Skip to main content

Anticancer effects of disulfiram: a systematic review of in vitro, animal, and human studies

Abstract

Background and objectives

Cancer morbidity and mortality rates remain high, and thus, at present, considerable efforts are focused on finding drugs with higher sensitivity against tumor cells and fewer side effects. Disulfiram (DSF), as an anti-alcoholic drug, kills the cancer cells by inducing apoptosis. Several preclinical and clinical studies have examined the potential of repurposing DSF as an anticancer treatment. This systematic review aimed to assess evidence regarding the antineoplastic activity of DSF in in vitro and in vivo models, as well as in humans.

Methods

Two authors independently conducted this systematic review of English and Chinese articles from the PubMed, Embase, and the Cochrane Library databases up to July 2019. Eligible in vitro studies needed to include assessments of the apoptosis rate by flow cytometry using annexin V/propidium iodide, and studies in animal models and clinical trials needed to examine tumor inhibition rates, and progression-free survival (PFS) and overall survival (OS), respectively. Data were analyzed using descriptive statistics.

Results

Overall, 35 studies, i.e., 21 performed in vitro, 11 based on animal models, and three clinical trials, were finally included. In vitro and animal studies indicated that DSF was associated with enhanced apoptosis and tumor inhibition rates, separately. Human studies showed that DSF prolongs PFS and OS. The greatest anti-tumor activity was observed when DSF was used as combination therapy or as a nanoparticle-encapsulated molecule. There was no noticeable body weight loss after DSF treatment, which indicated that there was no major toxicity of DSF.

Conclusions

This systematic review provides evidence regarding the anti-tumor activity of DSF in vitro, in animals, and in humans and indicates the optimal forms of treatment to be evaluated in future research.

Peer Review reports

Introduction

Cancer is expected to be the leading cause of death and the foremost contributor to decreased life expectancy in every country worldwide during the twenty-first century and beyond [1]. Although comprehensive therapies prolong survival and improve the quality of life of cancer patients, approximately 96,000,000 cancer deaths occurred in 2018 worldwide [1]. The global community is well aware that new drug development, discovery, and synthesis are a time-consuming process, which involves intensive work and appraisal of the cost-effectiveness of the drug under development [2]. As a result, researchers are allocating considerable efforts for repurposing existing drugs such as disulfiram (DSF).

In the 1800s, DSF was used as an industrial catalyst in the production of rubber [3]. In 1948, DSF was approved by the Food and Drug Administration for treating alcoholism [4]. In 1988, DSF was associated with a decrease in the occurrence of occasional infections in symptomatic patients with human immunodeficiency virus infection [5], prompting the conduct of a wealth of clinical trials, some of which are still ongoing (www.clinicaltrials.gov). The antineoplastic activity of DSF was first recorded in 1977 by Dr. Lewison in a 35-year-old female breast cancer patient with systemic metastases who received DSF for her severe alcoholic syndrome and remained clinically free of cancer for 10 years without receiving any form of anticancer therapy [6]. This observation was noted in an era in which the anticancer effect of DSF was being researched. In recent years, a large number of preclinical studies and clinical trials (www.clinicaltrials.gov) of DSF have been conducted to explore the anticancer activities of this drug. Nonetheless, the antitumor effectiveness of DSF remains uncertain owing to existing heterogeneity across different studies with cell lines, animals, and humans. Currently, a systematic review of these studies to assess and clarify the anticancer potential of DSF is lacking.

It is worthy to explore whether there are substantial differences and are appropriate for clinical proposals. Therefore, this study aimed to perform a systematic review of published data on the antitumor activity of DSF. Specifically, this review aimed to assess the apoptosis and tumor inhibition rates of DSF based on data from studies in cell lines and animal models, respectively, and examine the benefit of DSF on progression-free survival (PFS) and overall survival (OS) based on results from clinical studies, regardless of the study design or type of cancer investigated. Meanwhile, it is important for evaluating the anti-tumor effect of disulfiram to include in the side effects. The side effects of disulfiram will be covered in this article.

Materials and methods

The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed to conduct this systematic review [7].

Search strategy

PubMed, Embase, and Cochrane Library databases were searched for relevant studies from their inception to the end of July 2019. The search was performed with a combination of Medical Subject Headings and free words as follows: (neoplasia OR neoplasm OR tumor OR cancer OR malignancy OR malignant neoplasm), and [disulfiram OR bis (diethylthiocarbamoyl) disulfide OR tetraethylthioperoxydicarbonic diamide OR tetraethylthiuram disulfide OR tetraethylthiuram OR antabus OR antabuse teturam OR dicupral OR esperal OR alcophobin OR anticol]. The details of the search strategy are presented in the supplement.

Study selection

Studies that implemented the below criteria were included: (1) solid cancer cell lines, animals, or patients treated with DSF; (2) in vitro studies focusing on parameters of the apoptosis index (early apoptosis or early apoptosis plus late apoptosis) using annexin V-fluorescein isothiocyanate/propidium iodide double-staining analysis by flow cytometry, in vivo studies evaluating the tumor inhibition rate in cell-line-derived xenograft animal models, or studies in humans, which included OS and PFS as endpoints, to assess the effect of DSF in cancer patients; and (3) studies published in the English and Chinese language. There were no restrictions on the type of cancer studied. To avoid duplication of data, only the most recent and most comprehensive articles were included. Studies with incomplete data or conference abstracts were excluded. Two investigators (Ling Wang, Run Wan) independently screened the databases for studies based on the eligibility criteria. Any discrepancies were resolved by consulting a third researcher (Cong Zhou).

Data derivation

Two investigators (Ling Wang, Cong Zhou) independently extracted data from the inclusive studies. Inconsistencies between the two investigators were resolved by consulting a third reviewer (Run Wan). When required, we contacted the authors of the research for further information. A pre-designed structured outline was used to abstract data. The outline included the following fields: study type (in vitro, in vivo, clinical study, or case series); general information (first author, publication year, country, and study design); supplement used; anticancer treatment used; and outcomes (i.e., apoptosis rate, tumor inhibition rate, OS and PFS, as applicable). The results of each study included were summarized. Descriptive statistics were used for data analysis. Meta-analysis was not performed owing to substantial heterogeneity across studies.

Results

Study characteristics

The initial search yielded a total of 1278 studies. After excluding 274 irrelevant and duplicate studies, the full texts of 1004 studies were screened. Of these, 148 were considered eligible based on the availability of full texts as well as the description of target outcomes. Ultimately, 113 articles were removed (no full texts, n = 43; no target outcomes, n = 70), and 35 studies were selected. A detailed description of the steps followed during the retrieval process is provided in Fig. 1.

Fig. 1
figure 1

Flow diagram of literature search process

Of 35 selected studies, 21 were in vitro studies (Table 1), 11 were in vivo studies with animal models (Table 2), and three were clinical trials (Table 3). In in vitro studies, the most studied cancer was breast cancer (five studies) [8,9,10,11,12], while the A549 non-small cell lung cancer (NSCLC) cell line was the one most commonly used cell line (four studies) [13,14,15,16]. Three studies examined DSF as a single agent [17,18,19], and 17 studies examined DSF in combination with metal ions (Cu, Ag), chemotherapy, or radiation therapy [8,9,10,11,12,13,14,15,16, 20,21,22,23,24,25,26,27]. In addition, DSF was encapsulated in nanoparticles (DSF-NPs) in three studies [12, 16, 25].

Table 1 Effects of disulfiram on cell apoptosis rates from in vitro studies
Table 2 Effects of disulfiram on tumor inhibition rates from animal studies
Table 3 Effects of disulfiram on progression-free survival and overall survival from human studies

Of 11 animal studies, Balb/C nude mice were utilized in nine [28,29,30,31,32,33,34,35,36], whereas the remaining studies used KunMing or female SCID mice [37, 38]. Ten studies used subcutaneous tumor models by injecting cancer cell lines [26, 29, 31, 32, 34,35,36,37,38], and one study used an in situ tumor model [33]. Eleven studies had assessed the dimensions of tumor volume (V) using the same formula (V = 0.5 × length × width2) [28,29,30,31,32,33,34,35,36,37,38], nine studies assessed changes in body weight in mice [26,27,28,29,30,31,32,33,34, 37, 38], and six studies contained data regarding the toxicity of DSF [28, 29, 32,33,34,35]. In addition, eight of the animal studies used DSF by re-synthesizing the molecule with nanomaterials [28,29,30,31,32,33,34, 37].

The three human studies included participants with differing characteristics and cancer types. All three clinical trials investigated DSF as a combination therapy with chemotherapy or/and radiation therapy [39,40,41], while two studies reported on adverse events [39, 40].

Outcomes

Three cell lines and one animal study showed that treatment with DSF as a single agent induced apoptosis and increased the rate of tumor inhibition [17,18,19, 35]. Although the sensitivity between the various cell lines varied, dose-dependency was consistently observed.

The concentration-dependent increase in apoptosis and tumor inhibition rates was augmented by a combination therapy of DSF adding metal ions [copper (Cu), silver (Ag)] in 10 in vitro [8,9,10,11, 13,14,15, 20, 26, 42] and three in vivo studies [36,37,38]. The synergistic effect of Cis, DOX, TMZ, PTX, Gy, and DSF in induced apoptosis was significantly higher than that of DSF or Cis or DOX or TMZ or Gy alone [8,9,10, 21, 24, 42]. Tumor cell growth was significantly inhibited when DSF, chemotherapy, and radiation therapy were used simultaneously, as shown in the examined in vivo studies [30, 31, 35, 37].

Compared with free molecule, DSF encapsulated with nanomaterials significantly induced selective death-dependent apoptosis, especially in acidic conditions (pH = 6.5) in cancer cell lines. Eleven animal studies demonstrated that DSF modified by particular nanomaterials increased the tumor inhibition rate and that the anticancer activity was more obvious when chemotherapy (Cis) was combined with nanoencapsulated DSF [32].

Changes in body weight during the whole study period were analyzed in nine animal studies. With the exception of three reports of weight changes in DSF-treated or DSF-modified groups [30, 33, 36], other studies recorded that there was no noticeable body weight loss after DSF treatment or no significant difference in body weight changes across different groups [28,29,30,31,32, 34, 36, 38], which indicated that there was no major toxicity of DSF [28, 29, 32,33,34,35].

Many clinical trials have mentioned the use of DSF for solid tumors (www.clinicaltrials.gov). One study clearly analyzed the difference in PFS (5.9 versus 4.9 months) and OS (10.0 versus 7.1 months) between control and experimental groups [42]. PFS and OS both improved in the experimental groups. Two studies described PFS and OS of the entire research cohort, and the treatment efficacy seemed to be in contrast to historical data [39, 40]. Our systemic review included two single-arm trials in glioblastoma (GBM) patients and a randomized controlled trial in NSCLC patients. Although the two single-arm clinical trials did not compare treatment with a control group, positive effects were observed; e.g., a 40-year-old woman with unmethylated isocitrate dehydrogenase wild-type GBM had good health without any signs of tumor recurrence 33 months after study initiation.

Among the reported adverse effects, none were serious, and they were of grades 2–3. Adverse effects were reported in two studies and included diarrhea, nausea, dizziness, vomiting, motor neuropathy, and elevated alanine transaminase levels. Symptoms resolved quickly when the dose was reduced [39, 40].

All three studies show that DSF is safe and seems to prolong survival of cancer patients. Because of individual differences in patients, the response to DSF was also varied [39, 40, 42]. The optimal concentration and sensitivity type should be further explored by in vitro and animal studies.

Discussion

DSF is decomposed into diethyldithiocarbamate in the body and exhibits anticancer activities [43]. Considering that the loss of cellular proliferation control leads to the development of cancer, effective clinical therapies of cancer have been developed based on the principle of inducing apoptosis [44]. In the included animal studies, the tumor inhibition rate was utilized to evaluate antitumor efficiency by calculating tumor volume. Most studies included in this review revealed enhanced apoptosis and tumor inhibition rates with DSF treatment (Table 4).

Table 4 The summary of the findings

In recent years, metal-based complexes have been reported to exhibit anticancer activity [45]. Silver complexes demonstrate anti-tumor activity and display low toxicity in humans. The mechanism of action is related to their interaction with nucleic acids and proteins [46]. Metabolites of DSF chelate with metal ions, leading to alterations in the intracellular levels of metal ions, enhancement of oxidative stress, inhibition of the activities of superoxide dismutase or matrix metalloproteinases, inactivation of essential sulfhydryl groups by protein carbamoylation, and alteration of cancer cell invasion, tumor angiogenesis, and metastasis [47, 48]. The observation that the combination of DSF with metal ions (Cu, Ag) leads to enhanced anticancer effectiveness is in accordance with the observations of in vitro and animal experiments [11, 14].

In different cancer cell lines, the lethal concentration of DSF was different. The lethal concentration was reduced when DSF combined with metal ions or nano-reconstructed DSF.

The additive/synergistic action of DSF with other chemotherapy agents in inhibiting tumor cell growth and cytotoxicity is mediated through the enhancement of cellular oxidative stress, inhibition of P glycol-protein (P-gp) activity, and dysregulation of the NF-κB signaling pathway [8, 49, 50].

In the examined studies, anti-tumor activity, as evidenced by higher apoptosis and tumor inhibition rates, was enhanced with DSF-NPs in various ways. At the pH of 7.4, the half-life of DSF is 1–1.5 min [47]. The half-life was improved by nanomaterial packaging of DSF, with the anti-tumor effects increasing under acidic conditions (pH = 6.5) [51]. DSF-NPs enhanced cellular uptake, induced high levels of reactive oxygen species, activated the MAP-kinase pathway, sustained drug supply, and blocked copolymer micelles, such as the P-gp inhibitor [14, 20, 52]. Evidence supports that DSF-NPs ameliorate the instability and low treatment efficacy of free DSF.

Event-free survival (EFS) means that there are no adverse events since the start of treatment, including change of regimen, adverse side effects, intolerance, disease progression, and patient death. EFS represents a direct measure of the ability of the treatment to achieve a response, the durability of the response achieved, and its capacity to prolong life [53]. It was found that the doses of disulfiram significantly increased EFS [39].

Although our results may be more reliable than those of single studies, the present study has certain limitations. First, only articles published in English and Chinese were included; the non-inclusion of articles published in other languages may have had an effect on the results. Second, only some solid tumors were included, not referred to non-solid tumor (hematological malignancy). Third, the scarcity of the studies in general (35 in total) and the fact that they are performed on different cancers may make any specific conclusions difficult. Finally, no quality evaluation was conducted, and the majority of studies were animal and cell experiments; thus, the translation of these results to benefits in the clinic needs to be determined.

In conclusion, many studies have investigated the antineoplastic activity of DSF. This systematic review provides evidence of the antineoplastic activity of DSF in vitro, in in vivo animal models, and in humans. DSF could induce cancer cell apoptosis in cell experiments and inhibit cancer cell growth in animal experiments. Administration of DSF as a combination therapy or as a nanoparticle-encapsulated molecule seems to enhance its effectiveness. Meanwhile, DSF hardly affect the animal weight. Above of all, DSF is effectiveness and safety. These findings may serve as the basis for designing clinical studies of DSF in the future.

Availability of data and materials

Not applicable.

Abbreviations

DSF:

Disulfiram

PFS:

Progression-free survival

OS:

Overall survival

EFS:

Event-free survival

TIR:

Tumor inhibition rate

References

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.

    Article  PubMed  Google Scholar 

  2. Kaitin KI. Deconstructing the drug development process: the new face of innovation. Clin Pharmacol Ther. 2010;87(3):356–61.

    Article  CAS  PubMed  Google Scholar 

  3. Suh JJ, Pettinati HM, Kampman KM, O’Brien CP. The status of disulfiram: a half of a century later. J Clin Psychopharmacol. 2006;26(3):290–302.

    Article  CAS  PubMed  Google Scholar 

  4. Eneanya DI, Bianchine JR, Duran DO, Andresen BD. The actions of metabolic fate of disulfiram. Annu Rev Pharmacol Toxicol. 1981;21:575–96.

    Article  CAS  PubMed  Google Scholar 

  5. Gotzsche PC. Ditiocarb in HIV infection. Lancet. 1988;2(8618):1024.

    Article  CAS  PubMed  Google Scholar 

  6. Lewison EF. Spontaneous regression of breast cancer. Prog Clin Biol Res. 1977;12:47–53.

  7. Moher D, Liberati A, Tetzlaff J, Altman DG, PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. J Clin Epidemiol. 2009;62(10):1006–12.

    Article  PubMed  Google Scholar 

  8. 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;113:108727.

    Article  CAS  PubMed  Google Scholar 

  9. Wu L, Meng F, Dong L, 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.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Yang Y, Zhang K, Wang Y, et al. Disulfiram chelated with copper promotes apoptosis in human breast cancer cells by impairing the mitochondria functions. Scanning. 2016;38(6):825–36.

    Article  CAS  PubMed  Google Scholar 

  11. Kim JY, Cho Y, Oh E, et al. Disulfiram targets cancer stem-like properties and the HER2/Akt signaling pathway in HER2-positive breast cancer. Cancer Lett. 2016;379(1):39–48.

    Article  CAS  PubMed  Google Scholar 

  12. Duan X, Xiao J, Yin Q, et al. Multi-targeted inhibition of tumor growth and lung metastasis by redox-sensitive shell crosslinked micelles loading disulfiram. Nanotechnology. 2014;25(12):125102.

    Article  PubMed  CAS  Google Scholar 

  13. 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.

    Article  CAS  PubMed  Google Scholar 

  14. Chen W, Yang W, Chen PY, Huang YZ, Li F. Disulfiram copper nanoparticles prepared with a stabilized metal ion ligand complex method for treating drug-resistant prostate cancers. ACS Appl Mater Interfaces. 2018;10(48):41118–28.

    Article  CAS  PubMed  Google Scholar 

  15. Butcher K, Kannappan V, Kilari RS, et al. Investigation of the key chemical structures involved in the anticancer activity of disulfiram in A549 non-small cell lung cancer cell line. BMC Cancer. 2018;18(1):753.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Wang C, Yang J, Han H, et al. Disulfiram-loaded porous PLGA microparticle for inhibiting the proliferation and migration of non-small-cell lung cancer. Int J Nanomedicine. 2017;12:827–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sharma V, Verma V, Lal N, et al. Disulfiram and its novel derivative sensitize prostate cancer cells to the growth regulatory mechanisms of the cell by re-expressing the epigenetically repressed tumor suppressor-estrogen receptor beta. Mol Carcinog. 2016;55(11):1843–57.

    Article  CAS  PubMed  Google Scholar 

  18. Rezkk Y, Yang S, Bai K, et al. Disulfiram’s antineoplastic effects on ovarian cancer bulk tumor cells and the stem cell population: a study in ovarian cancer cell lines and in rodents. Gynecol Oncol. 2013;130(1):e135–6.

    Article  Google Scholar 

  19. Dastjerdi MN, Babazadeh Z, Rabbani M, Gharagozloo M, Esmaeili A, Narimani M. Effects of disulfiram on apoptosis in PANC-1 human pancreatic cancer cell line. Res Pharm Sci. 2014;9(4):287–94.

    PubMed  PubMed Central  Google Scholar 

  20. Guo F, Yang Z, Kulbe H, Albers AE, Sehouli J. Inhibitory effect on ovarian cancer ALDH+ stem-like cells by Disulfiram and Copper treatment through ALDH and ROS modulation. Biomed Pharmacother. 2019;118:109371.

    Article  CAS  PubMed  Google Scholar 

  21. Kaufmann, Albers AE, Yao WH, Qian X, Kinghammer K, Ochsenreither S. Disulfiram (Antabuse®) acts as potent radio-chemo sensitizer of HNSCC and derived stem cells in vitro. Laryngo Rhino Otol. 2018;97(S 02):S73.

    Google Scholar 

  22. Yang Y, Li M, Sun X, et al. The selective cytotoxicity of DSF-Cu attributes to the biomechanical properties and cytoskeleton rearrangements in the normal and cancerous nasopharyngeal epithelial cells. Int J Biochem Cell Biol. 2017;84:96–108.

    Article  CAS  PubMed  Google Scholar 

  23. Abu-Serie MM, El-Rashidy FH. In vitro collapsing colon cancer cells by selectivity of disulfiram-loaded charge switchable nanoparticles against cancer stem cells. Recent Pat Anticancer Drug Discov. 2017;12(3):260–71.

    Article  CAS  PubMed  Google Scholar 

  24. Zhao Y, Xiao Z, Chen WN, Yang JS, Li T, Fan B. Disulfiram sensitizes pituitary adenoma cells to temozolomide by regulating O6-methylguanine-DNA methyltransferase expression. Mol Med Rep. 2015;12(2):2313–22.

    Article  CAS  PubMed  Google Scholar 

  25. Zhang L, Tian B, Li Y, et al. A copper-mediated disulfiram-loaded pH-triggered PEG-shedding TAT peptide-modified lipid nanocapsules for use in tumor therapy. ACS Appl Mater Interfaces. 2015;7(45):25147–61.

    Article  CAS  PubMed  Google Scholar 

  26. Han JB, Liu LM, Yue XQ, Chang JJ, Shi WD, Hua YQ. A binuclear complex constituted by diethyldithiocarbamate and copper(I) functions as a proteasome activity inhibitor in pancreatic cancer cultures and xenografts. Toxicol Appl Pharmacol. 2013;273(3):477–83.

    Article  CAS  PubMed  Google Scholar 

  27. Cen DZ, Gonzalez RI, Buckmeier JA, Kahlon RS, Tohidian NB, Meyskens FL. Disulfiram induces apoptosis in human melanoma cells: a redox-related process. Mol Cancer Ther. 2002;1(3):197–204.

    CAS  PubMed  Google Scholar 

  28. Peng X, Pan Q, Zhang B, et al. Highly stable, coordinated polymeric nanoparticles loading copper (II) diethyldithiocarbamate for combinational chemo/chemodynamic therapy of cancer. Biomacromolecules. 2019;20(6):2372–83.

    Article  CAS  PubMed  Google Scholar 

  29. Banerjee P, Geng T, Mahanty A, Li TT, Zong L, Wang B. Integrating the drug, disulfiram into the vitamin E-TPGS-modified PEGylated nanostructured lipid carriers to synergize its repurposing for anti-cancer therapy of solid tumors. Int J Pharm. 2019;557:374–89.

    Article  CAS  PubMed  Google Scholar 

  30. Ji YB, Liu B, Yu RQ, et al. Preparation of disulfiram naonosuspensions and their anti-tumor efficacy in vitro and in vivo. Acta Pharm Sin. 2019;54(3):565–73.

    Google Scholar 

  31. Tao X, Gou J, Zhang Q, et al. Synergistic breast tumor cell killing achieved by intracellular co-delivery of doxorubicin and disulfiram via core-shell-corona nanoparticles. Biomater Sci. 2018;6(7):1869–81.

    Article  CAS  PubMed  Google Scholar 

  32. Song W, Tang Z, Lei T, et al. Stable loading and delivery of disulfiram with mPEG-PLGA/PCL mixed nanoparticles for tumor therapy. Nanomedicine. 2016;12(2):377–86.

    Article  CAS  PubMed  Google Scholar 

  33. Fasehee H, Dinarvand R, Ghavamzadeh A, et al. Delivery of disulfiram into breast cancer cells using folate-receptor-targeted PLGA-PEG nanoparticles: in vitro and in vivo investigations. J Nanobiotechnology. 2016;14:32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Song W, Tang Z, Shen N, et al. Combining disulfiram and poly(l-glutamic acid)-cisplatin conjugates for combating cisplatin resistance. Control Release. 2016;231:94–102.

    Article  CAS  Google Scholar 

  35. Choi SA, Choi JW, Wang KC, et al. Disulfiram modulates stemness and metabolism of brain tumor initiating cells in atypical teratoid/rhabdoid tumors. Neuro Oncol. 2015;17(6):810–21.

    Article  CAS  PubMed  Google Scholar 

  36. Cheriyan VT, Wang Y, Muthu M, et al. Disulfiram suppresses growth of the malignant pleural mesothelioma cells in part by inducing apoptosis. PLoS One. 2014;9(4):e93711.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Zhou L, Yang L, Yang C, et al. Membrane loaded copper oleate PEGylated liposome combined with disulfiram for improving synergistic antitumor effect in vivo. Pharm Res. 2018;35(7):147.

    Article  PubMed  CAS  Google Scholar 

  38. Allensworth JL, Evans MK, Bertucci F, 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Huang J, Chaudhary R, Cohen AL, et al. A multicenter phase II study of temozolomide plus disulfiram and copper for recurrent temozolomide-resistant glioblastoma. J Neurooncol. 2019;142(3):537–44.

    Article  CAS  PubMed  Google Scholar 

  40. Huang J, Campian JL, Gujar AD, 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.

    Article  CAS  PubMed  Google Scholar 

  41. You SY, Rui W, Chen ST, et al. Process of immunogenic cell death caused by disulfiram as the anti-colorectal cancer candidate. Biochem Biophys Res Commun. 2019;513(4):891–7.

    Article  CAS  PubMed  Google Scholar 

  42. 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):366–7.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Ekinci E, Rohondia S, Khan R, Dou QP. Repurposing disulfiram as an anti-cancer agent: updated review on literature and patents. Recent Pat Anticancer Drug Discov. 2019;14(2):113–32.

    Article  CAS  PubMed  Google Scholar 

  44. D’Arcy MS. Cell death: a review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int. 2019;43(6):582–92.

    Article  PubMed  Google Scholar 

  45. Schmitt SM, Frezza M, Dou QP. New applications of old metal-binding drugs in the treatment of human cancer. Front Biosci (Schol Ed). 2012;4:375–91.

    Article  Google Scholar 

  46. Banti CN, Hadjikakou SK. Anti-proliferative and anti-tumor activity of silver(I) compounds. Metallomics. 2013;5(6):569–96.

    Article  CAS  PubMed  Google Scholar 

  47. Agarwal RP, Phillips M, McPherson RA, Hensley P. Serum albumin and the metabolism of disulfiram. Biochem Pharmacol. 1986;35(19):3341–7.

    Article  CAS  PubMed  Google Scholar 

  48. Salem K, McCormick ML, Wendlandt E, Zhan FH, Goel A. Copper-zinc superoxide dismutase-mediated redox regulation of bortezomib resistance in multiple myeloma. Redox Biol. 2015;4:23–33.

    Article  CAS  PubMed  Google Scholar 

  49. Majera D, Skrott Z, Bouchal J, 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.

    Article  CAS  PubMed  Google Scholar 

  50. Calderon-Aparicio A, Cornejo A, Orue A, Rieber M. Anticancer 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.

    PubMed  PubMed Central  Google Scholar 

  51. Miao L, Su J, Zhuo X, 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.

    Article  CAS  PubMed  Google Scholar 

  52. Estey E, Othus M, Lee SJ, Appelbaum FR, Gale RP. New drug approvals in acute myeloid leukemia: what’s the best end point? Leukemia. 2016;30(3):521–5.

    Article  CAS  PubMed  Google Scholar 

  53. Huo Q, Zhu J, Niu 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by grants from the National Natural Science Foundation of China (Grant No. 31770537), the International Science and Technology Cooperation Project of Gansu Province (Grant No. 18YF1WA113), and Fundamental Research Funds for the Central Universities (lzujbky-2021-ct18).

Author information

Authors and Affiliations

Authors

Contributions

All authors read and approved the final version of this article. Ling Wang conceptualized the review, conducted the literature search, and was responsible for data extraction, data analysis, and for writing the original draft of this manuscript. Cong Zhou was responsible for data extraction. Run Wan conducted the literature search and served as an independent reviewer of the data extracted from the studies, responsible for solving any inconsistencies between Ling Wang and Cong Zhou. Yang Yu contributed to the analysis and interpretation of study data. Yumin Li conceptualized the review, supervised the process, and was responsible for project administration and manuscript review.

Corresponding author

Correspondence to Yumin Li.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

Ling Wang, Yang Yu, Cong Zhou, Run Wan, and Yumin Li declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, L., Yu, Y., Zhou, C. et al. Anticancer effects of disulfiram: a systematic review of in vitro, animal, and human studies. Syst Rev 11, 109 (2022). https://doi.org/10.1186/s13643-021-01858-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13643-021-01858-4

Keywords