Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
Add more filters

Publication year range
1.
Int J Pharm ; 652: 123800, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38218507

ABSTRACT

The ancient anti-alcohol drug disulfiram (DSF) has gained widespread attention for its highly effective anti-tumor effects in cancer treatment. Our previous studies have developed liposome of Cu (DDC)2 to overcome the limitations, like the poor water solubility. However, Cu (DDC)2 liposomes still have shown difficulties in severe hemolytic reactions at high doses and systemic toxicity, which have limited their clinical use. Therefore, this study aims to exploratively investigate the feasibility of using DSF or DDC in combination also can chelate Zn2+ to form zinc diethyldithiocarbamate (Zn (DDC)2). Furthermore, this study prepared stable and homogeneous Zn (DDC)2 liposomes, which were able to be released in the tumor microenvironment (TME). The released Zn (DDC)2 was converted to Cu (DDC)2 with the help of endogenous Cu2+-switch enriched in the TME, which has a higher stability constant compared with Zn (DDC)2. In other words, the Cu2+-switch is activated at the tumor site, completing the conversion of the less cytotoxic Zn (DDC)2 to the more cytotoxic Cu (DDC)2 for effective tumor therapy so that the Zn (DDC)2 liposomes in vivo achieved the comparable therapeutic efficacy and provided a safer alternative to Cu (DDC)2 liposomes in cancer therapy.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Liposomes/therapeutic use , Ditiocarb/therapeutic use , Disulfiram , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Zinc , Copper/therapeutic use , Tumor Microenvironment , Aromatic-L-Amino-Acid Decarboxylases/therapeutic use
2.
ACS Biomater Sci Eng ; 9(11): 6425-6437, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37874613

ABSTRACT

Melanoma is a malignancy of the skin that is resistant to conventional treatment, necessitating the development of effective and safe new therapies. The percutaneous microneedle (MN) system has garnered increasing interest as a viable treatment option due to its high efficacy, minimal invasiveness, painlessness, and secure benefits. In this investigation, a sensitive MN system with multiple functions was created to combat melanoma effectively. This MN system utilized polyvinylpyrrolidone (PVP) as microneedle substrates and biocompatibility panax notoginseng polysaccharide (PNPS) as microneedle tips, which encapsulated PVP-stabilized CuO2 nanoparticles as a therapeutic agent and disulfiram-containing F127 micelles to enhance the tumor treatment effect. The MN system had sufficient mechanical properties to pierce the skin, and the excellent water solubility of PNPS brought high-speed dissolution properties under the bio conditions, allowing the MNs to effectively penetrate the skin and deliver the CuO2 nanoparticles as well as the drug-loaded micelles to the melanoma site. CuO2 nanoparticles released by the MN system generated Cu2+ and H2O2 in the tumor acidic environment to achieve self-supply of hydrogen peroxide to chemodynamic therapy (CDT). In addition, Cu2+ was chelated with disulfiram to produce CuET, which killed tumor cells. And the MN system had excellent near-infrared (NIR) photothermal properties due to the loading of CuO2 nanoparticles and induced localized thermotherapy in the melanoma region to further inhibit tumor growth. Thus, the designed MN system accomplished effective tumor suppression and minimal side effects in vivo via combined therapy, offering patients a safe and effective option for melanoma treatment.


Subject(s)
Disulfiram , Melanoma , Humans , Disulfiram/pharmacology , Disulfiram/therapeutic use , Photothermal Therapy , Micelles , Hydrogen Peroxide , Melanoma/drug therapy , Povidone
3.
Front Biosci (Landmark Ed) ; 28(8): 186, 2023 08 28.
Article in English | MEDLINE | ID: mdl-37664913

ABSTRACT

BACKGROUND: The complex formed by disulfiram (DSF) and copper (Cu) is safe and effective for the prevention and treatment of triple-negative breast cancer (TNBC). Although previous studies have shown that DSF/Cu induces ferroptosis, the mechanism remains unclear. METHODS: The mitochondrial morphology of TNBC treated with DSF/Cu was observed by transmission microscopy, and intracellular levels of iron, lipid reactive oxygen species (ROS), malondialdehyde, and glutathione were evaluated to detect the presence of ferroptosis. Target genes for the DSF/Cu-activated ferroptosis signaling pathway were examined by transcriptome sequencing analysis. Expression of the target gene, HOMX1, was detected by qRT-PCR, immunofluorescence and western blot. RESULTS: The mitochondria of TNBC cells were significantly atrophied following treatment with DSF/Cu for 24 h. Addition of DSF/Cu supplement resulted in significant up-regulation of intracellular iron, lipid ROS and malondialdehyde levels, and significant down-regulation of glutathione levels, all of which are important markers of ferroptosis. Transcriptome analysis confirmed that DSF/Cu activated the ferroptosis signaling pathway and up-regulated several ferroptosis target genes associated with redox regulation, especially heme oxygenase-1 (HMOX-1). Inhibition of ferroptosis by addition of the ROS scavenger N-acetyl-L-cysteine (NAC) significantly increased the viability of DSF/Cu-treated TNBC cells. CONCLUSIONS: These results show that DSF/Cu increases lipid peroxidation and causes a sharp increase in HMOX1 activity, thereby inducing TNBC cell death through ferroptosis. DSF/Cu is a promising therapeutic drug for TNBC and could lead to ferroptosis-mediated therapeutic strategies for human cancer.


Subject(s)
Ferroptosis , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Copper/pharmacology , Disulfiram/pharmacology , Ferroptosis/genetics , Reactive Oxygen Species , Cell Line , Glutathione , Lipids
4.
Clin Ther ; 45(11): 1151-1154, 2023 11.
Article in English | MEDLINE | ID: mdl-37722955

ABSTRACT

Alcohol consumption, even minimal, can exacerbate the disulfiram-like reaction (also referred to as acetaldehyde syndrome) that occurs with the use of medications that impede the breakdown of acetaldehyde. Such medications include Ginaton, a proprietary tablet formulation of Ginkgo biloba extract commonly used in Europe, Asia, and the United States for cardiovascular and nervous system health. This article details such a case from China. Healthcare providers should be proactive in educating patients about the potential adverse reactions related to using Ginaton and the importance of avoiding alcohol consumption while using it. Patients should also be advised to disclose their alcohol-consumption habits and seek medical advice before initiating treatment with any medication or supplement during treatment with Ginaton.


Subject(s)
Disulfiram , Drugs, Chinese Herbal , Humans , Acetaldehyde , Disulfiram/adverse effects , Drugs, Chinese Herbal/adverse effects , Heart
5.
Int J Pharm ; 640: 123036, 2023 Jun 10.
Article in English | MEDLINE | ID: mdl-37169106

ABSTRACT

Disulfiram (DS) is an anti-alcoholism drug capable of acting against important and hard-to-treat cancers. The drug's relative instability and variable absorption/distribution have led to its variable pharmacokinetics and suboptimal exposure. Hence, it was hypothesised that a nano-enabled form of DS might be able to overcome such limitations. Encapsulation of the labile DS was achieved with quaternary ammonium palmitoyl glycol chitosan (GCPQ) to form a high-capacity, soybean oil-based DS-GCPQ nanoemulsion. DS-GCPQ showed capability of oil-loading up to 50% v/v for a stable entrapment of high drug content. With increasing oil content (10 to 50% v/v), the mean particle size and polydispersity index were also increased (166 to 351 nm and 0.14 to 0.22, respectively) for a given amount of GCPQ. Formulations showed a highly positive particle surface charge (50.9 ± 1.3 mV), contributing to the colloidal stability of the individual particles. DS-GCPQ showed marked cytotoxicity against pancreatic cancer cell lines with enhanced activity in the presence of copper. An intravenous pharmacokinetic study of DS-GCPQ in vivo showed improved plasma drug stability with a DS half-life of 17 min. Prolonged survival was seen in tumour-bearing animals treated with DS-GCPQ supplemented with copper. In conclusion, DS-GCPQ nanoemulsion has the potential to be developed further for cancer therapeutic purposes.


Subject(s)
Chitosan , Disulfiram , Nanoparticles , Animals , Copper , Drug Compounding , Emulsions , Micelles , Particle Size , Chitosan/chemistry , Nanoparticle Drug Delivery System
6.
Cell Death Differ ; 30(7): 1666-1678, 2023 07.
Article in English | MEDLINE | ID: mdl-37142656

ABSTRACT

Drug repurposing is a versatile strategy to improve current therapies. Disulfiram has long been used in the treatment of alcohol dependency and multiple clinical trials to evaluate its clinical value in oncology are ongoing. We have recently reported that the disulfiram metabolite diethyldithiocarbamate, when combined with copper (CuET), targets the NPL4 adapter of the p97VCP segregase to suppress the growth of a spectrum of cancer cell lines and xenograft models in vivo. CuET induces proteotoxic stress and genotoxic effects, however important issues concerning the full range of the CuET-evoked tumor cell phenotypes, their temporal order, and mechanistic basis have remained largely unexplored. Here, we have addressed these outstanding questions and show that in diverse human cancer cell models, CuET causes a very early translational arrest through the integrated stress response (ISR), later followed by features of nucleolar stress. Furthermore, we report that CuET entraps p53 in NPL4-rich aggregates leading to elevated p53 protein and its functional inhibition, consistent with the possibility of CuET-triggered cell death being p53-independent. Our transcriptomics profiling revealed activation of pro-survival adaptive pathways of ribosomal biogenesis (RiBi) and autophagy upon prolonged exposure to CuET, indicating potential feedback responses to CuET treatment. The latter concept was validated here by simultaneous pharmacological inhibition of RiBi and/or autophagy that further enhanced CuET's tumor cytotoxicity, using both cell culture and zebrafish in vivo preclinical models. Overall, these findings expand the mechanistic repertoire of CuET's anti-cancer activity, inform about the temporal order of responses and identify an unorthodox new mechanism of targeting p53. Our results are discussed in light of cancer-associated endogenous stresses as exploitable tumor vulnerabilities and may inspire future clinical applications of CuET in oncology, including combinatorial treatments and focus on potential advantages of using certain validated drug metabolites, rather than old, approved drugs with their, often complex, metabolic profiles.


Subject(s)
Disulfiram , Neoplasms , Animals , Humans , Cell Line, Tumor , Disulfiram/metabolism , Neoplasms/metabolism , Ribosomes/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish/metabolism
7.
Biomolecules ; 12(11)2022 10 31.
Article in English | MEDLINE | ID: mdl-36358950

ABSTRACT

Oxidative stress plays a central role in the pathophysiology of melanoma. Curcumin (CUR) is a polyphenolic phytochemical that stimulates reactive oxygen species (ROS) production, while disulfiram (DSS) is a US FDA-approved drug for the treatment of alcoholism that can act by inhibiting the intracellular antioxidant system. Therefore, we hypothesized that they act synergistically against melanoma cells. Herein, we aimed to study the antitumor potential of the combination of CUR with DSS in B16-F10 melanoma cells using in vitro and in vivo models. The cytotoxic effects of different combination ratios of CUR and DSS were evaluated using the Alamar Blue method, allowing the production of isobolograms. Apoptosis detection, DNA fragmentation, cell cycle distribution, and mitochondrial superoxide levels were quantified by flow cytometry. Tumor development in vivo was evaluated using C57BL/6 mice bearing B16-F10 cells. The combinations ratios of 1:2, 1:3, and 2:3 showed synergic effects. B16-F10 cells treated with these combinations showed improved apoptotic cell death and DNA fragmentation. Enhanced mitochondrial superoxide levels were observed at combination ratios of 1:2 and 1:3, indicating increased oxidative stress. In vivo tumor growth inhibition for CUR (20 mg/kg), DSS (60 mg/kg), and their combination were 17.0%, 19.8%, and 28.8%, respectively. This study provided data on the potential cytotoxic activity of the combination of CUR with DSS and may provide a useful tool for the development of a therapeutic combination against melanoma.


Subject(s)
Antineoplastic Agents , Curcumin , Melanoma, Experimental , Mice , Animals , Curcumin/pharmacology , Curcumin/therapeutic use , Disulfiram/pharmacology , Cell Line, Tumor , Superoxides/metabolism , Mice, Inbred C57BL , Melanoma, Experimental/metabolism , Apoptosis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Oxidative Stress
8.
Biomater Sci ; 10(21): 6267-6281, 2022 Oct 25.
Article in English | MEDLINE | ID: mdl-36128848

ABSTRACT

Conventional treatments for cancer, such as chemotherapy, surgical resection, and radiotherapy, have shown limited therapeutic efficacy, with severe side effects, lack of targeting and drug resistance for monotherapies, which limit their clinical application. Therefore, combinatorial strategies have been widely investigated in the battle against cancer. Herein, we fabricated a dual-targeted nanoscale drug delivery system based on EpCAM aptamer- and lactic acid-modified low-polyamidoamine dendrimers to co-deliver the FDA-approved agent disulfiram and photosensitizer indocyanine green, combining the imaging and therapeutic functions in a single platform. The multifunctional nanoparticles with uniform size had high drug-loading payload, sustained release, as well as excellent photothermal conversion. The integrated nanoplatform showed a superior synergistic effect in vitro and possessed precise spatial delivery to HepG2 cells with the dual-targeting nanocarrier. Intriguingly, a robust anticancer response of chemo-phototherapy was achieved; chemotherapy combined with the efficacy of phototherapy to cause cellular apoptosis of HepG2 cells (>35%) and inhibit the regrowth of damaged cells. Furthermore, the theranostic nanosystem displayed fluorescence imaging in vivo, attributed to its splendid accumulation in the tumor site, and it provided exceptional tumor inhibition rate against liver cancer cells (>76%). Overall, our research presents a promising multifunctional theranostic nanoplatform for the development of synergistic therapeutics for tumors in further applications.


Subject(s)
Dendrimers , Hyperthermia, Induced , Nanoparticles , Neoplasms , Humans , Indocyanine Green/pharmacology , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Epithelial Cell Adhesion Molecule , Doxorubicin/pharmacology , Delayed-Action Preparations , Precision Medicine , Disulfiram , Drug Delivery Systems/methods , Neoplasms/therapy , Lactic Acid , Hyperthermia, Induced/methods , Drug Liberation , Theranostic Nanomedicine/methods , Cell Line, Tumor
9.
Cell Death Dis ; 13(3): 203, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35246527

ABSTRACT

Despite several approved therapeutic modalities, multiple myeloma (MM) remains an incurable blood malignancy and only a small fraction of patients achieves prolonged disease control. The common anti-MM treatment targets proteasome with specific inhibitors (PI). The resulting interference with protein degradation is particularly toxic to MM cells as they typically accumulate large amounts of toxic proteins. However, MM cells often acquire resistance to PIs through aberrant expression or mutations of proteasome subunits such as PSMB5, resulting in disease recurrence and further treatment failure. Here we propose CuET-a proteasome-like inhibitor agent that is spontaneously formed in-vivo and in-vitro from the approved alcohol-abuse drug disulfiram (DSF), as a readily available treatment effective against diverse resistant forms of MM. We show that CuET efficiently kills also resistant MM cells adapted to proliferate under exposure to common anti-myeloma drugs such as bortezomib and carfilzomib used as the first-line therapy, as well as to other experimental drugs targeting protein degradation upstream of the proteasome. Furthermore, CuET can overcome also the adaptation mechanism based on reduced proteasome load, another clinically relevant form of treatment resistance. Data obtained from experimental treatment-resistant cellular models of human MM are further corroborated using rather unique advanced cytotoxicity experiments on myeloma and normal blood cells obtained from fresh patient biopsies including newly diagnosed as well as relapsed and treatment-resistant MM. Overall our findings suggest that disulfiram repurposing particularly if combined with copper supplementation may offer a promising and readily available treatment option for patients suffering from relapsed and/or therapy-resistant multiple myeloma.


Subject(s)
Antineoplastic Agents , Multiple Myeloma , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bortezomib/pharmacology , Bortezomib/therapeutic use , Cell Line, Tumor , Disulfiram/pharmacology , Drug Repositioning , Drug Resistance, Neoplasm , Humans , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/drug therapy , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use
10.
Biomater Sci ; 10(5): 1384-1392, 2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35142760

ABSTRACT

Disulfiram (DSF) is nontoxic and exerts anticancer activity by forming highly toxic chelates via its metabolite diethyldithiocarbamate with transition metal ions. However, there are not enough such ions in the human body to maintain the therapeutic effect. Herein, we report nanogels that complex copper ions or cis-platinum (CDDP) for tumor metal delivery to potentiate DSF's antitumor efficacy. We synthesized zwitterionic poly[N-(3-(methacryloyloxy-2-hydroxy)propyl)]-N-methyl glycine (PGMA-SAR) capable of chelating copper ions or CDDP and formed nanogels with suitable size and zeta potential. The intravenously injected nanogels circulated long in the blood compartment and delivered a high concentration of metal ions to the tumor. Separately administered DSF could sequester the metal ions from the nanogels and form highly cytotoxic complexes with potent in vitro and in vivo anticancer activity. This study provides a new strategy to potentiate DSF in anticancer treatment.


Subject(s)
Copper , Disulfiram , Cell Line, Tumor , Cisplatin/pharmacology , Disulfiram/pharmacology , Ditiocarb/pharmacology , Humans , Nanogels
11.
Biomaterials ; 281: 121335, 2022 02.
Article in English | MEDLINE | ID: mdl-34979419

ABSTRACT

Disulfiram (DSF) has been used as an alcoholism drug for 70 years. Recently, it has attracted increasing attention owing to the distinguished anticancer activity, which can be further potentiated by the supplementation of Cu2+. Although encouraging anticancer results are obtained in lab, the clinical outcomes of oral DSF are not satisfactory, which urges an in-depth understanding of the underlying mechanisms, bottlenecks, and proposal of potential methods to address the dilemma. In this review, a critical summarization of various molecular biological anticancer mechanisms of DSF/Cu2+ is provided and the predicament of orally delivering DSF in clinical oncotherapy is explained by the metabolic barriers. We highlight the recent advances in the DSF/Cu2+ delivery strategies and the emerging treatment regimens for cancer treatment. Last but not the least, we summarize the clinical trials regarding DSF and make a prospect of DSF/Cu-based cancer therapy.


Subject(s)
Disulfiram , Neoplasms , Cell Line, Tumor , Copper/pharmacology , Disulfiram/pharmacology , Disulfiram/therapeutic use , Humans , Neoplasms/drug therapy
12.
J Nanobiotechnology ; 19(1): 290, 2021 Sep 27.
Article in English | MEDLINE | ID: mdl-34579711

ABSTRACT

BACKGROUND: In comparison with traditional therapeutics, it is highly preferable to develop a combinatorial therapeutic modality for nanomedicine and photothermal hyperthermia to achieve safe, efficient, and localized delivery of chemotherapeutic drugs into tumor tissues and exert tumor-activated nanotherapy. Biocompatible organic-inorganic hybrid hollow mesoporous organosilica nanoparticles (HMONs) have shown high performance in molecular imaging and drug delivery as compared to other inorganic nanosystems. Disulfiram (DSF), an alcohol-abuse drug, can act as a chemotherapeutic agent according to its recently reported effectiveness for cancer chemotherapy, whose activity strongly depends on copper ions. RESULTS: In this work, a therapeutic construction with high biosafety and efficiency was proposed and developed for synergistic tumor-activated and photothermal-augmented chemotherapy in breast tumor eradication both in vitro and in vivo. The proposed strategy is based on the employment of HMONs to integrate ultrasmall photothermal CuS particles onto the surface of the organosilica and the molecular drug DSF inside the mesopores and hollow interior. The ultrasmall CuS acted as both photothermal agent under near-infrared (NIR) irradiation for photonic tumor hyperthermia and Cu2+ self-supplier in an acidic tumor microenvironment to activate the nontoxic DSF drug into a highly toxic diethyldithiocarbamate (DTC)-copper complex for enhanced DSF chemotherapy, which effectively achieved a remarkable synergistic in-situ anticancer outcome with minimal side effects. CONCLUSION: This work provides a representative paradigm on the engineering of combinatorial therapeutic nanomedicine with both exogenous response for photonic tumor ablation and endogenous tumor microenvironment-responsive in-situ toxicity activation of a molecular drug (DSF) for augmented tumor chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Drug Therapy/methods , Nanomedicine , Nanoparticles/therapeutic use , Photothermal Therapy/methods , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Copper , Disulfiram/pharmacology , Ditiocarb , Female , Mice, Inbred BALB C , Mice, Nude , Particle Size , Phototherapy , Tumor Microenvironment/drug effects
13.
Drug Alcohol Depend ; 227: 108964, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34518028

ABSTRACT

BACKGROUND: Pharmacotherapy for alcohol use disorders (AUD) is effective. However, knowledge about utilization of, and patient characteristics associated with prescriptions is scarce. The aim is to investigate prescriptions of pharmacotherapy for AUD in Sweden across time, sociodemographics, domicile and comorbid conditions. METHOD: This is a national cohort study, comprising 132 733 adult patients with AUD diagnosis between 2007 and 2015. The exposure variables were age, sex, income, education, family constellation, domicile, origin, concurrent psychiatric and somatic co-morbid diagnoses. Logistic regression analyses were used to obtain odds ratios (OR) for any filled prescription of AUD pharmacotherapy; Acamprosate, Disulfiram, Naltrexone or Nalmefene during 12 months after AUD diagnosis. RESULTS: During the study period, the proportion of individuals who received pharmacotherapy ranged between 22.80 and 23.94 % (χ2(64) = 72.00, p = .23). Female sex, age 31-45, higher education and income, living in a big city, co-habiting and born in Sweden, bar Norway, Denmark and Iceland, were associated with higher odds of pharmacotherapy. Concurrent somatic diagnosis was associated with lower odds of pharmacotherapy but psychiatric diagnosis higher (aOR = 0.61 95 % CI 0.59-0.63 and aOR = 1.61 95 % CI 1.57-1.66 respectively). CONCLUSIONS: Pharmacotherapy for AUD is underutilized. The proportion of individuals with a prescription did not change between 2007 and 2015. Provision of treatment is unequal across different groups in society, where especially older age, lower income and education, and co-morbid somatic diagnosis were associated with lower odds of prescription. There is a need to develop treatment provision, particularly for individuals with co-morbid somatic conditions.


Subject(s)
Alcoholism , Acamprosate , Adult , Aged , Alcoholism/drug therapy , Alcoholism/epidemiology , Cohort Studies , Disulfiram , Female , Humans , Middle Aged , Sweden/epidemiology
14.
Toxicol Appl Pharmacol ; 426: 115642, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34242567

ABSTRACT

Disulfiram (DSF), a sulfur-containing compound, has been used to treat chronic alcoholism and cancer for decades by inactivating aldehyde dehydrogenase (ALDH). Hydrogen sulfide (H2S) is a new gasotransmitter and regulates various cellular functions by S-sulfhydrating cysteine in the target proteins. H2S exhibits similar properties to DSF in the sensitization of cancer cells. The interaction of DSF and H2S on ALDH activity and liver cancer cell survival are not clear. Here it was demonstrated that DSF facilitated H2S release from thiol-containing compounds, and DSF and H2S were both capable of regulating ALDH through inhibition of gene expression and enzymatic activity. The supplement of H2S sensitized human liver cancer cells (HepG2) to DSF-inhibited cell viability. The expression of cystathionine gamma-lyase (a major H2S-generating enzyme) was lower but ALDH was higher in mouse liver cancer stem cells (Dt81Hepa1-6) in comparison with their parental cells (Hepa1-6), and H2S was able to inhibit liver cancer stem cell adhesion. In conclusion, these data point to the potential of combining DSF and H2S for inhibition of cancer cell growth and tumor development by targeting ALDH.


Subject(s)
Acetaldehyde Dehydrogenase Inhibitors/pharmacology , Alcohol Deterrents/pharmacology , Aldehyde Dehydrogenase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Disulfiram/pharmacology , Hydrogen Sulfide/metabolism , Liver Neoplasms/drug therapy , Aldehyde Dehydrogenase/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Copper/pharmacology , Humans , Hydrogen-Ion Concentration , Liver/drug effects , Liver/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Temperature
15.
J Orthop Surg Res ; 16(1): 132, 2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33579316

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is a kind of systemic musculoskeletal disorder and a most important factor for causing disability and physical painfulness. Nevertheless, due to the fact that OA can be triggered by multiple etiological factors, this disease is hard to be cured. Therefore, it is of great necessity for us to find novel targets or drugs for OA treatment. MATERIALS AND METHODS: The chondrocytes were treated with lipopolysaccharide (LPS) and adenosine triphosphate (ATP) to induce pyroptosis in OA. The cell proliferation was detected by Cell Counting Kit-8 assay (CCK-8 assay). Enzyme-linked immunosorbent assay (ELISA) was used for the detection of pyroptosis-related inflammatory factors. Then, the antagonists for gasdermin D (GSDMD) (disulfiram) and high mobility group box 1 (HMGB1) (glycyrrhizic acid) were used to treat the cell model to observe the effects of disulfiram and glycyrrhizic acid on the proliferation of chondrocytes in OA. The protein levels of pyroptosis-related inflammatory factors were measured by western blot, and the levels of aldehyde dehydrogenase (ALDH) and reactive oxygen species (ROS) were measured by corresponding commercial kits. RESULTS: After chondrocytes were induced by LPS and ATP, the cell proliferation was decreased and the expressions of pyroptosis-related inflammatory factors were increased. Disulfiram and glycyrrhizic acid treatment led to enhanced cell proliferation and increased expressions of pyroptosis-related inflammatory factors, while disulfiram showed better alleviative effects on the inflammation in chondrocytes in OA. However, co-treatment with disulfiram at a high concentration and glycyrrhizic acid did not result in higher proliferation of chondrocytes and alleviated inflammation, but led to oxidative stress. CONCLUSION: In conclusion, co-treatment with disulfiram and glycyrrhizic acid at a standard concentration suppresses the inflammatory response of chondrocytes, which may provide guidance for the use of the drugs in the treatment of OA.


Subject(s)
Chondrocytes/drug effects , Chondrocytes/pathology , Disulfiram/pharmacology , Glycyrrhizic Acid/pharmacology , Inflammation/drug therapy , Inflammation/genetics , Osteoarthritis/pathology , Cell Proliferation/drug effects , Cells, Cultured , Chondrocytes/physiology , Disulfiram/therapeutic use , Dose-Response Relationship, Drug , Glycyrrhizic Acid/therapeutic use , Humans , Osteoarthritis/drug therapy , Phytotherapy , Pyroptosis/drug effects
16.
Nanomedicine ; 32: 102340, 2021 02.
Article in English | MEDLINE | ID: mdl-33227540

ABSTRACT

Copper(II) diethyldithiocarbamate complex (CuET), the metabolite of disulfiram complexed with copper, is the component responsible for cancer treatment efficacy of disulfiram. But the hydrophobic property of CuET limits its use in vivo, and an appropriate drug delivery system needs to be developed. Ultrasmall melanin nanoparticle (M-Dot) with excellent biosafety and biocompatibility properties has been synthesized in our previous studies. Herein we prepared CuET loaded with M-Dots through hydrophobic interaction, which could enhance the water solubility significantly. After the administration of M-Dots-CuET in mice tumor models, the nanoparticles showed good tumor accumulation as evidenced by the enhanced photoacoustic signal in tumor regions. M-Dots-CuET also displayed excellent tumor inhibition capability, and the tumor growth inhibition value (TGI) was 45.1%. When combined with photothermal therapy, the TGI reached up to 78.6%. In summary, M-Dots-CuET provide a new potential strategy for cancer theranostics.


Subject(s)
Copper/pharmacology , Disulfiram/pharmacology , Melanins/pharmacology , Neoplasms/therapy , Theranostic Nanomedicine , Animals , Cell Line, Tumor , Ditiocarb/chemistry , Female , Hyperthermia, Induced , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Photoacoustic Techniques , Phototherapy
17.
Theranostics ; 10(14): 6384-6398, 2020.
Article in English | MEDLINE | ID: mdl-32483459

ABSTRACT

To circumvent the huge cost, long R&D time and the difficulty to identify the targets of new drugs, repurposing the ones that have been clinically approved has been considered as a viable strategy to treat different diseases. In the current study, we outlined the rationale for repurposing disulfiram (DSF, an old alcohol-aversion drug) to treat primary breast cancer and its metastases. Methods: To overcome a few shortcomings of the individual administration of DSF, such as the dependence on copper ions (Cu2+) and limited capability in selective targeting, we here artificially synthesized the active form of DSF, diethyldithiocarbamate (DTC)-Cu complex (CuET) for cancer therapeutics. To achieve a greater efficacy in vivo, smart nanomedicines were devised through a one-step self-assembly of three functional components including a chemically stable and biocompatible phase-change material (PCM), the robust anticancer drug (CuET) and a near-infrared (NIR) dye (DIR), namely CuET/DIR NPs. A number of in vitro assays were performed including the photothermal efficacy, light-triggered drug release behavior, nuclear localization, DNA damage and induction of apoptosis of CuET/DIR NPs and molecular mechanisms underlying CuET-induced repression on cancer metastatic behaviors. Meanwhile, the mice bearing 4T1-LG12-drived orthotopic tumors were employed to evaluate in vivo biodistribution and anti-tumor effect of CuET/DIR NPs. The intravenous injection model was employed to reflect the changes of the intrinsic metastatic propensity of 4T1-LG12 cells responding to CuET/DIR NPs. Results: The rationally designed nanomedicines have self-traceability for bioimaging, long blood circulation time for enhanced drug accumulation in the tumor site and photo-responsive release of the anticancer drugs. Moreover, our data unearthed that CuET/DIR nanomedicines behave like "Trojan horse" to transport CuET into the cytoplasm, realizing substantial intracellular accumulation. Upon NIR laser irradiation, massive CuET would be triggered to release from the nanomedicines and reach a high local concentration towards the nucleus, where the pro-apoptotic effects were conducted. Importantly, our CuET/DIR nanomedicines revealed a pronounced capability to leash breast cancer metastases through inhibition on EMT. Additionally, these nanomedicines showed great biocompatibility in animals. Conclusion: These combined data unearthed a remarkably enhanced tumor-killing efficacy of our CuET nanomedicines through nuclear targeting. This work may open a new research area of repurposing DSF as innovative therapeutic agents to treat breast cancer and its metastases.


Subject(s)
Antineoplastic Agents/pharmacology , Copper , Disulfiram , Ditiocarb , Nanoparticles , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Cell Line, Tumor/drug effects , Cell Nucleus , Copper/chemistry , Copper/pharmacology , Disulfiram/chemistry , Disulfiram/pharmacology , Ditiocarb/chemistry , Ditiocarb/pharmacology , Drug Delivery Systems , Drug Liberation , Drug Repositioning , Female , Humans , Low-Level Light Therapy , Mice , Nanomedicine , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasm Metastasis/drug therapy , Neoplasms/drug therapy , Theranostic Nanomedicine/methods
18.
Nat Immunol ; 21(7): 736-745, 2020 07.
Article in English | MEDLINE | ID: mdl-32367036

ABSTRACT

Cytosolic sensing of pathogens and damage by myeloid and barrier epithelial cells assembles large complexes called inflammasomes, which activate inflammatory caspases to process cytokines (IL-1ß) and gasdermin D (GSDMD). Cleaved GSDMD forms membrane pores, leading to cytokine release and inflammatory cell death (pyroptosis). Inhibiting GSDMD is an attractive strategy to curb inflammation. Here we identify disulfiram, a drug for treating alcohol addiction, as an inhibitor of pore formation by GSDMD but not other members of the GSDM family. Disulfiram blocks pyroptosis and cytokine release in cells and lipopolysaccharide-induced septic death in mice. At nanomolar concentration, disulfiram covalently modifies human/mouse Cys191/Cys192 in GSDMD to block pore formation. Disulfiram still allows IL-1ß and GSDMD processing, but abrogates pore formation, thereby preventing IL-1ß release and pyroptosis. The role of disulfiram in inhibiting GSDMD provides new therapeutic indications for repurposing this safe drug to counteract inflammation, which contributes to many human diseases.


Subject(s)
Disulfiram/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Phosphate-Binding Proteins/antagonists & inhibitors , Pyroptosis/drug effects , Sepsis/drug therapy , Animals , Caspase 1/genetics , Caspase 1/metabolism , Caspase Inhibitors/pharmacology , Caspases/metabolism , Caspases, Initiator/genetics , Caspases, Initiator/metabolism , Cell Line, Tumor , Disulfiram/therapeutic use , Drug Evaluation, Preclinical , Drug Repositioning , Female , HEK293 Cells , High-Throughput Screening Assays , Humans , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Liposomes , Mice , Mutagenesis, Site-Directed , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Pyroptosis/immunology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sepsis/immunology , Sf9 Cells , Spodoptera
19.
Nanoscale ; 12(15): 8139-8146, 2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32236229

ABSTRACT

Recent studies have suggested that the anticancer activity of disulfiram (DSF, an FDA-approved alcohol-abuse drug) is Cu-dependent. Low system toxicity and explicit pharmacokinetic characteristics of DSF necessitate safe and effective Cu supplementation in local lesion for further applications. Herein, we presented a new conceptual 'nanosized coordination transport' strategy of Cu(ii) that was realized in porphyrin-based metal-organic frameworks, Sm-TCPP, with strong binding ability to Cu(ii) due to their coordination interactions. Sm-TCPP(Cu) was coated by hyaluronic acid (HA) that termed by Sm-TCPP(Cu)@HA, acting as 'beneficial horse' to target the tumor-localized HA receptor (CD44), thus liberating Cu(ii) ions in cellular overexpressed reductants. The CD44-mediated Cu(ii) accumulation efficiency of Sm-TCPP(Cu)@HA was benchmarked in vitro and vivo against the free TCPP (Cu) via ICP-MS analysis. More importantly, the sensitization effects of Sm-TCPP(Cu)@HA on the anticancer activity of DSF were demonstrated in vivo and in vitro. This study offered a new class of targeted Cu supplements to sensitize DSF for the effective treatment of cancer and established a versatile methodology for constructing a safe and specific delivery of metal ions within living organisms.


Subject(s)
Copper/administration & dosage , Disulfiram/administration & dosage , Drug Delivery Systems , Hyaluronan Receptors/metabolism , Nanostructures/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Animals , Cell Line, Tumor , Copper/chemistry , Drug Carriers , Female , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Mice , Nanostructures/chemistry , Porphyrins/chemistry , Samarium/chemistry , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
20.
Free Radic Biol Med ; 150: 1-11, 2020 04.
Article in English | MEDLINE | ID: mdl-32032663

ABSTRACT

Therapies for lung cancer patients initially elicit desirable responses, but the presence of hypoxia and drug resistant cells within tumors ultimately lead to treatment failure. Disulfiram (DSF) is an FDA approved, copper chelating agent that can target oxidative metabolic frailties in cancer vs. normal cells and be repurposed as an adjuvant to cancer therapy. Clonogenic survival assays showed that DSF (50-150 nM) combined with physiological levels of Cu (15 µM CuSO4) was selectively toxic to H292 NSCLC cells vs. normal human bronchial epithelial cells (HBEC). Furthermore, cancer cell toxicity was exacerbated at 1% O2, relative to 4 or 21% O2. This selective toxicity of DSF/Cu was associated with differential Cu ionophore capabilities. DSF/Cu treatment caused a >20-fold increase in cellular Cu in NSCLCs, with nearly two-fold higher Cu present in NSCLCs vs. HBECs and in cancer cells at 1% O2vs. 21% O2. DSF toxicity was shown to be dependent on the retention of Cu as well as oxidative stress mechanisms, including the production of superoxide, peroxide, lipid peroxidation, and mitochondrial damage. DSF was also shown to selectively (relative to HBECs) enhance radiation and chemotherapy-induced NSCLC killing and reduce radiation and chemotherapy resistance in hypoxia. Finally, DSF decreased xenograft tumor growth in vivo when combined with radiation and carboplatin. These results support the hypothesis that DSF could be a promising adjuvant to enhance cancer therapy based on its apparent ability to selectively target fundamental differences in cancer cell oxidative metabolism.


Subject(s)
Disulfiram , Lung Neoplasms , Cell Line, Tumor , Copper , Disulfiram/pharmacology , Humans , Hypoxia , Lung Neoplasms/drug therapy , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL