Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Br J Cancer ; 125(1): 55-64, 2021 07.
Article in English | MEDLINE | ID: mdl-33837299

ABSTRACT

BACKGROUND: The prognosis for high-risk childhood acute leukaemias remains dismal and established treatment protocols often cause long-term side effects in survivors. This study aims to identify more effective and safer therapeutics for these patients. METHODS: A high-throughput phenotypic screen of a library of 3707 approved drugs and pharmacologically active compounds was performed to identify compounds with selective cytotoxicity against leukaemia cells followed by further preclinical evaluation in patient-derived xenograft models. RESULTS: Auranofin, an FDA-approved agent for the treatment of rheumatoid arthritis, was identified as exerting selective anti-cancer activity against leukaemia cells, including patient-derived xenograft cells from children with high-risk ALL, versus solid tumour and non-cancerous cells. It induced apoptosis in leukaemia cells by increasing reactive oxygen species (ROS) and potentiated the activity of the chemotherapeutic cytarabine against highly aggressive models of infant MLL-rearranged ALL by enhancing DNA damage accumulation. The enhanced sensitivity of leukaemia cells towards auranofin was associated with lower basal levels of the antioxidant glutathione and higher baseline ROS levels compared to solid tumour cells. CONCLUSIONS: Our study highlights auranofin as a well-tolerated drug candidate for high-risk paediatric leukaemias that warrants further preclinical investigation for application in high-risk paediatric and adult acute leukaemias.


Subject(s)
Auranofin/administration & dosage , Cytarabine/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Reactive Oxygen Species/metabolism , Animals , Auranofin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Child , Cytarabine/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , Female , High-Throughput Screening Assays , Humans , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Small Molecule Libraries , Xenograft Model Antitumor Assays
2.
Lancet Respir Med ; 9(8): 897-908, 2021 08.
Article in English | MEDLINE | ID: mdl-33740465

ABSTRACT

BACKGROUND: Current tuberculosis treatments leave patients with clinically significant lung injury and increased all-cause mortality post-cure. Adjunctive host-directed therapies could protect the lungs, improve long-term survival, and shorten treatment duration; however, few have been tested clinically. Therefore, we aimed to assess the safety and preliminary efficacy of four host-directed therapies for tuberculosis. METHODS: In this prospective, open-label, phase 2, randomised controlled trial, patients with pulmonary tuberculosis were recruited at three clinical sites in South Africa. Eligible patients were aged 18-65 years, HIV-1-negative, and had rifampicin-susceptible Mycobacterium tuberculosis, a sputum Xpert cycle threshold of less than 20, and moderately advanced or far advanced disease on chest radiography. By use of numbers generated in blocks of ten and stratification by site, eligible patients were randomly assigned (1:1:1:1:1) to receive one of the four oral host-directed treatments plus standard tuberculosis treatment or standard treatment alone (the control group). Host-directed treatments were: CC-11050 (200 mg twice daily, taken with food; day 1-112); everolimus (0·5 mg/day; day 1-112); auranofin (3 mg/day for seven doses, then 6 mg/day; day 1-112); and ergocalciferol (5 mg on day 1, then 2·5 mg on day 28 and day 56). All study participants received oral rifabutin-substituted standard tuberculosis treatment for 180 days. Patients and clinicians were not masked to treatment assignment. Spirometry and sputum culture with solid and liquid media were done at baseline and up to 180 days at specified intervals throughout treatment. The primary endpoint was safety and tolerability up to day 210. Secondary preliminary efficacy endpoints were treatment effects on sputum microbiology (culture status at day 56 and the hazard ratio for stable culture conversion up to day 180) and lung function (FEV1 and forced vital capacity [FVC]) measured by spirometry at day 56, day 180, and day 540. Safety was analysed in the intention-to-treat population and preliminary efficacy primarily in the per-protocol population. The trial is registered at ClinicalTrials.gov, NCT02968927. Post-treatment follow-up was completed in 2020. FINDINGS: Between Nov 18, 2016, and Sept 27, 2018, 200 patients were screened and randomly assigned to different treatment groups (n=40 per group, apart from n=39 in the everolimus group after one patient withdrew consent). 11 treatment-emergent serious adverse events occurred either during treatment or within 30 days after treatment discontinuation, of which three were attributable to a host-directed treatment. Life-threatening thrombocytopenia occurred in an auranofin recipient; apparent intra-abdominal sepsis leading to death occurred in another auranofin recipient and was classified as a suspected unexpected serious adverse reaction. Tuberculous spondylitis occurred as an apparent paradoxical reaction in a patient receiving ergocalciferol. Two patients in the control group had life-threatening, treatment-attributable liver injury. No treatment-emergent, treatment-attributable serious adverse events occurred in patients receiving CC-11050 or everolimus. Mean FEV1 in the control group was 61·7% of predicted (95% CI 56·3-67·1) at baseline and 69·1% (62·3-75·8) at day 180. Patients treated with CC-11050 and everolimus had increased recovery of FEV1 at day 180 relative to the control group (mean difference from control group 6·30%, 95% CI 0·06-12·54; p=0·048; and 6·56%, 0·18-12·95; p=0·044, respectively), whereas auranofin and ergocalciferol recipients did not. None of the treatments had an effect on FVC during 180 days of follow-up or on measures of sputum culture status over the course of the study. INTERPRETATION: CC-11050 and everolimus were safe and reasonably well tolerated as adjunctive therapies for tuberculosis, and analysis of preliminary efficacy suggests they might also enhance the recovery of FEV1, a key measure of lung function and predictor of all-cause mortality. Further studies of these candidates are warranted. FUNDING: The Bill & Melinda Gates Foundation and the South African Medical Research Council.


Subject(s)
Antitubercular Agents/administration & dosage , Auranofin/administration & dosage , Ergocalciferols/administration & dosage , Everolimus/administration & dosage , Indoles/administration & dosage , Phosphodiesterase 4 Inhibitors/administration & dosage , Sulfones/administration & dosage , Tuberculosis, Pulmonary/drug therapy , Adult , Antitubercular Agents/adverse effects , Auranofin/adverse effects , Auranofin/pharmacology , Double-Blind Method , Drug Therapy, Combination , Ergocalciferols/adverse effects , Ergocalciferols/pharmacology , Everolimus/adverse effects , Everolimus/pharmacology , Female , Forced Expiratory Volume/drug effects , Humans , Indoles/adverse effects , Indoles/pharmacology , Male , Middle Aged , Mycobacterium tuberculosis , Phosphodiesterase 4 Inhibitors/adverse effects , Phosphodiesterase 4 Inhibitors/pharmacology , Prospective Studies , South Africa , Sputum/drug effects , Sputum/microbiology , Sulfones/adverse effects , Sulfones/pharmacology
3.
Yakugaku Zasshi ; 141(3): 315-321, 2021.
Article in Japanese | MEDLINE | ID: mdl-33642497

ABSTRACT

Gold compounds have been employed throughout history to treat various types of disease, from ancient times to the present day. In the year 1985, auranofin, a gold-containing compound, was approved by U.S. Food and Drug Administration (FDA) as a therapeutic agent to target rheumatoid arthritis that would facilitate easy oral drug administration as opposed to conventional intramuscular injection used in treatments. Furthermore, auranofin demonstrates promising results for the treatment of various diseases beyond rheumatoid arthritis, including cancer, neurodegenerative diseases, acquired immune deficiency syndrome, and bacterial and parasitic infections. Various potential novel applications for auranofin have been proposed for treating human diseases. Auranofin has previously been demonstrated to inhibit thioredoxin reductase (TrxR) involved within the thioredoxin (Trx) system that comprises one of the critical cellular redox systems within the body. TrxR comprises the sole known enzyme that catalyzes Trx reduction. With cancers in particular, TrxR inhibition facilitates an increase in cellular oxidative stress and suppresses tumor growth. In this review, we describe the potential of auranofin to serve as an anticancer agent and further drug repurposing to utilize this as a strategy for further appropriate drug developments.


Subject(s)
Antineoplastic Agents , Arthritis, Rheumatoid/drug therapy , Auranofin/administration & dosage , Auranofin/pharmacology , Drug Repositioning , Drug Development , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Oxidative Stress , Thioredoxin-Disulfide Reductase/antagonists & inhibitors
5.
Mol Ther ; 29(2): 775-787, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33091613

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) represents one of the deadliest forms of cancer with very few available therapeutic options. We previously reported that an engineered human enzyme, cyst(e)inase, which degrades L-cysteine (L-Cys) and cystine, inhibits growth of multiple cancer cells, including PDAC both in vitro and in vivo. Here, we show that cyst(e)inase treatment leads to increased clustered oxidative DNA damage, DNA single-strand breaks, apurinic/apyrimidinic sites, and DNA double-strand breaks (DSBs) in PDAC cells sensitive to intracellular depletion of L-Cys that is associated with reduced survival. BRCA2-deficient PDAC cells exhibited increased DSBs and enhanced sensitivity to cyst(e)inase. The blocking of a second antioxidant pathway (thioredoxin/thioredoxin reductase) using auranofin or inhibiting DNA repair using the poly (ADP-ribose) polymerase (PARP) inhibitor, olaparib, led to significant increases in DSBs following cyst(e)inase treatment in all PDAC cells examined. Cyst(e)inase plus olaparib also synergistically inhibited growth of sensitive and resistant PDAC cells in both xenograft and allograft tumor models. Collectively, these results demonstrate an important role for oxidative DNA damage and ultimately DNA DSBs in the anticancer action of cyst(e)inase. The data further show the potential for combining agents that target alternate antioxidant pathways or by targeting DNA repair pathways or genetic liabilities in DNA repair pathways to enhance the therapeutic action of cyst(e)inase for PDAC.


Subject(s)
Cysteine/metabolism , DNA Damage , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy , Animals , Auranofin/administration & dosage , DNA Damage/drug effects , Disease Models, Animal , Humans , Mice , Oxidative Stress , Pancreatic Neoplasms/etiology , Reactive Oxygen Species , Xenograft Model Antitumor Assays
6.
PLoS One ; 15(12): e0244540, 2020.
Article in English | MEDLINE | ID: mdl-33378390

ABSTRACT

Despite dramatic improvements in outcomes arising from the introduction of targeted therapies and immunotherapies, metastatic melanoma is a highly resistant form of cancer with 5 year survival rates of <35%. Drug resistance is frequently reported to be associated with changes in oxidative metabolism that lead to malignancy that is non-responsive to current treatments. The current report demonstrates that triphenylphosphonium(TPP)-based lipophilic cations can be utilized to induce cytotoxicity in pre-clinical models of malignant melanoma by disrupting mitochondrial metabolism. In vitro experiments demonstrated that TPP-derivatives modified with aliphatic side chains accumulated in melanoma cell mitochondria; disrupted mitochondrial metabolism; led to increases in steady-state levels of reactive oxygen species; decreased total glutathione; increased the fraction of glutathione disulfide; and caused cell killing by a thiol-dependent process that could be rescued by N-acetylcysteine. Furthermore, TPP-derivative-induced melanoma toxicity was enhanced by glutathione depletion (using buthionine sulfoximine) as well as inhibition of thioredoxin reductase (using auranofin). In addition, there was a structure-activity relationship between the aliphatic side-chain length of TPP-derivatives (5-16 carbons), where longer carbon chains increased melanoma cell metabolic disruption and cell killing. In vivo bio-distribution experiments showed that intratumoral administration of a C14-TPP-derivative (12-carbon aliphatic chain), using a slow-release thermosensitive hydrogel as a delivery vehicle, localized the drug at the melanoma tumor site. There, it was observed to persist and decrease the growth rate of melanoma tumors. These results demonstrate that TPP-derivatives selectively induce thiol-dependent metabolic oxidative stress and cell killing in malignant melanoma and support the hypothesis that a hydrogel-based TPP-derivative delivery system could represent a therapeutic drug-delivery strategy for melanoma.


Subject(s)
Auranofin/administration & dosage , Buthionine Sulfoximine/administration & dosage , Melanoma/drug therapy , Mitochondria/metabolism , Organophosphorus Compounds/administration & dosage , Animals , Auranofin/pharmacology , Buthionine Sulfoximine/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Delayed-Action Preparations , Drug Synergism , Female , Humans , Hydrogels/chemistry , Melanoma/metabolism , Mice , Mitochondria/drug effects , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacology , Oxidative Stress/drug effects , Structure-Activity Relationship , Temperature , Xenograft Model Antitumor Assays
7.
Nat Commun ; 11(1): 5263, 2020 10 16.
Article in English | MEDLINE | ID: mdl-33067430

ABSTRACT

Global emergence of Gram-negative bacteria carrying the plasmid-borne resistance genes, blaMBL and mcr, raises a significant challenge to the treatment of life-threatening infections by the antibiotics, carbapenem and colistin (COL). Here, we identify an antirheumatic drug, auranofin (AUR) as a dual inhibitor of metallo-ß-lactamases (MBLs) and mobilized colistin resistance (MCRs), two resistance enzymes that have distinct structures and substrates. We demonstrate that AUR irreversibly abrogates both enzyme activity via the displacement of Zn(II) cofactors from their active sites. We further show that AUR synergizes with antibiotics on killing a broad spectrum of carbapenem and/or COL resistant bacterial strains, and slows down the development of ß-lactam and COL resistance. Combination of AUR and COL rescues all mice infected by Escherichia coli co-expressing MCR-1 and New Delhi metallo-ß-lactamase 5 (NDM-5). Our findings provide potential therapeutic strategy to combine AUR with antibiotics for combating superbugs co-producing MBLs and MCRs.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Auranofin/administration & dosage , Carbapenems/pharmacology , Colistin/pharmacology , Escherichia coli Infections/drug therapy , beta-Lactamase Inhibitors/administration & dosage , Animals , Drug Resistance, Multiple, Bacterial , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Infections/microbiology , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , beta-Lactamases/genetics , beta-Lactamases/metabolism
8.
Biomedica ; 40(Supl. 1): 89-101, 2020 05 01.
Article in English, Spanish | MEDLINE | ID: mdl-32463611

ABSTRACT

INTRODUCTION: Leishmaniasis remains one of the neglected tropical diseases. Repurposing existing drugs has proven to be successful for treating neglected tropical diseases while combination therapy is a strategic alternative for the treatment of infectious diseases. Auranofin, lopinavir/ritonavir, and sorafenib are FDA approved drugs used in the treatment of diverse diseases by acting on different essential biological enzymes. OBJECTIVE: To evaluate the effects of monotherapy and combined therapies with the three drugs against Leishmania infantum. MATERIALS AND METHODS: We compared the leishmanicidal effects of the three drugs on promastigotes in vitro as regards the parasite count, the drug concentration providing a half-maximal response, and the ultrastructural changes of the parasite. We determined the fractional inhibitory concentration index of combined drugs in two ways, as well as the activity of the three drugs together to establish their synergetic effect. RESULTS: The monotherapy with the three drugs was effective with auranofin showing the best leishmanicidal effect (EC50=1.5 µM), whereas sorafinib reduced parasite growth at EC50=2.5 µM. The scanning electron microscopy of promastigotes from all treated media showed distortion in the shape with loss of flagella and bleb formation. Acidocalcinosis was evident by transmission electron microscopy with all treatments suggesting apoptosis. Treatment with lopinavir/ritonavir showed signs of autophagy. The two-way combination of the drugs led to additive interactions while the combination of the three drugs showed synergistic action. CONCLUSION: Each drug when used as monotherapy against Leishmania spp. was effective, but the combination therapy was more effective than the individual drugs due to the additive or synergistic effects.


Introducción. La leishmaniasis sigue siendo una de las enfermedades tropicales desatendidas. La reutilización de medicamentos existentes ha demostrado ser exitosa para tratar enfermedades tropicales desatendidas y la terapia combinada es una alternativa estratégica para el tratamiento de enfermedades infecciosas. Auranofin, lopinavir/ritonavir y sorafenib son medicamentos aprobados por la Food and Drug Administration (FDA) de Estados Unidos utilizados en el tratamiento de diversas enfermedades, pues actúan sobre diferentes enzimas biológicas esenciales. Objetivo. Evaluar los efectos terapéuticos de la monoterapia y de los tres fármacos combinados contra Leishmania infantum. Materiales y métodos. Los efectos leishmanicidas de los tres fármacos sobre los promastigotes se compararon in vitro en cuanto al recuento de parásitos, la concentración del fármaco que proporcionara una respuesta semimáxima y los cambios ultraestructurales del parásito. Se calculó el índice de concentración inhibitoria de fracciones de fármacos combinados de dos maneras y la actividad de los tres fármacos juntos para determinar el efecto sinérgico. Resultados. La monoterapia con los tres medicamentos fue efectiva, pero la auranofina tuvo el mejor efecto antileishmanicida con un CE50 de 1,5 µM, en tanto que el sorafinib redujo el crecimiento del parásito con un CE50 de 2,5 µM. La microscopía electrónica de barrido de promastigotes de todos los medios tratados mostró una distorsión en la forma, con pérdida de flagelos y formación de ampollas. La acidocalcinosis fue evidente por microscopía electrónica de transmisión con todos los tratamientos, lo que sugiere apoptosis. El tratamiento con lopinavir/ritonavir mostró signos de autofagia. La combinación de dos medicamentos condujo a interacciones aditivas, mientras que la combinación de las tres drogas produjo una acción sinérgica. Conclusión. Los tres medicamentos usados como monoterapia contra Leishmania spp. fueron efectivos, pero el tratamiento combinado lo fue en mayor medida debido a los efectos aditivos o sinérgicos.


Subject(s)
Auranofin/administration & dosage , Leishmania infantum/drug effects , Lopinavir/administration & dosage , Ritonavir/administration & dosage , Sorafenib/administration & dosage , Auranofin/pharmacology , Drug Combinations , Drug Synergism , Lopinavir/pharmacology , Parasitology/methods , Ritonavir/pharmacology , Sorafenib/pharmacology
9.
Biochem Pharmacol ; 171: 113714, 2020 01.
Article in English | MEDLINE | ID: mdl-31738894

ABSTRACT

Brain tumours are among the deadliest tumours being highly resistant to currently available therapies. The proliferative behaviour of gliomas is strongly influenced by ion channel activity. Small-conductance calcium-activated potassium (SK/KCa) channels are a family of ion channels that are associated with cell proliferation and cell survival. A combined treatment of classical anti-cancer agents and pharmacological SK channel modulators has not been addressed yet. We used the gold-derivative auranofin to induce cancer cell death by targeting thioredoxin reductases in combination with CyPPA to activate SK channels in neuro- and glioblastoma cells. Combined treatment with auranofin and CyPPA induced massive mitochondrial damage and potentiated auranofin-induced toxicity in neuroblastoma cells in vitro. In particular, mitochondrial integrity, respiration and associated energy generation were impaired. These findings were recapitulated in patient-derived glioblastoma neurospheres yet not observed in non-cancerous HT22 cells. Taken together, integrating auranofin and SK channel openers to affect mitochondrial health was identified as a promising strategy to increase the effectiveness of anti-cancer agents and potentially overcome resistance.


Subject(s)
Auranofin/pharmacology , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Neuroblastoma/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/agonists , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Auranofin/administration & dosage , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Spheroids, Cellular/drug effects , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism
10.
J Cancer Res Clin Oncol ; 145(6): 1495-1507, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31028540

ABSTRACT

PURPOSE: Constructed from a theoretical framework, the coordinated undermining of survival paths in glioblastoma (GBM) is a combination of nine drugs approved for non-oncological indications (CUSP9; aprepitant, auranofin, captopril, celecoxib, disulfiram, itraconazole, minocycline, quetiapine, and sertraline) combined with temozolomide (TMZ). The availability of these drugs outside of specialized treatment centers has led patients to embark on combination treatments without systematic follow-up. However, no experimental data on efficacy using the CUSP9 strategy in GBM have been reported. METHODS: Using patient-derived glioblastoma stem cell (GSC) cultures from 15 GBM patients, we described stem cell properties of individual cultures, determined the dose-response relationships of the drugs in the CUSP9, and assessed the efficacy the CUSP9 combination with TMZ in concentrations clinically achievable. The efficacy was evaluated by cell viability, cytotoxicity, and sphere-forming assays in both primary and recurrent GSC cultures. RESULTS: We found that CUSP9 with TMZ induced a combination effect compared to the drugs individually (p < 0.0001). Evaluated by cell viability and cytotoxicity, 50% of the GSC cultures displayed a high sensitivity to the drug combination. In clinical plasma concentrations, the effect of the CUSP9 with TMZ was superior to TMZ monotherapy (p < 0.001). The Wnt-signaling pathway has been shown important in GSC, and CUSP9 significantly reduces Wnt-activity. CONCLUSIONS: Adding experimental data to the theoretical rationale of CUSP9, our results demonstrate that the CUSP9 treatment strategy can induce a combination effect in both treatment-naïve and pretreated GSC cultures; however, predicting response in individual cultures will require further profiling of GSCs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Animals , Aprepitant/administration & dosage , Aprepitant/pharmacology , Auranofin/administration & dosage , Auranofin/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Captopril/administration & dosage , Captopril/pharmacology , Celecoxib/administration & dosage , Celecoxib/pharmacology , Disulfiram/administration & dosage , Disulfiram/pharmacology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Itraconazole/administration & dosage , Itraconazole/pharmacology , Mice , Mice, SCID , Minocycline/administration & dosage , Minocycline/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Quetiapine Fumarate/administration & dosage , Quetiapine Fumarate/pharmacology , Reproducibility of Results , Sertraline/administration & dosage , Sertraline/pharmacology , Signal Transduction/drug effects , Temozolomide/administration & dosage , Temozolomide/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Invest New Drugs ; 37(6): 1166-1176, 2019 12.
Article in English | MEDLINE | ID: mdl-30825105

ABSTRACT

Small cell lung cancer (SCLC) is a highly lethal malignancy with the 5-year survival rate of less than 7%. Chemotherapy-resistance is a major challenge for SCLC treatment in clinic. In the study, we developed a high-throughput drug screen strategy to identify new drugs that can enhance the sensitivity of chemo-drug cisplatin in SCLC. This screen identified auranofin, a US Food and Drug Administration (FDA)-approved drug used therapeutically for rheumatoid arthritis, as a sensitizer of cisplatin. Further study validated that auranofin synergistically enhanced the anti-tumor activity of cisplatin in chemo-resistant SCLC cells, which was accompanied by the enhanced induction of cell cycle arrest and apoptosis. The synergistic action of auranofin and cisplatin was through ROS overproduction, thereby leading to mitochondrial dysfunction and DNA damage. Furthermore, in vivo study demonstrated that the combination treatment of auranofin and cisplatin dramatically inhibited tumor growth in SCLC. Therefore, our study provides a rational basis for further clinical study to test whether auranofin could enhance the sensitivity of cisplatin-based therapy in SCLC patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Auranofin/administration & dosage , Cisplatin/administration & dosage , Lung Neoplasms/drug therapy , Small Cell Lung Carcinoma/drug therapy , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Humans , Lung Neoplasms/metabolism , Mice, Nude , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Small Cell Lung Carcinoma/metabolism
12.
J Natl Cancer Inst ; 111(6): 597-608, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30779852

ABSTRACT

BACKGROUND: Cancer cells from different origins exhibit various basal redox statuses and thus respond differently to intrinsic or extrinsic oxidative stress. These intricate characteristics condition the success of redox-based anticancer therapies that capitalize on the ability of reactive oxygen species to achieve selective and efficient cancer cell killing. METHODS: Redox biology methods, stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics, and bioinformatics pattern comparisons were used to decipher the underlying mechanisms for differential response of lung and breast cancer cell models to redox-modulating molecule auranofin (AUF) and to combinations of AUF and vitamin C (VC). The in vivo effect of AUF, VC, and two AUF/VC combinations on mice bearing MDA-MB-231 xenografts (n = 5 mice per group) was also evaluated. All statistical tests were two-sided. RESULTS: AUF targeted simultaneously the thioredoxin and glutathione antioxidant systems. AUF/VC combinations exerted a synergistic and hydrogen peroxide (H2O2)-mediated cytotoxicity toward MDA-MB-231 cells and other breast cancer cell lines. The anticancer potential of AUF/VC combinations was validated in vivo on MDA-MB-231 xenografts in mice without notable side effects. On day 14 of treatments, mean (SD) tumor volumes for the vehicle-treated control group and the two AUF/VC combination-treated groups (A/V1 and A/V2) were 197.67 (24.28) mm3, 15.66 (10.90) mm3, and 10.23 (7.30)mm3, respectively; adjusted P values of the differences between mean tumor volumes of vehicle vs A/V1 groups and vehicle vs A/V2 groups were both less than .001. SILAC proteomics, bioinformatics analysis, and functional experiments linked prostaglandin reductase 1 (PTGR1) expression levels with breast cancer cell sensitivity to AUF/VC combinations. CONCLUSION: The combination of AUF and VC, two commonly available drugs, could be efficient against triple-negative breast cancer and potentially other cancers with similar redox properties and PTGR1 expression levels. The redox-based anticancer activity of this combination and the discriminatory potential of PTGR1 expression are worth further assessment in preclinical and clinical studies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Triple Negative Breast Neoplasms/drug therapy , A549 Cells , Animals , Ascorbic Acid/administration & dosage , Auranofin/administration & dosage , Cell Line, Tumor , Female , Glutathione/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Oxidative Stress/drug effects , Proteome/metabolism , Random Allocation , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
13.
Cell Death Dis ; 8(10): e3067, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28981107

ABSTRACT

Rhabdomyosarcoma (RMS) cells have recently been reported to be sensitive to oxidative stress. Therefore, we investigated whether concomitant inhibition of the two main antioxidant defense pathways, that is, the thioredoxin (TRX) and the glutathione (GSH) systems, presents a new strategy to trigger cell death in RMS. In this study, we discover that GSH-depleting agents, i.e. γ-glutamylcysteine synthetase inhibitor, buthionine sulfoximine (BSO) or the cystine/glutamate antiporter inhibitor erastin (ERA), synergize with thioredoxin reductase (TrxR) inhibitor auranofin (AUR) to induce cell death in RMS cells. Interestingly, AUR causes accumulation of ubiquitinated proteins when combined with BSO or ERA, in line with recent reports showing that AUR inhibits the proteasome besides TrxR. Consistently, AUR/BSO or AUR/ERA cotreatment increases ubiquitination and expression of the short-lived proteins NOXA and MCL-1, accompanied by increased binding of NOXA to MCL-1. Notably, NOXA knockdown significantly rescues RMS cells from AUR/BSO- or AUR/ERA-induced cell death. In addition, AUR acts together with BSO or ERA to stimulate BAX/BAK and caspase activation. Of note, BSO or ERA abolish the AUR-stimulated increase in GSH levels, leading to reduced GSH levels upon cotreatment. Although AUR/BSO or AUR/ERA cotreatment enhances reactive oxygen species (ROS) production, only thiol-containing antioxidants (i.e., N-acetylcysteine (NAC), GSH), but not the non-thiol-containing ROS scavenger α-Tocopherol consistently suppress AUR/BSO- and AUR/ERA-stimulated cell death in both cell lines. Importantly, re-supply of GSH or its precursor NAC completely prevents AUR/ERA- and AUR/BSO-induced accumulation of ubiquitinated proteins, NOXA upregulation and cell death, indicating that GSH depletion rather than ROS production is critical for AUR/BSO- or AUR/ERA-mediated cell death. Thus, by demonstrating that GSH-depleting agents enhance the antitumor activity of AUR, we highlight new treatment options for RMS by targeting the redox homeostasis.


Subject(s)
Auranofin/administration & dosage , Glutathione/metabolism , Oxidative Stress/drug effects , Rhabdomyosarcoma/drug therapy , Thioredoxins/metabolism , Antioxidants/metabolism , Apoptosis/drug effects , Buthionine Sulfoximine/administration & dosage , Cell Line, Tumor , Glutathione/antagonists & inhibitors , Humans , Oxidation-Reduction/drug effects , Oxidative Stress/genetics , Piperazines/administration & dosage , Reactive Oxygen Species/metabolism , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxins/antagonists & inhibitors , gamma-Glutamylcyclotransferase/antagonists & inhibitors
14.
Oncol Rep ; 38(4): 2417-2425, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28765909

ABSTRACT

Previously, we reported that auranofin induces apoptosis in SKOV3 cells via regulation of the IKKß/FOXO3 pathway. In the present study, we reveal that the anticancer activity of auranofin in SKOV3 cells could be enhanced by the attenuation of MUC4 through the regulation of the Her2/Akt/FOXO3 pathway. Compared to the control-siRNA, siRNA transfection against MUC4 into SKOV3 cells accelerated the protein degradation of Her2. Under the same conditions, the expression level of phosphorylated Akt was also downregulated leading to an increase of FOXO3 in the nucleus. Notably, auranofin treatment in SKOV3 cells also resulted in the downregulation of the expression levels of both Her2 and phosphorylated Akt. Thus, Her2 was identified as the common molecular target protein by siRNA transfection against MUC4. Western blot analysis of total and nuclear fraction lysates from SKOV3 cells revealed that attenuation of MUC4 combined with auranofin treatment in SKOV3 cells synergistically activated FOXO3 translocation from the cytoplasm to the nucleus through the regulation of the Her2/Akt/FOXO3 pathway. Attenuation of MUC4 by siRNA transfection potentiated the antitumor effect of auranofin which was examined by performing in vitro assays such as WST-1, cell counting, colony formation, TUNEL and Annexin V staining. In addition, western blot analysis of the apoptosis­related proteins such as PARP1, caspase-3, Bim extra large (EL), Bax and Bcl2 revealed that the attenuation of MUC4 by siRNA transfection potentiates the pro-apoptotic activity of auranofin in SKOV3 cells. Collectively, auranofin could regulate the Her2/Akt/FOXO3 signaling pathway in SKOV3 cells and be used as a potential antitumor agent considering the expression of MUC4 in ovarian cancer patients.


Subject(s)
Forkhead Box Protein O3/genetics , Mucin-4/genetics , Ovarian Neoplasms/drug therapy , Receptor, ErbB-2/genetics , Apoptosis/drug effects , Auranofin/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/genetics , Signal Transduction , Transfection
15.
Gen Physiol Biophys ; 36(2): 117-128, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28218611

ABSTRACT

Mammalian thioredoxin reductase (TrxR) plays a vital role in restoring cellular redox balance disrupted by reactive oxygen species (ROS) generation and oxidative damage. Here, we evaluated whether auranofin, a selective inhibitor of TrxR, could serve as a potential anti-cancer agent through its selective targeting of TrxR activity in Hep3B hepatocellular carcinoma cells. Auranofin treatment reduced the TrxR activity of these cells and induced apoptosis, which were accompanied by up-regulation of death receptors (DRs) and activation of caspases, as well as promotion of proteolytic degradation of poly(ADP-ribose)-polymerase. Treatment with a pan-caspase inhibitor reversed the auranofin-induced apoptosis and growth suppression, indicating that auranofin may induce apoptosis through a caspase-dependent mechanism involving both the intrinsic and extrinsic apoptotic pathways. Auranofin also significantly altered mitochondrial function, promoting mitochondrial membrane permeabilization and cytochrome c release by regulating Bcl-2 family proteins; these events were accompanied by an accumulation of ROS. Inhibition of ROS generation with the ROS quencher significantly attenuated the inactivation of TrxR in auranofin-treated cells and almost completely suppressed the auranofin-induced up-regulation of DRs and activation of caspases, thereby preventing auranofin-induced apoptosis and loss of cell viability. Taken together, these findings indicate that auranofin inhibition of TrxR activity in Hep3B cells activates ROS- and caspase-dependent apoptotic signaling pathways and triggers cancer cell death.


Subject(s)
Apoptosis/drug effects , Auranofin/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Reactive Oxygen Species/metabolism , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Thioredoxin-Disulfide Reductase/metabolism , Treatment Outcome
16.
Antioxid Redox Signal ; 27(2): 106-114, 2017 07 10.
Article in English | MEDLINE | ID: mdl-27733046

ABSTRACT

AIMS: The glutathione (GSH), thioredoxin (Trx), and Nrf2 systems represent a major defense against reactive oxygen species (ROS), the cellular imbalance of which in cancer promotes growth and therapeutic resistance. This study investigated whether targeting the GSH, Trx, and Nrf2 antioxidant systems effectively eliminated head and neck cancer (HNC). RESULTS: At high concentrations, auranofin, but not buthionine sulfoximine (BSO) alone, decreased the viability of HNC, whereas even at low concentrations, auranofin plus BSO synergized to kill HNC cells. Dual silencing of the genes for GCLM and TrxR1 induced GSH depletion, Trx activity inhibition, and ROS accumulation, synergistically killing HNC cells. Inhibition of the GSH and Trx systems resulted in activation of the Nrf2-antioxidant response element (ARE) pathway, which may result in suboptimal GSH and Trx inhibition where HNC is resistant. Genetic inhibition of Nrf2 and/or HO-1 or trigonelline enhanced growth suppression, ROS accumulation, and cell death from GSH and Trx inhibition. The in vivo effects of GSH, Trx, and Nrf2 system inhibition were confirmed in a mouse HNC xenograft model by achieving growth inhibition >60% compared with those of control. Innovations: This study is the first to show that triple inhibition of GSH, Trx, and Nrf2 pathways could be an effective method to overcome the resistance of HNC. CONCLUSIONS: Inhibition of the Nrf2-ARE pathway in addition to dual inhibition of the GSH and Trx antioxidant systems can effectively eliminate resistant HNC. Antioxid. Redox Signal. 27, 106-114.


Subject(s)
Auranofin/administration & dosage , Buthionine Sulfoximine/administration & dosage , Glutathione/metabolism , Head and Neck Neoplasms/drug therapy , NF-E2-Related Factor 2/metabolism , Thioredoxins/metabolism , Animals , Auranofin/pharmacology , Buthionine Sulfoximine/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/metabolism , Humans , Mice , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
17.
Support Care Cancer ; 25(3): 833-838, 2017 03.
Article in English | MEDLINE | ID: mdl-27838777

ABSTRACT

PURPOSE: Paclitaxel causes the paclitaxel-induced acute pain (PIAP) syndrome. Based on preclinical data, we hypothesized that the protein kinase C (PKC) iota inhibitor, auranofin (a gold salt used for other pain conditions), palliates this pain. METHODS: In a randomized, double-blinded manner, patients who had suffered this syndrome were assigned a one-time dose of auranofin 6 mg orally on day #2 of the chemotherapy cycle (post-paclitaxel) versus placebo. Patients completed the Brief Pain Inventory and a pain diary on days 2 through 8 and at the end of the cycle. The primary endpoint was pain scores, as calculated by area under the curve, in response to "Please rate your pain by circling the one number that best describes your pain at its worse in the last 24 hours." RESULTS: Thirty patients were enrolled. For the primary endpoint, mean area under the curve of 55 units (standard deviation 19) and 61 units (standard deviation 22) were observed in auranofin-treated and placebo-exposed patients, respectively (p = 0.44). On day 8 and at the end of the cycle, pain scores in auranofin-treated patients were more favorable, although differences were not statistically significant. CONCLUSIONS: In the dose schedule studied, auranofin did not palliate the PIAP syndrome, but delayed beneficial trends suggest further study for this indication.


Subject(s)
Acute Pain/chemically induced , Acute Pain/drug therapy , Auranofin/administration & dosage , Isoenzymes/antagonists & inhibitors , Paclitaxel/adverse effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , Acute Pain/enzymology , Administration, Oral , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/therapeutic use , Double-Blind Method , Female , Humans , Male , Middle Aged , Paclitaxel/therapeutic use , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Syndrome
18.
PLoS One ; 11(11): e0166386, 2016.
Article in English | MEDLINE | ID: mdl-27846303

ABSTRACT

Abnormal survival of retinal pigment epithelium (RPE) cells contributes to the pathogenesis of proliferative vitreoretinopathy (PVR), a sight-threatening disease. In this study, we explored the effect of the anti-rheumatic agent auranofin (AF) on RPE cell survival and studied the underlying signaling mechanisms in vitro. Our results showed that AF inhibited ARPE-19 cell survival in a dose and time-dependent manner. Application of AF induced several effects: a significant decrease in total epidermal growth factor receptor (EGFR) and an increase in phosphorylated EGFR and mitogen-activated protein kinase (MAPK), including extracellular signal-regulated kinase (ERK), P38 mitogen-activated protein kinase (P38MAPK), c-Jun N-terminal kinase (JNK), c-Jun, mitogen activated protein kinase activated protein kinase 2(MAPKAPK2), and heat shock protein 27 (HSP27). AF also inhibited epidermal growth factor (EGF)-dependent cell proliferation and migration through affecting EGFR/MAPK signaling. The antioxidant N-acetylcysteine (NAC) blocked the AF-induced increase of reactive oxygen species (ROS) production, the reduction of total EGFR, and the phosphorylation of multiple nodes in EGFR/MAPK signaling pathway. P38MAPK inhibitor SB203580, but not inhibitors of EGFR (erlotinib), ERK (FR180204) and JNK (SP600125), suppressed AF-induced phosphorylation of EGFR/p38MAPK/MAPKAPK2/Hsp27. In conclusion, the ROS-dependent phosphorylation of EGFR/MAPK is an important signaling pathway for AF-induced inhibition of RPE cell survival, and AF may have the potential for treatment of abnormal survival of RPE cells in PVR.


Subject(s)
Auranofin/administration & dosage , ErbB Receptors/genetics , Retinal Pigment Epithelium/drug effects , Vitreoretinopathy, Proliferative/drug therapy , p38 Mitogen-Activated Protein Kinases/genetics , Cell Line , Cell Proliferation/drug effects , Cell Survival , Humans , MAP Kinase Signaling System/drug effects , Phosphorylation , Reactive Oxygen Species/metabolism , Vitreoretinopathy, Proliferative/genetics , Vitreoretinopathy, Proliferative/pathology
19.
Free Radic Biol Med ; 96: 334-46, 2016 07.
Article in English | MEDLINE | ID: mdl-27101737

ABSTRACT

The most common methods of measuring mitochondrial hydrogen peroxide production are based on the extramitochondrial oxidation of a fluorescent probe such as amplex ultra red (AUR) by horseradish peroxidase (HRP). These traditional HRP-based assays only detect H2O2 that has escaped the matrix, raising the potential for substantial underestimation of production if H2O2 is consumed by matrix antioxidant pathways. To measure this underestimation, we characterized matrix consumers of H2O2 in rat skeletal muscle mitochondria, and developed specific means to inhibit these consumers. Mitochondria removed exogenously added H2O2 (2.5µM) at rates of 4.7 and 5.0nmol min(-1) mg protein(-1) when respiring on glutamate+malate and succinate+rotenone, respectively. In the absence of respiratory substrate, or after disrupting membranes by cycles of freeze-thaw, rates of H2O2 consumption were negligible. We concluded that matrix consumers are respiration-dependent (requiring respiratory substrates), suggesting the involvement of either the thioredoxin (Trx) and/or glutathione (GSH)-dependent enzymatic pathways. The Trx-reductase inhibitor auranofin (2µM), and a pre-treatment of mitochondria with 35µM of 1-chloro-2,4-dintrobenzene (CDNB) to deplete GSH specifically compromise these two consumption pathways. These inhibition approaches presented no undesirable "off-target" effects during extensive preliminary tests. These inhibition approaches independently and additively decreased the rate of consumption of H2O2 exogenously added to the medium (2.5µM). During traditional HRP-based H2O2 efflux assays, these inhibition approaches independently and additively increased apparent efflux rates. When used in combination (double inhibition), these inhibition approaches allowed accumulation of (endogenously produced) H2O2 in the medium at a comparable rate whether it was measured with an end point assay where 2.5µM H2O2 is initially added to the medium or with traditional HRP-based efflux assays. This finding confirms that a high degree of inhibition of all matrix consumers is attained with the double inhibition. Importantly, this double inhibition of the matrix consumers allowed revealing that a large part of the H2O2 produced in muscle mitochondria is consumed before escaping the matrix during traditional HRP-based efflux assays. The degree of this underestimation was substrate dependent, reaching >80% with malate, which complicates comparisons of substrates for their capacity to generate H2O2 in normal conditions i.e. when matrix consumers are active. Our results also urge caution in interpreting changes in H2O2 efflux in response to a treatment; when HRP-based assays are used, large changes in apparent H2O2 efflux may come from altered capacity of the matrix consumers.


Subject(s)
Glutathione/metabolism , Hydrogen Peroxide/metabolism , Mitochondria, Muscle/metabolism , Thioredoxins/metabolism , Animals , Antioxidants/metabolism , Auranofin/administration & dosage , Dinitrochlorobenzene/administration & dosage , Humans , Oxidative Stress/genetics , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
20.
Cell Cycle ; 15(4): 559-72, 2016.
Article in English | MEDLINE | ID: mdl-26743692

ABSTRACT

Multiple myeloma (MM) is a B-cell malignancy characterized by an accumulation of abnormal clonal plasma cells in the bone marrow. Introduction of the proteasome-inhibitor bortezomib has improved MM prognosis and survival; however hypoxia-induced or acquired bortezomib resistance remains a clinical problem. This study highlighted the role of thioredoxin reductase 1 (TrxR1) in the hypoxia-induced and acquired bortezomib resistance in MM. Higher TrxR1 gene expression correlated with high-risk disease, adverse overall survival, and poor prognosis in myeloma patients. We demonstrated that hypoxia induced bortezomib resistance in myeloma cells and increased TrxR1 protein levels. Inhibition of TrxR1 using auranofin overcame hypoxia-induced bortezomib resistance and restored the sensitivity of hypoxic-myeloma cells to bortezomib. Hypoxia increased NF-Ðºß subunit p65 nuclear protein levels and TrxR1 inhibition decreased hypoxia-induced NF-Ðºß p65 protein levels in the nucleus and reduced the expression of NF-кß-regulated genes. In addition, higher TrxR1 protein levels were observed in bortezomib-resistant myeloma cells compared to the naïve cells, and its inhibition using either auranofin or TrxR1-specific siRNAs reversed bortezomib resistance. TrxR1 inhibition reduced p65 mRNA and protein expression in bortezomib-resistant myeloma cells, and also decreased the expression of NF-кß-regulated anti-apoptotic and proliferative genes. Thus, TrxR1 inhibition overcomes both hypoxia-induced and acquired bortezomib resistance by inhibiting the NF-Ðºß signaling pathway. Our findings demonstrate that elevated TrxR1 levels correlate with the acquisition of bortezomib resistance in MM. We propose considering TrxR1-inhibiting drugs, such as auranofin, either for single agent or combination therapy to circumvent bortezomib-resistance and improve survival outcomes of MM patients.


Subject(s)
Drug Resistance, Neoplasm/genetics , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Thioredoxin Reductase 1/genetics , Apoptosis/drug effects , Auranofin/administration & dosage , Bortezomib/administration & dosage , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Multiple Myeloma/pathology , NF-kappa B/genetics , Proteasome Inhibitors/administration & dosage , Signal Transduction/drug effects , Thioredoxin Reductase 1/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...