N-acetyl-l-cysteine for the treatment of non-small cell lung cancer through regulating NF-κB signalling pathway without neurotoxicity

, , , , , , , , , , , & show all
Received 11 Mar 2024, Accepted 24 Jun 2024, Published online: 12 Jul 2024

References

  • Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–249. doi: 10.3322/caac.21660.
  • Schein CH. Repurposing approved drugs on the pathway to novel therapies. Med Res Rev. 2020;40(2):586–605. doi: 10.1002/med.21627.
  • Kannappan V, Ali M, Small B, et al. Recent advances in repurposing disulfiram and disulfiram derivatives as copper-dependent anticancer agents. Front Mol Biosci. 2021;8:741316.
  • Wu X, Xue X, Wang L, et al. Suppressing autophagy enhances disulfiram/copper-induced apoptosis in non-small cell lung cancer. Eur J Pharmacol. 2018;827:1–12. doi: 10.1016/j.ejphar.2018.02.039.
  • Zhang T, Kephart J, Bronson E, et al. Prospective clinical trial of disulfiram plus copper in men with metastatic castration-resistant prostate cancer. Prostate. 2022;82(7):858–866. doi: 10.1002/pros.24329.
  • Triscott J, Rose Pambid M, Dunn SE. Concise review: bullseye: targeting cancer stem cells to improve the treatment of gliomas by repurposing disulfiram. Stem Cells. 2015;33(4):1042–1046. doi: 10.1002/stem.1956.
  • Johansson B. A review of the pharmacokinetics and pharmacodynamics of disulfiram and its metabolites. Acta Psychiatr Scand Suppl. 1992;369:15–26. doi: 10.1111/j.1600-0447.1992.tb03310.x.
  • Angelova PR, Abramov AY. Role of mitochondrial ROS in the brain: from physiology to neurodegeneration. FEBS Lett. 2018;592(5):692–702. doi: 10.1002/1873-3468.12964.
  • Peddawad D, Nagendra S, Chatterjee R, et al. Fulminant encephalopathy with unusual brain imaging in disulfiram toxicity. Neurology. 2018;90(11):518–519. doi: 10.1212/WNL.0000000000005125.
  • Stone SL, Cartwright MS, Panea OR, et al. Neuromuscular ultrasound findings in polyneuropathy secondary to disulfiram. J Clin Neurophysiol. 2014;31(6):e18–e20. doi: 10.1097/WNP.0000000000000101.
  • Chen W, Ercal N, Huynh T, et al. Characterizing N-acetylcysteine (NAC) and N-acetylcysteine amide (NACA) binding for lead poisoning treatment. J Colloid Interface Sci. 2012;371(1):144–149. doi: 10.1016/j.jcis.2011.12.052.
  • Deepmala, Slattery J, Kumar N, et al. Clinical trials of N-acetylcysteine in psychiatry and neurology: a systematic review. Neurosci Biobehav Rev. 2015;55:294–321. doi: 10.1016/j.neubiorev.2015.04.015.
  • Zhang X, Wang Y-N, Zhu J-J, et al. N-acetylcysteine negatively regulates Notch3 and its malignant signaling. Oncotarget. 2016;7(21):30855–30866. doi: 10.18632/oncotarget.8806.
  • Kretzmann NA, Chiela E, Matte U, et al. N-acetylcysteine improves antitumoural response of Interferon alpha by NF-kB downregulation in liver cancer cells. Comp Hepatol. 2012;11(1):4. doi: 10.1186/1476-5926-11-4.
  • Auf der Maur P, Trefny MP, Baumann Z, et al. N-acetylcysteine overcomes NF1 loss-driven resistance to PI3Kα inhibition in breast cancer. Cell Rep Med. 2023;4(4):101002. doi: 10.1016/j.xcrm.2023.101002.
  • Lv H, Yang H, Duan Y, et al. A disulfiram derivative against lung cancer via the Notch signaling pathway without neurotoxicity and hepatotoxicity. Naunyn Schmiedebergs Arch Pharmacol. 2023;397(7):4747–4760.
  • Lee BH, Lee S, Kim YS, et al. Chemopreventive effects of S-(N,N-diethyldithiocarbamoyl)-N-acetyl-l-cysteine against benzo[a]pyrene. Mutat Res. 1997;377(2):167–175. doi: 10.1016/s0027-5107(97)00063-8.
  • Hu W, Wang R, Sun B. Meteorin-like ameliorates β cell function by inhibiting β cell apoptosis of and promoting β cell proliferation via activating the WNT/β-catenin pathway. Front Pharmacol. 2021;12:627147. doi: 10.3389/fphar.2021.627147.
  • Justus CR, Leffler N, Ruiz-Echevarria M, et al. In vitro cell migration and invasion assays. J Vis Exp. 2014;2014(88):51046. doi: 10.3791/51046.
  • Żyro D, Śliwińska A, Szymczak-Pajor I, et al. Light stability, pro-apoptotic and genotoxic properties of silver (I) complexes of metronidazole and 4-hydroxymethylpyridine against pancreatic cancer cells in vitro. Cancers. 2020;12(12):3848. doi: 10.3390/cancers12123848.
  • Lindén S, Singh MK, Wegner KD, et al. Terbium-based time-gated Förster resonance energy transfer imaging for evaluating protein–protein interactions on cell membranes. Dalton Trans. 2015;44(11):4994–5003. doi: 10.1039/c4dt02884h.
  • Zheng Z, Wang Y, Yu H, et al. Salvianolic acid B inhibits ototoxic drug-induced ototoxicity by suppression of the mitochondrial apoptosis pathway. J Cell Mol Med. 2020;24(12):6883–6897. doi: 10.1111/jcmm.15345.
  • Piña R, Santos-Díaz AI, Orta-Salazar E, et al. Ten approaches that improve immunostaining: a review of the latest advances for the optimization of immunofluorescence. Int J Mol Sci. 2022;23(3):1426.
  • Ye W, Lv H, Zhang Q, et al. A cisplatin and disulphiram co-loaded inclusion complex overcomes drug resistance by inhibiting cancer cell stemness in non-small cell lung cancer. J Drug Target. 2024;32(2):159–171. doi: 10.1080/1061186X.2023.2298844.
  • Onn A, Isobe T, Itasaka S, et al. Development of an orthotopic model to study the biology and therapy of primary human lung cancer in nude mice. Clin Cancer Res. 2003;9(15):5532–5539.
  • Ren X, Li Y, Zhou Y, et al. Overcoming the compensatory elevation of NRF2 renders hepatocellular carcinoma cells more vulnerable to disulfiram/copper-induced ferroptosis. Redox Biol. 2021;46:102122. doi: 10.1016/j.redox.2021.102122.
  • Zorzo C, Higarza SG, Méndez M, et al. High frequency repetitive transcranial magnetic stimulation improves neuronal activity without affecting astrocytes and microglia density. Brain Res Bull. 2019;150:13–20. doi: 10.1016/j.brainresbull.2019.05.004.
  • Zuo W, Zhang W, Han N, et al. Compound IMM-H004, a novel coumarin derivative, protects against CA1 cell loss and spatial learning impairments resulting from transient global ischemia. CNS Neurosci Ther. 2015;21(3):280–288. doi: 10.1111/cns.12364.
  • O’Leary K, Shia A, Schmid P. Epigenetic regulation of EMT in non-small cell lung cancer. Curr Cancer Drug Targets. 2018;18(1):89–96. doi: 10.2174/1568009617666170203162556.
  • Yang Q, Yao Y, Li K, et al. An updated review of disulfiram: molecular targets and strategies for cancer treatment. Curr Pharm Des. 2019;25(30):3248–3256. doi: 10.2174/1381612825666190816233755.
  • Gao P, Zhang H, Dinavahi R, et al. HIF-dependent antitumorigenic effect of antioxidants in vivo. Cancer Cell. 2007;12(3):230–238. doi: 10.1016/j.ccr.2007.08.004.
  • Wang W, McLeod HL, Cassidy J. Disulfiram-mediated inhibition of NF-kappaB activity enhances cytotoxicity of 5-fluorouracil in human colorectal cancer cell lines. Int J Cancer. 2003;104(4):504–511. doi: 10.1002/ijc.10972.
  • Ho MY, Mackey JR. Presentation and management of docetaxel-related adverse effects in patients with breast cancer. Cancer Manag Res. 2014;6:253–259. doi: 10.2147/CMAR.S40601.
  • Tagawa N, Sugiyama E, Tajima M, et al. Comparison of adverse events following injection of original or generic docetaxel for the treatment of breast cancer. Cancer Chemother Pharmacol. 2017;80(4):841–849. doi: 10.1007/s00280-017-3425-3.
  • Beam DR, Knight JM. Posterior reversible encephalopathy syndrome instigated by off-label disulfiram use for metastatic melanoma. Psychosomatics. 2020;61(3):302–306. doi: 10.1016/j.psym.2019.09.003.
  • Kwon HS, Koh SH. Neuroinflammation in neurodegenerative disorders: the roles of microglia and astrocytes. Transl Neurodegener. 2020;9(1):42. doi: 10.1186/s40035-020-00221-2.
  • Olude MA, Mouihate A, Mustapha OA, et al. Astrocytes and microglia in stress-induced neuroinflammation: the African perspective. Front Immunol. 2022;13:795089. doi: 10.3389/fimmu.2022.795089.
  • Xie Y, Lu W, Wang S, et al. Validation of the 12-gene predictive signature for adjuvant chemotherapy response in lung cancer. Clin Cancer Res. 2019;25(1):150–157. doi: 10.1158/1078-0432.CCR-17-2543.
  • Zheng Y, Wang R, Song H-Z, et al. Epigenetic downregulation of RUNX3 by DNA methylation induces docetaxel chemoresistance in human lung adenocarcinoma cells by activation of the AKT pathway. Int J Biochem Cell Biol. 2013;45(11):2369–2378. doi: 10.1016/j.biocel.2013.07.013.
  • Zou M, Hu X, Xu B, et al. Glutathione S‑transferase isozyme alpha 1 is predominantly involved in the cisplatin resistance of common types of solid cancer. Oncol Rep. 2019;41(2):989–998. doi: 10.3892/or.2018.6861.
  • Staff NP, Grisold A, Grisold W, et al. Chemotherapy-induced peripheral neuropathy: a current review. Ann Neurol. 2017;81(6):772–781. doi: 10.1002/ana.24951.
  • Nechushtan H, Hamamreh Y, Nidal S, et al. A phase IIb trial assessing the addition of disulfiram to chemotherapy for the treatment of metastatic non-small cell lung cancer. Oncologist. 2015;20(4):366–367. doi: 10.1634/theoncologist.2014-0424.
  • Chen D, Cui QC, Yang H, et al. Disulfiram, a clinically used anti-alcoholism drug and copper-binding agent, induces apoptotic cell death in breast cancer cultures and xenografts via inhibition of the proteasome activity. Cancer Res. 2006;66(21):10425–10433. doi: 10.1158/0008-5472.CAN-06-2126.
  • Kim JY, Lee N, Kim Y-J, et al. Disulfiram induces anoikis and suppresses lung colonization in triple-negative breast cancer via calpain activation. Cancer Lett. 2017;386:151–160. doi: 10.1016/j.canlet.2016.11.022.
  • Skrott Z, Mistrik M, Andersen KK, et al. Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature. 2017;552(7684):194–199. doi: 10.1038/nature25016.
  • Yip NC, Fombon IS, Liu P, et al. Disulfiram modulated ROS-MAPK and NFκB pathways and targeted breast cancer cells with cancer stem cell-like properties. Br J Cancer. 2011;104(10):1564–1574. doi: 10.1038/bjc.2011.126.
  • Aruoma OI, Halliwell B, Hoey BM, et al. The antioxidant action of N-acetylcysteine: its reaction with hydrogen peroxide, hydroxyl radical, superoxide, and hypochlorous acid. Free Radic Biol Med. 1989;6(6):593–597. doi: 10.1016/0891-5849(89)90066-x.
  • Rota C, Bergamini S, Daneri F, et al. N-Acetylcysteine negatively modulates nitric oxide production in endotoxin-treated rats through inhibition of NF-kappaB activation. Antioxid Redox Signal. 2002;4(1):221–226. doi: 10.1089/152308602753625988.
  • Zafarullah M, Li WQ, Sylvester J, et al. Molecular mechanisms of N-acetylcysteine actions. Cell Mol Life Sci. 2003;60(1):6–20. doi: 10.1007/s000180300001.
  • Smaga I, Frankowska M, Filip M. N-acetylcysteine as a new prominent approach for treating psychiatric disorders. Br J Pharmacol. 2021;178(13):2569–2594. doi: 10.1111/bph.15456.
  • Faria M, Prats E, Gómez-Canela C, et al. Therapeutic potential of N-acetylcysteine in acrylamide acute neurotoxicity in adult zebrafish. Sci Rep. 2019;9(1):16467. doi: 10.1038/s41598-019-53154-w.
  • Cirone M, D’Orazi G. NRF2 in cancer: cross-talk with oncogenic pathways and involvement in gammaherpesvirus-driven carcinogenesis. Int J Mol Sci. 2022;24(1):24. doi: 10.3390/ijms24010595.
  • Khusnurrokhman G, Wati FF. Tumor-promoting inflammation in lung cancer: a literature review. Ann Med Surg. 2022;79:104022. doi: 10.1016/j.amsu.2022.104022.
  • Rasmi RR, Sakthivel KM, Guruvayoorappan C. NF-κB inhibitors in treatment and prevention of lung cancer. Biomed Pharmacother. 2020;130:110569. doi: 10.1016/j.biopha.2020.110569.
  • Yan M, Fan X, Si H, et al. Association between gene polymorphism and adverse effects in cancer patients receiving docetaxel treatment: a meta-analysis. Cancer Chemother Pharmacol. 2022;89(2):173–181. doi: 10.1007/s00280-021-04374-3.
  • Tofthagen C, McAllister RD, Visovsky C. Peripheral neuropathy caused by paclitaxel and docetaxel: an evaluation and comparison of symptoms. J Adv Pract Oncol. 2013;4(4):204–215.
  • Ahmad S, Singh V, Gautam HK, et al. Multisampling-based docking reveals imidazolidinyl urea as a multitargeted inhibitor for lung cancer: an optimisation followed multi-simulation and in-vitro study. J Biomol Struct Dyn. 2024;42(5):2494–2511. doi: 10.1080/07391102.2023.2209673.
  • Ahmad S, Raza K. Identification of 5-nitroindazole as a multitargeted inhibitor for CDK and transferase kinase in lung cancer: a multisampling algorithm-based structural study. Mol Divers. 2023. doi: 10.1007/s11030-023-10648-0.

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.