Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 395
Filter
1.
Biochem Pharmacol ; 212: 115537, 2023 06.
Article in English | MEDLINE | ID: mdl-37019184

ABSTRACT

Lung cancer is characterized by high incidence and mortality. 90% of cancer deaths are caused by metastases. The epithelial-mesenchymal transition (EMT) process in cancer cells is a prerequisite for the metastatic process. Ethacrynic acid (ECA) is a loop diuretic that inhibits the EMT process in lung cancer cells. EMT has been related to the tumour immunemicroenvironment. However, the effect of ECA on immune checkpoint molecules in the context of cancer has not been fully identified. In the present study, we found that sphingosylphosphorylcholine (SPC) and TGF-ß1, awell-known EMT inducer, induced the expression of B7-H4 in lung cancer cells. We also investigated the involvement of B7-H4 in the SPC-induced EMT process. Knockdown of B7-H4 suppressed SPC-induced EMT, while B7-H4 overexpression enhanced EMT of lung cancer cells. ECA inhibited SPC/TGF-ß1-induced B7-H4 expression via suppression of STAT3 activation. Moreover, ECA inhibits the colonization of mice lung by tail vein-injected LLC1 cells. ECA-treated mice increased the CD4-positive T cells in lung tumour tissues. In summary, these results suggested that ECA inhibits B7-H4 expression via STAT3 inhibition, leading to SPC/TGF-ß1-induced EMT. Therefore, ECA might be an immune oncological drug for B7-H4-positive cancer, especially lung cancer.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Animals , Mice , Transforming Growth Factor beta1/metabolism , Ethacrynic Acid/pharmacology , Ethacrynic Acid/therapeutic use , Epithelial-Mesenchymal Transition , Cell Line, Tumor , Cell Movement , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism
2.
Dis Markers ; 2021: 5530673, 2021.
Article in English | MEDLINE | ID: mdl-34122668

ABSTRACT

Background. Despite afatinib as a new first-line treatment for EGFR L858R and exon 19 deletion or other rare EGFR-mutation patients, the acquired resistance or toxic effects associated with it limited its use clinically. The controlling of acquired resistance or optimization of the afatinib dosage in EGFR/T790M mutation-positive non-small-cell lung cancer (NSCLC) is still an important fundamental problem. Ethacrynic acid (EA) has been proved as a dual inhibitor of GST and WNT, and the α, ß-unsaturated-keto structure of it is similar to that of irreversible tyrosine kinase inhibitors (TKIs). However, these beneficial effects of EA combined with afatinib have never been reported in NSCLC. Therefore, the antitumor effects of afatinib combined with EA in EGFR L858R/T790M-mutated NSCLC cells and related mechanisms were analyzed. Our in vitro and in vivo results showed that EA has strong synergistic antitumor effects with afatinib in EGFR L858R/T790M-mutated NSCLC cells, but has no cytotoxic effects in NSCLC cells when used it alone, i.e., the cytotoxic effects of afatinib (IC30) plus EA (IC30) were stronger than the effects of afatinib (IC50) alone. Our functional studies found that the antitumor mechanisms of afatinib when combined with EA mainly occurred by inhibiting WNT/ß-catenin pathway activation and suppression of the secretion of anti-inflammatory factors. These results revealed that combination of afatinib with EA derivatives not only provided a new therapeutic approach for EGFR/T790M-mutated NSCLC patients but also offered a new idea for developing new drugs or optimizing the dose of afatinib in clinical use in future antitumor therapy.


Subject(s)
Afatinib/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Ethacrynic Acid/pharmacology , Lung Neoplasms/drug therapy , Wnt Signaling Pathway/drug effects , A549 Cells , Afatinib/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance, Neoplasm/genetics , Drug Synergism , Ethacrynic Acid/therapeutic use , Genes, erbB-1 , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mutation , Random Allocation
3.
Biochem Pharmacol ; 183: 114339, 2021 01.
Article in English | MEDLINE | ID: mdl-33189676

ABSTRACT

Lung cancer is one of the leading causes of death in cancer patients. Epithelial-mesenchymal transition (EMT) plays an important role in lung cancer progression. Therefore, for lung cancer treatment, it is crucial to find substances that inhibit EMT. Ethacrynic acid (ECA) is a diuretic that inhibits cellular ion flux and exerts anticancer effects. However, the effects of ECA on EMT in lung cancer remain unclear. We examined the effects of ECA on sphingosylphosphorylcholine (SPC) or TGF-ß1-induced EMT process in A549 and H1299 cells via reverse transcription polymerase chain reaction and Western blotting. We found that ECA inhibited SPC-induced EMT and SPC-induced WNT signalling in EMT. We observed that SPC induces the expression of NDP [Norrie disease protein] and WNT-2, whereas ECA suppressed their expression. SPC-induced WNT activation, EMT, migration, and invasion were suppressed by NDP small-interfering RNA (siNDP), but NDP overexpression (pNDP) enhanced these events in A549 and H1299 cells. Accordingly, NDP expression may influence lung cancer prognosis. In summary, our results revealed that ECA inhibited SPC or TGF-ß1-induced EMT in A549 and H1299 lung cancer cells by downregulating NDP expression and inhibiting WNT activation. Therefore, ECA might be a new drug candidate for lung cancer treatment.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Ethacrynic Acid/pharmacology , Eye Proteins/pharmacology , Lung Neoplasms/metabolism , Nerve Tissue Proteins/pharmacology , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Wnt Signaling Pathway/drug effects , A549 Cells , Animals , Cell Movement/drug effects , Cell Movement/physiology , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/physiology , Ethacrynic Acid/therapeutic use , Eye Proteins/antagonists & inhibitors , Eye Proteins/biosynthesis , Humans , Lung Neoplasms/drug therapy , Male , Mice , Mice, Inbred NOD , Mice, SCID , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , RNA, Small Interfering/pharmacology , Sodium Potassium Chloride Symporter Inhibitors/therapeutic use , Wnt Signaling Pathway/physiology
4.
Chemistry ; 26(72): 17525-17535, 2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33252170

ABSTRACT

A facile route to PtII complexes doubly functionalized with bioactive molecules through a bipyridine-type ligand is described. Initially, ligands LEE (containing two ethacrynic acid units), LEF (ethacrynic acid+flurbiprofen) and LEB (ethacrynic acid+biotin) were obtained in moderate to good yields from 2,2'-bipyridine-4,4'-dicarboxylic acid. Subsequent reaction of the ligands with [PtCl2 (DMSO)2 ] afforded complexes [PtCl2 (LEE )] (2), [PtCl2 (LEF )] (3) and [PtCl2 (LEB )] (4) in high yields. All compounds were fully characterized by analytical and spectroscopic methods. Complexes 2-4 are highly stable in water/DMSO solution at 37 °C after 72 h, whereas progressive release of the bioactive fragments was detected in a cell culture medium. The compounds were assessed for their in vitro antiproliferative activity towards tumorigenic A2780, A2780cisR and Y79 cells and non-tumourigenic HEK293 cells. In particular, the combination of ethacrynic acid and flurbiprofen in 3 overcomes cisplatin-based resistance and provides strong cancer cell selectivity. Enzyme inhibition assays on human GST P1 and human COX-2 and cross-experiments with complex 1, analogous to 2-4 but lacking bio-groups, revealed a clear synergy between the PtII frame and the bioactive organic components.


Subject(s)
2,2'-Dipyridyl/chemistry , Antineoplastic Agents , Cisplatin/pharmacology , Ethacrynic Acid/pharmacology , Ovarian Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cisplatin/therapeutic use , Ethacrynic Acid/therapeutic use , Female , Flurbiprofen/therapeutic use , HEK293 Cells , Humans , Ovarian Neoplasms/drug therapy
5.
Biochem Pharmacol ; 175: 113920, 2020 05.
Article in English | MEDLINE | ID: mdl-32201212

ABSTRACT

To identify signal transducer and activator of transcription factor 3 (STAT3) inhibitors, we generated STAT3-dependent gene expression signature by analyzing gene expression profiles of DU145 cancer cells treated with STAT3 inhibitor, piperlongumine and 2-hydroxycinnamaldehyde. Then we explored gene expression signature-based strategies using a connectivity map database and identified several STAT3 inhibitors, including ethacrynic acid (EA). EA is currently used as a diuretic drug. EA inhibited STAT3 activation in DU145 prostate cancer cells and consequently decreased the levels of STAT3 target genes such as cyclin A and MCL-1. Furthermore, EA treatment inhibited tumor growth in mice xenografted with DU145 cells and decreased p-STAT3 expression in tumor tissues. Knockdown of Src homology region 2 domain-containing phosphatase-2 (SHP2) or Protein tyrosine phosphatase 1B (PTP1B) gene expression by siRNA suppressed the ability of EA to inhibit STAT3 activation. When EA was combined with an activator of SHP2 or PTP1B, p-STAT3 expression was synergistically decreased; when EA was combined with an inhibitor of SHP2 or PTP1B, p-STAT3 expression was rescued. By using an affinity pulldown assay with biotinyl-EA, EA was shown to associate with SHP2 and PTP1B in vitro. Additionally, the drug affinity responsive target stability (DARTS) assay confirmed the direct binding of EA to SHP2 and PTP1B. SHP2 is activated by EA through active phosphorylation at Y580 and direct binding to SHP2. Collectively, our results suggest that EA inhibits STAT3 activity through the modulation of phosphatases such as SHP2 and PTP1B and may be a potential anticancer drug to target STAT3 in cancer progression.


Subject(s)
Ethacrynic Acid/pharmacology , Prostatic Neoplasms/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Ethacrynic Acid/therapeutic use , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Xenograft Model Antitumor Assays/methods
6.
ChemMedChem ; 13(12): 1210-1217, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29637702

ABSTRACT

The cytotoxicity of cisplatin (cDDP) is enhanced when co-administered with ethacrynic acid (EA), a glutathione S-transferase (GST) inhibitor. A PtIV -EA conjugate containing a cDDP core and two axial ethacrynate ligands (compound 1) was shown to be an excellent inhibitor of GST, but did not readily release a PtII species to exert a synergistic cytotoxic effect. In this study, a redesigned PtIV construct composed of a cDDP core with one axial ethacrynate ligand and one axial hydroxido ligand (compound 2) was prepared and shown to overcome the limitations of compound 1. The EA ligand in 2 is readily released in vitro together with a cytotoxic PtII species derived from cisplatin, working together to inhibit cell proliferation in cDDP-resistant human ovarian cancer cells. The in vitro activity translates well in vivo with 2, showing effective (∼80 %) inhibition of tumor growth in a human ovarian carcinoma A2780 tumor model, while showing considerably lower toxicity than cisplatin, thus validating the new design strategy.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Glutathione Transferase/antagonists & inhibitors , Organoplatinum Compounds/therapeutic use , Prodrugs/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chickens , Chorioallantoic Membrane/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Enzyme Assays , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Ethacrynic Acid/analogs & derivatives , Ethacrynic Acid/chemical synthesis , Ethacrynic Acid/pharmacology , Ethacrynic Acid/therapeutic use , Humans , Ligands , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacology , Prodrugs/chemical synthesis , Prodrugs/chemistry , Prodrugs/pharmacology
7.
Biomaterials ; 144: 119-129, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28834763

ABSTRACT

Platinum-based DNA-adducting agents are used extensively in the clinic for cancer chemotherapy. However, the anti-tumor efficacy of these drugs is severely limited by cisplatin resistance, and this can lead to the failure of chemotherapy. One of cisplatin resistance mechanisms is associated with overexpression of glutathione S-transferases (GSTs), which would accelerate the deactivation of cisplatin and decrease its antitumor efficiency. Nanoscale micelles encapsulating ethacraplatin, a conjugate of cisplatin and ethacrynic acid (an effective GSTs inhibitor), can enhance the accumulation of active cisplatin in cancer cells by inhibiting the activity of GSTs and circumventing deactivation of cisplatin. In vitro and in vivo results provide strong evidence that GSTs inhibitor-modified cisplatin prodrug combined with nanoparticle encapsulation favor high effective platinum accumulation, significantly enhanced antitumor efficacy against cisplatin-resistant cancer and decreased system toxicity. It is believed that these ethacraplatin-loaded micelles have the ability of overcoming resistance of cancers toward cisplatin and will improve the prospects for chemotherapy of cisplatin-resistant cancers in the near future.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/therapeutic use , Ethacrynic Acid/analogs & derivatives , Glutathione Transferase/antagonists & inhibitors , Neoplasms/drug therapy , Organoplatinum Compounds/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cisplatin/administration & dosage , Cisplatin/pharmacology , Delayed-Action Preparations/chemistry , Drug Delivery Systems , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Ethacrynic Acid/administration & dosage , Ethacrynic Acid/pharmacology , Ethacrynic Acid/therapeutic use , Female , Glutathione Transferase/metabolism , Humans , Mice, Inbred NOD , Mice, SCID , Micelles , Neoplasms/metabolism , Neoplasms/pathology , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/pharmacology
8.
Oncotarget ; 7(36): 58038-58050, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27487128

ABSTRACT

Prolonged treatment of breast cancer with epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) often results in acquired resistance and a narrow therapeutic index. One strategy to improve the therapeutic effects of EGFR TKIs is to combine them with drugs used for other clinical indications. Ethacrynic acid (EA) is an FDA approved drug that may have antitumor effects and may enhance the cytotoxicity of chemotherapeutic agents by binding to glutathione and inhibiting WNT signaling. While the α,ß-unsaturated-keto structure of EA is similar to that of irreversible TKIs, the mechanism of action of EA when combined with irreversible EGFR TKIs in breast cancer remains unknown. We therefore investigated the combination of irreversible EGFR TKIs and EA. We found that irreversible EGFR TKIs and EA synergistically inhibit breast cancer both in vitro and in vivo. The combination of EGFR TKIs and EA induces necrosis and cell cycle arrest and represses WNT/ß-catenin signaling as well as MAPK-ERK1/2 signaling. We conclude that EA synergistically enhances the antitumor effects of irreversible EGFR TKIs in breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Ethacrynic Acid/pharmacology , Protein Kinase Inhibitors/pharmacology , Afatinib , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Breast/pathology , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Ethacrynic Acid/therapeutic use , Female , Humans , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Mice, Inbred BALB C , Protein Kinase Inhibitors/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Quinolines/pharmacology , Quinolines/therapeutic use , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays
10.
BMJ Case Rep ; 20152015 Mar 05.
Article in English | MEDLINE | ID: mdl-25743872

ABSTRACT

Patients with glucose-6-phosphate dehydrogenase (G6PD) deficiency are not represented in clinical trials for heart failure. Moreover, many of the recommended medications can cause haemolysis in this group of patients. We present the case of a 71-year-old woman with G6PD deficiency admitted for acute non-ischemic heart failure with reduced ejection fraction. Our experience showed that a combination of ethacrynic acid and spironolactone is safe and effective for relief of volume overload in this group of patients. Studies are needed to determine whether the morbidity and mortality benefits of established heart failure regimens extend to patients with G6PD deficiency.


Subject(s)
Glucosephosphate Dehydrogenase Deficiency/complications , Heart Failure/drug therapy , Heart Failure/etiology , Aged , Diuretics/therapeutic use , Drug Therapy, Combination , Ethacrynic Acid/therapeutic use , Female , Humans , Losartan/therapeutic use , Metoprolol/therapeutic use , Spironolactone/therapeutic use
11.
Crit Care ; 19: 2, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25563826

ABSTRACT

INTRODUCTION: Clinical effects of furosemide (F) and ethacrynic acid (EA) continuous infusion on urine output (UO), fluid balance, and renal, cardiac, respiratory, and metabolic function were compared in infants undergoing surgery for congenital heart diseases. METHODS: A prospective randomized double-blinded study was conducted. Patients received 0.2 mg/kg/h (up to 0.8 mg/kg/h) of either F or EA. RESULTS: In total, 38 patients were enrolled in the F group, and 36, in the EA group. No adverse reactions were recorded. UO at postoperative day (POD) 0 was significantly higher in the EA group, 6.9 (3.3) ml/kg/h, compared with the F group, 4.6 (2.3) ml/kg/h (P = 0.002) but tended to be similar in the two groups thereafter. Mean administered F dose was 0.33 (0.19) mg/kg/h compared with 0.22 (0.13) mg/kg/h of EA (P < 0.0001). Fluid balance was significantly more negative in the EA group at postoperative day 0: -43 (54) ml/kg/h versus -17 (32) ml/kg/h in the F group (P = 0.01). Serum creatinine, cystatin C and neutrophil gelatinase-associated lipocalin levels and incidence of acute kidney injury did not show significant differences between groups. Metabolic alkalosis occurred frequently (about 70% of cases) in both groups, but mean bicarbonate level was higher in the EA group: 27.8 (1.5) M in the F group versus 29.1 (2) mM in the EA group (P = 0.006). Mean cardiac index (CI) values were 2.6 (0.1) L/min/m(2) in the F group compared with 2.98 (0.09) L/min/m(2) in the EA group (P = 0.0081). Length of mechanical ventilation was shorter in the EA group, 5.5 (8.8) days compared with the F group, 6.7 (5.9) (P = 0.06). Length of Pediatric Cardiac Intensive Care Unit (PCICU) admission was shorter in the EA group: 14 (19) days compared with 16 (15) in the F group (P = 0.046). CONCLUSIONS: In cardiac surgery infants, EA produced more UO compared with F on POD0. Generally, a smaller EA dose is required to achieve similar UO than F. EA and F were safe in terms of renal function, but EA caused a more-intense metabolic alkalosis. EA patients achieved better CI, and shorter mechanical ventilation and PCICU admission time. TRIAL REGISTRATION: Clinicaltrials.gov NCT01628731. Registered 24 June 2012.


Subject(s)
Cardiac Surgical Procedures/methods , Diuretics/therapeutic use , Ethacrynic Acid/therapeutic use , Furosemide/therapeutic use , Heart Defects, Congenital/surgery , Double-Blind Method , Female , Humans , Infant , Infant, Newborn , Intention to Treat Analysis , Kidney/drug effects , Kidney/physiology , Male , Postoperative Period , Prospective Studies , Urine
12.
Am J Perinatol ; 32(1): 49-56, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24801161

ABSTRACT

OBJECTIVE: Diuretics are often prescribed off-label to premature infants, particularly to prevent or treat bronchopulmonary dysplasia. We examined their use and safety in this group. STUDY DESIGN: Retrospective cohort study of infants < 32 weeks gestation and < 1,500 g birth weight exposed to diuretics in 333 neonatal intensive care units from 1997 to 2011. We examined use of acetazolamide, amiloride, bumetanide, chlorothiazide, diazoxide, ethacrynic acid, furosemide, hydrochlorothiazide, mannitol, metolazone, or spironolactone combination. Respiratory support and fraction of inspired oxygen on the first day of each course of diuretic use were identified. RESULTS: About 37% (39,357/107,542) infants were exposed to at least one diuretic; furosemide was the most commonly used (93% with ≥ 1 recorded dose), followed by spironolactone, chlorothiazide, hydrochlorothiazide, bumetanide, and acetazolamide. About 74% patients were exposed to one diuretic at a time, 19% to two diuretics simultaneously, and 6% to three diuretics simultaneously. The most common combination was furosemide/spironolactone, followed by furosemide/chlorothiazide and chlorothiazide/spironolactone. Many infants were not receiving mechanical ventilation on the first day of each new course of furosemide (47%), spironolactone (69%), chlorothiazide (61%), and hydrochlorothiazide (68%). Any adverse event occurred on 42 per 1,000 infant-days for any diuretic and 35 per 1,000 infant-days for furosemide. Any serious adverse event occurred in 3.8 for any diuretic and 3.2 per 1,000 infant-days for furosemide. The most common laboratory abnormality associated with diuretic exposure was thrombocytopenia. CONCLUSION: Despite no Food and Drug Administration (FDA) indication and little safety data, over one-third of premature infants in our population were exposed to a diuretic, many with minimal respiratory support.


Subject(s)
Bronchopulmonary Dysplasia/prevention & control , Diuretics/therapeutic use , Respiration, Artificial/statistics & numerical data , Thrombocytopenia/epidemiology , Acetazolamide/therapeutic use , Amiloride/therapeutic use , Bronchopulmonary Dysplasia/drug therapy , Chlorothiazide/therapeutic use , Cohort Studies , Diazoxide/therapeutic use , Drug Therapy, Combination , Ethacrynic Acid/therapeutic use , Female , Furosemide/therapeutic use , Humans , Hydrochlorothiazide/therapeutic use , Infant, Extremely Premature , Infant, Newborn , Infant, Premature , Male , Mannitol/therapeutic use , Metolazone/therapeutic use , Off-Label Use , Retrospective Studies , Spironolactone/therapeutic use
13.
Leuk Res ; 36(5): 598-600, 2012 May.
Article in English | MEDLINE | ID: mdl-22386728

ABSTRACT

It was recently confirmed that the diuretic agent ethacrynic acid (EA) inhibits Wnt/beta catenin signaling in myeloma. This study investigated the antitumor effect of EA in vivo in a murine myeloma model. In vivo, tumor growth was significantly reduced and overall survival significantly prolonged in mice treated with EA as compared to untreated mice. Interestingly, this effect was higher as compared to the effect by lenalidomide, a commonly used drug against myeloma. These results reveal a significant in vivo effect by EA against myeloma.


Subject(s)
Diuretics/therapeutic use , Ethacrynic Acid/therapeutic use , Multiple Myeloma/drug therapy , Animals , Cell Line, Tumor , Disease Models, Animal , Drug Therapy, Combination , Ethacrynic Acid/administration & dosage , Humans , Lenalidomide , Mice , Mice, Inbred BALB C , Multiple Myeloma/mortality , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Wnt Signaling Pathway , beta Catenin/physiology
14.
Chemistry ; 17(28): 7806-16, 2011 Jul 04.
Article in English | MEDLINE | ID: mdl-21681839

ABSTRACT

Platinum-based cancer drugs, such as cisplatin, are highly effective chemotherapeutic agents used extensively for the treatment of solid tumors. However, their effectiveness is limited by drug resistance, which, in some cancers, has been associated with an overexpression of pi class glutathione S-transferase (GST P1-1), an important enzyme in the mercapturic acid detoxification pathway. Ethacraplatin (EA-CPT), a trans-Pt(IV) carboxylate complex containing ethacrynate ligands, was designed as a platinum cancer metallodrug that could also target cytosolic GST enzymes. We previously reported that EA-CPT was an excellent inhibitor of GST activity in live mammalian cells compared to either cisplatin or ethacrynic acid. In order to understand the nature of the drug-protein interactions between EA-CPT and GST P1-1, and to obtain mechanistic insights at a molecular level, structural and biochemical investigations were carried out, supported by molecular modeling analysis using quantum mechanical/molecular mechanical methods. The results suggest that EA-CPT preferentially docks at the dimer interface at GST P1-1 and subsequent interaction with the enzyme resulted in docking of the ethacrynate ligands at both active sites (in the H-sites), with the Pt moiety remaining bound at the dimer interface. The activation of the inhibitor by its target enzyme and covalent binding accounts for the strong and irreversible inhibition of enzymatic activity by the platinum complex.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Glutathione S-Transferase pi/metabolism , Platinum/chemistry , Platinum/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cisplatin/chemistry , Cisplatin/therapeutic use , Crystallography, X-Ray , Dimerization , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/therapeutic use , Ethacrynic Acid/chemistry , Ethacrynic Acid/metabolism , Ethacrynic Acid/therapeutic use , Glutathione S-Transferase pi/chemistry , Glutathione S-Transferase pi/genetics , Humans , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Molecular Structure , Neoplasms/drug therapy , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
15.
Bioorg Med Chem Lett ; 21(3): 912-5, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21227691

ABSTRACT

A series of ethacrynic acid analogues, lacking the α,ß-unsaturated carbonyl unit, was synthesized and subsequently evaluated for their ability to inhibit the migration of human breast cancer cells, Hs578Ts(i)8 as well as of human prostate cancer cells, C4-2B. These cell lines provide a good model system to study migration and invasion, since they represent metastatic cancer. Our studies show that ethacrynic acid analogues with methyl substituents at the aromatic ring demonstrate no inhibitory effect on the migration of both cancer cell lines, whereas a precursor in the synthesis of these ethacrynic acid analogues (II-1, a para-acylated m-cresol) is an excellent inhibitor of the migration of both cancer cell lines.


Subject(s)
Antineoplastic Agents/chemistry , Ethacrynic Acid/analogs & derivatives , Ketones/chemistry , Phenols/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Movement , Ethacrynic Acid/therapeutic use , Ethacrynic Acid/toxicity , Humans , Male , Prostatic Neoplasms/drug therapy
16.
Pharmacol Rep ; 62(5): 808-13, 2010.
Article in English | MEDLINE | ID: mdl-21098864

ABSTRACT

The aim of this study was to determine whether ethacrynic acid (EA), a loop diuretic with anticonvulsant activity, would affect the protective action of the conventional antiepileptics (AEDs) carbamazepine (CBZ), phenytoin (PHT), valproate (VPA) and phenobarbital (PB) in the mouse maximal electroshock seizure (MES) model. The effects of acute and chronic treatment with EA on these AEDs were examined. At a single dose of 100 mg/kg ip, EA enhanced the antielectroshock activity of VPA, decreasing its ED50 value from 225.6 to 146.6 mg/kg (p < 0.05), but enhancement was not observed following continuous administration of EA (12.5 mg/kg) for seven days. Combined treatment of EA with other AEDs had no effect on their ED50 values. The observed interaction between EA and VPA was pharmacodynamic in nature as EA did not alter free plasma (non-protein-bound) and total brain concentrations of VPA. Taking into consideration the clinical use of both drugs, this interaction between EA and VPA can be important for patients receiving these drugs.


Subject(s)
Anticonvulsants/therapeutic use , Diuretics/therapeutic use , Ethacrynic Acid/therapeutic use , Seizures/drug therapy , Animals , Anticonvulsants/pharmacokinetics , Anticonvulsants/pharmacology , Carbamazepine/pharmacokinetics , Carbamazepine/pharmacology , Carbamazepine/therapeutic use , Diuretics/pharmacokinetics , Diuretics/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Electroshock , Ethacrynic Acid/pharmacokinetics , Ethacrynic Acid/pharmacology , Male , Mice , Phenobarbital/pharmacokinetics , Phenobarbital/pharmacology , Phenobarbital/therapeutic use , Phenytoin/pharmacokinetics , Phenytoin/pharmacology , Phenytoin/therapeutic use , Seizures/etiology , Valproic Acid/pharmacokinetics , Valproic Acid/pharmacology , Valproic Acid/therapeutic use
17.
Antivir Chem Chemother ; 20(5): 201-5, 2010 Apr 14.
Article in English | MEDLINE | ID: mdl-20413827

ABSTRACT

BACKGROUND: Aurintricarboxylic acid (ATA) and ethacrynic acid (ECA) have been reported to exhibit antiviral activity against vaccinia virus infections in cell culture by inhibiting early and late gene transcription, respectively. The purpose of this work was to determine if these inhibitors would effectively treat vaccinia virus infections in mice, which has not previously been studied. METHODS: ECA was investigated by cell culture plaque reduction assay for the inhibition of cowpox and vaccinia virus infections to clarify issues regarding its potency and selectivity. Mice infected intranasally with vaccinia virus were treated by intraperitoneal route twice daily for 5 days with ATA (10 and 30 mg/kg/day) and ECA (15 and 30 mg/kg/day) or once daily for 2 days with cidofovir (100 mg/kg/day). RESULTS: ECA caused 50% inhibition of virus plaque formation at 20-79 muM in four cultured cell lines, with 50% cytotoxicity at 84-173 muM, giving low (1.3-4.2) selectivity index values. Preliminary toxicity tests in uninfected mice indicated that ATA and ECA were both overtly toxic at 100 mg/kg/day. No protection from mortality was afforded by treatment of vaccinia virus infections with ATA or ECA, but 100% survival was achieved in the cidofovir group. ATA- and ECA-treated mice died significantly sooner than placebo-treated animals, indicating that these compounds exacerbated the infection. CONCLUSIONS: Both ATA and ECA lack antiviral potency and selectivity in cell culture. The compounds were ineffective in treating mice at intraperitoneal doses of

Subject(s)
Antiviral Agents/pharmacology , Aurintricarboxylic Acid/pharmacology , Ethacrynic Acid/pharmacology , Respiratory Tract Infections/drug therapy , Vaccinia virus/drug effects , Vaccinia/drug therapy , Animals , Antiviral Agents/therapeutic use , Aurintricarboxylic Acid/therapeutic use , Cell Line , Cidofovir , Cytosine/analogs & derivatives , Cytosine/pharmacology , Ethacrynic Acid/therapeutic use , Mice , Mice, Inbred BALB C , Organophosphonates/pharmacology , Respiratory Tract Infections/virology , Time Factors
18.
Am J Ther ; 16(1): 86-92, 2009.
Article in English | MEDLINE | ID: mdl-19142159

ABSTRACT

Ethacrynic acid (Edecrin) is a loop diuretic that produces a prompt and profound diuresis. The primary action of ethacrynic acid is the inhibition of the activity of the Na⁺-K⁺-2Cl⁻ symporter in the thick ascending limb of the loop of Henle. The onset of action is usually within 30 minutes after an oral dose and within 5 minutes after an intravenous injection. After oral administration, peak diuretic effect occurs in about 2 hours and the effect lasts about 6-8 hours. After intravenous administration, peak diuretic effect occurs within 30 minutes and the diuretic effect is virtually completed in 2-4 hours. The bioavailability of ethacrynic acid approximates 100%, with maximal blood level between 40 and 92 minutes. The elimination half-life has been reported to be less than 1 hour, but highly variable (average 30 minutes with a range of 12-160 minutes). Intravenous ethacrynic acid has a prompt venous dilatory effect and immediately relieves symptoms of pulmonary congestion, before a diuresis can occur. Ethacrynic acid is effective in all types of edema whether there is clinical acidosis, alkalosis, or electrolyte imbalance. Most side effects of ethacrynic acid can be attributed to its effectiveness (volume depletion); however, it may cause metabolic alkalosis that is preventable by KCl replacement. Ethacrynic acid has ototoxic effect that occasionally results in temporally or permanent deafness. Despite limitations, ethacrynic acid has been employed in the treatment of congestive heart failure and other edematous states, especially in patients allergic to sulfa-containing drugs because all the other loop diuretics have a sulfa moiety.


Subject(s)
Diuretics/pharmacology , Edema/drug therapy , Ethacrynic Acid/pharmacology , Animals , Biological Availability , Diuretics/adverse effects , Diuretics/therapeutic use , Edema/pathology , Ethacrynic Acid/adverse effects , Ethacrynic Acid/therapeutic use , Half-Life , Heart Failure/drug therapy , Heart Failure/physiopathology , Humans , Time Factors
20.
Acta Paediatr ; 95(2): 214-23, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16449030

ABSTRACT

AIM: To determine relationships between ototoxic drugs and 4-y sensorineural hearing loss (SNHL) in near-term and term survivors of severe neonatal respiratory failure. METHODS: All 81 survivors of the Canadian arm of the Neonatal Inhaled Nitric Oxide Study (mortality 32, loss to follow-up 9) received loop diuretics, aminoglycosides, and neuromuscular blockers (NMB), and 50 received vancomycin as neonates. Prospective, longitudinal secondary outcome using audiological tests diagnosed late-onset, progressive SNHL in 43 (53%); not flat (sloping) in 29, flat (severe to profound) in 14. Risk for SNHL was determined. RESULTS: A combination of duration of diuretic use of >14 d and average NMB dose of >0.96 mg/kg/d contributed to SNHL among survivors (odds ratio 5.2; 95% CI 1.6, 16.7). Markers of illness severity did not contribute. Dosage or duration of aminoglycosides use did not relate to SNHL. Cumulative dosages and duration of use of diuretics; NMB; use of vancomycin; and overlap of diuretics with NMB, aminoglycosides, and vancomycin individually linked to SNHL (p<0.001). CONCLUSION: Overuse of loop diuretics and/or NMB contributes to SNHL after neonatal respiratory failure; markers of illness severity or the appropriate administration of aminoglycosides do not.


Subject(s)
Aminoglycosides/adverse effects , Anti-Infective Agents/adverse effects , Diuretics/adverse effects , Hearing Loss, Sensorineural/chemically induced , Respiratory Insufficiency/drug therapy , Respiratory Insufficiency/physiopathology , Amikacin/adverse effects , Amikacin/therapeutic use , Aminoglycosides/therapeutic use , Anti-Infective Agents/therapeutic use , Audiometry, Pure-Tone/methods , Auditory Threshold/physiology , Diuretics/therapeutic use , Ethacrynic Acid/adverse effects , Ethacrynic Acid/therapeutic use , Female , Furosemide/adverse effects , Furosemide/therapeutic use , Gentamicins/adverse effects , Gentamicins/therapeutic use , Hearing Loss, Sensorineural/diagnosis , Humans , Infant, Newborn , Male , Pancuronium/adverse effects , Pancuronium/therapeutic use , Respiratory Insufficiency/diagnosis , Severity of Illness Index , Tobramycin/adverse effects , Tobramycin/therapeutic use , Vancomycin/adverse effects , Vancomycin/therapeutic use , Vecuronium Bromide/adverse effects , Vecuronium Bromide/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...