8,196 results on '"DISULFIRAM"'
Search Results
2. Endpoints for Pharmacotherapy Trials for Alcohol Use Disorder.
- Author
-
Belnap, Malia, McManus, Kaitlin, Grodin, Erica, and Ray, Lara
- Subjects
Humans ,Alcoholism ,Alcohol Deterrents ,Acamprosate ,Disulfiram ,Naltrexone ,Clinical Trials as Topic ,Treatment Outcome ,United States ,Endpoint Determination ,Biomarkers ,Self Report - Abstract
Alcohol use disorder (AUD) is a debilitating disorder, yet currently approved pharmacotherapies to treat AUD are under-utilized. The three medications approved by the US Food and Drug Administration (FDA) for the indication of AUD are disulfiram, acamprosate, and naltrexone. The current landscape of pharmacotherapies for AUD suggests opportunities for improvement. Clinical trials investigating novel pharmacotherapies for AUD traditionally use abstinence-based drinking outcomes or no heavy drinking days as trial endpoints to determine the efficacy of pharmacotherapies. These outcomes are typically measured through patient self-report endorsements of their drinking. Apart from these traditional outcomes, there have been recent developments in novel endpoints for AUD pharmacotherapies. These novel endpoints include utilizing the World Health Organization (WHO) risk drinking level reductions to promote a harm-reduction endpoint rather than an abstinence-based endpoint. Additionally, in contrast to patient self-report measurements, biological markers of alcohol use may serve as objective endpoints in AUD pharmacotherapy trials. Lastly, the National Institute on Alcohol Abuse and Alcoholism (NIAAA) definition of recovery from AUD and patient-oriented outcomes offer new frameworks to consider endpoints associated with more than alcohol consumption itself, such as the provider-patient experiences with novel pharmacotherapies. These recent developments in new endpoints for AUD pharmacotherapies offer promising future opportunities for pharmacotherapy development, so long as validity and reliability measures are demonstrated for the endpoints. A greater breadth of endpoint utilization may better capture the complexity of AUD symptomatology.
- Published
- 2024
3. In vitro activities of Dithiocarbamate Derivatives against Echinococcus multilocularis metacestode vesicles
- Author
-
Kaethner, Marc, Rennar, Georg, Kampfer, Tobias, Hemphill, Andrew, Mader, Patrick, Luque-Gomez, Ana, Schlitzer, Martin, and Lundstrom-Stadelmann, Britta
- Published
- 2023
4. Disulfiram and Chemotherapy in Treating Patients With Refractory Solid Tumors or Metastatic Pancreatic Cancer
- Author
-
National Cancer Institute (NCI)
- Published
- 2024
5. The Effects of Disulfiram (Antabuse®) on Visual Acuity in Patients With Retinal Degeneration
- Author
-
Alex Levin, Chief, Pediatric Ophthalmology and Ocular Genetics
- Published
- 2024
6. Phase II Trial of Disulfiram With Copper in Metastatic Breast Cancer (DISC)
- Author
-
University Hospital Olomouc
- Published
- 2024
7. Disulfiram With Copper Gluconate and Liposomal Doxorubicin in Treatment-Refractory Sarcomas
- Published
- 2024
8. Combined targeting of GPX4 and BCR-ABL tyrosine kinase selectively compromises BCR-ABL+ leukemia stem cells.
- Author
-
Zeng, Chengwu, Nie, Dingrui, Wang, Xianfeng, Zhong, Shuxin, Zeng, Xiangbo, Liu, Xin, Qiu, Kangjie, Peng, Xueting, Zhang, Wenyi, Chen, Shengting, Zha, Xianfeng, Chen, Cunte, Chen, Zhenhua, Wang, Weizhang, and Li, Yangqiu
- Abstract
Background: In the ongoing battle against BCR-ABL+ leukemia, despite significant advances with tyrosine kinase inhibitors (TKIs), the persistent challenges of drug resistance and the enduring presence of leukemic stem cells (LSCs) remain formidable barriers to achieving a cure. Methods: In this study, we demonstrated that Disulfiram (DSF) induces ferroptosis to synergize with TKIs in inhibiting BCR-ABL+ cells, particularly targeting resistant cells and LSCs, using cell models, mouse models, and primary cells from patients. We elucidated the mechanism by which DSF promotes GPX4 degradation to induce ferroptosis through immunofluorescence, co-immunoprecipitation (CO-IP), RNA sequencing, lipid peroxidation assays, and rescue experiments. Results: Here, we present compelling evidence elucidating the sensitivity of DSF, an USA FDA-approved drug for alcohol dependence, towards BCR-ABL+ cells. Our findings underscore DSF's ability to selectively induce a potent cytotoxic effect on BCR-ABL+ cell lines and effectively inhibit primary BCR-ABL+ leukemia cells. Crucially, the combined treatment of DSF with TKIs selectively eradicates TKI-insensitive stem cells and resistant cells. Of particular note is DSF's capacity to disrupt GPX4 stability, elevate the labile iron pool, and intensify lipid peroxidation, ultimately leading to ferroptotic cell death. Our investigation shows that BCR-ABL expression induces alterations in cellular iron metabolism and increases GPX4 expression. Additionally, we demonstrate the indispensability of GPX4 for LSC development and the initiation/maintenance of BCR-ABL+ leukemia. Mechanical analysis further elucidates DSF's capacity to overcome resistance by reducing GPX4 levels through the disruption of its binding with HSPA8, thereby promoting STUB1-mediated GPX4 ubiquitination and subsequent proteasomal degradation. Furthermore, the combined treatment of DSF with TKIs effectively targets both BCR-ABL+ blast cells and drug-insensitive LSCs, conferring a significant survival advantage in mouse models. Conclusion: In summary, the dual inhibition of GPX4 and BCR-ABL presents a promising therapeutic strategy to synergistically target blast cells and drug-insensitive LSCs in patients, offering potential avenues for advancing leukemia treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. AuCePt porous hollow cascade nanozymes targeted delivery of disulfiram for alleviating hepatic insulin resistance.
- Author
-
Shen, Huawei, Fu, Yafei, Liu, Feifei, Zhang, Wanliang, Yuan, Yin, Yang, Gangyi, Yang, Mengliu, and Li, Ling
- Abstract
As the pathophysiological basis of type 2 diabetes mellitus (T2DM), insulin resistance (IR) is closely related to oxidative stress (OS) and inflammation, while nanozymes have a good therapeutic effect on inflammation and OS by scavenging reactive oxygen species (ROS). Hence, AuCePt porous hollow cascade nanozymes (AuCePt PHNs) are designed by integrating the dominant enzymatic activities of three metallic materials, which exhibit superior superoxide dismutase/catalase-like activities, and high drug loading capacity. In vitro experiments proved that AuCePt PHNs can ultra-efficiently scavenge endogenous and exogenous ROS. Moreover, AuCePt PHNs modified with lactobionic acid (LA) and loaded with disulfiram (DSF), named as AuCePt PHNs-LA@DSF, can significantly improve glucose uptake and glycogen synthesis in IR hepatocytes by regulating the insulin signaling pathways (IRS-1/AKT) and gluconeogenesis signaling pathways (FOXO-1/PEPCK). Intravenous administration of AuCePt PHNs-LA@DSF not only showed high liver targeting efficiency, but also reduced body weight and blood glucose and improved IR and lipid accumulation in high-fat diet-induced obese mice and diabetic ob/ob mice. This research elucidates the intrinsic activity of AuCePt PHNs for cascade scavenging of ROS, and reveals the potential effect of AuCePt PHNs-LA@DSF in T2DM treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Tumor specific in situ synthesis of therapeutic agent for precision cancer therapy.
- Author
-
Zhou, Zhixin, Zhou, Cheng, Liu, Jia, Yuan, Ye, Yao, Chundong, Liu, Miaodeng, Deng, Lixue, Sun, Jia, Chen, Zuoyu, Wang, Lin, and Wang, Zheng
- Subjects
- *
COPPER , *ALCOHOLISM , *COPPER ions , *IMMUNE checkpoint proteins , *COMPLEX ions - Abstract
Background: Traditional chemotherapeutic agents suffer from a lack of selectivity, poor targeting ability, and drug resistance. Developing tumor-specific therapies is crucial for precisely eliminating tumors while circumventing toxicity to normal tissues. Disulfiram (DSF), an FDA-approved drug for treating alcohol dependence, exhibits antitumor effect by forming complexes with copper ions (Cu(DDC)2). Here, we developed a Cu-doped polydopamine-based nanosystem (DSF@CuPDA-PEGM) to achieve in situ generation of toxic Cu(DDC)2. Results: In cancer cells with elevated H2O2 contents, CuPDA responsively degrades to release Cu ions and DSF, allowing on-site synthesis of Cu(DDC)2 with potent antitumor activity. DSF@CuPDA-PEGM exhibits excellent therapeutic efficacy against both drug-sensitive and drug-resistant cancer cells while minimizing toxicity to noncancerous cells. Moreover, DSF@CuPDA-PEGM promotes the immune response by inducing cancer cell immunogenic death, thereby augmenting anti-PD-1-based immune checkpoint blockade therapy. Conclusion: A tumor-specifically degradable Cu-doped polydopamine-based nanosystem is developed to achieve in situ synthesis of antitumor compounds, providing a promising approach to precisely eliminate tumors and heighten chemo-immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Nucleus‐Spike 3D Hierarchical Superstructures via a Lecithin‐Mediated Biomineralization Approach.
- Author
-
Rui, Jiaxin, Wu, Tingting, Zhang, Zhiwei, Lu, Wei, Shi, Xuzhi, Liu, Ying, Han, Xiaolin, Dang, Meng, Su, Xiaodan, and Teng, Zhaogang
- Subjects
- *
RAMAN scattering , *BIOMINERALIZATION , *LECITHIN , *DISULFIRAM , *MORPHOLOGY - Abstract
3D hierarchical superstructures (3DHSs) are key products of nature's evolution and have raised wide interest. However, the preparation of 3DHSs composed of building blocks with different structures is rarely reported, and regulating their structural parameters is challenging. Herein, a simple lecithin‐mediated biomineralization approach is reported for the first time to prepare gold 3DHSs composed of 0D nucleus and 1D protruding dendritic spikes. It is demonstrated that a hydrophobic complex by coordination of disulfiram (DSF) with a share of chloroauric acid is the key to forming the 3DHSs. Under the lecithin mediation, chloroauric acid is first reduced to form the 0D nucleus, followed by the spike growth through the reduction of the hydrophobic complex. The prepared 3DHSs possess well‐defined morphology with a spike length of ≈95 nm. Notably, the hierarchical spike density is systematically manipulated from 38.9% to 74.3% by controlling DSF concentrations. Moreover, the spike diameter is regulated from 9.2 to 12.9 nm by selecting different lecithin concentrations to tune the biomineralization process. Finite‐difference time‐domain (FDTD) simulations reveal that the spikes form “hot spots”. The dense spike structure endows the 3DHSs with sound performance in surface‐enhanced Raman scattering (SERS) applications. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. The potential of disulfiram as a drug to improve the prognosis after the onset of subarachnoid hemorrhage.
- Author
-
Itani, Masahiko, Okada, Akihiro, Arakawa, Yoshiki, Terashima, Yuya, and Aoki, Tomohiro
- Subjects
- *
INTRACRANIAL aneurysm ruptures , *SUBARACHNOID hemorrhage , *INTRATHECAL injections , *SUBARACHNOID space , *ANIMAL mortality , *INTRACRANIAL pressure - Abstract
• The FROUNT inhibitor, Disulfiram, suppressed inflammatory cell infiltration. • Disulfiram suppressed microglia activation after subarachnoid hemorrhage in animals. • Disulfiram suppressed the death after experimental subarachnoid hemorrhage. • Disulfiram prevented the neuronal death after experimental subarachnoid hemorrhage. • Disulfiram prevented the vasospasm after experimental subarachnoid haemorrhage. Subarachnoid hemorrhage due to rupture of intracranial aneurysms has a poor outcome, making this disease being the social problem. Inflammation evoked by the increase in intracranial pressure and the clot in the subarachnoid space after the onset of SAH exacerbates neuronal death and vasospasm, resulting in the poor outcome and severe aftereffects. Here, FROUNT mediates CCR2 and CCR5 signaling as an intracellular molecule binding to these chemoattractant receptors which facilitate the migration of inflammatory cells, such as macrophages, in situ to trigger inflammation there. Animal model of subarachnoid hemorrhage was established in rats through intrathecal injection of autologous blood. The effect of the FROUNT inhibitor, disulfiram, on survival rate, neuronal death in hippocampus or vasospasm was then examined. The intrathecal administration of disulfiram significantly suppressed the infiltration of CD68-positive macrophages and myeloperoxidase-positive neutrophils toward the clot in the cistern in situ. In this condition, disulfiram ameliorated the death of animals after the onset of subarachnoid hemorrhage in rats. In addition, disulfiram suppressed both the two major events after subarachnoid hemorrhage, the neuronal death in hippocampus and vasospasm. The pharmacological inhibition of CCR2 and CCR5 signaling by disulfiram could thus be the therapeutic strategy to improve the outcome of subarachnoid hemorrhage. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. A fibroblast activation protein α-activatable nanoagent co-delivering diethyldithiocarbamate and copper for tumor therapy and imaging.
- Author
-
Ding, Yaqing, Huang, Zeqian, Luo, Yong, Lin, Huanxin, Wang, Jue, Zeng, Zishan, Zhang, Tao, Chen, Yiwei, Gong, Yujun, Zhang, Mingxia, and Zhao, Chunshun
- Subjects
COPPER ,CANCER chemotherapy ,ANTINEOPLASTIC agents ,DISULFIRAM ,DIETHYLDITHIOCARBAMATE ,CYANINES - Abstract
Disulfiram (DSF), an FDA-approved drug for treating alcoholism, has been verified with Cu
2+ -dependent anticancer activity by forming Cu(DTC) 2 , the complex of one of its metabolites diethyldithiocarbamate (DTC) and Cu2+ . Nevertheless, the antitumor effect is limited by insufficient Cu(DTC) 2 formation in suit and off-target system toxicity. Herein, we developed a fibroblast activation protein α (FAPα) activatable nanoagent (HfD-HID-Cu) for co-delivery of DTC polymeric prodrug and exogenous Cu2+ to achieve enhanced cancer-specific therapy and activatable in situ fluorescence imaging meanwhile. HfD-HID-Cu was simply constructed through the co-assembly of the DTC polymeric prodrug (HA-fap-DTC) and the copper-loaded IR808-conjugated polymer (HA-IR-DPA-Cu), which could serve as the "OFF-to-ON" switch for chemotherapy and fluorescence. With the high expression of FAPα in tumor tissues, HA-fap-DTC could be activated specifically to release DTC, while maintaining inactive in normal tissues. The liberated DTC within tumor tissues could contend for Cu2+ from HA-IR-DPA-Cu, resulting in the formation of highly cytotoxic Cu(DTC) 2 in situ for chemotherapy, concomitant with the fluorescence recovery of cyanine dye for tumor imaging. This work provides an effective strategy for co-delivery of DTC prodrug and Cu2+ for tumor theranostic with improved selectivity and minimal side effects. DSF-based antitumor therapy is highly dependent on Cu2+ . However, the non-synchronous distribution of DSF/DTC and Cu2+ in tumor tissues attenuates the antitumor efficacy. The insufficient Cu(DTC) 2 formation in suit and off-target distribution greatly limit the anti-tumor application. This study provides a nanoagent for co-delivery of DTC polymeric prodrug and Cu2+ by simple co-assembly to achieve their synchronous tumor distribution. It can be selectively activated by FAPα, forming cytotoxic Cu(DTC) 2 in suit for tumor-specific chemotherapy and reducing the systemic toxicity. In addition to chemotherapy, the nanoagent can emit fluorescence under the sequential triggering of FAPα and released DTC for tumor imaging. Overall, this study renders a promising strategy for improved Cu(DTC) 2 -based antitumor therapy and imaging. [Display omitted] [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
14. Antibiotics-free compounds for managing carbapenem-resistant bacteria; a narrative review.
- Author
-
Shariati, Aref, Kashi, Milad, Chegini, Zahra, and Hosseini, Seyed Mostafa
- Subjects
CARBAPENEM-resistant bacteria ,PENICILLIN-binding proteins ,ANTIMICROBIAL peptides ,GRAM-negative bacteria ,DISULFIRAM - Abstract
Carbapenem-resistant (CR) Gram-negative bacteria have become a significant public health problem in the last decade. In recent years, the prevalence of CR bacteria has increased. The resistance to carbapenems could result from different mechanisms such as loss of porin, penicillin-binding protein alteration, carbapenemase, efflux pump, and biofilm community. Additionally, genetic variations like insertion, deletion, mutation, and post-transcriptional modification of corresponding coding genes could decrease the susceptibility of bacteria to carbapenems. In this regard, scientists are looking for new approaches to inhibit CR bacteria. Using bacteriophages, natural products, nanoparticles, disulfiram, N-acetylcysteine, and antimicrobial peptides showed promising inhibitory effects against CR bacteria. Additionally, the mentioned compounds could destroy the biofilm community of CR bacteria. Using them in combination with conventional antibiotics increases the efficacy of antibiotics, decreases their dosage and toxicity, and resensitizes CR bacteria to antibiotics. Therefore, in the present review article, we have discussed different aspects of non-antibiotic approaches for managing and inhibiting the CR bacteria and various methods and procedures used as an alternative for carbapenems against these bacteria. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Repurposing FDA-approved disulfiram for targeted inhibition of diphtheria toxin and the binary protein toxins of Clostridium botulinum and Bacillus anthracis.
- Author
-
Borho, Joscha, Kögel, Merle, Eckert, Amelie, and Barth, Holger
- Subjects
BOTULINUM toxin ,BOTULINUM A toxins ,DIPHTHERIA toxin ,PATHOGENIC bacteria ,BACILLUS anthracis ,TOXINS ,BACTERIAL toxins - Abstract
Many bacteria act pathogenic by the release of AB-type protein toxins that efficiently enter human or animal cells and act as enzymes in their cytosol. This leads to disturbed cell functions and the clinical symptoms characteristic for the individual toxin. Therefore, molecules that directly target and neutralize these toxins provide promising novel therapeutic options. Here, we found that the FDA-approved drug disulfiram (DSF), used for decades to treat alcohol abuse, protects cells from intoxication with diphtheria toxin (DT) from Corynebacterium diphtheria, the causative agent of diphtheria, lethal toxin (LT) from Bacillus anthracis, which contributes to anthrax, and C2 enterotoxin from Clostridium botulinum when applied in concentrations lower than those found in plasma of patients receiving standard DSF treatment for alcoholism (up to 20 µM). Moreover, this inhibitory effect is increased by copper, a known enhancer of DSF activity. LT and C2 are binary toxins, consisting of two non-linked proteins, an enzyme (A) and a separate binding/transport (B) subunit. To act cytotoxic, their proteolytically activated B subunits PA63 and C2IIa, respectively, form barrelshaped heptamers that bind to their cellular receptors and form complexes with their respective A subunits LF and C2I. The toxin complexes are internalized via receptor-mediated endocytosis and in acidified endosomes, PA63 and C2IIa form pores in endosomal membranes, which facilitate translocation of LF and C2I into the cytosol, where they act cytotoxic. In DT, A and B subunits are located within one protein, but DT also forms pores in endosomes that facilitate translocation of the A subunit. If cell binding, membrane translocation, or substrate modification is inhibited, cells are protected from intoxication. Our results implicate that DSF neither affects cellular binding nor the catalytic activity of the investigated toxins to a relevant extend, but interferes with the toxin pore-mediated translocation of the A subunits of DT, LT and C2 toxin, as demonstrated by membranetranslocation assays and toxin pore conductivity experiments in the presence or absence of DSF. Since toxin translocation across intracellular membranes represents a central step during cellular uptake of many bacterial toxins, DSF might neutralize a broad spectrum of medically relevant toxins. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. RETRACTED: Disulfiram Chelated With Copper Inhibits the Growth of Gastric Cancer Cells by Modulating Stress Response and Wnt/β-catenin Signaling.
- Author
-
Ling Wang, Xiaoke Chai, Run Wan, Hong Zhang, Cong Zhou, Lin Xiang, Paul, Maswikiti Ewetse, and Yumin Li
- Subjects
DNA repair ,COPPER ,REACTIVE oxygen species ,WNT proteins ,OXIDATIVE phosphorylation - Abstract
Disulfiram (DSF) is a well-known drug for alcohol abuse. In recent decades, DSF has been demonstrated to exhibit anti-tumor activity; DSF chelated with copper shows enhanced anti-tumor effect. Our goal was to explore the effect of DSF/Cu complex on the growth and metastasis of gastric cancer (GC) in vitro and in vivo. DSF/Cu complex suppressed the proliferation, migration of MKN-45 and BGC-823 GC cells. Furthermore, DSF/Cu treatment reduced the tumor volume in GC mouse models with a tumor suppression rate of 48.24%. Additionally, DSF/Cu induced apoptosis in vitro in MKN-45 and BGC-823 GC cells in a dose- and time-dependent manner as well as in vivo in the xenograft tumor mouse model. Furthermore, DSF/Cu induced autophagy and autophagic flux in MKN-45 and BGC-823 cells, increased the expression of autophagy-related Beclin-1 and LC3 proteins in vivo. Additionally, DSF/Cu suppressed aerobic glycolysis and oxidative phosphorylation by reducing oxygen consumption rate and extracellular acidification rate, respectively, in MKN-45 and BGC-823 cells. Treatment with DSF/Cu induced oxidative stress and DNA damage response by elevating the reactive oxygen species levels; increasing the expression of P53, P21, and g-H2AX proteins; and inhibiting Wnt/bcatenin signaling in vitro and in vivo. Thus, DSF/Cu suppressed the growth and metastasis of GC cells via modulating the stress response and Wnt/b-catenin signaling. Hence, DSF may be used as a potential therapeutic agent for the treatment of GC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Common anesthetic used in preclinical PET imaging inhibits metabolism of the PET tracer [18F]3F4AP.
- Author
-
Ramos‐Torres, Karla, Sun, Yang, Takahashi, Kazue, Zhou, Yu‐Peng, and Brugarolas, Pedro
- Subjects
- *
CYTOCHROME P-450 CYP2E1 , *POSITRON emission tomography , *DISULFIRAM , *DIAGNOSTIC imaging ,BRAIN metabolism - Abstract
Positron emission tomography (PET) imaging studies in laboratory animals are almost always performed under isoflurane anesthesia to ensure that the subject stays still during the image acquisition. Isoflurane is effective, safe, and easy to use, and it is generally assumed to not have an impact on the imaging results. Motivated by marked differences observed in the brain uptake and metabolism of the PET tracer 3‐[18F]fluoro‐4‐aminopyridine [(18F]3F4AP) between human and nonhuman primate studies, this study investigates the possible effect of isoflurane on this process. Mice received [18F]3F4AP injection while awake or under anesthesia and the tracer brain uptake and metabolism was compared between groups. A separate group of mice received the known cytochrome P450 2E1 inhibitor disulfiram prior to tracer administration. Isoflurane was found to largely abolish tracer metabolism in mice (74.8 ± 1.6 vs. 17.7 ± 1.7% plasma parent fraction, % PF) resulting in a 4.0‐fold higher brain uptake in anesthetized mice at 35 min post‐radiotracer administration. Similar to anesthetized mice, animals that received disulfiram showed reduced metabolism (50.0 ± 6.9% PF) and a 2.2‐fold higher brain signal than control mice. The higher brain uptake and lower metabolism of [18F]3F4AP observed in anesthetized mice compared to awake mice are attributed to isoflurane's interference in the CYP2E1‐mediated breakdown of the tracer, which was confirmed by reproducing the effect upon treatment with the known CYP2E1 inhibitor disulfiram. These findings underscore the critical need to examine the effect of isoflurane in PET imaging studies before translating tracers to humans that will be scanned without anesthesia. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Synthesis and Catalytic Activity of Novel Complexes Based on Cyano-Substituted Phthalocyanines as Promising Drug Conversion Agents.
- Author
-
Erzunov, Dmitry, Tonkova, Svetlana, Sarvin, Ilya, and Vashurin, Arthur
- Subjects
- *
CATALYTIC activity , *ALCOHOLISM , *CATALYST structure , *ZINC compounds , *DISULFIRAM - Abstract
This paper presents the results of obtaining new cobalt and zinc complexes based on dicyanophenoxy-substituted carboxypthalocyanine. The original method of synthesis and isolation of the compound is shown; its spectroscopic and photophysical characteristics are studied. Studies show the absence of aggregation processes in organic media for solutions of complexes in working concentration ranges. This shows the possibility of the practical application of structures as catalysts. The high catalytic activity of cobalt complexes with dicyanophenoxy-substituted carboxyphthalocyanine ligand in the conversion reaction of sodium diethyldithiocarbamate to disulfiram, which is an active component of drugs for the treatment of alcohol dependence, is determined. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. GSH exhaustion via inhibition of xCT-GSH-GPX4 pathway synergistically enhanced DSF/Cu-induced cuproptosis in myelodysplastic syndromes.
- Author
-
Li, Huanjuan, Li, Yanchun, Yu, Yanhua, Ren, Xueying, Yang, Chen, Jin, Weidong, Li, Keyi, Zhou, Yi, Wu, Cuiyun, Shen, Yuhuan, Hu, Wanye, Liu, Yingchao, Yu, Lingyan, Tong, Xiangmin, Du, Jing, and Wang, Ying
- Subjects
- *
APOPTOSIS , *COPPER , *ACUTE myeloid leukemia , *MYELODYSPLASTIC syndromes , *OLDER people - Abstract
The clinical application of the therapeutic approach in myelodysplastic syndromes (MDS) remains an insurmountable challenge for the high propensity for progressing to acute myeloid leukemia and predominantly affecting elderly individuals. Thus, the discovery of molecular mechanisms underlying the regulatory network of different programmed cell death holds great promise for the identification of therapeutic targets and provides insights into new therapeutic avenues. Herein, we found that disulfiram/copper (DSF/Cu) significantly repressed the cell viability, increased reactive oxygen species (ROS) accumulation, destroyed mitochondrial morphology, and altered oxygen consumption rate. Further studies verified that DSF/Cu induces cuproptosis, as evidenced by the depletion of glutathione (GSH), aggregation of lipoylated DLAT, and induced loss of Fe–S cluster–containing proteins, which could be rescued by tetrathiomolybdate and knockdown of ferredoxin 1 (FDX1). Additionally, GSH contributed to the tolerance of DSF/Cu-mediated cuproptosis, while pharmacological chelation of GSH triggered ROS accumulation and sensitized cell death. The xCT-GSH-GPX4 axis is the ideal downstream component of ferroptosis that exerts a powerful protective mechanism. Notably, classical xCT inhibitors were capable of leading to the catastrophic accumulation of ROS and exerting synergistic cell death, while xCT overexpression restored these phenomena. Simvastatin, an inhibitor of HMG-CoA (3-hydroxy-3-methylglutaryl-coenzyme A) reductase, has beneficial effects in repurposing for inhibiting GPX4. Similarly, the combination treatment of DSF/Cu and simvastatin dramatically decreased the expression of GPX4 and Fe–S proteins, ultimately accelerating cell death. Moreover, we identified that the combination treatment of DSF/Cu and simvastatin also had a synergistic antitumor effect in the MDS mouse model, with the reduced GPX4, increased COX-2 and accumulated lipid peroxides. Overall, our study provided insight into developing a novel synergistic strategy to sensitize MDS therapy by targeting ferroptosis and cuproptosis. [Display omitted] • DSF/Cu causes the accumulation of ROS accompanied by the exhausting of GSH in the MDS cell lines. • GSH contributed to the tolerance of DSF/Cu-induced cuproptosis. • Targeting the xCT-GSH-GPX4 pathway can enhance the effectiveness of DSF/Cu-induced cell death in MDS cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. GSDMD-Dependent Neutrophil Extracellular Traps Mediate Portal Vein Thrombosis and Associated Fibrosis in Cirrhosis.
- Author
-
Che, Ying, Chien, Youjung, Zhu, Yuli, Huang, Xiaoquan, Wu, Ling, Ai, Yingjie, Jiang, Siyu, Li, Feng, and Chen, Shiyao
- Subjects
- *
PORTAL vein , *CIRRHOSIS of the liver , *NEUTROPHILS , *DISULFIRAM , *THIOACETAMIDE - Abstract
Portal vein thrombosis (PVT) is a challenging and controversial complication of cirrhosis. Experimental models that reproduce cirrhotic PVT and effective pharmacological therapies are limited. We aimed to investigate the nature course and mechanisms of PVT in cirrhosis. A novel PVT model was developed via two-step total portal vein ligation in healthy and thioacetamide (TAA)-cirrhotic rats. Circulating and liver-infiltrating neutrophils were isolated from individuals with cirrhosis to examine neutrophil extracellular traps (NETs) and explore their unique characteristics and implications in PVT-associated fibrosis in cirrhosis. We further validated macrophage–myofibroblast transition (MMT) via multiplex immunofluorescence and single-cell sequencing. In the experimental model, cirrhosis promoted PVT development and portal vein intimal thickening. Interestingly, cirrhosis promoted spontaneous resolution of PVT due to instability of thrombus structure, along with pulmonary and intrahepatic clots. NETs-MMT mediate cirrhotic PVT and PVT-associated fibrosis, including fibrotic thrombus remodeling and increased hepatic collagen deposition. Mechanistically, caspase-4-dependent activation of neutrophils and GSDMD mediated the formation of NETs. The extracellular DNA of NETs promoted TGF-β1/Smad3-driven MMT. Inhibiting GSDMD with disulfiram suppressed cirrhotic PVT and prevented associated fibrosis. The cirrhotic PVT model reflected the following three main characteristics of cirrhotic PVT: spontaneous resolution, immunothrombosis, and intimal fibrosis. Targeting NETs with GSDMD inhibitors may serve as a new therapeutic concept to treat cirrhotic PVT. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. 双硫仑联合二价铜离子诱导铜死亡抑制肝癌细胞Hep3B增殖、迁移 和侵袭.
- Author
-
唐敏, 黄娟婵, 韦杏雯, 罗雪文, and 赵伟
- Abstract
Objective To investigate the effects of disulfiram (DSF) combined with Cu2+ on the proliferation, migration and invasion of hepatoma Hep3B cells and the underlying mechanism. Methods Hepatoma Hep3B cells were cultured in vitro. DSF (30 nmol/L) solution, Cu2+ (1 μmol/L) solution and copper chelating agent ammonium tetrathiomolybdate Ⅵ (ATTM) (30 nmol/L) solution were used individually or combinedly for intervention. The cells treated with dimethyl sulfoxide (DMSO) (30 nmol/ L) were used as the control group. CCK-8, scratch test and Transwell cell test were used to detect the proliferation, migration and invasion ability of cells. The expression of copper death related proteins dihydrolipoamide S-acetyltransferase (DLAT) and ferredoxin 1 (FDX1) were detected by immunofluorescence assay. Results Compared with the control group, the proliferation, migration and invasion ability of Hep3B cells were significantly decreased after DSF or Cu2+ or DSF+Cu2+ combined intervention (all P<0.05), and the decrease was more significant in DSF+Cu2+ combined intervention (all P<0.0001). After addition of DSF combined with Cu2+ to ATTM, the inhibitory effects of DSF combined with Cu2+ on the proliferation, migration and invasion of Hep3B cells were reversed, and the proliferation, migration and invasion ability cells were enhanced compared with DSF+Cu2+ group (all P<0.05). Compared with the control group, the fluorescence intensity of copper death related proteins DLAT and FDX1 showed no significant increase after DSF or Cu2+ intervention, but the fluorescence intensity of DLAT protein increased while that of FDX1 protein decreased after DSF +Cu2+ combined intervention, and the expression trend of DLAT and FDX1 proteins was reversed after the addition of ATTM. Conclusions DSF combined with Cu2+ can inhibit the proliferation, migration and invasion of Hep3B cells, probably through the induction of cuproptosis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Inhalable nanoparticles with enhanced cuproptosis and cGAS–STING activation for synergistic lung metastasis immunotherapy.
- Author
-
Yan, Chongzheng, Lv, Huaiyou, Feng, Yafei, Li, Yuhan, and Zhao, Zhongxi
- Subjects
INHALATION administration ,LUNG diseases ,INTRAVENOUS injections ,DENDRITIC cells ,NATURAL immunity - Abstract
Due to the insufficient Cu
+ accumulation, Cu+ efflux mechanism, and highly immunosuppressive tumor microenvironment (TME) in lung metastasis, the cuproptosis efficacy is limited. Herein, an inhalable nanodevice (CLDCu) is constructed to successfully overcome the drawbacks of cuproptosis. CLDCu consists of a Cu2+ -chitosan shell and low molecular weight heparin-tocopherol succinate (LMWH-TOS, LT) core with disulfiram (DSF) loading. The prepared CLDCu can be inhaled and accumulate in large amounts in lung lesions (63.6%) with 56.5 times higher than intravenous injection. Within tumor cells, the mild acidity triggers the co-release of DSF and Cu2+ , thus generating bis(diethyldithiocarbamate)-copper (CuET) to block Cu+ efflux protein ATP7B and forming toxic Cu+ , leading to enhanced cuproptosis. Meanwhile, the released chitosan cooperates with CLDCu-induced cuproptosis to activate stimulator of interferon genes (STING) pathway, which significantly potentiates dendritic cells (DCs) maturation, as wells as evokes innate and adaptive immunity. In lung metastatic mice model, CLDCu is found to induce cuproptosis and reverse the immunosuppressive TME by inhalation administration. Moreover, CLDCu combined with anti-programmed cell death protein ligand-1 antibody (aPD-L1) provokes stronger antitumor immunity. Therefore, nanomedicine that combines cuproptosis with STING activation is a novel strategy for tumor immunotherapy. An inhalable nanoformulation (CLDCu) co-delivering disulfiram and Cu2+ was designed to enhance lung metastasis immunotherapy via cuproptosis and cGAS–STING activation. Moreover, CLDCu collaborated with aPD-L1 provoked more powerful antitumor immunity. [Display omitted] [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
23. Carrier‐Free Disulfiram Based Nanomedicine for Enhanced Cancer Therapy.
- Author
-
Dang, Meng, Lu, Nan, Shi, Xuzhi, Li, Qiang, Lin, Bin, Dong, Heng, Han, Xiaolin, Rui, Jiaxin, Sun, Junfen, Luo, Wei, Teng, Zhaogang, and Su, Xiaodan
- Subjects
REACTIVE oxygen species ,DISULFIRAM ,NANORODS ,CYTOTOXINS ,ANTINEOPLASTIC agents - Abstract
Numerous nanomedicines have been developed to improve the efficiency and safety of conventional anticancer drugs. However, the carrier materials and intricate nature of multifunctional design always hindered the clinical transformation of nanomedicines. Herein, a novel carrier‐free anticancer nanomedicine (CFDC) with tailored morphologies including nanodots, nanorod and nanosheet were prepared using the clinically approved anti‐alcoholism drug disulfiram (DSF) via supramolecular assembly process. Our study reveals that CFDC induces the production of reactive oxygen species and activates the downstream apoptosis‐related c‐Jun N‐terminal kinase (JNK) and p‐38 pathway. In addition, the CFDC effectively counteract the inhibitory effect of NF‐κB expression on ROS‐induced cellular cytotoxicity, ultimately resulting in enhanced cell apoptosis, which is not achievable by pure DSF and the simply mixing of DSF and Cu2+ (DSF+Cu). Notably, the CFDC exhibits 3.1‐, 3.0‐folds increased on cancer cell DNA damage compared with the DSF, and DSF+Cu groups. In vivo experiments conducted on breast‐ or prostate‐bearing mice modals demonstrated that the CFDC exhibits a higher efficacy in suppressing the tumor growth. The remarkable drug delivery efficiency and better anticancer effect of CFDC nanodrug provide promising prospects for the clinical transformation of DSF based nanodrug in cancer therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. AuCePt porous hollow cascade nanozymes targeted delivery of disulfiram for alleviating hepatic insulin resistance
- Author
-
Huawei Shen, Yafei Fu, Feifei Liu, Wanliang Zhang, Yin Yuan, Gangyi Yang, Mengliu Yang, and Ling Li
- Subjects
Insulin resistance ,Oxidative stress ,Nanozymes ,Disulfiram ,Targeted drug delivery ,Biotechnology ,TP248.13-248.65 ,Medical technology ,R855-855.5 - Abstract
Abstract As the pathophysiological basis of type 2 diabetes mellitus (T2DM), insulin resistance (IR) is closely related to oxidative stress (OS) and inflammation, while nanozymes have a good therapeutic effect on inflammation and OS by scavenging reactive oxygen species (ROS). Hence, AuCePt porous hollow cascade nanozymes (AuCePt PHNs) are designed by integrating the dominant enzymatic activities of three metallic materials, which exhibit superior superoxide dismutase/catalase-like activities, and high drug loading capacity. In vitro experiments proved that AuCePt PHNs can ultra-efficiently scavenge endogenous and exogenous ROS. Moreover, AuCePt PHNs modified with lactobionic acid (LA) and loaded with disulfiram (DSF), named as AuCePt PHNs-LA@DSF, can significantly improve glucose uptake and glycogen synthesis in IR hepatocytes by regulating the insulin signaling pathways (IRS-1/AKT) and gluconeogenesis signaling pathways (FOXO-1/PEPCK). Intravenous administration of AuCePt PHNs-LA@DSF not only showed high liver targeting efficiency, but also reduced body weight and blood glucose and improved IR and lipid accumulation in high-fat diet-induced obese mice and diabetic ob/ob mice. This research elucidates the intrinsic activity of AuCePt PHNs for cascade scavenging of ROS, and reveals the potential effect of AuCePt PHNs-LA@DSF in T2DM treatment. Graphical abstract
- Published
- 2024
- Full Text
- View/download PDF
25. Tumor specific in situ synthesis of therapeutic agent for precision cancer therapy
- Author
-
Zhixin Zhou, Cheng Zhou, Jia Liu, Ye Yuan, Chundong Yao, Miaodeng Liu, Lixue Deng, Jia Sun, Zuoyu Chen, Lin Wang, and Zheng Wang
- Subjects
Cancer therapy ,Disulfiram ,Tumor microenvironment ,In situ synthesis of antitumor agent ,Nanomedicine ,Biotechnology ,TP248.13-248.65 ,Medical technology ,R855-855.5 - Abstract
Abstract Background Traditional chemotherapeutic agents suffer from a lack of selectivity, poor targeting ability, and drug resistance. Developing tumor-specific therapies is crucial for precisely eliminating tumors while circumventing toxicity to normal tissues. Disulfiram (DSF), an FDA-approved drug for treating alcohol dependence, exhibits antitumor effect by forming complexes with copper ions (Cu(DDC)2). Here, we developed a Cu-doped polydopamine-based nanosystem (DSF@CuPDA-PEGM) to achieve in situ generation of toxic Cu(DDC)2. Results In cancer cells with elevated H2O2 contents, CuPDA responsively degrades to release Cu ions and DSF, allowing on-site synthesis of Cu(DDC)2 with potent antitumor activity. DSF@CuPDA-PEGM exhibits excellent therapeutic efficacy against both drug-sensitive and drug-resistant cancer cells while minimizing toxicity to noncancerous cells. Moreover, DSF@CuPDA-PEGM promotes the immune response by inducing cancer cell immunogenic death, thereby augmenting anti-PD-1-based immune checkpoint blockade therapy. Conclusion A tumor-specifically degradable Cu-doped polydopamine-based nanosystem is developed to achieve in situ synthesis of antitumor compounds, providing a promising approach to precisely eliminate tumors and heighten chemo-immunotherapy. Graphical Abstract
- Published
- 2024
- Full Text
- View/download PDF
26. Disulfiram attenuates cell and tissue damage and blood‒brain barrier dysfunction after intracranial haemorrhage by inhibiting the classical pyroptosis pathway
- Author
-
Chen Xu, Fangchao Jiang, Yuanfu Mao, Wan Wei, Jihe Song, Feihong Jia, Xinshu Du, Di Zhong, and Guozhong Li
- Subjects
Intracerebral haemorrhage ,Disulfiram ,Pyroptosis ,GSDMD ,Blood‒brain barrier ,Medicine ,Science - Abstract
Abstract No single treatment significantly reduces the mortality rate and improves neurological outcomes after intracerebral haemorrhage (ICH). New evidence suggests that pyroptosis-specific proteins are highly expressed in the perihaematomal tissues of patients with ICH and that the disulfiram (DSF) inhibits pyroptosis. An ICH model was established in C57BL/6 mice by intracranial injection of collagenase, after which DSF was used to treat the mice. Cell model of ICH was constructed, and DSF was used to treat the cells. HE, TUNEL, Nissl, FJC and IF staining were performed to evaluate the morphology of brain tissues; Western blotting and ELISA were performed to measure the protein expression of NOD-like receptor protein 3 (NLRP3)/Caspase-1/gasdermin D (GSDMD) classical pyroptosis pathway and Toll-likereceptor4 (TLR4)/nuclear factor-kappaB (NF-κB) inflammatory signaling pathway and blood‒brain barrier-associated factoes, and the wet/dry weight method was used to determine the brain water content. The expression of proteins related to the NLRP3/Caspase-1/GSDMD pathway and the TLR4/NF-κB pathway was upregulated in tissues surrounding the haematoma compared with that in control tissues; Moreover, the expression of the blood–brain barrier structural proteins occludin and zonula occludens-1 (ZO-1) was downregulated, and the expression of Aquaporin Protein-4 (AQP4) and matrix metalloprotein 9 (MMP-9) was upregulated. DSF significantly inhibited these changes, reduced the haematoma volume, decreased the brain water content, reduced neuronal death and degeneration and improved neurological function after ICH. ICH activated the classical pyroptosis pathway and TLR4/NF-κB inflammatory pathway, disruped the expression of blood–brain barrier structural proteins, and exacerbated brain injury and neurological dysfunction. DSF inhibited these changes and exerted the therapeutic effects on pathological changes and dysfunction caused by ICH.
- Published
- 2024
- Full Text
- View/download PDF
27. Desperate measures of involuntary deaddiction: A case report of acute disulfiram poisoning
- Author
-
Nanma Livingstone and Arya Jith
- Subjects
disulfiram ,direct toxic effects ,adult ,covert use ,Psychiatry ,RC435-571 - Abstract
Disulfiram is an effective deterrent drug for alcohol use in patients who have undergone detoxification and who are motivated to remain abstinent. Disulfiram poisoning is not common; only a few cases have been reported in the literature. Although most patients tolerate disulfiram well, severe toxic side effects have also been reported including hepatitis, encephalopathy, psychosis, optic neuropathy, and peripheral neuropathy. This is a case report of disulfiram toxicity in a 30-year-old man who developed mental status changes, pruritic rashes, and blurred vision after being given one gram of disulfiram per day covertly. He was managed conservatively and the symptoms subsided gradually. This case report highlights the importance of creating awareness about the dangers of covert administration of medications for alcohol use.
- Published
- 2024
- Full Text
- View/download PDF
28. Inhalable nanoparticles with enhanced cuproptosis and cGAS–STING activation for synergistic lung metastasis immunotherapy
- Author
-
Chongzheng Yan, Huaiyou Lv, Yafei Feng, Yuhan Li, and Zhongxi Zhao
- Subjects
Cuproptosis ,cGAS–STING activation ,Disulfiram ,Inhalation administration ,Tumor microenvironment ,PD-L1 checkpoint blockade ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Due to the insufficient Cu+ accumulation, Cu+ efflux mechanism, and highly immunosuppressive tumor microenvironment (TME) in lung metastasis, the cuproptosis efficacy is limited. Herein, an inhalable nanodevice (CLDCu) is constructed to successfully overcome the drawbacks of cuproptosis. CLDCu consists of a Cu2+-chitosan shell and low molecular weight heparin-tocopherol succinate (LMWH-TOS, LT) core with disulfiram (DSF) loading. The prepared CLDCu can be inhaled and accumulate in large amounts in lung lesions (63.6%) with 56.5 times higher than intravenous injection. Within tumor cells, the mild acidity triggers the co-release of DSF and Cu2+, thus generating bis(diethyldithiocarbamate)-copper (CuET) to block Cu+ efflux protein ATP7B and forming toxic Cu+, leading to enhanced cuproptosis. Meanwhile, the released chitosan cooperates with CLDCu-induced cuproptosis to activate stimulator of interferon genes (STING) pathway, which significantly potentiates dendritic cells (DCs) maturation, as wells as evokes innate and adaptive immunity. In lung metastatic mice model, CLDCu is found to induce cuproptosis and reverse the immunosuppressive TME by inhalation administration. Moreover, CLDCu combined with anti-programmed cell death protein ligand-1 antibody (aPD-L1) provokes stronger antitumor immunity. Therefore, nanomedicine that combines cuproptosis with STING activation is a novel strategy for tumor immunotherapy.
- Published
- 2024
- Full Text
- View/download PDF
29. Reporting on Adverse Clinical Events.
- Subjects
ADVERSE health care events ,DISULFIRAM ,SPASMS ,DRUG side effects ,HYPOTHYROIDISM ,FLUMAZENIL - Published
- 2024
- Full Text
- View/download PDF
30. Disulfiram and Chelated Zinc for the Rx of Disseminated Mets Mel That Has Failed First Line Therapy
- Published
- 2023
31. Disulfiram for Treatment of Retinal Degeneration
- Author
-
Debarshi Mustafi, Assistant Professor: Dept of Ophthamology
- Published
- 2023
32. V9302-loaded copper-polyphenol hydrogel for enhancing the anti-tumor effect of disulfiram.
- Author
-
Sun, Zhongquan, Wei, Shenyu, Guo, Quanshi, Ouyang, Hanxiang, Mao, Zhengwei, Wang, Weilin, Tong, Zongrui, and Ding, Yuan
- Subjects
- *
DRUG delivery systems , *REACTIVE oxygen species , *ANIMAL experimentation , *SCHIFF bases , *COPPER - Abstract
A copper-polyphenol hydrogel loaded with glutamine uptake inhibitor (V9302) was designed for a local drug delivery platform. Three low-toxicity elements, released Cu2+, V9302, and orally administrated disulfiram caused in situ synergistic anti-tumor effect by formation of CuET and intracellular glutathione depletion. [Display omitted] • An injectable hydrogel crosslinked by PA-Cu were designed as local drug delivery system. • The sustained copper from PA-Cu/HPCS hydrogel can coordinate with disulfiram cause local toxicity transformation. • We verified the synergistic effect of PA-Cu/HPCS/V9302 and DSF to hepatocellular carcinoma. Disulfiram (DSF) is a safe drug with negligible toxicity and Cu-dependent anti-tumor efficacy. However, the accumulation and combination of DSF and Cu in non-tumor tissues leads to systemic toxicity owing to the formation of highly poisonous diethyldithiocarbamate (CuET). In addition, CuET-mediated tumor-killing reactive oxygen species may be weakened by intra-tumoral glutathione (GSH). Herein, a synergistic treatment was developed that utilized the oral delivery of DSF and an injectable polyphenol-copper (PA-Cu) hydrogel loaded with the glutamine uptake inhibitor 2-amino-4-bis(phenoxymethyl)aminobutane (V9302). The injectable hydrogels were synthesized by the Schiff base reaction of hydroxypropyl chitosan (HPCS) with a PA-Cu reversible cross-linking agent. Because of the dynamic coordination between PA and Cu, the PA-Cu/HPCS hydrogel gradually releases Cu2+, forming CuET with DSF. The released V9302 inhibits glutamine uptake, thereby suppressing GSH synthesis and enhancing the therapeutic efficacy of the in situ formed CuET. The synergistic effect of PA-Cu/HPCS/V9302 and DSF in eliminating intracellular GSH and killing tumor cells was validated by in vitro cell experiments. Animal experiments further confirmed that PA-Cu/HPCS/V9302 and DSF have an inhibitory effect on tumor growth while maintaining the biosafety of main organs. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
33. Disulfiram-loaded CuO2 nanocarriers for enhanced synergistic chemodynamic chemotherapy.
- Author
-
Hu, Wei, Yang, Lianlian, Liao, Hongtao, Sun, Deguan, Ouyang, Xiao-kun, Wang, Nan, and Yang, Guocai
- Subjects
- *
REACTIVE oxygen species , *DISULFIRAM , *CELL death , *DIETHYLDITHIOCARBAMATE , *NANOCARRIERS - Abstract
[Display omitted] Disulfiram (DSF) metabolites exhibit antitumor properties when bound to Cu2+. This combination also promotes the generation of reactive oxygen species (ROS), ultimately leading to tumor cell death. In this study, CuO 2 served as a carrier for DSF, forming a dual-drug delivery system with Cu2+ and DSF encapsulated in polydopamine (PDA). In the final delivery system, CuO 2 (DSF-CuO 2 @PDA) was hydrolyzed at the tumor site, releasing both Cu2+ and H 2 O 2. Cu2+ reacts with DSF metabolites to form Bis(diethyldithiocarbamate)-Cu (CuET), which triggers a Fenton-like reaction that generates ROS. Chemotherapy and chemodynamic therapy exhibited significant tumor-suppressive capabilities, with an inhibition rate of 61 %. In addition, the DSF-CuO 2 @PDA complex demonstrated superlative tumor-targeting ability and biocompatibility. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. A simple and rapid in vitro assay for identification of direct inhibitors of O6-methylguanine-DNA methyltransferase
- Author
-
Vahid Khalaj, Solmaz AghaAmiri, Sukhen C Ghosh, Servando Hernandez Vargas, Majid Momeny, and Ali Azhdarinia
- Subjects
disulfiram ,GFP ,Lomeguatrib ,MGMT inhibitor ,O6-benzylguanine ,O6-methylguanine methyl transferase ,Biology (General) ,QH301-705.5 - Abstract
O6-Methylguanine-DNA methyltransferase (MGMT) is a DNA repair enzyme that is overexpressed in certain tumors and is associated with resistance to the DNA alkylating agent temozolomide. MGMT inhibitors show potential in combating temozolomide resistance, but current assays for MGMT enzyme activity and inhibition, primarily oligonucleotide-based and fluorescent probe-based, are laborious and costly. The clinical relevance of temozolomide therapy calls for more convenient methodologies to study MGMT inhibition. Here, we extended the application of SNAP-Capture magnetic beads to develop a novel MGMT inhibition assay that demonstrated efficacy not only with known MGMT inhibitors, but also with the aldehyde dehydrogenase inhibitor, disulfiram. The assay uses standard fluorescence microscopy as a simple and reliable detection method, and is translationally applicable in drug discovery programs.
- Published
- 2024
- Full Text
- View/download PDF
35. Repurposing disulfiram with CuET nanocrystals: Enhancing anti-pyroptotic effect through NLRP3 inflammasome inhibition for treating inflammatory bowel diseases
- Author
-
Xueming Xu, Yuanfeng Han, Jiali Deng, Shengfeng Wang, Shijie Zhuo, Kai Zhao, and Wenhu Zhou
- Subjects
Drug repurposing ,Pyroptosis ,Nanoparticles ,Bioavailability ,Disulfiram ,DSS-induced colitis ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Drug repurposing offers a valuable strategy for identifying new therapeutic applications for existing drugs. Recently, disulfiram (DSF), a drug primarily used for alcohol addiction treatment, has emerged as a potential treatment for inflammatory diseases by inhibiting pyroptosis, a form of programmed cell death. The therapeutic activity of DSF can be further enhanced by the presence of Cu2+, although the underlying mechanism of this enhancement remains unclear. In this study, we investigated the mechanistic basis of Cu2+-induced enhancement and discovered that it is attributed to the formation of a novel copper ethylthiocarbamate (CuET) complex. CuET exhibited significantly stronger anti-pyroptotic activity compared to DSF and employed a distinct mechanism of action. However, despite its potent activity, CuET suffered from poor solubility and limited permeability, as revealed by our druggability studies. To overcome these intrinsic limitations, we developed a scalable method to prepare CuET nanocrystals (CuET NCs) using a metal coordination-driven self-assembly approach. Pharmacokinetic studies demonstrated that CuET NCs exhibited a 6-fold improvement in bioavailability. Notably, CuET NCs exhibited high biodistribution in the intestine, suggesting their potential application for the treatment of inflammatory bowel diseases (IBDs). To evaluate their therapeutic efficacy in vivo, we employed a murine model of DSS-induced colitis and observed that CuET NCs effectively attenuated inflammation and ameliorated colitis symptoms. Our findings highlight the discovery of CuET as a potent anti-pyroptotic agent, and the development of CuET NCs represents a novel approach to enhance the druggability of CuET.
- Published
- 2024
- Full Text
- View/download PDF
36. Combination of disulfiram and Copper−Cysteamine nanoparticles induces mitochondria damage and promotes apoptosis in endometrial cancer
- Author
-
Lijun Yang, Cancan Yao, Zhenning Su, Yihao Fang, Nil Kanatha Pandey, Eric Amador, Tian Diao, Guo Bao, Derong Cao, Xihua Chen, Xiangbo Xu, Bin He, Yufeng Zheng, and Wei Chen
- Subjects
Combination ,Disulfiram ,Copper−Cysteamine nanoparticles ,Mitochondria damage ,Promotes apoptosis in Endometrial Cancer ,Materials of engineering and construction. Mechanics of materials ,TA401-492 ,Biology (General) ,QH301-705.5 - Abstract
Endometrial cancer (EC) stands as one of the most prevalent gynecological malignancies affecting women, with its incidence and disease-related mortality steadily on the rise. Disulfiram (DSF), an FDA-approved medication primarily used for treating alcohol addiction, has exhibited promising anti-tumor properties. Studies have revealed DSF's capacity for enhanced anti-tumor activity, particularly when combined with copper. The novel Copper-Cysteamine (CuCy) compound, Cu3Cl(SR)2 (RCH2CH2NH2), showcases photodynamic effects and demonstrates significant anti-tumor potential under various conditions, including exposure to ultraviolet light, X-ray, microwave, and ultrasound. This study delves into exploring the synergistic anti-tumor effects and underlying mechanisms by utilizing copper-cysteamine in conjunction with DSF against endometrial cancer. The investigation involved comprehensive analyses encompassing in vitro experiments utilizing Ishikawa cells, in vivo studies, and transcriptomic analyses. Remarkably, the combined administration of both compounds at a low dose of 0.5 μM exhibited pronounced efficacy in impeding tumor growth, inhibiting blood vessel formation, and stimulating cell apoptosis. Notably, experiments involving transplanted tumors in nude mice vividly demonstrated the significant in vivo anti-tumor effects of this combination treatment. Detailed examination through transmission electron microscopy unveiled compelling evidence of mitochondrial damage, cellular swelling, and rupture, indicative of apoptotic changes in morphology due to the combined treatment. Moreover, transcriptomic analysis unveiled substantial downregulation of mitochondrial-related genes at the molecular level, coupled with a significant hindrance in the DNA repair pathway. These findings strongly suggest that the combined application of CuCy and DSF induces mitochondrial impairment in Ishikawa cells, thereby fostering apoptosis and ultimately yielding potent anti-tumor effects.
- Published
- 2024
- Full Text
- View/download PDF
37. Many human pharmaceuticals are weak inhibitors of the cytochrome P450 system in rainbow trout (Oncorhynchus mykiss) liver S9 fractions.
- Author
-
Pihlaja, Tea, Oksanen, Timo, Vinkvist, Netta, and Sikanen, Tiina
- Subjects
RAINBOW trout ,CYTOCHROME P-450 ,FLECAINIDE ,DISULFIRAM ,BIMATOPROST - Abstract
Introduction: Pharmaceutical residues are widely detected in aquatic environment and can be taken up by nontarget species such as fish. The cytochromes P450 (CYP) represent an important detoxification mechanism in fish, like in humans. In the present study, we assessed the correlation of the substrate selectivities of rainbow trout CYP1A and CYP3A homologues with those of human, through determination of the half-maximal inhibitory concentrations (IC50) of a total sixteen human pharmaceuticals toward CYP1A-like ethoxyresorufin O-deethylase (EROD) and CYP3A-like 7-benzyloxy-4-trifluoromethylcoumarin O-debenzylase (BFCOD) in rainbow trout (Oncorhynchus mykiss) liver S9 fractions (RT-S9). Methods: The inhibitory impacts (IC50) of atomoxetine, atorvastatin, azelastine, bimatoprost, clomethiazole, clozapine, desloratadine, disulfiram, esomeprazole, felbinac, flecainide, orphenadrine, prazosin, quetiapine, sulpiride, and zolmitriptan toward the EROD and BFCOD activities in RT-S9 were determined using the IC50 shift assay, capable of identifying time-dependent inhibitors (TDI). Additionally, the nonspecific binding of the test pharmaceuticals to RT-S9 was assessed using equilibrium dialysis. Results: Most test pharmaceuticals were moderate to weak inhibitors of both EROD and BFCOD activity in RT-S9, even if most are noninhibitors of human CYP1A or CYP3A. Only bimatoprost, clomethiazole, felbinac, sulpiride, and zolmitriptan did not inhibit either activity in RT-S9. EROD inhibition was generally stronger than that of BFCOD and some substances (atomoxetine, flecainide, and prazosin) inhibited selectively only EROD activity. The strongest EROD inhibition was detected with azelastine and esomeprazole (unbound IC50 of 3.8 ± 0.5 µM and 3.0 ± 0.8 µM, respectively). None of the test substances were TDIs of BFCOD, but esomeprazole was a TDI of EROD. Apart from clomethiazole and disulfiram, the nonspecific binding of the test pharmaceuticals to the RT-S9 was extensive (unbound fractions <0.5) and correlated well (R2 = 0.7135) with their water-octanol distribution coefficients. Discussion: The results indicate that the P450 interactions in RT-S9 cannot be explicitly predicted based on human data, but the in vitro data reported herein can shed light on the substrate selectivity of rainbow trout CYP1A1 and CYP3A27 in comparison to their human homologues. The IC50 concentrations are however many orders of magnitude higher than average environmental concentrations of pharmaceuticals. The time-dependent EROD inhibition by esomeprazole could warrant further research to evaluate its possible interlinkages with hepatotoxic impacts on fish. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. Preclinical and Clinical Investigations of Potential Drugs and Vaccines for COVID-19 Therapy: A Comprehensive Review With Recent Update.
- Author
-
Mia, Md. Easin, Howlader, Mithu, Akter, Farzana, and Hossain, Md. Murad
- Subjects
- *
CONVALESCENT plasma , *INVESTIGATIONAL drugs , *VACCINE effectiveness , *COVID-19 vaccines , *PATIENT care , *MESSENGER RNA , *PROTEASE inhibitors , *ANTIVIRAL agents , *MONOCLONAL antibodies , *DRUG approval , *DRUG efficacy , *ALTERNATIVE medicine , *OSELTAMIVIR , *DRUG development , *PEPTIDE antibiotics , *STEM cells , *COVID-19 , *COVID-19 pandemic , *ANTIPARASITIC agents , *DISULFIRAM - Abstract
The COVID-19 pandemic-led worldwide healthcare crisis necessitates prompt societal, ecological, and medical efforts to stop or reduce the rising number of fatalities. Numerous mRNA based vaccines and vaccines for viral vectors have been licensed for use in emergencies which showed 90% to 95% efficacy in preventing SARS-CoV-2 infection. However, safety issues, vaccine reluctance, and skepticism remain major concerns for making mass vaccination a successful approach to treat COVID-19. Hence, alternative therapeutics is needed for eradicating the global burden of COVID-19 from developed and low-resource countries. Repurposing current medications and drug candidates could be a more viable option for treating SARS-CoV-2 as these therapies have previously passed a number of significant checkpoints for drug development and patient care. Besides vaccines, this review focused on the potential usage of alternative therapeutic agents including antiviral, antiparasitic, and antibacterial drugs, protease inhibitors, neuraminidase inhibitors, and monoclonal antibodies that are currently undergoing preclinical and clinical investigations to assess their effectiveness and safety in the treatment of COVID-19. Among the repurposed drugs, remdesivir is considered as the most promising agent, while favipiravir, molnupiravir, paxlovid, and lopinavir/ritonavir exhibited improved therapeutic effects in terms of elimination of viruses. However, the outcomes of treatment with oseltamivir, umifenovir, disulfiram, teicoplanin, and ivermectin were not significant. It is noteworthy that combining multiple drugs as therapy showcases impressive effectiveness in managing individuals with COVID-19. Tocilizumab is presently employed for the treatment of patients who exhibit COVID-19-related pneumonia. Numerous antiviral drugs such as galidesivir, griffithsin, and thapsigargin are under clinical trials which could be promising for treating COVID-19 individuals with severe symptoms. Supportive treatment for patients of COVID-19 may involve the use of corticosteroids, convalescent plasma, stem cells, pooled antibodies, vitamins, and natural substances. This study provides an updated progress in SARS-CoV-2 medications and a crucial guide for inventing novel interventions against COVID-19. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
39. SOD1 inhibition enhances sorafenib efficacy in HBV‐related hepatocellular carcinoma by modulating PI3K/Akt/mTOR pathway and ROS‐mediated cell death.
- Author
-
Lee, Jooyoung, Kim, Jiye, Lee, Ryunjin, Lee, Eunkyeong, An, Hye‐In, Kwon, Yong‐Jae, Jin, Hana, Pack, Chan‐Gi, Kim, Inki, Yoon, Young‐In, Park, Gil‐Chun, Jwa, Eun‐Kyoung, Kwon, Jae Hyun, Namgoong, Jung‐Man, Song, Gi‐Won, Hwang, Shin, Tak, Eunyoung, and Lee, Sung‐Gyu
- Subjects
HEPATITIS B virus ,SUPEROXIDE dismutase ,REACTIVE oxygen species ,HEPATOCELLULAR carcinoma ,CANCER invasiveness ,SORAFENIB ,CELL death - Abstract
Hepatitis B Virus (HBV) infection significantly elevates the risk of hepatocellular carcinoma (HCC), with the HBV X protein (HBx) playing a crucial role in cancer progression. Sorafenib, the primary therapy for advanced HCC, shows limited effectiveness in HBV‐infected patients due to HBx‐related resistance. Numerous studies have explored combination therapies to overcome this resistance. Sodium diethyldithiocarbamate (DDC), known for its anticancer effects and its inhibition of superoxide dismutase 1 (SOD1), is hypothesized to counteract sorafenib (SF) resistance in HBV‐positive HCCs. Our research demonstrates that combining DDC with SF significantly reduces HBx and SOD1 expressions in HBV‐positive HCC cells and human tissues. This combination therapy disrupts the PI3K/Akt/mTOR signalling pathway and promotes apoptosis by increasing reactive oxygen species (ROS) levels. These cellular changes lead to reduced tumour viability and enhanced sensitivity to SF, as evidenced by the synergistic suppression of tumour growth in xenograft models. Additionally, DDC‐mediated suppression of SOD1 further enhances SF sensitivity in HBV‐positive HCC cells and xenografted animals, thereby inhibiting cancer progression more effectively. These findings suggest that the DDC‐SF combination could serve as a promising strategy for overcoming SF resistance in HBV‐related HCC, potentially optimizing therapy outcomes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
40. Disulfiram downregulates ferredoxin 1 to maintain copper homeostasis and inhibit inflammation in cerebral ischemia/reperfusion injury.
- Author
-
Yang, Shuai, Li, Xudong, Yan, Jinhong, Jiang, Fangchao, Fan, Xuehui, Jin, Jing, Zhang, Weihua, Zhong, Di, and Li, Guozhong
- Subjects
- *
COPPER , *REPERFUSION injury , *CEREBRAL ischemia , *DISULFIRAM , *COPPER proteins , *REPERFUSION , *HOMEOSTASIS - Abstract
In the current study, we aimed to investigate whether disulfiram (DSF) exerts a neuroprotective role in cerebral ischemiareperfusion (CI-RI) injury by modulating ferredoxin 1 (FDX1) to regulate copper ion (Cu) levels and inhibiting inflammatory responses. To simulate CI-RI, a transient middle cerebral artery occlusion (tMCAO) model in C57/BL6 mice was employed. Mice were administered with or without DSF before and after tMCAO. Changes in infarct volume after tMCAO were observed using TTC staining. Nissl staining and hematoxylin–eosin (he) staining were used to observe the morphological changes of nerve cells at the microscopic level. The inhibitory effect of DSF on initial inflammation was verified by TUNEL assay, apoptosis-related protein detection and iron concentration detection. FDX1 is the main regulatory protein of copper death, and the occurrence of copper death will lead to the increase of HSP70 stress and inflammatory response. Cuproptosis-related proteins and downstream inflammatory factors were detected by western blotting, immunofluorescence staining, and immunohistochemistry. The content of copper ions was detected using a specific kit, while electron microscopy was employed to examine mitochondrial changes. We found that DSF reduced the cerebral infarction volume, regulated the expression of cuproptosis-related proteins, and modulated copper content through down regulation of FDX1 expression. Moreover, DSF inhibited the HSP70/TLR-4/NLRP3 signaling pathway. Collectively, DSF could regulate Cu homeostasis by inhibiting FDX1, acting on the HSP70/TLR4/NLRP3 pathway to alleviate CI/RI. Accordingly, DSF could mitigate inflammatory responses and safeguard mitochondrial integrity, yielding novel therapeutic targets and mechanisms for the clinical management of ischemia–reperfusion injury. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
41. Pyroptosis-mediator GSDMD promotes Parkinson's disease pathology via microglial activation and dopaminergic neuronal death.
- Author
-
Zhang, Xiaoshuang, Zhang, Yunhe, Wang, Boya, Xie, Chuantong, Wang, Jinghui, Fang, Rong, Dong, Hongtian, Fan, Guangchun, Wang, Mengze, He, Yongtao, Shen, Chenye, Duan, Yufei, Zhao, Jiayin, Liu, Zhaolin, Li, Qing, Ma, Yuanyuan, Yu, Mei, Wang, Jian, Fei, Jian, and Xiao, Lei
- Subjects
- *
PARKINSON'S disease , *PATHOLOGY , *DOPAMINERGIC neurons , *ANIMAL diseases , *MICROGLIA - Abstract
• GSDMD-mediated pyroptosis is activated in pro-inflammatory microglia and degenerating dopaminergic neurons in PD models. • Gsdmd deficiency suppresses neuroinflammation in PD mouse models, and also inhibits MPTP-induced neuropathology. • Gsdmd deficiency inhibits the release of NO in LPS-treated primary microglia and death of MPP+-loaded primary neurons. • Pyroptosis inhibitor disulfiram alleviates MPTP-induced damages in PD mice. GSDMD-mediated pyroptosis occurs in the nigrostriatal pathway in Parkinson's disease animals, yet the role of GSDMD in neuroinflammation and death of dopaminergic neurons in Parkinson's disease remains elusive. Here, our in vivo and in vitro studies demonstrated that GSDMD, as a pyroptosis executor, contributed to glial reaction and death of dopaminergic neurons across different Parkinson's disease models. The ablation of the Gsdmd attenuated Parkinson's disease damage by reducing dopaminergic neuronal death, microglial activation, and detrimental transformation. Disulfiram, an inhibitor blocking GSDMD pore formation, efficiently curtailed pyroptosis, thereby lessening the pathology of Parkinson's disease. Additionally, a modification in GSDMD was identified in the blood of Parkinson's disease patients in contrast to healthy subjects. Therefore, the detected alteration in GSDMD within the blood of Parkinson's disease patients and the protective impact of disulfiram could be promising for the diagnostic and therapeutic approaches against Parkinson's disease. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
42. Endpoints for Pharmacotherapy Trials for Alcohol Use Disorder.
- Author
-
Belnap, Malia A., McManus, Kaitlin R., Grodin, Erica N., and Ray, Lara A.
- Subjects
- *
ALCOHOLISM , *DRUG therapy , *ALCOHOL drinking , *BIOMARKERS , *DISULFIRAM - Abstract
Alcohol use disorder (AUD) is a debilitating disorder, yet currently approved pharmacotherapies to treat AUD are under-utilized. The three medications approved by the US Food and Drug Administration (FDA) for the indication of AUD are disulfiram, acamprosate, and naltrexone. The current landscape of pharmacotherapies for AUD suggests opportunities for improvement. Clinical trials investigating novel pharmacotherapies for AUD traditionally use abstinence-based drinking outcomes or no heavy drinking days as trial endpoints to determine the efficacy of pharmacotherapies. These outcomes are typically measured through patient self-report endorsements of their drinking. Apart from these traditional outcomes, there have been recent developments in novel endpoints for AUD pharmacotherapies. These novel endpoints include utilizing the World Health Organization (WHO) risk drinking level reductions to promote a harm-reduction endpoint rather than an abstinence-based endpoint. Additionally, in contrast to patient self-report measurements, biological markers of alcohol use may serve as objective endpoints in AUD pharmacotherapy trials. Lastly, the National Institute on Alcohol Abuse and Alcoholism (NIAAA) definition of recovery from AUD and patient-oriented outcomes offer new frameworks to consider endpoints associated with more than alcohol consumption itself, such as the provider-patient experiences with novel pharmacotherapies. These recent developments in new endpoints for AUD pharmacotherapies offer promising future opportunities for pharmacotherapy development, so long as validity and reliability measures are demonstrated for the endpoints. A greater breadth of endpoint utilization may better capture the complexity of AUD symptomatology. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
43. High throughput screening identifies dasatinib as synergistic with trametinib in low grade serous ovarian carcinoma.
- Author
-
Hollis, Robert L., Elliott, Richard, Dawson, John C., Ilenkovan, Narthana, Matthews, Rosie M., Stillie, Lorna J., Oswald, Ailsa J., Kim, Hannah, Llaurado Fernandez, Marta, Churchman, Michael, Porter, Joanna M., Roxburgh, Patricia, Unciti-Broceta, Asier, Gershenson, David M., Herrington, C. Simon, Carey, Mark S., Carragher, Neil O., and Gourley, Charlie
- Subjects
- *
DASATINIB , *HIGH throughput screening (Drug development) , *HISTONE deacetylase inhibitors , *PROTEIN-tyrosine kinase inhibitors , *ALDEHYDE dehydrogenase , *PROTEASOME inhibitors - Abstract
Low grade serous ovarian carcinoma (LGSOC) is a distinct histotype of ovarian cancer characterised high levels of intrinsic chemoresistance, highlighting the urgent need for new treatments. High throughput screening in clinically-informative cell-based models represents an attractive strategy for identifying candidate treatment options for prioritisation in clinical studies. We performed a high throughput drug screen of 1610 agents across a panel of 6 LGSOC cell lines (3 RAS/RAF-mutant, 3 RAS/RAF-wildtype) to identify novel candidate therapeutic approaches. Validation comprised dose-response analysis across 9 LGSOC models and 5 high grade serous comparator lines. 16 hits of 1610 screened compounds were prioritised for validation based on >50% reduction in nuclei counts in over half of screened cell lines at 1000 nM concentration. 11 compounds passed validation, and the four agents of greatest interest (dasatinib, tyrosine kinase inhibitor; disulfiram, aldehyde dehydrogenase inhibitor; carfilzomib, proteasome inhibitor; romidepsin, histone deacetylase inhibitor) underwent synergy profiling with the recently approved MEK inhibitor trametinib. Disulfiram demonstrated excellent selectivity for LGSOC versus high grade serous ovarian carcinoma comparator lines (P = 0.003 for IC50 comparison), while the tyrosine kinase inhibitor dasatinib demonstrated favourable synergy with trametinib across multiple LGSOC models (maximum zero interaction potency synergy score 46.9). The novel, highly selective Src family kinase (SFK) inhibitor NXP900 demonstrated a similar trametinib synergy profile to dasatinib, suggesting that SFK inhibition is the likely driver of synergy. Dasatinib and other SFK inhibitors represent novel candidate treatments for LGSOC and demonstrate synergy with trametinib. Disulfiram represents an additional treatment strategy worthy of investigation. • We performed a high throughput screen of 1610 compounds across six low grade serous ovarian carcinoma cell lines • Of 16 prioritised screen hits, 11 compounds passed dose-response validation using cell viability assays • The most promising hits underwent synergy profiling with the trametinib (dasatinib, disulfiram, carfilzomib, romidepsin) • Disulfiram demonstrated excellent selectivity for LGSOC versus high grade serous ovarian carcinoma comparator lines • Dasatinib demonstrated favourable synergy with the MEK inhibitor trametinib [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
44. Exploring Disulfiram's Anticancer Potential: PLGA Nano-Carriers for Prolonged Drug Delivery and Potential Improved Therapeutic Efficacy.
- Author
-
Dumbuya, Ibrahim, Pereira, Ana Maria, Tolaymat, Ibrahim, Al Dalaty, Adnan, Arafat, Basel, Webster, Matt, Pierscionek, Barbara, Khoder, Mouhamad, and Najlah, Mohammad
- Subjects
- *
DISULFIRAM , *SIZE reduction of materials , *TREATMENT effectiveness , *DRUG delivery systems , *BREAST cancer - Abstract
Disulfiram (DS) has been shown to have potent anti-cancer activity; however, it is also characterised by its low water solubility and rapid metabolism in vivo. Biodegradable polylactic-co-glycolic acid (PLGA) polymers have been frequently employed in the manufacturing of PLGA nano-carrier drug delivery systems. Thus, to develop DS-loaded PLGA nanoparticles (NPs) capable of overcoming DS's limitations, two methodologies were used to formulate the NPs: direct nanoprecipitation (DNP) and single emulsion/solvent evaporation (SE), followed by particle size reduction. The DNP method was demonstrated to produce NPs of superior characteristics in terms of size (151.3 nm), PDI (0.083), charge (−37.9 mV), and loading efficiency (65.3%). Consequently, NPs consisting of PLGA and encapsulated DS coated with mPEG2k-PLGA at adjustable ratios were prepared using the DNP method. Formulations were then characterised, and their stability in horse serum was assessed. Results revealed the PEGylated DS-loaded PLGA nano-carriers to be more efficient; hence, in-vitro studies testing these formulations were subsequently performed using two distinct breast cancer cell lines, showing great potential to significantly enhance cancer therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
45. Pharmacological treatments for alcohol dependence: Evidence on uptake, inequalities and comparative effectiveness from a UK population‐based cohort.
- Author
-
Manca, Francesco, Zhang, Lisong, Fitzgerald, Niamh, Ho, Frederick, Innes, Hamish, Jani, Bhautesh, Katikireddi, Srinivasa Vittal, McAuley, Andrew, Sharp, Clare, and Lewsey, Jim
- Subjects
- *
ALCOHOLISM , *DRUG therapy , *DRUG prescribing , *DISULFIRAM , *ODDS ratio - Abstract
Introduction: We assessed the prevalence of prescribing of certain medications for alcohol dependence and the extent of any inequalities in receiving prescriptions for individuals with such a diagnosis. Further, we compared the effectiveness of two of the most prescribed medications (acamprosate and disulfiram) for alcohol dependence and assessed whether there is inequality in prescribing either of them. Methods: We used a nationwide dataset on prescriptions and hospitalisations in Scotland, UK (N = 19,748). We calculated the percentage of patients receiving alcohol dependence prescriptions after discharge, both overall and by socio‐economic groups. Binary logistic regressions were used to assess the odds of receiving any alcohol‐dependence prescription and the comparative odds of receiving acamprosate or disulfiram. Comparative effectiveness in avoiding future alcohol‐related hospitalisations (N = 11,239) was assessed using Cox modelling with statistical adjustment for potential confounding. Results: Upto 7% of hospitalised individuals for alcohol use disorder received prescriptions for alcohol dependence after being discharged. Least deprived socio‐economic groups had relatively more individuals receiving prescriptions. Inequalities in prescribing for alcohol dependence existed, especially across sex and comorbidities: males had 12% (odds ratio [OR] 0.88, 95% confidence interval [CI] 0.81–0.96) and those with a history of mental health hospitalisations had 10% (OR 0.90, 95% CI 0.82–0.98) lower odds of receiving prescriptions after an alcohol‐related hospitalisation. Prescribing disulfiram was superior to prescribing acamprosate in preventing alcohol‐related hospitalisations (hazard ratio ranged between 0.60 and 0.81 across analyses). Disulfiram was relatively less likely prescribed to those from more deprived areas. Discussion and Conclusions: Inequalities in prescribing for alcohol dependence exists in Scotland with lower prescribing to men and disulfiram prescribed more to those from least deprived areas. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
46. A disulfiram derivative against lung cancer via the Notch signaling pathway without neurotoxicity and hepatotoxicity.
- Author
-
Lv, Huaiyou, Yang, Huatian, Duan, Yifei, Sha, Hongyu, and Zhao, Zhongxi
- Subjects
NOTCH signaling pathway ,LUNG cancer ,DISULFIRAM ,HEPATOTOXICOLOGY ,NEUROTOXICOLOGY - Abstract
The exploration of novel anti-lung cancer small-molecule drugs is important for drug resistance and adverse effects of chemotherapeutic drugs in current clinics. Disulfiram (DSF), as an antidote, has been proven to have excellent antitumor effects in combination with copper (Cu). However, the risk for potential neurotoxicity and hepatotoxicity in clinical use, as well as its poor water solubility, limits its use. In this study, we identified a DSF derivative, S-(N,N-diethyldithiocarbamoyl)-N-acetyl-L-cysteine, which could greatly increase the water solubility by converting it to a calcium salt (DS-NAC). The anti-lung cancer pharmacodynamic studies in vitro of DS-NAC were evaluated and a mouse model of lung cancer in situ was established to explore the therapeutic effects of DS-NAC compared with DSF and oxaliplatin (OXA). The results demonstrated that DS-NAC combined with Cu had superior cytotoxicity to DSF and OXA in the CCK8 assay against lung cancer cells, and exhibited potent anti-metastatic, epithelial-mesenchymal transition inhibition. In addition, DS-NAC showed better antitumor effects than DSF and comparable effects to OXA in lung cancer in situ model. In terms of the antitumor mechanism, we discovered that DS-NAC in combination with Cu exerted a greater inhibitory effect on the Notch pathway than DSF, which may account for its excellent antitumor effects. Finally, we verified the safety of DS-NAC in vivo, showing lower hepatotoxicity and neurotoxicity compared with DSF and OXA. DS-NAC is a promising anti-lung cancer drug with a favorable safety profile. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
47. Design and Characterization of Chitosan-Based Smart Injectable Hydrogel for Improved Sustained Release of Antinarcotics.
- Author
-
Aftab, Maryam, Javed, Fatima, Haider, Sajjad, Khan, Rawaiz, Khan, Salah Uddin, Alam, Kamran, Amir, Afreenish, Ullah, Faheem, and Shah, Naseer Ali
- Subjects
- *
GUAR gum , *FOURIER transform infrared spectroscopy , *HYDROGELS - Abstract
The treatment adherence of narcotics-addicted individuals with reduced incidences of relapse can be enhanced by a sustained drug release formulation of antinarcotics. So far, different drug formulations have been reported with sustained drug release periods of 28 and 35 days. To further enhance this duration, different formulations of injectable hydrogels (IHs) have been developed by combining low molecular weight (LMW) and high molecular weight (HMW) chitosan (CS) with guar gum (GG) and crosslinking them by sodium bi phosphate dibasic. The structural, morphological, and physicochemical properties of LMW-CS IH, and HMW-CS IH were evaluated using Fourier transform infrared spectroscopy (FT-IR), thermo-gravimetric analysis (TGA), scanning electron microscopy (SEM), and rheological, swelling, and biodegradation analysis. The HMW-CS IH showed high crosslinking, increased thermal stability, high mechanical strength, elevated swelling, and low biodegradation. The antinarcotic drugs naltrexone (NTX) and disulfiram (DSF) were loaded separately into the HMW-CS IH and LMW-CS IH. The release of NTX and DSF was investigated in phosphate buffer saline (PBS) and ethanol (0.3%, 0.4%, and 0.5%) over a 56-day period using an UV spectrophotometer. The drug release data were tested in zero-order, first-order, and Korsemeyer–Peppas mathematical models. In PBS, all prepared formulations followed non-Fickian drug release, while in ethanol, only NTX HMW-CS IH followed non-Fickian release in all three different concentrations of ethanol. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
48. Repurposing disulfiram with CuET nanocrystals: Enhancing anti-pyroptotic effect through NLRP3 inflammasome inhibition for treating inflammatory bowel diseases.
- Author
-
Xu, Xueming, Han, Yuanfeng, Deng, Jiali, Wang, Shengfeng, Zhuo, Shijie, Zhao, Kai, and Zhou, Wenhu
- Subjects
INFLAMMATORY bowel diseases ,NLRP3 protein ,DISULFIRAM ,INFLAMMASOMES ,NANOCRYSTALS - Abstract
Drug repurposing offers a valuable strategy for identifying new therapeutic applications for existing drugs. Recently, disulfiram (DSF), a drug primarily used for alcohol addiction treatment, has emerged as a potential treatment for inflammatory diseases by inhibiting pyroptosis, a form of programmed cell death. The therapeutic activity of DSF can be further enhanced by the presence of Cu
2+ , although the underlying mechanism of this enhancement remains unclear. In this study, we investigated the mechanistic basis of Cu2+ -induced enhancement and discovered that it is attributed to the formation of a novel copper ethylthiocarbamate (CuET) complex. CuET exhibited significantly stronger anti-pyroptotic activity compared to DSF and employed a distinct mechanism of action. However, despite its potent activity, CuET suffered from poor solubility and limited permeability, as revealed by our druggability studies. To overcome these intrinsic limitations, we developed a scalable method to prepare CuET nanocrystals (CuET NCs) using a metal coordination-driven self-assembly approach. Pharmacokinetic studies demonstrated that CuET NCs exhibited a 6-fold improvement in bioavailability. Notably, CuET NCs exhibited high biodistribution in the intestine, suggesting their potential application for the treatment of inflammatory bowel diseases (IBDs). To evaluate their therapeutic efficacy in vivo , we employed a murine model of DSS-induced colitis and observed that CuET NCs effectively attenuated inflammation and ameliorated colitis symptoms. Our findings highlight the discovery of CuET as a potent anti-pyroptotic agent, and the development of CuET NCs represents a novel approach to enhance the druggability of CuET. Mass-produced CuET nanocrystals (NCs) were developed to solve CuET druggability limitations for inflammatory bowel disease (IBD) therapy and the anti-pyroptotic mechanism of CuET through NLRP3 inflammasome inhibition was elucidated. [Display omitted] [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
49. Changes in Histone Code Regulation during the Initiation of Paraptosis-Like Death of HEp-2 Tumor Cells by Oxidized Disulfiram Derivatives.
- Author
-
Solovieva, M. E., Shatalin, Yu. V., and Akatov, V. S.
- Abstract
Disulfiram (DSF) and its oxidized derivatives (DSFoxy) are currently being investigated as potential anticancer agents. We previously found that DSFoxy initiate the paraptosis-like death of tumor cells, which is of potential interest for the treatment of tumors resistant to the initiation of apoptosis. Based on bioinformatics analysis of mass spectrometric data on protein ubiquitination, we formulated a conception about the important role of disruption of the retrograde transport of damaged proteins from the endoplasmic reticulum to the cytosol in the mechanism of initiation of paraptosis-like cell death. In the present work, it has been found that DSFoxy, in the process of initiating paraptosis-like death of human adenocarcinoma HEp-2 cells, also enhances the ubiquitination of histones and histone code enzymes. In particular, this applies to the ubiquitination of histone H2BC12, histone methyltransferases responsible for transcription and repair of damaged DNA, as well as acetylating and ubiquitin-conjugating proteins. Bioinformatics analysis of changes in ubiquitination of cell nuclear proteins using the STRING database revealed during this process an increase in the occurrence of ubiquitinated proteins (functional enrichment) of cell cycle regulation, cell response to DNA damage and DNA repair, the regulation of which also depends on the histone code. This directly indicates damage to the cell nucleus and is consistent with confocal microscopy data. These results indicate that paraptosis-like death initiated by DSFoxy is accompanied, along with impairment of retrograde transport and ER stress, also by a change in regulation of the histone code, which points to a pleiotropic mechanism of cell death induction. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
50. Immunohistochemical evidences of anticancer actions of metformin with other repurposed drug combinations and correlation with hamster fibrosarcoma tumor size.
- Author
-
POPOVIĆ, JOVAN K., POPOVIĆ, DUŠICA J., POPOVIĆ, KOSTA J., MILJKOVIĆ, DEJAN, LALOŠEVIĆ, DUŠAN, DOLIĆANIN, ZANA, and ČAPO, IVAN
- Subjects
- *
METFORMIN , *ITRACONAZOLE , *FIBROSARCOMA , *ANTINEOPLASTIC agents , *DISULFIRAM , *HAMSTERS , *GOLDEN hamster , *NEOVASCULARIZATION - Abstract
The aim was to detect and correlate anticancer effects of metformin in combinations with other repurposed drugs, already registered for other indications, which may be immediately applied and clinically investigated in oncology, reducing the time and cost of research for new cancer treatments. Immunohistochemistry was performed for tumors treated by dual drug combinations containing metformin with deoxycholic acid, caffeine, itraconazole, nitroglycerin, disulfiram or diclofenac. The drugs were applied in Syrian golden hamsters (6 animals per group) with the inoculated BHK21/C13 fibrosarcoma in doses equivalent to usual human doses, <50 % LD50. The anticancer effects were assessed by: p53 (mutational status); Ki-67 and PCNA (tumor proliferation); CD34 and CD31 (neoangiogenesis); GLUT1 (glucose metabolism); iNOS (NO metabolism); COX4, Cytochrome C and caspase 3 (apoptosis); immunohistochemical markers. Also, biophysical characteristics of fibrosarcoma, animal blood samples and the toxicity on main organs were analyzed. Treatments significantly (P < 0.05) reduced mutational status, tumor proliferation, neoangiogenesis, glucose metabolism, NO metabolism and modulated apoptosis, in correlation with tumor size, without toxicity and influence on biochemical blood and hematological tests. The administration of metformin in two-drug combination with deoxycholic acid, caffeine, itraconazole, nitroglycerin, disulfiram or diclofenac may be recommended for further clinical investigations in oncology. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.