Generic placeholder image

Recent Patents on Anti-Cancer Drug Discovery

Editor-in-Chief

ISSN (Print): 1574-8928
ISSN (Online): 2212-3970

Review Article

Repurposing Disulfiram as An Anti-Cancer Agent: Updated Review on Literature and Patents

Author(s): Elmira Ekinci, Sagar Rohondia, Raheel Khan and Qingping P. Dou*

Volume 14, Issue 2, 2019

Page: [113 - 132] Pages: 20

DOI: 10.2174/1574892814666190514104035

Price: $65

Abstract

Background: Despite years of success of most anti-cancer drugs, one of the major clinical problems is inherent and acquired resistance to these drugs. Overcoming the drug resistance or developing new drugs would offer promising strategies in cancer treatment. Disulfiram, a drug currently used in the treatment of chronic alcoholism, has been found to have anti-cancer activity.

Objective: To summarize the anti-cancer effects of Disulfiram through a thorough patent review.

Methods: This article reviews molecular mechanisms and recent patents of Disulfiram in cancer therapy.

Results: Several anti-cancer mechanisms of Disulfiram have been proposed, including triggering oxidative stress by the generation of reactive oxygen species, inhibition of the superoxide dismutase activity, suppression of the ubiquitin-proteasome system, and activation of the mitogen-activated protein kinase pathway. In addition, Disulfiram can reverse the resistance to chemotherapeutic drugs by inhibiting the P-glycoprotein multidrug efflux pump and suppressing the activation of NF-kB, both of which play an important role in the development of drug resistance. Furthermore, Disulfiram has been found to reduce angiogenesis because of its metal chelating properties as well as its ability to inactivate Cu/Zn superoxide dismutase and matrix metalloproteinases. Disulfiram has also been shown to inhibit the proteasomes, DNA topoisomerases, DNA methyltransferase, glutathione S-transferase P1, and O6- methylguanine DNA methyltransferase, a DNA repair protein highly expressed in brain tumors. The patents described in this review demonstrate that Disulfiram is useful as an anti-cancer drug.

Conclusion: For years the FDA-approved, well-tolerated, inexpensive, orally-administered drug Disulfiram was used in the treatment of chronic alcoholism, but it has recently demonstrated anti-cancer effects in a range of solid and hematological malignancies. Its combination with copper at clinically relevant concentrations might overcome the resistance of many anti-cancer drugs in vitro, in vivo, and in patients.

Keywords: Angiogenesis, cancer, cancer stem cells, cytotoxicity, disulfiram, inflammation, proteasome.

[1]
Ashburn TT, Thor KB. Drug repositioning: Identifying and developing new uses for existing drugs. Nat Rev Drug Discov 2004; 3(8): 673-83.
[http://dx.doi.org/10.1038/nrd1468] [PMID: 15286734]
[2]
Walker I, Newell H. Do molecularly targeted agents in oncolo-gy have reduced attrition rates? Nat Rev Drug Discov 2009; 8(1): 15-6.
[http://dx.doi.org/10.1038/nrd2758] [PMID: 19008887]
[3]
Schmitt SM, Frezza M, Dou QP. New applications of old met-al-binding drugs in the treatment of human cancer. Front Biosci (Schol Ed) 2012; 4: 375-91.
[http://dx.doi.org/10.2741/s274] [PMID: 22202066]
[4]
Spillier Q, Vertommen D, Ravez S, Marteau R, Themans Q, Corbet C, et al. Anti-alcohol abuse drug disulfiram inhibits human PHGDH via disruption of its active tetrameric form through a specific cysteine oxidation. Sci Rep 2019; 9(1): 4737.
[http://dx.doi.org/10.1038/s41598-019-41187-0] [PMID: 30894617]
[5]
Cvek B. Antabuse repurposing: We need more knowledge and wide international support. Int J Cancer 2011; 129: 1286-7.
[http://dx.doi.org/10.1002/ijc.25779]
[6]
Krampe H, Stawicki S, Wagner T, Bertels C, Aust C, Ruther E, et al. Follow-up of 180 alcoholic patients for up to 7 years after outpatient treatment: Impact of alcohol deterrents on outcome. Alcohol Clin Exp Res 2006; 30(1): 86-95.
[http://dx.doi.org/10.1111/j.1530-0277.2006.00013.x] [PMID: 16433735]
[7]
Wang Y, Li W, Patel SS, Cong J, Zhang N, Sabbatino F, et al. Blocking the formation of radiation-induced breast cancer stem cells. Oncotarget 2014; 5(11): 3743-55.
[http://dx.doi.org/10.18632/oncotarget.1992] [PMID: 25003837]
[8]
Cen D, Brayton D, Shahandeh B, Meyskens FL Jr, Farmer PJ. Disulfiram facilitates intracellular Cu uptake and induces apoptosis in human melanoma cells. J Med Chem 2004; 47(27): 6914-20.
[http://dx.doi.org/10.1021/jm049568z] [PMID: 15615540]
[9]
Available at. https://clinicaltrials.gov/ [Accessed on January 4, 2019]
[10]
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.
[http://dx.doi.org/10.1016/j.ctrv.2008.07.004] [PMID: 18774652]
[11]
Fabris C, Farini R, Del Favero G, Gurriera G, Piccolo A, Sturniolo GC, et al. Copper, zinc and cop-per/zinc ratio in chronic pancreatitis and pancreatic cancer. Clin Biochem 1985; 18(6): 373-5.
[http://dx.doi.org/10.1016/S0009-9120(85)80078-3] [PMID: 4092355]
[12]
Sauna ZE, Shukla S, Ambudkar SV. Disulfiram, an old drug with new potential therapeutic uses for human cancers and fungal infections. Mol Biosyst 2005; 1(2): 127-34.
[http://dx.doi.org/10.1039/b504392a] [PMID: 16880974]
[13]
Cen D, Gonzalez RI, Buckmeier JA, Kahlon RS, Tohidian NB, Meyskens FL Jr. Disulfiram induces apoptosis in human mel-anoma cells: A redox-related process. Mol Cancer Ther 2002; 1(3): 197-204.
[PMID: 12467214]
[14]
Iljin K, Ketola K, Vainio P, Halonen P, Kohonen P, Fey V, et al. High-throughput cell-based screening of 4910 known drugs and drug-like small molecules identifies disulfiram as an inhibitor of prostate cancer cell growth. Clin Cancer Res 2009; 15(19): 6070-8.
[http://dx.doi.org/10.1158/1078-0432.CCR-09-1035] [PMID: 19789329]
[15]
Viola-Rhenals M, Patel KR, Jaimes-Santamaria L, Wu G, Liu J, Dou QP. Recent advances in antabuse (Disulfiram): The importance of its metal-binding ability to its anticancer activi-ty. Curr Med Chem 2018; 25(4): 506-24.
[http://dx.doi.org/10.2174/0929867324666171023161121] [PMID: 29065820]
[16]
Zha J, Chen F, Dong H, Shi P, Yao Y, Zhang Y, et al. Disulfi-ram targeting lymphoid malignant cell lines via ROS-JNK acti-vation as well as Nrf2 and NF-kB pathway inhibition. J Transl Med 2014; 12: 163.
[http://dx.doi.org/10.1186/1479-5876-12-163] [PMID: 24915933]
[17]
Hassani S, Ghaffari P, Chahardouli B, Alimoghaddam K, Ghavamzadeh A, Alizadeh S, et al. Disulfiram/copper causes ROS levels alteration, cell cycle inhibition, and apoptosis in acute myeloid leukaemia cell lines with modulation in the ex-pression of related genes. Biomed Pharmacother 2018; 99: 561-9.
[http://dx.doi.org/10.1016/j.biopha.2018.01.109] [PMID: 29902866]
[18]
Yip NC, Fombon IS, Liu P, Brown S, Kannappan V, Armesil-la AL, 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): 1564-74.
[http://dx.doi.org/10.1038/bjc.2011.126] [PMID: 21487404]
[19]
Cvek B. Targeting malignancies with disulfiram (Antabuse): Multidrug resistance, angiogenesis, and proteasome. Curr Cancer Drug Targets 2011; 11(3): 332-7.
[http://dx.doi.org/10.2174/156800911794519806] [PMID: 21247389]
[20]
Liu P, Wang Z, Brown S, Vinodh K, Patricia ET, Wenguo J, et al. Liposome encapsulated Disulfiram inhibits NFκB pathway and targets breast cancer stem cells in vitro and in vivo. Oncotarget 2014; 5(17): 7471-85.
[http://dx.doi.org/10.18632/oncotarget.2166] [PMID: 25277186]
[21]
Marikovsky M, Nevo N, Vadai E, Harris-Cerruti C. Cu/Zn superoxide dismutase plays a role in angiogenesis. Int J Cancer 2002; 97(1): 34-41.
[http://dx.doi.org/10.1002/ijc.1565] [PMID: 11774241]
[22]
Li Y, Fu SY, Wang LH, Wang FY, Wang NN, Cao Q, et al. Copper improves the anti-angiogenic activity of disulfiram through the EGFR/Src/VEGF pathway in gliomas. Cancer Lett 2015; 369(1): 86-96.
[http://dx.doi.org/10.1016/j.canlet.2015.07.029] [PMID: 26254539]
[23]
Goto K, Arai J, Stephanou A, Kato N. Novel therapeutic fea-tures of disulfiram against hepatocellular carcinoma cells with inhibitory effects on a disintegrin and metalloproteinase 10. Oncotarget 2018; 9(27): 18821-31.
[http://dx.doi.org/10.18632/oncotarget.24568] [PMID: 29721164]
[24]
Yakisich JS, Sidén A, Eneroth P, Cruz M. Disulfiram is a potent in vitro inhibitor of DNA topoisomerases. Biochem Biophys Res Commun 2001; 289(2): 586-90.
[http://dx.doi.org/10.1006/bbrc.2001.6027] [PMID: 11716515]
[25]
Lin J, Haffner MC, Zhang Y, Lee BH, Brennen WN, Britton J, et al. Disulfiram is a DNA demethylating agent and inhibits prostate cancer cell growth. Prostate 2011; 71(4): 333-43.
[http://dx.doi.org/10.1002/pros.21247] [PMID: 20809552]
[26]
Madala HR, Punganuru SR, Ali-Osman F, Zhang R, Srivenu-gopal KS. Brain- and brain tumor-penetrating disulfiram na-noparticles: Sequence of cytotoxic events and efficacy in hu-man glioma cell lines and intracranial xenografts. Oncotarget 2017; 9(3): 3459-82.
[PMID: 29423059]
[27]
Zha J, Chen F, Dong H, Shi P, Yao Y, Zhang Y, et al. Disulfi-ram targeting lymphoid malignant cell lines via ROS-JNK acti-vation as well as Nrf2 and NF-kB pathway inhibition. J Transl Med 2014; 12: 163.
[http://dx.doi.org/10.1186/1479-5876-12-163] [PMID: 24915933]
[28]
Paranjpe A, Zhang R, Ali-Osman F, Bobustuc GC, Srivenu-gopal KS. Disulfiram is a direct and potent inhibitor of human O6-methylguanine-DNA methyltransferase (MGMT) in brain tumor cells and mouse brain and markedly increases the al-kylating DNA damage. Carcinogenesis 2014; 35(3): 692-702.
[http://dx.doi.org/10.1093/carcin/bgt366] [PMID: 24193513]
[29]
Takahashi C, Sheng Z, Horan TP, Kitayama H, Maki M, Hitomi K, et al. Regulation of matrix metalloproteinase-9 and inhibition of tumor invasion by the membrane-anchored gly-coprotein RECK. Proc Natl Acad Sci USA 1998; 95(22): 13221-6.
[http://dx.doi.org/10.1073/pnas.95.22.13221] [PMID: 9789069]
[30]
Noda M, Oh J, Takahashi R, Kondo S, Kitayama H, Takahashi C. RECK: A novel suppressor of malignancy link-ing oncogenic signaling to extracellular matrix remodeling. Cancer Metastasis Rev 2003; 22(2-3): 167-75.
[http://dx.doi.org/10.1023/A:1023043315031] [PMID: 12784995]
[31]
Noda M, Takahashi C. Recklessness as a hallmark of aggres-sive cancer. Cancer Sci 2007; 98(11): 1659-65.
[http://dx.doi.org/10.1111/j.1349-7006.2007.00588.x] [PMID: 17725805]
[32]
Sasahara RM, Takahashi C, Noda M. Involvement of the Sp1 site in ras-mediated downregulation of the RECK metastasis suppressor gene. Biochem Biophys Res Commun 1999; 264(3): 668-75.
[http://dx.doi.org/10.1006/bbrc.1999.1552] [PMID: 10543990]
[33]
Oh J, Takahashi R, Kondo S, Mizoguchi A, Adachi E, Sasaha-ra RM, et al. The membrane-anchored MMP inhibitor RECK is a key regulator of extracellular matrix integrity and angio-genesis. Cell 2001; 107(6): 789-800.
[http://dx.doi.org/10.1016/S0092-8674(01)00597-9] [PMID: 11747814]
[34]
Murai R, Yoshida Y, Muraguchi T, Nishimoto E, Morioka Y, Kitayama H, et al. A novel screen using the Reck tumor sup-pressor gene promoter detects both conventional and metasta-sis-suppressing anticancer drugs. Oncotarget 2010; 1(4): 252-64.
[PMID: 21304177]
[35]
Tesson M, Anselmi G, Bell C, Mairs R. Cell cycle specific radiosensitisation by the disulfiram and copper complex. Oncotarget 2017; 8(39): 65900-16.
[http://dx.doi.org/10.18632/oncotarget.19539] [PMID: 29029481]
[36]
Koh HK, Seo SY, Kim JH, Kim HJ, Chie EK, Kim SK, et al. Disulfiram, a re-positioned aldehyde dehydrogenase inhibitor, enhances radiosensitivity of human glioblastoma cells in vitro. Cancer Res Treat 2019; 51(2): 696-705.
[http://dx.doi.org/10.4143/crt.2018.249] [PMID: 30121967]
[37]
Triscott J, Lee C, Hu K, Fotovati A, Berns R, Pambid M, et al. Disulfiram, a drug widely used to control alcoholism, suppresses the self-renewal of glioblastoma and overrides resistance to temozolomide. Oncotarget 2012; 3(10): 1112-23.
[http://dx.doi.org/10.18632/oncotarget.604] [PMID: 23047041]
[38]
Triscott J, Rose Pambid M, Dunn SE. Concise review: Bull-seye: Targeting cancer stem cells to improve the treatment of gliomas by repurposing disulfiram. Stem Cells 2015; 33(4): 1042-6.
[http://dx.doi.org/10.1002/stem.1956] [PMID: 25588723]
[39]
Jiao Y, Hannafon BN, Zhang RR, Fung KM, Ding WQ. Do-cosahexaenoic acid and disulfiram act in concert to kill cancer cells: A mutual enhancement of their anticancer actions. Oncotarget 2017; 8(11): 17908-20.
[http://dx.doi.org/10.18632/oncotarget.14702] [PMID: 28107189]
[40]
Cong J, Wang Y, Zhang X, Zhang N, Liu L, Soukup K, et al. A novel chemoradiation targeting stem and nonstem pancreatic cancer cells by repurposing disulfiram. Cancer Lett 2017; 409: 9-19.
[http://dx.doi.org/10.1016/j.canlet.2017.08.028] [PMID: 28864067]
[41]
Nechushtan H, Hamamreh Y, Nidal S, Gotfried M, Baron A, Shalev Y, 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): 366-7.
[http://dx.doi.org/10.1634/theoncologist.2014-0424] [PMID: 25777347]
[42]
Wang Z, Tan J, McConville C, Kannappan V, Tawari PE, Brown J, et al. Poly lactic-co-glycolic acid controlled delivery of disulfiram to target liver cancer stem-like cells. Nanomedicine (Lond) 2017; 13(2): 641-57.
[http://dx.doi.org/10.1016/j.nano.2016.08.001] [PMID: 27521693]
[43]
Chiba T, Suzuki E, Yuki K. Disulfiram eradicates tumor-initiating hepatocellular carcinoma cells in ROS-p38 MAPK pathway-dependent and -independent manners. PLoS One 2014; 9(1)e84807
[http://dx.doi.org/10.1371/journal.pone.0084807] [PMID: 24454751]
[44]
Bista R, Lee DW, Pepper OB, Azorsa DO, Arceci RJ, Aleem E. Disulfiram overcomes bortezomib and cytarabine resistance in Down-syndrome-associated acute myeloid leukemia cells. J Exp Clin Cancer Res 2017; 36(1): 22.
[http://dx.doi.org/10.1186/s13046-017-0493-5] [PMID: 28143565]
[45]
O’Brien A, Barber JE, Reid S, Niknejad N, Dimitroulakos J. Enhancement of cisplatin cytotoxicity by disulfiram involves activating transcription factor 3. Anticancer Res 2012; 32(7): 2679-88.
[PMID: 22753726]
[46]
Huo Q, Zhu J, Niu Y, Shi H, Gong Y, Li Y, et al. pH-Triggered surface charge-switchable polymer micelles for the co-delivery of paclitaxel/disulfiram and overcoming multidrug resistance in cancer. Int J Nanomedicine 2017; 12: 8631-47.
[http://dx.doi.org/10.2147/IJN.S144452] [PMID: 29270012]
[47]
Wiggins HL, Wymant JM, Solfa F, Hiscox SE, Taylor KM, Westwell AD, et al. Disulfiram-induced cytotoxicity and endo-lysosomal sequestration of zinc in breast cancer cells. Biochem Pharmacol 2015; 93(3): 332-42.
[http://dx.doi.org/10.1016/j.bcp.2014.12.014] [PMID: 25557293]
[48]
Huang H, Liao Y, Liu N, Xianliang H, Jianyu C, Changshan Y, et al. Two clinical drugs deubiquitinase inhibitor auranofin and aldehyde dehydrogenase inhibitor disulfiram trigger synergistic anti-tumor effects in vitro and in vivo. Oncotarget 2016; 7(3): 2796-808.
[http://dx.doi.org/10.18632/oncotarget.6425] [PMID: 26625200]
[49]
Yang Z, Guo F, Albers AE, Sehouli J, Kaufmann AM. Disulfiram modulates ROS accumulation and overcomes synergistically cisplatin resistance in breast cancer cell lines. Biomed Pharmacother 2019.113108727
[http://dx.doi.org/10.1016/j.biopha.2019.108727] [PMID: 30870721]
[50]
Calderon-Aparicio A, Cornejo A, Orue A, Rieber M. Anti-cancer response to disulfiram may be enhanced by co-treatment with MEK inhibitor or oxaliplatin: Modulation by tetrathiomolybdate, KRAS/BRAF mutations and c-MYC/p53 status. Ecancermedicalscience 2019; 13: 890.
[http://dx.doi.org/10.3332/ecancer.2019.890] [PMID: 30792807]
[51]
Wu L, Meng F, Dong L, Block CJ, Mitchell AV, Wu J, et al. Disulfiram and BKM120 in Combination with Chemotherapy Impede Tumor Progression and Delay Tumor Recurrence in Tumor Initiating Cell-Rich TNBC. Sci Rep 2019; 9(1): 236.
[http://dx.doi.org/10.1038/s41598-018-35619-6] [PMID: 30659204]
[52]
Majera D, Skrott Z, Bouchal J, Bartkova J, Simkova D, Ga-chechiladze M, et al. Targeting genotoxic and proteotoxic stress-response pathways in human prostate cancer by clinically available PARP inhibitors, vorinostat and disulfiram. Prostate 2019; 79(4): 352-62.
[http://dx.doi.org/10.1002/pros.23741] [PMID: 30499118]
[53]
Huang J, Campian JL, Gujar AD, Tsien C, Ansstas G, Tran DD, et al. Final results of a Phase I dose-escalation, dose-expansion study of adding disulfiram with or without copper to adjuvant temozolomide for newly diagnosed glioblastoma. J Neurooncol 2018; 138(1): 105-11.
[http://dx.doi.org/10.1007/s11060-018-2775-y] [PMID: 29374809]
[54]
Banerjee P, Geng T, Mahanty A, Li T, Zong L, Wang B. Inte-grating the drug, disulfiram into the vitamin E-TPGS-modified PEGylated nanostructured lipid carriers to synergize its re-purposing for anti-cancer therapy of solid tumors. Int J Pharm 2019; 557: 374-89.
[http://dx.doi.org/10.1016/j.ijpharm.2018.12.051] [PMID: 30610896]
[55]
Chen W, Yang W, Chen P, Huang Y, Li F. Disulfiram copper nanoparticles prepared with a stabilized metal ion ligand com-plex method for treating drug-resistant prostate cancers. ACS Appl Mater Interfaces 2018; 10(48): 41118-28.
[http://dx.doi.org/10.1021/acsami.8b14940] [PMID: 30444340]
[56]
Duan X, Xiao J, Yin Q, Zhiwen Z, Haijun Y, Shirui M, et al. Multi-targeted inhibition of tumor growth and lung metastasis by redox-sensitive shell crosslinked micelles loading disulfi-ram. Nanotechnology 2014; 25(12)125102
[http://dx.doi.org/10.1088/0957-4484/25/12/125102] [PMID: 24576956]
[57]
Agarwal RP, McPherson RA, Phillips M. Rapid degradation of disulfiram by serum albumin. Res Commun Chem Pathol Pharmacol 1983; 42(2): 293-310.
[PMID: 6318285]
[58]
Agarwal RP, Phillips M, McPherson RA, Hensley P. Serum albumin and the metabolism of disulfiram. Biochem Pharmacol 1986; 35(19): 3341-7.
[http://dx.doi.org/10.1016/0006-2952(86)90433-8] [PMID: 3021167]
[59]
Gessner T, Jakubowski M. Diethyldithiocarbamic acid methyl ester: A metabolite of disulfiram. Biochem Pharmacol 1972; 21(2): 219-30.
[http://dx.doi.org/10.1016/0006-2952(72)90272-9] [PMID: 4645555]
[60]
Kaslander J. Formation of an S-glucuronide from tetraethyl-thiuram disulfide (Antabuse) in man. Biochim Biophys Acta 1963; 71: 730-1.
[http://dx.doi.org/10.1016/0006-3002(63)91149-1] [PMID: 13962691]
[61]
Prickett CS, Johnston CD. The in vivo production of carbon disulfide from tetraethylthiuramdisulfide (antabuse). Biochim Biophys Acta 1953; 12(4): 542-6.
[http://dx.doi.org/10.1016/0006-3002(53)90186-3] [PMID: 13140263]
[62]
Wang W. The in vivo production of carbon disulfide from tetraethylthiuramdisulfide (antabuse). Biochim Biophys Acta 1953; 12: 542-6.
[63]
Miao L, Su J, Zhuo X, Luo L, Kong Y, Gou J, et al. mPEG5k- b-PLGA2k/PCL3.4k/MCT mixed micelles as carriers of disulfiram for improving plasma stability and antitumor effect in vivo. Mol Pharm 2018; 15(4): 1556-64.
[http://dx.doi.org/10.1021/acs.molpharmaceut.7b01094] [PMID: 29505711]
[64]
Liu P, Kumar IS, Brown S, Kannappan V, Tawari PE, Tang JZ, et al. Disulfiram targets cancer stem-like cells and reverses resistance and cross-resistance in acquired paclitaxel-resistant triple-negative breast cancer cells. Br J Cancer 2013; 109(7): 1876-85.
[http://dx.doi.org/10.1038/bjc.2013.534] [PMID: 24008666]
[65]
Skrott Z, Mistrik M, Andersen KK, Friis S, Majera D, Gursky J, et al. Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature 2017; 552(7684): 194-9.
[http://dx.doi.org/10.1038/nature25016] [PMID: 29211715]
[66]
Wu X, Xue X, Wang L, Wang W, Han J, Sun X, et al. Sup-pressing autophagy enhances disulfiram/copper-induced apoptosis in non-small cell lung cancer. Eur J Pharmacol 2018; 827: 1-12.
[http://dx.doi.org/10.1016/j.ejphar.2018.02.039] [PMID: 29547841]
[67]
Choi SA, Choi JW, Wang KC, Phi JH, Lee JY, Park KD, et al. Disulfiram modulates stemness and metabolism of brain tu-mor initiating cells in atypical teratoid/rhabdoid tumors. Neuro-oncol 2015; 17(6): 810-21.
[http://dx.doi.org/10.1093/neuonc/nou305] [PMID: 25378634]
[68]
Wickström M, Danielsson K, Rickardson L, Gullbo J, Nygren P, Isaksson A, et al. Pharmacological profiling of disulfiram using human tumor cell lines and human tumor cells from patients. Biochem Pharmacol 2007; 73(1): 25-33.
[http://dx.doi.org/10.1016/j.bcp.2006.08.016] [PMID: 17026967]
[69]
Brar SS, Grigg C, Wilson KS, Holder WD Jr, Dreau D, Austin C, et al. Disulfiram inhibits activating transcription factor/cyclic AMP-responsive element binding protein and human melanoma growth in a metal-dependent manner in vitro, in mice and in a patient with metastatic disease. Mol Cancer Ther 2004; 3(9): 1049-60.
[PMID: 15367699]
[70]
Chen D, Cui QC, Yang H, Dou QP. 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): 10425-33.
[http://dx.doi.org/10.1158/0008-5472.CAN-06-2126] [PMID: 17079463]
[71]
You SY, Rui W, Chen ST, Chen HC, Liu XW, Huang J, et al. Process of immunogenic cell death caused by disulfiram as the anti-colorectal cancer candidate. Biochem Biophys Res Commun 2019; pii: S0006-291X (19): 30599-6.
[http://dx.doi.org/10.1016/j.bbrc.2019.03.192]
[72]
Mohammad IS, He W, Yin L. A smart paclitaxel-disulfiram nanococrystals for efficient mdr reversal and enhanced apoptosis. Pharm Res 2018; 35(4): 77.
[http://dx.doi.org/10.1007/s11095-018-2370-0] [PMID: 29488114]
[73]
Burkitt MJ, Bishop HS, Milne L, Tsang SY, Provan GJ, Nobel CS, et al. Dithiocarbamate toxicity toward thymocytes involves their copper-catalyzed conversion to thiuram disulfides, which oxidize glutathione in a redox cycle without the release of reactive oxygen species. Arch Biochem Biophys 1998; 353(1): 73-84.
[http://dx.doi.org/10.1006/abbi.1998.0618] [PMID: 9578602]
[74]
Chen D, Peng F, Cui QC, Daniel KG, Orlu S, Liu J, et al. Inhibition of prostate cancer cellular proteasome activity by a pyrrolidine dithiocarbamate-copper complex is associated with suppression of proliferation and induction of apoptosis. Front Biosci 2005; 10: 2932-9.
[http://dx.doi.org/10.2741/1749] [PMID: 15970547]
[75]
Chen J, Du C, Kang J, Wang J. Cu2+ is required for pyrrolidine dithiocarbamate to inhibit histone acetylation and induce human leukemia cell apoptosis. Chem Biol Interact 2008; 171(1): 26-36.
[http://dx.doi.org/10.1016/j.cbi.2007.09.004] [PMID: 17961528]
[76]
Erl W, Weber C, Hansson GK. Pyrrolidine dithiocarbamate-induced apoptosis depends on cell type, density, and the presence of Cu2+ and Zn2+. Am J Physiol Cell Physiol 2000; 278(6): C1116-25.
[http://dx.doi.org/10.1152/ajpcell.2000.278.6.C1116] [PMID: 10837339]
[77]
Watanabe T, Yoshizumi M, Akishita M, Eto M, Toba K, Hashimoto M, et al. Induction of nuclear orphan receptor NGFI-B gene and apoptosis in rat vascular smooth muscle cells treated with pyrrolidinedithiocarbamate. Arterioscler Thromb Vasc Biol 2001; 21(11): 1738-44.
[http://dx.doi.org/10.1161/hq1101.098550] [PMID: 11701459]
[78]
Masquelier M, Vitols S. Drastic effect of cell density on the cytotoxicity of daunorubicin and cytosine arabinoside. Biochem Pharmacol 2004; 67(9): 1639-46.
[http://dx.doi.org/10.1016/j.bcp.2003.12.034] [PMID: 15081863]
[79]
Kobayashi H, Takemura Y, Holland JF, Ohnuma T. Vincristine saturation of cellular binding sites and its cytotoxic activity in human lymphoblastic leukemia cells: mechanism of inoculum effect. Biochem Pharmacol 1998; 55(8): 1229-34.
[http://dx.doi.org/10.1016/S0006-2952(97)00615-1] [PMID: 9719477]
[80]
Slapak CA, Lecerf JM, Daniel JC, Levy SB. Energy-dependent accumulation of daunorubicin into subcellular compartments of human leukemia cells and cytoplasts. J Biol Chem 1992; 267(15): 10638-44.
[PMID: 1350280]
[81]
Navrátilová J, Jungová P, Vanhara P, Preisler J, Kanicky V, Smarda J. Copper ions regulate cytotoxicity of disulfiram to myeloid leukemia cells. Int J Mol Med 2009; 24(5): 661-70.
[PMID: 19787200]
[82]
Park YM, Go YY, Shin SH, Cho JG, Woo JS, Song JJ. Anti-cancer effects of disulfiram in head and neck squamous cell carcinoma via autophagic cell death. PLoS One 2018; 13(9)e0203069
[http://dx.doi.org/10.1371/journal.pone.0203069] [PMID: 30212479]
[83]
Hothi P, Martins TJ, Chen L, Deleyrolle L, Yoon JG, Reynolds B, et al. High-throughput chemical screens identify disulfiram as an inhibitor of human glioblastoma stem cells. Oncotarget 2012; 3(10): 1124-36.
[http://dx.doi.org/10.18632/oncotarget.707] [PMID: 23165409]
[84]
Mimeault M, Batra SK. Recent advances in the development of novel anti‐cancer drugs targeting cancer stem/progenitor cells. Drug Dev Res 2008; 69: 415-30.
[http://dx.doi.org/10.1002/ddr.20273]
[85]
Jin N, Zhu X, Cheng F, Zhang L. Disulfiram/copper targets stem cell-like ALDH+ population of multiple myeloma by inhibition of ALDH1A1 and Hedgehog pathway. J Cell Biochem 2018; 119(8): 6882-93.
[http://dx.doi.org/10.1002/jcb.26885] [PMID: 29665144]
[86]
Corti S, Locatelli F, Papadimitriou D, Donadoni C, Salani S, Del BR, et al. Identification of a primitive brain-derived neural stem cell population based on aldehyde dehydrogenase activity. Stem Cells 2006; 24(4): 975-85.
[http://dx.doi.org/10.1634/stemcells.2005-0217] [PMID: 16293577]
[87]
Deleyrolle LP, Harding A, Cato K, Siebzehnrubl FA, Rahman M, Azari H, et al. Evidence for label-retaining tumour-initiating cells in human glioblastoma. Brain 2011; 134(Pt 5): 1331-43.
[http://dx.doi.org/10.1093/brain/awr081] [PMID: 21515906]
[88]
Greco N, Schott T, Mu X, Rothenberg A, Voigt C, McGough RL, et al. ALDH activity correlates with metastatic potential in primary sarcomas of bone. J Cancer Ther 2014; 5(4): 331-8.
[http://dx.doi.org/10.4236/jct.2014.54040] [PMID: 25328803]
[89]
Marikovsky M, Ziv V, Nevo N, Harris-Cerruti C, Mahler O. Cu/Zn superoxide dismutase plays important role in immune response. J Immunol 2003; 170(6): 2993-3001.
[http://dx.doi.org/10.4049/jimmunol.170.6.2993] [PMID: 12626552]
[90]
Forman HJ, York JL, Fisher AB. Mechanism for the potentiation of oxygen toxicity by disulfiram. J Pharmacol Exp Ther 1980; 212(3): 452-5.
[PMID: 6244385]
[91]
Allensworth JL, Evans MK, Bertucci F, Aldrich AJ, Festa RA, Finetti P, et al. Disulfiram (DSF) acts as a copper ionophore to induce copper-dependent oxidative stress and mediate anti-tumor efficacy in inflammatory breast cancer. Mol Oncol 2015; 9(6): 1155-68.
[http://dx.doi.org/10.1016/j.molonc.2015.02.007] [PMID: 25769405]
[92]
Failla ML, Hopkins RG. Is low copper status immunosup-pressive? Nutr Rev 1998; 56(1 Pt 2): S59-64.
[PMID: 9481125]
[93]
Gengelbach GP, Ward JD, Spears JW, Brown TT Jr. Effects of copper deficiency and copper deficiency coupled with high dietary iron or molybdenum on phagocytic cell function and response of calves to a respiratory disease challenge. J Anim Sci 1997; 75(4): 1112-8.
[http://dx.doi.org/10.2527/1997.7541112x] [PMID: 9110227]
[94]
Beckman JS, Beckman TW, Chen J, Marshall PA, Freeman BA. Apparent hydroxyl radical production by peroxynitrite: Implications for endothelial injury from nitric oxide and superoxide. Proc Natl Acad Sci USA 1990; 87(4): 1620-4.
[http://dx.doi.org/10.1073/pnas.87.4.1620] [PMID: 2154753]
[95]
Brockhaus F, Brüne B. Overexpression of CuZn superoxide dismutase protects RAW 264.7 macrophages against nitric oxide cytotoxicity. Biochem J 1999; 338(Pt 2): 295-303.
[http://dx.doi.org/10.1042/bj3380295] [PMID: 10024504]
[96]
Teixeira HD, Schumacher RI, Meneghini R. Lower intracellular hydrogen peroxide levels in cells overexpressing CuZn-superoxide dismutase. Proc Natl Acad Sci USA 1998; 95(14): 7872-5.
[http://dx.doi.org/10.1073/pnas.95.14.7872] [PMID: 9653107]
[97]
Elroy-Stein O, Bernstein Y, Groner Y. Overproduction of human Cu/Zn-superoxide dismutase in transfected cells: Ex-tenuation of paraquat-mediated cytotoxicity and enhancement of lipid peroxidation. EMBO J 1986; 5(3): 615-22.
[http://dx.doi.org/10.1002/j.1460-2075.1986.tb04255.x] [PMID: 3011416]
[98]
Marikovsky M. Thiram inhibits angiogenesis and slows the development of experimental tumours in mice. Br J Cancer 2002; 86(5): 779-87.
[http://dx.doi.org/10.1038/sj.bjc.6600078] [PMID: 11875743]
[99]
Rengarajan J, Mowen KA, McBride KD, Smith ED, Singh H, Glimcher LH. Interferon regulatory factor 4 (IRF4) interacts with NFATc2 to modulate interleukin 4 gene expression. J Exp Med 2002; 195(8): 1003-12.
[http://dx.doi.org/10.1084/jem.20011128] [PMID: 11956291]
[100]
Tominaga N, Ohkusu-Tsukada K, Udono H, Abe R, Matsuyama T, Yui K. Development of Th1 and not Th2 immune responses in mice lacking IFN-regulatory factor-4. Int Immunol 2003; 15(1): 1-10.
[http://dx.doi.org/10.1093/intimm/dxg001] [PMID: 12502720]
[101]
Zhao M, Sun D, Guan Y, Wang Z, Sang D, Liu M, et al. Disulfiram and Diphenhydramine Hydrochloride Upregulate miR-30a to Suppress IL-17-Associated Autoimmune Inflammation. J Neurosci 2016; 36(35): 9253-66.
[http://dx.doi.org/10.1523/JNEUROSCI.4587-15.2016] [PMID: 27581464]
[102]
Folkman J, Klagsbrun M. Angiogenic factors. Science 1987; 235(4787): 442-7.
[http://dx.doi.org/10.1126/science.2432664] [PMID: 2432664]
[103]
Folkman J. What is the evidence that tumors are angiogenesis dependent? J Natl Cancer Inst 1990; 82(1): 4-6.
[http://dx.doi.org/10.1093/jnci/82.1.4] [PMID: 1688381]
[104]
Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med 1995; 1(1): 27-31.
[http://dx.doi.org/10.1038/nm0195-27] [PMID: 7584949]
[105]
Folkman J, D’Amore PA. Blood vessel formation: What is its molecular basis? Cell 1996; 87(7): 1153-5.
[http://dx.doi.org/10.1016/S0092-8674(00)81810-3] [PMID: 8980221]
[106]
Ellis LM, Fidler IJ. Angiogenesis and metastasis. Eur J Cancer 1996; 32A(14): 2451-60.
[http://dx.doi.org/10.1016/S0959-8049(96)00389-9] [PMID: 9059333]
[107]
Fidler IJ, Kumar R, Bielenberg DR, Ellis LM. Molecular determinants of angiogenesis in cancer metastasis. Cancer J Sci Am 1998; 4(Suppl. 1): S58-66.
[PMID: 9619272]
[108]
Shweiki D, Itin A, Soffer D, Keshet E. Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis. Nature 1992; 359(6398): 843-5.
[http://dx.doi.org/10.1038/359843a0] [PMID: 1279431]
[109]
Terada LS, Willingham IR, Rosandich ME, Leff JA, Kindt GW, Repine JE. Generation of superoxide anion by brain endothelial cell xanthine oxidase. J Cell Physiol 1991; 148(2): 191-6.
[http://dx.doi.org/10.1002/jcp.1041480202] [PMID: 1652587]
[110]
Chen D, Dou QP. New uses for old copper-binding drugs: converting the pro-angiogenic copper to a specific cancer cell death inducer. Expert Opin Ther Targets 2008; 12(6): 739-48.
[http://dx.doi.org/10.1517/14728222.12.6.739] [PMID: 18479220]
[111]
Ratych RE, Chuknyiska RS, Bulkley GB. The primary localization of free radical generation after anoxia/reoxygenation in isolated endothelial cells. Surgery 1987; 102(2): 122-31.
[PMID: 3039675]
[112]
Zweier JL, Kuppusamy P, Lutty GA. Measurement of endothelial cell free radical generation: evidence for a central mechanism of free radical injury in postischemic tissues. Proc Natl Acad Sci USA 1988; 85(11): 4046-50.
[http://dx.doi.org/10.1073/pnas.85.11.4046] [PMID: 2836868]
[113]
Inauen W, Payne DK, Kvietys PR, Granger DN. Hypox-ia/reoxygenation increases the permeability of endothelial cell monolayers: role of oxygen radicals. Free Radic Biol Med 1990; 9(3): 219-23.
[http://dx.doi.org/10.1016/0891-5849(90)90031-D] [PMID: 2272529]
[114]
Lum H, Barr DA, Shaffer JR, Gordon RJ, Ezrin AM, Malik AB. Reoxygenation of endothelial cells increases permeability by oxidant-dependent mechanisms. Circ Res 1992; 70(5): 991-8.
[http://dx.doi.org/10.1161/01.RES.70.5.991] [PMID: 1568306]
[115]
Zweier JL, Broderick R, Kuppusamy P, Thompson-Gorman S, Lutty GA. Determination of the mechanism of free radical generation in human aortic endothelial cells exposed to anoxia and reoxygenation. J Biol Chem 1994; 269(39): 24156-62.
[PMID: 7929072]
[116]
Zweier JL, Kuppusamy P, Thompson-Gorman S, Klunk D, Lutty GA. Measurement and characterization of free radical generation in reoxygenated human endothelial cells. Am J Physiol 1994; 266(3 Pt 1): C700-8.
[http://dx.doi.org/10.1152/ajpcell.1994.266.3.C700] [PMID: 8166233]
[117]
Terada LS. Hypoxia-reoxygenation increases O2-. efflux which injures endothelial cells by an extracellular mechanism. Am J Physiol 1996; 270(3 Pt 2): H945-50.
[PMID: 8780189]
[118]
Matsubara T, Ziff M. Increased superoxide anion release from human endothelial cells in response to cytokines. J Immunol 1986; 137(10): 3295-8.
[PMID: 3021851]
[119]
Malmström BG. Enzymology of oxygen. Annu Rev Biochem 1982; 51: 21-59.
[http://dx.doi.org/10.1146/annurev.bi.51.070182.000321] [PMID: 6287915]
[120]
Huang P, Feng L, Oldham EA, Keating MJ, Plunkett W. Superoxide dismutase as a target for the selective killing of cancer cells. Nature 2000; 407(6802): 390-5.
[http://dx.doi.org/10.1038/35030140] [PMID: 11014196]
[121]
Fotsis T, Zhang Y, Pepper MS, Adler CH, Montesano R, Nawroth PP, et al. The endogenous oestrogen metabolite 2-methoxyoestradiol inhibits angiogenesis and suppresses tumour growth. Nature 1994; 368(6468): 237-9.
[http://dx.doi.org/10.1038/368237a0] [PMID: 7511798]
[122]
Wambi-Kiéssé CO, Katusic ZS. Inhibition of copper/zinc superoxide dismutase impairs NO. mediated endothelium-dependent relaxations. Am J Physiol 1999; 276(3): H1043-8.
[PMID: 10070090]
[123]
Dimmeler S, Hermann C, Galle J, Zeiher AM. Upregulation of superoxide dismutase and nitric oxide synthase mediates the apoptosis-suppressive effects of shear stress on endothelial cells. Arterioscler Thromb Vasc Biol 1999; 19(3): 656-64.
[http://dx.doi.org/10.1161/01.ATV.19.3.656] [PMID: 10073970]
[124]
Hochstrasser M. Ubiquitin, proteasomes, and the regulation of intracellular protein degradation. Curr Opin Cell Biol 1995; 7(2): 215-23.
[http://dx.doi.org/10.1016/0955-0674(95)80031-X] [PMID: 7612274]
[125]
Dou QP, Li B. Proteasome inhibitors as potential novel anti-cancer agents. Drug Resist Updat 1999; 2(4): 215-23.
[http://dx.doi.org/10.1054/drup.1999.0095] [PMID: 11504494]
[126]
Ciechanover A. The ubiquitin-proteasome proteolytic pathway. Cell 1994; 79(1): 13-21.
[http://dx.doi.org/10.1016/0092-8674(94)90396-4] [PMID: 7923371]
[127]
Farshi P, Deshmukh RR, Nwankwo JO. Deubiquitinases (DUBs) and DUB inhibitors: a patent review. Expert Opin Ther Pat 2015; 25(10): 1191-208.
[http://dx.doi.org/10.1517/13543776.2015.1056737] [PMID: 26077642]
[128]
Jagannathan S, Abdel-Malek MA, Malek E, Vad N, Latif T, Anderson KC, et al. Pharmacologic screens reveal metformin that suppresses GRP78-dependent autophagy to enhance the anti-myeloma effect of bortezomib. Leukemia 2015; 29(11): 2184-91.
[http://dx.doi.org/10.1038/leu.2015.157] [PMID: 26108695]
[129]
Eriksson E, Wickström M, Perup LS, Johnsen JI, Eksborg S, Kogner P, et al. Protective role of humanin on bortezomib-induced bone growth impairment in anticancer treatment. J Natl Cancer Inst 2014; 106(3)djt459
[http://dx.doi.org/10.1093/jnci/djt459] [PMID: 24586107]
[130]
Daniel KG, Gupta P, Harbach RH, Guida WC, Dou QP. Organic copper complexes as a new class of proteasome inhibitors and apoptosis inducers in human cancer cells. Biochem Pharmacol 2004; 67(6): 1139-51.
[http://dx.doi.org/10.1016/j.bcp.2003.10.031] [PMID: 15006550]
[131]
Daniel KG, Chen D, Orlu S, Cui QC, Miller FR, Dou QP. Clioquinol and pyrrolidine dithiocarbamate complex with copper to form proteasome inhibitors and apoptosis inducers in human breast cancer cells. Breast Cancer Res 2005; 7(6): R897-908.
[http://dx.doi.org/10.1186/bcr1322] [PMID: 16280039]
[132]
An B, Goldfarb RH, Siman R, Dou QP. Novel dipeptidyl proteasome inhibitors overcome Bcl-2 protective function and selectively accumulate the cyclin-dependent kinase inhibitor p27 and induce apoptosis in transformed, but not normal, human fibroblasts. Cell Death Differ 1998; 5(12): 1062-75.
[http://dx.doi.org/10.1038/sj.cdd.4400436] [PMID: 9894613]
[133]
Voorhees PM, Orlowski RZ. The proteasome and proteasome inhibitors in cancer therapy. Annu Rev Pharmacol Toxicol 2006; 46: 189-213.
[http://dx.doi.org/10.1146/annurev.pharmtox.46.120604.141300] [PMID: 16402903]
[134]
White D, Whittle RR, Stowell GW, Whittall LB, Kennedy T. Method of treating cancer using dithiocarbamate derivatives. US20050096304 (2005).
[135]
Srivenugopal KS, Paranjpe AM. Disulfiram compositions and treatments for brain tumors. US20160346231 (2016).
[136]
Wang W. Disulfiram formulation and uses thereof. US2014/0037715 (2014).
[137]
Kennedy TP. Method of inhibiting ATF/CREB and cancer cell growth and pharmaceutical compositions for same. US7816403 (2010).
[138]
Xu P, He H. Polymeric prodrug of disulfiram and application thereof. US20160166706 (2016).
[139]
Chiba T, Suzuki E, Kanai F, Yokosuka S. Liver cancer stem cell inhibitor. JP2013100268 (2013).
[140]
Wang W, Wang Z. Method for treating pleuroperitoneal membrane cancers by locally injecting disulfiram preparation. WO2017177947 (2017).
[141]
Marikovsky M. Pharmaceutical compositions comprising disulfiram. US6288110 (2001).
[142]
Marikovsky M. Pharmaceutical compositions comprising disulfiram for inhibiting angiogenesis. WO1999034784 (1999).
[143]
Israeli-Shalev Y, Bar-On A. Disulfiram doses and treatment regimen suitable for treatment of angiogenesis-dependent. WO2008068746 (2008).
[144]
Devi G. Use of disulfiram for inflammatory breast cancer therapy. US20170020828 (2017).
[145]
Lai CS, Vassilev VP, Wang T. Conjugates of dithiocarba-mate disulfides with pharmacologically active agents and uses therefor. US6274627 (2001).
[146]
George KU, Doherty N. Method of inhibiting interleukin-1 release. EP0284879 (1990).
[147]
Terashima Y, Matsushima K, Toda E, Terasawa H, Yoshinaga S. Agent for controlling cells constituting cancer microenvironment or inflammatory microenvironment. US20180000755 (2018).
[148]
Gooberman LL. Implants containing disulfiram and an antiinflammatory agent. US20100196436 (2010).
[149]
Hacimuftuoglu A. The use of disulfiram in the treatment of gastrointestinal system ulcers. WO2008153511 (2008).
[150]
Wang W, Wang Z, Bian XW. Disulfiram formulation. WO2017077336 (2017).
[151]
Raha D, Settleman J, Wilson TR. Methods of treating cancer and preventing drug resistance. WO2014128235 (2014).
[152]
Burger AM, Westwell A. Anti-cancer therapeutic agents. WO2011097218 (2011).

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy